The field of multiple myeloma (MM) treatment has seen significant advancements in recent years, which have improved median survival rates for patients from 3 years two decades ago to around 10 years now. One of the most notable advancements in the treatment of MM is the introduction of immune-based therapies, specifically anti-CD38 monoclonal antibodies. These therapies have had a profound impact on the field and have caused significant shifts in the approach to MM treatment.

Building upon the success of immunotherapy, newer immunological treatments such as CAR-T cells and bispecific antibodies are currently being developed. These novel therapies hold great promise for improving patient outcomes. However, it is important to acknowledge that, despite these remarkable advancements, MM remains an incurable disease. This highlights the need for continuous research seeking new and diversified treatment options.

As new mechanisms are unraveled, new targets are being identified, leading to the development of new drugs and drug classes that hold great hope for the future of MM treatments. This article aims to discuss the top 7 latest advancements in MM treatment, including updates from the 2023 American Society of Clinical Oncology (ASCO) annual meeting. Our goal is to provide healthcare professionals with the most up-to-date information for optimal patient outcomes. This article provides a detailed overview of the following items: 

  1. Recent breakthroughs in therapy targeting CD38. 
  2. Latest developments in CAR T-cell therapies. 
  3. Latest insights on bispecific T-cell engagers.
  4. Exciting research findings on antibody–drug conjugates. 
  5. Exploring the potential of cereblon E3 ligase modulators in treating resistant MM. 
  6. Advancements in venetoclax (anti-BCL-2) and selinexor (anti-XPO1) therapies. 
  7. Updated recommendations for long-term maintenance therapy.

Immunotherapies have indeed transformed the field of MM treatment, bringing significant benefits to patients of all ages and disease stages. The introduction of elotuzumab, an anti-CS1/SLAMF7 therapy, marked the initial successful use of immunotherapy in the treatment of MM. However, it is the use of anti-CD38 monoclonal antibodies (mAbs) that has actually revolutionized the treatment of MM (1). These breakthroughs have been followed by the development of innovative immunotherapies such as CAR-T cells and bispecific antibodies (BsAbs), which have shown remarkable effectiveness and fewer side effects. Ongoing research is actively exploring new immune-based options like antibody–drug conjugates (ADCs), cereblon E3 ligase modulators (CELMoDs), and immune fusion proteins. Here, we explore the latest advancements in the field of MM immunotherapy made in the last few years. So let’s delve in.

Recent Breakthroughs in Therapy Targeting CD38

CD38 is a glycoprotein found on the surface of cells that regulates calcium signaling and immune cell migration to the tumor microenvironment. It is highly expressed on MM cells, making it an attractive target for MM therapy. Several anti-CD38 monoclonal antibodies (mAbs) are now widely used in MM treatment, including daratumumab (D), isatuximab, MOR2021, and TAK-0792 (1).

 

CD38 is a glycoprotein found on the surface of cells that regulates calcium signaling and immune cell migration to the tumor microenvironment

Newly Diagnosed Multiple Myeloma

In the treatment of newly diagnosed MM (NDMM), several clinical trials have shown promising results with the use of anti-CD38 mAbs. The phase III CASSIOPEIA trial demonstrated that combining daratumumab with bortezomib, thalidomide, and dexamethasone (D-VTd) resulted in better response depth and progression-free survival (PFS) compared to bortezomib, thalidomide, and dexamethasone alone in transplant-eligible patients (2). The phase II GRIFFIN trial also showed promising results with daratumumab combined with bortezomib, lenalidomide, and dexamethasone (D-VRd), leading to a high stringent complete response rate (3). The phase III PERSEUS study (ClinicalTrials.gov Identifier: NCT03710603) will further investigate D-VRd using lenalidomide instead of thalidomide.

Isatuximab is being studied in two phase III trials, isa-VRd  (NCT0367731) and isa-KRd  (NCT04483739), both for transplant-eligible NDMM. In non-transplant-eligible NDMM patients, the phase III MAIA trial demonstrated impressive median PFS with daratumumab combined with lenalidomide and dexamethasone (DRd). Another option for non-transplant-eligible NDMM is the combination of daratumumab with VMP (melphalan and prednisone), which showed a median PFS of 36.4 months in the ALCYONE trial (4). Isatuximab is also being investigated in the IMROZ trial, with results still expected, and in the ongoing BENEFIT/IFM 2020-05 trial (NCT04751877) with the objective of improving minimal residual disease rates.

Overall, anti-CD38 mAbs are making their way into frontline therapy for MM. The combination of anti-CD38 mAbs with VRd for non-transplant-eligible NDMM is being studied in the CEPHEUS trial, with results yet to be reported. These drugs offer new hope for patients with MM and continue to be studied in clinical trials to improve outcomes.

  • Latest Developments in CAR T-Cell Therapies 

CAR-T cell therapy is a form of immunotherapy that has revolutionized the treatment of blood cancers. This therapy involves genetically modifying T cells to express a chimeric antigen receptor (CAR) that targets tumor antigens. Two advanced CAR-T cell therapies, idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel), target B-cell maturation antigen (BCMA) found on malignant plasma cells.

Clinical Trials

In the phase II KARMMA-1 trial, ide-cel demonstrated an overall response rate (ORR) of 73%, with 33% of patients achieving a complete response (CR). The median progression-free survival (PFS) was 8.6 months, and the median overall survival (OS) was 24.8 months. Cilta-cel was evaluated in the CARTITUDE-1 study, which showed an ORR of 97.9%, with 80.4% of patients achieving a CR. The 18-month PFS was 66%.

Side Effects

The main side effect of CAR-T cell therapy is cytokine release syndrome (CRS), which occurs shortly after infusion of the CAR-T cells. The incidence of CRS was 84% in KARMMA-1 and 95% in CARTITUDE-1, mostly mild to moderate in severity. Another potential side effect is neurotoxicity, known as immune effector cell-associated neurotoxicity syndrome (ICANS). Its occurrence was 18% for ide-cel and 21% for cilta-cel.

Recent Updates

Recent data presented at ASCO 2023 provided updates on cilta-cel and ide-cel. In the phase III CARTITUDE-4 study (NCT04181827), cilta-cel was compared to standard treatments in patients with relapsed/refractory multiple myeloma (RRMM). The results showed that cilta-cel reduced the risk of disease progression by 74% compared to standard treatments. Additionally, 85% of patients responded to cilta-cel, and 73% achieved a complete response or better.

In the KARMMA-3 trial (NCT03651128), patient-reported outcomes on health-related quality of life were assessed in patients with RRMM who received ide-cel. The results showed significant improvements in cognitive functioning, fatigue, and pain reduction compared to standard treatment regimens.

Overall, these findings highlight the efficacy of both cilta-cel and ide-cel in treating RRMM. Cilta-cel showed superior results compared to standard treatments, indicating its potential as a new treatment option for patients with relapsed/refractory disease. Additionally, ide-cel demonstrated improvements in health-related quality of life, further supporting its benefit in this patient population.

PHE885: A Novel CAR T-Cell Therapy for Multiple Myeloma

PHE885 is a novel CAR T-cell therapy being developed for the treatment of B-cell maturation antigen (BCMA)-positive MM. This therapy has a shortened manufacturing time of less than two days, compared to several weeks for other CAR T-cell therapies.

You May Also Like::  Exploring the Urban–Rural Divide in Non-Hodgkin Lymphoma Mortality

Phase 1 Study Results

A phase 1 study conducted by Adam Sperling and colleagues at Dana-Farber Cancer Institute evaluated the safety and efficacy of PHE885. The updated results showed promising outcomes, with an overall response rate (ORR) of 98% among the 49 patients enrolled in the study. Cytokine release syndrome (CRS), a common side effect of CAR T-cell therapy, was observed in 96% of patients (5).

Ongoing Studies

Based on these positive results, a phase 2 study is currently underway to further evaluate the efficacy and safety of PHE885 in patients with relapsed/refractory multiple myeloma (RRMM). Additionally, the developers are planning to assess the therapy in earlier lines of therapy, potentially expanding its use to a broader patient population.

Future Developments

In the field of CAR T-cell therapy, there are ongoing advancements and developments. One avenue for improvement is the development of allogeneic CAR cells, which would allow for “off-the-shelf” availability. Another approach is to select more active T cells, potentially enhancing the therapeutic response. Additionally, the exploration of CAR natural killer (CAR-NK) cells could provide alternative cell sources for CAR therapy.

Overall, PHE885 has shown promising results in targeting BCMA-positive MM. Further studies are ongoing to establish its efficacy in treating RRMM, and future developments in the CAR T-cell therapy field hold potential for enhancing treatment outcomes and expanding its use to broader patient populations.

  • Latest Insights on Bispecific T-Cell Engagers 

Bispecific antibodies (BsAbs) are a type of antibody-based therapy that can link T cells and target cells to induce T-cell activation and cell lysis. Unlike CAR-T cell therapies, BsAbs do not require apheresis of lymphocytes from the patient and can be used “off-the-shelf”, allowing for faster delivery. The safety profiles of CAR-T cells and BsAbs are similar, with both therapies potentially causing immune reactions such as cytokine release syndrome (CRS) and neurotoxicity, although to a lesser extent overall. In terms of efficacy, BsAbs have shown promise in MM, but direct comparisons with CAR-T cell therapies are not available.

Types of BsAb Constructs

There are two types of BsAb constructs: bispecific T-cell engagers (BiTes), which lack the Fc fragment and have a short half-life requiring continuous administration, and “IgG-like” molecules with an Fc fragment, which can be administered intermittently. The first BsAbs targeting BCMA, similar to CAR-T cell therapies, are teclistamab and elranatamab.

Clinical Trial Results

 

In a phase I dose escalation study of teclistamab, an ORR of 65% was observed at the recommended phase 2 dose, with 40% of patients achieving at least a complete response (CR).

 

In a phase I dose escalation study of teclistamab, an ORR of 65% was observed at the recommended phase 2 dose, with 40% of patients achieving at least a complete response (CR). The most common adverse events were CRS and neutropenia. Elranatamab also showed promising results with an ORR of 83% in the phase 1 MagnetisMM-1 study, and a CRS of not more than grade 2 was observed in 83% of patients.

In addition to BCMA, BsAbs are being developed to target other antigens expressed by plasma cells (PCs), such as GPRC5D (talquetamab) and FcRH5 (cevostamab/BFCR4350A), as well as CD38 (GBR1342). In the first-in-human study of talquetamab, an ORR of 70% was observed at the recommended phase 2 dose. Cevostamab also showed activity in relapsed/refractory MM, with an ORR of 54.5% in the expansion cohort of the phase I study (6). These BsAbs have shown potential for rescuing progressive disease seen in patients treated with anti-BCMA BsAbs.

Tecvayli: The First Approved Off-the-Shelf BCMA-Directed BsAb

Tecvayli is the first approved off-the-shelf BCMA-directed BsAb for the treatment of patients with relapsed/refractory MM. The approval was based on data from the MajesTEC-1 study, which reported an ORR of 63% in patients treated with Tecvayli. In follow-up, it was observed that 39.4% of patients achieved a complete response or better, with a median duration of response of 18.4 months. The median progression-free survival was 11.3 months, and the median overall survival was not estimable. Key side effects included low white blood cell counts, CRS, respiratory infections, and low red blood cell counts. These long-term follow-up data support Tecvayli as a safe and effective off-the-shelf BCMA bispecific therapy for patients with relapsed/refractory MM.

Combination Therapies with Tecvayli and Talquetamab

In the RedirecTT-1 trial, Dr. Yael Cohen and researchers from Israel investigated the combination of Tecvayli and talquetamab, two bispecific antibodies targeting different immunotherapeutic targets (7). The trial results showed an overall response rate (ORR) of 84% across all dose levels of Tecvayli and talquetamab. Patients with extramedullary disease also responded well to the therapy, with an ORR of 86%. The most common side effect observed was cytokine release syndrome (CRS), affecting 81% of patients (7).

Combination Therapy with Talquetamab and Darzalex

In the TRiMM-2 study, Dr. Bhagirathbhai Dholaria and colleagues from Vanderbilt University Medical Center investigated the combined use of talquetamab and Darzalex in heavily pretreated patients with relapsed or refractory MM (RRMM) (8). The study results showed an ORR of 84% for patients receiving the higher dose of talquetamab and 71% for those receiving the lower dose (8).

Overall, combination therapies with bispecific antibodies have shown promising response rates in patients with RRMM. Combining Tecvayli and talquetamab or talquetamab and Darzalex demonstrated high response rates in heavily pretreated patients. Ongoing research will further investigate the potential of these therapies for the treatment of MM.

Talquetamab Monotherapy

The MonumenTAL-1 study, led by Dr. Carolina Schinke from the University of Arkansas for Medical Sciences, evaluated the efficacy of talquetamab as a monotherapy in patients with RRMM (9). The results showed that 73% of patients treated with the biweekly dose and 74% of patients treated with the weekly dose responded to the treatment (9). Notably, 65% of patients who had previously undergone T-cell redirection therapies, such as CAR T-cell or bispecific antibody therapy, responded to talquetamab.

Elranatamab Monotherapy

Elranatamab is an investigational bispecific antibody targeting BCMA and is being studied as a single-agent therapy for patients with relapsed or refractory multiple myeloma (RRMM). In the MagnetisMM-3 study, 61% of patients who hadn’t previously received a BCMA-directed therapy responded to elranatamab monotherapy (10). The most common side effects observed were cytokine release syndrome (CRS) in 65.1% of patients and low red blood cell counts in 59.3% of patients (10).

Linvoseltamab Monotherapy

Linvoseltamab is another investigational bispecific antibody targeting BCMA, intended for patients with RRMM who have received three or more prior treatments. In the phase 2 LINKER-MM1 trial, an overall response rate of 71% was observed for patients receiving the 200 mg dose and 50% for those receiving the 50 mg dose of linvoseltamab (11). The most common side effect observed in patients receiving either dosage was cytokine release syndrome.

Overall, elranatamab and linvoseltamab are both promising investigational bispecific antibodies targeting BCMA for the treatment of RRMM. Elranatamab has shown effectiveness as a monotherapy, particularly in patients who have not been exposed to prior BCMA-directed therapies. Furthermore, linvoseltamab demonstrated notable response rates in patients who had received multiple prior treatments. Both therapies were associated with cytokine release syndrome as the most common side effect. Further research, including phase 3 trials, will continue to evaluate their therapeutic potential. Additionally, talquetamab monotherapy showed positive response rates in patients with RRMM, including those previously treated with T-cell redirection therapies. 

  • Exciting Research Findings on Antibody–Drug Conjugates 

Belantamab mafodotin (BM) is an anti-BCMA antibody–drug conjugate (ADC) and the first of its kind. ADCs are a type of drug that consists of a monoclonal antibody (mAb) linked to a cytotoxic agent, delivering the cytotoxic payload directly to targeted cells and reducing off-target side effects. In the case of BM, the mAb is coupled with monomethyl auristatin F (MMAF), a cytotoxic drug, via a protease-resistant linker.

You May Also Like::  Soluble PD‐L1 and Classic Hodgkin Lymphoma

Clinical Trial Results

BM shows promise as a treatment option for patients with relapsed or refractory multiple myeloma (RRMM) who have been previously treated with all available anti-myeloma drug classes. In the phase II DREAMM-2 study, the overall response rate (ORR) with BM as a single agent was 31%, and the median progression-free survival (PFS) was 2.9 months. However, an extended follow-up of 13 months showed that responding patients who achieved a very good partial response had an estimated median PFS of 14 months (13).

Safety Concerns

Safety concerns associated with BM primarily include the frequent occurrence of corneal keratopathy and hematological toxicities, such as thrombocytopenia. Given the risk of corneal issues, regular and careful ophthalmological follow-up is necessary for patients receiving BM treatment.

Ongoing Studies

Ongoing or planned studies, such as DREAMM-3, DREAM-8, and DREAMM-9, aim to further investigate the efficacy and safety of belantamab mafodotin for RRMM.

Overall, belantamab mafodotin is a promising antibody-drug conjugate for the treatment of RRMM. The phase II DREAMM-2 study demonstrated its efficacy as a single agent in patients who have been previously treated with all available anti-myeloma drug classes. Safety concerns associated with BM primarily include corneal keratopathy and hematological toxicities. Ongoing research will continue to evaluate its therapeutic potential.

  • Exploring the Potential of Cereblon E3 Ligase Modulators in Treating Resistant Multiple Myeloma 

CELMoDs (cereblon E3 ubiquitin ligase modulators) are a newly developed class of drugs derived from IMiDs. Similar to IMiDs, they bind to the cereblon E3-ubiquitin ligase complex, but with a higher affinity for cereblon. This leads to an increased degradation of IKZF1 and IKZF3, which are transcription factors crucial for the survival of multiple myeloma (MM) cells. Iberdomide (CC-220) is one of the most advanced CELMoDs currently being studied.

Clinical Trial Results

The first-in-human multicohort phase I/II trial (CC-220-MM-001) evaluated iberdomide for the treatment of relapsed or refractory MM. In the dose expansion phase of the study, where iberdomide was combined with dexamethasone (dex), the overall response rate (ORR) was 26.2%, and the median progression-free survival (PFS) was 3 months. Additionally, when iberdomide was used in combination with other drugs, the ORR was 56% with iberdomide and bortezomib-dexamethasone (iber-Vd) and 45.9% with iberdomide and daratumumab-dexamethasone (iber-Dd) (1).

Safety Concerns

CELMoDs have a safety profile characterized mainly by hematological adverse events (AEs), particularly myelosuppression, which is consistent with the known safety profile of IMiDs. In the CC-220-MM-001 study, 82.2% of patients experienced grade 3 or 4 AEs, including neutropenia (44.9%), anemia (28%), thrombocytopenia (21.5%), and leukopenia (20.6%). Other AEs observed were gastrointestinal disorders (5.6%), fatigue (2.8%), and rash (1.9%).

Ongoing Studies

Ongoing or planned studies, such as DREAMM-3, DREAM-8, and DREAMM-9, aim to further investigate the efficacy and safety of belantamab mafodotin for RRMM.

Overall, CELMoDs are a new class of drugs derived from IMiDs that show promise in the treatment of MM. Iberdomide is one of the most advanced CELMoDs currently being studied and has demonstrated efficacy in combination with other drugs in clinical trials. CELMoDs have a safety profile characterized mainly by hematological adverse events, particularly myelosuppression. Ongoing research will continue to evaluate their therapeutic potential.

  • Advancements in Venetoclax (anti-BCL-2) and Selinexor (anti-XPO1) Therapies 

Venetoclax and selinexor are two novel agents that have emerged in recent years for the treatment of MM. Venetoclax is an anti-BCL-2 inhibitor that specifically targets the subgroup of patients with the t(11;14) translocation, while selinexor is an anti-XPO1 inhibitor that provides an alternative option for patients with relapsed/refractory MM (RRMM) who have already received all traditional MM drugs.

Venetoclax

Venetoclax is an oral inhibitor of BCL-2, a protein that promotes cell survival in MM. In the phase III BELLINI trial, the combination of venetoclax with bortezomib and dexamethasone (Ven-Vd) showed a median progression-free survival (PFS) of 23.4 months, compared to 11.4 months in the Vd group (14). However, the trial showed a higher risk of mortality in the venetoclax arm, mostly due to infections. Patients with the t(11;14) translocation had the highest response rates and longest PFS with Ven-Vd.

Selinexor

Selinexor is a selective inhibitor of nuclear export that targets exportin-1 (XPO1). In the phase IIb STORM trial, selinexor was combined with dexamethasone and evaluated in heavily pretreated patients with RRMM. The overall response rate (ORR) was 26%, median PFS was 3.7 months, and median overall survival (OS) was 8.6 months. Selinexor-based triplets are also being studied in RRMM patients.

Safety Concerns

There are concerns about the safety profile of selinexor. A retrospective pooled analysis of selinexor trials revealed high rates of non-hematological adverse events such as nausea, decreased appetite, diarrhea, and vomiting. Hematological events like thrombocytopenia and neutropenia are also common but can be dose-dependent.

Overall, venetoclax and selinexor are two novel agents that offer potential benefits for the treatment of MM. Venetoclax shows promise for patients with the t(11;14) translocation and has demonstrated improved response rates and longer PFS in this subgroup. Selinexor provides a novel therapeutic option with a different mechanism of action, although its safety profile requires careful management. Ongoing trials are further exploring the effectiveness and safety of these agents in the treatment of MM.

  • Updated Recommendations for Long-Term Maintenance Therapy 

Maintenance therapy with Revlimid is currently the standard of care for patients with standard-risk myeloma after induction therapy and autologous stem cell transplantation (ASCT). However, for patients with high-risk myeloma, current data suggest the use of a proteasome inhibitor such as Velcade, either alone or in combination with Revlimid, or a triplet regimen like Velcade, Revlimid, and dexamethasone (VRd).

 

Maintenance therapy with Revlimid is currently the standard of care for patients with standard-risk myeloma after induction therapy and autologous stem cell transplantation (ASCT).

KPd Maintenance Therapy

A phase 2 trial conducted by Dr. Ajay Nooka and colleagues from Emory University evaluated the efficacy of a maintenance therapy called KPd in patients with high-risk myeloma (15). The results of the trial showed that 89.7% of patients achieved a complete response (CR) or better with KPd maintenance therapy. The 36-month progression-free survival (PFS) rate was 63% and the 36-month overall survival (OS) rate was 72% in patients receiving KPd.

Safety Concerns

The most common side effects observed were fever, fatigue, and diarrhea (15). These findings suggest that KPd maintenance therapy deepened responses in patients with high-risk myeloma and may address an unmet treatment need in this specific patient population.

Overall, the phase 2 trial evaluating KPd maintenance therapy in patients with high-risk myeloma demonstrated promising results, with a high percentage of patients achieving a CR or better. The 36-month PFS and OS rates further support the potential benefit of KPd in this patient population. In addition, the trial included a significant number of Black patients, highlighting the importance of considering racial diversity in clinical studies.

You May Also Like::  Recommendations for Addressing Race-Based Disparities in Breast Cancer Screening

Conclusion

The future of MM treatment looks promising with the emergence of new therapies year after year. Immunological agents have revolutionized the field with their effectiveness and relatively low toxicity profiles. Novel immunotherapies like CAR-T cells and BsAbs show immense potential for heavily pretreated patients at the time of relapse.

The field of MM treatment continues to evolve, driven by advancements in immunological and non-immune-based therapies. The combination of these therapies, along with a better understanding of MM pathophysiology, holds great promise for improving patient outcomes and ultimately leading to a cure. However, it is important to note that a complete cure has not yet been achieved with immunotherapies alone, and the development of non-immunological drugs remains crucial for addressing relapses and expanding treatment options.

References

  1. Bobin, A., & Leleu, X. (2022). Recent advances in the treatment of multiple myeloma: a brief review. Faculty reviews, 11, 28. https://doi.org/10.12703/r/11-28
  2. Moreau, P., Attal, M., Hulin, C., Arnulf, B., Belhadj, K., Benboubker, L., Béné, M. C., Broijl, A., Caillon, H., Caillot, D., Corre, J., Delforge, M., Dejoie, T., Doyen, C., Facon, T., Sonntag, C., Fontan, J., Garderet, L., Jie, K. G., . . . Sonneveld, P. (2019). Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. The Lancet, 394(10192), 29–38. https://doi.org/10.1016/s0140-6736(19)31240-1
  3. Voorhees, P. M., Rodriguez, C., Reeves, B., Nathwani, N., Costa, L. J., Lutska, Y., Bobba, P., Hoehn, D., Pei, H., Ukropec, J., Qi, M., Lin, T. S., & Richardson, P. G. (2021). Daratumumab plus RVd for newly diagnosed multiple myeloma: final analysis of the safety run-in cohort of GRIFFIN. Blood Advances, 5(4), 1092–1096. https://doi.org/10.1182/bloodadvances.2020003642
  4. Mateos, M. V., Cavo, M., Bladé, J., Dimopoulos, M. A., Suzuki, K., Jakubowiak, A., Knop, S., Doyen, C., Lucio, P., Nagy, Z., Pour, L., Cook, M., Grosicki, S., Crepaldi, A. H., Liberati, A. M., Campbell, P., Shelekhova, T., Yoon, S. S., Iosava, G., . . . Miguel, J. F. S. (2020). Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): a randomised, open-label, phase 3 trial. The Lancet, 395(10218), 132–141. https://doi.org/10.1016/s0140-6736(19)32956-3
  5. Sperling, A. S., Derman, B. A., Nikiforow, S., Im, S., Ikegawa, S., Prabhala, R., Rodriguez, D. H., Li, Y., Quinn, D. S., Pearson, D. E., Bu, D., Mataraza, J., Liegel, J., D’Souza, A., Rispoli, L., Credi, M., Ritz, J., Jakubowiak, A., De Vita, S., & Munshi, N. C. (2023). Updated phase I study results of PHE885, a T-Charge manufactured BCMA-directed CAR-T cell therapy, for patients (pts) with r/r multiple myeloma (RRMM). Journal of Clinical Oncology, 41(16_suppl), 8004. https://doi.org/10.1200/jco.2023.41.16_suppl.8004
  6. Trudel, S., Cohen, A. D., Krishnan, A., Fonseca, R., Spencer, A., Berdeja, J. G., Lesokhin, A. M., Forsberg, P., Laubach, J. P., Costa, L. J., Rodriguez-Otero, P., Kaedbey, R., Richter, J., Mateos, M., Thomas, S. K., Wong, C., Li, M., Choeurng, V., Vaze, A., . . . Harrison, S. (2021). Cevostamab Monotherapy Continues to Show Clinically Meaningful Activity and Manageable Safety in Patients with Heavily Pre-Treated Relapsed/Refractory Multiple Myeloma (RRMM): Updated Results from an Ongoing Phase I Study. Blood, 138(Supplement 1), 157. https://doi.org/10.1182/blood-2021-147983
  7. Cohen, Y., Morillo, D., Gatt, M. E., Sebag, M., Kim, K., Min, C., Oriol, A., Ocio, E. M., Yoon, S. S., Mateos, M., Chu, M., Rodriguez-Otero, P., Avivi, I., Guo, Y., Krevvata, M., Peterson, M. R., Beelen, M. J., Vanák, J., Banerjee, A., & Magen, H. (2023). First results from the RedirecTT-1 study with teclistamab (tec) + talquetamab (tal) simultaneously targeting BCMA and GPRC5D in patients (pts) with relapsed/refractory multiple myeloma (RRMM). Journal of Clinical Oncology, 41(16_suppl), 8002. https://doi.org/10.1200/jco.2023.41.16_suppl.8002
  8. Dholaria, B., Weisel, K., Mateos, M., Goldschmidt, H., Martin, T., Morillo, D., Reece, D., Rodriguez-Otero, P., Bhutani, M., D’Souza, A., Oriol, A., Rosiñol, L., Bahlis, N. J., Bakshi, K., Kang, L., Vandenberk, L., Smit, M. D., Wäsch, R., Van De Donk, N. W., & Chari, A. (2023). Talquetamab (tal) + daratumumab (dara) in patients (pts) with relapsed/refractory multiple myeloma (RRMM): Updated TRIMM-2 results. Journal of Clinical Oncology, 41(16_suppl), 8003. https://doi.org/10.1200/jco.2023.41.16_suppl.8003
  9. Schinke, C., Touzeau, C., Minnema, M. C., Van De Donk, N. W., Rodriguez-Otero, P., Mateos, M., Rasche, L., Ye, J. C., Vishwamitra, D., Ma, X., Qin, X., Campagna, M., Masterson, T., Hilder, B., Tolbert, J. A., Renaud, T., Goldberg, J. D., Heuck, C., & Chari, A. (2023). Pivotal phase 2 MonumenTAL-1 results of talquetamab (tal), a GPRC5DxCD3 bispecific antibody (BsAb), for relapsed/refractory multiple myeloma (RRMM). Journal of Clinical Oncology, 41(16_suppl), 8036. https://doi.org/10.1200/jco.2023.41.16_suppl.8036
  10. Mohty, M., Tomasson, M. H., Arnulf, B., Bahlis, N. J., Prince, H. M., Niesvizky, R., Rodriguez-Otero, P., Martinez-Lopez, J., Koehne, G., Jethava, Y., Gabayan, A. E., Stevens, D. A., Nooka, A. K., Raje, N., Iida, S., Leip, E., Conte, U., Czibere, A., Viqueira, A., & Lesokhin, A. M. (2023). Elranatamab, a B-cell maturation antigen (BCMA)-CD3 bispecific antibody, for patients (pts) with relapsed/refractory multiple myeloma (RRMM): Extended follow up and biweekly administration from the MagnetisMM-3 study. Journal of Clinical Oncology, 41(16_suppl), 8039. https://doi.org/10.1200/jco.2023.41.16_suppl.8039
  11. Lee, H. C., Bumma, N., Richter, J., Dhodapkar, M. V., Hoffman, J. E., Suvannasankha, A., Zonder, J. A., Shah, M., Lentzsch, S., Maly, J., Ye, J. C., Wu, K. L., DeVeaux, M., Chokshi, D., Boyapati, A., Hazra, A., Rodriguez-Lorenc, K., Kroog, G. S., Houvras, Y., & Jagannath, S. (2023). LINKER-MM1 study: Linvoseltamab (REGN5458) in patients with relapsed/refractory multiple myeloma. Journal of Clinical Oncology, 41(16_suppl), 8006. https://doi.org/10.1200/jco.2023.41.16_suppl.8006
  12. Knop, S., Stuebig, T., Kull, M., Greil, R., Steiner, N., Bassermann, F., Nogai, A., Von Lilienfeld-Toal, M., Janjetovic, S., Trautmann-Grill, K., Bittrich, M., Engelhardt, M., Hoferer, A., Theurich, S., Binder, M., Zojer, N., Duerk, H. A., Brueggemann, M., Held, S., & Einsele, H. (2023). Carfilzomib, lenalidomide, and dexamethasone (KRd) versus elotuzumab and KRd in transplant-eligible patients with newly diagnosed multiple myeloma: Post-induction response and MRD results from an open-label randomized phase 3 study. Journal of Clinical Oncology, 41(16_suppl), 8000. https://doi.org/10.1200/jco.2023.41.16_suppl.8000
  13. Lonial, S., Lee, H. C., Badros, A., Trudel, S., Nooka, A. K., Chari, A., Abdallah, A. O., Callander, N. S., Sborov, D. W., Suvannasankha, A., Weisel, K., Voorhees, P. M., Womersley, L., Baron, J., Piontek, T., Lewis, E., Opalinska, J., Gupta, I., & Cohen, A. D. (2021). Longer term outcomes with single‐agent belantamab mafodotin in patients with relapsed or refractory multiple myeloma: 13‐month follow‐up from the pivotal DREAMM‐2 study. Cancer, 127(22), 4198–4212. https://doi.org/10.1002/cncr.33809
  14. Kumar, S., Harrison, S., Cavo, M., De La Rubia, J., Popat, R., Gasparetto, C., Hungria, V., Salwender, H., Suzuki, K., Kim, I., Onishi, M., Ku, G., Pothacamury, R. K., Sehgal, V., Masud, A. A., Ross, J. A., Dobkowska, E., & Moreau, P. (2021). Final Overall Survival Results from BELLINI, a Phase 3 Study of Venetoclax or Placebo in Combination with Bortezomib and Dexamethasone in Relapsed/Refractory Multiple Myeloma. Blood, 138(Supplement 1), 84. https://doi.org/10.1182/blood-2021-145757
  15. Derman, B. A., Zonder, J. A., Reece, D., Cole, C. E., Berdeja, J. G., Stefka, A., Major, A., Kin, A., Griffith, K. A., Jasielec, J., & Jakubowiak, A. (2023). Phase 1/2 study of carfilzomib, pomalidomide, dexamethasone with and without daratumumab in relapsed multiple myeloma. Blood Advances. https://doi.org/10.1182/bloodadvances.2022008866

 

Categories