HEMONC 178 17 June 2017 Hematol Oncol Stem Cell Ther (2017) xxx, xxx– xxx
No. of Pages 8, Model 6+
1
Available at www.sciencedirect.com
ScienceDirect
6 7
journal homepage: www.elsevier.com/locate/hemonc
5
Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances
8
Narendranath Epperla a, Mehdi Hamadani a,b,*
3
4
9 10
11
a b
Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA Center for International Blood and Marrow Transplant Research, Milwaukee, WI 53226, USA
Received 6 January 2017; accepted 28 February 2017
12
20 21 14 22 15 23 16 24 17 25 18 26 19 27 28 29 30 31 32 33 34 35 36 37 38 39 40 45 42 41 43 44
KEYWORDS Allo-HCT; Auto-HCT; Diffuse large B-cell lymphoma; Follicular lymphoma
Abstract Non-Hodgkin’s lymphoma (NHL) constitutes a collection of lymphoproliferative disorders with diverse biologic, histologic, and clinical features. With a better understanding of the molecular pathogenesis, recently there have been major advances in the treatment of NHLs including addition of novel monoclonal antibodies, targeted therapies, and immune activators to the therapy armamentarium. Despite these remarkable developments, autologous hematopoietic cell transplantation (auto-HCT) remains not only a standard-of-care curative option for aggressive NHL but also an important therapeutic option for indolent NHL. In NHL, for patients with high-risk features, including those heavily pretreated or with refractory disease or those experiencing failure after an auto-HCT, allogeneic HCT (allo-HCT) remains the only curative option. In this review, we briefly discuss the role of transplantation in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL). In DLBCL patients, we discuss the role of HCT in clinically and biologically defined ultra-high-risk disease. In FL patients, auto-HCT is best reserved for relapsed chemosensitive patients after two to three lines of prior chemoimmunotherapies, who are not candidates for allo-HCT, either because of donor unavailability, associated comorbidities, or patient preference. Reduced-intensity conditioning allo-HCT offers the curative option for patients with relapsed/refractory FL. Although the emergence of targeted, biologic, and immunological therapies is welcoming, it is currently unclear how these new therapies might enhance or replace allo-HCT. Until we have further definitive data, allo-HCT remains the only curative option. Ó 2017 King Faisal Specialist Hospital & Research Centre. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-ncnd/4.0/).
* Corresponding author at: Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Suite C5500, Milwaukee, WI 53226, USA. E-mail address:
[email protected] (M. Hamadani). http://dx.doi.org/10.1016/j.hemonc.2017.05.004 1658-3876/Ó 2017 King Faisal Specialist Hospital & Research Centre. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
HEMONC 178 17 June 2017
2 46
N. Epperla, M. Hamadani troversial. Several randomized studies evaluating the role of upfront auto-HCT consolidation in DLBCL in the prerituximab era reported conflicting results [18–21]. A meta-analysis of these pre-rituximab era trials, however, did not show the superiority of auto-HCT consolidation compared with the standard chemotherapy alone [22]. Therefore, auto-HCT was not adopted as standard therapy in the frontline setting for high-risk DLBCL in CR1. In the rituximab era, provocative findings of the Southwest Oncology Group S9704 [23] study reignited the discussion of upfront auto-HCT in DLBCL by showing a possible overall survival (OS) benefit in an unplanned/post hoc analysis for the high-risk IPI subgroup. However, DLBCL comprised only 60% of S9704 study population and not all DLBCL patients received rituximab-containing frontline therapies [23]. Of note, two recent randomized trials restricted to IPI defined intermediate–high or high-risk DLBCL, treated uniformly with rituximab-based therapies, did not show any benefit with upfront auto-HCT consolidation [24,25]. Based on these data, upfront auto-HCT in high-risk DLBCL in CR1, as defined by clinical prognostic markers, cannot be considered a standard option. Whether upfront auto-HCT can improve outcomes of biologically defined poor-risk DLBCL (e.g., DHL or DEL) is not known. Limited retrospective data suggest that upfront auto-HCT consolidation in DHL can provide durable disease control [11]. While outcomes of DHL when treated with RCHOP like therapies are poor [14], those achieving CR1 with more intensive frontline therapies can experience durable disease control with or without auto-HCT consolidation [26]. Considering the relative rarity of DHL, a randomized trial evaluating the role of auto-HCT in this setting is unlikely. Upfront auto-HCT should be strongly considered in DHL patients receiving R-CHOP as first-line treatment or those not achieving CR1 with more intensive induction therapies. Large registry studies are urgently needed to further define the role of upfront auto-HCT in DHL and DEL.
100
Auto-HCT in relapsed/refractory DLBCL
137
The landmark PARMA study firmly established auto-HCT consolidation as standard-of-care in relapsed/chemosensitive DLBCL, by demonstrating a survival benefit over salvage chemotherapy alone [27]. The CORAL study, while reaffirming the role of auto-HCT in relapsed DLBCL in the rituximab era, also hinted at the futility of this approach in patients experiencing early failure of rituximab-based therapies [28]. At the Center for International Blood and Marrow Transplant Research (CIBMTR), we evaluated the role of auto-HCT in relapsed DLBCL in the rituximab era, relative to the timing of chemoimmunotherapy failure and noted that DLBCL with early rituximab failure had a higher risk of disease relapse in the 1st year after transplantation. However, beyond that point no survival difference was noted [29]. Thus, auto-HCT remains the standard of care for chemosensitive relapsed/refractory DLBCL.
138
Upfront transplantation (in first complete remission)
Relapsed DLBCL unlikely to benefit from auto-HCT
154
Auto-HCT consolidation in DLBCL patients achieving first complete remission (CR1) after frontline therapies is con-
Prognostic markers able to identify relapsed/refractory DLBCL patients unlikely to benefit from auto-HCT are
155
Introduction
65
Non-Hodgkin’s lymphomas (NHLs) are a heterogeneous group of lymphoproliferative disorders with diverse biologic, histologic, and clinical features. Over the past decade, a better understanding of the molecular pathogenesis has led to major advances in the treatment of NHL, including the addition of monoclonal antibodies and other novel agents such as B-cell receptor signaling inhibitors, immunomodulatory agents, check-point inhibitors, and proteasome inhibitors. Despite these remarkable developments, autologous hematopoietic cell transplantation (auto-HCT) remains not only a standard-of-care option for relapsed aggressive NHL but also an important therapeutic option for indolent NHL. In NHL, for patients with highrisk features, including heavily pretreated or refractory disease or those experiencing failure after an auto-HCT, allogeneic HCT (allo-HCT) remains the only curative option. In this review, we discuss the current role of HCT in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL), relative to the novel prognostic tools available in the clinic.
66
Diffuse large B-cell lymphoma
47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64
94
DLBCL is the most common type of NHL. Like International Prognostic Index (IPI) [1], newer clinical prognostic models, such as Revised-IPI [2] or NCCN-IPI [3], can risk-stratify patients based on simple clinical parameters. In addition to these clinical prognostic models, a greater understanding of lymphomagenesis in DLBCL has allowed us to predict patient-specific outcomes based on biological predictors such as cell-of-origin (defined by gene expression profiling or immunohistochemistry) [4,5], and translocation and/or overexpression of the MYC and BCL-2 or BCL-6 oncogenes [6], potentially ushering in the era of precision medicine in NHL treatment. Rearrangement of the MYC proto-oncogene is seen in 12– 15% of DLBCL [7,8]. The concurrent rearrangement of MYC plus BCL2 and/or BCL6 genes leads to a clinically resistant form of lymphoma termed double-hit lymphoma (DHL). DHL comprises 5–7% of newly diagnosed DLBCL and carries a dismal prognosis with standard therapies [9–11]. The recently described double-expresser lymphoma (DEL) is DLBCL with overexpression of both the MYC and BCL2 (cutoff of 40% and >50%, respectively) proteins by immunohistochemistry and accounts for 21–34% of patients newly diagnosed with DLBCL [12,13]. Similar to DHL, patients with DEL also have poor outcomes after R-CHOP therapy, independent of DHL or cell-oforigin status [9,14–16]. DLBCL cases with dual DHL/DEL have grim prognosis [17]. The challenge in the year 2017 and beyond is to incorporate these biological prognostic markers in the decision-making algorithms of HCT.
95
Role of auto-HCT in DLBCL
67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93
96 97
98 99
No. of Pages 8, Model 6+
Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136
139 140 141 142 143 144 145 146 147 148 149 150 151 152 153
156
HEMONC 178 17 June 2017
No. of Pages 8, Model 6+
HCT in DLBCL and FL
177
urgently needed to investigate alternative therapies. In the recently reported REgistry of diFfuse large B-cell lymphoma with prImary treatmeNt failurE (REFINE) analysis, the presence of at least two out of three risk factors—(a) primary progressive disease, (b) MYC rearrangement, and (c) intermediate-high/high NCCN-IPI—in relapsed/refractory DLBCL predicted a 2-year OS of only 13% after a subsequent auto-HCT [30]. This simple-to-use prognostic score can be utilized as a benchmark tool to investigate novel therapeutic approaches in this ultra-high-risk DLBCL subgroup. The data for auto-HCT in relapsed/refractory DHL/DEL are sparse. This question was addressed in a recent multicenter retrospective study, limited to relapsed/chemosensitive DLBCL patients (non-DHL/DEL = 58, DEL = 47, DHL = 12) who underwent auto-HCT [17]. This study suggested that perhaps a subset of chemosensitive DEL (without DHL status) can experience durable disease control with auto-HCT (4-year progression-free survival [PFS] of 48%), but outcomes of chemosensitive DHL (or concurrent DHL/DEL) patients remained dismal, underscoring the need for alternative consolidation modalities of this subgroup.
178
Bottom line
179
Auto-HCT remains the standard of care for chemosensitive relapsed/refractory DLBCL patients. For the REFINEdefined ultra-high-risk patients or relapsed DHL patients, alternative consolidation modalities are urgently needed.
157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176
180 181 182
183
184 185 186 187 188 189 190 191 192 193 194 195
196 197
198 199 200 201 202 203 204 205 206 207 208 209
Role of allo-HCT in DLBCL Approximately 30–40% of DLBCL cases will experience relapse/progression after auto-HCT. In addition, auto-HCT is not appropriate for patients with refractory disease. For such patients, allo-HCT might be curative option. Table 1 summarizes recent studies of allo-HCT involving DLBCL patients (n 40). These studies, in general, show that reduced-intensity conditioning (RIC) allo-HCT can provide durable disease control (OS, 45–50%; PFS, 35–45%) in patients with relapsed/refractory DLBCL [31–34]. CIBMTR data show no clear benefit of myeloablative conditioning (MAC) over RIC allo-HCT in either chemosensitive [35] or refractory patients [36].
Identifying DLBCL patients likely to benefit from allo-HCT after prior autografting Easy-to-use prognostic models able to predict allo-HCT outcomes in DLBCL relapsing after a prior auto-HCT could be clinically useful. Unlike auto-HCT, pre allo-HCT positron emission tomography scans have not consistently been shown to predict transplantation outcomes [37]. CIBMTR recently reported a prognostic model that based on three prognostic variables (Karnofsky Performance Status < 80, chemoresistant disease at allo-HCT, and <1 year between auto-HCT and allo-HCT) was able to define four different risk groups: low, intermediate, high and very-high with 3year OS probabilities of 43%, 39%, 19%, and 11% respectively [38]. This model can be used to identify patients unlikely to
3 benefit from allo-HCT, or those where relapse prevention strategies postallografting should be evaluated. The role of allo-HCT in relapsed/refractory DHL/DEL warrants investigation. In a recent retrospective analysis of DLBCL patients undergoing allo-HCT, interestingly, neither DHL nor DEL seemed to impact outcomes (PFS, OS, nonrelapse mortality, and relapse), suggesting that allo-HCT may potentially abrogate the negative prognostic impact of this high-risk subset of DLBCL patients [17].
210
Bottom line
219
There is no clear benefit of MAC over RIC allo-HCT in patients with DLBCL. CIBMTR risk score can be used to identify patients likely to benefit from allo-HCT, after failing a prior autograft. Allo-HCT also seems to have a role in biologically high-risk patients (DHL and DEL) but needs further validation.
220
Follicular lymphoma
226
FL is a common indolent NHL, with disease course highlighted by frequent relapses and eventual evolution to refractory disease.
227
211 212 213 214 215 216 217 218
221 222 223 224 225
228 229
Role of auto-HCT in FL
230
Upfront transplant (in CR1)
231
The role of upfront auto-HCT consolidation was evaluated for FL patients in CR1 in an effort to improve outcomes. Three randomized studies (German Low-Grade Lymphoma Study Group [GLSG], Groupe d’Etude des Lymphomes de ´mies et Autres l’Adulte, and Groupe Ouest-Est des Leuce Maladies du Sang [GOELAMS]) conducted in the prerituximab era for advanced stage FL patients in CR1 compared consolidation strategy with auto-HCT or interferon maintenance [39–41]. While a PFS benefit was noted in GLSG and GOELAMS trials [39,41], none of these studies reported an OS benefit with auto-HCT consolidation. Moreover, there was a significantly increased risk of secondary malignancies (such as myelodysplastic syndrome and acute myeloid leukemia) in the auto-HCT consolidation arm. A major limitation of these three trials is that they were conducted in the pre-rituximab era, and hence their applicability and relevance are questioned in current practice. The only randomized study addressing this question in the rituximab era also showed results in line with the pre-rituximab era by demonstrating an improved PFS but not OS, along with a trend toward more second malignancies with upfront auto-HCT [42]. Routine use of auto-HCT for FL in CR1 is not recommended.
232
Auto-HCT in relapsed/refractory FL
255
Although FL patients generally respond to initial therapy, disease relapse remains common. The CUP trial conducted in the pre-rituximab era compared salvage chemotherapy alone with chemotherapy followed by either purged or unpurged auto-HCT consolidation, in relapsed/chemosensi-
256
Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254
257 258 259 260
HEMONC 178 17 June 2017
No. of Pages 8, Model 6+
4
N. Epperla, M. Hamadani Table 1
Studies reporting the outcomes of allogeneic HCT for DLBCL (>40 patients).
Author or registry (y)
N
Prior auto-HCT
Median age (range)
Conditioning regimen
NRM/TRM (y)
Relapse (y)
PFS (y)
OS (y)
Thomson et al [32] Sirvent et al [33] CIBMTR [66] van Kampen et al [34]
48 68 79a 101
69% 79% 0% 100%
46 48 46 46
32% 23% 43% 28%
(4) (1) (3) (3)
33% 41% 33% 30%
47% 44% 24% 42%
48% 49% 26% 54%
Rigacci et al [67]
165
100%
43 (16–65)
19–32% (2)
NR
32% (2)
39% (2)
CIBMTR [35]
396
32%
54 (18–66)
36–56% (5)
26–40% (5)
15–25% (5)
18–26% (5)
CIBMTR [36]
533b
25%
CIBMTR [38]
503
100%
46 (19–66) 53 (20–70) 52 (19–72)
RIC (100%) RIC (100%) MAC (100%) MAC (37%) RIC (63%) MAC (30%) RIC (70%) MAC (42%) RIC (58%) MAC (58%) RIC (42%) MAC (25%) RIC (75%)
53% (3) 42% (3) 31% (5)
28% (3) 35% (3) 40% (5)
19% (3) 23% (3) 29% (5)
19% (3) 28% (3) 34% (5)
(23–64) (17–66) (21–59) (18–66)
(4) (2) (3) (3)
(4) (2) (3) (3)
(4) (2) (3) (3)
Note. CIBMTR = Center for International Blood and Marrow Transplant Research; DLBCL = diffuse large B-cell lymphoma; FL = follicular lymphoma; HCT = hematopoietic cell transplantation; MAC = myeloablative conditioning; NR = not reported; NRM = nonrelapse mortality; OS = overall survival; PFS = progression-free survival; RIC = reduced intensity conditioning; TRM = treatment-related mortality. a Analysis restricted to patients undergoing myeloablative allo-HCT as first transplant. b Included 85% DLBCL and 15% FL Grade 3; all patients had chemo-resistant disease pretransplant.
261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286
287
288 289 290 291 292 293
tive FL. This trial was closed prematurely due to poor accrual, but despite the small number of patients enrolled (n = 89), it showed a significant PFS and OS benefit in favor of auto-HCT [43]. However, the current clinical relevance of this trial is questionable, because it was conducted before the advent of contemporary chemoimmunotherapies. To investigate whether auto-HCT offered any benefit over chemotherapy alone in the rituximab era, a post hoc analysis of patients enrolled in two consecutive French studies (Groupe d’Etude des Lymphomes Folliculaires [GELF]) was performed. In this analysis, in patients who received rituximab-based salvage therapies, auto-HCT was not associated with statistically significant OS benefit when compared with patients receiving salvage chemoimmunotherapy alone [44]. Of note, patients in GELF studies did not receive rituximab-based frontline therapies. In the combined St. Bart/Dana-Farber retrospective study of relapsed FL patients (again primarily from the prerituximab era), prolonged remissions were noted after auto-HCT, but this benefit was restricted mostly to patients in second CR [45]. However, a large set of registry data (European Society for Blood and Marrow Transplantation and CIBMTR) did not show a clear plateau in relapse rates postautografting in FL [46–49]. In addition, the risk of second malignancies after auto-HCT (5–15%) has historically dampened the enthusiasm for this modality [45,46].
Role of allo-HCT in relapsed/refractory FL Allo-HCT is a potentially curative option for FL. A CIBMTR study that compared auto-HCT with allo-HCT in the prerituximab era showed significantly reduced risk of relapse after allo-HCT with clear plateaus in relapse curves [60]. Subsequently, several small prospective and large registry studies have looked at the role of allo-HCT in relapsed/
refractory FL and are summarized in Table 2. These studies show that while transplant-related mortality with RIC alloHCT ranged from 8% to 17% at 1 year, PFS has been in the 51–83% range [32,48–58].
Auto-HCT vs. allo-HCT in relapsed/refractory FL The data discussed above have fueled the decades-old quest of defining the superiority of auto-HCT versus allo-HCT over each other. The Blood and Marrow Transplant Clinical Trials Network (BMT CTN) 0202 trial tried to answer this question by comparing auto-HCT with RIC allo-HCT in a randomized fashion in FL. Unfortunately, the trial was closed early due to poor accrual (N for auto-HCT = 22 and N for alloHCT = 8) [59]. For the 30 patients enrolled in the BMT CTN 0202 study, the 3-year OS was 73% with auto-HCT versus 100% following allo-HCT, and 3-year PFS was 63% in the auto-HCT group versus 86% in the allo-HCT cohort. Because there are no randomized data in the rituximab era, CIBMTR analyzed the auto-HCT versus RIC allo-HCT in FL treated in the rituximab era. This updated study suggested that autoHCT and allo-HCT, when applied as the first transplantation approach, provided comparable outcomes in FL; however, the risk of relapse was substantially lower, and nonrelapse mortality significantly higher after allo-HCT. In long-term disease-free survivors (>2 years), allo-HCT was associated with a survival benefit [48,49].
High-risk FL: What are the options? Choosing auto-HCT vs. allo-HCT in relapsed FL remains controversial and largely a factor of treating physician and transplant center preferences. Based on mostly pre-
Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
294 295 296 297
298 299
300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319
320
321 322 323
HEMONC 178 17 June 2017
No. of Pages 8, Model 6+
HCT in DLBCL and FL Table 2
5
Studies evaluating allogeneic HCT in relapsed/refractory FL (>30 patients).
Author/registry (y)
N
Conditioning regimen
Median age (range)
TRM/NRM (y)
Relapse (y)
PFS/EFS (y)
OS (y)
Rezvani et al [50] CIBMTR [51] Pinana et al [52] Thomson et al [68] Shea et al [57] Khouri et al [53] Evens et al [56] Robinson et al [54] CIBMTR [48] Yano et al [55] CIBMTR [49] BMT CTN [58]
46 208 37 82 44a 47 48 149 268 46 70 62
RIC MAC (58%) RIC (42%) RIC RIC RIC RIC NR RIC RIC RIC RIC RIC
54 44 50 45 53 53 50 51 52 48 53 55
42% (3) 23% (1) 37% (4) 15% (4) 9 15% (8) 24% (3) 22% (3) 26% (5) 23% (5) 27% (5) 13 (3)
14% (3) 8–17% (3) 8% (4) 26% (4) NR 4% (8) 16% (3) 20% (5) 20% (5) 15% (5) 20% (5) 16 (3)
43% (3) 55–67% (3) 57% (4) 76% (4) 75 83% (8) NR 57% (5) 68% (5) 75% (5) 51% (5) 71 (3)
52% (3) 62–71% (3) 57% (4) 76% (4) 81 85% (8) 61% (3) 67% (5) 66% (5) 81% (5) 54% (5) 82 (3)
(33–66) (27–70) 51 (27–700 (34–62) (26–65) (39–68) (33–68) (27–64) (33–66) (27–74) (34–66) (36–64) (29–74)
Note. BMT CTN = Blood and Marrow Transplant Clinical Trials Network; CIBMTR = Center for International Blood and Marrow Transplant Research; EFS = event-free survival; FL = follicular lymphoma; HCT = hematopoietic cell transplantation; MAC = myeloablative conditioning; NR = not reported; NRM = nonrelapse mortality; OS = overall survival; PFS = progression-free survival; RIC = reduced intensity conditioning; TRM = treatment-related mortality. a Of the 44 evaluable patients, there were only 16 FL patients.
Conclusion
Fig. 1 Overall survival by lines of chemotherapy prior to autologous hematopoietic cell transplantation.
324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341
rituximab era data showing greater benefit of auto-HCT early in the disease course, it is common practice in many centers to limit auto-HCT use in patients who have received two to three prior lines of therapy [45,60]. However, rituximab-era unpublished data from the CIBMTR show that lines of prior therapies might not be predictive of auto-HCT outcomes (unpublished data; Fig. 1). Perhaps in the rituximab era, investigation of auto-HCT consolidation in biologically high-risk cohort of FL cases must be performed. The National LymphoCare Study (NLCS) recently reported that early failure of first-line chemoimmunotherapies (<2 years) identified a biologically high-risk subset of FL patients with an extremely poor prognosis [61]. An ongoing collaborative study between CIBMTR and NLCS is investigating the role of auto-HCT in FL patients experiencing early failure of chemoimmunotherapies. Results from that study will hopefully clarify the role of auto-HCT in this high-risk group of patients.
Although rapid strides have been made in the treatment of NHL largely due to the better understanding of the molecular pathogenesis, there are still areas that need further investigation. Studies should be performed to identify high-risk FL patients who are likely to benefit from early application of auto-HCT and hopefully the ongoing NLCS/ CIBMTR study will inform this question. Effective salvage therapies, especially for REFINE ultra-high-risk DLBCL, are an unmet medical need. These patients should be ideally enrolled into clinical trials with experimental therapies such as the use of chimeric antigen receptor T cells as standalone therapy [62] or in the setting of auto-HCT [63] or allo-HCT [64]. Efforts need to be focused on evaluating novel post-HCT consolidation or maintenance strategies, possibly with agents not used in induction chemoimmunotherapies. Currently, there are several ongoing trials in the post-auto-HCT setting evaluating the combination of bortezomib and vorinostat (NCT00992446), everolimus and rituximab (NCT01665768), lenalidomide (NCT01241734), pembrolizumab (NCT02362997), and ibrutinib (BMT CTN/Alliance trial, which will be opened soon). Consolidation and/or maintenance with monoclonal antibodies (to cite a few anti-CD 79b [Polatuzumab vedotin], anti-CD19 [MEDI 551], and anti-CD20 [Obinutuzumab]), programmed death-ligand 1 (MPDL3280A), Bcl-2 inhibitors (ABT-199), and aurora A kinase inhibitors (Alisertib) in the post-auto-HCT setting seem to be a potential area of further investigation [65]. Although the emergence of targeted, biologic, and immunological therapies is exciting, it is currently unclear how these new therapies might enhance or replace HCT. Until we have further definitive data, allo-HCT remains the only curative option.
Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
342
343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375
HEMONC 178 17 June 2017
6
N. Epperla, M. Hamadani
376
Conflicts of interest
377
The authors have no conflicts of interest to declare.
378
References
379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438
No. of Pages 8, Model 6+
[1] The International Non-Hodgkin’s Lymphoma Prognostic Factors Project. A predictive model for aggressive non-Hodgkin’s lymphoma. N Engl J Med 1993;329:987–94. [2] Sehn LH, Berry B, Chhanabhai M, Fitzgerald C, Gill K, Hoskins P, et al. The revised International Prognostic Index (R-IPI) is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 2007;109:1857–61. [3] Zhou Z, Sehn LH, Rademaker AW, Gordon LI, Lacasce AS, Crosby-Thompson A, et al. An enhanced International Prognostic Index (NCCN-IPI) for patients with diffuse large B-cell lymphoma treated in the rituximab era. Blood 2014;123:837–42. [4] Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med 2002;346:1937–47. [5] Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, et al. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 2004;103:275–82. [6] Cheah CY, Oki Y, Westin JR, Turturro F. A clinician’s guide to double hit lymphomas. Brit J Haematol 2015;168:784–95. [7] Savage KJ, Johnson NA, Ben-Neriah S, Connors JM, Sehn LH, Farinha P, et al. MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy. Blood 2009;114:3533–7. [8] Barrans S, Crouch S, Smith A, Turner K, Owen R, Patmore R, et al. Rearrangement of MYC is associated with poor prognosis in patients with diffuse large B-cell lymphoma treated in the era of rituximab. J Clin Oncol 2010;28:3360–5. [9] Hu S, Xu-Monette ZY, Tzankov A, Green T, Wu L, Balasubramanyam A, et al. MYC/BCL2 protein coexpression contributes to the inferior survival of activated B-cell subtype of diffuse large B-cell lymphoma and demonstrates high-risk gene expression signatures: a report from the international DLBCL rituximab-CHOP consortium program. Blood 2013; 121: 4021– 31 [quiz 250]. [10] Oki Y, Noorani M, Lin P, Davis RE, Neelapu SS, Ma L, et al. Double hit lymphoma: the MD Anderson Cancer Center clinical experience. Brit J Haematol 2014;166:891–901. [11] Petrich AM, Gandhi M, Jovanovic B, Castillo JJ, Rajguru S, Yang DT, et al. Impact of induction regimen and stem cell transplantation on outcomes in double-hit lymphoma: a multicenter retrospective analysis. Blood 2014;124:2354–61. [12] Karube K, Campo E. MYC alterations in diffuse large B-cell lymphomas. Semin Hematol 2015;52:97–106. [13] Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127:2375–90. [14] Johnson NA, Slack GW, Savage KJ, Connors JM, Ben-Neriah S, Rogic S, et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol 2012;30:3452–9. [15] Horn H, Ziepert M, Becher C, Barth TF, Bernd HW, Feller AC, et al. MYC status in concert with BCL2 and BCL6 expression predicts outcome in diffuse large B-cell lymphoma. Blood 2013;121:2253–63.
[16] Perry AM, Alvarado-Bernal Y, Laurini JA, Smith LM, Slack GW, Tan KL, et al. MYC and BCL2 protein expression predicts survival in patients with diffuse large B-cell lymphoma treated with rituximab. Brit J Haematol 2014;165:382–91. [17] Herrera AF, Mei M, Low L, Kim HT, Griffin GK, Song JY, et al. Relapsed or refractory double-expressor and double-hit lymphomas have inferior progression-free survival after autologous stem-cell transplantation. J Clin Oncol 2017;35:24–31. [18] Vitolo U, Chiappella A, Angelucci E, Rossi G, Liberati AM, Cabras MG, et al. Dose-dense and high-dose chemotherapy plus rituximab with autologous stem cell transplantation for primary treatment of diffuse large B-cell lymphoma with a poor prognosis: a phase II multicenter study. Haematologica 2009;94:1250–8. [19] Martelli M, Gherlinzoni F, De Renzo A, Zinzani PL, De Vivo A, Cantonetti M, et al. Early autologous stem-cell transplantation versus conventional chemotherapy as front-line therapy in high-risk, aggressive non-Hodgkin’s lymphoma: an Italian multicenter randomized trial. J Clin Oncol 2003;21:1255–62. [20] Milpied N, Deconinck E, Gaillard F, Delwail V, Foussard C, Berthou C, et al. Initial treatment of aggressive lymphoma with high-dose chemotherapy and autologous stem-cell support. N Engl J Med 2004;350:1287–95. [21] Greb A, Bohlius J, Schiefer D, Schwarzer G, Schulz H, Engert A. High-dose chemotherapy with autologous stem cell transplantation in the first line treatment of aggressive non-Hodgkin lymphoma (NHL) in adults. Cochrane Database Syst Rev; 2008. CD004024. [22] Greb A, Bohlius J, Trelle S, Schiefer D, De Souza CA, Gisselbrecht C, et al. High-dose chemotherapy with autologous stem cell support in first-line treatment of aggressive nonHodgkin lymphoma—results of a comprehensive meta-analysis. Cancer Treat Rev 2007;33:338–46. [23] Stiff PJ, Unger JM, Cook JR, Constine LS, Couban S, Stewart DA, et al. Autologous transplantation as consolidation for aggressive non-Hodgkin’s lymphoma. N Engl J Med 2013;369:1681–90. [24] Schmitz N, Nickelsen M, Ziepert M, Haenel M, Borchmann P, Schmidt C, et al. Conventional chemotherapy (CHOEP-14) with rituximab or high-dose chemotherapy (MegaCHOEP) with rituximab for young, high-risk patients with aggressive B-cell lymphoma: an open-label, randomised, phase 3 trial (DSHNHL 2002–1). Lancet Oncol 2012;13:1250–9. [25] Cortelazzo S, Tarella C, Gianni AM, Ladetto M, Barbui AM, Rossi A, et al. Randomized Trial comparing R-CHOP versus high-dose sequential chemotherapy in high-risk patients with diffuse large B-cell lymphomas. J Clin Oncol 2016;34:4015–22. [26] Cohen JB, Geyer SM, Lozanski G, Zhao W, Heerema NA, Hall NC, et al. Complete response to induction therapy in patients with Myc-positive and double-hit non-Hodgkin lymphoma is associated with prolonged progression-free survival. Cancer 2014;120:1677–85. [27] Philip T, Guglielmi C, Hagenbeek A, Somers R, Van der Lelie H, Bron D, et al. Autologous bone marrow transplantation as compared with salvage chemotherapy in relapses of chemotherapy-sensitive non-Hodgkin’s lymphoma. N Engl J Med 1995;333:1540–5. [28] Gisselbrecht C, Glass B, Mounier N, Singh Gill D, Linch DC, Trneny M, et al. Salvage regimens with autologous transplantation for relapsed large B-cell lymphoma in the rituximab era. J Clin Oncol 2010;28:4184–90. [29] Hamadani M, Hari PN, Zhang Y, Carreras J, Akpek G, Aljurf MD, et al. Early failure of frontline rituximab-containing chemoimmunotherapy in diffuse large B cell lymphoma does not predict futility of autologous hematopoietic cell transplantation. Biol Blood Marrow Transplant 2014;20:1729–36. [30] Costa LJ, Maddocks K, Epperla N, Reddy NM, Karmali R, Umyarova E, et al. Diffuse large B-cell lymphoma with primary
Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506
HEMONC 178 17 June 2017
No. of Pages 8, Model 6+
HCT in DLBCL and FL 507 508 509
[31]
510 511 512 513 514
[32]
515 516 517 518
[33]
519 520 521 522 523 524 525
[34]
526 527 528 529 530 531 532
[35]
533 534 535 536
[36]
537 538 539 540 541 542
[37]
543 544 545 546 547 548
[38]
549 550 551 552
[39]
553 554 555 556 557 558
[40]
559 560 561 562 563 564 565
[41]
566 567 568 569 570 571 572 573 574
[42]
treatment failure: ultra-high risk features and benchmarking for experimental therapies. Am J Hematol 2017;92:161–70. Rezvani AR, Norasetthada L, Gooley T, Sorror M, Bouvier ME, Sahebi F, et al. Non-myeloablative allogeneic haematopoietic cell transplantation for relapsed diffuse large B-cell lymphoma: a multicentre experience. Brit J Haematol 2008;143:395–403. Thomson KJ, Morris EC, Bloor A, Cook G, Milligan D, Parker A, et al. Favorable long-term survival after reduced-intensity allogeneic transplantation for multiple-relapse aggressive nonHodgkin’s lymphoma. J Clin Oncol 2009;27:426–32. Sirvent A, Dhedin N, Michallet M, Mounier N, Faucher C, Yakoub-Agha I, et al. Low nonrelapse mortality and prolonged long-term survival after reduced-intensity allogeneic stem cell transplantation for relapsed or refractory diffuse large B cell lymphoma: report of the Societe Francaise de Greffe de Moelle et de Therapie Cellulaire. Biol Blood Marrow Transplant 2010;16:78–85. van Kampen RJ, Canals C, Schouten HC, Nagler A, Thomson KJ, Vernant JP, et al. Allogeneic stem-cell transplantation as salvage therapy for patients with diffuse large B-cell nonHodgkin’s lymphoma relapsing after an autologous stem-cell transplantation: an analysis of the European Group for Blood and Marrow Transplantation Registry. J Clin Oncol 2011;29:1342–8. Bacher U, Klyuchnikov E, Le-Rademacher J, Carreras J, Armand P, Bishop MR, et al. Conditioning regimens for allotransplants for diffuse large B-cell lymphoma: myeloablative or reduced intensity? Blood 2012;120:4256–62. Hamadani M, Saber W, Ahn KW, Carreras J, Cairo MS, Fenske TS, et al. Impact of pretransplantation conditioning regimens on outcomes of allogeneic transplantation for chemotherapyunresponsive diffuse large B cell lymphoma and grade III follicular lymphoma. Biol Blood Marrow Transplant 2013;19:746–53. Bachanova V, Burns LJ, Ahn KW, Laport GG, Akpek G, KharfanDabaja MA, et al. Impact of pretransplantation 18F-fluorodeoxy glucose-positron emission tomography status on outcomes after allogeneic hematopoietic cell transplantation for nonHodgkin lymphoma. Biol Blood Marrow Transplant 2015;21:1605–11. Fenske TS, Ahn KW, Graff TM, DiGilio A, Bashir Q, Kamble RT, et al. Allogeneic transplantation provides durable remission in a subset of DLBCL patients relapsing after autologous transplantation. Brit J Haematol 2016;174:235–48. Lenz G, Dreyling M, Schiegnitz E, Forstpointner R, Wandt H, Freund M, et al. Myeloablative radiochemotherapy followed by autologous stem cell transplantation in first remission prolongs progression-free survival in follicular lymphoma: results of a prospective, randomized trial of the German Low-Grade Lymphoma Study Group. Blood 2004;104:2667–74. Sebban C, Mounier N, Brousse N, Belanger C, Brice P, Haioun C, et al. Standard chemotherapy with interferon compared with CHOP followed by high-dose therapy with autologous stem cell transplantation in untreated patients with advanced follicular lymphoma: the GELF-94 randomized study from the Groupe d’Etude des Lymphomes de l’Adulte (GELA). Blood 2006;108:2540–4. Gyan E, Foussard C, Bertrand P, Michenet P, Le Gouill S, Berthou C, et al. High-dose therapy followed by autologous purged stem cell transplantation and doxorubicin-based chemotherapy in patients with advanced follicular lymphoma: a randomized multicenter study by the GOELAMS with final results after a median follow-up of 9 years. Blood 2009;113:995–1001. Ladetto M, De Marco F, Benedetti F, Vitolo U, Patti C, Rambaldi A, et al. Prospective, multicenter randomized GITMO/IIL trial comparing intensive (R-HDS) versus conven-
7
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
tional (CHOP-R) chemoimmunotherapy in high-risk follicular lymphoma at diagnosis: the superior disease control of R-HDS does not translate into an overall survival advantage. Blood 2008;111:4004–13. Schouten HC, Qian W, Kvaloy S, Porcellini A, Hagberg H, Johnsen HE, et al. High-dose therapy improves progressionfree survival and survival in relapsed follicular non-Hodgkin’s lymphoma: results from the randomized European CUP trial. J Clin Oncol 2003;21:3918–27. Sebban C, Brice P, Delarue R, Haioun C, Souleau B, Mounier N, et al. Impact of rituximab and/or high-dose therapy with autotransplant at time of relapse in patients with follicular lymphoma: a GELA study. J Clin Oncol 2008;26:3614–20. Rohatiner AZ, Nadler L, Davies AJ, Apostolidis J, Neuberg D, Matthews J, et al. Myeloablative therapy with autologous bone marrow transplantation for follicular lymphoma at the time of second or subsequent remission: long-term follow-up. J Clin Oncol 2007;25:2554–9. Montoto S, Canals C, Rohatiner AZ, Taghipour G, Sureda A, Schmitz N, et al. Long-term follow-up of high-dose treatment with autologous haematopoietic progenitor cell support in 693 patients with follicular lymphoma: an EBMT registry study. Leukemia 2007;21:2324–31. van Besien K, Loberiza Jr FR, Bajorunaite R, Armitage JO, Bashey A, Burns LJ, et al. Comparison of autologous and allogeneic hematopoietic stem cell transplantation for follicular lymphoma. Blood 2003;102:3521–9. Klyuchnikov E, Bacher U, Kroger NM, Hari PN, Ahn KW, Carreras J, et al. Reduced-intensity allografting as first transplantation approach in relapsed/refractory grades one and two follicular lymphoma provides improved outcomes in long-term survivors. Biol Blood Marrow Transplant 2015;21:2091–9. Klyuchnikov E, Bacher U, Woo Ahn K, Carreras J, Kroger NM, Hari PN, et al. Long-term survival outcomes of reducedintensity allogeneic or autologous transplantation in relapsed grade 3 follicular lymphoma. Bone Marrow Transplant 2016;51:58–66. Rezvani AR, Storer B, Maris M, Sorror ML, Agura E, Maziarz RT, et al. Nonmyeloablative allogeneic hematopoietic cell transplantation in relapsed, refractory, and transformed indolent non-Hodgkin’s lymphoma. J Clin Oncol 2008;26:211–7. Hari P, Carreras J, Zhang MJ, Gale RP, Bolwell BJ, Bredeson CN, et al. Allogeneic transplants in follicular lymphoma: higher risk of disease progression after reduced-intensity compared to myeloablative conditioning. Biol Blood Marrow Transplant 2008;14:236–45. Pinana JL, Martino R, Gayoso J, Sureda A, de la Serna J, DiezMartin JL, et al. Reduced intensity conditioning HLA identical sibling donor allogeneic stem cell transplantation for patients with follicular lymphoma: long-term follow-up from two prospective multicenter trials. Haematologica 2010;95:1176–82. Khouri IF, Saliba RM, Erwin WD, Samuels BI, Korbling M, Medeiros LJ, et al. Nonmyeloablative allogeneic transplantation with or without 90yttrium ibritumomab tiuxetan is potentially curative for relapsed follicular lymphoma: 12-year results. Blood 2012;119:6373–8. Robinson SP, Canals C, Luang JJ, Tilly H, Crawley C, Cahn JY, et al. The outcome of reduced intensity allogeneic stem cell transplantation and autologous stem cell transplantation when performed as a first transplant strategy in relapsed follicular lymphoma: an analysis from the Lymphoma Working Party of the EBMT. Bone Marrow Transplant 2013;48:1409–14. Yano S, Mori T, Kanda Y, Kato J, Nakaseko C, Fujisawa S, et al. Favorable survival after allogeneic stem cell transplantation with reduced-intensity conditioning regimens for relapsed/ refractory follicular lymphoma. Bone Marrow Transplant 2015;50:1299–305.
Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642
HEMONC 178 17 June 2017
No. of Pages 8, Model 6+
8 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677
N. Epperla, M. Hamadani
[56] Evens AM, Vanderplas A, LaCasce AS, Crosby AL, Nademanee AP, Kaminski MS, et al. Stem cell transplantation for follicular lymphoma relapsed/refractory after prior rituximab: a comprehensive analysis from the NCCN lymphoma outcomes project. Cancer 2013;119:3662–71. [57] Shea T, Johnson J, Westervelt P, Farag S, McCarty J, Bashey A, et al. Reduced-intensity allogeneic transplantation provides high event-free and overall survival in patients with advanced indolent B cell malignancies: CALGB 109901. Biol Blood Marrow Transplant 2011;17:1395–403. [58] Laport GG, Wu J, Logan B, Bachanova V, Hosing C, Fenske T, et al. Reduced-intensity conditioning with fludarabine, cyclophosphamide, and high-dose rituximab for allogeneic hematopoietic cell transplantation for follicular lymphoma: a phase two multicenter trial from the Blood and Marrow Transplant Clinical Trials Network. Biol Blood Marrow Transplant 2016;22:1440–8. [59] Tomblyn MR, Ewell M, Bredeson C, Kahl BS, Goodman SA, Horowitz MM, et al. Autologous versus reduced-intensity allogeneic hematopoietic cell transplantation for patients with chemosensitive follicular non-Hodgkin lymphoma beyond first complete response or first partial response. Biol Blood Marrow Transplant 2011;17:1051–7. [60] Vose JM, Bierman PJ, Loberiza FR, Lynch JC, Bociek GR, Weisenburger DD, et al. Long-term outcomes of autologous stem cell transplantation for follicular non-Hodgkin lymphoma: effect of histological grade and Follicular International Prognostic Index. Biol Blood Marrow Transplant 2008;14:36–42. [61] Casulo C, Byrtek M, Dawson KL, Zhou X, Farber CM, Flowers CR, et al. Early relapse of follicular lymphoma after rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone defines patients at high risk for death: an analysis from the National LymphoCare Study. J Clin Oncol 2015;33:2516–22. [62] Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, et al. Chemotherapy-
[63]
[64]
[65]
[66]
[67]
[68]
refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol 2015;33:540–9. Wang X, Popplewell LL, Wagner JR, Naranjo A, Blanchard MS, Mott MR, et al. Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL. Blood 2016;127:2980–90. Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 2013;122:4129–39. Epperla N, Fenske TS, Lazarus HM, Hamadani M. Post-autologous transplant maintenance therapies in lymphoid malignancies: are we there yet? Bone Marrow Transplant 2015;50:1393–404. Lazarus HM, Zhang MJ, Carreras J, Hayes-Lattin BM, Ataergin AS, Bitran JD, et al. A comparison of HLA-identical sibling allogeneic versus autologous transplantation for diffuse large B cell lymphoma: a report from the CIBMTR. Biol Blood Marrow Transplant 2010;16:35–45. Rigacci L, Puccini B, Dodero A, Iacopino P, Castagna L, Bramanti S, et al. Allogeneic hematopoietic stem cell transplantation in patients with diffuse large B cell lymphoma relapsed after autologous stem cell transplantation: a GITMO study. Ann Hematol 2012;91:931–9. Thomson KJ, Morris EC, Milligan D, Parker AN, Hunter AE, Cook G, et al. T-cell-depleted reduced-intensity transplantation followed by donor leukocyte infusions to promote graft-versuslymphoma activity results in excellent long-term survival in patients with multiply relapsed follicular lymphoma. J Clin Oncol 2010;28:3695–700.
712
Please cite this article in press as: Epperla N, Hamadani M, Hematopoietic cell transplantation for diffuse large B-cell and follicular lymphoma: Current controversies and advances, Hematol Oncol Stem Cell Ther (2017), http://dx.doi.org/10.1016/j.hemonc.2017.05.004
678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711