GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 5123-5123
    Abstract: Background: monoclonal gammopathy of undetermined significance (MGUS) has a prevalence of 1 to 3%. Although it has an indolent evolution, some patients (pts) will develop a malignant transformation. Thus, prognostic factors that may identify pts who will progress are important. We update our experience published before. Methods: we performed a retrospective analysis of 387 pts with MGUS diagnosed between 1982 and 2008 in our institution to identify simple haematological variables associated with progression. Actuarial progression free survival (PFS) and overall survival (OS) were also calculated. MGUS was diagnosed when monoclonal component (MC) was present at a concentration of 3 gr per decilitre or less, no or only moderate amounts of monoclonal light chains in the urine, absence of lytic bone lesions, anemia, hypercalcemia and renal insufficiency related to monoclonal protein; and if performed a proportion of plasma cells of 10 percent or less. MC was detected by agarose gel and/or cellulose acetate electrophoresis in serum and/or urine. The identification of the MC was performed by immunoelectrophoresis or immunofixation and the quantification of immunoglobulins, by radial immunodifussion. Progression to myeloma was defined by: MC & gt; 3 gr/dl, plasma cell infiltration & gt; 10%, or associated lytic bone lesions. Progression to another B-cell neoplasm was considered when there was histologic evidence of the disease. The identification of prognostic factors was made using Cox models. PFS and OS were calculated using the Kaplan Meier method and the curves were compared with the log-rank test. Results: The median (md) age at diagnosis was 62 years (range 24–89 years) and 18% of the pts were younger than 50. Anemia not related to the monoclonal protein was present in 18% of the pts. Albumin, beta 2 microglobulin and erythrocyte sedimentation rate (ESR) were normal in 88%, 50% and 41% of the pts, respectively. MC was 0.6 gr/dl (range 0.1–2.9). In 70% of the cases it was IgG, 15% IgA, 13% IgM and 2% biclonal. The light chain was Kappa in 65% of the pts. Bence Jones was detected in only 9%. Uninvolved immunoglobulins (UI) were reduced in 21% of the pts. Bone marrow analysis were performed in 79 pts (20%) and md plasma cell infiltration was 3 (range 0 – 10). The 387 pts were followed for 2340 person-years (md 4.75 years, range 0 – 31) during which 31 pts (8%) evolved to a malignancy (23 multiple myeloma, 7 NHL, and 1 amyloidosis) and 17 (4.6%) died (3 related to progressive disease, 6 related to non hematological malignancy, 2 associated to cardiovascular disorders, 2 secondary to infection and 4 of unknown cause). PFS and OS at 10 years were 89% and 91%, respectively. The overall risk of progression was 1.3% per year. Among all the variables analysed (age, gender, hemoglobin, albumin, beta 2 microglobulin, ESR, type and concentration of MC, Bence Jones and reduction of UI), only MC concentration (HR 4.81, CI 2.2 – 10.32, p = 0.00007) and ESR (HR 3.67, CI 1.7 – 7.88, p = 0.001) had prognostic value for progression. Conclusions: MC concentration and ESR at diagnosis identified a subgroup of pts with higher risk of transformation in our experience.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1739-1739
    Abstract: Introduction: Chemoimmunotherapy with 6 cycles of fludarabine, cyclophosphamide and rituximab (FCR) is considered standard therapy for physically fit patients with chronic lymphocytic leukemia (CLL). Due to treatment toxicity, some patients are unable to undergo standard 6 cycles of FCR. We evaluated safety and efficacy of abbreviating FCR treatment to 4 cycles in a cohort of 35 untreated physically fit CLL patients who achieved CR with negative minimal residual disease (MRD). Patients and methods: Within 150 physically fit CLL patients treated with FCR on 1st line at our Center, from April 2003 to November 2014, a subgroup of 35 patients interrupted treatment after achieving negative MRD at the end of the 4th cycle. Median age at start of treatment was 62.8 years (34-81). Binet A/B: 24pts and C: 11. CD38 expression was positive ( 〉 7% off cells) in 57.1% and negative in 9% of the pts. A bone marrow biopsy was performed at start of treatment and 1 month post 4th cycle. Response was assessed in peripheral blood (PB) or bone marrow (BM). Negative MRD was defined as 〈 10-4. We used NCI criteria for response modified for the evaluation of MRD by flow cytometry. Progression was defined according to the NCI recommendations. Overall survival (OS) was defined as the time of initiation of therapy until death or last follow-up and progression free survival (PFS) as the time to progression. Data analysis included frequency and contingency tables, survival curves were plotted by the Kaplan Meier method. Treatment schedule: Fludarabine 25 mg/m2 IV day 1-3, cyclophosphamide 250 mg/m2 IV day 1-3, rituximab 375 mg/m2 IV day 3 cycle 1 and day 1 cycles 2-4, in all cycles every 28 days. Results: All 35 patients had negative MRD in PB after one month post 4th cycle. In addition, 28 had bone marrow evaluation showing CR with negative MRD in all of them. No splenomegaly nor hepatomegaly, enlarged lymphadenopathies nor lymphocytosis was observed in all the patients with negative MRD. After a median follow-up of 57 months (7 -141), median PFS was 65.8 months, not being yet reached the median of OS. PFS and OS at 72 months was 46% and 68% respectively. A total of 10 pts ( 3.5%) died: 7 on progressive disease, 3 on secondary neoplasms. Patients who progressed before 24 months had a median of survival of 22 months; median not reached on the group who progressed after 24 months (p=0.0001). Neutropenia grade 3-4 and infectious events were observed in 25.7% and 9.1% during all cycles respectively. Grade 3-4 neutropenia showed to increase over time (Cycle 1: 24%, Cycle 4: 39%). There was no treatment related death. Conclusion: With a long median follow-up, abbreviating treatment to 4 courses of FCR in patients who obtained negative MRD showed durable remissions with high PFS and OS at 72 months, minimizing treatment related toxicity. Sixty five percent of the patients who progressed after 24 months are still alive. Large randomized trials will be necessary to confirm our data. Disclosures Pavlovsky: Bristol Myers Squibb: Honoraria, Speakers Bureau; Novartis: Consultancy, Honoraria, Speakers Bureau. Pavlovsky:Novartis: Honoraria, Speakers Bureau; Janssen: Honoraria, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 1619-1619
    Abstract: Abstract 1619 Background: Positron emission tomography using 18F-fluoro-2-deoxy-d-glucose (FDG-PET-CT) is an important tool for treatment response assessment in Hodgkin Lymphoma (HL) treated with ABVD. It can predict response and overall outcome. The negative predictive value for PET-CT in patients (pts.) with HL is 90–94%. New recommendations define complete remission (CR) for HL as the lack of signs and symptoms of lymphoma with a negative PET-CT. OBJECTIVES: Reduce therapy in pts. who achieve early CR with negative PET-CT. Intensify treatment, only in pts. with positive PET-CT after 3 cycles of ABVD. Achieve CR, event free survival (EFS) and overall survival (OS), as good as in our historical control, when we used 3 or 6 cycles of ABVD plus involved field radio therapy (IFRT) in all pts.(LH-96) PATIENTS AND METHOD: Since October 2005, 200 newly diagnosed pts. with HL have been included in a prospective multicenter clinical trial (LH-05) All pts. received 3 cycles of ABVD and were then evaluated with a PET-CT (PET-CT +3) Pts. with a negative PET-CT+3 and absence of other signs or symptoms of lymphoma were considered in CR and received no further therapy. Pts with more than 50% of anatomic reduction of initial masses but persistent hyper metabolic lesions were considered in partial response (PR) and completed 6 cycles of ABVD and IFRT on PET-CT positive areas. Pts with less than PR received high doses of chemotherapy and an autologous stem cell transplant (ASCT). All pts were re-evaluated at the end of treatment with a new PET CT. One hundred and ninety three pts. have been evaluated. The median age at diagnosis was 29 years. One hundred and twenty five (65%) had localized stage (I-II) non bulky and 68 (35%) presented with advanced stage (III-IV), or bulky disease, 33 (17%) had bulky disease. RESULTS: One hundred and forty-eight (77%) achieved CR with negative PET-CT + 3. Forty-five (21%) were PET-CT+3 positive, 5 showed progressive disease. The other 40 pts. were in PR and completed a total of 6 ABVD + IFRT in PET-CT positive areas. Twenty eight achieved CR and 12 persisted with hypermetabolic lesions. Three died of progressive disease. After finishing planned treatment 178 pts. (92%) were in CR. With a median follow up of 39 months the EFS and OS at 36 months is 80% and 97% respectively. Patients with negative PET-CT +3 have an EFS of 86% compared to 61% for pts. with positive PET-CT+3 (P=0,001). We perform a multivariate analysis for EFS which included age, stage, IPS, bulky disease, extranodal areas and the result of the PET –CT+ 3. This last parameter together with age were the only ones with statistical significance (p=0.001 and 0.046 respectively). When comparing the results LH-05 with LH-96 there is no difference in EFS and OS at 36 months (83% vs. 85% and 97 vs. 96%) but in LH-05 only 23% received 6 cycles of ABVD and IFRT compared to 61% and 100% in LH-96. This reduces the exposure to chemo and radiotherapy. CONCLUSION: With PET-CT adapted therapy after 3 cycles of ABVD, 148 pts.(77%) received only 3 cycles of ABVD as initial therapy with an EFS and OS of 80% and 97% at 36 months. In the Cox regression model, PET-CT at completion of treatment was the most significant factor associated to EFS. In this interim analysis of PET-CT adapted therapy to all stages of HL, treatment with 3 cycles of ABVD can be adequate for pts. with negative PET-CT+3. Continuing with ABVD after a positive PET-CT +3 can be considered insufficient. A longer follow-up and a larger number of pts. are necessary to confirm these results. 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4408-4408
    Abstract: Different treatment guidelines suggest that advanced follicular lymphoma (AFL) subjects should be treated only when meeting criteria treatment, such as GELF, are present. Conversely, when absent the watch and wait (W & W) approach is recommended. However, in our country, we had the impression that in real life, a high percentage of patients without the above-mentioned criteria were treated. With the purpose of unravelling the medical approach of AFL patients at diagnosis and subsequent evolution, the Lymphoma Subcommittee of the Argentinian Society of Haematology undertook this retrospective survey. Results: From years 2006 to 2014 305 patients from 23 institutions were included. GELF criteria were encountered in 62% of patients at diagnosis and all of them were treated with immunochemotherapy (ICT). Among the 116 (38%) patients without meeting GELF criteria (GELF negative group), in only 30 (26%) W & W was the approach chosen, while the rest received ICT. The survey questionnaire revealed that own assessment of the treating physician was the main reason for treating the GELF negative group. In the W & W group, 60% required ICT at a mean of 17 months, being 15% of them transformed to DLBCL at time of treatment. The 89% of cases (271/305) received ICT at some time; 66% R-CHOP, 29% R-CVP, and 5% other regimes. Patient median age receiving R-CHOP and R-CVP was 57 and 62 years (p 〈 .01), respectively. Rituximab maintenance (RM) was added to ICT in 64% of cases. For the whole group, with a median follow up of 36 months, the overall survival (OS) was 95% and progression free survival (PFS) 68%. Comparing GELF negative and positive groups, PFS was better for GELF negative group, 87% vs 61% (p 〈 .01). There was no difference in OS. Within the GELF negative group, OS was not different between patients treated at the time of diagnosis vs those in which a W & W approach was chosen. Conclusion: 1) When comparing with international reports, the percentage (62%) of patients with positive GELF criteria was higher at diagnosis. This fact may be due to delay in access to health care; 2) we found a remarkable discrepancy among guidelines recommendations and real life medical behaviour. Three out of four patients received treatment at diagnosis, when W & W ought to have been the guideline-recommended approach; 3) R-CHOP was the most used ICT scheme, while R-CVP was mostly reserved for the elderly. RM was indicated in the majority of patients, particularly after year 2011; 4) despite acknowledging the methodological limitations of this retrospective analysis, a high tumor mass (GELF positive) picture conferred a worse prognosis in term of PFS, while a W & W approach did not affect the OS for the GELF negative group. Disclosures Riveros: Roche: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 1454-1454
    Abstract: Background: The prognostic score for Hodgkin’s lymphoma was defined as the number of adverse prognostic factors presented at diagnosis. Seven factors had similar independent prognostic effects. This model was validated retrospectively in advanced disease using different therapeutic approaches (D Hasenclever et al N Eng J Med339:1506–14, 1998). Methods: From December 1996 up to October 2005, the GATLA completed a risk-adapted therapy with ABVD and IFRT. Patients with stages I-IIIA without bulky disease, who achieved complete remission (CR) after three cycles of ABVD, favorable group (FG) received only IFRT 25 GY to areas of & gt;2 cm at diagnosis. Patients with FG not in CR after three cycles of ABVD, slow responders (FGSR), all stages IIIB-IV and all bulky disease, unfavorable group (UG) received six cycles of ABVD and IFRT 30 GY at remaining areas after 3 cycles of ABVD. A total of 584 patients, completed therapy; of them 513 were evaluated with the IPS. Patients were divided in three groups according to the number of adverse prognostic factors 0–1, 2–3, and ≥ 4. Results: The number of patients, complete remission (CR) rate, event-free survival (EFS) and overall survival (OSV) at 5 years according to prognostic factors in the 513 patients were as follows: IPS # patients (%) # CR (%) % EFS % OSV 0–1 224 (44) 217 (97) 86 95 2–3 241 (47) 213 (88) 73 90 ≥4 48 (9) 40 (83) 65 72 P & lt; 0.020 0.001 0.001 A total of 200 patients with FG had a 5 years EFS and OSV of 89% and 98% while 53 patients with FGSR had an EFS and OSV of 66% and 88% respectively (P & lt;0.001). The IPS in FG and FGSR was 0–1 of 61% versus 49%, 2–3 of 38.5% versus 43% and ≥4 of 0.5% versus 8% respectively (p=0.003). In UG with an EFS and OSV of 72% and 87%, the incidence of IPS 0–1 was 29%, 2–3 was 54% and ≥4 was 17%. Conclusion: The IPS is an excellent tool to predict outcome. Patients with stages I-IIIA without bulky tumour who did not achieve CR after three cycles of ABVD (FGSR) had poorer IPS than FG. In spite of receiving six cycles of ABVD, those with FGSR instead of three of those with FG had statistically a poor outcome. In the PET-TC era, patients who remain positive after three cycles of ABVD will need an intensified therapy with the purpose of improving the bad prognosis.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 5445-5445
    Abstract: Introduction: Early reduction of BCR-ABL transcript level has been associated with improved outcomes in CML treatment. Inability to achieve early molecular response(MR) at 3 months (M3 〉 10%) is considered a predictor factor for unfavourable outcome. However, the kinetics of BCR-ABL transcript level reduction measured at early time points have shown to be an independent predictor of response.The aim of this analysis was to determine whether the "M3-M6" status is critical to categorize CML patients (pts) focusing in high-risk group. Method: Molecular monitoring was performed in all pts prior treatment (M0), at months 3 (M3), 6 (M6), 12 (M12) and every 6 months thereafter, applying Q-PCR method according international recommendations. Results of BCR-ABL1 transcript level were reported on the international scale as IS-BCR-ABL %. Optimal responses: M3≤10%, M6≤1%, M12≤0,1%. Deep responses (MR4.0): ≤0,01% or undetectable/10.000 ABL copies. Results: A total of 70 CML pts were included, median age 49 (19-82), female 39%. First line treatment: sustained branded 81% and generic 19% TKIs. Imatinib 59%, Dasatinib and Nilotinib 41%. Sokal risk score: low (L) 51%, intermediate (In) and high (H) 49%. Optimal responses at molecular milestones: 75% at M3, 72% at M6, 61% at M12 and 53% pts achieved MR4.0. Event-free survival (EFS) was evaluated according to time point M3: M3≤10% group had significantly better EFS compared with the M3 〉 10% (96% vs 70%; P=0.028). M3-M6 status defined 4 groups of pts: M3≤10%-M6≤1%, M3≤10%-M6 〉 1%, M3 〉 10%-M6≤1%, M3 〉 10%-M6 〉 1%. Molecular response evolution by M3-M6 status is described in Table 1. EFS stratified by groups according to combined M3-M6 responses showed significant differences: 92% for group 1, 87% for group 2, 68% for group 3, 54% for group 4. (P=0.002). M6 time point was shown to be critical in 32 high-risk pts (H+In): 17 pts with M6 ≤1% showed significant differences in MR4.0 achievement compared to 15 pts with M6 〉 1% (82% vs 27% P=0.02). Better EFS was observed in this high-risk group under branded vs generic TKIs treatment (97% vs 54% P=0.04). Statistical differences in deep responses and MMR at M12 were observed between branded and generic TKIs independently of Sokal risk (P=0.06, P=0.02). Conclusions: M3≤10% pts showed a favourable evolution with better EFS than M3 〉 10% group. However not all patients with M3 〈 10% had a similar behavior, showing lower rates of MMR at M12 and deep responses in those pts with M6 〉 1%. In pts with M3 〉 10% and optimal response at M6 also showed higher MR4.0 rate. Our study supports that M6 is a crucial endpoint to predict MMR at M12 and deep responses in CML pts.Pts with M3≤10% without optimal response at M6 ( 〉 1%) had a worse evolution than those slow responders who showed M3 〉 10% and M6≤1%.High-risk pts are still a challenge, observing better outcomes in those under branded TKIs treatment. The M3-M6 status would be a prognostic marker of responses and EFS in chronic phase CML pts treated with TKIs. Our data support the critical role of M6 response in non-optimal M3 〉 10% pts and intermediate and high risk Sokal score. Treatment adherence is mandatory for achieving and sustaining optimal responses. This multicentric Argentine study, reinforces the importance of clinical follow-up and molecular monitoring under IS standardization at early time points. Education on early molecular monitoring with adequate resources must continue to be an objective in our region. Table 1 Table 1. Disclosures Pavlovsky: Roche: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Speakers Bureau. Moiraghi:Novartis: Speakers Bureau; Bristol: Speakers Bureau. Varela:Novartis: Speakers Bureau; Bristol: Speakers Bureau. Enrico:Novartis: Honoraria, Patents & Royalties; Bristol Myers squib: Speakers Bureau. Brodsky:International PNH Registry: Other: -; Alexion Pharma Argentina: Speakers Bureau. Pavlovsky:Novartis: Speakers Bureau; Janssen: Speakers Bureau; Bristol Myers Squib: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 5149-5149
    Abstract: INTRODUCTION: Ponatinib is a potent TKI indicated in T315I mutated CML and not mutated CML resistant to other drugs. Deep responses were reported in PACE trial in patients who had failed all other TKIs in chronic and advanced phase CML with acceptable toxicity profile. Ponatinib use is authorized in United States and Europe but compassionate use allows access in Argentina with no previous reports of local results. OBJECTIVE: To analyze a cohort of CML resistant patients treated with ponatinib: clinical characteristics, outcome and adverse events. MATERIALS AND METHDOS: Retrospective, multicentric, observational, descriptive study. Data was collected from charts review of patients with resistant CML treated with ponatinib at 8 different centers. Mutational status was reported. Frequency of Complete Hematologic Response (CHR), Complete Cytogenetic Response (CCgR) and Major Molecular Response (MMR) at 3, 6 and 12 months, adverse events and death were analyzed. RESULTS Twenty three patients were included with median follow-up from CML diagnosis 119 months (m) (IQR: 12-215) and 6 m (IQR: 2-21) from ponatinib first dose. Median age at CML diagnosis was 41 year-old (5-61). At the moment of ponatinib first dose 74% (17/23) patients were in chronic phase (CP), 13% (3/23) in accelerated phase and (AP) and 13% (3/23) in blast crisis (BC). Ponatinib was indicated as second line treatment in 4% (1/23), third line treatment in 9% (2/23), fourth line 83% (19/23) and fifth line 4% (1/23). Mutations were detected in 83% (19/23) patients with presence of T315I mutation in 44% (10/23). Median time from mutation detection to ponatinib start was 6 m (1-12). Ponatinib dose was 45 mg/d in 48% (11/23) and 55% (6/11) of these patients required dose reduction, 52% (12/23) received 30mg/d. Treatment have been discontinued in 8% (2/23) due to safety reasons. All patients in CP with evaluable response achieved CHR with median time to CHR of 1m, 27% (3/11) achieved CCgR, and 18% (2/11) obtained MMR by 3 m y 6 m, and 37% (3/8) by 12 m. Of patients in AP/BC 83% (5/6) achieved no molecular response and only 1patient have received ponatinib associated with standard chemotherapy, obtained MR4.5 and underwent unrelated bone marrow transplantation and is alive --- months after BC T315I mutated CML. Disease progression occurred in 6% (1/17). Death occurred in 4/23 (17%) all advanced phase at start of ponatinib. Adverse events are reported in table 1:Table 1.ADVERSE EVENTINCIDENCETOXICITY GRADEHematologic21% (5/23)1-2- 3Dermatologic13% (3/23)1-2-3Hypertension9% (2/23)2- 3Mialgia4% (1/23)3Palpitations4% (1/23)2Hipertriglyceridemia4% (1/23)4Hepatotoxicity4% (1/23)2Disnea4% (1/23)3Arterial Thrombosis4% (1/23)4Grade 4 toxicities occurred in 2 patients receiving 30 mg/d. Arterial thrombosis was the cause of death of one patient and cardiac toxicity mandated drug interruption in another. CONCLUSION: This is the first report of ponatinib use in daily practice in Argentina.Response rates are lower than those reported in the literature. Drug was safe with low discontinuation rates although severe cardiac toxicity occurred as reported. Time to first dose in Argentina may be a relevant factor to explain the lower rates of response in patients who have failed other ITKs. Disclosures Varela: Bristol Myers Squibb: Speakers Bureau; Novartis: Speakers Bureau. Enrico:Novartis: Speakers Bureau; Bristol Myers Squibb: Speakers Bureau. Pavlovsky:Novartis: Consultancy, Honoraria, Speakers Bureau; Bristol Myers Squibb: Honoraria, Speakers Bureau. Bengio:Bristol Myers Squibb: Speakers Bureau; Novartis: Speakers Bureau. Pavlovsky:Janssen: Consultancy, Honoraria, Speakers Bureau; Novartis: Consultancy, Honoraria, Speakers Bureau. Moiraghi:Bristol Myers Squibb: Speakers Bureau; Novartis: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1590-1590
    Abstract: BCR-ABL transcript levels (IS%BCR-ABL) in CML patients (pts) allow to define molecular response (MR) to TKIs. MR at 3 months (M3 〉 10%) is the strongest predictor of bad outcome and a point of divergence for changing therapy. Emerging biomarkers are being proposed in order to optimize prognosis in addition to initial MR and other clinical markers. The rate of BCR-ABL decline measured as halving time (HT) for transcript level reduction (RT0.5) at month 3 in 1GTKI (imatinib) treated pts, and the fractional clearance as a relative BCR-ABL ratio (RR3) during the first 3 months in 2GTKI (dasatinib) treated pts were recently proposed as predictive markers of outcome. Optimal responses (OR) were defined upon %ISBCR-ABL values at 3, 6 and 12 months: M3 ≤10%, M6 ≤1% y M12 ≤0,1%. Early time point prognostication could allow better outcome predictions and rapid therapeutic interventions. Aim: to determine whether the rate of BCR-ABL decline as HT and the BCR-ABL transcripts reduction as relative ratio at M3 from baseline, have predictive value in CML pts treated with 2GTKIs. Method: BCR-ABL transcript level was measured by Q-PCR according international recommendations and results were reported as BCR-ABL/ABL on the international scale (IS BCR-ABL %). Molecular monitoring was performed prior treatment (M0) and at months 3 (M3), 6 (M6), 12 (M12) and every 6 months thereafter. RT0.5 from baseline to M3 was calculated as the time to halve by considering an exponential model of reduction during the period. The median RT0.5 and quartiles, were calculated to define rapid declines as those HT in the lowest quartile. RR3 was determined as a relative ratio of transcript clearance at M3 from baseline. The median RR3and quartiles, were considered to define high BCR-ABL clearance as those in the highest quartile. Fisher statistical test was used to determine whether the proportions of good responses were statistically different depending on the decline rate. Results: A total of 77 CML pts were included, first line treatment: Dasatinib (D) 28%, Nilotinib (N) 32% and Imatinib (I) 40%. Sokal risk score was available in 70% pts: low (L) 67%, intermediate (In) 22% and high (H) 11%. Molecular assessment: prior therapy M0 (90%), M3 (60%), M6 (79%), M12 (78%) and M0+M3+M12(39%). Deep responses (MR4.0) were considered as IS%BCR-ABL 〈 0,01 or no detectable transcripts in ≥10000 ABL copies. OR according to first line TKI (I vs D+N), were: M3≤10%: 75% vs 90% and M12 ≤0,1%: 50% vs 65%. RT0.5 median values for I vs D+N groups were 32d vs 16d and RR3 median values were 79% vs 98%. RT0.5 ≤13d and RR3 ≥99% were cut off values for fast responses and high clearance in pts treated with 2GTKI(83%). Milestone molecular result M3≤10% showed 20% pts with M12 〉 0,1% and without MR4.0. All of these pts showed RT0.5 〉 13d and RR3 〈 99%. From total pts with OR, 80% pts achieved MR4.0 and all showed RR3≥99% meanwhile the remaining 20% pts who never achieved MR4.0 had RR3 〈 99%. Higher frequencies of OR were observed in low Sokal risk pts, in RT0.5 ≤13d pts and in pts with RR3 ≥99%. High Sokal risk pts had poorer results at milestones molecular responses but those with rapid initial decline (RT0.5 ≤13d) and high BCR-ABL clearance (RR3 ≥99%) achieved molecular responses similar to those observed in the low risk sokal group. Conclusions: Faster BCR-ABL decline and high clearance were related to OR and MR4.0 achievement in 2GTKIs patients. No direct relation between Sokal Index and M12≤ 0,1% was observed. Patients with RT0.5 ≤13d showed higher frequencies of good responses. Statistical differences were not found probably due to low sample numbers. Our data suggests that RT0.5 in 2GTKIs (N+D) patients could have a similar predictive value as reported by Branford in 2014, even though it is known that BCR-ABL measurements tend to be less satisfactory when control gene is ABL, owing to possible non-linear ratios. Whilst there were no statistical correlations probably due to the low sample number, pts with OR but no MR4.0 had low reduction rate RR3 〈 99%. RR3 〉 99% could provide a stronger predictive value for milestone optimal responses achievement in high risk pts. It will be necessary to extend this study including a higher number of patients treated with 2G TKIs. Figure 1. Figure 1. Disclosures Varela: Bristol Myers Squibb: Speakers Bureau; Novartis: Speakers Bureau. Moiraghi:Novartis: Speakers Bureau; Bristol Myers Squibb: Speakers Bureau. Pavlovsky:Novartis: Honoraria, Speakers Bureau; Janssen: Honoraria, Speakers Bureau. Pavlovsky:Bristol Myers Squibb: Honoraria, Speakers Bureau; Novartis: Consultancy, Honoraria, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 5169-5169
    Abstract: Background Response to TKI is the most important predictor of survival in CML and its early acquisition predicts the achievement of deep molecular responses (MR) (Baccarani et al. Blood 2013). In current ELN recommendations, the optimal time point to achieve Major Molecular Response (MMR) is defined at 12 months (mo) after diagnosis of CML however, not achieving MMR is not a failure criterion at any time point (Saussele et al, Blood 2014). There are controversies about when CML therapy must be changed in pts who do not achieve MMR after 12 mo of treatment. We wonder if pts achieving late MMR are less likely to achieve deep MR or are at risk of progression to accelerated phase (AP) or blast crisis (BC). Objectives We aim to identify predictive factors for achievement of deep MR and improvement of failure-free survival (FFS). Other objective was to determine if delay in MMR acquisition predicts outcomes in order to justify an optimal landmark for changing TKI reducing the risk of failure. Methods Between 2000 and 2015, 122 consecutive adult pts with CP CML were treated with various TKI modalities. Adherence (ADH) monitoring occurred in every visit and was reported in medical records, optimal ADH defined as 〉 90% of dose completed. Cytogenetic and MR were defined according to ELN definitions and followed the procedures as described elsewhere (Cross et al. Leukemia 2012). In particular, BCR-ABL 〈 1% corresponds to complete cytogenetic remission (CCyR) (Lauseker et al. 2014). MMR: BCR-ABL transcript 〈 0.1% IS. Confirmed MR4.0: BCR-ABL detection ≤0.01% IS in two consecutive analyses. Failure was defined as loss of CCyR, AP or BC. Different models were performed for CCyR at 6mo, MMR at 12mo and MMR at 24mo as predictors of MR 4.0; in all cases age and Sokal were used as covariates. Failure-risk was evaluated with Cox regression including in the model: Time to MMR, ADH and Sokal as covariates. Results From 122 pts diagnosed with CP CML, 110 pts received 1st line imatinib (IM) 400mg daily and constitute the basis of the analysis. Median age was 48 years (14-82), 65% were males, 82% low Sokal score. With a median follow-up of 107 mo (IQR 69.5-141) 82% pts are still on initial IM, 4.5% died and 1.8% were lost to follow-up. A total of 20 pts (18%) discontinued Imatinib, 9% due to intolerance and 9% due to failure. At 15 years, FFS was 88% and OS 95%.The proportion of pts treated with IM who achieved CCyR at 6mo and MMR at 12mo was 84% and 38% respectively. Median-time to achieve MMR and MR4.0 was 2.6 and 3.6 years respectively. By logistic regression analysis CCyR at 6mo OR 5,6 (95% CI 1,6-19,00), MMR at 12mo OR 5,3 (95% CI 1,4-21,0), ADH OR 7,0 (95% CI 1,3-37,0) and not MMR at 24mo OR 2,5 (95% CI 0,9-7,2) resulted independent predictors for achieving MR4.0 and confirm earlier observations. In Cox model of proportional hazards for risk of failure, time to achieve MMR HR 1,02 (95% CI 1,01-1,04), ADH HR 6.1 (95% CI 1,38-26,8), and Sokal HR 4,7 (95% CI 1,12-19,3) were independent predictors of failure. Time to achieve MMR is a continuous variable, so the HR associated with it, is equivalent to a 2% monthly increase in the risk of failure. To calculate HR in other time intervals, the product of the coefficient model (B) and time of interest (in mo) were used as an exponent of the base of the natural logarithm (e). Not reaching MMR at 12 mo correlates with higher risk of failure: hazard-risk (HR) 1.30, (95% CI 1.28-1.30) and the risk increases with every passing year not achieving MMR (Table 1). Table 1.Time to MMR as predictor of progressionNo MMR achievement in different time intervalsHRCI 95%After 12 mo of TKI1,30(1,3-1,3)After 24 mo of TKI1,70(1,7-1,7)After 30 mo of TKI2,21(2,2-2,2)After 60 mo of TKI3,74(3,7-3,8) Conclusions The prognostic value of CCyR at 6mo and MMR at 12mo was confirmed. The association between early MMR and deep MR was confirmed. In this long-term follow-up Latin American real-world cohort of patients ADH was also an early predictor for achievement of deep MR. Although good responses are obtained with IM, some patients still progress, this is the reason why it is necessary to identify those who will relapse so as to consider a change in treatment to another TKI at a convenient time point. Time to achieve MMR, ADH, and Sokal Risk score were independent predictors of failure. Not achievement of MMR at 12mo correlates with increased risk of failure. The risk increases annually in proportion to the time delay. Patients who are delayed in reaching MMR should be controlled closely. Disclosures Pavlovsky: Novartis: Honoraria, Speakers Bureau; Janssen: Honoraria, Speakers Bureau. Pavlovsky:Novartis - Bristol: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 5247-5247
    Abstract: MF is a myeloproliferative neoplasm characterized by bone marrow fibrosis, splenomegaly, cytopenias and constitutional symptoms. Since the identification of JAK2V617F mutation and development of anti JAK molecules, the course of the disease has changed. Ruxolitinib is a JAK1 and JAK2 inhibitor recently approved for patients (pts) with primary and secondary MF. In Argentina, pts could access the ruxolitinib through CUP. Analysis of this multicenter protocol provides a preliminary data set, follow up and efficacy results. Objectives To assess the efficacy (reduction in spleen size/improvement in constitutional symptoms) and toxicity. To investigate risk groups modifications during follow-up. Methods In Argentina, 36 pts with myelofibrosis, including primary MF (PMF) and post polycythemia vera (PPV) received ruxolitinib through CUP regardless of JAK2 mutational status. Study period: September 2011 to June 2013. The participating physicians provided information of the disease characteristics by completing a data form. Splenomegaly was evaluated by physical exam, and constitutional symptoms were categorized as present or absent. Retrospective analysis was performed based on data at baseline (n 36), after 3 months (n 30), 6 months (n 24) and 12 months (n 14). Results Median follow-up is 10 months (1-20). The median age is 65 years (30-79), 64% were men and 36% women.  There were 72% positive for the JAK2 V617F mutation, 69% had PMF and 31% Post-PV. At admission 89% of pts had received ≥1 lines of therapy for MF prior to ruxolitinib: hydroxyurea 77%, thalidomide 26%, erythropoietin 23%, others: corticosteroids, interferon, anagrelide, danazol, busulfan, splenic radiation, cytarabine, and 6-mercaptopurine. The distribution of risk category according to DIPSS was: 61% high, 19% Intermediate 2, 17% Intermediate 1, and 3% low. Median value for spleen size was 15 cm (4-33) below the costal margin. Constitutional symptoms were present in 71% of the patients and 33% were transfusion dependent. ECOG (Eastern Cooperative Oncology Group) was zero 28%, one 44%, two 22%, three 3%, and four 3%. The median hemoglobin, leucocytes and platelets was 10 g/dl (4.8-15.2); 13, 4 x 109/L (3.6-64) and 217 x 109/L (75-850), respectively. Peripheral blood blasts ≥ 1% in 44% of pts. Ruxolitinib therapy was initiated at 40 mg/d in 16 pts (50%), 30 mg/day in 9 pts (28%), and at a lower dose in the minority of pts. The median value for spleen size at 3, 6 and 12 months was 10, 8 and 6.5 cm, representing decrease of 20%, 41% and 42% from baseline, respectively (fig 1.). In 13 of 29 pts (45%) spleen size decreased ≥ 50% in 3 months. Constitutional symptoms not present at 3, 6 and 12 months in 79%, 100% and 87% of pts, respectively. Improvement in ECOG class was noted in 73%, 69 % and 73% at the above mentioned time points, respectively. Persistence of transfusion dependence at 3, 6 and 12 months was seen in 33, 24 and 27% of the patients. Hemoglobin median levels during treatment (g/dL): at baseline 10.1, at 3 months 8.5, at 6 months 9.7, and at 12 months 9.5. During follow-up, 27% pts shifted to a lower risk group, 2 pts (7%) to a higher group due to worsening anemia, and 67% did not change its category. Hematologic adverse events (AE) were anemia and thrombocytopenia in 8 pts (Grade 1 and Grade 3 according to CTCAE v4.0), 31% of pts required dose adjustment. There were no suspensions of medication because of cytopenia. Others AE 〉 10% were: headache, musculoskeletal pain, and diarrhea ( 〈 G3). Treatment was discontinued in 7 pts (MF progression n= 2, tuberculosis n=1, death n=4 non related to ruxolitinib). Conclusion Ruxolitinib is an effective therapy that reduced spleen size and improved constitutional symptoms with an acceptable toxicity profile in this group of pts. During follow up at 12 months, a quarter of pts shifted to a lower risk group. Our data is consistent with previous reports Disclosures: Barreyro: GSK: Employment. Enrico:Bristol Myers Squibb: Speakers Bureau; Pfizer: Membership on an entity’s Board of Directors or advisory committees; Novartis: Membership on an entity’s Board of Directors or advisory committees. Lanari Zubiaur:Novartis: Membership on an entity’s Board of Directors or advisory committees. Pavlovsky:Novartis: Consultancy. Sackmann:Novartis: Membership on an entity’s Board of Directors or advisory committees. Bengió:Novartis: Member advisory Board Myelofibrosis 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...