Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Leukemia, Springer Science and Business Media LLC, Vol. 35, No. 8 ( 2021-08), p. 2332-2345
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2008023-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 3015-3015
    Abstract: Background: In France, as in many other countries, nationwide data on prevalence are rarely available and recent prevalence estimates of Chronic Myeloid Leukemia (CML) are scarce. Improved overall survival following the introduction of tyrosine kinase inhibitors (TKIs) is expected to have increased the prevalence of CML in Western countries. Aim: We sought to estimate and analyze the prevalence of CML in France for the year 2014 using a large health care claim-based dataset. Methods: Using the French national health insurance database that covers 98.8% of the French population (66 million people) we implemented a 3-step approach. First, focusing on the 2006-2014 period, we selected: 1) all patients treated with a TKI (ie, imatinib, dasatinib, nilotinib, bosutinib or ponatinib) and/or 2) identified by the ICD-10 diagnosis code C92.1 (Chronic Myeloid Leukemia, BCR/ABL-positive) among hospital discharge diagnoses and/or 3) identified by the ICD-10 diagnosis code C92 (myeloid leukemia) for coinsurance exemption. Then, we developed a claim-based algorithm to identify CML cases. Case definition was based on 1) identifying any TKI reimbursement lasting ≥ 2 months and 2) excluding patients receiving TKIs for diseases other than CML including Phi+ Acute Lymphoblastic Leukemia, Gastrointestinal Stromal Tumor, Stromal or other connective tissue tumor, and hypereosinophilic disease. Finally, prevalent CML cases were those identified by the algorithm above and having ≥ 1 healthcare reimbursement during the year 2014 and still alive on December, 31st 2014. The internal validity of the algorithm was tested on a random sample of 100 potential CML cases fulfilling ≥ 1/3 selection criteria in step 1 by comparing the results of the algorithm with the opinion of two hematologists (gold standard). For each individual, hematologists reviewed patient demographics and the sequence of care from 2006 to 2014 including healthcare resource utilization (ie, all hospitalizations and ICD-10 diagnosis codes, all medication use, all specialist consultations with date and specialist type). In addition, we assessed the external validity of the algorithm by comparing the number of incident CML patients in 2014 as identified in the French national health insurance database with the number of incident CML cases recorded in the French cancer registries for respective departments (i.e. ~ 20% of the French territory). Results: There were 10,789 prevalent CML cases in 2014 out of 68,067 individuals from the French national health insurance database who fulfilled the selection criteria for the overall 2006-2014 period. Eighty-nine percent of the prevalent CML cases were identified by at least two out of three selection criteria (TKI, ICD-10 code C92.1 among hospital discharge diagnoses, ICD-10 code C92 for coinsurance exemption). There was a 96% concordance rate (internal validity) between the algorithm and the opinion of the hematologists. For the year 2014, 162 and 150 incident CML patients were identified by the algorithm and the French cancer registries, respectively (high external validity). Median age [Inter-Quartile Range] of the prevalent population of CML patients was 63 years [51-73] , with slightly more males affected (55%). On December, 31st 2014, the crude prevalence of CML was estimated at 16.3 per 100,000 inhabitants [95% confidence interval (CI) 16.0-16.6]. The crude prevalence of CML was 18.5 per 100,000 in men (95% CI 18.0-19.0) and 14.2 per 100,000 in women (95% CI 13.8-14.6). The crude prevalence of CML was less than 1.6 per 100,000 (95% CI 1.2-2.0) before 20 years of age, progressively increasing to 19.4 per 100,000 (95% CI 18.1-20.7) among those with 50-54 and reaching a peak of 48.2 per 100,000 (95% CI 45.3-51.1) at 75-79 years. There was a male preponderance in CML prevalence in all age groups. The crude prevalence of CML varied in a ratio of one to two throughout the French territory (from 10.2 to 23.8 per 100,000 inhabitants). Conclusion: Healthcare claims data are increasingly used to estimate epidemiological parameters worldwide. This approach is particularly relevant for rare diseases and administrative databases with high population coverage. Countries without national cohorts or cancer registries could easily use our algorithm to estimate their prevalence of CML. Disclosures Cony-Makhoul: Pfizer: Consultancy; BMS: Consultancy, Speakers Bureau; Incyte: Other: Travels for attending to Congress; Novartis: Consultancy, Other: Writing support, Travels for attending to Congress. Guerci-Bresler:Pfizer: Other: Fees for symposiums and boards; Novartis: Consultancy, Other: Fees for symposiums and boards; Incyte: Other: Fees for symposiums and boards; BMS: Other: Fees for symposiums and boards; Pfizer: Other: Travel fees for Congress. Delord:Incyte: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Haematologica, Ferrata Storti Foundation (Haematologica), Vol. 107, No. 12 ( 2022-08-04), p. 2944-2949
    Type of Medium: Online Resource
    ISSN: 1592-8721 , 0390-6078
    Language: Unknown
    Publisher: Ferrata Storti Foundation (Haematologica)
    Publication Date: 2022
    detail.hit.zdb_id: 2186022-1
    detail.hit.zdb_id: 2030158-3
    detail.hit.zdb_id: 2805244-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Medicine, Wiley, Vol. 8, No. 6 ( 2019-06), p. 3296-3304
    Abstract: Data on Chronic Myeloid Leukemia (CML) prevalence are scarce. Here we provide an estimation of the prevalence of CML in France for the year 2014 using French national health insurance data. Methods We selected patients claiming reimbursement for tyrosine kinase inhibitors (TKI) or with hospital discharge diagnoses for CML, BCR/ABL‐positive or with full reimbursement of health care expenses for myeloid leukemia. We built an algorithm which we validated on a random sample of 100 potential CML patients by comparing the results obtained using the algorithm and the opinion of two hematologists who reviewed the patient demographics and sequence of care abstracted from claims data (internal validity). For external validity, we compared the number of incident CML patients identified using the algorithm with those recorded in French population‐based cancer registries in departments covered by such a registry. Results We identified 10 789 prevalent CML patients in 2014, corresponding to a crude prevalence rate of 16.3 per 100 000 inhabitants [95% confidence interval (CI) 16.0‐16.6]: 18.5 in men [18.0‐19.0] and 14.2 in women [13.8‐14.6]. The crude CML prevalence was less than 1.6 per 100 000 [1.2‐2.0] under age 20, increasing to a maximum of 48.2 [45.4‐51.2) at ages 75‐79. It varied from 10.2 to 23.8 per 100 000 across French departments. The algorithm showed high internal and external validity. Concordance rate between the algorithm and the hematologists was 96%, and the numbers of incident CML patients identified using the algorithm and the registries were 162 and 150, respectively. Conclusion We built and validated an algorithm to identify CML patients in administrative healthcare databases. In addition to prevalence estimation, the algorithm could be used for future economic evaluations or pharmaco‐epidemiological studies in this population.
    Type of Medium: Online Resource
    ISSN: 2045-7634 , 2045-7634
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2019
    detail.hit.zdb_id: 2659751-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cytometry Part A, Wiley, Vol. 81A, No. 11 ( 2012-11), p. 996-1004
    Type of Medium: Online Resource
    ISSN: 1552-4922
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 2180639-1
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: American Journal of Hematology, Wiley, Vol. 95, No. 11 ( 2020-11), p. 1314-1323
    Abstract: FIP1L1‐PDGFRA‐positive myeloid neoplasm with eosinophilia (F/P+ MN‐eo) is a rare disease: robust epidemiological data are lacking and reported issues are scarce, of low sample‐size and limited follow‐up. Imatinib mesylate (IM) is highly efficient but no predictive factor of relapse after discontinuation has yet been identified. One hundred and fifty‐one patients with F/P+ MN‐eo (143 males; mean age at diagnosis 49 years; mean annual incidence: 0.18 case per million population) were included in this retrospective nationwide study involving all French laboratories who perform the search of F/P fusion gene (study period: 2003‐2019). The main organs involved included the spleen (44%), skin (32%), lungs (30%), heart (19%) and central nervous system (9%). Serum vitamin B12 and tryptase levels were elevated in 74/79 (94%) and 45/57 (79%) patients, respectively, and none of the 31 patients initially treated with corticosteroids achieved complete hematologic remission. All 148 (98%) IM‐treated patients achieved complete hematologic and molecular (when tested, n = 84) responses. Forty‐six patients eventually discontinued IM, among whom 20 (57%) relapsed. In multivariate analysis, time to IM initiation (continuous HR: 1,01 [0.99‐1,03]; P = .05) and duration of IM treatment (continuous HR: 0,97 [0,95‐0,99]; P = .004) were independent factors of relapse after discontinuation of IM. After a mean follow‐up of 80 (56) months, the 1, 5‐ and 10‐year overall survival rates in IM‐treated patients were 99%, 95% and 84% respectively. In F/P+ MN‐eo, prompt initiation of IM and longer treatment durations may prevent relapses after discontinuation of IM.
    Type of Medium: Online Resource
    ISSN: 0361-8609 , 1096-8652
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2020
    detail.hit.zdb_id: 1492749-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2747-2747
    Abstract: Abstract 2747 Introduction Despite the major benefit of TKI in the treatment of Chronic Myeloid Leukemia (CML), patient response is heterogeneous and it is generally accepted that residual disease and relapse are due to persistent CML cells, considered as leukemic stem cells. Their resistance has been related to lower TKI uptake. The amount of drug penetrating the targeted cells is most likely a major parameter of targeted therapy efficacy since it is essential that the therapeutic molecule be as close as possible to the target molecule. We developed a flow cytometry technique to analyze primary cells. Method To evaluate intracellular imatinib (ICIM) uptake, we developed a patented method based on natural UV fluorescence related to chemical structure. Consequently, since the difference in UV fluorescence units between treated and control cells is proportional to the amount of intra-cellular drugs, we validated this method after incubating K562 and KCL22 cell lines with TKI. The flow cytometry technique was standardized by using Flow-Check Fluorosphere calibrated beads immediately before, and at the end of, each series of analyses with a Coulter Epics Elite™ flow cytometer (Beckman Coulter) equipped with an Innova I90C-4 UV laser (Coherent). Then we analyzed primary blood cells from CML patients in chronic phase before any treatment. After lysis of erythrocytes, nucleated cells were incubated at 1.106 cells/ml with different doses of imatinib (IMA) (n=22), Nilotinib (NIL) (n=20) and Dasatinib (DAS) (n=20) at different times. Whenever possible, CML stem cells were analyzed using CD34-FITC staining. Results In preliminary assays, we checked that there was a significant correlation between additional fluorescence measured by flow cytometry and the amount quantified by physico-chemical analysis after lysing a known number of cells (n=57, r2=0.73, p 〈 0.001), which enabled us to convert UV fluorescence into pg of IMA per cell. Then we confirmed that IMI rapidly penetrated K562 and KCL22 cells (from 5 minutes of incubation) and reached a stable influx in viable cells from 1 hour (T1h). We chose this incubation time for further experiments. Similarly, we choose T2h for second generation TKI. We observed a dose-dependent accumulation in the two cells lines, but with differences at the lowest extra-cellular concentrations (1–5 μM) and not correlated with any membrane pump expression (OCT-1, ABCG2, ABCB1 and ABCC1). ICIM at T1h was correlated with cell sensitivity to IM at T24h expressed by the proportion of dead cells (r2=0.93 and 0.88 for K562 and KCL22 cells, respectively). We then applied our method to primary CML blood cells in comparison with normal blood cells. TKI penetrated all cell subsets, but amounts varied depending on cell sizes (FS/SS characteristics). The first data obtained with IM showed ICIM levels in CML cells that were relatively heterogeneous from one patient to another, ranging from 0.9 to 4 pg/cell for an extracellular concentration of 5 μM, i.e. a higher concentration (x 300) than in culture medium. The ability of the granulocyte cell lineage to store IMA was related to the Sokal prognostic index (p=0.05). We detected variable ICIM levels in CML CD34+ cells from 10/16 patients (0.04–0.7 pg/cell) and no signal for 6/16 patients. Surprisingly, the ability of CD34+ to store second generation TKIs is variable and not necessarily correlated to IMA uptake. Discussion We developed a simple, rapid flow cytometry method directly applicable to primary cells and requiring only few cells which makes it possible to identify target cell subsets, such as CML stem cells. The strong correlation between the ICIM amount and the sensitivity of CML cell lines to TKIs validated the method and suggested that ICIM could be a relevant biomarker for predicting the sensitivity of the CML clone. In our CML series, we observed striking inter-patient variability of the capacity of primary CML cells to store TKI. A correlation with the Sokal score suggests possible predictive value with regard to in vivo CML response to IMA, which could be taken into account when choosing TKI for first-line therapy. Furthermore, we observed marked heterogeneity between CML CD34+ cells for storing TKI that could partially explain the heterogeneity of in vivo response. The relationship between the ability of untreated CML CD34+ cells to store TKI and complete molecular response has to be established. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2713-2713
    Abstract: Recent studies demonstrated that early molecular response was predictive for outcome in chronic phase chronic myeloid leukemia (CML) patients on imatinib (IM) (Hanfstein et al., Leukemia 2012, Marin et al., JCO 2012). It is thus essential to identify patients likely to achieve better long-term outcome if they are switched early to second-line therapy. In reference laboratories, BCR-ABL1 transcript levels are expressed as BCR-ABL/ABL ratios (%) according to the international scale (IS, Hughes et al., Blood 2006). Nevertheless, for high levels of BCR-ABL1, the copy number of total-ABL1 comprises a part of BCR-ABL1, which results in underestimation of the ratio at diagnosis. But the BCR-ABL1 levels for which this underestimation begins to be significant is currently unknown. Besides, since the cut-off value of 10% seems critical to predict outcome for patients treated with IM for 3 months, it stresses the necessity of a precise quantification at this level. In this study, we evaluated the ability of GUS, identified as another suitable control gene (CG) (Beillard et al., Leukemia 2003), to accurately define the relevant thresholds for known therapeutic decision levels (10% and major molecular response MMR: 0.1%) in a series of front line therapy patients on IM. Methods We collected 141 blood samples of CML patients between August 2000 and December 2012, including a first group of diagnosis samples (n= 83) and follow-up samples chosen according to their transcript value: the second group with a BCR-ABL1/ABL1 ratio between 6 and 14% (n=21) and the third group at the MMR threshold of 0.1% (n=37). The expression of BCR-ABL1, total-ABL1 and GUS were quantified as previously described (Gabert et al., Leukemia 2003). Results were expressed as percent ratios relative either to ABL1, with original values converted to IS, or to GUS, without transformation of the original values. Results The mean BCR-ABL1/ABL1 ratio was 55.91% (range: 34.83-98.01%) at diagnosis, 9.66% (range: 6.72-13.53%) in the second group (6-14% values) and 0.093% (range: 0.05-0.18%) in the third group (MMR patients). To determine the impact of GUS as CG on the MMR threshold, we compared in the third group the copy number ratios of BCR-ABL1 relative either to ABL1 (=0.093%) or GUS. The BCR-ABL1/GUS mean value was 0.036%. Thus, the translation of the MMR threshold from BCR-ABL1/ABL1 to BCR-ABL1/GUS would need the introduction of an additional conversion factor (Figure 1). This factor is defined as the ratio of (BCR-ABL1/ABL1)/(BCR-ABL1/GUS) =GUS/ABL1 for each measurement and its mean was valued at 2.774 (2.507-3.040, 95% confidence interval). To assess whether this factor was constant within the different disease time, we next calculated it for the two other groups (figure 2). In diagnosis samples, the mean value of the factor was 3.772 (3.554-3.991, 95%CI) whereas it valued at 2.668 (2.263-3.072, 95%CI) in samples with a 6-14% BCR-ABL1/ABL1 ratio. Comparison of means showed a significant difference between the 3 groups (p 〈 0.001, ANOVA test). This difference over time in the course of the disease was due to GUS overexpression in CML cells at diagnosis, GUS overexpression being superior to the overestimation of ABL1 at diagnosis due to the quantification test bias (Figure 3). This overexpression of GUS, previously reported (Beillard et al., Leukemia 2003), impacts the BCR-ABL1/GUS ratio at diagnosis and warrants the use of a unique translation factor for all disease levels. The number of copies of GUS did not differ significantly between the 6-14% samples and MMR samples (p=0.123, Student's T-Test), nor did the ABL1 copy number (p=0.06). This means that the quantification bias observed for diagnosis samples does not impact the values under 14%. Conclusion The use of GUS as CG for the MRD would require defining the factor allowing translation between BCR-ABL1/GUS and BCR-ABL1/ABL1 ratios, which implies a new standardization of the international scale. Nevertheless, this factor seems to depend on the transcripts levels, since the value of BCR-ABL1/GUS obtained at diagnosis is impacted by the higher GUS copy number in CML cells. As the copy number of ABL1 and GUS did not differ significantly between the groups of patients with 0.1% and 6-14% BCR-ABL1/ABL1 ratio, we concluded that there is no quantification bias at these critical therapeutic decision levels. As a consequence, ABL1 as GUS may be used efficiently as CG to assess early response to TKI. Disclosures: Cony-Makhoul: BMS: Honoraria. Michallet:MSD: Consultancy, Honoraria, Research Funding; Astellas: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding; Genzyme: Consultancy, Honoraria, Research Funding. Nicolini:Novartis, Ariad, Teva, BMS and Pfizer: Honoraria from Novartis, Ariad, Teva, BMS and Pfizer. Grants from Novartis. Other.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1971-1971
    Abstract: Prognosis of relapses is severe in elderly multiple myeloma (MM). In recent studies, median survival at progression after 1st line therapy was between 9 and 13 months (T. Facon, Lancet 2007; C. Hulin, J Clin Oncol 2009). Bortezomib (V) plus dexamethasone (D) is a major regimen in the treatment of relapses. Bendamustine (B) demonstrated to be highly active in advanced MM. The IFM 2009-01 trial evaluates the combination of B, V and D in elderly patients with progressive MM on or after 1stline treatment. Methods Phase 2 IFM 2009-01 trial was dedicated to patients older than 65 years in 1st relapse or refractory to 1st line therapy. Inclusion criteria were measurable disease, PS ECOG 0-2, ANC 〉 1.5x109/l, platelets 〉 100x109/l, serum creatinine level 〈 250 mcmol/l, AST and ALT 〈 3xULN. Pts with prior exposure to bortezomib were excluded. Treatment regimen was B 70 mg/m2 day 1-8, V 1.3 mg/m2 day 1-8-15-22 and D 20 mg day 1-8-15-22 every 28 days. 6 cycles were administered. Responders were assigned to receive maintenance treatment with 6 additional cycles administered 1 month out of 2. Response was evaluated according to IMWG criteria. Response rate was the primary objective. Progression-free survival (PFS), overall survival (OS) and toxicity were secondary end-points. Results From 03/2010 to 07/2011, 73 pts were included. Median age was 75.8 years (range 66-86). Median time from diagnosis to inclusion was 29 months. All pts received only 1 prior line of therapy: Melphalan-Prednisone (MP) in 12, MP-Thalidomide in 42, Lenalidomide-Dexamethasone (LD) in 14, other IMiD-based regimen in 5. Median treatment cycles administered was 7 (1-12). 51 pts (69.8%) achieved at least partial response [best response CR: 10 pts (13.6%), VGPR: 12 pts (16.5%), PR: 29 pts (39.7%), MR: 4 pts (5.5%), SD: 5 pts (6.8%), progression: 12 pts (16.5%), unrelated early death: 1 pt (1.3%)]. Median PFS was 10.8 months (95%CI: 7-18.2 months) and median OS 23 months (15.4-27.5). Adverse prognostic factors for PFS were PS ECOG 2 (p=0.0002), beta 2 microglobulin level 〉 3.5 mg/l (p=0.0006) and del17p (p=0.01). 26 pts (35.6%) completed the planned 12 cycles of treatment. Cause of treatment discontinuation was progressive MM in 30 pts (41.1%), failure to achieve PR in 5 pts (6.8%), adverse event in 10 pts (13.6%), patient refusal and unknown 1 pt (1.3%) each. 37 pts (50.6%) had died at time of final analysis. Cause of death was MM in 30 pts, sepsis in 5 pts, renal failure in 1 pt and unrelated in 1 pt. Grade 3-4 adverse events were neutropenia: 14 pts (19.1%), thrombocytopenia: 8 pts (10.9%), sepsis: 14 pts (19.1%), gastro-intestinal: 6 pts (8.2%), anaphylaxis: 2 pt (2.7%). Grade 2 peripheral neuropathy occurred in 8 pts (10.%) and grade 3 in 3 pts (4.1%). Conclusions In this elderly population with poor prognosis MM, BVD combination provides a high overall response rate and manageable toxicity. These results compare favourably with those achieved with VD or LD. Disclosures: Roussel: CELGENE: Honoraria; JANSSEN: Honoraria. Leleu:CELGENE: Honoraria; JANSSEN: Honoraria. Cony-Makhoul:BMS: Honoraria. Avet-Loiseau:CELGENE: Honoraria, Speakers Bureau; JANSSEN: Honoraria, Speakers Bureau. Moreau:Janssen: Honoraria; Janssen and Millennium: Membership on an entity’s Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4023-4023
    Abstract: Abstract 4023 Background: Treatment of CMML remains difficult, with no drug having shown a clear clinical benefit. AZA has demonstrated a survival benefit in higher risk MDS, in a study that included a small number of CMML (Lancet Oncol, 2009). Several small series of CMML treated by Decitabine (Wijermans, Leuk Res. 2008, Aribi A, Cancer. 2007 and Kantarjian H, Blood. 2007), and AZA (Scott, Br J Haematol. 2010) have been reported, but pts were often very heterogeneous in terms of risk factors and treatment, while in the AZA paper, patients also received etanercept in combination. Methods: The French health agency (AFSSAPS) designed, between 2004 and 2008, a pt named program (ATU) of AZA in higher risk MDS and poor risk AML. 38 pts with CMML or AML arising from CMML included in this program before April 08 and having completed ≥ 1 cycle of AZA (75 mg/m2/d during 7 days every 28 d) are analysed here. As CMML has features of both MDS and MPD, its risk factors are somewhat composite. Based on our previous experience (JCO 1988 6:1417, Blood 1996 88:2480) and on IPSS, pts with WBC 〈 13 G/L were classified according to IPSS, and, in those with WBC 〉 13 G/L, risk factors were based on: marrow blasts≥5%, Hb 〈 10g/dl, plts 〈 100G/l, abnormal cytogenetics, splenomegaly 〉 5cm below costal margin (SMG 〉 5cm) and extramedullary disease (EMD). Response was evaluated according to IWG 2006 criteria in pts with WBC 〈 13G/L, also took into account “proliferative” features of CMML (splenomegaly, increased WBC and blood monocytes, extra medullary disease) in pts with WBC 〉 13G/L, and IWG-AML 2003 criteria for AML. Results: median age was 71 y (range 50–87), M/F: 28/10. Median interval from diagnosis to treatment was 22 months (range 0.2–74 months). Previous treatment was low dose chemotherapy (CT) (n= 10, low dose Arac n=2, HU n=8), Intensive CT (n=12), allogeneic SCT (n=1), ATO (n=2). At inclusion, 9 pts had CMML-1, 17 CMML-2 and 12 AML secondary to CMML according to WHO. Karyotype was normal (n=16), isolated –7/7q- (n=2), +8 (n=1), del 20q (n=1), complex (n=2), -Y (n=2) and a failure (n=2). In the 14 CMML with WBC 〈 13G/L, IPSS was low in 1, int-1 in 3 pts, int-2 in 8 pts and High in 2 pts. In the 12 CMML with WBC 〉 13G/L, 10 had more than 3 risk factors defined in Blood 1996 88:2480. The median number of cycles of AZA administered was 4 (range 1–26). 6 pts received also HU during the first cycles to reduce WBC count. 9 pts received less than 4 cycles due to early death (n=4), progression (n=3) and haematological toxicity (n=2). 20 pts (53%) responded including 9 CR, 3 marrow CR,8 HI-E and 1 partial remission. 19 (68%) of the 28 pts who received more than 4 cycles responded, including 9 CR, 3 mCR, 1 PR and 6 HI. Of the 26 CMML without AML progression, 15 (58%) responded (7 CR, 2 marrow CR and 6 HI-E). Of the 12 AML arising from CMML, 5 (42%) responded (2 CR, 1 marrow CR, 1 PR and 1 HI-E). Median number of cycles of AZA to achieve best response was 4 (range 3–12). Age (p=0.38), WBC count (p=0.76), Hb level (p=0.987), platelet count (p=0.07), blood monocytes (p=0.4823), Sex (p=0.28), CMML 1 vs 2 (p=0.48), splenomegaly (p=0.7), normal karyotype (p=1), -7/del7q(p=0.65), concomitant treatment with HU (p=0.6), 〉 3 risk factors in CMML with WBC 〉 13 G/l, and previous treatment (p=0.506) had no impact on response. 9 of the 20 responders relapsed after a median of 10.6 months (range 3–23), including 4/9 (44%), 1/3 (33%), 0/1, 4/8 (50%) of the pts who had achieved CR, mCR, PR and HI respectively, 10 remained responders after a median of 26 months (16-35) and 1 pt with HI died without relapse. Median overall survival (OS) was 24 months in CMML compared to 7 months in AML arising from CMML (p= 0.0081). Presence of splenomegaly, WBC 〉 13 G/l, previous treatment (excluding ESA), Sex, -7/del7q and normal karyotype had no impact on OS. Conclusion: In this series of CMML which had on average more unfavourable prognostic factors than in previous series of CMML treated with Decitabine or AZA (10/14 with WBC 〈 13G/L were IPSS int 2 or high, 10/12 with WBC 〉 13 G/L had at least 3 risk factors, and 12 had progressed to AML), AZA showed clear efficacy, but mainly in pts who had not progressed to AML. Disclosures: Fenaux: CELGENE, JANSSEN CILAG, AMGEN, ROCHE, GSK, NOVARTIS, MERCK, CEPHALON: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages