Radiotherapy Post Autologous Stem Cell Transplantation in Patients with Lymphoma Not Reached Complete Response.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5473-5473
Author(s):  
Naibai Chang ◽  
Xichun Gu ◽  
Ling Zhu ◽  
Jianping Wei ◽  
Shengming Zhao ◽  
...  

Abstract Objective: Anthracycline-based chemotherapy induces 50%–70% of CR in patients with lymphoma, but only 30%–40% of long-term disease-free survival. Salvage chemotherapy with autologous stem cell rescue is required in patients with aggressive disease or never achieve CR with conventional chemotherapy,but the relapsed rate is still high. The purpose of this study was to evaluate radiotherapy post autologous stem cell transplantation in such group of patients. Methods: 15 patients who underwent autologous stem cell transplantation during 1992–1998 were enrolled in this study. Conditioning regimen was CBV (cyclophosphomide + carmustine + etoposide). Radiotherapy was started on day +50(31–90). All patients were followed up until January 2005. Kaplan-Mier survival analysis was made by using SPSS10.0 software. Results: There were 14 patients with non-Hodgkin lymphoma and 1 with Hodgkin disease enrolled. Male:female=11:4. Median age was 40 (30–64). At least 6 cycles of induction chemotherapy were given before transplantation. There were 3 patients in progression disease, 1 in stable disease, and 11 in partial remission before transplantation. Three patients received total lymph node irradiation (TLI). Seven patients received TBI(200cGY)+involved field irradiation therapy(IFIT). Five were treated with IFIT. All patients acheaved complete response(including 1 CRu) after radiotherapy. Three patients relapsed. One patient treated with TBI+IFIT relapsed at 6 months later. Two patients treated with IFIT relapsed at 8 and 36 months later respectively. The mean disease-free survival and overall survival were 10.84(SD1.37,95%CI) years and 11.89(SD1.35,95%CI) years respectively. The estimatrd 10-year disease-free survival and Overall survival were both 73%. One patient developed AML at 86 month. Grade III–IV hematologic toxicity was seen in 2 patients. Conclusions: Post ASCT radiotherapy is safe and tolerated. Relaps rate is low. Patients who received TLI or TBI+IFIT seem to have better outcome than that received IFIT only.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3115-3115
Author(s):  
Esa Jantunen ◽  
Ariane Boumendil ◽  
Alessandro Rambaldi ◽  
Marc Schapira ◽  
Jian Jian Luan ◽  
...  

Abstract Abstract 3115 Background: EATCL is a rare subtype of peripheral T-cell lymphoma frequently observed in patients with a history of celiac disease. EATCL is characterized by poor prognosis when treated with conventional chemotherapy with only 10–20 % of long-term survivors. Limited data are available on feasibility and efficacy of stem cell transplantation in this lymphoma entity. Patients and methods: The database of the EBMT was used to identify patients with EATCL who had received autologous and/or allogeneic stem cell transplantation in 2000–2007. All centres reporting these patients were contacted to receive confirmation to histopathology report/pathology review and to obtain information in regard to treatment prior to stem cell transplantation and more recent follow-up data. Results: Altogether 85 patients with EATCL were identified. Seventy-three patients had received ASCT and 12 patients allogeneic SCT. Histological report/review with additional follow-up data was available from 22 ASCT treated patients which are reported here. There were 14 females (64 %) and eight males with a median age of 55 years at the time of transplant. Half of the patients had a history of coeliac disease. The median number of treatment lines before ASCT was 1 and 50 % of the patients were in the first remission at the time of ASCT. BEAM was the most commonly used high-dose regimen (17 pts, 77 %) and all patents received blood stem cell grafts. The median time from the diagnosis to ASCT was 6 months. The median follow-up time for living patients was 45 months from ASCT. During the follow-up relapse has been observed in 13 patients (59 %), the median time was only four months from ASCT. The median disease-free survival and overall survival were nine months and 15 months, respectively. Two-year overall survival, disease-free survival, cumulative incidence of relapse and non-relase mortality (NRM) was 45%, 40%, 55% and 4%, respectively. Conclusions: ASCT is feasible in selected patients with EATCL with a low NRM. Of transplanted patients 40 % remained disease-free beyond 2 years. This seems to be superior when compared to historical experiences although selection factors should be taken into account. ASCT is a treatment option in transplant eligible EATCL patients who respond to initial therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 352-352 ◽  
Author(s):  
Nicolaus Kroeger ◽  
Victoria Panagiota ◽  
Tatjana Zabelina ◽  
Michelle Maria Araujo Cruz ◽  
Rabia Shahswar ◽  
...  

Abstract Introduction Primary or post-ET/post-PV myelofibrosis is one of the Philadelphia chromosome-negative chronic myeloproliferative neoplasia characterized by significantly reduced overall survival. More recently several mutated genes have been detected, which allow, in addition to clinical factors, to identify patients with a significantly shorter overall survival and a higher risk of transformation into acute leukemia. Allogeneic stem cell transplantation is still the only curative treatment option for patients with myelofibrosis, and due to the inherent risk of the treatment procedure careful selection of the patients is required. Molecular genetics may help to select patients for allogeneic stem cell transplantation. Patients and methods To determine the impact of mutated genes in patients with myelofibrosis who underwent allogeneic stem cell transplantation we analyzed samples from 169 patients with a median age of 58 years (r: 18 - 75) who received allogeneic stem cell transplantation either from related (n = 36) or unrelated (n = 133) donor. Stem cell source were more often peripheral blood stem cells (n = 165) than bone marrow (n = 4). The intensity of conditioning was mainly reduced intensity (n = 166), rather than myeloablative conditioning (n = 3). Patients suffered from primary myelofibrosis (n = 110), post-ET/PV myelofibrosis (n = 46), while 13 patients were in acceleration or had transformed into acute myeloid leukemia. According to dynamic IPSS (DIPSS) (n = 165) the patients were either low (n = 7), intermediate-1 (n = 35), intermediat-2 (n = 91), or high risk (n = 32). Regarding molecular genetics we found JAK2V617F mutations in 62%, calreticulin (CALR) mutations in 20%, MPL mutations in 4%, U2AF1 in 7%, SRSF2 in 10%, SF3B1 in 4%, ASXL1 in 29%, IDH1 in 2%, IDH2 in 3%, CBL in 1%, DNMT3A in 4%, TET2 in 10%, EZH2 in 4%, while none of the patients showed mutations in ETV6 and PTPN11. Overall, only in 11 patients no mutation could be detected. One mutation could be detected in 41%, 2 mutations in 30%, 3 mutations in 11%, 4 mutations in 5%. Results During follow-up 39 patients experienced relapse and 46 patients experienced non-relapse mortality. From the non-molecular factors regarding disease-free survival in univariate analysis age < 58 (p < 0.01), intermediate-1 and low risk according to DIPPS (p = 0.002), HLA-matched vs. mismatched (p = 0.04) were significant factors for improved disease-free survival. Regarding molecular markers improved disease-free survival was seen for patients with mutations in CALR (p = 0.005), while negative impact on disease-free survival was seen for mutations in U2AF1 (p = 0.035), ASXL1 (p = 0.05), IDH2 (p = 0.006), DNMT3A (p = 0.029). No significant difference could be seen for patients with EZH2, IDH1, SRSF2, and SF3B1 mutations. There was no difference in disease-free survival for patients without any mutation vs. 1, and more than 1 mutation (p = 0.12). Regarding the previously described unfavorable mutations ASXL1, SRSF2, EZH2, IDH1, and IDH2, we found 40 patients who had at least 1 of these unfavorable mutations, 11 had 2 of these mutation, and 1 had 3 of these unfavorable mutations. However, the estimated 5-year disease-free survival did not differ significantly between patients without any of these unfavorable mutations, with 1 or with 2 of them (47 vs. 40 vs. 41%, p = 0.5). Conclusions These results suggest that some molecular marker such as ASXL1, U2AF1, IDH2 and DNMT3A negatively influence DFS in myelofibrosis after allogeneic stem cell transplantation in a univariate analysis. In contrast, the poor prognosis of the recently described unfavorable mutated genes SRSF2, EZH2, and IDH1 was not observed and may therefore be overcome by allogeneic stem cell transplantation. Disclosures No relevant conflicts of interest to declare.


1999 ◽  
Vol 17 (5) ◽  
pp. 1551-1551 ◽  
Author(s):  
S. Vincent Rajkumar ◽  
Rafael Fonseca ◽  
Martha Q. Lacy ◽  
Thomas E. Witzig ◽  
Terry M. Therneau ◽  
...  

PURPOSE: To study the prognostic value of plasmablastic morphology after autologous stem-cell transplantation for relapsed or primary refractory myeloma. PATIENTS AND METHODS: Seventy-five patients were studied. Investigators blinded to the clinical details of the individual cases reviewed bone marrow aspirate slides to determine plasmablastic classification. Plasmablasts were defined using strict, well-described criteria. Plasmablastic morphology was considered to be present (plasmablastic myeloma) when 2% or more plasmablasts were present in the plasma-cell population. RESULTS: Patients underwent transplantation 5 to 88 months (median, 20 months) after the initial diagnosis of myeloma. Twenty-eight percent of patients had plasmablastic morphology. A significantly greater proportion of patients with plasmablastic morphology had abnormal cytogenetics compared with those with nonplasmablastic classification (73% v 31%, respectively; P = .003). The overall survival rate measured from the time of transplantation was significantly worse in patients with plasmablastic morphology compared with those without (median survival time, 5 months v 24 months, respectively; P < .001). Progression-free survival time was shortened also, with a median time of 4 months compared with 12 months, respectively (P < .001). In the multivariate analysis, plasmablastic classification was the most powerful prognostic factor after transplantation for both overall (P = .001) and progression-free survival rates (P < .001). We also identified three risk groups based on plasmablastic morphology: plasma-cell labeling index, lactate dehydrogenase, and cytogenetics. The median overall survival time was 38 months when none of these factors was abnormal, 17 months with one abnormal factor, and 8 months with two or more abnormal factors (P < .001). CONCLUSION: Plasmablastic morphology is a powerful independent predictor of poor survival rate after autologous stem-cell transplantation for relapsed or primary refractory myeloma.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 890-890 ◽  
Author(s):  
Julie M. Vose ◽  
Philip J. Bierman ◽  
James C. Lynch ◽  
Gregory Bociek ◽  
James O. Armitage

Abstract Several studies have now identified that the addition of rituximab (R) to CHOP chemotherapy has increased the complete response, EFS, and overall survival (OS) for patients with previously untreated DLBCL. However, concerns of increased resistance of DLBCL following failure of CHOP-R and the inability to salvage those patients with high-dose chemotherapy and autologous stem cell transplantation have been raised. In an attempt to address this issue, we evaluated 103 patients with high risk, relapsed, or refractory DLBCL receiving high-dose chemotherapy and autologous stem cell transplantation at our center between 1999 and 2003. Fifty-six (54%) of the patients received CHOP as their initial induction therapy and 47 (46%) received CHOP-R. The patients ranged in age from 20–73 years (median 50). Sixty patients were male and 43 female. Sixty two of the patients were transplanted < 12 months from diagnosis and 41 were transplanted ≥ 12 months from diagnosis. The patients had received a median of 2 prior therapies (range 1–4). The majority (81%) were chemotherapy sensitive at the time of transplantation. The median follow-up of surviving patients is 27 months ( range 3–74). All patients received a BEAM (C) - based regimen [carmustine, etoposide, cytarabine, melphalan or cycylophosphamide] +/− anti-CD20 monoclonal antibodies. There was no difference in the 2-year EFS for patients failing CHOP vs. CHOP-R (60% vs. 68%, p=0.49). In addition, there was no difference in the 2-year overall survival (OS) of patients failing CHOP vs. CHOP-R (72% vs. 68%, p=0.63). In a multivariate analysis, the only factor predicting for an event (relapse or death) post-transplant was having received ≥ 2 prior chemotherapies (Relative Risk (RR) 7.5, p=0.048) and predicting for death from any cause was chemotherapy resistance (RR 2.5, p=0.037) and an increased lactic dehydrogenase at transplant (RR 4.1, p=0.002). The additional use of a monoclonal antibody with the transplant regimen did not significantly impact outcome in this analysis. This study demonstrates that patients with DLBCL failing CHOP-R are able to be salvaged with high-dose chemotherapy and autologous stem cell transplantation and have a similar 2-year EFS and OS as patients being transplanted following CHOP failure.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3446-3446
Author(s):  
XiaoWen Tang ◽  
Marcos De Lima ◽  
Wei Wei ◽  
Alexandre Chiattone ◽  
Peter F. Thall ◽  
...  

Abstract Abstract 3446 Background: The achievement of a fully chimeric state, as opposed to mixed chimerism, has been associated with a more favorable outcome after allogeneic stem cell transplantation (allo-SCT) for leukemia. When using the reduced-toxicity IV Busulfan-Fludarabine (Bu-Flu) regimen (de Lima et al, BLOOD 2004;104:857-64) we were intrigued by a seemingly high incidence of early (day+30) mixed chimerism, yet a low incidence of serious toxicity, GvHD and high overall and disease-free survival, especially for patients transplanted in (any) remission (CR). We hypothesized that the introduction of highly sensitive PCR-based chimerism assessment technique, as well as separately assaying myeloid- and T-cell chimerism might provide more reliable data for assessing the prognostic value of chimerism in reference to overall (OS) and disease-free survival (DFS) after allo-SCT. Patients and methods: Chimerism assay was performed with PCR-based technique on informative loci, and multi-variate Cox models including chimerism and other covariates were fit for OS and DFS (See Table 1). Results: 206 AML/MDS patients were treated on two consecutive protocols with Flu at 40 mg/m2 daily for 4 days, each dose followed by IV Bu at 130 mg/m2 or pharmacokinetically targeted to an average systemic exposure of 6,000 mcMol-min. Recipients of an unrelated or one-Ag mismatched related graft received rabbit-ATG at a total dose of 4 mg/kg on days -3 to -1. GvHD prophylaxis was Tacrolimus with mini-dose MTX (5 mg/m2) on post-transplant days 1, 3, 6, and 11. There were 98 females and 108 males at a median age of 47 years (range 16–66). Sixty-six patients were in CR1, 48 in CR2, 18 had 1st chemotherapy-refractory relapse, 20 were in 1st or 2nd untreated relapse, 37 had primary induction failure, while 17 had high-risk MDS. One patient died before day 30, without chimerism studies, and 11 recovered with refractory leukemia. Median follow-up of patients still alive is 5.5 yrs (range 1.3–8.6). 193 patients who engrafted and were in CR on day +30 had chimerism analysis performed, 64% were full donor chimeras, and 36% had mixed chimerism (≥1% remaining host cell-derived DNA). As expected, being in CR prior to SCT and, if transplanted with active disease, to engraft and remain in CR or to achieve CR, respectively, were important predictors for survival. A cytogenetic “bad” prognostic subgroup (e.g. -5/-7), was of adverse importance. However, in the multivariate model neither higher age, up to age 65, or attainment of full vs. mixed donor chimerism by day +30 were of additional predictive value for either OS or DFS. (See Table 1). Conclusion: When the reduced-toxicity IV Bu-Flu regimen is used as conditioning therapy for AML/MDS only cytogenetic subgroup (Bad/others) and disease state (CR/No CR) at the start of conditioning therapy influenced DFS and OS. Neither patient age nor attaining complete chimerism on BMT day +30 were independently predictive of an altered prognosis in reference to OS and DFS. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2508-2508
Author(s):  
Norbert Claude Gorin ◽  
Myriam Labopin ◽  
Simona Piemontese ◽  
Fabio Ciceri ◽  
Arnon Nagler ◽  
...  

Abstract Adult patients with acute leukemia in need of a transplant but with no genoidentical donor, are nowadays usually considered upfront for an alternative donor transplant, while autologous stem cell transplantation is seldom done. The purpose of this study was to compare the outcome after T cell-replete haploidentical transplant and autologous transplant. We used data from the European Society for Blood and Marrow Transplantation (EBMT) registry. From January 2007 to December 2012, 234 adult patients with acute leukemia, transplanted with marrow from a haploidentical family donor, and 2259 autografted patients were reported to the EBMT registry. We performed a matched pair analysis on 188 haploidentical and 356 autologous transplants using age, diagnosis, disease status, cytogenetics, and interval from diagnosis to transplant as matching factors. The median follow-up was 15 months. The two-year non-relapse mortality rate was higher post-haploidentical transplant ( 27% versus 4%; p <10-5 ) [hazard ratio (HR): 0.16, p<0.0001], and the relapse incidence was higher after autologous transplantation ( 49% versus 31%, p <10-4) (HR: 1.69, p=0.02). Regarding LFS, there was no difference between haploidentical transplants and ASCT (Figure 1 A and 1 C). Overall survival was higher after autologous transplantation (HR: 0.69, p=0.02) with 63% versus 53% (p=0.01) in all patients, 67% versus 54% (p=0.006) in patients transplanted in first complete remission (Figure 1B), and 45% versus 47% in second complete remission (Figure 1 D). Thirty-six patients with Ph+ ALL received an autograft and 19 received a haploidentical transplant. ASCT was associated with better NRM (8% versus 46%; p=0.002), LFS (53% versus 22%; p= 0.01), and OS (70% versus 19%; p 0.001), while RI was identical (39% versus 32%; p=0.83). When comparing haploidentical transplants performed in “haplo expert” centers to ASCT, there was no difference in LFS (haplo: 53% versus ASCT 46%, p=0.17) and OS (haplo: 59% versus ASCT 63%, p=0.58) (Figure 2). These findings indicate that autologous stem cell transplantation remains a valuable therapeutic option for patients with no identical donors. Figure 1: Leukemia-free survival and overall survival in patients transplanted in first (A, B) and second remission (C, D). Overall survival in patients transplanted in CR1 (C) is significantly higher following autologous stem cell transplantation (p=0.006 ). Figure 1:. Leukemia-free survival and overall survival in patients transplanted in first (A, B) and second remission (C, D). Overall survival in patients transplanted in CR1 (C) is significantly higher following autologous stem cell transplantation (p=0.006 ). Figure 2: Leukemia-free survival of patients autografted and patients receiving a haploidentical transplant in “haplo expert” versus regular transplant centers. Figure 2:. Leukemia-free survival of patients autografted and patients receiving a haploidentical transplant in “haplo expert” versus regular transplant centers. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4267-4267
Author(s):  
Xiaoqin Feng ◽  
Zhiyong Peng ◽  
Yuelin He ◽  
Chunfu Li ◽  
Yongsheng Ruan ◽  
...  

OBJECTIVE: Pre-transplant chemotherapy can control progression of primary disease, alleviate disease and improve disease-free survival after transplantation in juvenile granulocyte leukemia (JMML).Methylation abnormalities play an important role in the development of JMML. This study was to explore the effectiveness of induction regimen including decitabine ,cytarabine and fludarabine before transplantation and to explore the affection to transplantation in JMML. METHODS: A retrospective analysis of the remission and survival of 33 children with JMML before and after stem cell transplantation in the Department of Pediatrics of Nanfang Hospital from 2014.2 to 2019.7. There were fourteen girls and 19 boys, median diagnosis age 23 months(2m-10 years old). Median white blood cell count,median hemoglobin level,median platelet count( WBC) was 29.3G/L (6.29-158.66G/L); 83g /L(41-113g/L,); 27.17G / L(4-431G / L) respectively. Median Hemoglobin F level was 34.16%( 1.56-78%). Median spleen level under the costal margin was 5cm(0-13.3cm); Median liver under the costal margin was 3.9 cm(0 -8.9 cm) .There were 26 cases with the pulmonary involvement (26/33, 78.8%). The original blast cells in bone marrow were 0-8.8%, with a median of 4.5%.Mutant genes including: 2 cases of KAS, 9 cases of NF1, 2 cases of NRAS, 16 cases of PTPN11, and 4 cases without common JMML gene .The first course of treatment after diagnosis is 20 mg/m2 × 5 days of decitabine. The second course of treatment is DA: decitabine 20 mg/m2 × 5 days + cytarabine 100 mg/m2 × 5 days or A-3V regimen: Ara-C 100 mg/m2/d CIV×7 days+Etoposide 100 mg/m2/d ×5 days+Vincristine 1.5 mg/m2/d ×1 day. The third course of treatment is decitabine 20 mg/m2 × 5 days,then FLAG regimen: Fludarabine 30mg / m2 × 5 days, Ara-C 1 g / m2 × 5 days, G-CSF 5μg / Kg × 6 days. Single drug of decitabine 20 mg/m2 × 5 days was adminstered 1-3 times per monthly during the period of waiting for transplantation. Comprehensive assessment was performed before transplantation. Survival outcomes were analyzed by Kaplan-Meier curves. RESULTS: At least 3 courses of chemotherapy were completed in 33 cases, including 1-5 courses of decitabine.The bone marrow was evaluated in 31 patients before transplantation: 12 patients got complete remission(CR) and 19 patients got partial remission(PR) .Peripheral blood evaluation: 23 cases achieved WBC CR; 17 cases achieved platelet CR, 6 cases achieved platelet PR, 7 cases had no improvement. Eighteen cases of spleen were evaluated, of which only 3 cases were CR, 13 cases were PR, and 2 cases did not improve. Overall assessment, only 1 case achieved CR before transplantation, 1 case did not improve, and 31 cases achieved PR .Allogeneic hematopoietic stem cell transplantation was performed in 33 cases, including 3 cases of nonrelated peripheral blood hematopoietic stem cell transplantation(PB HSCT), 30 cases of complementary transplantation (haploid identical PB HSCT plus non-related cord blood transplantation). The median follow-up time after transplantation was 22m(3-63m).There was no death in the period of chemotherapy before transplantation. Four patients relapsed after transplantation, and One patient died of transplantation related death. Three years EFS was 80.6%. Conclusion: Pre-transplant chemotherapy based on regimen of decitabine + cytarabine + fludarabine is safe, effective and feasible. The response rate of treatment is 97.0%, although the complete remission rate before transplantation is low, most of them are partial remission, but the disease-free survival after non-related HSCT or complementary transplantation could reached to 80.6%. These results indicated that pre-transplant chemotherapy based on regimen of decitabine + cytarabine + fludarabine was benefit to improved the survival of children with JMML after HSCT. Figure 1 Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document