Four Drugs Induction Therapy (fludarabine, cytarabine, idarubicin and gemtuzumab ozogamycin) for the Treatment of Elderly Acute Myeloid Leukemia Patients.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1027-1027
Author(s):  
Stefania Paolini ◽  
Sarah Parisi ◽  
Anna Candoni ◽  
Pier Paolo Piccaluga ◽  
Michele Gottardi ◽  
...  

Abstract Abstract 1027 Poster Board I-49 Background: Current therapeutic strategies for the treatment of elderly acute myeloid leukemia (AML) patients are still unsatisfactory. Induction regimens relying on anthracycline and cytarabine combination (3+7) still represent the standard reference therapy, with complete remission (CR) rate of 35-60% but no more than 10-20% of patients living more than three years from diagnosis. The addition of other drugs to overcome resistance and prevent subsequent relapse, has often shown greater toxicity without improving results. Aim We designed a phase II study aiming to assess toxicity and efficacy of My-FLAI regimen in patients with newly diagnosed AML aged more than 60 years. Fifty-one patients were enrolled with a median age of 68 years (range 60-76). The median leukocyte count was 10.2 ×109/L (range 0.7-222.7). Twenty-five patients had a secondary AML and 31% had a complex kariotype. Fludarabine, cytarabine and idarubicin were administered for three consecutive days at the daily dose of 25 mg/m2, 1 g/ m2 and 5 mg/ m2 respectively. Gemtuzumab ozogamycin (Mylotarg, My) was infused at day four at the dose of 5 mg. Patients achieving a CR after the induction course were planned to receive a second identical course of My-FLAI. Granulocyte colony-stimulating factor (G-CSF) was administered at physician's discretion to promote granulocytic recovery if clinically indicated. Results: Twenty-seven patients achieved a CR (one of these with an incomplete platelet recovery) and 4 obtained a partial response (marrow blast 5-19%) for an overall response rate (ORR) of 61%. Seventeen patients (33%) showed a resistant disease and three (6%) died during induction (DDI). Seventeen patients received an identical consolidation course. Seven patients were treated with cytarabine-based consolidation and the remaining 27 patients were not treated furthermore. The disease-free survival (DFS) and overall survival (OS) were 6 months (range 2.4-48.0) and 13 months (range 1.1-61.5) respectively. DFS appear to be more favorable in patients receiving additional therapy after two My-FLAI cycles (IDAC n=3; ASCT n= 4; experimental drugs n= 1; My n=4) in comparison to patients no further treated, being 12.8 (range 4.7-18.0) and 5.9 (range 2.4-48.0) months respectively. The median duration of G-CSF administration was 6 days (range 0-35) in induction and 8 days (range 0-15) in consolidation cycle. All patients developed a grade IV hematological toxicity. Median time to ANC recovery (>1 ×109/L) was 21 days (range 10-52) after induction course and 18 days (range 12-20) after consolidation course. Median time to PLT recovery (PLT>20 ×109/L and PLT>100 ×109/L) was 18 days (range 9-54) and 23 days (range 15-78) after induction and 18 days (range 12-20) and 32 days (range 21-110) after the consolidation course. Thirty-six patients had infectious complication; 33 experienced a microbiologically documented infection (n=20 gram+, n=12 gram- n=1 candidemia). Eighteen patients had a fever of unknown origin (FUO) and 12 patients suffered from pulmonary infection (n=2 bacterial; n=8 probable fungal; n=2 aspergillosis). One patient developed a maxillary sinus aspergillosis. Grade III/IV haemorrhagic complications were observed in 5 patients. No severe gastro-intestinal adverse events were observed. With a median follow-up of 8.8 months (range 0.5-61.5) 41 patients died. Five patients died in first CR for cardiac failure (n=2) and for infective complication (n=3). The remaining died for disease. Nine patients are alive, four of them in first CR. Conclusions: The four drug regimen My-FLAI is overall well tolerated in an elderly AML population. However, the efficacy did not appear to be superior to that of standard “3+7” regimen. Acknowledgments: Supported by BolognAIL, AIRC, European LeukemiaNET, COFIN, FIRB 2006, Fondazione del Monte di Bologna e Ravenna Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4735-4735
Author(s):  
Liu Xiaoli ◽  
Xu Na ◽  
DU Qingfeng ◽  
Xu Dan ◽  
Meng Fanyi ◽  
...  

Abstract Abstract 4735 Purpose: Monosomal karyotype (MK) refers to the presence of two or more distinct autosomal monosomies or a single monosomy associated with a structural abnormality. To analyze the prognosis of cytogenetic components of a complex karyotype or Monosomal Karyotype in acute myeloid leukemia (AML) except acute promyelocytic leukemia(APL). Patients and Methods:Cytogenetics and overall survival (OS), Disease free survival(DFS) were analyzed in 551 AML patients age 14 to 60 years in our center.Results: There ware 235 patiets with cytogenetic abnormalities, 25 cases with inv(16)(p13.1q22) or t(16;16)(p13.1;q22),and 63 cases with t(8;21); 31 cases (13.2%)met the criteria for MK and 39 cases (16.6%) had a complex karyotype without monosomies. OS was significantly inferior in patients with MK compared with those with a complex karyotype without monosomies (P<0.01;HR 1.85,95% confidence interval(95%CI),0.95-2.81). There was no difference between MK cases with complex karyotype cases in DFS (P>0.05□GHR 3.42,95% confidence interval(95%CI),2.96-6.70). There was significant difference in regardless of whether OS or DFS between MK+ patients with MK− patients (P<0.01). Conclusion: MK was one of independent risk factor in AML patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4294-4294
Author(s):  
Jianda Hu ◽  
Tingbo Liu ◽  
Chunxia Cai ◽  
Xinji Chen ◽  
Buyuan Chen

Abstract Abstract 4294 Advances in effective chemotherapy have improved clinical outcomes in acute leukemia in recent years. 5-year survival rate approaches 50–60% for acute myeloid leukemia (AML). However, the prognosis remains poor for patients who are relapsed or refractory to first-line therapy. Drug resistance and early disease recurrence are major contributing factors in the limited survival of patients with AML. The strategy for treating these patients is through reinduction chemotherapy followed by allogeneic stem cell transplantation. New combinations of different agents were employed in refractory patients to overcome drug resistance. The current study is to evaluate the efficacy of a MAH regimen comprising Mitoxantrone,Ara-C and Homoharringtonine in refractory or relapsed AML. 37 patients aged 14–65 years with refractory or relapsed AML (15 refractory AML patients, 22 relapsed AML patients) were treated with the MAH regimen(Mitoxantrone 10mg qd, iv.gtt, for 2□‘3 days;Ara-C 100mg bid, iv.gtt, for 5□‘7 days; Homoharringtonine 4mg qd iv.gtt, for 5□‘7 days). Chemotherapy duration lasted for 5 or 7days depended on bone marrow cellurarity. 15 (40.5%)and 1 (2.7%) patients achieved complete remission (CR) and partial remission (PR) respectively. The overall response rate was 43.2%. There was no relation between remission duration and previous chemotherapy. All patients who achieved CR received a consolidation and intensification therapy. The median overall survival (OS) for all patients was 97 days (range 18–487 d). For the patients who were in CR or PR,the median relapse-free survival(RFS) was 147 days(range 4 to 341 d). All patients experienced profound myelosuppression. The most common observed side effect of the regimen was infection because of grade ‡W neutropenia, which could be observed in 33 patients(89.1%). 4 patients died in aplasia due to severe infection and brain hemorrhage. In patients achieving remission, the median time to reach absolute neutrophil count (ANC) more than 0.5×109/l was (16.0±6.4)d. Platelet levels of more than 20×109/l were achieved in a median time of (12.7±6.2)d. Nonhematological side effects, consisting mainly of gastrointestinal toxicity(21/37,56.8%) and transient liver ALT and AST increase (4/37), were generally mild to moderate and tolerated. To a conclusion, MAH regimen can be employed in treatment of the refractory or relapsed AML patients who were not responded to other regimen. It is effective and is good tolerant.It could provide some refractory patients the chance to receive hematopoietic stem cell transplantation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 363-363
Author(s):  
Jing Qi ◽  
Qi Cai ◽  
Sandeep Singh ◽  
Ling Li ◽  
Hongjun Liu ◽  
...  

Abstract The inv(16)-created CBFβ-SMMHC fusion protein inhibits differentiation of hematopoietic stem and progenitor cells (HSPCs) and creates pre-leukemic populations predisposed to acute myeloid leukemia (AML) transformation. However, the molecular mechanism underlying the leukemogenic function of CBFβ-SMMHC has been elusive. Given the low TP53 mutation rate in AML, alternative mechanisms disrupting p53 function are expected. We showed thatCBFβ-SMMHC impairs p53 acetylation and p53 target gene activation through formation of an aberrant protein complex with p53 and HDAC8 (Blood, 120: A772; 122(21): 224). We now show that CBFβ-SMMHC binds to p53 and HDAC8 independently through distinct regions and that HDAC8 mediates the deacetylation of p53 associated with CBFβ-SMMHC. In addition, we generated mice carrying a floxed Hdac8 (Hdac8f) allele and crossed with Cbfb56M/+/Mx1-Cre (Kuo YH et al, Cancer Cell 2006). Deletion of Hdac8 signifiacntly (p<0.0001) reduced the incidence of AML and prolonged disease-free survival. Pharmacologic inhibition of HDAC8 activity with HDAC8-selective inhibitors (HDAC8i) reactivates p53 and selectively induces apoptosis of inv(16)+ AML CD34+ cells while sparing normal HSPCs. To test the effect of HDAC8i on LSC engraftment and leukemia-initiating capacity, we generated Cbfb56M/+/Mx1-Cre mice with a Cre-reporter line expressing tdTomato fluorescence protein following Cre-mediated recombination. AML cells (dTomato+/cKit+) treated with HDAC8i (22d) ex vivo showed reduced engraftment (p=0.025) and enhanced survival (p=0.025) in transplanted mice. To examine whether HDAC8i 22d treatment affects the engraftment capacity on surviving cells, we transplanted equal number (2 x 106) of AML cells treated with either 22d or vehicle in another cohort of mice (n=4). We show that HDAC8i 22d treatment reduced the engraftment of dTomato+/cKit+ AML cells and enhanced survival, suggesting that the engraftment capacity is altered in addition to reducing AML cell survival. We next performed preclinical studies to determine the efficacy of in vivo administration of HDAC8i 22d. AML transplanted mice were randomized into two groups, one group treated with vehicle and the other treated with HDAC8i 22d for 2 weeks. Flow cytometry analysis revealed significantly reduced frequency (p=0.0097) and number (p=0.0101) of dTomato+/cKit+ AML cells in the bone marrow and spleen of 22d treated mice compared to vehicle treated group. To further assess the impact on LSC activity, we transplanted bone marrow cells from these treated mice into secondary recipients and analyzed for AML engraftment. Significant reduction in the frequency (p<0.0001) and the number (p=0.0006) of dTomato+/cKit+ AML cells was observed in the bone marrow and spleen. Furthermore, HDAC8i 22d treated transplants showed no signs of leukemia while vehicle treated transplants are moribund with aggressive AML. These results indicate that HDAC8 inhibition by 22d treatment effectively eliminates engraftment and leukemia-initiating capacity of AML LSCs. In conclusion, our studies identify a novel post-translational p53-inactivating mechanism and demonstrate selective HDAC8 inhibition as a promising approach to target inv(16)+ AML LSCs. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 21-21
Author(s):  
Can Can ◽  
Lu Ding ◽  
Yuxin Tan ◽  
Balu Wu ◽  
Dongdong Zhang ◽  
...  

Objective: To investigate the efficacy and safety of recombinant human thrombopoietin(rhTPO) in the treatment of thrombocytopenia after chemotherapy for acute myeloid leukemia in vivo and in vitro. Methods: A retrospective collection of 95 patients with acute myeloid leukemia who suffered thrombocytopenia after chemotherapy in Zhongnan hospital from July 2014 to April 2019 were collected. The patients were divided into two groups according to different medications, one group was treated with reorganized human thrombopoietin, the other one with recombinant IL-11, the platelet counts between two groups was analyzed and the platelet recovery under different ages, medical conditions and other factors were analyzed; mice patient derived xenograft model of acute myeloid leukemia with thrombocytopenia after chemotherapy was established to verify the efficacy and the mechanism of rhTPO. Results: Since the platelet counts began to recover, the platelet increase in rhTPO group was significantly greater than that in IL-11 group, the platelet count is significantly higher than IL-11 group at Day42 (P&lt;0.05). Age and whether it is chemotherapy-naive patient has no significant effect on the function of rhTPO in restoring platelet counts. It is verified that rhTPO can increase platelet counts, but has no significant effect on leukocytes, erythrocytes. The injection of rhTPO leads to more synthesis of TPO in liver by activating the JAK2-STAT3 pathway. Conclusion: Our work has paved the way for further study on the clinical application of rhTPO in managing thrombocytopenia for AML after chemotherapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1003-1003 ◽  
Author(s):  
Isabel Granada ◽  
Salut Brunet ◽  
Montserrat Hoyos ◽  
Dolors Costa ◽  
Anna Aventín ◽  
...  

Abstract Abstract 1003 Poster Board I-25 Introduction: Recently, the cooperative group HOVON-SAKK has refined the prognostic impact of cytogenetic abnormalities in acute myeloid leukemia (AML) by introducing the concept of monosomal karyotype (MK). This consists of ≥ 2 autosomal monosomies or one autosomal monosomy in addition to a structural alteration. In their experience, MK would explain the poor prognosis of AML with a complex karyotype. Objective: To investigate the prognostic impact of MK in patients with primary (de novo) AML enrolled in the Spanish CETLAM group protocols (AML 94/99/03). Also, to determine whether considering MK added predictive value to the cytogenetic classification of the Medical Research Council (MRC). Methods: Retrospective analysis of data from 1149 AML patients. Chromosomal formula was centrally reviewed with karyotypes being classified by the presence of MK and allocated into the MRC risk categories. Complete remission (CR) rate, disease-free survival (DFS) and overall survival (OS) were calculated. Results: The karyotype was assessable in 904 (79%) of the 1149 cases. In 145 of the 904 cases (16%), abnormalities involving CBF gene were detected and in 437 (48%) the karyotype was normal (NK). In 253 (28%) additional patients the karyotype was not monosomal; of them, 61 (24%) belonged to the unfavorable MRC with 17 cases harboring a complex karyotype ≥ 5 abnormalities, 7 cases with rearrangements 3q, 13 cases with -7, 9 cases with 5q abnormalities and 16 cases with t(6;9)). The remaining 69 (7.7%) patients had a MK; of them, 59 (85.5%) were from the unfavorable MRC category and included 43 cases with complex karyotype ≥ 5 abnormalities, 6 cases with rearrangements 3q, 5 cases with -7, 5 cases with alterations of 5q). The following table summarizes the results in terms of CR rate, DFS and OS: Conclusions: The addition of MK to the MRC cytogenetic classification refines the prognostic prediction. In our series, the dismal outcome of patients with MK is confirmed; these patients had worse prognosis than those with adverse cytogenetics without MK. Alternative treatment strategies are mandatory for MK+ patients. Supported in part by grants: GR1-01075, ECO07/90065, PI080672 and RD06/0020/0101. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4932-4932
Author(s):  
Xuejie Jiang ◽  
Zhixiang Wang ◽  
Changxin Yin ◽  
Guopan Yu ◽  
Jieyu Ye ◽  
...  

Abstract Introduction. Most acute myeloid leukemia (AML) patients with adverse prognosis either fail to achieve complete remission (CR) or relapse after short-term remission, new strategies are needed to improve therapeutic effect in these patients. Gene silencing via DNA methylation are frequent and reversible in high-risk AML, it provides the possibility to treat AML patients with DNA-hypomethylating agent. Decitabine alone has limited anti-leukemia effect and minimal toxicity. Studies demonstrated that decitabine prior to chemotherapy was likely to increase efficacy in AML. In this study, we investigated the effect of decitabine on susceptibility to cytotoxic drugs in chemoresistant AML cells. Efficacy and safety of decitabine prior to HAA regimen was evaluated in refractory, relapsed and high-risk AML patients. Methods. HL60, HL60/ADR, Kasumi-1, and primary AML cells were pre-treated with 1.0 μM decitabine for 72 hours, Sensitivities to harringtonine, adriamycin, cytarabine and the combination was determined by MTT, apoptosis was analyzed by flow cytometry. Protein expression was detected by western blotting. In clinic, Twenty-three patients were enrolled in decitabine prior to HAA regimen, and twenty-four patients in HAA alone. CR ratio was used to evaluate efficacy. Durations of neutrocytopenia or thrombcytopemia and infection incidence were observed to assess the safety. All of patients were followed up to 36 monthes, overall survival (OS) and disease-free survival (DFS) were calculated. Result. Decitabine increased sensitivity and apoptosis induced by harringtonine in HL60/ADR, as well as adriamycin and cytarabine in HL60/ADR and Kasumi-1 cells. But no change in sensitivity to each drug was observed in HL60, and sensitivity to harringtonine in Kasumi-1. The similar changes in sensitivity to adriamycin and cytarabine were also observed in primary AML cells from refractory patients (n=6). Cytotoxic effect of HAA was alsoincreaed by decitabine in HL60/ADR and Kasumi-1 cells (p=0.004, 0.018). Western blotting showed that decitabine decreased expression of DNMT1, activation of p53 and inhibition of c-Myc, Survivin and Bcl-2. In clinic, 16 (69.6%) patients in decitabine plus HAA regimen responded to the first course of induction therapy: 14 CR (60.9%) and 2 PR (8.7%). 1 in 2 patients with PR achieved CR after second course induction. It brought the overall CR was 65.2%. Otherwise, 10 (45.8%) patients in HAA regimen alone responded to the first induction: 7 CR (29.2%) and 4 PR (16.7%), 3 in 4 patients with PR achieved CR after the second induction, and overall CR reached 41.7%. The first-cycle CR ratio in decitabine plus HAA was higher than that in HAA alone (p=0.041). Patients continued to receive chemotherapy alternatively combined with decitabine. The median OS was 14 months in chemotherapy plus decitabine, 6 months in chemotherapy alone, and median DFS were 18 and 5 months in the former and latter. No significant difference was observed in the time of neutrophil (p=0.832, 0.631) or platelet recovery (p=0.798, 0.544) after induction and intensification therapy. The incidences of infection were also similar (p=0.724, 0.697). The clinic data indicated that decitabine plus HAA regimenit was efficacy and well-tolerated to treat refractory AML patients. Conclusion. Our results demonstrated that decitabine increased cytotoxic effect against chemoresistant AML cells. Combination of decitabine and HAA was safe and efficacy to treat refractory/relapsed AML. These findings support clinic protocols based on decitabine prior to chemotherapy to overcome resistance and improve therapeutic efficacy in AML patients. Disclosures Carter: PrismBiolab: Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2595-2595
Author(s):  
Wenjuan Yu ◽  
Liping Mao ◽  
Jiejing Qian ◽  
Wenbin Qian ◽  
Haitao Meng ◽  
...  

Abstract Abstract 2595 Patients with refractory and relapsed acute myeloid leukemia (AML) have relatively unfavorable prognoses and require innovative therapeutic approaches. We evaluated the efficacy and toxicity profiles of homoharringtonine (HHT), cytarabine (Ara-c) and aclarubicin combination chemotherapy in 46 refractory and/or relapsed AML patients between February 2004 and December 2009. Eleven cases were primary refractory disease and rest of them had relapsing disease. After one course of HAA treatment, complete remission (CR) was achieved in 35 of 46 patients (76.1%), while 3 cases showed partial remission (PR) (6.5%) and 8 cases (17.4%) had resistant to this regimen. One PR and one NR patient obtained CR after second course of HAA, yielding a total CR rate was 80.4%. Hematological toxicity was the most prominent toxicity of this regimen. Grade IV hematologic toxicity occurred in 91.3% cases. Neutrophil recovery time was 13 (5–60) days. Nonhematologic complications were mild to moderate nausea, vomiting, and mucositis and could be controlled by routine measures. Early death (ED) did not occur in these 46 patients. There was no correlation between response rate and leukemia subtype (AML FAB subtype), leukocyte, blast count in bone marrow, platelet count and sex (P >0.05). However, poor karyotype and age (≥45 years old) were factors associated with increased risk of treatment failure. Of the 46 patients, one patient, who upon achieving CR after the first re-induction therapy refused further treatment, was ultimately lost to follow-up. The median overall survival rate (OS) was 29 (2–86) months in 45 cases. Relapse-free survival (RFS) was 36 (0.5–85) months in 36 CR cases. By the time of closeout, the estimated 3-year OS rate was 42% for all 45 cases, and 43% estimated RFS at 3 years for the 36 cases of CR. OS and RFS were influenced by leukemia-related factors and disease status: Hb <10g/dl and poor karyotype in both univariate and multivariate analyses. Allogeneic stem cell transplantation was performed in 7 patients and all achieved CR, but 1 patient died of GVHD, 2 died of pneumonia, 1 relapsed followed by death and another 3 are still alive with CR. At present, 19 patients are still alive and 18 of these are in continuous remission with one alive with leukemia. The rest (26/45) of the patients are all deceased. In conclusion, HAA is a good choice in cases with refractory/relapsing AML for salvage chemotherapy, preferably with a high efficacy and low-toxicity profile. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3122-3122
Author(s):  
William Arcese ◽  
Raffaella Cerretti ◽  
Stella Santarone ◽  
Gottardo De Angelis ◽  
Pasqua Bavaro ◽  
...  

Abstract Abstract 3122 The outcome of high-risk acute myeloid leukemia (AML) patients not undergoing an allogeneic transplantation is extremely poor. Therefore transplantation from haploidentical donor represents an alternative option for these patients on urgency to be transplanted. We report the results of a study on unmanipulated, G-CSF primed, haploidentical bone marrow (BM) transplantation in patients with high-risk AML lacking a suitable HLA-identical donor. Materials and methods: Between August 2005 to December 2011 58 patients (median age: 44 yrs, range 5–71) with very high-risk AML (CR1=32; CR2=16; advanced stage=10) underwent BM transplant from haploidentical donor. As pretransplant regimens, 43 patients were conditioned with a myeloablative regimen (MAC), while 15 patients received a reduced intensity conditioning (RIC). Of the 58 patients, 43 received the chemotherapy based regimen consisting of Thiotepa, i.v. Busulphan and Fludarabine (TBF MAC or RIC protocol). All 42 patients received an identical GvHD prophylaxis consisting of pretransplant ATG combined with CSA, MTX, MMF and Basiliximab, an anti-CD25 monoclonal antibody. Donors were primed with G-CSF at 4 microgr/Kg/d for 7 consecutive days. BM was harvested on day 0 and infused unmanipulated. Results: The median number of total nucleated, CD34+ and CD3+ cells infused was 7.2 (1–28)x108/kg, 2.04 (0.8–11)x106/Kg and 2.9 (0.9–6.7)x107/Kg, respectively. Five patients died early. All 53 evaluable patients engrafted at a median of 21 (13–29) days and the cumulative incidence (CI) of neutrophil engraftment was 100% at 30 days. For 53 evaluable patients, acute GVHD was absent or just grade I in 25 (47%). The 100-day CI of II-IV and III-IV grade acute GVHD was 34+/−0.4% and 12+/−0.2% respectively. Extensive chronic GVHD occurred in 4 (8%) out of 49 evaluable patients and the 2-year CI of extensive chronic GVHD was 13+/−0.4%. The 1 and 5-year CI of transplant-related mortality (TRM) was 32+/−0.4% and 34+/−0.4% respectively. The overall CI of relapse was 20+/−0.4% at 1-year and 34+/−0.7% at 5-year. The overall and disease-free survival probability was 61+/−6% and 54+/−7% at 1 year, 49+/−7% and 42+/−7% at 5 years. For patients in early stage of disease (CR1+CR2: n=48) the 1 and 3-year probability of overall survival was 70+/−7% and 58+/−8%. Conclusions: Haploidentical transplant using G-CSF primed, unmanipulated BM is correlated with high engraftment rate, low incidence of acute and chronic GVHD, acceptable TRM and favorable outcome. This approach represents a valid and feasible alternative to transplant from matched unrelated donor or cord blood for high-risk AML patients particularly on urgency to be transplanted. Disclosures: No relevant conflicts of interest to declare.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 6602-6602
Author(s):  
Carlos Enrique Vigil ◽  
Elizabeth A. Griffiths ◽  
James E Thompson ◽  
Wei Tan ◽  
Jessica Greene ◽  
...  

6602 Background: The outcome of acute myeloid leukemia (AML) patients (pts) ≥60 years (yrs) old remains poor, with a median overall survival of a few months. Neither the addition of other drugs during induction of the traditional 7+3 regimen nor an increase of cytarabine or daunorubicin doses has improved their overall survival. We have therefore designed a combination of clofarabine and daunorubicin. Aims: Determine the safety and efficacy of clofarabine and daunorubicin in pts ≥ 60 yrs of age. Methods: Induction-clofarabine 20 mg/m2 /d in a 1-hour IV for 5 days followed 3 hrs later by daunorubicin 50 mg/m2 IV over 5 minutes on days 1, 3, and 5. Pts who did not achieve complete remission (CR) or CR without platelet recovery (CRp) were eligible for a second round of induction as above. Consolidation - 2 cycles of clofarabine 20mg/m2 /d for 3 days and daunorubicin 50 mg/m2/day on days 1 and 3. Results: Twenty-one pts were enrolled. The median age was 69 (range 60-85) yrs. Seven pts (34%) had secondary AML and 52% had complex karyotype. Twenty-one pts completed one induction cycle, one pt received a second induction due to residual disease and 6 pts underwent consolidation therapy. The principal toxicity was grade ≥3 infections and prolonged thrombocytopenia and neutropenia, which occurred in 18% and 21% of pts, respectively. Three deaths were documented from treatment complications (sepsis and fungal infections). Eight (38.1%) pts achieved either a CR or CRp. Responses were observed in 5/11 (45.5%) pts with high risk features (complex karyotype and/or presence of FLT3). The median disease free-survival was 1.9 (range, 0.9-8) months and the median overall survival was 6.5 (range, 4.2-14.2) months. The 1-yr overall survival was 35%. Conclusions: These preliminary results suggest that clofarabine in combination with daunorubicin is safe, well-tolerated with minimal toxicity, and active for the upfront treatment of ≥60 yrs old AML pts, including those with adverse features. Future trials will explore additional modifications to this regimen in order to optimize therapy for this group of pts with high risk features and frequent treatment failure.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2045-2045
Author(s):  
Ronit Gurion ◽  
Yulia Belnik-Plitman ◽  
Anat Gafter-Gvili ◽  
Mical Paul ◽  
Liat Vidal ◽  
...  

Abstract Abstract 2045 Poster Board II-22 Myeloid colony-stimulating factors (M-CSFs) are recommended as primary prophylaxis for the prevention of febrile neutropenia (FN) in patients who are at high risk for developing it. Since acute myeloid leukemia (AML) cells express functional growth factor receptors on their surface, concerns have been raised regarding the effect of M-CSFs on these cells. We assessed the safety of M-CSFs in AML patients. Systematic review and meta-analysis of all randomized controlled trials that compared the addition of M-CSFs during and following chemotherapy to chemotherapy alone in patients with AML was conducted. Trials evaluating the role of M-CSFs administered for the purpose of stem cell collection and/or priming e.g., before and/ or only for the duration of chemotherapy, were excluded. Both patients with and without neutropenia and/ or fever on admission were included. Two reviewers appraised the quality of trials and extracted data independently. The primary outcome was all-cause mortality at defined points in time. Secondary outcomes included complete remission (CR), disease free survival (DFS), relapse and infection rates. Relative risks (RR) with 95% confidence intervals (CIs) were estimated and pooled. The search yielded 17 trials including 4800 patients (range 53-803 per trial). 14 trials included patients undergoing induction chemotherapy, two trials included patients undergoing consolidation and one trial included patients undergoing salvage chemotherapy. The addition of M-CSFs to chemotherapy yielded no difference in all-cause mortality at 30 days, 1 year and at the end of follow-up [RR 1.00 (95% CI.0.82-1.22), RR 1.07 (95% CI 0.97-1.19) and RR 1.02 (95% CI 0.98-1.05), respectively] (Fig.1). There was no difference with regard to CR [RR 1.03 (95% CI 0.98-1.07)], relapse [RR 0.99 (95% CI 0.91-1.08)] and DFS [HR 1.00 (95% CI 0.90-1.13)]. M-CSFs did not decrease the occurrence of bacteremias, RR 0.96 (95% CI 0.82-1.12) or invasive fungal infections, RR 1.40 (95% CI 0.90-2.19). The addition of M-CSFs to chemotherapy does not affect all-cause mortality and leukemia associated clinical end points, e.g. CR, relapse rate and DFS. Hence, the use of M-CSFs for prevention of FN in patients with AML is safe. However, since prevention of neutropenia does not result in improved survival, they should not be used on a routine basis. Their use in the individual patient can be considered according to the clinical situation. Fig. 1: All Cause Mortality Fig. 1:. All Cause Mortality Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document