Open access peer-reviewed chapter

Treatment of Multiple Myeloma in the First Relapse

Written By

Ahmad Alhuraiji, Dina Abd El Razik and Shaza A.A. Elkourahy Omar

Submitted: 05 July 2022 Reviewed: 01 August 2022 Published: 30 October 2022

DOI: 10.5772/intechopen.106895

From the Edited Volume

Recent Updates on Multiple Myeloma

Edited by Khalid Ahmed Al-Anazi

Chapter metrics overview

111 Chapter Downloads

View Full Metrics

Abstract

The treatment scope for relapsed myeloma has been expanded considerably in the last few years, by virtue of the advent of numerous novel agents with new mechanisms of actions. This has resulted in increasing responses and prolonging survival even in advanced diseases. The wealth of novel regimens comes with the challenges of balancing toxicities and aligning a regimen with the biology of myeloma and the nature of relapse in conjunction with the patient’s treatment history, comorbidities, and personal preference. The second-line treatment in myeloma includes new generation of proteasome inhibitors and immunomodulators, CD38 monoclonal antibodies, Panobinostat, and Elotuzumab. Recent randomized trials have shown that triplet combinations incorporating CD38 monoclonal antibodies, dexamethasone along with either proteasome inhibitor or immunomodulator were superior to doublet combinations in terms of response rate and progression-free survival. The choice of the second-line therapy is determined by lenalidomide/bortezomib exposure and resistance and access to new agents. Furthermore, autologous transplantation should be considered in selected cases. Here, we will be discussing the optimal management of multiple myeloma in the first relapse.

Keywords

  • multiple myeloma
  • relapse
  • novel agents in myeloma

1. Introduction

Multiple myeloma (MM) is a neoplastic proliferation of plasma cells accounting for 10% of hematologic malignancies [1]. An induction regimen using a combination of immunomodulatory drugs, proteasome inhibitors, and dexamethasone followed by autologous stem cell transplantation (ASCT) is considered standard treatment for newly diagnosed myeloma in physically fit patients [2]. In the era of novel therapies, several randomized trials have proved improved progression-free survival (PFS) and overall survival (OS) in favor of use of novel therapies in a combination of ASCT with maintenance therapy [3]. Despite these advances, MM remains an incurable disease and the majority of patients continue to relapse and will require additional treatment [4]. Factors related to poor outcomes include lack of response, high-risk cytogenetics, stage, age, presence of extramedullary disease, and circulating plasma cells, and co-morbidities and functional status are linked to bad prognosis [5].

Advertisement

2. Definitions of relapsed and relapsed/refractory myeloma

The International myeloma working group (IMWG) published and revised the definitions of relapsed MM in 2015. Relapsed MM is defined as a recurrence of disease after prior response on the basis of objective laboratory and radiological criteria:

  • ≥25% increase of the monoclonal protein (M-protein) in serum (absolute increase ≥0.5 g/dl) or urine (absolute increase ≥200 mg /d) or

  • ≥25% difference between involved and uninvolved serum free light chains (absolute increase >10 g/L) or

  • >10% increase of the absolute percentage of the bone marrow plasma cells or

  • Development of new (extramedullary) plasmacytomas or hypercalcemia.

Relapsed/refractory MM (RRMM) is defined as a disease that becomes nonresponsive or progressive on therapy or within 60 days of the last treatment in patients who had achieved a minimal response or better on prior therapy [6]. Furthermore, the IMWG consensus defined the relapse of MM based on the clinical aggressiveness as shown in Table 1.

Non-aggressive relapseAggressive relapse
Biochemical relapseSymptomatic relapse
Progression based on increased M-protein
No associated symptoms or myeloma related organ dysfunction
Slowly increasing M protein and slow onset of clinical symptoms
Progressive disease with prominent symptoms
Short duration of response or progression while on therapy
Aggressive clinical progression includes:
Rapid onset of symptoms
Extensive disease on radiologic, laboratory, or pathologic findings
Circulatory plasma cells
ISS stage II/III at relapse
Isotype transformation (light chain disease or hypo secretory disease)
Adverse cytogenetic abnormalities;
t 14;4, del 17p, hypodiploidy
High B2 microglobulin (>5.5 g/L) or low albumin (<3.5 g/ L), high LDH
Presence of extramedullary disease
Disease-associated organ impairment

Table 1.

The IMWG consensus defined the relapse of MM based on the clinical aggressiveness.

Advertisement

3. Diagnosis of relapse

At relapse, the diagnostic assessment should include the full routine workup of MM, including complete blood count and differential, serum electrolyte, renal and liver function, serum and urine electrophoresis with immunofixation, serum free light chain assay, and 24-hour urine for protein. Bone marrow evaluations are highly recommended (especially in non or oligosecretory MM). BM examination should include morphology and fluorescence in situ hybridization (FISH) on CD138 selected plasma cells to detect cytogenetically unfavorable abnormalities that require an intensive approach with a combination of maintenance therapy and other abnormalities that predict response to therapy (Venetoclax) such as t(11;14) [7].

Imaging evaluation is recommended to all patients (Pts) at relapse and this includes low dose whole body computed tomography (CT) scan or whole spine magnetic resonance imaging (MRI) in cases of relapsed smoldering MM to detect any focal lesion or FDG positron emission tomography combined with computed tomography (PET/CT) in cases of suspected extramedullary relapse [8].

Advertisement

4. Predictive factors for early relapse

The result of Pourmoussa et al study in 2019 has shown that achievement of complete response (CR) before transplant may help to prevent early relapse or progression of the disease, which was in accordance with prior observations where achievement of CR or very good partial response before autologous stem cell transplantation translated to a better long-term outcome [9]. There is a strong association between high-risk cytogenetic by FISH results such as del(17p) and/or t(4;14) and/or t(14;16), high lactate dehydrogenase (LDH) and serum albumin (<3.5 g/L) are predictive of early relapse [10]. Furthermore, there was a strong relation between Freiberg comorbidity index (FCI) and early relapse and progression partly due to poor tolerance to treatment [11]. Minimal residual disease (MRD) positivity at the end of induction and post consolidation or transplantation is strongly associated with inferior outcomes and early relapse [12].

Advertisement

5. Prognostic factors at time of relapse

There are several prognostic markers indicative of an aggressive relapse as shown in Table 2. Patients who experienced a primary refractory disease, or relapsed within 112 months of initial diagnosis, usually have a poor prognosis [13]. Relapse with prior lenalidomide exposure usually indicates a poor prognosis and short progression-free survival (PFS) [14]. Patients with extramedullary or secondary plasma cell leukemia (sPCL) tend to have dismal outcomes [15, 16].

Parameters
Disease-related parameters (parameters associated with poor prognosis)
  • High ISS

  • High risk cytogenetics (t (4;14), t(14;16), 17p del

  • Extramedullary disease

  • Short response to prior therapy (less than 12 months)

  • Aggressive relapse (rapid onset hypercalcemia, renal failure)

Patient and treatment-related
  • Age

  • Co-morbidities

  • Performance status (EGOG or frailty index)

  • Failure to achieve VGPR or more

  • Detectable MRD

  • Previous ASCT or not

  • Lenalidomide/Bortezomib refractoriness or intolerance

  • Number and toxicity of prior lines of treatment

Table 2.

prognostic markers indicative of an aggressive relapse.

Advertisement

6. Management of early/first relapse

6.1 General considerations

Myeloma treatment has evolved during the past decade to include multiple immunomodulatory agents, proteasome inhibitors, and monoclonal antibodies. The choices of treatment can be guided by disease biology and the nature of relapse (biochemical vs clinically aggressive) and prior lines of treatment.

Autologous stem cell transplantation remains a mainstay for patients who elect to defer transplantation as initial therapy [17]. The main classes of drugs in multiple myeloma include proteasome inhibitors, immunomodulatory agents, and monoclonal antibodies primarily anti CD38 monoclonal antibodies (daratumumab and Isatuximab) and Elotuzumab (targets SLAMF7). The choice of regimen depends on response and prior therapies. It is preferable to class switch if needed or uses next generation of the same class.

Evaluating indolent versus aggressive relapse is critical since patient with mild biochemical relapse might not require switching therapy as discussed before. Patients who experience a biochemical relapse may be treated by increasing the medication dose if they are on maintenance lenalidomide, reintroducing dexamethasone, and/or adding another agent. While patients who develop aggressive relapses, such as extramedullary disease, may require special approach with multiagent chemoimmunotherapy.

Assessing frailty and comorbidities is crucial in deciding the choice of therapy. It is generally recommended to use a triple combination; however, this might not be appropriate in extremely frail patients, therefore, a doublet combination might be used.

Psychosocial issues and access to care are important in the relapsed setting, especially in older patients or with relapsed myeloma with comorbidities. Patients who have no access to transportation can be treated at home with oral treatment whenever possible [18].

6.1.1 Indications of treatment at relapse

The goal of relapse treatment is to relieve disease symptoms, prevent new organ damage, and achieve a second lasting disease remission. Second and later remissions tend to be shorter because the disease may be more aggressive owing to the presence of different clones, which represent refractory disease [19, 20].

Indications to start treatment at relapse have been defined as clinical or significant relapse as defined by the IMWG [21] as shown in Table 3. The choice of salvage regimen is based on lenalidomide/bortezomib resistance, CD38 monoclonal antibody availability, and access. ASCT is done in specific scenarios as per standard recommendations (to be discussed below).

Type of relapseIndications
Clinical relapseDevelopment of new soft-tissue plasmacytomas or bone lesions
  • Definite increase (≥50%) in size of existing plasmacytomas or bone lesions

  • Hypercalcemia (≥11.5 mg/dL; 2.875 mmol/L)

  • Decrease in hemoglobin of ≥2 g/dL (1.25 mmol/L) or of10 g/dL because of myeloma

  • Rise in serum creatinine by ≥2 mg/dL or more (≥177 mmol/L), due to myeloma

  • Hyperviscosity requiring therapeutic intervention

Significant biochemical relapse in patients without clinical relapseDoubling of the M-component in 2 consecutive measurements separated by 2 months with the reference value of 5 g/L, or
- In 2 consecutive measurements, any of the following increases:
  • The absolute levels of serum M-protein by ≥10 g/L, or

  • An increase of urine M-protein by ≥500 mg per 24 h, or

  • An increase of involved FLC level by $20 mg/dL (plus an abnormal FLC ratio) or a 25% increase (whichever is greater)

Table 3.

Indications to start treatment at relapse.

There are different protocols used in the first relapse refractory cases as summarized in Table 4. Incorporating CD38 monoclonal antibodies into the backbone of salvage therapy has been shown to be superior to historical controls in many clinical trials.

Study (median FU)Median PFSSafetyHazard ratioComments
CANDOR (27.8 m)
- D-Kd vs Kd
28.6 vs 15.2 monsHTN: 21 vs 15%
↓PLT: 25 vs 16%
PNA: 15 vs 9%
HR 0.59
p<0·0001
1–3 lines, HR CG 15% (50% failed CG !!), GFR ≳20, LVEF ≳40%
Len refractory 39%
POLLUX (44.3 m)
- D-Rd vs Rd
SR: NR vs 18.6 mons
HR: 26.3 vs 8.3 mons
-SR: 0.43; P < 0.0001
HR: 0.34; P = 0.0035
≳1 lines (1–11,), GFR > 30,
1 line (52%), 80% no prior len
CASTOR (40 m)
- D-Vd vs Vd
16.7 vs 7.1 mons-HR 0.31
P < .0001
1–9 lines (median 2)
len refractory 25%,
prior Velcade 65%
IKEMA (20.7 m)
- Isa-Kd vs Kd
NR vs 19 mons-HR 0·53
p=0·0007
1–3 lines, GFR ≳15, LVEF ≳40%
primary refractory excluded, HR CG 23%
No K or Dara, Len refractory 25%
OPTIMISUM (15.7 m)
- P-Vd vs Vd
ITT: 11.2 mons
1L Rx: 20.7 mons
↑neutropenia
↑thrombocytopenia
↑infection
ITT: 0.61; P < 0.0001
1L: 0.54; P = .0027
≳1 lines, GFR > 30
TOURMALINE
- Ixa-Rd vs Rd
ITT: 20.6 mons
1L Rx: 20.6 mons
-ITT: 0.73
1L: 0.83
≳1 lines, GFR > 30
No G2PN, Exc. Len or PI ref
ELOQUENT-2
- Elo-Rd vs Rd
N=321 pts
19.4 vs 14.9 mons
  • Lymphocytopenia, neutropenia, fatigue, and pneumonia

  • Infusion-related reactions (IRR) 10% mostly Grade 1/2

Table 4.

Cross trial comparison of different protocols.

Patients who are lenalidomide exposed or sensitive seem to have the best outcomes from daratumumab, lenalidomide, and dexamethasone (D-Rd) combination as shown by POLLUX trial [22], with a median follow-up of 44.3 mons, the median progression-free survival (PFS) not reached and 25.3 months for standard risk and high-risk patients, respectively. In patients with lenalidomide refractoriness, the use of isatuximab (IKEMA, Isa-Kd) [23] and daratumumab (CANDOR trial, Dara-Kd) [24] based combination with carfilzomib and dexamethasone best outcomes. In CANDOR trial, with a median follow-up of 27.8 mons the median PFS was 28.6 mons (Hazard ratio HR 0.59, P < 0.0001%), while the IKEMA trial has shown a median PFS not reached with a median follow up of 20.7 mons. Daratumumab, bortezomib, and dexamethasone (Dara-Vd) as per the CASTOR trial, with a median follow-up of 40 mons, the median PFS was 16.7 months, and HR of 0.31 (P < 0.0001) [25].

Another group of monoclonal antibodies called anti SLAMF7 (signal lymphocyte activation molecule F7) has been evaluated in a phase 3 trial, Elotuzumab, lenalidomide, and dexamethasone (Elo-Rd) vs Rd have shown a median PFS benefit of 19.4 mons vs 14.9 mons (HR 0.70, P<0.001%) [26].

In phase 3 trial, evaluating the use of pomalidomide, bortezomib, and dexamethasone (P-Vd) vs bortezomib and dexamethasone (Vd) at a median follow-up of 15.7 mons, median PFS for patients who had one prior line of therapy was 20.7 mons in favor of P-Vd (HR 0.54, P=0.0027) [27]. TOURMALINE trial, which evaluated Ixazomib, lenalidomide, and dexamethasone (I-Rd) vs Rd showed a median PFS of 20.6 months in favor of I-Rd with an HR of 0.83 [28], although there was no statistically significant difference in overall survival with the addition of ixazomib to the combination, this might be confounded by the subsequent therapies [29].

High-dose chemotherapy and ASCT can be used in the first relapse for fit patients who experienced a prolonged PFS after the first transplant or those who never had a transplant before as summarized in Table 5 [7, 13, 30].

ESMO 2021:
  • Initial remission duration of ≥ 36 months.

mSMART 2020: if eligible
  • Consider salvage auto SCT who have not had it before;

  • Consider 2nd auto SCT

  • Remission ≥ 18 months unmaintained or

  • Maintained response to first ASCT of ≥ 36 months

EBMT/ASTCT 2015:
  • Initial remission duration of ≥ 18 months

Table 5.

Role of autologous stem cell transplant in the first relapse.

Advertisement

7. Treatment of relapse in special scenarios

7.1 Renal failure

Renal failure is commonly seen in patients with multiple myeloma at the first diagnosis, however, it is less common in the relapse if the patient was followed up regularly because antecedent biochemical relapse is seen before clinical relapse. However, If the patient has renal impairment, it is crucial to note that almost all clinical trials have excluded patients with renal impairment (estimated glomerular filtration rate eGFR) [31].

Proteasome inhibitors do not need dose modification except for Ixazomib. Immunomodulators (pomalidomide and thalidomide) do not need dose modification but lenalidomide does. CD38 monoclonal antibodies do not need dose modification. Alkylating agents do need dose modification. In summary, we need to check the dosing schedule as per eGFR for the patient.

7.2 Extramedullary relapse (EMD) or secondary plasma cell leukemia (sPCL)

EMD and sPCL usually indicate an aggressive disease and carry a dismal prognosis with a median overall survival (OS) of 6 mons (EMD) [15] and 4.3 mon (sPCL) [16]. It required incorporating chemotherapy i.e. VTD-PACE followed by ASCT if meets the criteria as discussed before. The use of CD38 monoclonal antibodies such as daratumumab has not shown to improve outcomes in this group of patients [32].

Central nervous system (CNS) involvement at relapse correlates with poor outcomes due to its resistance to several treatments. The frequency of CNS involvement is only approximately 1% [33]. Immunomodulators (IMIDs) and daratumumab have been shown to have a good penetration to the blood-brain barrier (BBB) and are effective in CNS myeloma cases as shown in Table 6 [34]. Use of intrathecal chemotherapy has shown efficacy in combination with anti-myeloma treatment, however, dosing, frequency, and duration of therapy are not well defined [35]. Myeloma cells are usually radiosensitive [36], therefore combining radiotherapy with chemotherapy can be more effective than if used alone [37].

Class of therapyAgentsBBB penetrationCNS Myeloma
ImmunomodulatorThalidomide
Lenalidomide
Pomalidomide
Good
Good
Good
Effective
Effective
Effective
Proteasome inhibitorBortezomib
Carfilzomib
Ixazomib
Poor
Poor
Poor
Ineffective
No data
No data
CD38 monoclonalDaratumumab
Isatuximab
Good
No data
Effective
No data

Table 6.

Anti-myeloma efficacy in CNS myeloma.

Advertisement

8. Summary and recommendations

  • Patients with multiple myeloma at the first relapse should undergo a full clinical, laboratory, and radiological evaluation.

  • It is crucial to differentiate between clinical and biochemical relapse and determine the aggressiveness of the relapse.

  • The choice of treatment is based on the disease biology, prior treatment, and aggressiveness of the relapse.

  • ASCT can be offered to a specific group of patients who are fit and had a prolonged duration of remission with the first transplant or those who are transplant naïve.

  • The use of monoclonal antibodies yields the best outcomes in the first relapse of multiple myeloma patients

  • For lenalidomide refractory patients, Dara-KD, Isa-KD, KPD, or PVD can be used.

  • For lenalidomide sensitive patients: Dara-Rd, Dara-KD, Dara-Pd, Isa-Kd, Isa-Pd, KRd, KPd, or Elo-Rd

References

  1. 1. Sychra V, Esser D, Kosmehl H, Herold M. Unusual manifestation of a multiple myeloma in the hyoid bone. Dento Maxillo Facial Radiology. 2013;42(3):27101530
  2. 2. Kumar SK, Callander NS, Alsina M, Atanackovic D, Biermann JS, Chandler JC, et al. Multiple myeloma, Version 3.2017, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network. 2017;15(2):230-269
  3. 3. Attal M, Lauwers-Cances V, Marit G, Caillot D, Moreau P, Facon T, et al. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. The New England Journal of Medicine. 2012;366(19):1782-1791
  4. 4. Sonneveld P. Management of multiple myeloma in the relapsed/refractory patient. Hematology. American Society of Hematology. Education Program. 2017;2017(1):508-517
  5. 5. Saad A, Mahindra A, Zhang MJ, Zhong X, Costa LJ, Dispenzieri A, et al. Hematopoietic cell transplant comorbidity index is predictive of survival after autologous hematopoietic cell transplantation in multiple myeloma. Biology of Blood and Marrow Transplantation. 2014;20(3):402-408
  6. 6. Anderson KC, Kyle RA, Rajkumar SV, Stewart AK, Weber D, Richardson P, et al. Clinically relevant end points and new drug approvals for myeloma. Leukemia. 2008;22(2):231-239
  7. 7. Dimopoulos MA, Moreau P, Terpos E, Mateos MV, Zweegman S, Cook G, et al. Multiple myeloma: EHA-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up(dagger). Annals of Oncology. 2021;32(3):309-322
  8. 8. Caers J, Garderet L, Kortum KM, O’Dwyer ME, van de Donk N, Binder M, et al. European Myeloma Network recommendations on tools for the diagnosis and monitoring of multiple myeloma: What to use and when. Haematologica. 2018;103(11):1772-1784
  9. 9. Gertz MA, Kumar S, Lacy MQ , Dispenzieri A, Dingli D, Hayman SR, et al. Stem cell transplantation in multiple myeloma: Impact of response failure with thalidomide or lenalidomide induction. Blood. 2010;115(12):2348-2353
  10. 10. 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
  11. 11. Pourmoussa AM, Spielberger R, Cai J, Khoshbin O, Farol L, Cao T, et al. Predictive factors for early relapse in multiple myeloma after autologous hematopoietic stem cell transplant. The Permanente Journal. 2019;23
  12. 12. Mina R, Bonello F, Oliva S. Minimal residual disease in multiple myeloma: Ready for prime time? Cancer Journal. 2021;27(3):247-255
  13. 13. Dingli D, Ailawadhi S, Bergsagel PL, Buadi FK, Dispenzieri A, Fonseca R, et al. Therapy for relapsed multiple myeloma: Guidelines from the Mayo stratification for myeloma and risk-adapted therapy. Mayo Clinic Proceedings. 2017;92(4):578-598
  14. 14. Lecat CSY, Taube JB, Wilson W, Carmichael J, Parrish C, Wallis G, et al. Defining unmet need following lenalidomide refractoriness: Real-world evidence of outcomes in patients with multiple myeloma. Frontiers in Oncology. 2021;11:703233
  15. 15. Avivi I, Cohen YC, Suska A, Shragai T, Mikala G, Garderet L, et al. Hematogenous extramedullary relapse in multiple myeloma – A multicenter retrospective study in 127 patients. American Journal of Hematology. 2019;94(10):1132-1140
  16. 16. Jurczyszyn A, Castillo JJ, Avivi I, Czepiel J, Davila J, Vij R, et al. Secondary plasma cell leukemia: A multicenter retrospective study of 101 patients. Leukemia & Lymphoma. 2019;60(1):118-123
  17. 17. Attal M, Lauwers-Cances V, Hulin C, Leleu X, Caillot D, Escoffre M, et al. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. The New England Journal of Medicine. 2017;376(14):1311-1320
  18. 18. Nathwani N, Bertamini L, Banerjee R, Gay F, Shah N, Krishnan A. When and how to treat relapsed multiple myeloma. American Society of Clinical Oncology Educational Book. 2021;41:358-375
  19. 19. Borrello I. Can we change the disease biology of multiple myeloma? Leukemia Research. 2012;36(Suppl. 1):S3-S12
  20. 20. Richardson PG, San Miguel JF, Moreau P, Hajek R, Dimopoulos MA, Laubach JP, et al. Interpreting clinical trial data in multiple myeloma: Translating findings to the real-world setting. Blood Cancer Journal. 2018;8(11):109
  21. 21. Moreau P, Kumar SK, San Miguel J, Davies F, Zamagni E, Bahlis N, et al. Treatment of relapsed and refractory multiple myeloma: Recommendations from the International Myeloma Working Group. The Lancet Oncology. 2021;22(3):e105-ee18
  22. 22. Kaufman JL, Dimopoulos MA, White D, Benboubker L, Cook G, Leiba M, et al. Daratumumab, lenalidomide, and dexamethasone in relapsed/refractory myeloma: A cytogenetic subgroup analysis of POLLUX. Blood Cancer Journal. 2020;10(11):111
  23. 23. Moreau P, Dimopoulos MA, Mikhael J, Yong K, Capra M, Facon T, et al. Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): A multicentre, open-label, randomised phase 3 trial. Lancet. 2021;397(10292):2361-2371
  24. 24. Usmani SZ, Quach H, Mateos MV, Landgren O, Leleu X, Siegel D, et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): Updated outcomes from a randomised, multicentre, open-label, phase 3 study. The Lancet Oncology. 2022;23(1):65-76
  25. 25. Mateos MV, Sonneveld P, Hungria V, Nooka AK, Estell JA, Barreto W, et al. Daratumumab, bortezomib, and dexamethasone versus bortezomib and dexamethasone in patients with previously treated multiple myeloma: Three-year Follow-up of CASTOR. Clinical Lymphoma, Myeloma & Leukemia. 2020;20(8):509-518
  26. 26. Lonial S, Dimopoulos M, Palumbo A, White D, Grosicki S, Spicka I, et al. Elotuzumab therapy for relapsed or refractory multiple myeloma. The New England Journal of Medicine. 2015;373(7):621-631
  27. 27. Richardson PG, Oriol A, Beksac M, Liberati AM, Galli M, Schjesvold F, et al. Pomalidomide, bortezomib, and dexamethasone for patients with relapsed or refractory multiple myeloma previously treated with lenalidomide (OPTIMISMM): A randomised, open-label, phase 3 trial. The Lancet Oncology. 2019;20(6):781-794
  28. 28. Moreau P, Masszi T, Grzasko N, Bahlis NJ, Hansson M, Pour L, et al. Oral Ixazomib, lenalidomide, and dexamethasone for multiple myeloma. The New England Journal of Medicine. 2016;374(17):1621-1634
  29. 29. Richardson PG, Kumar SK, Masszi T, Grzasko N, Bahlis NJ, Hansson M, et al. Final overall survival analysis of the TOURMALINE-MM1 Phase III Trial of Ixazomib, lenalidomide, and dexamethasone in patients with relapsed or refractory multiple myeloma. Journal of Clinical Oncology. 2021;39(22):2430-2442
  30. 30. Giralt S, Garderet L, Durie B, Cook G, Gahrton G, Bruno B, et al. American Society of Blood and Marrow Transplantation, European Society of Blood and Marrow Transplantation, Blood and Marrow Transplant Clinical Trials Network, and International Myeloma Working Group Consensus Conference on Salvage Hematopoietic Cell Transplantation in Patients with Relapsed Multiple Myeloma. Biology of Blood and Marrow Transplantation. 2015;21(12):2039-2051
  31. 31. Heher EC, Rennke HG, Laubach JP, Richardson PG. Kidney disease and multiple myeloma. Clinical Journal of the American Society of Nephrology. 2013;8(11):2007-2017
  32. 32. Jelinek T, Sevcikova T, Zihala D, Popkova T, Kapustova V, Broskevicova L, et al. Limited efficacy of daratumumab in multiple myeloma with extramedullary disease. Leukemia. 2022;36(1):288-291
  33. 33. Nieuwenhuizen L, Biesma DH. Central nervous system myelomatosis: Review of the literature. European Journal of Haematology. 2008;80(1):1-9
  34. 34. Hotta M, Ito T, Konishi A, Yoshimura H, Nakanishi T, Fujita S, et al. Multiple myeloma with central nervous system relapse early after autologous stem cell transplantation: A Case Report and Literature Review. Internal Medicine. 2021;60(3):463-468
  35. 35. Lee D, Kalff A, Low M, Gangatharan S, Ho P, Bajel A, et al. Central nervous system multiple myeloma--potential roles for intrathecal therapy and measurement of cerebrospinal fluid light chains. British Journal of Haematology. 2013;162(3):371-375
  36. 36. Quach H, Ryan G, Ganju V, Prince HM. Effective treatment of leptomeningeal multiple myeloma with total craniospinal irradiation supported by second allogeneic donor stem cell infusion. Bone Marrow Transplantation. 2005;35(4):423-424
  37. 37. Gozzetti A, Cerase A, Lotti F, Rossi D, Palumbo A, Petrucci MT, et al. Extramedullary intracranial localization of multiple myeloma and treatment with novel agents: A retrospective survey of 50 patients. Cancer. 2012;118(6):1574-1584

Written By

Ahmad Alhuraiji, Dina Abd El Razik and Shaza A.A. Elkourahy Omar

Submitted: 05 July 2022 Reviewed: 01 August 2022 Published: 30 October 2022