Comparison of Prognostic Factors and Outcomes of Patients with Secondary Acute Myeloid Leukemia (AML) Following Myelodysplastic Syndromes (MDS), Myeloproliferative Disorders (MPD), or Therapy-Related AML (t-AML).

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4292-4292 ◽  
Author(s):  
Sanjay Mohan ◽  
Paul Elson ◽  
Cristina Rodriguez ◽  
Rachid Baz ◽  
Matt Kalaycio ◽  
...  

Abstract Background: Response rates to induction chemotherapy and survival are poorer for patients (pts) with secondary AML compared to those with de novo AML. Within the category of secondary AML, no studies have compared the outcomes of induction chemotherapy in pts with AML arising from antecedent MDS vs. from MPD vs. t-AML. Methods: We conducted a retrospective review of all pts with newly diagnosed, pathologically-confirmed AML at a single institution between 1997 and 2005 and identified pts who also were diagnosed with an antecedent MDS, MPD, or t-AML. Data were collected on baseline characteristics and outcome, and controlled for in stepwise multivariable analyses. All pts were treated with anthracycline-based induction regimens. The primary endpoints were: complete remission (CR) rate (as defined by the IWG criteria) and survival from time of AML diagnosis, to determine whether those with AML arising from MDS, from MPD, or t-AML had different outcomes; and to define predictors of outcome among pts with secondary AML. Results: Of 457 AML patients, 281 were treated with remission induction therapy, of whom 66 had AML arising from MDS, MPD, or t-AML. Thirty-one (47%) had antecedent MDS, 20 (30%) an MPD, and 15 (23%) had t-AML. Twenty-six pts (39%) were female. The median age at the time of AML diagnosis for those with MDS, MPD, and t-AML was 67, 61, and 57 years, respectively (range 36–82, p=0.03 for all). Time from antecedent diagnosis/event was 7, 42, and 38 months, respectively (p=0.001). Cytogenetic risk categories (per CALGB 8461) were favorable in 3 pts (5%), intermediate in 30 (45%), unfavorable in 22 (33%), and unknown in 11 (17%). Neither cytogenetics (p=0.19) nor FAB/WHO AML classification (p=0.43) differed among groups. Median WBC at AML diagnosis was lower for pts with AML from MDS (3.7k/uL, vs. 9.9 k/uL for AML from MPD and 7.2 k/uL for t-AML, p=0.04), as were the percentage of peripheral blasts (6%, vs. 30% for AML from MPD and 23% for t-AML, p=0.005) and incidence of splenomegaly (6%, vs. 50% for AML from MPD and 0% for t-AML, p<0.001). CR rate was not significantly lower for patients with AML from MDS (35%) than for those with AML from MPD (55%) or t-AML (53%, p=0.33). Overall median survival was 8.0 months, with no significant difference among the 3 groups (7.7 months for AML from MDS, 11.1 months for AML from MPD, and 5.2 months for t-AML, p=0.23). Gender, interval from diagnosis of the antecedent disorder to diagnosis of AML, and marrow blast count did not correlate with CR rate or survival. In multivariable analyses, the antecedent diagnosis remained non-predictive of response rate or survival. Favorable- or intermediate-risk cytogenetics, however, compared to poor-risk, were significant predictors of higher CR rates (p= .005) and longer survival (p<.001). Lower peripheral blast percentage and age <60 years were favorable prognostic factors for CR. Conclusions: Contrary to expectations, pts with AML from MDS, from MPD, or t-AML had similar outcomes. The most important predictors of response to induction chemotherapy were cytogenetic risk group, age, and peripheral blast percentage. Only cytogenetic risk group was predictive of survival.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 921-921 ◽  
Author(s):  
Sanjay R. Mohan ◽  
Paul Elson ◽  
Cristina Rodriguez ◽  
Rachid Baz ◽  
Matt Kalaycio ◽  
...  

Abstract Background: Patients (pts) with sAML are evaluated together in large AML trials regardless of whether their sAML arises from antecedent MDS vs. from MPD vs. t-AML. Prognostic factors and outcomes may differ among these subgroups, and a prognostic scoring system would be helpful in determining which patients would benefit from induction chemotherapy. Methods: We conducted a retrospective review of all pts with newly diagnosed, pathologically-confirmed AML at Cleveland Clinic between 1997 and 2007 to identify sAML pts treated with cytarabine-based induction chemotherapy. Data on known prognostic factors (age, white blood cell count (WBC) at diagnosis, cytogenetic risk groups (as defined by CALGB 8461), and AML etiology) were collected as baseline characteristics and controlled for in stepwise multivariable analyses. Complete response (CR) and overall survival (OS) were analyzed. A prognostic scoring system for OS was developed based on the number of poor prognostic features present, derived from significant multivariable factors. Pts received 1 point for adverse cytogenetics, 1 point for having 1-10% peripheral blasts, and 1 point for AML arising from MDS or MPD. Pts with 0 points were favorable, 1 point intermediate, 2 or more points unfavorable. Results: Of 584 AML pts identified, 361 were treated with remission induction therapy, of whom 90 had AML arising from MDS, MPD, or t-AML. Thirty-nine (43%) had antecedent MDS, 21 (23%) an MPD, and 30 (33%) had t-AML, and 47% were female. Pts with AML arising from MDS were older at AML diagnosis (median of 67 years) vs. from MPD (61 years) and t-AML (60 years) (p=.02) but a shorter time from antecedent diagnosis/event (7 months, vs. 47 and 37 months, respectively (p<0.001)). Cytogenetic risk categories were favorable in 9 pts (10%), intermediate in 38 (42%), adverse in 27 (30%), and unknown in 16 (18%) and were similar among groups (p=.28). Median WBC at AML diagnosis was also lower for pts with AML from MDS (3.7k/uL, vs. 9.9 k/uL for AML from MPD and 8.9 k/uL for t-AML, p=.04), as were peripheral blasts (11%, vs. 29% for AML from MPD and 23% for t-AML, p=.01). The overall CR rate was 51% and was qualitatively lower for pts with AML from MDS (38%) than from MPD (62%) or t-AML (60%), but not significantly (p=0.11). The overall reinduction rate was 21% and did not differ among groups (p=.76). Median OS was significantly longer for pts with t-AML (15 months) vs. AML from MDS (8 months) or from MPD (11 months, p=.02). Available SNP karyotyping data on a subset of pts (6 from MDS, 1 from MPD, 4 with t-AML) did not reveal any shared or cryptic lesions that would distinguish responders from non-responders. In multivariable analyses, secondary AML etiology remained non-predictive of CR but was an independent prognostic factor for OS. Pts with t-AML had improved OS compared to AML from MDS (p=.01) or from MPD (p=.08). Favorable- or intermediate-risk cytogenetics, compared to adverse, were significant predictors of CR (p<.001) and OS (p<.001). Age <60 years (compared to 360 years) was a significant predictor of CR (p=.02), but not OS. Patients with 1–10% peripheral blasts had worse OS vs. 0% or >10% blasts (p=.01). Using the prognostic scoring system described above, favorable pts (n=16) had a median OS of 40 months vs. 12 months for intermediate pts (n=33) and 4 months for unfavorable patients (n=27, p<.001). Conclusions: Among sAML patients undergoing induction chemotherapy, pts with t-AML have a longer OS than AML from MDS or from MPD. Other important predictors of CR or OS were cytogenetics, age, and peripheral blast %. The sAML prognostic scoring system distinguished patients with improved survival from induction chemotherapy.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4846-4846
Author(s):  
Yeo-Kyeoung Kim ◽  
Hee-Je Kim ◽  
Woo-Sung Min ◽  
Jong- Ho Won ◽  
Deog-Yeon Jo ◽  
...  

Abstract Background: Although the most powerful prognostic factor of acute myeloid leukemia (AML) patients is the karyotype of the leukemic blast, data have not been obtained almost entirely in patients with heterogeneous cytogenetics. Further, some patients with favorable cytogenetics may show the poor treatment outcomes. Previous reports suggested that the single nucleotide polymorphisms of genes coding drug detoxification enzymes such as cytochrome P450 family or DNA repair system may influence the treatment outcomes in the patients with AML. We evaluated the role of polymorphisms in XRCC1, XRCC4, CYP1A1, GST-T1, GST-M1, NOQ1, and NAT2*6A in predicting therapeutic outcomes of adults with AML. Methods: XRCC1 (rs25487), XRCC4 (rs1056503), NQO1 (rs1800566), CYP-4501A1*2B (rs1048943), NAT2*6A (rs1799930) gene polymorphisms and deletion of GST-M1/GST-T1 were evaluated in 460 bone marrow (BM) samples obtained at initial diagnosis from de novo AML patients. Genotyping method is pyrosequencing using genomic DNA from BM samples. Homozygous deletions of GST-M1 and GST-T1 genes were detected with a multiplex PCR technique. All patients except APL (acute promyelocytic leukemia) received one or two rounds of intensive induction chemotherapy consisting of 3 days of idarubicin and 7 days of cytarabine. APL patients treated with AIDA regimen consisting of 45 days of ATRA (all-trans retinoic acid) and 3 days of idarubicin. Results: Of total 460 patients, ninety-nine patients (21.5%) were APL. Seventy-one (15.4%) were AML with t(8;21), twenty-three (5%) were AML with inv(16), and 179 patients (38.9%) showed normal cytogenetics. The median age of patients was 44 years (range, 14–75 years). In all cytogenetic risk group, the patients carrying homozygous NQO1 gene polymorphism (TT) showed significantly lower rate of complete remission (CR) than in those with negative or heterogyzous polymorphisms (TT: 72.7% vs. CC/CT: 85.9%, p=0.03). There was no significant difference in relapse rate, leukemia-free survival (LFS) and overall survival between homo- and heterozygote groups in these polymorphsims. In subgroup analysis, APL patients carrying TT genotype in NQO1 also showed lower rate of CR (TT: 77.8% vs. CC/CT: 95.4%, p=0.04). In AML patients except APL, NQO1 homozygous polymorphsim (TT) was also associated with lower CR rate (TT: 69.6% vs. CC/CT: 84.2%, p=0.005). In normal cytogenetics, the patients with del GST-M1 showed shorter LFS compared with those carrying GST-M1 (18.0 ± 5.7ms. vs. 34.6 ± NA. p=0.04). Conclusions: This study revealed an association between NQO1 polymorphism and GST-M1 deletion and the treatment outcomes for AML patients. Further study and larger sample size are needed to reach the definite conclusion on these associations. However, a stratified treatment plan in remission induction chemotherapy such as augmentation or addition of other chemotherapeutic agents may be warranted for AML patients harvoring homozygous NQO1 polymorphism (TT) or del GST-M1.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3523-3523
Author(s):  
Yunsuk Choi ◽  
Sungdoo Kim ◽  
Young-Hun Park ◽  
Jae Seok Lee ◽  
Dae-Young Kim ◽  
...  

Abstract Abstract 3523 Introduction: Secondary AML that has evolved from MDS shows different clinical features and outcomes compared to de novo AML. Prognostic implications of immunophenotypes have been studied in de novo AML, whereas those have not well been defined in secondary AML from MDS. Methods: This retrospective study involved analysis of data from 65 adult patients, 37 males and 28 females, who were diagnosed with AML arising from MDS at a single institute. Data for baseline clinico-pathological features, treatments, and outcomes were collected from medical records of each patient. Immunophenotyping was performed for the markers including TdT, CD34, CD13, CD33, CD117, CD14, CD56, HLA-DR, CD3, CD7, CD10, and CD19 using flow cytometry. Results: At the time of MDS diagnosis, the WHO subtype was RA/RARS in 5, RCMD in 10, RAEB-1 in 17, RAEB-2 in 29, and unknown in 4. For the treatment of MDS, hypomethylating agents were given to 17 patients and 2 patients underwent allogeneic hematopoietic cell transplantation (HCT). Median duration of MDS prior to diagnosis of AML was 4.9 months (range, 0.3–91.1). At the time of AML evolution, median age was 50.7 years (range, 18–80), and cytogenetic risk group was good-risk in 1, intermediate-risk in 45, and poor-risk in 18. Proportion of positivity of each immunophenotype marker was as follows: TdT (5%), CD34 (65%), CD13 (98%), CD33 (97%), CD117 (90%), CD14 (22%), CD56 (10%), HLA-DR (93%), CD3 (2%), CD7 (35%), CD10 (8%), and CD19 (2%). After the evolution to AML, 52 patients received induction chemotherapy consisted of cytarabine plus idarubicin or daunorubicin and 8 patients underwent allogeneic HCT as initial treatment of AML. Complete remission (CR) was induced in 27 patients after treatment. At a median follow-up time of 29.2 months (range, 2.6–116.2) among surviving patients, 49 patients died, 13 relapsed, and 53 died or relapsed. Median overall survival (OS), relapse-free survival (RFS), and event-free survival (EFS) were 7.6, 26.1, and 5.4 months, respectively. Of immunophenotype markers, CD14 positivity only showed prognostic implications at the univariate analyses: lower CR rate after induction chemotherapy (P=0.034) and shorter survivals (OS, P<0.001; RFS, P=0.078, and EFS, P<0.001). Differences in OS and EFS remained significant after adjustment for other variables (OS, HR, 4.49, 95% CI, 2.16–9.87, P<0.001; EFS, HR, 4.06, 95% CI, 2.03–8.13, P<0.001). Other prognostic variables included age of 60 years or older (shorter OS [P=0.003] and EFS [P=0.020]), WBC over 60,000/mcl (shorter OS [P<0.001] and EFS [P=0.001]), and poor cytogenetic risk group (shorter OS [P=0.005]). Conclusions: Surface expression of CD14 on leukemic blasts was an independent prognostic factor for survivals in the patients with AML arising from MDS. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1920-1920
Author(s):  
Cristina Rodriguez ◽  
Rachid Baz ◽  
Rony Abou Jawde ◽  
Lisa Rybicki ◽  
Christopher Lowe ◽  
...  

Abstract Background: Socioeconomic status (SES) and distance from a treatment center are prognostic factors in chronic malignancies, such as head and neck or breast cancer, where relative disease latency enables patients with greater means to travel to receive their care. Little is known about the influence of these demographics on patient outcomes in rapidly-growing cancers, such as AML. Methods: We conducted a retrospective review of all patients receiving remission induction therapy for AML at the Cleveland Clinic between January 1997 and December 2005. Race and residential zipcodes were obtained from information disclosed by patients in their electronic medical records. Average annual household income in a zipcode and distance from the treatment center were obtained using web-based databases (www.melissadata.com, www.zip-codes.com). Data on known prognostic factors (age, WBC at diagnosis, cytogenetic risk groups (as defined by CALGB 8461) and AML etiology (de novo vs. secondary AML)) were collected and controlled for in multivariable analyses. Survival was estimated using the Kaplan-Meier method, and the association between distance and income assessed using the Spearman rank correlation. Results: Anthracycline-based remission induction chemotherapy was administered to 281 patients: 132 (47%) were female, with a median age at diagnosis of 60 years (range 17–80). Median WBC at diagnosis was 9.9 k/uL (range:0.4–550 k/uL). Cytogenetics were favorable in 33 patients (11.7%, 10 of whom had t(15;17)), intermediate in 137 (48.8%), unfavorable in 73 (26%), and unknown in 38 (13.5%). Ninety patients (32%) had secondary AML. The ethnic distribution was consistent with other AML series: 252 (90.6%) were Caucasian (C), 22 (7.9%) were African American (AA), and 4 (1.4%) were neither (non-AA non-C). The median distance from the treatment center was 24.4 miles (range: 0.9–2058), and median average household income was $38,972 (range: $17,496-$143,220). Overall survival (OS) was 30.2% at a median of 22.6 months of follow-up. There was no significant correlation between distance and income. In both univariable and multivariable analyses, age ≥60 years, unfavorable cytogenetics, increased WBC count at presentation and secondary AML were all found to adversely affect survival (p<0.001, p<0.001, p=0.035, and p=0.010, respectively). OS was similar for AA and non-AA non-C patients compared to C (HR=1.12 [95% CI=.61–2.07, p=.71], and HR=.87 [CI=.21–3.62, p=.84], respectively). Neither distance from treatment facility (HR=1.00 [95%CI = .98–1.02 p =.96] for every 20 mile increase in distance) nor SES (HR=1.02 [95%CI=.92–1.13, p=.77] per $10000 increase) had an impact on OS. Similar findings held for CR rates. Conclusion: Contrary to their impact on chronic malignancies, neither SES nor distance from treatment facility affect outcome in patients with AML treated at the same hospital. This supports referring patients for treatment to tertiary facilities skilled at managing AML, even when they live at great distances from those facilities.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4492-4492
Author(s):  
Rachid Baz ◽  
Cristina Rodriguez ◽  
Rony Abou Jawde ◽  
Lisa Rybicki ◽  
Matt Kalaycio ◽  
...  

Abstract Background: AML in older adults poses therapeutic challenges, as many patients are not candidates for intensive chemotherapy (IC), and it is not clear that IC provides a survival benefit compared to NIC/BSC. While risk factors and outcomes of patients treated with IC are well-defined, few data support similar prognostication in patients treated with NIC or BSC. Methods: We conducted a retrospective review of all patients with AML receiving NIC or BSC at the Cleveland Clinic between January 1997 and December 2005. NIC consisted of treatment with hydroxyurea, low-dose cytarabine, or azacitidine. The choice and decision of therapy was dictated by patient and physician preferences. Data on known prognostic factors (age, white blood cell count (WBC) at diagnosis, cytogenetic risk groups (as defined by CALGB 8461) and AML etiology (de novo vs. secondary AML)) were collected and controlled for in multivariable analyses, and survival determined using the log-rank test, measured from the time of diagnosis. Race and residential zipcodes were obtained from information disclosed by patients in their electronic medical records. Average annual household income in a zipcode was obtained using a web-based database, www.melissadata.com. Results: Fifty-three patients were identified. The median age was 74 years (range 33–92), and 26 patients (49%) were female. Eight patients (15%) were African American (AA), while 44 patients were Caucasian (C) and 1 patient was neither AA nor C. The median WBC at presentation was 6.0 ×103/μL. Cytogenetics were favorable in 1 patient (2%), intermediate in 10 (19%), unfavorable in 19 (36%), and unknown in 23 (43%). Most patients (60%) had secondary AML. The median average annual income was $39,332 (range 23,375–82,806). The median survival was 54 days (95% CI 33–80 days) for all patients, and the 1 year survival was 7%. Known prognostic factors for AML (WBC at presentation, de novo vs. secondary AML, cytogenetics, and age at diagnosis) were not predictors of overall survival in multivariable analyses in patients not receiving intensive chemotherapy, though higher WBC count was predictive in univariable analyses. Race was a predictor for outcomes, with AA patients experiencing a worse median survival than their C counterparts (22 days versus 64 days, log rank p=0.03, figure 1) despite similar baseline characteristics. SES, however, had no impact on outcome. Conclusions: AML patients receiving NIC or BSC have a median survival of almost 8 weeks, with AA patients experiencing worse outcomes. A small but significant percentage of patients remain alive at one year. Prognostic variables useful in treatment decision-making in patients receiving IC are less valuable for NIC/BSC patients. This information can be used for patient counseling and to help guide treatment decisions. Figure Figure


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 6046-6046
Author(s):  
Sik-Kwan Chan ◽  
Cheng Lin ◽  
Shao Hui Huang ◽  
Tin Ching Chau ◽  
Qiaojuan Guo ◽  
...  

6046 Background: The eighth edition TNM (TNM-8) classified de novo metastatic (metastatic disease at presentation) nasopharyngeal carcinoma (NPC) as M1 without further subdivision. However, survival heterogeneity exists and long-term survival has been observed in a subset of this population. We hypothesize that certain metastatic characteristics could further segregate survival for de novo M1 NPC. Methods: Patients with previously untreated de novo M1 NPC prospectively treated in two academic institutions (The University of Hong Kong [n = 69] and Provincial Clinical College of Fujian Medical University [n = 114] between 2007 and 2016 were recruited and re-staged based on TNM-8 in this study. They were randomized in 2:1 ratio to generate a training cohort (n = 120) and validation cohort (n = 63) respectively. Univariable and multivariable analyses (MVA) were performed for the training cohort to identify the anatomic prognostic factors of overall survival (OS). We then performed recursive partitioning analysis (RPA) which incorporated the anatomic prognostic factors identified in multivariable analyses and derived a new set of RPA stage groups (Anatomic-RPA groups) which predicted OS in the training cohort. The significance of Anatomic-RPA groups in the training cohort was then validated in the validation cohort. UVA and MVA were performed again on the validation cohorts to identify significant OS prognosticators. Results: The training and the validation cohorts had a median follow-up of 27.2 months and 30.2 months, respectively, with the 3-year OS of 51.6% and 51.1%, respectively. Univariable analysis (UVA) and multivariable analysis (MVA) revealed that co-existing liver and bone metastases was the only factor prognostic of OS. Anatomic-RPA groups based on the anatomic prognostic factors identified in UVA and MVA yielded good segregation (M1a: no co-existing liver and bone metastases and M1b: co-existing both liver and bone metastases; median OS 39.5 and 23.7 months respectively; P =.004). RPA for the validation set also confirmed good segregation with co-existing liver and bone metastases (M1a: no co-existing liver and bone metastases and M1b: co-existing liver and bone metastases), with median OS 47.7 and 16.0 months, respectively; P =.008). It was also the only prognostic factor in UVA and MVA in the validation cohort. Conclusions: Our Anatomic-RPA M1 stage groups with anatomical factors provided better subgroup segregation for de novo M1 NPC. The study results provide a robust justification to refine M1 categories in future editions of TNM staging classification.


2013 ◽  
Vol 1 (2) ◽  
pp. 70-73
Author(s):  
Alina M Gridjac ◽  
Cristian Daniel Pirlog ◽  
Anca Simona Bojan

Background: Acute myeloid leukemia (AML) is a malignant disease with significant identified prognostic factors. Therefore our aim was to develop an Assessment Scheme of Prognosis in AML based on prognostic factors. In some counties, such as Romania or other less-highly developed countries, this scheme would be beneficial particularly when cytogenetic testing is unavailable or time-intensive. Methods: We analyzed 119 adult patients with AML during a five year-period from a single-center in Romania. We retrospectively collected and analyzed data with Epi Info and Excel using patient medical records. Results: According to age, the group A1 (<60 years) had a 40 months survival, in contrast with the group B1 (≥60 years) with a survival of 19 months (p=0,0063). The group A2 (secondary AML) survived 15 months, whereas the group B2 (AML de novo) survived 40 months (p=0.0021). Additionally, the group A3 (mild comorbidities) achieved a 40 months survival, the group B3 (moderate comorbidities) survived 19 months, whereas the group C3 (severe comorbidities) survived 7 months (p=0,0059). According to WBC and blast number, the group A4 (high levels) had a 25 months survival, whereas the group B4 (low levels) survived 40 months (p=0,0057). Conclusion: The prognostic factors studied are useful to identify the risk level of AML disease for each patient at diagnosis. We developed an assessment scheme of prognosis with three risk groups according to age, secondary AML, comorbidity, WBC and blasts and cytogenetic examination.


2021 ◽  
Author(s):  
Ádám Jóna ◽  
Anna Kenyeres ◽  
Sándor Barna ◽  
Árpád Illés ◽  
Zsófia Simon

Abstract Introduction: Follicular lymphoma (FL) is an indolent yet heterogeneous B-cell lymphoproliferative disorder. Most people respond to treatment well. However, a particular group of patients has a poor prognosis, and these patients are difficult to define.Patients and methods: We retrospectively analyzed FL patients treated at the University of Debrecen in the past 20 years. We investigated prognostic factors that may influence the survival of FL patients.Results: We found a standardized uptake value (SUV)max cut-off value of 9.85 at the staging PET/CT to significantly separate FL patients’ progression-free survival (PFS) (p=0.0003, HR: 0.2560, 95%CI: 0.1232-0.5318). Lymphocyte/ monocyte (Ly/Mo) ratio of 3.45 drawn at diagnosis also significantly predicted PFS (p=0.0324, HR: 1.806, 95% CI: 1.051-3.104). Combining patients’ with staging SUVmax >9.85 and Ly/Mo < 3.45 a high-risk group of FL patients can be identified (p<0.0001, HR: 0.1033, 95%CI: 0.03719-0.2868). Similarly, a significant difference was shown with a SUVmax cut-off of 3.15 at the interim PET/CT (p<0.0001, HR: 0.1535, 95%CI: 0.06329-0.3720). Combining patients with staging SUVmax >9.85 and interim SUVmax >3.15, a high-risk group of FL patients can be identified (p<0.0001, HR: 0.1037, 95%CI: 0.03811-0.2824). The PFS difference is translated into overall survival advantage (p=0.0506, HR: 0.1187, 95%CI: 0.01401-1.005).Discussion: Biological prognostic factors, such as the Ly/ Mo ratio, may improve the prognostic assessment of staging PET/CT. Nevertheless, PFS difference is translated into OS when using a combination of staging and interim SUVmax. We consider investigating additional biological prognostic factors while currently highlighting PET/CT's role in FL.


Cancers ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 3334
Author(s):  
Safaa M. Ramadan ◽  
Stefan Suciu ◽  
Marian J. P. L. Stevens-Kroef ◽  
Roelof Willemze ◽  
Sergio Amadori ◽  
...  

We report the outcomes of secondary acute myeloid leukemia (s-AML) patients included in one of 13 European Organisation for Research and Treatment of Cancer (EORTC) collaborative AML trials using intensive remission-induction chemotherapy. Among 8858 patients treated between May 1986 and January 2008, 960 were identified as having s-AML, either after MDS (cohort A; n = 508), occurring after primary solid tumors or hematologic malignancies other than MDS (cohort B; n = 361), or after non-malignant conditions or with a history of toxic exposure (cohort C; n = 91). Median age was 64 years, 60 years and 61 years in cohort A, B and C, respectively. Among patients ≤60 years and classified in the cohorts A or B (n = 367), the 5-year overall survival (OS) rate was 28%. There was a systematic improvement in the 5-year OS rate over three time periods (p < 0.001): 7.7% (95% CI: 1.3–21.7%) for patients treated before 1990 (period 1: n = 26), 23.3% (95% CI: 17.1–30.0%) for those treated between 1990 and 2000 (period 2: n = 188) and 36.5% (95% CI: 28.7–44.3%) for those treated in 2000 or later (period 3: n = 153). In multivariate analysis, male gender (HR = 1.39; p = 0.01), WBC ≥ 25 × 109/L (HR = 2.00; p < 0.0001), age 46-60 years (HR = 1.65; p < 0.001) and poor-risk cytogenetics (HR = 2.17; p < 0.0001) were independently associated with shorter OS, while being treated during period 2 (HR = 0.50, p = 0.003) or period 3 (HR = 0.43; p = 0.0008). Having received high-dose cytarabine (HD-AraC) (n = 48) in the induction chemotherapy (HR = 0.54, p = 0.012) was associated with a longer OS. In contrast, among patients >60 years of age (n = 502), the OS was dismal, and there was no improvement over time.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 866-866 ◽  
Author(s):  
Alberto Bosi ◽  
Jeffrey Szer ◽  
Jeannine Kassis ◽  
Jorge Sierra ◽  
Claire Desborough ◽  
...  

Abstract BACKGROUND: Heil et al (Blood ‘97) demonstrated that the duration of neutropenia and its clinical consequences following induction chemotherapy for AML were significantly reduced by the addition of filgrastim, with no increased risk of death, second malignancy or relapse (ASH ‘99). A single injection of pegfilgrastim has been shown to be comparable to daily injections of filgrastim in the management of chemotherapy-induced neutropenia. The primary aim of this trial was to estimate the difference in time to recovery from severe neutropenia (SN, ANC < 0.5 x109/L) in the first induction chemotherapy cycle (Induction 1) in AML subjects treated with pegfilgrastim or filgrastim. METHODS: Subjects with de novo AML received 1 or 2 courses of induction chemotherapy (idarubicin 12mg/m2 IV days 1–3, cytarabine 100mg/m2 IV 12 hourly days 1–7 [IA 3+7]) then, if in remission, consolidation chemotherapy (cytarabine 2 [subjects < 55 years] or 3g/m2 [subjects ≥ 55 years] IV 12 hourly days 1, 3, 5). Subjects received either single administration 6μg pegfilgrastim or daily 5μg/kg filgrastim starting 24 hours after completion of chemotherapy until neutrophil recovery. Time to recovery from SN was defined as the number of days from the first day of chemotherapy until the first of two ANC consecutive values after the nadir that were ≥ 0.5 x 109/L whereas duration of SN was defined as the total number of days during the cycle with an ANC < 0.5 x 109/L. RESULTS: Of 84 subjects randomised into the study, 83 received study drug (42 pegfilgrastim, 41 filgrastim). The treatment groups were generally well balanced for demographics and baseline characteristics. The median time to recovery from SN (ANC < 0.5 x109/L) in Induction 1 was 22 days in both treatment groups (95% CI for treatment difference: −1.9, 1.9). There was also no statistically significant difference in the median duration of SN between the 2 groups (21 days pegfilgrastim, 20 days filgrastim). Subjects in the filgrastim group required a median of 16 daily injections compared to a single administration of pegfilgrastim in the second group. Median serum concentration of pegfilgrastim in Induction 1 remained above clinically relevant concentrations until 21 days after the start of chemotherapy, results that are consistent with the neutrophil-mediated clearance of pegfilgrastim. The incidence of serious adverse events was comparable between the 2 groups except for infectious complications, which were higher in the filgrastim group (5 subjects [12%] pegfilgrastim versus 9 subjects [22%] filgrastim). CONCLUSION: In the setting of the first cycle of IA 3+7 induction chemotherapy in AML patients, once per cycle administration of 6 mg pegfilgrastim or daily administration of 5mg/kg filgrastim result in similar median time to recovery to ANC ≥ 0.5 x109/L. Pegfilgrastim is safe and well tolerated in this subject population.


Sign in / Sign up

Export Citation Format

Share Document