Skip to main content
Erschienen in: BMC Cancer 1/2023

Open Access 01.12.2023 | Research

Stem cell collection after lenalidomide, bortezomib and dexamethasone plus elotuzumab or isatuximab in newly diagnosed multiple myeloma patients: a single centre experience from the GMMG-HD6 and -HD7 trials

verfasst von: Joseph Kauer, Emma P. Freundt, Anita Schmitt, Niels Weinhold, Elias K. Mai, Carsten Müller-Tidow, Hartmut Goldschmidt, Marc S. Raab, Katharina Kriegsmann, Sandra Sauer

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

Background

While quadruplet induction therapies deepen responses in newly diagnosed multiple myeloma patients, their impact on peripheral blood stem cell (PBSC) collection remains incompletely understood. This analysis aims to evaluate the effects of prolonged lenalidomide induction and isatuximab- or elotuzumab-containing quadruplet induction therapies on PBSC mobilization and collection.

Methods

A total of 179 transplant-eligible patients with newly diagnosed MM treated at a single academic center were included. The patients were evaluated based on PBSC mobilization and collection parameters, including overall collection results, CD34+ cell levels in peripheral blood, leukapheresis (LP) delays, overall number of LP sessions, and the rate of rescue mobilization with plerixafor. The patients underwent four different induction regimens: Lenalidomide, bortezomib, and dexamethasone (RVd, six 21-day cycles, n = 44), isatuximab-RVd (six 21-day cycles, n = 35), RVd (four 21-day cycles, n = 51), or elotuzumab-RVd (four 21-day cycles, n = 49).

Results

The patients' characteristics were well balanced across the different groups. Collection failures, defined as the inability to collect three sufficient PBSC transplants, were rare (n = 3, 2%), with no occurrences in the isatuximab-RVd and elotuzumab-RVd groups. Intensified induction with six 21-day cycles of RVd did not negatively impact the overall number of collected PBSCs (9.7 × 106/kg bw versus 10.5 × 106/kg bw, p = 0.331) compared to four 21-day cycles of RVd. Plerixafor usage was more common after six cycles of RVd compared to four cycles (16% versus 8%). Addition of elotuzumab to RVd did not adversely affect overall PBSC collection (10.9 × 106/kg bw versus 10.5 × 106/kg bw, p = 0.915). Patients treated with isatuximab-RVd (six cycles) had lower numbers of collected stem cells compared to those receiving RVd (six cycles) induction (8.8 × 106/kg bw versus 9.7 × 106/kg bw, p = 0.801), without experiencing significant delays in LP or increased numbers of LP sessions in a multivariable logistic regression analysis. Plerixafor usage was more common after isatuximab plus RVd compared to RVd alone (34% versus 16%).

Conclusions

This study demonstrates that stem cell collection is feasible after prolonged induction with isatuximab-RVd without collection failures and might be further explored as induction therapy.

Trial registration

Patients were treated within the randomized phase III clinical trials GMMG-HD6 (NCT02495922, 24/06/2015) and GMMG-HD7 (NCT03617731, 24/07/2018). However, during stem cell mobilization and -collection, no study-specific therapeutic intervention was performed.
Hinweise
Katharina Kriegsmann and Sandra Sauer contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

In transplant-eligible patients with newly diagnosed multiple myeloma (MM), induction therapy followed by peripheral blood stem cell (PBSC) collection with granulocyte-colony-stimulating factor (G-CSF), high-dose chemotherapy (HDCT) with melphalan and autologous blood stem cell transplantation (ABSCT) is a standard-of-care [13].
While a single treatment with HDCT/ABSCT prolongs overall survival (OS) [4, 5], tandem treatment might improve outcomes even further and is used in some countries (i.e., Germany) [6, 7]. Patients that achieve a remission of more than 18 months after upfront HDCT/ABSCT may also benefit from salvage HDCT/ABSCT [810]. Therefore, up to three HDCT/ABSCTs may be performed during the treatment course of a MM patient. Accordingly, at our institution, PBSC collection by leukapheresis (LP) is considered successful if three sufficient transplants containing at least ≥ 2.0 × 106 CD34+ cells/kg body weight (bw) have been collected [11, 12]. PBSC mobilization should be performed after induction therapy to ensure collection of a sufficient number of cells.
A variety of factors, such as higher age, melphalan-containing induction or previous radiotherapy involving haematopoietic bone marrow are associated with impaired PBSC collection results or increased rates of collection failure [1315]. In contrast, the impact of lenalidomide induction on stem cell yield is a matter of debate [1621].
Anti-CD38 monoclonal antibodies (mAb), such as daratumumab and isatuximab, significantly improve efficacy and outcomes after induction therapy [2224]. However, various studies demonstrated a negative impact of daratumumab on PBSC collection [2529]. The GMMG-HD7 multicentre study showed impaired overall stem cell collection after Isatuximab-RVd versus RVd (7.71 versus 9.54 × 106/kg CD34+) without further detailed analyses [24]. Herein we report in-depth data on the effect of the anti-CD38 mAb isatuximab on PBSC collection. We further assessed the impact of intensified induction therapy with lenalidomide, bortezomib and dexamethasone (RVd, six 21-day versus four 21-day cycles) and the addition of the anti-SLAMF7 mAb elotuzumab to RVd on PBSC mobilization and collection parameters in patients treated within the randomized phase III clinical trials GMMG-HD6 (NCT02495922, 24/06/2015) and GMMG-HD7 (NCT03617731, 24/07/2018) [24, 30].

Methods

Patient selection and data collection

MM patients that were subjected to autologous PBSC collection at the Department of Haematology, Oncology and Rheumatology at the University Hospital Heidelberg within the clinical trials GMMG-HD6 and GMMG-HD7 between 2015 and 2021 were included (n = 179; HD6 = 100 patients, HD7 = 79 Patients). Patients underwent PSBC collection after mobilization chemotherapy with cyclophosphamide, doxorubicin, and dexamethasone (CAD) or cyclophosphamide. Details for each regimen are given in Table 1.
Table 1
Induction and mobilization therapy
Induction protocol
Dose
Application
Treatment days
(isatuximab)-RVd (21 days/cycle, 6 cycles)
  (Isatuximab)
10 mg/kg
iv
cycle 1: 1, 8, 15
cycle 2: 1, 8
cycle 3, 5: 1, 15
cycle 4, 6: 1
 Lenalidomide
25 mg
po
1–14
 Bortezomib
1.3 mg/qm
sc
1, 4, 8, 11
 Dexamethasone
20 mg
po
cycle 1,3,5: 1, 2, 4, 5, 8, 9, 11, 12, 15
cycle 2,4,6: 1, 2, 4, 5, 8, 9, 11, 12
(elotuzumab)-RVd (21 days/cycle, 4 cycles)
  (Elotuzumab)
10 mg/kg
iv
cycle 1: 1, 8, 15
cycle 3–4: 1, 11
 Lenalidomide
25 mg
po
1–14
 Bortezomib
1.3 mg/qm
sc
1, 4, 8, 11
 Dexamethasone
20 mg
po
1, 2, 4, 5, 8, 9, 11, 12, 15,
Mobilization protocol
CAD (28 days/cycle, 1 cycle)
  Cyclophosphamide
1000 mg/qm
iv
1
  Doxorubicin
15 mg/qm
Iv
1–4
  G-CSF
10 µg/kg bw
iv
9,10,11,12,13,14
Cyclophosphamide mono (28 days/cycle, 1 cycle)
  Cyclophosphamide
1000 mg/qm
iv
1,2
  G-CSF
10 µg/kg bw
iv
9,10,11,12,13,14
G-CSF Granulocyte colony-stimulating factor, iv Intravenous, po Per os, qm Square meter, sc Subcutaneous, RVd Lenalidomide, bortezomib, dexamethasone
Patients characteristics at first diagnosis, first line treatment, remission status, and detailed assessment of PBSC mobilization and collection results were collected retrospectively from routine medical records. Patient characteristics from the GMMG-HD7 trial were collected from study records.

PBSC mobilization and collection

PBSC mobilization and collection by LP was performed according to protocols as previously described [31]. Mobilization protocols are shown in Table 1. Collection of three transplants comprised of ≥ 2.0 × 106 CD34+ cells/kg bw was defined as successful collection. G-CSF (10 µg/kg bw) was applied on days 9—14. On day 14, the first PB CD34+ cell measurement was conducted. LP was initiated if the PB CD34+ cell count exceeded 10/µl. In the absence of infection or other limiting factors, the following LPs were conducted until collection of three transplants comprised of ≥ 2.0 × 106 CD34+ cells/kg bw. In case of collection failure, reflected by insufficient PB CD34+ cell counts or insufficient collection, plerixafor was applied. In short, PB CD34+  < 10/µl after continued G-CSF stimulation until the day after the first planned measurement triggered pre-emptive plerixafor application. At PB CD34+ 10/µl—20/µl, plerixafor was used per treating physician’s discretion. Rescue mobilization was applied if less than 2.0 × 106 CD34+ cells/kg bw were collected during LP1. Key metrics for evaluation of PBSC mobilization and collection include CD34+ cell counts/µl in the peripheral blood, collection delays due to poor mobilization, increased number of LP sessions due to insufficient collection results, collection of CD34+ cells/ kg bw upon the first session and CD34+ cell collection result upon all sessions.

Procedures and definitions

Patients aged ≥ 18 years with untreated multiple myeloma requiring systemic therapy according to International Myeloma Working Group (IMWG) criteria [32] were included in the above mentioned trials. Detailed eligibility criteria are listed in the manuscript by Goldschmidt et al. reporting the primary end point of the GMMG-HD7 trial [24]. Response assessment was conducted according to IMWG criteria with near complete response as additional criterion [33]. Cytogenetic abnormalities were classified as high-risk in case of del(17)(p13), t(4;14)(p16;q32), or t(14;16)(q32;q23) in ≥ 10% of cells.

Statistical analysis

Descriptive statistics were performed by R-Studio (R version 4.0.0, 2020–04-24) and SPSS (SPSS version 27). Data are depicted as absolute numbers and percentages, medians and ranges or means and standard deviations (SD). Categorical variables were compared using the Chi-Square test. Group means of continuous variables were compared by an analysis of variance (ANOVA). Median values of not normally distributed variables were compared by Kruskal–Wallis tests. Multivariable logistic regression analysis was performed with SPSS using the following dependent variables: Overall CD34+ collection results (≥ 10 × 106/kg bw versus < 10), CD34+ cells in PB ((≥ 50/µl versus < 50/µl), LP delay (≥ 1 day versus 0 days), LP sessions (≥ 2 versus 1). The following independent variables were included: Age (> 60 versus ≤ 60 years), High-risk cytogenetic (yes versus no), ISS (3 versus 1–2), Induction six cycles of RVd vs. other, Induction Isa-RVd vs. other, Induction Elo-RVd vs. other and remission prior to mobilization.
(≥ VGPR versus < VGPR). P values < 0.05 were considered statistically significant.

Results

Patients’ characteristics and first line treatment

In this study, 179 patients that underwent induction therapy for newly diagnosed MM with subsequent PBSC mobilization and collection at our institution were included. Patients were grouped according to induction therapy: RVd (six 21-day cycles, n = 44), isatuximab-RVd (six 21-day cycles, n = 35), RVd (four 21-day cycles, n = 51), or elotuzumab-RVd (four 21-day cycles, n = 49). We aimed to assess the effect of intensified induction (six versus four cycles RVd), the addition of isatuximab to RVd (6 cycles) and the addition of elotuzumab to RVd (4 cycles) on PBSC mobilization and collection.
Patients characteristics at first diagnosis were equally distributed among groups, including gender, age, heavy chain type, light chain type, ISS stage, Salmon and Durie stage, and cytogenetic risk profile (Table 2). Response to induction therapy differed significantly between groups in favour of isatuximab-RVd (p = 0.006, Table 3).
Table 2
Patients` characteristics at first diagnosis
Variable
Overall cohort
RVd (six 21-day cycles)
Isatuximab –RVd (six 21-day cycles)
RVd (four 21-day cycles)
elotuzumab-RVd (four 21-day cycles)
p value
n
%
n
%
n
%
n
%
n
%
Patient number
179
100
44
100
35
100
51
100
49
100
/
Gender
          
0.808
  Male
114
62
30
68
23
66
30
59
31
63
  Female
65
38
14
32
12
34
21
41
18
37
FIRST DIAGNOSIS
          
/
Diagnosis
          
  MM
178
99
44
100
35
100
51
100
48
98
  Plasma cell leukaemia
1
1
0
0
0
0
0
0
1
2
Mean age at diagnosis, years (SD)
58
8
58
9
58
7
56
9
58
7
0.615
  Heavy chain type
          
0.523a
   IgG
116
65
25
57
25
71
32
63
34
69
   IgA
32
18
11
25
5
14
6
12
10
20
   IgM
3
2
1
2
1
3
1
2
0
0
   IgD
0
0
0
0
0
0
0
0
0
0
   Double gammopathy
2
1
1
2
0
0
1
2
0
0
   Light chain only
26
15
6
14
4
11
11
22
5
10
   Non-secretory
0
0
0
0
0
0
0
0
0
0
  Light chain type
          
0.117b
   Lambda
64
36
11
25
18
51
15
29
20
41
   Kappa
114
64
32
73
17
49
36
71
29
59
   Double gammopathy
1
1
1
2
0
0
0
0
0
0
   Non-secretory
0
0
0
0
0
0
0
0
0
0
ISS stage
          
0.139
 I
97
54
25
57
14
40
28
55
30
61
 II
41
23
7
16
14
40
11
22
9
18
 III
36
20
12
27
7
20
9
18
8
16
 NA
5
3
0
0
0
0
3
6
2
4
Cytogenetic profile
          
0.793
 High-risk
51
28
11
25
9
26
17
33
14
29
 Standard risk
114
64
29
66
24
69
30
59
31
63
 NA
14
8
4
9
2
6
4
8
4
8
Ig Immunoglobulin, ISS International Staging System, MM Multiple myeloma, NA Not available, PBSC Peripheral blood stem cells, SD Standard deviation
aIgG versus IgA,IgD,IgM, Double Gammopathy versus Light chain only
bLambda versus Kappa
Table 3
First line treatment
Variable
Overall cohort
RVd (six 21-day cycles)
isatuximab-RVd (six 21-day cycles)
RVd (four 21-day cycles)
elotuzumab-RVd (four 21-day cycles)
p value
n
%
n
%
n
%
n
%
n
%
Patient number
179
100
44
100
35
100
51
100
49
100
/
Remission post induction
          
0.006a
 CR and nCR
52
29
22
50
12
34
10
20
8
16
 VGPR
76
42
10
23
16
46
25
49
25
51
 PR or worse
51
29
12
27
7
20
16
32
16
33
Median number of induction cycles (range)
4
(4–6)
6
(4–6)
6
(4–6)
4
(3–4)
4
(3–4)
/
aCR + nCR versus VGPR versus PR or worse
ABSCT Autologous blood stem cell transplantation, CR Complete response, HDCT High-dose chemotherapy, MR Minimal response, NA Not available, n.a. Not applicable, nCR Near complete response, PBSC Peripheral blood stem cells, PD Progressive disease, PR Partial response, RVd Lenalidomide, bortezomib, dexamethasone, SD Stable disease, VGPR Very good partial response, VCD Bortezomib, cyclophosphamide, dexamethasone

PBMC mobilization and collection

For the overall cohort the following PBSC mobilization and collection metrics were observed: either CAD (n = 167) or cyclophosphamide (n = 12) followed by G-CSF was applied for chemotherapy mobilization. Plerixafor application was performed, in general due to a delayed mobilization, in 25 patients (Table 4). Leukapheresis collection was considered successful if three transplants with a sufficient number of CD34+ cells (> 2 × 106/ kg body weight) were collected. Collection failure occurred in three patients, with two of them being treated with six cycles RVd and one with 4 cycles RVd. Main outcome variables for PBSC collection were CD34+ cells in peripheral blood at first collection day, number of LP sessions, the need for plerixafor, LP delay due to poor mobilization as well as CD34+ collection results in the first session and all sessions.
Table 4
PBSC mobilization and collection
Variable
Overall cohort
RVd (six 21-day cycles)
isatuximab-RVd (six 21-day cycles)
RVd (four 21-day cycles)
elotuzumab-RVd (four 21-day cycles)
p value
n
%
n
%
n
%
n
%
n
%
Patient number
179
100
44
100
35
100
51
100
49
100
/
Mobilization regimen
          
/
 CAD
167
93
39
89
28
80
51
100
49
100
 Cyclophosphamide
12
7
5
11
7
20
0
0
0
0
G-CSF dosage
          
 < 0.001
 5 µg/kg bw/day
95
53
1
2
0
0
46
90
48
98
 10 µg/kg bw/day
84
47
43
98
35
100
5
10
1
2
Plerixafor application
          
 < 0.001
 Yes
25
14
7
16
12
34
4
8
2
4
 No
154
86
37
84
23
66
47
92
47
96
Prolonged mobilization
 Median delay, days (range)
0
(0–21)
0
(0–5)
0
(0–5)
0
(0–3)
0
(0–21)
0.785
 Patients with distinct number of days in delay
          
0.777a
  0
135
75
31
70
26
74
39
76
39
80
  1
28
16
9
20
5
14
9
18
5
10
  ≥ 2
16
9
4
9
4
11
3
6
5
10
Blood count prior to first LP
 Mean leukocyte count /nl (SD)
20
(12)
21
15
27
14
17
(8)
16
(8)
 < 0.001
 Mean PB CD34+ cells/µl (SD)
103
(75)
116
80
80
60
111
(91)
97
(57)
0.143
First LP session
 Mean CD34+ cells × 106/kg (SD)
8,2
(4.2)
7,6
3,6
5,8
2,8
8,8
(4,8)
9,8
(4,1)
0.004
 Mean processed blood volume, l (SD)
15,7
(3.8)
15,0
3,8
16,4
3,4
15,4
(4,2)
16,2
(3,3)
0.436
Overall PBSC collection result
 Mean CD34+ cells × 106/kg
10,1
(2.9)
9,7
2,6
8,8
1,8
10,5
(3,5)
10,9
(3,0)
 < 0.001
 Collection failure, n (%)
3
2
2
5
0
0
1
2
0
0
0.301
LP sessions
 Median, n (range)
1
(1–3)
1
(1–3)
2
(1–3)
1
(1–3)
1
(1–3)
0.008
 Patients with distinct number of LP sessions
          
0.017b
  1
110
61
25
57
14
40
34
67
37
76
 
  2
59
33
18
41
17
49
14
27
10
20
 
  3
10
6
1
2
4
11
3
6
2
4
 
bw Body weight, CAD Cyclophosphamide, doxorubicin, dexamethasone, CR Complete response, G-CSF Granulocyte-colony stimulating factor, LP Leukapheresis, MR Minimal response, NA Not available, PBSC Peripheral blood stem cells, PD Progressive disease, PR Partial response, RVd Lenalidomide, bortezomib, dexamethasone, SD Stable disease, VCD Bortezomib, cyclophosphamide, dexamethasone, VGPR Very good partial response
a0 days versus 1 day versus 2 or more days
b1 LP versus 2 LPs versus ≥ 3 LPs

Effect of intensified induction on PBSC mobilization/collection

First, we focused on the effect of intensified induction with prolonged RVd (six 21-day cycles) versus standard RVd (four 21-day cycles). Mean PB CD34+ cell count (116/µl versus 111/µl, p = 0.999), number of LP sessions (median 1 (range 1–3) versus 1 (range 1–3), plerixafor use (16% versus 8%), LP delay (median 0 (range 0–5) versus 0 days (range 0–3), p = 0.999), mean CD34+ cell collection result upon first LP (7.6 × 106/kg bw, interquartile range [IQR] 5.1) versus 8.8 × 106/kg bw, IQR 6.7, p = 0.335) and overall CD34+ cell collection result (9.7 × 106/kg bw, IQR 3.1, versus 10.5 × 106/kg bw, IQR 3.9, p = 0.331) did not significantly differ between these groups (Figs. 1, 2 and Table 4).

Impact of quadruplet therapy on PBSC mobilization/collection

Next, the impact of addition of isatuximab to RVd (six 21-day cycles) was evaluated. Mean PB CD34+ cell count (80/µl versus 116/µl, p = 0.424), number of LP sessions (median 2 (range 1–3) versus 1 (range 1–3), p = 0.401), plerixafor use (34% versus 16%, p = 0.176), LP delay (median 0 (range 0–5) versus 0 days (range 0–5, p = 0.999), mean CD34+ cell collection result upon first LP (5.8 × 106/kg bw, IWR 3.7 versus 7.6 × 106/kg bw, IQR 5.1, p = 0.460) and overall CD34+ cell collection result (8.8 × 106/kg bw, IQR 1.8 versus 9.7 × 106/kg bw, IQR 3.1, p = 0.801) did not significantly differ between groups (Figs. 1, 2 and Table 4).
Addition of elotuzumab to RVd (four 21-day cycles) did not hamper PBSC collection results. Mean PB CD34+ cell count (97/µl versus 111/µl, p = 0.807), number of LP sessions (median 1 (range 1–3) versus 1 (range 1–3)), plerixafor use (4% versus 8%), LP delay (median 0 days (range 0–21) versus 0 days (range 0–3)), mean CD34+ cell collection result upon first LP (9.8 × 106/kg bw, IQR 5.4 versus 8.8 × 106/kg bw, IQR 6.7, p = 0.625) and overall CD34+ cell collection result (10.9 × 106/kg bw, IQR 3.8 versus 10.5 × 106/kg bw, IQR 3.9, p = 0.915) did not significantly differ between groups (Figs. 1, 2 and Table 4).

Multivariate analysis

Multivariable logistic regression analysis was performed regarding the outcome variables mobilization delay, number of LP sessions, peripheral blood CD34+ levels and overall CD34+ cell collection results. Age > 60, High-risk cytogenetics, ISS stage 3 (versus 1–2), induction regimen and remission after induction (≥ VGPR versus < VGPR) had no significant impact on LP delay, LP sessions and peripheral blood CD34+ levels (Table 5). Induction with isatuximab-RVd significantly reduced the rate of exceptionally high overall collection results (≥ 107/kg bw, Odds ratio 0.17, 95% CI 0.05–0.51, p = 0.002). However, no relative collection failure, defined by the inability to collect three sufficient PBSC transplants, was observed with isatuximab-RVd.
Table 5
Multivariable logistic regression analysis
Variable
Overall CD34+ collection results (≥ 10 × 106/kg bw versus < 10)
CD34+ cells in PB ((≥ 50/µl versus < 50/µl)
Odds ratio
[95% CI]
p value
Odds ratio
[95% CI]
p value
Age (> 60 versus ≤ 60 years)
0.89
[0.45 -1.76]
0.743
0.68
[0.34 -1.38]
0.289
High-risk cytogenetic (yes versus no)
0.95
[0,41 -2.20]
0.904
1.14
[0.47 -2.80]
0.773
ISS (3 versus 1–2)
1.50
[0.72 -3.09]
0.279
1.07
[0.50 -2.30]
0.854
Induction 6 × RVd vs. other
0.88
[0.37 -2.09]
0.770
1.22
[0.49 -3.03]
0.671
Induction 6 × Isa-RVd vs. other
0.17
[0.05 -0.51]
0.002
1.53
[0.57 -4.07]
0.399
Induction 4 × Elo-RVd vs. other
1.24
[0.54 -2.88]
0.611
2.59
[0.97 -6.89]
0.057
Remission prior to mobilization
(≥ VGPR versus < VGPR)
1.39
[0.66 -2.90]
0.384
1.34
[0.62 -2.90]
0.460
Variable
LP delay (≥ 1 day versus 0 days)
LP sessions ((≥ 2 versus 1)
Odds ratio
[95% CI]
p value
Odds ratio
[95% CI]
p value
Age (> 60 versus ≤ 60 years)
1.71
[0.79 – 3.70]
0.172
1.03
[0.52 -2.04]
0.929
High-risk cytogenetic (yes versus no)
2.37
[0.98 -5.73]
0.055
0.80
[0.34 -1.87]
0.594
ISS (3 versus 1–2)
1.20
[0.51 -2.80]
0.677
1.55
[0.74 -3.26]
0.247
Induction 6 × RVd vs. other
1.72
[0.62 -4.72]
0.296
1.69
[0.69 -4.12]
0.252
Induction 6 × Isa-RVd vs. other
1.30
[0.43 -3.92]
0.646
2.47
[0.97 -6.30]
0.058
Induction 4 × Elo-RVd vs. other
0.80
[0.27 -2.41]
0.693
0.61
[0.24 -1.57]
0.305
Remission prior to mobilization
(≥ VGPR versus < VGPR)
0.76
[0.32 -1.78]
0.528
0.79
[0.38 -1.66]
0.531
CI Confidence interval, Elo Elotuzumab, Isa Isatuximab, ISS International Staging System, LP Leukapheresis, PB Peripheral blood, RVd Lenalidomide, bortezomib, dexamethasone, VGPR Very good

Discussion

This academic single centre study provides novel data on the impact of different state-of-the-art induction regimen on PBSC mobilization and collection metrics in patients with newly diagnosed MM. While several factors have been described as being harmful to PBSC collection [1315], no such data are available for quadruplet induction therapies comprising isatuximab and comparisons of different lengths of RVd induction. This study was able to assess important direct and indirect parameters of successful stem cell collection such as LP delay, number of LP sessions, plerixafor utilization, and overall collection results.
Quadruplet induction therapies such as daratumumab, lenalidomide, bortezomib, and dexamethasone (Dara-RVd) or daratumumab, bortezomib, thalidomide, and dexamethasone (Dara-VTd) are now standard-of-care for transplant-eligible patients with newly diagnosed multiple myeloma [22, 23]. Since CD38 is expressed on CD34+ progenitor cells [34], concerns regarding impaired stem cell mobilization after CD38-targeting antibody therapy have been raised. A negative impact of daratumumab on PBSC mobilization and collection has been described in the setting of several clinical trials. Within the phase III CASSIOPEIA trial, overall stem cell collection was impaired after Dara-VTd compared to VTd (6.7 vs. 10.0 × 106/kg bw), additionally mirrored by increased utilization of plerixafor (21.7 vs. 7.9%) and higher rates of relative collection failure (reported as collection < 5 × 106/kg bw, 24.6% vs. 11.4%) [35]. Though our clinical practice is similar and includes cyclophosphamide-based mobilization chemotherapy and a rescue policy including plerixafor, the collection failure rate in the CASSIOPEIA trial was higher compared to our study.
In the phase II GRIFFIN trial, lower stem cell yield (8.3 versus 9.4 × 106/kg bw) and higher utilization of plerixafor (72% vs. 55%) was seen after daratumumab plus RVd versus RVd alone [26]. However, institutional practice regarding plerixafor rescue or upfront application differed between participating centres, with some using steady-state mobilisation. Furthermore, cyclophosphamide mobilization chemotherapy was only permitted after unsuccessful mobilization with G-CSF with or without plerixafor. In patients that underwent a rescue plerixafor strategy similar to the strategy employed at our centre, 41% of patients received plerixafor after daratumumab-RVd versus 27% after RVd [26].
In contrast to the data on daratumumab, our results suggest that isatuximab does not increase the risk for relative collection failure although the total number of collected stem cells is lowered. Furthermore, utilization of plerixafor was required in a minority of patients (34%) and upfront application to all patients might not be necessary after 18 weeks of isatuximab-RVd, thus limiting the economic burden of this regimen. However, a direct comparison between daratumumab and isatuximab regarding stem cell collection yield cannot be drawn from our data. The observation is in line with an extensive in vitro study, in which isatuximab did not induce bone marrow toxicity in vitro while effectively lysing MM cells [36]. The multicentre data of the GMMG-HD7 trial showed a significantly impaired overall collection after induction therapy isatuximab-RVd versus RVd alone (7.71 versus 9.54 × 106/kg CD34+). The smaller gap in overall collection rate at our centre after isatuximab-RVd versus RVd alone (8.8 versus 9.7 × 106/kg CD34+) might be explained by the extensive experience and high patient volume. Some patient characteristics might have been beneficial for stem cell collection in our cohort, such as the relatively low median age (58 years) and the use of mobilization chemotherapy in all patients. Furthermore, collection was considered successful after collection of at least 6 × 106/kg bw, which might be lower than collection goals in other studies.
This study also aimed to compare induction regimen in newly diagnosed MM patients receiving lenalidomide in either a standard regimen (4 cycles, 25 mg/day for 14 days) or a prolonged regimen (6 cycles, 25 mg/d for 14 days). No significant differences regarding PBSC mobilization and collection metrics were observed in our study. The previously reported data on lenalidomide treatment prior to PBSC collection reveals contradictory results. Initially, Kumar et al. suggested a negative impact of lenalidomide on stem cell collection in patients treated with lenalidomide-dexamethasone [16]. This was confirmed by Bhutani et al., who found that lenalidomide application over eight or more courses correlates with poor collection results and increased number of LP sessions [17]. More recent data on lenalidomide in a small cohort receiving also RVd suggests delayed mobilization and increased numbers of LP sessions [18]. Another recent study, in contrast, did not reveal any negative effects of prolonged lenalidomide exposure (> 6 cycles) on LP results [20]. Of note, the latter study comprised a cohort of patients treated with a variety of different induction regimens containing lenalidomide, which hampers direct comparisons. The standardized lenalidomide-containing induction therapies in our cohort ensure comparability and allow for multivariate analyses, thus reducing confounders.
The SLAMF7 antibody elotuzumab is an established therapeutic option in relapsed MM [37, 38]. While being present on MM cells, SLAMF7 is not expressed on other bone marrow cells [39]. Detailed data on the impact of elotuzumab treatment prior to PBSC transplantation are missing. We here provide evidence that elotuzumab does not affect PBSC mobilization and collection metrics, which is in line with the multicentre data from the GMMG-HD6 trial [40]. Likely, due to the negative results of the SWOG-1211 and the GMMG-HD6 trials, elotuzumab will not be utilized in the front-line setting combined with RVd in transplant-eligible patients with newly diagnosed MM [30, 41]. However, studies combining elotuzumab with other regimen in the front-line setting are ongoing.
Limitations of our study include its single centre design and its retrospective nature. While patients were treated within randomized trials, stem cell collection was not an endpoint of either trial. Comparisons with outcomes in other trials or other centres might therefore be impaired. The collection results presented represent outcomes after quadruplet therapy followed by cyclophosphamide-based mobilization chemotherapy and might not be transferable to steady-state mobilization.

Conclusions

In summary, our study demonstrated that stem cell collection is feasible after prolonged induction with isatuximab-RVd and did not lead to collection failure in this academic single centre cohort. Moreover, induction therapy with RVd (21-days) for four or six cycles did not negatively impact overall collection results.

Author information

Not applicable.

Acknowledgements

The German Speaking Myeloma Multicenter Group (GMMG) initiated and designed the HD6 and HD7 trial. Sanofi, Chugai and Bristol Myers Squibb reviewed the manuscript. For the publication fee we acknowledge financial support by Deutsche Forschungsgemeinschaft within the funding programme “Open Access Publikationskosten” as well as by Heidelberg University.

Declarations

All patients provided written informed consent. The trial was conducted in accordance with the International Conference on Harmonisation Good Clinical Practice guidelines, the Declaration of Helsinki principles, and local legal and regulatory requirements and approved by the ethics committee of the University of Heidelberg.
Not applicable.

Competing interests

Sandra Sauer: travel grants or honoraria for presentations for Celgene, BMS, Janssen, Takeda and Amgen. Anita Schmitt: travel grants from Hexal and Jazz Pharmaceuticals. Research grant from Therakos/Mallinckrodt. Consultant by Janssen-Cilag and BMS. Anita Schmitt is co-founder of TolerogenixX LtD. Anita Schmitt is part-time employee of TolerogenixX Ltd.Hartmut Goldschmidt: Grants and/or provision of Investigational Medicinal Product: Amgen, Array Biopharma/Pfizer, BMS/Celgene, Chugai, Dietmar-Hopp-Foundation, Janssen, Johns Hopkins University, Mundipharma GmbH, Sanofi. Research Support: Amgen, BMS, Celgene, GlycoMimetics Inc., GSK, Heidelberg Pharma, Hoffmann-La Roche, Karyopharm, Janssen, Incyte Corporation, Millenium Pharmaceuticals Inc., Molecular Partners, Merck Sharp and Dohme (MSD), MorphoSys AG, Pfizer, Sanofi, Takeda, Novartis. Advisory Boards: Amgen, BMS, Janssen, Sanofi, Adaptive Biotechnology. Honoraria: Amgen, BMS, Chugai, GlaxoSmithKline (GSK), Janssen, Novartis, Sanofi, Pfizer. Support for attending meetings and/or travel: Amgen, BMS, GlaxoSmithKline (GSK), Janssen, Novartis, Sanofi, Pfizer. Katharina Kriegsmann: Research funding from Bristol Myers Squibb and Sanofi-Aventis Deutschland GmbH.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Dimopoulos MA, Moreau P, Terpos E, et al. Multiple myeloma: EHA-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32(3):309–22.CrossRefPubMed Dimopoulos MA, Moreau P, Terpos E, et al. Multiple myeloma: EHA-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32(3):309–22.CrossRefPubMed
2.
Zurück zum Zitat Cavo M, Gay F, Beksac M, et al. Autologous haematopoietic stem-cell transplantation versus bortezomib–melphalan–prednisone, with or without bortezomib–lenalidomide–dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): A multicentre, randomised, open-label, phase 3 study. The Lancet Haematology. 2020;7(6):e456–68.CrossRefPubMed Cavo M, Gay F, Beksac M, et al. Autologous haematopoietic stem-cell transplantation versus bortezomib–melphalan–prednisone, with or without bortezomib–lenalidomide–dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): A multicentre, randomised, open-label, phase 3 study. The Lancet Haematology. 2020;7(6):e456–68.CrossRefPubMed
3.
Zurück zum Zitat Attal M, Lauwers-Cances V, Hulin C, et al. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med. 2017;376(14):1311–20.CrossRefPubMedPubMedCentral Attal M, Lauwers-Cances V, Hulin C, et al. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med. 2017;376(14):1311–20.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Attal M, Harousseau J-L, Stoppa A-M, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. N Engl J Med. 1996;335(2):91–7.CrossRefPubMed Attal M, Harousseau J-L, Stoppa A-M, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. N Engl J Med. 1996;335(2):91–7.CrossRefPubMed
5.
Zurück zum Zitat Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med. 2003;348(19):1875–83.CrossRefPubMed Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med. 2003;348(19):1875–83.CrossRefPubMed
6.
Zurück zum Zitat Attal M, Harousseau J-L, Facon T, et al. Single versus double autologous stem-cell transplantation for multiple myeloma. N Engl J Med. 2003;349(26):2495–502.CrossRefPubMed Attal M, Harousseau J-L, Facon T, et al. Single versus double autologous stem-cell transplantation for multiple myeloma. N Engl J Med. 2003;349(26):2495–502.CrossRefPubMed
7.
Zurück zum Zitat Regelink JC, van Roessel C, van Galen K, Ossenkoppele GJ, Huijgens PC, Zweegman S. Long-term follow-up of tandem autologous stem-cell transplantation in multiple myeloma. J Clin Oncol. 2010;28(35):e741-743 author reply e744.CrossRefPubMed Regelink JC, van Roessel C, van Galen K, Ossenkoppele GJ, Huijgens PC, Zweegman S. Long-term follow-up of tandem autologous stem-cell transplantation in multiple myeloma. J Clin Oncol. 2010;28(35):e741-743 author reply e744.CrossRefPubMed
8.
Zurück zum Zitat Auner HW, Szydlo R, Rone A, et al. Salvage autologous stem cell transplantation for multiple myeloma relapsing or progressing after up-front autologous transplantation. Leuk Lymphoma. 2013;54(10):2200–4.CrossRefPubMed Auner HW, Szydlo R, Rone A, et al. Salvage autologous stem cell transplantation for multiple myeloma relapsing or progressing after up-front autologous transplantation. Leuk Lymphoma. 2013;54(10):2200–4.CrossRefPubMed
9.
Zurück zum Zitat Michaelis LC, Saad A, Zhong X, et al. Salvage second hematopoietic cell transplantation in myeloma. Biol Blood Marrow Transplant. 2013;19(5):760–6.CrossRefPubMed Michaelis LC, Saad A, Zhong X, et al. Salvage second hematopoietic cell transplantation in myeloma. Biol Blood Marrow Transplant. 2013;19(5):760–6.CrossRefPubMed
10.
Zurück zum Zitat Lemieux E, Hulin C, Caillot D, et al. Autologous stem cell transplantation: an effective salvage therapy in multiple myeloma. Biol Blood Marrow Transplant. 2013;19(3):445–9.CrossRefPubMed Lemieux E, Hulin C, Caillot D, et al. Autologous stem cell transplantation: an effective salvage therapy in multiple myeloma. Biol Blood Marrow Transplant. 2013;19(3):445–9.CrossRefPubMed
11.
Zurück zum Zitat Huijgens P, Dekker-Van Roessel H, Jonkhoff A, et al. High-dose melphalan with G-CSF-stimulated whole blood rescue followed by stem cell harvesting and busulphan/cyclophosphamide with autologous stem cell transplantation in multiple myeloma. Bone Marrow Transplant. 2001;27(9):925–31.CrossRefPubMed Huijgens P, Dekker-Van Roessel H, Jonkhoff A, et al. High-dose melphalan with G-CSF-stimulated whole blood rescue followed by stem cell harvesting and busulphan/cyclophosphamide with autologous stem cell transplantation in multiple myeloma. Bone Marrow Transplant. 2001;27(9):925–31.CrossRefPubMed
12.
Zurück zum Zitat Hübel K, de la Rubia J, Azar N, Corradini P. Current status of haematopoietic autologous stem cell transplantation in lymphoid malignancies: a European perspective. Eur J Haematol. 2015;94(1):12–22.CrossRefPubMed Hübel K, de la Rubia J, Azar N, Corradini P. Current status of haematopoietic autologous stem cell transplantation in lymphoid malignancies: a European perspective. Eur J Haematol. 2015;94(1):12–22.CrossRefPubMed
13.
Zurück zum Zitat Perseghin P, Terruzzi E, Dassi M, et al. Management of poor peripheral blood stem cell mobilization: incidence, predictive factors, alternative strategies and outcome. A retrospective analysis on 2177 patients from three major Italian institutions. Transfus Apher Sci. 2009;41(1):33–7.CrossRefPubMed Perseghin P, Terruzzi E, Dassi M, et al. Management of poor peripheral blood stem cell mobilization: incidence, predictive factors, alternative strategies and outcome. A retrospective analysis on 2177 patients from three major Italian institutions. Transfus Apher Sci. 2009;41(1):33–7.CrossRefPubMed
14.
Zurück zum Zitat Wuchter P, Ran D, Bruckner T, et al. Poor mobilization of hematopoietic stem cells—definitions, incidence, risk factors, and impact on outcome of autologous transplantation. Biol Blood Marrow Transplant. 2010;16(4):490–9.CrossRefPubMed Wuchter P, Ran D, Bruckner T, et al. Poor mobilization of hematopoietic stem cells—definitions, incidence, risk factors, and impact on outcome of autologous transplantation. Biol Blood Marrow Transplant. 2010;16(4):490–9.CrossRefPubMed
15.
Zurück zum Zitat Sauer S, Erdmann K, Jensen AD, et al. Local radiation therapy before and during induction delays stem cell mobilization and collection in multiple myeloma patients. Transplant Cell Ther. 2021;27(10):e871-876. e811.CrossRef Sauer S, Erdmann K, Jensen AD, et al. Local radiation therapy before and during induction delays stem cell mobilization and collection in multiple myeloma patients. Transplant Cell Ther. 2021;27(10):e871-876. e811.CrossRef
16.
Zurück zum Zitat Kumar S, Dispenzieri A, Lacy M, et al. Impact of lenalidomide therapy on stem cell mobilization and engraftment post-peripheral blood stem cell transplantation in patients with newly diagnosed myeloma. Leukemia. 2007;21(9):2035–42.CrossRefPubMed Kumar S, Dispenzieri A, Lacy M, et al. Impact of lenalidomide therapy on stem cell mobilization and engraftment post-peripheral blood stem cell transplantation in patients with newly diagnosed myeloma. Leukemia. 2007;21(9):2035–42.CrossRefPubMed
17.
Zurück zum Zitat Bhutani D, Zonder J, Valent J, et al. Evaluating the effects of lenalidomide induction therapy on peripheral stem cells collection in patients undergoing autologous stem cell transplant for multiple myeloma. Support Care Cancer. 2013;21(9):2437–42.CrossRefPubMed Bhutani D, Zonder J, Valent J, et al. Evaluating the effects of lenalidomide induction therapy on peripheral stem cells collection in patients undergoing autologous stem cell transplant for multiple myeloma. Support Care Cancer. 2013;21(9):2437–42.CrossRefPubMed
18.
Zurück zum Zitat Partanen A, Valtola J, Silvennoinen R, et al. Impact of lenalidomide-based induction therapy on the mobilization of CD34+ cells, blood graft cellular composition, and post-transplant recovery in myeloma patients: a prospective multicenter study. Transfusion. 2017;57(10):2366–72.CrossRefPubMed Partanen A, Valtola J, Silvennoinen R, et al. Impact of lenalidomide-based induction therapy on the mobilization of CD34+ cells, blood graft cellular composition, and post-transplant recovery in myeloma patients: a prospective multicenter study. Transfusion. 2017;57(10):2366–72.CrossRefPubMed
19.
Zurück zum Zitat Dosani T, Covut F, Pinto R, et al. Impact of lenalidomide on collected hematopoietic myeloid and erythroid progenitors: peripheral stem cell collection may not be affected. Leuk Lymphoma. 2019;60(9):2199–206.CrossRefPubMed Dosani T, Covut F, Pinto R, et al. Impact of lenalidomide on collected hematopoietic myeloid and erythroid progenitors: peripheral stem cell collection may not be affected. Leuk Lymphoma. 2019;60(9):2199–206.CrossRefPubMed
20.
Zurück zum Zitat Cowan AJ, Stevenson PA, Green DJ, et al. Prolonged lenalidomide therapy does not impact autologous peripheral blood stem cell mobilization and collection in multiple myeloma patients: a single-center retrospective analysis. Transplant Cell Ther. 2023;27(8):661-e1. Cowan AJ, Stevenson PA, Green DJ, et al. Prolonged lenalidomide therapy does not impact autologous peripheral blood stem cell mobilization and collection in multiple myeloma patients: a single-center retrospective analysis. Transplant Cell Ther. 2023;27(8):661-e1.
21.
Zurück zum Zitat Laurent V, Fronteau C, Antier C, et al. Autologous stem-cell collection following VTD or VRD induction therapy in multiple myeloma: a single-center experience. Bone Marrow Transplant. 2021;56(2):395–9.CrossRefPubMed Laurent V, Fronteau C, Antier C, et al. Autologous stem-cell collection following VTD or VRD induction therapy in multiple myeloma: a single-center experience. Bone Marrow Transplant. 2021;56(2):395–9.CrossRefPubMed
22.
Zurück zum Zitat Voorhees PM, Kaufman JL, Laubach J, et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial. Blood. 2020;136(8):936–45.CrossRefPubMedPubMedCentral Voorhees PM, Kaufman JL, Laubach J, et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial. Blood. 2020;136(8):936–45.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet. 2019;394(10192):29–38.CrossRefPubMed Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet. 2019;394(10192):29–38.CrossRefPubMed
24.
Zurück zum Zitat Goldschmidt H, Mai EK, Bertsch U, et al. Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone as induction therapy for newly diagnosed, transplantation-eligible patients with multiple myeloma (GMMG-HD7): part 1 of an open-label, multicentre, randomised, active-controlled, phase 3 trial. Lancet Haematol. 2022;9(11):e810–21.CrossRefPubMed Goldschmidt H, Mai EK, Bertsch U, et al. Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone as induction therapy for newly diagnosed, transplantation-eligible patients with multiple myeloma (GMMG-HD7): part 1 of an open-label, multicentre, randomised, active-controlled, phase 3 trial. Lancet Haematol. 2022;9(11):e810–21.CrossRefPubMed
25.
Zurück zum Zitat Manjappa S, Fox R, Reese J, et al. Impact of daratumumab on stem cell collection, graft composition and engraftment among multiple myeloma patients undergoing autologous stem cell transplant. Blood. 2020;136:35–7.CrossRef Manjappa S, Fox R, Reese J, et al. Impact of daratumumab on stem cell collection, graft composition and engraftment among multiple myeloma patients undergoing autologous stem cell transplant. Blood. 2020;136:35–7.CrossRef
26.
Zurück zum Zitat Chhabra S, Callander N, Watts NL, et al. Stem cell mobilization yields with daratumumab-and lenalidomide-containing quadruplet induction therapy in newly diagnosed multiple myeloma: findings from the MASTER and GRIFFIN trials. Transplant Cell Ther. 2023;29(3):174. e171-174. e110.CrossRef Chhabra S, Callander N, Watts NL, et al. Stem cell mobilization yields with daratumumab-and lenalidomide-containing quadruplet induction therapy in newly diagnosed multiple myeloma: findings from the MASTER and GRIFFIN trials. Transplant Cell Ther. 2023;29(3):174. e171-174. e110.CrossRef
27.
Zurück zum Zitat Chhabra S, Callander NS, Costa LJ, et al. Stem cell collection with daratumumab (DARA)-based regimens in transplant-eligible newly diagnosed multiple myeloma (NDMM) Patients (pts) in the griffin and master studies. Blood. 2021;138:2852.CrossRef Chhabra S, Callander NS, Costa LJ, et al. Stem cell collection with daratumumab (DARA)-based regimens in transplant-eligible newly diagnosed multiple myeloma (NDMM) Patients (pts) in the griffin and master studies. Blood. 2021;138:2852.CrossRef
28.
Zurück zum Zitat Lemonakis K, Tätting L, Lisak M, et al. Impact of daratumumab-based induction on stem cell collection parameters in Swedish myeloma patients. Haematologica. 2023;108(2):610.CrossRefPubMed Lemonakis K, Tätting L, Lisak M, et al. Impact of daratumumab-based induction on stem cell collection parameters in Swedish myeloma patients. Haematologica. 2023;108(2):610.CrossRefPubMed
29.
Zurück zum Zitat Sauer S, Kriegsmann K, Nientiedt C, et al. Autologous stem cell collection after daratumumab, bortezomib, thalidomide, and dexamethasone versus bortezomib, cyclophosphamide, and dexamethasone in newly diagnosed multiple myeloma. Transfus Med Hemotherapy. 2023;50:371–80. Sauer S, Kriegsmann K, Nientiedt C, et al. Autologous stem cell collection after daratumumab, bortezomib, thalidomide, and dexamethasone versus bortezomib, cyclophosphamide, and dexamethasone in newly diagnosed multiple myeloma. Transfus Med Hemotherapy. 2023;50:371–80.
30.
Zurück zum Zitat Goldschmidt H, Mai EK, Bertsch U, et al. Elotuzumab in combination with lenalidomide, bortezomib, dexamethasone and autologous transplantation for newly-diagnosed multiple myeloma: results from the randomized phase III GMMG-HD6 trial. Blood. 2021;138:486.CrossRef Goldschmidt H, Mai EK, Bertsch U, et al. Elotuzumab in combination with lenalidomide, bortezomib, dexamethasone and autologous transplantation for newly-diagnosed multiple myeloma: results from the randomized phase III GMMG-HD6 trial. Blood. 2021;138:486.CrossRef
31.
Zurück zum Zitat Kriegsmann K, Schmitt A, Kriegsmann M, et al. Orchestration of chemomobilization and G-CSF administration for successful hematopoietic stem cell collection. Biol Blood Marrow Transplant. 2018;24(6):1281–8.CrossRefPubMed Kriegsmann K, Schmitt A, Kriegsmann M, et al. Orchestration of chemomobilization and G-CSF administration for successful hematopoietic stem cell collection. Biol Blood Marrow Transplant. 2018;24(6):1281–8.CrossRefPubMed
32.
Zurück zum Zitat Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538–48.CrossRefPubMed Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538–48.CrossRefPubMed
33.
Zurück zum Zitat Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17(8):e328–46.CrossRefPubMed Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17(8):e328–46.CrossRefPubMed
34.
Zurück zum Zitat Campana D, Suzuki T, Todisco E, Kitanaka A. CD38 in hematopoiesis. Chem Immunol. 2000;75:169–88.PubMed Campana D, Suzuki T, Todisco E, Kitanaka A. CD38 in hematopoiesis. Chem Immunol. 2000;75:169–88.PubMed
35.
Zurück zum Zitat Hulin C, Offner F, Moreau P, et al. Stem cell yield and transplantation in transplant-eligible newly diagnosed multiple myeloma patients receiving daratumumab plus bortezomib/thalidomide/dexamethasone in the phase III CASSIOPEIA study. Haematologica. 2021;106(8):2257.CrossRefPubMedPubMedCentral Hulin C, Offner F, Moreau P, et al. Stem cell yield and transplantation in transplant-eligible newly diagnosed multiple myeloma patients receiving daratumumab plus bortezomib/thalidomide/dexamethasone in the phase III CASSIOPEIA study. Haematologica. 2021;106(8):2257.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Zhu C, Song Z, Wang A, et al. Isatuximab acts through Fc-dependent, independent, and direct pathways to kill multiple myeloma cells. Front Immunol. 2020;11:1771.CrossRefPubMedPubMedCentral Zhu C, Song Z, Wang A, et al. Isatuximab acts through Fc-dependent, independent, and direct pathways to kill multiple myeloma cells. Front Immunol. 2020;11:1771.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Dimopoulos MA, Lonial S, White D, et al. Elotuzumab, lenalidomide, and dexamethasone in RRMM: final overall survival results from the phase 3 randomized ELOQUENT-2 study. Blood Cancer J. 2020;10(9):1–10.CrossRef Dimopoulos MA, Lonial S, White D, et al. Elotuzumab, lenalidomide, and dexamethasone in RRMM: final overall survival results from the phase 3 randomized ELOQUENT-2 study. Blood Cancer J. 2020;10(9):1–10.CrossRef
38.
Zurück zum Zitat Lonial S, Dimopoulos M, Palumbo A, et al. Elotuzumab therapy for relapsed or refractory multiple myeloma. N Engl J Med. 2015;373(7):621–31.CrossRefPubMed Lonial S, Dimopoulos M, Palumbo A, et al. Elotuzumab therapy for relapsed or refractory multiple myeloma. N Engl J Med. 2015;373(7):621–31.CrossRefPubMed
39.
Zurück zum Zitat Soh KT, Tario Jr JD, Hahn T, et al. CD319 (SLAMF7) an alternative marker for detecting plasma cells in the presence of daratumumab or elotuzumab. Cytometry B Clin Cytom. 2021;100(4):497–508. Soh KT, Tario Jr JD, Hahn T, et al. CD319 (SLAMF7) an alternative marker for detecting plasma cells in the presence of daratumumab or elotuzumab. Cytometry B Clin Cytom. 2021;100(4):497–508.
40.
Zurück zum Zitat Wuchter P, Bertsch U, Salwender H-J, et al. Evaluation of stem cell mobilization in patients with multiple myeloma after lenalidomide-based induction chemotherapy within the GMMG-HD6 trial. Blood. 2016;128(22):3373.CrossRef Wuchter P, Bertsch U, Salwender H-J, et al. Evaluation of stem cell mobilization in patients with multiple myeloma after lenalidomide-based induction chemotherapy within the GMMG-HD6 trial. Blood. 2016;128(22):3373.CrossRef
41.
Zurück zum Zitat Usmani SZ, Hoering A, Ailawadhi S, et al. Bortezomib, lenalidomide, and dexamethasone with or without elotuzumab in patients with untreated, high-risk multiple myeloma (SWOG-1211): primary analysis of a randomised, phase 2 trial. Lancet Haematol. 2021;8(1):e45–54.CrossRefPubMed Usmani SZ, Hoering A, Ailawadhi S, et al. Bortezomib, lenalidomide, and dexamethasone with or without elotuzumab in patients with untreated, high-risk multiple myeloma (SWOG-1211): primary analysis of a randomised, phase 2 trial. Lancet Haematol. 2021;8(1):e45–54.CrossRefPubMed
Metadaten
Titel
Stem cell collection after lenalidomide, bortezomib and dexamethasone plus elotuzumab or isatuximab in newly diagnosed multiple myeloma patients: a single centre experience from the GMMG-HD6 and -HD7 trials
verfasst von
Joseph Kauer
Emma P. Freundt
Anita Schmitt
Niels Weinhold
Elias K. Mai
Carsten Müller-Tidow
Hartmut Goldschmidt
Marc S. Raab
Katharina Kriegsmann
Sandra Sauer
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-11507-9

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.