Open access peer-reviewed chapter

An Update on Hematopoietic Stem Cell Transplantation in Patients with Multiple Myeloma

Written By

Khalid Ahmed Al-Anazi, Ziyad Alshaibani and Panagiotis Kalogianidis

Submitted: 26 May 2022 Reviewed: 17 November 2022 Published: 09 January 2023

DOI: 10.5772/intechopen.109059

From the Edited Volume

Recent Updates on Multiple Myeloma

Edited by Khalid Ahmed Al-Anazi

Chapter metrics overview

128 Chapter Downloads

View Full Metrics

Abstract

Over the past two decades, treatment of multiple myeloma (MM) has advanced dramatically. However, despite the introduction of several lines of novel therapeutics, autologous hematopoietic stem cell transplantation (HSCT) followed by maintenance therapy is the current standard of care in transplant eligible patients. Autologous HSCT can be performed with or without cryopreservation with equivalent short-term and long-term outcomes. In patients with MM, performance of autologous HSCT at outpatient setting is safe, feasible and has a number of advantages such as saving hospital beds and reducing treatment costs. Autologous HSCT can be safely performed in patients with MM having renal dysfunction or failure although particular attention should be made to the timing of administering medications and stem cells with respect to hemodialysis and dose reduction of specific medications according to creatinine clearance. Tandem autologous HSCT is of value in younger patients with adverse cytogenetics and extramedullary disease. Allogeneic HSCT is the only potentially curative therapeutic modality in MM, but it can only be performed in a small fraction of highly selected patients due to the relatively high treatment-related morbidity and mortality. Despite its valuable role in the treatment of MM, autologous HSCT has its own short-term as well as long-term complications.

Keywords

  • multiple myeloma
  • hematopietic stem cell transplantation
  • cryopreservation
  • maintenance therapy

1. Introduction

MM accounts for 1% of all cancers and 10–15% of all hematologic malignancies [12]. It is a disease of old age with the median age at diagnosis ranging between 65 and 74 years in the United States of America (USA) and Europe [1, 2, 3, 4]. The 5 years survival not only in the USA but also globally has more than doubled over the past decades due to the availability of several lines of novel therapeutic agents, HSCT, advancements in diagnostic techniques, and general improvement in health care [4, 5, 6]. The diagnostic criteria for multiple myeloma (MM) and staging of the disease according to the revised international staging system (RISS) are shown in Tables 1 and 2, respectively [1, 2, 4]. Presence of the following cytogenetic and molecular abnormalities: del(17p), t(4;14), t(14;16), t(14;20), gain 1q, or p53 mutation implies high-risk (HR) MM. Additionally, the presence of any two HR factors is considered double-hit myeloma; three or more HR factors are triple-hit MM [1].

1.≥ 10% clonal bone marrow (BM) plasma cells or a biopsy-proven plasmacytoma
and
2.Evidence of one or more multiple myeloma-defining events namely:
  1. CRAB (hypercalcemia, renal failure, anemia, or lytic bone lesions)

    features felt related to the plasma cell disorder

  2. BM clonal plasmacytosis ≥60%

  3. Serum involved/uninvolved free light chain ratio (FLC) ≥ 100

    (provided that involved FLC is ≥100 mg/L)

  4. More than one focal lesion on magnetic resonance imaging

Table 1.

Diagnostic criteria for multiple myeloma.

Stage IAll of the following:
  1. Serum albumin ≥3.5 g/ dL

  2. Serum beta 2 microglobulin (B2M) < 3.5 mg/L

  3. Normal serum lactic dehydrogenase (LDH)

  4. No high-risk cytogenetic abnormalities.

Stage II
  1. Not fitting stages I and III.

  2. Serum B2M: 3.5–5.5 mg/L.

Stage IIIAll of the following:
  1. Serum B2M > 5.5 mg/L

  2. High-risk cytogenetic abnormalities or elevated serum LDH level

Table 2.

Staging of multiple myeloma according to the revised international staging system (RISS).

The treatment of MM has advanced dramatically in the past two decades [7]. Induction therapy with a proteasome inhibitor, an immunomodulatory agent, and dexamethasone followed by autologous hematopoietic stem cell transplantation (HSCT), and maintenance therapy with lenalidomide are among the treatments that are considered the standard care for standard risk (SR) and eligible patients [89]. The triplet regimen of bortezomib, lenalidomide, and dexamethasone (VRd) is recommended as the standard first-line treatment, although the addition of a fourth drug can improve efficacy and survival. In transplant-eligible patients, 3–4 cycles of VRd induction therapy can be administered prior to HSCT while in HR patients, daratumumab, bortezomib, lenalidomide, dexamethasone (Dara-VRd) is an alternative to VRd [1, 10, 11, 12, 13, 14]. Selected SR patients can receive additional cycles of induction, and delay in transplant until the first relapse [1]. After autologous HSCT, SR patients need lenalidomide maintenance, while bortezomib-based maintenance is needed for patients with HR myeloma [15, 16]. The role of a 4-drug induction regimen is still being defined but can be considered for patients with HR disease [1, 7, 10, 12]. For patients who are eligible to undergo HSCT, this option is of value in case the transplant is delayed or refused by the patient [8]. Patients who are not candidates for transplant are typically treated with VRd, for approximately 8–12 cycles followed by lenalidomide maintenance. Alternatively, these patients can be treated with the triplet regimen: daratumumab, lenalidomide, dexamethasone (DRd), or the quadruplet regimen: daratumumab, bortezomib, melphalan, and prednisolone (D-VMP) [1, 17, 18, 19].

Unfortunately, nearly all MM patients ultimately relapse, even those who experience a complete response (CR) to initial therapy [2]. In patients with relapsed disease, it is important to switch treatment to new drug classes; for this, multiple combinations can be recommended [8]. Management of the relapsed disease remains a critical aspect of MM care and an important area of ongoing research [2]. In case of refractory disease, most patients require a triplet regimen at relapse, with the choice of regimen varying with each successive relapse [1]. The updated National Comprehensive Cancer Network (NCCN) guidelines include new drugs for refractory disease such as selinexor and belantamab mafodotin which are listed as other regimens [8]. For relapsed/refractory myeloma (RRMM) patients, novel agents such as selinexor and venetoclax are superior to bortezomib. Also, chimeric antigen receptor (CAR)-T cells and other cell-surface-targeted therapies appear promising [7].

Advertisement

2. Autologous HSCT in patients with MM

Since the mid-1990s and despite the recent availability of several lines of novel agents, high-dose (HD) melphalan followed by autologous HSCT is still the standard of care for newly diagnosed patients with MM who are eligible for autologous HSCT [20, 21, 22, 23]. The long-term outcome of patients with MM subjected to autologous HSCT has improved significantly over the last three decades [1, 24]. Nishimura KK et al. reported the long-term outcomes of a total of 4329 patients with newly diagnosed MM treated with autologous HSCT using cryopreserved stem cells at the university of Arkansas in the USA between 1989 and 2014 [24]. The 5 years progression-free survival (PFS) for the entire population of autologous HSCT recipients had improved from 29–68% and the overall survival (OS) for the entire population of autologous HSCT recipients had improved over that period of time from 47–70%, respectively [24]. Eligibility for autologous HSCT is determined by age, performance status, presence and severity of comorbid medical conditions, and frailty score as frailty has been shown to be a predictor of short survival and is considered an exclusion criterion for autologous HSCT [25, 26, 27]. Cryopreservation of hematopoietic stem cells is routinely employed in the setting of autologous HSCT [23].

Melphalan is the standard chemotherapeutic agent that is used in conditioning therapy prior to autologous HSCT in MM [20, 23]. According to creatinine clearance, the dose ranges between 140 and 200 mg/m2, and the drug is administered intravenously (IV) [23, 28, 29]. However, large interpatient variability in melphalan exposure exists among MM patients undergoing autologous HSCT. Additionally, higher melphalan exposure has been shown to improve survival at the expense of increased but acceptable transplant-related toxicities. So, it is recommended to apply pharmacokinetic testing and individualized dosing of melphalan in MM patients undergoing autologous HSCT [30]. In patients with MM having renal impairment, several studies have shown that: (1) conditioning therapy with melphalan 140 mg/m2 has acceptable toxicity and is equally effective to a melphalan dose of 200 mg/m2, and (2) melphalan dose adjustment is not needed in patients having renal failure subjected to autologous HSCT [31, 32, 33, 34, 35, 36, 37, 38]. However, in patients with MM having renal impairment subjected to autologous HSCT: (1) it is advisable to reduce the dose of melphalan by 25% in patients having creatinine clearance between 10 and 45 ml/minute and by 15% in patients having creatinine clearance between 46 and 60 ml/min, respectively; and (2) melphalan dose of 200 mg/m2 is safe and effective in patients having creatinine clearance between 30 and 60 ml/minute [30, 39]. In patients with MM having end-stage renal disease (ESRD) receiving hemodialysis, careful evaluation prior to autologous HSCT with the involvement of a multidisciplinary team should be made and dose adjustment for all drugs that adversely affect renal function should be taken into consideration [40, 41].

The doses of granulocyte colony-stimulating factor (G-CSF) and plerixafor; which is used in case of poor mobilization in heavily pretreated patients with MM; in stem cell mobilization prior to autologous HSCT are as follows: (1) G-CSF: 5 μg per kilogram (kg) body weight twice daily subcutaneously (SC) twice daily [ie 10 μg/kg/day] for 4–5 days, and (2) plerixafor: 0.24 mg/kg SC, one dose to be given the night before stem cell collection [42, 43, 44, 45]. The doses of cyclophosphamide in stem cell mobilization prior to autologous HSCT are as follows: (1) low dose: 1.0 to 1.5 g/m2 IV; (2) intermediate dose: 3.0 to 4.0 g/m2 IV; and (3) high dose: 5.0 to 7.0 g/m2 IV [23, 46, 47, 48, 49].

Advertisement

3. Autologous HSCT without cryopreservation

Melphalan is cleared from plasma and urine in 1 and 6 hours, respectively. Hence, stem cells can be safely infused as early as 8–24 hours following melphalan administration [23, 50]. Since 1957, there have been preclinical data supporting the use of non-cryopreserved HSCs. Also, studies on mice have reported successful rescue after the administration of lethal doses of total body irradiation and reinfusion of BM cells that had been stored for 11 days at 25°C [50]. Studies have shown that: (1) peripheral blood stem cells can be stored safely at 4°C for at least 5 days, while the patient receives HD chemotherapy; and (2) the viability of stem cells decreases progressively from day 5 onwards [23, 51]. Three studies that compared immediate cryopreservation of peripheral blood progenitor cell products and overnight storage showed that there was no statistically significant difference between the two groups regarding: viability of stem cells, neutrophil and platelet engraftment days, safety, and even long-term outcome of the primary disease. Additional benefits of overnight storage of stem cells were a reduction in costs and processing time [52, 53, 54].

Several studies and one meta-analysis have shown that non-cryopreserved autologous HSCT for MM is simple, safe, and cost-effective and gives results that are at least equivalent to autologous HSCT with cryopreservation [23, 50, 55]. Treatment-related mortality (TRM) at day 100 post-HSCT using non-cryopreserved autologous stem cells has ranged between 0.0 and 3.4% [28, 55]. Non-cryopreserved stem cells can be infused till day 5 post-apheresis without viability loss provided they are stored at +4°C in a conventional blood bank refrigerator [23, 28, 50]. In a systematic review that included 16 studies having 560 patients with various hematologic malignancies (HMs) including MM, hematopoietic engraftment was universal and only one graft failure was reported [23, 50]. Several old and more recent studies have shown that the median times of engraftment following non-cryopreserved autografts were 9–14 days for neutrophils and 13–25 days for platelets [23, 50, 56, 57, 58, 59, 60, 61, 62, 63]. Transplantation of non-cryopreserved stem cells may be of value in two scenarios: (1) use in medical institutions from areas with limited economic resources, that is, having the infrastructure to treat HMs but not cryopreservation facilities, and (2) use in medical institutions treating HMs and in the process of establishing an HSCT program that will eventually have cryopreservation [28, 50, 55, 64].

HSCT without cryopreservation has several advantages including (1) allowing autologous HSCT to be performed entirely as an outpatient due to the simplicity of its implementation, (2) decreasing transplantation costs and the time between the last induction therapy and HD chemotherapy, (3) prevention of dimethyl sulfoxide toxicity, (4) no significant loss of viability of the collected HSCs provided stem cell infusion is made within 5 days of apheresis, (5) expansion of the number of medical institutions performing stem cell therapies, and (6) potent engraftment syndrome and autologous graft versus host disease (GVHD) [23, 28, 50, 55, 65, 66, 67, 68]. HSCT without cryopreservation has the following disadvantages: (1) plenty of coordination is needed between various teams regarding the timing of stem cell mobilization, apheresis, administration of conditioning therapy, and infusion of stem cells; (2) limitation of the use of standard HD chemotherapy schedules such as BEAM (BCNU, etoposide, cytarabine, and melphalan) employed in the autologous HSCT for lymphoma, and (3) inability to store part of the collection and reserving it for a second autologous HSCT in case a rich product is obtained [23, 28, 50, 55].

Advertisement

4. Autologous HSCT in patients with MM having renal failure

Renal failure is one of the most common and most serious complications of MM that is associated with high mortality [41, 69, 70, 71, 72]. Renal impairment has been reported in 30–50% of patients with newly diagnosed MM, while renal failure occurs in 20–30% of MM patients. Additionally, renal impairment develops in 50% of patients with MM during the course of the disease, and approximately 5–10% of MM patients having renal failure at diagnosis are dialysis-dependent [34, 36, 37, 73, 74, 75]. Causes of renal dysfunction/failure in patients with MM include: light chain-induced proximal tubular damage, cast nephropathy, interstitial nephritis, dehydration, hypercalcemia, hyperuricemia, amyloid deposition, plasma cell infiltration, hyperviscosity, various infections, nephrotoxic drugs, and contrast media [36, 37, 72, 76]. The modalities of treatment of renal dysfunction/failure in MM patients include: hydration, treatment of infectious complications, withholding nephrotoxic drugs and contrast media, renal replacement therapy such as hemodialysis, removal of serum-free light chains by plasma exchange, use of high cut-off dialyzers, administration of anti-myeloma chemotherapy, HSCT for patients with controlled disease, and renal transplantation [37, 71, 75, 76, 77].

In patients with MM having dialysis-dependent renal failure, the use of induction therapy with novel agents and high cut-off dialyzers has resulted in an improvement of renal function due to the removal of large quantities of serum-free light chains [37, 75, 77]. Factors associated with a high probability of dialysis independence in patients with newly diagnosed MM having dialysis-dependent renal failure include: shorter duration of kidney disease, achieving at least very good partial response (VGPR), low beta 2 microglobulin at diagnosis, and low level of free light chains at diagnosis [70]. In patients with MM having renal dysfunction/failure, recovery of renal function depends on: the elimination of causes of renal dysfunction/failure, and timely induction therapy using novel agents such as bortezomib in addition to corticosteroids followed by autologous HSCT once the disease is under control [37, 70, 71, 73, 76]. In patients with MM having dialysis-dependent renal failure, HD chemotherapy and autologous HSCT have traditionally been contraindicated due to the following reasons: lower survival rates, higher short-term mortality, greater susceptibility to infectious complications, longer duration of hospitalization, greatly compromised quality of life, as well as predilection for the following complications: mucositis, cardiac arrhythmias, bleeding, and encephalopathy [34, 37, 72, 74, 78, 79].

Patients with MM having renal dysfunction and even those having ESRD receiving hemodialysis should not be excluded from autologous HSCT as several studies have proven not only the safety but also the efficacy of HD chemotherapy and autologous HSCT in this group of patients [35, 37, 40, 69, 74, 79, 80, 81]. Historically, the fist autologous HSCT performed for a patient with MM having renal insufficiency was reported in the year 1997 [82]. In patients with MM having renal impairment, studies have shown that: (1) induction therapy with almost all the combinations of novel agents such as VRd and bortezomib, cyclophosphamide and dexamethasone (VCD) results in the reversal of renal impairment in the majority of patients, and (2) despite the acceptable toxicity, consolidation with autologous HSCT can overcome the adverse impact of renal impairment on survival and may further improve renal function in at least one-third of patients [34, 37, 83, 84]. Factors associated with a high probability of recovery of renal function in patients having renal failure subjected to autologous HSCT include: being on hemodialysis for less than 6 months, and pre-transplant creatinine clearance >10 ml/minute [71].

In patients with MM having ESRD receiving hemodialysis, careful evaluation prior to HSCT with the involvement of a multidisciplinary team should be made and dose adjustment for all drugs that adversely affect renal function should be taken into consideration. In patients with MM having ESRD on hemodialysis, it is recommended to perform hemodialysis before and 24 hours after the administration of HD melphalan [78]. Additionally, in patients with MM having ESRD, combined HSCT and renal transplantation can be performed either simultaneously or sequentially after controlling MM by appropriate chemotherapy [37, 71, 85, 86].

The prognostic factors that imply good prognosis in patients with MM having severe renal impairment subjected to autologous HSCT include: (1) good performance status, (2) higher albumin concentration, (3) chemotherapy-responsive disease in the pre-HSCT period, (4) adjustment of melphalan dose to that of chronic kidney disease, and (5) intensive supportive care post-transplantation [87]. Autologous HSCT is a safe and effective therapeutic modality in patients with ESRD even those on regular hemodialysis [87, 88]. However, patients who demonstrate renal deterioration at one-year post-HSCT should be monitored closely as this predicts poor long-term survival [88]. In patients with MM subjected to autologous HSCT, autologous transplantation does not adversely affect renal function [89]. One study showed that peritoneal dialysis is safe in patients with MM having ESRD subjected to autologous HSCT [90].

In patients with MM having renal impairment, two studies have demonstrated that the use of bortezomib-containing therapeutic regimens in induction treatment as well as in maintenance therapy after autologous HSCT can overcome the negative prognostic impact of renal impairment in this group of patients [91, 92]. However, two other studies have shown the superiority of carfilzomib-based therapeutic regimens as compared to bortezomib-based treatment not only in improving renal function but also in offering better survival outcomes in patients with RRMM having various degrees of renal impairment [93, 94]. Novel agents have helped to widen the treatment options that are available for patients with renal impairment and RRMM, since dose adjustments are unnecessary with dexamethasone, bortezomib, carfilzomib, panobinostat, elotuzumab, pomalidomide, or daratumumab in patients with renal impairment [39, 95]. Pretransplant hemoglobin level and creatinine clearance represent important determinants of clinical outcomes after autologous HSCT conditioned with melphalan dose of 200 mg/m2. Patients having lower hemoglobin levels and creatinine clearance were reported to achieve longer treatment-free survival despite experiencing increased toxicity, likely due to higher melphalan exposure [96]. Finally, studies have shown that despite the requirement of hemodialysis at the time of autologous HSCT, patients with MM having ESRD may recover their renal function at least partially [97, 98]. So, in patients with MM having ESRD, it is recommended to perform autologous HSCT as early as possible [98].

Advertisement

5. Tandem autologous HSCT in MM

In the 1990s and in an era where conventional chemotherapy was the only available drug, the concept of up-front treatment with a tandem autologous HSCT was attempted to improve PFS and OS [99, 100]. Previous randomized trials had demonstrated improved outcomes with tandem transplantation in terms of PFS and OS even in patients who had not achieved a VGPR after the first transplant [20, 100].

In the era of novel drugs, clinical trials such as EMN02/HO 95 and StaMINA are needed to evaluate the impact of tandem transplantation [101, 102]. Although the results have to be interpreted with caution due to high drop-out rates, lack of use of novel therapy, and lack of subgroup analysis, the long-term analysis of the GMMG-HD2 trial that compared single versus tandem transplantation with conditioning with melphalan (200 mg/m2) showed non-inferiority of single transplantation compared to tandem in the sense that OS and EFS did not significantly differ and that the CR rates were significantly improved after the second transplantation [103]. The EMN02/HO95 trial which explored the result of tandem versus single transplantation in newly diagnosed MM patients showed that tandem transplantation improved the depth of the response by 25% with more than 50% of the patients achieving at least a CR and that PFS and OS were significantly improved after a second transplant, with approximately 30% reduction in the risk of death and progression [102]. Updated results of the EMN02/HO95 confirmed the improved 3-year PFS in tandem autologous HSCTs and showed the positive effect of tandem autologous HSCT in HR groups [102, 104]. So, the analysis concluded that double frontline autologous HSCT was superior to single autologous HSCT in terms of PFS and OS in all patients, particularly poor prognosis subgroups of patients [102, 104]. However, the StaMINA trial failed to show the superiority of tandem versus single transplant in the era of novel agents although more than 30% of patients randomized to tandem transplant did not receive the second transplant [101]. Overall, with the currently available data, a second autologous HSCT may be beneficial in HR patients including patients with adverse cytogenetics and RISS stage III disease [20].

In patients with newly diagnosed MM having HR cytogenetics and extramedullary disease, tandem autologous HSCT has been shown to overcome the expected poor outcome [105]. As compared with a single autologous HSCT after HD chemotherapy, tandem transplantation improves OS among patients with myeloma, especially those who do not have a VGPR after undergoing the first transplantation [106]. In comparison with a single autologous HSCT as up-front therapy for newly diagnosed MM, double autologous HSCT achieved superior CR or near CR (nCR) rate, relapse-free survival (RFS), and event-free survival (EFS), but failed to significantly prolong OS. Benefits offered by double autologous HSCT were particularly evident among patients who failed to achieve at least nCR after one auto-transplantation [107]. Whether tandem autologous transplantation will continue to provide benefits in this HR population with an extramedullary disease in an era of highly active induction regimens, cellular therapeutics, and effective maintenance therapy is an open question, but Gagelmann and colleagues have provided evidence that outcomes with a tandem transplant are superior to standard induction and a single transplant alone and should be weighed as an option taking into consideration the following factors: patient and disease characteristics, trial availability, and access to active triplet and quadruplet induction regimens [108]. A tandem autologous HSCT approach should be considered for all patients, although the benefit from the second autologous HSCT in patients who are in CR or experience a VGPR should be answered in a clinical trial. Recent results with the new induction regimens indicate that there is a role for tandem autologous HSCT in the presence of adverse cytogenetic abnormalities [109].

Tandem HSCT; with autologous HSCT followed by non-myeloablative allogeneic HSCT; is an effective therapy for HR or relapsed MM [110]. Planned allogeneic HSCT after autologous HSCT has not been found to be superior in the majority of studies and is not recommended outside a clinical trial. However, single or tandem autologous HSCT are both appropriate options and participation in prospective clinical trials should be encouraged to resolve the debate in the era of novel agents for MM [109]. After a median follow-up of more than 11 years, the prospective, randomized phase III trial (GMMG-HD2) that aimed to demonstrate non-inferiority of single versus tandem HD melphalan followed by autologous transplantation with regard to 2-year EFS in newly-diagnosed MM and which included 358 evaluable patients showed that HD melphalan followed by single autologous HSCT was non-inferior to tandem transplantation in newly diagnosed patients with MM [103].

In a phase II trial that evaluated, for the first time, the safety and efficacy of bendamustine plus HD melphalan as a conditioning regimen before the second autologous HSCT in previously untreated MM patients, it was shown that bendamustine plus HD melphalan is feasible as conditioning regimen for second autologous HSCT in MM patients [111]. In a study exploring the safety and efficacy of combining dose-intensified bendamustine (200 mg/m2 on days −4/−3) with HD melphalan (100 mg/m2 on days −2/−1) before a second (tandem) autologous HSCT in adverse risk MM patients after the first HD melphalan and autologous HSCT, dose-intensified bendamustine with melphalan conditioning was shown to be safe [112]. Additionally, thiotepa/melphalan is another feasible and safe conditioning regimen for autologous HSCT in MM and should be explored for efficacy in a phase III study [111, 113].

A systematic review and a meta-analysis that included all phase 3 randomized clinical trials evaluating the role of HD therapy followed by autologous HSCT showed that: (1) both HD therapy followed by tandem autologous HSCT and HD therapy followed by single autologous HSCT plus bortezomib, lenalidomide, and dexamethasone were superior to HD therapy followed by single autologous HSCT alone and standard-dose therapy (SDT) for PFS, and (2) for PFS, HD therapy followed by tandem autologous HSCT had the most favorable hazard ratio followed by HD therapy and single autologous HSCT plus bortezomib, lenalidomide, and dexamethasone [114].

However, in the era of novel agents; where novel anti-myeloma drugs are used in induction as well as maintenance therapy; the use of novel therapies might decrease the need for a second transplant and tandem transplantation may not improve OS or PFS in either SR MM or HR MM patients compared to a single transplant [115, 116]. Additionally, the alternative treatment approach to tandem autologous HSCT which is the total therapy 3 (TT3) that includes induction, tandem autologous HSCT, consolidation, and maintenance, has allowed one of the best outcomes in terms of CR/nCR, OS, and PFS [117]. Therefore, induction therapy with novel agents followed by single autologous HCT and maintenance therapy should remain as the standard of care for newly diagnosed MM patients who are transplant eligible [115, 116].

Advertisement

6. Outpatient autologous HSCT in MM

While historically, due to logistic issues and concerns regarding toxicities and infections, most of the autologous HSCTs were performed in inpatient setting, the swift recovery after peripheral autologous HSCT and improvements in supportive care have enabled patients to receive autologous HSCT at outpatient [60, 118]. It has been reported that outpatient autologous HSCT is safe and feasible in patients with: lymphoma, tumors of the central nervous system, and breast cancer [60, 119, 120, 121].

Several studies have shown that; with daily outpatient clinical evaluation and intensive supportive care; outpatient autologous HSCT is safe, feasible, and cost-effective and it can lead to excellent short-term as well as long-term outcomes in carefully selected patients with MM and lymphoma [13, 56, 57, 59, 60, 61, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135]. However, a multidisciplinary approach with close follow-up is required to guarantee a successful outcome of the autologous outpatient HSCT program [59, 122, 123, 131, 136]. 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 short period of neutropenia [56, 135, 137].

The inclusion criteria of outpatient HSCT include: (1) availability of full-time caregiver; (2) residence within 20–30 minutes-drive from the hospital; (3) favorable performance status and comorbidity profile; (4) stable psychology and expected compliance; and (5) patient preference and signed written consent [60, 124, 125, 132, 134, 135]. On the other hand, the exclusion criteria of outpatient HSCT include: (1) age more than 65 years; (2) performance status >1; (3) severe comorbid medical conditions and severe impairment of organ functions; (4) severe recent or incompletely eradicated infection and colonization with multidrug-resistant micro-organisms; (5) lack of caregiver and living >1-hour drive distance from the hospital; and (6) advanced disease such as MM or lymphoma [60, 61, 63, 118].

Indications for admission in recipients of outpatient HSCT include: (1) febrile neutropenia, pneumonia, sepsis, or arrhythmia; (2) severe mucositis and poor oral intake; and (3) declining performance status of the patient and inability of family or caregiver to cope [57, 61, 118, 123, 129, 130, 138]. Between 8% and 84%. of recipients of outpatient autologous HSCT require hospitalization in the first 100 days post-HSCT and the duration of hospitalization ranges between 4 and 9 days [57, 59, 61, 122, 123, 127, 129, 130]. The median time to engraftment in patients with MM receiving autologous HSCT at outpatient is: 9–14 days for neutrophils and 12–19 days for platelets, while the reported transplant-related mortality is ≤1.1% [57, 59, 61, 123, 124, 126, 127, 129, 130, 132, 136]. Outpatient autologous HSCT has the following advantages: (1) significant reduction in costs and saving beds; (2) patient convenience and high patient satisfaction; (3) lower rate of infections; and (4) lower morbidity and TRM [56, 59, 61, 122, 134, 136, 139, 140, 141].

Advertisement

7. Allogeneic HSCT in MM

Despite the current advances in the treatment of MM including the introduction of several classes of novel agents, MM remains incurable and eventually most patients develop progressive disease [142, 143, 144, 145]. Currently, allogeneic HSCT represents the only potentially curative therapy for patients with MM [146, 147, 148, 149]. In MM patients, allogeneic HSCT exerts its therapeutic efficacy mainly through its graft versus myeloma (GVM) [143, 144, 146]. It is reasonable to consider allogeneic HSCT as the treatment strategy for younger patients with MM having HR disease as several studies have shown that allogeneic HSCT can potentially overcome the adverse prognosis of HR cytogenetics [143, 146, 147, 148]. In MM patients, the use of myeloablative conditioning (MAC) in allogeneic HSCT is associated with high treatment-related mortality (TRM) mainly due to the regimen-related toxicities and GVHD which are translated into considerable transplant-related morbidity and mortality while the use of reduced intensity conditioning (RIC) in allogeneic HSCT is associated with high relapse rates [144, 145, 147, 148, 149]. Nevertheless, allogeneic HSCT offers a potentially curative option in 10–20% of patients with RR MM [142]. A study performed at MD Andersen Cancer Centre that included 149 patients with MM subjected to allogeneic HSCT [38 MAC; and 110 RIC] showed that predictors of prolonged survival included: chemosensitive disease in the pre-transplant period in addition to the absence of HR cytogenetics [150].To minimize treatment-related toxicity while allowing the GVM effect, some clinical trials have used RIC-allogeneic HSCT as a tandem approach following autologous HSCT, that is, autologous-RIC allogeneic HSCT in patients with MM who are eligible for HSCT [144, 149]. In patients with RR MM, allogeneic HCT with an RIC regimen is associated with acceptable toxicity as well as durable remissions and long-term survival and the use of novel agents as maintenance therapy following RIC-allogeneic HSCT can reduce the rate of relapse and disease progression [142, 145, 149, 151]. Haploidentical HSCT with post-transplant cyclophosphamide is a feasible option in patients with HR-MM eligible for allogeneic HSCT but lacking HLA-identical donors [146]. The use of CD34-selected stem cells in allogeneic HSCT in patients with MM is safe and effective, although the outcome of CD34-selected HSCT is influenced by the following: age of the patient, extramedullary disease, and disease status prior to CD34-selected HSCT [152]. Whole-body imaging is an appropriate and highly recommended diagnostic approach for the detection of prognostically relevant lesions before and after allogeneic HSCT in patients with MM [153]. The utilization of minimal residual disease evaluation prior to allogeneic HSCT could allow the identification of subgroups of patients who are likely to benefit from allogeneic HSCT [154]. Finally, the role of allogeneic HSCT in patients should be complementary to other available therapeutic options such as: monoclonal antibodies, bispecific T-cell engagers (BiTe), and CAR T-cell therapy [149, 154].

Advertisement

8. Complications of autologous HSCT in patients with MM

8.1 Engraftment syndrome and autologous GVHD

During the neutrophilic recovery following HSCT, a constellation of clinical manifestations that include fever, erythematous skin rash, nausea, vomiting, diarrhea, and noncardiogenic pulmonary edema may occur [67, 155]. These clinical features are usually referred to as engraftment syndrome which may be a manifestation of graft versus host reaction. This syndrome reflects cellular and cytokine interactions and may be associated with significant transplant-related mortality and morbidity due to pulmonary leak syndrome and multiorgan failure [155, 156, 157, 158]. Early recognition of this syndrome is vital in order to administer appropriate GVHD therapy which includes HD corticosteroids, alemtuzumab, infliximab, daclizumab, and etanercept [67, 155, 156, 157, 158, 159].

GVHD is a common complication of allogeneic HSCT [160]. A similar autoimmune syndrome, termed auto-aggregation syndrome or autologous GVHD, has been reported in the setting of autologous HSCT [160, 161]. Autologous GVHD represents the extreme or severe form of engraftment syndrome [68]. The incidence of autologous GVHD is 5–20% [162]. The predisposing factors for autologous GVHD include: MM as the primary disease; second auto-HSCT; heavily pretreated patients; high CD34+ cells infused; HLA B55 expression; low percentages of CD3+ and CD8+ T-cells; and achievement of high levels of absolute lymphocyte counts after HSCT [68, 155, 156, 157, 158, 159, 160, 163].

In autologous GVHD, there is dysregulation of the immune responses due to: the primary disease such as MM, HD melphalan used the conditioning therapy before HSCT; and the use of immunomodulatory agents in the treatment of MM [163]. The clinical and histological manifestations of autologous GVHD are similar to those encountered in acute GVHD following allogeneic HSCT although the clinical features tend to be milder and self-limited in most cases [68, 160, 161, 164, 165, 166]. Autologous GVHD can involve the: skin, liver, and gastrointestinal tract [68, 160, 162, 164, 165, 166]. Treatment is usually symptomatic although immunosuppression with corticosteroids is usually needed in severe cases [68, 160, 164, 162, 165]. Death as a consequence of infectious complications has been reported in severe forms of autologous GVHD [165].

8.2 Other complications of autologous HSCT in MM patients

Autologous HSCT in patients with MM has several complications that can be classified as early or late complications. Early complications occur before day 100 post-HSCT, while late complications are usually encountered after day 100 post-transplantation. The early and late complications are shown in Table 3 [167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182] and Table 4 [183, 184, 185, 186, 187, 188], respectively. The predisposing factors for the complications of autologous HSCT in patients with MM include: (1) the disease itself; (2) presence of other comorbid medical conditions; (3) old age; (4) renal failure; and (5) drugs used in the treatment of patients with MM such as: corticosteroids, cyclophosphamide, HD melphalan, thalidomide, lenalidomide use before and after HSCT, as well as bortezomib use before and after autologous transplantation [167, 171, 172, 173, 174, 175, 176, 177, 179, 180, 181, 182, 183, 184, 185, 187].

1.Febrile neutropenia
2.Sepsis; bacteremia with multidrug-resistant organisms
3.Pneumonia with Streptococcus pneumoniae
4.Cellulitis
5.Neutropenic colitis
6.Infections with Candida species
7.Clostridium difficile infections
8.Oral mucositis
9.Electrolytic disturbances particularly hypokalemia and hypophosphatemia
10.Thromboses related to central venous catheters
11.Acute renal failure
12.Acute respiratory failure requiring endotracheal intubation and mechanical ventilation

Table 3.

Early complications of autologous hematopoietic stem cell transplantation in patients with multiple myeloma.

(1)Reactivation of cytomegalovirus and hepatitis-B infections
(2)Infection with herpes simplex and varicella-zoster viruses
(3)Pneumocystis jeroveci infections
(4)Infections with Aspergillus species
(5)Infection with multidrug-resistant organisms
(6)Therapy-related myelodysplasia and acute myeloid leukemia
(7)Second primary malignancies such as solid tumors and skin cancer
(8)Chronic pulmonary complications: lung dysfunction and pneumonitis
(9)Sexual dysfunction
(10)Hypothyroidism
(11)Cataract
(12)Osteopenia and osteoporosis
(13)Avascular necrosis of bone
(14)Hypertension
(15)Cardiomyopathy and congestive cardiac failure
(16)Post-traumatic stress disorders: anxiety; and depression

Table 4.

Late complications of autologous hematopoietic stem cell transplantation in patients with multiple myeloma.

Advertisement

9. Maintenance therapy after autologous HSCT in patients with MM

In patients with MM, autologous HSCT has been shown to improve OS and PFS but it is not curative [189]. The residual disease is almost always present after autologous HSCT and is responsible for relapse [190]. Maintenance therapy after autologous HSCT has been shown to deepen and prolong responses and increase OS and PFS [190, 191]. Thalidomide was the first immunomodulatory agent to be used in maintenance therapy after autologous HSCT in MM patients [192, 193]. The use of lenalidomide in the maintenance therapy following autologous HSCT in patients with newly diagnosed MM has been investigated in four phase III randomized control studies [193, 194]. These clinical trials and other studies have shown that lenalidomide maintenance therapy until disease progression prolongs OS, PFS, and EFS in patients with MM [189, 193, 195, 196, 197].

In patients with MM, bortezomib induction and maintenance therapy after autologous HSCT improves rates of CR and achieves superior OS and PFS [198]. Bortezomib alone or in combination with other drugs such as dexamethasone, thalidomide, and pomalidomide has been shown to be feasible, well tolerated, and beneficial in maintenance therapy following autologous HSCT in patients with: (1) HR MM such as patients with 17 p deletion; (2) renal insufficiency; (3) previous history of another cancer; and (4) inability to tolerate lenalidomide [16, 199, 200]. However, in patients with newly diagnosed MM lenalidomide maintenance therapy after HD melphalan and autologous HSCT has become the standard of care [190, 193, 196, 199, 201].

Advertisement

10. Continuous therapy after autologous HSCT in MM patients

In younger patients with MM, long-term maintenance therapy after autologous HSCT has been shown to significantly improve OS and PFS compared to observation [202]. Consolidation therapy with VRd regimen followed by lenalidomide maintenance improves PFS and the depth of response in patients with newly diagnosed MM compared to maintenance therapy alone [203]. Compared to the traditional fixed-duration therapy, maintenance therapy approaches in MM offer prolonged disease control and improved outcomes. In patients with newly diagnosed MM, multiple agents have been investigated as long-term options and these include: immunomodulatory agents such as thalidomide and lenalidomide; proteasome inhibitors such as bortezomib, carfizomib, and ixazomib; and monoclonal antibodies such as daratumumab, elotuzumab, and isatuximab [204].

Continuous therapy has become a key strategy in patients with MM as it has been shown to prolong the duration of remission and significantly improve OS and PFS [205, 206, 207]. Continuous therapy represents the standard approach for patients with MM both at diagnosis and at relapse as it provides better disease control [202]. However, risk-adapted therapy is recommended as patients having HR-MM may benefit from more intensive maintenance treatment than patients with SR-MM [205].

11. Conclusions and future directions

Autologous HSCT followed by maintenance therapy till relapse or disease progression remains the standard of care in patients with MM who are transplant eligible. Autologous HSCT can safely be performed with or without cryopreservation in inpatient or outpatient settings as well as in patients having renal failure. Allogeneic HSCT and tandem autologous HSCT are indicated in a highly selected group of patients with MM particularly younger patients with HR features such as adverse cytogenetics. The recent developments in the treatment of patients with MM include: induction therapy with four drugs; continuous therapy even in transplanted patients; and the use of CAR T-cell therapy, bispecific antibodies, and other novel agents in the treatment of patients having RR-MM. The timing of the incorporation of novel agents, stem cell therapies, and new immunotherapies will be determined by the results of the ongoing clinical trials.

References

  1. 1. Rajkumar SV. Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. American Journal of Hematology. 2020;95(5):548-567. DOI: 10.1002/ajh.25791
  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. 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
  4. 4. 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
  5. 5. 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 2016 Nov 10
  6. 6. 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 2019 Apr 23
  7. 7. Du J, Zhuang J. Major advances in the treatment of multiple myeloma in American Society of Hematology Annual Meeting 2020. Chronic Diseases and Translational Medicine. 2021;7(4):220-226. DOI: 10.1016/j.cdtm.2021.08.003
  8. 8. 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
  9. 9. 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
  10. 10. 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
  11. 11. 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
  12. 12. 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
  13. 13. Cavo M, Tacchetti P, Zamagni E. Front-line treatment of multiple myeloma. Hemasphere. 2019;3(Suppl):127-130. DOI: 10.1097/HS9. 0000000000000242
  14. 14. 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
  15. 15. Chakraborty R, Muchtar E, Kumar SK, Buadi FK, Dingli D, Dispenzieri A, et al. Outcomes of maintenance therapy with lenalidomide or bortezomib in multiple myeloma in the setting of early autologous stem cell transplantation. Leukemia. 2018;32(3):712-718. DOI: 10.1038/leu.2017.256 Epub 2017 Aug 14
  16. 16. 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
  17. 17. Korst CLBM, van de Donk NWCJ. Should all newly diagnosed MM patients receive CD38 antibody-based treatment? Hematology. 2020;2020(1):259-263. DOI: 10.1182/hematology. 2020000161
  18. 18. 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
  19. 19. 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
  20. 20. 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
  21. 21. Bazarbachi AH, Al Hamed R, Malard F, Bazarbachi A, Harousseau JL, Mohty M. Induction therapy prior to autologous stem cell transplantation (ASCT) in newly diagnosed multiple myeloma: An update. Blood Cancer Journal. 2022;12(3):47. DOI: 10.1038/s41408-022-00645-1
  22. 22. Rajkumar SV, Kumar S. Multiple myeloma current treatment algorithms. Blood Cancer Journal. 2020;10(9):94. DOI: 10.1038/s41408-020-00359-2
  23. 23. 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
  24. 24. Nishimura KK, Barlogie B, van Rhee F, Zangari M, Walker BA, Rosenthal A, et al. Long-term outcomes after autologous stem cell transplantation for multiple myeloma. Blood Advances. 2020;4(2):422-431. DOI: 10.1182/bloodadvances.2019000524
  25. 25. Rajkumar SV. In: Kyle RA, Connor RF, editors. Clinical Features, Laboratory Manifestations, and Diagnosis of Multiple Myeloma. Up to Date 2018
  26. 26. Belotti A, Ribolla R, Cancelli V, Crippa C, Bianchetti N, Ferrari S, et al. Transplant eligibility in elderly multiple myeloma patients: Prospective external validation of the international myeloma working group frailty score and comparison with clinical judgment and other comorbidity scores in unselected patients aged 65-75 years. American Journal of Hematology. 2020;95(7):759-765. DOI: 10.1002/ajh.25797 Epub 2020 Apr 23
  27. 27. Mina R, Lonial S. Is there still a role for stem cell transplantation in multiple myeloma? Cancer. 2019;125(15):2534-2543. DOI: 10.1002/cncr. 32060 Epub 2019 Apr 15
  28. 28. Ramzi M, Zakerinia M, Nourani H, Dehghani M, Vojdani R, Haghighinejad H. Noncryopreserved hematopoietic stem cell transplantation in multiple myeloma, a single center experience. Clinical Transplantation. 2012;26(1):117-122
  29. 29. Sivaraj D, Bacon W, Long GD, Rizzieri DA, Horwitz ME, Sullivan KM, et al. High-dose BCNU/Melphalan conditioning regimen before autologous stem cell transplantation in newly diagnosed multiple myeloma. Bone Marrow Transplantation. 2018;53(1):34-38. DOI: 10.1038/bmt.2017.208 Epub 2017 Oct 30
  30. 30. Sweiss K, Patel S, Culos K, Oh A, Rondelli D, Patel P. Melphalan 200 mg/m2 in patients with renal impairment is associated with increased short-term toxicity but improved response and longer treatment-free survival. Bone Marrow Transplantation. 2016;51(10):1337-1341. DOI: 10.1038/bmt.2016.136 Epub 2016 May 16
  31. 31. Srour SA, Milton DR, Bashir Q , Nieto Y, Saini N, Daher M, et al. Melphalan dose intensity for autologous stem cell transplantation in multiple myeloma. Haematologica. 2021;106(12):3211-3214. DOI: 10.3324/haematol.2021.279179
  32. 32. Mikhael JR, Dingli D, Roy V, Reeder CB, Buadi FK, Hayman SR, et al. Management of newly diagnosed symptomatic multiple myeloma: Updated mayo stratification of myeloma and risk-adapted therapy (mSMART) consensus guidelines 2013. Mayo Clinic Proceedings. 2013;88:360-376. DOI: 10.1016/j.mayocp. 2013.01.019
  33. 33. Tricot G, Alberts DS, Johnson C, Roe DJ, Dorr RT, Bracy D, et al. Safety of autotransplants with high-dose melphalan in renal failure: A pharmacokinetic and toxicity study. Clinical Cancer Research. 1996;2(6):947-952
  34. 34. Parikh GC, Amjad AI, Saliba RM, Kazmi SM, Khan ZU, Lahoti A, et al. Autologous hematopoietic stem cell transplantation may reverse renal failure in patients with multiple myeloma. Biology of Blood and Marrow Transplantation. 2009;15(7):812-816. DOI: 10.1016/j.bbmt.2009.03.021
  35. 35. El Fakih R, Fox P, Popat U, Nieto Y, Shah N, Parmar S, et al. Autologous hematopoietic stem cell transplantation in dialysis-dependent myeloma patients. Clinical Lymphoma, Myeloma & Leukemia. 2015;15(8):472-476. DOI: 10.1016/j.clml.2015.03.003 Epub 2015 Mar
  36. 36. Mahindra A, Hari P, Fraser R, Fei M, Huang J, Berdeja J, et al. Autologous hematopoietic cell transplantation for multiple myeloma patients with renal insufficiency: A center for international blood and marrow transplant research analysis. Bone Marrow Transplantation. 2017;52(12):1616-1622. DOI: 10.1038/bmt. 2017.198 Epub 2017 Sep 18
  37. 37. Al-Anazi KA, Mokhtar N, Kawari M, AlHashmi H, Abduljalil O, Alshaibani E, et al. A young patient with refractory multiple myeloma and dialysis-dependent renal failure has been cured by non-cryopreserved autologous stem cell transplantation followed by live-related kidney transplantation. Journal of Stem Cell Biology and Transplantation. 2017;1(2:13). DOI: 10.21767/2575-7725.100013
  38. 38. Katragadda L, McCullough LM, Dai Y, Hsu J, Byrne M, Hiemenz J, et al. Effect of melphalan 140 mg/m(2) vs 200 mg/m(2) on toxicities and outcomes in multiple myeloma patients undergoing single autologous stem cell transplantation-a single center experience. Clinical Transplantation. 2016;30(8):894-900. DOI: 10.1111/ctr.12762 Epub 2016 Jun 29
  39. 39. Wanchoo R, Abudayyeh A, Doshi M, Edeani A, Glezerman IG, Monga D, et al. Renal toxicities of novel agents used for treatment of multiple myeloma. Clinical Journal of the American Society of Nephrology. 2017;12(1):176-189. DOI: 10.2215/CJN. 06100616 Epub 2016 Sep 21
  40. 40. Knudsen LM, Nielsen B, Gimsing P, Geisler C. Autologous stem cell transplantation in multiple myeloma: Outcome in patients with renal failure. European Journal of Haematology. 2005;75(1):27-33. DOI: 10.1111/j.1600-0609.2005.00446.x
  41. 41. Antlanger M, Dust T, Reiter T, Böhm A, Lamm WW, Gornicec M, et al. Impact of renal impairment on outcomes after autologous stem cell transplantation in multiple myeloma: A multi-center, retrospective cohort study. BMC Cancer. 2018;18(1):1008. DOI: 10.1186/s12885-018-4926-0
  42. 42. Russell N, Douglas K, Ho AD, Mohty M, Carlson K, Ossenkoppele GJ, et al. Plerixafor and granulocyte colony-stimulating factor for first-line steady-state autologous peripheral blood stem cell mobilization in lymphoma and multiple myeloma: Results of the prospective PREDICT trial. Haematologica. 2013;98(2):172-178. DOI: 10.3324/haematol.2012.071456 Epub 2012 Sep 14
  43. 43. Lin TL, Wang PN, Kuo MC, Hung YH, Chang H, Tang TC. Cyclophosphamide plus granulocyte-colony stimulating factor for hematopoietic stem cell mobilization in patients with multiple myeloma. Journal of Clinical Apheresis. 2016;31(5):423-428. DOI: 10.1002/jca.21421 Epub 2015 Sep 5
  44. 44. Nademanee AP, DiPersio JF, Maziarz RT, Stadtmauer EA, Micallef IN, Stiff PJ, et al. Plerixafor plus granulocyte colony-stimulating factor versus placebo plus granulocyte colony-stimulating factor for mobilization of CD34(+) hematopoietic stem cells in patients with multiple myeloma and low peripheral blood CD34(+) cell count: Results of a subset analysis of a randomized trial. Biology of Blood and Marrow Transplantation. 2012;18(10):1564-1572. DOI: 10.1016/j.bbmt. 2012.05.017 Epub 2012 Jun 6
  45. 45. Li J, Hamilton E, Vaughn L, Graiser M, Renfroe H, Lechowicz MJ, et al. Effectiveness and cost analysis of “just-in-time” salvage plerixafor administration in autologous transplant patients with poor stem cell mobilization kinetics. Transfusion. 2011;51(10):2175-2182. DOI: 10.1111/j.1537-2995.2011.03136.x Epub 2011 Apr 14
  46. 46. Hamadani M, Kochuparambil ST, Osman S, Cumpston A, Leadmon S, Bunner P, et al. Intermediate-dose versus low-dose cyclophosphamide and granulocyte colony-stimulating factor for peripheral blood stem cell mobilization in patients with multiple myeloma treated with novel induction therapies. Biology of Blood and Marrow Transplantation. 2012;18(7):1128-1135. DOI: 10.1016/j.bbmt.2012.01.005 Epub 2012 Jan 14
  47. 47. Tuchman SA, Bacon WA, Huang LW, Long G, Rizzieri D, Horwitz M, et al. Cyclophosphamide-based hematopoietic stem cell mobilization before autologous stem cell transplantation in newly diagnosed multiple myeloma. Journal of Clinical Apheresis. 2015;30(3):176-182. DOI: 10.1002/jca.21360 Epub 2014 Oct 8
  48. 48. Bacon WA, Long GD, Rizzieri DA, Horwitz ME, Chute JP, Sullivan KM, et al. Impact of high dose cyclophosphamide on the outcome of autologous stem cell transplant in patients with newly diagnosed multiple myeloma. Blood. 2011;118(21):4127. DOI: 10.1182/blood.V118.21.4127.4127
  49. 49. Awan F, Kochuparambil ST, Falconer DE, Cumpston A, Leadmon S, Watkins K, et al. Comparable efficacy and lower cost of PBSC mobilization with intermediate-dose cyclophosphamide and G-CSF compared with plerixafor and G-CSF in patients with multiple myeloma treated with novel therapies. Bone Marrow Transplantation. 2013;48(10):1279-1284. DOI: 10.1038/bmt.2013.52 Epub 2013 Apr 15
  50. 50. 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
  51. 51. Hechler G, Weide R, Heymanns J, Köppler H, Havemann K. Storage of noncryopreserved periphered blood stem cells for transplantation. Annals of Hematology. 1996;72(5):303-306. DOI: 10.1007/s002770050176
  52. 52. Parkins MD, Bahlis N, Brown C, Savoie L, Chaudhry A, Russell JA, et al. Overnight storage of autologous stem cell apheresis products before cryopreservation does not adversely impact early or long-term engraftment following transplantation. Bone Marrow Transplantation. 2006;38(9):609-614. DOI: 10.1038/sj.bmt.1705501 Epub 2006 Sep 18
  53. 53. Donmez A, Cagirgan S, Saydam G, Tombuloglu M. Overnight refrigerator storage of autologous peripheral progenitor stem cells without cryopreservation. Transfusion and Apheresis Science. 2007;36(3):313-319. DOI: 10.1016/j.transci.2007.03.011 Epub 2007 Jun 13
  54. 54. Lazarus HM, Pecora AL, Shea TC, Koç ON, White JM, Gabriel DA, et al. CD34+ selection of hematopoietic blood cell collections and auto-transplantation in lymphoma: Overnight storage of cells at 4 degrees C does not affect outcome. Bone Marrow Transplantation. 2000;25(5):559-566. DOI: 10.1038/sj.bmt.1702175
  55. 55. Jasuja SK, Kukar(jasuja) N, Jain R, Bhateja A, Jasuja A, Jain R. A simplified method at lowest cost for autologous, non-cryopreserved, unmanipulated, peripheral hematopoietic stem cell transplant in multiple myeloma and non-Hodgkin’s lymphoma: Asian scenario. Journal of Clinical Oncology. 2010;28(15):ė18545
  56. 56. 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 Apr 4, 2016
  57. 57. Jagannath S, Vesole DH, Zhang M, Desikan KR, Copeland N, Jagannath M, et al. Feasibility and cost-effectiveness of outpatient autotransplants in multiple myeloma. Bone Marrow Transplantation. 1997;20(6):445-450. DOI: 10.1038/sj. bmt. 1700900
  58. 58. Ferrara F, Palmieri S, Viola A, Copia C, Schiavone EM, De Simone M, et al. Outpatient-based peripheral blood stem cell transplantation for patients with multiple myeloma. The Hematology Journal. 2004;5(3):222-226. DOI: 10.1038/sj.thj. 6200349
  59. 59. Holbro A, Ahmad I, Cohen S, Roy J, Lachance S, Chagnon M, et al. Safety and cost effectiveness of outpatient autologous stem cell transplantation in patients with multiple myeloma. Biology of Blood and Marrow Transplantation. 2013;19(4):547-551. DOI: 10.1016/j.bbmt.2012.12.006 Epub Dec 16, 2012
  60. 60. Graff TM, Singavi AK, Schmidt W, Eastwood D, Drobyski WR, Horowitz M, et al. Safety of outpatient autologous hematopoietic cell transplantation for multiple myeloma and lymphoma. Bone Marrow Transplantation. 2015;50(7):947-953. DOI: 10.1038/bmt. 2015.46 Epub Apr 13, 2015
  61. 61. Lisenko K, Sauer S, Bruckner T, Egerer G, Goldschmidt H, Hillengass J, et al. High dose chemotherapy and autologous stem cell transplantation of patients with multiple myeloma in an outpatient setting. BMC Cancer. 2017;17(1):151. DOI: 10.1186/s12885-017-3137-4
  62. 62. Kroll TM, Singavi A, Schmidt W, Eastwood D, Drobski W, Horowitz MM, et al. Safety of outpatient autologous hematopoietic cell transplantation (AuHCT) for multiple myeloma and lymphoma. Biology of Blood and Marrow Transplantation. 2014;20(Suppl. 2):S114. DOI: 10.1016/j.bbmt.2013.12.166
  63. 63. Frey P, Stinson T, Siston A, Knight SJ, Ferdman E, Traynor A, et al. Lack of care-givers limits use of outpatient hematopoietic stem cell transplant program. Bone Marrow Transplantation. 2002;30(11):741-748. DOI: 10.1038/sj. bmt.1703676
  64. 64. Lopez-Otero A, Ruiz-Delgado GJ, Ruiz-Arguelles GJ, A simplified method for stem cell autografting in multiple myeloma: A single institution experience. Bone Marrow Transplantation. 2009; 44(11): 715-719
  65. 65. Kayal S, Sharma A, Iqbal S, Tejomurtula T, Cyriac SL, Raina V. High-dose chemotherapy and autologous stem cell transplantation in multiple myeloma: A single institution experience at All India Institute of Medical Sciences, New Delhi, using noncryopreserved peripheral blood stem cells. Clinical Lymphoma, Myeloma & Leukemia. 2014;14(2):140-147. DOI: 10.1016/j.clml. 2013.09.001 Epub 2013 Sep 28
  66. 66. Bekadja MA, Brahimi M, Osmani S, Arabi A, Bouhass R, Yafour N, et al. A simplified method for autologous stem cell transplantation in multiple myeloma. Hematology/Oncology and Stem Cell Therapy. 2012;5(1):49-53. DOI: 10.5144/1658-3876.2012.49
  67. 67. Mutahar E, Al-Anazi KA. Engraftment syndrome: An updated review. Journal of Stem Cell Biology and Transplantation. 2017;1(3):16. DOI: 10.21767/2575-7725.100016
  68. 68. Kanfar S, Al-Anazi KA. Autologous graft versus host disease: An updated review. Annals of Stem Cells and Regenerative Medicine. 2018;1(1):1002
  69. 69. Ho PJ, Moore EM, McQuilten ZK, Wellard C, Bergin K, Augustson B, et al. Renal impairment at diagnosis in myeloma: Patient characteristics, treatment, and impact on outcomes. Results from the Australia and New Zealand Myeloma and Related Diseases Registry. Clinical Lymphoma, Myeloma & Leukemia. 2019;19(8):e415-e424. DOI: 10.1016/j.clml.2019.05.010 Epub 2019 May 16
  70. 70. Song J, Jiang F, Liu H, Ding K, Ren Y, Li L, et al. Effect factors related to a high probability of hemodialysis independence in newly diagnosed multiple myeloma patients requiring hemodialysis. Journal of Clinical Laboratory Analysis. 2020;34(2):e23057. DOI: 10.1002/jcla.23057 Epub 2019 Oct 30
  71. 71. Kundu S, Jha SB, Rivera AP, Flores Monar GV, Islam H, et al. Multiple myeloma and renal failure: Mechanisms, diagnosis, and management. Cureus. 2022;14(2):e22585. DOI: 10.7759/cureus.22585
  72. 72. Zhong H, Xie X, Xu G. Autologous stem cell transplantation in multiple myeloma with renal failure: Friend or foe? Stem Cells International. 2019;2019:9401717. DOI: 10.1155/2019/9401717
  73. 73. Firsova MV, Mendeleeva LP, Rekhtina IG, Solovev MV, Pokrovskaya OS, Gemdzhian E, et al. Autologous haematopoietic stem cell transplantation in patients with multiple myeloma complicated by dialysis-dependent renal failure. Blood. 2018;132(Suppl 1):5765. DOI: 10.1182/blood-2018-99-115880
  74. 74. Tosi P, Zamagni E, Ronconi S, Benni M, Motta MR, Rizzi S, et al. Safety of autologous hematopoietic stem cell transplantation in patients with multiple myeloma and chronic renal failure. Leukemia. 2000;14(7):1310-1313. DOI: 10.1038/sj.leu.2401819 PMID: 10914557
  75. 75. Wirk B. Renal failure in multiple myeloma: A medical emergency. Bone Marrow Transplantation. 2011;46(6):771-783. DOI: 10.1038/bmt.2011.8 Epub 2011 Feb 21
  76. 76. Ludwig H, Drach J, Graf H, Lang A, Meran JG. Reversal of acute renal failure by bortezomib-based chemotherapy in patients with multiple myeloma. Haematologica. 2007;92(10):1411-1414. DOI: 10.3324/haematol. 11463 PMID: 17768111
  77. 77. Hutchison CA, Bradwell AR, Cook M, Basnayake K, Basu S, Harding S, et al. Treatment of acute renal failure secondary to multiple myeloma with chemotherapy and extended high cut-off hemodialysis. Clinical Journal of the American Society of Nephrology. 2009;4(4):745-754. DOI: 10.2215/CJN.04590908 Epub 2009 Apr 1
  78. 78. Amit O, Ram R. Autologous hematopoietic cell transplantation for dialysis-dependent myeloma: More efficient, less toxic. Acta Haematologica. 2018;139(2):104-105. DOI: 10.1159/000486890 Epub 2018 Feb 7
  79. 79. San Miguel JF, Lahuerta JJ, García-Sanz R, Alegre A, Bladé J, Martinez R, et al. Are myeloma patients with renal failure candidates for autologous stem cell transplantation? The Hematology Journal. 2000;1(1):28-36. DOI: 10.1038/sj.thj. 6200003
  80. 80. Ballester OF, Tummala R, Janssen WE, Fields KK, Hiemenz JW, Goldstein SC, et al. High-dose chemotherapy and autologous peripheral blood stem cell transplantation in patients with multiple myeloma and renal insufficiency. Bone Marrow Transplantation. 1997 Oct;20(8):653-656. DOI: 10.1038/sj.bmt.1700950
  81. 81. Bird JM, Fuge R, Sirohi B, Apperley JF, Hunter A, Snowden J, et al. British society of blood and marrow transplantation. The clinical outcome and toxicity of high-dose chemotherapy and autologous stem cell transplantation in patients with myeloma or amyloid and severe renal impairment: A British Society of Blood and Marrow Transplantation study. British Journal of Haematology. 2006;134(4):385-390. DOI: 10.1111/j.1365-2141.2006.06191.x Epub 2006 Jul 5
  82. 82. Krejcí M, Doubek M, Adam Z, Hájek R, Vorlícek J, Mayer J. Autologous transplantation of peripheral hematopoietic cells in a patient with multiple myeloma and renal insufficiency. Vnitr̆ní Lékar̆ství. 1997;43(11):756-758
  83. 83. Kumar L, Chellapuram SK, Dev R, Varshneya A, Pawar S, Sharma A, et al. Induction therapy with novel agents and autologous stem cell transplant overcomes the adverse impact of renal impairment in multiple myeloma. Clinical Hematology International. 2019;1(4):205-219. DOI: 10.2991/chi.d.190805.003
  84. 84. Bachmann F, Schreder M, Engelhardt M, Langer C, Wolleschak D, Mügge LO, et al. Kinetics of renal function during induction in newly diagnosed multiple myeloma: Results of two prospective studies by the German Myeloma Study Group DSMM. Cancers (Basel). 2021;13(6):1322. DOI: 10.3390/cancers13061322 Epub 2021 Mar 16
  85. 85. Heher EC, Spitzer TR. Hematopoietic stem cell transplantation in patients with chronic kidney disease. Seminars in Nephrology. 2010;30(6):602-614. DOI: 10.1016/j.semnephrol. 2010.09.008
  86. 86. Le TX, Wolf JL, Peralta CA, Webber AB. Kidney transplantation for kidney failure due to multiple myeloma: Case reports. American Journal of Kidney Diseases. 2017;69(6):858-862. DOI: 10.1053/j.ajkd.2016.12.023 Epub 2017 Mar 18
  87. 87. Waszczuk-Gajda A, Vesole DH, Małyszko J, Jurczyszyn A, Wróbel T, Drozd-Sokołowska J, et al. Real-world prognostic factors in autotransplanted multiple myeloma patients with severe renal impairment: Study of the Polish Myeloma Study Group. Archives of Medical Science. 2020. DOI: 10.5114/aoms.2020.93442
  88. 88. Lazana I, Floro L, Christmas T, Shah S, Bramham K, Cuthill K, et al. Autologous stem cell transplantation for multiple myeloma patients with chronic kidney disease: A safe and effective option. Bone Marrow Transplantation. 2022;57(6):959-965. DOI: 10.1038/s41409-022-01657-y. Epub 2022 Apr 12
  89. 89. Abudayyeh A, Lin H, Mamlouk O, Abdelrahim M, Saliba R, Rondon G, et al. Impact of autologous stem cell transplantation on long term renal function and associated progression-free and overall survival in multiple myeloma. Leukemia & Lymphoma. 2020;61(13):3101-3111. DOI: 10.1080/10428194.2020.1797719 Epub 2020 Jul 29
  90. 90. Tyszkiewicz A, Benitoa MH, Rodriguezb GU, Búac BR, Gonzálezc MB, Sánchez-Jáuregui CM. Multiple myeloma with chronic kidney disease dependent on peritoneal dialysis and autologous stem cell transplant. Nephrologia. 2022;42(1):108-109. DOI: 10.1016/j.nefro.2020.07.014. Epub ahead of print
  91. 91. Scheid C, Sonneveld P, Schmidt-Wolf IG, van der Holt B, el Jarari L, Bertsch U, et al. Bortezomib before and after autologous stem cell transplantation overcomes the negative prognostic impact of renal impairment in newly diagnosed multiple myeloma: A subgroup analysis from the HOVON-65/GMMG-HD4 trial. Haematologica. 2014;99(1):148-154. DOI: 10.3324/haematol.2013.087585 Epub 2013 Aug 30
  92. 92. San Miguel JF. Bortezomib just for induction or also for maintenance in myeloma patients with renal impairment? Haematologica. 2014;99(1):5-6. DOI: 10.3324/haematol.2013.100982
  93. 93. Kumar S, Fu A, Niesvizky R, Jagannath S, Boccia R, Raje N. Renal response in real-world carfilzomib - vs bortezomib-treated patients with relapsed or refractory multiple myeloma. Blood Advances. 2021;5(2):367-376. DOI: 10.1182/bloodadvances.2019001059
  94. 94. Dimopoulos M, Siegel D, White DJ, Boccia R, Iskander KS, Yang Z, et al. Carfilzomib vs bortezomib in patients with multiple myeloma and renal failure: A subgroup analysis of ENDEAVOR. Blood. 2019;133(2):147-155. DOI: 10.1182/blood-2018-06-860015 Epub 2018 Nov 26
  95. 95. Bozic B, Rutner J, Zheng C, Ruckser R, Selimi F, Racz K, et al. Advances in the treatment of relapsed and refractory multiple myeloma in patients with renal insufficiency: Novel agents, immunotherapies and beyond. Cancers (Basel). 2021;13(20):5036. DOI: 10.3390/cancers13205036
  96. 96. Sweiss K, Calip GS, Johnson JJ, Rondelli D, Patel PR. Pretransplant hemoglobin and creatinine clearance correlate with treatment-free survival after autologous stem cell transplantation for multiple myeloma. Bone Marrow Transplantation. 2019;54(12):2081-2087. DOI: 10.1038/s41409-019-0628-8 Epub 2019 Aug 6
  97. 97. Lee CK, Zangari M, Barlogie B, Fassas A, van Rhee F, Thertulien R, et al. Dialysis-dependent renal failure in patients with myeloma can be reversed by high-dose myeloablative therapy and autotransplant. Bone Marrow Transplantation. 2004;33(8):823-828. DOI: 10.1038/sj.bmt.1704440
  98. 98. Tauro S, Clark FJ, Duncan N, Lipkin G, Richards N, Mahendra P. Recovery of renal function after autologous stem cell transplantation in myeloma patients with end-stage renal failure. Bone Marrow Transplantation. 2002;30(7):471-473. DOI: 10.1038/sj.bmt.1703713
  99. 99. Barlogie B, Jagannath S, Vesole DH, Naucke S, Cheson B, Mattox S, et al. Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma. Blood. 1997;89(3):789-793
  100. 100. Putkonen M, Rauhala A, Itälä M, Kauppila M, Pelliniemi TT, Remes K. Double versus single autotransplantation in multiple myeloma; a single center experience of 100 patients. Haematologica. 2005;90(4):562-563
  101. 101. Stadtmauer EA, Pasquini MC, Blackwell B, Hari P, Bashey A, Devine S, et al. Autologous transplantation, consolidation, and maintenance therapy in multiple myeloma: Results of the BMT CTN 0702 Trial. Journal of Clinical Oncology. 2019;37(7):589-597. DOI: 10.1200/JCO.18.00685 Epub 2019 Jan 17
  102. 102. Cavo M, Petrucci MT, Di Raimondo F, Zamagni E, Gamberi B, Crippa C, et al. Upfront single versus double autologous stem cell transplantation for newly diagnosed multiple myeloma: An intergroup, multicenter, phase III study of the European Myeloma Network (EMN02/HO95 MM Trial). Blood. 2016;128:991. DOI: 10.1182/blood.V128.22.991.991
  103. 103. Mai EK, Benner A, Bertsch U, Brossart P, Hänel A, Kunzmann V, et al. Single versus tandem high-dose melphalan followed by autologous blood stem cell transplantation in multiple myeloma: Long-term results from the phase III GMMG-HD2 trial. British Journal of Haematology. 2016;173(5):731-741. DOI: 10.1111/bjh. 13994 Epub 2016 Mar 17
  104. 104. Cavo M, Gay FM, Patriarca F, Zamagni E, Montefusco V, Dozza L, et al. Double autologous stem cell transplantation significantly prolongs progression-free survival and overall survival in comparison with single autotransplantation in newly diagnosed multiple myeloma: An analysis of phase 3 EMN02/H095 study. Blood. 2017;130(Suppl. 1):401. DOI: 10.1182/blood.V130. Suppl_1.401.401
  105. 105. Gagelmann N, Eikema DJ, Koster L, Caillot D, Pioltelli P, Lleonart JB, et al. Tandem autologous stem cell transplantation improves outcomes in newly diagnosed multiple myeloma with extramedullary disease and high-risk cytogenetics: A study from the Chronic Malignancies Working Party of the European Society for Blood and Marrow Transplantation. Biology of Blood and Marrow Transplantation. 2019;25(11):2134-2142. DOI: 10.1016/j.bbmt.2019.07.004 Epub 2019 Jul 6
  106. 106. Attal M, Harousseau JL, Facon T, Guilhot F, Doyen C, Fuzibet JG, et al. InterGroupe Francophone du Myélome. Single versus double autologous stem-cell transplantation for multiple myeloma. The New England Journal of Medicine. 2003;349(26):2495-2502. DOI: 10.1056/NEJMoa032290
  107. 107. Cavo M, Tosi P, Zamagni E, Cellini C, Tacchetti P, Patriarca F, et al. Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: Bologna 96 clinical study. Journal of Clinical Oncology. 2007;25(17):2434-2441. DOI: 10.1200/JCO. 2006.10.2509 Epub 2007 May 7
  108. 108. Larsen JT. Think twice: Doubling back to tandem autologous stem cell transplant in newly diagnosed multiple myeloma with extramedullary Disease. Biology of Blood and Marrow Transplantation. 2019;25(11):e317-e318. DOI: 10.1016/j.bbmt. 2019.09.020 Epub 2019 Sep 19
  109. 109. Martino M, Recchia AG, Fedele R, Neri S, Vincelli ID, Moscato T, et al. The role of tandem stem cell transplantation for multiple myeloma patients. Expert Opinion on Biological Therapy. 2016;16(4):515-534. DOI: 10.1517/14712598.2016.1136285 Epub 2016 Feb 6
  110. 110. Nair AP, Walker P, Kalff A, Bergin K, Hocking J, Avery S, et al. Adverse impact of high donor CD3+ cell dose on outcome following tandem auto-NMA allogeneic transplantation for high-risk myeloma. Bone Marrow Transplantation. 2017;52(6):839-845. DOI: 10.1038/bmt.2017.37 Epub 2017 Mar 20
  111. 111. Martino M, Tripepi G, Messina G, Vincelli ID, Console G, Recchia AG, et al. A phase II, single-arm, prospective study of bendamustine plus melphalan conditioning for second autologous stem cell transplantation in de novo multiple myeloma patients through a tandem transplant strategy. Bone Marrow Transplantation. 2016;51(9):1197-1203. DOI: 10.1038/bmt.2016.94 Epub 2016 Apr 18
  112. 112. Farag S, Jeker B, Bacher U, Mansouri Taleghani B, Mueller BU, et al. Dose-intensified bendamustine and melphalan (BenMel) conditioning before second autologous transplantation in myeloma patients. Hematological Oncology. 2018;36(4):671-678. DOI: 10.1002/hon.2546 Epub 2018 Sep 11
  113. 113. Musso M, Messina G, Marcacci G, Crescimanno A, Console G, Donnarumma D, et al. High-dose melphalan plus thiotepa as conditioning regimen before second autologous stem cell transplantation for “de novo” multiple myeloma patients: A phase II study. Biology of Blood and Marrow Transplantation. 2015;21(11):1932-1938. DOI: 10.1016/j. bbmt.2015.06.011 Epub 2015 Jun 19
  114. 114. Dhakal B, Szabo A, Chhabra S, Hamadani M, D’Souza A, Usmani SZ, et al. Autologous transplantation for newly diagnosed multiple myeloma in the era of novel agent induction: A systematic review and meta-analysis. JAMA Oncology. 2018;4(3):343-350. DOI: 10.1001/jamaoncol.2017.4600
  115. 115. Sing K, Sabo R, O'Bryan J, Risendal M, Roberts CH, Toor AA. Risk stratified tandem vs single autologous stem cell transplantation for multiple myeloma yields equivalent survival. Blood. 2020;136(1):23-24. DOI: 10.1182/blood-2020-136800
  116. 116. Malkan UY, Demiroglu H, Buyukasik Y, Karatas A, Aladag E, Goker H. Comparison of single and double autologous stem cell transplantation in multiple myeloma patients. Open Medicine. 2021;16(1):192-197. DOI: 10.1515/med-2021-0216
  117. 117. Barlogie B, Anaissie E, van Rhee F, Haessler J, Hollmig K, Pineda-Roman M, et al. Incorporating bortezomib into upfront treatment for multiple myeloma: Early results of total therapy 3. British Journal of Haematology. 2007;138(2):176-185. DOI: 10.1111/j.1365-2141.2007.06639.x
  118. 118. Larsen K, Spencer H, Mohan M, Bailey C, Hill K, Kottarathara M, et al. Feasibility of outpatient stem cell transplantation in multiple myeloma and risk factors predictive of hospital admission. Journal of Clinical Medicine. 2022;11(6):1640. DOI: 10.3390/jcm11061640
  119. 119. Leger C, Sabloff M, McDiarmid S, Bence-Bruckler I, Atkins H, Bredeson C, et al. Outpatient autologous hematopoietic stem cell transplantation for patients with relapsed follicular lymphoma. Annals of Hematology. 2006;85(10):723-729. DOI: 10.1007/s00277-006-0149-6 Epub 2006 Jul 11
  120. 120. Peters WP, Ross M, Vredenburgh JJ, Hussein A, Rubin P, Dukelow K, et al. The use of intensive clinic support to permit outpatient autologous bone marrow transplantation for breast cancer. Seminars in Oncology. 1994;21(4 Suppl. 7):25-31
  121. 121. Koo J, Silverman S, Nuechterlein B, Keating AK, Verneris MR, Foreman NK, et al. Safety and feasibility of outpatient autologous stem cell transplantation in pediatric patients with primary central nervous system tumors. Bone Marrow Transplantation. 2019;54(10):1605-1613. DOI: 10.1038/s41409-019-0479-3 Epub 2019 Feb 19
  122. 122. 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
  123. 123. Gertz MA, Ansell SM, Dingli D, Dispenzieri A, Buadi FK, Elliott MA, et al. Autologous stem cell transplant in 716 patients with multiple myeloma: Low treatment-related mortality, feasibility of outpatient transplant, and effect of a multidisciplinary quality initiative. Mayo Clinic Proceedings. 2008;83(10):1131-1138. DOI: 10.4065/83.10.1131
  124. 124. Martino M, Paviglianiti A, Memoli M, Martinelli G, Cerchione C. Multiple myeloma outpatient transplant program in the era of novel agents: State-of-the-art. Frontiers in Oncology. 2020;10:592487. DOI: 10.3389/fonc.2020.592487
  125. 125. Khouri J, Majhail NS. Advances in delivery of ambulatory autologous stem cell transplantation for multiple myeloma. Current Opinion in Supportive and Palliative Care. 2017;11(4):361-365. DOI: 10.1097/SPC.0000000000000305
  126. 126. Marini J, Maldonado A, Weeda ER, Hashmi H, Neppalli AK, Edwards K. Efficacy, safety and cost implications of outpatient autologous hematopoietic stem cell transplant for multiple myeloma: A single center experience. Blood. 2020;136(Suppl. 1):31. DOI: 10.1182/blood-2020-143137
  127. 127. Obiozor C, Subramaniam DP, Divine C, Shune L, Singh AK, Lin TL, et al. Evaluation of performance status and hematopoietic cell transplantation specific comorbidity index on unplanned admission rates in patients with multiple myeloma undergoing outpatient autologous stem cell transplantation. Biology of Blood and Marrow Transplantation. 2017;23(10):1641-1645. DOI: 10.1016/j.bbmt.2017.06.001 Epub 2017 Jun 8
  128. 128. 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
  129. 129. Martino M, Russo L, Martinello T, Gallo GA, Fedele R, Moscato T, et al. A home-care, early discharge model after autografting in multiple myeloma: Results of a three-arm prospective, non-randomized study. Leukemia & Lymphoma. 2015;56(3):801-804. DOI: 10.3109/10428194.2014.931952 Epub 2014 Jul 17
  130. 130. Ferrara F, Izzo T, Criscuolo C, Riccardi C, Viola A, Delia R, et al. Comparison of fixed dose pegfilgrastim and daily filgrastim after autologous stem cell transplantation in patients with multiple myeloma autografted on a outpatient basis. Hematological Oncology. 2011;29(3):139-143. DOI: 10.1002/hon.978 Epub 2010 Nov 30
  131. 131. Shah N, Cornelison AM, Saliba R, Ahmed S, Nieto YL, Bashir Q , et al. Inpatient vs outpatient autologous hematopoietic stem cell transplantation for multiple myeloma. European Journal of Haematology. 2017;99(6):532-535. DOI: 10.1111/ejh. 12970 Epub 2017 Oct 8
  132. 132. Paul TM, Liu SV, Chong EA, Luger SM, Porter DL, Schuster SJ, et al. Outpatient autologous stem cell transplantation for patients with myeloma. Clinical Lymphoma, Myeloma & Leukemia. 2015;15(9):536-540. DOI: 10.1016/j.clml.2015.05.006 Epub 2015 Jun 6
  133. 133. Martino M, Pitino A, Tripepi G, Paviglianiti A, Russo L, Cusumano G, et al. The burden in caregivers of multiple myeloma patients undergoing outpatient autologous stem-cell transplantation compared to inpatient transplantation. Clinical Lymphoma, Myeloma & Leukemia. 2021;21(4):e402-e409. DOI: 10.1016/j.clml.2020.11.011 Epub 2020 Nov 20
  134. 134. 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
  135. 135. 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
  136. 136. 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. Biology of Blood and Marrow Transplantation. 2018;24:S25-S118
  137. 137. Dytfeld D, Łojko-Dankowska A, Nowicki A, Matuszak M, Wache A, Gil L. Safety and cost effectiveness of outpatient autologous hematopoietic stem cell transplantation for multiple myeloma - single-center experience of a pilot Early Discharge Program. Acta Haematologica Polonica. 2021;52(3):178-181. DOI: 10.5603/AHP.a2021.0029·
  138. 138. Faucher C, Le Corroller Soriano AG, Esterni B, Vey N, Stoppa AM, Chabannon C, et al. Randomized study of early hospital discharge following autologous blood SCT: Medical outcomes and hospital costs. Bone Marrow Transplantation. 2012;47(4):549-555. DOI: 10.1038/bmt.2011.126 Epub 2011 Jul 4
  139. 139. Owattanapanich W, Suphadirekkul K, Kunacheewa C, Ungprasert P, Prayongratana K. Risk of febrile neutropenia among patients with multiple myeloma or lymphoma who undergo inpatient versus outpatient autologous stem cell transplantation: A systematic review and meta-analysis. BMC Cancer. 2018;18(1):1126. DOI: 10.1186/s12885-018-5054-6
  140. 140. 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
  141. 141. Fernández-Avilés F, Carreras E, Urbano-Ispizua A, Rovira M, Martínez C, Gaya A, et al. Case-control comparison of at-home to total hospital care for autologous stem-cell transplantation for hematologic malignancies. Journal of Clinical Oncology. 2006;24(30):4855-4861. DOI: 10.1200/JCO. 2006.06.4238 Epub 2006 Sep 25
  142. 142. Efebera YA, Qureshi SR, Cole SM, Saliba R, Pelosini M, Patel RM, et al. Reduced-intensity allogeneic hematopoietic stem cell transplantation for relapsed multiple myeloma. Biology of Blood and Marrow Transplantation. 2010;16(8):1122-1129. DOI: 10.1016/j.bbmt. 2010.02.015 Epub 2010 Feb 21
  143. 143. Yin X, Tang L, Fan F, Jiang Q , Sun C, Hu Y. Allogeneic stem-cell transplantation for multiple myeloma: A systematic review and meta-analysis from 2007 to 2017. Cancer Cell International. 2018;18:62. DOI: 10.1186/s12935-018-0553-8
  144. 144. Greil C, Engelhardt M, Finke J, Wäsch R. Allogeneic stem cell transplantation in multiple myeloma. Cancers (Basel). 2021;14(1):55. DOI: 10.3390/cancers14010055
  145. 145. Pawarode A, Mineishi S, Reddy P, Braun TM, Khaled YA, Choi SW, et al. Reducing treatment-related mortality did not improve outcomes of allogeneic myeloablative hematopoietic cell transplantation for high-risk multiple myeloma: A university of Michigan prospective series. Biology of Blood and Marrow Transplantation. 2016;22(1):54-60. DOI: 10.1016/j.bbmt.2015.07.021 Epub 2015 Jul 26
  146. 146. Castagna L, Mussetti A, Devillier R, Dominietto A, Marcatti M, Milone G, et al. Haploidentical allogeneic hematopoietic cell transplantation for multiple myeloma using post-transplantation cyclophosphamide graft-versus-host disease prophylaxis. Biology of Blood and Marrow Transplantation. 2017;23(9):1549-1554. DOI: 10.1016/j.bbmt. 2017.05.006 Epub 2017 May 10
  147. 147. Kaloyannidis P, Apostolidis J. Allogeneic stem cell transplantation in patients with high-risk multiple myeloma: Utopia or continuous challenge in aiming for cure? Current Treatment Options in Oncology. 2021;22(8):65. DOI: 10.1007/s11864-021-00864-x
  148. 148. Greil C, Engelhardt M, Ihorst G, Schoeller K, Bertz H, Marks R, et al. Allogeneic transplantation of multiple myeloma patients may allow long-term survival in carefully selected patients with acceptable toxicity and preserved quality of life. Haematologica. 2019;104(2):370-379. DOI: 10.3324/haematol. 2018.200881 Epub 2018 Sep 20
  149. 149. Gahrton G, Iacobelli S, Garderet L, Yakoub-Agha I, Schönland S. Allogeneic transplantation in multiple myeloma-does it still have a place? Journal of Clinical Medicine. 2020;9(7):2180. DOI: 10.3390/jcm9072180
  150. 150. Bashir Q , Khan H, Orlowski RZ, Amjad AI, Shah N, Parmar S, et al. Predictors of prolonged survival after allogeneic hematopoietic stem cell transplantation for multiple myeloma. American Journal of Hematology. 2012;87(3):272-276. DOI: 10.1002/ajh.22273 Epub 2012 Jan 9
  151. 151. Alsina M, Becker PS, Zhong X, Adams A, Hari P, Rowley S, et al. Resource for clinical investigation in blood and marrow transplantation. Lenalidomide maintenance for high-risk multiple myeloma after allogeneic hematopoietic cell transplantation. Biology of Blood and Marrow Transplantation. 2014;20(8):1183-1189. DOI: 10.1016/j.bbmt. 2014.04.014 Epub 2014 Apr 21
  152. 152. Smith E, Devlin SM, Kosuri S, Orlando E, Landau H, Lesokhin AM, et al. CD34-selected allogeneic hematopoietic stem cell transplantation for patients with relapsed, high-risk multiple myeloma. Biology of Blood and Marrow Transplantation. 2016;22(2):258-267. DOI: 10.1016/j.bbmt.2015.08.025 Epub 2015 Aug 30
  153. 153. Mosebach J, Shah S, Delorme S, Hielscher T, Goldschmidt H, Schlemmer HP, et al. Prognostic significance of tumor burden assessed by whole-body magnetic resonance imaging in multiple myeloma patients treated with allogeneic stem cell transplantation. Haematologica. 2018;103(2):336-343. DOI: 10.3324/haematol.2017.176073 Epub 2017 Dec 7
  154. 154. 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
  155. 155. Spitzer TR. Engraftment syndrome following hematopoietic stem cell transplantation. Bone Marrow Transplantation. 2001;27(9):893-938. DOI: 10.1038/sj.bmt.1703015
  156. 156. Cogbill CH, Drobyski WR, Komorowski RA. Gastrointestinal pathology of autologous graft-versus-host disease following hematopoietic stem cell transplantation: A clinicopathological study of 17 cases. Modern Pathology. 2011;24(1):117-125. DOI: 10.1038/modpathol.2010.163 Epub 2010 Oct 15
  157. 157. Nellen RG, van Marion AM, Frank J, Poblete-Gutiérrez P, Steijlen PM. Eruption of lymphocyte recovery or autologous graft-versus-host disease? International Journal of Dermatology. 2008;47(Suppl. 1):32-34. DOI: 10.1111/j.1365-4632.2008.03956.x
  158. 158. Drobyski WR, Hari P, Keever-Taylor C, Komorowski R, Grossman W. Severe autologous GVHD after hematopoietic progenitor cell transplantation for multiple myeloma. Bone Marrow Transplantation. 2009;43(2):169-177. DOI: 10.1038/bmt.2008.295 Epub 2008 Sep 1
  159. 159. Porrata LF. Clinical evidence of autologous graft versus tumor effect. American Journal of Immunology. 2009;5(1):1-7. DOI: 10.3844/ajisp.2009.1.7
  160. 160. Lee SE, Yoon JH, Shin SH, Park G, Min CK. Skin graft-versus-host disease following autologous stem cell transplantation for multiple myeloma. Immune Network. 2013;13(3):107-110. DOI: 10.4110/in.2013.13.3.107 Epub 2013 Jun 30
  161. 161. Kline J, Subbiah S, Lazarus HM, van Besien K. Autologous graft-versus-host disease: Harnessing anti-tumor immunity through impaired self-tolerance. Bone Marrow Transplantation. 2008;41(6):505-513. DOI: 10.1038/sj.bmt.1705931 Epub 2007 Nov 19
  162. 162. Hammami MB, Talkin R, Al-Taee AM, Schoen MW, Goyal SD, Lai JP. Autologous graft-versus-host disease of the gastrointestinal tract in patients with multiple myeloma and hematopoietic stem cell transplantation. Gastroenterology Research. 2018;11(1):52-57. DOI: 10.14740/gr925w Epub 2018 Feb 23
  163. 163. Batra A, Cottler-Fox M, Harville T, Rhodes-Clark BS, Makhoul I, Nakagawa M. Autologous graft versus host disease: An emerging complication in patients with multiple myeloma. Bone Marrow Research. 2014;2014:891427. DOI: 10.1155/2014/891427 Epub 2014 May 4
  164. 164. Fidler C, Klumpp T, Mangan K, Martin M, Sharma M, Emmons R, et al. Spontaneous graft versus host disease occurring in a patient with multiple myeloma after autologous stem cell transplant. American Journal of Hematology. 2012;87(2):219-221. DOI: 10.1002/ajh.22227 Epub 2011 Dec 21
  165. 165. Włodarczyk M, Wachowiak A, Wieczorek K, Toborek M, Wieczorkiewicz-Kabut A, Kata D, et al. Graft-versus-host disease as an unusual complication following autologous stem cell transplantation. Acta Haematologica Polonica. 2020;51(1):47-50. DOI: 10.2478/ahp-2020-0010
  166. 166. Krishna SG, Barlogie B, Lamps LW, Krishna K, Aduli F, Anaissie E. Recurrent spontaneous gastrointestinal graft-versus-host disease in autologous hematopoietic stem cell transplantation. Clinical Lymphoma, Myeloma & Leukemia. 2010;10(1):E17-E21. DOI: 10.3816/CLML.2010.n.012
  167. 167. Satlin MJ, Vardhana S, Soave R, Shore TB, Mark TM, Jacobs SE, et al. Impact of prophylactic levofloxacin on rates of bloodstream infection and fever in neutropenic patients with multiple myeloma undergoing autologous hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2015;21(10):1808-1814. DOI: 10.1016/j.bbmt. 2015.06.017 Epub 2015 Jul 3
  168. 168. Rahman S, Rybicki L, Ky Hamilton B, Pohlman B, Jagadeesh D, Cober E, et al. Early infectious complications after autologous hematopoietic cell transplantation for multiple myeloma. Transplant Infectious Disease. 2019;21(4):e13114. DOI: 10.1111/tid.13114 Epub 2019 Jun 1
  169. 169. Waszczuk-Gajda A, Drozd-Sokołowska J, Basak GW, Piekarska A, Mensah-Glanowska P, Sadowska-Klasa A, et al. Infectious complications in patients with multiple myeloma after high-dose chemotherapy followed by autologous stem cell transplant: Nationwide study of the infectious complications study group of the Polish Adult Leukemia Group. Transplantation Proceedings. 2020;52(7):2178-2185. DOI: 10.1016/j.transproceed.2020.02.068 Epub 2020 Mar 23
  170. 170. Anandan A, Kolk M, Ferrari N, Copley M, Driscoll J, Caimi P, et al. Serum electrolyte dynamics in multiple myeloma patients undergoing autologous haematopoietic stem cell transplantation. Nephrology. 2020;25(6):450-456. DOI: 10.1111/nep.13712 Epub 2020 Mar 23
  171. 171. Batlle M, Morgades M, Vives S, Ferrà C, Oriol A, Sancho JM, et al. Usefulness and safety of oral cryotherapy in the prevention of oral mucositis after conditioning regimens with high-dose melphalan for autologous stem cell transplantation for lymphoma and myeloma. European Journal of Haematology. 2014;93(6):487-491. DOI: 10.1111/ejh.12386 Epub 2014 Jun 21
  172. 172. Chen J, Seabrook J, Fulford A, Rajakumar I. Icing oral mucositis: Oral cryotherapy in multiple myeloma patients undergoing autologous hematopoietic stem cell transplant. Journal of Oncology Pharmacy Practice. 2017;23(2):116-120. DOI: 10.1177/1078155215620920 Epub 2016 Jul 9
  173. 173. Vera-Llonch M, Oster G, Ford CM, Lu J, Sonis S. Oral mucositis and outcomes of autologous hematopoietic stem-cell transplantation following high-dose melphalan conditioning for multiple myeloma. The Journal of Supportive Oncology. 2007;5(5):231-235
  174. 174. Grazziutti ML, Dong L, Miceli MH, Krishna SG, Kiwan E, Syed N, et al. Oral mucositis in myeloma patients undergoing melphalan-based autologous stem cell transplantation: Incidence, risk factors and a severity predictive model. Bone Marrow Transplantation. 2006;38(7):501-506. DOI: 10.1038/sj.bmt.1705471
  175. 175. Al-Jasser A, Al-Anazi K. Infections in patients with multiple myeloma in the era of novel agents and stem cell therapies. In: Al-Anazi K, editor. Update on Multiple Myeloma. London: Intech Open; 2018. DOI: 10.5772/intechopen.81683
  176. 176. Sanchez L, Sylvester M, Parrondo R, Mariotti V, Eloy JA, Chang VT. In-hospital mortality and post-transplantation complications in elderly multiple myeloma patients undergoing autologous hematopoietic stem cell transplantation: A population-based study. Biology of Blood and Marrow Transplantation. 2017;23(7):1203-1207. DOI: 10.1016/j.bbmt.2017.03.012 Epub 2017 Mar 9
  177. 177. Park H, Youk J, Kim HR, Koh Y, Kwon JH, Yoon SS, et al. Infectious complications in multiple myeloma receiving autologous stem cell transplantation in the past 10 years. International Journal of Hematology. 2017;106(6):801-810. DOI: 10.1007/s12185-017-2313-2 Epub 2017 Aug 20
  178. 178. Marchesi F, Mengarelli A, Giannotti F, Tendas A, Anaclerico B, Porrini R, et al. Rome Transplant Network. High incidence of post-transplant cytomegalovirus reactivations in myeloma patients undergoing autologous stem cell transplantation after treatment with bortezomib-based regimens: A survey from the Rome transplant network. Transplant Infectious Disease. 2014;16(1):158-164. DOI: 10.1111/tid.12162 Epub 2013 Nov 12
  179. 179. Marchesi F, Pimpinelli F, Dessanti ML, Gumenyuk S, Palombi F, Pisani F, et al. Evaluation of risk of symptomatic cytomegalovirus reactivation in myeloma patients treated with tandem autologous stem cell transplantation and novel agents: A single-institution study. Transplant Infectious Disease. 2014;16(6):1032-1038. DOI: 10.1111/tid.12309 Epub 2014 Nov 5
  180. 180. Kim JH, Goulston C, Sanders S, Lampas M, Zangari M, Tricot G, et al. Cytomegalovirus reactivation following autologous peripheral blood stem cell transplantation for multiple myeloma in the era of novel chemotherapeutics and tandem transplantation. Biology of Blood and Marrow Transplantation. 2012;18(11):1753-1758. DOI: 10.1016/j.bbmt.2012.06.008 Epub 2012 Jun 19
  181. 181. Belmoufid N, Daghri S, Driouich S, Nadi A, Bouanani N. Neutropenic enterocolitis as a complication of autologous stem cell transplant in patients with multiple myeloma: A case series. Cureus. 2022;14(4):e24475. DOI: 10.7759/cureus.24475
  182. 182. Hoppe A, Rupa-Matysek J, Małecki B, Dytfeld D, Hoppe K, Gil L. Risk factors for catheter-related thrombosis in multiple myeloma patients undergoing autologous stem cell transplantation. Medicina (Kaunas, Lithuania). 2021;57(10):1020. DOI: 10.3390/medicina57101020
  183. 183. Steingrimsdottir H, Gruber A, Kalin M, Björkholm M. Late infections after blood progenitor cell transplantation in patients with multiple myeloma. The American Journal of Medicine. 2001;110(4):329-330. DOI: 10.1016/s0002-9343(00)00725-7
  184. 184. Burns LJ. Late effects after autologous hematopoietic cell transplantation. Biology of Blood and Marrow Transplantation. 2009;15(1 Suppl):21-24. DOI: 10.1016/j.bbmt. 2008.10.009
  185. 185. Scarlata S, Annibali O, Santangelo S, Tomarchio V, Ferraro S, Armiento D, et al. Pulmonary complications and survival after autologous stem cell transplantation: Predictive role of pulmonary function and pneumotoxic medications. The European Respiratory Journal. 2017;49(3):1601902. DOI: 10.1183/13993003.01902-2016
  186. 186. Yamasaki S, Yoshimoto G, Kohno K, Henzan H, Aoki T, Tanimoto K, et al. Fukuoka blood and marrow transplantation group. Risk of secondary primary malignancies in multiple myeloma patients with or without autologous stem cell transplantation. International Journal of Hematology. 2019;109(1):98-106. DOI: 10.1007/s12185-018-2538-8 Epub 2018 Sep 24
  187. 187. Musto P, Anderson KC, Attal M, Richardson PG, Badros A, Hou J, et al. International Myeloma Working Group. Second primary malignancies in multiple myeloma: An overview and IMWG consensus. Annals of Oncology. 2017;28(2):228-245. DOI: 10.1093/annonc/mdw606
  188. 188. Georges GE, Bar M, Onstad L, Yi JC, Shadman M, Flowers ME, et al. Survivorship after autologous hematopoietic cell transplantation for lymphoma and multiple myeloma: Late effects and quality of life. Biology of Blood and Marrow Transplantation. 2020;26(2):407-412. DOI: 10.1016/j.bbmt.2019.10.002 Epub 2019 Oct 9
  189. 189. Uyl-de Groot CA, Ramsden R, Lee D, Boersma J, Zweegman S, Dhanasiri S. Lenalidomide as maintenance treatment for patients with multiple myeloma after autologous stem cell transplantation: A pharmaco-economic assessment. European Journal of Haematology. 2020;105(5):635-645. DOI: 10.1111/ejh.13497 Epub 2020 Sep 12
  190. 190. 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
  191. 191. 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
  192. 192. Manasanch EE. Recommend maintenance therapy with lenalidomide in multiple myeloma. Seminars in Oncology. 2016;43(6):712-713. DOI: 10.1053/j.seminoncol.2016.11.002 Epub 2016 Nov 5
  193. 193. Goldschmidt H, Mai EK, Dürig J, Scheid C, Weisel KC, Kunz C, et al. German-speaking Myeloma Multicenter Group (GMMG). Response-adapted lenalidomide maintenance in newly diagnosed myeloma: Results from the phase III GMMG-MM5 trial. Leukemia. 2020;34(7):1853-1865. DOI: 10.1038/s41375-020-0724-1 Epub 2020 Feb 7
  194. 194. Holstein SA, Suman VJ, Hillengass J, McCarthy PL. Future directions in maintenance therapy in multiple myeloma. Journal of Clinical Medicine. 2021;10(11):2261. DOI: 10.3390/jcm10112261
  195. 195. Syed YY. Lenalidomide: A review in newly diagnosed multiple myeloma as maintenance therapy after ASCT. Drugs. 2017;77:1473-1480. DOI: 10.1007/s40265-017-0795-0
  196. 196. Baertsch MA, Mai EK, Hielscher T, Bertsch U, Salwender HJ, Munder M, et al. German-Speaking Myeloma Multicenter Group (GMMG). Lenalidomide versus bortezomib maintenance after frontline autologous stem cell transplantation for multiple myeloma. Blood. Cancer Journal. 2021;11(1):1. DOI: 10.1038/s41408-020-00390-3
  197. 197. 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
  198. 198. Sonneveld P, Schmidt-Wolf IG, van der Holt B, El Jarari L, Bertsch U, Salwender H, et al. Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: Results of the randomized phase III HOVON-65/GMMG-HD4 trial. Journal of Clinical Oncology. 2012;30(24):2946-2955. DOI: 10.1200/JCO.2011.39.6820 Epub 2012 Jul 16
  199. 199. Bird SA, Jackson GH, Pawlyn C. Maintenance strategies post-autologous stem cell transplantation for newly diagnosed multiple myeloma. Clinical Hematology International. 2020;2(2):59-68. DOI: 10.2991/chi.d.200502.001
  200. 200. Sahebi F, Frankel PH, Farol L, Krishnan AY, Cai JL, Somlo G, et al. Sequential bortezomib, dexamethasone, and thalidomide maintenance therapy after single autologous peripheral stem cell transplantation in patients with multiple myeloma. Biology of Blood and Marrow Transplantation. 2012;18(3):486-492. DOI: 10.1016/j.bbmt.2011.12.580 Epub 2011 Dec 22
  201. 201. Richardson PG, Holstein SA, Schlossman RL, Anderson KC, Attal M, McCarthy PL. Lenalidomide in combination or alone as maintenance therapy following autologous stem cell transplant in patients with multiple myeloma: A review of options for and against. Expert Opinion on Pharmacotherapy. 2017;18(18):1975-1985. DOI: 10.1080/14656566.2017.1409207 Epub 2017 Dec 1
  202. 202. 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
  203. 203. Sonneveld P, Dimopoulos MA, Beksac M, van der Holt B, Aquino S, Ludwig H, et al. Consolidation and maintenance in newly diagnosed multiple myeloma. Journal of Clinical Oncology. 2021;39(32):3613-3622. DOI: 10.1200/JCO.21.01045 Epub 2021 Sep 14
  204. 204. 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
  205. 205. 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
  206. 206. 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
  207. 207. Ozaki S, Handa H, Koiso H, Saitoh T, Sunami K, Ishida T, et al. Propensity-score matched analysis of the efficacy of maintenance/continuous therapy in newly diagnosed patients with multiple myeloma: A multicenter retrospective collaborative study of the Japanese Society of Myeloma. Journal of Cancer Research and Clinical Oncology. 2022;148(1):191-203. DOI: 10.1007/s00432-021-03668-6 Epub 2021 Jun 2

Written By

Khalid Ahmed Al-Anazi, Ziyad Alshaibani and Panagiotis Kalogianidis

Submitted: 26 May 2022 Reviewed: 17 November 2022 Published: 09 January 2023