Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Apr 1.
Published in final edited form as: Transplant Cell Ther. 2022 Jan 23;28(4):187.e1–187.e10. doi: 10.1016/j.jtct.2022.01.017

Outcomes of Allogeneic Hematopoietic Cell Transplantation in T-cell Prolymphocytic Leukemia: A Contemporary Analysis from the Center for International Blood and Marrow Transplant Research

Hemant S Murthy 1, Kwang Woo Ahn 2,3, Noel Estrada-Merly 2, Hassan B Alkhateeb 4, Susan Bal 5, Mohamed A Kharfan-Dabaja 1, Bhagirathbhai Dholaria 6, Francine Foss 7, Lohith Gowda 8, Deepa Jagadeesh 9, Craig Sauter 10,11, Muhammad Bilal Abid 12, Mahmoud Aljurf 13, Farrukh T Awan 14, Ulrike Bacher 15, Sherif M Badawy 16,17, Minoo Battiwalla 18, Chris Bredeson 19, Jan Cerny 20, Saurabh Chhabra 2, Abhinav Deol 21, Miguel Angel Diaz 22, Nosha Farhadfar 23, César Freytes 24, James Gajewski 25, Manish J Gandhi 26, Siddhartha Ganguly 27, Michael R Grunwald 28, Joerg Halter 29, Shahrukh Hashmi 30,31, Gerhard C Hildebrandt 32, Yoshihiro Inamoto 33, Antonio Martin Jimenez-Jimenez 34, Matt Kalaycio 35, Rammurti Kamble 36, Maxwell M Krem 32, Hillard M Lazarus 37, Aleksandr Lazaryan 38, Joseph Maakaron 39, Pashna N Munshi 40, Reinhold Munker 32, Aziz Nazha 9, Taiga Nishihori 38, Olalekan O OIuwole 6, Guillermo Ortí 41, Dorothy C Pan 42, Sagar S Patel 43, Attaphol Pawarode 44, David Rizzieri 45, Nakhle S Saba 46, Bipin Savani 47, Sachiko Seo 48, Celalettin Ustun 49, Marjolein van der Poel 50, Leo F Verdonck 51, John L Wagner 52, Baldeep Wirk 53, Betul Oran 54, Ryotaro Nakamura 55, Bart Scott 56, Wael Saber 2
PMCID: PMC8977261  NIHMSID: NIHMS1784251  PMID: 35081472

Abstract

Background:

T-cell prolymphocytic leukemia (T-PLL) is a rare, aggressive malignancy with limited treatment options and poor long-term survival. Previous studies of allogeneic hematopoietic cell transplantation (alloHCT) for T-PLL are limited by small numbers, and descriptions of patient and transplant characteristics and outcomes after alloHCT are sparse.

Objective:

To describe outcomes of alloHCT in T-PLL and identify predictors of post-transplant relapse and survival.

Study Design:

We conducted an analysis of data using the Center for International Blood and Marrow Transplant Research (CIBMTR) database on 266 patients with T-PLL who underwent alloHCT during 2008–2018.

Results:

The 4-year rates of overall survival (OS), disease-free survival (DFS), relapse, and treatment-related mortality (TRM) were 30.0% (95% CI, 23.8–36.5%), 25.7% (95% CI, 20–32%), 41.9% (95% CI, 35.5–48.4%), and 32.4% (95% CI, 26.4–38.6%), respectively. In multivariable analyses, three variables were associated with inferior OS: myeloablative conditioning (MAC) (hazard ratio [HR] 2.18, p<0.0001); age older than 60 years (HR 1.61, p=0.0053); and suboptimal performance status defined by Karnofsky Performance Status (KPS) <90 (HR 1.53, p=0.0073). MAC also was associated with increased TRM (HR 3.31, p<0.0001), increased cumulative incidence of grade 2–4 acute graft-versus-host disease (GVHD) (HR 2.94, p=0.0011) and an inferior disease-free survival (HR 1.86, p=0.0004). Conditioning intensity was not associated with relapse; however stable disease/progression correlated with increased risk of relapse (HR 2.13, p=0.0072). Both in vivo T cell depletion (TCD) as part of conditioning and KPS <90 were associated with worse TRM and inferior DFS. Total Body Irradiation was not found to have any significant effect on OS, DFS or TRM.

Conclusion:

Our data showed that reduced-intensity conditioning without in vivo T-cell depletion (that is, without ATG or alemtuzumab) prior to alloHCT was associated with long-term disease-free survival in patients with T-PLL who were 60 or younger or who had KPS >90 or had chemo-sensitive disease.

INTRODUCTION

T-cell prolymphocytic leukemia (T-PLL) is a rare aggressive malignancy, representing approximately 2% of mature lymphocytic leukemias in adults[1,2]. Patients tend to be older, with a median age of 65 years at diagnosis. Typically, T-PLL presents with signs such as marked leukocytosis, hepatosplenomegaly, lymphadenopathy, and cutaneous lesions. Treatment options are generally limited, and outcomes are poor, with a reported median survival of 19 months[3]. Alemtuzumab, an anti-CD52 humanized monoclonal antibody, is often used in the front line in T-PLL. While complete remission (CR) rates with alemtuzumab are high (60–80%), most responses are brief, and the relapse rate remains high[4,5]. Survival of patients with relapsed T-PLL is dismal, as responses to second-line therapies are limited and generally short-lived [2,6].

Allogeneic hematopoietic cell transplantation (alloHCT) is a potential curative therapy for T-PLL and has been reported to yield durable remissions, notably in patients who are in complete remission prior to transplantation[712]. AlloHCT aided small subsets of patients with T-PLL, according to studies by the Center for International Blood and Marrow Transplant Research (CIBMTR) [10], European Society for Blood and Marrow Transplantation (EBMT) [7,13], the Francophone Society of Bone Marrow Transplantation and Cellular Therapy (SFGM-TC) [9], and the Japanese Society for Transplantation and Cellular Therapy (JSTCT) [14]. The benefits of alloHCT are limited by high rates of non-relapse mortality (NRM), ranging from 28–40%. In addition, there exists high risk of post-transplant relapse, many occurring within 2 years of alloHCT [10,15]. Because these studies were relatively small, researchers were unable to identify factors associated with sustained remission and improved overall survival (OS). Hence, using CIBMTR Research Database, the aim of this study is to evaluate the effectiveness of alloHCT in T-PLL and to identify predictors of post-transplant relapse and survival.

METHODS

Data sources

The CIBMTR is a nonprofit research collaboration of the National Marrow Donor Program (NMDP)/Be The Match and the Medical College of Wisconsin (MCW). More than 300 medical centers worldwide submit clinical data to the CIBMTR about HCT and other cellular therapies. Participating centers are required to report all transplantations consecutively. The CIBMTR ensures data quality through computerized checks for discrepancies, physicians’ review of submitted data, and on-site audits of participating centers. The CIBMTR complies with federal regulations that protect human research participants. The Institutional Review Boards of MCW and NMDP approved this study.

Patient selection

Adults (aged 18 and older) who underwent first alloHCT for T-PLL during 2008–2018 were included in this analysis. Graft sources included peripheral blood stem cells (PBSC) and bone marrow. Eligible donors included human leukocyte antigen (HLA)-identical sibling donors or unrelated donors (URD) matched at the allele-level at HLA-A, -B, -C, and -DRB1, and alternative donor transplantation (haploidentical, mismatched unrelated donor). Cord blood and ex vivo T-cell depleted grafts were excluded, as were patients who received syngeneic transplants. AlloHCT recipients who received in vivo T-cell depletion (TCD) with anti-thymocyte globulin (ATG) or alemtuzumab were included.

Definitions and study endpoints

Disease response was defined based on National Cancer Institute-Sponsored Working Group guidelines for chronic lymphocytic leukemia. [16] The intensity of conditioning regimens was defined using published consensus criteria.[17] The primary endpoint was OS. Death from any cause was considered an event, and surviving patients were censored at the time of last follow-up. Secondary endpoints included cumulative incidence of acute graft-versus-host disease (aGVHD), chronic graft-versus-host disease (cGVHD), treatment related mortality (TRM), progression/relapse, and disease-free survival (DFS). TRM was defined as death without preceding disease relapse/progression; relapse and progression were considered competing events. Progressive disease or recurrences of T-PLL were defined as progression after alloHCT or recurrence following CR; TRM was considered competing event. DFS was defined as survival following alloHCT without relapse or progression. Patients who survived without evidence of disease relapse or progression were censored at last follow-up. The causes of death were reported in accordance to the methodology described previously. [18]

Statistical analysis

Cumulative incidence of GVHD, relapse/progression, and TRM were calculated using the cumulative incidence estimator to accommodate for competing risks. Probabilities of OS and DFS were calculated using the Kaplan-Meier method for a univariable analysis. Multivariable regression analysis was performed using logistic regression for aGVHD, the proportional cause-specific hazards model for chronic GVHD, relapse, and TRM, and the Cox proportional hazards model for DFS and OS. The assumption of proportional hazards for each factor was tested for the proportional hazards and cause-specific hazards models, and a forward stepwise selection was used to select significant risk factors. In the final model, we retained factors with statistical significance of < 5%. We examined the interaction between the main effect and the other significant variables and found no center effect based on the score test of homogeneity[19]. The variables that were considered in the multivariable models included: recipient age, Karnofsky Performance Status (KPS), Hematopoietic Cell Transplantation Comorbidity Index (HCT-CI), disease status at transplant, intensity of conditioning regimen, use of total body irradiation (TBI) in conditioning, time from diagnosis to transplant, recipients’ cytomegalovirus (CMV) serostatus, GVHD prophylaxis, donor type, graft source, use of ATG/alemtuzumab, and year of transplant. Adjusted probabilities [20,21] were calculated based on the final regression models for OS, DFS, relapse, and TRM.

RESULTS

Baseline characteristics

The study included 266 adults who received alloHCT for T-PLL. The median follow-up was 49 months (range 3.32–116.84). The baseline patient-, disease-, and transplantation-related characteristics are described in (Tables & Figures Table 1, Supplementary Table 1). Participants’ median age at the time of alloHCT was 59.1 years (range 25.0–76.3); 53% were male; and 58% had a KPS ≥90. The majority of alloHCT recipients were white (87%). Disease status at the time of HCT was CR, partial remission (PR) and chemo-refractory disease in 56%, 30% and 11%, respectively. Most patients received PBSC grafts (89%) and calcineurin-based GVHD prophylaxis (80%). Matched related donors (30%) and 8/8 matched unrelated donors (43%) were the most common types of donors. Reduced intensity and non-myeloablative conditioning (RIC/NMA) and myeloablative conditioning (MAC) were used in 70% and 30% of cases, respectively. Commonly utilized MAC regimens included cyclophosphamide-TBI (n=33) and busulfan-fludarabine (n=20) while commonly utilized RIC/NMA regimens included fludarabine-melphalan (n=55), fludarabine-busulfan (n=33), and fludarabine-TBI (n=32). A total of 49 patients (18%) received in vivo TCD with anti-thymocyte globulin (n=47) or alemtuzumab (n=2).

Table 1.

Baseline characteristics of patients who had first alloHCT for T-PLL, 2000–2018

Characteristic No. (%)
No. patients 266
No. centers 87
Sex
 Male 140 (53)
 Female 126 (47)
Age, y
Median age (range), y 59.1 (25.01–76.26)
 18–29 1 (0)
 30–39 7 (3)
 40–49 38 (14)
 50–59 98 (37)
 60–69 101 (38)
 ≥ 70 21 (8)
Karnofsky Performance Status score
 90–100 153 (58)
 < 90 101 (38)
 Not reported 12 (4)
HCT-CI
 0 73 (27)
 1–2 84 (31)
 3–4 77 (25)
 ≥ 5 28 (11)
 Not reported 4 (6)
Remission status at HCT
 Complete remission 149 (56)
 Partial response 80 (30)
 No response/ stable/ progression 31 (11)
 Not reported 6 (2)
Graft source
 Bone marrow 30 (11)
 Peripheral blood 236 (89)
Time from diagnosis to HCT
 Median (range) 7.85 (2.07–81.74)
 < 6 months 82 (31)
 6–11 months 103 (39)
 ≥ 12 months 81 (30)
Donor type
 HLA-identical sibling 80 (30)
 Haploidentical 30 (11)
 URD 8/8 115 (43)
 URD 7/8 33 (12)
 Other related 8 (3)
Conditioning regimen intensity a
 Myeloablative with TBI 44 (17)
 Myeloablative without TBI 34 (13)
 Reduced-intensity with TBI 75 (28)
 Reduced-intensity without TBI 113 (42)
GVHD prophylaxis
 CNI + MMF ± others (except PTCy) 68 (26)
 CNI + MTX ± others (except MMF, PTCy) 123 (46)
 CNI + others (except MMF, MTX, PTCy) 20 (8)
 Other prophylaxis b 55 (21)
In vivo T cell depletion (ATG/alemtuzumab) c
 Yes 49 (18)
 No 217 (82)
Median follow-up (range), months 49 (3.32–116.84)

Abbreviations: alloHCT, allogeneic hematopoietic cell transplantation; ATG, anti-thymocyte globulin; CNI, calcineurin inhibitor; GVHD, graft-versus-host disease; HCT, hematopoietic cell transplantation; HCT-CI, Hematopoietic Cell Transplantation Comorbidity Index; HLA, human leukocyte antigen; MMF, mycophenolate mofetil; MTX, methotrexate; PTCy, post-transplant cyclophosphamide; TBI, total body irradiation; T-PLL, T-cell prolymphocytic leukemia; URD, unrelated donor.

a

Refer to Supplementary Table 1 for full conditioning list

b

Other: CNI alone (12), CNI + PTCy + MMF (32), PTCy-MMF (1), sirolimus + PTCy (2), MTX alone (3), sirolimus-MMF-PTCy (1), monoclonal antibody + MMF (3), PTCy alone (1)

c

ATG n=47, alemtuzumab n=2

Overall survival and disease-free survival

The 4-year OS and DFS were 30.0% (95% CI, 23.8–36.5%) and 25.7% (95% CI, 20–32%), respectively (Supplementary Table 2). The 4-year OS based on donor for HLA matched sibling donor (MSD), 8/8 matched unrelated donor (MUD), haploidentical donor (haplo) and 7/8 mismatch unrelated donor (MMUD) was 40.1% (95% CI, 28.9–51.8%), 24.6% (95% CI, 16.2–34.2%), 33.9% (95% CI, 15–56%) and 26.8% (95% CI, 9.6–48.9%) respectively. The 4-year DFS based on donor for MSD, MUD, haplo and MMUD was 34.9% (95% CI, 24.4–46.3%), 19.6% (95% CI, 12–28.5%), 23.4% (95% CI, 8.2–43.3%), and 28.9% (95% CI, 10.4–52.1%) respectively (Supplementary Table 3).

On multivariate analyses, RIC/NMA conditioning regimen was significantly associated with longer DFS (hazard ratio [HR] 1.86; 95%CI, 1.32–2.61; p=0.0004) and OS (HR 2.18; 95% CI, 1.53–3.09; p<0.0001) when compared with MAC. (Figures 1, 2). Performance status (KPS <90%) was associated with both inferior DFS (HR 1.51; 95% CI, 1.12–2.05; p=0.0075) and OS (HR 1.53; 95% CI, 1.12–2.08; p=0.0073), as was recipient age >60 years, which was associated with inferior DFS (HR 1.41; 95% CI, 1.03–1.93; p=0.0337) and OS (HR 1.61; 95% CI, 1.15–2.24; p=0.0053). Use of in vivo TCD resulted in inferior DFS (HR 1.50; 95% CI, 1.05–2.15; p=0.0276), but had no significant effect on OS (Table 2). Time from diagnosis to transplant did not have any significant effect on DFS or OS.

Figure 1. Adjusted overall survival by conditioning intensity (P<0.0001).

Figure 1

MAC, myeloablative conditioning; RIC/NMA, reduced-intensity conditioning/nonmyeloablative conditioning.

Figure 2. Adjusted disease-free survival, by conditioning intensity (P= 0.0004).

Figure 2

MAC, myeloablative conditioning; RIC/NMA, reduced-intensity conditioning/nonmyeloablative conditioning.

Table 2.

Multivariable regression analysis

Factors N OR/HR (95% CI) P-value Overall P-value
Overall survival
 Conditioning regimen
  RIC/NMA 188 1.00 (Reference) < 0.0001
  MAC 78 2.18 (1.53– 3.09) < 0.0001
 Age
  ≤ 60 142 1.00 (Reference) 0.0053
  > 60 122 1.61 (1.15– 2.24) 0.0053
 KPS
  ≥ 90% 153 1.00 (Reference) 0.0272
  < 90% 101 1.53 (1.12– 2.08) 0.0073
  Not reported 12 1.23 (0.60– 2.54) 0.573
Disease-free survival
 Conditioning regimen
  RIC/NMA 77 1.00 (Reference) 0.0004
  MAC 187 1.86 (1.32–2.61) 0.0004
 Age
  ≤ 60 142 1.00 Reference) 0.0337
  > 60 122 1.41 (1.03– 1.93) 0.0337
 KPS
  ≥ 90% 152 1.00 (Reference) 0.0075
  < 90% 101 1.51 (1.12– 2.05) 0.0075
  Not reported 4 11 1.13 (0.53–2.44) 0.7507
 In vivo T-cell depletion
  No 215 1.00 (Reference) 0.0253
  Yes 49 1.50 (1.05–2.13) 0.0253
Treatment-related mortality
 Conditioning regimen
  RIC/NMA 187 1.00 (Reference) < 0.0001
  MAC 77 3.31 (2.01–5.45) < 0.0001
 Age
  ≤ 60 142 1.0 (Reference) 0.0108
  > 60 122 1.87 (1.16– 3.04) 0.0108
 KPS
  ≥ 90% 152 1.00 (Reference) 0.0142
  < 90% 101 1.98 (1.25– 3.14) 0.0036
  Not reported 11 1.18 (0.36–3.83) 0.7811
 In vivo T-cell depletion
  No 215 1.00 (Reference) 0.0263
  Yes 49 1.79 (1.07–2.98) 0.0263
Acute GVHD
 Conditioning regimen
  RIC/NMA 172 1.00 (Reference) 0.0011
  MAC 75 2.94 (1.54– 5.62) 0.0011
 GVHD prophylaxis
  CNI + MMF 65 1.00 (Reference) 0.0093
  CNI + MTX 114 0.56 (0.28–1.14) 0.1077
  CNI + others (except MMF, MTX, PTCy) 18 0.36 (0.11–1.17) 0.0902
  PTCy ± others 33 0.26 (0.10–0.71) 0.0082
  Other prophylaxis 17 2.17 (0.71– 6.60) 0.174
Chronic GVHD
 GVHD prophylaxis
  CNI + MMF ± others (except PTCy) 67 1.00 (Reference) 0.0015
  CNI + MTX ± others (except MMF, PTCy) 121 1.06 (0.68– 1.65) 0.8045
  CNI + others (except MMF, MTX, PTCy) 20 2.35 (1.31– 4.20) 0.0041
  PTCy ± others 37 0.44 (0.19– 1.05) 0.0645
  Other prophylaxis 17 0.65 (0.25– 1.66) 0.3677
 Year of transplant
  2008–2011 50 1.00 (Reference) 0.0216
  2012–2015 110 0.62 (0.39–0.97) 0.0382
  2016–2018 102 0.48 (0.28–0.82) 0.0069
Relapse
 Disease status at HCT
  CR 149 1.00 (Reference) 0.0486
  PR 80 1.40 (0.91–2.17) 0.1257
  No response/ SD/ PD 31 2.13 (1.23–3.71) 0.0072
  Not reported 6 0.94 (0.23– 3.87) 0.932

Abbreviations: CNI, calcineurin inhibitor; CR, complete remission; GVHD, graft-versus-host disease; HCT, hematopoietic cell transplantation; HR, hazard ratio; KPS, Karnofsky Performance Status; MAC, myeloablative conditioning; MMF, mycophenolate mofetil; MTX, methotrexate; OR, odds ratio; PD, progressive disease; PR, partial remission; PTCy, post-transplant cyclophosphamide; RIC/NMA, reduced-intensity conditioning/nonmyeloablative conditioning; SD, stable disease;

TBI effect on OS and DFS was analyzed as part of conditioning intensity (Supplementary Table 7). When comparing MAC without TBI (MAC-Chemo) to MAC with TBI, TBI did not have any significant effect on OS (HR 0.83 (95% CI, 0.49–1.41; p=0.0073) or DFS (HR 1.01 (95% CI, 0.60–1.71; p=0.9628). Performing the same analysis with RIC comparing RIC with TBI to RIC without TBI (RIC-Chemo), TBI did not have any significant effect on OS (HR 1.22 (95% CI, 0.81–1.82; p=0.3437) or DFS (HR 1.17 (95% CI, 0.79–1.72; p=0.4390).

Treatment-related mortality

The 1-year and 4-year cumulative incidence of TRM were 21.5% (95% CI, 16.7–26.7), and 32.4% (95% CI, 26.4–38.6), respectively. The 4-year TRM based on donor for MSD, MUD, haplo and MMUD was 20.4% (95% CI, 11.8–30.7%), 36.6% (95% CI, 27.3–46.4%), 31.6% (95% CI, 15.5–50.3%), and 42.1% (95% CI, 23.2–62.4%) respectively (Supplementary Table 3). On multivariate analysis, MAC resulted in increased cumulative incidence of TRM (HR 3.31; 95% CI 2.01–5.45; p<0.0001) when compared to RIC (Figure 3). Additionally, performance status (KPS < 90%) (HR 1.98; 95% CI, 1.25–3.14; p=0.0036) and use of in vivo TCD (HR 1.79; 95% CI, 1.07–2.98; p=0.0263) resulted in increased incidence of TRM (Table 2).

Figure 3. Adjusted treatment-related mortality, by conditioning intensity (P<0.0001).

Figure 3

MAC, myeloablative conditioning; RIC/NMA, reduced-intensity conditioning/nonmyeloablative conditioning.

The effect of TBI on TRM was analyzed as part of conditioning intensity (Supplementary Table 7). When comparing MAC without TBI (MAC-Chemo) to MAC with TBI, TBI did not have any significant effect on TRM (HR 0.48 (95% CI, 0.22–1.05; p=0.0662). Comparing RIC with TBI to RIC without TBI (RIC-Chemo), TBI did not have any significant effect on TRM (HR 1.39 (95% CI, 0.74–2.64; p=0.3068).

Acute and chronic GVHD

The cumulative incidence of grades II-IV aGVHD at day 180 post alloHCT was 22.5% (95% CI, 16.8–28.9) while cumulative incidence of grades III-IV aGVHD at day 180 post alloHCT was 5.3% (95% CI, 2.8–8.6). (Supplementary Table 4). The cumulative incidence of grades II-IV aGVHD at day 180 based on donor for MSD, MUD, haplo and MMUD was 14.3% (95% CI, 6.4–24.7%), 25.7% (95% CI, 16.6–36%), 36.4% (95% CI, 17.5–57.8%) and 20% (95% CI, 5.6–40.4%) respectively (Supplementary Table 3). On multivariate analysis, MAC was predictive for increased risk of grades II-IV aGVHD (OR 2.94; 95% CI, 1.54–5.62; p=0.0011), while post-transplant cyclophosphamide (PTCy) predicted for reduced grades II-IV aGVHD (OR 0.26; 95% CI, 0.10–0.71; p=0.0082) (Table 2). In vivo TCD did not have a significant effect on aGVHD. When comparing MAC with TBI to MAC without TBI as well as RIC with TBI to RIC without TBI, TBI did not have any significant effect on aGVHD (Supplemental Table 7).

The cumulative incidences of chronic GVHD (cGVHD) at 1 year and 2 years post-transplant were 38.8% (95% CI, 32.9–44.9) and 45.5% (95% CI, 39.2–51.8), respectively. Among those with cGVHD at 1 year, 71% had extensive cGVHD and 29% with limited cGVHD, while at 2 years, cGVHD was extensive in 72% and limited in 28% of recipients with cGVHD. The cumulative incidence of cGVHD at 2 years post-transplant based on donor for MSD, MUD, haplo and MMUD was 47.5% (95% CI, 35.8–59.3%), 47.6% (95% CI, 37.9–57.4%), 33.9% (95% CI, 16.6–53.9%) and 49.1% (95% CI, 31.5–66.8%) respectively (Supplementary Table 3). Age, conditioning intensity and in vivo TCD had no significant effect on chronic GVHD. PTCy-based GVHD prophylaxis was associated with less chronic GVHD when compared to calcineurin based GVHD prophylaxis (Table 2). We also observed alloHCT performed before 2011 was associated with increased incidence of cGVHD than those performed after 2011. (Supplementary Table 6).

Relapse

The cumulative incidence of relapse/progression at 1 year and 4 years was 27.6% (95% CI, 22.3–33.2%) and 41.9% (95% CI, 35.5–48.4%). Based on the multivariate analyses (Table 2), age and conditioning intensity were not associated with rate of relapse. Stable or progressive disease at time of alloHCT was associated with increased incidence of relapse (HR 2.13; 95%CI 1.23–3.71; p=0.0072) when compared to CR. However, the depth of response at HCT (PR vs CR), in vivo TCD and TBI-based conditioning were not associated with the incidence of relapse.

Causes of death

The most common cause of death was relapse of the primary disease (52%), followed by infection (15%) and GVHD (13%). (Supplementary Table 5).

DISCUSSION

Using the CIBMTR database, we showed that long-term disease-free survival can be achieved in patients with T-PLL. We observed that RIC/NMA conditioning regimens are associated with reduced TRM and improved DFS and OS. Our analysis also found that the use of in vivo TCD strategies (ATG and/or alemtuzumab) resulted in an increased TRM and inferior DFS. Disease relapse continues to pose a challenge, with a 4-year relapse incidence of 41%. Patients with chemo-sensitive disease prior to transplant had a reduced incidence of relapse.

Data from this analysis are consistent with previous registry studies from the SFGM and the JSHCT (Table 3). The SFGM study retrospectively reported 3-year OS and DFS estimates at 36% and 26% in 27 patients with median follow-up of 33 months, while the JSHCT reported 3-year OS and PFS of 39.8% and 33.5% respectively in 20 patients with median follow-up of 51 months [9,14]. The EBMT study, a prospective observational study amongst recipients age 65 and younger with median follow-up of 50 months, reported 4-year OS and PFS of 42% and 30%, respectively[13]. However, in the EBMT series, the oldest patient was 59 years, whereas in this current CIBMTR study, 42% of patients were older than 60 years, which more closely reflects the median age of T-PLL diagnosis in the US.

Table 3.

Selected studies of alloHCT in T-PLL

Publication Study No. patients Remission status at alloHCT (N) Donor type Regimen intensity (N) Outcomes
Wiktor-Jedrzejczak et al. 13 EBMT 37 a CR=22
PR=10
Other=5
MRD=15
MUD=22
MAC=13
RIC=24
4 year OS: 42%
4 year NRM: 32%
4 year relapse: 38%
Kalaycio et al.10 CIBMTR 47 (21 T-PLL)b CR=16
PR=8
Other=21
MRD=11
MUD=19
Other: 13
MAC=19
NMA=14
1 year OS: 48%
1 year NRM: 28%
1 year relapse: 28%
Guillaume et al. 9 SFGM-TC 27 CR=14
PR=10
Other=3
MRD=10
MUD=17
MAC=10
NMA=17
3 year OS: 36%
3 year NRM: 31%
3 year relapse: 47%
Dholaria et al. 8 Moffitt
Cancer
Center
11 CR=9
PR=1
Other=1
MRD =5
MUD=3
Other=3
MAC=8
RIC=3
4 year OS: 56%
4 year NRM: 34%
4 year relapse: 21%
Yamasaki et al. 14 JSHCT 20 CR=6
PR=1
Other=13
MRD =5
MUD=6
Haplo=2
MMUD=7
UCB: 2
MAC=10
RIC=10
3 year OS: 39.8%
1 year NRM: 20.9%
3 year relapse: 69.6%
Murthy et al (current study) CIBMTR 266 CR=149
PR= 80
Other=37
MRD =80
MUD=115
Haplo=30
MMUD=33
Other=8
MAC=78
RIC=188
4 year OS: 30%
4 year TRM: 32.4%
4 year relapse: 41.9%

Abbreviations: B-PLL, B cell prolymphocytic leukemia; CIBMTR, Center for International Blood and Marrow Transplant Research; CR, complete remission; EBMT, European Society for Blood and Marrow Transplantation; haplo, haploidentical donor; HCT, hematopoietic cell transplantation; JSHCT, Japan Society for Hematopoietic Cell Transplantation (now known as the Japanese Society for Transplantation and Cellular Therapy; MRD, matched related donor; MMUD, mismatched unrelated donor; MUD, matched unrelated donor; NRM, nonrelapse mortality; OS, overall survival; PR, partial response; SFGM-TC, Francophone Society of Bone Marrow Transplantation and Cellular Therapy; T-PLL, T-cell prolymphocytic leukemia; UCB, umbilical cord blood.

a

Data available for 36 patients

b

B-PLL and T-PLL

The intensity of conditioning regimens across these three studies was comparable. RIC/NMA regimens were utilized in 70% patients in the current study, compared to 60% in SFGM, 50% in JSHCT and 65% in EBMT. RIC/NMA conditioning in younger patients was associated with reduced TRM and improved DFS and OS compared to younger patients receiving MAC conditioning. The survival benefit offered with RIC/NMA conditioning may be explained by graft-versus-leukemia (GVL) effect. A study by Sellner and colleagues evaluated a longitudinal quantitative minimal residual disease using clone-specific T-cell receptor (TCR)-based real-time quantitative polymerase chain reaction (PCR): They demonstrated minimal residual disease responses post-alloHCT were associated with a shift from a clonal, T-PLL-driven profile to a polyclonal signature, effectively validating GVL effect in T-PLL[22]. In our analysis, a surrogate marker of GVL, which is the impact of in vivo TCD on relapse, was not evident. The use of in vivo TCD was associated with inferior DFS due to increased risk of TRM.

High incidences of TRM have been reported in prior studies of alloHCT for T-PLL. The 4-year TRM of 32.4% is similar to reports by the EBMT (4-year NRM 32%) and SFGM (3-year TRM 31%). Predictably, we observed reduced TRM and reduced incidence of aGVHD with the use of RIC/NMA conditioning regimens. We observed that in vivo TCD was linked to increased TRM. In the current study, 18% of patients received in vivo TCD, mostly with ATG, compared to the EBMT study, in which 51% received TCD. AlloHCT with TCD has been associated with delayed immune reconstitution and increased risk of infection[2325]. Infection was reported as the second most common cause of death. Ongoing T-cell depletion caused by pre-transplant alemtuzumab therapy might influence TRM. Additionally, one could hypothesize that ongoing T cell depletion from pre-transplant alemtuzumab therapy, in addition to the use of RIC/NMA conditioning regimens and PTCy GVHD prophylaxis, could explain the low incidence of aGVHD and severe aGVHD observed. However, we could not answer this question conclusively in this analysis, because data for time from last alemtuzumab dose to transplant nor T cell reconstitution data were available.

Outcomes were by donor type were also reviewed (supplementary table 3). Although small in numbers, it is worth mentioning that we observed both haploidentical and mismatch unrelated transplants as feasible and effective in patients with T-PLL. Haploidentical transplants in particular we found to have less cGVHD and TRM w/comparable 4 year relapse, DFS and OS when comparing to MUD and MMUD transplants. It is important to note that donor type was not found to be significant on multivariate analyses and these findings are on univariate analysis only, so it is difficult to draw significant conclusions regarding choice of ideal donor. However, with increased utilization of haploidentical transplantation [26] and feasibility and effectiveness of PTCy in allo-HCT with MMUD [27], allo-HCT should be considered for patients with T-PLL even in the absence of a HLA matched donor.

Controlling disease and preventing relapse remain difficult in patients after alloHCT. Achieving complete remission prior to alloHCT was associated with less relapse, but only when compared to stable or progressive disease and not when compared to partial remission, suggesting that chemoresponsive disease prior to alloHCT is more significant than the depth of remission. Additionally, in this analysis, we investigated the role of total body irradiation. A prospective study by the EBMT identified TBI dose of 6 Gy or more as predictive of a reduced relapse risk in a univariable analysis[13]. We looked specifically whether adding TBI to both MAC and RIC would affect OS, DFS or TRM. When comparing MAC with TBI to MAC without TBI as well as RIC with TBI to RIC without TBI, we did not appreciate any significant effect on OS, DFS and TRM. Our analysis showed differences in survival outcomes with respect to pre-transplant conditioning were more attributed to comparing conditioning intensity (MAC vs RIC) rather than use of TBI

We found that relapse rates increased over time. Incidence of relapse increased, from 27.6% at 1 year, to 41.9% at 4 years. Unfortunately, there is no standard minimal residual disease test for T-PLL, and such a test potentially could help forecast early relapse. Late relapse may reflect waning GVL effect over time. Post-transplant immune modulation strategies may help prevent late relapse. Venetoclax [28], histone deacetylase (HDAC) inhibitors [29], p53 reactivators [30,31], and Janus kinase/signal transducers and activators of transcription (JAK/STAT) inhibitors [3235] have previously demonstrated some pre-clinical and/or clinical activity in T-PLL, and warrant further investigation for post-transplant maintenance.

This study has limitations inherent to a retrospective registry study. As this data was obtained from a transplant registry, we could not compare outcomes with patients who did not undergo alloHCT. Another limitation was the lack of pertinent pre-transplant information, such as cytogenetics, mutation data and details of therapies prior to alloHCT. Details of pre-HCT induction therapy were not available for most of our study participants, so we did not include this information in our analyses. The lack of consensus disease response criteria is a notable limitation. The CIBMTR registry defined T-PLL response criteria based on international consensus response criteria for chronic lymphocytic leukemia[16]. Only recently in 2019 were consensus T-PLL response guidelines were published[12]. Given that patients included in this analysis date back to 2008, utilizing the updated criteria was not feasible. Finally, detailed data were not available regarding the timing and severity of infections, as well as immune reconstitution.

CONCLUSION

In summary, alloHCT results in durable remissions and disease control in some patients with T-PLL. Relapse continues to remain a barrier to long-term survival. Reduced-intensity conditioning and avoidance of in vivo TCD are associated with improved outcomes. Molecular monitoring of patients for recurrence after transplant could be undertaken to identify early relapses for treatment and potentially donor lymphocyte therapy. Other novel approaches combined with alloHCT warrant investigation to further improve outcomes of alloHCT in T-PLL.

Supplementary Material

1
2

Supplementary Table 1: Full conditioning regimen list

Supplementary Table 2: Univariate analysis

Supplementary Table 3: Univariate analysis stratified by donor

Supplementary Table 4: Cumulative incidence of graft failure and GVHD

Supplementary Table 5: Causes of death

Supplementary Table 6: Full multivariate analysis

Supplementary Table 7: Multivariate analysis (Conditioning intensity +/− TBI)

Highlights.

  • Allogeneic HCT is effective in yielding durable remissions in patients with T-PLL

  • Myeloablative conditioning, age greater than 60 and KPS <90 were all associated with reduced OS

  • Reduced intensity conditioning and avoidance of in vivo T cell depletion correlated with better DFS and less TRM

  • TBI was not found to any significant effect on OS, DFS or TRM

ACKNOWLEDGEMENTS

The CIBMTR is supported primarily by Public Health Service U24CA076518 from the National Cancer Institute (NCI), the National Heart, Lung and Blood Institute (NHLBI) and the National Institute of Allergy and Infectious Diseases (NIAID); HHSH250201700006C from the Health Resources and Services Administration (HRSA); and N00014-20-1-2705 and N00014-20-1-2832 from the Office of Naval Research; Support is also provided by Be the Match Foundation, the Medical College of Wisconsin, the National Marrow Donor Program, and from the following commercial entities: AbbVie; Accenture; Actinium Pharmaceuticals, Inc.; Adaptive Biotechnologies Corporation; Adienne SA; Allovir, Inc.; Amgen Inc.; Astellas Pharma US Inc.; bluebird bio, inc.; Bristol Myers Squibb Co.; CareDx; CSL Behring; CytoSen Therapeutics, Inc.; Daiichi Sankyo Co., Ltd.; Eurofins Viracor; ExcellThera; Fate Therapeutics; Gamida-Cell, Ltd.; Genentech Inc.; Gilead; GlaxoSmithKline; Incyte Corporation; Janssen/Johnson & Johnson; Jasper Therapeutics; Jazz Pharmaceuticals, Inc.; Karyopharm Therapeutics; Kiadis Pharma; Kite, a Gilead Company; Kyowa Kirin; Legend; Magenta Therapeutics; Medac GmbH; Medexus; Merck & Co.; Millennium, the Takeda Oncology Co.; Miltenyi Biotec, Inc.; MorphoSys; Novartis Pharmaceuticals Corporation; Omeros Corporation; Oncopeptides, Inc.; Orca Biosystems, Inc.; Ossium Health, Inc.; Pfizer, Inc.; Pharmacyclics, LLC; Priothera; Sanofi Genzyme; Seagen, Inc.; Stemcyte; Takeda Pharmaceuticals; Tscan; Vertex; Vor Biopharma; Xenikos BV.

Footnotes

Conflict of Interest:

Dr. Awan reports personal fees from Genentech, personal fees from Astrazeneca, personal fees from Abbvie, personal fees from Janssen, personal fees from Pharmacyclics, personal fees from Gilead sciences, personal fees from Kite pharma, personal fees from Celgene, personal fees from Karyopharm, personal fees from MEI Pharma, personal fees from Verastem, personal fees from Incyte, personal fees from Beigene, personal fees from Johnson and Johnson, personal fees from Dava Oncology, personal fees from BMS, personal fees from Merck, personal fees from Cardinal Health, personal fees from ADCT therapeutics, outside the submitted work.

Dr. Dholaria reports and institutional research support from Takeda, Janssen, Angiocrine, Pfizer, Poseida.

Dr. Deol reports personal fees from Kite/Gilead, personal fees from Jannsen/Johnson& Johnson, personal fees from Navartis, outside the submitted work.

Dr. Grunwald reports personal fees from Abbvie, personal fees from Agios, personal fees from Amgen, personal fees from Cardinal Health, personal fees from BMS, personal fees from Daiichi Sankyo, personal fees and other from Incyte, personal fees from Merck, personal fees from Pfizer, personal fees from Premier, personal fees from Karius, other from Forma Therapeutics, other from Genentech/Roche, other from Janssen, personal fees from Astellas, personal fees from Trovagene, personal fees from Stemline, personal fees from Gilead, outside the submitted work.

Dr. Inamoto reports personal fees from Novartis, personal fees from Janssen, from Meiji Seika Pharma, outside the submitted work.

Dr. Munshi reports personal fees from Kite Pharma, personal fees from Incyte, outside the submitted work.

Dr. Oluwole reports personal fees from Gilead, personal fees from Pfizer, personal fees from Spectrum, personal fees from Bayer, personal fees from Curio Science, outside the submitted work.

Dr. Nishihori reports other from Novartis, other from Karyopharm, outside the submitted work.

Dr. Ortí reports personal fees from Bristol Myers Squibb, personal fees from Novartis, personal fees and non-financial support from Incyte, personal fees and non-financial support from Pfizer, outside the submitted work.

Dr. Seo reports personal fees from Janssen Pharmaceutical K.K., outside the submitted work.

Dr. Patel reports personal fees from Kite Pharma, outside the submitted work.

Dr. Rizzieri reports personal fees from Abbvie, personal fees from Agios, personal fees from AROG, personal fees from Bayer, personal fees from Celgene, personal fees and other from Celltrion/Teva, personal fees from Gilead, personal fees from Incyte, personal fees from Jazz, personal fees from Kadmon, personal fees from Kite, personal fees from Morphosys, personal fees from Mustang, personal fees from Novartis, personal fees from Pfizer, personal fees from Sanofi, personal fees from Seattle Genetics, personal fees and other from Stemline, personal fees from Amgen, personal fees from Acrobiotech, personal fees from UCART, personal fees from Chimerix, INC, personal fees from Pharmacyclics, outside the submitted work.

Dr. Sauter reports grants from Juno Therapeutics, grants from Celgene, grants from Bristol-Myers Squibb, grants from Precision Biosciences, personal fees from Precision Biosciences, grants from Sanofi Genzyme, personal fees from Sanofi Genzyme, personal fees from Juno Therapeutics, personal fees from Spectrum Pharmaceuticals, personal fees from Novartis, personal fees from Genmab, personal fees from Kite, a Gilead Company, personal fees from Celgene, personal fees from Gamida Cell, personal fees from Karyopharm Therapeutics, personal fees from GlaxoSmithKline, outside the submitted work.

Dr. Cerny reports personal fees from Jazz Pharmaceuticals Inc., personal fees from Daiichi-Sankyo Inc., personal fees from Pfizer Inc., personal fees from Amgen, personal fees from Allovir, outside the submitted work; and I own stocks of Actinium Pharmaceuticals, Bluebird Bio Inc., Dynavax Pharma, Atyr Pharmac, Gamida Cell, Miragen Therapeutics, Mustang Bio, Novavax, Ovid Therapeutics, Sorrento Therapeutics, TG Therapeutics, Vaxart Inc, and Veru Inc..

Dr. Hildebrandt reports other from Incyte, other from Jazz Pharmaceuticals, other from Incyte, other from Morphosys, other from Alexion Pharmaceuticals, other from Karyopharm Therapeutics, other from Takeda, other from Jazz Pharmaceuticals, other from Pharmacyclics, other from Incyte, other from AstraZeneca, other from Jazz Pharmaceuticals, other from Astellas Parma, other from Incyte, other from Falk Foundation, other from Takeda, outside the submitted work.

Dr. Bal reports grants from Amyloidosis Foundation, outside the submitted work.

Dr. Ustun reports other from Novartis, other from Blueprint, outside the submitted work.

Dr. Foss reports in addition has a patent Photopheresis for modulation of dendritic cells issued.

DATA USE STATEMENT

CIBMTR supports accessibility of research in accord with the National Institutes of Health (NIH) Data Sharing Policy and the National Cancer Institute (NCI) Cancer Moonshot Public Access and Data Sharing Policy. The CIBMTR only releases de-identified datasets that comply with all relevant global regulations regarding privacy and confidentiality.

Publisher's Disclaimer: This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

BIBLIOGRAPHY

  • 1.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. May 19;127:2375–2390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sud A, Dearden C: T-cell Prolymphocytic Leukemia. Hematol Oncol Clin North Am 2017;31:273–283. [DOI] [PubMed] [Google Scholar]
  • 3.Jain P, Aoki E, Keating M, Wierda WG, O’Brien S, Gonzalez GN, et al. : Characteristics, outcomes, prognostic factors and treatment of patients with T-cell prolymphocytic leukemia (T-PLL). Ann Oncol 2017. Jul 1;28:1554–1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Dearden CE, Khot A, Else M, Hamblin M, Grand E, Roy A, et al. : Alemtuzumab therapy in T-cell prolymphocytic leukemia: comparing efficacy in a series treated intravenously and a study piloting the subcutaneous route. Blood 2011. Nov 24;118:5799–5802. [DOI] [PubMed] [Google Scholar]
  • 5.Dearden C: Management of prolymphocytic leukemia. Hematology Am Soc Hematol Educ Program 2015;2015:361–367. [DOI] [PubMed] [Google Scholar]
  • 6.Dearden C: How I treat prolymphocytic leukemia. Blood 2012. Jul 19;120:538–551. [DOI] [PubMed] [Google Scholar]
  • 7.Wiktor-Jedrzejczak W, Dearden C, de Wreede L, van Biezen A, Brinch L, Leblond V, et al. : Hematopoietic stem cell transplantation in T-prolymphocytic leukemia: a retrospective study from the European Group for Blood and Marrow Transplantation and the Royal Marsden Consortium. Leukemia 2012. May;26:972–976. [DOI] [PubMed] [Google Scholar]
  • 8.Dholaria BR, Ayala E, Sokol L, Nishihori T, Chavez JC, Hussaini M, et al. : Allogeneic hematopoietic cell transplantation in T-cell prolymphocytic leukemia: A single-center experience. Leuk Res 2018. Apr;67:1–5. [DOI] [PubMed] [Google Scholar]
  • 9.Guillaume T, Beguin Y, Tabrizi R, Nguyen S, Blaise D, Deconinck E, et al. : Allogeneic hematopoietic stem cell transplantation for T-prolymphocytic leukemia: a report from the French society for stem cell transplantation (SFGM-TC). Eur J Haematol 2015. Mar;94:265–269. [DOI] [PubMed] [Google Scholar]
  • 10.Kalaycio ME, Kukreja M, Woolfrey AE, Szer J, Cortes J, Maziarz RT, et al. : Allogeneic hematopoietic cell transplant for prolymphocytic leukemia. Biol Blood Marrow Transplant 2010. Apr;16:543–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Krishnan B, Else M, Tjonnfjord GE, Cazin B, Carney D, Carter J, et al. : Stem cell transplantation after alemtuzumab in T-cell prolymphocytic leukaemia results in longer survival than after alemtuzumab alone: a multicentre retrospective study. Br J Haematol 2010. Jun;149:907–910. [DOI] [PubMed] [Google Scholar]
  • 12.Staber PB, Herling M, Bellido M, Jacobsen ED, Davids MS, Kadia TM, et al. : Consensus criteria for diagnosis, staging, and treatment response assessment of T-cell prolymphocytic leukemia. Blood 2019. Oct 3;134:1132–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wiktor-Jedrzejczak W, Drozd-Sokolowska J, Eikema DJ, Hoek J, Potter M, Wulf G, et al. : EBMT prospective observational study on allogeneic hematopoietic stem cell transplantation in T-prolymphocytic leukemia (T-PLL). Bone Marrow Transplant 2019. Jan 21;54:1391–1398. [DOI] [PubMed] [Google Scholar]
  • 14.Yamasaki S, Nitta H, Kondo E, Uchida N, Miyazaki T, Ishiyama K, et al. : Effect of allogeneic hematopoietic cell transplantation for patients with T-prolymphocytic leukemia: a retrospective study from the Adult Lymphoma Working Group of the Japan Society for hematopoietic cell transplantation. Ann Hematol 2019. Sep;98:2213–2220. [DOI] [PubMed] [Google Scholar]
  • 15.Collignon A, Wanquet A, Maitre E, Cornet E, Troussard X, Aurran-Schleinitz T: Prolymphocytic leukemia: new insights in diagnosis and in treatment. Curr Oncol Rep 2017. Apr;19:29. [DOI] [PubMed] [Google Scholar]
  • 16.Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Döhner H, et al. : Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood 2008. Jun 15;111:5446–5456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bacigalupo A, Ballen K, Rizzo D, Giralt S, Lazarus H, Ho V, et al. : Defining the intensity of conditioning regimens: working definitions. Biol Blood Marrow Transplant 2009. Dec;15:1628–1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Copelan E, Casper JT, Carter SL, van Burik J-AH, Hurd D, Mendizabal AM, et al. : A scheme for defining cause of death and its application in the T cell depletion trial. Biol Blood Marrow Transplant 2007. Dec;13:1469–1476. [DOI] [PubMed] [Google Scholar]
  • 19.Commenges D, Andersen PK: Score test of homogeneity for survival data. Lifetime Data Anal 1995;1:145–56; discussion 157. [DOI] [PubMed] [Google Scholar]
  • 20.Zhang X, Loberiza FR, Klein JP, Zhang M-J: A SAS macro for estimation of direct adjusted survival curves based on a stratified Cox regression model. Comput Methods Programs Biomed 2007. Nov;88:95–101. [DOI] [PubMed] [Google Scholar]
  • 21.Zhang X, Zhang M-J: SAS macros for estimation of direct adjusted cumulative incidence curves under proportional subdistribution hazards models. Comput Methods Programs Biomed 2011. Jan;101:87–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sellner L, Brüggemann M, Schlitt M, Knecht H, Herrmann D, Reigl T, et al. : GvL effects in T-prolymphocytic leukemia: evidence from MRD kinetics and TCR repertoire analyses. Bone Marrow Transplant 2017. Apr;52:544–551. [DOI] [PubMed] [Google Scholar]
  • 23.Soiffer RJ, Kim HT, McGuirk J, Horwitz ME, Johnston L, Patnaik MM, et al. : Prospective, Randomized, Double-Blind, Phase III Clinical Trial of Anti-T-Lymphocyte Globulin to Assess Impact on Chronic Graft-Versus-Host Disease-Free Survival in Patients Undergoing HLA-Matched Unrelated Myeloablative Hematopoietic Cell Transplantation. J Clin Oncol 2017. Dec 20;35:4003–4011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Walker I, Panzarella T, Couban S, Couture F, Devins G, Elemary M, et al. : Pretreatment with anti-thymocyte globulin versus no anti-thymocyte globulin in patients with haematological malignancies undergoing haemopoietic cell transplantation from unrelated donors: a randomised, controlled, open-label, phase 3, multicentre trial. Lancet Oncol 2016. Feb;17:164–173. [DOI] [PubMed] [Google Scholar]
  • 25.Bacigalupo A, Lamparelli T, Bruzzi P, Guidi S, Alessandrino PE, di Bartolomeo P, et al. : Antithymocyte globulin for graft-versus-host disease prophylaxis in transplants from unrelated donors: 2 randomized studies from Gruppo Italiano Trapianti Midollo Osseo (GITMO). Blood 2001. Nov 15;98:2942–2947. [DOI] [PubMed] [Google Scholar]
  • 26.D’Souza A, Fretham C, Lee SJ, Arora M, Brunner J, Chhabra S, et al. : Current use of and trends in hematopoietic cell transplantation in the united states. Biol Blood Marrow Transplant 2020. Aug;26:e177–e182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Shaw BE, Jimenez-Jimenez AM, Burns LJ, Logan BR, Khimani F, Shaffer BC, et al. : National Marrow Donor Program-Sponsored Multicenter, Phase II Trial of HLA-Mismatched Unrelated Donor Bone Marrow Transplantation Using Post-Transplant Cyclophosphamide. J Clin Oncol 2021. Apr 27;:JCO2003502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boidol B, Kornauth C, van der Kouwe E, Prutsch N, Kazianka L, Gültekin S, et al. : First-in-human response of BCL-2 inhibitor venetoclax in T-cell prolymphocytic leukemia. Blood 2017. Dec 7;130:2499–2503. [DOI] [PubMed] [Google Scholar]
  • 29.Hasanali ZS, Saroya BS, Stuart A, Shimko S, Evans J, Vinod Shah M, et al. : Epigenetic therapy overcomes treatment resistance in T cell prolymphocytic leukemia. Sci Transl Med 2015. Jun 24;7:293ra102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Andersson EI, Pützer S, Yadav B, Dufva O, Khan S, He L, et al. : Discovery of novel drug sensitivities in T-PLL by high-throughput ex vivo drug testing and mutation profiling. Leukemia 2018;32:774–787. [DOI] [PubMed] [Google Scholar]
  • 31.Schrader A, Braun T, Herling M: The dawn of a new era in treating T-PLL. Oncotarget 2019. Jan 18;10:626–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wahnschaffe L, Braun T, Timonen S, Giri AK, Schrader A, Wagle P, et al. : JAK/STAT-Activating Genomic Alterations Are a Hallmark of T-PLL. Cancers (Basel) 2019. Nov 21;11. DOI: 10.3390/cancers11121833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gomez-Arteaga A, Margolskee E, Wei MT, van Besien K, Inghirami G, Horwitz S: Combined use of tofacitinib (pan-JAK inhibitor) and ruxolitinib (a JAK1/2 inhibitor) for refractory T-cell prolymphocytic leukemia (T-PLL) with a JAK3 mutation. Leuk Lymphoma 2019. Apr 18;60:1626–1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wei M, Koshy N, van Besien K, Inghirami G, Horwitz SM: Refractory T-Cell Prolymphocytic Leukemia with JAK3 Mutation: In Vitro and Clinical Synergy of Tofacitinib and Ruxolitinib. Blood 2015. Dec 3;126:5486–5486. [Google Scholar]
  • 35.Orlova A, Wagner C, de Araujo ED, Bajusz D, Neubauer HA, Herling M, et al. : Direct targeting options for STAT3 and STAT5 in cancer. Cancers (Basel) 2019. Dec 3;11. DOI: 10.3390/cancers11121930 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

Supplementary Table 1: Full conditioning regimen list

Supplementary Table 2: Univariate analysis

Supplementary Table 3: Univariate analysis stratified by donor

Supplementary Table 4: Cumulative incidence of graft failure and GVHD

Supplementary Table 5: Causes of death

Supplementary Table 6: Full multivariate analysis

Supplementary Table 7: Multivariate analysis (Conditioning intensity +/− TBI)

RESOURCES