Open access peer-reviewed chapter

Multiple Myeloma in the Era of Novel Agents and Stem Cell Therapies

Written By

Khalid Ahmed Al-Anazi

Submitted: 08 June 2022 Reviewed: 01 December 2022 Published: 09 January 2023

DOI: 10.5772/intechopen.109279

From the Edited Volume

Recent Updates on Multiple Myeloma

Edited by Khalid Ahmed Al-Anazi

Chapter metrics overview

101 Chapter Downloads

View Full Metrics

Abstract

The recent availability of several lines of novel therapeutic agents such as immunomodulatory agents, proteasome inhibitors, and monoclonal antibodies; the widespread utilization of hematopoietic stem cell transplantation; the use of advanced diagnostic techniques that allow risk stratification and monitoring of treatment responses; and the general improvement in health care have revolutionized treatment of patients with multiple myeloma and this has translated into significant improvements in survival outcomes. Monitoring of minimal residual disease can guide the intensity of treatment, and the efficient application of modern diagnostic tools in monitoring treatment responses in real-world clinical practice can hopefully be achieved in the near future. The recent use of quadruplet regimens in the treatment of patients with multiple myeloma has translated into unprecedented treatment responses and survival outcomes. Also, chimeric antigen receptor T-cell therapy and bispecific antibodies represent a new dimension in the precision medicine in MM. Additionally, our ability to induce deep responses has improved, and the treatment goal in myeloma patients tolerating the recommended therapy has moved from delay of disease progression to induction of the deepest possible response.

Keywords

  • multiple myeloma
  • proteasome inhibitors
  • immunomodulatory agents
  • monoclonal antibodies
  • bispecific antibodies
  • chimeric antigen receptor T-cell therapy
  • hematopoietic stem cell transplantation
  • maintenance therapy

1. Introduction

Multiple myeloma (MM), which accounts for 10–15% of all hematologic malignancies, arises from a terminally differentiated postgerminal center plasma cells in the bone marrow (BM) and is characterized by a monoclonal proliferation of plasma cells resulting in the production of monoclonal antibodies and end-organ damage [1, 2, 3, 4]. MM is a disease of old age with the median age at diagnosis ranging between 65 and 74 years in western countries [1, 2, 3, 5]. The risk factors for MM include old age; certain races such as African Americans and living in certain geographic locations such as Australia, Western Europe, and the United States of America (USA); male gender; and family history [1, 3, 5]. However, ionizing radiation, pesticides and benzene, obesity and chronic infection, genetic factors, chronic antigenic stimulation, and environmental as well as occupational factors play a role in the pathogenesis of MM [5, 6, 7, 8]. The recent advances in diagnostics and therapeutics have translated into an increase in the median survival of patients with MM by approximately 6 years [1, 9]. The global 5 years survival is more than double over the past decades due to the availability of several lines of novel therapeutic agents and hematopoietic stem cell transplantation (HSCT), the recent advancements in diagnostic techniques, and the general improvement in health care [3, 10, 11].

Advertisement

2. Diagnosis and staging of MM

The diagnosis of MM requires: (1) ≥10% clonal BM plasma cells or a biopsy proven plasmacytoma; and (2) evidence of one or more of MM defining events namely: [A] CRAB (hypercalcemia, renal failure, anemia, or lytic bone lesions) features felt related to the plasma cell disorder, [B] BM clonal plasmacytosis ≥ 60%, [C] serum involved/uninvolved free light chain ratio (FLC) ≥ 100 (provided involved FLC is ≥ 100 mg/L), or [D] >1 focal lesion on magnetic resonance imaging (MRI) [1, 2, 3]. Based on the revised international staging system (RISS), MM is usually classified into three stages: (1) stage I: all the following: serum albumin ≥ 3.5 g/ dL, serum beta 2 microglobulin (B2M) < 3.5 mg/L, normal serum lactic dehydrogenase (LDH) and no high-risk (HR) cytogenetics; (2) stage II: not fitting stages I and III with serum B2M: 3.5–5.5 mg/L; and (3) stage III: all the following: serum B2M > 5.5 mg/L and HR cytogenetics or elevated serum LDH level [1, 2, 3]. According to the RISS, which was developed based on a study of 11 international trials, the 5 years survival rates among the patients with stages I, II, and III RISS are 82%, 62%, and 40%, respectively [3, 12]. The RISS combines elements of tumor burden as well as disease biology and allows the use of (1) specific biomarkers to define the disease in addition to the established CRAB features and (2) modern imaging tools to diagnose MM bone disease and clarify several other diagnostic requirements [2, 3, 13]. The presence of del(17p), t(4;14), t(14;16), t(14;20), gain 1q, or p53 mutation is considered HR-MM. Additionally, the presence of any two HR factors is considered double-hit myeloma; while triple-hit myeloma is defined by the presence of ≥ 3 HR features [1].

Advertisement

3. General treatment outline

In transplant-eligible patients, 3–4 cycles of induction therapy that consist of either bortezomib, lenalidomide, dexamethasone (VRd) or bortezomib, cyclophosphamide, dexamethasone (VCD), or bortezomib, thalidomide, dexamethasone (VTD) are usually given followed by single autologous HSCT [1, 2, 3]. However, for patients with HR-MM, it is recommended to give induction therapy with either daratumumab, bortezomib, lenalidomide, dexamethasone (Dara-VRd), or carfilzomib, lenalidomide, dexamethasone (KRd) as alternatives to VRd followed by single or tandem autologous HSCT [1, 2]. Selected standard risk (SR) patients can receive additional cycles of induction and delay in transplant until the first relapse [1]. Patients who are not eligible for HSCT are typically treated with 8–12 cycles of VRd followed by lenalidomide maintenance. Alternatively, these patients can be treated with daratumumab, lenalidomide, and dexamethasone (DRd) [1, 2, 3]. After autologous HSCT, SR patients need lenalidomide maintenance, while bortezomib-based maintenance is needed for patients with HR-MM [1, 3]. In case of refractory disease, most patients require a triplet regimen at relapse, with the choice of regimen varying with each successive relapse [1, 3]. The old, current, and future therapeutic modalities in MM are shown in Table 1 [14, 15, 16, 17, 18, 19].

(1)Melphalan
(2)VAD (vincristine, doxorubicin, dexamethasone) regimen of chemotherapy
(3)Corticosteroids: prednisolone and dexamethasone
(4)Immunomodulatory agents: thalidomide, lenalidomide, pomalidomide
(5)Proteasome inhibitors: bortezomib, carfilzomib, ixazomib
(6)Monoclonal antibodies:
(a) Anti-CD 38 (daratumumab, elotuzumab, isatuximab, and MOR202)
(b) Anti-CD138 (indatuximab ravtansine) (c) Anti-interleukin-6 (siltuximab)
(d) Anti-RANKL (denosumab)       (e) Anti-KIR2DL1/2/4 (IPH2101)
(7)Histone deacetylase inhibitors: panobinostat, vorinostat, romidepsin, and ricolinostat
(8)mTOR inhibitors: everolimus, temsirolimus.
(9)Checkpoint (programmed cell death protein 1) inhibitors: nivolumab, pembrolizumab
(10)Bruton’s tyrosine kinase inhibitors: ibrutinib
(11)BCL2 antagonists (BH3 mimetics): venetoclax, obatoclax, and navitoclax
(12)Cyclin-dependent kinase inhibitors: dinaciclib
(13)BRAF and BRAF/MEK inhibitors: selumetinib
(14)Kinesin spindle protein 1 inhibitors: filanesib, array 520
(15)Selective inhibitors of nuclear-cytoplasmic transport: selinexor, exportin
(16)Phosphoinositide 3-kinase-Akt inhibitors: perifosine, afuresertib
(17)PIM kinase inhibitors: LGH 447
(18)Kinesin spindle protein inhibitors: the peptide drug conjugate melfuflen
(19)Vaccines: (A) Multiple myeloma cell/dendritic cell fusion Vaccines
     (B) Peptide-based vaccines
(20)Bispecific antibodies and bispecific T-cell engagers: AMG-4209; AMG-701;
CC-93269; Teclistamab; Talquetumab; Cevostamab (BFCR4350A); Blinatumomab
(21)Antibody-drug conjugates:
Belantamab mafodotin directed against B-cell maturation antigen (BCMA).
(22)Chimeric antigen receptor T cells (CAR T cells) that are directed against:
CD-19; CD-38; B-cell maturation antigen; and cell surface glycoprotein

Table 1.

Old, current, and future therapeutic modalities in multiple myeloma.

Advertisement

4. Risk stratification, prognosis, and minimal residual disease

Definition of HR-MM includes the following features: (1) HR cytogenetics and molecular mutations, such as del 17p; t4,14; t14,16; t14,20; 1q21 amplification; and TP53; (2) plasma cell leukemia; (3) extramedullary disease (EMD); (4) 5–20% circulating plasma cells; (5) renal failure; (6) relapsed MM; (7) MM refractory to treatment; (8) advanced disease, stage III; and (9) frailty [16, 20]. In patients with HR-MM (double-hit or triple-hit myeloma), it is recommended to adopt the following line of treatment, induction therapy with 3–4 cycles of VRd followed by autologous HSCT, and then maintenance therapy with bortezomib-based regimen, that is, bortezomib every 2 weeks or low-intensity VRd regimen till disease progression [120, 21, 22, 23]. Alternatively, patients can be treated with either (1) 3–4 cycles of KRd followed by early autologous HSCT, followed by carfilzomib-based or bortezomib-based maintenance therapy, or (2) the combination of daratumumab + VRd [16, 20, 22, 23, 24, 25]. The details of prognostication in MM are shown in Table 2 [25, 26, 27, 28, 29, 30].

(1) Risk stratification
  1. Staging of MM such as the revised international staging system

  2. Plasma cell labeling index

  3. Cytogenetics; fluorescence in situ hybridization

  4. Molecular mutations and single nucleotide polymorphism assay

  5. Gene expression profiling

  6. Serum biomarkers: β-2 microglobulin and lactic dehydrogenase

(2) Monitoring response to treatment:
  1. Serum-free light chain assay

  2. Serum heavy/light chain assay

  3. Advanced imaging techniques: magnetic resonance imaging and positron emission tomography

(3) Minimal residual disease (MRD) monitoring:
  1. Circulating plasma cells

  2. MRD monitoring techniques: flowcytometry and next-generation sequencing

  3. Value of depth of response

  4. Proteomic evaluation of MRD

(4) Novel prognostic markers:
  1. Proteomic- and glycomic-based platforms

  2. Biomarkers of drug resistance

Table 2.

Prognostication in multiple myeloma (MM).

In patients with MM, the presence of circulating clonal plasma cells is associated with aggressive disease and poor prognosis [31]. Several studies have shown that detection and quantification of circulating plasma cells as well as circulating tumor cell-free DNA by flow cytometry, next-generation sequencing, and whole exome sequencing, which are less invasive than performing BM biopsies can be used as biomarkers of prognosis and risk stratification in patients with either newly diagnosed MM or in patients with MM on treatment to monitor disease response or progression [31, 32, 33, 34, 35, 36, 37, 38, 39]. Two groups of scientists have proposed two separate risk score models, each composed of five genes: EPAS1, ERC2, PRC1, CSGALNACT1, CCND1, and FAM53B, TAPBPL, REPIN1, DDX11, CSGALNCT1, in order to predict prognosis and overall survival (OS) in patients with MM [40, 41]. Another group of scientists has used 15 gene-signature to predict prognosis and OS in MM patients [42].

Minimal residual disease (MRD) detection represents a sensitive tool to appropriately measure the response obtained with therapies, and it can independently predict prognosis during MM treatment [43, 44]. In 2016, the International Myeloma Working Group (IMWG) updated MM response categories defining MRD-negative responses both in the BM and outside the BM. Hence, our ability to induce deep responses has improved and the treatment goal in patients tolerating treatment has moved from delay of disease progression to the induction of the deepest possible response [44]. Intensive treatment regimens administered after establishing the diagnosis of MM can lead to MRD negativity in up to 70% of patients, although the current proportion of curable patients is still unknown [45]. Additionally, using combinations of novel therapies, MRD-negative status can be achieved in a fairly high proportion of patients [44]. In patients who achieve complete response (CR), several high-sensitivity techniques are available for the detection of MRD, including (1) techniques that can detect residual monoclonal plasma cells within the BM, such as next-generation sequencing, and next-generation flow cytometry; and (2) techniques which can detect disease outside the BM by imaging techniques, such as computerized axial tomography scans, positron emission tomography, and MRI or by techniques that detect circulating plasma cells and disease markers in the peripheral blood [45]. Utilization of these advanced techniques allows the determination of the efficacy of antimyeloma treatments and early detection of MRD that can drive clinical relapse [43, 45]. Consequently, high-sensitivity techniques to detect MRD have been developed and validated [44, 46].

The achievement of MRD negativity after therapy, which is considered prognostically important for MM patients, has superseded the conventional CR and has been proposed as a surrogate endpoint for progression-free survival (PFS) and OS as confirmed by data from clinical trials and meta-analyses [43, 45]. So, MRD monitoring can guide treatment intensity, but the efficient application of tools used in monitoring in real-world clinical practice and their potential role to guide treatment-decision making are still open issues [44, 45, 46]. In clinical practice, MRD evaluation is usually performed prior to autologous HSCT, before starting maintenance chemotherapy, and then yearly whilst on maintenance treatment [24].

Advertisement

5. Treatment of relapsed and refractory MM

The choice of treatment regimen at relapse of MM is complicated and is affected by several factors, including the timing of relapse, response to prior therapy, aggressiveness of the relapse, and performance status of the patient [47]. The treatment choices in patients with relapsed MM include (1) salvage with the classical triplet regimens: VRd, VCD, and VTD; (2) daratumumab combinations: daratumumab, bortezomib, dexamethasone; daratumumab, pomalidomide, dexamethasone; DRd; (3) other drug combinations: KRd; ixazomib, lenalidomide, dexamethasone (IRD); elotuzumab, lenalidomide, dexamethasone (ERD); pomalidomide, daratumumab, dexamethasone; and pomalidomide, carfilzomib, dexamethasone; (4) other drugs (panobinostat, bendamustine, venetoclax, pembrolizumab) in various combinations; (5) other single-agent regimens: isatuximab, selinexor, and LGH-447 (pan PIM kinase inhibitor); (6) new immunotherapies, such as chimeric antigen receptor (CAR) T-cells; and (7) salvage or second autologous HSCT in patients relapsing after the first autologous HSCT [1, 47, 48].

Approval of several novel agents in the last decade has substantially changed the landscape of relapsed and refractory (RR-MM) [49]. During the past 2 decades, agents with novel mechanisms of action, such as monoclonal antibodies (MAbs) and histone deacetylase inhibitors (HDACs), have been applied to treat RR-MM [50]. Many clinical trials have assessed the effect and safety of MAbs in combination with proteasome inhibitors (PIs) or immunomodulatory agents (IMiDs) plus dexamethasone/prednisone for the treatment of MM [51]. The choice of therapy for RR-MM requires careful consideration of patient factors including age, frailty, comorbidities, and disease factors, such as symptom burden or biology, as well as treatment-related factors, including drug toxicities and responses to previous therapies. Also, a critical factor in selecting a certain agent is the patient’s sensitivity to lenalidomide and bortezomib at the time of relapse [49].

Combinatory strategies with carfilzomib, plus dexamethasone with or without lenalidomide have shown promising efficacy for patients with RR-MM in pivotal clinical trials [52]. The KRd regimen has been approved for the treatment of RR-MM based on ASPIRE clinical trial as the regimen has been shown to be effective and well tolerated in RR-MM patients [53, 54]. Additionally, a longer PFS was shown in patients achieving a very good partial response (VGPR), in patients who are lenalidomide naïve, and in those relapsing after previous autologous HSCT. Hence, previous autologous HSCT should not hamper the option for KRd therapy [53].

Daratumumab has demonstrated efficacy as monotherapy and combination therapy across several indications, both among newly diagnosed and refractory patients with MM. Daratumumab-based regimens are an effective treatment option across all lines of therapy, with highest response rate in first-line [55]. Daratumumab triplet regimen (DRd) has been shown to be superior to other triplet regimens for the treatment of RR-MM, and daratumumab monotherapy has been shown to be more effective than either single agent in heavily pretreated MM patients, suggesting that daratumumab is effective in the treatment of RR-MM [56]. The EQUULEUS and CANDOR clinical trials have established the efficacy of the DKd regimen (daratumumab, carfilzomib, and dexamethasone) in the landscape of bortezomib and lenalidomide refractory patients. Additionally, the split dosing schedule of the first dose of daratumumab, which was approved by the food and drug administration (FDA) in the USA based on the EQUULEUS trial, has significantly improved patient convenience [57]. Thus, novel and effective regimens are needed in patients with RR-MM who inevitably relapse after treatment containing PIs and IMiDs [57].

Despite the availability of several treatment options, most patients with MM will ultimately become refractory to the three classes of therapy that currently comprise the standard of care for MM: PIs, IMiDs, and MoAbs [58, 59]. Patients who are refractory to the three classes of antimyeloma drugs have poor survival [58, 59]. The current therapeutic approaches of triple-class refractory disease are limited with short-lived efficacy, and they include conventional chemotherapy; salvage autologous HSCT; and recycling of the previous regimens [58, 59]. Salvage high-dose chemotherapy (HDCT) and autologous HSCT are the treatment options for RR-MM [53, 60]. The deep remissions achieved with KRd translate into prolonged PFS, following salvage HDCT and autologous HSCT, and are enhanced by maintenance treatment [53, 60]. It is anticipated that selinexor, CAR T-cell therapy, and next-generation MoAbs will be available for triple-refractory disease in the near future [58, 59].

Advertisement

6. New therapeutic modalities in MM

6.1 CAR T-cell therapy

CAR T-cell therapy is a new cellular immunotherapy that can target and recognize antigens and kill tumor cells, but the efficacy and safety of this therapeutic modality are variable in different studies [61]. Treatment with CAR T-cells has dramatically changed the therapeutic effectiveness in high-grade (HG) B-cell malignancies [62]. However, safety and efficacy of CAR T-cell therapy are affected by the types of costimulatory molecules and CAR T-cell antigens [61].

In recent years, several novel therapeutic agents have improved the prognosis in patients with RR-MM but the prognosis of patients with EMD remains poor [63]. CAR T-cell therapy has demonstrated efficacy and safety in patients with RR-MM with B-cell maturation antigen (BCMA)-targeted and anti-BCMA-contained regimens with superior effectiveness [62, 64]. Despite the HG cytokine release syndrome (CRS) and immune effector cell-associated neurotoxic syndrome encountered, anti-BCMA CAR T-cell therapy allows a remission time for RR-MM patients with EMD, which could be maintained by bridging to HSCT and other therapies [63]. In patients with RR-MM having HR cytogenetics, anti-BCMA CAR T-cell treatment can improve the outcome, particularly if this form of therapy is given early in the course of the disease [64]. Primary resistance and relapse occur with single-target immunotherapy, but humanized bispecific BM38 CAR T-cells (that target both BCMA and CD38) have been shown to be feasible, safe, and significantly effective in patients with RR-MM [65].

6.2 Bispecific antibodies (BsAbs)

One of the hallmarks of MM is immune dysfunction and tumor-permissive immune microenvironment. Hence, ameliorating immune paresis can lead to improved outcomes [66]. However, the OS of triplet-class refractory MM remains poor [67].

BsAbs are novel immunotherapeutic approaches that are designed to bind antigens on malignant plasma cells and cytotoxic effector cells, such as T-cells and natural killer cells [67, 68]. The use of BsAbs early in clinical trials has shown a favorable safety profile and impressive preliminary efficacy in heavily pretreated patients with MM with response rates ranging between 61% and 83% [67, 68, 69]. However, CRS and neurotoxicity have been reported and resistance mechanisms were found to be related to the following: tumor-related features, T-cell characteristics, and impact of components of the immune suppressive tumor microenvironment [66, 69].

Various clinical trials are currently evaluating combining BsAbs with other agents, such as CD38 monoclonal antibodies, and immunomodulatory agents such as pomalidomide to further improve the duration and depth of responses [69]. Together with CAR T-cells, BsAbs represent a new dimension in precision medicine in MM [68].

6.3 Selinexor in the treatment of MM

Selinexor, which is an oral inhibitor of the nuclear export protein exportin-1, has been shown to be safe, tolerable, and effective in the treatment of RR-MM, particularly when combined with either dexamethasone alone or bortezomib and dexamethasone [70, 71, 72, 73, 74].

6.4 Venetoclax in the treatment of MM

B-cell lymphoma-2 (BCL-2) protein is an antiapoptotic protein expressed on clonal plasma cells in patients with MM [75]. Venetoclax is a highly selective, potent, oral BCL-2 inhibitor that can induce apoptosis in MM cells [76]. MM subsets with t11,14 have overexpression of BCL-2 and can benefit from venetoclax when used either alone or in combination with other chemotherapeutic agents with an overall response rate of 40–100% [75]. However, the following side effects have been reported: gastrointestinal disturbances, cytopenias, infectious complications, and death [75, 76]. Venetoclax and dexamethasone combination has demonstrated efficacy and manageable safety in heavily pretreated patients with RR-MM having t11,14 [77]. Additionally, the combination of venetoclax, bortezomib, and dexamethasone has shown encouraging clinical efficacy with acceptable safety and tolerability in a phase-I trial [76].

6.5 Iberdomide in the treatment of MM

Cereblon is the essential binding protein of IMiDs [78, 79]. Almost one-third of MM patients have genetic alterations in cereblon by the time they become refractory to pomalidomide [78]. Three cereblon genetic aberrations that are associated with inferior outcomes to pomalidomide-based regimens have been described in patients who are already refractory to lenalidomide [78]. The biochemical activity of iberbomide, a potent cereblon E3 ligase modulator, translates into greater anti-MM activity than lenalidomide or pomalidomide in IMiD-sensitive and IMiD-resistant MM cell lines [80]. In patients with heavily pretreated RR-MM, the following combinations: iberbomide, daratumumab, dexamethasone; iberbomide, bortezomib, dexamethasone; and iberbomide, carfilzomib, dexamethasone have shown tolerable safety profile and promising efficacy [79, 81].

6.6 Melflufen in the treatment of MM

Melflufen, a peptide-drug conjugate that relies on a novel drug-delivery platform, has 50 times higher cytotoxicity than melphalan and it received accelerated approval by the FDA in the USA after showing potent antimyeloma activity based on the Horizon trial in February 2021, but it was withdrawn from the USA market in October 2021, based on the results of the Ocean trial, which showed inferior survival in patients treated with melflufen [82, 83].

Advertisement

7. Conclusions

The recent advances in therapeutics and diagnostics have revolutionized the management of patients with MM and have significantly improved survival outcomes. The introduction of quadruplet regimens in the treatment of patients with MM has translated into unprecedented therapeutic responses and survival outcomes. The current and future use of new therapeutic modalities such as CAR T-cells, BsAbs, selinexor, venetoclax, and iberdomide represents a new dimension in the era of precision medicine in MM.

References

  1. 1. Rajkumar SV. Multiple myeloma: 2022 update on diagnosis, risk stratification, and management. American Journal of Hematology. 2022;97(8):1086-1107. DOI: 10.1002/ajh.26590. Epub: 23 May 2022
  2. 2. Charliński G, Jurczyszyn A. Multiple myeloma-2020 update on diagnosis and management. NOWOTWORY Journal of Oncology. 2020;70:173-183. DOI: 10.5603/ NJO.a2020.0035
  3. 3. Padala SA, Barsouk A, Barsouk A, Rawla P, Vakiti A, Kolhe R, et al. Epidemiology, staging, and management of multiple myeloma. Medical Sciences (Basel). 2021;9(1):3. DOI: 10.3390/ medsci9010003
  4. 4. Jurczyszyn A, Suska A. Multiple myeloma. Encyclopedia in Biomedical Gerontology. 2020;2:461-478. DOI: 10.1016/B978-0-12-801238-3.11412-6. Epub: 29 Nov 2019
  5. 5. Gerecke C, Fuhrmann S, Strifler S, Schmidt-Hieber M, Einsele H, Knop S. The diagnosis and treatment of multiple myeloma. Deutsches Ärzteblatt International. 2016;113(27-28):470-476. DOI: 10.3238/arztebl.2016. 0470
  6. 6. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375-2390. DOI: 10.1182/blood-2016-01-643569. Epub: 15 Mar 2016
  7. 7. Bębnowska D, Hrynkiewicz R, Grywalska E, Pasiarski M, Sosnowska-Pasiarska B, Smarz-Widelska I, et al. Immunological prognostic factors in multiple myeloma. International Journal of Molecular Sciences. 2021;22(7):3587. DOI: 10.3390/ijms22073587
  8. 8. Qian J, Jin J, Luo H, Jin C, Wang L, Qian W, et al. Analysis of clinical characteristics and prognostic factors of multiple myeloma: A retrospective single-center study of 787 cases. Hematology. 2017;22(8):472-476. DOI: 10.1080/10245332.2017.1309493. Epub: 2 May 2017
  9. 9. Durie BGM, Hoering A, Abidi MH, Rajkumar SV, Epstein J, Kahanic SP, et al. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): A randomised, open-label, phase 3 trial. Lancet. 2017;389(10068):519-527. DOI: 10.1016/ S0140-6736(16)31594-X. Epub: 23 Dec 2016
  10. 10. Kazandjian D. Multiple myeloma epidemiology and survival: A unique malignancy. Seminars in Oncology. 2016;43(6):676-681. DOI: 10.1053/ j.seminoncol.2016.11.004. Epub: 10 Nov 2016
  11. 11. Joshua DE, Bryant C, Dix C, Gibson J, Ho J. Biology and therapy of multiple myeloma. The Medical Journal of Australia. 2019;210(8):375-380. DOI: 10.5694/mja2.50129. Epub: 23 Apr 2019
  12. 12. Rajkumar SV. Updated diagnostic criteria and staging system for multiple myeloma. American Society of Clinical Oncology Educational Book. 2016;35:e418-e423. DOI: 10.1200/EDBK_159009
  13. 13. Palumbo A, Avet-Loiseau H, Oliva S, Lokhorst HM, Goldschmidt H, Rosinol L, et al. Revised international staging system for multiple myeloma: A report from International Myeloma Working Group. Journal of Clinical Oncology. 2015;33(26):2863-2869. DOI: 10.1200/JCO.2015.61.2267. Epub: 3 Aug 2015
  14. 14. Pinto V, Bergantim R, Caires HR, Seca H, Guimarães JE, Vasconcelos MH. Multiple myeloma: Available therapies and causes of drug resistance. Cancers (Basel). 2020;12(2):407. DOI: 10.3390/cancers12020407
  15. 15. Offidani M, Corvatta L, Morè S, Olivieri A. Novel experimental drugs for treatment of multiple myeloma. Journal of Experimental Pharmacology. 2021;13:245-264. DOI: 10.2147/JEP.S265288
  16. 16. Yang Y, Li Y, Gu H, Dong M, Cai Z. Emerging agents and regimens for multiple myeloma. Journal of Hematology & Oncology. 2020;13(1):150. DOI: 10.1186/s13045-020-00980-5
  17. 17. Gulla A, Anderson KC. Multiple myeloma: The revolution of current therapy and a glance into future. Haematologica. 2020;105(10):2358-2367. DOI: 10.3324/haematol.2020.247015
  18. 18. Leow CC, Low MSY. Targeted therapies for multiple myeloma. Journal of Persian Medicine. 2021;11(5):334. DOI: 10.3390/jpm11050334
  19. 19. Al-Anazi KA. Hematopoietic stem cell transplantation in multiple myeloma in the era of novel therapies. In: Al-Anazi KA, editor. Update on Multiple Myeloma. London, UK: Intech Open; 5 Nov 2018. DOI: 10.5772/ intechopen.79999
  20. 20. Caro J, Al Hadidi S, Usmani S, Yee AJ, Raje N, Davies FE. How to treat high-risk myeloma at diagnosis and relapse. American Society of Clinical Oncology Educational Book. 2021;41:291-309. DOI: 10.1200/ EDBK_320105
  21. 21. Chng WJ, Dispenzieri A, Chim CS, Fonseca R, Goldschmidt H, Lentzsch S, et al. International Myeloma Working Group consensus on risk stratification in multiple myeloma. Leukemia. 2014;28(2):269-277. DOI: 10.1038/leu.2013.247. Epub: 26 Aug 2013
  22. 22. Perrot A, Corre J, Avet-Loiseau H. Risk stratification and targets in multiple myeloma: From genomics to the bedside. American Society of Clinical Oncology Educational Book. 2018;38:675-680. DOI: 10.1200/EDBK_200879
  23. 23. Hanamura I. Multiple myeloma with high-risk cytogenetics and its treatment approach. International Journal of Hematology. 2022;115(6):762-777. DOI: 10.1007/s12185-022-03353-5. Epub: 9 May 2022
  24. 24. Marneni N, Chakraborty R. Current approach to managing patients with newly diagnosed high-risk multiple myeloma. Current Hematologic Malignancy Reports. 2021;16(2):148-161. DOI: 10.1007/s11899-021-00631-7. Epub: 19 Apr 2021
  25. 25. Woldu M, Fentie A, Tadesse T. Treatment approaches of multiple myeloma. In: Fuchs O, editor. Multiple Myeloma. London: Intech Open; 6 May 2021. DOI: 10.5772/intechopen.97390.
  26. 26. Lê GN, Bones J, Coyne M, Bazou D, Dowling P, O’Gorman P, et al. Current and future biomarkers for risk-stratification and treatment personalisation in multiple myeloma. Molecular Omics. 2019;15(1):7-20. DOI: 10.1039/c8mo00193f
  27. 27. Hanbali A, Hassanein M, Rasheed W, Aljurf M, Alsharif F. The evolution of prognostic factors in multiple myeloma. Advances in Hematology. 2017;2017:4812637. DOI: 10.1155/2017/4812637. Epub: 21 Feb 2017
  28. 28. Pawlyn C, Davies FE. Toward personalized treatment in multiple myeloma based on molecular characteristics. Blood. 2019;133(7):660-675. DOI: 10.1182/blood-2018-09-825331. Epub: 26 Dec 2018
  29. 29. Fonseca R, Monge J, Dimopoulos MA. Staging and prognostication of multiple myeloma. Expert Reviews of Hematology. 2014;7(1):21-31. DOI 10.1586/17474086.2014.882224
  30. 30. Ho M, Bianchi G, Anderson KC. Proteomics-inspired precision medicine for treating and understanding multiple myeloma. Expert Review of Precision Medicine Drug Development. 2020;5(2):67-85. DOI: 10.1080/23808993.2020.1732205. Epub: 24 Feb 2020
  31. 31. Han W, Jin Y, Xu M, Zhao SS, Shi Q, Qu X, et al. Prognostic value of circulating clonal plasma cells in newly diagnosed multiple myeloma. Hematology. 2021;26(1):510-517. DOI: 10.1080/16078454.2021.1948208
  32. 32. Galieni P, Travaglini F, Vagnoni D, Ruggieri M, Caraffa P, Bigazzi C, et al. The detection of circulating plasma cells may improve the Revised International Staging System (R-ISS) risk stratification of patients with newly diagnosed multiple myeloma. British Journal of Haematology. 2021;193(3):542-550. DOI: 10.1111/bjh.17118. Epub: 1 Apr 2021
  33. 33. Mack EKM, Hartmann S, Ross P, Wollmer E, Mann C, Neubauer A, et al. Monitoring multiple myeloma in the peripheral blood based on cell-free DNA and circulating plasma cells. Annals of Hematology. 2022;101(4):811-824. DOI: 10.1007/ s00277-022-04771-5. Epub: 1 Feb 2022
  34. 34. Sanoja-Flores L, Flores-Montero J, Puig N, Contreras-Sanfeliciano T, Pontes R, Corral-Mateos A, et al. Blood monitoring of circulating tumor plasma cells by next generation flow in multiple myeloma after therapy. Blood. 2019;134(24):2218-2222. DOI: 10.1182/blood.2019002610
  35. 35. Sanoja-Flores L, Flores-Montero J, Garcés JJ, Paiva B, Puig N, García-Mateo A, et al. Euroflow consortium. Next generation flow for minimally-invasive blood characterization of MGUS and multiple myeloma at diagnosis based on circulating tumor plasma cells (CTPC). Blood Cancer Journal. 2018;8(12):117. DOI: 10.1038/s41408-018-0153-9
  36. 36. Rustad EH, Coward E, Skytøen ER, Misund K, Holien T, Standal T, et al. Monitoring multiple myeloma by quantification of recurrent mutations in serum. Haematologica. 2017;102(7):1266-1272. DOI: 10.3324/haematol.2016.160564. Epub: 6 Apr 2017
  37. 37. Yasui H, Kobayashi M, Sato K, Kondoh K, Ishida T, Kaito Y, et al. Circulating cell-free DNA in the peripheral blood plasma of patients is an informative biomarker for multiple myeloma relapse. International Journal of Clinical Oncology. 2021;26(11):2142-2150. DOI: 10.1007/s10147-021-01991-z. Epub: 14 Jul 2021
  38. 38. Manier S, Park J, Capelletti M, Bustoros M, Freeman SS, Ha G, et al. Whole-exome sequencing of cell-free DNA and circulating tumor cells in multiple myeloma. Nature Communications. 2018;9(1):1691. DOI: 10.1038/s41467-018-04001-5
  39. 39. Deshpande S, Tytarenko RG, Wang Y, Boyle EM, Ashby C, Schinke CD, et al. Monitoring treatment response and disease progression in myeloma with circulating cell-free DNA. European Journal of Haematology. 2021;106(2):230-240. DOI: 10.1111/ ejh.13541. Epub: 10 Nov 2020
  40. 40. Chen X, Liu L, Chen M, Xiang J, Wan Y, Li X, et al. A five-gene risk score model for predicting the prognosis of multiple myeloma patients based on gene expression profiles. Frontiers in Genetics. 2021;12:785330. DOI: 10.3389/fgene.2021.785330
  41. 41. Qi T, Qu J, Tu C, Lu Q, Li G, Wang J, et al. Super-enhancer associated five-gene risk score model predicts overall survival in multiple myeloma patients. Frontiers in Cell and Developmental Biology. 2020;8:596777. DOI: 10.3389/fcell.2020. 596777
  42. 42. Liu L, Qu J, Dai Y, Qi T, Teng X, Li G, et al. An interactive nomogram based on clinical and molecular signatures to predict prognosis in multiple myeloma patients. Aging (Albany NY). 2021;13(14):18442-18463. DOI: 0.18632/aging.203294. Epub: 14 Jul 2021
  43. 43. Mina R, Oliva S, Boccadoro M. Minimal residual disease in multiple myeloma: State of the art and future perspectives. Journal of Clinical Medicine. 2020;9(7):2142. DOI: 10.3390/ jcm9072142
  44. 44. Oliva S, D’Agostino M, Boccadoro M, Larocca A. Clinical applications and future directions of minimal residual disease testing in multiple myeloma. Frontiers in Oncology. 2020;10:1. DOI: 10.3389/fonc.2020. 00001
  45. 45. Bertamini L, D’Agostino M, Gay F. MRD assessment in multiple myeloma: Progress and challenges. Current Hematologic Malignancy Reports. 2021;16(2):162-171. DOI: 10.1007/ s11899-021-00633-5. Epub: 5 May 2021
  46. 46. Bonello F, Cani L, D’Agostino M. Risk stratification before and during treatment in newly diagnosed multiple myeloma: From clinical trials to the real-world setting. Frontiers in Oncology. 2022;12:830922. DOI: 10.3389/fonc.2022. 830922
  47. 47. Rajkumar SV. Multiple myeloma: Every year a new standard? Hematological Oncology. 2019;37(Suppl. 1):62-65. DOI: 10.1002/hon.2586
  48. 48. Chim CS, Kumar SK, Orlowski RZ, Cook G, Richardson PG, Gertz MA, et al. Management of relapsed and refractory multiple myeloma: Novel agents, antibodies, immunotherapies and beyond. Leukemia. 2018;32(2):252-262. DOI: 10.1038/leu. 2017.329. Epub: 16 Nov 2017
  49. 49. Sanchez L, Barley K, Richter J, Franz J, Cho HJ, Jagannath S, et al. Immunomodulatory drug- and proteasome inhibitor-backbone regimens in the treatment of relapsed multiple myeloma: An evidence-based review. Expert Reviews of Hematology. 2020;13(9):943-958. DOI: 10.1080/17474086.2020.1804356. Epub: 30 Aug 2020
  50. 50. Zheng Y, Shen H, Xu L, Feng J, Tang H, Zhang N, et al. Monoclonal antibodies versus histone deacetylase inhibitors in combination with bortezomib or lenalidomide plus dexamethasone for the treatment of relapsed or refractory multiple myeloma: An indirect-comparison meta-analysis of randomized controlled trials. Journal of Immunology Research. 2018;2018:7646913. DOI: 10.1155/2018/7646913
  51. 51. Ye W, Wu X, Liu X, Zheng X, Deng J, Gong Y. Comparison of monoclonal antibodies targeting CD38, SLAMF7 and PD-1/PD-L1 in combination with bortezomib/immunomodulators plus dexamethasone/ prednisone for the treatment of multiple myeloma: An indirect-comparison meta-analysis of randomised controlled trials. BMC Cancer. 2021;21(1):994. DOI: 10.1186/s12885-021-08588-9
  52. 52. Kawaji-Kanayama Y, Kobayashi T, Muramatsu A, Uchiyama H, Sasaki N, Uoshima N, et al. Kyoto Clinical Hematology Study Group (KOTOSG) Investigators. Prognostic impact of resistance to bortezomib and/or lenalidomide in carfilzomib-based therapies for relapsed/refractory multiple myeloma: The Kyoto Clinical Hematology Study Group, multicenter, pilot, prospective, observational study in Asian patients. Cancer Report (Hoboken). 2022;5(2):e1476. DOI: 10.1002/cnr2.1476. Epub: 14 Jun 2021
  53. 53. Mele A, Prete E, De Risi C, Citiso S, Greco G, Falcone AP, et al. Carfilzomib, lenalidomide, and dexamethasone in relapsed/refractory multiple myeloma patients: The real-life experience of Rete Ematologica Pugliese (REP). Annals of Hematology. 2021;100(2):429-436. DOI: 10.1007/s00277-020-04329-3. Epub: 7 Nov 2020
  54. 54. Rocchi S, Tacchetti P, Pantani L, Mancuso K, Rizzello I, di Giovanni BC, et al. A real-world efficacy and safety analysis of combined carfilzomib, lenalidomide, and dexamethasone (KRd) in relapsed/refractory multiple myeloma. Hematological Oncology. 2021;39(1):41-50. DOI: 10.1002/hon.2820. Epub: 1 Nov 2020
  55. 55. Atrash S, Thompson-Leduc P, Tai MH, Kaila S, Gray K, Ghelerter I, et al. Treatment patterns and effectiveness of patients with multiple myeloma initiating Daratumumab across different lines of therapy: A real-world chart review study. BMC Cancer. 2021;21(1):1207. DOI: 10.1186/s12885-021-08881-7
  56. 56. Zhang T, Wang S, Lin T, Xie J, Zhao L, Liang Z, et al. Systematic review and meta-analysis of the efficacy and safety of novel monoclonal antibodies for treatment of relapsed/refractory multiple myeloma. Oncotarget. 2017;8(20):34001-34017. DOI: 10.18632/oncotarget.16987
  57. 57. Richard S, Jagannath S, Cho HJ, Parekh S, Madduri D, Richter J, et al. A comprehensive overview of daratumumab and carfilzomib and the recently approved daratumumab, carfilzomib and dexamethasone regimen in relapsed/refractory multiple myeloma. Expert Review of Hematology. 2021;14(1):31-45. DOI: 10.1080/17474086.2021.1858790. Epub: 17 Dec 2020
  58. 58. Mikhael J. Treatment options for triple-class refractory multiple myeloma. Clinical Lymphoma, Myeloma & Leukemia. 2020;20(1):1-7. DOI: 10.1016/j.clml.2019.09.621. Epub: 9 Oct 2019
  59. 59. Costa LJ, Hungria V, Mohty M, Mateos MV. How I treat triple-class refractory multiple myeloma. British Journal of Haematology. 2022;198(2):244-256. DOI: 10.1111/bjh.18185. Epub: 3 Apr 2022
  60. 60. Baertsch MA, Fougereau M, Hielscher T, Sauer S, Breitkreutz I, Jordan K, et al. Carfilzomib, lenalidomide, and dexamethasone followed by salvage autologous stem cell transplant with or without maintenance for relapsed or refractory multiple myeloma. Cancers (Basel). 2021;13(18):4706. DOI: 10.3390/cancers13184706
  61. 61. Li J, Tang Y, Huang Z. Efficacy and safety of chimeric antigen receptor (CAR)-T cell therapy in the treatment of relapsed and refractory multiple myeloma: A systematic-review and meta-analysis of clinical trials. Translational Cancer Research. 2022;11(3):569-579. DOI: 10.21037/tcr-22-344
  62. 62. Yang Q, Li X, Zhang F, Yang Q, Zhou W, Liu J. Efficacy and safety of CAR-T therapy for relapse or refractory multiple myeloma: A systematic review and meta-analysis. International Journal of Medical Sciences. 2021;18(8):1786-1797. DOI: 10. 7150/ ijms.46811
  63. 63. Deng H, Liu M, Yuan T, Zhang H, Cui R, Li J, et al. Efficacy of humanized anti-BCMA CAR T cell therapy in relapsed/refractory multiple myeloma patients with and without extramedullary disease. Frontiers in Immunology. 2021;12:720571. DOI: 10.3389/fimmu.2021.720571
  64. 64. Zhang L, Shen X, Yu W, Li J, Zhang J, Zhang R, et al. Comprehensive meta-analysis of anti-BCMA chimeric antigen receptor T-cell therapy in relapsed or refractory multiple myeloma. Annals of Medicine. 2021;53(1):1547-1559. DOI: 10.1080/ 07853890. 2021.1970218
  65. 65. Mei H, Li C, Jiang H, Zhao X, Huang Z, Jin D, et al. A bispecific CAR-T cell therapy targeting BCMA and CD38 in relapsed or refractory multiple myeloma. Journal of Hematology & Oncology. 2021;14(1):161. DOI: 10.1186/s13045-021-01170-7
  66. 66. Swan D, Routledge D, Harrison S. The evolving status of immunotherapies in multiple myeloma: The future role of bispecific antibodies. British Journal of Haematology. 2022;196(3):488-506. DOI: 10.1111/ bjh.17805. Epub: 1 Sep 2021
  67. 67. Lancman G, Sastow DL, Cho HJ, Jagannath S, Madduri D, Parekh SS, et al. Bispecific antibodies in multiple myeloma: Present and future. Blood Cancer Discovery. 2021;2(5):423-433. DOI: 10.1158/2643-3230.BCD-21-0028
  68. 68. Zhou X, Einsele H, Danhof S. Bispecific antibodies: A new era of treatment for multiple myeloma. Journal of Clinical Medicine. 2020;9(7):2166. DOI: 10.3390/jcm9072166
  69. 69. Hosny M, Verkleij CPM, van der Schans J, Frerichs KA, Mutis T, Zweegman S, et al. Current state of the art and prospects of T cell-redirecting bispecific antibodies in multiple myeloma. Journal of Clinical Medicine. 2021;10(19):4593. DOI: 10.3390/ jcm10194593
  70. 70. Vogl DT, Dingli D, Cornell RF, Huff CA, Jagannath S, Bhutani D, et al. Selective inhibition of nuclear export with oral selinexor for treatment of relapsed or refractory multiple myeloma. Journal of Clinical Oncology. 2018;36(9):859-866. DOI: 10.1200/JCO.2017.75.5207. Epub: 30 Jan 2018
  71. 71. Bahlis NJ, Sutherland H, White D, Sebag M, Lentzsch S, Kotb R, et al. Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. Blood. 2018;132(24):2546-2554. DOI: 10.1182/blood-2018-06-858852. Epub: 23 Oct 2018
  72. 72. Chari A, Vogl DT, Gavriatopoulou M, Nooka AK, Yee AJ, Huff CA, et al. Oral selinexor-dexamethasone for triple-class refractory multiple myeloma. The New England Journal of Medicine. 2019;381(8):727-738. DOI: 10.1056/ NEJMoa1903455
  73. 73. Grosicki S, Simonova M, Spicka I, Pour L, Kriachok I, Gavriatopoulou M, et al. Once-per-week selinexor, bortezomib, and dexamethasone versus twice-per-week bortezomib and dexamethasone in patients with multiple myeloma (BOSTON): A randomised, open-label, phase 3 trial. Lancet. 2020;396(10262):1563-1573. DOI: 10.1016/S0140-6736(20)32292-3
  74. 74. Peterson TJ, Orozco J, Buege M. Selinexor: A first-in-class nuclear export inhibitor for management of multiply relapsed multiple myeloma. The Annals of Pharmacotherapy. 2020;54(6):577-582. DOI: 10.1177/1060028019892643. Epub: 2 Dec 2019
  75. 75. Ehsan H, Wahab A, Shah Z, Sana MK, Masood A, Rafae A, et al. Role of Venetoclax in the treatment of relapsed and refractory multiple myeloma. Journal of Hematology. 2021;10(3):89-97. DOI: 10.14740/ jh844. Epub: 16 Jun 2021
  76. 76. Kumar SK, Harrison SJ, Cavo M, de la Rubia J, Popat R, Gasparetto C, et al. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): A randomised, double-blind, multicentre, phase 3 trial. The Lancet Oncology. 2020;21(12):1630-1642. DOI: 10.1016/S1470-2045(20)30525-8. Epub: 29 Oct 2020
  77. 77. Kaufman JL, Gasparetto C, Schjesvold FH, Moreau P, Touzeau C, Facon T, et al. Targeting BCL-2 with venetoclax and dexamethasone in patients with relapsed/refractory t(11;14) multiple myeloma. American Journal of Hematology. 2021;96(4):418-427. DOI: 10.1002/ajh.26083. Epub: 19 Jan 2021
  78. 78. Gooding S, Ansari-Pour N, Towfic F, Ortiz Estévez M, Chamberlain PP, Tsai KT, et al. Multiple cereblon genetic changes are associated with acquired resistance to lenalidomide or pomalidomide in multiple myeloma. Blood. 2021;137(2):232-237. DOI: 10.1182/blood.2020007081
  79. 79. Thakurta A, Pierceall WE, Amatangelo MD, Flynt E, Agarwal A. Developing next generation immunomodulatory drugs and their combinations in multiple myeloma. Oncotarget. 2021;12(15):1555-1563. DOI: 10.18632/oncotarget.27973
  80. 80. Bjorklund CC, Kang J, Amatangelo M, Polonskaia A, Katz M, Chiu H, et al. Iberdomide (CC-220) is a potent cereblon E3 ligase modulator with antitumor and immunostimulatory activities in lenalidomide- and pomalidomide-resistant multiple myeloma cells with dysregulated CRBN. Leukemia. 2020;34(4):1197-1201. DOI: 10.1038/s41375-019-0620-8. Epub: 12 Nov 2019
  81. 81. Lonial S, Richardson PG, Popat R, Stadtmauer EA, Larsen JT, Oriol A, et al. OAB-013: Iberdomide (IBER) in combination with dexamethasone (DEX) and daratumumab (DARA), bortezomib (BORT), or carfilzomib (CFZ) in patients (pts) with relapsed/refractory multiple myeloma (RRMM). Clinical Lymphoma, Myeloma & Leukemia. 2021;21(Suppl 2):S9. DOI: 10.1016/ S2152-2650(21)02087-5
  82. 82. Kapoor P, Gonsalves WI. Melflufen for multiple myeloma: A promise unfulfilled? Lancet Haematology. 2022;9(2):e82-e84. DOI: 10.1016/ S2352-3026(22)00002-3. Epub: 12 Jan 2022
  83. 83. Olivier T, Prasadb V. The approval and withdrawal of melphalan flufenamide (melflufen): Implications for the state of the FDA. Translational Oncology. 2022;18:101374. DOI: 10.1016/j.tranon. 2022.101374

Written By

Khalid Ahmed Al-Anazi

Submitted: 08 June 2022 Reviewed: 01 December 2022 Published: 09 January 2023