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: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3870-3870
    Abstract: Background: Allogeneic hematopoietic cell transplantation (HCT) is an effective therapy in Acute Myeloid Leukemia and Myelodysplastic Syndrome (AML/MDS). There is controversy over whether reduced intensity conditioning (RIC) results in similar outcomes to myeloablative conditioning (MAC), especially regarding relapse risk. It is difficult to identify the specific cause of the transplant failure rate in RIC patients amongst the multiple possible factors including relapse risk due to disease characteristics of older pts with AML/MDS, or multiple comorbidities in the population receiving RIC, resulting in higher morbidity and mortality, versus expected lower risk of regimen-related toxicity. In order to overcome this, we used a propensity score matching analysis in this study. Methods: A total of 248 patients transplanted for AML or MDS at the Princess Margret Cancer Center between 2009 and 2013 were included in this analysis. Inclusion was restricted to patients receiving Fludarabine/Busulfan plus low dose total body irradiation (TBI) with either RIC conditioning (Fludarabine 30mg/m2/day for 4 days, Busulfan 3.2mg/kg/day for 2 days and TBI 200 cGy n=121) or MAC conditioning (Fludarabine 50mg/m2/day for 4 days, Busulfan 3.2mg/kg/day for 4 days and TBI 400 cGy; n=127). The RIC and MAC groups were compared for overall survival (OS), non-relapse mortality (NRM) and relapse. Propensity score matching (PSM) analysis is used to adjust for the risk factors which affect the choice of treatment between different treatment options. Using PSM analysis, we performed a case-control study with well-balanced pairs of RIC and MAC patients. Pre-transplant variables included in the PSM were age at HCT, HCT-Comorbidity Index (HCT-CI), complete remission status (CR) at HCT, diagnosis (AML vs MDS), cytogenetic risk group (high-risk vs others), donor type (related vs unrelated) and period effect (transplant year). A total of 39 case-control pairs were selected within 0.2 of a difference in propensity score. Paired analysis was adopted throughout the PSM analysis for survival. RESULTS: With a median follow-up of 18 months among survivors in the overall population (n=248), the 2-year OS, NRM and relapse incidence rates were 48.0±3.6%, 34.6±3.6% and 24.8±3.5% respectively There was no difference between the 2 groups in OS (45.2±5.0% in RIC vs 51.7±5.2% in MAC at 2 years; p=0.541) or NRM (32.9±5.2% in RIC vs 35.7±4.9% in MAC at 2 years; p=0.504). However, there was a higher incidence of relapse in the RIC group (31.5±5.1% in RIC vs 18.2±4.8% in MAC at 2 years; p=0.033) Demographic and transplant characteristics were imbalanced between the 2 groups within the overall population, including older age (P= 〈 0.001), higher HCT-CI score (p=0.002) and more related donors in the RIC group (p=0.02). However, no differences were observed in CR status at HCT (p=0.110), subtype of diagnosis (AML vs MDS, p=0.174), or cytogenetic risk group (p=0.278). To overcome baseline imbalances we used a PSM analysis, and 39 case-control pairs (n=78) were selected. All pre-transplant variables became well balanced after propensity score matching, i.e. there were no differences in age (p=0.537), HCT-CI (p=0.931), CR status at HCT (p=0.655), diagnosis (p=0.774), cytogenetic risk group (p=0.784), donor type (p=0.496) or period effect (p=0.984). In the propensity score matched patients, there were no differences in OS (58.0±8.8% in RIC vs 50.9±8.1% in MAC at 2 years; p=0.554), NRM (28.0±8.2% in RIC vs 32.8±7.8% in MAC at 2 years; p=0.688), or relapse (17.8±6.7% in RIC vs 18.0±6.8% in MAC at 2 years; p=0.635). Conclusion: These results suggest, based on a propensity score matching analysis, that the outcomes of a Fludarabine/Busulfan plus low dose TBI based‎ RIC HCT for AML/MDS are equivalent to a Fludarabine/Busulfan plus low dose TBI based MAC with regards to the risk of relapse, NRM, and OS. 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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3139-3139
    Abstract: Introduction: Chronic graft versus host disease (cGVHD) is one of the major complications after allogeneic hematopoietic cell transplantation (allo-HCT). Several prognostic factors have been proposed to predict the outcomes of cGVHD including progressive type onset, extensive skin involvement, thrombocytopenia and NIH global score (NIH GS). Most studies have been focusing on the factors at the diagnosis of cGVHD without consideration of baseline characteristics prior to allo-HCT. We attempted to evaluate the prognostic factors for the outcomes of cGVHD treatment including the characteristics at the start of cGVHD treatment as well as prior to HCT. Method: We retrospectively reviewed 668 consecutive patients who underwent allo-HCT between 2004 and 2012 at the Princess Margaret Cancer Centre, Toronto, Canada, among whom 277 patients diagnosed as cGVHD and received systemic corticosteroids as a frontline cGVHD therapy. Chronic GVHD was classified and graded using the NIH consensus criteria. We evaluated non-relapse mortality (NRM), relapse and failure-free survival (FFS). FFS was defined as time to a switch in systemic therapy, NRM or relapse. The Kaplan-Meier method was used for FFS. The cumulative incidences of NRM, relapse and the treatment switch (TS) were calculated considering competing risks. Multivariate analysis was performed using the Cox proportional hazard regression model for FFS. Results: With a median follow-up duration of 26 months, the median time from HCT to cGVHD treatment was 183 days (range, 61-828). 102 patients (36.8%) were classified as classical cGVHD and 175 (63.2%) as overlap syndrome. At the start of cGVHD treatment 25 patients (9.0%) had mild cGVHD by the NIH GS, 189 (68.2%) moderate and 63 (22.7%) severe. Median age at allo-HCT was 51 year-old (range, 19-70). 162 patients (58.5%) were males and 65 (23.5%) patients were gender match of female donor to male recipient. 257 patients (92.8%) received peripheral blood stem cells (PBSC).175 grafts (63.2%) were from matched sibling donors (MSD). 180 patients (65%) received myeloablative conditioning. GVHD prophylaxis was calcineurin inhibitor (CNI) and methotrexate (n=82, 29.6%), CNI and mycophenolate mofetil (n=141, 50.9%), CNI and T-cell depletion (n=37, 13.5%) or others (n=17, 6.1%). The median FFS was 255 days (95% CI, 218-321). The severity of cGVHD, NIH GS correlated with FFS: median FFS was 164 days in severe vs 238 days in moderate vs 304 days in mild (p=0.001). The overlap syndrome was associated with a shorter FFS than classical cGVHD (223 vs 329 days, p=0.015). Patients receiving MSD graft showed longer FFS (329 days) than unrelated donor (196 days; p=0.004). The cumulative incidence of TS was 47.7% at 1 year. The NRM was 7.1% and relapse rate was 6.8% at 1 year. The MSD was associated with a lower 1-year NRM than the unrelated donors (4.2% vs 12.3%, p=0.003) while no difference between 2 groups for TS (p=0.731) or relapse at 1 year (p=0.565). Patients with overlap syndrome had higher NRM at 1 year than with classical cGVHD (10.0% vs 2.2%, p=0.009), but no differences in TS or relapse at 1 year (p=0.167 and p=0.138). Chronic GVHD severity by NIH GS showed a significant correlation with TS (28% in mild, 51.9% in moderate, and 43.8% in severe grade at 1 year, p=0.02) and NRM (4% in mild, 3.6% in moderate, and 19.1% in severe grade at 1 year, p 〈 0.001), but with relapse (p=0.784). Multivariate analysis for FFS confirmed that the use of unrelated donor showed a worse FFS (hazard ratio (HR) 1.660, p=0.001). FFS was also associated with the severity of cGVHD, NCC GS (mild vs moderate vs severe; HR 1 vs 2.1 vs 2.9, p=0.002) and the cGVHD subtype (classical vs overlap, HR 1 vs 1.39, p=0.028). We then assigned score 0 for NIH GS mild, 1 for moderate, and 2 for severe; for NIH subtype, score 0 for classical and 1 for overlap; for donor types, score 0 for MSD and 1 for unrelated donors. After summation of the scores, we regrouped them into low (score 0, n=11, 3.9%), intermediate (score 1-2, n=168, 60.6%), and high risk groups (score 3-4, n=98, 35.3%). The risk stratification model correlated nicely with FFS (FFS duration, 1977 days in low vs 341 days in intermediate, and 150 days in high risk group, p 〈 0.001). Conclusion: the use of unrelated donor, overlap subtype of chronic GVHD and severe grade of chronic GVHD at the time of initiation of chronic GVHD treatment affect adversely on failure-free survival. Disclosures Kim: Novartis Pharmaceuticals: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 5927-5927
    Abstract: Several studies have suggested a beneficial effect of CMV seropositivity and or reactivation on relapse risk after allogeneic hematopoietic cell transplantation (HCT) in myeloid malignancies. Inability to replicate this finding in T cell depleted HCT is suggestive of a potential effect modification between TCD and CMV in this setting, but this has not been addressed in previously published studies. We have retrospectively analyzed transplant outcomes in 192 patients with myeloid malignancies who have undergone unrelated donor HCT at our center during January 2006 –November 2013. Among this 111 patients received in vivo TCD with low dose (30 mg) alemtuzumab and 81 patients received non-TCD transplants. Recipients were CMV seropositive in 57% of TCD transplants and 59% of non-TCD transplants. Analysis showed a significant effect modification (p=0.03 for interaction) between alemtuzumab and CMV serostatus on relapse risk, and stratified analysis based on recipient CMV status showed inferior outcomes with alemtuzumab in CMV seropositive recipients but not in CMV seronegative recipients. In CMV seropositive recipients alemtuzumab was an independent predictor of higher relapse (Hazard Ratio=13.95, p=0.008), inferior relapse free survival (HR=2.30, p=0.002), and inferior overall survival (HR= 2.04, p=0.008). There was no difference in NRM between CMV seropositive and CMV seronegative groups, or between TCD and non-TCD transplants. These findings suggest the presence of an effect modification by TCD on CMV's effect on relapse, resulting in inferior transplant outcomes with low dose Alemtuzumab in CMV seropositive recipients, but not in seronegative recipients. Figure 1. CIR, NRM, RFS, and OS showing differential effects of T cell depletion between CMV seropositive and seronegative recipients (A-B) Cumulative incidence of relapse, (C-D) Non-relapse mortality, (E-F) Relapse free survival, and (G-H) Over all survival. Figure 1. CIR, NRM, RFS, and OS showing differential effects of T cell depletion between CMV seropositive and seronegative recipients. / (A-B) Cumulative incidence of relapse, (C-D) Non-relapse mortality, (E-F) Relapse free survival, and (G-H) Over all survival. Disclosures Off Label Use: Alemtuzumab for GVHD prophylaxis.
    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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3056-3056
    Abstract: Abstract 3056 Background: The pathogenesis of GVHD is not fully understood. Alloreactive T-lymphocytes are believed to be key mediators of GVHD. However, it is not clear if the pathobiology of GHVD is similar in each target organ GVHD. We aimed to identify predictive single nucleotide polymorphisms (SNP) markers associated with the risk of acute or chronic graft versus host disease (GVHD) as well as organ specific GVHD in 394 transplant recipients and donors. Methods: A total of 259 SNPs were genotyped in 53 genes, and evaluated for the risk of acute/chronic GVHD and organ specific GVHD. Predictive models were generated using both clinical factors and genetic SNP markers confirmed by multivariate analyses. Patients were stratified by quartile (25%) according to their risk score, and the risk of overall and organ specific GVHD were compared among the 3 risk groups (low, intermediate and high risk). C-statistic analysis was also performed to compare the stratification power of the predictive model generated using clinical and genetic factors with a model obtained using only clinical factors. Results: Several SNP markers in the cytokine-, apoptosis-, TGF-¥â or PDGF-mediated pathways were identified as predictive markers of acute/chronic GVHD. The risk of acute GVHD was associated with clinical factors such as HLA disparity and patient age. In addition, recipient FAS genotype (rs2234978), EDN1 genotype (rs4714384), and TGFB genotype (rs1800469), and donor TNFRII genotype (rs3397) were also strong predictive markers for acute GVHD. Significant predictive risk factors forchronic GVHD were the source of stem cells, a previous episode of acute GVHD and the donor IL1R1 genotype (rs3917225). Each organ specific GVHD shared common biologic pathways such as cytokine, TGF-¥â or PDGF-mediated pathways. However, different SNP markers were identified as predictive for individual organ-specific GVHD. Multivariate analyses identified several SNP markers may predict the risk of organ specific acute GVHD in combination with clinical factors. For skin acute GVHD, recipient PDGFD (rs10895534), donor NOS2A (rs3730017), TNFRII (rs3397) and TGFB1 (rs1800469) genotypes were predictive together with clinical factors such as HLA disparity. Donor's genotype for TNFRII (rs3397) was predictive not only for overall acute GVHD but also for skin acute GVHD. No clinical factors were identified for the risk of liver or gut acute GVHD, but several SNP markers were found including recipient PDGFRB (rs2302273), IFNGR1 (rs2234711) and donor PTGS1 (rs10306114), NOS1 (rs9658254), IL1R1 (rs2192752) genotypes for liver acute GVHD and recipient IL4 (rs2243248), donor PDGFD (rs1053861), TGFBR1 (rs420549), IL12A (rs2243115) genotypes for gut acute GVHD. In summary, there are no overlapping SNP markers for the risk prediction of organ specific acute GVHD. For organ specific chronic GVHD, 2 clinical risk factors were predictive including source of stem cells and a preceding history of acute GVHD. In addition, several SNP markers were also identified: recipient PDGFC (rs1425486), donor NFKB1 (rs1805034) and NOS2A (rs3730017) for skin chronic GVHD; recipient IL10RB (rs8178561) and PDGFRB (rs22229562), and donor TGFBR1 (rs868) for eye chronic GVHD; recipient IL12RB1 (rs3746190) and donor FCGR2A (rs1801274) for oral chronic GVHD; and donor IL4R (rs2057768), FAS (rs2234767) and TGFB1 (rs1800469) for lung chronic GVHD. Again, In no overlapping SNP markers were observed for organ-specific chronic GVHD risk. Although this predictive model could not stratify patients according to their risk of overall chronic GVHD (p=0.0763), the predictive models per each organ specific chronic GVHD enabled to stratify the patients according to their risks of each organ specific GVHD (p 〈 0.0001 for skin chronic GVHD, p=0.0033 for eye chronic GVHD, p=0.003 for oral chronic GVHD and p=0.0036 for lung chronic GVHD).Predictive models incorporating clinical and genetic factors improved the stratification power by 11.1% compared to models only including clinical factors. Conclusion: Our study suggests that SNP based approaches can predict the risk of organ-specific GVHD. These SNP markers need to be validated in other series. These SNPs may help focus studies into pathobiology and targeted therapy of GVHD in the future. 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: 2012
    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. 120, No. 21 ( 2012-11-16), p. 4184-4184
    Abstract: Abstract 4184 Background: Chronic graft-versus-host disease (cGVHD) risk score by Arora et al (Blood 2011) was successfully demonstrated to stratify patients with cGVHD according to their transplantation outcomes. In this risk score model, the following variables were included: recipient age, donor type, sex mismatch, GVHD prophylaxis, prior acute GVHD, time from HSCT to cGVHD, disease status before HSCT, platelet count at cGVHD diagnosis, serum bilirubin at cGVHD onset and performance status at diagnosis of cGVHD. However, this risk score system needs to be validated in an independent cohort. Methods: A total of 425 consecutive patients who survived beyond 100 days following allogeneic hematopoietic stem cell transplantation (HSCT) at the Princess Margaret Hospital from Jan 1996 to Oct 2007, were reviewed retrospectively and evaluated for the cGVHD risk score[J1]. Results: Out of 425 patients, 317 patients developed cGVHD according to the NIH consensus criteria of GVHD. Ten patients were removed from the analysis due to missing variables. Long-term transplant outcomes of the remaining 307 patients were evaluated using the cGVHD risk score system. First, overall survival (OS) according to the risk group (RG) were 82.5±11.3 (RG1), 76.4±3.4 (RG2), 69.4±6.5 (RG3), and 27.3±13.4 (RG4) (p 〈 0.001), while non-relapse mortality (NRM) were 10.0±9.5 (RG1), 10.4±2.4 (RG2), 21.4±5.8 (RG3), and 69.3±14.7 (RG4) (p 〈 0.001). Significantly lower OS and higher NRM was noted in RG4. However, there is no statistical difference of the transplant outcomes among 3 groups of RG1, RG2 and RG3 for OS and NRM. Next, in order to improve a stratification power of cGVHD risk score model, two other clinical variables were incorporated into the model, absolute lymphocyte count (ALC) and eosinophil count (EOS) at the onset of cGVHD. The patients with lower (ALC, 〈 1.0·109/L) and lower eosinophil counts (EOS, 〈 0.5·109/L) at the onset of cGVHD were associated with inferior OS and higher risk of NRM. In a multivariate analysis, lower ALC was confirmed to be an independent variable predicting OS (HR 1.94, 95% CI 1.14–3.28, p=0.014) and NRM (HR 2.87, 95% CI 1.35–6.08, p=0.006). Similarly, EOS was also confirmed to be an independent variable predicting OS (HR 3.27, 95% CI 1.28–8.38, p=0.014). Accordingly, these two variables (i.e. ALC and EOS) were tested in a revised cGVHD risk score model and patients were stratified into revised 4 groups: rRG1 (score 0–3), rRG2 (score 4–6), rRG3 (score 7–9), rRG4 (score °Ã10). Overall survival (OS) according to the revised risk group (rRG) were 93.3±6.4 (rRG1), 81.4±4.1 (rRG2), 68.2±4.6 (rRG3), and 32.0±1.7 (rRG4) (p 〈 0.001). Non-relapse mortality (NRM) were 0.0 (rRG1), 7.9±2.8 (rRG2), 17.2±3.8 (rRG3), and 62.2±11.8 (rRG4) (p 〈 0.001). The revised cGVHD-risk score appears to better discriminatepatients according to their transplant outcomes (particularly OS and NRM). Conclusion: The original cGVHD risk score model has been successfully validated to stratify transplant patients according to their risk of cGVHD[J2]. In addition, revised cGVHD risk score model including ALC and EOS at cGVHD onset improved prognostic stratification of the patients according to their transplantation outcomes. 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: 2012
    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. 1237-1237
    Abstract: Allogeneic hematopoietic cell transplantation (HCT) is a curative treatment option for acute myeloid leukemia (AML) when indicated. Numerous pre-transplant risk scores have been developed to predict post-transplant outcome, utilizing a variety of parameters. The purpose of this single-center study was to retrospectively develop and validate a prognostic score based on known significant pre-transplant variables for outcomes of 747 patients that underwent HCT for AML between 1978 and 2013. Median age of all patients at transplant was 44 years (range 17-71 years), 391 patients (52%) were female. HCT was performed in first complete remission (CR1) for 497 patients (67%) and in second complete remission (CR2) or advanced disease for 250 patients (33%). Donors were related for 538 patients (72%) and unrelated for 209 patients (28%). Peripheral blood stem cells (PBSC) were used as a graft source in 367 patients (49%). Myeloablative conditioning (MAC) was administered to 615 (82%) patients, 132 (18%) received reduced-intensity conditioning (RIC) regimens. HCT was performed over the time periods 1978-1990 (n=139), 1991-1999 (n=192), 2000-2006 (n=183) and 2007-2013 (n=233). Median follow-up of survivors was 90 months. Patients were assigned a combined score based on patient age, disease status and donor status. For disease status CR1, age 〈 45 years and related donors, each parameter received a score 0. For disease status CR2 or advanced stage, age ≥45 and unrelated donors, each parameter received a score 1. Patients demonstrated a cumulative score of 0 (n=197), 1 (n=326), 2 (n=179) or 3 (n=45). All 747 patients were randomized into two groups, a test cohort (n=373) and a validation cohort (n=374). Univariate analysis for the test cohort demonstrated a favorable risk group with score 0 (n=92), an intermediate risk group of score 1-2 (n=255) and an unfavorable risk group with score 3 (n=26) with a 5-year overall survival (OS) of 61%, 35% and 20% respectively (p=0.0001)(Figure). Cumulative incidence of relapse (CIR) demonstrated a marginally significant difference between groups (p=0.05) with 5-year CIR 14%, 28% and 23% respectively. Non-relapse mortality (NRM) was marginally different (p=0.07) with 5-year NRM 27%, 39% and 54% respectively. Multivariable analysis of the test cohort for OS demonstrated that the presented scoring system remained significantly prognostic (p 〈 0.0001) accounting for year of transplant as a continuous variable in the analysis (p=0.001, HR for transplant year 0.97, 95%CI 0.96-0.98). For OS, HR was 2.3 and 4.0 for the intermediate and high risk group respectively compared to favorable risk. For CIR, HR was 2.1 and 1.8 for intermediate and high risk patients respectively (p=0.05). For NRM, HR was 1.5 and 2.8 for intermediate and high risk patients respectively (p=0.01). Analysis of the validation cohort confirmed significant stratification for OS on univariate (p 〈 0.0001, 5-year OS 68%, 36% and 30% for the three risk groups respectively) and multivariable analysis (p 〈 0.0001). For CIR, no significant difference was seen on univariate (p=0.3, 5-year CIR 15%, 23% and 17% respectively) or multivariable analysis (p=0.3). For NRM, the validation cohort confirmed significant stratification between the risk groups on univariate (p 〈 0.0001, 5-year NRM 19%, 42% and 48% respectively) and multivariable analysis (p 〈 0.0001). In the presented study we developed and validated this readily calculable pre-transplant scoring system involving age, CR status and donor type which is highly prognostic for OS and NRM of patients undergoing allogeneic HCT for AML. We also demonstrated that the year transplant was performed over the last three decades had no influence on the prognostic significance of the scoring system. Figure 1 Figure 1. Disclosures Messner: Otsuka Pharmaceuticals Inc: Research Funding.
    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. 124, No. 21 ( 2014-12-06), p. 1179-1179
    Abstract: Late effects occurring post allogeneic hematopoietic cell transplantation (HCT) include the development of secondary malignancies (SM). The purpose of this single-center study was to retrospectively assess the incidence of SM in 2200 patients that consecutively underwent HCT for a variety of hematological conditions between 1970 and 2013, and to investigate parameters associated with occurrence of SM and survival post diagnosis. Median patient age at transplant was 42 years (range 17-71). Graft was bone marrow in 1310 patients (60%), peripheral blood stem cells (PBSC) in 890 patients (40%). Related donors were used in 1624 patients (74%), unrelated in 576 patients (26%). Transplants were performed for acute myeloid leukemia (AML, n=756, 34%), chronic myeloid leukemia (CML, n=486, 22%), acute lymphoblastic leukemia (ALL, n=275, 13%), non-Hodgkin lymphoma (n=174, 8%), myelodysplastic syndrome (n=159, 7%), aplastic anemia (n=128, 6%) and other conditions (n=222, 10%). Conditioning regimen was myeloablative in 1903 patients (87%) and reduced intensity in 297 patients (13%). Concerning total body irradiation (TBI) dose, 556 patients (25%) did not receive TBI, 1060 patients (48%) received 200-500 cGy and 584 patients (27%) received 〉 500 cGy. Median follow-up of survivors was 120 months (range 1-398). SM was observed in 155 patients (7% of total). Of the patients that developed SM, 39 (25% of SM) were with skin malignancy (30 with non-metastatic squamous and basal cell carcinoma), 25 (16%) with gynecological malignancies, 18 (12%) with gastrointestinal, 19 (12%) with hematological, 20 (13%) with oral squamous carcinoma, 14 (9%) with prostate, 6 (4%) with lung, 4 (3%) with thyroid and 10 (6%) with other forms of SM. Excluding patients with non-metastatic squamous and basal cell carcinoma of skin, the remaining 125 patients demonstrated a median time to SM of 99 months (range 1-393). Median time to development of SM was 4, 7 and 12 years for the age groups 〉 55, 41-55 and 〈 41 years of age respectively (p=0.0004). At 5 years post-HCT, 2.5% of patients had developed SM (95%CI 1.9-3.3), at 10 years 3.9% developed SM (95%CI 3.1-4.9) and at 15 years 6.0% of survivors had developed SM (95%CI 4.8-7.3). Analysis was performed for the effect of age at HCT, graft source, donor type, time period transplant was performed, conditioning intensity and TBI dose. In both the univariate and multivariable analysis, none of the aforementioned variables influenced cumulative incidence of SM. Concerning the survival of the 125 patients following diagnosis of SM (excluding non-metastatic squamous and basal cell carcinoma of skin), median follow-up of survivors was 37 months (range 1-344 months), survival at 10 years post-SM was 49% (SE ±6.6%). 25% of patients died of causes related to SM. Univariate analysis demonstrated a significant influence of ECOG score at diagnosis of SM (p 〈 0.0001), while age at SM, coexistence of GvHD at diagnosis, existence of other co-morbidities and time period from HCT to diagnosis of SM did not significantly influence survival. Multivariable analysis demonstrated ECOG score at diagnosis of SM as the only independent predictor of survival post diagnosis, with HR 2.8 for ECOG score 1 and HR 7.0 for ECOG score 2-4 compared to ECOG score 0 (p 〈 0.0001). In conclusion, incidence of SM post-HCT does not seem to be related to dose of TBI or conditioning intensity. Younger patients develop SM significantly later post-HCT, while a higher ECOG score at diagnosis of SM is independently prognostic of poor survival. Disclosures Messner: Otsuka Pharmaceuticals Inc: Research Funding.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1917-1917
    Abstract: Introduction: Nontuberculous mycobacteria (NTM) are ubiquitous environmental organisms that are increasingly recognized as clinically significant pathogens in the allogenic hematopoietic cell transplanted (alloHCT) population. The incidence of NTM infection post alloHCT has increased from 0.49-1.0% in early studies to 2.8-8.7% in more recent investigations, possibly due to improvements in NTM detection, varying pre-transplant conditioning regimens and regional epidemiology of different NTM species. We investigated incidence and risk factors of NTM infection after alloHCT. Methods & Patients: Medical records for 1097 consecutive patients who underwent alloHCT at Princess Margaret Cancer Centre from 2000 to 2013 were reviewed to determine the frequency, risk factors and outcomes associated with NTM infections. Clinically significant NTM infection was differentiated from colonization according to the American Thoracic Society guidelines, and was classified as pulmonary, non-pulmonary, or disseminated. Acute and chronic graft versus host disease (aGVHD and cGVHD) were diagnosed and graded using established and NIH consensus criteria respectively. The cumulative incidence of NTM was calculated considering competing risks of death. Multivariate analysis comprised Cox proportional hazards regression, modeling NTM risk. Statistical analyses were performed using EZR software (Saitama, Japan). Results: Of 1097 patients, NTM were isolated in 45 (4.1%) and judged clinically significant in 30 (2.7%). The incidence of NTM infection by competing risk analysis was 2.8% at 5 years (95% CI, 1.9-4.0%). The median (range) time to diagnosis was 343 (19-1967) days, and in 83% of patients, was diagnosed within 2 years of alloHCT. Of the 30 clinically significant NTM infections, 28 (93.3%) were pulmonary and 2 (6.7%) were disseminated. With respect to the latter group, one patient had NTM isolated from blood, while the second case was presumed disseminated based on characteristic skin findings, but with no confirmed microbiologic diagnosis. The most common species/groups isolated were Mycobacterium avium complex (n=11, 36.7%), M. xenopi (n=5, 16.7%), and M. fortuitum (n=5, 16.7%). 22/30 patients (73.3%) were on systemic immunosuppression at the time of diagnosis, and 95.7% had concurrent infections (30.4% pulmonary, 17.3% extra-pulmonary, and 47.8% both), with fungal infections occurring most frequently (53.3%). Significant risk factors (HR 95% CI) for NTM included aGVHD grades 2-4 (3.25 [1.33-7.96] p=0.036), cGVHD (3.20 [1.06-9.68] p=0.010), age (1.05 [1.02-1.07], p 〈 0.001), and CMV viremia (4.64 [1.90-11.37] p=0.001). 76.7% of patients with clinically significant NTM had a diagnosis of cGVHD (23/30), in comparison to 47.4% (520/1097) of patients without a diagnosis of NTM infection (p=0.003), and cGVHD severity by NIH global score correlated with NTM risk. Among all patients with cGVHD, severe cGVHD was present in 39% (9/23) of NTM patients, versus 17% (89/520) of non-NTM patients (p=0.012). Pre-alloHCT diagnosis (p=0.34), conditioning regimen (p=0.81), T-cell depletion (p=0.66), HLA matching (p=0.62), or donor type (p=0.63), did not reach statistical significance. Median survival duration after a diagnosis of clinically significant NTM was 398 (range, 20-764) days, with a survival rate of 40.8±10.8% at 2 years. Conclusion: Clinically significant NTM infection after alloHCT was relatively common in our study population. GVHD (acute and chronic), age, and CMV bacteremia were significant risk factors. Given a median survival of approximately 1 year following diagnosis, NTM infection may be of greater clinical significance than previously thought. A high index of suspicion for NTM infection in patients with pulmonary symptoms, particularly within 2 years after HCT and in the presence of cGVHD, may lead to prompt diagnosis and treatment, and potentially better outcomes. Disclosures Lipton: Pfizer: Consultancy, Research Funding; Ariad: Consultancy, Research Funding; Teva: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding; Novartis Pharmaceuticals: Consultancy, Research Funding. Kim:Novartis Pharmaceuticals: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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. 124, No. 21 ( 2014-12-06), p. 3928-3928
    Abstract: Introduction: The reconstitution of the immune system after allogeneic hematopoietic stem cell transplantation (allo-HCT) depends on multiple factors such as the conditioning regimen, age, stem cell source, and graft-versus-host disease (GVHD). Patient with chronic GVHD (cGVHD) can remain B-cell deficient even 1 year after allo-HCT. Although hypogammaglobulinemia post-transplant has been suggested as a poor prognostic factor for survival and transplant-related mortality (TRM), very little data has been published in the recent decade in the field of reconstitution of B-cell repertoires and hypogammaglobulinemia, particularly after the introduction of peripheral blood stem cell allo-HCT and in vivo T-cell depletion. This study aimed to identify the risk factors for hypogammaglobulinemia and evaluate the association between hypogammaglobulinemia and transplant outcomes in adult allo-HCT population. Methods: We retrospectively reviewed 339 consecutive patients who received allo-HCT between 2009 and 2012 at the Princess Margaret Cancer Centre, Toronto, Canada. At lest one measurement of immunoglobulin level was available in 157 patients between 3 months and 1 year post allo-HCT. IgG hypogammaglobulinemia (hypo-IgG) was defined as IgG 〈 7g/L. The Kaplan-Meier method was used for OS, and cumulative incidences considering competing risks were calculated for NRM and relapse. The independent student’s t-test was used to compare mean IgG levels in each subgroup. The Χ2 test was used to identify the risk factors for hypo-IgG in univariate analysis and binary logistic regression was used in multivariate analysis. Multivariate analysis for OS was performed using the time-dependent cox proportional hazard model with cGVHD as a time-dependent covariate. Fine-Gray proportional hazard regression for competing events were used for NRM in multivariate analysis. Results: The mean values of IgG were 6.67±0.41g/L (n=103) at 3 months, 6.93±0.53g/L (n=94) at 6 months, and 8.34±0.49g/L (n=136) at 1 year post allo-HCT. The proportions of patients with hypo-IgG ( 〈 7g/L) among those with available IgG level at the select time-points were 59.2% at 3 months, 59.6% at 6 months and 44.9% at 1 year. Patients with lymphoid malignancies showed a lower IgG level at 3 months than those with other diseases (5.12g/L vs 7.25gL, p=0.041). Non-T-cell depletion (non-TCD) was associated with lower IgG levels at 6 months and at 1 year: 5.83g/L vs 8.98g/L at 6 months (p=0.004) and 6.97g/L vs 10.94g/L at 1 year (p 〈 0.001). The presence of previous acute GVHD (aGVHD) grades 2-4 at 6 months was associated with a lower IgG level at 6 months (5.21g/L vs 8.66 g/L, p=0.001), but cGVHD at 6 months was not. However, a lower level of IgG at 1 year was observed among patients who developed cGVHD by 1 year than those who did not (7.79 g/L vs 10.38g/L, p=0.031). The proportion of patients with hypo-IgG at 6 months was significantly higher in the lymphoid malignancies group (78% vs 56%, p=0.049), in the non-TCD group (71% vs 39%, p=0.003), in patients with aGVHD grades 2-4 (81% vs 38%, p 〈 0.001) and with cGVHD (71% vs 50%, p=0.056). Binary logistic regression identified the following variables as independent risk factors for hypo-IgG at 6 months; non-TCD (hazard ratio (HR) of 3.61, p=0.16), aGVHD grades 2-4 (HR 8.45, p 〈 0.001) and cGVHD (HR 3.31, p=0.025). Overall survival at 2 years post allo-HCT was significantly lower in the group with hypo-IgG (n=56) than those with a normal IgG level (n=38) at 6 months (54.5% vs 86.6%, p=0.001). NRM at 2 years was also significantly higher in the hypo-IgG group than in the normal IgG group at 6 months (44.0% vs 3.6%, p 〈 0.001). There was no difference in the relapse rate at 2 years between the two groups. Multivariate analysis demonstrated that hypo-IgG at 6 months (HR 6.10, p=0.006) and aGVHD grade 2-4 (HR 3.23, p=0.31) were adverse prognostic factors while reduced-intensity conditioning (HR 0.27, p=0.028) and time-dependent cGVHD (HR 0.194, p=0.001) were associated with better OS. Furthermore, cGVHD was associated with lower NRM (HR 0.27, p=0.004) and hypo-IgG at 6 months was associated with higher NRM (HR 20.0, p=0.004). Conclusion: A significant number of patients remain hypogammaglobulinemic at 6 months and even 1 year post allo-HCT. Non-TCD and acute and chronic GVHD were identified as risk factors for hypogammaglobulinemia. Hypogammaglobulinemia at 6 months was found to adversely affect OS and NRM. 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1945-1945
    Abstract: Introduction Large granular lymphocytes (LGL) are a morphologically recognizable subpopulation of lymphocytes comprising an immunophenotypically heterogeneous population of activated CD3+ T cells and CD3- natural killer (NK) cells that mediate non-MHC-restricted cytotoxicity. Increased number of circulating LGL can be found as a response to viral infections, autoimmune disease or malignant neoplasms, as a result of chronic antigenic stimulation. Of interest, LGL lymphocytosis has been reported to occur following hematopoietic cell transplantation (HCT), with a variable incidence of up to 20%. This population display improved transplant outcomes with a lower incidence of non-relapse mortality and relapse (Kim, BMT, 2013; Nann-Rütti, BBMT, 2012). The aim of the present study is to determine the risk factors associated with the development of LGL lymphocytosis after allogeneic HCT. Methods A total of 826 patients who underwent an allogeneic HCT at Princess Margaret Cancer Centre, Toronto, Canada from 2000 to 2012 were retrospectively analyzed. LGL lymphocytosis was defined as the presence of at least two of the followings: 1) Sustained peripheral blood lymphocyte count ≥3.0 x 109/L observed in at least three consecutive determinations over a period of 2-3 months; 2) Predominance (≥30%) of LGL lymphocytes in the peripheral blood, as assessed by morphologic or immunophenotypic criteria; 3) T-cell receptor monoclonality assessed by PCR. The patient population was divided into discovery and replication sets using 2 different methods: stratified randomization and propensity score matching, using relevant baseline variables such as donor type, CMV serostatus, conditioning, T-cell depletion. Results No significant imbalances were found between the discovery and replication sets in terms of relevant baseline characteristics and clinical outcomes, for both the randomly divided patients and the propensity score matched groups. The overall incidence of LGL lymphocytosis was 14.5% at 3 years. The incidence of LGL lymphocytosis was similar across all subgroups of patients, both for the randomly divided groups and the propensity score matching (P-value not significant). A multivariable analysis of the risk factors for the development of LGL lymphocytosis was performed, including the following variables: grades 3-4 acute graft-versus-host-disease (GVHD), chronic GVHD, CMV viremia, CMV serostatus of the recipient, donor type, transplant year and T-cell depletion (TCD) for GVHD prophylaxis. In the stratified randomization analysis, the following risk factors were identified: 1) Discovery set: chronic GVHD (Hazard Ratio: 8.3, 95% CI: 3.1-22.6, P 〈 0.001), CMV viremia (HR: 2.7, 95% CI: 1.5-4.7, P 〈 0.001) and use of an unrelated donor (HR: 2.1, 95% CI: 1.2-3.7, P=0.01). 2) Replication set: chronic GVHD (HR: 38.9, 95% CI: 5.3-284.9, P 〈 0.001) and CMV viremia (HR 3.8, 95% CI: 2.2-6.6, P 〈 0.001). For the propensity score matching analysis the risk factors for the development of LGL lymphocytosis were the following: 1) Discovery set: chronic GVHD (HR: 22.9, 95% CI: 3.2-169.3, P=0.002) and CMV viremia (HR: 2.2, 95% CI: 1.1-4.2, P=0.02). 2) Replication set: chronic GVHD (HR: 28.9, 95% CI: 3.9-212.5, P 〈 0.001), CMV viremia (HR: 3.6, 95% CI: 1.9-6.7, P 〈 0.001). Conclusions Chronic GVHD and CMV viremia are strongly associated with the development of LGL lymphocytosis following allogeneic HCT. This may reflect a chronic antigenic stimulation in the setting of GVHD and CMV infection, leading to the expansion of LGL. However, the underlying mechanisms of LGL activation and expansion in the allogeneic HCT setting still remain unclear. Thus, further investigations are needed to elucidate these mechanisms in particular in the setting of GVHD. Disclosures Lipton: Pfizer: Consultancy, Research Funding; Ariad: Consultancy, Research Funding; Teva: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding; Novartis Pharmaceuticals: Consultancy, Research Funding. Kim:Bristol-Myers Squibb: Consultancy, Research Funding; Novartis Pharmaceuticals: Consultancy, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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...