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. 120, No. 21 ( 2012-11-16), p. 2555-2555
    Abstract: Abstract 2555 Background: The prognosis for patients with chronic myeloid leukaemia (CML) has vastly improved through the use of tyrosine kinase inhibitors (TKIs), such as imatinib. However, a proportion of patients will not respond, or will lose their initial response, and the biological mechanisms underlying this heterogeneity are poorly understood. Human organic cation transporter-1 (hOCT-1 or SLC22A1), the main transporter for imatinib, has been proposed as one determinant. This study set out to assess the prognostic value of hOCT-1 expression and the presence of polymorphisms in the hOCT-1 gene. Methods: hOCT-1 mRNA levels in 153 diagnostic whole blood samples from two different patient cohorts (one from a trial population, and the other from patients treated only at our institution) were measured by RT-qPCR (normalised against two control genes). hOCT-1 exon 7 DNA (and cDNA transcripts) were sequenced in 156 patients, and four cells lines, to identify insertions/deletions or single nucleotide polymorphisms (SNPs). Fragment length analysis using gene-scanner technology was also used to identify patients with insertions/deletions and to correlate genotypes with cDNA transcript lengths. Time to each endpoint (remission/imatinib failure) was compared according to the level of expression for each gene, or according to genotype, using Kaplan-Meier analyses and the log-rank test. Results and Conclusions: No significant differences in outcomes were found when comparing patients with high or low hOCT-1 expression (whether defined using the median or other cut-offs). The 408V 〉 M (g.1222G 〉 A) SNP in hOCT-1 exon 7 was found in 102/156 patients (22 homozygotes and 80 heterozygotes) and was associated in all cases with an eight base-pair insertion at the exon-intron boundary (rs113569197). This insertion was found to create an alternative splice site, leading to the transcription of an additional RNA/cDNA transcript in these patients, the sequence of which contains a premature stop codon soon after the splice site. Additionally, M420del was found in 52 patients (three homozygotes and 49 heterozygotes) and was not found in alleles containing the eight base pair insertion. The six possible combinations of these three alleles (N=no insertion/deletion, 8+=8bp insertion and 3−=3bp deletion) were found to give rise to five possible combinations of RNA transcript lengths, since patients with 8+8+ produce identical transcript lengths to those with 8+N (i.e. the normal transcript and one with an additional 8bp and premature stop). In the trial cohort (n=109), significant differences in time to 10% (p=0.01), 1% (p=0.0002) and 0.1% (p=0.0003) molecular responses (by the international scale) and time to imatinib failure (p=0.02) were seen when patients with 8+8+/8+N were compared to those with the remaining four genotypes (NN/N3−/3−8+/3−3−). However, this association was not replicated in the internal cohort, which was smaller (n=47) and more heterogeneous in terms of baseline characteristics and management. These findings may explain discrepancies in the results of previous studies that have examined the association between hOCT-1 expression and outcome, since a number used primer/probe sets that would be affected by the presence/absence of these polymorphisms. Our results suggest that while hOCT-1 expression is not a determinant of response, alterations in splice sites or amino acid sequence due to insertions/deletions may be. Further work is required to clarify the impact of these polymorphisms on hOCT-1 protein levels, function and responses to imatinib. Disclosures: White: Novartis Oncology: Honoraria, Research Funding; BMS: Research Funding; CSL: Research Funding. Marin:Novartis: Research Funding; BMS: Research Funding. Apperley:Novartis, Bristol Myers-Squibb, and ARIAD: Honoraria, Research Funding. Goldman:Novartis, Bristol Myers-Squibb, and Amgen: Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    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: Transplantation, Ovid Technologies (Wolters Kluwer Health), Vol. 59, No. 9 ( 1995-05), p. 1302-1307
    Type of Medium: Online Resource
    ISSN: 0041-1337
    RVK:
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 1995
    detail.hit.zdb_id: 2035395-9
    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. 1680-1680
    Abstract: Abstract 1680 We studied BCR-ABL1 transcript levels in patients with CML in chronic phase at 3, 6 and 12 months after starting imatinib to identify molecular milestones that would predict for overall survival and other outcomes more reliably than serial marrow cytogenetics. We analyzed 282 patients with CML-CP who received imatinib 400 mg/day as first line therapy followed by dasatinib or nilotinib if they failed imatinib. The median age was 46.3 years (range 13–86.4), 157 (55.7%) patients were male. The Sokal risk distribution was: 31.8% low, 40.1% intermediate and 28.1% high. The median follow-up was 69 months (range 17–131). BCR-ABL1 transcripts were analyzed in the peripheral blood at 12 week intervals using RQ-PCR. Results were expressed as percentage ratios relative to an ABL internal control and converted to the international scale. Complete molecular response (CMR) was defined as two consecutive samples with no detectable transcripts with the ABL1 control 〉 40,000 copies (the median ABL control in the CMR samples was 84,000). We employed a ROC curve to identify the cut-offs in transcript levels at 3, 6 and 12 months that would best predict patient outcome. Patients with transcript levels 〉 9.84% (n=68) at 3 months had significantly lower 8-year probabilities of overall survival (OS) (56.9% vs 93.3%, p 〈 0.0001), progression-free survival, cumulative incidence of CCyR and CMR than those with higher transcript levels. Similarly transcript levels 〉 1.67% (n=87) at 6 months and 〉 0.53% (n=93) at 12 months identified poor risk patients. However transcript levels at 3 months were the most strongly predictive for the various outcomes. When we compared OS for the groups defined molecularly at 6 and 12 months with use of the usual cytogenetic milestones, categorization by transcript numbers was the only independent predictor for OS (RR= 0.207, p 〈 0.0001 and RR=0.158, p 〈 0.0001). We validated our results by classifying 95 patients treated with imatinib first line at the Royal Liverpool University Hospital according to their transcript levels at 3, 6 and 12 months after converting their local their RQ-PCR results to the international scale. At each time point the high risk patients had significantly poorer OS, namely 69.1% vs 98.3% (p=0.003); 98.1% vs 71.2% (p=0.009) and 98.0% vs 74.4 (p= 0.016) than the good risk patients. The various transcript cut-offs could be refined in order to better identify those patients more likely to achieve CMR. The 57 patients who had a 3-month transcript ratio ≤0.61% had a 8-year CI of CMR of 84.7%, while the 222 patients with a ratio 〉 0.61% had a CI of CMR of only 1.5% (p 〈 0.0001). Similar thresholds with high predictive power could be identified for 6 and 12 months. We also tried to identify levels of transcripts at 6 and 12 months above which patients in CCyR were destined to fare worse than those with lower values. At 6 months and 12 months, 23 of the 109 and 20 of the 166 patients who were in CCyR had a transcript level higher than 0.53% (the value that we had previously identified as defining the 12 months high risk group); these patients had significantly worse OS (65.8% vs 95.9%, p=0.0002 and 81.5% vs 94.9%, p=0.01) than the 104 and 140 patients with lower transcript levels. During follow-up 118 patients failed imatinib and required alternative therapy. In order to explore further the impact of early measurements of transcript levels on outcome irrespective of the use of subsequent rescue treatment, we calculated the ‘current CCyR survival’ (c-CCyRS, i.e. the probability of being alive and in CCyR at a given time point). The 8-year c-CCyRS for the whole population was 76.6%. High risk patients had a significantly worse 8-year probability of c-CCyRS than good risk patients, namely 47.0% vs 91.1% at 3 months (p=0.0002) and 53.1% vs 91.3% (p=0.0001) at 6 months (Figure 1). These findings indicate that the prognostic value of early measurement of the residual leukemia burden retains its value even for patients requiring treatment with second generation tyrosine kinase inhibitors. In conclusion, a single measurement of BCR-ABL1 transcripts performed at 3 months is the best way to identify patients destined to fare poorly, thereby allowing early clinical intervention.Figure 1Figure 1. 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: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 30, No. 3 ( 2012-01-20), p. 232-238
    Abstract: We studied BCR-ABL1 transcript levels in patients with chronic myeloid leukemia in chronic phase (CML-CP) at 3, 6, and 12 months after starting imatinib to identify molecular milestones that would predict for overall survival (OS) and other outcomes more reliably than serial marrow cytogenetics. Patients and Methods We analyzed 282 patients with CML-CP who received imatinib 400 mg/d as first-line therapy followed by dasatinib or nilotinib if treatment with imatinib failed. We used a receiver operating characteristic curve to identify the cutoffs in transcript levels at 3, 6, and 12 months that would best predict patient outcome. We validated our findings in an independent cohort of 95 patients treated elsewhere. Results Patients with transcript levels of more than 9.84% (n = 68) at 3 months had significantly lower 8-year probabilities of OS (56.9% v 93.3%; P 〈 .001), progression-free survival, cumulative incidence of complete cytogenetic response, and complete molecular response than those with higher transcript levels. Similarly, transcript levels of more than 1.67% (n = 87) at 6 months and more than 0.53% (n = 93) at 12 months identified high-risk patients. However, transcript levels at 3 months were the most strongly predictive for the various outcomes. When we compared OS for the groups defined molecularly at 6 and 12 months with the usual cytogenetic milestones, categorization by transcript numbers was the only independent predictor for OS (relative risk, 0.207; P 〈 .001 and relative risk, 0.158; P 〈 .001, respectively). Conclusion A single measurement of BCR-ABL1 transcripts performed at 3 months is the best way to identify patients destined to fare poorly, thereby allowing early clinical intervention.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2012
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 868-868
    Abstract: The NF-κB transcription factor pathway is aberrantly activated in multiple myeloma (MM) and many other cancers, where it promotes malignancy by upregulating survival genes, thus providing a compelling rationale for therapeutically targeting this pathway in MM. However, despite aggressive efforts to develop a specific NF-κB or IκB kinase (IKK)β inhibitor, no such inhibitor has been approved, due to the preclusive toxicities associated with the general suppression of NF-κB. As a key pathogenetic activity of NF-κB in MM is to block apoptosis through the induction of target genes, an attractive alternative to globally inhibiting NF-κB would be to therapeutically target the non-redundant, cancer-specific downstream effectors of the NF-κB survival pathway. Recently, we identified the interaction between the NF-κB-regulated antiapoptotic factor, GADD45β, and the JNK kinase, MKK7, as a pathogenically critical and cancer cell-restricted survival module downstream of NF-κB and novel therapeutic target in MM. Further, we developed a D-tripeptide inhibitor of the GADD45β/MKK7 complex, DTP3, which effectively kills MM cells by inducing MKK7/JNK-dependent apoptosis and, importantly, is not toxic to normal tissues. Due to this cancer-cell specificity, DTP3 has similar anti-cancer efficacy to bortezomib, but more than 100-fold higher cancer-cell specificity in patient MM cells, ex vivo. DTP3 also displays potent and cancer-selective activity against MM in preclinical animal models, with no apparent side-effects, and far greater therapeutic index than existing treatments. DTP3 further displays synergistic activity with conventional MM therapies, such as bortezomib, suggesting a clinical utility as frontline combination therapy. Additionally, it retains therapeutic efficacy in MM cells that are resistant to most common MM treatments, suggesting further clinical utility. We currently aim to conduct a phase I/IIa clinical trial of DTP3 in MM to deliver clinical Proof of Concept for a cancer-selective NF-κB-targeting strategy as a highly effective novel therapy. This first-in-man study will commence in late 2015. We report here the results from the regulatory pharmacodynamics (PD), safety pharmacology, pharmacokinetic (PK), and toxicology studies. 28-day intravenous (i.v.) repeat dose toxicology studies in rat and dog demonstrate that DTP3 is well tolerated, with no significant target organs of toxicity at up to 17 times the optimal exposure in mouse efficacy models. Toxicokinetic (TK) analyses indicate that DTP3 does not accumulate on repeat dosing. Safety pharmacology studies indicate no adverse effect on the central nervous, cardiovascular or respiratory systems. In an Ames assay, DTP3 was not mutagenic. In the rat, i.v. DTP3 rapidly and extensively distributes to tissues, does not pass the blood-brain barrier, and is readily eliminated in urine (30%) and faeces (60%). No major metabolites have been identified. In vitro, DTP3 did not inhibit or induce major human cytochrome P450 isoforms, suggesting no potential for drug interactions via P450. In vitro, DTP3 is neither a substrate for nor inhibitor of P-gp or BCRP, nor is it a substrate or inhibitor of most transporters evaluated. However, DTP3 is a substrate for the uptake transporters, MATE1, MATE2-K and OATP1B3, with some inhibitory potential for MATE1 and MATE2-K at high concentrations. PD studies in a mouse xenograft model show that i.v. bolus injection of at least 10 mg/kg of DTP3 over 2 weeks, daily, every other day, or every 3 days, is highly effective in causing complete tumour regression. The PK and TK evaluation of DTP3 in rat and dog identified long plasma half-lives, modelled into a half-life of 20-24 hr in man. On these bases, we have proposed an initial clinical dosing regimen of DTP3 of three times per week i.v., testing doses from 0.5 to 20 mg/kg. The data also support a subcutaneous route of administration, and an i.v. bolus regimen of twice per week. Collectively, the preclinical package demonstrates the outstanding selective pharmacology and efficacy of DTP3, combined with excellent i.v. tolerability, wide therapeutic window, and favourable PK profile, and supports progression into clinical development. A companion biomarker programme has also been developed in order to inform patient stratification, demonstrate pathway-specific PD activity and proof of mechanism, and so maximise the chance of a positive clinical response. Disclosures Tornatore: Kesios Therapeutics Ltd.: Consultancy, Equity Ownership, Honoraria, Patents & Royalties. Adams:Kesios Therapeutics Ltd.: Consultancy. Kelly:Kesios Therapeutics Ltd.: Consultancy. Ruvo:Kesios Therapeutics Ltd.: Equity Ownership, Patents & Royalties. Oakervee:Celgene: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Novartis: Consultancy, Honoraria. Schey:Celgene Corporation: Honoraria. Apperley:Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; BMS: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; ARIAD: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Franzoso:Kesios Therapeutics Ltd.: Consultancy, Equity Ownership, Honoraria, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties.
    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 ...
  • 6
    In: Pesticide Science, Wiley, Vol. 16, No. 6 ( 1985-12), p. 673-683
    Type of Medium: Online Resource
    ISSN: 0031-613X
    Language: English
    Publisher: Wiley
    Publication Date: 1985
    detail.hit.zdb_id: 2003455-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 5987-5987
    Abstract: Introduction: Critically ill onco-hematology patients (pts) admitted to intensive care units (ICU) have poor prognosis. Mechanical ventilation, multiple organ failures and severe sepsis are factors associated with high mortality. Current literature identifies day 5 in ICU as a specific time point at which ceilings of care should be re-addressed. Patients and methods: We retrospectively reviewed all consecutive onco-haematology patients admitted to the ICU between October 2010 and December 2015. We classified pts according to the reason for ICU admission in 5 groups: a) respiratory failure without mechanical ventilation during the first 24h; b) respiratory failure and mechanical ventilation in the first 24h; c) sepsis without respiratory failure and without renal replacement therapy in the first 24h; d) renal replacement therapy without respiratory failure regardless of septic status; and e) needing hemodynamic support without respiratory failure, sepsis or renal replacement therapy in the first 24h. After 5 days of full intensive therapy we defined a successful 5-day ICU trial for each of the five groups as follows: a) no mechanical ventilation during 5 days; b) neutrophils 〉 1.0 or ² 2 organ failures by day 5; c) C-reactive protein decreased by 50% or normalised lactate by day 5; d) off renal replacement therapy by day 5; and e) no inotropic support on day 5. Patients who died during the first 5 days of ICU admission were considered failures and pts who were discharged from the ICU before day 5 were considered successes. Results: 166 pts were identified, with 202 ICU admissions. The median number of ICU admissions was 1 (1-4), with 138 (84%) having 1 admission, 20 (12%) 2 admissions, 4 (2.4%) had 3 admissions and 3 (2%) 4 admissions respectively. The median length of stay in ICU was 6 days (1-95). The median duration of hospital stay prior to ICU admission was 14 days (0-104). The diagnoses were: AML 28% (n= 57), ALL 8% (n=16), CML 8% (n=16), myelofibrosis 4% (n=7), MDS 4% (n=7), myeloma 11% (n=23), NHL 30% (n=61) and Hodgkin's lymphoma 2% (n=4). Regarding pre ICU treatment, 44% (n=88) received chemotherapy, 11% (n=22) underwent autologous stem cell transplantation and 40% (n=81) allogeneic stem cell transplantation. Of those, 30% had myeloablative and 70% reduced intensity conditioning and 29 (35%) were from HLA identical sibling, 47 (58%) unrelated and 6 (7%) haplo-identical donors. The disease status was complete remission (n=77, 38%), partial remission (n=28, 14%) and stable disease (n=96, 48%). The reason for admission to ICU was respiratory failure in 53% (n=107), 19% sepsis (n=39), 16% renal failure (n=32) and 11% hemodynamic failure (n=22). The median APACHE II score was 24 (10-51), the median SOFA score was 10 (2-21) and the median SAPS-II score was 45 (0-100). APACHE II and SOFA scores were significantly greater in non-survivors vs survivors (p 〈 0.0001). Overall 101(50%) pts survived their ICU admission and were discharged to the hematology ward. Of these, 31 (30%) died in hospital and 70 (70%) were discharged home. Estimated overall survival was 15% (95% CI 10-23) at 3 years post ICU admission. For the 5-day ICU trial we selected 138 pts with one admission. The distribution according to the different groups was: a) 56; b) 34; c) 17; d) 17 and e) 14. Overall 58 (42%) successfully passed the trial: a) 30 (53%); b) 14 (41%); c) 4 (23%); d) 7 (41%) and e) 3 (21%). Overall 41 (30%) pts failed the trial and were alive on day 5 and 39 (28%) died before day 5. The overall survival (Figure 1) for the 58 pts who passed the trial was 28% at 3 years. The overall mortality in ICU was 33% (19/58) for those who successfully passed the 5-day ICU trial, and was 71% (29/41) for those who failed. The overall survival for pts that successfully completed the 5-day ICU trial and were discharged to the hematology ward (n=39), was 49% at 3 years. Conclusions: In this study, 50% of onco-hematologic patients survived their ICU admission. The long-term overall survival was 15% at 3 years. Patients could be stratified according to the reason for admission and given an individualised 5-day trial: those who successfully completed their trial (42%) had a low ICU mortality (33%) and those who were subsequently discharged home had a long-term survival of 49% at 3 years. This study raises the possibility of offering a short-term ICU trial to onco-hematologic patients and perhaps allows for the ceiling of intensive care for those who fail the trial. Figure Figure. Disclosures MacDonald: Gilead Sciences: Speakers Bureau. Milojkovic:Ariad: Honoraria; Novartis: Honoraria; BMS: Honoraria; Pfizer: Honoraria. Apperley:Incyte: Speakers Bureau; Pfizer: Honoraria, Speakers Bureau; Novartis: Honoraria, Speakers Bureau; Ariad: Honoraria, Speakers Bureau; Bristol Myers Squibb: Honoraria, 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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 93-93
    Abstract: In CML, rapid reductions in tumor load, as defined by at least 65% Ph-negativity and/or a RT-qPCR 〈 10%IS at 3 mths, are associated with an improved probability of CCyR and better overall and progression free survivals. However some 15-20% of patients experience cytopenias shortly after starting TKI treatment and are managed by drug interruptions and/or dose reduction. The dilemma is whether these early periods of altered treatment should be considered in the interpretation of the 3 mth results. We investigated the impact of missing days of therapy and average dosing over the first 3 mths on the 3 mth RT-qPCR levels, CCyR at 12 mths and ability to remain on study, in patients treated in a phase III randomized study of imatinib (IM) versus dasatinib (DA) in newly diagnosed patients. RT-qPCR results at 3 mths were available for 585 (IM-292, DA-293) of 632 patients who completed 3 mths of therapy. Patients were divided according to their randomized drug and the amount missed: those who did not miss any days (IM0 [243], DA0 [211] ), those who missed 1-14 days (IM1-14 [38], DA1-14 [37] ) and those who missed 〉 14 days (IM 〉 14 [11], DA 〉 14 [45]). More patients on DA missed days of dosing (28%) than on IM (17%) with a median number of missed days for DA of 16 (range 1-62) compared to 12.5 (range 1-42) for IM p=0.008. Achievement of a RT-qPCR 〈 10%IS at 3 mths for IM0, IM1-14 and IM 〉 14 was 78.6%, 63.2% and 63.5% p=0.033 and 93.8%, 91.9% and 77.8% for DA-0, DA1-14 and DA 〉 14 respectively p=0.001. Predictably the chance of a RT-qPCR 〈 10%IS at 3 mths is higher with DA than IM but DA is less well tolerated in the early months. RT-qPCR 〈 10%IS at 3 mths occurred in 78.7% and 93. 8% of patients who took more than 95% of the standard doses of IM and DA respectively in contrast to 60% (IM) and 84% (DA) in patients who missed more than 20% of the prescribed doses. Thus missing drug or dose reductions are associated with a reduced chance of achieving a RT-qPCR 〈 10%IS at 3 mths, although the effect is not seen for missing 〈 14 days of DA, and is less marked for reduced average dosing of DA, suggesting that the higher potency of DA compensates for the impact of missing a few days of drug or dose reduction. 107/632 (17%) patients had discontinued the study by 12 mths and RT-qPCR results are available for all remaining patients. Using RT-qPCR 〈 1% (MR2) as a surrogate for CCyR, the 12 mth MR2 rates for patients on IM were 78%, 78% and 90% in the IM0, IM1-14 and IM 〉 14 cohorts p=0.5, and 96%, 88.6% and 79.5% p=0.026 for the DA0, DA1-4 and DA 〉 14 cohorts, confirming superiority of DA over IM for MR2 and the potential impact of missing early doses of DA. Whether this is related to inadequate dosing or whether failure to tolerate the recommended dose is indicative of higher risk disease is not yet clear. We then studied whether missing drug in the first 3 months predicts the ability to stay on trial. The proportion of patients able to take the study drug consistently through mths 3-12 were 91%, 71% and 57% in the IM0/DA0, IM1-14/DA1-14 and IM 〉 14/DA 〉 14 groups respectively. This correlated with discontinuing the study (treatment failure) for 12.5%, 38.1% and 35.7% of patients in the IM0, IM1-14 and IM 〉 14 cohorts and 4.4%, 12.8% and 18.8% in the equivalent DA cohorts. Thus the ability to tolerate daily drug in the first 3 mths predicts future tolerability and efficacy over the subsequent 9 mths and long-term compliance with the first line therapy. RT-qPCR monitoring was provided long-term for all patients entered into the study irrespective of their continuation in the study, so we were able to study the achievement of MR3 at 12 mths in an intention to treat analysis in all patients. With respect to average dosing over the first 3 mths MR3 was seen in 50%, 52.8% and 54.9% p=0.17 of patients who took 〉 95%, 80-95% and 〈 80% of the prescribed dose of IM and 68.1% ( 〉 95%), 59.4% (80-95%) and 53.1% ( 〈 80%) p=0.038 for the equivalent doses of DA. Similar results were obtained using the number of days of missed drug (data not shown), suggesting that early failure to tolerate IM as 1st line does not impact on subsequent responses as effective alternative therapy is available for the majority of patients. In contract failure to tolerate DA is associated with a reduced chance of MR3 at 12 mths. Patients who experience drug cessation/reduction of either drug in the first 3 mths are less likely to obtain RT-qPCR 〈 10%IS at 3 mths and are more likely to require a change of drug in the longer-term and therefore require close observation during the first year. Disclosures: Apperley: Ariad: Honoraria; Bristol Myers Squibb: Honoraria; Novartis: Honoraria, Research Funding; Pfizer: Honoraria. Clark:Novartis : Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding, Speakers Bureau; BMS: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Pfizer: Honoraria, Membership on an entity’s Board of Directors or advisory committees. O'Brien:Bristol Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria; Pfizer: Honoraria.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 107, No. 1 ( 2006-01-01), p. 205-212
    Abstract: Although most patients with chronic myeloid leukemia (CML) have the same initial molecular abnormality, the BCR-ABL fusion gene, the duration of chronic phase (CP) varies widely. To identify the possible molecular basis of this heterogeneity, we studied CD34+ cells collected at diagnosis from 68 patients with CML-CP. By using oligonucleotide microarray screening, we performed gene-expression profiling on 2 subsets of patients, one comprising patients with an “aggressive disease” who developed blastic transformation (BT) within 3 years of diagnosis (n = 10) and, at the other extreme, patients with an “indolent disease” whose BT occurred 7 or more years from diagnosis (n = 9). This screening revealed 20 genes differentially expressed in patients with aggressive and indolent disease, which were validated by quantitative reverse transcriptase/polymerase chain reaction (Q-RT/PCR). A multivariate Cox regression model identified the combination of low CD7 expression with high expression of proteinase 3 or elastase as associated with longer survival in the complete cohort of 68 patients. This differential pattern of gene expression probably reflects the intrinsic heterogeneity of the disease; if so, assessing expression levels of selected genes at diagnosis may be valuable in predicting duration of survival in patients treated with imatinib and the newer tyrosine kinase inhibitors.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    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. 112, No. 5 ( 2008-09-01), p. 2163-2166
    Abstract: Expression of CD7, ELA-2, PR-3, and the polycomb group gene BMI-1 reflects the intrinsic heterogeneity and predicts prognosis of patients with chronic myeloid leukemia (CML) who were not treated with allogeneic stem cell transplantation (allo-SCT). This study investigated whether expression of these genes determined outcome following allo-SCT in a cohort of 84 patients with chronic-phase (CP) CML. We found that patients expressing BMI-1 at a “high” level before allo-SCT had an improved overall survival (P = .005) related to a reduced transplantation-related mortality. In multivariate analysis, when adjusted for the European Group for Blood and Marrow Transplantation (EBMT)–Gratwohl score and other prog-nostic factors, there was an independent association between BMI-1 expression and grades 2 to 4 acute graft-versus-host disease (relative risk [RR] = 2.85; 95% confidence interval [CI] , 1.3-6.4; P = .011), suggesting that BMI-1 measured prior to allo-SCT can serve as a biomarker for predicting outcome in patients with CP-CML receiving allo-SCT, and may thus contribute to better therapeutic decisions.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...