Next Article in Journal
T-Cell Repertoire in Tumor Radiation: The Emerging Frontier as a Radiotherapy Biomarker
Next Article in Special Issue
Pretransplant Systemic Lipidomic Profiles in Allogeneic Stem Cell Transplant Recipients
Previous Article in Journal
Concomitant Hepatic Artery Resection for Advanced Perihilar Cholangiocarcinoma: A Narrative Review
Previous Article in Special Issue
Improvement in Post-Autologous Stem Cell Transplant Survival of Multiple Myeloma Patients: A Long-Term Institutional Experience
 
 
cancers-logo
Article Menu

Article Menu

Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Allogeneic Stem Cell Transplantation in Mantle Cell Lymphoma; Insights into Its Potential Role in the Era of New Immunotherapeutic and Targeted Therapies: The GETH/GELTAMO Experience

by
Antonio Gutierrez
1,*,
Leyre Bento
1,
Silvana Novelli
2,
Alejandro Martin
3,
Gonzalo Gutierrez
4,
Maria Queralt Salas
4,
Mariana Bastos-Oreiro
5,
Ariadna Perez
6,
Rafael Hernani
6,
Maria Cruz Viguria
7,
Oriana Lopez-Godino
8,
Juan Montoro
9,
Jose Luis Piñana
9,
Christelle Ferra
10,
Rocio Parody
11,
Carmen Martin
12,
Ignacio Español
13,
Lucrecia Yañez
14,
Guillermo Rodriguez
15,
Joud Zanabili
16,
Pilar Herrera
17,
Maria Rosario Varela
18,
Antonia Sampol
1,
Carlos Solano
6,
Dolores Caballero
3 and
on behalf of the Grupo Español de Trasplante de Progenitores Hematopoyéticos (GETH) and Grupo Español de Linfoma y Trasplante Autólogo (GELTAMO)
add Show full author list remove Hide full author list
1
Son Espases University HospitaI, IdISBa, 07120 Palma, Spain
2
Hospital Sant Creu i Sant Pau, Service of Hematology, 08025 Barcelona, Spain
3
Hospital Universitario Salamanca, IBSAL, CIBERONC, 37007 Salamanca, Spain
4
Hospital Clinic, ICMHO, 08036 Barcelona, Spain
5
Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
6
Hospital Clínico Valencia, 46010 Valencia, Spain
7
Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
8
Hospital Morales Meseguer, 30008 Murcia, Spain
9
Hospital La Fe, Facultad de Medicina, Universidad Catolica de Valencia, 46026 Valencia, Spain
10
ICO-IJC-Hospital Germans Trias i Pujol, 08916 Badalona, Spain
11
Institut Català d’Oncologia (ICO), 08908 L’Hospitalet de Llobregat, Spain
12
Hospital Reina Sofía, 14004 Córdoba, Spain
13
Hospital Universitario Virgen de Arrixaca, 30120 Murcia, Spain
14
Hospital Marques de Valdecilla, IDIVAL, 39008 Santander, Spain
15
Hospital Virgen del Rocío, 41013 Sevilla, Spain
16
Hospital Asturias, 33011 Oviedo, Spain
17
Hospital Ramón y Cajal, 28034 Madrid, Spain
18
Complejo Hospital Juan Canalejo, 15006 Coruña, Spain
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(11), 2673; https://doi.org/10.3390/cancers14112673
Submission received: 17 April 2022 / Revised: 16 May 2022 / Accepted: 25 May 2022 / Published: 27 May 2022
(This article belongs to the Special Issue Autologous and Allogeneic Stem Cell Transplant in Cancer Therapy)

Abstract

:

Simple Summary

We present the long-term results of patients receiving allogeneic stem cell transplantation (allo-SCT) for relapsed/refractory mantle cell lymphoma (R/R MCL) in the last 25 years in Spain. We conclude that allo-SCT may be a curative option in R/R MCL with a low cumulative incidence (CI) of relapse, although non-relapse mortality (NRM) is still high, which is mainly secondary to acute graft-versus-host disease (aGVHD). Results are better for fit patients, using HLA-identical (related or unrelated) or haploidentical related donors and without previous ASCT. However, the arrival of new highly effective and low toxic immunotherapeutic or targeted therapies inevitably will relegate allo-SCT to those fit patients who fail these therapies, being administered far away from the optimal timing.

Abstract

Allo-SCT is a curative option for selected patients with relapsed/refractory (R/R) MCL, but with significant NRM. We present the long-term results of patients receiving allo-SCT in Spain from March 1995 to February 2020. The primary endpoints were EFS, OS, and cumulative incidence (CI) of NRM, relapse, and GVHD. We included 135 patients, most (85%) receiving RIC. After a median follow-up of 68 months, 5-year EFS and OS were 47 and 50%, respectively. Overall and CR rates were 86 and 80%. The CI of relapse at 1 and 3 years were 7 and 12%. NRM at day 100 and 1 year were 17 and 32%. Previous ASCT and Grade 3–4 aGVHD were associated with a higher NRM. Grade 3–4 aGVHD, donor type (mismatch non-related), and the time-period 2006–2020 were independently related to worse EFS. Patients from 1995–2005 were younger, most from HLA-identical sibling donors, and were pretreated less. Our data confirmed that allo-SCT may be a curative option in R/R MCL with low a CI of relapse, although NRM is still high, being mainly secondary to aGVHD. The arrival of new, highly effective and low toxic immunotherapeutic or targeted therapies inevitably will relegate allo-SCT to those fit patients who fail these therapies, far away from the optimal timing of treatment.

1. Introduction

Mantle cell lymphoma (MCL) is an uncommon B-cell lymphoma that generally has a poor prognosis, with high rates of chemorefractoriness and an advanced median age at diagnosis [1]. For transplant-eligible patients, clinical outcome improves using intensive cytarabine-based induction chemotherapy, followed by autologous stem cell transplant (ASCT) [2,3] consolidation. Recently, new approaches with anti-CD20 maintenance [4,5], new target drugs [6,7], or new ways of immunotherapy such as CARTs [8] are changing the front and salvage therapeutic lines.
Allogeneic stem cell transplantation (allo-SCT) is a potential curative option for selected patients, mediated by a well-demonstrated graft versus lymphoma (GVL) effect in this lymphoma [9]. However, allo-SCT in MCL has also been associated with significant non-relapse mortality (NRM) [10,11]. The efficacy and toxicity of allo-SCT should be well-balanced, considering that MCL typically affects an older population with potentially higher rates of comorbidities, and there is an increasing number of emerging effective and manageable alternative therapeutic approaches. For these reasons, there is a need to clarify candidates’ selection, and which is the optimal target population for allo-SCT. We should especially consider that most previously published studies that focused on investigating the role of allo-SCT in MCL are retrospective and conclusions are limited by a reduced sample size of patients. Furthermore, this is particularly important with the outstanding efficacy and manageable toxicity associated with new immunotherapies based on CAR-Ts in relapsed/refractory (R/R) MCL [8,12].
Our objective was to analyze the long-term results of MCL patients undergoing allo-SCT in Spain, trying to define better its current role in the era of new immunotherapeutic and targeted therapies, focusing on candidates’ selection.

2. Materials and Methods

2.1. Study Design and Patient Eligibility

We designed a retrospective multicenter study including all registered patients from centers’ members of the Spanish Group of Hematopoietic Transplantation (GETH)/Spanish Group of Lymphoma (GELTAMO) with relapsed or refractory (R/R) MCL consolidated with allo-SCT. For this purpose, all patients who had undergone an allo-SCT in the abovementioned centers from March 1995 to February 2020 that was reported to the EBMT registry were eligible. The primary endpoints were event-free survival (EFS) and overall survival (OS). Secondary outcomes were cumulative incidence (CI) of NRM, relapse, and graft versus host disease (GVHD). The study was performed in compliance with the Declaration of Helsinki and approved by research ethics committees and institutional review boards at each participating institution. As part of the EBMT registration, all patients signed informed consent.

2.2. Data Recovery and Study Definitions

The histological diagnosis was based on a local review, and patients were staged according to the Ann Arbor system. Disease status was assessed by the local team according to the Revised Response Criteria for Malignant Lymphoma [13] and/or Lugano Classification [14]. Myeloablative conditioning was defined as a regimen containing either total body irradiation (TBI) with a dose greater than 6 Gy, a total dose of oral busulfan greater than 8 mg/kg, or a total dose of intravenous busulfan greater than 6.4 mg/kg. All other regimens were defined as reduced-intensity conditioning (RIC) [15]. The diagnosis and grading of acute and chronic graft versus host disease (aGVHD and cGVHD) were performed by the transplant centers using the standard criteria [16].

2.3. Statistical Analysis

All outcome measures were assessed from the time of allo-SCT. OS was defined as the time to death. Event-free survival (EFS) was defined as the time to relapse, progression, or death from any cause. NRM was defined as the time to death without previous disease relapse or progression (considering relapse as a competing event). CI of relapse was defined as the time from relapse or progression (considering death without relapse as a competing event).
Qualitative or binomial variables are expressed as frequencies and percentages. Comparisons between qualitative variables were made using the Fisher Exact Test or the Chi-squared test. Comparisons between quantitative and qualitative variables were performed through non-parametric tests (U of Mann–Whitney or Kruskal–Wallis). The binary logistic regression was used to find out the risk factors associated with day 100 complete response (CR) or NRM. Time to event variables were estimated according to the Kaplan–Meier method and comparisons between variables of interest were performed by the log-rank test. Multivariate analysis with the variables that appeared to be significant in the univariate analysis was carried out according to the Cox proportional hazard regression model (stepwise forward likelihood ratio selection). Those variables not available at transplant were included as time-dependent variables. All p values reported were 2-sided, and statistical significance was defined at p < 0.05. To analyze the impact of time periods on the survival of transplanted patients, we segmented the full range of follow-up (1995–2020) of our series using MAXTAT for disease progression or death. The statistical analysis was performed using SPSS software (SPSS version 28.0; IBM, Chicago, IL, USA) and RStudio (Version 1.3.959; RStudio, PBC, Boston, MA, USA).

3. Results

3.1. Patient and Allo-SCT Characteristics

A total of 135 patients with R/R MCL that fulfilled the inclusion criteria were included in the study. Table 1 summarizes the main patient and allo-SCT information. Briefly, the median age of the study cohort at the time of the allo-SCT was 56 years (32–70), with 27% of patients being older than 60 years. A proportion of 66% of patients had classic, 27% blastic, and 7% indolent MCL. The median time from diagnosis to allo-SCT was 33 months (3–164), and the median number of previous lines of therapy before allo-SCT was two (one to eight), including previous autologous SCT (ASCT) in 49% of patients. Disease status before allo-SCT was as follows: complete response (CR) in 86 patients (64%), partial response (PR) in 35 (26%) patients, and SD/PD in 13 (10%).
Overall, 85% adults underwent RIC allo-SCT, and most patients received grafts from HLA-matched, related and unrelated donors (76%) followed by haploidentical donors (13%), and 9/10 mismatched unrelated donors (10%).

3.2. Main Outcome Data

After a median follow-up of 68 months (2–247), median EFS and OS were 30 (95% CI: 0–72) and 45 (95% CI: 3–86) months, respectively (Figure 1A,B). Eighteen (13%) patients had a progression of lymphoma and 71 (53%) died. Overall and complete response rates (ORR and CR) at day 100 were 84 and 80%, respectively. Most patients (95%) with CR before allo-SCT maintained CR at day 100, while only 2 and 1%, respectively, had a PR or a disease progression. On the other hand, 73 and 50% of patients with previous PR or SD/PD achieved CR at day 100, with only 9 and 17%, respectively, showing progressive disease at day 100. Factors that were significantly associated with CR at day 100 were CR pre-allo-SCT (relative risk (RR) 9.6; 95% CI: 2.9–31.3; p < 0.001) and less than three prior lines (RR 3.1; 95% CI: 1.1–8.9; p = 0.036). The CIs of relapse at 1 and 3 years were 7% (95% CI: 3–12) and 12% (95% CI: 7–18), respectively (Figure 1C).

3.3. GVHD and NRM

Seventy-four (55%) patients developed aGVHD (Grade 1–2: 42 (31%) and Grade 3–4: 32 (24%)) at a median of 31 days post-allo-SCT. The CI of overall acute, acute Grade 2-4, and acute Grade 3-4 GVHD at day 100 were 53% (95% CI: 45–62), 48% (95% CI: 39–57), and 29% (95% CI: 62–80), respectively. Forty-eight (36%) patients developed cGVHD (13% mild, 11% moderate, 10% severe). The CIs of overall chronic and chronic moderate/severe GVHD at 3 years were 57% (CI 95%: 45–68) and 42% (CI 95%: 29–55), respectively.
The CIs of day 100 and one-year NRM were 17% (95% CI: 11–24) and 32% (95% CI: 24–40), respectively (Figure 1D). When analyzing all pretransplant factors included in Table 1, as well as aGVHD incidence, to assess their contribution to NRM at day 100, we observed that patients having a previous ASCT (RR 3; 95% CI: 1.1–8.1; p = 0.03) and those who developed Grade 3–4 aGVHD (RR 5.4; 95% CI: 2.1–13.7; p < 0.001) were significantly and independently related to a higher NRM. In contrast, the NRM was not influenced by age, ECOG PS, conditioning regimen, or donor type (Table 2).

3.4. Survival Analysis

One and 5-year-EFS were 61% (95% CI: 57–65) and 47% (95% CI: 39–56), respectively. On the other hand, one and 5-year-OS were 63% (95% CI: 59–67) and 50% (95% CI: 41–59), respectively. Univariate analysis showed that EFS and OS were influenced by the type of donor (mismatch), ECOG PS at allo-SCT, pre-transplant response, GVHD prophylaxis, response to allo-SCT, aGVHD, cGVHD, and time-period (Table 3). In the multivariate analysis, three variables showed an independent prognostic value for a worse EFS: grade 3–4 aGVHD, donor type (mismatch non-related), and time period 2006–2020.
The main causes of death were associated with NRM: 32 due to GVHD (45%), most of them were grade 3–4, 15 to infections (21%), 2 sinusoidal obstruction syndromes (SOS) (3%), 2 thrombotic microangiopathy (3%) and 8 other causes (11%). Progression of lymphoma was the cause of death in 12 (17%) patients. Most patients suffering NRM were in CR (69%), 8% in PR and 22% died of NRM before response evaluation.

3.5. Impact in Results of Allo-SCT Candidates’ Selection along Decades

We focused on the impact of time periods on the survival of transplanted patients. For this purpose, we segmented the full range of follow-up (1995–2020) of our series using MAXTAT for EFS, obtaining two cutoffs at 15 and 9 years: 2005 and 2011. As shown in Table 3 and Figure 2, the best outcomes in terms of EFS were obtained from 1995 to 2005; with the worst observed from 2006 to 2011. Since 2012, the results slightly improved again but still did not achieve previous levels. However, the 5-year CIs of relapse were similar between all three time periods from 1995 to 2020: 14, 15, and 19% (p = 0.89).
We compared patient characteristics along time periods to discover the causes of these results. As shown in Table 4, patients from 1995 to 2005 were younger, with a much shorter interval from diagnosis to allo-SCT; most of them were from HLA-identical sibling donors and with much less previous therapy. In other words, from 1995 to 2005, patients were much more selected for and transplanted earlier than after 2005. This translated into a lower NRM and a better EFS and OS in these patients (Table 3 and Table 4).

4. Discussion

Our study presents real-world evidence from GETH and GELTAMO Spanish centers about the role of allo-SCT in MCL, confirming its efficacy as a potentially curative option but, at the same time, highlighting its major handicap in terms of potential toxicity and high NRM. However, the most important point is that our series provides a great insight into the importance of candidates’ selection for allo-SCT, which limits its current role in the era of new immunotherapeutic and targeted therapies in MCL.
Most previously published series about the role of allo-SCT in MCL are retrospective and generally small. Our retrospective series with 135 patients compares favorably with most of them in terms of size. It was obtained from all patients reported to the EBMT registry from Spanish centers, with a 5-year EFS and OS of 47 and 50%, respectively. This is in the range between 30–60% of the previously published studies [9,18,19].
All these works and our series provide convincing evidence of the existence of an allogeneic GVL effect, suggesting a curative potential, although this is weaker than in indolent lymphoma [9,18,19,20]. This is also illustrated by the better EFS in patients having cGVHD and the high number of responses in our patients: overall 80% CR, which was higher in patients with previous CR (95%) but also in cases with previous PR (73%) or even SD/PD (50%), demonstrating the graft-versus-MCL effect. Furthermore, the incidence of cGVHD was significantly higher in those patients with less than a CR at pretransplant, who then obtained a CR posttransplant (43%), vs those not achieving a CR (0%) (p = 0.027). Other works have reported that chemorefractoriness is not a major risk factor for disease control in MCL after allo-SCT [21]. In our series, the relapse rate was not a major challenge (only 7 and 12% at 1 and 3 years, respectively). This contrasts with other RIC series in which relapse was reported in up to 40% [18]. However, the high rate of NRM might reduce the number of patients at risk of relapse in our series.
Most previously published studies in allo-SCT in MCL share high rates of toxicity in terms of 10 to more than 50% of NRM, as well as high rates (30–40%) of acute or chronic GVHD. Of course, the higher rates of NRM have been reported to be associated with myeloablative conditioning regimens as well as in more pretreated patients, particularly those failing ASCT [10,11], as was seen in our patients. These high rates of NRM may be lower (below 30%) with a similar efficacy using RIC, as shown in several retrospective [18,19,21,22,23] or even prospective clinical trials [24]. New strategies of GVHD prophylaxis such as cyclophosphamide post-allo-SCT could also improve NRM results in these patients [25,26]. However, only 9% of our patients received this prophylaxis in some of our last haploidentical transplants. In our series, we had 17 and 32% CI of NRM at 100 days and 1 year, respectively, which was independently associated with Grade 3–4 aGVHD incidence and previous ASCT, but not with other transplant characteristics. This may be influenced by the fact that most of our patients received an RIC regimen (85%).
For these reasons, once one demonstrates efficacy, this should be balanced against significant rates of NRM, and is when patient selection and other therapeutic options should be considered. In our series, we observed worse outcome results in terms of EFS and OS in patients with Grade 3–4 aGVHD who were transplanted with unrelated mismatched donors (related mismatched transplants, including haploidentical ones, fared similarly to HLA-identical procedures) and, unexpectedly, in the period 2006 to 2020, this was clearly related to a change in the pattern of candidate selection towards older and more pretreated patients beyond 2006. This is the logical consequence of having much better alternatives to allo-SCT in this population of patients, with also better results.
Nowadays, frontline high-dose cytarabine-containing programs followed by ASCT have been considered the standard of care for young and fit patients [3,4]. When comparing the abovementioned time-periods, in the older cohort (1995–2005), less than half of the patients received an ASCT (19 vs. 56% beyond 2006) as the value of frontline ASCT in MCL was first reported in 2005 [2]. Maintenance with rituximab has been shown to prolong PFS and OS, both in fit or unfit patients [4,5]. New, non-cross resistant chemotherapeutic drugs have shown interesting activity in MCL such as oxaliplatin [27,28,29], bortezomib [30], or bendamustine [31,32,33]. Targeted therapies such as BTK inhibitors [6,34,35] or venetoclax [7,36] obtain impressive results with manageable toxicity, representing good salvage options that may also delay the decision of allo-SCT. Finally, outstanding results have been reported with new immunotherapies with anti-CD19 CAR-T [8,12], which led to the approval of brexucabtagene autoleucel as salvage therapy for R/R MCL patients. Points favoring CAR-T cell therapy are that it is more effective than allo-SCT in patients with active disease, with much lower rates of toxicity and NRM. On the other hand, the follow-up with CAR-T cell therapy is still short when compared with allo-SCT, so it is not known for its long-term curative potential; there is also a relevant economic impact, and limited accessibility. Considering the sequencing of both approaches, if CAR-T cell therapy fails, patients could still receive an allo-SCT but in a more pretreated status that we know would further reduce the efficacy and increase the toxicity of this procedure.
The most important updated guidelines from the American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, European Society for Blood and Marrow Transplantation [37], and National Comprehensive Cancer Network (NCCN) recommend CAR-T cell therapies in MCL from the third line for patients who are intolerant to or relapse after at least one BTKi. NCCN guidelines also recommend considering allo-SCT as consolidation for high-risk, young, and fit responders to a second line. Consequently, if there is an increase in the accessibility of CAR-T cell therapy, we hypothesize that CAR-T cell therapies will probably delay allo-SCT to later lines of therapy, far away from the optimal timing evidenced in our series.
Our work has several limitations. First, it is a retrospective study that implies real world evidence but lacks homogeneity in terms of frontline or salvage therapies, supportive care strategies, and GVHD prophylaxis and management. We do not have some important diagnostic or prognostic information, such as molecular high-risk profiles or TP53 mutation status that could modify therapeutic decisions for allo-SCT candidates [38,39]. Addressing the abovementioned lack of some new strategies for GVHD prophylaxis such as cyclophosphamide post-allo-SCT, we could also mention a few patients receiving ibrutinib before and after allo-SCT, which may provide a benefit in terms of survival [40]. However, in our series, we only had 12 (9%) patients with cyclophosphamide post-allo-SCT prophylaxis and 19 (10%) having received ibrutinib.
Of note, while the older cohort (1995–2005) comprised essentially of identical siblings in contrast with the later cohorts, GVHD incidence was not significantly different and Grade 3–4 aGVHD was only 10% lower in the older cohort. This may illustrate the improvement in GVHD prophylaxis and therapy with the time. At the same time, in our study there were several patients at a high risk for poor outcomes, in which allo-SCT usually is contraindicated: ECOG PS 2–4 (5%) and pretransplant SD/PD (10%). In our series, 5 y-PFS was respectively 0 and 15%, illustrating the potential for disease control of allo-SCT even in selected cases with active disease at transplant, as well as the need of a good ECOG PS to avoid NRM.

5. Conclusions

Taking all this data together, allo-SCT is a feasible and effective therapy in MCL with a well-demonstrated GVL effect favored by cGVHD even in situations of active disease at allo-SCT, but still with high rates of toxicity and NRM. As concluded from our time-period analysis, allo-SCT may be a better approach for young, fit, high-risk patients consolidated early (i.e., second line) that probably is linked to lower rates of severe aGVHD and NRM. Improved outcomes may be obtained using HLA-identical (related or unrelated) or haploidentical related donors, which are better than mismatched, unrelated donors. However, the arrival of new highly effective and low toxic immunotherapeutic or targeted therapies inevitably will relegate allo-SCT to fit patients who fail these treatments, which would then be administered far away from the optimal timing.

Author Contributions

Conceptualization and methodology: A.G., L.B. and D.C.; data curation, A.G., L.B., S.N., A.M., G.G., M.Q.S., M.B.-O., A.P., R.H., M.C.V., O.L.-G., J.M., J.L.P., C.F., R.P., C.M., I.E., L.Y., G.R., J.Z., P.H., M.R.V., A.S., C.S. and D.C.; formal analysis and writing-original draft preparation, A.G., L.B., A.M., M.B.-O., C.S. and D.C. All authors contributed to writing-review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was performed in compliance with the Declaration of Helsinki and approved by research ethics committees and institutional review boards at each participating institution. While the inclusion of data in the EBMT registry had a different committee for every institution, this study was approved by Comité de Ética de la Investigación de las Islas Baleares, 25 July 2018. Reference: IB 3746/18 EPA.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We thank Angel Cedillo for his contribution in obtaining the clinical data of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Smith, A.; Crouch, S.; Lax, S.; Li, J.; Painter, D.; Howell, D.; Patmore, R.; Jack, A.; Roman, E. Lymphoma incidence, survival and prevalence 2004–2014: Sub-type analyses from the UK’s Haematological Malignancy Research Network. Br. J. Cancer 2015, 112, 1575–1584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Dreyling, M.; Lenz, G.; Hoster, E.; Van Hoof, A.; Gisselbrecht, C.; Schmits, R.; Metzner, B.; Truemper, L.; Reiser, M.; Steinhauer, H.; et al. Early consolidation by myeloablative radiochemotherapy followed by autologous stem cell transplantation in first remission significantly prolongs progression-free survival in mantle-cell lymphoma: Results of a prospective randomized trial of the European. Blood 2005, 105, 2677–2684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hermine, O.; Hoster, E.; Walewski, J.; Bosly, A.; Stilgenbauer, S.; Thieblemont, C.; Szymczyk, M.; Bouabdallah, R.; Kneba, M.; Hallek, M.; et al. Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL Younger): A randomised, open-label, phase 3 trial of the European Mantle Cell Lymphoma N. Lancet 2016, 388, 565–575. [Google Scholar] [CrossRef]
  4. Le Gouill, S.; Thieblemont, C.; Oberic, L.; Moreau, A.; Bouabdallah, K.; Gyan, E.; Damaj, G.; Ribrag, V.; Feugier, P.; Casasnovas, O.; et al. Rituximab Maintenance after Autologous Stem Cell Transplantation Prolongs Survival in Younger Patients with Mantle Cell Lymphoma: Final Results of the Randomized Phase 3 LyMa Trial of the Lysa/Goelams Group. Blood 2016, 128, 145a. [Google Scholar] [CrossRef]
  5. Kluin-Nelemans, H.C.; Hoster, E.; Hermine, O.; Walewski, J.; Trneny, M.; Geisler, C.H.; Stilgenbauer, S.; Thieblemont, C.; Vehling-Kaiser, U.; Doorduijn, J.K.; et al. Treatment of older patients with mantle-cell lymphoma. N. Engl. J. Med. 2012, 367, 520–531. [Google Scholar] [CrossRef] [Green Version]
  6. Rule, S.; Jurczak, W.; Jerkeman, M.; Rusconi, C.; Trneny, M.; Offner, F.; Caballero, D.; Joao, C.; Witzens-Harig, M.; Hess, G.; et al. Ibrutinib versus temsirolimus: 3-year follow-up of patients with previously treated mantle cell lymphoma from the phase 3, international, randomized, open-label RAY study. Leukemia 2018, 32, 1799–1803. [Google Scholar] [CrossRef] [Green Version]
  7. Eyre, T.A.; Walter, H.S.; Iyengar, S.; Follows, G.; Cross, M.; Fox, C.P.; Hodson, A.; Coats, J.; Narat, S.; Morley, N.; et al. Efficacy of venetoclax monotherapy in patients with relapsed, refractory mantle cell lymphoma after Bruton tyrosine kinase inhibitor therapy. Haematologica 2019, 104, e68–e71. [Google Scholar] [CrossRef] [Green Version]
  8. Wang, M.; Munoz, J.; Goy, A.; Locke, F.L.; Jacobson, C.A.; Hill, B.T.; Timmerman, J.M.; Holmes, H.; Jaglowski, S.; Flinn, I.W.; et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N. Engl. J. Med. 2020, 382, 1331–1342. [Google Scholar] [CrossRef]
  9. Sandoval-Sus, J.D.; Faramand, R.; Chavez, J.; Puri, S.; Parra, P.; Sokol, L.; Kharfan-Dabaja, M.A.; Shah, B.; Ayala, E. Allogeneic hematopoietic cell transplantation is potentially curative in mantle cell lymphoma: Results from a single institution study. Leuk. Lymphoma 2019, 60, 309–316. [Google Scholar] [CrossRef]
  10. Kasamon, Y.L.; Jones, R.J.; Diehl, L.F.; Nayer, H.; Borowitz, M.J.; Garrett-Mayer, E.; Ambinder, R.F.; Abrams, R.A.; Zhang, Z.; Flinn, I.W. Outcomes of autologous and allogeneic blood or marrow transplantation for mantle cell lymphoma. Biol. Blood Marrow Transplant. 2005, 11, 39–46. [Google Scholar] [CrossRef] [Green Version]
  11. Tsai, T.; Goodman, S.; Saez, R.; Schiller, G.; Adkins, D.; Callander, N.; Wolff, S.; Freytes, C.O. Allogeneic bone marrow transplantation in patients who relapse after autologous transplantation. Bone Marrow Transplant. 1997, 20, 859–863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Palomba, M.; Gordon, L.; Siddiqi, T.; Abramson, J.; Kamdar, M.; Lunning, M.; Maloney, D.; Andreadis, C.; Arnason, J.; Ghosh, N.; et al. Safety and Preliminary Efficacy in Patients with Relapsed/Refractory Mantle Cell Lymphoma Receiving Lisocabtagene Maraleucel in Transcend NHL 001. Blood 2020, 136, 10–11. [Google Scholar] [CrossRef]
  13. Cheson, B.D.; Pfistner, B.; Juweid, M.E.; Gascoyne, R.D.; Specht, L.; Horning, S.J.; Coiffier, B.; Fisher, R.I.; Hagenbeek, A.; Zucca, E.; et al. Revised response criteria for malignant lymphoma. J. Clin. Oncol. 2007, 25, 579–586. [Google Scholar] [CrossRef] [PubMed]
  14. Cheson, B.D.; Fisher, R.I.; Barrington, S.F.; Cavalli, F.; Schwartz, L.H.; Zucca, E.; Lister, T.A.; Eastern Cooperative Oncology Group; European Mantle Cell Lymphoma Consortium. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: The Lugano classification. J. Clin. Oncol. 2014, 32, 3059–3068. [Google Scholar] [CrossRef] [PubMed]
  15. Bacigalupo, A.; Ballen, K.; Rizzo, D.; Giralt, S.; Lazarus, H.; Ho, V.; Apperley, J.; Slavin, S.; Pasquini, M.; Sandmaier, B.M.; et al. Defining the intensity of conditioning regimens: Working definitions. Biol. Blood Marrow Transplant. 2009, 15, 1628–1633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Harris, A.C.; Young, R.; Devine, S.; Hogan, W.J.; Ayuk, F.; Bunworasate, U.; Chanswangphuwana, C.; Efebera, Y.A.; Holler, E.; Litzow, M.; et al. International, Multicenter Standardization of Acute Graft-versus-Host Disease Clinical Data Collection: A Report from the Mount Sinai Acute GVHD International Consortium. Biol. Blood Marrow Transplant. 2016, 22, 4–10. [Google Scholar] [CrossRef] [Green Version]
  17. Orchard, J.; Garand, R.; Davis, Z.; Babbage, G.; Sahota, S.; Matutes, E.; Catovsky, D.; Thomas, P.W.; Avet-Loiseau, H.; Oscier, D. A subset of t(11;14) lymphoma with mantle cell features displays mutated IgVH genes and includes patients with good prognosis, nonnodal disease. Blood 2003, 101, 4975–4981. [Google Scholar] [CrossRef]
  18. Robinson, S.P.; Boumendil, A.; Finel, H.; Peggs, K.S.; Chevallier, P.; Sierra, J.; Finke, J.; Poiré, X.; Maillard, N.; Milpied, N.; et al. Long-term outcome analysis of reduced-intensity allogeneic stem cell transplantation in patients with mantle cell lymphoma: A retrospective study from the EBMT Lymphoma Working Party. Bone Marrow Transplant. 2018, 53, 617–624. [Google Scholar] [CrossRef] [Green Version]
  19. Vaughn, J.E.; Sorror, M.L.; Storer, B.E.; Chauncey, T.R.; Pulsipher, M.A.; Maziarz, R.T.; Maris, M.B.; Hari, P.; Laport, G.G.; Franke, G.N.; et al. Long-term sustained disease control in patients with mantle cell lymphoma with or without active disease after treatment with allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. Cancer 2015, 121, 3709–3716. [Google Scholar] [CrossRef]
  20. Cook, G.; Smith, G.M.; Kirkland, K.; Lee, J.; Pearce, R.; Thomson, K.; Morris, E.; Orchard, K.; Rule, S.; Russell, N.; et al. Outcome following Reduced-Intensity Allogeneic Stem Cell Transplantation (RIC AlloSCT) for relapsed and refractory mantle cell lymphoma (MCL): A study of the British Society for Blood and Marrow Transplantation. Biol. Blood Marrow Transplant. 2010, 16, 1419–1427. [Google Scholar] [CrossRef] [Green Version]
  21. Maris, M.B.; Sandmaier, B.M.; Storer, B.E.; Chauncey, T.; Stuart, M.J.; Maziarz, R.T.; Agura, E.; Langston, A.A.; Pulsipher, M.; Storb, R.; et al. Allogeneic hematopoietic cell transplantation after fludarabine and 2 Gy total body irradiation for relapsed and refractory mantle cell lymphoma. Blood 2004, 104, 3535–3542. [Google Scholar] [CrossRef] [PubMed]
  22. Mussetti, A.; Devlin, S.M.; Castro-Malaspina, H.R.; Barker, J.N.; Giralt, S.A.; Zelenetz, A.D.; Sauter, C.S.; Perales, M.-A. Non-myeloablative allogeneic hematopoietic stem cell transplantation for adults with relapsed and refractory mantle cell lymphoma: A single-center analysis in the rituximab era. Bone Marrow Transplant. 2015, 50, 1293–1298. [Google Scholar] [CrossRef] [PubMed]
  23. Tam, C.S.; Bassett, R.; Ledesma, C.; Korbling, M.; Alousi, A.; Hosing, C.; Kebraei, P.; Harrell, R.; Rondon, G.; Giralt, S.A.; et al. Mature results of the M.D. Anderson Cancer Center risk-adapted transplantation strategy in mantle cell lymphoma. Blood 2009, 113, 4144–4152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Krüger, W.H.; Hirt, C.; Basara, N.; Sayer, H.G.; Behre, G.; Fischer, T.; Grobe, N.; Maschmeyer, G.; Niederwieser, D.; Dölken, G. Allogeneic stem cell transplantation for mantle cell lymphoma—Final report from the prospective trials of the East German Study Group Haematology/Oncology (OSHO). Ann. Hematol. 2014, 93, 1587–1597. [Google Scholar] [CrossRef]
  25. Martínez, C.; Gayoso, J.; Canals, C.; Finel, H.; Peggs, K.; Dominietto, A.; Castagna, L.; Afanasyev, B.; Robinson, S.; Blaise, D.; et al. Post-Transplantation Cyclophosphamide-Based Haploidentical Transplantation as Alternative to Matched Sibling or Unrelated Donor Transplantation for Hodgkin Lymphoma: A Registry Study of the Lymphoma Working Party of the European Society for Blood and Marr. J. Clin. Oncol. 2017, 35, 3425–3432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sanz, J.; Galimard, J.-E.; Labopin, M.; Afanasyev, B.; Angelucci, E.; Ciceri, F.; Blaise, D.; Cornelissen, J.J.; Meijer, E.; Diez-Martin, J.L.; et al. Post-transplant cyclophosphamide after matched sibling, unrelated and haploidentical donor transplants in patients with acute myeloid leukemia: A comparative study of the ALWP EBMT. J. Hematol. Oncol. 2020, 13, 46. [Google Scholar] [CrossRef]
  27. Rodríguez, J.; Gutierrez, A.; Palacios, A.; Navarrete, M.; Blancas, I.; Alarcón, J.; Caballero, M.D.; De Mattos, S.F.; Gines, J.; Martínez, J.; et al. Rituximab, gemcitabine and oxaliplatin: An effective regimen in patients with refractory and relapsing mantle cell lymphoma. Leuk. Lymphoma 2007, 48, 2172–2178. [Google Scholar] [CrossRef]
  28. Obrador-Hevia, A.; Serra-Sitjar, M.; Rodríguez, J.; Belayachi, L.; Bento, L.; García-Recio, M.; Sánchez, J.M.; Villalonga, P.; Gutiérrez, A.; Fernández de Mattos, S. Efficacy of the GemOx-R regimen leads to the identification of Oxaliplatin as a highly effective drug against Mantle Cell Lymphoma. Br. J. Haematol. 2016, 174, 899–910. [Google Scholar] [CrossRef] [Green Version]
  29. Tessoulin, B.; Chiron, D.; Thieblemont, C.; Oberic, L.; Bouadballah, K.; Gyan, E.; Damaj, G.; Ribrag, V.; Gressin, R.; Feugier, P.; et al. Oxaliplatin before autologous transplantation in combination with high-dose cytarabine and rituximab provides longer disease control than cisplatin or carboplatin in patients with mantle-cell lymphoma: Results from the LyMA prospective trial. Bone Marrow Transplant. 2021, 56, 1700–1709. [Google Scholar] [CrossRef]
  30. Robak, T.; Huang, H.; Jin, J.; Zhu, J.; Liu, T.; Samoilova, O.; Pylypenko, H.; Verhoef, G.; Siritanaratkul, N.; Osmanov, E.; et al. Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma. N. Engl. J. Med. 2015, 372, 944–953. [Google Scholar] [CrossRef] [Green Version]
  31. Rummel, M.; Kaiser, U.; Balser, C.; Stauch, M.; Brugger, W.; Welslau, M.; Niederle, N.; Losem, C.; Boeck, H.-P.; Weidmann, E.; et al. Bendamustine plus rituximab versus fludarabine plus rituximab for patients with relapsed indolent and mantle-cell lymphomas: A multicentre, randomised, open-label, non-inferiority phase 3 trial. Lancet Oncol. 2016, 17, 57–66. [Google Scholar] [CrossRef]
  32. Flinn, I.W.; van der Jagt, R.; Kahl, B.; Wood, P.; Hawkins, T.; MacDonald, D.; Simpson, D.; Kolibaba, K.; Issa, S.; Chang, J.; et al. First-Line Treatment of Patients With Indolent Non-Hodgkin Lymphoma or Mantle-Cell Lymphoma with Bendamustine Plus Rituximab Versus R-CHOP or R-CVP: Results of the BRIGHT 5-Year Follow-Up Study. J. Clin. Oncol. 2019, 37, 984–991. [Google Scholar] [CrossRef] [PubMed]
  33. Visco, C.; Chiappella, A.; Nassi, L.; Patti, C.; Ferrero, S.; Barbero, D.; Evangelista, A.; Spina, M.; Molinari, A.; Rigacci, L.; et al. Rituximab, bendamustine, and low-dose cytarabine as induction therapy in elderly patients with mantle cell lymphoma: A multicentre, phase 2 trial from Fondazione Italiana Linfomi. Lancet Haematol. 2017, 4, e15–e23. [Google Scholar] [CrossRef] [Green Version]
  34. Song, Y.; Zhou, K.; Zou, D.; Zhou, J.; Hu, J.; Yang, H.; Zhang, H.; Ji, J.; Xu, W.; Jin, J.; et al. Treatment of Patients with Relapsed or Refractory Mantle-Cell Lymphoma with Zanubrutinib, a Selective Inhibitor of Bruton’s Tyrosine Kinase. Clin. Cancer Res. 2020, 26, 4216–4224. [Google Scholar] [CrossRef] [PubMed]
  35. Witzig, T.E.; Inwards, D. Acalabrutinib for mantle cell lymphoma. Blood 2019, 133, 2570–2574. [Google Scholar] [CrossRef]
  36. Tam, C.S.; Anderson, M.A.; Pott, C.; Agarwal, R.; Handunnetti, S.; Hicks, R.J.; Burbury, K.; Turner, G.; Di Iulio, J.; Bressel, M.; et al. Ibrutinib plus Venetoclax for the Treatment of Mantle-Cell Lymphoma. N. Engl. J. Med. 2018, 378, 1211–1223. [Google Scholar] [CrossRef]
  37. Munshi, P.N.; Hamadani, M.; Kumar, A.; Dreger, P.; Friedberg, J.W.; Dreyling, M.; Kahl, B.; Jerkeman, M.; Kharfan-Dabaja, M.A.; Locke, F.L.; et al. American Society of Transplantation and Cellular Therapy, Center of International Blood and Marrow Transplant Research, and European Society for Blood and Marrow Transplantation Clinical Practice Recommendations for Transplantation and Cellular Therapies. Transplant. Cell. Ther. 2021, 27, 720–728. [Google Scholar] [CrossRef]
  38. Lin, R.J.; Ho, C.; Hilden, P.D.; Barker, J.N.; Giralt, S.A.; Hamlin, P.A.; Jakubowski, A.A.; Castro-Malaspina, H.R.; Robinson, K.S.; Papadopoulos, E.B.; et al. Allogeneic haematopoietic cell transplantation impacts on outcomes of mantle cell lymphoma with TP53 alterations. Br. J. Haematol. 2019, 184, 1006–1010. [Google Scholar] [CrossRef]
  39. Lew, T.E.; Cliff, E.R.S.; Dickinson, M.; Tam, C.S.; Seymour, J.F.; Blombery, P.; Bajel, A.; Ritchie, D.; Khot, A. T-cell replete allogeneic stem cell transplant for mantle cell lymphoma achieves durable disease control, including against TP53-mutated disease. Bone Marrow Transplant. 2021, 56, 2857–2859. [Google Scholar] [CrossRef]
  40. Dreger, P.; Michallet, M.; Bosman, P.; Dietrich, S.; Sobh, M.; Boumendil, A.; Nagler, A.; Scheid, C.; Cornelissen, J.; Niederwieser, D.; et al. Ibrutinib for bridging to allogeneic hematopoietic cell transplantation in patients with chronic lymphocytic leukemia or mantle cell lymphoma: A study by the EBMT Chronic Malignancies and Lymphoma Working Parties. Bone Marrow Transplant. 2019, 54, 44–52. [Google Scholar] [CrossRef]
Figure 1. Main outcome data of the patients including event-free survival (A), overall survival (B), CI of relapse (C)and CI of NRM (D).
Figure 1. Main outcome data of the patients including event-free survival (A), overall survival (B), CI of relapse (C)and CI of NRM (D).
Cancers 14 02673 g001
Figure 2. Impact of time-periods in EFS (A) and OS (B) after allo-SCT.
Figure 2. Impact of time-periods in EFS (A) and OS (B) after allo-SCT.
Cancers 14 02673 g002
Table 1. Main characteristics of patients at diagnostic and before allo-SCT.
Table 1. Main characteristics of patients at diagnostic and before allo-SCT.
Characteristics at DiagnosisN (%)Missing Data (%)
Median age, years (range)52 (31–67)0 (0%)
Sex (M/F)108 (80%)/27 (20%)0 (0%)
Ann Arbor stage: 2 (1%)
I–II8 (6%)-
III–IV125 (94%)-
B-symptoms:44 (38%)20 (15%)
CNS involvement:2 (2%)7 (5%)
Bone marrow involvement:98 (75%)4 (3%)
Number of extranodal sites: 7 (5%)
0–181 (63%)-
>147 (37%)-
Mantle cell lymphoma histology: 33 (24%)
Indolent [17]7 (7%)-
Classic67 (66%)-
Blastic28 (27%)-
Characteristics at allo-SCT -
Median previous lines of therapy (range)2 (1–8)2 (1%)
Previous ASCT66 (49%)0 (0%)
Previous ibrutinib19 (14%)2 (1%)
Median months from diagnosis to allo-SCT (range)33 (3–164)0 (0%)
Median age, years (range)56 (32–70)0 (0%)
Age >60 years36 (27%)0 (0%)
Donor type: 0 (0%)
HLA-id sibling74 (55%)-
HLA-id unrelated29 (21%)-
Haploidentical18 (13%)-
Mismatch unrelated14 (10%)-
Median donor age (range)46 (19–72)2 (1%)
Donor sex: 0 (0%)
Male82 (61%)-
Female53 (39%)-
ECOG PS: 12 (9%)
074 (60%)-
143 (35%)-
2–46 (5%)-
HCT-CI: 11 (8%)
0–170 (56%)-
228 (23%)-
≥326 (21%)-
Disease status: 1 (1%)
CR86 (64%)-
PR35 (26%)-
SD/PD13 (10%)-
NE1 (1%)-
Conditioning regimen: 0 (0%)
Myeloablative20 (15%)-
RIC115 (85%)-
Myeloablative conditioning: 0 (0%)
Cy + TBI10 (50%)-
FLUBU5 (25%)-
FLUMEL1 (5%)-
BEAM1 (5%)-
Other3 (15%)-
RIC: 0 (0%)
FLUMEL90 (78%)-
FLUBU15 (13%)-
Cy + TBI4 (4%)-
Other6 (5%)-
GVHD prophylaxis: 0 (0%)
CsA/Tacro-MTX45 (33%)-
CsA /Tacro-MMF31 (23%)-
Timoglobulin based prophylaxis24 (18%)-
Tacro-sirolimus19 (14%)-
Cy-post12 (9%)-
Other4 (3%)-
CMV recipient/donor relation: 2 (1%)
R−/D−12 (9%)-
R−/D+11 (8%)-
R+/D−35 (26%)-
R+/D−75 (56%)-
Stem cell source: 1 (1%)
PB127 (95%)-
BM5 (4%)-
UCB *2 (2%)-
Median CD34 + cells (range) (×106/kg)5.1 (0.1–18.1)5 (4%)
M: male, F: female, CNS: central nervous system, allo-SCT: allogeneic stem cell transplantation, ASCT: autologous stem cell transplantation, HLA-id: HLA identical, ECOG PS: Eastern Cooperative Oncology Group performance status, HCT-CI: hematopoietic cell transplantation-specific comorbidity index, CR: complete response, PR: partial response, SD/PD: stable disease/progression of disease, NE: not evaluable, RIC: reduced-intensity conditioning, GVHD: graft versus host disease, CsA: cyclosporine A, Tacro: tacrolimus, MTX: methotrexate, MMF: Mofetil mycophenolate, Cy: Cyclophosphamide, CMV: cytomegalovirus, R: receptor, D: donor, PB: peripheral blood, BM: bone marrow, UCB: umbilical cord blood. * Included one as haploidentical and the other as mismatch unrelated.
Table 2. Analysis of clinical factors influencing non-relapse mortality.
Table 2. Analysis of clinical factors influencing non-relapse mortality.
CharacteristicsDay100-NRM (%)p
Age at allo-SCT: 1
18–5614 (20%)
>56 years12 (19%)
Months from diagnosis to allo-SCT: 0.51
0–3311 (16%)
>3315 (22%)
Previous lines: 0.65
0–213 (17%)
>212 (21%)
Previous ASCT: 0.008
No7 (10%)
Yes19 (29%)
Donor type: 0.38
HLA-id sibling12 (16%)
HLA-id unrelated 5 (17%)
Mismatch unrelated5 (36%)
Haploidentical4 (22%)
Donor type: 0.2
HLA-id17 (16%)
Mismatch related4 (22%)
Mismatch unrelated5 (36%)
Donor age: 0.38
18–4615 (22%)
>4610 (15%)
Donor sex: 0.82
Male15 (18%)
Female11 (21%)
ECOG PS at allo-SCT 0.31
0–121 (18%)
2–42 (33%)
Pretransplant HCT-CI: 0.58
0–218 (18%)
≥36 (23%)
Conditioning regimen: 0.76
Myeloablative3 (15%)
RIC23 (20%)
GVHD prophylaxis: 0.25
CsA/Tacro-MTX20 (22%)
Other6 (13%)
CMV recipient/donor relation: 0.26
R−/D−1 (8%)
R−/D+2 (18%)
R+/D−4 (11%)
R+/D−19 (25%)
Stem cell source: 0.54
PB24 (19%)
BM1 (20%)
UCB1 (50%)
CD34+ infused cells (range) (×106/kg): 1
0.1–5.113 (20%)
>5.113 (20%)
aGVHD: <0.001
No aGVHD or grade 1–211 (11%)
Grade 3–415 (43%)
NRM: non-relapse mortality, allo-SCT: allogeneic stem cell transplantation, ASCT: autologous stem cell transplantation, HLA-id: HLA identical, ECOG PS: Eastern Cooperative Oncology Group performance status, HCT-CI: hematopoietic cell transplantation-specific comorbidity index, RIC: reduced-intensity conditioning, GVHD: graft versus host disease, CsA: cyclosporine A, Tacro: tacrolimus, MTX: methotrexate, CMV: cytomegalovirus, R: receptor, D: donor, PB: peripheral blood, BM: bone marrow, UCB: umbilical cord blood, aGVHD: acute GVHD.
Table 3. Univariate and multivariate survival analysis.
Table 3. Univariate and multivariate survival analysis.
Univariate Analysis
Characteristics5y-EFS (95% CI)p5y-OS (95% CI)p
Age at allo-SCT: 0.7 0.66
0–6050% (40–60)52% (42–63)
>6039% (21–57)42% (24–60)
Median months from diagnosis to allo-SCT 0.53 0.77
0–3351% (39–63)52% (40–64)
>3344% (31–56)47% (34–60)
Donor type: 0.032 0.2
HLA-id sibling52% (40–64)55% (43–67)
HLA-id non-related50% (31–69)49% (30–68)
Mismatch non-related21% (0–45)35% (7–63)
Haploidentical44% (21–67)42% (17–66)
Mismatch: 0.018 0.044
Yes35% (18–52)39% (20–57)
No51% (41–61)53% (43–63)
ECOG PS at allo-SCT: 0.035 0.021
0–151% (42–60)54% (45–63)
2–40% (NA)0% (NA)
HCT-CI: 0.41 0.42
0–249% (39–60)52% (42–63)
3 or more31% (13–50)35% (16–54)
Previous lines of therapy: 0.11 0.4
1–255% (43–66)55% (43–66)
>237% (24–50)42% (28–56)
Previous ASCT: 0.061 0.02
Yes39% (27–52)39% (25–52)
No55% (43–67)59% (48–71)
Previous ibrutinib: 0.66 0.89
Yes44% (18–69)28% (0–69)
No48% (39–57)50% (41–60)
Conditioning regimen: 0.69 0.99
Myeloablative48% (26–70)48% (25–70)
RIC47% (38–57)50% (40–60)
Response pre-allo-SCT: 0.005 0.004
CR54% (43–65)58% (47–69)
PR40% (23–57)43% (25–60)
SD/PD15% (0–35)15% (0–35)
GVHD prophylaxis: 0.021 0.008
CsA/Tacro-MTX62% (47–76)66% (52–80)
Other40% (30–51)41% (30–52)
Time-period: 0.02 0.023
1995–200569% (51–87)69% (51–87)
2006–202042% (32–52)45% (35–55)
Time-period: 0.023 0.03
1995–200569% (51–87)69% (51–87)
2006–201133% (20–47)38% (24–52)
2012–202049% (37–62)50% (37–64)
Time-dependent variables (univariate)EFSpOSp
HR (95% CI)HR (95% CI)
Grade 3–4 aGVHD5.1 (3.2–8.1)<0.0016 (3.7–9.8)<0.001
Chronic GVHD1 (0.5–2)0.971 (0.5–2)0.92
Multivariate analysis
Grade 3–4 aGVHD:7.6 (4.5–12.8)<0.0018.9 (5.1–15.3)<0.001
Mismatch non-related:3 (1.5–6.2)0.002--
Time-period 2006–2020:2.7 (1.1–6.4)0.0233.2 (1.3–8)0.014
EFS: event-free survival, OS: overall survival, allo-SCT: allogeneic stem cell transplantation, HLA-id: HLA identical, ECOG PS: Eastern Cooperative Oncology Group performance status, HCT-CI: hematopoietic cell transplantation-specific comorbidity index, ASCT: autologous stem cell transplantation, RIC: reduced-intensity conditioning, CR: complete response, PR: partial response, SD/PD: stable disease/progression of disease, GVHD: graft versus host disease, CsA: cyclosporine A, Tacro: tacrolimus, MTX: methotrexate, aGVHD: acute GVHD.
Table 4. Analysis of patient characteristics according to time periods.
Table 4. Analysis of patient characteristics according to time periods.
CharacteristicsGlobal Series (N = 135)2006–20201995–2005p
(N = 109)(N = 26)
Median age at allo-SCT, years (range)56 (32–70)57 (32–70)52 (34–68)0.047
Median months diagnosis to allo-SCT (range)33 (3–164)39 (3–164)13 (5–84)0.001
Frontline therapy cytarabine-based93 (70%)76 (70%)17 (68%)0.81
More than 33 months from diagnosis to allo-SCT67 (50%)46 (42%)5 (19%)<0.001
Donor type: <0.001
HLA-id sibling74 (55%)50 (46%)24 (92%)
HLA-id non-related29 (21%)28 (26%)1 (4%)
Haploidentical18 (13%)18 (16%)0 (0%)
Mismatch non-related14 (10%)13 (12%)1 (4%)
ECOG PS at allo-SCT: 1
0–1117 (95%)98 (95%)19 (95%)
2–46 (5%)5 (5%)1 (5%)
HCT-CI at allo-SCT: 0.11
0–170 (56%)56 (53%)14 (78%)
228 (23%)25 (24%)3 (17%)
3 or more26 (21%)25 (24%)1 (6%)
>2 previous lines:57 (43%)52 (48%)5 (20%)0.013
Previous ASCT:66 (49%)61 (56%)5 (19%)0.001
Response pre-allo-SCT: 0.92
CR86 (64%)70 (64%)16 (61%)
PR35 (26%)28 (26%)7 (27%)
SD/PD13 (10%)101 (9%)3 (11%)
NE1 (1%)1 (1%)
Conditioning: 0.067
Myeloablative20 (15%)13 (12%)7 (27%)
RIC115 (85%)96 (88%)19 (73%)
GVHD prophylaxis: <0.001
CsA/Tacro-MTX45 (33%)81 (74%)9 (35%)
Other90 (67%)28 (26%)17 (65%)
Donor median age (range)46 (19–72)45 (19–72)53 (25–70)0.052
Stem cell source: 0.26
PB127 (95%)104 (96%)23 (88%)
BM5 (4%)3 (3%)2 (8%)
UCB2 (2%)1 (1%)1 (4%)
Median de CD34 + cells (range) (×106/kg)5.1 (0.1–18.1)5.1 (0.1–13)4.3 (2.1–18.1)0.45
aGVHD at + 100:74 (55%)70 (64%)13 (50%)0.5
aGVHD at + 100: 0.37
No aGVHD60 (45%)39 (36%)13 (50%)
1–242 (31%)38 (35%)8 (31%)
3–432 (24%)32 (29%)5 (19%)
Chronic GVHD (%):48 (36%)36 (33%)12 (46%)0.26
Chronic GVHD (%): 0.59
No87 (64%)73 (68%)14 (54%)
Mild17 (13%)14 (13%)3 (12%)
Moderate15 (11%)11 (10%)4 (16%)
Severe14 (10%)10 (9%)4 (16%)
Overall NRM (%): 61 (45%)54 (49%)7 (27%)0.048
NRM at +100 (%):27 (20%)24 (22%)3 (11%)0.28
NRM at 1 year (%)45 (33%)40 (37%)5 (19%)0.11
Causes of death: 0.16
aGVHD32 (45%)30 (49%)2 (20%)
Infection15 (21%)11 (18%)4 (40%)
Lymphoma12 (17%)9 (15%)3 (30%)
Other12 (17%)11 (18%)1 (10%)
Response post-allo-SCT: 0.12
CR110 (81%)88 (81%)22 (85%)
PR6 (4%)6 (5%)0 (0%)
SD/PD6 (4%)4 (4%)2 (8%)
NE13 (10%)11 (10%)2 (8%)
Allo-SCT: allogeneic stem cell transplantation, HLA-id: HLA identical, ECOG PS: Eastern Cooperative Oncology Group performance status, HCT-CI: hematopoietic cell transplantation-specific comorbidity index, ASCT: autologous stem cell transplantation, CR: complete response, PR: partial response, SD/PD: stable disease/progression of disease, NE: not evaluable, RIC: reduced-intensity conditioning, GVHD: graft versus host disease, CsA: cyclosporine A, Tacro: tacrolimus, MTX: methotrexate, PB: peripheral blood, BM: bone marrow, UCB: umbilical cord blood, aGVHD: acute GVHD; NRM: non-relapse mortality.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gutierrez, A.; Bento, L.; Novelli, S.; Martin, A.; Gutierrez, G.; Queralt Salas, M.; Bastos-Oreiro, M.; Perez, A.; Hernani, R.; Cruz Viguria, M.; et al. Allogeneic Stem Cell Transplantation in Mantle Cell Lymphoma; Insights into Its Potential Role in the Era of New Immunotherapeutic and Targeted Therapies: The GETH/GELTAMO Experience. Cancers 2022, 14, 2673. https://doi.org/10.3390/cancers14112673

AMA Style

Gutierrez A, Bento L, Novelli S, Martin A, Gutierrez G, Queralt Salas M, Bastos-Oreiro M, Perez A, Hernani R, Cruz Viguria M, et al. Allogeneic Stem Cell Transplantation in Mantle Cell Lymphoma; Insights into Its Potential Role in the Era of New Immunotherapeutic and Targeted Therapies: The GETH/GELTAMO Experience. Cancers. 2022; 14(11):2673. https://doi.org/10.3390/cancers14112673

Chicago/Turabian Style

Gutierrez, Antonio, Leyre Bento, Silvana Novelli, Alejandro Martin, Gonzalo Gutierrez, Maria Queralt Salas, Mariana Bastos-Oreiro, Ariadna Perez, Rafael Hernani, Maria Cruz Viguria, and et al. 2022. "Allogeneic Stem Cell Transplantation in Mantle Cell Lymphoma; Insights into Its Potential Role in the Era of New Immunotherapeutic and Targeted Therapies: The GETH/GELTAMO Experience" Cancers 14, no. 11: 2673. https://doi.org/10.3390/cancers14112673

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop