Multiple Myeloma: Targeted Therapy and Immunotherapy

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Therapy".

Deadline for manuscript submissions: closed (31 August 2021) | Viewed by 43029

Special Issue Editor


E-Mail Website
Guest Editor
Department of Haematology and Stem Cell-Transplantation, South-Pest Central Hospital-National Institute for Hematology and Infectious Diseases, 1097 Budapest, Hungary
Interests: hemopoietic stem cell; transplantation; autologous; allogeneic; haploidentical; graft versus tumor; graft versus host; consolidation therapy; maintenance therapy
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

A cure for multiple myeloma, the second most common hematologic malignancy, remains elusive. Dysregulation of the immune system is a hallmark of this disease both at onset and during progression. Immune therapy — initially in the form of maintenance interferon therapy — has been around for at least four decades. Nevertheless, a highly effective therapy— at least in part — through the rectification of the compromised immune component of the bone marrow niche has gained general acceptance in the form of proteasome inhibitors, immune-modulatory agents (IMIDs), and more recently, monoclonal antibodies.

The further development of immune-based therapies has recently increased the clinical options for relapsed and/or refractory myeloma, which not only include “bare” monoclonal antibodies (alone or in combination), but also promising antibody–drug conjugates, bispecific antibodies, and bispecific T-cell engagers. Furthermore, chimeric antigen T-cell therapy is fast gaining approval in the treatment of multiple myeloma. Inhibition of the immune checkpoint blockade in relapsed myeloma has been hindered by unexpected toxicity. If the toxic side effect problem can be solved, the substantial antimyeloma activity seen in early trials may also provide breakthroughs. It is also expected that these novel therapeutics — in the field of newly diagnosed disease — will lead to further improvements in clinical results and eventually to a cure in a substantial number of patients.

This Special Issue will highlight one of the most rapidly developing fields of hematological cancer therapy, which may also serve as a model for other areas. Basic and clinical aspects will be covered to increase our understanding of targeted and immunotherapy in myeloma to facilitate progress from the bench to the bedside.

Dr. Gabor Mikala
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Published Papers (12 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review, Other

4 pages, 198 KiB  
Editorial
Myeloma: A Lot of Progress, Still a Long Way to Go
by Gábor Mikala and Gergely Varga
Cancers 2021, 13(23), 6087; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13236087 - 02 Dec 2021
Cited by 1 | Viewed by 1395
Abstract
It was Bart Barlogie who made a clear point by stating in one of his lectures that any myeloma that is not cured will eventually turn into a resistant disease with aggressive clinical behaviour [...] Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)

Research

Jump to: Editorial, Review, Other

8 pages, 577 KiB  
Article
Salvage Autologous Stem Cell Transplantation in Daratumumab-Refractory Multiple Myeloma
by Lakshmi Yarlagadda, Sravani Gundarlapalli, Richa Parikh, Reid D. Landes, Mathew Kottarathara, Yetunde Ogunsesan, Shadiqul Hoque, Angel A. Mitma, Clyde Bailey, Kerri M. Hill, Sharmilan Thanendrarajan, Monica Graziutti, Meera Mohan, Maurizio Zangari, Frits van Rhee, Guido Tricot and Carolina Schinke
Cancers 2021, 13(16), 4019; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13164019 - 10 Aug 2021
Cited by 10 | Viewed by 2744
Abstract
Daratumumab, a CD38-targeting monoclonal antibody, has significantly improved survival rates in multiple myeloma (MM), yet patients who progress on Daratumumab have dismal clinical outcomes with an overall median of less than 10 months. While emerging novel modalities have shown promising results, the current [...] Read more.
Daratumumab, a CD38-targeting monoclonal antibody, has significantly improved survival rates in multiple myeloma (MM), yet patients who progress on Daratumumab have dismal clinical outcomes with an overall median of less than 10 months. While emerging novel modalities have shown promising results, the current study explores the use of high-dose chemotherapy followed by autologous stem cell transplantation (ASCT) in heavily pretreated Daratumumab-refractory MM patients. We retrospectively investigated the outcome of 69 consecutive patients who received upfront ASCT. The median progression-free survival (PFS) for the entire patient cohort was 7.2 months with a median overall survival (OS) of 19.3 months. For patients with ≥very good partial response (VGPR), median PFS and OS improved to 9 months and 34 months, respectively. Achievement of MRD negativity in ≥VGPR did not further improve the outcome. A better performance status, younger age, longer time interval from initial MM diagnosis/initial ASCT to salvage ASCT and low-risk GEP70 were all associated with improved PFS and OS after salvage ASCT. Our results suggest a role for salvage ASCT in selected heavily pretreated and Daratumumab-refractory patients. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

24 pages, 5624 KiB  
Article
Stromal Cells Serve Drug Resistance for Multiple Myeloma via Mitochondrial Transfer: A Study on Primary Myeloma and Stromal Cells
by Zsolt Matula, Gábor Mikala, Szilvia Lukácsi, János Matkó, Tamás Kovács, Éva Monostori, Ferenc Uher and István Vályi-Nagy
Cancers 2021, 13(14), 3461; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13143461 - 10 Jul 2021
Cited by 27 | Viewed by 3648
Abstract
Recently, it has become evident that mitochondrial transfer (MT) plays a crucial role in the acquisition of cancer drug resistance in many hematologic malignancies; however, for multiple myeloma, there is a need to generate novel data to better understand this mechanism. Here, we [...] Read more.
Recently, it has become evident that mitochondrial transfer (MT) plays a crucial role in the acquisition of cancer drug resistance in many hematologic malignancies; however, for multiple myeloma, there is a need to generate novel data to better understand this mechanism. Here, we show that primary myeloma cells (MMs) respond to an increasing concentration of chemotherapeutic drugs with an increase in the acquisition of mitochondria from autologous bone marrow stromal cells (BM-MSCs), whereupon survival and adenosine triphosphate levels of MMs increase, while the mitochondrial superoxide levels decrease in MMs. These changes are proportional to the amount of incorporated BM-MSC-derived mitochondria and to the concentration of the used drug, but seem independent from the type and mechanism of action of chemotherapeutics. In parallel, BM-MSCs also incorporate an increasing amount of MM cell-derived mitochondria accompanied by an elevation of superoxide levels. Using the therapeutic antibodies Daratumumab, Isatuximab, or Elotuzumab, no similar effect was observed regarding the MT. Our research shows that MT occurs via tunneling nanotubes and partial cell fusion with extreme increases under the influence of chemotherapeutic drugs, but its inhibition is limited. However, the supportive effect of stromal cells can be effectively avoided by influencing the metabolism of myeloma cells with the concomitant use of chemotherapeutic agents and an inhibitor of oxidative phosphorylation. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

16 pages, 2888 KiB  
Article
Telomere Architecture Correlates with Aggressiveness in Multiple Myeloma
by Aline Rangel-Pozzo, Pak Lok Ivan Yu, Sadhana LaL, Yasmin Asbaghi, Luiza Sisdelli, Pille Tammur, Anu Tamm, Mari Punab, Ludger Klewes, Sherif Louis, Hans Knecht, Adebayo Olujohungbe and Sabine Mai
Cancers 2021, 13(8), 1969; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13081969 - 19 Apr 2021
Cited by 11 | Viewed by 2732
Abstract
The prognosis of multiple myeloma (MM), an incurable B-cell malignancy, has significantly improved through the introduction of novel therapeutic modalities. Myeloma prognosis is essentially determined by cytogenetics, both at diagnosis and at disease progression. However, for a large cohort of patients, cytogenetic analysis [...] Read more.
The prognosis of multiple myeloma (MM), an incurable B-cell malignancy, has significantly improved through the introduction of novel therapeutic modalities. Myeloma prognosis is essentially determined by cytogenetics, both at diagnosis and at disease progression. However, for a large cohort of patients, cytogenetic analysis is not always available. In addition, myeloma patients with favorable cytogenetics can display an aggressive clinical course. Therefore, it is necessary to develop additional prognostic and predictive markers for this disease to allow for patient risk stratification and personalized clinical decision-making. Genomic instability is a prominent characteristic in MM, and we have previously shown that the three-dimensional (3D) nuclear organization of telomeres is a marker of both genomic instability and genetic heterogeneity in myeloma. In this study, we compared in a longitudinal prospective study blindly the 3D telomeric profiles from bone marrow samples of 214 initially treatment-naïve patients with either monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), or MM, with a minimum follow-up of 5 years. Here, we report distinctive 3D telomeric profiles correlating with disease aggressiveness and patient response to treatment in MM patients, and also distinctive 3D telomeric profiles for disease progression in smoldering multiple myeloma patients. In particular, lower average intensity (telomere length, below 13,500 arbitrary units) and increased number of telomere aggregates are associated with shorter survival and could be used as a prognostic factor to identify high-risk SMM and MM patients. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

13 pages, 495 KiB  
Article
Kinetics of Renal Function during Induction in Newly Diagnosed Multiple Myeloma: Results of Two Prospective Studies by the German Myeloma Study Group DSMM
by Friederike Bachmann, Martin Schreder, Monika Engelhardt, Christian Langer, Denise Wolleschak, Lars Olof Mügge, Heinz Dürk, Kerstin Schäfer-Eckart, Igor Wolfgang Blau, Martin Gramatzki, Peter Liebisch, Matthias Grube, Ivana v Metzler, Florian Bassermann, Bernd Metzner, Christoph Röllig, Bernd Hertenstein, Cyrus Khandanpour, Tobias Dechow, Holger Hebart, Wolfram Jung, Sebastian Theurich, Georg Maschmeyer, Hans Salwender, Georg Hess, Max Bittrich, Leo Rasche, Annamaria Brioli, Kai-Uwe Eckardt, Christian Straka, Swantje Held, Hermann Einsele and Stefan Knopadd Show full author list remove Hide full author list
Cancers 2021, 13(6), 1322; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13061322 - 16 Mar 2021
Cited by 5 | Viewed by 2262
Abstract
Background: Preservation of kidney function in newly diagnosed (ND) multiple myeloma (MM) helps to prevent excess toxicity. Patients (pts) from two prospective trials were analyzed, provided postinduction (PInd) restaging was performed. Pts received three cycles with bortezomib (btz), cyclophosphamide, and dexamethasone (dex; VCD) [...] Read more.
Background: Preservation of kidney function in newly diagnosed (ND) multiple myeloma (MM) helps to prevent excess toxicity. Patients (pts) from two prospective trials were analyzed, provided postinduction (PInd) restaging was performed. Pts received three cycles with bortezomib (btz), cyclophosphamide, and dexamethasone (dex; VCD) or btz, lenalidomide (len), and dex (VRd) or len, adriamycin, and dex (RAD). The minimum required estimated glomerular filtration rate (eGFR) was >30 mL/min. We analyzed the percent change of the renal function using the International Myeloma Working Group (IMWG) criteria and Kidney Disease: Improving Global Outcomes (KDIGO)-defined categories. Results: Seven hundred and seventy-two patients were eligible. Three hundred and fifty-six received VCD, 214 VRd, and 202 RAD. VCD patients had the best baseline eGFR. The proportion of pts with eGFR <45 mL/min decreased from 7.3% at baseline to 1.9% PInd (p < 0.0001). Thirty-seven point one percent of VCD versus 49% of VRd patients had a decrease of GFR (p = 0.0872). IMWG-defined “renal complete response (CRrenal)” was achieved in 17/25 (68%) pts after VCD, 12/19 (63%) after RAD, and 14/27 (52%) after VRd (p = 0.4747). Conclusions: Analyzing a large and representative newly diagnosed myeloma (NDMM) group, we found no difference in CRrenal that occurred independently from the myeloma response across the three regimens. A trend towards deterioration of the renal function with VRd versus VCD may be explained by a better pretreatment “renal fitness” in the latter group. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

27 pages, 1198 KiB  
Review
Keeping Myeloma in Check: The Past, Present and Future of Immunotherapy in Multiple Myeloma
by James Ackley, Miguel Armenta Ochoa, Delta Ghoshal, Krishnendu Roy, Sagar Lonial and Lawrence H. Boise
Cancers 2021, 13(19), 4787; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13194787 - 24 Sep 2021
Cited by 13 | Viewed by 3715
Abstract
Multiple myeloma is an incurable disease of malignant plasma cells and an ideal target for modern immune therapy. The unique plasma cell biology maintained in multiple myeloma, coupled with its hematological nature and unique bone marrow microenvironment, provide an opportunity to design specifically [...] Read more.
Multiple myeloma is an incurable disease of malignant plasma cells and an ideal target for modern immune therapy. The unique plasma cell biology maintained in multiple myeloma, coupled with its hematological nature and unique bone marrow microenvironment, provide an opportunity to design specifically targeted immunotherapies that selectively kill transformed cells with limited on-target off-tumor effects. Broadly defined, immune therapy is the utilization of the immune system and immune agents to treat a disease. In the context of multiple myeloma, immune therapy can be subdivided into four main categories: immune modulatory imide drugs, targeted antibodies, adoptive cell transfer therapies, and vaccines. In recent years, advances in all four of these categories have led to improved therapies with enhanced antitumor activity and specificity. In IMiDs, modified chemical structures have been developed that improve drug potency while reducing dose limiting side effects. Targeted antibody therapies have resulted from the development of new selectively expressed targets as well as the development of antibody drug conjugates and bispecific antibodies. Adoptive cell therapies, particularly CAR-T therapies, have been enhanced through improvements in the manufacturing process, as well as through the development of CAR constructs that enhance CAR-T activation and provide protection from a suppressive immune microenvironment. This review will first cover in-class breakthrough therapies for each of these categories, as well as therapies currently utilized in the clinic. Additionally, this review will explore up and coming therapeutics in the preclinical and clinical trial stage. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

18 pages, 1024 KiB  
Review
Rapid Progress in the Use of Immunomodulatory Drugs and Cereblon E3 Ligase Modulators in the Treatment of Multiple Myeloma
by Grzegorz Charliński, David H. Vesole and Artur Jurczyszyn
Cancers 2021, 13(18), 4666; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13184666 - 17 Sep 2021
Cited by 13 | Viewed by 3731
Abstract
Over the past two decades, the improvement in our understanding of the biology of MM and the introduction of new drug classes, including immunomodulatory drugs (IMiDs), proteasome inhibitors (PI), and monoclonal antibodies (MoAb), have significantly improved outcomes. The first IMiD introduced to treat [...] Read more.
Over the past two decades, the improvement in our understanding of the biology of MM and the introduction of new drug classes, including immunomodulatory drugs (IMiDs), proteasome inhibitors (PI), and monoclonal antibodies (MoAb), have significantly improved outcomes. The first IMiD introduced to treat MM was thalidomide. The side effects observed during treatment with thalidomide initiated work on the synthesis of IMiD analogs. Subsequently, lenalidomide and pomalidomide were developed, both with different safety profiles, and they have better tolerability than thalidomide. In 2010, the cereblon (CRBN) protein was discovered as a direct target of IMiDs. By binding to CRBN, IMiDs change the substrate specificity of the CRBN E3 ubiquitin ligase complex, which results in the breakdown of internal Ikaros and Aiolos proteins. Most clinical trials conducted, both in newly diagnosed, post-transplant maintenance and relapsed/refractory MM, report a beneficial effect of IMiDs on the extension of progression-free survival and overall survival in patients with MM. Due to side effects, thalidomide is used less frequently. Currently, lenalidomide is used at every phase of MM treatment. Lenalidomide is used in conjunction with other agents such as PIs and MoAb as induction and relapsed therapy. Pomalidomide is currently used to treat relapsed/refractory MM, also with PIs and monoclonal antibodies. Current clinical trials are evaluating the efficacy of IMiD derivatives, the CRBN E3 ligase modulators (CELMoDs). This review focuses on the impact of IMiDs for the treatment of MM. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

24 pages, 2179 KiB  
Review
Harnessing the Immune System to Fight Multiple Myeloma
by Jakub Krejcik, Mike Bogetofte Barnkob, Charlotte Guldborg Nyvold, Thomas Stauffer Larsen, Torben Barington and Niels Abildgaard
Cancers 2021, 13(18), 4546; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13184546 - 10 Sep 2021
Cited by 9 | Viewed by 3390
Abstract
Multiple myeloma (MM) is a heterogeneous plasma cell malignancy differing substantially in clinical behavior, prognosis, and response to treatment. With the advent of novel therapies, many patients achieve long-lasting remissions, but some experience aggressive and treatment refractory relapses. So far, MM is considered [...] Read more.
Multiple myeloma (MM) is a heterogeneous plasma cell malignancy differing substantially in clinical behavior, prognosis, and response to treatment. With the advent of novel therapies, many patients achieve long-lasting remissions, but some experience aggressive and treatment refractory relapses. So far, MM is considered incurable. Myeloma pathogenesis can broadly be explained by two interacting mechanisms, intraclonal evolution of cancer cells and development of an immunosuppressive tumor microenvironment. Failures in isotype class switching and somatic hypermutations result in the neoplastic transformation typical of MM and other B cell malignancies. Interestingly, although genetic alterations occur and evolve over time, they are also present in premalignant stages, which never progress to MM, suggesting that genetic mutations are necessary but not sufficient for myeloma transformation. Changes in composition and function of the immune cells are associated with loss of effective immune surveillance, which might represent another mechanism driving malignant transformation. During the last decade, the traditional view on myeloma treatment has changed dramatically. It is increasingly evident that treatment strategies solely based on targeting intrinsic properties of myeloma cells are insufficient. Lately, approaches that redirect the cells of the otherwise suppressed immune system to take control over myeloma have emerged. Evidence of utility of this principle was initially established by the observation of the graft-versus-myeloma effect in allogeneic stem cell-transplanted patients. A variety of new strategies to harness both innate and antigen-specific immunity against MM have recently been developed and intensively tested in clinical trials. This review aims to give readers a basic understanding of how the immune system can be engaged to treat MM, to summarize the main immunotherapeutic modalities, their current role in clinical care, and future prospects. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

18 pages, 1847 KiB  
Review
Review of Myeloma Therapies and Their Potential for Oral and Maxillofacial Side Effects
by Sophie Beaumont, Simon Harrison and Jeremy Er
Cancers 2021, 13(17), 4479; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13174479 - 06 Sep 2021
Cited by 2 | Viewed by 5349
Abstract
Myeloma is a common haematological malignancy in which adverse skeletal related events are frequently seen. Over recent years, treatment for myeloma has evolved leading to improved survival. Antiresorptive therapy is an important adjunct therapy to reduce the risk of bone fractures and to [...] Read more.
Myeloma is a common haematological malignancy in which adverse skeletal related events are frequently seen. Over recent years, treatment for myeloma has evolved leading to improved survival. Antiresorptive therapy is an important adjunct therapy to reduce the risk of bone fractures and to improve the quality of life for myeloma patients; however, this has the potential for unwanted side effects in the oral cavity and maxillofacial region. Osteonecrosis of the jaw related to antiresorptive medications and other myeloma therapies is not uncommon. This review serves to highlight the risk of osteonecrosis of the jaw for myeloma patients, with some suggestions for prevention and management. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

17 pages, 798 KiB  
Review
Minimal Residual Disease in Multiple Myeloma: Something Old, Something New
by Carlos Bravo-Pérez, María Sola, Raúl Teruel-Montoya, María Dolores García-Malo, Francisco José Ortuño, Vicente Vicente, Felipe de Arriba and Andrés Jerez
Cancers 2021, 13(17), 4332; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13174332 - 27 Aug 2021
Cited by 6 | Viewed by 2483
Abstract
The game-changing outcome effect, due to the generalized use of novel agents in MM, has cre-ated a paradigm shift. Achieving frequent deep responses has placed MM among those neoplasms where the rationale for assessing MRD is fulfilled. However, its implementation in MM has [...] Read more.
The game-changing outcome effect, due to the generalized use of novel agents in MM, has cre-ated a paradigm shift. Achieving frequent deep responses has placed MM among those neoplasms where the rationale for assessing MRD is fulfilled. However, its implementation in MM has raised specific questions: how might we weight standard measures against deep MRD in the emerging CAR-T setting? Which high sensitivity method to choose? Are current response criteria still useful? In this work, we address lessons learned from the use of MRD in other neoplasms, the steps followed for the harmonization of current methods for comprehensively measuring MRD, and the challenges that new therapies and concepts pose in the MM clinical field. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

19 pages, 1020 KiB  
Review
CART-Cell Therapy: Recent Advances and New Evidence in Multiple Myeloma
by Massimo Martino, Filippo Antonio Canale, Caterina Alati, Iolanda Donatella Vincelli, Tiziana Moscato, Gaetana Porto, Barbara Loteta, Virginia Naso, Massimiliano Mazza, Fabio Nicolini, Andrea Ghelli Luserna di Rorà, Giorgia Simonetti, Sonia Ronconi, Michela Ceccolini, Gerardo Musuraca, Giovanni Martinelli and Claudio Cerchione
Cancers 2021, 13(11), 2639; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13112639 - 27 May 2021
Cited by 19 | Viewed by 6090
Abstract
Despite the improvement in survival outcomes, multiple myeloma (MM) remains an incurable disease. Chimeric antigen receptor (CAR) T-cell therapy targeting B-cell maturation antigen (BCMA) represents a new strategy for the treatment of relapsed/refractory MM (R/R). In this paper, we describe several recent advances [...] Read more.
Despite the improvement in survival outcomes, multiple myeloma (MM) remains an incurable disease. Chimeric antigen receptor (CAR) T-cell therapy targeting B-cell maturation antigen (BCMA) represents a new strategy for the treatment of relapsed/refractory MM (R/R). In this paper, we describe several recent advances in the field of anti-BCMA CAR T-cell therapy and MM. Currently, available data on anti-BCMA CART-cell therapy has demonstrated efficacy and manageable toxicity in heavily pretreated R/R MM patients. Despite this, the main issues remain to be addressed. First of all, a significant proportion of patients eventually relapse. The potential strategy to prevent relapse includes sequential or combined infusion with CAR T-cells against targets other than BCMA, CAR T-cells with novel dual-targeting vector design, and BCMA expression upregulation. Another dark side of CART therapy is safety. Cytokine release syndrome (CRS) andneurologic toxicity are well-described adverse effects. In the MM trials, most CRS events tended to be grade 1 or 2, with fewer patients experiencing grade 3 or higher. Another critical point is the extended timeline of the manufacturing process. Allo-CARs offers the potential for scalable manufacturing for on-demand treatment with shorter waiting days. Another issue is undoubtedly going to be access to this therapy. Currently, only a few academic centers can perform these procedures. Recognizing these issues, the excellent response with BCMA-targeted CAR T-cell therapy makes it a treatment strategy of great promise. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

Other

18 pages, 1661 KiB  
Systematic Review
A Systematic Review of Cost-Effectiveness Analyses of Novel Agents in the Treatment of Multiple Myeloma
by Maarten R. Seefat, David G. J. Cucchi, Stijn Dirven, Kaz Groen, Sonja Zweegman and Hedwig M. Blommestein
Cancers 2021, 13(22), 5606; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13225606 - 09 Nov 2021
Cited by 5 | Viewed by 4106
Abstract
Background: Novel therapies for multiple myeloma (MM) promise to improve outcomes but are also associated with substantial increasing costs. Evidence regarding cost-effectiveness of novel treatments is necessary, but a comprehensive up-to-date overview of the cost-effectiveness evidence of novel treatments is currently lacking. Methods: [...] Read more.
Background: Novel therapies for multiple myeloma (MM) promise to improve outcomes but are also associated with substantial increasing costs. Evidence regarding cost-effectiveness of novel treatments is necessary, but a comprehensive up-to-date overview of the cost-effectiveness evidence of novel treatments is currently lacking. Methods: We searched Embase, Medline via Ovid, Web of Science and EconLIT ProQuest to identify all cost-effectiveness evaluations of novel pharmacological treatment of MM reporting cost per quality-adjusted life year (QALY) and cost per life year (LY) gained since 2005. Quality and completeness of reporting was assessed using the Consolidated Health Economic Evaluation Reporting Standards. Results: We identified 13 economic evaluations, comprising 32 comparisons. Our results show that novel agents generate additional LYs (range: 0.311–3.85) and QALYs (range: 0.1–2.85) compared to backbone regimens and 0.02 to 1.10 LYs and 0.01 to 0.91 QALYs for comparisons between regimens containing two novel agents. Lifetime healthcare costs ranged from USD 60,413 to 1,434,937 per patient. The cost-effectiveness ratios per QALY gained ranged from dominating to USD 1,369,062 for novel agents compared with backbone therapies and from dominating to USD 618,018 for comparisons between novel agents. Conclusions: Cost-effectiveness ratios of novel agents were generally above current willingness-to-pay thresholds. To ensure access, cost-effectiveness should be improved or cost-effectiveness ratios above current thresholds should be accepted. Full article
(This article belongs to the Special Issue Multiple Myeloma: Targeted Therapy and Immunotherapy)
Show Figures

Figure 1

Back to TopTop