Open access

Introductory Chapter: Update on Multiple Myeloma

Written By

Khalid Ahmed Al-Anazi

Published: 15 March 2023

DOI: 10.5772/intechopen.110335

From the Edited Volume

Recent Updates on Multiple Myeloma

Edited by Khalid Ahmed Al-Anazi

Chapter metrics overview

81 Chapter Downloads

View Full Metrics

1. Introduction

Multiple myeloma (MM) is a heterogeneous and an incurable disease that is characterized by periods of remission alternating with relapses or progressions that ultimately lead to refractory disease [1, 2]. High-risk (HR) MM is defined by the presence of specific cytogenetic and molecular abnormalities [3, 4]. Double-hit myeloma refers to the presence of ≥2 HR features, while triple-hit MM refers to the presence of ≥3 HR abnormalities [3, 4].

Over the last two decades, the utilization of various novel therapies such as proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies (MoAbs) in the treatment of patients with MM has improved the depth and duration of disease response and has eventually translated into improved overall survival (OS) [5, 6]. The therapeutic modalities of MM include alkylating agents such as melphalan; corticosteroids including dexamethasone; anthracyclines such as liposomal doxorubicin; IMiDs such as lenalidomide, and pomalidomide; PIs including bortezomib, and carfilzomib; MoAbs such as daratumumab; histone deacetylase inhibitors such as panobinostat; exportin-1 inhibitors such as selinexor; BCL2 inhibitors such as venetoclax; chimeric antigen receptor (CAR) T-cells; and bispecific T-cell engaging (BiTE) therapy [1, 4].

For standard risk (SR) and transplant-eligible patients with MM, induction therapy with a PI, an IMiD, and dexamethasone followed by autologous hematopoietic stem cell transplantation (HSCT) represent the standard care [7, 8]. In SR patients, three-four cycles of the triplet regimen bortezomib, lenalidomide, and dexamethasone (VRd) are recommended while in HR patients daratumumab is added to VRd [3, 9, 10, 11, 12, 13]. Patients who are not candidates for transplant are treated with 8–12 cycles of VRd, followed by lenalidomide maintenance. Alternative regimens include daratumumab, lenalidomide, dexamethasone (DRd) or daratumumab, bortezomib, melphalan, and prednisolone (D-VMP) [3, 14, 15, 16].

Autologous HSCT is still considered the standard of care in the treatment of patients with MM who are eligible for transplantation [5, 17, 18, 19, 20]. The standard conditioning regimen for patients with MM undergoing autologous HSCT is high-dose (HD) melphalan but in patients with renal dysfunction or failure, reductions in melphalan doses according to creatinine clearance are required [4, 5, 17, 18, 19]. Cryopreservation of the harvested stem cells is routinely employed prior to autologous HSCT [17, 20, 21]. However, autologous HSCT using non-cryopreserved stem cells has been shown to be safe and cost-effective and leads to short-term and long-term results that are at least equivalent to autologous HSCT using cryopreserved stem cells [17, 21, 22, 23]. Patients with MM are ideal candidates for outpatient autologous HSCT due to the ease of administration of HD melphalan, the relatively low extra-hematological toxicity, and the brief period of neutropenia [24, 25]. Outpatient HSCT has certain inclusion criteria and exclusion criteria as well as several advantages that include: significant reduction in costs; saving hospital beds; lower rate of infections; and lower morbidity and treatment-related mortality [24, 26, 27, 28].

In patients with MM, maintenance therapy after autologous HSCT has been shown to deepen and prolong responses and increase OS and progression-free survival (PFS) [29]. Lenalidomide maintenance given after autologous HSCT till disease progression is the standard of care in patients with SR MM while bortezomib maintenance therapy after autologous HSCT is preferable in MM patients having: HR cytogenetics, renal insufficiency, inability to tolerate lenalidomide, and previous history of another cancer [30, 31, 32]. Continuous therapy has been shown to significantly improve OS and PFS [33, 34]. Continuous therapy till disease progression has become a key strategy in the treatment of patients with MM as it has been shown to improve duration of remission and it represents the standard approach for patients with MM both at diagnosis and at relapse [35, 36].

Unfortunately, nearly all MM patients ultimately relapse, even those who experience a complete response (CR) to initial therapy [19]. Management of the relapsed disease remains a critical aspect of MM care and an important area of ongoing research [19]. New treatment strategies and therapeutic modalities are needed to treat MM in relapse, particularly in case of triple-refractory disease [1]. Treatment of relapsed MM should depend on: the number of relapses encountered; the previous anti-myeloma treatment; the presence of de novo or acquired drug resistance; aggressiveness of disease relapse particularly in case of extramedullary disease, plasma cell leukemia, or clonal evolution [3, 37].

Minimal residual disease (MRD) is an important factor that can independently predict the prognosis of MM during treatment as undetectable MRD has been shown to improve PFS and OS regardless: disease status, prior transplant, or cytogenetic risk [38]. Flow cytometry has become a valuable tool to monitor MRD and evaluate the depth of CR. However, next-generation flow cytometry is more sensitive than the standard flow cytometry in detecting MRD in patients with MM [39]. Finally, the development of novel targeting therapies with different mechanisms of action is needed to achieve deep and durable responses in an attempt to cure MM while identification of tumor intrinsic and extrinsic resistance mechanisms may direct the design of combinations of novel drugs that prevent or overcome drug resistance so as to improve patient survival [40, 41].

References

  1. 1. Stalker ME, Mark TM. Clinical management of triple-class refractory multiple myeloma: A review of current strategies and emerging therapies. Current Oncology. 2022;29(7):4464-4477. DOI: 10.3390/curroncol29070355
  2. 2. 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
  3. 3. 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 2022 May 23
  4. 4. Rajkumar SV, Kumar S. Multiple myeloma current treatment algorithms. Blood Cancer Journal. 2020;10(9):94. DOI: 10.1038/s41408-020-00359-2
  5. 5. Parrondo RD, Ailawadhi S, Sher T, Chanan-Khan AA, Roy V. Autologous stem-cell transplantation for multiple myeloma in the era of novel therapies. Journal of Oncology Practice. 2020;16(2):56-66. DOI: 10.1200/JOP.19.00335
  6. 6. Gonsalves WI, Buadi FK, Ailawadhi S, Bergsagel PL, Chanan Khan AA, Dingli D, et al. Utilization of hematopoietic stem cell transplantation for the treatment of multiple myeloma: A Mayo stratification of Myeloma and risk-adapted therapy (mSMART) consensus statement. Bone Marrow Transplantation. 2019;54(3):353-367. DOI: 10.1038/s41409-018-0264-8. Epub 2018 Jul 9
  7. 7. Kumar SK, Callander NS, Adekola K, Anderson L, Baljevic M, Campagnaro E, et al. Multiple Myeloma, version 3.2021, NCCN clinical practice guidelines in oncology. Journal of the National Comprehensive Cancer Network. 2020;18(12):1685-1717. DOI: 10.6004/jnccn.2020.0057
  8. 8. Cowan AJ, Green DJ, Kwok M, Lee S, Coffey DG, Holmberg LA, et al. Diagnosis and management of multiple myeloma: A review. Journal of the American Medical Association. 2022;327(5):464-477. DOI: 10.1001/jama.2022.0003
  9. 9. 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
  10. 10. Berbari HE, Kumar SK. Initial therapeutic approaches to patients with multiple myeloma. Advances in Therapy. 2021;38(7):3694-3711. DOI: 10.1007/s12325-021-01824-5. Epub 2021 Jun 18
  11. 11. Offidani M, Corvatta L, Morè S, Nappi D, Martinelli G, Olivieri A, et al. Daratumumab for the management of newly diagnosed and relapsed/refractory multiple myeloma: Current and emerging treatments. Frontiers in Oncology. 2021;10:624661. DOI: 10.3389/fonc.2020.624661
  12. 12. Cavo M, Tacchetti P, Zamagni E. Front-line treatment of multiple myeloma. HemaSphere. 2019;3(Suppl):127-130. DOI: 10.1097/HS9.0000000000000242
  13. 13. Moreau P, Hebraud B, Facon T, Leleu X, Hulin C, Hashim M, et al. Front-line daratumumab-VTd versus standard-of-care in ASCT-eligible multiple myeloma: Matching-adjusted indirect comparison. Immunotherapy. 2021;13(2):143-154. DOI: 10.2217/imt-2020-0266. Epub 2020 Nov 24
  14. 14. Korst CLBM, van de Donk NWCJ. Should all newly diagnosed MM patients receive CD38 antibody-based treatment? Hematology. American Society of Hematology Education Program. 2020;2020(1):259-263. DOI: 10.1182/hematology.2020000161
  15. 15. Grant SJ, Mian HS, Giri S, Boutin M, Dottorini L, Neuendorff NR, et al. Transplant-ineligible newly diagnosed multiple myeloma: Current and future approaches to clinical care: A young international society of geriatric oncology review paper. Journal of Geriatric Oncology. 2021;12(4):499-507. DOI: 10.1016/j.jgo.2020.12.001. Epub 2020 Dec 17
  16. 16. Facon T, Kumar SK, Plesner T, Orlowski RZ, Moreau P, Bahlis N, et al. Daratumumab, lenalidomide, and dexamethasone versus lenalidomide and dexamethasone alone in newly diagnosed multiple myeloma (MAIA): Overall survival results from a randomised, open-label, phase 3 trial. The Lancet Oncology. 2021;22(11):1582-1596. DOI: 10.1016/S1470-2045(21)00466-6. Epub 2021 Oct 13
  17. 17. Al-Anazi K. Hematopoietic stem cell transplantation in multiple myeloma in the era of novel therapies. In: Al-Anazi K, editor. Update on Multiple Myeloma. London: Intech Open; 2018. DOI: 10.5772/intechopen.79999
  18. 18. Al Hamed R, Bazarbachi AH, Malard F, Harousseau JL, Mohty M. Current status of autologous stem cell transplantation for multiple myeloma. Blood Cancer Journal. 2019;9(4):44. DOI: 10.1038/s41408-019-0205-9
  19. 19. Charliński G, Jurczyszyn A. Multiple myeloma - 2020 update on diagnosis and management. Nowotwory Journal of Oncology. 2020;70:85-91
  20. 20. Al-Anazi KA. Autologous hematopoietic stem cell transplantation for multiple myeloma without cryopreservation. Bone Marrow Research. 2012;2012:917361. DOI: 10.1155/2012/917361. Epub 2012 May 28
  21. 21. Piriyakhuntorn P, Tantiworawit A, Rattanathammethee T, Hantrakool S, Chai-Adisaksopha C, Rattarittamrong E, et al. Outcomes of non-cryopreserved versus cryopreserved peripheral blood stem cells for autologous stem cell transplantation in multiple myeloma. Annals of Transplantation. 2020;25:e927084. DOI: 10.12659/AOT.927084
  22. 22. Sarmiento M, Ramírez P, Parody R, Salas MQ , Beffermann N, Jara V, et al. Advantages of non-cryopreserved autologous hematopoietic stem cell transplantation against a cryopreserved strategy. Bone Marrow Transplantation. 2018;53(8):960-966. DOI: 10.1038/s41409-018-0117-5. Epub 2018 Feb 13
  23. 23. Wannesson L, Panzarella T, Mikhael J, Keating A. Feasibility and safety of autotransplants with noncryopreserved marrow or peripheral blood stem cells: a systematic review. Annals of Oncology. 2007;18(4):623-632. DOI: 10.1093/annonc/mdm069. Epub 2007 Mar 12
  24. 24. Martino M, Lemoli RM, Girmenia C, Castagna L, Bruno B, Cavallo F, et al. Italian consensus conference for the outpatient autologous stem cell transplantation management in multiple myeloma. Bone Marrow Transplantation. 2016;51(8):1032-1040. DOI: 10.1038/bmt.2016.79. Epub 2016 Apr 4
  25. 25. Martino M, Montanari M, Bruno B, Console G, Irrera G, Messina G, et al. Autologous hematopoietic progenitor cell transplantation for multiple myeloma through an outpatient program. Expert Opinion on Biological Therapy. 2012;12(11):1449-1462. DOI: 10.1517/14712598.2012.707185. Epub 2012 Jul 13
  26. 26. Abid MB, Christopher D, Abid MA, Poon ML, Tan LK, Koh LP, et al. Safety and cost-effectiveness of outpatient autologous transplantation for multiple myeloma in Asia: Single-center perspective from Singapore. Bone Marrow Transplantation. 2017;52(7):1044-1046. DOI: 10.1038/bmt.2017.77. Epub 2017 May 8
  27. 27. Kodad SG, Sutherland H, Limvorapitak W, Abou Mourad Y, Barnett MJ, Forrest D, et al. Outpatient autologous stem cell transplants for multiple myeloma: Analysis of safety and outcomes in a tertiary care center. Clinical Lymphoma, Myeloma & Leukemia. 2019;19(12):784-790. DOI: 10.1016/j.clml. 2019.09.619. Epub 2019 Oct 9
  28. 28. Meisenberg BR, Ferran K, Hollenbach K, Brehm T, Jollon J, Piro LD. Reduced charges and costs associated with outpatient autologous stem cell transplantation. Bone Marrow Transplantation. 1998;21(9):927-932. DOI: 10.1038/sj. bmt.1701191
  29. 29. Vaxman I, Gertz M. Risk adapted post-transplant maintenance in multiple myeloma. Expert Review of Hematology. 2019;12(2):107-118. DOI: 10.1080/17474086.2019.1576521
  30. 30. Attal M, Lauwers-Cances V, Marit G, Caillot D, Moreau P, Facon T, et al. IFM investigators. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. The New England Journal of Medicine. 2012;366(19):1782-1791. DOI: 10.1056/NEJMoa1114138
  31. 31. McCarthy PL, Holstein SA, Petrucci MT, Richardson PG, Hulin C, Tosi P, et al. Lenalidomide maintenance after autologous stem-cell transplantation in newly diagnosed multiple myeloma: A meta-analysis. Journal of Clinical Oncology. 2017;35(29):3279-3289. DOI: 10.1200/JCO.2017.72.6679. Epub 2017 Jul 25
  32. 32. Sivaraj D, Green MM, Li Z, Sung AD, Sarantopoulos S, Kang Y, et al. Outcomes of maintenance therapy with bortezomib after autologous stem cell transplantation for patients with multiple myeloma. Biology of Blood and Marrow Transplantation. 2017;23(2):262-268. DOI: 10.1016/j.bbmt.2016.11.010. Epub 2016 Nov 14
  33. 33. Dimopoulos MA, Jakubowiak AJ, McCarthy PL, Orlowski RZ, Attal M, Bladé J, et al. Developments in continuous therapy and maintenance treatment approaches for patients with newly diagnosed multiple myeloma. Blood Cancer Journal. 2020;10(2):17. DOI: 10.1038/s41408-020-0273-x
  34. 34. Palumbo A, Gay F, Cavallo F, Di Raimondo F, Larocca A, Hardan I, et al. Continuous therapy versus fixed duration of therapy in patients with newly diagnosed multiple myeloma. Journal of Clinical Oncology. 2015;33(30):3459-3466. DOI: 10.1200/JCO.2014.60.2466. Epub 2015 Aug 17
  35. 35. D’Agostino M, De Paoli L, Conticello C, Offidani M, Ria R, Petrucci MT, et al. Continuous therapy in standard- and high-risk newly-diagnosed multiple myeloma: A pooled analysis of 2 phase III trials. Critical Reviews in Oncology/Hematology. 2018;132:9-16. DOI: 10.1016/j.critrevonc.2018.09.008. Epub 2018 Sep 14
  36. 36. Bonello F, Cetani G, Bertamini L, Gay F, Larocca A. Moving toward continuous therapy in multiple myeloma. Clinical Hematology International. 2019;1(4):189-200. DOI: 10.2991/chi.d.191101.001
  37. 37. Suzuki K, Nishiwaki K, Yano S. Treatment strategies considering micro-environment and clonal evolution in multiple myeloma. Cancers (Basel). 2021;13(2):215. DOI: 10.3390/cancers13020215
  38. 38. 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
  39. 39. Flores-Montero J, Sanoja-Flores L, Paiva B, Puig N, García-Sánchez O, Böttcher S, et al. Next generation flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia. 2017;31(10):2094-2103. DOI: 10.1038/leu.2017.29. Epub 2017 Jan 20
  40. 40. Nishida H. Rapid progress in immunotherapies for multiple myeloma: an updated comprehensive review. Cancers (Basel). 2021;13(11):2712. DOI: 10.3390/cancers13112712
  41. 41. Swamydas M, Murphy EV, Ignatz-Hoover JJ, Malek E, Driscoll JJ. Deciphering mechanisms of immune escape to inform immunotherapeutic strategies in multiple myeloma. Journal of Hematology & Oncology. 2022;15(1):17. DOI: 10.1186/s13045-022-01234-2

Written By

Khalid Ahmed Al-Anazi

Published: 15 March 2023