At the end of every month, the International Myeloma Foundation Newsroom will feature a wrap-up of some of the most fascinating studies about multiple myeloma from medical journals. Here is the April 2025 edition.
The IMF team of medical editors has provided overviews of key studies. Yet, we encourage you to visit the actual articles in the journals for full details and to increase your understanding. Check the IMF Newsroom monthly for updates like this one.
In the Journals (Key Myeloma Research in April 2025)
"N6-methyladenosine-mediated upregulation of H19 promotes resistance to bortezomib by modulating the miR-184/CARM1 axis in multiple myeloma"
Source
Wang, G., Wu, W., He, D. et al. N6-methyladenosine-mediated upregulation of H19 promotes resistance to bortezomib by modulating the miR-184/CARM1 axis in multiple myeloma. Clin Exp Med 25, 102 (2025). https://doi.org/10.1007/s10238-025-01624-z April 1, 2025.
Overview
Velcade® (bortezomib) is a drug used to treat multiple myeloma (MM), but over time, many patients stop responding to it. Scientists are still trying to understand why this happens.
Recent research found that a molecule called METTL3 may play a role. METTL3 adds a special chemical tag (called m6A) to another molecule called lncRNA H19. This change helps MM cells resist bortezomib. H19 also blocks the activity of a tumor-fighting molecule called miR-184. Normally, miR-184 helps stop cancer growth by lowering the levels of a protein called CARM1.
The study shows that the METTL3–H19–miR-184–CARM1 pathway is important in how MM cells respond to bortezomib. Targeting this pathway may help make BTZ work better for patients with MM.
"Individualized dynamic risk assessment and treatment selection for multiple myeloma"
Source
Murie, C., Turkarslan, S., Patel, A.P. et al. Individualized dynamic risk assessment and treatment selection for multiple myeloma. Br J Cancer (2025). https://doi.org/10.1038/s41416-025-02987-6 April 1, 2025.
Overview
Doctors need to make personalized treatment plans for people with multiple myeloma (MM), but this can be hard because the disease behaves differently depending on a person’s genetic makeup.
Researchers created a tool called mmSYGNAL that uses computer analysis and genetic data from MM patients to better understand how the disease progresses. They used machine learning to build models that predict how high a patient’s risk is and how well they might respond to certain treatments—based on their unique genetic features.
When tested on over 1,300 patients, these models worked better than current methods for predicting how long patients stay free from disease after diagnosis, transplant, and even after relapses. The models also matched patients to drugs that were most likely to work for them, including those used in standard treatment and clinical trials.
This new tool can help doctors make more accurate, personalized treatment decisions for MM patients at every stage of the disease.
"Treatment of myeloma bone disease: When, how often, and for how long?"
Source
Michael Tveden Gundesen, Fredrik Schjesvold, Thomas Lund, Treatment of myeloma bone disease: When, how often, and for how long?, Journal of Bone Oncology, 2025, 100680,ISSN 2212-1374, https://doi.org/10.1016/j.jbo.2025.100680. April 1, 2025.
Overview
The way doctors treat multiple myeloma (MM) has changed a lot in recent years. New and better treatments are helping patients live longer and go into deeper remission. Because of this, patients may need bone-protecting treatments for a longer time—or possibly not as much.
Better imaging tools now make it easier to find bone damage, and updated guidelines (called SLiM-CRAB) have changed how bone disease is measured in clinical trials.
New research also compares two bone-protecting drugs: denosumab and zoledronic acid (ZOL). Studies are looking at how long patients should stay on these treatments, how often they should get them, what happens if treatment is stopped, and whether it's helpful to take a break from the drug before dental work.
As treatments improve and patients live longer, doctors are rethinking how best to protect bones in people with MM.
"Acevaltrate Overcomes Myeloma Resistance to Bortezomib via Pyroptosis by Promoting BAX Translocalization to Mitochondria"
Source
Yaner Wang, Yaoli Cui, Ziyang Liu, Longlong Liu, Zhenqian Huang, Qi Wang, Xinliang Mao, Acevaltrate Overcomes Myeloma Resistance to Bortezomib via Pyroptosis by Promoting BAX Translocalization to Mitochondria, European Journal of Pharmacology,2025,177572, ISSN 0014-2999, https://doi.org/10.1016/j.ejphar.2025.177572. April 1, 2025.
Overview
Over time, some myeloma patients may develop resistance to treatment with Velcade (bortezomib). This study shows that acevaltrate, a natural compound from traditional Chinese medicine, may help fight this problem.
Researchers found that acevaltrate kills bortezomib-resistant myeloma cells through a process called pyroptosis, a form of cell death. It does this by activating proteins called Caspase-3 and GSDME. If these proteins are blocked, the cell death doesn’t happen—proving they’re essential to the process.
Acevaltrate also causes stress in the cell by increasing harmful molecules called reactive oxygen species and damaging the mitochondria (the cell’s energy source). It helps release a protein called BAX, which moves to the mitochondria and starts the death process.
Another protein, IFIT3, doesn’t cause cell death on its own, but it makes acevaltrate’s effects even stronger by blocking a protein that normally stops BAX from working.
Acevaltrate not only kills myeloma cells that resist bortezomib, but it also helps make them sensitive to the drug again. Because of its strong effects and safety record, acevaltrate may be a promising new treatment to help overcome drug resistance in multiple myeloma.
"Enhancer Extrachromosomal Circular DNA ANKRD28 Elicits Drug Resistance via POU2F2-Mediated Transcriptional Network in Multiple Myeloma"
Source
B. Chen, J. Liu, Y. Zhang, C. Shi, D. Zhu, G. Zhang, F. Xiao, L. Zhong, M. Zhang, L. G. Ng, H. Huang, T. Lu, J. Hou, Enhancer Extrachromosomal Circular DNA ANKRD28 Elicits Drug Resistance via POU2F2-Mediated Transcriptional Network in Multiple Myeloma. Adv. Sci. 2025, 2415695. https://doi.org/10.1002/advs.202415695 April 1, 2025.
Overview
Multiple myeloma (MM) is hard to cure mainly because cancer cells often become resistant to treatment. Scientists are still working to understand why this happens.
This study looked at small circular pieces of DNA called eccDNAs, which are found outside the chromosomes in cancer cells. These DNA pieces can come from both coding and non-coding parts of the genome. One of these, called eccANKRD28, was found in higher amounts in MM patients who didn’t respond well to standard treatment with bortezomib, lenalidomide, and dexamethasone (VRd).
In lab tests and animal models, raising the levels of eccANKRD28 made the myeloma cells more resistant to treatment. The researchers found that eccANKRD28 acts like a powerful "on switch" (enhancer) that boosts the activity of genes that help cancer cells survive and resist treatment. It works through a protein called POU2F2, which forms a complex with other proteins to activate cancer-promoting genes like IRF4, JUNB, BCL2, and others.
This study shows that eccANKRD28 plays an important role in drug resistance and may be a new target for future MM treatments.
"GART promotes multiple myeloma malignancy via tumor stemness mediated by activating the HSP90α/CDK6/β-catenin axis"
Source
Jinjun Qian, Han Meng, Ze Wang, Yi Sun, Xiaoning Xu, Hui Shi, Cheng Wang, Lianxin Zhou, Xinyu Lv, Ye Yang, Chunyan Gu, GART promotes multiple myeloma malignancy via tumor stemness mediated by activating the HSP90α/CDK6/β-catenin axis, European Journal of Pharmacology, 2025, 177584, ISSN 0014-2999, https://doi.org/10.1016/j.ejphar.2025.177584. April 2, 2025.
Overview
Multiple myeloma (MM) is a type of blood cancer that can become harder to treat as the cancer cells start acting more like stem cells, which helps them grow and survive longer.
This study looked at a protein called GART, which has been linked to cancer growth in other tumors, to see what it does in MM. The researchers found that MM patients with high levels of GART had worse outcomes. In lab and animal studies, GART helped myeloma cells grow faster, form more colonies, and move through the cell cycle more quickly.
They also discovered how GART works: it helps stabilize another protein called HSP90α, which increases the levels of CDK6—a protein that drives cancer cell growth. GART also turns on a growth pathway called Wnt/β-catenin, which boosts the cancer cells’ ability to grow and act like stem cells.
Finally, a drug called pemetrexed (PEM), which blocks GART, was able to slow down tumor growth in animal models.
GART helps myeloma cells grow and become more aggressive, and blocking it could be a promising new treatment for MM.
"Treatment refractoriness and response rates in patients with relapsed/refractory multiple myeloma: a retrospective analysis of real-world data"
Source
Ning Lyu, Zahra Majd, Bilqees Fatima, Zhen Zeng, Hua Chen, Susan Abughosh, Treatment refractoriness and response rates in patients with relapsed/refractory multiple myeloma: a retrospective analysis of real-world data, Cancer Treatment and Research Communications,2025,100921, ISSN 2468-2942, https://doi.org/10.1016/j.ctarc.2025.100921. April 2, 2025.
Overview
Even though treatments for multiple myeloma (MM) have improved, many patients still relapse, and their cancer becomes harder to treat over time. This study looked at real-world data from 2022 to understand how well treatments are working for patients with relapsed or treatment-resistant MM.
Researchers studied 283 adults with MM who had received at least five different types of treatment, including a proteasome inhibitor, an immunomodulatory drug (IMiD), and a CD38-targeting antibody. Most of these patients were heavily pretreated and had tough-to-treat disease.
The study found that:
- Only about 36% of patients responded to treatment.
- The average time that the treatment worked was just over 6 months.
- Many patients were resistant to multiple types of treatment—including penta-refractory (resistant to 5 drug types), which was the most common group.
This study shows that a large number of myeloma patients don’t respond well to treatment after several rounds of therapy. New and better options are urgently needed for these patients.
"Epigenetic reprogramming in multiple myeloma—Challenges and opportunities"
Source
Kumar S, Kats LM, Gruber E. Epigenetic reprogramming in multiple myeloma—Challenges and opportunities. Int J Cancer. 2025; 1-10. doi:10.1002/ijc.35426 April 2, 2025.
Overview
Cancer cells change in many ways to help them grow and survive. Some of these changes happen in their DNA (mutations), while others happen in how their genes are turned on or off—this is called epigenetic reprogramming.
In multiple myeloma (MM), there are many differences between patients and even within the same patient’s cancer cells. These differences can be genetic or epigenetic, and they help the cancer adapt and resist treatment.
This review looks at the most common epigenetic changes seen in myeloma and how they affect the disease. It also explores new ways to treat myeloma by trying to reverse these harmful changes in how genes are controlled.
"Efficacy and Safety of Quadruplet Therapy in Newly Diagnosed Transplant-Eligible Multiple Myeloma: A Systematic Review and Meta-Analysis"
Source
Channar, A., Naqvi, S.A.A., Khan, M.A., Bibi, A., Saxena, A., Tripathi, N., Iftikhar, A., Raina, A., Khakwani, K.Z.R., Riaz, I.B. and Husnain, M. (2025), Efficacy and Safety of Quadruplet Therapy in Newly Diagnosed Transplant-Eligible Multiple Myeloma: A Systematic Review and Meta-Analysis. Cancer Reports, 8: e70171. https://doi.org/10.1002/cnr2.70171 April 2, 2025.
Overview
The treatment for multiple myeloma (MM) has been improving. Recently, adding an anti-CD38 monoclonal antibody (mAb) to the standard triplet therapy (which includes a proteasome inhibitor, an immunomodulatory agent, and a steroid) has shown promising results for newly diagnosed MM patients who are eligible for a transplant.
This study looked at the effectiveness and safety of quadruplet therapy (which includes the anti-CD38 mAb) compared to the standard triplet therapy. Researchers reviewed data from five clinical trials with nearly 3,000 patients.
The results showed:
- Progression-free survival (PFS), or how long patients stay without the cancer getting worse, was significantly better with quadruplet therapy.
- The response rate and measurable residual disease (MRD) negativity were also higher in the quadruplet therapy group.
- There was no significant difference in overall survival (OS) between the two therapies.
- While quadruplet therapy did not increase the risk of severe side effects (like anemia or lymphopenia), it did raise the risk of thrombocytopenia (low platelets), neutropenia (low white blood cells), and infections.
Quadruplet therapy appears to be a more effective treatment for newly diagnosed MM patients who are transplant-eligible and could become the new standard of care.
"Single-Cell Transcriptome Analysis Reveals the Effect of MIF on Myeloid-Derived Suppressor Cells in Multiple Myeloma"
Source
Zhao, X., Zhou, J., Yao, J., Shi, Q., Su, J. and Hu, N. (2025), Single-Cell Transcriptome Analysis Reveals the Effect of MIF on Myeloid-Derived Suppressor Cells in Multiple Myeloma. Int J Lab Hematol. https://doi.org/10.1111/ijlh.14473 April 2, 2025.
Overview
Myeloid-derived suppressor cells (MDSCs) play an important role in multiple myeloma (MM) by affecting myeloma cells in the bone marrow. One key factor, macrophage migration inhibitory factor (MIF), is a protein involved in controlling immune responses and preventing macrophages (a type of immune cell) from moving freely. This study looked at how MIF interacts with MDSCs in the bone marrow of MM patients.
Researchers used a special technique called single-cell RNA sequencing to study the cells in MM and normal bone marrow. They found two groups of MDSCs based on the level of MIF: MIF+MDSCs and MIF−MDSCs. They observed that these two groups behaved differently in MM patients compared to normal individuals.
The study showed that MIF+MDSCs had lower signaling with plasma cells (a type of immune cell) in a specific pathway, while MIF−MDSCs had stronger signaling with plasma cells. They also found higher levels of certain proteins (RETN, Arg-1, and iNOS) in MDSCs from MM patients compared to normal ones, with MIF+MDSCs showing even higher levels. Additionally, the protein CAP1 in plasma cells was higher in MM patients.
In conclusion, MIF is highly expressed in MM patients and is linked to MDSCs in the bone marrow, suggesting it could play an important role in the disease's progression.
"Nanostructures: An Efficient Drug Delivery Platform for Therapy of Multiple Myeloma"
Source
Teodora Eliana Petcov, Vadim V. Silberschmidt, Mădălina Andreea Pandele, Elena Alina Chiticaru, Mariana Ioniță, Marius Manole, Nanostructures: An Efficient Drug Delivery Platform for Therapy of Multiple Myeloma, European Journal of Medicinal Chemistry Reports, 2025, 100263, ISSN 2772-4174, https://doi.org/10.1016/j.ejmcr.2025.100263. April 2, 2025.
Overview
Recent advancements in nanotechnology have brought new possibilities for diagnosing and treating multiple myeloma (MM). Traditional treatments often do not last, making the need for more effective therapies urgent. Nanotechnology-based drug delivery systems (DDS) are showing promise because they can target drugs directly to cancer cells, reducing side effects and improving treatment effectiveness.
This review focuses on various nanostructures used in drug delivery, such as liposomes, micelles, polymeric nanoparticles, and DNA nanostructures. These systems help deliver chemotherapy more precisely to tumor cells. The review also highlights how these nanostructures are taken up by cancer cells, improving the accuracy and success of treatments.
However, there are challenges. The safety and effectiveness of these new technologies must be carefully studied. Nanostructures can interact with the body in unpredictable ways, potentially causing side effects. Moreover, designing nanostructures that can bypass the body’s defenses while delivering the drug efficiently is complex. Researchers are working on improving these systems, making them safer and more effective for patients with MM.
In the future, personalized medicine that uses nanotechnology could revolutionize MM treatment. Machine learning and artificial intelligence may help create even better drug delivery systems tailored to each patient. While there are hurdles to overcome, the potential of nanotechnology in MM treatment is very promising and could lead to better outcomes for patients.
"Early identification of functional high-risk multiple myeloma patients after transplant: the predictive power of fat fraction and Response Assessment Category score in diffusion-weighted whole-body magnetic resonance imaging"
Source
Belotti A, Frittoli B, Terlizzi S, Ribolla R, Crippa C, Saeli C, Ferrari S, Bianchetti N, Bottelli C, Cattaneo C, Carbone C, Gullino A, Chiarini M, Giustini V, Roccaro A, Grazioli L, Tucci A. Early identification of functional high-risk multiple myeloma patients after transplant: the predictive power of fat fraction and Response Assessment Category score in diffusion-weighted whole-body magnetic resonance imaging. Haematologica; https://doi.org/10.3324/haematol.2025.287409 [Early view]. April 3, 2025.
Overview
Functional high-risk (FHR) multiple myeloma (MM) patients are those who relapse early even after receiving the best initial treatment. These patients need better ways to predict their outcomes. One tool that is increasingly used in managing MM is diffusion-weighted whole-body MRI (DW-MRI), which is good at detecting how well treatments are working. A system called MY-RADS helps assess how patients are responding to treatment using a 5-point scale, where a score of 1 means a complete response and 5 means disease is getting worse.
In this study, researchers looked at how combining MY-RADS scores with a measurement from DW-MRI called relative Fat Fraction (rFF) could help predict which patients are at risk of relapsing early. The study found that an rFF value of 17.2% was a strong predictor for early relapse. Patients with rFF higher than this had much better survival and progression-free survival (PFS) after treatment compared to those with lower rFF values.
Using both the MY-RADS scale and rFF can help doctors better identify high-risk MM patients who might relapse soon after treatment. This approach could lead to more tailored treatment plans to improve outcomes for these patients.
"Perspectives on Talquetamab and its Utility in the Treatment of Multiple Myeloma: Safety, Efficacy and Place in Therapy"
Source
Miller KC, Hamadeh I, Tan CR. Perspectives on Talquetamab and its Utility in the Treatment of Multiple Myeloma: Safety, Efficacy and Place in Therapy. Cancer Manag Res. 2025;17:743-756
https://doi.org/10.2147/CMAR.S441550 April 3, 2025.
Overview
Despite progress in treating multiple myeloma, many patients eventually develop resistance to current treatments, highlighting the need for new therapies. G protein-coupled receptor class C group 5 member D (GPRC5D) has become a promising new target for treating myeloma cells. Talquetamab, a medication designed to target GPRC5D, is the first of its kind and was approved by the FDA in August 2023.
While talquetamab is effective in treating many patients, it can cause side effects like cytokine release syndrome, nerve problems, and blood cell issues. It also often leads to problems in the mouth, nails, and skin, which can impact a patient's quality of life and sometimes cause people to stop treatment. However, for many patients, talquetamab shows strong results, and in some cases, these results last for a long time.
There are now several clinical trials testing talquetamab in different situations and in combination with other treatments for multiple myeloma. Future research will focus on understanding how resistance to talquetamab develops and how to reduce its common side effects, which are important for improving its use in treating multiple myeloma.
"LY6E as a new prognostic biomarker of multiple myeloma-related bone disease"
Source
Shi, M., Li, J., Wang, J. et al. LY6E as a new prognostic biomarker of multiple myeloma-related bone disease. Sci Rep 15, 11431 (2025). https://doi.org/10.1038/s41598-025-91413-1 April 3, 2025.
Overview
Osteolytic bone disease, which weakens bones and impacts quality of life, is a common complication of multiple myeloma (MM). In this study, researchers used bioinformatics tools to identify genes linked to MM-related bone disease (MBD). Their analysis found that the LY6E molecule was strongly associated with the progression of MM, poor patient outcomes, and the development of MBD. They also confirmed in lab experiments that higher levels of LY6E helped MM cells grow and promoted the development of bone-destroying cells (osteoclasts). These findings suggest that LY6E plays a key role in MBD and could be a new target for treating multiple myeloma.
"Real-world evaluation of teclistamab for the treatment of relapsed/refractory multiple myeloma (RRMM): an International Myeloma Working Group Study"
Source
Tan, C.R., Asoori, S., Huang, CY. et al. Real-world evaluation of teclistamab for the treatment of relapsed/refractory multiple myeloma (RRMM): an International Myeloma Working Group Study. Blood Cancer J. 15, 53 (2025). https://doi.org/10.1038/s41408-025-01259-z April 3, 2025.
Overview
Teclistamab is a new antibody treatment that targets multiple myeloma, a type of blood cancer. In a study of 210 patients from 9 academic centers, teclistamab showed strong results, even in patients who had already tried several treatments. The study found that 67% of patients had a positive response to the treatment, and 55% of them had a significant improvement. After 6 months, about half of the patients were still progression-free, and 73% were alive. Patients who had already been treated with other BCMA-targeted therapies had slightly lower response rates. The most common side effects were cytokine release syndrome (CRS) in 54% of patients and infections in 56%, with some infections being severe. Despite these side effects, teclistamab showed strong effectiveness and safety, similar to earlier studies.
"Ambroxol induces myeloma cell death by inhibiting autophagy"
Source
Yutaka Hattori, Hiromu Sugiyama, Yamato Miyashita, Shinsuke Shibata, Taiga Okaue, Yoshinao Matsumoto, Taketo Yamada, Tomofumi Yamamoto, Takashi Yamaguchi, Kohei Yamazaki, Hisako Kunieda, Hideyuki Saya, Maiko Matsushita; Ambroxol induces myeloma cell death by inhibiting autophagy. Blood Neoplasia 2025; 100100. doi: https://doi.org/10.1016/j.bneo.2025.100100 April 3, 2025
Overview
Over the past decade, new treatments have improved the outlook for patients with multiple myeloma (MM), but most patients eventually relapse, and serious side effects can make treatment difficult. Researchers have been exploring new ways to boost the effectiveness of existing treatments while reducing side effects. One promising discovery is that ambroxol hydrochloride, a drug originally used for other purposes, can cause MM cells to die. Ambroxol works by blocking a key process called autophagy, which is important for the survival of MM cells. When ambroxol was used with the drug panobinostat, which normally triggers autophagy, the combination was even more effective in fighting MM and helped reduce the diarrhea that panobinostat can cause. In lab tests, ambroxol also slowed tumor growth on its own and worked even better when used with panobinostat. These findings suggest that targeting autophagy could be a new and promising way to treat MM.
"Prognostic value of [18F]fluorodeoxyglucose-PET/MRI(CT) novel interpretation criteria (IMPeTUs) in multiple myeloma"
Source
Zhang, M., Cai, T., Jin, S. et al. Prognostic value of [18F]fluorodeoxyglucose-PET/MRI(CT) novel interpretation criteria (IMPeTUs) in multiple myeloma. Eur J Nucl Med Mol Imaging (2025). https://doi.org/10.1007/s00259-025-07219-w April 3, 2025.
Overview
[18F]fluorodeoxyglucose (FDG)-PET scans are important for evaluating how well multiple myeloma (MM) is progressing. A new set of criteria, called IMPeTUs, has been developed to standardize how PET scans are interpreted in MM. However, until now, the value of the IMPeTUs score for predicting patient outcomes wasn’t clear.
In this study, 58 newly diagnosed MM patients underwent both PET/MRI and PET/CT scans and were followed for about 42 months. The researchers compared the predictive power of the IMPeTUs score to other common tests. The results showed that the PET/MRI IMPeTUs score was better at predicting how long patients would live without their disease progressing (progression-free survival, PFS) compared to other tests. A higher IMPeTUs score was linked to worse outcomes, with a score of 13 or higher being an independent risk factor for poor progression-free survival.
The IMPeTUs score is a helpful tool for predicting prognosis in MM and is more reliable than other traditional tests.
"HLA-mismatched stem cell microtransplant prolonged overall survival and promoted immunological reconstitution for multiple myeloma"
Source
Lei Y, Cai B, Liu Z, Xie A, Qiao J, Wang Y, Chen X, Peng F, Zhao Y, Chen J, Guan W, Yu C, Lou X, Hu K, Zhang A, Sun Q, Huang Y, Ai H, Guo M. HLA-mismatched stem cell microtransplant prolonged overall survival and promoted immunological reconstitution for multiple myeloma. Front Immunol. 2025 Apr 4;16:1509588. doi: 10.3389/fimmu.2025.1509588.
Overview
Microtransplant (MST), a treatment used in other blood cancers, hasn’t been explored much for multiple myeloma—until now.
In this study, 20 multiple myeloma patients who received MST at a medical center were analyzed. The results showed that 17 out of 20 patients responded to the treatment. The 6-year overall survival (OS) rate was 64.7%, and the progression-free survival (PFS) rate was 35.3%. There were no serious side effects like graft-versus-host disease, and only a few patients experienced mild symptoms like controlled fever or Grade I cytokine release syndrome.
The study found that certain factors, such as earlier stages of the disease or better response to treatment before MST, were linked to longer survival. MST also improved immune function, with an increase in specific types of immune cells.
MST appears to help extend survival and improve immune system function in multiple myeloma patients, particularly those with high-risk disease, making it a promising treatment option
"A novel cereblon variant with both exon 8 and 10 deletions in newly diagnosed and relapsed multiple myeloma"
Source
Yuan Xiao Zhu, Soumya Dutta, Gregory J. Ahmann, Laura Bruins, Mariano Arribas, Xianfeng Chen, Abhigna Polikarpov, Jonathan Chapman, Issac Lopez, Marta Chesi, P. Leif Bergsagel, Abhishek Singharoy, Rafael Fonseca, Lisa M. Rimsza; A novel cereblon variant with both exon 8 and 10 deletions in newly diagnosed and relapsed multiple myeloma. Blood Neoplasia 2025; 100099. doi: https://doi.org/10.1016/j.bneo.2025.100099 April 4, 2025.
Overview
In multiple myeloma (MM), patients may develop resistance to treatment, especially to immunomodulatory drugs (IMiDs). IMiDs work by binding to a protein called cereblon (CRBN), helping the body target harmful proteins. However, changes in the CRBN gene can make MM cells resistant to these drugs. One such change involves the deletion of part of the CRBN gene, specifically in exon 10, which is linked to IMiD resistance.
In this study, the researchers created a new test to detect this exon 10 deletion in the CRBN gene. They analyzed 74 MM samples, including both newly diagnosed and relapsed cases, and found that four samples had the deletion. Interestingly, two of these samples also showed an additional deletion in exon 8 of the gene. The researchers used advanced techniques to study how these genetic changes affect CRBN and its ability to work with other proteins to target harmful substances in the cell.
The study developed a reliable method to detect this genetic change and found that the CRBN variant could appear early in the disease. This could help explain why some patients are resistant to IMiDs and may have different impacts on treatment effectiveness
"A novel cereblon variant with both exon 8 and 10 deletions in newly diagnosed and relapsed multiple myeloma"
Source
Yuan Xiao Zhu, Soumya Dutta, Gregory J. Ahmann, Laura Bruins, Mariano Arribas, Xianfeng Chen, Abhigna Polikarpov, Jonathan Chapman, Issac Lopez, Marta Chesi, P. Leif Bergsagel, Abhishek Singharoy, Rafael Fonseca, Lisa M. Rimsza; A novel cereblon variant with both exon 8 and 10 deletions in newly diagnosed and relapsed multiple myeloma. Blood Neoplasia 2025; 100099. doi: https://doi.org/10.1016/j.bneo.2025.100099 April 4, 2025.
Overview
In multiple myeloma (MM), patients may develop resistance to treatment, especially to immunomodulatory drugs (IMiDs). IMiDs work by binding to a protein called cereblon (CRBN), helping the body target harmful proteins. However, changes in the CRBN gene can make MM cells resistant to these drugs. One such change involves the deletion of part of the CRBN gene, specifically in exon 10, which is linked to IMiD resistance.
In this study, the researchers created a new test to detect this exon 10 deletion in the CRBN gene. They analyzed 74 MM samples, including both newly diagnosed and relapsed cases, and found that four samples had the deletion. Interestingly, two of these samples also showed an additional deletion in exon 8 of the gene. The researchers used advanced techniques to study how these genetic changes affect CRBN and its ability to work with other proteins to target harmful substances in the cell.
The study developed a reliable method to detect this genetic change and found that the CRBN variant could appear early in the disease. This could help explain why some patients are resistant to IMiDs and may have different impacts on treatment effectiveness
"MicroRNA-665 and its potential role in drug response and survival outcomes in multiple myeloma: a preliminary study"
Source
Bergantim R, Peixoto da Silva S, Pinto V, Pereira JM, Sousa D, Trigo F, Matthiesen R, Guimarães JE, Vasconcelos MH. MicroRNA-665 and its potential role in drug response and survival outcomes in multiple myeloma: a preliminary study. Front Pharmacol. 2025 Apr 4;16:1465814. doi: 10.3389/fphar.2025.1465814.
Overview
Multiple myeloma is the second most common blood cancer, and understanding its genetic risk factors is important for improving treatment. In this study, researchers looked at data from over 3,400 multiple myeloma patients and more than 14,700 healthy controls. They focused on certain genetic changes, known as single-nucleotide polymorphisms (SNPs), that may be linked to a higher risk of the disease.
The study found that several genetic variations were associated with an increased risk of multiple myeloma. For example, the DNAH11, ULK4, DTNB, and VDR gene variations were linked to higher chances of developing the disease. These findings suggest that these genes could be used as markers to predict who is at higher risk for multiple myeloma, helping doctors make more informed treatment decisions.
"Dynamic monitoring of M-protein quantification by immunotyping using capillary zone electrophoresis during the chemotherapy of patients with multiple myeloma"
Source
Jia, Z., Lu, Q. Dynamic monitoring of M-protein quantification by immunotyping using capillary zone electrophoresis during the chemotherapy of patients with multiple myeloma. Sci Rep 15, 11541 (2025). https://doi.org/10.1038/s41598-025-96565-8 April 4, 2025.
Overview
In this study, researchers used a special technique to measure M-proteins (monoclonal immunoglobulin proteins) in patients with multiple myeloma (MM) during chemotherapy. They tracked how these proteins changed while patients were receiving treatment. The results showed that the technique, called immunotyping by immunosubtraction (ISUB), was accurate in measuring M-proteins and could help doctors see how well chemotherapy was working.
Out of 21 patients, 15 were treated with a drug called bortezomib. In some cases, the M-protein levels dropped by more than 50% after just one round of treatment. The study suggests that measuring M-protein levels regularly during treatment can help doctors assess how well the drugs are working and guide decisions on which medications to use. Tracking even small changes in M-protein is important for monitoring the disease and adjusting treatments.
"Talquetamab in Heavily Pretreated Multiple Myeloma Patients, Including BCMA-Refractory"
Source
Andrew J Vegel, Bradley T Loeffler, Jonathan Lochner, Reed Friend, Alma Habib, Marshall Mckenna, Jordan Snyder, Kimberly Green, Rachel Dileo, Gina Patrus, Prerna Mewawalla, Muhammad Umair Mushtaq, Christopher Strouse, Yun Kyoung Ryu Tiger, Nausheen Ahmed, Abdullah M. Khan, Hamza Hashmi, Barry Paul, Shebli Atrash, Al-Ola Abdallah, Hira Shaikh; Talquetamab in Heavily Pretreated Multiple Myeloma Patients, Including BCMA-Refractory. Blood Immunology & Cellular Therapy 2025; 100001. doi: https://doi.org/10.1016/j.bict.2025.100001 April 4, 2024.
Overview
Talquetamab is a treatment for relapsed multiple myeloma that targets a protein called GPRC5D. It was approved based on the MonumenTAL-1 trial. In this new study, researchers looked at the safety and effectiveness of talquetamab in 68 patients across seven U.S. academic centers. They found that 40% of patients would not have qualified for the MonumenTAL-1 trial.
The study showed that infections were less common than in the original trial and other treatments, but side effects like a strange taste in the mouth (dysgeusia) were more frequent. Overall, 71% of patients had a good response to treatment, with 51% having a very good partial response or better, and 25% achieving a complete response. After about 4.5 months, 74% of patients were still alive.
The study also found that patients who had already been treated with other bispecific antibodies or had cancer outside the bone marrow had worse outcomes. Patients who had received BCMA-targeted treatments before talquetamab had lower response rates and shorter survival. Overall, the safety and effectiveness of talquetamab in this real-world setting were similar to what was seen in the MonumenTAL-1 trial.
"A novel cereblon variant with both exon 8 and 10 deletions in newly diagnosed and relapsed multiple myeloma"
Source
Yuan Xiao Zhu, Soumya Dutta, Gregory J. Ahmann, Laura Bruins, Mariano Arribas, Xianfeng Chen, Abhigna Polikarpov, Jonathan Chapman, Issac Lopez, Marta Chesi, P. Leif Bergsagel, Abhishek Singharoy, Rafael Fonseca, Lisa M. Rimsza; A novel cereblon variant with both exon 8 and 10 deletions in newly diagnosed and relapsed multiple myeloma. Blood Neoplasia 2025; 100099. doi: https://doi.org/10.1016/j.bneo.2025.100099 April 4, 2025.
Overview
With multiple myeloma (MM), patients may become resistant to treatments called immunomodulatory drugs (IMiDs). IMiDs work by binding to a protein called cereblon (CRBN) and helping it target other proteins to be broken down. However, changes in the CRBN gene can cause resistance to IMiDs. One such change is a deletion of part of the CRBN gene called exon 10.
In this study, researchers created a test to detect this deletion and studied samples from 74 MM patients, including those who were newly diagnosed and those with advanced or relapsed disease. They found that 4 patients had a 40% deletion of exon 10. Interestingly, two samples also had an additional deletion in exon 8. These changes affect how CRBN works and may interfere with the drug’s ability to target the disease.
The findings suggest that these genetic changes, present from the time of diagnosis, could play a role in how patients become resistant to treatment. Detecting these changes could help doctors understand and manage drug resistance in MM more effectively.
"Venetoclax-based treatment combinations in relapsed/refractory multiple myeloma: practice patterns and impact of secondary cytogenetic abnormalities on outcomes"
Source
Bolarinwa, A., Nagaraj, M., Zanwar, S. et al. Venetoclax-based treatment combinations in relapsed/refractory multiple myeloma: practice patterns and impact of secondary cytogenetic abnormalities on outcomes. Blood Cancer J. 15, 57 (2025). https://doi.org/10.1038/s41408-025-01264-2 April 4, 2025.
Overview
Venetoclax (Ven) is a drug that targets a protein called BCL-2 and has shown potential in treating patients with relapsed or refractory multiple myeloma (RRMM) who have certain genetic changes, like the t(11;14) translocation or high BCL-2 levels. However, the results from clinical trials have been mixed.
This study looked at 232 multiple myeloma patients treated at Mayo Clinic from January 2015 to December 2023. Most patients had the t(11;14) genetic change, while a smaller group had high BCL-2 expression. The patients received venetoclax in combination with other drugs, such as dexamethasone, proteasome inhibitors, or daratumumab.
The study found that 57% of patients responded to treatment. For patients with the t(11;14) change, 64% showed a positive response, while only 26% of those without the change responded. The average time before the disease progressed was 9.4 months for all patients, but it was much longer for patients with t(11;14) (11.8 months) compared to those without it (2.9 months). Additionally, for patients with t(11;14), the presence of certain other high-risk genetic changes, like del(17p) or 1q gain/amplification, reduced the time before disease progression.
The study concludes that venetoclax-based treatments work well for patients with the t(11;14) genetic change but are less effective for those without it. Patients with additional high-risk genetic features may experience shorter periods of effectiveness.
"Real-World Evidence Evaluating Teclistamab in Patients with Relapsed/Refractory Multiple Myeloma: A Systematic Literature Review"
Source
Derman, B., Tan, C., Steinfield, I., Wilson, F. R., Lin, D., Wu, B., Fernandez, M., Fowler, J., Paner-Straseviciute, A., Kim, N., Doyle, M., Marshall, A., Cheadle, J., Keeping, S., & Liu, J. J. (2025). Real-World Evidence Evaluating Teclistamab in Patients with Relapsed/Refractory Multiple Myeloma: A Systematic Literature Review. Cancers, 17(7), 1235. https://doi.org/10.3390/cancers17071235 April 5, 2025.
Overview
Teclistamab is a type of bispecific antibody used to treat patients with relapsed or refractory multiple myeloma who have not responded to previous treatments. Initially studied in the MajesTEC-1 clinical trial, teclistamab has been increasingly used in practice since it was approved in October 2022. This study conducts a systematic review of existing literature to evaluate the drug's effectiveness, safety, healthcare resource use, and prescribing trends in real-world settings. The findings suggest that patients treated with teclistamab in everyday clinical practice experience similar outcomes to those seen in clinical trials. Given the high disease burden and limited treatment options for patients with relapsed or refractory multiple myeloma, this study provides valuable insights into the therapeutic benefits and safety of teclistamab, helping inform healthcare providers and policymakers.
"Solitary plasmacytoma: single-institution experience, and systematic review and meta-analysis of clinical outcomes"
Source
Charalampos Charalampous, Jean-Sebastien Claveau, Prashant Kapoor, Moritz Binder, Francis K. Buadi, Joselle Cook, David Dingli, Angela Dispenzieri, Amie L. Fonder, Morie A. Gertz, Wilson Gonsalves, Suzanne R. Hayman, Miriam A. Hobbs, Yi L. Hwa, Taxiarchis Kourelis, Martha Q. Lacy, Nelson Leung, Yi Lin, Rahma Warsame, Robert A. Kyle, S. Vincent Rajkumar, Shaji K. Kumar; Solitary plasmacytoma: single-institution experience, and systematic review and meta-analysis of clinical outcomes. Blood Adv 2025; 9 (7): 1559–1570. doi: https://doi.org/10.1182/bloodadvances.2024013355 April 8, 2025.
Overview
This study analyzed data from 147 patients with solitary plasmacytomas treated at the Mayo Clinic and expanded the investigation with a review of 62 studies involving 3487 patients from 1960 to 2022. The research found that patients with up to 10% clonal plasma cells in their bone marrow (BM) had a much shorter disease-free survival (DFS) of 15.7 months, compared to 79 months for patients with no clonal plasma cells in the BM. Key risk factors for shorter DFS included the presence of clonal plasma cells in the marrow and a large difference in free light chains. The study also confirmed that radiation therapy was the main treatment. For patients with plasmacytomas, the DFS rates at 3, 5, and 10 years were 66.9%, 55%, and 42.1%, respectively. Patients with extramedullary plasmacytomas (tumors outside the bone) had better survival rates than those with bone plasmacytomas. This research emphasizes the importance of disease staging at diagnosis and the location of the tumor in predicting patient outcomes.
"Identifying potential prognosis markers in relapsed multiple myeloma via integrated bioinformatics analysis and biological experiments"
Source
Yong Xu, Xinya Cao, He Zhou, Han Xu, Bing Chen, Hua Bai, Identifying potential prognosis markers in relapsed multiple myeloma via integrated bioinformatics analysis and biological experiments, Current Research in Translational Medicine, Volume 73, Issue 2, 2025, 103495, ISSN 2452-3186, https://doi.org/10.1016/j.retram.2025.103495. April-June 2025.
Overview
This study focused on identifying key genes involved in relapsed multiple myeloma (MM) using bioinformatics and biological experiments. Researchers analyzed gene data from baseline and relapsed MM patients to find genes linked to disease progression. They discovered four important genes—CENPE, ASPM, TOP2A, and FANCI—that play roles in cell division and growth. These genes were used to create a "relapsed gene score" (RGS) model, which can help predict the risk of relapse and patient prognosis. The study also found that an inhibitor of CENPE (called GSK923295) had a positive effect on MM cells by promoting cell death and damage. This research suggests that targeting these genes, particularly CENPE, could help develop new treatments for MM.
"Updates on myeloma in older adults: Conference summary from the 2024 International Myeloma Society annual meeting"
Source
Sandra Frimpong, Aminatta Tejan-Kamara Mitchell, Hira Mian, Tanya M. Wildes, Updates on myeloma in older adults: Conference summary from the 2024 International Myeloma Society annual meeting, Journal of Geriatric Oncology, Volume 16, Issue 3, 2025, 102199, ISSN 1879-4068, https://doi.org/10.1016/j.jgo.2025.102199. April 2025.
Overview
Multiple myeloma (MM) mainly affects older adults, with the average age at diagnosis being around 70. As more attention is given to how frailty and other vulnerabilities impact treatment and outcomes in older patients, research focusing on MM in this age group has grown. The 2024 Annual Meeting of the International Myeloma Society held in Rio de Janeiro presented new findings on this topic. Researchers reviewed 506 abstracts and highlighted 17 that focused on frailty, aging, and MM treatment in older adults.
These studies explored topics such as new treatments for newly diagnosed MM in older patients, including clinical trials with new drug regimens and stem cell transplants. Some studies found that selected older patients could benefit from treatments like autologous stem cell transplants and CAR T therapy, which were previously mainly tested in younger patients. The meeting also emphasized the importance of assessing frailty, using tools like the IMWG frailty score, to predict outcomes and treatment risks. Overall, the research presented at the meeting shows promise for improving MM treatment in older adults by considering their unique needs and vulnerabilities.
"Updates on myeloma in older adults: Conference summary from the 2024 International Myeloma Society annual meeting"
Source
Sandra Frimpong, Aminatta Tejan-Kamara Mitchell, Hira Mian, Tanya M. Wildes, Updates on myeloma in older adults: Conference summary from the 2024 International Myeloma Society annual meeting, Journal of Geriatric Oncology, Volume 16, Issue 3, 2025, 102199, ISSN 1879-4068, https://doi.org/10.1016/j.jgo.2025.102199. April 2025.
Overview
Multiple myeloma (MM) mainly affects older adults, with the average age at diagnosis being around 70. As more attention is given to how frailty and other vulnerabilities impact treatment and outcomes in older patients, research focusing on MM in this age group has grown. The 2024 Annual Meeting of the International Myeloma Society held in Rio de Janeiro presented new findings on this topic. Researchers reviewed 506 abstracts and highlighted 17 that focused on frailty, aging, and MM treatment in older adults.
These studies explored topics such as new treatments for newly diagnosed MM in older patients, including clinical trials with new drug regimens and stem cell transplants. Some studies found that selected older patients could benefit from treatments like autologous stem cell transplants and CAR T therapy, which were previously mainly tested in younger patients. The meeting also emphasized the importance of assessing frailty, using tools like the IMWG frailty score, to predict outcomes and treatment risks. Overall, the research presented at the meeting shows promise for improving MM treatment in older adults by considering their unique needs and vulnerabilities.
"HRP2 regulating MICU1-mediated Ca2+ overload to dictate chemoresistance of multiple myeloma"
Source
Qian Li, Ziyi Peng, Li Lin, Zhiying Zhang, Jing Ma, Lin Chen, Su Liu, Shuang Gao, Linchuang Jia, Jingjing Wang, Zeng Cao, Xingli Zhao, Zhiqiang Liu, Yafei Wang, HRP2 regulating MICU1-mediated Ca2+ overload to dictate chemoresistance of multiple myeloma, Neoplasia, Volume 62, 2025, 101150, ISSN 1476-5586, https://doi.org/10.1016/j.neo.2025.101150. April 2025.
Overview
Velcade (bortezomib)-based chemotherapy is effective in treating multiple myeloma (MM), but many patients develop resistance over time, especially those who initially respond well. In this study, researchers found that a protein called hepatoma-derived growth factor-related protein-2 (HRP2) plays a key role in this resistance. When HRP2 expression was reduced in MM cells, the cells became more resistant to bortezomtreatment. The study also showed that lower levels of HRP2 in patients predicted shorter survival rates for those undergoing bortezomib-based therapies.
The researchers discovered that reducing HRP2 led to changes in the cells’ gene activity, particularly increasing the expression of a gene called MICU1, which helps control calcium levels in cells. This helped protect the cells from damage caused by high calcium and reactive oxygen species (ROS), which can lead to cell death. By suppressing MICU1, the researchers improved the cells' sensitivity to bortezomib and reduced tumor growth in mice.
This study provides new insights into how HRP2 affects calcium balance in MM cells and contributes to drug resistance. These findings could help develop better treatments for patients who become resistant to bortezomib-based therapies.
"A novel CCL3-HMGB1 signaling axis regulating osteocyte RANKL expression in multiple myeloma"
Source
Anloague A, Sabol HM, Kaur J, Khan S, Ashby C, Schinke C, Barnes CL, Alturkmani F, Ambrogini E, Gundesen MT, Lund T, Amstrup AK, Andersen TL, Diaz-delCastillo M, Roodman GD, Bellido T, Delgado-Calle J. A novel CCL3-HMGB1 signaling axis regulating osteocyte RANKL expression in multiple myeloma. Haematologica 2025;110(4):952-966; https://doi.org/10.3324/haematol.2024.286484. April 2025.
Overview
One of the key features of multiple myeloma (MM) is bone destruction, which is caused by an increase in osteoclasts (cells that break down bone tissue) and higher levels of RANKL, a protein that controls osteoclast formation. Previous research showed that osteocytes, the most common cells in bone, contribute to this problem by producing too much RANKL.
In this study, researchers found that CCL3, a protein produced by MM cells, increases RANKL levels in osteocytes. Using different models and clinical data, they showed that blocking CCL3, either through genetics or drugs, reduced RANKL production in osteocytes and helped prevent bone loss caused by MM. The study also revealed that CCL3 triggers osteocytes to release another protein called HMGB1, which is necessary for the increase in RANKL.
This research uncovers a new signaling pathway (CCL3-HMGB1) that drives bone damage in MM and may offer new targets for treating MM-related bone disease.
"Teclistamab in relapsed refractory multiple myeloma: a multi-institutional real-world study from the French early access program"
Source
Perrot A, Hulin C, Boumendil A, Manjra H, Leveque A, Croizier C, Dony A, Mohty M, Roussel M, Manier S, Orsini-Piocelle F, Bauschert L, Bobin A, Frenzel L, Vincent L, Breal C, Eveillard JR, Gerome T, Tiab M, Chalayer E, Belkhir R, Mariette C, Moyer P, Chalopin T, Cherel B, Montes L, Coste A, Tabrizi R, Karlin L, Robu D, Huguet A, Harel S, Moreau P. Teclistamab in relapsed refractory multiple myeloma: a multi-institutional real-world study from the French early access program. Haematologica 2025;110(4):990-994; https://doi.org/10.3324/haematol.2024.286118. April 2025.
Overview
Teclistamab is a new treatment for relapsed and refractory multiple myeloma (RRMM), a form of cancer that is resistant to multiple therapies. It works by targeting two proteins, BCMA and CD3, and was approved after the results of the MajesTEC-1 clinical trial. In this study, we looked at how teclistamab performed in real-world settings in France, where it was used by 303 patients at 30 centers from October 2022 to September 2023.
The patients in this study had already tried several treatments, with many being resistant to commonly used therapies like immunomodulatory drugs and proteasome inhibitors. The results showed that 68.8% of patients responded to teclistamab, with 61.4% achieving at least a very good partial response. The average progression-free survival (PFS) for all patients was 11.3 months, and for patients who responded well, it was 17 months. However, patients with more advanced disease, like extramedullary disease or those with certain genetic changes, had shorter PFS.
While infections were the most common side effect, the safety profile was similar to what was observed in the clinical trial. This study provides real-world evidence that teclistamab is an effective and safe treatment for patients with RRMM, even those who were excluded from the original clinical trials. The findings support the continued use of teclistamab for these patients, particularly with the addition of immunoglobulin (Ig) therapy to manage side effects.
"Isatuximab, pomalidomide, and dexamethasone as salvage therapy for patients with multiple myeloma: the Italian, multicenter, retrospective clinical experience with 270 cases outside of controlled clinical trials"
Source
Martino EA, Derudas D, Rossi E, Terlizzi S, Reddiconto G, Stefanoni P, Micozzi J, Mangiacavalli S, Zamagni E, Offidani M, Furlan A, Buda G, Lotti F, Liberatore C, Lazzaro A, Pepa RD, Bertuglia G, Barbieri E, Conticello C, De Magistris C, De Paoli L, Bongarzoni V, Cafro AM, Mele A, Benvenuti P, Cerchione C, Botta C, Antonioli E, Sgherza N, Aquino S, Mele G, Barilà G, Palmieri S, Annibali O, Bianco R, Febbo MA, Casaluci GM, Rago A, Fontana R, Farina F, Vigna E, Bruzzese A, Mancuso K, Nappi D, Morè S, Rivolti E, Califano C, Amendola A, Roccotelli D, Lombardo A, Citro A, Uccello G, Zambello R, Maggi A, Neri S, Monachesi M, Gozzetti A, Montefusco V, Brunori M, Cotzia E, Pietrantuono G, Quinto AM, Amico V, Di Renzo N, Coscia M, Galli M, De Stefano V, Petrucci MT, Neri A, Di Raimondo F, Morabito F, Musto P, Gentile M. Isatuximab, pomalidomide, and dexamethasone as salvage therapy for patients with multiple myeloma: the Italian, multicenter, retrospective clinical experience with 270 cases outside of controlled clinical trials. Haematologica 2025;110(4):1028-1033; https://doi.org/10.3324/haematol.2024.286658. April 2025.
Overview
A recent study evaluated the effectiveness and safety of the isatuximab, pomalidomide, and dexamethasone (IsaPd) combination in patients with relapsed or refractory multiple myeloma (RRMM). The results confirmed the treatment's safety and effectiveness, matching findings from a major clinical trial, ICARIA-MM.
The study involved 270 RRMM patients from 51 centers in Italy. The combination of IsaPd showed a high response rate, with 74.1% of patients responding to treatment. This response was even higher in patients with good kidney function and lower-stage disease. The median progression-free survival (PFS) was 15.7 months, which is higher than the results of the ICARIA-MM trial. The median overall survival (OS) was not yet reached, with a 2-year survival rate of 64.2%.
However, patients who had been treated with daratumumab before starting IsaPd had a shorter PFS. The study also showed that patients with high-risk cytogenetics had a worse outcome compared to those with standard-risk genetics.
Side effects were manageable, with 56.3% of patients experiencing severe neutropenia, and 47.8% developing infections. Despite these issues, the treatment was generally well-tolerated.
Overall, IsaPd is a promising treatment for RRMM patients who have already received at least two prior therapies. However, more options like bispecific antibodies and CAR T-cell therapies are needed for high-risk patients or those who are refractory to daratumumab.
"Deciphering Genomic Complexity of Multiple Myeloma Using Optimized Optical Genome Mapping"
Source
Hélène Guermouche, Pauline Roynard, Francesca Servoli, Valentin Lestringant, Benoît Quilichini, Christine Terré, Sabine Defasque, Catherine Roche-Lestienne, Dominique Penther, Agnès Daudignon, Deciphering Genomic Complexity of Multiple Myeloma Using Optimized Optical Genome Mapping, The Journal of Molecular Diagnostics, Volume 27, Issue 4, 2025, Pages 306-322, ISSN 1525-1578, https://doi.org/10.1016/j.jmoldx.2025.01.003. April 2025.
Overview
A recent study evaluated the effectiveness and safety of the isatuximab, pomalidomide, and dexamethasone (IsaPd) combination in patients with relapsed or refractory multiple myeloma (RRMM). The results confirmed the treatment's safety and effectiveness, matching findings from a major clinical trial, ICARIA-MM.
The study involved 270 RRMM patients from 51 centers in Italy. The combination of IsaPd showed a high response rate, with 74.1% of patients responding to treatment. This response was even higher in patients with good kidney function and lower-stage disease. The median progression-free survival (PFS) was 15.7 months, which is higher than the results of the ICARIA-MM trial. The median overall survival (OS) was not yet reached, with a 2-year survival rate of 64.2%.
However, patients who had been treated with daratumumab before starting IsaPd had a shorter PFS. The study also showed that patients with high-risk cytogenetics had a worse outcome compared to those with standard-risk genetics.
Side effects were manageable, with 56.3% of patients experiencing severe neutropenia, and 47.8% developing infections. Despite these issues, the treatment was generally well-tolerated.
Overall, IsaPd is a promising treatment for RRMM patients who have already received at least two prior therapies. However, more options like bispecific antibodies and CAR T-cell therapies are needed for high-risk patients or those who are refractory to daratumumab.
"Serum ferritin can serve as a biomarker for the prognosis and increased the prognostic predictive value of ISS/RISS in multiple myeloma patients"
Source
Dong, M., Zhang, J., Yang, L., Li, Y., He, J., & Cai, Z. (2025). Serum ferritin can serve as a biomarker for the prognosis and increased the prognostic predictive value of ISS/RISS in multiple myeloma patients. Biomarkers, 1–13. https://doi.org/10.1080/1354750X.2025.2485142 April 6, 2025.
Overview
This study explored the role of serum ferritin levels in predicting the outcome of multiple myeloma (MM), a type of cancer that affects plasma cells. The study included 302 patients and found that higher ferritin levels were linked to poorer survival and disease progression.
The analysis showed that MM patients with higher ferritin levels had shorter progression-free survival (PFS) and overall survival (OS). The study also confirmed that ferritin levels were an independent factor that could help predict these outcomes, making it a valuable addition to the current staging systems, such as the International Staging System (ISS) and the Revised International Staging System (RISS).
This finding suggests that measuring serum ferritin could improve the ability to predict prognosis in MM patients and supports the need for better biomarkers to help guide treatment decisions.
"Opportunities and challenges for MRD assessment in the clinical management of multiple myeloma"
Source
Paiva, B., Shi, Q., Puig, N. et al. Opportunities and challenges for MRD assessment in the clinical management of multiple myeloma. Nat Rev Clin Oncol (2025). https://doi.org/10.1038/s41571-025-01017-x April 7, 2025.
Overview
Measurable residual disease (MRD) assessment is becoming an important tool for managing multiple myeloma (MM). Advanced tests, like next-generation flow cytometry and sequencing, help doctors evaluate how well a patient is responding to treatment and predict their outlook. MRD negativity, which means no detectable cancer cells, is increasingly accepted as a key goal in treatment and has been used to speed up the approval of new MM therapies.
Studies show that treatments that lead to MRD-negative responses are linked to longer survival for patients. However, challenges still exist in making MRD testing a routine part of patient care. Despite its potential, there are technical and practical issues, and some experts remain cautious about using MRD as a stand-in for treatment success.
This review highlights the current research and suggests ways to overcome these challenges to make MRD testing a more effective part of MM treatment.
"18F-FDG PET/CT Findings in a Case of Plasmablastic Myeloma: A Rare Entity"
Source
Kundu, Nivedita MBBS, MD; Mahalik, Aparna MD; Khan, Dikhra MD; Kumar, Rakesh PhD. 18F-FDG PET/CT Findings in a Case of Plasmablastic Myeloma: A Rare Entity. Clinical Nuclear Medicine ():10.1097/RLU.0000000000005857, April 7, 2025. | DOI: 10.1097/RLU.0000000000005857
Overview
Plasmablastic myeloma (PBM) is a rare form of multiple myeloma that can be difficult to distinguish from plasmablastic lymphoma (PBL), as they share similar symptoms and imaging results. This distinction is important because the treatments for PBM and PBL are different. In this case, PBM developed in a patient with chronic light chain myeloma and showed extensive disease spread outside the bone marrow, including pleural deposits, pleural effusion, and fat around the kidneys. The study highlights the usefulness of 18F-FDG PET/CT scans to differentiate PBM from PBL, assess how far the disease has spread, and identify critical areas that need immediate treatment.
"A Real-World International Staging System (RW-ISS) for patients with newly diagnosed multiple myeloma"
Source
Brieghel, C., Slørdahl, T.S., Andersen, M.N. et al. A Real-World International Staging System (RW-ISS) for patients with newly diagnosed multiple myeloma. Blood Cancer J. 15, 59 (2025). https://doi.org/10.1038/s41408-025-01268-y April 7, 2025.
Overview
The second version of the Revised International Staging System (R2-ISS) for multiple myeloma (MM) was tested and shown to help predict overall survival (OS) in clinical trial patients. However, it was less effective for younger patients. To improve this, a new real-world staging system (RW-ISS) was developed using data from 2,929 Danish patients. The RW-ISS considers factors like age, performance status, certain genetic changes, and lab results to better predict survival. In testing, patients with higher RW-ISS scores had shorter survival times. The RW-ISS performed better than the R2-ISS and original R-ISS in predicting survival. The RW-ISS is recommended for use in everyday clinical practice to help doctors manage newly diagnosed MM patients.
"Elranatamab versus physician's choice of treatment in patients with triple-class exposed/refractory multiple myeloma: an updated matching-adjusted indirect comparison"
Source
Mol I, Hu Y, LeBlanc TW, Cappelleri JC, Chu H, Nador G, Aydin D, Cruz IP, Hlavacek P. Elranatamab versus physician's choice of treatment in patients with triple-class exposed/refractory multiple myeloma: an updated matching-adjusted indirect comparison. J Comp Eff Res. 2025 Apr 7:e240236. doi: 10.57264/cer-2024-0236. Epub ahead of print.
Overview
This study aimed to compare the effectiveness of elranatamab, a treatment for triple-class exposed/refractory multiple myeloma, against the physician's choice of treatment. Using updated data from two studies (MagnetisMM-3 and LocoMMotion), the results showed that elranatamab significantly improved progression-free survival (PFS) and overall survival (OS) compared to the standard treatment. Specifically, patients treated with elranatamab had better survival outcomes, with a 68% lower risk of disease progression and a 50% lower risk of death. The findings support the value of elranatamab as an effective treatment option for these patients.
"A novel indirubin- 3-monoxime derivative I3MV- 8b exhibits remarkable cytotoxicity against multiple myeloma by targeting TRIM28"
Source
Fang, T., Liu, L., Sun, H. et al. A novel indirubin- 3-monoxime derivative I3MV- 8b exhibits remarkable cytotoxicity against multiple myeloma by targeting TRIM28. Biomark Res 13, 57 (2025). https://doi.org/10.1186/s40364-025-00773-3 April 7, 2025.
Overview
This study explored a new treatment for multiple myeloma (MM) called I3MV-8b, which combines two actions to fight cancer: inhibiting the proteasome and blocking autophagy (a process that helps cells survive). I3MV-8b was tested in lab and animal models, showing it could stop MM cell growth and trigger cell death. When used with other proteasome inhibitors, it worked even better by attacking both survival pathways at once. The study also found that I3MV-8b lowers the expression of key genes involved in proteasome and autophagy, making MM cells more sensitive to treatment. This dual-action approach could be a promising new strategy for treating multiple myeloma.
"The butyrate derived from probiotic Clostridium butyricum exhibits an inhibitory effect on multiple myeloma through cell death induction"
Source
Konishi, H., Saito, T., Takahashi, S. et al. The butyrate derived from probiotic Clostridium butyricum exhibits an inhibitory effect on multiple myeloma through cell death induction. Sci Rep 15, 11919 (2025). https://doi.org/10.1038/s41598-025-97038-8 April 7, 2025.
Overview
Some probiotics—"good" bacteria that live in our gut—can make substances that slow down tumor growth, but it’s not clear if they can help with blood cancers like multiple myeloma (MM).
In this study, researchers tested 24 different probiotic strains to see if any could help kill MM cells. One strain, Clostridium butyricum, stood out. It made a substance called butyrate, which reduced the number of MM cells in the lab.
Butyrate was more toxic to cancer cells than to healthy blood cells, which is a good sign. In mice, it also helped shrink tumors. Scientists found that butyrate caused cancer cells to stop dividing and eventually die.
Even more promising: when butyrate was combined with bortezomib, a drug already used to treat MM, the two worked better together than either did alone.
Probiotics—or the substances they make—might someday be used alongside existing treatments to fight blood cancers like MM.
"Dihydromyricetin restores lysosomal function in Schwann cells to alleviate bortezomib-induced peripheral neuropathy via ERK/TFEB signaling"
Source
Liu, X., Zhang, X., Li, X. et al. Dihydromyricetin restores lysosomal function in Schwann cells to alleviate bortezomib-induced peripheral neuropathy via ERK/TFEB signaling. Arch Toxicol (2025). https://doi.org/10.1007/s00204-025-04030-2 April 7, 2025.
Overview
Velcade® (bortezomib) is a key drug used to treat multiple myeloma, a type of blood cancer. But a common side effect—nerve damage (called peripheral neuropathy)—can be painful and hard to treat. Scientists are still trying to understand exactly why it happens.
In this study, researchers looked at a natural plant compound called dihydromyricetin (DHM) to see if it could protect nerves from bortezomib’s side effects. They tested DHM in mice and in lab-grown nerve cells. Here’s what they found:
- Mice given DHM had less nerve pain and better nerve function after bortezomib treatment.
- DHM helped nerve cells (called Schwann cells) clear out waste properly by restoring a process called autophagy, which bortezomib had blocked.
- DHM worked by activating a protein called TFEB, which helps control how cells clean and recycle parts of themselves.
- DHM also blocked a damaging signal (called ERK) triggered by bortezomib.
- Importantly, DHM did not interfere with BTZ’s ability to kill myeloma cells.
DHM might help prevent or treat nerve pain caused by myeloma treatment, without making the treatment less effective. More research is needed, but it could be a helpful add-on therapy in the future.
"Optimal MRD-based end point to support response-adapted treatment cessation in newly diagnosed multiple myeloma"
Source
Smith Giri, Binod Dhakal, Natalie S Callander, Eva Medvedova, Kelly Godby, Bhagirathbhai Dholaria, Susan Bal, Gayathri Ravi, Saurabh Chhabra, Rebecca Silbermann, Luciano J. Costa; Optimal MRD-based end point to support response-adapted treatment cessation in newly diagnosed multiple myeloma. Blood 2025; blood.2024027674. doi: https://doi.org/10.1182/blood.2024027674 April 7, 2025.
Overview
For people newly diagnosed with multiple myeloma, new treatments are helping more patients reach a deep response to therapy. Some newly diagnosed multiple myeloma patients fare so well that doctors are exploring stopping treatment after a certain point. But how can we know when it’s safe to stop?
This study looked at patients who received a strong first treatment (called quadruplet therapy) followed by a stem cell transplant. After that, patients stopped treatment if they had no signs of cancer in their blood using a sensitive test called minimal residual disease (MRD) testing.
Researchers tested five ways to measure how well patients were doing. The best predictor of long-term success was being MRD-negative at two timepoints at least one year apart (called sustained MRD-negative at less than 1 in 100,000 cells, or S-MRD<10⁻⁵).
Patients who stayed MRD-negative at this level had the lowest risk of the cancer coming back and the best progression-free survival.
Staying MRD-negative over time—especially below a certain threshold—may be the best way to decide when it’s safe to stop treatment for multiple myeloma.
"Outcomes of older adults and frail patients receiving idecabtagene vicleucel: a CIBMTR study"
Source
Hamza Hashmi, Surbhi Sidana, Nausheen Ahmed, Matthew Bye, Doris Hansen, Christopher Ferreri, Binod Dhakal, Devender Dhanda, Melanie Harrison, Amani Kitali, Heather Landau, Abu-Sayeef Mirza, Jinalben Patel, Pallavi Patwardhan, Muzaffar Qazilbash, Krina Patel, Taiga Nishihori, Siddhartha Ganguly, Lohith Gowda, Larry D. Anderson, Marcelo C. Pasquini, Saad Usmani, Ciara Louise Freeman; Outcomes of older adults and frail patients receiving idecabtagene vicleucel: a CIBMTR study. Blood Adv 2025; 9 (7): 1587–1592. doi: https://doi.org/10.1182/bloodadvances.2024014970 April 8, 2025.
Overview
CAR T-cell therapy, like Abecma® (ide-cel), is changing the way doctors treat people with relapsed or hard-to-treat multiple myeloma (MM). But there’s been a big question: Is it safe and effective for older or frail patients, who are often left out of clinical trials?
This study looked at real-world data from U.S. patients treated with ide-cel between 2021 and 2023. It focused on patients aged 70 and older, and those who were considered frail using a special frailty score.
The study found the following:
- Older patients (70+) had higher rates of certain side effects, including ICANS (a type of brain-related side effect) and more severe cases of CRS (cytokine release syndrome).
- Even though older patients had more side effects, they also had lower relapse rates and better survival.
- Frail patients had more ICANS and infections, but their survival and cancer outcomes were similar to non-frail patients.
- Importantly, treatment-related death rates did not increase with age or frailty.
Being older or frail shouldn’t automatically rule someone out from getting ide-cel treatment. With careful monitoring, many older and frail patients do well. Still, doctors should watch closely for side effects, especially in frail patients.
"Anxiety and depression among patients newly diagnosed with lymphoma and myeloma"
Source
Oreofe O. Odejide, Angel M. Cronin, Tamryn F. Gray, Tsotso Ablorh, Anna Ying, Amelia Yang, Cheyenne Ashley, Anna Tidswell, Eleanor Shi, Miryam Yusufov, Gregory A. Abel, Lizabeth Roemer; Anxiety and depression among patients newly diagnosed with lymphoma and myeloma. Blood Adv 2025; 9 (7): 1618–1629. doi: https://doi.org/10.1182/bloodadvances.2024014821 April 8, 2025.
Overview
Lymphoma and myeloma are serious blood cancers that take a toll not just on the body, but also on mental health. A recent study looked at 200 adults who had been diagnosed with one of these cancers in the last 6 months. Researchers wanted to know how many of them had signs of anxiety or depression, and what factors might be linked to these feelings.
More than half of the patients (56%) had symptoms of anxiety or depression that were strong enough to be considered clinically significant. About 52% had anxiety, and 28% had depression.
The study found that people who were less satisfied with their finances were more likely to feel anxious or depressed. Patients who had less trust in the medical system were more likely to feel anxious, while those with less social support were more likely to feel depressed.
Depression had a strong negative effect on patients’ quality of life. These results show that emotional health is a big part of living with lymphoma or myeloma—and that more mental health support is needed for people facing these cancers.
"LRP8-dependent cholesterol metabolism modulates mTORC1 signaling and apoptotic pathways in multiple myeloma"
Source
Wang, Y., Lan, T., Zhou, C. et al. LRP8-dependent cholesterol metabolism modulates mTORC1 signaling and apoptotic pathways in multiple myeloma. Cell Death Dis 16, 263 (2025). https://doi.org/10.1038/s41419-025-07625-w April 8, 2025.
Overview
Cholesterol isn’t just important for heart health—it also plays a key role in cancer, including multiple myeloma (MM). A new study looked at 703 patients newly diagnosed with MM and found that low cholesterol levels in the blood were linked to worse outcomes. This drop in cholesterol may be caused by myeloma cells using up extra cholesterol to help them grow.
Researchers discovered that a protein called LRP8 may be behind this increased cholesterol use. Myeloma cells with high levels of LRP8 absorb and use more cholesterol. Lab and animal studies showed that blocking LRP8 caused the cancer cells to die and stop growing. It also slowed down a key cancer-related pathway that depends on cholesterol.
Blocking LRP8 also triggered a process called autophagy, where cells clean out damaged parts to survive. But if this backup process was also blocked, the myeloma cells died even faster.
This research shows that LRP8 is a major player in how myeloma cells use cholesterol. Targeting LRP8 might be a promising new way to treat multiple myeloma.
"Paradigm shift in age of multiple myeloma patients: a study from a tertiary care government oncology hospital in Pakistan"
Source
Habib, M., Aslam, W. & Aziz, S. Paradigm shift in age of multiple myeloma patients: a study from a tertiary care government oncology hospital in Pakistan. BMC Res Notes 18, 147 (2025). https://doi.org/10.1186/s13104-025-07223-1 April 8, 2025.
Overview
A recent study from a government hospital in Islamabad, Pakistan looked at 81 patients diagnosed with myeloma over eight years to better understand how the disease appears in this setting.
The average age at diagnosis was 53, and nearly 80% of patients were 60 or younger—much younger than patients typically seen in wealthier countries. Most patients were men. The most common first symptom was bone pain, followed by tiredness and frequent infections.
Doctors found that the most common type of myeloma involved a protein called IgG. Many patients were already in advanced stages of the disease by the time they were diagnosed—only 12% were in early stage (Stage I), while most were in Stage II or III.
This is the first study from a public hospital in Pakistan to look at how myeloma affects patients there. The authors say more research is needed from both public and private hospitals to understand why patients in Pakistan are being diagnosed younger and often at more advanced stages.
"Changing lanes: extending CAR T-cell therapy to high-risk plasma cell dyscrasias"
Source
Morgan HT, Derman BA, Ma H, Kumar SK. Changing lanes: extending CAR T-cell therapy to high-risk plasma cell dyscrasias. Front Immunol. 2025 Apr 8;16:1558275. doi: 10.3389/fimmu.2025.1558275.
Overview
CAR T-cell therapy has shown great success in blood cancers like leukemia, lymphoma, and especially multiple myeloma. Now, researchers are exploring whether this powerful therapy could also help people with two other tough-to-treat plasma cell disorders: AL amyloidosis and plasma cell leukemia (PCL).
AL amyloidosis happens when abnormal plasma cells produce harmful proteins that build up in organs like the heart and kidneys, leading to serious damage. Plasma cell leukemia is a rare and aggressive form of blood cancer with poor survival rates, even with intensive treatment.
Both diseases are usually treated with chemotherapy and, for some patients, a stem cell transplant. But many people with AL amyloidosis aren’t healthy enough for a transplant, and people with PCL often relapse after treatment.
So far, clinical trials of CAR T-cell therapy have mostly excluded patients with AL amyloidosis and PCL. But because CAR T has worked well in multiple myeloma—which also involves plasma cells—scientists believe it might help these patients too.
This review highlights the urgent need for research into using CAR T-cell therapy for plasma cell diseases beyond myeloma, offering hope for better, shorter, and more effective treatment options.
"Teclistamab for Patients with Heavily Pretreated Relapsed/Refractory Multiple Myeloma and Renal Impairment"
Source
Danai Dima, Aimaz Afrough, Utkarsh Goel, Ariel Grajales-Cruz, Jack Khouri, Kelley Julian, Oren Pasvolsky, Rahul Banerjee, Beatrice Razzo, Christopher J Ferreri, Mariola Alejandra Vazquez Martinez, James A Davis, Aishwarya Sannareddy, Omar Castaneda Puglianini, Shahzad Raza, Andrew J. Portuguese, Mahmoud R. Gaballa, Masooma Rana, Alex Lieberman-Cribbin, Shaun DeJarnette, Rebecca Gonzalez, Anna Chen, Megan M. Herr, Lakha Mikkilineni, Hitomi Hosoya, Evguenia Ouchveridze, Gurbakhash Kaur, Adriana C Rossi, Leyla Shune, Faiz Anwer, Yi Lin, Shambavi Richard, Douglas W Sborov, Rachid C Baz, Alfred Garfall, Hans C. Lee, Larry D. Anderson, Andrew J Cowan, Krina K. Patel, Peter M Voorhees, Surbhi Sidana, Doris K Hansen, Shebli Atrash, Sandra P Susanibar-Adaniya; Teclistamab for Patients with Heavily Pretreated Relapsed/Refractory Multiple Myeloma and Renal Impairment. Blood Adv 2025; bloodadvances.2025016059. doi: https://doi.org/10.1182/bloodadvances.2025016059 April 8, 2025.
Overview
Many people with relapsed or hard-to-treat multiple myeloma also have kidney problems, but they’re often left out of clinical trials. A new study examined how well Tecvayli® (teclistamab), a type of bispecific antibody, works in these patients.
Out of 384 patients treated with teclistamab, 81 had renal impairment (meaning their kidneys weren’t working well), and 18 of those were on dialysis. These patients were a bit older and had tried more treatments in the past.
Side effects like cytokine release syndrome (CRS) and neurotoxicity were about the same in patients with and without kidney problems. Some blood-related side effects, like low red blood cells and low platelets, were more common in patients with kidney issues, but their kidney function didn’t get worse unless their cancer progressed.
Importantly, response rates and survival times were similar between patients with and without kidney problems. This suggests teclistamab is safe and effective, even for those on dialysis.
This study offers hope for people with multiple myeloma and kidney issues, showing they, too, may benefit from this promising new treatment.
"Longitudinal analysis of gut microbiome and metabolome correlates of response and toxicity with idecabtagene vicleucel"
Source
Satabdi Saha, Lubna Rehman, Abdur Rehman, Faezeh Darbaniyan, Donna M Weber, Melody Becnel, Mahmoud R. Gaballa, Sheeba K Thomas, Hans C. Lee, Chia-Chi Chang, Reetakshi Arora, Meghan Menges, Salvatore Corallo, Marco L. Davila, Frederick L. Locke, Mark R Tanner, Sattva S. Neelapu, Elizabeth J. Shpall, Christopher R. Flowers, Robert Z Orlowski, Robert R Jenq, Michael D Jain, Christine Peterson, Doris K Hansen, Neeraj Y Saini, Krina K. Patel; Longitudinal analysis of gut microbiome and metabolome correlates of response and toxicity with idecabtagene vicleucel. Blood Adv 2025; bloodadvances.2024014476. doi: https://doi.org/10.1182/bloodadvances.2024014476 April 8, 2025
Overview
Researchers are learning that the gut microbiome—the mix of bacteria in your digestive system—might affect how well patients respond to CAR-T therapy and how likely they are to have side effects. A recent study looked at 33 multiple myeloma patients treated with Abecma® (idecabtagene vicleucel, or ide-cel), a type of CAR-T therapy.
The team collected 117 stool samples and used DNA testing to analyze the bacteria. After treatment, patients had less gut bacteria diversity, and certain bacteria were linked to better or worse outcomes.
Patients who responded well to ide-cel had higher levels of specific bacteria, including Flavonifractor plautii and Bacteroides thetaiotaomicron. But when one type of bacteria dominated the gut, especially Enterococcus, patients were more likely to experience serious side effects like cytokine release syndrome (CRS)—a common and sometimes dangerous reaction to CAR-T therapy.
The researchers also found that certain bacteria were tied to helpful stool chemicals, which might play a role in how the immune system reacts to treatment.
These findings suggest that the gut microbiome could become an important tool for predicting how patients will respond to CAR-T therapy and may even be something doctors can adjust to help improve outcomes and reduce risks.
"A Randomized Controlled ‘REAL-FITNESS’ Trial to Evaluate Physical Activity in Patients With Newly Diagnosed Multiple Myeloma"
Source
Dreyling, E., Räder, J., Möller, M.-D., Ihorst, G., Wenger, S., Pahl, A., Arends, J., Herget, G., Deibert, P., Wäsch, R. and Engelhardt, M. (2025), A Randomized Controlled ‘REAL-FITNESS’ Trial to Evaluate Physical Activity in Patients With Newly Diagnosed Multiple Myeloma. Journal of Cachexia, Sarcopenia and Muscle, 16: e13793. https://doi.org/10.1002/jcsm.13793 April 8, 2025.
Overview
A new study shows that staying active during treatment can help people with multiple myeloma (MM) feel better and manage side effects.
In this study, 34 patients with newly diagnosed MM were split into two groups. One group followed World Health Organization (WHO) exercise guidelines—150 minutes of aerobic activity and two strength training sessions per week. The other group continued with their usual level of activity. The patients were treated with a common drug combination (VCd: Velcade, or bortezomib; Cytoxan, or cyclophosphamide; and dexamethasone) and tracked using smartwatches and diaries.
After three months:
- The exercise group was twice as active as the control group.
- No exercise-related injuries were reported.
- People who exercised had fewer side effects from treatment, were less likely to be hospitalized, and needed fewer dose changes.
- They also had less fatigue (6% vs. 75%), less depression (6% vs. 44%), and better mobility and strength by the end of treatment.
Although some markers like blood tests and treatment response didn’t show big differences, the people who exercised clearly felt and functioned better.
Exercise is safe and beneficial for people with multiple myeloma during treatment. It can reduce side effects and boost quality of life. More programs are needed to support physical activity in MM care.
"Real-world treatment patterns for teclistamab and talquetamab in multiple myeloma (MM): experience from 609 patients"
Source
Chunara, F., Lugo, C., Osinski, K. et al. Real-world treatment patterns for teclistamab and talquetamab in multiple myeloma (MM): experience from 609 patients. Blood Cancer J. 15, 61 (2025). https://doi.org/10.1038/s41408-025-01254-4 April 8, 2025
Overview
Tecvayli® (teclistamab) and Talvey® (talquetamab) are two bispecific antibody treatments approved by the FDA for people with relapsed or hard-to-treat multiple myeloma (MM). These drugs work by directing the immune system to attack myeloma cells and have shown strong results in early clinical trials. Typically, patients begin treatment with a “step-up dosing” phase: three lower doses given a few days apart to help the body adjust. After that, teclistamab is usually given once a week, but can be switched to every two weeks if the patient responds well. Talquetamab is typically given either once a week or every two weeks.
This study looked at how these drugs are being used in real-world settings by analyzing data from over 600 patients across 40 U.S. healthcare centers. Most patients were able to complete the step-up phase safely. While many still receive this phase in the hospital, there’s a growing interest in shortening hospital stays by giving the step-up doses more closely together, every 48 hours instead of every few days. In most cases, doctors did not need to repeat the step-up process, even when treatment was paused or delayed.
For teclistamab, weekly dosing was common at first, but many patients eventually switched to every-other-week or even once-a-month dosing after several months. This trend was also seen in talquetamab treatment, where every-other-week dosing was the most common schedule throughout. Less frequent dosing may be enough to maintain remission for many patients while lowering the risk of side effects and easing the burden of treatment.
These findings suggest that more flexible dosing schedules may be both safe and effective for some patients. Doctors are now exploring whether these treatments can be tailored based on how well a patient is responding. The ultimate goal is to maintain strong responses while reducing the number of doses, improving quality of life, and making treatment more manageable for people living with multiple myeloma.
"Psychometric properties of the Chinese version of the M.D. Anderson symptom Inventory-Multiple Myeloma Module: a translation and validation study"
Source
Wang, T., Chen, W., Lin, Y. et al. Psychometric properties of the Chinese version of the M.D. Anderson symptom Inventory-Multiple Myeloma Module: a translation and validation study. BMC Cancer 25, 640 (2025). https://doi.org/10.1186/s12885-025-14054-7 April 8, 2025.
Overview
Researchers tested a Chinese version of a symptom-tracking tool called the MDASI-MM, which helps doctors understand the symptoms of people with multiple myeloma. This tool was originally developed at MD Anderson Cancer Center and includes questions about both common cancer symptoms and those specific to multiple myeloma. The goal of the study was to make sure the Chinese version was accurate, easy to understand, and reliable for patients in China.
To do this, the tool was carefully translated using a multi-step process that included both forward and back translation, followed by a test run with patients to make sure it made sense culturally and linguistically. The researchers then tested the tool with 500 multiple myeloma patients from three hospitals in Zhejiang Province to check how well it measured symptoms and whether the results were consistent.
The final Chinese version includes 26 questions that cover common symptoms, multiple myeloma–specific symptoms, and how these symptoms affect daily life. The results showed that the Chinese version was both valid and reliable. In other words, it accurately measures what it's supposed to and produces consistent results.
This means healthcare providers in China can confidently use the MDASI-MM to better understand their patients’ symptoms. With this tool, they can offer more targeted care and develop better strategies to help manage symptoms and improve quality of life for people living with multiple myeloma.
"Multiple Myeloma Experiences and Preferences: A Mixed Methods Study of Patients and Care Partners in the United States"
Source
Flora DR, Byrd R, Platt DA, Hlavacek P, Hoag Goldman E, Cappelleri JC, Kennedy CT, LeBlanc TW. Multiple Myeloma Experiences and Preferences: A Mixed Methods Study of Patients and Care Partners in the United States. Patient Prefer Adherence. 2025;19:963-979
https://doi.org/10.2147/PPA.S491263 April 8, 2025.
Overview
This study explored how patients with multiple myeloma (MM) nd their care partners make these decisions as the disease progresses and treatment options change.
The study involved 32 MM patients and 10 care partners, focusing on their treatment experiences. The researchers found that the treatment landscape for MM has become more complex, with patients seeking specialist care earlier in their journey. When deciding on treatment, patients and their care partners prioritized extending survival, improving quality of life, and maintaining remission. As patients moved further along in their treatment, decision-making became more collaborative, with patients taking on more of an active role in advocating for their care.
The study also revealed that many patients struggled to find clear, trustworthy, and easy-to-understand information about their treatment options. Younger patients, in particular, had unique concerns, such as reproductive health and long-term financial stability. Support groups and advocacy organizations were essential in providing reliable information that patients found empowering.
The findings emphasize the importance of collaborative discussions between doctors, patients, and care partners. These discussions help build trust and give patients more control over their treatment. Ensuring that patients have access to accurate information and support resources—especially for younger patients—is critical in addressing their evolving needs and improving treatment outcomes.
"A phase 1 trial of fully human BCMA CAR-T therapy for relapsed/refractory multiple myeloma with 5-year follow-up"
Source
Sherilyn A Tuazon, Andrew J. Portuguese, Margot J Pont, Andrew J Cowan, Gabriel O Cole, Blythe D Sather, Xiaoling Song, Sushma Thomas, Brent L. Wood, Michelle Lee Blake, Melissa G Works, Mazyar Shadman, Emily C. Liang, Qian Wu, Jenna M Voutsinas, Ted A Gooley, Cameron J Turtle, Brian G. Till, David G Coffey, David G Maloney, Stanley R. Riddell, Damian J Green; A phase 1 trial of fully human BCMA CAR-T therapy for relapsed/refractory multiple myeloma with 5-year follow-up. Blood 2025; blood.2024027681. doi: https://doi.org/10.1182/blood.2024027681 April 8, 2025.
Overview
FCARH143 is a new CAR-T cell therapy designed to target BCMA, a protein found on multiple myeloma cells. It was tested in a phase I trial (NCT03338972) to treat relapsed/refractory multiple myeloma (RRMM). In the study, patients received chemotherapy to prepare their bodies for the CAR-T cells, followed by increasing doses of the therapy. The main goal of the trial was to assess the safety of FCARH143, with secondary goals including how well it worked in treating the disease.
The trial included 28 patients, with a median age of 64, and most had already received multiple treatments for their myeloma. Among the 25 patients who received the treatment, 84% experienced cytokine release syndrome (CRS), though only 8% had severe (grade 3-4) cases. About 24% had neurotoxicity, but no patient experienced life-threatening side effects. After a median follow-up of 67.3 months, all treated patients responded to the therapy, with 64% achieving a stringent complete response. The median progression-free survival (PFS) was 15.5 months, and the overall survival (OS) was 32.1 months. Even when considering all patients, including those who didn't receive treatment, the response rate remained high at 89.3%, with an OS of 30.2 months.
FCARH143 showed strong effectiveness in treating RRMM, with a 100% response rate and manageable side effects. This promising therapy warrants further testing, especially for high-risk patients.
"Economic Impact of Elranatamab for Treatment of Patients with Relapsed or Refractory Multiple Myeloma"
Source
Shah B, Sandin R, Liu Y, Bobolts LR, Hu Y, Mol I, Schepart A, Hughes DM, Hart J, Hlavacek P. Economic Impact of Elranatamab for Treatment of Patients with Relapsed or Refractory Multiple Myeloma. Clinicoecon Outcomes Res. 2025;17:289-302
https://doi.org/10.2147/CEOR.S501404 April 8, 2025.
Overview
A recent study looked at the financial impact of adding Elrexffio® (elranatamab)—a treatment for relapsed or refractory multiple myeloma (RRMM)—to U.S. health insurance plans. The researchers focused on adults who had already received at least four different treatments, including a proteasome inhibitor, an immunomodulatory drug, and an anti-CD38 monoclonal antibody. The goal was to see how elranatamab compares in terms of total cost and value, especially in relation to how long patients stay free from disease progression (called progression-free survival, or PFS).
Using data from clinical trials, government sources, and real-world studies, the researchers built an economic model to predict the cost of adding elranatamab to both commercial and Medicare insurance plans. They found that about 14 commercial and 60 Medicare patients per year would qualify for the drug. Over three years, the estimated cost increase would be just $0.05 per member per month (PMPM) for commercial plans and $0.20 PMPM for Medicare, indicating a small budget impact.
When comparing the cost of care per month of progression-free survival, elranatamab was found to be more cost-effective than most other treatments, including Tecvayli® (teclistamab), talquetamab, and CAR T-cell therapies like idecabtagene vicleucel. Only one other therapy, ciltacabtagene autoleucel, had a lower monthly cost.
Adding elranatamab to treatment options for RRMM is expected to have a low financial impact on health plans while offering good value for the money, especially when compared to other available treatments.
"Early detection of cardiac amyloidosis in patients with multiple myeloma by echocardiography: Is it suitable?"
Source
Yu Fu Ferrari Chen, Alberto Aimo, Michele Emdin, Early detection of cardiac amyloidosis in patients with multiple myeloma by echocardiography: Is it suitable?, International Journal of Cardiology, 2025, 133253, ISSN 0167-5273, https://doi.org/10.1016/j.ijcard.2025.133253. April 9, 2025.
Overview
Many people with multiple myeloma (MM) also develop a condition called light-chain amyloidosis (AL), where abnormal proteins build up in organs like the heart. When these proteins collect in the heart, it’s called cardiac amyloidosis (CA). This heart involvement happens in nearly half to almost all MM patients, depending on how it's diagnosed.
Having heart problems from amyloidosis makes MM more dangerous and can change how doctors treat the disease. But spotting heart involvement early is tough because there’s no clear, standard way to screen for it. Blood tests that look at heart biomarkers can help rule out heart problems, but high levels can also be caused by other issues, like kidney disease, which makes the results harder to understand. There's also no clear rule on when these tests should be done.
Cardiac MRI is a very accurate way to detect amyloidosis in the heart, but it’s not always available and can’t be used in people with kidney problems. This makes early diagnosis of heart involvement in MM a real challenge.
"Bortezomib, melphalan, and prednisone with or without daratumumab in transplant-ineligible patients with newly diagnosed multiple myeloma (ALCYONE): final analysis of an open-label, randomised, multicentre, phase 3 trial"
Source
Mateos, Maria-Victoria et al. Bortezomib, melphalan, and prednisone with or without daratumumab in transplant-ineligible patients with newly diagnosed multiple myeloma (ALCYONE): final analysis of an open-label, randomised, multicentre, phase 3 trial. The Lancet Oncology, Volume 0, Issue 0. DOI: 10.1016/S1470-2045(25)00018-X. April 9, 2025.
Overview
A large clinical trial called ALCYONE looked at whether adding the drug daratumumab to a standard treatment mix (bortezomib, melphalan, and prednisone, or VMP) could help people with newly diagnosed multiple myeloma who can’t get a stem cell transplant, usually because of age or other health problems.
The final results show that the group who got daratumumab lived much longer, with a median survival of 83 months compared to 53.6 months for those on standard treatment alone.
The safety profile was similar between the two groups, though some serious side effects did occur in both. These findings confirm that daratumumab is a powerful frontline option for patients who aren’t able to receive a stem cell transplant.
"Lenalidomide-based triplets in relapsed/refractory multiple myeloma: analysis of the Czech Myeloma Group"
Source
Minarik, J., Pour, L., Latal, V. et al. Lenalidomide-based triplets in relapsed/refractory multiple myeloma: analysis of the Czech Myeloma Group. BMC Cancer 25, 651 (2025). https://doi.org/10.1186/s12885-025-14087-y April 9, 2025.
Overview
Even though treatment for multiple myeloma (MM) has improved a lot over the past 20 years, most patients eventually relapse and need new options. This study looked at how well three common treatment combinations work for people with relapsed or hard-to-treat MM. Each combo included the drug Revlimid (lenalidomide) plus one of three other drugs: Darzalex (daratumumab), Kyprolis (carfilzomib), or Ninlaro (ixazomib).
Researchers studied 538 patients who were being treated for their first to third relapse. They compared how many people responded to each treatment, how long they stayed in remission (progression-free survival), and how long they lived overall (overall survival).
The Darzalex, Revlimid, and dexamethasone (DRd) combination had the best results, with the highest response rate (91.4%) and the longest time without the disease getting worse (about 24 months). Kyporlis, Revlimid, and dexamethasone (KRd) and Ninlaro, Revlimid, and dexamethasone (IRD) had lower results, especially for patients who got IRD later in their treatment journey.
The study also showed that patients with high-risk disease didn’t benefit as much from any of the combinations.
This research supports using the DRD combo for better, longer-lasting results in real-world settings.
"Tumor burden quantified by soluble B-cell maturation antigen and metabolic tumor volume determines myeloma CAR-T outcomes"
Source
Ciara L. Freeman, Jerald Noble, Meghan Menges, Ricardo Villanueva, Justyn Y. Nakashima, Nicholas B. Figura, Rolf Petter Tonseth, Dietrich Werner Idiaquez, Lawrence Skelson, Eric Smith, Julieta Abraham-Miranda, Salvatore Corallo, Gabriel De Avila, Omar A. Castaneda Puglianini, Hien Liu, Melissa Alsina, Taiga Nishihori, Kenneth H. Shain, Rachid Baz, Brandon Blue, Ariel Grajales-Cruz, John M. Koomen, Reginald M. Atkins, Doris K. Hansen, Ariosto S. Silva, Jongphil Kim, Yoganand Balagurunathan, Frederick L. Locke; Tumor burden quantified by soluble B-cell maturation antigen and metabolic tumor volume determines myeloma CAR-T outcomes. Blood 2025; 145 (15): 1645–1657. doi: https://doi.org/10.1182/blood.2024024965 April 10, 2025.
Overview
CAR T-cell therapy is an exciting new treatment for people with relapsed or hard-to-treat multiple myeloma. But it’s not easy to give, and it doesn’t work for everyone. That’s why researchers are looking for ways to predict who will benefit the most from it.
In this study, scientists looked at two possible clues in the body:
- sBCMA – a protein found in the blood
- MTV – a measure of tumor size seen on special PET-CT scans
They studied 183 patients before they received CAR T-cell therapy. They found that:
- High sBCMA levels were linked to more cancer in the body, more side effects, and shorter time before the cancer came back.
- High MTV was also a sign of worse outcomes, including shorter survival.
Interestingly, some patients had low sBCMA but high MTV. These patients often had very little of the BCMA target on their cancer cells—and they didn’t respond well to treatment.
These results suggest that using both sBCMA and MTV can help doctors better choose who should get CAR T therapy and maybe even improve how it’s used in the future.
"sBCMA and MTV prediction of outcome for CAR T"
Source
Hermann Einsele, K. Martin Kortüm; sBCMA and MTV prediction of outcome for CAR T. Blood 2025; 145 (15): 1594–1595. doi: https://doi.org/10.1182/blood.2024027446 April 10, 2025.
Overview
CAR T-cell therapy is a powerful treatment for people with relapsed or hard-to-treat multiple myeloma (MM). It works by using a patient’s own immune cells to fight cancer. While it can lead to deep responses, it's complex to give, can cause serious side effects, and doesn't work for everyone. Researchers are trying to find better ways to predict who will benefit most from this therapy.
A new study looked at two possible clues:
- sBCMA: a protein in the blood linked to myeloma cells
- MTV: a scan that measures how much cancer is in the body
They studied 183 patients who received BCMA-targeted CAR T-cell therapy. Here’s what they found:
- High sBCMA and high MTV levels before treatment were linked to worse outcomes, like shorter survival and more side effects.
- Some patients had high MTV but low sBCMA. These patients often had very little BCMA on their cancer cells—and didn’t respond well to treatment.
- High sBCMA may interfere more with other treatments like bispecific antibodies, but not as much with CAR T-cells, which seem to stay effective even at high levels of sBCMA.
The study suggests that testing sBCMA and MTV before treatment can help doctors make better choices about how to treat each patient. For patients with high tumor burden, giving treatment to reduce cancer before CAR T (called bridging therapy) may make the main treatment safer and more effective.
This research could lead to more personalized, safer, and stronger treatment plans for people with multiple myeloma.
"Efficacy and safety of daratumumab in intermediate/high-risk smoldering multiple myeloma: final analysis of CENTAURUS"
Source
Ola Landgren, Ajai Chari, Yael C. Cohen, Andrew Spencer, Peter M. Voorhees, Irwindeep Sandhu, Matthew W. Jenner, Dean Smith, Michele Cavo, Niels W. C. J. van de Donk, Meral Beksac, Philippe Moreau, Hartmut Goldschmidt, Diego Vieyra, Linlin Sha, Liang Li, Els Rousseau, Robyn Dennis, Robin Carson, Craig C. Hofmeister; Efficacy and safety of daratumumab in intermediate/high-risk smoldering multiple myeloma: final analysis of CENTAURUS. Blood 2025; 145 (15): 1658–1669. doi: https://doi.org/10.1182/blood.2024025897 April 10, 2025.
Overview
Smoldering multiple myeloma (SMM) is an early form of myeloma that hasn’t yet caused symptoms. Yet, some people with higher-risk SMM will go on to develop full-blown multiple myeloma (MM). The big question: Can early treatment delay or even prevent that from happening?
The CENTAURUS study looked at whether using a medicine called daratumumab early on could help. Daratumumab is already used to treat active myeloma. In this study, 123 people with intermediate- or high-risk SMM got daratumumab through an IV in one of three dosing schedules:
- Long-intense
- Intermediate
- Short-intense
After about 7 years of follow-up:
- People in the long-intense and intermediate groups responded better to treatment than those in the short-intense group.
- Around half of the patients in the long-intense and intermediate groups saw their disease improve.
- Progression to full-blown myeloma or death happened less often in the longer treatment groups.
- Some patients continued treatment beyond the main study and tolerated it well.
- No new safety concerns were found.
These results suggest that daratumumab is safe over the long term and may help delay the progression of SMM in people at higher risk. More research is ongoing to see if this early treatment approach should become standard
"Daratumumab: a game changer in early myeloma"
Source
María-Victoria Mateos, Verónica González-Calle; Daratumumab: a game changer in early myeloma. Blood 2025; 145 (15): 1595–1596. doi: https://doi.org/10.1182/blood.2024027932 April 10, 2025
Overview
Researchers are looking at new ways to treat smoldering multiple myeloma (SMM)—a precursor stage of myeloma where the disease is present but hasn’t caused symptoms yet. One drug being studied is Darzalex® (daratumumab), which is already approved and widely used for active multiple myeloma.
A recent study called CENTAURUS looked at using daratumumab earlier in people with higher-risk SMM. The study tested three different treatment plans using daratumumab alone. The “long-intense” plan worked best and followed a similar schedule already used in regular myeloma care. After nearly 7 years, daratumumab showed promising results with manageable side effects. However, since this was only a phase II study, more research is needed before this approach can become a standard option.
A phase III study called AQUILA has already shown that daratumumab can significantly delay the time it takes for SMM to turn into full-blown myeloma. But that raises new questions: Should we compare new treatments like daratumumab to doing nothing (just watching), or to other early treatment options like Revlimid® (lenalidomide) and dexamethasone (Rd), which have already shown strong results?
Other more intense treatment options are also being studied. These include combination therapies, stem cell transplants, and newer BCMA-targeted therapies like CAR T-cell therapy. These treatments may even offer the chance of a cure. Yet, they also come with more risks and side effects. Choosing the right option may depend on the patient’s age, health, and personal preferences.
Another challenge is figuring out when a patient with SMM should start treatment. Doctors usually wait for clear signs of disease progression (like anemia or bone damage), but this may be too late for some. On the flip side, starting treatment too early might expose patients to side effects they don’t yet need.
In the end, the commentary highlights three key takeaways:
- We need better tools, like genetic tests, to predict who’s truly at risk of progressing to myeloma.
- For high-risk patients, we should aim for the most effective treatment—maybe even a cure.
- Most importantly, patients should be part of the decision-making process. Their feelings about starting treatment before symptoms appear really matter.
"Daratumumab, Lenalidomide, and Dexamethasone Versus Bortezomib, Lenalidomide, and Dexamethasone in Transplant-Ineligible Newly Diagnosed Multiple Myeloma: A Systematic Literature Review and Meta-Analysis."
Source
Gordan, L.N., Medhekar, R., Fu, A.Z., Shokoohi, M., Oliva Ramirez, A., Bonar, N., Nguyen, B.-N., Spence, M., McTavish, R., Disher, T., Gautam, S., Gupta-Werner, N., Kaila, S. and Patel, A.J. (2025), Daratumumab, Lenalidomide, and Dexamethasone Versus Bortezomib, Lenalidomide, and Dexamethasone in Transplant-Ineligible Newly Diagnosed Multiple Myeloma: A Systematic Literature Review and Meta-Analysis. Hematological Oncology, 43: e70061. https://doi.org/10.1002/hon.70061 April 10, 2025.
Overview
For people with newly diagnosed multiple myeloma who can’t have a stem cell transplant, doctors often start treatment with one of two combinations:
- Darzalex® (daratumumab), Revlimid® (lenalidomide), and dexamethasone (DRd)
- Velcade® (bortezomib), Revlimid (lenalidomide), and dexamethaone (VRd)
Both are recommended by medical guidelines. But which one works better?
Researchers looked at recent studies to compare DRd and VRd in patients who are transplant-ineligible (TIE). They reviewed medical databases and conference data, and carefully selected three studies for a deeper analysis.
Since there are no direct head-to-head trials comparing DRd and VRd, the researchers used a method called meta-analysis to combine the data from different studies. Here’s what they found:
Patients who received DRd had a lower risk of disease progression or death compared to those who received VRd.
Even after adjusting for differences in study quality, DRd still came out ahead, with a hazard ratio of 0.56. This means patients on DRd were 44% less likely to see their disease worsen or die during the study period.
While more research is still needed—especially direct comparison trials—this analysis suggests that DRd may be the better first treatment for people with multiple myeloma who aren’t eligible for a transplant.
"Myeloma mesenchymal stem cells’ bioenergetics afford a novel selective therapeutic target"
Source
Komemi, O., Orbuch, E., Jarchowsky-Dolberg, O. et al. Myeloma mesenchymal stem cells’ bioenergetics afford a novel selective therapeutic target. Oncogenesis 14, 9 (2025). https://doi.org/10.1038/s41389-025-00554-5 April 11, 2025.
Overview
Multiple myeloma (MM) grows in the bone marrow. Scientists know that the cancer doesn’t grow alone; it gets help from nearby bone marrow stem cells, called BM-MSCs. These helper cells give energy and support to the cancer cells, helping them survive treatments.
In this study, researchers looked at the mitochondria (the energy centers of cells) in BM-MSCs from both healthy people and people with myeloma. They found that in people with myeloma, the BM-MSCs had damaged mitochondria and showed signs of stress. To cope, the cells activated backup systems that helped them keep producing energy.
One key discovery: these support cells in myeloma used a protein called STAT3 to keep their energy going. When researchers blocked this energy backup system using a drug called Venetoclax, the BM-MSCs lost their ability to support the cancer cells. Even better, when Venetoclax was combined with the standard myeloma drug Velcade® (bortezomib), it made the treatment work more effectively.
Targeting the energy system of BM-MSCs could be a new way to fight multiple myeloma. It can do this not by attacking the cancer cells directly, but by weakening the cells that help them.
"Persistent opioid use is associated with a shorter myeloma survival"
Source
Sweiss, K., Rodriguez, A.M.A., Quigley, J.G. et al. Persistent opioid use is associated with a shorter myeloma survival. Support Care Cancer 33, 374 (2025). https://doi.org/10.1007/s00520-025-09407-8 April 11, 2025.
Overview
Opioids are strong painkillers that are often used to help manage pain in multiple myeloma (MM), especially during and after treatment like stem cell transplants. But there are concerns about how long-term opioid use (called chronic opioid use, or COU) may affect overall health, especially once the cancer is under control.
In this study, doctors reviewed the records of 174 people with multiple myeloma who were treated at a large city hospital. Most of the patients were people of color, and all had received a stem cell transplant as part of their treatment.
They found that over half of the patients were already using opioids regularly before their transplant, even if they didn’t have bone damage from the cancer. After the transplant, most patients were given opioids in the hospital. At 6 months after transplant, about 4 in 10 patients were still using opioids regularly. People who were sent home with opioids were more likely to still be using them at the 6-month mark. Importantly, those still using opioids at 6 months had a lower overall survival rate.
This study suggests that long-term opioid use may be linked to worse survival in people with myeloma, even though it doesn’t seem to affect how long the cancer stays in remission. It highlights the need to better understand the risks of opioid use and to find safer, more effective ways to manage pain in myeloma patients.
"Practical guidance on the prevention and management of infection in multiple myeloma patients: A case-based approach"
Source
A. Kannan, K. Jeffrey, S. Misbah, K. Ramasamy, Practical guidance on the prevention and management of infection in multiple myeloma patients: A case-based approach, Blood Reviews, 2025, 101287, ISSN 0268-960X, https://doi.org/10.1016/j.blre.2025.101287. April 11, 2025.
Overview
Myeloma patients are at a greater risk for infections for these reasons:
- The disease itself weakens the immune system.
- Most patients are older, which means the immune system may already be less effective due to age.
- Other health conditions common in older adults can also increase the risk.
New treatments, including advanced therapies like T-cell therapy, are helping people live longer with better cancer control. Yet, some of these treatments can make the immune system even weaker.
Doctors are now seeing cases where some older patients are more at risk of dying from infections than from the cancer itself. The good news is that there are steps you and your care team can take to help protect you from serious infections. This includes:
- Preventive medications (prophylaxis)
- Regular monitoring for early signs of infection
- Personalized infection prevention plans based on your treatment, age, and overall health
By staying informed and working closely with your care team, you can reduce your risk and stay focused on living well with multiple myeloma.
"Establishment of patient-derived xenograft models in Chinese patients with multiple myeloma: Insights into therapeutic responsiveness and molecular subtyping"
Source
Xueju Wang, Di Zhou, Chanjuan Jin, Zhangzhen Shi, Danyi Wen, Lintao Bi, Establishment of patient-derived xenograft models in Chinese patients with multiple myeloma: Insights into therapeutic responsiveness and molecular subtyping, Translational Oncology, Volume 56, 2025, 102385, ISSN 1936-5233, https://doi.org/10.1016/j.tranon.2025.102385. April 11, 2025.
Overview
In this study, scientists in China tested a new way to better understand and treat multiple myeloma by using patient-derived xenograft (PDX) models. These are special lab models made by taking cancer cells from a real patient and placing them into mice with very weak immune systems.
They found the following:
- They successfully created PDX models from three different patients.
- These models reacted to treatment in the same way the patients did, showing similar sensitivity or resistance to certain drugs.
- Using advanced testing, the researchers confirmed that the PDX models closely matched the patients’ specific types of myeloma.
This research shows that PDX models could help doctors test different treatments in the lab before trying them on patients. It could lead to more personalized care, where treatments are tailored to the exact needs of each person based on their unique type of myeloma.
This study is a step toward better, more targeted options for people living with multiple myeloma, especially in China, where this research took place.
"Stem Cell Mobilization Yields with Daratumumab (Dara) and Lenalidomide (Len)-Containing Quadruplet Induction Therapy in Patients with Newly Diagnosed Multiple Myeloma (NDMM): A Real-World Experience at Two Institutes"
Source
Cindy Varga, Myra Robinson, Vikas Gupta, Craig C. Hofmeister, Ajay K. Nooka, Jonathan L. Kaufman, Madhav V. Dhodapkar, Sagar Lonial, Shanice Borden, Christopher Ferreri, Barry Paul, Shebli Atrash, Manisha Bhutani, Peter M. Voorhees, Nisha S. Joseph, Stem Cell Mobilization Yields with Daratumumab (Dara) and Lenalidomide (Len)-Containing Quadruplet Induction Therapy in Patients with Newly Diagnosed Multiple Myeloma (NDMM): A Real-World Experience at Two Institutes, Clinical Lymphoma Myeloma and Leukemia, 2025, ISSN 2152-2650, https://doi.org/10.1016/j.clml.2025.04.003. April 11, 2025.
Overview
For people newly diagnosed with multiple myeloma (NDMM) who are eligible for a stem cell transplant, treatment often starts with a combination of four medicines. This includes Darzaelex® (daratumumab), Revlimid® (lenalidomide), Velcade® (bortezomib), and dexamethasone, known together as DRVd.
Some people worry that using this powerful treatment might make it harder to collect stem cells for a future transplant. But this new study shows that stem cell collection still works very well, even outside of clinical trials.
Researchers at two major cancer centers looked at 423 myeloma patients who received DRVd and went on to collect stem cells for a possible transplant. They checked:
- How many stem cells were collected
- How often extra help was needed to collect them (like extra medicine)
- How many people had trouble collecting enough
Researchers learned the following:
- 94% of patients collected enough stem cells for two transplants.
- Only 1 person out of 423 had a complete failure to collect stem cells.
- There was no major difference between patients who had different treatment schedules or who received slightly different stem cell boosting strategies.
- The average number of stem cells collected was even better than in some earlier clinical tria
The study shows that adding daratumumab to the usual drug combination does not harm the body’s ability to collect stem cells for transplant. Most people in this study had excellent results, showing that this treatment approach is safe and effective even in everyday care settings.
"Daratumumab for newly diagnosed multiple myeloma: pooled analysis of patients aged ≥65 years from GRIFFIN and PERSEUS"
Source
Paula Rodriguez-Otero, Peter M. Voorhees, Mario Boccadoro, Jacob Laubach, Hermann Einsele, Douglas W. Sborov, Meletios A. Dimopoulos, Annemiek Broijl, Roberto Mina, Andrew Spencer, Fredrik Schjesvold, Rebecca Silbermann, Francesca Gay, Luciano J. Costa, Aurore Perrot, Yanfang Liu, Jianping Wang, Anna Sitthi-Amorn, Robin Carson, Annelore Cortoos, Saad Z. Usmani, Paul G. Richardson, Philippe Moreau, Pieter Sonneveld, Jonathan L. Kaufman, Daratumumab for newly diagnosed multiple myeloma: pooled analysis of patients aged ≥65 years from GRIFFIN and PERSEUS, Clinical Lymphoma Myeloma and Leukemia, 2025, ISSN 2152-2650, https://doi.org/10.1016/j.clml.2025.04.007. April 11, 2025.
Overview
For older adults with newly diagnosed multiple myeloma (NDMM), finding the right treatment is critical. Research from the GRIFFIN and PERSEUS studies shows that adding the targeted therapy Darazalex® (daratumumab) to the standard treatment combination of Velcade® (bortezomib), Revlimid® (lenalidomide), and dexamethasone (VRd) can improve treatment outcomes, even for patients aged 65 and older.
The trials found the following:
- Progression-free survival (PFS), which means the time patients go without their disease getting worse, showed a trend towards improvement when daratumumab was added to VRd in patients aged 65 and older.
- Complete response (when no signs of myeloma are found) was seen in 82.8% of patients who received D-VRd, compared to 67.0% with VRd alone.
- Minimal residual disease negativity (meaning almost no detectable disease) was achieved by 66.4% of patients on D-VRd, compared to 41.7% with VRd.
These results suggest that older patients benefit from the addition of daratumumab to their treatment, achieving deeper responses and better outcomes.
Also, the safety profile of adding Daratumumab was similar to standard VRd therapy, with no new safety concerns noted.
For older adults with multiple myeloma who are eligible for a stem cell transplant, adding daratumumab to VRd is a promising treatment approach. This combination deepens the response to treatment and can be considered as a standard option for patients up to age 70.
"Cardiovascular adverse events and outcomes after anti-BCMA CAR-T for relapsed and refractory multiple myeloma"
Source
Palfi, S., Peres, L.C., Koelmeyer, H. et al. Cardiovascular adverse events and outcomes after anti-BCMA CAR-T for relapsed and refractory multiple myeloma. Blood Cancer J. 15, 63 (2025). https://doi.org/10.1038/s41408-025-01261-5 April 14, 2025.
Overview
For patients with relapsed or refractory multiple myeloma (RRMM), CAR-T therapies like Abecma® (idecabtagene vicleucel, or ide-cel) and Carvykti® (ciltacabtagene autoleucel, or cilta-cel) have shown promising results. However, these therapies can also cause serious side effects, including heart-related issues.
Heart problems, or cardiovascular adverse events (CVAEs), are common with CAR-T treatments. These can include arrhythmias (irregular heartbeats), heart failure, and even cardiovascular death. In one study of 164 patients treated with ide-cel or cilta-cel, 12.2% developed CVAEs, with the most common being atrial arrhythmias (irregular heartbeats in the upper chambers of the heart).
Patients with more advanced stages of myeloma were more likely to experience heart problems. Those with higher levels of certain blood markers, like ferritin and C-reactive protein (CRP), also had a higher risk. Interestingly, patients' previous heart conditions didn't seem to be a major factor in the development of CVAEs.
A key finding in the study was that patients who developed severe cytokine release syndrome (CRS), a common side effect of CAR-T therapy, were more likely to experience heart issues. CRS can cause inflammation in the body, which may affect the heart. The study showed that high-grade CRS was a strong risk factor for CVAEs.
While heart problems were linked to early deaths in some patients, they weren't the main cause of death. Most early deaths were due to CRS or infections, not directly from heart issues. After adjusting for CRS severity, heart problems didn't seem to have a major impact on long-term survival.
The study suggests that better management of CRS and careful cardiovascular monitoring can help reduce the risks of heart-related issues. It's also important to personalize treatment based on a patient's condition to minimize these risks.
This research emphasizes the need for careful monitoring and support during CAR-T therapy to improve patient outcomes, particularly for those who develop severe side effects like CRS.
"Cytogenetic risk stratification combined with minimal residual disease status influences the therapeutic outcome and prognosis of multiple myelomas"
Source
Mao, J., Xue, L., Wang, H. et al. Cytogenetic risk stratification combined with minimal residual disease status influences the therapeutic outcome and prognosis of multiple myelomas. Sci Rep 15, 12545 (2025). https://doi.org/10.1038/s41598-025-97125-w April 12, 2025.
Overview
In multiple myeloma (MM), doctors can use a combination of genetic testing and minimal residual disease (MRD) status to help predict how well treatment will work and what the future may hold for patients. MRD refers to the small number of cancer cells that may remain in a patient’s body after treatment, even if the patient is in remission. The presence or absence of MRD can tell doctors how likely the cancer is to return.
This study looked at 73 newly diagnosed MM patients between 2017 and 2023. Researchers checked their genetic risks (using a test called FISH) and MRD status, then followed up with them for an average of 30 months.
The study found the following:
- Progression-Free Survival (PFS): On average, patients who were MRD-negative (no detectable cancer cells) lived longer without their cancer worsening (45 months) compared to MRD-positive patients (30 months).
- Overall Survival (OS): MRD-negative patients also lived longer overall (59 months) compared to MRD-positive patients (34 months).
When combining MRD status and genetic risk (high risk vs. standard risk), the study found:
- Patients with high-risk genetics and MRD-positive status had the worst outcomes, with a median PFS of just 20 months and OS of 33 months.
- Those with standard-risk genetics and MRD-negative status did the best, with no median PFS reached and an OS of 59 months.
The study suggests that MRD status is a strong predictor of how well treatment will work and how long patients will live after treatment. However, genetic risk plays an even more important role. Combining both factors can help doctors create more personalized treatment plans to improve outcomes for MM patients. This information emphasizes how MRD and genetic testing are important tools in determining the best treatment and prognosis for multiple myeloma patients.
"LncRNA HCG11 regulates selinexor sensitivity in multiple myeloma"
Source
Yue Zhang, Xin Wang, Hongmei Luo, Xiang Liu, Jingcao Huang, Ziyue Mi, Siyao He, Jingjing Wen, Qianwen Gao, Haonan Yang, Yu Feng, Linfeng Li, Xinyu Zhai, Fangfang Wang, Yang Dai, Li Zhang, Ting Niu, Chunyan Sun, Yuhuan Zheng, LncRNA HCG11 regulates selinexor sensitivity in multiple myeloma, Biochemical Pharmacology, 2025, 116948, ISSN 0006-2952, https://doi.org/10.1016/j.bcp.2025.116948. April 12, 2025.
Overview
One treatment option for multiple myeloma (MM) is Xpovio (selinexor), a drug that targets a protein called XPO1, which helps control how substances move in and out of cells.
However, not all MM patients respond well to selinexor, and scientists are working to understand why. In this study, researchers discovered that a long noncoding RNA (lncRNA) called HCG11 plays an important role in how MM cells grow and respond to selinexor. They found that:
- HCG11 is found in both MM cell lines and MM patient samples.
- High levels of HCG11 in MM cells were linked to poorer survival for patients.
- When HCG11 was reduced in MM cells, these cells became more sensitive to selinexor, meaning they were more likely to die off after treatment.
HCG11 affects selinexor sensitivity in these ways:
- HCG11 helps XPO1 stay stable in the cell by binding to its mRNA (a type of genetic material). This leads to more XPO1 being produced.
- When HCG11 was removed, XPO1 levels dropped, and the MM cells became more sensitive to the effects of selinexor.
- In an animal model, MM cells with reduced HCG11 were more sensitive to selinexor, showing that HCG11 plays a role in how well the drug works.
This research suggests that HCG11 might influence how well selinexor works in treating MM. Understanding this could help doctors identify which patients may benefit most from selinexor and develop new treatment strategies in the future.
"Anti-GPRC5D CAR T-cell therapy as a salvage treatment in patients with progressive multiple myeloma after anti-BCMA CAR T-cell therapy: a single-centre, single-arm, phase 2 trial"
Source
Jieyun Xia, Qian Sun, Dian Zhou, Hujun Li, Ying Wang, Yuekun Qi, Jiang Cao, Zhiling Yan, Depeng Li, Hai Cheng, Wei Sang, Feng Zhu, Haiying Sun, Wei Chen, Kunming Qi, Dongmei Yan, Tingting Qiu, Tingyu Hu, Weiying Gu, Jun Qian, Fan Xia, Na Qi, Congqian Jin, Yang Liu, Xue Wang, Yanlei Zhang, Shuixiu Peng, Zhenyu Li, Alex H Chang, Kailin Xu, Anti-GPRC5D CAR T-cell therapy as a salvage treatment in patients with progressive multiple myeloma after anti-BCMA CAR T-cell therapy: a single-centre, single-arm, phase 2 trial, The Lancet Haematology, 2025, ISSN 2352-3026, https://doi.org/10.1016/S2352-3026(25)00048-1. April 12, 2025.
Overview
For patients with multiple myeloma who no longer respond to anti-BCMA CAR T-cell therapy, finding effective treatment options is challenging. A new study tested a different type of CAR T-cell therapy, targeting GPRC5D, as a possible treatment for these patients.
This phase II clinical trial involved patients whose multiple myeloma had returned after they received anti-BCMA CAR T-cell therapy. The study looked at how well anti-GPRC5D CAR T-cells worked and how safe they were.
A total of 37 patients were treated with a single dose of anti-GPRC5D CAR T-cells, and the goal was to see how many patients had a positive response, including complete or partial remission.
The study found the following:
- 84% of patients responded to the treatment, with 35% showing a complete response or better.
- The most common side effects were related to the blood, including low white blood cell counts and anemia.
- 70% of patients had cytokine release syndrome (CRS), a common side effect of CAR T-cell therapy, but it was manageable. Only a few patients experienced more severe side effects like neurotoxicity.
- There were no treatment-related deaths during the study.
Anti-GPRC5D CAR T-cell therapy showed promising results in treating multiple myeloma patients who had progressed after anti-BCMA therapy. While the treatment is effective and generally safe, more research is needed to fully understand the long-term benefits and risks of this new approach.
This study suggests that anti-GPRC5D CAR T-cell therapy might be a helpful option for patients who have run out of treatment choices after anti-BCMA CAR T-cell therapy.
"Sequential CAR T-cell therapy in myeloma: going from BCMA to GPRC5D"
Source
Niels W C J van de Donk, Carlos Fernandez de Larrea, Sequential CAR T-cell therapy in myeloma: going from BCMA to GPRC5D, The Lancet Haematology, 2025, ISSN 2352-3026 https://doi.org/10.1016/S2352-3026(25)00074-2. April 12, 2025.
Overview
For patients with multiple myeloma who have already received multiple types of treatments, including BCMA-directed therapies (such as CAR T-cells, bispecific antibodies, and antibody-drug conjugates), these treatments have greatly improved survival. However, patients whose disease comes back after using these treatments, and who are also resistant to other therapies like immunomodulatory drugs, proteasome inhibitors, and CD38-targeting antibodies, tend to have poor outcomes.
Recently, there is growing evidence that using a second type of T-cell therapy that targets a different protein may offer a new treatment option and significantly improve clinical results for these patients. This approach could provide hope for those with relapsed disease who have limited treatment options.
"Efficacy of maintenance or consolidation therapy for multiple myeloma based on proteasome inhibitors: a meta-analysis"
Source
Wu, D., Wang, F., Yang, X. et al. Efficacy of maintenance or consolidation therapy for multiple myeloma based on proteasome inhibitors: a meta-analysis. Discov Onc 16, 519 (2025). https://doi.org/10.1007/s12672-025-02304-w April 12, 2025.
Overview
Proteasome inhibitors (PIs) have been studied for their effectiveness in maintenance and consolidation therapies for multiple myeloma (MM). Researchers analyzed data from 10 clinical trials with a total of 4,711 patients to assess how well these treatments work.
The findings showed that PI-based therapies significantly improved progression-free survival (PFS) and overall survival (OS) compared to standard treatments. For consolidation therapy, PIs were especially effective in extending PFS. In maintenance therapy, PIs also improved PFS and OS when compared to observation (no treatment) groups. Additionally, PIs helped more patients achieve a complete response (CR) and very good partial response (≥ VGPR) than other treatments.
However, while these therapies improved outcomes, they were linked to a higher risk of serious side effects (grade 3 adverse events). This suggests that while PI-based therapies can be effective, they may come with some additional risks
"Lipin1-dependent transcriptional inactivation of SREBPs contributes to selinexor sensitivity in multiple myeloma"
Source
Wang, Jy., Chen, Mp., Jiang, Jx. et al. Lipin1-dependent transcriptional inactivation of SREBPs contributes to selinexor sensitivity in multiple myeloma. Acta Pharmacol Sin (2025). https://doi.org/10.1038/s41401-025-01553-3 April 14, 2025.
Overview
Xpovio® (selinexor) is a treatment used for people with relapsed multiple myeloma. While it’s been shown to work for some patients, scientists are still figuring out exactly how it works—and why some people respond better than others.
In this study, researchers looked closely at how SEL affects myeloma cells, focusing on changes inside the cell’s nucleus (the cell’s control center). They discovered that selinexor blocks a protein called XPO1, which usually moves certain molecules out of the nucleus. When XPO1 is blocked, a molecule called Lipin1 gets stuck inside the nucleus instead of being moved out.
Once inside the nucleus, Lipin1 acts as a brake on another set of molecules called SREBPs, which are important for making fats and cholesterol. By stopping SREBPs, selinexor reduces the production of fats that myeloma cells need to grow and survive.
The researchers found that myeloma cells with high levels of Lipin1 were more sensitive to selinexor. This suggests that Lipin1 might help predict who will respond best to selinexor treatment.
This study reveals a new way that selinexor fights myeloma—by blocking fat production in cancer cells—and shows that Lipin1 might be used as a biomarker to identify patients who could benefit most from this treatment.
"Developments in the treatment of multiple myeloma: overview and nursing implications"
Source
Toland S. Developments in the treatment of multiple myeloma: overview and nursing implications. Nurs Stand. 2025 Apr 14. doi: 10.7748/ns.2025.e12403. Epub ahead of print. April 14, 2025.
Overview
Multiple myeloma begins in the bone marrow. In the past, treatment options were very limited, mainly relying on stem cell transplants. But in the last 20 years, major advances in treatment have greatly improved survival for people with myeloma.
Today, many people with multiple myeloma live much longer. Instead of being quickly life-threatening, it has become more of a relapsing and remitting disease, meaning patients go through periods of remission (when the cancer is under control) and relapse (when it comes back).
However, longer life comes with new challenges. Patients often go through long and complex treatments and may deal with lasting side effects from both the disease and the treatments. These can seriously affect their quality of life, even when the cancer is not active.
Nurses play a key role in caring for people with multiple myeloma. They help monitor symptoms, manage side effects, and provide support throughout the treatment journey.
While people with multiple myeloma are living longer than ever before, they also face new challenges. Ongoing support—especially from nurses—is essential to help manage the impact of both the disease and its treatments.
"Real-world evidence supports venetoclax as an additional option for patients with t (11;14) myeloma"
Source
Jules Higué, Gaetan Basile, Titouan Cazaubiel, Julie Gay, Guillaume Béziat, Martin Gauthier, Miguel Carreiro, Noémie Gadaud, François Lifermann, Reza Tabrizi, Benjamin Hébraud, Miguel Granell, Leopoldine Lapierre, Pierre-Luc Mouchel, Pierre Bories, Jill Corre, Cyrille Hulin, Aurore Perrot, Real-world evidence supports venetoclax as an additional option for patients with t (11;14) myeloma. Clinical Lymphoma Myeloma and Leukemia, 2025, ISSN 2152-2650, https://doi.org/10.1016/j.clml.2025.04.005. April 14, 2025.
Overview
Even with new treatments, some people with multiple myeloma see their cancer return or stop responding to therapy. This is especially true for patients with a specific genetic feature called t(11;14).
Researchers looked back at the cases of 46 people with this type of multiple myeloma who were treated with venetoclax, a drug that targets a protein cancer cells need to survive. Most of these patients had already stopped responding to two common treatments—lenalidomide and daratumumab.
On average, their cancer stayed under control for 16 months, and they lived about 26 months after starting treatment. Side effects were manageable, but some patients developed serious infections, showing the importance of preventing these early.
These results suggest that venetoclax could be a helpful option for people with t(11;14) multiple myeloma, but more research is needed to confirm how well it works.
"Real-world treatment patterns, outcomes, and economic costs by lines of therapy in patients with newly diagnosed multiple myeloma: a nationwide population-based cohort study in South Korea"
Source
Park, SS., Park, Y., Yoon, S. et al. Real-world treatment patterns, outcomes, and economic costs by lines of therapy in patients with newly diagnosed multiple myeloma: a nationwide population-based cohort study in South Korea. Blood Res. 60, 26 (2025). https://doi.org/10.1007/s44313-025-00069-3 April 15, 2025.
Overview
Even with new treatments, some people with multiple myeloma see their cancer return or stop responding to therapy. This is especially true for patients with a specific genetic feature called t(11;14).
Researchers looked back at the cases of 46 people with this type of multiple myeloma who were treated with venetoclax, a drug that targets a protein cancer cells need to survive. Most of these patients had already stopped responding to two common treatments—lenalidomide and daratumumab.
On average, their cancer stayed under control for 16 months, and they lived about 26 months after starting treatment. Side effects were manageable, but some patients developed serious infections, showing the importance of preventing these early.
These results suggest that venetoclax could be a helpful option for people with t(11;14) multiple myeloma, but more research is needed to confirm how well it works.
"Deciphering N‑Glycosylation Dynamics of Serum Monoclonal Immunoglobulins in Multiple Myeloma via EThcD-sceHCD-MS/MS"
Source
Huixian Li, Wanhong Lu, Qian Jin, Jiping Sun, Li Gao, Juanjuan Hu, Yingying Ling, Wenyu Zhao, Yong Zhang, Xinfang Xie, Deciphering N‑Glycosylation Dynamics of Serum Monoclonal Immunoglobulins in Multiple Myeloma via EThcD-sceHCD-MS/MS, Journal of Proteome Research, 2025, ISSN 1535-3907, https://doi.org/10.1021/acs.jproteome.5c00253. April 15, 2025.
Overview
In multiple myeloma (MM), abnormal proteins called M-proteins build up in the blood. These proteins often have sugar molecules attached to them—a process called glycosylation. Changes in these sugar patterns may give clues about how the disease is behaving.
In this study, researchers compared the sugar patterns on M-proteins from MM patients with those from healthy people. They looked at two types of MM—IgA-MM and IgG-MM—and found that M-proteins from MM patients had more of certain sugars, including fucose, sialic acid, and mannose, than the proteins from healthy people.
They also found that some specific sugar patterns were linked to important signs of disease, like low albumin levels, kidney function, and how well a patient might respond to treatment.
These results suggest that measuring sugar changes on M-proteins could help doctors better understand and manage multiple myeloma in the future.
"Bortezomib, Mitoxantrone Hydrochloride Liposome, and Dexamethasone for Relapsed/Refractory Multiple Myeloma: A Multi-Center, Open-Label Phase I Trial"
Source
Zhou, Y.-L., Zhang, J.-Q., Wang, W., Bao, L., Fang, B.-J., Gao, D., Su, L.-P., Chen, W.-M. and Yang, G.-Z. (2025), Bortezomib, Mitoxantrone Hydrochloride Liposome, and Dexamethasone for Relapsed/Refractory Multiple Myeloma: A Multi-Center, Open-Label Phase I Trial. Cancer Med, 14: e70890. https://doi.org/10.1002/cam4.70890 April 15, 2025.
Overview
A new combination treatment using liposomal mitoxantrone (Lipo-MIT) with Velcade (bortezomib) and dexamethasone—called VMitD—was tested in people with relapsed/refractory multiple myeloma.
In this early-phase study, 20 patients tried different doses of Lipo-MIT alongside the two other drugs. The main goal was to see how safe the treatment was and how well it worked.
The most serious side effect was pneumonia, seen in 20% of patients. Blood-related side effects were more common, with 70% experiencing low platelets, 55% with low white cells (neutropenia), and some with anemia or low lymphocyte levels.
Despite these side effects, the treatment showed encouraging results. Out of 15 people who were checked for response, 9 had a very good partial response or better, and 13 responded overall—an 86.7% response rate.
This study suggests the VMitD combo could be a promising option for patients with relapsed or refractory multiple myeloma. More research is needed to compare it with other treatment options.
"Enhancing risk stratification and treatment decision in multiple myeloma with SKY92 gene expression profiling in real-world data"
Source
Biran N, Dhakal B, Niesvizky R, Lentzsch S, Bhutani D, McKay JT, et al. Enhancing risk stratification and treatment decision in multiple myeloma with SKY92 gene expression profiling in real-world data. Br J Haematol. 2025; 00: 1–12. https://doi.org/10.1111/bjh.20050 April 15, 2025.
Overview
Doctors use risk markers to help decide how aggressive a patient’s multiple myeloma (MM) is and what treatment may work best. But these traditional markers can sometimes give mixed results. A newer test called SKY92, which looks at gene activity in myeloma cells, may offer a clearer picture.
In a recent study of 251 newly diagnosed MM patients, researchers compared doctors’ risk assessments with and without the SKY92 test. Before seeing the SKY92 results, doctors classified 51% of patients as high-risk. But SKY92 identified only 28% as truly high-risk. This difference showed that the test could more accurately identify which patients are at higher risk of their cancer progressing.
Once doctors saw the SKY92 results, they said they would change their treatment plans for half of the patients and felt more confident in their choices for 40% of them.
The study shows that SKY92 may help doctors better understand each patient’s risk and make more personalized treatment decisions.
"Enhancing risk stratification and treatment decision in multiple myeloma with SKY92 gene expression profiling in real-world data"
Source
Biran N, Dhakal B, Niesvizky R, Lentzsch S, Bhutani D, McKay JT, et al. Enhancing risk stratification and treatment decision in multiple myeloma with SKY92 gene expression profiling in real-world data. Br J Haematol. 2025; 00: 1–12. https://doi.org/10.1111/bjh.20050 April 15, 2025.
Overview
Doctors use risk markers to help decide how aggressive a patient’s multiple myeloma (MM) is and what treatment may work best. But these traditional markers can sometimes give mixed results. A newer test called SKY92, which looks at gene activity in myeloma cells, may offer a clearer picture.
In a recent study of 251 newly diagnosed MM patients, researchers compared doctors’ risk assessments with and without the SKY92 test. Before seeing the SKY92 results, doctors classified 51% of patients as high-risk. But SKY92 identified only 28% as truly high-risk. This difference showed that the test could more accurately identify which patients are at higher risk of their cancer progressing.
Once doctors saw the SKY92 results, they said they would change their treatment plans for half of the patients and felt more confident in their choices for 40% of them.
The study shows that SKY92 may help doctors better understand each patient’s risk and make more personalized treatment decisions.
"Distress and symptom burden in patients with monoclonal gammopathy of undetermined significance and smoldering myeloma"
Source
Elizabeth K. O’Donnell, Jennifer E. Carroll, Jaqueline Perry, Katherine Padgett, Benjamin Harris, Vidhi Patel, Omar Nadeem, Areej El-Jawahri, Irene M. Ghobrial, Catherine R. Marinac; Distress and symptom burden in patients with monoclonal gammopathy of undetermined significance and smoldering myeloma. Blood Adv 2025; 9 (8): 1984–1987. doi: https://doi.org/10.1182/bloodadvances.2024015575 April 22, 2025.
Overview
Many people are diagnosed with conditions like MGUS (monoclonal gammopathy of undetermined significance) or smoldering multiple myeloma (SMM) before developing multiple myeloma. These early conditions don’t cause symptoms and are often described as “low-risk” or “benign.” But a new study shows that patients living with MGUS or SMM often experience real emotional and physical challenges, long before they are diagnosed with full-blown multiple myeloma.
Researchers surveyed 467 adults with MGUS or SMM and found that:
- 27% had signs of anxiety
- 14% showed signs of depression
- 24% had symptoms of post-traumatic stress (PTSD)
- 31% reported moderate to severe symptoms such as tiredness, pain, or poor well-being
- 21% experienced moderate to severe fatigue
These symptoms were just as common in people with MGUS as in those with SMM—even though SMM carries a higher risk of progressing to myeloma. Patients reported feeling tired, unwell, or drowsy, though it’s unclear if these were caused by MGUS or SMM directly. The emotional toll of "watching and waiting" for possible progression to cancer may be one of the biggest burdens.
Some groups reported higher distress levels:
- Women were more likely to feel anxious.
- People with lower incomes had higher levels of depression and fatigue.
- Older adults reported less anxiety and distress, but more fatigue.
This study suggests that even without physical symptoms, the emotional burden of having a “precancer” condition is real and significant. Patients may benefit from more support—both emotional and physical—while living with MGUS or SMM. Future research and treatment plans should focus not just on preventing progression, but also on improving quality of life for patients at every stage.
"Dynamics of Mutant Hematopoietic Progenitor Cells Are Not Associated with Clinical Outcomes in Multiple Myeloma"
Source
Camila Guerrero, Maria-Jose Larrayoz, Irache Erdozain, Noemi Puig, Maria-Teresa Cedena, Catarina Maia, Amagoia Mañu, Natalia Gordillo, Oihane Churruca, Diego Alignani, Sarai Sarvide, Iria Vazquez, Cristina Perez, Albert Oriol, Laura Rosinol, Rafael Ríos-Tamayo, Marta-Sonia Gonzalez-Perez, Ana-Pilar Gonzalez-Rodriguez, Felipe de Arriba, Jose M. Moraleda, Jesus Martin, Luis Palomera, Valentin Cabañas, Maria-Jose Calasanz, Joaquin Martinez-Lopez, Maria-Victoria Mateos, Joan Bladé, Juan-Jose Lahuerta, Jesus F. San-Miguel, Bruno Paiva; on behalf of the Programa Para el Estudio de la Terapéutica en Hemopatías Malignas/Grupo Español de Mieloma (PETHEMA/GEM) cooperative group, Dynamics of Mutant Hematopoietic Progenitor Cells Are Not Associated with Clinical Outcomes in Multiple Myeloma. Blood Cancer Discov 2025; https://doi.org/10.1158/2643-3230.BCD-24-0274 April 15, 2025.
Overview
Some treatments for multiple myeloma—like high-dose melphalan and Revlimid® (lenalidomide)—can increase the risk of developing a second blood cancer (called a second primary malignancy, or SPM). Scientists wanted to know if testing for certain mutations in blood-forming stem cells (called hematopoietic progenitor cells, or HPCs) might help predict this risk.
In a recent study, researchers looked at samples from 348 multiple myeloma patients treated with lenalidomide. They found that:
- About 1 in 4 patients (23%) already had mutated HPCs at diagnosis.
- These mutations were linked to higher tumor burden and changes in how blood cells were made, but not to worse survival or more SPMs.
- Some mutations—like TP53—became more common during maintenance treatment, rising from 1% to 7%.
While this suggests we should keep an eye on certain mutations during treatment, the study did not find enough evidence to support routine genetic testing of HPCs in myeloma patients right now. More research is needed to determine if monitoring these mutations could help prevent second cancers in the future.
"Treatment pathways and clinical outcomes in newly diagnosed multiple myeloma outside Europe and North America: The INTEGRATE study"
Source
Kim, K., Verburgh, E., Mitina, T. et al. Treatment pathways and clinical outcomes in newly diagnosed multiple myeloma outside Europe and North America: The INTEGRATE study. Int J Hematol (2025). https://doi.org/10.1007/s12185-025-03972-8 April 15, 2025.
Overview
This study looked at how people with multiple myeloma (MM) were treated in eight countries outside of Europe and North America, including Argentina, China, South Korea, and others. Researchers reviewed the medical records of over 1,500 adults who were newly diagnosed with MM between 2010 and 2011.
Key findings:
- Most patients were around 59 years old.
- One-third had an advanced stage of MM.
- The most common first treatments included chemotherapy with drugs like bortezomib and thalidomide. Lenalidomide was used more often in later treatments.
- The average time before needing the next treatment was about 3 years and 3 months.
- Only about one-third of patients had a stem cell transplant (SCT) at diagnosis. Those who did lived longer and stayed in remission longer than those who didn’t.
Teatment in these countries mostly followed current medical guidelines. But more access to stronger treatments and earlier use of stem cell transplants could help improve outcomes for patients.
"Outpatient Management of Bispecific Related Toxicities: An Observational Study of Safety Outcomes and Resource Utilization"
Source
Puttkammer JR, Barreto JN, Jensen CJ, Nedved AN, Wilson-Miller JL, Cole KC, Holmes LM, Kosobud AR, Kapoor P, Gertz MA, Dingli DM, Gonsalves WI, Kumar SK, Hayman SR, Kourelis TV, Warsame R, Binder M, Cook J, Lin Y, Sandahl TB. Outpatient Management of Bispecific Related Toxicities: An Observational Study of Safety Outcomes and Resource Utilization. JCO Oncol Pract. 2025 Apr 15:OP2400930. doi: 10.1200/OP-24-00930. Epub ahead of print. April 15, 2025.
Overview
Some newer treatments for multiple myeloma, called bispecific antibodies (BsAbs), can cause a side effect known as cytokine release syndrome (CRS). CRS is common but usually mild. Serious cases are rare. Because of this, doctors are looking at ways to safely give these treatments without needing to admit patients to the hospital.
This study followed 34 people with multiple myeloma who received BsAb treatments at an outpatient clinic between August 2023 and March 2024. Half received teclistamab, and half got talequetamab. Nearly half of the patients stayed outpatient the entire time. While some patients did develop CRS, none of them needed to be hospitalized. Most cases were mild (grade 1 or 2), and all were safely managed with medications like steroids and tocilizumab—even at home when needed.
The study also showed that giving these treatments outside the hospital could save a lot of money—over $100,000 in medication-related costs.
Giving bispecific antibody treatments in outpatient clinics appears to be safe, even if mild side effects occur, and it may also help lower costs. More research is needed to predict which patients are at higher risk for side effects so care can be personalized.
"Cost-Effectiveness Analysis of Daratumumab Monotherapy and Subsequent Therapies in Heavily Treated Relapsed/Refractory Multiple Myeloma: A Feasible Methodology using a Korean Nationwide Population Cohort"
Source
Park SS, Choi S, Jung S, Han S, Lee C, Han J, Kim S, Kim K, Min CK. Cost-Effectiveness Analysis of Daratumumab Monotherapy and Subsequent Therapies in Heavily Treated Relapsed/Refractory Multiple Myeloma: A Feasible Methodology using a Korean Nationwide Population Cohort. Cancer Res Treat. 2025 Apr 15. doi: 10.4143/crt.2025.046. Epub ahead of print.
Overview
As new, more expensive treatments for multiple myeloma become available, it’s important to understand both how well they work and how much they cost.
This study looked at medical data from over 11,000 people in one country who were newly diagnosed with multiple myeloma between 2010 and 2019. The number of new patients increased over time. Researchers used a special method to track how patients moved through different lines of therapy (LoT)—meaning different treatment stages as the disease progressed.
They found that:
- As patients moved to later treatments, their time until needing the next treatment and overall survival both got shorter.
- Medical costs went up with each new treatment stage.
- Daratumumab, a newer drug, helped patients live longer and delayed the need for another treatment compared to standard options—even when taking cost into account.
The team also created a formula to help predict how much future treatments might cost, based on how much extra life they’re expected to give.
This study offers a way to evaluate new myeloma treatments by comparing how well they work and how much they cost. It can help doctors, health systems, and policymakers make smarter decisions about care.
"Treatment patterns, efficacy, and tolerability of belantamab mafodotin in patients with relapsed and/or refractory multiple myeloma: a real-world analysis"
Source
Mihir N. Patel, Susan C. Locke, Caroline Falvey, Jesse D. Troy, Kris W. Herring, Amanda Bolgioni-Smith, Cherie Elcock, Laura Iadeluca, Cristiana Costa Chase, Cristina Gasparetto, Mark S. Newman, Thomas W. LeBlanc, Treatment patterns, efficacy, and tolerability of belantamab mafodotin in patients with relapsed and/or refractory multiple myeloma: a real-world analysis, Clinical Lymphoma Myeloma and Leukemia,2025, ISSN 2152-2650, https://doi.org/10.1016/j.clml.2025.04.008. April 15, 2025.
Overview
Belantamab mafodotin is a treatment for people with relapsed or refractory multiple myeloma (RRMM)—meaning their cancer came back or didn’t respond to other treatments. This study looked at how well it worked and what side effects it caused in 30 adults treated at Duke Cancer Institute between 2020 and 2022.
Here’s what they found:
- Most patients had already gone through 4 or more other treatments, and nearly 90% had disease that didn’t respond to multiple standard drugs.
- About 2 out of 3 patients (67%) responded to belantamab mafodotin.
- On average, the cancer did not get worse for about 9.5 months.
- Vision-related side effects were common: 93% had some level of eye problems, and 90% had changes in vision. However, only a small number had serious (grade 3 or higher) eye issues.
- Eye problems caused many patients to delay or lower their dose. About 13% stopped treatment because of it.
Belantamab mafodotin can help patients with hard-to-treat multiple myeloma, but eye side effects are common and often require adjusting or delaying treatment. Most side effects were manageable, and many patients saw benefits from the drug.
"Impact of blood pressure variability on the prediction of cancer therapy-related cardiovascular toxicity in patients with multiple myeloma"
Source
Morimoto, S., Ichihara, A. Impact of blood pressure variability on the prediction of cancer therapy-related cardiovascular toxicity in patients with multiple myeloma. Hypertens Res (2025). https://doi.org/10.1038/s41440-025-02215-x April 16, 2025.
Overview
Some treatments for multiple myeloma—like proteasome inhibitors—can affect the heart and blood vessels. These side effects, called cancer therapy-related cardiovascular toxicity (CTR-CVT), can make treatment harder and increase health risks.
This study looked at a simple way to help spot patients at higher risk: tracking changes in blood pressure over time, known as blood pressure variability (BPV). Using a tool called ambulatory blood pressure monitoring (ABPM), researchers measured patients’ BP—especially at night—and found that those with more ups and downs in their blood pressure were more likely to develop heart-related side effects during treatment.
Key points of this study include the following:
- Patients with high nighttime BPV had more heart problems, even if they didn’t have other heart risk factors like high cholesterol or diabetes.
- ABPM is widely available and easy to use, making it a good option for monitoring.
- Catching BPV changes early could lead to better heart care during cancer treatment, like adjusting medications or adding cardiology follow-ups.
Watching how your blood pressure changes—especially at night—may help your care team spot risks before heart problems happen. More research is needed, but this could become part of routine care for multiple myeloma patients getting treatments that can affect the heart.
"Impact of blood pressure variability on the prediction of cancer therapy-related cardiovascular toxicity in patients with multiple myeloma"
Source
Morimoto, S., Ichihara, A. Impact of blood pressure variability on the prediction of cancer therapy-related cardiovascular toxicity in patients with multiple myeloma. Hypertens Res (2025). https://doi.org/10.1038/s41440-025-02215-x April 16, 2025.
Overview
Multiple myeloma can be difficult to treat, even with new medications. This study looked at a specific protein in myeloma cells called HDAC6, which helps cancer cells survive and resist treatment. HDAC6 does more than just one job—it also helps get rid of damaged proteins inside the cell through a process called autophagy.
Researchers focused on a part of HDAC6 called the ZnF-UBP binding domain. This part helps the protein recognize and bind to broken proteins so they can be destroyed. Scientists created a version of HDAC6 with a small change in this domain, which stopped it from working properly. They found that cancer cells with the non-working version of this domain:
- Grew more slowly
- Struggled to clear out damaged proteins
- Showed unusual gene activity
- These effects were even stronger than in cells where HDAC6 was completely removed.
- Based on this, the researchers designed and tested new drug-like molecules that block this part of HDAC6. The goal is to develop a new treatment that targets this weak spot in multiple myeloma cells.
Blocking a specific part of the HDAC6 protein may be a promising new way to treat multiple myeloma, especially in cases that don’t respond well to current therapies.
"Cure for multiple myeloma?"
Source
Podar, K. Cure for multiple myeloma?. memo (2025). https://doi.org/10.1007/s12254-025-01030-x April 16, 2025.
Overview
Thanks to major advances in treatment, people with multiple myeloma are living longer than ever before. But many are now asking: Can multiple myeloma actually be cured? New research shows that for some patients, the answer may be yes—or at least, close to it.
What Does “Cure” Mean?
Doctors talk about three types of cures:
- True cure: The cancer is completely gone, doesn’t come back, and the patient lives a normal life without side effects.
- Functional cure: The cancer is still there in tiny amounts but doesn’t grow or cause problems.
- Relative cure: The cancer is gone, but side effects from treatment affect quality of life.
In multiple myeloma, some patients—around 30%—are still doing well more than 8 years after treatment, with no signs that the disease is getting worse. That’s a big deal.
What’s Helping Myeloma Patients Live Longer?
- Stronger combination therapies using 3 or 4 drugs at once
- Stem cell transplants followed by more treatment
- Newer therapies like CAR T-cell therapy and bispecific antibodies
- Personalized medicine based on the biology of your myeloma
- People treated with two or more modern drugs upfront are now living an average of 12 years or longer. Some are living 20 years or more.
The Power of MRD Testing
MRD stands for minimal residual disease, or the tiny number of myeloma cells that may still be in the body after treatment. If doctors can’t find any MRD using highly sensitive tests (like next-generation sequencing or flow cytometry), that’s a very good sign. Even better? Staying MRD-negative for a long time. That’s now one of the best predictors of long-term survival.
The FDA recently approved MRD testing as a way to fast-track new myeloma treatments.
What’s Next in the Multiple Myeloma Landscape?
- Liquid biopsies (blood tests that detect cancer cells or DNA)
- Mass spectrometry to track myeloma proteins in the blood
- Advanced imaging like PET-MRI and whole-genome sequencing
- AI tools to personalize treatment and catch relapse early
- New therapies like trispecific antibodies, protein degraders, and new CAR T-cell options
So—Can Myeloma Be Cured?
For now, myeloma isn’t curable, But we’re getting closer—especially for people diagnosed early, treated aggressively, and who reach MRD negativity. Ongoing research aims to raise the number of long-term survivors and help more patients live full, cancer-free lives.
"Earlier onset of chemotherapy-induced neuropathic pain in females by ICAM-1–mediated accumulation of perivascular macrophage"
Source
Li Chen et al., Earlier onset of chemotherapy-induced neuropathic pain in females by ICAM-1–mediated accumulation of perivascular macrophages.Sci.n Adv.11,eadu2159(2025).DOI:10.1126/sciadv.adu2159 April 16, 2025.
Overview
Velcade® (bortezomib) is a common treatment for multiple myeloma, but one of its side effects is nerve pain, also known as peripheral neuropathy. This study looked at whether men and women experience this side effect differently—and the answer is yes.
Researchers reviewed patient records and found that women developed nerve-related symptoms earlier than men after starting bortezomib. In lab studies with mice, they discovered why: Bortezomib caused certain immune cells to enter the spinal cord more quickly in females. These cells released signals that triggered nerve cells and increased brain support cell (astrocyte) activity—both of which led to pain sooner.
What this means: Women may be more likely to feel nerve pain from bortezomib earlier than men. Understanding this difference could help doctors better personalize treatment and manage side effects more effectively for female patients.
"ATF4:p52 Complex Activates Oncogenic Enhancers in Multiple Myeloma via p300/CBP Recruitment to Regulate BACH1"
Source
Daniel Aron Ang, Nathan Harmston, Yinghui Li, ATF4:p52 Complex Activates Oncogenic Enhancers in Multiple Myeloma via p300/CBP Recruitment to Regulate BACH1, Cancer Letters, 2025, 217727, ISSN 0304-3835, https://doi.org/10.1016/j.canlet.2025.217727. April 16, 2025.
Overview
Multiple myeloma makes up about 1 in 5 cases of blood-related cancers. People with a more aggressive form of this disease often have a certain cell process turned on all the time, which helps the cancer grow. Scientists already knew that a protein called p52 plays a role in this process by turning on cancer-related genes.
In this study, researchers looked at cancer cells to find out how p52 works with other proteins. They discovered that p52 teams up with another protein called ATF4. Together, they turn on certain switches in the DNA, called enhancers, that help cancer grow. They also found that this pair activates a specific gene called BACH1, which makes cancer cells grow and divide faster.
This research helps explain how certain proteins make the cancer worse and could lead to better ways to treat multiple myeloma in the future.
"Belantamab Mafodotin in Patients with Relapsed/Refractory Multiple Myeloma: a Real-World Experience"
Source
Malin Hultcrantz, Andriy Derkach, Hani Hassoun, Neha Korde, Kylee MacLachlan, Sham Mailankody, Dhwani Patel, Urvi A Shah, Carlyn R Tan, David J Chung, Oscar B Lahoud, Heather Landau, Michael Scordo, Gunjan L Shah, David Nemirovsky, Ross S Firestone, Theresia Akhlaghi, Tala Shekarkhand, Sergio Giralt, Alexander M Lesokhin, Saad Z Usmani; Belantamab Mafodotin in Patients with Relapsed/Refractory Multiple Myeloma: a Real-World Experience. Blood Neoplasia 2025; 100103. doi: https://doi.org/10.1016/j.bneo.2025.100103 April 16, 2025.
Overview
Blenrep (belantamab mafodotin) is a treatment for multiple myeloma that targets a protein called BCMA found on myeloma cells. This study looked at how well it worked and how safe it was for patients treated at Memorial Sloan Kettering Cancer Center between 2020 and 2023.
The study included 94 patients who had already gone through many other treatments. Most had high-risk disease and were resistant to standard therapies. Even so, 43% of patients responded to belantamab mafodotin, and about 1 in 5 had a strong response.
On average, patients went about 4 months without their cancer getting worse, and overall survival was around 17 months. In patients who had already received other BCMA-targeted treatments, the response rate was lower, but some still benefited.
A common side effect was eye problems—about two-thirds of patients had some level of eye irritation or damage to the cornea (called keratopathy). Serious cases were less common, and all improved over time. Most patients were able to keep taking the drug with adjusted dosing. Some also had mild infections, but serious infections were rare.
Belantamab mafodotin helped many patients whose cancer was hard to treat, with manageable side effects, especially related to the eyes.
"Belantamab Mafodotin in Patients with Relapsed/Refractory Multiple Myeloma: a Real-World Experience"
Source
Malin Hultcrantz, Andriy Derkach, Hani Hassoun, Neha Korde, Kylee MacLachlan, Sham Mailankody, Dhwani Patel, Urvi A Shah, Carlyn R Tan, David J Chung, Oscar B Lahoud, Heather Landau, Michael Scordo, Gunjan L Shah, David Nemirovsky, Ross S Firestone, Theresia Akhlaghi, Tala Shekarkhand, Sergio Giralt, Alexander M Lesokhin, Saad Z Usmani; Belantamab Mafodotin in Patients with Relapsed/Refractory Multiple Myeloma: a Real-World Experience. Blood Neoplasia 2025; 100103. doi: https://doi.org/10.1016/j.bneo.2025.100103 April 16, 2025.
Overview
Blenrep (belantamab mafodotin) is a treatment for multiple myeloma that targets a protein called BCMA found on myeloma cells. This study looked at how well it worked and how safe it was for patients treated at Memorial Sloan Kettering Cancer Center between 2020 and 2023.
The study included 94 patients who had already gone through many other treatments. Most had high-risk disease and were resistant to standard therapies. Even so, 43% of patients responded to belantamab mafodotin, and about 1 in 5 had a strong response.
On average, patients went about 4 months without their cancer getting worse, and overall survival was around 17 months. In patients who had already received other BCMA-targeted treatments, the response rate was lower, but some still benefited.
A common side effect was eye problems—about two-thirds of patients had some level of eye irritation or damage to the cornea (called keratopathy). Serious cases were less common, and all improved over time. Most patients were able to keep taking the drug with adjusted dosing. Some also had mild infections, but serious infections were rare.
Belantamab mafodotin helped many patients whose cancer was hard to treat, with manageable side effects, especially related to the eyes.
"Clinical benefit loss in myeloma patients declining autologous stem cell transplantation: a real-world study"
Source
Wang, Y., Xiong, Y., Gu, S. et al. Clinical benefit loss in myeloma patients declining autologous stem cell transplantation: a real-world study. Discov Onc 16, 534 (2025). https://doi.org/10.1007/s12672-025-02356-y April 16, 2025
Overview
A stem cell transplant using a patient’s own cells—called autologous stem cell transplant (ASCT)—is a common treatment for multiple myeloma. With new drugs available today, doctors are rethinking when and for whom ASCT is most helpful.
This study looked at over 1,000 people in China diagnosed with multiple myeloma between 2012 and 2022. About half were healthy enough to get a stem cell transplant. Of those, some received ASCT and others did not.
The results showed that patients who had a stem cell transplant went longer without their cancer getting worse. However, there was no major difference in overall survival (how long people lived) between the two groups.
ASCT worked best for patients who were 60 or younger, had more advanced disease, or had certain genetic markers. It didn’t show the same benefits for older patients or those with early-stage disease, kidney problems, or no leftover cancer after treatment.
This study supports using ASCT for some patients with multiple myeloma, but not everyone may need it. Choosing who should get a transplant can depend on age, disease stage, and other health factors.
"Perspectives of Healthcare Providers and Patients with Relapsed/Refractory Multiple Myeloma on Treatment Priorities and Novel Therapies. Patient Prefer Adherence"
Source
Ailawadhi S, Biru Y, Clavreul S, San Miguel M, Cormier N, Efebera Y, Merz M, Sato A, Zeanah C, Watkins JL, Farrell J, Goldman EH, Popat R. Perspectives of Healthcare Providers and Patients with Relapsed/Refractory Multiple Myeloma on Treatment Priorities and Novel Therapies. Patient Prefer Adherence. 2025;19:1089-1104 https://doi.org/10.2147/PPA.S496106 April 16, 2025.
Overview
As new treatments like CAR-T cell therapy and bispecific antibodies (BsAbs) become more available for relapsed or hard-to-treat multiple myeloma, doctors and patients are facing new challenges in making treatment decisions.
This large international survey included over 2,200 people—patients and healthcare providers—from 7 countries. It looked at what matters most to patients, what challenges exist in using new therapies, and how decisions are made.
Patients said their top priorities were slowing down the cancer, avoiding side effects, and living longer. Doctors focused more on stopping the disease and increasing survival. Younger patients cared more about convenience and not having to travel to other hospitals for treatment.
Many doctors said it was hard to offer newer treatments like CAR-T or BsAbs because of logistical issues—like complex setup or hospital limitations. In the U.S., these therapies were offered more often than in Europe or Japan, but very few patients anywhere remembered being offered them at all.
Patients who got BsAbs liked that they were effective and didn’t take up too much time or money. Those who had CAR-T chose it because they had heard success stories and believed it could work for them with little financial stress.
The study shows a gap between what doctors and patients focus on when choosing treatments. It highlights the importance of talking openly and making decisions together, especially with newer options for treating multiple myeloma.
"The de novo DNA methyltransferase 3B is a novel epigenetic regulator of MYC in multiple myeloma, representing a promising therapeutic target to counter relapse"
Source
Muylaert, C., Van Hemelrijck, L.A., Van der Vreken, A. et al. The de novo DNA methyltransferase 3B is a novel epigenetic regulator of MYC in multiple myeloma, representing a promising therapeutic target to counter relapse. J Exp Clin Cancer Res 44, 125 (2025). https://doi.org/10.1186/s13046-025-03382-y April 17, 2025.
Overview
A protein called DNMT3B has been found at higher levels in patients with more aggressive or relapsed multiple myeloma (MM), but its exact role wasn’t clear until now.
This study looked at how DNMT3B affects MM and whether blocking it could help. Researchers used patient data and lab tests to see what happens when DNMT3B is turned off, either by using a drug called Nanaomycin A or through genetic techniques.
They found that MM cells with high DNMT3B levels were linked to worse outcomes. When DNMT3B was blocked, cancer cell growth slowed down, and more cells died. This treatment worked in both newly diagnosed and relapsed patient cells. It also made the cancer more sensitive to other MM drugs like bortezomib, melphalan, and anti-CD38 antibodies (such as daratumumab and isatuximab).
In short, the study shows that DNMT3B helps MM grow and survive—especially in high-risk patients—and that targeting it could lead to better treatments.
"Salvage autologous transplant in relapsed multiple myeloma: long-term follow-up of the phase 3 GMMG ReLApsE trial"
Source
Marc-Andrea Baertsch, Jana Schlenzka, Thomas Hielscher, Marc S. Raab, Sandra Sauer, Maximilian Merz, Elias Karl Mai, Carsten Müller-Tidow, Steffen Luntz, Anna Jauch, Peter Brossart, Martin Goerner, Stefan Klein, Bertram Glass, Peter Reimer, Ullrich Graeven, Roland Fenk, Mathias Haenel, Ivana von Metzler, Hans W. Lindemann, Christof Scheid, Igor-Wolfgang Blau, Hans J. Salwender, Richard Noppeney, Britta Besemer, Katja C. Weisel, Hartmut Goldschmidt; for the German-Speaking Myeloma Multicenter Group, Salvage autologous transplant in relapsed multiple myeloma: long-term follow-up of the phase 3 GMMG ReLApsE trial. Blood 2025; 145 (16): 1780–1787. doi: https://doi.org/10.1182/blood.2024027342 April 17, 2025
Overview
The GMMG ReLApsE trial looked at whether a second stem cell transplant (called salvage high-dose chemotherapy and ASCT, or sHDCT/ASCT) helps people with relapsed or refractory multiple myeloma who already had a stem cell transplant earlier in their treatment.
In the study, some patients received the second transplant followed by maintenance therapy. Others stayed on regular medication (lenalidomide and dexamethasone). After over 8 years of follow-up, the results showed no major difference in how long patients lived or how long they stayed in remission between the two groups.
Even among patients who had more time before their cancer came back after their first transplant (a factor doctors often use to decide on a second transplant), the second transplant didn’t lead to better outcomes.
Also, nearly a third of patients in the transplant group didn’t end up getting the second transplant, and some in the non-transplant group got it later, which made it harder to measure true differences. On top of that, new treatments like CAR T-cell therapy and triplet drug regimens are now available and may offer better results with fewer side effects.
The study suggests that doing a second transplant at relapse isn’t clearly better than staying on newer medicines, especially given the temporary drop in quality of life after a transplant. However, some patients may still choose this option based on personal health and preferences, and they should be fully informed about what to expect.
For most people whose myeloma has come back after an earlier transplant, newer treatments may be a better option than repeating the transplant.
"The decline of transplant for relapsed myeloma"
Source
Aurore Perrot, Cyrille Touzeau; The decline of transplant for relapsed myeloma. Blood 2025; 145 (16): 1711–1712. doi: https://doi.org/10.1182/blood.2024028275 April 17, 2025.
Overview
A new expert commentary reviewed long-term results from the ReLApsE trial, which compared two treatment options for people with relapsed or hard-to-treat multiple myeloma who had already had a stem cell transplant earlier in their treatment.
The trial looked at whether doing a second stem cell transplant (called salvage ASCT) followed by maintenance therapy worked better than staying on lenalidomide and dexamethasone (LEN/DEX) alone. After nearly 8 years of follow-up, there was no difference in how long patients lived or how long they stayed in remission between the two groups, even in patients whose cancer stayed away for a long time after their first transplant.
The study also showed that many patients in the transplant group didn’t make it to the second transplant, often due to side effects or disease progression. Some patients in the non-transplant group ended up getting a transplant later, which made the results harder to compare. Importantly, newer and more effective treatments—like CAR-T cell therapy, anti-CD38 antibody combinations, and carfilzomib-based regimens—are now available and may offer longer remission with fewer side effects.
These findings challenge the idea that a second transplant should be routine in relapsed multiple myeloma, especially when better options exist. However, in places with fewer resources or for patients looking for shorter treatment plans, a second transplant could still be considered, with maintenance therapy and full discussion of possible side effects.
For most people, newer treatment options are now preferred over a second transplant when myeloma returns
"The decline of transplant for relapsed myeloma"
Source
Aurore Perrot, Cyrille Touzeau; The decline of transplant for relapsed myeloma. Blood 2025; 145 (16): 1711–1712. doi: https://doi.org/10.1182/blood.2024028275 April 17, 2025.
Overview
A new expert commentary reviewed long-term results from the ReLApsE trial, which compared two treatment options for people with relapsed or hard-to-treat multiple myeloma who had already had a stem cell transplant earlier in their treatment.
The trial looked at whether doing a second stem cell transplant (called salvage ASCT) followed by maintenance therapy worked better than staying on lenalidomide and dexamethasone (LEN/DEX) alone. After nearly 8 years of follow-up, there was no difference in how long patients lived or how long they stayed in remission between the two groups, even in patients whose cancer stayed away for a long time after their first transplant.
The study also showed that many patients in the transplant group didn’t make it to the second transplant, often due to side effects or disease progression. Some patients in the non-transplant group ended up getting a transplant later, which made the results harder to compare. Importantly, newer and more effective treatments—like CAR-T cell therapy, anti-CD38 antibody combinations, and carfilzomib-based regimens—are now available and may offer longer remission with fewer side effects.
These findings challenge the idea that a second transplant should be routine in relapsed multiple myeloma, especially when better options exist. However, in places with fewer resources or for patients looking for shorter treatment plans, a second transplant could still be considered, with maintenance therapy and full discussion of possible side effects.
For most people, newer treatment options are now preferred over a second transplant when myeloma returns.
"A German multicenter real-world analysis of talquetamab in 138 patients with relapsed/refractory multiple myeloma"
Source
Frenking, J.H., Riedhammer, C., Teipel, R., Bassermann, F., Besemer, B., Bewarder, M., Braune, J., Brioli, A., Brunner, F., Dampmann, M., Fenk, R., Gezer, D.N., Goldman-Mazur, S., Hanoun, C., Högner, M., Khandanpour, C., Kolditz, K., Kos, I., Krönke, J., Kull, M., Landrin, V., Leitner, T., Merz, M., von Metzler, I., Michel, C.S., Müller-Tidow, C., Theurich, S., Trautmann-Grill, K., Wäsch, R., Zukovs, R., Hänel, M., Rasche, L. and Raab, M.S. (2025), A German multicenter real-world analysis of talquetamab in 138 patients with relapsed/refractory multiple myeloma. HemaSphere, 9: e70114. https://doi.org/10.1002/hem3.70114 April 17, 2025.
Overview
Talvey (talquetamab) is a new type of immunotherapy called a bispecific T-cell engager (BTCE) that helps the immune system fight multiple myeloma. It targets a protein called GPRC5D on myeloma cells and has shown strong results in clinical trials. It’s now approved in both the U.S. and Europe for treating relapsed or hard-to-treat multiple myeloma.
This study looked at how well talquetamab works in real-world settings, outside of clinical trials, by reviewing the cases of 138 patients treated in Germany. Many of these patients had more advanced disease than those typically included in trials. For example, almost half had high-risk genetics, over a third had disease outside the bones, and most had already gone through six or more previous treatments.
Even with these challenges, 65% of patients responded to talquetamab. On average, the treatment kept the cancer from getting worse for about 6.4 months. Serious side effects like cytokine release syndrome and nerve problems were rare, each happening in only a small number of patients.
Bottom line: Talquetamab is effective and generally safe, even for people with advanced or high-risk multiple myeloma. It may be used as a short-term or long-term treatment option in real-world care
"Infection-related adverse events comparison of Bortezomib, Carfilzomib and Ixazomib: A pharmacovigilance study based on FAERS"
Source
Wang, T., Xiang, C., Guo, R., Ti, J., & Zhang, S. (2025). Infection-related adverse events comparison of Bortezomib, Carfilzomib and Ixazomib: A pharmacovigilance study based on FAERS. Expert Opinion on Drug Safety. https://doi.org/10.1080/14740338.2025.2494692 April 17, 2025.
Overview
Proteasome inhibitors (PIs) like Velcade (bortezomib), Kyprolis (carfilzomib), and Ninlaro (ixazomib) are common treatments for multiple myeloma. This study looked at side effect reports from a large U.S. database (FAERS) to see what kinds of problems people had with these drugs in real-world use.
Here’s what the researchers found:
- Bortezomib was most often linked to nerve damage (peripheral neuropathy) and cytomegalovirus (CMV) infections.
- Carfilzomib was most often linked to kidney problems and serious bloodstream infections (bacteremia).
- Ixazomib was most often linked to vomiting and eye infections (conjunctivitis).
The study shows that each PI has its own set of risks, especially when it comes to infections. Knowing these risks can help doctors and patients make safer treatment choices. However, more research is needed to confirm these findings.
"Infection-related adverse events comparison of Bortezomib, Carfilzomib and Ixazomib: A pharmacovigilance study based on FAERS"
Source
Wang, T., Xiang, C., Guo, R., Ti, J., & Zhang, S. (2025). Infection-related adverse events comparison of Bortezomib, Carfilzomib and Ixazomib: A pharmacovigilance study based on FAERS. Expert Opinion on Drug Safety. https://doi.org/10.1080/14740338.2025.2494692 April 17, 2025.
Overview
Proteasome inhibitors (PIs) like Velcade (bortezomib), Kyprolis (carfilzomib), and Ninlaro (ixazomib) are common treatments for multiple myeloma. This study looked at side effect reports from a large U.S. database (FAERS) to see what kinds of problems people had with these drugs in real-world use.
Here’s what the researchers found:
- Bortezomib was most often linked to nerve damage (peripheral neuropathy) and cytomegalovirus (CMV) infections.
- Carfilzomib was most often linked to kidney problems and serious bloodstream infections (bacteremia).
- Ixazomib was most often linked to vomiting and eye infections (conjunctivitis).
The study shows that each PI has its own set of risks, especially when it comes to infections. Knowing these risks can help doctors and patients make safer treatment choices. However, more research is needed to confirm these findings.
"Prognostic value of minimal residual disease detected by EuroFlow next-generation flow cytometry and next-generation sequencing in patients with multiple myeloma achieving complete response and receiving lenalidomide maintenance after autotransplant: a prospective comparison study"
Source
Yoroidaka T, Takamatsu H, Urushihara R, Itagaki M, Yoshihara S, Sato K, Takezako N, Ozaki S, Suzuki K, Kohno K, Muta T, Matsumoto M, Terasaki Y, Yamashita T, Fuchida S- ichi, Sakamoto J, Ishida T, Suzuki K, Murakami H, Durie BG, Shimizu K. Prognostic value of minimal residual disease detected by EuroFlow next-generation flow cytometry and next-generation sequencing in patients with multiple myeloma achieving complete response and receiving lenalidomide maintenance after autotransplant: a prospective comparison study. Haematologica; https://doi.org/10.3324/haematol.2025.287411 [Early view]. April 17, 2025.
Overview
New treatments for multiple myeloma are helping patients reach deeper, longer-lasting responses. One way doctors check how well treatment is working is by measuring minimal residual disease (MRD)—the tiny number of cancer cells that may still be in the body after treatment.
This study in Japan looked at 52 newly diagnosed myeloma patients who had a stem cell transplant followed by maintenance therapy with lenalidomide. Researchers used two advanced methods—flow cytometry (NGF) and genetic testing (NGS)—to check for MRD at several time points up to two years after transplant.
They found the two MRD tests gave nearly identical results. Patients who stayed MRD-negative (no signs of remaining cancer) for more than 6 months had much better outcomes. Specifically, those patients were less likely to have their cancer come back within 3 years.
Staying MRD-negative for 6 months or more after treatment is a strong sign of long-term success. New testing methods are helping doctors track this more accurately
"Prognostic value of minimal residual disease detected by EuroFlow next-generation flow cytometry and next-generation sequencing in patients with multiple myeloma achieving complete response and receiving lenalidomide maintenance after autotransplant: a prospective comparison study"
Source
Yoroidaka T, Takamatsu H, Urushihara R, Itagaki M, Yoshihara S, Sato K, Takezako N, Ozaki S, Suzuki K, Kohno K, Muta T, Matsumoto M, Terasaki Y, Yamashita T, Fuchida S- ichi, Sakamoto J, Ishida T, Suzuki K, Murakami H, Durie BG, Shimizu K. Prognostic value of minimal residual disease detected by EuroFlow next-generation flow cytometry and next-generation sequencing in patients with multiple myeloma achieving complete response and receiving lenalidomide maintenance after autotransplant: a prospective comparison study. Haematologica; https://doi.org/10.3324/haematol.2025.287411 [Early view]. April 17, 2025.
Overview
New treatments for multiple myeloma are helping patients reach deeper, longer-lasting responses. One way doctors check how well treatment is working is by measuring minimal residual disease (MRD)—the tiny number of cancer cells that may still be in the body after treatment.
This study in Japan looked at 52 newly diagnosed myeloma patients who had a stem cell transplant followed by maintenance therapy with lenalidomide. Researchers used two advanced methods—flow cytometry (NGF) and genetic testing (NGS)—to check for MRD at several time points up to two years after transplant.
They found the two MRD tests gave nearly identical results. Patients who stayed MRD-negative (no signs of remaining cancer) for more than 6 months had much better outcomes. Specifically, those patients were less likely to have their cancer come back within 3 years.
Staying MRD-negative for 6 months or more after treatment is a strong sign of long-term success. New testing methods are helping doctors track this more accurately
"An oncolytic adenovirus targeting SLAMF7 demonstrates anti-myeloma efficacy"
Source
Stewart, G., Tazzyman, S., Sun, Y. et al. An oncolytic adenovirus targeting SLAMF7 demonstrates anti-myeloma efficacy. Leukemia (2025). https://doi.org/10.1038/s41375-025-02617-3 April 17, 2025.
Overview
Researchers are testing a new type of virus-based therapy to treat multiple myeloma. This therapy uses a modified virus called Ad[CE1A] that is designed to infect and kill only myeloma cells while leaving healthy cells alone.
Here’s what they found:
- The virus was able to infect and kill myeloma cells in lab tests but did not harm healthy donor cells.
- The virus triggers a unique kind of cell death, which might also alert the immune system to attack the cancer.
- In mice, the virus shrank tumors and worked even better when combined with common myeloma drugs like Velcade (bortezomib) and melphalan.
- One drug, melphalan, even made the virus work harder by helping it replicate inside the cancer cells.
This virus therapy could be a powerful new option for people with multiple myeloma, especially when used alongside current treatments. It not only kills cancer cells directly but may also boost the immune system to fight back.
"Gene Expression Profiling Identifies CAV1, CD44, and TFRC as Potential Diagnostic Markers and Therapeutic Targets for Multiple Myeloma"
Source
Ali, A., Ali, S.L., Ullah, W. et al. Gene Expression Profiling Identifies CAV1, CD44, and TFRC as Potential Diagnostic Markers and Therapeutic Targets for Multiple Myeloma. Cell Biochem Biophys (2025). https://doi.org/10.1007/s12013-025-01743-0 April 17, 2025.
Overview
Researchers are testing a new type of virus-based therapy to treat multiple myeloma. This therapy uses a modified virus called Ad[CE1A] that is designed to infect and kill only myeloma cells while leaving healthy cells alone.
Here’s what they found:
- The virus was able to infect and kill myeloma cells in lab tests but did not harm healthy donor cells.
- The virus triggers a unique kind of cell death, which might also alert the immune system to attack the cancer.
- In mice, the virus shrank tumors and worked even better when combined with common myeloma drugs like Velcade (bortezomib) and melphalan.
- One drug, melphalan, even made the virus work harder by helping it replicate inside the cancer cells.
This virus therapy could be a powerful new option for people with multiple myeloma, especially when used alongside current treatments. It not only kills cancer cells directly but may also boost the immune system to fight back.
"Gene Expression Profiling Identifies CAV1, CD44, and TFRC as Potential Diagnostic Markers and Therapeutic Targets for Multiple Myeloma"
Source
Ali, A., Ali, S.L., Ullah, W. et al. Gene Expression Profiling Identifies CAV1, CD44, and TFRC as Potential Diagnostic Markers and Therapeutic Targets for Multiple Myeloma. Cell Biochem Biophys (2025). https://doi.org/10.1007/s12013-025-01743-0 April 17, 2025.
Overview
Researchers are testing a new type of virus-based therapy to treat multiple myeloma. This therapy uses a modified virus called Ad[CE1A] that is designed to infect and kill only myeloma cells while leaving healthy cells alone.
Here’s what they found:
- The virus was able to infect and kill myeloma cells in lab tests but did not harm healthy donor cells.
- The virus triggers a unique kind of cell death, which might also alert the immune system to attack the cancer.
- In mice, the virus shrank tumors and worked even better when combined with common myeloma drugs like Velcade (bortezomib) and melphalan.
- One drug, melphalan, even made the virus work harder by helping it replicate inside the cancer cells.
This virus therapy could be a powerful new option for people with multiple myeloma, especially when used alongside current treatments. It not only kills cancer cells directly but may also boost the immune system to fight back.
"Addressing practical challenges with bispecific antibody therapy in multiple myeloma. Expert Opinion on Biological Therapy"
Source
Vasudevan, S., Mohan Lal, B., Vojjala, N., & Mohan, M. (2025). Addressing practical challenges with bispecific antibody therapy in multiple myeloma. Expert Opinion on Biological Therapy. https://doi.org/10.1080/14712598.2025.2495984 April 17, 2025.
Overview
Bispecific antibodies (bsAbs) have shown strong results for people with relapsed or hard-to-treat multiple myeloma. They work by helping the immune system find and kill cancer cells. These drugs are now being tested for use earlier in treatment, even in newly diagnosed patients.
Doctors and researchers are still figuring out the best way to use bsAbs. Questions include:
- Which bsAb is best for each patient?
- What’s the safest and most effective way to give the drug?
- How can side effects like cytokine release syndrome (CRS) and nerve issues (ICANS) be managed?
- What should be done if the cancer comes back after bsAb treatment?
To manage side effects, medicines like tocilizumab and steroids are being used, which might make it possible to give bsAbs safely outside of the hospital. Infections are a known risk, so patients often receive preventive treatments, but new ways to reduce infection risk—like giving the drug less often or for a limited time—are being explored.
Researchers are also studying how to use bsAbs alongside or before other treatments like CAR T-cell therapy.
BsAbs are a promising new option for multiple myeloma, but doctors are still learning how to use them in the safest and most effective way.
"Plasma Cell Enrichment and New Genomic Approaches in Multiple Myeloma: A Scoping Review"
Source
Juan Javier López Rivera, Natalia Gomez-Lopera, Diana Jennifer Moreno-Garcia, Rocío Orduz-Rodriguez, Juan F Combariza-Vallejo, Mario Isaza-Ruget, Plasma Cell Enrichment and New Genomic Approaches in Multiple Myeloma: A Scoping Review, The Journal of Applied Laboratory Medicine, 2025;, jfaf044, https://doi.org/10.1093/jalm/jfaf044 April 18, 2025.
Overview
Multiple myeloma is a complex cancer with many different genetic changes that can affect how the disease behaves and how it's treated. To understand each patient’s specific case, doctors use special lab tests—like FISH, PCR, and next-generation sequencing (NGS)—to find these genetic changes in bone marrow samples.
However, these tests don’t always work well if there aren’t enough myeloma (plasma) cells in the sample. That’s why plasma cell enrichment—a method that separates and concentrates the cancer cells—is important. It helps doctors get a clearer picture of what’s going on.
This review looked at nearly 400 studies and found that using enrichment techniques increased the detection of important genetic changes from 61% to over 95%. FISH is still the most common method, but newer tools like NGS are becoming more popular because they can spot changes more accurately.
Improving how myeloma cells are prepared for testing helps doctors find key genetic information, leading to better diagnosis, risk assessment, and treatment plans.
"Enhanced CAR-T Function and Mitochondrial Fitness from Earlier Unfractionated Stem Cell Product in Multiple Myeloma"
Source
Ciara L. Freeman, Julieta Abraham-Miranda, Meghan Menges, Reginald M. Atkins, Jerald Noble, Hien Liu, Salvatore Corallo, Luis A. Cuadrado Delgado, Albert J. Ribickas, Constanza Savid-Frontera, Gabriel de Avila, Omar A. Castaneda Puglianini, Jose Ochoa-Bayona, Doris K. Hansen, Melissa Alsina, Rachid Baz, Taiga Nishihori, Kenneth H. Shain, Frederick L. Locke, Enhanced CAR-T Function and Mitochondrial Fitness from Earlier Unfractionated Stem Cell Product in Multiple Myeloma, Molecular Therapy, 2025, ISSN 1525-0016, https://doi.org/10.1016/j.ymthe.2025.04.019. April 18, 2025.
Overview
CAR-T cell therapy targeting BCMA is a powerful treatment for people with relapsed or hard-to-treat multiple myeloma. But as the disease progresses and patients go through many treatments, their T-cells can become weak, making it harder to create strong, effective CAR-T cells.
This study looked at whether using T-cells collected and frozen earlier, for example, before a stem cell transplant, could make better CAR-T cells later. Researchers compared these early samples to the ones usually taken years later, just before patients receive CAR-T therapy.
They found that earlier-collected T-cells were healthier, expanded better in the lab, released more helpful immune signals, and were more powerful at killing myeloma cells. These cells also had better energy function and fewer signs of “exhaustion.”
Saving T-cells earlier in the disease—when they’re still strong—may lead to more effective CAR-T therapy later on. This could become an important step in long-term myeloma care planning.
"Trends and projections of the global and regional burden of multiple myeloma in adults aged 40 and over, 1990–2044"
Source
Liu, X., Zhuang, H., Li, F. et al. Trends and projections of the global and regional burden of multiple myeloma in adults aged 40 and over, 1990–2044. Sci Rep 15, 13595 (2025). https://doi.org/10.1038/s41598-025-96981-w April 19, 2025.
Overview
CAR-T cell therapy targeting BCMA is a powerful treatment for people with relapsed or hard-to-treat multiple myeloma. But as the disease
This study looked at global data on multiple myeloma from 1990 to 2021 to see how the disease has changed over time in people aged 40 and older. Researchers studied how often people are diagnosed (incidence), how many people are living with the disease (prevalence), how many are dying from it (mortality), and how much it affects daily life and health overall (disability-adjusted life years, or DALYs).
Here’s what they found:
- Multiple myeloma is on the rise worldwide—more people are being diagnosed and living with the disease.
- The number of deaths and the overall health burden have also gone up, but more slowly.
- Countries with middle levels of income and development have seen the fastest increase in new cases, more than four times the global average.
- These trends are expected to continue through 2044, especially in areas with fewer healthcare resources.
Multiple myeloma is becoming more common, especially in middle-income countries. This highlights the need for better awareness, early detection, and treatment options worldwide.
"Efficacy and Safety of Isatuximab, Carfilzomib, and Dexamethasone (IsaKd) in Multiple Myeloma Patients at the First Relapse After Autologous Stem Cell Transplantation and Lenalidomide Maintenance: Results from the Multicenter, Real-Life AENEID Study"
Source
Sgherza, N., Battisti, O., Curci, P., Conticello, C., Palmieri, S., Derudas, D., Germano, C., Martino, E. A., Mele, G., Pepa, R. D., Fazio, F., Mele, A., Rossini, B., Palazzo, G., Roccotelli, D., Rasola, S., Petrucci, M. T., Pastore, D., Tarantini, G., ... Musto, P. (2025). Efficacy and Safety of Isatuximab, Carfilzomib, and Dexamethasone (IsaKd) in Multiple Myeloma Patients at the First Relapse After Autologous Stem Cell Transplantation and Lenalidomide Maintenance: Results from the Multicenter, Real-Life AENEID Study. Pharmaceuticals, 18(4), 595. https://doi.org/10.3390/ph18040595 April 19, 2025.
Overview
This study looked at how well the drug combination of Sarclisa® (isatuximab}, Kyprolis® (carfilzomib), and dexamethasone (IsaKd) works for people with multiple myeloma who relapsed after their first round of treatment, including stem cell transplant and lenalidomide maintenance. These patients were not well represented in earlier clinical trials like IKEMA.
Researchers followed 82 patients in real-world clinics (not part of a trial). All had become resistant to lenalidomide. After about a year of follow-up:
- 79% of patients responded to IsaKd treatment.
- Over half had a very good partial response or better.
- The time before the cancer started to grow again (progression-free survival) was just over 24 months.
- 1-year survival was 85%, and the treatment’s safety matched earlier trials.
Some patients had disease outside the bone marrow, which made treatment harder and affected outcomes. Even so, IsaKd proved to be a good option for this group, especially since they are often not eligible for newer treatments like CAR-T or bispecific antibodies.
For patients whose myeloma returns during lenalidomide maintenance, IsaKd offers a strong and safe treatment option.
"Analysis of Ubiquitin-Conjugating Enzyme E2T (UBE2T) Protein Levels in the Bone Marrow Biopsy Specimens of Patients With Multiple Myeloma"
Source
Wang F, Yu N, Xia R (April 19, 2025) Analysis of Ubiquitin-Conjugating Enzyme E2T (UBE2T) Protein Levels in the Bone Marrow Biopsy Specimens of Patients With Multiple Myeloma. Cureus 17(4): e82571. doi:10.7759/cureus.82571 April 19, 2025.
Overview
This study looked at a protein called UBE2T, which may play a role in multiple myeloma. Earlier research suggested that levels of UBE2T are higher in myeloma, but most of that was based on computer models. This study aimed to find out if UBE2T is also higher at the protein level in real patient bone marrow samples.
Researchers tested bone marrow biopsies from 77 patients with multiple myeloma and compared them to samples from 16 people without blood cancers. They found that UBE2T levels were much higher in the myeloma group.
They also discovered that:
- UBE2T levels were higher in more advanced stages of myeloma (Stage II and III).
- Higher UBE2T levels were linked to higher blood levels of beta-2 microglobulin (β2-MG) and LDH, which are known to be markers of more aggressive disease.
- Patients with lower UBE2T levels had better one-year outcomes—87% were still in remission after a year, compared to 71% of those with higher levels.
UBE2T may be a helpful marker to predict disease severity and could become a new target for future treatments. More research is needed, but this protein might help doctors better understand and treat multiple myeloma.
"Analysis of Ubiquitin-Conjugating Enzyme E2T (UBE2T) Protein Levels in the Bone Marrow Biopsy Specimens of Patients With Multiple Myeloma"
Source
Wang F, Yu N, Xia R (April 19, 2025) Analysis of Ubiquitin-Conjugating Enzyme E2T (UBE2T) Protein Levels in the Bone Marrow Biopsy Specimens of Patients With Multiple Myeloma. Cureus 17(4): e82571. doi:10.7759/cureus.82571 April 19, 2025.
Overview
This study looked at a protein called UBE2T, which may play a role in multiple myeloma. Earlier research suggested that levels of UBE2T are higher in myeloma, but most of that was based on computer models. This study aimed to find out if UBE2T is also higher at the protein level in real patient bone marrow samples.
Researchers tested bone marrow biopsies from 77 patients with multiple myeloma and compared them to samples from 16 people without blood cancers. They found that UBE2T levels were much higher in the myeloma group.
They also discovered that:
- UBE2T levels were higher in more advanced stages of myeloma (Stage II and III).
- Higher UBE2T levels were linked to higher blood levels of beta-2 microglobulin (β2-MG) and LDH, which are known to be markers of more aggressive disease.
- Patients with lower UBE2T levels had better one-year outcomes—87% were still in remission after a year, compared to 71% of those with higher levels.
UBE2T may be a helpful marker to predict disease severity and could become a new target for future treatments. More research is needed, but this protein might help doctors better understand and treat multiple myeloma.
"Large-scale human myeloma cell line small molecule compound screen dataset"
Source
Hughitt, V.K., Simmons, J.K., Gorjifard, S. et al. Large-scale human myeloma cell line small molecule compound screen dataset. Sci Data 12, 661 (2025). https://doi.org/10.1038/s41597-025-04989-8 April 19, 2025.
Overview
While treatments for multiple myeloma (MM) have improved and people are living longer, the disease is still not curable, and relapses are common.
To find better treatments and understand why some drugs work better for certain patients, researchers tested 1,912 different drugs on 47 different myeloma cell lines in the lab. Each drug was tested at multiple doses to see how the cancer cells responded.
The goal of this large study was to:
- Discover new treatment options
- Identify biomarkers (signals in the cells) that might predict which patients will respond best to certain drugs
- All the results and tools from the study are publicly available online so other researchers can use the data to help move myeloma research forward.
This research could lead to more personalized and effective treatments for people with multiple myeloma.
"Efficacy and safety of isatuximab monotherapy to treat relapsed or refractory multiple myeloma: a pooled analysis of clinical trials"
Source
Dimopoulos, M., Sunami, K., Leleu, X. et al. Efficacy and safety of isatuximab monotherapy to treat relapsed or refractory multiple myeloma: a pooled analysis of clinical trials. Ann Hematol (2025). https://doi.org/10.1007/s00277-025-06343-9 April 21, 2025.
Overview
This study looked at how well Sarclisa (isatuximab), a type of antibody therapy, works on its own (monotherapy) for people with relapsed or refractory multiple myeloma who had already been through an average of four previous treatments.
Here’s what researchers found:
- About 26% of patients responded to isatuximab alone.
- On average, it took just 1 month to see a response, and the response lasted about 10 months.
- People lived an average of 5.6 months without disease worsening and had an average overall survival of 20.2 months.
The treatment was generally safe. The most common side effect was an infusion reaction (happened in about 46% of patients), mostly mild and usually during the first treatment.
Interestingly, patients aged 75 and older did as well or better than younger patients, showing that age did not affect the safety or effectiveness of the treatment.
Isatuximab is a promising option for people with relapsed multiple myeloma, even as a standalone treatment, and it may work well for older adults too.
"Cytomegalovirus Retinitis During Idecabtagene Vicleucel Therapy in Patients With Relapsed/Refractory Multiple Myeloma"
Source
Kikuchi, T., Watanabe, M., Tsukada, N., Matsumoto, C., Nomura-Yogo, M., Kunisada, K., Sawada, H., Sato, K., Takei, T., Ogura, M., Abe, Y., Suzuki, K., Hosoya, O. and Ishida, T. (2025), Cytomegalovirus Retinitis During Idecabtagene Vicleucel Therapy in Patients With Relapsed/Refractory Multiple Myeloma. Transpl Infect Dis e70040. https://doi.org/10.1111/tid.70040 April 21, 2025.
Overview
CAR-T cell therapy, like Abecma (idecabtagene vicleucel, or ide-cel), has helped improve outcomes for people with relapsed or refractory multiple myeloma. But like many powerful treatments, it can come with side effects—including infections.
One rare but serious infection is CMV retinitis, which affects the eyes and can lead to vision loss. This study from Japan looked at 53 patients who received ide-cel to see how often CMV retinitis occurred and how it presented.
Here’s what they found:
- About 9% of patients developed CMV retinitis within 6 months of treatment.
- Most cases began around 2 months after receiving ide-cel.
- Three of the four patients with CMV retinitis had vision problems, while one had no symptoms.
- One person had CMV retinitis even though their blood test was negative, showing that standard CMV tests may miss it.
CMV retinitis is a rare but serious complication of CAR-T therapy. Patients should have regular eye checkups, especially if they notice changes in vision, even if blood tests for CMV are normal.
"A systematic review of direct oral anticoagulants for thromboprophylaxis in multiple myeloma"
Source
Catalina Codreanu, Tessa Elling, Nic J.G. M. Veeger, Wilfried W.H. Roeloffzen, Karina Meijer, A systematic review of direct oral anticoagulants for thromboprophylaxis in multiple myeloma, Research and Practice in Thrombosis and Haemostasis, 2025, 102865, ISSN 2475-0379, https://doi.org/10.1016/j.rpth.2025.102865. April 22, 2025.
Overview
People with multiple myeloma have a higher risk of developing blood clots, also known as venous thromboembolism (VTE). To lower this risk, current guidelines recommend using aspirin or blood thinners like low molecular weight heparin, depending on how high the risk is.
This study reviewed whether direct oral anticoagulants (DOACs)—a newer type of blood thinner—might be a good option for preventing clots in myeloma patients. Researchers looked at 7 studies with a total of 416 patients, most of whom were newly diagnosed and receiving lenalidomide-based treatments, which are known to increase clot risk.
Here’s what they found:
- In some studies, no blood clots occurred, while in others, rates went up to 23.5%.
- Bleeding (a common side effect of blood thinners) happened in some cases, but serious bleeding was rare.
- Only a few studies reported death rates, and the overall quality of the evidence was very low.
While DOACs might help prevent blood clots in people with multiple myeloma, more research is needed to know how safe and effective they are for routine use.
"Multiple myeloma with 1q gain/amplification exhibits reduced CD38 expression via interleukin-6 receptor overexpression"
Source
Kuroki W, Kitadate A, Takahashi Y, Iwama S, Yamada M, Kobayashi T, et al. Multiple myeloma with 1q gain/amplification exhibits reduced CD38 expression via interleukin-6 receptor overexpression. Br J Haematol. 2025; 00: 1–12. https://doi.org/10.1111/bjh.20106 April 22, 2025.
Overview
Some people with multiple myeloma have a genetic change called 1q21 gain or amplification (1q+), which makes their cancer less responsive to daratumumab, a commonly used treatment. This study looked at why that happens.
Researchers found that in patients with 1q+ myeloma, the levels of CD38—the protein that daratumumab targets—were much lower than in patients without the 1q+ change. They discovered this is likely caused by high levels of the interleukin-6 receptor (IL6R), which is located in the same 1q21 region.
Here's what they learned:
- The IL-6 protein triggers a chain reaction in 1q+ cells that lowers CD38 levels, making daratumumab less effective.
- This doesn't happen in patients without the 1q+ change.
- But when the researchers blocked the IL-6 pathway using drugs like ruxolitinib or tocilizumab, CD38 levels went back up—suggesting a possible way to make daratumumab work better in these patients.
People with 1q+ myeloma may not respond well to daratumumab because of lower CD38 levels caused by IL-6. Blocking this pathway might improve treatment in the future.
"Multiple myeloma with 1q gain/amplification exhibits reduced CD38 expression via interleukin-6 receptor overexpression"
Source
Kuroki W, Kitadate A, Takahashi Y, Iwama S, Yamada M, Kobayashi T, et al. Multiple myeloma with 1q gain/amplification exhibits reduced CD38 expression via interleukin-6 receptor overexpression. Br J Haematol. 2025; 00: 1–12. https://doi.org/10.1111/bjh.20106 April 22, 2025.
Overview
Some people with multiple myeloma have a genetic change called 1q21 gain or amplification (1q+), which makes their cancer less responsive to daratumumab, a commonly used treatment. This study looked at why that happens.
Researchers found that in patients with 1q+ myeloma, the levels of CD38—the protein that daratumumab targets—were much lower than in patients without the 1q+ change. They discovered this is likely caused by high levels of the interleukin-6 receptor (IL6R), which is located in the same 1q21 region.
Here's what they learned:
- The IL-6 protein triggers a chain reaction in 1q+ cells that lowers CD38 levels, making daratumumab less effective.
- This doesn't happen in patients without the 1q+ change.
- But when the researchers blocked the IL-6 pathway using drugs like ruxolitinib or tocilizumab, CD38 levels went back up—suggesting a possible way to make daratumumab work better in these patients.
People with 1q+ myeloma may not respond well to daratumumab because of lower CD38 levels caused by IL-6. Blocking this pathway might improve treatment in the future.
"Targeting the RBM39-MEK5 axis synergizes with bortezomib to inhibit the malignant growth of multiple myeloma"
Source
Jia Liu, Zilu Zhang, Wenbin Xu, Mingyuan Jia, Xinyi Zeng, Chengyu Wu, Ze Fu, Xiaoguang Xu, Chenjing Ye, Chao Wu, Hanzhang Xu, Hu Lei, Yingli Wu, Hua Yan; Targeting the RBM39-MEK5 axis synergizes with bortezomib to inhibit the malignant growth of multiple myeloma. Blood Adv 2025; 9 (8): 1991–2005. doi: https://doi.org/10.1182/bloodadvances.2025015815 April 22, 2025.
Overview
This study looked at a protein called RBM39, which helps control how genes are "spliced"—a process that edits gene messages before they’re used to make proteins. In multiple myeloma, RBM39 is found at higher-than-normal levels, and this has been linked to worse outcomes.
Researchers discovered that myeloma cells rely on RBM39 to grow and survive. When they blocked or removed RBM39, the cancer cells stopped growing. A drug called indisulam, which causes the body to break down RBM39, showed strong anti-cancer effects in lab and animal tests.
They also found that blocking RBM39 affected another protein called MEK5, which is important for keeping myeloma cells alive. When MEK5 is not made correctly, it loses its function and is destroyed by the body.
Even more promising, targeting RBM39 or MEK5 made standard myeloma treatment with bortezomib work better, suggesting a new way to boost treatment effectiveness.
RBM39 is a key driver of myeloma cell growth. Blocking it with drugs like indisulam—and combining that with existing treatments—could lead to more effective therapies in the future.
"Age-related mesenchymal stromal cell senescence is associated with progression from MGUS to multiple myeloma"
Source
Plakhova, N., Panagopoulos, V., Cantley, M.D. et al. Age-related mesenchymal stromal cell senescence is associated with progression from MGUS to multiple myeloma. Leukemia (2025). https://doi.org/10.1038/s41375-025-02621-7 April 22, 2025.
Overview
People with monoclonal gammopathy of undetermined significance (MGUS) have a small risk of developing multiple myeloma over time. This risk gets higher with age, but the reason hasn’t been fully understood.
This study suggests that as we age, certain cells in the bone marrow—called mesenchymal stromal cells (MSCs)—become senescent, meaning they grow old and stop working properly. These aging cells show signs like slower growth and changes in shape. The study found that these “old” MSCs may actually help myeloma cells grow, which could explain why older adults with MGUS are more likely to develop multiple myeloma.
Researchers also found that senescent MSCs produce more of a substance called Gremlin1, which seems to support the growth of cancer cells.
Aging bone marrow cells may play a role in pushing MGUS to become multiple myeloma. Understanding this process could help find ways to delay or prevent disease progression in the future.
"Establishing measurable residual disease trajectories for patients on treatment for newly diagnosed multiple myeloma as benchmark for deployment of T-cell redirection therapy"
Source
Bal, S., Magnusson, T., Ravi, G. et al. Establishing measurable residual disease trajectories for patients on treatment for newly diagnosed multiple myeloma as benchmark for deployment of T-cell redirection therapy. Blood Cancer J. 15, 73 (2025). https://doi.org/10.1038/s41408-025-01252-6 April 23, 2025.
Overview
Autologous stem cell transplant (ASCT) is a common next step after initial treatment for people with newly diagnosed multiple myeloma (NDMM). It helps deepen the treatment response but can come with short- and long-term side effects. One way doctors measure how well treatment is working is by checking for minimal residual disease (MRD)—tiny amounts of cancer that may still be in the body.
This study looked at how much ASCT helps lower MRD levels, especially after triplet or quadruplet drug combinations given as the first treatment (induction). MRD was measured before and after the transplant in 330 patients.
Here’s what they found:
- After quadruplet induction, 29% of patients had very low MRD levels. That number increased to 59% after ASCT.
- Even for patients who still had MRD after induction, 69% saw a big drop in MRD levels after the transplant.
- ASCT had the most impact in patients with high-risk genetic features.
- Whether patients received triplet or quadruplet therapy beforehand, ASCT still helped reduce MRD.
ASCT remains an effective way to lower MRD in newly diagnosed multiple myeloma. This study sets a new standard to help researchers compare how well newer treatments work as alternatives to transplant.
"Downexpression of miR- 17 - 5p and miR- 125a- 5p is Potentially Associated with the Renal Impairment Through STAT- 3 and CD69 in Multiple Myeloma Adult Patients"
Source
Haroun RA, Eweida SM, Elsaid DS, Al-Dosoky MA, Elbedewy TA, Keshk RA, Gamal-Eldin SM. Downexpression of miR- 17 - 5p and miR- 125a- 5p is Potentially Associated with the Renal Impairment Through STAT- 3 and CD69 in Multiple Myeloma Adult Patients. Biochem Genet. 2025 Apr 23. doi: 10.1007/s10528-025-11094-3. Epub ahead of print.
Overview
Multiple myeloma (MM) can cause kidney problems, which can make treatment more difficult. In this study, researchers looked at certain molecules and proteins in the blood to see if they could predict the risk of kidney issues in MM patients. They focused on two microRNAs (miR-17-5p and miR-125a-5p) and two proteins (STAT3 and CD69). They found that levels of miR-17-5p and miR-125a-5p were lower in MM patients with kidney problems, while STAT3 and CD69 were higher. These markers were able to predict kidney issues with high accuracy. Patients with lower miR-17-5p and miR-125a-5p and higher STAT3 and CD69 had a poorer outlook and shorter survival. The study suggests these markers could help doctors predict kidney problems and overall survival in MM patients.
"Coronary artery calcification score as a prognostic factor in younger patients with multiple myeloma undergoing autologous stem cell therapy"
Source
Meyer, HJ., Pönisch, W., Borggrefe, J. et al. Coronary artery calcification score as a prognostic factor in younger patients with multiple myeloma undergoing autologous stem cell therapy. Cardio-Oncology 11, 38 (2025). https://doi.org/10.1186/s40959-025-00338-1 April 23, 2025.
Overview
A study looked at whether coronary artery calcification (CAC), which is a buildup of calcium in the heart's arteries, could help predict overall survival in people with multiple myeloma (MM). The researchers examined 127 MM patients who had stem cell transplants between 2009 and 2019. They used CT scans to measure the CAC score for each patient. However, the study found no clear link between the CAC score and survival. The number of deaths in patients with or without coronary calcification was similar, and the CAC score didn’t seem to predict how long patients would live after treatment. In conclusion, while coronary calcification can indicate heart problems, it doesn’t appear to be a strong factor in predicting survival for younger MM patients undergoing stem cell therapy.
"Effect of Intravenous Immunoglobulin (IVIG) Supplementation on infection-free survival in recipients of BCMA-directed bispecific antibody therapy for multiple myeloma"
Source
Mohan, M., Szabo, A., Cheruvalath, H. et al. Effect of Intravenous Immunoglobulin (IVIG) Supplementation on infection-free survival in recipients of BCMA-directed bispecific antibody therapy for multiple myeloma. Blood Cancer J. 15, 74 (2025). https://doi.org/10.1038/s41408-025-01282-0 April 23, 2025.
Overview
This study looked at how primary intravenous immunoglobulin (IVIG) therapy affects the health of people with multiple myeloma who are being treated with BCMA-directed bispecific antibodies (bsAb), like teclistamab. Infections are a major concern for these patients, and IVIG is often used to help prevent infections. The study included 225 patients and compared those who started IVIG before their first infection to those who did not.
The results showed that while the overall rate of infections was similar in both groups, those who received IVIG had fewer severe infections and stayed infection-free for longer. Additionally, patients on IVIG had better progression-free survival (PFS) and overall survival (OS) compared to those without IVIG. The study also found that IVIG was linked to improved overall survival, particularly for those with less aggressive disease.
IVIG therapy was associated with better clinical outcomes, including fewer infections and longer survival, for multiple myeloma patients treated with BCMA-directed bispecific antibodies.
"Spatial imaging unlocks the potential of charting multiple myeloma and extramedullary disease"
Source
Desantis, V., Andriano, A., Düking, T. et al. Spatial imaging unlocks the potential of charting multiple myeloma and extramedullary disease. J Hematol Oncol 18, 47 (2025). https://doi.org/10.1186/s13045-025-01699-x April 23, 2025.
Overview
Extramedullary disease (EMD) in multiple myeloma (MM) is a serious challenge because it’s not fully understood, especially how it behaves differently from disease in the bone marrow. In this study, researchers compared 10 matched samples from both bone marrow and EMD sites to better understand these differences. They used advanced imaging techniques to look at specific biomarkers that could guide personalized treatments for MM.
The study found that EMD plasma cells had higher levels of BCL-2, a protein that prevents cell death, suggesting that BCL-2 inhibitors could be used to target this pathway. EMD also showed higher levels of EZH2, a protein that helps tumors stay aggressive, pointing to the potential use of epigenetic therapies. Additionally, there was a significant increase in the distance between T-cells (immune cells) and the plasma cells in EMD, indicating that the immune system has a harder time recognizing and fighting the disease in these areas.
Another key finding was that CD38 expression, a target for some therapies, was much lower in EMD plasma cells. This suggests that EMD may be resistant to certain treatments. Overall, the study highlights the biological differences between bone marrow and EMD in MM, which may require new approaches to treatment, especially in cases where EMD is present. The insights from this research could lead to better-targeted therapies for this challenging form of the disease.
"Bridging intensity is associated with impaired hematopoietic recovery following BCMA CAR-T therapy for multiple myeloma"
Source
Jan Hendrik Frenking, Xiang Zhou, Kai Rejeski, Vivien Wagner, Patrick Costello, Thomas Hielscher, Lilan Gatti, Joseph Kauer, Omar Nadeem, Elias K Mai, Christian S Michel, Mirco J Friedrich, David Nikolov Sedloev, Niels Weinhold, Hartmut Goldschmidt, Klaus Herfarth, Anita Schmitt, Michael Hundemer, Michael Schmitt, Carsten Müller-Tidow, Max S. Topp, Hermann Einsele, Peter Dreger, Nikhil C. Munshi, Adam S. Sperling, Leo Rasche, Sandra Sauer, Marc S. Raab; Bridging intensity is associated with impaired hematopoietic recovery following BCMA CAR-T therapy for multiple myeloma. Blood Adv 2025; bloodadvances.2024015732. doi: https://doi.org/10.1182/bloodadvances.2024015732 April 23, 2025
Overview
CAR T-cell therapy has become an important treatment for people with relapsed or refractory multiple myeloma. However, because it takes time to prepare CAR T cells (usually weeks), patients often need other treatment—called "bridging therapy"—to control their disease while they wait.
This study looked at how different types of bridging therapy affect blood cell recovery after CAR T-cell treatment in 158 patients. Researchers grouped the bridging therapies by how intense they were: no chemotherapy (non-CTX), moderate chemotherapy (1–2 drugs), or intensive chemotherapy (3 or more drugs or high-dose therapy with a stem cell transplant).
They found that the more intense the chemotherapy, the harder it was for patients' blood counts to recover after CAR T-cell therapy. Patients who received intensive chemotherapy had slower recovery of neutrophils and platelets, more infections, and a higher chance of low blood counts (called cytopenias) later on.
The study suggests that less intense or targeted treatments may be better for bridging in some patients. It also highlights the need for extra care and monitoring for those who receive stronger chemotherapy before their CAR T-cell infusion.
"Immune profiling of smoldering multiple myeloma patients treated in a phase lb study of PVX-410 vaccine targeting XBP1/CD138/CS1 antigens, and citarinostat, a histone deacetylase inhibitor (HDACi) with and without lenalidomide"
Source
Cirstea, D., Shome, R., Samur, M. et al. Immune profiling of smoldering multiple myeloma patients treated in a phase lb study of PVX-410 vaccine targeting XBP1/CD138/CS1 antigens, and citarinostat, a histone deacetylase inhibitor (HDACi) with and without lenalidomide. Blood Cancer J. 15, 77 (2025). https://doi.org/10.1038/s41408-025-01272-2 April 24, 2025.
Overview
Smoldering multiple myeloma (SMM) is an early, symptom-free stage of myeloma that may eventually progress to active disease. Researchers are exploring ways to boost the immune system to slow or stop this progression.
This study tested a vaccine called PVX-410, which helps the immune system recognize and attack myeloma cells. The vaccine was given with a drug called citarinostat—and in some patients, also with Revlimid (lenalidomide), a medication that boosts immune function.
Fifteen patients with SMM were treated and followed for over a year. The vaccine combinations appeared safe and successfully activated several types of immune cells that fight cancer. Only one patient progressed to full myeloma during the study.
However, because there was no comparison group (patients who got the vaccine alone), it's unclear exactly how much the added drugs helped. Also, while the immune response was strong, the actual clinical benefit was modest.
The results suggest that cancer vaccines like PVX-410 may work better when used earlier in the disease or combined with other therapies like CAR T-cells or T-cell engagers. More research is needed to understand the best way to use vaccines in treating or preventing myeloma.
"USP5 motivates immunosuppressive microenvironment in multiple myeloma by activating STAT2-PFKFB4-mediated glycolysis"
Source
Long, S., Ding, T., Zheng, Y. et al. USP5 motivates immunosuppressive microenvironment in multiple myeloma by activating STAT2-PFKFB4-mediated glycolysis. Cancer Immunol Immunother 74, 180 (2025). https://doi.org/10.1007/s00262-025-04031-1 April 24, 2025.
Overview
In multiple myeloma, cancer cells often use a process called glycolysis to grow and survive. This process creates a substance called lactate, which can weaken the immune system and help cancer hide from it.
Researchers found that a protein called USP5 is more active than normal in people with multiple myeloma. USP5 helps myeloma cells stay alive and boosts glycolysis, leading to more lactate. This lactate then encourages immune cells called macrophages to shift into a type (called M2-like) that protects the cancer instead of fighting it.
When researchers reduced the amount of USP5 in lab and animal models, the myeloma cells died more easily, made less lactate, and were less able to create this immune-suppressing environment. They discovered that USP5 does this by affecting another protein called STAT2, which controls a gene involved in glycolysis.
These findings suggest that blocking USP5 could be a new way to treat multiple myeloma—by both killing cancer cells and helping the immune system fight back.
"Targeting Enterobacter cloacae attenuates osteolysis by reducing ammonium in multiple myeloma"
Source
Qin Yang, Yinghong Zhu, Xingxing Jian, Yi Qiu, Yan Zhu, Lia Zhao, Yanjuan He, Gang An, Lugui Qiu, Jiaojiao Guo, Nihan He, Huerxidan Abudumijiti, Cong Hu, Xun Chen, Siqing Huang, Xiangling Feng, Xin Li, Jing Liu, Yajing Xu, Wen Zhou; Targeting Enterobacter cloacae attenuates osteolysis by reducing ammonium in multiple myeloma. Blood 2025; 145 (17): 1876–1889. doi: https://doi.org/10.1182/blood.2024025694 April 24, 2025.
Overview
Multiple myeloma can cause bone loss, known as osteolysis, which leads to pain, fractures, and worse overall health. A new study has found that a common gut bacterium called Enterobacter cloacae may play a role in this bone damage.
Researchers found that this bacterium is more common in the guts of people with myeloma-related bone disease. In mouse studies, they showed that E. cloacae increases levels of a chemical called ammonium in the blood. This ammonium makes bone-destroying cells (called osteoclasts) more active and stable, which leads to more bone loss.
When scientists used a special version of E. cloacae that couldn’t make ammonium, and gave mice probiotics (beneficial bacteria), the bone damage was less severe.
This research suggests that gut bacteria—and the chemicals they produce—may affect how myeloma damages bones. In the future, targeting certain gut bacteria or lowering ammonium levels could become a new way to help protect bone health in people with multiple myeloma.
"Recycling is “bad to the bone” in patients with myeloma"
Source
Kevin C. Miller, Alexander M. Lesokhin; Recycling is “bad to the bone” in patients with myeloma. Blood 2025; 145 (17): 1832–1833. doi: https://doi.org/10.1182/blood.2024027940 April 24, 2025.
Overview
Researchers are discovering that gut bacteria may play a surprising role in bone damage caused by multiple myeloma.
A recent study found that certain gut bacteria, especially Enterobacter cloacae and Klebsiella pneumoniae, can produce a substance called ammonium by recycling nitrogen. In lab models, this ammonium increased the activity of bone-destroying cells called osteoclasts. As a result, the bones broke down more easily—a serious issue for people with multiple myeloma, since more than 80% already have bone damage when they’re diagnosed.
The researchers also found a possible solution. They gave mice a probiotic mix that reduced the amount of harmful bacteria and slowed down the bone damage caused by myeloma. This adds to earlier work from the same group showing that gut bacteria can support myeloma growth by recycling nitrogen into nutrients that feed cancer cells.
These findings suggest a two-way relationship: myeloma cells help certain gut bacteria grow, and in turn, those bacteria produce substances that make bone disease worse. This discovery may open the door to new treatments—like probiotics, antibiotics, or diet changes—that target the gut to help protect the bones and slow down disease progression.
Clinical trials are already underway to test whether improving gut health can delay or prevent multiple myeloma. Researchers are also exploring whether certain bacteria in the gut can help predict how well a person might respond to treatment.
"Comprehensive evaluation of clinical prognostic parameters in a real-world cohort of 812 patients with multiple myeloma"
Source
Wang, Y., Lan, T., Zhou, C. et al. Comprehensive evaluation of clinical prognostic parameters in a real-world cohort of 812 patients with multiple myeloma. Clin. Cancer Bull. 4, 4 (2025). https://doi.org/10.1007/s44272-025-00031-5 April 24, 2025.
Overview
A recent study from Zhongshan Hospital at Fudan University in China analyzed data from 812 patients newly diagnosed with multiple myeloma between 2010 and 2021. The researchers aimed to improve the accuracy of predicting patient outcomes by examining a wide range of factors.
They discovered ten important factors that can help determine a patient's prognosis:
- ECOG Performance Status: Measures a patient's daily living abilities.
- Extramedullary Lesions: Presence of cancer outside the bone marrow.
- Platelet Count: Number of platelets in the blood.
- Reticulocyte Count: Indicates bone marrow activity.
- Anion Gap: A blood test that can signal metabolic issues.
- Hypercalcemia: High calcium levels in the blood.
- Complement C3 Levels: Part of the immune system that may influence disease progression.
- β2-Microglobulin Levels: A protein associated with tumor burden.
- Cytogenetic Abnormalities: Genetic changes in cancer cells.
- Cancer Network
- Interleukin-2 Receptor Levels: A marker related to immune system activity.
Using these factors, the researchers developed a new risk scoring system that more accurately predicts patient outcomes compared to existing methods.
This advancement could lead to more personalized treatment plans and better management of multiple myeloma.
"High-risk features of early relapse in newly-diagnosed multiple myeloma: The impact of cytogenetics and response to initial therapy"
Source
Manubens, A., Paiva, B., Gutiérrez, N.C., Fernandez, M., Calasanz, M.-J., Rosiñol, L., Oriol, A., Blanchard, M.J., Carrillo, E., Benavente, C., Martínez-López, J., Bargay, J., Hernández, M.T., de la Rubia, J., González, Y., Paricio, M., de Arriba, F., Ocio, E.M., Teruel, A.I., de la Guia, A.L., Sirvent, M., Gironella, M., Sampol, A., Arguiñano, J.M., Cabrera, C., Alegre, A., Granell, M., Cabañas, V., Núñez-Córdoba, J.M., Mateos, M.V., Lahuerta, J.J., San Miguel, J.F., Bladé, J. and Rodriguez-Otero, P. (2025), High-risk features of early relapse in newly-diagnosed multiple myeloma: The impact of cytogenetics and response to initial therapy. HemaSphere, 9: e70127. https://doi.org/10.1002/hem3.70127 April 24, 2025.
Overview
A recent study involving over 1,200 patients with newly diagnosed multiple myeloma found that about 22% experienced an early relapse within 18 months of starting treatment. These early relapses were linked to significantly shorter overall survival—19 months compared to 114 months for those who did not relapse early.
Researchers developed a risk scoring system to better predict which patients are at risk for early relapse. This system considers factors such as performance status, disease stage, calcium levels, and specific genetic markers. Patients were categorized into low-, intermediate-, and high-risk groups, with early relapse rates of 2%, 24.5%, and 59%, respectively.
Identifying patients at higher risk for early relapse allows healthcare providers to consider alternative treatment strategies early on, potentially improving outcomes.
If you have multiple myeloma, you may want to discuss your risk factors with your doctor and whether this new scoring system might inform your treatment plan.
"Targeting MTHFD2 alters metabolic homeostasis and synergizes with bortezomib to inhibit multiple myeloma"
Source
Jia, M., Fu, Z., Ye, C. et al. Targeting MTHFD2 alters metabolic homeostasis and synergizes with bortezomib to inhibit multiple myeloma. Cell Death Discov. 11, 201 (2025). https://doi.org/10.1038/s41420-025-02498-6 April 25, 2025.
Overview
Researchers are looking for new ways to treat multiple myeloma, especially when the cancer becomes resistant to current drugs. A recent study looked at a protein called MTHFD2, which is found at high levels in many myeloma cells. Patients with more of this protein tend to have worse outcomes.
The scientists found that blocking MTHFD2 slowed the growth of myeloma cells in lab tests and in mice. It worked by disrupting how the cancer cells make energy and how they respond to stress.
They also discovered that blocking MTHFD2 made myeloma cells more sensitive to bortezomib, a common drug used to treat this cancer. When used together, the two treatments worked better than either one alone.
This research suggests that MTHFD2 could be a promising new target for treating multiple myeloma, especially in combination with existing therapies.
"Real-World Experience of Weekly Carfilzomib in Combination with Cyclophosphamide and Dexamethasone in Multiple Myeloma Relapsed/Refractory to Bortezomib and Lenalidomide"
Source
Yang C, Kim C, Kwak K, Kang KW, Park Y, Kim BS, Jeong SH, Park JS, Choi YS. Real-World Experience of Weekly Carfilzomib in Combination with Cyclophosphamide and Dexamethasone in Multiple Myeloma Relapsed/Refractory to Bortezomib and Lenalidomide. Cancer Res Treat. ;0.0. doi: 10.4143/crt.2025.418 April 25, 2025.
Overview
This study looked at how well a weekly treatment combination—Kyprolis (carfilzomib), Cytoxan (cyclophosphamide), and dexamethasone (KCd)—works for people with relapsed or refractory multiple myeloma (RRMM) who had already been treated with bortezomib and lenalidomide. Most of the patients in the study no longer responded to lenalidomide.
In total, 33 patients received the KCd combination. Carfilzomib was given once a week, and patients took cyclophosphamide and dexamethasone by mouth.
Here are the study's key findings:
- About 2 out of 3 patients (66.7%) responded to the treatment.
- Around 15% had a complete response (no signs of myeloma), and over 40% had a very good partial response or better.
- The treatment kept the disease from getting worse for a median of 13.5 months.
- The most common serious side effect was low white blood cell counts (neutropenia), but most side effects were manageable.
The weekly KCd regimen worked well and was generally safe in patients who had already tried several other treatments. It may be a good option, especially for those whose myeloma no longer responds to lenalidomide.
"Impact of single dose of pegfilgrastim on peripheral blood stem cell harvest in patients with multiple myeloma or malignant lymphoma"
Source
Goto, H., Sawa, M., Fujiwara, Si. et al. Impact of single dose of pegfilgrastim on peripheral blood stem cell harvest in patients with multiple myeloma or malignant lymphoma. Sci Rep 15, 14523 (2025). https://doi.org/10.1038/s41598-025-98453-7 April 25, 2025.
Overview
This study looked at whether a single dose of pegfilgrastim, a long-acting medication, could work as well as daily doses of filgrastim to help move stem cells into the bloodstream. This process, called stem cell mobilization, is important for patients with multiple myeloma (MM) or lymphoma who are preparing for a stem cell transplant.
Researchers compared pegfilgrastim and filgrastim in patients with multiple myeloma, and also tested pegfilgrastim alone in some lymphoma patients.
Here are the study's key findings for myeloma patients:
- 100% of patients who received pegfilgrastim reached the needed stem cell count for transplant.
- 96.7% of those on daily filgrastim reached the same goal.
- Both drugs were similarly safe, with no major side effects.
Pegfilgrastim worked just as well as filgrastim for collecting enough stem cells—but with only one dose instead of daily injections. This could make the process easier and more convenient for patients.
"Research progress on N6-methyladenosine and non-coding RNA in multiple myeloma"
Source
Sun, X., Zhou, Y., Zhu, W. et al. Research progress on N6-methyladenosine and non-coding RNA in multiple myeloma. Discov Onc 16, 615 (2025). https://doi.org/10.1007/s12672-025-02386-6 April 25, 2025.
Overview
Scientists are studying two key molecules—m6A and non-coding RNA (ncRNA)—that may play important roles in how multiple myeloma (MM) grows, spreads, and becomes resistant to treatment. These molecules can affect how myeloma cells behave, including how fast they grow, how they avoid dying, and how they respond to treatment. High levels of these molecules have also been linked to worse outcomes for patients.
Although more research is needed, early studies suggest that targeting m6A and ncRNA could lead to new treatment options. Researchers are looking into three possible approaches:
- Lowering ALKBH5 could help kill myeloma cells.
- Reducing METTL3 might slow down bone damage from the disease.
- Blocking FTO could help make myeloma cells respond better to current drugs.
While these are still experimental ideas, they show promise. Scientists are now working on ways to make these treatments more precise, safer, and better able to reach the bone marrow. Future studies using advanced tools like single-cell sequencing and 3D organoid models may help tailor these treatments to individual patients
"Impact of induction regimens on stem cell mobilization yields in newly diagnosed multiple myeloma"
Source
Kwon, S., Park, H.Y., Byun, J.M. et al. Impact of induction regimens on stem cell mobilization yields in newly diagnosed multiple myeloma. Ann Hematol (2025). https://doi.org/10.1007/s00277-025-06372-4 April 25, 2025.
Overview
Autologous stem cell transplant (ASCT) is a key part of treatment for people with newly diagnosed multiple myeloma. But some of the drugs used before transplant—called induction therapy—can make it harder to collect enough stem cells.
This study looked at how three common drug combinations affect stem cell collection and transplant outcomes:
- VTd: Velcade (bortezomib), Thalomid (thalidomide), and dexamethasone
- VRd: Velcade (bortezomib), Revlimid (lenalidomide), and dexamethasone
- DVTd: Darzalex (daratumumab), Velcade (bortezomib), Thalomid (thalidomide), and dexamethasone
The study had these key findings:
- Patients on VTd had the highest number of collected stem cells and needed fewer extra treatments to boost stem cell collection.
- Patients on VRd had more difficulty collecting stem cells and often needed extra help with drugs like plerixafor or a second round of mobilization.
- Although DVTd was better than VRd in many ways, it still collected fewer stem cells than VTd.
- Time to blood count recovery after transplant was slightly slower in the VRd group, but similar across all groups overall.
In the VRd combination, Revlimid (lenalidomide) can make it harder to collect stem cells, so it’s important to plan for early stem cell collection when this drug is used. Other combinations, like VTd, may make stem cell collection easier.
"A phase 2 randomized study of modakafusp alfa as a single agent in patients with relapsed/refractory multiple myeloma"
Source
Sarah A Holstein, Shebli Atrash, Hira Mian, Meletios Athanasios Dimopoulos, Fredrik Schjesvold, Rakesh Popat, Nishi Shah, Moshe E Gatt, Christian B. Gocke, Laurent Frenzel, Cyrille Touzeau, Meral Beksac, Salomon Manier, Hila Magen, Patrick Travis, Omar Nadeem, Kaveri Suryanarayan, Cheryl Li, Shuli Li, Allison Nelson, Dasha Cherepanov, Xavier Parot, Dan T Vogl; A phase 2 randomized study of modakafusp alfa as a single agent in patients with relapsed/refractory multiple myeloma. Blood 2025; blood.2024027873. doi: https://doi.org/10.1182/blood.2024027873 April 25, 2025.
Overview
Modakafusp alfa is an experimental drug that helps the immune system fight multiple myeloma by delivering interferon directly to myeloma cells. In this study, researchers tested two doses of the drug in people whose myeloma had come back or stopped responding to treatment after at least three prior therapies.
- 147 patients were divided into two groups:
- One group received 120 mg of modakafusp alfa every 4 weeks.
- The other group received 240 mg every 4 weeks.
Most patients had already been treated with several other myeloma drugs, and nearly half had received anti-BCMA therapy. The study was stopped early for business reasons, not because of safety concerns.
Here are key findings of this study:
- The overall response rate (ORR) was 32% for the lower dose and 41% for the higher dose.
- People who had not been treated with anti-BCMA therapy responded better (46%) than those who had (29%).
- The most common side effects were low platelet and white blood cell counts, which were often severe (grade 3 or higher).
- Serious side effects occurred in about 4 out of 10 patients in both groups.
Even though the study ended early, results show that modakafusp alfa may help some patients with heavily pretreated myeloma. More research is needed to fully understand its benefits and risks.
"MLN0905, a new inhibitor of Polo-like kinase 1 (PLK1), enhances the efficiency of lenalidomide in promoting the apoptosis of multiple myeloma cell lines"
Source
Piri, R., Shahidi, M., Pooraskari, Z. et al. MLN0905, a new inhibitor of Polo-like kinase 1 (PLK1), enhances the efficiency of lenalidomide in promoting the apoptosis of multiple myeloma cell lines. Invest New Drugs (2025). https://doi.org/10.1007/s10637-025-01531-w April 25, 2025.
Overview
Researchers are exploring new ways to treat multiple myeloma, especially when the cancer becomes resistant to standard treatments. This lab study looked at a drug called MLN0905, which blocks a protein called Polo-like kinase 1 that helps cancer cells grow. They tested MLN0905 alone and with Revlimid (lenalidomide) in a type of myeloma cell grown in the lab.
Here are the study's key findings:
- Both drugs reduced the number of myeloma cells in a dose-dependent way—the higher the dose, the stronger the effect.
- When the drugs were used together, they killed more cancer cells than when used alone.
- The combo worked best at 40 μM lenalidomide and 50 nM MLN0905, causing over 50% of the cells to die.
- The drugs also changed the activity of certain genes that control cell survival and death, including BCL2, p21, and PUMA.
The combination of MLN0905 and Revlimid (lenalidomide) may work better than either drug alone at stopping myeloma cells from growing and helping them die. While this was an early-stage lab study, it shows potential for new treatment strategies in the future.
"Role of NK Cells in Progression and Treatment of Multiple Myeloma"
Source
Iman Chanchiri, Emil Birch Christensen, Niels Abildgaard, Torben Barington, Thomas Lund, Jakub Krejcik. Role of NK Cells in Progression and Treatment of Multiple Myeloma. Front. Biosci. (Landmark Ed) 2025, 30(4), 26205. https://doi.org/10.31083/FBL26205 April 25, 2025.
Overview
While newer therapies have improved survival for multiple myeloma (MM) patients, the disease is still considered incurable. One reason it keeps returning is that the immune system, which normally helps fight cancer, doesn't work as well in people with myeloma.
Most of today’s immune-based treatments focus on T cells, but scientists are now looking closely at another type of immune cell called natural killer (NK) cells. NK cells are experts at identifying and destroying cancer cells without needing prior training, like built-in cancer fighters.
This review highlights the growing interest in using NK cells as a new way to treat myeloma, especially when other therapies stop working. It explains how NK cells can support or even enhance current treatments and discusses exciting new research aimed at boosting their power to fight cancer.
NK cell-based therapies could open up new treatment options and help improve long-term outcomes for people living with multiple myeloma
"Recombinant human holo-lactoferrin in complex with oleic acid suppresses the growth of solid myeloma more efficiently than its apo-form"
Source
Dr. Anna Yu Elizarova, Dr. Alexey V. Sokolov, Dr. Valeria A. Kostevich, Dr. Nikolay P. Gorbunov, Dr. Igor V. Kudryavtsev, Dr. Yulia M Berson, Dr. Valery N Yermalitsky, Dr. Alexander Budevich, and Prof. Vadim B. Vasilyev. Recombinant human holo-lactoferrin in complex with oleic acid suppresses the growth of solid myeloma more efficiently than its apo-form. Biochemistry and Cell Biology. Just-IN https://doi.org/10.1139/bcb-2024-0159 April 26, 2025.
Overview
Researchers are studying a new treatment made from two natural substances: lactoferrin, a protein found in the body, and oleic acid, a type of healthy fat. When these are combined in a specific way—especially using iron-rich lactoferrin (called holo-lactoferrin)—they seem to work together to help stop tumor growth.
In this study, mice with solid myeloma tumors were treated for 10 days with this new combination. After 15 days, the tumors were 93% smaller than those in untreated mice. The treatment worked much better than either ingredient on its own.
While this research is still in the early stages and was only done in animals, it shows that combining holo-lactoferrin with oleic acid may one day help in the fight against multiple myeloma.
"A thematic analysis of prognostic, diagnostic, and therapeutic of circulating miRNA biomarkers in bortezomib-resistant multiple myeloma"
Source
Bamfield-Cummings S, Silva J, Karim ZA. A thematic analysis of prognostic, diagnostic, and therapeutic of circulating miRNA biomarkers in bortezomib-resistant multiple myeloma. SAGE Open Medicine. 2025;13. doi:10.1177/20503121251328486 April 26, 2025.
Overview
Researchers are learning that miRNAs—tiny molecules found in the blood—could become powerful tools in managing multiple myeloma, especially for patients whose disease no longer responds to Velcade (bortezomib).
This review looked at 20 studies published between 2014 and 2024. The studies found that certain circulating miRNAs could help:
- Diagnose multiple myeloma earlier,
- Predict how the disease might progress, and
- Guide treatment, especially when standard drugs stop working.
Because miRNAs are stable in the blood and easy to detect, they may offer a noninvasive way to monitor the disease over time. This could lead to better outcomes and improved quality of life for patients.
"Second Primary Malignancies Post Autologous Stem-cell Transplant in Multiple Myeloma from India – Knowing the Unknown"
Source
Mirgh, S., Surendran, A., Punatar, S. et al. Second Primary Malignancies Post Autologous Stem-cell Transplant in Multiple Myeloma from India – Knowing the Unknown. Indian J Hematol Blood Transfus (2025). https://doi.org/10.1007/s12288-025-01984-2 April 26, 2025.
Overview
This study looked at how often patients with multiple myeloma develop a second cancer after receiving an autologous stem cell transplant (ASCT). Researchers followed 178 patients who had a transplant between 2007 and 2022.
Here are the study's key findings:
- About 3% of patients developed a second cancer (called a second primary malignancy or SPM), often years after their transplant.
- These included both solid tumors (like those in organs) and blood cancers.
- The second cancers appeared about 8 to 9 years after diagnosis and transplant.
- Many of the patients had been treated with Revlimid (lenalidomide) for a few years before developing the second cancer.
- Half of the patients with a second cancer were still alive at the time of the report.
While second cancers after ASCT are rare, they can happen even many years later. That’s why lifelong follow-up is important for myeloma patients after a stem cell transplant.
"Changes in sagittal spinopelvic alignment are associated with increased pain and impaired function in multiple myeloma patients"
Source
Kylies, J., Dirks, M., Brauneck, E. et al. Changes in sagittal spinopelvic alignment are associated with increased pain and impaired function in multiple myeloma patients. Eur Spine J (2025). https://doi.org/10.1007/s00586-025-08878-x April 26, 2025. .
Overview
In multiple myeloma, it's common for a patient's bones in the spine to be affected. This study looked at 86 patients over several years to see how these changes affect pain and movement.
Here’s what the researchers found:
- Over time, the curve in the upper back (called thoracic kyphosis) became more pronounced.
- This change in spine shape was linked to more back pain and higher use of pain medications.
- Patients whose spine curved more than 9 extra degrees had the worst pain scores.
- Surprisingly, new spine fractures didn’t have as much effect on pain as the overall change in spine shape.
As multiple myeloma progresses, spinal alignment—especially increasing upper-back curvature—can lead to more pain. This study suggests that treating or correcting this spine curve may help manage pain better in some patients.
"Managing Toxicities of Antibody-Drug Conjugates, Bispecific Antibodies, and CAR T-Cell Therapies"
Source
Michael D. Jain et al. Easy as ABC: Managing Toxicities of Antibody-Drug Conjugates, Bispecific Antibodies, and CAR T-Cell Therapies. Am Soc Clin Oncol Educ Book 45, e473916(2025). DOI:10.1200/EDBK-25-473916. April 28, 2025.
Overview
Antibody-drug conjugates (ADCs), bispecific T-cell engagers (BsAbs), and CAR T-cell therapies are becoming more common in treating multiple myeloma and other cancers. These immunotherapies are designed to help your immune system find and attack cancer cells more effectively.
- These therapies are very powerful and targeted, which helps make them effective.
- But they also come with unique side effects that are different from standard chemotherapy or older immunotherapies.
- Doctors are learning more every day about how to recognize and manage these side effects to keep patients safe.
As these treatments become more widely used, both doctors and patients will need to be aware of their benefits and risks. Learning about the “ABCs” of these new therapies can help you make informed decisions about your care
"Revised free light chain reference intervals enhance risk stratification in monoclonal gammopathy of undetermined significance and reduce overdiagnosis"
Source
Maeng, C.V., Rögnvaldsson, S., Einarsson Long, T. et al. Revised free light chain reference intervals enhance risk stratification in monoclonal gammopathy of undetermined significance and reduce overdiagnosis. Blood Cancer J. 15, 80 (2025). https://doi.org/10.1038/s41408-025-01289-7 April 28, 2025.
Overview
Doctors use a lab test called the free light chain (FLC) ratio to help estimate a person’s risk of developing multiple myeloma from monoclonal gammopathy of undetermined significance (MGUS). Recently, researchers created new FLC reference ranges that take into account a person’s age and kidney function. These updated ranges help reduce the number of people who are wrongly labeled as high-risk.
In this study of nearly 7,000 people with MGUS:
- About 1 in 3 people who were previously told they had an abnormal FLC ratio were reclassified as normal using the new guidelines.
- People in this reclassified group had no higher risk of developing myeloma than those who always had normal results.
- People still classified as abnormal with the new ranges did have a higher risk of progression.
Thanks to these new guidelines, doctors can more accurately tell who is truly at risk—and avoid unnecessary worry or treatment for those who are not.