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
Filter
  • American Society of Hematology  (43)
  • 1
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1827-1827
    Abstract: Background: HBI-8000 is an orally bioavailable member of the benzamide class of histone deacetylase inhibitors (HDACi), that inhibits cancer-associated HDAC enzymes (Class I and IIb). HBI-8000 has anti-tumor activity through various mechanisms of action, including epigenetic reprogramming and immunomodulation. It was recently approved by the Chinese FDA under the name chidamide (Epidaza) for relapsed or refractory (R/R) peripheral T-cell lymphoma (PTCL) with a recommended dose of 30 mg twice weekly (BIW). HBI-8000 is also being manufactured in the USA for clinical development outside of China. The preliminary results of a phase I trial of HBI-8000 to confirm the safety and maximum tolerated dose (MTD) in Japanese patients (pts) with advanced NHL are presented (NCT02697552). Methods: This is a multicenter, prospective phase I trial in Japan. Inclusion criteria: patients are eligible if they have histologically or cytologically proven NHL and no other standard therapy is available. The primary endpoint is the MTD based on the frequency of dose-limiting toxicities (DLTs) observed within 28 days of the first dose. Secondary endpoints include pharmacokinetic (PK) profile and anti-tumor activity. At the time of this abstract submission, the trial is still ongoing. Results: Thirteen out of 14 pts were eligible for the 1st cycle DLT assessment (6 pts in the 30 mg, 7 pts in the 40 mg cohort). Median age was 68 years, gender well balanced, and the majority of pts had ≥ 2 prior treatment regimens. Five pts had the diagnosis of adult T-cell leukemia-lymphoma (ATL), 2 pts presented with PTCL, 3 with diffuse large B-cell lymphoma (DLBLC), 2 with follicular lymphoma (FL), 1 with cutaneous T-cell lymphoma (CTCL), and 1 with marginal zone lymphoma. Overall, the treatment was well tolerated, and adverse drug reactions (ADRs) were predominantly hematologic, consistent with the previous experiences. There were 7 pts in the 40 mg dose cohort because one of the first 3 pts had to be replaced for incomplete dosing due to grade 3 hypertriglyceridemia which was not regarded as DLT by the Data Monitoring Safety Committee (DMC/SMC). In the 40 mg cohort, 2 pts were considered as DLTs by definition in the protocol: grade 4 neutropenia and grade 3 alanine transaminase (ALT) increase. Both pts were asymptomatic. The grade 4 neutropenia promptly resolved with the administration of G-CSF and the grade 3 ALT elevation resolved with dose interruption. The 30 mg dose cohort completed with no DLT after the 1st cycle in 6 pts. The following hematologic grade 3/4 toxicities were noted in the 40 mg dose cohort (N=7): leukopenia (2 pts, 29%), neutropenia (3 pts, 43%), and thrombocytopenia (3 pts, 43%). Non-hematologic ADRs included fatigue, nausea, diarrhea, decreased appetite, erythema and pyrexia. The preliminary pharmacokinetic (PK) results from the 3 patients in the 30 mg cohort, and 7 patients in the 40 mg dose cohort show inter-patient variability as expected of an oral agent. Mean half-life (t ½ ) was between16.5 and 20 hours (h) with a Tmax between 2.5 and 3.5h and consistent with previous findings. Mean Cmax and AUC increased with dose (30 mg: 210 ng/mL; 3660 h*ng/mL and 40 mg: 590 ng/mL; 7200 h*ng/mL). The patient with neutropenia as DLT presented with the highest exposure. Cardiovascular assessments including serial ECGs and troponin assessments did not reveal clinically relevant findings. Best overall response was noted in 40 mg BIW cohort (N=7): 1 CR (10%), 5 PR (30%), 1 SD (20%). Four of the partial responders were ATL patients. In the 30 mg BIW dose cohort, 4/6 patients had stable disease after the 1st cycle. Summary: In this phase l trial evaluating the safety of twice weekly 30 mg and 40 mg doses, HBI-8000 was well tolerated with expected toxicities that could be managed with dose interruptions/reductions. Tumor response results in pts who completed at least one cycle of treatment indicate some clinical benefit especially in pts who started with the 40 mg dose level. The DMC/SMC has provided an opinion that the 2 observed DLTs with HBI-8000 in the phase I trial were clinically manageable and that 40 mg BIW would be recommended as the dosage for subsequent phase II studies. Registration enabling phase II trials to evaluate efficacy and safety in R/R ATL pts (Japan) and R/R PTCL pts (Japan and Korea) are being initiated. Disclosures Ando: SymBio Pharmaceuticals: Research Funding. Yoshimitsu:HUYA Bioscience International: Research Funding. Ishida:Kyowa Hakko Kirin, Co., Ltd.: Honoraria, Research Funding; Celgene KK: Research Funding; Bayer Pharma AG: Research Funding. Hidaka:Chugai-pharm: Research Funding. Nagashima:HUYA Bioscience International: Employment. Miyazato:HUYA Bioscience International: Employment. Schupp:HUYA Bioscience International: Employment. Rolland:HUYA Bioscience International: Employment. Gillings:HUYA Bioscience International: Employment. Lee:HUYA Bioscience International: Employment. Tobinai:Eisai: Honoraria, Research Funding; GlaxoSmithKline: Research Funding; HUYA Bioscience: Honoraria; Janssen Pharmaceuticals: Honoraria, Research Funding; Kyowa Hakko Kirin: Research Funding; Mundipharma: Honoraria, Research Funding; Ono Pharmaceuticals: Research Funding; Servier: Research Funding; Takeda: Honoraria, Research Funding; Zenyaku Kogyo: Honoraria; Chugai Pharma: Research Funding; Celgene: Research Funding; Abbvie: Research Funding.
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 5347-5347
    Abstract: Chemotherapy with Rituximab is widely used to treat patients with various B-cell lymphomas and auto-transplantation with Rituximab is promising strategy due to the potential for in vivo purging. However, the possibility of late onset neutropenia and immunoglobulin suppression after auto-transplantation with Rituximab has been indicated. We studied the frequency and degree of these phenomena. We performed a retrospective analysis on 26 consecutive patients at three centers during the period of January 1998 to March 2005. Thirteen patients (Follicular 8, Marzinal zone B cell lymphoma 2, Diffuse large B 3) received auto-transplantation without Rituximab (R-) compared with 13 patients (Follicular 8, MZBCL 2, DLB 3) received auto-transplantation with Rituximab (R+). In R+ patients pripheral blood stem cells were harvested after High dose AraC followed by three times of Rituximab (375mg/m2 day -2 of AraC, day7, 14). Conditioning regimen consisted of MCEC (MCNU, CBDCA, ETOP, Cy) or TBI+Cy followed by three times of Rituximab (375mg/m2 day 0, 7, 14). Mean immunoglobulin concentration one month after transplantation was 890 mg/dl for R- vs. 470 mg/dl for R+ (P=0.04). Lowest neutrophil numbers over 4 weeks after transplantation was 1.24X109/L for R- vs. 0.36X109/L for R+ (p=0.02). Late onset neutropenia ( & lt;0.5X109/L) were seen in three cases of R+ group, but no case in R- group. Therapy related death was seen one case in R+ group. This case showed low immunoglobulin level after transplantation and died of Pneumocystis Carinii. These data, although preliminary, indicates that the addition of Rituximab to auto-transplantation leads to decrease in immmunoglobulin and neutrophil levels after transplantation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    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. 4580-4580
    Abstract: Abstract 4580 Many reports were seen about reactivation of human herpes virus 6 (HHV6) after stem cell transplantation (SCT) in adult patients, and this reactivation sometimes induce severe condition of patients. However, few reports were seen about pediatric patients. Therefore, we examined HHV 6 reactivation after stem cell transplantation in patients with children, retrospectively. The cases were 80 patients, 48 male, 32 female, and the median age was 6 years old (range 0–20 years old). Transplantations were 23 related bone marrow transplantations (BMT) or peripheral blood stem cell transplantations (PBSCT), 18 unrelated BMT, one related cord blood transplantation (CBT), 31 unrelated CBT, and seven autologous BMT or PBSCT. We analyzed HHV6 DNA samples of serum with these patients before SCT, 20 days and 40 days after SCT using PCR method. In addition, we analyzed relationship between HHV6 reactivation and syndrome of inappropriate antidiuretic hormone secretion (SIADH). In samples of 20 days after SCT, 35.0% of samples were positive for HHV6 DNA. On the other hand, 2.5% and 5.0% were positive before SCT and 40 days after SCT, respectively. From 24 out of 28 samples, over 10E3 of HHV6 DNA were detected in positive samples. Factors associated with HHV6 reactivation were CBT, unrelated donor, malignant diseases, use of total body irradiation as conditioning, cyclosporine and methyl prednisolone as GVHD prophylaxis, acute GVHD ( 〉 grade 2), chronic GVHD and use of steroid using univariate analysis. Moreover, CBT was an only risk factor of HHV6 reactivation using multivariate analysis. In 14 patients with SIADH, 78.6% of patients had HHV6 reactivation. On the other hand, 25.8% of patients had HHV6 reactivation in 66 patients without SIADH. This result was statistically significant (p 〈 0.001). From this analysis, we can understand HHV6 reactivation was seen in many patients with children. In addition, we thought about the possibility of SIADH is one symptom of the encephalopathy by the HHV6 reactivation. 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. 128, No. 22 ( 2016-12-02), p. 4408-4408
    Abstract: Background: Several studies have demonstrated that aberrant expression of microRNAs in multiple myeloma (MM) cells is associated with the pathogenesis and development of MM. Recently, circulating serum microRNAs have been recognized as novel biomarkers in tumor biology and have predictive value in determining the efficacy of various drugs. However, little is known regarding the role of circulating serum microRNAs in patients with MM in terms of MM biology and the clinical efficacy of anti-MM drugs. In this study, we evaluated the expression levels of serum microRNAs in patients with MM, including newly diagnosed (ND) and relapsed and/or refractory (RR) cases. We also evaluated the correlation of the expression levels of serum microRNAs with the clinical efficacy of bortezomib (BTZ)-containing treatment. Materials & Methods: Fifteen serum samples from healthy donors and 62 from 10 patients with NDMM and 52 patients with RRMM were collected and subjected to comprehensive microRNA analysis using next-generation sequencing (NGS). First, we compared the microRNA expression levels between healthy donors and patients with MM. Next, using 52 serum samples collected from patients with NDMM and RRMM who received BTZ plus low-dose dexamethasone (Bd) therapy, the correlation between the response to Bd therapy and specific serum microRNA expression profiles was determined. Results: Approximately 150-250 microRNAs were detected by small RNA analysis of serum samples using NGS. The expression levels of 32 serum microRNAs were higher in MM than in healthy donors (Mann-Whitney U test, P 〈 0.05). Among them, 5 microRNAs (mir-10a, 10b, 92a, 378a, and 378d) had higher expression in RRMM than in NDMM. These microRNAs are involved in the biology and oncogenesis of several solid tumors, including MM. The mir-92a expression level has been associated with the response to chemotherapy and disease progression in MM. Regarding the correlation between microRNA expression levels and the clinical efficacy of Bd therapy, expression levels of 14 microRNAs were associated with progression-free survival (PFS) in Bd therapy (Spearmanfs rho 〈 -0.2, P 〈 0.05). Among them, 5 microRNAs (mir-22, 146a, 193b, 584, and 1307) showed high correlation with PFS (Spearmanfs rho 〈 -0.4, P 〈 0.002). These microRNAs are involved in angiogenesis, proliferation, and apoptosis in several solid tumors and MM. Next, we divided the 52 samples into two groups according to PFS: short ( 〈 6 months; n = 27) or long (≥6 months; n = 25). The short-PFS group showed lower expression of 5 microRNAs (mir-22, 146a, 193b, 320b, and 320c) than the long-PFS group did (Mann-Whitney U test, P 〈 0.01). Among them, mir-146a can regulate TRAF6, NF-kB, and TNF-axis, and is regulated by the c-Myc at the transcriptional level. c-Myc-mediated mir-146a overexpression can reduce CXCR4 expression. Several studies suggest that CXCR4 expression is an important factor for MM cells to migrate and interact with stromal cells; lower expression is recognized as a poor prognostic factor in the survival of patients with MM. Therefore, we hypothesized that BTZ-insensitive clone has high mir-146a expression along with low CXCR4 expression, suggesting that low dependence on stromal cells may contribute to the resistance to BTZ activity. Conclusion: We have demonstrated that the expression levels of several serum microRNAs are associated with the progression of MM and may serve as predictive markers in BTZ-containing therapies in MM. Further validation studies in a larger number of patients is needed and the origin of these serum microRNAs, together with the functional consequences of aberrant expression, must be pursued. Our findings can contribute in developing circulating microRNA analysis as a potential strategy in determining useful biomarkers for diagnosis and therapeutic outcomes in MM. Figure Figure. Disclosures Ishida: Celgene KK: Research Funding; Kyowa Hakko Kirin, Co., Ltd.: Honoraria, Research Funding; Bayer Pharma AG: Research Funding. Iida:Celgene: Honoraria, Research Funding; Janssen Pharmaceuticals: Honoraria, Research Funding.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 711-711
    Abstract: Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative therapy for patients with myelodysplastic syndromes (MDS), whose benefit, however, is frequently offset by accompanying mortality and morbidity, underscoring the importance of accurate prognostication before the therapeutic choice. For this purpose, several systems, such as the International Prognostic Scoring System (IPSS), are being successfully applied to clinics, and recent genome profiling studies indicate that molecular diagnostics can further improve the prediction. Nevertheless, existing systems are based on the observation from those patients who were untreated or only supportively treated and therefore, may not successfully be applied to the prognostication of the patients who are actually treated by HSCT. Methods: We analyzed patients with MDS (N = 719) from a cohort of Japan Marrow Donor Program (JMDP) who were treated by unrelated HSCT between 2006 and 2013. Peripheral blood DNA was subjected to targeted deep sequencing in 68 major driver genes for the detection of both somatic mutations and copy number variations (CNVs) with accurate determination of their allelic burdens. Results: The median age at HSCT and observation period were 53 years old (20-66) and 372 days (2-3001), respectively. At the diagnosis, 63, 203, 163 and 65 patients have low, intermediate-1, intermediate-2 and high risk classified on the basis of IPSS, respectively (IPSS data was not available for 250 patients). The median time from diagnosis to HSCT was 274 (9-10900) days. Mutations were observed in 75% of the patients, of which TP53 was most frequently mutated (14.3%), followed by U2AF1 (13.2%), RUNX1 (12.2%), ASXL1 (11.0%) and DNMT3A (9.3%). The mean number of mutations was 2.1 per patient and the mean allelic burden was 23.4%. To evaluate karyotyping we combined metaphase cytogenetics and copy number variations using targeted sequencing data. Complex karyotype, chromosome 7 anomaly, deletion 5q, and deletion 20q were observed in 174 (24.4%), 173 (24.3%), 91 (12.8%), and 50 (7.0%) of the patients, respectively. Combined, 86.6% of the patients had one or more genetic lesions. Patients with one or more mutations or CNVs showed unfavorable overall survival (Hazard Ratio (HR) 2.46, P = 2.12 x 10-5). Univariate analysis for each gene identified mutations in TP53 (HR 2.85, P 〈 2.0 x 10-16), NRAS (HR 1.90, P = 5.4 x 10-4), ETV6 (HR 1.54, P = 0.029), CBL (HR 2.25, P = 5.3 x10-5), EZH2 (HR 1.74, P = 0.014), KRAS (HR 2.01, P = 2.0 x 10-3), U2AF2 (HR 1.97, P = 0.027), JARID2 (HR 2.09, P = 0.039), and RIT1 (HR 2.16, P = 0.023) as the unfavorable factors for the overall survival. Besides, mutations in PRPF8 had a favorable effect on overall survival (HR 0.50, P = 0.029). Then, we performed multivariate analysis with stepwise model selection of these significant mutations and clinical parameters. Mutations in TP53 (HR 2.31, P=0.015), and ETV6 (HR 2.57, P=0.015) remained significant together with complex karyotype (HR 2.15, P = 0.0063), grade of acute graft versus host disease (GVHD) (Grade I or II: HR 1.95, P = 0.011, Grade III or IV: HR 4.18, P = 7.94 x 10-5), and the number of red blood cell transfusion received before HSCT ( 〉 =10 times: HR 2.64, P = 0.027). Next, we analyzed the impact of mutations on relapse in cases who achieved complete response after HSCT (N = 423 (58.8%)). Patients with mutations in one or more genetic lesions showed unfavorable relapse free survival (HR 2.27, P = 1.65 x 10-4). Univariate analysis for each gene revealed mutations in TP53 (HR 3.09, P = 9.5 x 10-16), NRAS (HR 2.21, P = 0.0019), ETV6 (HR 1.90, P = 0.012), PRPF8 (HR 0.40, P = 0.046), and WT1 (HR 2.24, P = 0.013) were significant for the relapse free survival. Multivariate analysis and stepwise model selection identified ETV6 (HR 2.98, P = 0.011), WT1 (HR 4.01, P = 0.014), complex karyotype (HR 2.39, P = 0.0083), IPSS High (HR 6.22, P = 0.0053), and Grade III or IV acute GVHD (HR 2.91, P = 0.0071) as unfavorable factors. Conclusions: This large study of MDS cases treated by unrelated HSCT demonstrated that somatic mutations of several driver genes were novel prognostic factors for overall and relapse free survival. These genetic factors were independent of well-known prognostic makers, and therefore could be used to better guide therapy for MDS patients. 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: 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: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4112-4112
    Abstract: Introduction Primary central nervous system lymphoma (PCNSL) is a rare subtype of non-Hodgkin's lymphoma. Although most cases (~95%) show histology of diffuse large B-cell lymphomas (DLBCLs), PCNSL shows very different biological and clinical characteristics from systemic DLBCL. Nevertheless, our knowledge about the molecular pathogenesis of PCNSL and genetic differences between both lymphomas are still incomplete. Method To obtain a comprehensive view of the genetic alterations, including mutations in non-coding regions as well as structural variants (SVs), we performed whole-genome sequencing (WGS) of 22 PCNSL cases. Subsequently, to unravel the genetic differences between PCNSL and systemic DLBCL, we re-analyzed WGS data from systemic DLBCL cases (N = 47) generated by the Cancer Genome Atlas Network (TCGA) and Cancer Genome Characterization Initiative (CGCI) using our in-house pipeline. The mean depth of WGS for tumor samples were 49X and 37X for PCNSL and DLBCL cases, respectively. Whole-exome sequencing (WES) was also performed for an additional 37 PCNSL cases to reliably capture driver alterations and also to analyze mutational signatures in PCNSL, which were compared to those obtained from the WES data for DLBCL from TCGA (N = 49). Results WGS identified 10.5 and 5.6 mutations per mega-base on average in PCNSL and DLBCL, respectively. We first explored the density of somatic mutations and identified 64 and 33 genomic loci showing significantly high mutation densities in PCNSL and DLBCL, respectively. In PCNSL, most of these loci corresponded to known targets of somatic hypermutations (SHMs) induced by activation-induced cytidine deaminase (AID), including those for IG genes (IGK, IGH and IGL), BCL6, and PIM1, as well as those for known driver genes, such as MYD88 and CD79B. Although most of the hypermutated regions were overlapped between PCNSL and DLBCL, some regions were differentially affected by hypermutations between both lymphoma types. For example, BCL2 and SGK1 loci were frequently affected by SHMs in germinal center B-cell (GCB) DLBCL, while not in PCNSL. In terms of non-coding driver mutations, we identified frequent mutations in a PAX5 enhancer region in 8/22 (36%) of PCNSL and 18/47 (38%) of DLBCL cases. SVs were common in both lymphoma types, where 104 (PCNSL) and 57 (DLBCL) SVs were detected per sample. SV clusters were identified in 34 (PCNSL) and 13 (DLBCL) regions, of which several clusters were commonly seen in both PCNSL and DLBCL, and included IG loci, BCL6, FHIT, TOX and CDKN2A. In PCNSL, SVs were clustered within the loci for known targets of SHMs, such as BCL6, BTG2 and PIM1. As was the case with somatic mutations, the SV cluster corresponding to BCL2 was only seen in DLBCL. We then analyzed these clustered breakpoints for their proximity to known sequence motifs targeted by AID (CpG and WGCW). Breakpoints of SVs found in the targets of SHMs, including PIM1, BCL6, BTG2 and BCL2, showed an enrichment at or near the CpG, supporting the involvement of AID in the generation of these SVs. By analyzing these SV clusters, we identified several novel driver genes in PCNSL. For example, WGS and WES identified an enrichment of breakpoints of deletions (7/22) and loss-of-function mutations (6/37) in GRB2, strongly indicating its tumor suppressor role in PCNSL. We also analyzed pentanucleotide signatures of mutations in coding sequences detected by WES of PCNSL and DLBCL, taking into consideration the two adjacent bases 3' and 5' of the substitutions as well as transcription strand biases. Two predominant mutational signatures were identified in PCNSL: the AID signature characterized by C 〉 T mutations within the WRCY motif targeted by SHMs and the age-related signature involving C 〉 T transition at CpG dinucleotides. For DLBCL, an additional signature (signature 17 according to Alexandrov et al.) was detected as well, which had been reported in DLBCL with an unknown mechanistic basis. Conclusions Comprehensive genomic analyses of a large cohort of PCNSL and DLBCL cases have revealed the major targets of somatic mutations and SVs, including novel driver genes. In both PCNSL and systemic DLBCL, an enhanced AID activity is thought to be associated with generation of both SHMs and SVs, although the activity and targets of AID seem to substantially differ between both lymphoma types, suggesting distinct pathogenesis therein. Disclosures Kataoka: Boehringer Ingelheim: Honoraria; Yakult: Honoraria; Kyowa Hakko Kirin: Honoraria. Ogawa:Takeda Pharmaceuticals: Consultancy, Research Funding; Kan research institute: Consultancy, Research Funding; Sumitomo Dainippon Pharma: Research Funding.
    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. 129, No. 17 ( 2017-04-27), p. 2347-2358
    Abstract: TP53 and RAS-pathway mutations predict very poor survival, when seen with CK and MDS/MPNs, respectively. For patients with mutated TP53 or CK alone, long-term survival could be obtained with stem cell transplantation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2017
    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. 108, No. 11 ( 2006-11-16), p. 4525-4525
    Abstract: T-cell acute lymphoblastic leukemia (T-ALL) and T-cell lymphoblastic lymphoma (T-LBL) account for most of the childhood T-lymphoid malignancy(LM). T-ALL is usually treated by the same protocol to the B-progenitor ALL. It is obvious that biologically, T-cell behaviors are different from those of B-progenitor cell, and cell origins are same among T-ALL and T-LBL. Thus we conducted a strategy initiating T-ALL specific protocol, differing from protocols for B-progenitor ALL. Furthermore, we indicated the same protocol to advanced T-LBL. The aims of this study were to evaluate the efficacy and safety of indicating the same protocol for childhood T-ALL and advanced T-LBL, and to reveal the prognostic factors of childhood T-LM. 70 eligibleT-ALL patients enrolled in the JACLS ALL-97 study between 1997-2001, and 32 eligible patients with stage III and IV T-LBL enrolled in the JACLS NHL-T98 trial between 1998-2002 were analyzed. Median age was 9y8m (2y~15y3m) for T-ALL and 11y11m (3y~15y4m) for T-LBL. Male/female ratio was 46/24, 26/6, respectively. Mediastinal mass was found in 31/70(44.3%) for T-ALL, 19/21(90.5%) for stageIII and 6/11(54.5%) for stageIV T-LBL. The treatment for 2 years consisted of the induction therapy (VCR, HD-MTX, CA, PSL, ASP), the 5-drug consolidation therapy A and B, both including high dose of ASP, and maintenance therapy with block-rotated treatment using drugs above. Complete remission(CR) at the end of induction therapy was obtained 65/70(92.9%) for T-ALL, 15/21(71.4%) for stage III and 10/11(90.9%) for stage IV T-NHL. 5-year overall survival(OAS) rate for T-ALL, stage III and stage IV T-LBL was 81.1%, 63.9% and 81.8%, respectively. 5-year event free survival (EFS) rate was 72.9%, 47.6% and 72.3%, respectively. Relapse after CR occurred in 12/65 with T-ALL, 6/15 with stage III and 1/10 with stage IV T-LBL. Single variant analysis revealed that there were no significant difference in OAS or EFS for T-ALL patients based on WBC, NCI index, but statistical difference in OAS or EFS based on age(older than 10y worse), the existence of mediastinal mass(absence, worse) In T-LBL, there were no statistical differences based on age, existence of mediastinal mass. Multivariate analysis revealed, for T-ALL and T-LBL patients as a whole, that age 〉 10 years was a risk factor in both OAS and EFS, absence of mediastinal mass and stage III T-LBL were risk factors in OAS. Our data shows that indicating same T-cell specific protocol, for T-ALL and advanced T-LBL has a potential to improve the prognosis of T-LM. The older age, and stage III T-LBL appeared as prognostic factors. Moreover, mediastinal mass with bone marrow involvement was a favorable factor for childhood T-LM. Although some risk factors were documented, it is needed to clarify unknown prognostic factors and develop the more effective, stratified T-cell specific protocols.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 1874-1874
    Abstract: BACKGROUND: With current intensified chemotherapy, more than 70% of children with acute lymphoblastic leukemia (ALL) are curable. However, prognosis of the patients at higher risk has been unsatisfied. More appropriate risk assignment and innovative treatment is expected to be developed. We conducted a clinical trial of JACLS ALL-97 including multi-agent block therapy followed by hematopoietic stem cell transplantation (SCT) for the selected higher risk patients. PATIENTS AND METHODS: Between April 1997 and March 2002, 674 patients aged 1 to 15 years with newly diagnosed ALL (excluding mature B-cell ALL) were enrolled on the JACLS ALL-97 protocol. Excluding 75 T-ALL, 26 mixed lineage leukemia other than B-precursor ALL with myeloid markers, 9 acute unclassified leukemia and 1 B-precursor ALL with pretreatment, 563 patients with B-precursor ALL were eligible for this analysis. Treatment group was divided 5 groups according to the modified National Cancer Institute (NCI) workshop criteria. Standard risk (SR) and IR (intermediate risk) divided by WBC10 × 103 account for NCI-SR. High risk (HR) and ER (extremely high risk) divided by WBC 100 × 103 account for NCI-HR. The patients with ALL positive for Philadelphia chromosome and for translocation with chromosome11q23 were assigned to the highest risk group F. The patients in complete remission (CR) at day 35 with M2/M3 at the day 14 marrow were assigned to shift higher risk after induction therapy. Treatment of ALL-97 consists of early phase and maintenance phase. Early phase includes induction therapy, consolidation therapy, sanctuary therapy, and re-induction therapy for 19 to 26 weeks dependent on risk group. Early phase in IR/HR protocol is identical to the SR protocol except the addition of two doses of DNR and three doses of CPM. Maintenance phase contains standard MTX and MP with monthly VCR and PSL intensification for SR, and rotational therapy of a set of MTX and MP with a set of VCR, PSL, ASP, and (DNR or CPM) for IR/HR/ER/F. The IR protocol is identical to the HR protocol without cranial irradiation. Treatment duration is 24 months for any risk. The patients assigned ER and F were candidate for allogeneic stem cell transplantation by the end of early phase. Transplant procedures depended on the institute. RESULTS: Number of patients at each risk was 204 for SR, 158 for IR, 128 for HR, 36 for ER, 27 for Ph+ and 10 for 11q23. Six of them were treated with incorrect risk protocol. Thirty-four patients received SCT in first CR. Following the induction therapy, 550 of 564 patients (97.5%) achieved CR. 23 patients(8 in SR, 9 in IR, 4 in HR and 2 in ER, 4.4% of all) were shift to higher risk due to the findings of day 14 marrow. Five-year overall survival rate (OS) and event-free survival rate (EFS) for all patients was 90.6% and 77.0%, respectively. Five-year EFS for NCI-SR and HR was 81.6% and 67.6%, respectively. According to risk group, 5-year EFS for SR, IR, HR, ER, Ph+ ALL, and ALL with 11q23 were 86.6%, 77.0%, 71.9%, 68.7%, 40.7%, and 70.0%, respectively. 5-year OS for them were 94.0%, 93.9%, 92.1%, 83.8%, 55.6%, and 68.6%, respectively. CONCLUSIONS: By the risk-adapted therapy in the JACLS ALL-97 trial, high cure rate could be achieved for children with B-precursor ALL.
    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. 90, No. 11 ( 1997-12-01), p. 4363-4368
    Abstract: We recently showed that c-kit signal synergizes with glycoprotein (gp)130 signal mediated by a complex of interleukin (IL)-6 and soluble IL-6 receptor (IL-6/sIL-6R) to stimulate the expansion of human primitive hematopoietic progenitor cells and erythropoietin-independent erythropoiesis. In the present study, we examined the effect of a ligand for Flt3 (FL), whose receptor tyrosine kinase is closely related to c-kit, in combination with IL-6/sIL-6R on human hematopoiesis in vitro. In serum-containing methylcellulose clonal culture of cord blood CD34+ cells, whereas FL alone stimulated only granulocyte-macrophage (GM) colony formation, erythroid bursts and mixed colonies in addition to GM colonies were induced by FL with IL-6/sIL-6R, but not IL-6/sIL-6R alone. In suspension culture, CD34+ cells generated a small number of myeloid cells in the presence of FL or IL-6/sIL-6R alone. However, the addition of IL-6/sIL-6R to the culture with FL induced the generation of a significant number of erythroid cells and megakaryocytes in addition to myeloid cells. The combination of FL and IL-6/sIL-6R also induced a remarkable expansion of GM colony- and erythroid burst-forming cells and multipotential progenitors, although FL or IL-6/sIL-6R alone induced the generation of only a small number of progenitors for GM colonies. The synergistic effects of FL and IL-6/sIL-6R were confirmed in serum-free clonal and suspension cultures. In addition, the addition of anti-human gp130 monoclonal antibodies abrogated the synergistic action. These results indicate that Flt3 signal, as well as c-kit signal, synergizes with gp130 signal to stimulate human myelopoiesis, erythropoiesis and megakaryopoiesis, and the expansion of primitive multipotential hematopoietic progenitor cells.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1997
    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...