Feasibility of Cyclosporine and Short-Term Methotrexate for Graft-Vs.-Host disease Prophylaxis in T-Cell-Replete, Reduced-Intensity Allogeneic Bone Marrow Transplantation From Unrelated Donors.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4347-4347
Author(s):  
Takahiko Nakane ◽  
Hirohisa Nakamae ◽  
Hideo Koh ◽  
Mika Nakamae ◽  
Yoshiki Hayashi ◽  
...  

Abstract Abstract 4347 Optimal graft-vs.-host disease (GVHD) prophylaxis in T-cell-replete, reduced-intensity allogeneic stem cell transplantation from an unrelated donor (u-RIST) has not been established yet. Tacrolimus-based acute GVHD prophylaxis has been widely used in allogeneic stem cell transplantation from unrelated donors. In addition, T-cell-depletion conditioning regimen containing anti-thymocyte globulin (ATG), or alemtuzumab with/without 2-4 Gy total body irradiation (TBI) has been reportedly applied in u-RIST. However, in-vivo T cell depletion using ATG or alemtuzumab may cause increased risk of infection or relapse after transplantation. TBI may also possibly cause increased incidence of GVHD via tissue damage. Here we report the feasibility of using cyclosporine and short-term methotrexate for GVHD prophylaxis in u-RIST. The conditioning regimen consisted of fludarabine and busulfan (Flu+Bu) without T-cell depletion, such as ATG or alemtuzumab, and without TBI for u-RIST in patients who received prior chemotherapy. In this study, we retrospectively analyzed 30 consecutive patients with hematologic malignancies (median age: 53; range: 22 to 69 years; AML: 11; ALL: 9; MDS: 2; NHL: 5; ATL/L: 3) who received unrelated-donor bone marrow transplantation from HLA 6/6 (A, B, DRB1), either allele-matched (n = 24 patients), one DRB1 allele-mismatched (n = 5 patients), or one A allele-mismatched (n=1) donor at our institution between September 2002 and June 2009. All patients received at least one cycle of prior chemotherapy (median cycle: 6; range: 1 to 19). Of these, 15 patients (50%) were at high risk of relapse. For all, acute GVHD prophylaxis consisted of cyclosporine and short-term methotrexate (day 1: 10mg/m2; day 3 and 5: 7 mg/m2). The reduced intensity conditioning consisted of fludarabine 180mg/m2 and oral busulfan 8 mg/kg for 12 patients; the other 18 patients' conditioning consisted of fludarabine 180 mg/m2 and intravenous busulfan 8mg/kg. Neutrophil and platelet engraftments were achieved in 100% (30/30) and 93% (28/30) of patients, respectively. The median times for neutrophil and platelet engraftments were 16 days (range: 12 to 26 days) and 24.5 days (range: 18 to 291 days), respectively. In patients receiving oral and intravenous busulfan for conditioning, the achievement rates of complete T-cell chimerism by day 100 were 75% (9/12) and 100% (18/18), respectively (p = 0.03). The median period of follow-up was 280 days (range: 34∼1987). During the follow-up our observational periods, 19 patients were alive and 11 patients died of relapse (n = 8) or transplant-related mortality (n = 3). The cumulative incidences of acute GVHD of grade II to IV and III to IV at day 100 were 40% and 17%, respectively. Extensive type of chronic GVHD was 30% in 23 evaluable patients. The 2-year overall survival (OS) and event-free survival rates were 58% (low/standard risk group: 78%; high risk group: 43%; p = 0.04) and 52% (low/standard risk group: 72%; high risk group: 33%; p = 0.01), respectively. In multivariate analysis, grade III to IV acute GVHD and disease status at high relapse were significantly associated with worse OS (p = 0.03 and 0.008, respectively). Day 100 and 2-year TRM were 3% and 10%, respectively. Cyclosporine and short-term methotrexate for GVHD prophylaxis without T-cell deplete-conditioning allowed acceptable incidence of GVHD even in u-RIST. Additionally, only Flu+Bu conditioning for uRIST ensured high rate of complete T-cell chimerism especially in patients receiving intravenous busulfan. Our results suggested that we might be required to determine optimal conditioning and GVHD prophylaxis regimens based on patient characteristics, including a history of prior chemotherapy. Disclosures: Hino: Kyowa Hakko Kirin Co., Ltd. : Research Funding.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3077-3077
Author(s):  
Kyoo-Hyung Lee ◽  
Seong-Jun Choi ◽  
Jung-Hee Lee ◽  
Ho-Jin Shin ◽  
Young-Shin Lee ◽  
...  

Abstract Animal hematopoietic cell transplantation (HCT) models and several small clinical trials showed that successful engraftment can be achieved across HLA-haplotype difference after reduced-intensity conditioning (RIC). Furthermore, decreased graft-versus-host disease (GVHD) and transplantation-related mortality (TRM) after RIC was shown in a swine leukocyte antigen-haploidentical HCT experiment. Therefore, a protocol investigating the role of RIC in HLA-haploidentical familial donor HCT was initiated in April 2004 and 20 patients [13 male and 7 female; median age 26.5 years (16–65)] without HLA-matched donor enrolled until June 2007. The diagnosis were AML (n=9), ALL (n=4), acute biphenotypic leukemia (n=1), MDS (n=4), and SAA (n=2), and all patients had high-risk features, i.e. first complete remission (CR) but with high-risk chromosomal abnormality (n=1), first CR after salvage (n=1), second CR (n=6), recurrent/refractory state (n=7), immunotherapy failure (n=4), and high-risk MDS (RAEB-1, n=1). The RIC included iv busulfan 3.2 mg/kg × 2, fludarabine 30 mg/m2 × 6, plus anti-thymocyte globulin [Thymoglobuline 3 mg/kg (n=17) or Lymphoglobuline 15 mg/kg (n=3)] × 4. After receiving G-CSF, the donors (13 mothers; 5 offsprings; and 2 HLA-haploidentical siblings) underwent 2 or 3 daily leukapheresis, and the collected cells were given to patients without T cell depletion [medians of; 7.9 (3.7–12.1)×108/kg MNC, 6.9 (3.6–73.5)×106/kg CD34+ cells, and 4.6 (1.8–8.5)×108/kg CD3+ cells]. GVHD prophylaxis was cyclosporine 3 mg/kg/day iv from day -1 and a short course of methotrexate. As a part of separate phase 1 study, the two most-recently enrolled patients received additional donor CD34+ cell-derived NK cells 6 weeks after HCT. Except one patients with SAA who died due to K. pneumoniae sepsis on day 18, all 19 evaluable patients engrafted with ANC> 500/μl median 17 days (12–53) and platelet> 20,000/μl median 23 days (12–100) after HCT. Eight patients experienced acute GVHD (grades I, II, III, and IV; 2, 3, 2, and 1, respectively). Cumulative incidences (CI) of overall and grade II-IV acute GVHD were 40 and 30%, respectively. Eight patients experienced chronic GVHD (limited, 4; extensive, 4; CI, 51%). Fourteen showed positive CMV antigenemia, while 2 suffered CMV colitis, which resolved after treatment. As early as 2 weeks after HCT, 15 of 16 evaluable patients, and, by 4 weeks, all of 17 evaluable patients showed donor chimerism ≥95% on STR-PCR, which was maintained until 24 weeks in all 11 patients tested. Thirteen patients are alive after median follow-up of 13.6 months (1.5–37.9; Kaplan-Meier survival, 55.6%). Of 16 patients with acute leukemia and high-risk MDS, 8 remain alive without recurrence (event-free-survival, 40.9%). Two patients died of K. pneumoniae sepsis and grade IV acute GVHD, respectively (CI of TRM, 11%). Immune recovery in 10 patients without relapse for > 6 months showed robust lymphocyte contents and immunoglobulin levels at 6 months (means of; 1,060/ul CD3+, 222/ul CD4+, 767/ul CD8+ cells, and 1,317 mg/dl IgG) and 12 months. After RIC, consistent engraftment and durable complete donor hematopoietic chimerism can be achieved from HLA-haploidentical familial donor. The frequencies of GVHD and TRM were low.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1206-1206
Author(s):  
Olivia Laverick ◽  
Amy Publicover ◽  
Laura Jardine ◽  
Kile Green ◽  
Alan Potter ◽  
...  

Abstract Many variables influence the risk of graft versus host disease following hematopoietic stem cell transplantation. Comparison between preparative regimens is hampered by the use of many different combinations of chemotherapy and radiotherapy, varying intensity of conditioning, use of T cell depletion and donors who are either siblings or unrelated volunteers. Many reduced intensity regimens also incorporate enhanced GVHD prophylaxis with in vivo T cell depletion. Here we describe a cohort of patients prepared in a modular fashion with either reduced or full intensity conditioning combined with a uniform GVHD prophylaxis regimen for all transplants with sibling donors (alemtuzumab 30mg) and for all with unrelated donors (UD; alemtuzumab 60mg). Thus it was possible to dissect independently the effect of conditioning intensity and sibling or UD type upon GVHD risk in this settig of in vivo T cell depletion. Patients and analysis: the study was a retrospective analysis of 258 sequential transplants performed in adults with hematological malignancy between September 2005 and September 2013 at a single UK institution. Reduced intensity conditioning (n = 221) included fludarabine 150mg/m2 plus melphalan 140mg/m2 or fludarabine 150mg/m2 plus busulfan 9.6mg/kg. Full intensity transplants (n = 37) received 12Gy TBI plus melphalan 140mg/m2, 12Gy TBI plus cyclophosphamide 120mg/kg, or busulfan 16mg/kg plus cyclophosphamide 120mg/kg. All patients with sibling donors received 30mg alemtuzumab and those with UD received a 60mg of alemtuzumab. UD matching was similar in both reduced intensity and full intensity cohorts (92.2% and 86.5% 10/10 matches, respectively) but patients receiving reduced intensity were older than those receiving full intensity conditioning (median age 51 vs 31; p < 0.001). Outcome was analyzed according to EBMT guidelines. Relapse, non-relapse mortality and cGVHD were treated as competing risks and analysed as cumulative incidence. Outcome: the incidence of acute GVHD grades I-IV was comparable between reduced intensity and full intensity sibling transplants (45% vs 45%; p = NS) indicating a lack of effect of conditioning intensity upon GVHD risk in this setting. There was a slight increase in the risk of GVHD between reduced intensity UD compared with reduced intensity sibling donor transplants (57% vs 45%; p = NS) but a marked synergistic increase between UD transplants performed with full intensity compared with reduced intensity conditioning (100% vs 57%; p = < 0.001). The incidence of grades III-IV acute GVHD was also higher in full intensity UD transplants (16%) compared with reduced intensity UD transplants (5%). The incidence of chronic GVHD was also highest in full intensity UD transplants but both conditioning intensity and UD contributed in an additive manner: the rate of chronic GVHD progressed from 33% to 44% in reduced intensity and full intensity sibling transplants respectively and from 57% to 75% for reduced and full intensity UD transplants, respectively. Two year overall survival was comparable in all groups, ranging from 55% to 70%. In keeping with the higher rates of acute GVHD in full intensity transplants performed with UD, this group experienced the lowest relapse risk (15% vs 29% for all the other groups combined; p = 0.04) but the highest non-relapse mortality, reaching 41% at 2 years compared with 28% for all the other groups combined (p = 0.08). Conclusion: these results show that alemtuzumab provides good protection from acute GVHD in reduced intensity transplantation from sibling and UD. In sibling transplants given identical GVHD prophylaxis, full intensity conditioning does not increase the risk of GVHD. In contrast, a slight increase in GVHD risk with UD transplants seen with reduced intensity conditioning, is amplified in a synergistic manner by full intensity conditioning. This is associated with a high non-relapse mortality, even though the median age of full intensity patients is more than 20 years younger than those receiving reduced intensity conditioning. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 547-547 ◽  
Author(s):  
John Koreth ◽  
Kwang Woo Ahn ◽  
Joseph Pidala ◽  
James L. Gajewski ◽  
Hailin Wang ◽  
...  

Abstract In myeloablative unrelated donor allogeneic hematopoietic cell transplantation (HCT) a 1-locus HLA-mismatch (-A, -B, -C, -DRB1) is associated with lower survival compared to fully matched pairs. However data in reduced-intensity and non-myeloablative conditioning (together called RIC) HCT are limited. We analyzed adult AML/ALL/CML/MDS recipients of first 8/8 HLA-matched or 1-locus mismatched unrelated donor (MUD, MMUD) RIC HCT performed in the period 1999-2011 and registered in the CIBMTR. HLA-A, -B, -C and -DRB1 loci were typed in all pairs at high resolution; -DQB1 and -DPB1 loci could not be evaluated in all pairs. Transplants involving ex-vivo T-cell depletion, CD34+ selection, or post-transplant cyclophosphamide were excluded. Overall survival (OS) was the primary outcome. Secondary outcomes included non-relapse mortality (NRM), relapse, disease-free survival (DFS) and acute and chronic GVHD. Individual locus mismatch was also assessed. Apart from HLA matching, variables related to patient (age, race, sex, KPS, diagnosis, disease-risk), donor (age, parity), both (sex match/ABO match/CMV match) treatment (conditioning intensity, TBI use, in-vivo T-cell depletion (ATG), graft source (PB, BM) and GVHD prophylaxis (CyA-, Tac-based)) were considered. 2588 RIC HCT (8/8 MUD: 2025; 7/8 MMUD: 563) from 144 centers and 12 countries were analyzed. Median follow up in 8/8 MUD and 7/8 MMUD was 38 and 48 months respectively. Diagnoses were AML (65%), ALL (8%), CML (7%), MDS (20%). Conditioning intensity was RIC (79%), NMA (21%). 58% received in-vivo T-cell depletion. Graft source was PBSC (85%), BM (15%). GVHD prophylaxis was Tac-based (70%), CyA-based (27%). Mismatches involved HLA-A (188), -B (81), -C (219), and -DRB1 (75); with -DPB1 and -DQB1 typing available in 1382 and 2502 cases respectively. Compared to 8/8 MUD, 7/8 MMUD recipients were more likely to be younger and ethnic minorities and to have older and parous donors. In univariate analyses DQB1- and -DPB1 mismatch was not associated with worse OS, DFS, or NRM and was not further evaluated. There was a trend toward more grade II-IV acute GVHD in -DPB1 double (p=0.02) but not single mismatches. In multivariate models 7/8 MMUD RIC HCT had worse grade II-IV and III-IV acute GVHD, NRM, DFS and OS, but not relapse or chronic GVHD (Table). No significant interactions were identified between degree of HLA matching and other clinical variables. Adjusted 1- and 3-year NRM for 8/8 MUD vs. 7/8 MMUD was 20.4% vs. 28.9% (p<0.0001) and 29.2% vs. 38.1% (p<0.0007) respectively. Adjusted 1- and 3-year OS was 54.7% vs. 48.8% (p=0.01) and 37.4% vs. 30.9% (p=0.005) respectively (Figure). There was no difference between allele and antigen mismatches. HLA-A, -B, -C, and -DRB1 locus mismatches were each associated with 1 or more impaired outcomes (acute GVHD, NRM, DFS, and/or OS). Table 1 7/8 vs. 8/8 HLA HR (95% CI) p-value Acute GVHD II-IV 1.29 (1.09-1.53) 0.003 Acute GVHD III-IV 1.69 (1.00-3.36) 0.05 Chronic GVHD 1.11 (0.96-1.28) 0.15 Relapse 1.01 (0.87-1.17) 0.92 NRM 1.52 (1.29-1.79) <0.0001 DFS 1.20 (1.07-1.34) 0.0015 OS 1.25 (1.11-1.40) 0.0001 Compared to 8/8 MUD, both 7/8 allele and antigen MMUD RIC HCT have greater treatment toxicity and worse survival, of a magnitude similar to that seen in myeloablative transplantation. An isolated mismatch at HLA-A, -B, -C, or -DRB1 was associated with 1 or more adverse outcomes. In unrelated donor RIC HCT, matching for all alleles of HLA-A, -B, -C and -DRB1 loci results in superior outcomes. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5759-5759
Author(s):  
Feras Alfraih ◽  
Amjad Alhussaini ◽  
Farhan Anjum ◽  
Ghuzayel Mubarak Aldawsari ◽  
Fahad Alsharif ◽  
...  

Abstract Introduction : Hemorrhagic cystitis (HC) is one of common complications after allogeneic hematopoietic SCT (HSCT) with reported incidence varying from 7 to 70%. Several reports have shown that BK is strongly associated with HC (BK-HC) following HSCT. We conducted an institutional retrospective study to analyze the incidence and clinical factors associated with BK-HC following HSCT. Methods : A total of 517 consecutive patients above the age of 14 years receiving HSCT from 2009 and June 2015 were included in this retrospective analysis and evaluated for HC and urinary BK. HC was defined as documented hematuria of any grade and BK viruria was defined as positive at any level by BKV quantitative PCR testing in urine. Patients were stratified, based on hematuria and urinary BK virus, into the following groups (a) BK virus positive hemorrhagic cystitis (BK+HC), (b) BK virus negative hemorrhagic cystitis (BK-HC) and (c) Non-hemorrhagic cystitis (HC-). Screening for microscopic hematuria was performed only for patients with any kind of urinary symptoms. Results: 479 patients (92.6%) were matched related donor and 308 (60%) were male with a median age of 24 (range 14 to 66). Diagnoses were AML for 195 (38%), ALL for 183 (35%) and bone marrow failure for 44 (8.5%). Conditioning regimen was cyclophosphamide based in 427 (82.6%) patients (97%) versus others in 90 (17.4%) patients. GVHD prophylaxis was CSA/MTX for 456 (88.4%) however, T cell depletion was used in 13%. Peripheral blood stem cells were used for 56% of patients. With a median follow-up of 60 months for survivors (range 2 to 116.5 months), 43 (8%) patients showed BK+HC, 264 (51%) BK- HC while 209 (41%) did not have any hematuria (HC- group). Median time from transplantation to BK+ HC was 67 days (range 7 to 1261 days). Univariate analysis for risk factors of BK+ HC showed male, use of T-cell depletion and AML diagnosis were statistically significant factors. Other factors like age, conditioning regimen, GVHD prophylaxis, stem cell source, mismatched and remission status were not statistically significant. BK+ HC group was associated with higher incidence of other infections like CMV viremia (p=0.01) and fungal infection (p<0.01). Incidence of acute GVHD was 62.8% in BK+ HC group, 43% in HC- BK and 33.3% in HC- group (p=<0.01), suggesting higher incidence of acute GVHD in BK+ HC group. There was no difference in incidence of chronic GVHD. Conclusion: Hematuria following allogeneic bone marrow transplantation occurs in almost half of patients (51%) while BK associated HC develops in 8% of patients. Factors associated with BK+ HC were male gender, use of T-cell depletion and AML diagnosis. BK+HC is usually associated with other infections like CMV viremia and fungal infections. Further studies are needed to minimize or prevent BK+ HC following HSCT especially in high risk group. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3362-3362
Author(s):  
ZiYi Lim ◽  
Sarah Bennett ◽  
Aloysius YL Ho ◽  
Austin G Kulasekararaj ◽  
Sunil Gupta ◽  
...  

Abstract Abstract 3362 Poster Board III-250 Several groups have used in-vivo T-cell depletion in conjunction with RIC HSCT protocols in an attempt to improve engraftment and reduce the incidence of graft-versus-host disease. However, the potential benefits of this approach are potentially offset by an attenuation of the graft-versus-leukaemia effect. While, anti-thymocyte globulin(ATG) and alemtuzumab are commonly used in RIC HSCT protocols, there is limited data available on the comparative effects of either form of T-cell depletion on clinical outcomes. We performed a retrospective analysis on 65 patients with high risk AML/MDS who underwent a HLA-matched sibling donor RIC HSCT receiving either ATG or alemtuzumab. All patients received fludarabine (30mg/m2 x 5 days intravenously), and either oral busulphan (4mg/kg x 2 days) or IV busulphan (3.2mg/kg x 2 days). From Jan 2002-Jun 2005, 41 patients received GvHD prophylaxis with alemtuzumab (20mg x 5 days intravenously) followed by cyclosporin A post-transplant. From Jun 2005 onwards, 21 patients received GvHD prophylaxis with ATG (Thymoglobulin, Genzyme) (total 6mg/kg over 3 days intravenously) followed by methotrexate and cyclosporine A post-transplant. All patients had either MDS RAEB I/II (n=26), or de novo/secondary AML (n=39). The median recipient age was 57 years (range:39–65) for ATG and 56.0yrs (39-72) for alemtuzumab. All ATG patients received PBSC with a median CD34 cell dose of 5.15×106/kg (2.04 – 9.23). 37/41 alemtuzumab patients received PSBC with median CD34 cell dose of 5.10×106/kg (1.86 – 13.9). Both groups were matched for donor/recipient age, disease type, cytogenetic risk, disease stage at transplantation, cell dose. Median follow-up (for survivors) was 721 days (range: 187–1138) for ATG group and 2082 days (1094-3233) for the alemtuzumab group. Median time to neutrophil (>0.5×10 9/kg) and platelet (>20×10 9/kg) regeneration was 13 days and 14 days respectively, with no significant difference between both arms. There was 1 case of primary graft failure with ATG but none in patients receiving alemtuzumab. Lymphoid (CD3) engraftment was significantly slower in the alemtuzumab arm, with only 41% vs 75% patients attaining full donor chimerism(>95% donor) at day 100. There was no significant difference in the incidence of grade II-IV acute GvHD (ATG: 15%, Alemtuzumab 9%). However, patients who received ATG had a significantly higher incidence of chronic extensive GvHD (34% vs 6%, p=0.03). In addition, a significantly larger proportion of patients receiving alemtuzumab required subsequent DLI therapy (68% vs 19%). On univariate analysis, there was no significant difference in 2-year OS, and TRM between the alemtuzumab vs ATG group (56.1%+/-8% vs 73.7%+/-10%, p=0.25), and (19.5%+/-7% vs 10.6% +/- 7%, p=0.43). However, the use of ATG was associated with a significantly lower 2-year relapse incidence (28.4%+/-15% vs 51.5%+/-8%, p=0.04), and superior DFS (63.6%+/-14% vs 39.0%+/-8%, p=0.03). In summary, our experience indicates that in patients with high risk MDS or AML undergoing a sibling donor RIC HSCT, use of ATG confers a higher incidence of extensive chronic GvHD with a consequently lower relapse incidence and more favourable DFS when compared with alemtuzumab. In the setting of sibling RIC HSCT, the use of ATG for T-cell depletion may thus be more suitable for patients with more aggressive disease. In contrast the use of alemtuzumab at the dose reported may be more suitable for treatment of patients with stable/lower-risk disease, or for the treatment of non-haematological malignancies. Further studies with regards to dose titration of both ATG and alemtuzumab in specific disease settings are warranted. Disclosures: Off Label Use: fludarabine, alemtuzumab- used off-label for transplant conditioning. Marsh:Genzyme: Consultancy, Honoraria.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4594-4594
Author(s):  
Maria Queralt Salas ◽  
Wilson Lam ◽  
Arjun Law ◽  
Fotios V. Michelis ◽  
Dennis Dong Hwan Kim ◽  
...  

Introduction The combination of anti-thymoglobulin (ATG), post-transplant cyclophosphamide (PTCy) and cyclosporine (CsA) provides an effective control of graft-versus host disease (GVHD) in allo-HSCT using peripheral blood stem cell (PBSC) grafts, as has been reported by Dr. Viswabandya et al. We aim to report a large, single center experience in reduced intensity conditioning (RIC) allo-HSCT using dual T-cell depletion with ATG and PTCy combined with CsA for GVHD prophylaxis using grafts from 10/10 matched unrelated donors. Patients and methods Between October 2015 and April 2019, 167 adult patients diagnosed with hematological malignancies underwent first 10/10 MUD RIC allo-HSCT. RIC regimen was composed by fludarabine, busulfan, and 200cGy of total body irradiation. For GVHD prophylaxis all recipients received rabbit-ATG, PTCy 50mg/m2/day on day +3 and +4, and CsA since day +5. One hundred sixteen (69.5%) recipients, transplanted between 2015 and May 2018, received a total dose of 4.5mg/kg of rabbit-ATG (given on day -3,-2 and -1). In May 2018, the dose of ATG was lowered to a total of 2mg/kg (given on day -3 and -2). A total of 51 (30.5%) recipients received the lowered dose of ATG. The median follow-up of the entire cohort was 14 months (range: 0.4-44.5). For those patients who got a higher dose of ATG was 20 months and for those who received a lower dose of ATG was 8.8 months. Data was collected retrospectively and updated on July 2019. Cumulative incidence (Cum.Inc) of GVHD analysis was assessed accounting relapse and death as competing events. Results Baseline and post-transplant information are summarized in the Table 1 and 2. Ninety-three (55.7%) recipients were diagnosed with acute myeloid leukemia (AML). The cum.Inc of grade II-IV and grade III-IV acute GVHD at day +100 was respectively 15.6% (95% confidence interval (CI) 10.6-21.6) and 3.6% (95% CI 1.5-7.3). The cum.Inc of acute GVHD was not significantly affected by the dose of ATG (P>0.05). The cum.Inc of chronic GVHD was 10.9% (95% CI 6.6-16.4). Due to the shorter median follow up of the cohort that received a lower dose of ATG, the impact of the reduction of the dose in the cum.Inc of chronic GVHD was not explored. Overall, 48 (28.7%) recipients died and 35 (20.4%) relapsed. Main causes of death were relapse (14.4%) and infection (9.6%). Outcome information is reported in the Table 2 and Plot A, B and C. One-year overall survival (OS), relapse-free survival (RFS) and GVHD-free/RFS (GFRFS) were respectively 75.6%, 70.3%, 60.4%. Table 3 summarizes the impact of the use of a different dose of ATG in acute GVHD and post-transplant outcome. No significant differences were found between the two groups that receive a different dose of ATG. However, median follow-up was shorter in the cohort that received 2mg/kg of ATG. Table 4 reports the main post-allo-HSCT information of patients diagnosed with AML. One-year OS, RFS and GRFRS for this subgroup of patients were 76.8%, 71.9% and 65.9%. Conclusion The unique and modern combination of RIC PB allo-HSCT using ATG, PTCY and CsA for GVHD prophylaxis results in impressive post-transplant outcomes using 10/10 MUD. The use of dual T-cell depletion with ATG and PTCy is safe and provides an extraordinary control of GVHD with acceptable relapse rates using PB stem cell 10/10 MUD grafts. ATG of only 2mg/kg when it is combined with PTCy and CsA, results in an effective control of acute GVHD rates. The optimal dose of ATG for GVHD prophylaxis is not well established. Further investigations need to be done to determine the efficacy of a lower dose of ATG controlling chronic GVHD in this setting. For patients diagnosed with AML, this protocol is safe and an effective approach when a 10/10 MUD is available. Disclosures Michelis: CSL Behring: Other: Financial Support. Mattsson:Celgene: Honoraria; Gilead: Honoraria; Therakos: Honoraria.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2053-2053
Author(s):  
Kirsty J. Thomson ◽  
Karl S. Peggs ◽  
Emma C. Morris ◽  
Asim Khwaja ◽  
David C. Linch ◽  
...  

Abstract The role of T cell depletion in allogeneic stem cell transplantation for acute myeloid leukemia (AML) remains controversial. This study describes the results of an alemtuzumab-containing myeloablative regimen used to transplant 51 consecutive patients with hematological malignancy. Diagnoses were AML (n=48) of which 8 were secondary (6 transformed MDS; 2 therapy-related AML) or MDS (n= 3). Median age at transplant was 38yrs (range 13–57), and 29 patients had unrelated donors. Sixteen patients were HLA-mismatched at up to 3/10 loci (8x1; 7x2; 1x3). The majority of patients (39/51; 76%) were high-risk for relapse, as defined by induction failure (>15% blasts after one course of induction chemotherapy or >5% blasts after 2 courses), adverse cytogenetics, >CR1, or secondary disease. Six patients (12%) had residual detectable disease at the time of transplant despite at least 3 courses of induction therapy. Patients received cyclophosphamide 120 mg/kg and fractionated total body irradiation (14.4 Gy). Those with unrelated donors also received fludarabine 90 mg/m2. Stem cell source was bone marrow in 11 patients and PBSC in the remaining 40. Stem cells underwent in vitro T cell depletion using 20mg alemtuzumab added to the bag. All patients also received cyclosporin A for GvHD prophylaxis. Median follow-up for the surviving patients is 29 months (range 3–49). The estimated event-free survival is 70% at 1yr and 58% at 3yrs. Acute GvHD grade II occurred in 8% (no grade III–IV) and extensive chronic GvHD in 22%. Non-relapse mortality was 17% at 1yr and 25% at 3yrs, and relapse has occurred in 9 patients, giving an estimated relapse risk of 21% at 3yrs (24% in high-risk patients, n=39). For EFS and NRM, the only significant variable is age >45yrs at transplant, with no significance for donor type or presence of HLA mismatch. For patients aged ≤ 45yrs at transplant (n=42), of whom 81% were high risk for relapse, the outlook is very good with estimated NRM of 15% at 3yrs, and EFS of 69%. Use of this regimen therefore permits the successful transplantation of younger patients with high-risk disease and HLA-matched or mismatched unrelated donors, with minimal acute GvHD, low non-relapse mortality and no evidence of an excessive relapse rate, when compared to regimens without T cell depletion.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3533-3533 ◽  
Author(s):  
Heeje Kim ◽  
Woo-Sung Min ◽  
Ki-Seong Eom ◽  
Byung-Sik Cho ◽  
Seung-Ah Yahng ◽  
...  

Abstract Abstract 3533 Some groups have continuously tried to reveal the possible role of full-haplotype mismatch stem cell transplantation (SCT) in patients who lack a compatible donor. However, the long-term event-free survival (EFS) rate after high-grade mismatch SCT using conventional myeloablative conditioning with T-cell depletion (TCD), even in complete remission (CR), is still only in the range of 10–30%. Most patients who were transplanted had heavily pretreated acute leukemia. The most important obstacle to overcome is the high rate of infection-related mortality associated with delayed immunological recovery in the setting of full-haplotype mismatch SCT, specifically by way of complete depletion of donor T cells in the graft. In contrast, by using unmanipulated donor cells and less aggressive conditioning regimens without TCD, most groups in Asia have reported EFS >40%. We investigated the role of reduced-intensity conditioning without TCD in HLA-mismatched related-donor SCT, specifically in patients with high-risk acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) who received unmanipulated granulocyte colony stimulating factor (G-CSF)-mobilized peripheral blood stem cells (PBSCs). SCT was performed in 32 consecutive patients. We compared two different protocols: one with more intensive conventional conditioning with TCD (group 1), and one with reduced intensity without TCD (group 2). The protocols differed between the two groups with respect to total body irradiation (1200 cGy in group 1 vs. 800 cGy in group 2) as well as busulfex, fludarabine, and anti-thymocyte globulin (ATG; thymoglobulin, Genzyme) treatments. No patient in group 1 received post-transplant graft versus host disease (GvHD) prophlyaxis or G-CSF. In contrast, all patients in group 2 received 1.25 mg/kg/day ATG for 4 consecutive days, together with our standard GvHD prophylaxis regimen of methotrexate (5 mg/m2 intravenous bolus on days 1, 3, 6, and 11) and tacrolimus starting at day –1. All patients received G-CSF-mobilized PBSCs. The median number of CD34+ cells infused was 14.2 (range, 4.3–20.6) × 106/kg for group 1 and 6.0 (range, 3.6–8.5) × 106/kg for group 2. The median numbers of CD3+ cells infused were 0.03 (range, 0.015–0.057) × 106/kg and 870 (range, 84–1260) × 106/kg, respectively. G-CSF was administered subcutaneously to all patients in group 2 at a dose of 5 mg/kg/day, from day 7 after transplantation until neutrophil recovery. GvHD, relapse, non-relapse mortality (NRM), overall survival, EFS, reconstitution of immunity, and natural killer (NK) cell alloreactivity were compared between the groups. The median patient age was 33 (range, 17–48) years for group 1 and 39 (range, 19–63) years for group 2. The median follow-up for surviving patients was 18 months (range, 8–100). The majority of patients had intermediate or unfavorable cytogenetic features. The transplanted patients were all successfully engrafted. The median times to neutrophil (>0.5 × 109/kg) and platelet (>20 × 109/kg) recovery were 12 and 13 days in group 1, and 11 and 10 days in group 2. The overall rates of acute and chronic GvHD were 50% and 33%, and 80% and 92% in groups 1 and 2, respectively. Of note, NRM differed significantly between the groups: 46.2% in group 1 vs. 9.5% in group 2 (P = 0.0014). The estimated probability of EFS at 2 years was 15.3% for group 1 vs. 59.2% for group 2 (P = 0.05). We did not find NK alloreactivity in any of the patients, based on the NK-killer cell immunoglobulin-like receptor (KIR) disparity between donor and recipient. However, we noted a significant difference in EFS between C1/C1 homozygotes and C1/C2 heterozygotes according to the donor NK-KIR ligands (P = 0.0175). Recovery of CD4+ cell numbers at 3 months after SCT showed a markedly different pattern between the groups, with a median of 4 cells/μl (range, 2–7) in group 1 vs. 311 cells/μl (range, 56–1226) in group 2. Our findings suggest that full-haplotype mismatch SCT using a Korean-adapted protocol is a feasible therapeutic strategy for patients with high-risk AML/MDS, specifically in CR, provided that there is a further defined plan for the investigation of NK/T-cell alloreactivity. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4527-4527
Author(s):  
James L. Slack ◽  
Jose F. Leis ◽  
Craig B. Reeder ◽  
Joseph R. Mikhael ◽  
Raoul Tibes ◽  
...  

Abstract Abstract 4527 Introduction: The prognosis for pts with intermediate or high risk AML remains dismal, with relapse rates typically in the 60–80% range after treatment with chemotherapy alone. While allo-SCT can decrease the risk of relapse to 10–20%, widespread use of this modality is limited by relatively high rates of non-relapse mortality (NRM), often due to severe acute and/or chronic GVHD. Attenuation of severe GVHD, without a concomitant increase in relapse or NRM, should improve outcomes and result in cure of a larger fraction of AML pts. We therefore investigated the use of in-vivo T-cell depletion with r-ATG in pts with intermediate- or high-risk AML but without active leukemia at the time of allo-SCT. Patients and Methods: Pts (n = 43) were included in this retrospective analysis if they were between 18 and 65 years of age and had no evidence of active AML at the time of allo-SCT (see Table). All pts had 1 or more high-risk features: 1) adverse or intermediate risk cytogenetics (without NPM1 mutation if cytogenetically normal); 2) therapy-related or secondary AML; 3) high WBC count at diagnosis; 4) failure to achieve CR after 1 cycle of induction; or 5) not in CR1 at allo-SCT. Among the 43 pts, 10 received grafts from related donors, 14 from 10/10 matched unrelated donors (URDs), and 19 from mismatched URDs (9/10, n = 11; 8/10, n = 8). All pts received r-ATG according to institutional standard operating policy, with doses ranging from 2.5 – 10 mg/kg depending on donor type and degree of mismatch. All transplants were performed using PBSC. Additional GVHD prophylaxis included tacrolimus plus either methotrexate or mycophenolate mofetil. Results: The median age was 47 (range 20 – 65), and median follow-up for surviving pts is 12 (range 1 – 66) months. As of 8/5/11, 39 pts were alive, and 4 had died from multiorgan failure (n = 1), relapse (n = 1), GVHD (n = 1), and veno-occlusive disease (n = 1). The 2-year estimate of PFS is 84.7% (Fig. 1). The 2-year cumulative incidence of relapse is 6.8% (2 pts, days 97 and 147), and of non-relapse mortality 9.4%. Three pts developed severe (grades III-IV) acute GVHD by day 100 (cumulative incidence 4.6% at day 100) with no additional cases of severe acute GVHD beyond day 100. To date, 4 pts have developed moderate/severe chronic GVHD (cumulative incidence 16.8% at 2 yrs), with one death at day 344 related to complications of acute and chronic GVHD. CMV reactivation occurred in 29 pts (56%), with no deaths related to CMV. Three pts have reactivated EBV, with one case of PTLD (all treated with Rituximab). Conclusions: In this retrospective analysis of single center data, the inclusion of r-ATG in the GVHD prophylactic strategy appeared to significantly attenuate the incidence and severity of both acute and chronic GVHD. Although follow-up is relatively early, the incidence of relapse and NRM does not appear to be increased compared to contemporaneous pts treated without ATG. Given that almost half of the pts received grafts from mismatched URDs, this abrogation in risk of GVHD is significant and clinically relevant. While randomized studies are needed, these data suggest that in-vivo T-cell depletion with r-ATG ameliorates severe GVHD, without increasing relapse or non-relapse mortality, in AML pts without overt leukemia at the time of allo-SCT. Using this strategy, cure rates of 70 – 80% may be realistic and attainable for younger (</= age 65) AML patients who achieve a leukemia-free state and who have a reasonably matched related or unrelated donor. Disclosures: Reeder: Celgene: Research Funding; Millennium Pharmaceuticals Inc.: Research Funding; Novartis: Research Funding.


2021 ◽  
Vol 12 ◽  
pp. 204062072110637
Author(s):  
Jeongmin Seo ◽  
Dong-Yeop Shin ◽  
Youngil Koh ◽  
Inho Kim ◽  
Sung-Soo Yoon ◽  
...  

Background: Allogeneic stem cell transplantation (alloSCT) offers cure chance for various hematologic malignancies, but graft- versus-host disease (GVHD) remains a major impediment. Anti-thymocyte globulin (ATG) is used for prophylactic T-cell depletion and GVHD prevention, but there are no clear guidelines for the optimal dosing of ATG. It is suspected that for patients with low absolute lymphocyte counts (ALCs), current weight-based dosing of ATG can be excessive, which can result in profound T-cell depletion and poor transplant outcome. Methods: The objective of the study is to evaluate the association of low preconditioning ALC with outcomes in patients undergoing matched unrelated donor (MUD) alloSCT with reduced-intensity conditioning (RIC) and ATG. We conducted a single-center retrospective longitudinal cohort study of acute leukemia and myelodysplastic syndrome patients over 18 years old undergoing alloSCT. In total, 64 patients were included and dichotomized into lower ALC and higher ALC groups with the cutoff of 500/μl on D-7. Results: Patients with preconditioning ALC <500/μl were associated with shorter overall survival (OS) and higher infectious mortality. The incidence of acute GVHD and moderate-severe chronic GVHD as well as relapse rates did not differ according to preconditioning ALC. In multivariate analyses, low preconditioning ALC was recognized as an independent adverse prognostic factor for OS. Conclusion: Patients with lower ALC are exposed to excessive dose of ATG, leading to profound T-cell depletion that results in higher infectious mortality and shorter OS. Our results call for the implementation of more creative dosing regimens for patients with low preconditioning ALC.


Sign in / Sign up

Export Citation Format

Share Document