GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Hematology, Informa UK Limited, Vol. 19, No. 2 ( 2014-03), p. 63-72
    Type of Medium: Online Resource
    ISSN: 1607-8454
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2014
    detail.hit.zdb_id: 2035573-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research and Treatment, Korean Cancer Association, Vol. 43, No. 3 ( 2011-09-30), p. 195-198
    Type of Medium: Online Resource
    ISSN: 1598-2998 , 2005-9256
    Language: English
    Publisher: Korean Cancer Association
    Publication Date: 2011
    detail.hit.zdb_id: 2514151-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3503-3503
    Abstract: Abstract 3503 Background: Aplastic anemia (AA) is a life-threatening bone marrow failure disorder. Therefore, many patients with AA require blood transfusions as supportive management. Regular transfusions of packed red cell (PRC) lead to the development of iron overload, which is known to increase the risk of complications after stem cell transplantation (SCT). However, the prognostic impact of pretransplant transfusion history of PRC on outcome in AA has not been completely analyzed. We investigated the impact of pretransplant transfusion amount of PRC on outcome after allogeneic SCT in severe AA (SAA). Methods: 221 adult patients with SAA who underwent allogeneic SCT between January 1995 and August 2007 who had not received optimal iron chelating therapy were selected for retrospective analysis. Results: 221 patients were divided into two groups according to the mean amount of pretransplant transfusion (32 PRC units): receiving less than 32 PRC units (n=164), 〉 32 PRC units (n=57) before SCT. The median follow-up duration of survivors after SCT was 47.9 (39.5-56.2) months in ≤32 PRC units of transfusion group and 42.8 (39.7-45.9) months in 〉 32 PRC units of transfusion group. Primary engraftment was achieved in all, but 13 patients (9/164 patient, 5.5% in the ≤32 PRC units of transfusion group, 4/57 patients, 7% in the 〉 32 PRC units of transfusion group, P=0.745) developed secondary graft failure. Acute GVHD (grade II-IV) developed in 27.4% in ≤32 PRC units of transfusion group and 42.1% in 〉 32 PRC units of transfusion group (P=0.04), and extensive type of chronic GVHD occurred in 20.7% and 26.3% among evaluable patients, respectively. In the comparison between two groups, higher pretransplant transfusion group has significantly increased risk of transplant-related mortality (TRM) ( 〈 32 PRC units of transfusion: 8.2% vs 〉 32 PRC units of transfusion: 25.2%, P= 〈 0.000), and lower overall survival (OS) (91.8% vs 75.2%, P=0.001) than those with lesser transfusion history. Multivariate analysis revealed that higher pretransplant PRC amount [HR (95% CI) 3.09 (1.44-6.63), P=0.004] and donor type (related vs unrelated) [HR 2.41 (95% CI) (1.10-5.27), P=0.028] were independent prognostic factor for affecting OS. Conclusion: These results indicate that higher pre-transplant transfusion history of PRC was associated with increased TRM and decreased OS, and it has shown to have a negative impact on outcome after SCT in SAA. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3430-3430
    Abstract: Abstract 3430 Background: Antithymocyte globulin (ATG) is the drug of choice for immunosuppressive therapy (IST) in patients with aplastic anemia (AA) unsuitable for hematopoietic stem cell transplantation. The standard ATG preparation in AA had been horse ATG because of the larger experience and the results already reported with this preparation. Due to the unavailability of the horse ATG since 2006, rabbit ATG became the only available ATG preparation in Korea. But, there are only limited data about the therapeutic efficacy of rabbit ATG as first-line IST in AA. Method: We retrospectively investigated the outcome of 58 evaluable patients among 62 patients with AA treated with IST using rabbit ATG as front line between March 2006 and April 2010 at our institution. 70.7% of enrolled patients were very severe (n=18) or severe AA (n=23). All patients received rabbit ATG (Thymoglobulin®, 2.5mg/kg per day for 5 days) with methylprednisolone and cyclosporine A. Response rate (RR) was assessed at 3, 6, 9, 12 and 18 months after IST. Results: After IST, overall RR was 27.8%, 50.8%, 52.8%, 52.8% and 56.7% after 3, 6, 9, 12 and 18 months, respectively. Complete response (CR) rates were 0.8%, 1.8%, 5.6%, 9.6% and 21.2% after 3, 6, 9, 12 and 18 months, respectively. Median time to achieve partial response (PR) and CR were 93 (range; 12∼977 days) and 381 (range; 12–614) days. Among 31 responders, 10 patients (32.3%) relapsed. Median time between response and relapse were 396 days (range; 254∼681 days). Estimated overall survival and failure free survival at 3 years from ATG treatment were 85.8% and 42.8%, respectively. Age ( 〉 45) at the use of ATG was an independent predictor of overall survival and overall response (p=0.033 and 0.027) in univariate analysis. Other factors such as disease severity, pre-ATG hematological parameters (absolute lymphocyte count and absolute reticulocyte count) were not associated with overall survival and failure free survival. Conclusion: These data indicate that rabbit ATG was as effective as horse ATG based on other recently published data. But, the response time of rabbit ATG treatment is longer than that of horse ATG treatment. Especially, more than half of patients who achieved CR required time over 1 year after ATG treatment. Rabbit ATG can be effective IST regimen comparable to horse ATG, but it takes longer time to achieve sufficient response. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2355-2355
    Abstract: Abstract 2355 Background: The graft-versus-leukemia effect in adult acute lymphoblastic leukemia (ALL) has now been definitely confirmed from the ‘matched related donor (RD) versus no donor' comparisons. However, no definite conclusions can be extracted from these data as to whether or not there is a survival advantage to RD-stem cell transplantation (SCT) over other therapeutic modalities for both high-risk and standard-risk patients with Philadelphia (Ph)-negative ALL. In addition, ‘RD versus no donor' approach is becoming outmoded, as in many studies those previously in a ‘no donor' category are now undergoing unrelated donor (URD)-SCT. Aims: We report long-term outcomes of total body irradiation-based myeloablative SCT in 292 consecutive adults with Ph-negative ALL who received transplants at our center between 1995 and 2008 (median follow-up of survivors, 85 months). This study focused on following questions: (1) How different are the outcomes of SCT according to the donor sources? (2) Which factors are important to determination of transplantation outcome? (3) Which URD should be chosen? (4) Is there a role of autologous (AUTO)-SCT plus maintenance chemotherapy? Methods: Median age was 25 years (range, 15–63 years). Overall, 237 (81.2%) of 292 patients had one or more high-risk features, including adverse cytogenetics [t(4;11), t(8;14), complex ( 〉 =5 abnormalities), Ho-Tr], older age ( 〉 =35 years), high leukocyte counts ( 〉 =30×109/L for B-ALL, 〉 =100×109/L for T-ALL), or delayed first complete remission (CR1; 〉 28 days). Two hundred and forty-one patients (82.5%) were transplanted in CR1; 22 (7.6%) in CR2; and 29 (9.9%) in advanced status. URD sources were classified as well-matched (WM), partially matched (PM), and mismatched (MM) based on a new proposed guideline from the NMDP-CIBMTR. Donor sources were RD (n=132), WM-URD (n=30), PM-URD (n=19), MM-URD (n=19), and AUTO (n=92). All patients and donors provided written informed consent, and the treatment protocol was approved by the institutional review board of The Catholic University of Korea. Results: Cumulative incidence of relapse at 5 years was 48.5% for AUTO versus 32.6% for RD, 19.4% for WM-URD, 32.3% for PM-URD, and 51.0% for MM-URD (RR, 2.70; 95% CI, 1.65 to 4.42; p 〈 0.001). In multivariate analyses, other factors associated with higher relapse risk included transplantation in CR2 or later (p 〈 0.001), T-lineage ALL (p=0.020), and adverse cytogenetics (p=0.038). Cumulative incidence of non-relapse mortality (NRM) at 5 years was 40.5% for MM-URD versus 19.6% for SD, 20.3% for WM-URD, 15.8% for PM-URD, and 9.8% for AUTO (RR, 3.09; 95% CI, 1.32 to 7.25; p=0.010). Patients older than 35 years had higher NRM (p=0.007). As a result, disease-free survival (DFS) at 5 years was inferior using AUTO (46.1%; RR, 1.69; 95% CI, 1.14 to 2.51; p=0.010) or MM-URD (26.3%; RR, 2.03; 95% CI, 1.05 to 3.95; p=0.036), compared to RD sources, while DFS from all other donor sources was approximately equivalent (53.5% for RD, 63.3% for WM-URD, and 57.0% for PM-URD). Transplantation in CR2 or later (p 〈 0.001), older age (p=0.020), and adverse cytogenetics (p=0.041) were associated with poorer DFS. In a pairwise comparison of outcomes between RD-SCT and AUTO-SCT for patients in CR1, the inferiority of AUTO-SCT was observed, particularly in high-risk patients. Conversely, in standard-risk patients, AUTO-SCT yielded comparable outcomes to RD-SCT. Summary/Conclusions: Our long-term data suggest that outcomes are similar for transplantation using SD, WM-URD, or PM-URD sources, and these may be considered the best donor sources for adults with Ph-negative ALL, especially for those with high-risk features. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4398-4398
    Abstract: Background:Paroxysmal nocturnal hemoglobinuria (PNH) is a nonmalignant clonal disorder of hematopoietic stem cells characterized by a somatic mutation in the PIG-A gene, encoding the glycosyl phosphatidylinositol (GPI) moiety. PNH clones lack GPI-anchored proteins (GPI-AP) which inhibit the activation and cytolytic functions of complement. Recently, Eculizumab, humanized monoclonal antibody directed against complement component C5, has used increasingly for the patients with hemolytic PNH. However, the patients with PNH clone and bone marrow failure syndrome (i.e. aplastic anemia) should be treated as their predominant clinical manifestation. Allogeneic stem cell transplantation (SCT) can be curative treatment option especially for PNH patients with combined aplastic anemia (AA). The aim of the present study was to evaluate long-term outcome of allogeneic SCT in patients with AA/PNH. Methods: Total of 27 patients with PNH clones underwent allogeneic SCT at our institution between Jan 1998 and Mar 2014. Among them, seven patients had classic PNH and 20 patients with cytopenia had AA/PNH (with bone marrow evidence of a concomitant AA). We analyzed long-term transplant outcomes in 20 patients with AA/PNH. Results: There were 12 male and 8 female patients with a median age of 34 years (range, 13-51 years). The median interval from the diagnosis to transplantation was 8 months (range; 1-201 months). The median transfusions prior to SCT were 33 units (range; 8-208 units). Pre-transplant GPI-AP deficient neutrophils and erythrocytes were 46% (0-99) and 15.6% (0-88), respectively. Median white blood cell, absolute neutrophil count, hemoglobin, and platelet at transplant were 2.3×109/L, 0.7×109/L, 7.9 g/dL, and 21×109/L, respectively. Median LDH level was 714 U/L (range; 273-6499 U/L) and 11 (55%) patients had LDH ≥1.5x upper normal limit. PNH patients with SAA (n=14), VSAA (n=4), or non-SAA (n=2) received SCT from sibling (s) donor (n=15) or unrelated (u) donor (n=5). The conditioning regimen for s-SCT consisted of fludarabine (180 mg/m2) + cyclophosphamide (CY, 100 mg/kg) + ATG (10 mg/kg) (n=11), or busulfex (12.8 mg/kg) + CY (120mg/kg) (n=4). The conditioning regimen for u-SCT was TBI (fractionated, 800 cGy) + CY (100-120 mg/kg) ± ATG (2.5 mg/kg). GVHD prophylaxis consisted of CsA + MTX in s-SCT and FK506 + mini-MTX in u-SCT, respectively. After a median follow-up of 57 months (range 4.7-122.1), the 5-year estimated OS rates were 90.0 ± 6.7%. Two patients died of treatment-related mortality (TRM), including acute GVHD (n=1) and cerebral hemorrhage (n=1), respectively. Except one patient with early TRM, 19 patients engrafted with no secondary graft-failure. The cumulative incidence of acute GVHD (≥grade II) and chronic GVHD was 25.0 ± 1.0% and 26.3 ± 10.4%, respectively. PNH clones disappeared at median 1.8 months (range 0.9-11.9) after SCT and reemerging of PNH clone was not observed in all patients. Conclusion: This study showed that long-term transplant outcome in patients with AA/PNH were comparable to that of allogeneic SCT in SAA (the 3-year estimated OS rates were 92.7 and 89 % for s-SCT and u-SCT, respectively) at our institution (ASH Annual Meeting Abstracts 2012;120:4151). Therefore, application of allogeneic SCT should be considered in PNH patients with AA in case of availability of well matched donor. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 492-492
    Abstract: Abstract 492 Diagnosis and therapeutic decision of gastrointestinal (GI)-graft-versus-host disease (GVHD) mainly rely upon clinical symptoms and exclusion of other causes of GI dysfunction. Particularly, the presence of CMV-GI-disease should be examined with immunostaining of biopsy. However, there have been few reports about the occurrence of CMV-GI-disease in patients with GI-GVHD in the era of preemptive therapy for CMV infection with real-time quantitative polymerase chain reaction (RQ-PCR) and its related outcomes. Between January 2008 to December 2010, 392 adult patients with AML, ALL, MDS, and CML underwent allogeneic stem cell transplantation (SCT) and all had serial monitoring for CMV DNA based on RQ-PCR followed by initiation of preemptive therapy. Ninety-seven percent of recipients and 84% of donors were CMV-seropositive before SCT. Clinical GI-GVHD based on symptoms, stool studies, and endoscopic findings developed in 114 patients (29%) and they were treated by high-dose systemic steroid. Of these patients, 103 patients performed endoscopic biopsies not only at the time of onset of clinical symptoms but also during the clinical course of GI-GVHD in the absence of response to steroid, and all samples from biopsies were immunohistochemically stained. The median age of the 103 patients was 40 years (range, 15–70). These patients consisted of AML (n=59), ALL (n=29), MDS (n=12), and CML (n=3) transplanted from sibling (n=45), unrelated (n=48), familial-mismatched donors (n=8), and umbilical cord donors (n=2). Conditioning regimens consisted of myeloablative (76%) and reduced-intensity regimens (24%). Anti-thymocyte globulin was given as a part of conditioning in 41 patients (40%). GVHD prophylaxis was based on administering calcineurin inhibitor with short-course methotrexate. GI-GVHD was developed as a manifestation of not only acute GVHD (aGVHD, 87%) but also overlap syndrome (13%). Isolated GI-GVHD was 30%. Eighty-nine patients (86%) initially presented with GI-GVHD, whereas 14 patients (14%) developed GI-GVHD after the treatment of GVHD of other organs. Of the 103 patients, 60 (58%) had histologic findings for GI-GVHD (lower gut, n=41, 40%; lower gut + upper gut, n=10, 10%; upper gut, n=9, 8%) and 26 (25%) had positive immunohistochemical stain for CMV-GI-disease (lower gut, n=13, 13%; lower gut + upper gut, n=4, 4%; upper gut, n=9, 8%). The CMV-GI-disease developed at a median time of 56 days after SCT (range, 20–403). Multivariate analyses revealed that older age over 30 years (RR 4.42, 95%CI 1.10–17.84), HLA-mismatched donors (RR 3.41, 95%CI 1.21–9.60), and higher grade of GVHD (grade III+IV aGVHD or severe cGVHD, RR 3.24, 95% CI 1.22–8.60) were significant risk factors for CMV-GI-disease. Most patients (73%) developed CMV-GI-disease after the treatment of GI-GVHD or other organs of GVHD, whereas CMV-GI-disease developed concurrently with GI-GVHD in other 27%. After the anti-viral therapy with ganciclovir or foscarnet, 14 patients (54%) showed complete resolution of CMV-GI-disease, while 12 patients (46%) failed to show any response. There were 4 patterns of development of pathologically proven GI-GVHD and CMV-GI-disease (Figure). GVHD-specific survival of patients with CMV-GI-disease was significantly lower than patients without CMV-GI-disease (36.2% vs. 79.3%, P = 0.002). Patients with both GI-GVHD and CMV-GI-disease (n=18) had significantly inferior GVHD-specific survival than other 3 patterns of development of GI-GVHD and CMV-GI-disease (24.5% vs. 78.2%, P = 0.001). Our data demonstrate 25 percent of patients with clinical GI-GVHD developed CMV-GI-disease during the treatment or at the time of diagnosis of GI-GVHD even in the era of preemptive therapy for CMV disease based on RQ-PCR, especially in older patients, HAL-mismatched transplants, and higher grade of GVHD. Patients with CMV-GI-disease, especially who concurrently had pathologically proven GVHD, had inferior GVHD-specific survival. Thus, we reemphasize the need of endoscopic biopsy with immunohistochemical stain for CMV disease if ever possible at the diagnosis of GI-GVHD as well as in the absence of response to treatment of GVHD, especially in high risk groups, which can facilitate proper selection of treatment. Additionally, novel prophylactic measures such as immunotherapy and drug prophylaxis should be considered in this specific group of patients. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2343-2343
    Abstract: Abstract 2343 Background. The National Institutes of Health consensus criteria (NCC) for chronic graft-versus-host disease (GVHD) are based on clinical manifestations rather than onset time after transplantation. Previous studies about the feasibility of the new criteria have been performed in patients who survived beyond 100 days after transplantation and had GVHD presenting after day 100. In order to investigate and compare the clinical impact of acute and chronic GVHD by NCC on survival, a cohort including patients with GVHD presenting ‘before' as well as ‘after' day 100 is needed. Additionally, a proper statistical tool should be applied to clarify the effect of GVHD on survival outcomes, because the occurrence of GVHD is a well known time-dependent event. In this context, we examined a cohort including all patients who underwent allogeneic stem cell transplantation (SCT) in order to investigate clinical impact of acute or chronic GVHD by NCC on survival outcomes. Methods. We retrospectively investigated 771 patients who underwent allogeneic SCT between January 2002 to December 2008. To study a homogenous cohort, patients received 2 allogeneic SCT and/or donor-lymphocyte-infusion was excluded. We used time-dependent analyses to reveal the effect of GVHD on survival outcomes. In particular, to approach the effects of GVHD as a time-dependent covariate on competing risks [relapse or transplant-related mortality (TRM)], we analyzed cause-specific hazards by Cox proportional hazards regression model (Cartese G, Andersen PK. Biometrical Journal 2009;51:138–158). Results. The median age was 36 years (range, 15–68). Patients had various hematologic malignancies (AML/ALL/CML/MDS/MM, 361/226/86/67/31, respectively) and were transplanted from matched sibling (n=485), well-matched unrelated donors (n=154), partially-matched unrelated donors (n=101), and mismatched unrelated donors (n=31). Conditioning regimens consisted of myeloablative (n=536) and reduced-intensity regimens (n=235). GVHD prophylaxis consisted of cyclosporine and short-course methotrexate for related SCT and tacrolimus and short-course methotrexate for unrelated SCT. Among 771 patients, 540 patients were diagnosed with GVHD after transplantation. According to onset time of GVHD, in 348 patients GVHD developed within 100 days after SCT, whereas in 156 patients GVHD occurred more than 100 days after SCT. Using the NCC, we classified patients as three categories regardless of onset time: (1) acute GVHD (n=215), if patients had only acute features (no chronic features) during the course of GVHD, (2) acute GVHD with following chronic GVHD (n=118), if patients had any chronic features after the occurrence of acute GVHD, and (3) chronic GVHD (n=207), if patients had any chronic features at the onset of GVHD. Multivariate analyses using cause-specific hazards revealed that acute GVHD was significantly associated with lower relapse incidence [HR (95% CI) 0.65 (0.44–0.94), P=0.023]. However, it did not influence disease-free survival (DFS, P=0.602) and overall survival (OS, P=0.294) due to higher TRM in patients with acute GVHD [HR (95% CI) 1.74 (1.15–2.61), P=0.008] . Acute GVHD with following chronic GVHD was also associated with lower relapse incidence [HR (95% CI) 0.31 (0.15–0.66), P=0.002] and higher TRM [HR (95% CI) 2.89 (1.65–5.04), P 〈 0.001], which did not influence DFS (P=0.567) and OS (P=0.820). On the other hand, the occurrence of chronic GVHD significantly reduced relapse rates [HR (95% CI) 0.46 (0.29–0.75), P=0.002] without increasing TRM (P=0.177). Indeed, this effect was translated into improved DFS [HR (95% CI) 0.67 (0.48–0.95), P=0.023]. Conclusions. Our data show that both acute and chronic GVHD by NCC reduced the risk of relapse, demonstrating the presence of graft-versus-tumor (GVT) effect by the occurrence of GVHD and no difference according to the clinical features (acute or chronic) by NCC. However, survival benefit was only observed in chronic GVHD by NCC compared to acute GVHD with/without following chronic GVHD by NCC due to higher TRM. This study represents the first one demonstrating the GVT effect of GVHD defined by NCC in a cohort including GVHD presenting ‘before' as well as ‘after' day 100 using appropriate time-dependent analyses by cause-specific hazards by Cox proportional hazards regression model. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4559-4559
    Abstract: Abstract 4559 Patients with acute myeloid leukemia (AML) and t(8;21) generally have a favorable prognosis with a higher rate of first complete remission (CR1) and higher rate of cure with high-dose cytarabine (HDARA-C). However, some studies comparing stem cell transplantation (SCT) and conventional consolidation chemotherapy raised questions regarding improved survival with autologous SCT (ASCT) in these patients. Moreover, there has been some concern that the rate of relapse after HDARA-C is higher than previously reported and resultingly, only 50 – 60% of these patients are cured with contemporary treatment. Role of reduced intensity conditioning (RIC)-SCT has not been determined in this population. To date, few studies have analyzed the outcome of RIC-SCT for this population specifically. From this point of view, we performed a prospective phase II study to evaluate the efficacy of RIC-SCT for AML with t(8;21) and also compared the results with those of the historical cohort who treated with ASCT or myeloablative conditioning (MC)-SCT in our institution. We included adult patients aged between 16 and 65 with AML who had t(8;21) with or without additional aberration at the time of diagnosis. Since Mar 2007, we transplanted 19 consecutive adult patients with AML and t(8;21) after RIC from matched sibling (n = 12) or unrelated (n = 7) donor. Patients in RIC-SCT group were given a RIC regimen consisting of fludarabine (30 mg/m2/d, days –6, –5, –4, –3, and –2), busulfan (3.2 mg/kg/d, days –6 and –5) and low dose total body irradiation (400 cGy). Their clinical features were compared with historical patients who were transplanted with autografting (n = 47) or MC (n = 38) in our institution from 2001. The pretransplant characteristics among the three groups were well balanced except for patient age and time to SCT. For recipients of RIC, probability of overall survival (OS) and disease free survival (DFS) was 77.5 ± 9.9% and 78.2 ± 9.7%, respectively, with a median follow-up of 30 months (range: 9.5+ – 41.2+ months) for surviving patients (Fig 1a and b). 2-year cumulative incidence of relapse (CIR) and nonrelapse mortality (NRM) was 11.9 ± 7.9%, respectively (Fig. 1c and d). OS and DFS in this group were not different from those of ASCT and MC-SCT group. CIR appears to be lower than that of ASCT, but failing to demonstrate statistical difference. Patients in ASCT group showed clearly lower NRM (P = .049). By univariate analysis, age (£ 50 vs 〉 50 years) seems to be a dominant factor affecting DFS and OS in RIC-SCT and ASCT group (P = 0.25 and 0.29, respectively, for RIC-SCT group, and P = 0.36 and 0.13, respectively, for ASCT group). In ASCT group, presence of –Y was associated with inferior DFS (p = 0.049, Fig. 2a), whereas this difference disappeared when RIC or MC was applied for these patients (Fig. 2b and c). CIR was significantly lower in patients without –Y than with –Y (P = 0.012, Fig. 2d), while there was no difference in NRM between the two subgroups. Of 41 male patients with –Y, probability of DFS of allogeneic SCT recipients was slightly better than that of ASCT, although failing to demonstrate a statistical difference (Fig. 2e). RIC-SCT is effective treatment modality in terms of relapse and survival, judging from no difference in CIR and survival in comparison with those of MC-SCT, but greater effort should be put to reduce NRM further. Because ASCT was associated with extremely low NRM and demonstrated comparable efficacy to those of allo-SCT, this modality might be a preferable treatment option for this disease except for the subgroup of patients with –Y. Several previous studies reported that –Y was associated with shorter survival. In our study, patients with –Y demonstrated inferior survival only in the setting of ASCT, and these differences disappeared in allo-SCT groups, justifying that allo-SCT should be considered in this subgroup of patients. Superior survival in patients without –Y seems to be result of lowered CIR rather than improved NRM. Finally, MC-SCT is not recommended in patients with t(8;21) in CR1 because it has no advantages in terms of relapse and NRM.Figure 1.Overall transplant outcome. probability of (a) DFS, and (b) OS, (c) cumulative incidence of relapse, (d) cumulative incidence of NRMFigure 1. Overall transplant outcome. probability of (a) DFS, and (b) OS, (c) cumulative incidence of relapse, (d) cumulative incidence of NRMFigure 2.Outcome according to –Y status. (a) DFS of ASCT group (b) DFS of RIC-SCT group, (c) DFS of MC-SCT group, (d) CIR by –Y status (e) DFS according to transplant modalityFigure 2. Outcome according to –Y status. (a) DFS of ASCT group (b) DFS of RIC-SCT group, (c) DFS of MC-SCT group, (d) CIR by –Y status (e) DFS according to transplant modality Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2034-2034
    Abstract: Abstract 2034 Background: Despite the survival improvements with the advent of hypomethylating treatment (HMT), stem cell transplantation (SCT) remains the only curative treatment strategy in patients with myelodysplastic syndrome (MDS). Recent reports suggest that HMT could be a feasible bridge to SCT, thereby stabilizing the natural course of the disease. In the current study, we analyzed the influences of treatment response of HMT on the transplant outcome to clarify the optimal time point to proceed to SCT during HMT. Methods: We retrospectively analyzed 56 consecutive patients at a median 49 (18–70) years who received grafts from 23 sibling, 24 unrelated, or 9 alternative donors following a median 4 (1–13) cycles of HMT for MDS. Response assessment followed the standard criteria of IWG 2006 but two criteria were applied for disease progression (DP), for which blast counts at the start or of HMT (BL-start) or maximal response (BL-max) were used as reference blast level. Nineteen patients received SCT in continuous response (COR), which included 2 CR, 15 mCR with or without HI, and 2 HI. For remaining 37 patients, 21 were in stable disease (SD-start) and 16 in DP (DP-start) when BL-start was used whereas SD-start was reclassified to DP in 6 patients when BL-max was applied, who were designated as loss of response (LOR). DP included 12 AML, of whom 5 were in mCR at SCT. Results: Successful engraftment was achieved in all cases, and the cumulative incidence of acute GVHD (°ÃII) at day 100 and chronic GHVD at 2 years among evaluable patients were 32.1% and 66.0%, respectively. After a median follow-up of 18.3 months for survivors, overall survival (OS), disease free survival (DFS), cumulative incidence of relapse (CIR) and treatment related mortality (CITRM) at 2 years were 60.8%, 55.6%, 32.2, and 22.2%, respectively. Although statistically significant differences in survival were demonstrated whether BL-start or BL-max was used (Fig. A & B), the latter response model was better predictive and used for following analysis. When patients were dichotomized into COR/SD-max versus LOR/DP-start, the latter group was a significantly poor predictor for 2-year OS (77.1% versus 34.8%, p= 〈 .0001), DFS (74.5% versus 26.0%, p= 〈 .0001) and CIR (9.1% vs 64.5%, p= 〈 .0001) while CITRM was non-significant (16.3% versus 26.8%, p=.270). Multivariate analysis by adjusting for BM blast counts, cytogenetic risks at SCT, CD 34+ cell dose infused, hematopoietic cell transplantation specific comorbidity index and history of AML, DPmax and poor cytogenetic risk were powerful factors predicting worse DFS: DPmax, HR=4.09, p=.009,; HR=3.65, p=.004). This effect of response remained significant when analysis was confined in cohort excluding the patients with history of AML. Unexpected predictive power of response in the comparison of blast count at SCT was strengthened by the significant differences in DFS between COR/SD-max and LOR/DP-start in patients having the same blast level (5–19%). When influences of response were analyzed according to the IPSS risk group at HMT, COR showed better DFS compared to SD-max, LOR, or PD-start in INT-2/high group (Fig.C). Conclusion: This analysis demonstrates response to HMT and karyotype are the significant predictors in allogeneic SCT with pre-transplant HMT for MDS rather than the disease status before HMT or BM blast count at the time of SCT. Our data also suggests that carrying out SCT in MDS should be performed before LOR as well as PD and that DP is not just a progressed disease phase exerted by increased BM blast count but a condition led by evolved MDS clones with alternated characteristic features affected by HMT. This current study demonstrates the significance of response to HMT on transplant outcomes and therefore necessitates continued studies to further illustrate the significance. Disclosures: Kim: Janssen: Family, Honoraria, Research Funding; Celgene: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...