Next Article in Journal
Systemic Disease and Ocular Comorbidity Analysis of Geographically Isolated Federally Recognized American Indian Tribes of the Intermountain West
Next Article in Special Issue
Risk Factors and Outcome of C. difficile Infection after Hematopoietic Stem Cell Transplantation
Previous Article in Journal
Clinical Significance of Low-Flow Time in Patients Undergoing Extracorporeal Cardiopulmonary Resuscitation: Results from the RESCUE Registry
Previous Article in Special Issue
Dynamic Changes of Inhibitory Killer-Immunoglobulin-Like Receptors on NK Cells after Allogeneic Hematopoietic Stem Cell Transplantation: An Initial Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Haploidentical Stem Cell Transplantation in Lymphomas—Expectations and Pitfalls

1
Bone Marrow Transplant Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
2
Humanitas Clinical and Research Center, Humanitas University, 20089 Rozzano, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2020, 9(11), 3589; https://doi.org/10.3390/jcm9113589
Submission received: 29 September 2020 / Revised: 23 October 2020 / Accepted: 2 November 2020 / Published: 7 November 2020
(This article belongs to the Special Issue Clinical Outcomes of Stem Cell Transplants in Cancer Treatment)

Abstract

:
T-cell replete Haploidentical stem cell transplantation (Haplo-SCT) with Post-transplant cyclophosphamide (PT-Cy) is an emerging therapeutic option for patients with advanced relapsed or refractory lymphoma. The feasibility of this platform is supported by several retrospective studies showing a toxicity profile that is improved relative to umbilical cord blood and mismatched unrelated donor (UD) transplant and comparable to matched unrelated donor transplant. In particular, cumulative incidence of chronic graft-versus-host disease (GVHD) is reduced after Haplo-SCT relative to UD and matched related donor (MRD) transplant thanks to PT-Cy employed as GVHD prophylaxis. This achievement, together with a similar incidence of acute GVHD and disease relapse, results in a promising advantage of Haplo-SCT in terms of relapse-free/GVHD free survival. Unmet needs of the Haplo-SCT platform are represented by the persistence of a not negligible rate of non-relapse mortality, especially due to infections and disease relapse. Future efforts are warranted in order to reduce life-threatening infections and to employ Halo-SCT with PT-Cy as a platform to build new immunotherapeutic strategies.

1. Introduction

Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) represents a potential curative option for patients with refractory or relapsed lymphomas thanks to the immune-mediated graft-versus-lymphoma (GVL) effect. The existence of a GVL effect is supported first by the evidence of a reduced incidence of relapse for patients with Hodgkin (HL) and non-Hodgkin (NHL) lymphoma after Allo-HSCT (ranging between 6% and 29%) relative to autologous transplant (ranging between 35% and 69%) [1,2,3,4,5]. The GVL effect was more convincingly demonstrated by resolution of residual or progressive disease after Allo-HSCT with withdrawal of immunosuppression or donor lymphocyte infusion (DLI). In particular, disease response has been achieved in about 30–56% of patients with HL [6,7,8], 40–56% of diffuse large B cell (DLBCL) NHL [9,10], 75–90% for follicular NHL (FL) [11]. Unfortunately, the benefits of a GVL effect have been jeopardized by the occurrence of transplant complications resulting in a high rate of non-relapse mortality (NRM).
The development of new drugs for relapsed/refractory lymphomas has challenged the role of allogeneic transplant in this setting. Therapeutic chances comprise monoclonal antibodies [12], check-point inhibitors [13,14] chimeric-antigen-receptor (CAR)-T cells for specific histological subtypes [15,16]. Despite the high overall response rates (ORR) achieved with new drugs, long-term outcome is still a matter of debate: only 9 out of 102 patients were apparently cured at 5-years by brentuximab in relapsed/refractory (R/R) HL [17]; median duration of response was 16.5 months for R/R HL patients treated with pembrolizumab [18] and 2-year progression free survival (PFS) was 39% in the ZUMA-1 trial for patients with DLBCL treated with CAR-T [19]. Due to the short follow-up of these studies and the lack of principle of representing a curative procedure, allo-HSCT still represents the most powerful curative tool for patients with R/R lymphomas, even if the benefits in terms of OS and PFS are hampered by a not negligible risk of NRM. Therefore, choosing the right timing and integrating new therapeutic options with Allo-HSCT is a matter of debate [20].
According to the recommendations of the European Society for Blood and Marrow Transplantation (EBMT) [21], Allo-HSCT from matched related or unrelated donor is the standard of care for lymphomas relapsing after auto-HCT. Unfortunately, HLA-identical donors, both related and unrelated, are not available for all patients and alternative donors (mismatched unrelated donor, haploidentical donor and umbilical cord blood) are searched. In the last ten years, a growing numbers of Allo-HSCT have been performed using haploidentical donor without T-cell depletion. Although several T-cell replete platforms are available with different graft-versus-host-disease (GVHD) prophylaxis, the most frequently used in US and in Europe is represented by high dose post-transplantation cyclophosphamide (PT-Cy), as pioneered by the John Hopkins and Seattle groups [22]. In the pivotal study, using a truly non-myeloablative conditioning (NMAC) regimen, the feasibility and tolerability of this platform was confirmed with low incidence of GVHD and non-relapse mortality (NRM) [22]. This achievement has contributed to the worldwide extension of haploidentical transplant (Haplo-SCT) with PT-Cy, with different levels of conditioning intensity and graft source [23,24]. Given the widespread use of this platform, this review aims at summarizing the current evidences concerning the efficacy and debated issues/unmet needs of the Haplo-SCT with PT–Cy platform (from now on called Haplo-SCT, unless differently specify) in the contest of relapsed/refractory lymphoma.

2. What Is the Role of Haplo-SCT vs. Other Donor Types in Hematological Malignancies?

Two large meta-analysis comparing Haplo-SCT relative to other donors’ transplants for patients with any hematologic malignancy have recently been published and comprise most of the published reports since 2013. In the first one, Meybodi et al. [25] compared the outcomes of 1410 Haplo-SCT with those of 6396 matched related donor (MRD) transplants, while in the second one Gagelmann et al. [26] analyzed a cohort of 22,974 patients receiving either a Haplo-SCT or a MRD or a matched-unrelated (MUD) or a mismatched unrelated (MMUD) donor transplant (Table 1). Main diseases were represented by acute leukemias and lymphomas. Relative to MMUD, Haplo-SCT had a superior outcome in terms of overall survival (OS) (hazard ratio (HR): 0.79, 95% confidential intervals (CI): 0.65–0.97) due to a reduced risk of NRM. This was mainly due to a reduced risk of acute and chronic GVHD (Table 1). Haploidentical and MUD transplants had comparable results in terms of OS (HR: 1.06, 95% CI: 0.96–1.18) but this was the result of opposite effects. On one side, Haplo-SCT had lower incidence of both acute and chronic GVHD resulting in reduced NRM (HR: 0.75, 95% CI: 0.61–0.92) and improved GVHD/relapse free survival (GRFS). On the other side, relapse risk was higher after Haplo-SCT compared to MUD transplants (Table 1). Both studies agreed on the fact that Haplo-SCT had an inferior OS (HR. 1.17–1.18) compared with MRD transplant, even if the difference was statistically significant only in one report. Worse outcome with Haplo-SCT was mainly related to a higher risk of NRM (HR: 1.20–1.36), probably due to increased risk of infections and delayed immune reconstitution according to Meybodi et al. [25] or to an increased risk of grade II-IV but not III-IV, acute GVHD (aGVHD) in the report by Gagelman et al. [26]. Notably, GRFS did not differ between the two donor types due to the fact that the Haplo-SCT platform was endowed by a 50% lower risk of chronic GVHD (cGVHD) with a similar relapse incidence. Put together, these two large metanalysis suggest that Haplo-SCT represent a favorable option compared with MMUD, has a better toxicity profile relative to MUD but has still a worse outcome compared with MRD transplants.

3. What Is the Role of Haplo-SCT vs. Other Donor Types in Lymphomas?

There are three main retrospective analyses focusing only on patients with lymphomas that were published by the Center for International Blood and Marrow Transplant Research (CIBMTR) [27,28,29]. In the first seminal report, Kanate et al. [27] compared the outcomes of 185 patients undergoing Haplo-SCT with those of 732 recipients of unrelated donor (UD) transplant with or without antithymocyte globulin (ATG). Haplo-SCT had similar results in terms of OS, progression-free survival (PFS) and NRM relative to UD transplants (Table 1) but was surprisingly more advantageous relative to UD both in terms of grade III-IV acute and chronic GVHD (HR: 0.17–0.27). Unfortunately, GRFS was not analyzed in this report. Gosh et al. [28] documented similar outcomes between Haplo (n = 180) and MRD (n = 807) transplants in terms of OS and PFS, while Haplo-SCT was hampered by significantly higher risk of NRM and aGVHD, as reported also in the metanalysis by Meybody and Gagelmann [25,26]. Consistent with Kanate et al. [27], Haplo-SCT was endowed by a strikingly lower incidence of cGVHD compared with MRD transplants (HR: 0.06; 1-year cGVHD: 12% vs. 45%, p < 0.001) but again data on a survival endpoint such as GRFS was unfortunately not available. More recently, Fatobene et al. [29] compared the outcome of 526 Haplo-SCTs with 214 umbilical cord blood (UCB) transplants (approximately 60–70% NHL) and found a significant advantage of Haplo-SCT in terms of OS and PFS due to a reduced NRM rate (Table 1). Moreover, the incidence of grade II-IV aGVHD (20% vs. 40%, p < 0.001) and cGVHD (11% vs. 17%, p = 0.04) were lower after Haplo-SCT relative to UCB transplants when the graft source was represented by bone marrow (BM) cells but similar when peripheral blood stem cells (PBSC) were used.
Even with the limits of retrospective registry-based studies due to the high heterogeneity in terms of disease subtypes, conditioning regimens and graft sources, all these analysis (Table 1) strongly support that Haplo-SCT with PT-Cy should be considered an acceptable option for patients with lymphoma when a MRD or MUD is not available, as recently confirmed by a position statement of the EBMT [30]. In particular, Haplo-SCT is more advantageous relative to MMUD and UCB transplants, while outcomes are very similar to MUD and MRD transplants. Moreover, given the shorter timing to identify a Haplo donor relative to a MUD, Halpo-SCT may be advantageous when disease control is a major concern. In addition, relative to the search of an UD, the availability of a haploidentical family donor has the potential benefits of lowering costs [31] and providing higher chances of transplants for patients of underrepresented minorities in international registries. Reduced incidence of cGVHD did not influence an increased risk of disease relapse suggesting that the GVT effect after Haplo-SCT is independent of chronic GVHD. Because the lower incidence of cGVHD might be due to the fact that bone marrow (BM) was the predominant graft source in Haplo-SCT, recent studies, either retrospective [32] or prospective [33], have shown that the PT-Cy platform brings similar results in terms of cGVHD and GRFS among different donor types.

4. Results of Haplo-SCT in HL

Three main registry-based studies analyzed the outcomes of HL patients after Haplo-SCT and performed a comparison with other donor sources. Relative to UD transplants, Kanate et al. [27] performed a sub analysis within the aforementioned study and reported similar OS and PFS between the two platforms: 3-year OS was 82% for Haplo-SCT vs. 83–79% after UD with/without ATG and 3-year PFS was 63% vs. 61–60%, respectively. Consistently, Martinez et al. [34] for the Lymphoma working party of the EBMT group and Ahmed et al. [35] for the CIBMTR described similar outcomes between Haplo-SCT and MRD transplants for HL patients in terms of OS and PFS (Table 1). A trend for higher NRM after Haplo-SCT was described in both reports. This was probably due to a higher incidence of grade II-IV aGVHD, at least in the CIBMTR analysis. Results from other single or multi-center studies concerning the Haplo-SCT platform in HL patients are summarized in Table 2. Long-term results of these reports are quite promising: 3-/4-year OS ranges between 54% and 77%, PFS is 38–66%, NRM rate is acceptable ranging between 4% and 26%, that is in line with that reported after MRD or MUD transplants.
The CIMBTR report found that Haplo-SCT was endowed by a significantly lower risk (HR: 0.45) of cGVHD relative to MRD transplants. This strikingly different incidence of cGVHD in HL patients translated in a significant improvement in terms of GRFS or extensive cGVHD/relapse free survival (CRFS). These composite endpoints are very useful to evaluate the long-term efficacy of a transplant procedure since they estimate the frequency of patients free of disease and of immunosuppression, thus reflecting potentially cured subjects experiencing a better quality of life. In details, Martinez et al. [34] found that the 1-y CRFS was 43% after Haplo-SCT, relative to 28% after MRD transplants. Consistently Gauthier et al. [36] for the Francophone Society of Bone Marrow Transplantation and Cellular Therapy described a 3-year GRFS of 37% after Haplo-SCT vs. 15% for MRD transplants (Table 1 and Table 2). Such interesting low rates of cGVHD and promising results in terms of GRFS are confirmed by other single or multi-center non registry based studies summarized in Table 2.
Another important and unexpected observation coming both from the EMBT and the CIBMTR studies is that the risk of HL relapse is significantly reduced after Haplo-SCT relative to MRD transplants (HR: 0-69-0.74, Table 1). This result is confirmed by other single center reports. Burroughs et al. [37] for the Fred Hutchinson Institute described a 2-year relapse incidence (RI) of 40% for haploidentical transplants (Table 2) relative to 56% for MRD and 63% for MUD transplants, resulting into an improved 2-year PFS (51% vs. 29% vs. 23%). Consistently, we have described [38] that 3-year RI was significantly lower after Haplo-SCT (13%, Table 2) relative to HLA identical transplants (62%) and that this was independent of pre-transplant disease status. Taken together, these results suggest a preserved or even stronger immunological activity after haploidentical transplant. Given the importance of cytokines and chemokines interactions at the level of HL microenvironment providing the Reed Stenberg cells with an anti-apoptotic and tumor-escape phenotype [39], we may speculate that the enhanced GVL effect observed after Haplo-SCT may be due to a more potent graft versus microenvironment activity.
To summarize, Haplo-SCT is an effective strategy for HL patients, its efficacy is comparable to MRD and MUD transplants and hold promises to be more advantageous in terms of reduced cGVHD and RI. Due to the lack of prospective clinical trials, a recent EBMT consensus [33] recommended a haploidentical donor transplant within the PT-Cy platform only when a MRD or a MUD is not available or when there is an urgency to find a donor if a MRD is not present. At our center, our policy is to employ a haploidentical first or second degree relative when a MRD is not available, without looking for a MUD. Without a prospective clinical trial, it is impossible to recommend a haploidentical donor over a MRD to reduce the incidence of relapse given the higher NRM rates reported after Haplo-SCT. Because NRM after Haplo-SCT is not related to cGVHD, future efforts are warranted to reduce mortality mainly due to infections or aGVHD.

5. Risk Factors and Unmet Needs of Haplo-SCT for HL

Similar to other donor sources, the most commonly described risk factors affecting the outcome of patients undergoing Haplo-SCT comprise pre-transplant disease status (either CR vs. not in CR or CR/PR vs. active disease), patient performance status (Karnofsky) and hematopoietic stem cell transplantation comorbidity index (HCT-CI) (either 0 vs. >0 or <3 vs. ≥3) (Table 3). In 2013, Raiola et al. [40] (Table 2) reported encouraging outcomes in 25 R/R HL patients receiving BM Haplo-SCT after the Baltimore non-myeloablative conditioning regimen (NMAC) and described pre-transplant disease status as the main variable affecting PFS (Table 3). In 2017, Castagna et al. [41] described the outcome of 62 advanced HL patients receiving a BM graft after either the Baltimore NMAC or a reduced-intensity conditioning (RIC) comprising higher dose of cyclophosphamide and thiotepa (Table 2). The intensification of the conditioning was planned to overcome the relevant rate of lymphoma relapse in the original study by Luznkik et al. [22] with the aim of testing whether stronger conditioning could ameliorate disease control. Results were consistent with those by Raiola [40] both in terms of main outcomes (Table 2) and for identifying pre-transplant disease status as the main predictor not only of PFS but also of OS, NRM and RI (Table 3). The Spanish experience (Table 2) was reported by Gayoso et al. [42], where the original Baltimora NMAC was modified into a RIC regimen with total body irradiation (TBI) replaced by busulfan at a non-myeloablative dose. Again pre-transplant disease status was the main independent prognostic variable (Table 3). In a recent update of the Genoa experience [43] comprising 41 patients, disease status assessed by fluordeoxyglucose- positron emission tomography (FDG-PET) scan (Deauville score ≥ 4) and HCT-CI ≥ 3 were the only factors associated with a worse outcome in terms of RI, PFS and GRFS (Table 3). Moreover, HCT-CI ≥ 3 was associated with increased NRM and lower OS. This study underlines the importance of achieving a deeper level of disease response before transplant to ameliorate post-transplant outcome. New drugs have shown promising results: brentuximab was associated with improved outcome in patients receiving Allo-HSCT [44]; checkpoint inhibitors (CPI), such as nivolumab, resulted in a lower chance of relapse after transplant compared with subjects not treated with CPI before transplant either from our experience (0% vs. 20%) [45] and from the John Hopkins group (PFS 90% vs. 65%) [46]. Employment of CPI was associated with increased risk of GVHD after HLA identical Hallo-SCT [47]. Such increased risk was not confirmed after Haplo-SCT employing PT-Cy for GVHD prophylaxis [45,46] suggesting that this platform is capable of reducing post-transplant alloreactivity without losing GVT response.
Choice of the better graft source, either bone marrow (BM) or peripheral blood stem cells (PBSC) for patients receiving Haplo-SCT is still matter of debate, with retrospective analysis showing either an increased risk of GVHD [48,49] with PBSC or no change [50,51]. In two different reports focused on HL, we have described [41,52] that PBSC may be preferable to BM cells because this graft source is associated with enhanced OS, PFS and GRFS. This effect is probably related to a lower chance of relapse (Table 3) mediated by the higher lymphocyte content of PBSC relative to BM. This potential advantage of PBSC for HL in terms of reduced relapse rate without increasing NRM due GVHD needs to be confirmed in a randomized clinical trial.
Choice of the best haploidentical donor may represent another strategy to improve the outcome of Haplo-SCT. In a recent publication from a multicenter retrospective analysis we have shown that choice of a younger haploidentical donor results in lower chance of aGVHD and reduced NRM, while a sibling should be preferable to a parent donor in terms of relapse incidence and PFS and a father donor is better than the mother both for OS and PFS [53].
The impact of the conditioning regimen on the outcome of transplanted patients with HL has been a matter of debate. Sureda et al. [54], in a prospective clinical trial by GEL/TAMO and EBMT comprising 46 subjects with HL receiving a MRD or MUD transplantation, described a promising 1-year NRM rate of 15%, that is lower than that described after MAC. This finding was consistent with a previous retrospective report by the EBMT [55] in HLs identifying a significantly lower 1-year NRM after RIC (23%) relative to MAC (46%) regimens. On the contrary, a more recent retrospective analysis by the EBMT lymphoma working party [56] analyzed a cohort of 312 HL recipients of MAC or RIC Allo-HSCT and described no significant increase of NRM after MAC, that was indeed advantageous in terms of reduced RI. Ciurea et al. [57] recently analyzed the effect of two variants of RIC regimens on the outcome of 15 refractory HL with active disease at the time of transplant. Indeed, an intensified RIC employing melphalan 140 mg/mq instead of melphalan 100 mg/mq was associated with a lower RI and improved PFS. A recent retrospective analysis by the CIBMTR [58] compared three different RIC regimens (fludarabine/busulfan vs. fludarabine/melphalan 140 mg/mq vs. fludarabine/cyclophosphamide) in 492 HL patients receiving either a MRD or MUD transplant. The authors found that the intensity of the conditioning did not influence disease relapse, NRM or PFS but the combination fludarabina/cyclophosphamide was associated with a higher NRM beyond 11 months from Halo-HSCT. The lack of differences in terms of OS and NRM between a more intense RIC approach with Flu/Mel 140 and the other ones might be a specific feature of HL patients who have a younger median age compared with NHL subjects and may better tolerate more intensive chemotherapy. Within the Haplo-SCT platform, no specific report was focused on comparing different conditioning regimens in HL patients. In a recent collaborative report between our institution and the Paoli-Calmettes Hospital in Marseille [59], we analyzed the outcome of 147 lymphoma patients, of whom 73 with HL received the Baltimora NMAC regimen and found an encouraging low rate of 1-year NRM (12%) and a 2-year OS and PFS of 77% and 65%, respectively.
To summarize, strategies aimed at improving disease control (by enhancing pre-transplant response, using PBSC as graft source, improving conditioning regimen when active disease is present) and reducing transplant related complications such as aGVHD and infections (by choosing the best available donor or a less toxic conditioning regimen and by adopting better strategies to prevent infections and GVHD) are still debated and need to be explore by future randomized clinical trials. Achievement of these targets may result in a significant enhancement of the main outcomes such as OS, PFS and NRM in patients receiving Haplo-SCT.

6. Results of Haplo-SCT in NHL

A few retrospective studies analyzed the outcomes of NHL patients after Haplo-SCT and performed a comparison with other donor sources. Relative to UD transplants, Kanate et al. [27] performed a sub analysis within the aforementioned study and reported similar OS and PFS between the two platforms for the 4 main NHL histologies comprising follicular (FL), diffuse large B cell (DLBCL), mantle cell (MCL) and peripheral T cell (PTCL) lymphomas. In a retrospective analysis by EBMT, Dietrich et al. [60] analyzed 59 NHL patients receiving a Haplo-SCT and compared their outcomes with those of patients transplanted from a MRD (n = 2024) or MUD (n = 437) during the same time interval. Similar to HL retrospective studies, no differences were identified in terms of OS, PFS, NRM and RI (Table 1). Moreover, aGVHD and cGVHD rates were similar among different donor sources with a tendency for a lower frequency of extensive cGVHD after Haplo-SCT compared with other transplant types. Of note, 2-year OS, PFS, RI and NRM after Haplo-SCT were in agreement with those reported for MRD transplants: 56%, 50%, 27% and 23% respectively (Table 4). Garciaz et al. [61] retrospectively analyzed the outcome of 79 NHL patients receiving either Haplo-SCT (n = 26) vs. UD (n = 28) or MRD (n = 25) transplants and described similar rates of 2-year OS (77% vs. 71% vs. 83%), PFS (65% vs. 68% vs. 80%) and GRFS (65% vs. 54% vs. 66%) among the 3 donor sources (Table 4).
Several studies, both single-center and registry based, have focused on the impact of Haplo-SCT with PT-Cy on different NHL histologies summarized in Table 4 [60,61,62,63,64,65,66].
Regarding DLBCL subtype, Kanate et al. [27] showed again similar outcomes between Haplo-SCT and UD transplants in terms of 3-year OS and PFS. In particular, 3-year OS after Haplo-SCT were 58% and 44%, respectively (Table 4). Dreger et al. [62] performed a large retrospective analysis for the CIBMTR comparing the outcomes of 132 Haplo-SCT with 525 MRD and 781 MUD transplant for patients with R/R DLBCL. All patients received a NMA/RIC regimen and most Haplo-SCT recipients had a TBI-based conditioning followed by BM cells as graft source. Consistent with previous retrospective studies on unspecified lymphoma populations, 3-year OS, PFS, NRM and RI, after Haplo-SCT were comparable with those of other donor types and they were 46%, 38%, 22% and 41%, respectively (Table 4). Moreover, Haplo-SCT was associated with a lower incidence of cGVHD and an improved 2-year GRFS compared with other transplant types. Of note, a subset analysis showed that the benefit in terms of GRFS might be related to the more-prevalent usage of BM instead of PBSC in the Haplo-SCT platform. One of the most relevant observations of this study is that Haplo-SCT was not affected by a higher incidence of disease relapse, despite the very modest anti-lymphoma activity of the conditioning regimen employed and the reduced sensitivity of DLBCL to the GVL effect compared with other NHL subtypes. Therefore, the Haplo-SCT platform may have a stronger GVL effect compensating for the reduced activity of the conditioning regimen and despite the lower incidence of cGVHD.
The main report focused on MCL comes from the sub analysis of the CIBMTR where Kanate et al. [27] again reported similar 3-year OS and PFS between Haplo-SCT (Table 4) and UD transplants with or without ATG: 60% vs. 49% vs. 54% and 51% vs. 33% and 41%, respectively.
A few studies have analyzed the outcome of T cell lymphoma patients receiving Haplo-SCT. Kalankry et al. [63] for the Baltimora group. We recently retrospectively analyzed 29 patients with PTCL receiving a Haplo-SCT with PT-Cy at our institution and at the Paoli Calmettes hospital in Marseille between 2010 and 2019 (manuscript accepted) [66] and compared their outcome with that of 20 MRD and 19 UD transplants in the same time period. Two-year OS, PFS and RI for all 69 patients were 70%, 51% and 21% with no differences according to donor type. In particular, Haplo-SCT recipients had a 2-year OS, PFS, GRFS and RI of 76%, 72%, 59%, 21%, respectively. Haplo-SCT was affected by a significantly lower risk of grade II-IV aGVHD compared with MRD and UD transplants (25% vs. 35% vs. 58%) and with a lower risk of moderate/severe cGVHD (10% vs. 26%) relative to UD transplants. Consistently, receiving a UD transplant was associated with the higher risk of NRM.
To summarize, results from studies focused on different NHL histologies confirm the efficacy and feasibility of Haplo-SCT that should be the transplant of choice when a MRD or MUD is not available, in accordance with the recent position statement of the EBMT [30]. Consistent with findings in HL, Haplo-SCT seems to be characterized by a reduced incidence of cGVHD and improved GRFS compared with MRD or MUD transplants.

7. Risk Factors and Unmet Needs of Haplo-SCT in NHL

Risk factors affecting the outcome of NHL patients receiving Haplo-SCT, summarized in Table 5, are similar to the well-known variables identified in the setting of allogeneic transplant from other donor types. The two large studies by Ghosh [28] and Kanate [27] found that both disease related factors, such as pre-transplant disease status (being not in CR), intermediate or high disease-risk index (DRI), histology different from FL, bulky or extranodal disease and patient related factors, such as age, Karnofsky performance status, HCT-CI, were the main independent variables affecting OS, PFS, NRM and RI. These data need to be considered with caution because they originate from a mixed population comprising either haploidentical or MRD or MUD transplants and given the presence of HL and NHL patients, even if NHL represented more than 70% of the subjects. Other reports focused only on patients receiving Haplo-SCT were less powerful to identify risk factors and further analysis based on large number of patients are needed to better explore unmet needs related to this platform. For instance, Kanakry et al. [63] identified timing of remission (first vs. beyond first) and occurrence of GVHD as the only independent predictors of OS and RI, respectively. Results from these studies mainly point out that improving pre-transplant disease control and developing strategies to reduce post-transplant relapse incidence are the most important unmet needs for NHL patients receiving Haplo-SCT.
Better disease control need to be pursued especially for histologies different from FL or for patients at high-risk of relapse. New immunotherapeutic strategies, comprising CAR-T [20], new drug-conjugate antibodies targeting CD19 expressing cells [67] or bispecific T cell engagers [68], represent powerful tools to bring patients with refractory NHL, until now convicted to palliative treatments, to allogeneic transplant. Allogeneic transplant should be reserved to patients not reaching CR with CAR-T, while the other mentioned strategies may be employed as a bridge to transplant given the short median time of response duration.
Role of conditioning regimen for NHL patients is still a matter of debate. In our recent report [59] employing a NMAC regimen for patients with advanced lymphomas, of whom 74 had NHL, resulted in a low 2-year NRM rate (12%) and a promising 2-year OS and PFS of 69% and 65%, respectively. NMAC regimen apparently did not increase the chance of disease relapse (2-year RI: 18%). More recently, Ghosh et al. [69] reported a large retrospective analysis (n = 1823 patients) comparing three different RIC-NMA conditioning regimens ranging from a high (fludarabina/melphalan 140 mg/mq) to intermediate (fludarabina-busulfan 6.4 mg/Kg; fludarabina/cyclophosphamide) to lower (fludarabina/cyclophosphamide with TBI 2 Gy) intensity spectrum. Patients had all NHL and transplants were from a UD, either MRD or MUD. Differently from previous CIBMTR [70,71] studies reporting no differences between MAC or RIC-NMA conditioning regimen for R/R NHL, Ghosh et al. [69] found that the more intense Flu-Mel 140 mg/mq was associated with a higher incidence of cGVHD, resulting in a higher NRM rate (26% vs. 17%, p < 0.001) and lower OS (49% vs. approximately 60%, p < 0.001). This observation is somehow different from a similar comparison in patients with HL [58] and is probably due to the fact that NHL patients are usually older and are less tolerant to more intense conditioning. We recently performed a multicenter prospective phase II clinical trial (manuscript submitted) employing a more intensive RIC regimen, comprising active drugs against lymphoma, such as thiotepa, cyclophosphamide and fludarabina with the aim of reducing RI. We found an interesting low 4-year RI (28%) without any increase in 4-year NRM (15%), resulting in a 4-year OS and PFS of 65% and 54%, respectively (personal observations). While the potential benefits and pitfalls of a myeloablative relative to reduced intensity conditioning regimen before Haplo-SCT have been recently reported [72] in patients with acute leukemia and myelodysplastic syndrome, the role of conditioning regimen intensity in the Haplo-SCT setting needs to be better characterized in lymphomas to address. In particular, we need to address whether it is possible to improve OS by enhancing disease control with more intense conditioning without impacting on NRM or whether it is more advantageous to adopt truly NMA condition to rely on the GVL effect with low incidence of NRM.
Post-transplant strategies comprise prophylactic DLI or usage of new drugs, such as bispecific antibodies or checkpoint blockades that may rescue donor T-cells from tolerance. Up to date the largest published experience on strategies aimed at improving post-transplant disease control for patients with lymphoma concerns DLI [6,7,8,9,10,11,73]. Few studies have been published so fare in the setting of Haplo-SCT either for the treatment of disease relapse or to prevent disease relapse in patients considered at higher risk. Zeidan et al. [74] reported the John Hopkins experience for therapeutic DLI (tDLI) on 40 patients relapsing after Haplo-SCT, of whom 11 had lymphoma. Twelve patients (30%) achieved CR with a median duration of response of 11.8 months. Grade II-IV aGVHD developed in 25% of the subjects and 3 patients had chronic GVHD. The authors suggested that 1 × 10^6 CD3/Kg cells as a safe starting dose. Ghiso et al. [75] reported the Genoa experience on tDLI on 40 patients, of whom 10 with HL where DLI were preceded by 1–3 cycles of chemotherapy. Cumulative incidence of grade II-IV aGVHD was 14% and OS was 52% for the whole population. Results were particularly promising in the HL population with 70% ORR rate (40% CR), 2-year OS of 80% and a low incidence of aGVHD (10%). Cauchois et al. [76] recently reported the experience of our center together with the Paolo-Calmettes hospital of 36 patients, of whom 6 had lymphoma and were receiving prophylactic DLI (pDLI) because they were considered at high risk of relapse. Cumulative incidence of moderate-severe chronic GVHD was 33% with 2 patients dying of cGVHD and one of septic shock in the absence of GVHD resulting into a 9% 1-year NRM. One-year RI was 16% with a promising 1-year PFS of 83% and OS of 76%. Considering the high disease risk of this population, these reported relapse rates and survivals after pDLI are promising, justifying the prospective evaluation of pDLI in further randomized prospective studies. The role of checkpoint inhibitors (CPI) or other immunomodulating strategies is still matter of debate. CPI, such as those harnessing the PD-1/PD-L1 axis, may be highly effective in patients with HL relapsing after allo-HSCT but their effect is jeopardized by significantly increased risk of GVHD-related morbidity and mortality. A recent review of the literature reported aGVHD 14%, cGVHD 9%, ORR rate 54% (33% CR) [47]. Less is known on CPI in disease other than HL. A recent publication [77] retrospectively analyzed the outcome of 21 patients receiving CPI (nivolumab or ipilimumab) for myeloid malignancies or NHL (n = 5). Consistently with HL reports, ORR was interesting (43%) but hampered by a high rate of toxicity due to 48% grade II-IV aGVHD and 29% moderate-severe cGVHD. Interestingly, combination of CPI with DLI resulted in a strikingly higher degree of response (80%). Combinations of DLI with bispecific or trispecific T cell engagers have not been published so far, even if phase I/II trial have been announced in the past [78].
A recent consensus form the acute leukemia working party of EBMT [79] has summarized current evidences on tDLI, pre-emptive and pDLI after Haplo-SCT. They concluded that risk of GvHD after unmanipulated DLI in the Haplo-SCT with PT-Cy platform is comparable to an unmanipulated DLI from an HLA-matched donor and that patients with high-risk myeloid malignancies may benefit from a prophylactic haplo-DLI, which should be used in the setting of a clinical trial. Accordingly, clinical trials are needed to explore the efficacy of pDLI for patients with lymphoma after Haplo-SCT and to evaluate potential combination of DLI with other immunotherapeutic modalities.

8. Conclusions

Haplo-SCT with PT-Cy as GVHD prophylaxis is a promising platform to cure patients with R/R lymphoma when a HLA identical donor, either matched related or matched unrelated, is not available. Moreover, compared with a MUD transplant, Haplo-SCT has important advantages such as better timing to find a donor and increased chances of post-transplant immune-modulation. Improving disease control and reducing NRM are warranted in order to further improve the outcomes of lymphoma patients.

Author Contributions

J.M. wrote the paper, S.B. and A.S. revised the paper, L.C. wrote and revised the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jones, R.J.; Ambinder, R.F.; Piantadosi, S.; Santos, G.W. Evidence of a graft-versus-lymphoma effect associated with allogeneic bone marrow transplantation. Blood 1991, 77, 649–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ratanatharathorn, V.; Uberti, J.; Karanes, C.; Abella, E.; Lum, L.G.; Momin, F.; Cummings, G.; Sensenbrenner, L.L. Prospective comparative trial of autologous versus allogeneic bone marrow transplantation in patients with non-Hodgkin’s lymphoma. Blood 1994, 84, 1050–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Schimmer, A.D.; Jamal, S.; Messner, H.; Keating, A.; Meharchand, J.; Huebsch, L.; Walker, I.; Benger, A.; Gluck, S.; Smith, A. Allogeneic or autologous bone marrow transplantation (BMT) for non-Hodgkin’s lymphoma (NHL): Results of a provincial strategy. Ontario BMT Network, Canada. Bone Marrow Transpl. 2000, 26, 859–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. van Besien, K.; Loberiza, F.R., Jr.; Bajorunaite, R.; Armitage, J.O.; Bashey, A.; Burns, L.J.; Freytes, C.O.; Gibson, J.; Horowitz, M.M.; Inwards, D.J.; et al. Comparison of autologous and allogeneic hematopoietic stem cell transplantation for follicular lymphoma. Blood 2003, 102, 3521–3529. [Google Scholar] [CrossRef]
  5. Peniket, A.J.; Ruiz de Elvira, M.C.; Taghipour, G.; Cordonnier, C.; Gluckman, E.; de Witte, T.; Santini, G.; Blaise, D.; Greinix, H.; Ferrant, A.; et al. European Bone Marrow Transplantation (EBMT) Lymphoma Registry. An EBMT registry matched study of allogeneic stem cell transplants for lymphoma: Allogeneic transplantation is associated with a lower relapse rate but a higher procedure-related mortality rate than autologous transplantation. Bone Marrow Transpl. 2003, 31, 667–678. [Google Scholar]
  6. Peggs, K.S.; Hunter, A.; Chopra, R.; Parker, A.; Mahendra, P.; Milligan, D.; Craddock, C.; Pettengell, R.; Dogan, A.; Thomson, K.J.; et al. Clinical evidence of a graft-versus-Hodgkin’s-lymphoma effect after reduced-intensity allogeneic transplantation. Lancet 2005, 365, 1934–1941. [Google Scholar] [CrossRef]
  7. Anderlini, P.; Swanston, N.; Rashid, A.; Bueso-Ramos, C.; Macapinlac, H.A.; Champlin, R.E. Evidence of a graft-versus-Hodgkin lymphoma effect in the setting of extensive bone marrow involvement. Biol. Blood Marrow Transpl. 2008, 14, 478–480. [Google Scholar] [CrossRef] [Green Version]
  8. Anderlini, P.; Acholonu, S.A.; Okoroji, G.J.; Andersson, B.S.; Couriel, D.R.; De Lima, M.J.; Donato, M.L.; Khouri, I.F.; Giralt, S.A.; Ueno, N.T.; et al. Donor leukocyte infusions in relapsed Hodgkin’s lymphoma following allogeneic stem cell transplantation: CD3+ cell dose, GVHD and disease response. Bone Marrow Transpl. 2004, 34, 511–514. [Google Scholar] [CrossRef] [PubMed]
  9. Bishop, M.R.; Dean, R.M.; Steinberg, S.M.; Odom, J.; Pavletic, S.Z.; Chow, C.; Pittaluga, S.; Sportes, C.; Hardy, N.M.; Gea-Banacloche, J.; et al. Clinical evidence of a graft-versus-lymphoma effect against relapsed diffuse large B-cell lymphoma after allogeneic hematopoietic stem-cell transplantation. Ann. Oncol. 2008, 19, 1935–1940. [Google Scholar] [CrossRef] [PubMed]
  10. Thomson, K.J.; Morris, E.C.; Bloor, A.; Cook, G.; Milligan, D.; Parker, A.; Clark, F.; Yung, L.; Linch, D.C.; Chakraverty, R.; et al. Favorable long-term survival after reduced-intensity allogeneic transplantation for multiple-relapse aggressive non-Hodgkin’s lymphoma. J. Clin. Oncol. 2009, 27, 426–432. [Google Scholar] [CrossRef]
  11. Bloor, A.J.; Thomson, K.; Chowdhry, N.; Verfuerth, S.; Ings, S.J.; Chakraverty, R.; Linch, D.C.; Goldstone, A.H.; Peggs, K.S.; Mackinnon, S. High response rate to donor lymphocyte infusion after allogeneic stem cell transplantation for indolent non-Hodgkin lymphoma. Biol. Blood Marrow Transpl. 2008, 14, 50–58. [Google Scholar]
  12. Moskowitz, C.H.; Walewski, J.; Nademanee, A.; Masszi, T.; Agura, E.; Holowiecki, J.; Abidi, M.H.; Chen, A.I.; Stiff, P.; Viviani, S.; et al. Five-year PFS from the AETHERA trial of brentuximab vedotin for Hodgkin lymphoma at high risk of progression or relapse. Blood 2018, 132, 2639–2642. [Google Scholar] [PubMed] [Green Version]
  13. Chen, R.; Zinzani, P.L.; Fanale, M.A.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molini, D.; Vassilakopoulos, T.P.; et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 2125–2132. [Google Scholar] [PubMed]
  14. Westin, J.R.; Chu, F.; Zhang, M.; Fayad, L.E.; Kwak, L.W.; Fowler, N.; Romaguera, J.; Hagemeister, F.; Fanale, M.; Samaniego, F.; et al. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: A single group, open-label, phase 2 trial. Lancet Oncol. 2014, 15, 69–77. [Google Scholar] [PubMed] [Green Version]
  15. Schuster, S.J.; Svoboda, J.; Chong, E.A.; Nasta, S.D.; Mato, A.R.; Anak, Ö.; Brogdon, J.L.; Pruteanu-Malinici, L.; Bhoj, V.; Landsburg, D.; et al. Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N. Engl. J. Med. 2017, 377, 2545–2554. [Google Scholar] [CrossRef]
  16. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar]
  17. Chen, R.; Gopal, A.K.; Smith, S.E.; Ansell, S.M.; Rosenblatt, J.D.; Savage, K.J.; Connors, J.M.; Engert, A.; Larsen, E.K.; Huebner, D.; et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood 2016, 128, 1562–1566. [Google Scholar]
  18. Chen, R.; Zinzani, P.L.; Lee, H.J.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood 2019, 134, 1144–1153. [Google Scholar]
  19. Locke, F.L.; Ghobadi, A.; Jacobson, C.A.; Miklos, D.B.; Lekakis, L.J.; Oluwole, O.O.; Lin, Y.; Braunschweig, I.; Hill, B.T.; Timmerman, J.M.; et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): A single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019, 20, 31–42. [Google Scholar]
  20. Jain, T.; Bar, M.; Kansagra, A.J.; Chong, E.A.; Hashmi, S.K.; Neelapu, S.S.; Byrne, M.; Jacoby, E.; Lazaryan, A.; Jacobson, C.A.; et al. Use of Chimeric Antigen Receptor T Cell Therapy in Clinical Practice for Relapsed/Refractory Aggressive B Cell Non-Hodgkin Lymphoma: An Expert Panel Opinion from the American Society for Transplantation and Cellular Therapy. Biol. Blood Marrow Transpl. 2019, 25, 2305–2321. [Google Scholar]
  21. Duarte, R.F.; Labopin, M.; Bader, P.; Basak, G.W.; Bonini, C.; Chabannon, C.; Corbacioglu, S.; Dreger, P.; Dufour, C.; European Society for Blood and Marrow Transplantation (EBMT); et al. Indications for haematopoietic stem cell transplantation for haematological diseases, solid tumours and immune disorders: Current practice in Europe, 2019. Bone Marrow Transpl. 2019, 54, 1525–1552. [Google Scholar] [CrossRef]
  22. Luznik, L.; O’Donnell, P.V.; Symons, H.J.; Chen, A.R.; Leffell, M.S.; Zahurak, M.; Gooley, T.A.; Piantadosi, S.; Kaup, M.; Ambinder, R.F.; et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol. Blood Marrow Transpl. 2008, 14, 641–650. [Google Scholar] [CrossRef] [Green Version]
  23. Solomon, S.R.; Solh, M.; Morris, L.E.; Holland, H.K.; Bashey, A. Myeloablative Conditioning with PBSC Grafts for T Cell-Replete Haploidentical Donor Transplantation Using Posttransplant Cyclophosphamide. Adv. Hematol. 2016, 2016, 9736564. [Google Scholar] [CrossRef] [Green Version]
  24. Bacigalupo, A.; Dominietto, A.; Ghiso, A.; Di Grazia, C.; Lamparelli, T.; Gualandi, F.; Bregante, S.; Van Lint, M.T.; Geroldi, S.; Luchetti, S.; et al. Unmanipulated haploidentical bone marrow transplantation and post-transplant cyclophosphamide for hematologic malignanices following a myeloablative conditioning: An update. Bone Marrow Transpl. 2015, 50 (Suppl. 2), S37–S39. [Google Scholar] [CrossRef]
  25. Meybodi, M.A.; Cao, W.; Luznik, L.; Bashey, A.; Zhang, X.; Romee, R.; Saber, W.; Hamadani, M.; Weisdorf, D.J.; Chu, H.; et al. HLA-haploidentical vs. matched-sibling hematopoietic cell transplantation: A systematic review and meta-analysis. Blood Adv. 2019, 3, 2581–2585. [Google Scholar] [CrossRef] [PubMed]
  26. Gagelmann, N.; Bacigalupo, A.; Rambaldi, A.; Hoelzer, D.; Halter, J.; Sanz, J.; Bonifazi, F.; Meijer, E.; Itälä-Remes, M.; Marková, M.; et al. Haploidentical Stem Cell Transplantation with Posttransplant Cyclophosphamide Therapy vs. Other Donor Transplantations in Adults with Hematologic Cancers: A Systematic Review and Meta-analysis. JAMA Oncol. 2019, 5, 1739–1748. [Google Scholar] [CrossRef] [PubMed]
  27. Kanate, A.S.; Mussetti, A.; Kharfan-Dabaja, M.A.; Ahn, K.W.; DiGilio, A.; Beitinjaneh, A.; Chhabra, S.; Fenske, T.S.; Freytes, C.; Gale, R.P.; et al. Reduced-intensity transplantation for lymphomas using haploidentical related donors vs. HLA-matched unrelated donors. Blood 2016, 127, 938–947. [Google Scholar] [CrossRef]
  28. Kanate, A.S.; Mussetti, A.; Kharfan-Dabaja, M.A.; Ahn, K.W.; DiGilio, A.; Beitinjaneh, A.; Chhabra, S.; Fenske, T.S.; Freytes, C.; Gale, R.P.; et al. Reduced-Intensity Transplantation for Lymphomas Using Haploidentical Related Donors Versus HLA-Matched Sibling Donors: A Center for International Blood and Marrow Transpl. Research Analysis. J. Clin. Oncol. 2016, 34, 3141–3149. [Google Scholar]
  29. Fatobene, G.; Rocha, V.; St Martin, A.; Hamadani, M.; Robinson, S.; Bashey, A.; Boumendil, A.; Brunstein, C.; Castagna, L.; Dominietto, A.; et al. Nonmyeloablative Alternative Donor Transplantation for Hodgkin and Non-Hodgkin Lymphoma: From the LWP-EBMT, Eurocord, and CIBMTR. J. Clin. Oncol. 2020, 38, 1518–1526. [Google Scholar] [CrossRef]
  30. Dietrich, S.; Dreger, P.; Hermine, O.; Kyriakou, C.; Montoto, S.; Robinson, S.; Schmitz, N.; Schouten, H.C.; Sureda, A.; Tanase, A. Haploidentical stem cell transplantation for patients with lymphoma: A position statement from the Lymphoma Working Party-European Society for Blood and Marrow Transplantation. Bone Marrow Transpl. 2020, 55, 317–324. [Google Scholar] [CrossRef]
  31. Debals-Gonthier, M.; Siani, C.; Faucher, C.; Touzani, R.; Lemarié-Basset, C.; Chabannon, C.; Furst, S.; Devillier, R.; Harbi, S.; Castagna, L.; et al. Cost-effectiveness analysis of haploidentical vs matched unrelated allogeneic hematopoietic stem cells transplantation in patients older than 55 years. Bone Marrow Transpl. 2018, 53, 1096–1104. [Google Scholar] [CrossRef]
  32. McCurdy, S.R.; Kasamon, Y.L.; Kanakry, C.G.; Bolaños-Meade, J.; Tsai, H.L.; Showel, M.M.; Kanakry, J.A.; Symons, H.J.; Gojo, I.; Smith, B.D.; et al. Comparable composite endpoints after HLA-matched and HLA-haploidentical transplantation with post-transplantation cyclophosphamide. Haematologica 2017, 102, 391–400. [Google Scholar] [CrossRef] [Green Version]
  33. Bolaños-Meade, J.; Reshef, R.; Fraser, R.; Fei, M.; Abhyankar, S.; Al-Kadhimi, Z.; Alousi, A.M.; Antin, J.H.; Arai, S.; Bickett, K.; et al. Three prophylaxis regimens (tacrolimus, mycophenolate mofetil, and cyclophosphamide; tacrolimus, methotrexate, and bortezomib; or tacrolimus, methotrexate, and maraviroc) versus tacrolimus and methotrexate for prevention of graft-versus-host disease with haemopoietic cell transplantation with reduced-intensity conditioning: A randomised phase 2 trial with a non-randomised contemporaneous control group (BMT CTN 1203). Lancet Haematol. 2019, 6, e132–e143. [Google Scholar]
  34. Martínez, C.; Gayoso, J.; Canals, C.; Finel, H.; Peggs, K.; Dominietto, A.; Luca, C.; Boris, A.; Stephen, R.; Didier, B.; 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 Marrow Transplantation. JCO 2017, 35, 3425–3432. [Google Scholar]
  35. Ahmed, S.; Kanakry, J.A.; Ahn, K.W.; Litovich, C.; Abdel-Azim, H.; Aljurf, M.; Bacher, V.U.; Bejanyan, N.; Cohen, J.B.; Farooq, U.; et al. Lower Graft-versus-Host Disease and Relapse Risk in Post-Transplant Cyclophosphamide-Based Haploidentical versus Matched Sibling Donor Reduced-Intensity Conditioning Transplant for Hodgkin Lymphoma. Biol. Blood Marrow Transpl. 2019, 25, 1859–1868. [Google Scholar] [CrossRef] [PubMed]
  36. Gauthier, J.; Poiré, X.; Gac, A.C.; Leclerc, M.; Guillaume, T.; Chalandon, Y.; Nguyen, S.; Forcade, E.; Régny, C.; Bay, J.O.; et al. Better outcome with haploidentical over HLA-matched related donors in patients with Hodgkin’s lymphoma undergoing allogeneic haematopoietic cell transplantation-a study by the Francophone Society of Bone Marrow Transplantation and Cellular Therapy. Bone Marrow Transpl. 2018, 53, 400–409. [Google Scholar] [CrossRef]
  37. Burroughs, L.M.; O’Donnell, P.V.; Sandmaier, B.M.; Storer, B.E.; Luznik, L.; Symons, H.J.; Bruno, B.; Maziarz, R.T.; Pulsipher, M.A.; Petersen, F.B.; et al. Comparison of outcomes of HLA-matched related, unrelated, or HLA-haploidentical related hematopoietic cell transplantation following nonmyeloablative conditioning for relapsed or refractory Hodgkin lymphoma. Biol. Blood Marrow Transpl. 2008, 14, 1279–1287. [Google Scholar] [CrossRef] [Green Version]
  38. Mariotti, J.; Devillier, R.; Bramanti, S.; Sarina, B.; Furst, S.; Granata, A.; Faucher, C.; Harbi, S.; Morabito, L.; Chabannon, C.; et al. Cell-Replete Haploidentical Transplantation with Post-Transplantation Cyclophosphamide for Hodgkin Lymphoma Relapsed after Autologous Transplantation: Reduced Incidence of Relapse and of Chronic Graft-versus-Host Disease Compared with HLA-Identical Related Donors. Biol. Blood Marrow Transpl. 2018, 24, 627–632. [Google Scholar]
  39. Carbone, A.; Gloghini, A.; Castagna, L.; Santoro, A.; Carlo-Stella, C. Primary refractory and early-relapsed Hodgkin’s lymphoma: Strategies for therapeutic targeting based on the tumour microenvironment. J. Pathol. 2015, 237, 4–13. [Google Scholar] [CrossRef] [Green Version]
  40. Raiola, A.; Dominietto, A.; Varaldo, R.; Ghiso, A.; Galaverna, F.; Bramanti, S.; Todisco, E.; Sarina, B.; Giordano, L.; Ibatici, A.; et al. Unmanipulated haploidentical BMT following non-myeloablative conditioning and post-transplantation CY for advanced Hodgkin’s lymphoma. Bone Marrow Transpl. 2014, 49, 190–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Castagna, L.; Bramanti, S.; Devillier, R.; Sarina, B.; Crocchiolo, R.; Furst, S.; El-Cheikh, J.; Granata, A.; Faucher, C.; Harbi, S.; et al. Haploidentical transplantation with post-infusion cyclophosphamide in advanced Hodgkin lymphoma. Bone Marrow Transpl. 2017, 52, 683–688. [Google Scholar] [CrossRef]
  42. Gayoso, J.; Balsalobre, P.; Pascual, M.J.; Castilla-Llorente, C.; López-Corral, L.; Kwon, M.; Serrano, D.; Piñana, J.L.; Herrera, P.; Ferrá, C.; et al. Busulfan-based reduced intensity conditioning regimens for haploidentical transplantation in relapsed/refractory Hodgkin lymphoma: Spanish multicenter experience. Bone Marrow Transpl. 2016, 51, 1307–1312. [Google Scholar] [CrossRef] [Green Version]
  43. Marani, C.; Raiola, A.M.; Morbelli, S.; Dominietto, A.; Ferrarazzo, G.; Avenoso, D.; Giannoni, L.; Varaldo, R.; Gualandi, F.; Grazia, D.; et al. Angelucci EHaploidentical Transplants with Post-Transplant Cyclophosphamide for Relapsed or Refractory Hodgkin Lymphoma: The Role of Comorbidity Index and Pretransplant Positron Emission Tomography. Biol. Blood Marrow Transpl. 2018, 24, 2501–2508. [Google Scholar] [CrossRef] [Green Version]
  44. Chen, R.; Palmer, J.M.; Thomas, S.H.; Tsai, N.C.; Farol, L.; Nademanee, A.; Forman, S.J.; Gopal, A.K. Brentuximab vedotin enables successful reduced-intensity allogeneic hematopoietic cell transplantation in patients with relapsed or refractory Hodgkin lymphoma. Blood 2012, 119, 6379–6381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. De Philippis, C.; Legrand-Izadifar, F.; Bramanti, S.; Giordano, L.; Montes de Oca, C.; Duléry, R.; Bouabdallah, R.; Granata, A.; Devillier, R.; Mariotti, J.; et al. Checkpoint inhibition before haploidentical transplantation with posttransplant cyclophosphamide in Hodgkin lymphoma. Blood Adv. 2020, 4, 1242–1249. [Google Scholar] [CrossRef]
  46. Paul, S.; Zahurak, M.; Luznik, L.; Ambinder, R.F.; Fuchs, E.J.; Bolaños-Meade, J.; Wagner-Johnston, N.; Swinnen, L.J.; Schoch, L.; Varadhan, R.; et al. Gladstone DENon-Myeloablative Allogeneic Transplantation with Post-Transplant Cyclophosphamide after Immune Checkpoint Inhibition for Classic Hodgkin Lymphoma: A Retrospective Cohort Study. Biol. Blood Marrow Transpl. 2020, 26, 1679–1688. [Google Scholar] [CrossRef]
  47. Ijaz, A.; Khan, A.Y.; Malik, S.U.; Faridi, W.; Fraz, M.A.; Usman, M.; Tariq, M.J.; Durer, S.; Durer, C.; Russ, A.; et al. Significant Risk of Graft-versus-Host Disease with Exposure to Checkpoint Inhibitors before and after Allogeneic Transplantation. Biol. Blood Marrow Transpl. 2019, 25, 94–99. [Google Scholar] [CrossRef] [Green Version]
  48. Bashey, A.; Zhang, M.J.; McCurdy, S.R.; St Martin, A.; Argall, T.; Anasetti, C.; Ciurea, S.O.; Fasan, O.; Gaballa, S.; Hamadani, M.; et al. Mobilized Peripheral Blood Stem Cells Versus Unstimulated Bone Marrow As a Graft Source for T-Cell-Replete Haploidentical Donor Transplantation Using Post-Transpl. Cyclophosphamide. J. Clin. Oncol. 2017, 35, 3002–3009. [Google Scholar] [CrossRef]
  49. Ruggeri, A.; Labopin, M.; Bacigalupo, A.; Gülbas, Z.; Koc, Y.; Blaise, D.; Bruno, B.; Irrera, G.; Tischer, J.; Diez-Martin, J.L.; et al. Bone marrow versus mobilized peripheral blood stem cells in haploidentical transplants using posttransplantation cyclophosphamide. Cancer 2018, 124, 1428–1437. [Google Scholar] [CrossRef]
  50. Bradstock, K.; Bilmon, I.; Kwan, J.; Blyth, E.; Micklethwaite, K.; Huang, G.; Deren, S.; Byth, K.; Gottlieb, D. Influence of Stem Cell Source on Outcomes of Allogeneic Reduced-Intensity Conditioning Therapy Transplants Using Haploidentical Related Donors. Biol. Blood Marrow Transpl. 2015, 21, 1641–1645. [Google Scholar] [CrossRef] [Green Version]
  51. Jacque, N.; Lambert, J.; Yakoub-Agha, I.; Tabrizi, R.; Michallet, M.; Deconinck, E.; Thepot, S.; Meunier, M.; De Latour, R.P.; Suarez, F.; et al. Comparison of mobilized peripheral blood stem cells versus unmanipulated bone marrow haploidentical transplantation using post-transplant cyclophosphamide: A retrospective study of SFGM-TC in 176 patients. In Proceedings of the 59th Annual Meeting 2017, abs 2039, Atlanta, GA, USA, 9–12 December 2017. [Google Scholar]
  52. Mariotti, J.; Devillier, R.; Bramanti, S.; Giordano, L.; Sarina, B.; Furst, S.; Granata, A.; Maisano, V.; Pagliardini, T.; De Philippis, C.; et al. Peripheral Blood Stem Cells versus Bone Marrow for T Cell-Replete Haploidentical Transplantation with Post-Transplant Cyclophosphamide in Hodgkin Lymphoma. Biol. Blood Marrow Transpl. 2019, 25, 1810–1817. [Google Scholar] [CrossRef]
  53. Mariotti, J.; Raiola, A.M.; Evangelista, A.; Carella, A.M.; Martino, M.; Patriarca, F.; Risitano, A.; Bramanti, S.; Busca, A.; Giaccone, L.; et al. Impact of donor age and kinship on clinical outcomes after T-cell-replete haploidentical transplantation with PT-Cy. Blood Adv. 2020, 4, 3900–3912. [Google Scholar] [CrossRef]
  54. Sureda, A.; Canals, C.; Arranz, R.; Caballero, D.; Ribera, J.M.; Brune, M.; Passweg, J.; Martino, R.; Valcárcel, D.; Besalduch, J.; et al. Allogeneic stem cell transplantation after reduced intensity conditioning in patients with relapsed or refractory Hodgkin’s lymphoma. Results of the HDR-ALLO study—A prospective clinical trial by the Grupo Español de Linfomas/Trasplante de Médula Osea (GEL/TAMO) and the Lymphoma bWorking Party of the European Group for Blood and Marrow Transplantation. Haematologica 2012, 97, 310–317. [Google Scholar]
  55. Sureda, A.; Robinson, S.; Canals, C.; Carella, A.M.; Boogaerts, M.A.; Caballero, D.; Hunter, A.E.; Kanz, L.; Slavin, S.; Cornelissen, J.J.; et al. Reduced-intensity conditioning compared with conventional allogeneic stem-cell transplantation in relapsed or refractory Hodgkin’s lymphoma: An analysis from the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. J. Clin. Oncol. 2008, 26, 455–462. [Google Scholar] [CrossRef]
  56. Genadieva-Stavrik, S.; Boumendil, A.; Dreger, P.; Peggs, K.; Briones, J.; Corradini, P.; Bacigalupo, A.; Socié, G.; Bonifazi, F.; Finel, H.; et al. Myeloablative versus reduced intensity allogeneic stem cell transplantation for relapsed/refractory Hodgkin’s lymphoma in recent years: A retrospective analysis of the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. Ann. Oncol. 2016, 27, 2251–2257. [Google Scholar] [CrossRef]
  57. Ciurea, S.O.; Kongtim, P.; Srour, S.; Saini, N.; Im, J.; Ramdial, J.; Khouri, I.; Anderlini, P.; Popat, U.; Hosing, C.; et al. Can we cure refractory Hodgkin’s lymphoma with transplantation? Bone Marrow Transpl. 2020. [Google Scholar] [CrossRef] [PubMed]
  58. Ahmed, S.; Ghosh, N.; Ahn, K.W.; Khanal, M.; Litovich, C.; Mussetti, A.; Chhabra, S.; Cairo, M.; Mei, M.; William, B.; et al. Impact of type of reduced-intensity conditioning regimen on the outcomes of allogeneic haematopoietic cell transplantation in classical Hodgkin lymphoma. Br. J. Haematol. 2020. [Google Scholar] [CrossRef]
  59. de Oca, C.M.; Castagna, L.; De Philippis, C.; Bramanti, S.; Schiano, J.M.; Pagliardini, T.; Collignon, A.; Harbi, S.; Mariotti, J.; Granata, A.; et al. Non-myeloablative conditioning regimen before T-cell replete haploidentical transplantation with post-transplant cyclophosphamide for advanced Hodgkin’s and non-Hodgkin’s lymphomas. Biol. Blood Marrow Transpl. 2020, S1083–S8791, 30506–30601. [Google Scholar]
  60. Dietrich, S.; Finel, H.; Martinez, C.; Tischer, J.; Blaise, D.; Chevallier, P.; Castagna, L.; Milpied, N.; Bacigalupo, A.; Corradini, P.; et al. Post-transplant cyclophosphamide-based haplo-identical transplantation as alternative to matched sibling or unrelated donor transplantation for non-Hodgkin lymphoma: A registry study by the European society for blood and marrow transplantation. Leukemia 2016, 30, 2086–2089. [Google Scholar] [CrossRef]
  61. Garciaz, S.; Castagna, L.; Bouabdallah, R.; Fürst, S.; Bramanti, S.; Coso, D.; Crocchiolo, R.; El-Cheikh, J.; Broussais, F.; Chabannon, C.; et al. Familial haploidentical challenging unrelated donor Allo-SCT in advanced non-Hodgkin lymphomas when matched related donor is not available. Bone Marrow Transpl. 2015, 50, 865–867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Dreger, P.; Sureda, A.; Ahn, K.W.; Eapen, M.; Litovich, C.; Finel, H.; Boumendil, A.; Gopal, A.; Herrera, A.F.; Schmid, C.; et al. PTCy-based haploidentical vs matched related or unrelated donor reduced-intensity conditioning transplant for DLBCL. Blood Adv. 2019, 3, 360–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kanakry, J.A.; Kasamon, Y.L.; Gocke, C.D.; Tsai, H.L.; Davis-Sproul, J.; Ghosh, N.; Symons, H.; Bolaños-Meade, J.; Gladstone, D.E.; Swinnen, L.J.; et al. Outcomes of related donor HLA-identical or HLA-haploidentical allogeneic blood or marrow transplantation for peripheral T cell lymphoma. Biol. Blood Marrow Transpl. 2013, 19, 602–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kanakry, J.A.; Gocke, C.D.; Bolaños-Meade, J.; Gladstone, D.E.; Swinnen, L.J.; Blackford, A.L.; Fuchs, E.J.; Huff, C.A.; Borrello, I.; Matsui, W.H.; et al. Phase II Study of Nonmyeloablative Allogeneic Bone Marrow Transplantation for B Cell Lymphoma with Post-Transplantation Rituximab and Donor Selection Based First on Non-HLA Factors. Biol. Blood Marrow Transpl. 2015, 2, 2115–2122. [Google Scholar] [CrossRef] [Green Version]
  65. .Zoellner, A.K.; Fritsch, S.; Prevalsek, D.; Engel, N.; Hubmann, M.; Reibke, R.; Rieger, C.T.; Hellmuth, J.C.; Haas, M.; Mumm, F.; et al. Sequential therapy combining clofarabine and T-cell-replete HLA-haploidentical haematopoietic SCT is feasible and shows efficacy in the treatment of refractory or relapsed aggressive lymphoma. Bone Marrow Transpl. 2015, 50, 679–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Castagna, L.; Pagliardini, T.; Schiano, J.M. Haploidentical stem cell transplantation with post-transplantation cylcophosphamide in peripheral T-cell lymphoma. Bone Marrow Transpl. submitted.
  67. Kahl, B.S.; Hamadani, M.; Radford, J.; Carlo-Stella, C.; Caimi, P.; Reid, E.; Feingold, J.M.; Ardeshna, K.M.; Solh, M.; Heffner, L.T.; et al. A Phase I Study of ADCT-402 (Loncastuximab Tesirine), a Novel Pyrrolobenzodiazepine-Based Antibody-Drug Conjugate, in Relapsed/Refractory B-Cell Non-Hodgkin Lymphoma. Clin Cancer Res. 2019, 25, 6986–6994. [Google Scholar] [CrossRef] [Green Version]
  68. Gaballa, M.R.; Ramos, C.A. Cellular Immunotherapy in Lymphoma: Beyond CART Cells. Curr. Treat Opt. Oncol. 2020, 21, 21. [Google Scholar] [CrossRef]
  69. Ghosh, N.; Ahmed, S.; Ahn, K.W.; Khanal, M.; Litovich, C.; Aljurf, M.; Bacher, V.U.; Bredeson, C.; Epperla, N.; Farhadfar, N.; et al. Association of Reduced-Intensity Conditioning Regimens with Overall Survival among Patients with Non-Hodgkin Lymphoma Undergoing Allogeneic Transplant. JAMA Oncol. 2020, 6, 1011–1018. [Google Scholar] [CrossRef]
  70. Hamadani, M.; Saber, W.; Ahn, K.W.; Carreras, J.; Cairo, M.S.; Fenske, T.S.; Gale, R.P.; Gibson, J.; Hale, G.A.; Hari, P.N.; et al. Impact of pretransplantation conditioning regimens on outcomes of allogeneic transplantation for chemotherapy-unresponsive diffuse large B cell lymphoma and grade III follicular lymphoma. Biol. Blood Marrow Transpl. 2013, 19, 746–753. [Google Scholar] [CrossRef] [Green Version]
  71. Bacher, U.; Klyuchnikov, E.; Le-Rademacher, J.; Carreras, J.; Armand, P.; Bishop, M.R.; Bredeson, C.N.; Cairo, M.S.; Fenske, T.S.; Freytes, C.O.; et al. Lymphoma Working Committee of the CIBMTR Conditioning regimens for allotransplants for diffuse large B-cell lymphoma: Myeloablative or reduced intensity? Blood 2012, 120, 4256–4262. [Google Scholar] [CrossRef] [Green Version]
  72. Solomon, S.R.; St Martin, A.; Shah, N.N.; Fatobene, G.; Al Malki, M.M.; Ballen, K.K.; Bashey, A.; Bejanyan, N.; Bolaños Meade, J.; Brunstein, C.G.; et al. Myeloablative vs reduced intensity T-cell-replete haploidentical transplantation for hematologic malignancy. Blood Adv. 2019, 3, 2836–2844. [Google Scholar]
  73. Castagna, L.; Sarina, B.; Bramanti, S.; Perseghin, P.; Mariotti, J.; Morabito, L. Donor lymphocyte infusion after allogeneic stem cell transplantation. Transfus. Apher. Sci. 2016, 54, 345–355. [Google Scholar] [PubMed]
  74. Zeidan, A.M.; Forde, P.M.; Symons, H.; Chen, A.; Smith, B.D.; Pratz, K.; Carraway, H.; Gladstone, D.E.; Fuchs, E.J.; Luznik, L.; et al. HLA-haploidentical donor lymphocyte infusions for patients with relapsed hematologic malignancies after related HLA-haploidentical bone marrow transplantation. Biol. Blood Marrow Transpl. 2014, 20, 314–318. [Google Scholar]
  75. Ghiso, A.; Raiola, A.M.; Gualandi, F.; Dominietto, A.; Varaldo, R.; Van Lint, M.T.; Bregante, S.; Di Grazia, C.; Lamparelli, T.; Galaverna, F.; et al. DLI after haploidentical BMT with post-transplant CY. Bone Marrow Transpl. 2015, 50, 56–61. [Google Scholar]
  76. Cauchois, R.; Castagna, L.; Pagliardini, T.; Harbi, S.; Calmels, B.; Bramanti, S.; Granata, A.; Lemarie, C.; Maisano, V.; Legrand, F.; et al. Prophylactic donor lymphocyte infusions after haploidentical haematopoietic stem cell transplantation for high risk haematological malignancies: A retrospective bicentric analysis of serial infusions of increasing doses of CD3+ cells. Br. J. Haematol. 2019, 185, 570–573. [Google Scholar]
  77. Holderried, T.A.W.; Fraccaroli, A.; Schumacher, M.; Heine, A.; Brossart, P.; Stelljes, M.; Klobuch, S.; Kröger, N.; Apostolova, P.; Finke, J.; et al. The role of checkpoint blockade after allogeneic stem cell transplantation in diseases other than Hodgkin’s Lymphoma. Bone Marrow Transpl. 2019, 54, 1662–1667. [Google Scholar]
  78. Buhmann, R.; Michael, S.; Juergen, H.; Horst, L.; Peschel, C.; Kolb, H.J. Immunotherapy with FBTA05 (Bi20), a trifunctional bispecific anti-CD3 x anti-CD20 antibody and donor lymphocyte infusion (DLI) in relapsed or refractory B-cell lymphoma after allogeneic stem cell transplantation: Study protocol of an investigator-driven, open-label, non-randomized, uncontrolled, dose-escalating Phase I/II-trial. J. Transl. Med. 2013, 11, 160–169. [Google Scholar]
  79. Dholaria, B.; Savani, B.N.; Labopin, M.; Luznik, L.; Ruggeri, A.; Mielke, S.; Al Malki, M.M.; Kongtim, P.; Fuchs, E.; Huang, X.J.; et al. Clinical applications of donor lymphocyte infusion from an HLA-haploidentical donor: Consensus recommendations from the Acute Leukemia Working Party of the EBMT. Haematologica 2020, 105, 47–58. [Google Scholar]
Table 1. Main studies comparing by multivariable analysis the outcomes of hematologic malignancies receiving a MRD, MUD or CBU transplants vs. Haplo-SCT with PT-Cy.
Table 1. Main studies comparing by multivariable analysis the outcomes of hematologic malignancies receiving a MRD, MUD or CBU transplants vs. Haplo-SCT with PT-Cy.
StudyN° Patients (Haplo-SCT)ComparisonOS
(HR, 95% CI)
PFS or DFS
(HR, 95% CI)
NRM
(HR, 95% CI)
aGVHD
(HR, 95% CI)
cGVHD
(HR, 95% CI)
RI
(HR, 95% CI)
GRFS
(HR, 95% CI)
Meybodi 20197806 (1410)Haplo vs. MRD1.18 (0.92–1.20)1.03 (0.78–1.38)1.36 (1.12–1.66)1.14 (0.82–1.59)0.55 (0.41–0.74)0.88 (0.66–1.18)1.00 (0.50–2.00)
Gagelmann 201922,974 *Haplo vs. MRD1.17 (1.05–1.30)-1.20 (1.04–1.40)1.32 (1.07–1.62)0.46 (0.33–0.62)1.01 (0.86–1.17)0.87 (0.66–1.15)
Gagelmann 201922,974 *Haplo vs. MUD1.06 (0.96–1.18)-0.75 (0.61–0.92)0.76 (0.62–0.93)0.49 (0.34–0.71)1.20 (1.03–1.40)0.69 (0.52–0.93)
Gagelmann 201922,974 *Haplo vs. MMUD0.79 (0.65–0.97)-0.51 (0.25–1.02)0.51 (0.32–0.81)0.74 (0.54–1.03)1.06 (0.77–1.47)0.99 (0.62–1.58)
Kanate 2016917 (185)Haplo vs. UD w/o ATG1.20 (0.90–1.61)1.11 (0.86–1.40)0.97 (0.64–1.47)0.82 (0.57–1.16)0.17 (0.11–0.25)1.25 (0.92–1.69)-
Kanate 2016917 (185)Haplo vs. UD with ATG0.80 (0.59–1.08)0.86 (0.66–1.13)0.65 (0.41–1.02)0.79 (0.52–1.19)0.27 (0.17–0.42)1.05 (0.75–1.49)-
Gosh 2016987 (180)Haplo vs. MRD1.14 (0.87–1.49)0.98 (0.77–1.23)1.52 (0.99–2.34)1.40 (0.99–1.99)0.06 (0.01–0.42)0.80 (0.61–1.04)-
Fatobene 2020740 (526)Haplo BM vs. CBU0.64 (0.49–0.84)0.69 (0.54–0.87)0.52 (0.37–0.62)0.54 (0.40–0.73)0.65 (0.46–0.91)0.90 (0.65–1.25)-
Fatobene 2020740 (526)Haplo PBSC vs. CBU0.62 (0.45–0.86)0.53 (0.40–0.72)0.44 (0.28–0.68)0.96 (0.69–1.31)0.99 (0.67–1.44)0.66 (0.44–0.97)-
Martinez 2017709 (98)Haplo vs. MRD1.24 (0.84–1.82)0.82 (0.61–1.12)1.38 (0.79–2.39)--0.69 (0.48–0.99)-
Ahmed 2019596 (139)Haplo vs. MRD1.07 (0.79–1.45)0.86 (0.68–1.10)1.65 (0.99–2.77)1.73 (1.16–2.59)0.45 (0.32–0.64)0.74 (0.56–0.97)-
Dietrich 20162520 (59)Haplo vs. MRD0.98 (0.64–1.50)0.73 (0.48–1.12)0.86 (0.47–1.57)--1.04 (0.66–1.63)
Gauthier 2018151 (60)Haplo vs. MRD------0.33 (0.19–0.58)
Table legend: Haplo-SCT: haploidentical stem cell transplantation; MRD: matched related donor transplant; MUD: matched unrelated donor transplant; MMUD: mismatched unrelated donor transplant; UD: unrelated donor transplant; CBU: cord blood unit transplant; BM: bone marrow; PBSC: peripheral blood stem cells; HR: hazard ratio; OS: overall survival; PFS: progression free survival; DFS: disease free survival; NRM: non-relapse mortality; aGVHD: grade IIi–IV acute graft-versus-host-disease, cGVHD: chronic graft-versus-host-disease either mild-moderate-severe or moderate-severe according to different publications; RI: relapse incidence; GRFS: GVHD/relapse-free survival; * total number of patients, n° of Haplo-SCT varies according to explored outcome. Bold indicates statistically significant differences between graft sources.
Table 2. Results from main studies employing the Haplo-SCT with PT-Cy platform for patients with R/R HL.
Table 2. Results from main studies employing the Haplo-SCT with PT-Cy platform for patients with R/R HL.
StudyN° PatientsOSPFSNRMRIaGVHDcGVHDGRFS
Burroughs 20089058519404335-
Raiola 2013257763431249-
Kanate 2016466845-----
Gayoso 201643584821243919-
Castagna 201762635920-2316-
Martinez 20179867431739332640
Gauthier 20181517566925281557
Mariotti 2018645444261329347
Ahmed 2019139633811324523-
Marani 20194176447.555211239
Mariotti 2019916758222024752
Table legend: OS: overall survival; PFS: progression free survival; NRM: non-relapse mortality; aGVHD: grade II-IV acute graft-versus-host-disease, cGVHD: chronic graft-versus-host-disease either mild-moderate-severe or moderate-severe according to different publications; RI: relapse incidence; GRFS: GVHD/relapse-free survival.
Table 3. Risk factors affecting the outcome of Halo-SCT with PT-Cy in HL patients.
Table 3. Risk factors affecting the outcome of Halo-SCT with PT-Cy in HL patients.
StudyOSPFSRINRMGRFS
Raiola 2013-Disease Status---
Gayoso 2016Disease StatusDisease StatusDisease Status--
Castagna 2017Disease Status
HCT-CI > 0
PBSC
Disease StatusDisease StatusDisease Status
HCT-CI > 0
-
Martinez 2017Age ≥ 40
KPS
Refractory Dis
Age ≥ 40
KPS
Refractory Dis
KPS
Refractory Dis
Age ≥ 40
PS
Refractory Dis
-
Gauthier 2018--Disease status
PET status
-Disease status
Mariotti 2018Disease StatusDisease StatusDisease StatusRIC/MAC > NMADisease Status
NMA > RIC/MAC
Ahmed 2019Age ≥ 50
PS
Disease status
KPS
Disease status
KPS
Disease status
KPS
HCT-CI ≥ 3
-
Marani 2019HCT-CI ≥ 3HCT-CI ≥ 3
PET (DS ≥ 4)
HCT-CI ≥ 3
PET (DS ≥ 4)
HCT-CI ≥ 3HCT-CI ≥ 3
PET (DS ≥ 4)
Mariotti 2019Disease status
PBSC > BM
HCT-CI ≥ 3
Disease status
PBSC > BM
HCT-CI ≥ 3
Disease status -Disease status
PBSC > BM
HCT-CI ≥ 3
Table legend: OS: overall survival; PFS: progression free survival; NRM: non-relapse mortality; RI: relapse incidence; GRFS: GVHD/relapse-free survival; HCT-CI: hematopoietic cell transplant-comorbidity index; KPS: karnofsky performance status; Dis: disease; PS: performance status; DS: Deuville score; PBSC: peripheral blood stem cell; BM: bone marrow.
Table 4. Results from main studies employing Haplo-SCT with PT-Cy in patients with NHL.
Table 4. Results from main studies employing Haplo-SCT with PT-Cy in patients with NHL.
StudyN° PatientsNHL SubtypeOSPFSNRMRIaGVHDcGVHDGRFS
Kanakry 201344PTCL4340939---
Kanakry 201569B-cell736310204513-
Garciaz 201526Indol/Agg
B o T NHL
77651519-15-
Zoellner 201516Aggressive
B o T NHL
695019363725-
Dietrich 201659Indol/Agg
B o T NHL
56502327---
Kanate 2016129FL7066-----
Kanate 201693DLBCL5844-----
Kanate 201682MCL6051-----
Kanate 201679PTCL3632-----
Dreger 2019132DLBCL46382241341836
Castagna 202029PTCL7672721241059
Table legend: OS: overall survival; PFS: progression free survival; NRM: non-relapse mortality; RI: relapse incidence; aGVHD: grade II-IV acute graft-versus-host-disease, cGVHD: chronic graft-versus-host-disease either mild-moderate-severe or moderate-severe according to different publications; GRFS: GVHD/relapse-free survival; PTCL: peripheral T cell lymphomas, NHL: non-Hodgkin lymphoma, DLBCL. Diffuse large B cell lymphoma; Indol/Agg: indolent and aggressive NHL; FL: follicular lymphoma, MCL: mantle cell lymphoma.
Table 5. Risk factors affecting the outcome of Halo-SCT with PT-Cy in NHL patients.
Table 5. Risk factors affecting the outcome of Halo-SCT with PT-Cy in NHL patients.
StudyOSPFSRINRM
Kanakry 2013First vs. beyond first remission-aGVHD/cGVHD
Kanate 2016Disease status
Age ≥ 60
Not FL
DRI Int/High
Disease status
Age ≥ 60
Not FL
DRI Int/High
Disease status
Not FL
Extranodal Disease
DRI Int/High
Age > 40
KPFS < 90
HCT-CI ≥ 3
Ghosh 2016Disease status
Not FL
DRI Int/High
Bulky Disease
Disease status
Not FL
DRI Int/High
Extranodal/Bulky Disease
Disease status
Not FL
DRI Int/High
Extranodal/Bulky Disease
KPFS < 90
HCT-CI ≥ 3
Dietrich 2016Prior Auto-Tx
Aging
--Prior Auto-Tx
Aging
Dreger 2019Disease status
KPS < 90
HCT-CI ≥ 3
Age ≥ 60
Disease status
KPS < 90
Disease status
KPS < 90
Time from diagnosis ≥ 24 mm
HCT-CI ≥ 3
Age ≥ 60
Castagna 2020Older age---
Table legend: OS: overall survival; PFS: progression free survival; NRM: non-relapse mortality; RI: relapse incidence; HCT-CI: hematopoietic cell transplant-comorbidity index; KPS: karnofsky performance status; FL: follicular lymphoma; DRI: disease risk index; aGVHD/cGVHD: acute and chronic graft-versus-host-disease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mariotti, J.; Bramanti, S.; Santoro, A.; Castagna, L. Haploidentical Stem Cell Transplantation in Lymphomas—Expectations and Pitfalls. J. Clin. Med. 2020, 9, 3589. https://doi.org/10.3390/jcm9113589

AMA Style

Mariotti J, Bramanti S, Santoro A, Castagna L. Haploidentical Stem Cell Transplantation in Lymphomas—Expectations and Pitfalls. Journal of Clinical Medicine. 2020; 9(11):3589. https://doi.org/10.3390/jcm9113589

Chicago/Turabian Style

Mariotti, Jacopo, Stefania Bramanti, Armando Santoro, and Luca Castagna. 2020. "Haploidentical Stem Cell Transplantation in Lymphomas—Expectations and Pitfalls" Journal of Clinical Medicine 9, no. 11: 3589. https://doi.org/10.3390/jcm9113589

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