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. 126, No. 23 ( 2015-12-03), p. 2839-2839
    Abstract: Early therapeutic decision-making is crucial in patients with higher-risk MDS where median survival is only around one year. Azacitidine prolongs survival for these patients (Fenaux et al, Lancet Oncology 2009) but clinically relevant biomarkers remain to be identified. We evaluated retrospectively, the impact of clinical parameters and mutational profiles in 134 consecutive patients treated with a median number of 7 cycles of Azacitidine (range 1-45), in accordance to European guidelines. The vast majority (n=114) had higher-risk disease i.e. MDS with IPSS int-2 or high, AML with multilinear dysplasia and 20-30% blasts or CMML-II. We combined an initial cohort from Karolinska University Hospital (n=89) with a validating cohort from King's College Hospital, London (n=45). Studied endpoints were response, as defined by the IWG criteria, and survival. Since prolonged survival is the main goal for this cohort of patients, we believe that survival is the most relevant endpoint, supported by the fact that even non-responding patients have a survival benefit from Azacitidine (Gore et al, Haematologica, 2013). While neither clinical parameters nor mutations had a significant impact on response rate, both karyotype and mutational profile were strongly associated with survival from the start of treatment, see Table 1 and Figure 1-2. IPSS high-risk cytogenetics was negatively associated with survival (median 20 vs 10 months; p 〈 0.001), whereas mutations in histone modulators (ASXL1, EZH2, MLL) were associated with prolonged survival (22 vs 12 months, p=0.001). This positive association was present in both cohorts and remained highly significant in the multivariate cox model. Importantly, patients with mutations in histone modulators lacking high-risk cytogenetics showed a survival of 29 months (response rate 73%) compared to 10 months (response rate 49%) in patients with the opposite pattern, see Figure 3. While TP53 was negatively associated with survival in the univariate analysis, neither RUNX1-mutations nor the number of mutations, previously reported as negative prognostic markers, appeared to influence survival in this cohort. In contrast, disease duration and cellularity showed a weak negative correlation with survival. We propose a model combining histone modulator mutational screening with cytogenetics in the clinical decision-making process for higher-risk MDS eligible for treatment with Azacitidine. Table 1. Variables associated with survival. Univariate analyses used the log-rank test. The cox model included all listed variables except response rate in a multivariate analyses. Estimated median survival (months) Univariate p-value Cox regression p-value Hazard ratio (95% CI) Response rate: CR / mCR vs PR/HI vs SD/PD 20 vs 20 vs 10 〈 0.001 IPSS cytogenetic risk group: Favorable vs Int vs Adverse 20 vs 20 vs 10 〈 0.001 〈 0.001* 3.00 (1.9-4.7) Disease duration ≥ 4 months: Yes vs No 14 vs 17 0.44 0.05** 1.01 (1.00-1.02) Marrow blasts ≥ 11%: Yes vs No 14 vs 14 0.7 Cellularity ≥ 70%: Yes vs No 14 vs 20 0.2 0.02 1.013 (1.002-1.023)** ANC ≥ 1.3: Yes vs No 14 vs 17 0.32 Platelets ≥ 60: Yes vs No 17 vs 12 0.07 Transfusion dependent: Yes vs No 13 vs 17 0.43 Therapy related: Yes vs No 17 vs 14 0.44 Number of mutations: 0 vs 1 vs ≥ 2 17 vs 12 vs 17 0.64 Epigenetic mutation: Yes vs No 19 vs 12 0.03 DNA methylation mutation: Yes vs No 14 vs 14 0.64 Histone modulator mutation: Yes vs No 22 vs 12 0.001 0.007 0.499 (0.3-0.83) Splicing factor mutation: Yes vs No 13 vs 17 0.31 ASXL1 mutation: Yes vs No 29 vs 12 0.03 TET2 mutation: Yes vs No 13 vs 16 0.45 EZH2 mutation: Yes vs No 20 vs 14 0.37 SF3B1 mutation: Yes vs No 13 vs 16 0.35 RUNX1 mutation: Yes vs No 17 vs 14 0.76 SRSF2 mutation: Yes vs No 20 vs 14 0.5 TP53 mutation: Yes vs No 9 vs 17 〈 0.001 *Comparing adverse cytogenetics vs the other groups. ** Disease duration, marrow blasts, cellularity, ANC and TPK were analyzed as a continuous variable in the cox model Figure 1. Kaplan-Meier estimated survival stratified for response and pre-treatment parameters Figure 1. Kaplan-Meier estimated survival stratified for response and pre-treatment parameters Figure 2. Forest plot indicating hazard ratio including confidence interval for all pre-treatment variables. The hazard ratios were retrieved using cox univariate regression models for each variable analyzed separately. Figure 2. Forest plot indicating hazard ratio including confidence interval for all pre-treatment variables. The hazard ratios were retrieved using cox univariate regression models for each variable analyzed separately. Figure 3. Kaplan-Meier estimated survival stratified for the two dominant predictors in the cox regression model: Adverse cytogenetics and histone modulator mutations Figure 3. Kaplan-Meier estimated survival stratified for the two dominant predictors in the cox regression model: Adverse cytogenetics and histone modulator mutations Disclosures McLornan: Novartis: Research Funding, Speakers Bureau. Jädersten:Celgene: Other: speakers fee. Kulasekararaj:Alexion: Consultancy. Mufti:Celgene: Consultancy, Other: Speakers fee. Hellström-Lindberg:Celgene Corporation: Research Funding.
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 92, No. 1 ( 1998-07-01), p. 68-75
    Abstract: Treatment with erythropoietin (epo) may improve the anemia of myelodysplastic syndromes (MDS) in approximately 20% of patients. Previous studies have suggested that treatment with the combination of granulocyte colony-stimulating factor (G-CSF) and epo may increase this response rate. In the present phase II study, patients with MDS and anemia were randomized to treatment with G-CSF + epo according to one of two alternatives; arm A starting with G-CSF for 4 weeks followed by the combination for 12 weeks, and arm B starting with epo for 8 weeks followed by the combination for 10 weeks. Fifty evaluable patients (10 refractory anemia [RA], 13 refractory anemia with ring sideroblasts [RARS] , and 27 refractory anemia with excess blasts [RAEB]) were included in the study, three were evaluable only for epo as monotherapy and 47 for the combined treatment. The overall response rate to G-CSF + epo was 38%, which is identical to that in our previous study. The response rates for patients with RA, RARS, and RAEB were 20%, 46%, and 37%, respectively. Response rates were identical in the two treatment groups indicating that an initial treatment with G-CSF was not neccessary for a response to the combination. Nine patients in arm B showed a response to the combined treatment, but only three of these responded to epo alone. This suggests a synergistic effect in vivo by G-CSF + epo. A long-term follow-up was made on 71 evaluable patients from both the present and the preceding Scandinavian study on G-CSF + epo. Median survival was 26 months, and the overall risk of leukemic transformation during a median follow-up of 43 months was 28%. Twenty patients entered long-term maintenance treatment and showed a median duration of response of 24 months.The international prognostic scoring system (IPSS) was effective to predict survival, leukemic transformation, and to a lesser extent, duration of response, but had no impact on primary response rates.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1998
    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. 140, No. Supplement 1 ( 2022-11-15), p. 9785-9788
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    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. 112, No. 11 ( 2008-11-16), p. 223-223
    Abstract: Around 50% of patients with high-risk MDS or MDS-AML may enter CR after induction chemotherapy, but CR duration, as well as overall survival is usually short. To address this clinical problem the Nordic MDS Group designed a prospective multicenter phase II study, which assessed the clinical feasibility and utility of long-term maintenance treatment with azaciditine. Sixty patients with high-risk MDS (IPSS intermediate-2 or high) (n=23) or AML following a previous known MDS (n=37) were enrolled between 2004 and 2006. The mean age was 68 (54–83) and patients should not be eligible for stem cell transplantation. Induction treatment consisted of standard doses of daunorubicin and ara-C. Patients in CR received low dose azacitidine subcutaneously 5/28 days until relapse, unless unacceptable toxicity developed. Methylation status of the P15ink4b (P15), E-cadherine (CDH) and Hypermethylated in Cancer 1 (HIC) gene was analysed at study start, in CR and in some patients during follow up. Last follow up was on August 1 2008, 24 months after the last CR was reported. Twenty-four patients (40%) reached CR and 23 of these started maintenance treatment with azacitidine. The initial dose of azacitidine was 75 mg/m2 but as four of the first five enrolled patients developed grade 4 cytopenia, the starting dose was lowered to 60 mg/m2, and was allowed to be reduced to 45 or 30 mg/m2 to avoid severe cytopenias. The mean dose of azacitidine was 54.3 mg/m2. Azacitidine was well tolerated. In 52% of the cases no side effects at all were reported. The most commonly reported side effect was mild rashes at the injection site (35%). Twenty-two percent developed fever or some kind of infection, mostly mild. Myelosuppression (grade 1–3) was seen in 22% of the cases. As previously reported, the probability of reaching CR was negatively correlated to promoter hypermethylation of CDH (p=0.008) and none of the 6 patients hypermethylated on all 3 genes reached CR (p=0.03) and hence only four patients hypermethylated on other genes than P15 received demethylating therapy. The median CR duration for the azacididine treated group was 13.5 months (2–49+) and median survival time from time of inclusion in the study for the same group was 20 months (4–52+). Four of 23 patients (17%) had a CR exceeding 24 months (32–52+). The two patients hypermethylated on CDH pre-induction had CR durations of only 2 and 5 months respectively. By last follow up 3 patients were still in CR. Of 10 patients without any methylation pre-treatment, all but one maintained this pattern in CR. Of the nine patients with pre treatment methylation of at least one gene, only one remained hypermethylated in CR. This patient had a CR duration of only 5 months. One patient showed development of P15 hypermethylation in the bone marrow sampled at 12 months and relapsed at 15 months. These findings support previous reports on P15 hypermethylation as a marker for minimal residual disease (MRD) and threatening relapse. In the whole group, survival was significantly shorter in patients with CDH methylation (3 vs 9 months, p=0.005), while pre-treatment p15 methylation status did not affect CR duration or overall survival. In conclusion, we show for the first time that maintenance treatment with azacytidine is feasible and associated with a median CR duration of 13.5 months, and very mild side effects. However azacytidine does not seem to prevent relapse in the majority of patients, including those with hypermethylation pre-treatment and/or in CR. Hypermethylation of multiple genes is a strong negative factor for survival, probability of CR, and CR duration. We observe a subset of patients, 17%, with a CR duration of & gt;24 months; but no persistent pattern regarding cytogenetics, methylation or morphology could be identified in this group. The strong negative impact of E-Cadherin methylation, a gene involved in adhesion, warrants further investigation.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 818-818
    Abstract: Patients with high-risk myelodysplastic syndromes (MDS) or acute myeloid leukaemia (AML) secondary to MDS may reach complete remission (CR) following intensive chemotherapy but the CR-duration is usually very short. Promoter hypermethylation of e.g. tumour suppressor genes is considered to be important in cancerogenesis and a negative risk factor for survival in patients with MDS. We designed a prospective clinical trial to assess the impact of methylation status on the outcome of induction chemotherapy and to evaluate the effect of maintenance treatment with 5-azacytidine (aza) on CR duration. Sixty patients with a median age of 68 years (54–83) with high risk MDS (n=23) or AML (n=37) following MDS were treated with standard doses of daunorubicin and ara-C. Standard prognostic variables, and methylation status of the P15ink4b (P15), E-cadherin (CDH) and Hypermethylated in Cancer 1 (HIC) genes were analyzed before treatment. Patients in CR were given low dose aza subcutaneously for 5 days/4 weeks until relapse. The CR rate was 40%; significantly lower in patients with high white blood cell counts (P=0.03) and high CD34 expression on bone marrow cells (P=0.02). While P15 status alone was not associated with CR rate (P=0.25) patients with CDH methylation showed a lower CR rate (P=0.008). Moreover, no patient with hypermethylation of all three genes achieved CR (P=0.03). CDH methylation retained its prognostic value in the multivariate analysis. Hypermethylation was associated with increased CD34 expression but not with blast count or cytogenetic risk group. Median duration of CR was 13 months (2 to +37) and 7/ 23 patients (30%) had a CR duration 〉 20 months. Three of four patients with trisomy 8 had CR 〉 20 months. The overall survival of patients reaching CR was 17 months (6 to +40). Methylation status before treatment did not correlate with CR duration. We demonstrate for the first time a significant impact of methylation status on the outcome of conventional chemotherapy in high-risk MDS and MDS-AML and propose that this variable should be further evaluated in prospective studies. CR duration is promising, with a substantial portion of patients showing durable responses.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    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. 116, No. 21 ( 2010-11-19), p. 2491-2491
    Abstract: Abstract 2491 While much has been discovered in the laboratory regarding the mechanisms of multidrug resistance (MDR), based on phase III clinical trials of ATP-binding cassette (ABC) transporter modulators, these findings have not resulted in appreciable clinical benefit in acute myeloid leukemia (AML) patients. In this pilot study, we investigated the clinical relevance of 380 genes related to MDR in 31 samples from AML patients who had undergone conventional treatment, with 11 patients contributing samples at diagnosis and relapse, using TaqMan Low Density Arrays (TLDA). We and others have shown that the TaqMan-based qRT-PCR is superior to microarrays and SYBR-green-based qRT-PCR because of higher sensitivity and specificity in determining expression of highly homologous gene family members, such as the ABC transporters. 380 genes involved in metabolism, signal transduction, DNA repair, stress response, tumor suppressor activity, oncogenic transformation, apoptosis, and drug uptake or efflux were curated based on their potential role in MDR in the current literature. We found expression of four genes previously unreported in AML in both diagnostic and relapsed patient samples to correlate with duration of complete remission (CR) of induction chemotherapy: SLC7A8 and HSPE1 correlate negatively while GBP1 and ABCD2 correlate positively with CR duration ( 〉 2-fold change relative to the median, p 〈 0.05). SLC7A8 is an L-type amino acid transporter that has not previously been found to be expressed in tumor cells, while GBP1 is a guanylate binding protein that might have antiproliferative and antiangiogenic effects. HSPE1 and ABCD2 are poorly characterized members of heat shock protein and ABC transporter families, respectively. NFKB1A, involved in the NFKB pathway, was found to correlate positively with CR duration in both samples at diagnosis and relapse ( 〉 2-fold change relative to the median, p 〈 0.05). In a grouped comparison of all samples at relapse relative to diagnosis, GSTM5, a glutathione-S-transferase isoform involved in drug metabolism, was upregulated while TNF, tumor necrosis factor, was downregulated ( 〉 2 fold change compared to diagnosis, p 〈 0.05). ABCB1, the best studied ABC transporter for which many modulators have been developed, was upregulated in some patients, while downregulated in others at relapse ( 〉 2 fold change compared to diagnosis, p 〈 0.05). A patient-by-patient analysis of the paired samples revealed that each had a unique set of genes that was upregulated or downregulated at relapse relative to diagnosis, confirming the heterogeneity of AML with respect to acquired MDR. Finally, we assessed the ability of 15 cell lines to accurately depict MDR-linked gene expression in patients. The gene expression patterns of all cell lines were quite different from the patient samples, signifying the need to develop appropriate preclinical models for the study of MDR in AML. 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: 2010
    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, ( 2023-04-05)
    Abstract: Although tyrosine kinase inhibitors are effective for treating chronic myeloid leukemia (CML), they often fail to eradicate the leukemia-initiating stem cells (LSCs), causing disease persistence and relapse. Evidence indicates that LSC persistence may be due to bone marrow (BM) niche protection. However, little is known about the underlying mechanisms. We here molecularly and functionally characterized BM niches in CML patients at diagnosis and revealed the altered niche composition and function in the CML patients. Long-term culture initiating cell (LTC-IC) assay showed that the mesenchymal stem cells from CML patients displayed an enhanced supporting capacity for normal and CML BM CD34+CD38- cells. Molecularly, RNA sequencing detected dysregulated cytokine and growth factor expression in CML patient BM cellular niches. Among them, CXCL14 was lost in the BM cellular niches in contrast to its expression in healthy BM. Restoring CXCL14 significantly inhibited CML LSC maintenance and enhanced their response to imatinib in vitro, and CML engraftment in vivo in NSG-SGM3 mice. Importantly, CXCL14 treatment dramatically inhibited CML engraftment in xenografted NSG-SGM3 mice, even to a greater degree than imatinib, and this inhibition persisted in patients with suboptimal TKI response. Mechanistically, CXCL14 upregulated inflammatory cytokine signaling but downregulated mTOR signaling and oxidative phosphorylation in CML LSCs. Together, we have discovered a suppressive role of CXCL14 in CML LSC growth. CXCL14 might offer a treatment option targeting CML LSCs.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2023
    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. 2619-2619
    Abstract: In this study we aimed to identify biomarkers predictive of clinical response in acute myeloid leukemia (AML). For this purpose mRNA was isolated from diagnostic samples from 42 AML patients (“training cohort”) and subjected to Affymetrix® gene expression analysis. All patients entered complete remission (CR) after high-dose induction chemotherapy, reaching a median CR duration of 161 (range 12-3701) days. Samples from patients with “short CR duration” ( 〈 6 months, n=24) and “long CR duration” ( 〉 6 months n=18), respectively, were pooled and compared. Gene expression analyses revealed 383 genes to be up-regulated and 610 genes down-regulated more than two fold in samples from patients with short, as compared to those with long CR duration. Ten genes were found to be up-regulated 〉 30 times, with the runt-related transcription factor 1; translocated to 1 (cyclin D-related) (RUNX1T1) gene showing the highest differential expression (116-fold), while annexin 1 (ANXA1) was the most down-regulated gene (58-fold). Significantly higher transcript levels of RUNX1T1 were confirmed in the poor outcome group when performing quantitative real time polymerase chain reaction (qRT-PCR) on individual samples (n=20, p 〈 0.002). Subjecting our data to pathway analysis (Ingenuity®) comparing the same groups, we focused on RUNX1T1 and created a network of RUNX1T1 interacting molecules. Utilizing the IPA database to create a network over interacting molecules of RUNX1T1, we identified a majority of these to be transcriptional regulators and among them the transcription factor 3 (TCF3) to be up-regulated 5-fold in patients with short CR duration. Our training cohort data were validated in silico extracting information from an independent study by Metzeler et al, publicly available from Oncomine® (www.oncomine.org) and encompassing diagnostic samples from 162 AML patients. Among genes differentially and similarly regulated in poor responders in both the training and validation cohorts we observed TCF3, chemokine (C-X-C motif) ligand 3 (CXCL3), Zinc finger, MIZ-type containing 1 (ZMIZ1) (up-regulated) and Peroxiredoxin 2 (PRDX2) (down-regulated). Analyses of clinical outcome revealed that AML patients with a high ZMIZ1 expression had a significantly decreased overall survival (OS) as compared to that of patients with a low ZMIZ1 expression (p 〈 0.03). ZMIZ1 has been reported to be involved in tumor growth in general and suggested to interact with activated NOTCH in inducing leukemia, but its more precise role in AML is still unclear. In conclusion, we report clear differences in gene expression in diagnostic samples from AML patients with subsequent poor vs. better long-term clinical outcome to therapy, thus to constitute possible novel predictive biomarkers for response. In our training set RUNX1T1 was the most differentially expressed gene, while ZMIZ1 was upregulated in both the training and validation sets and significantly associated with survival. Further, functional studies of differentially expressed genes in clinical subsets of AML patients appear warranted. 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: 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. 126, No. 23 ( 2015-12-03), p. 710-710
    Abstract: Background: Myelodysplastic Syndrome (MDS) and Chronic Myelomonocytic Leukaemia (CMML) are haematological disorders that develop in haematopoietic stem or progenitor cells (HSPCs) and are characterised by ineffective haematopoiesis. 5'-Azacitidine (AZA), a DNA demethylating agent, is the primary drug for the treatment of high-risk MDS and CMML and response is associated with improved survival benefits. However, only half of treated patients will ever respond to AZA and the molecular basis for poor response is currently unknown. There are few alternative therapies for the non-responders. Additionally, AZA response is rarely sustained and a substantial fraction of responders will eventually relapse. The in vivo effect of AZA therapy on dysplastic cells in responders is unclear and there are no predictive markers for impending relapse in responders. Methods: To address these fundamental questions, we enrolled 18 high-risk MDS and CMML patients on a compassionate access program for AZA in Australia. Bone marrow was collected at seven different points - before treatment; through 6 cycles of treatment; and at up to two years after initiation - and we isolated high-purity CD34+ HSPCs (Figure A). 10 patients had a complete response while 8 were poorer responders. We performed RNA-seq to query the transcriptomes (and validated by Fluidigm-based PCR) and deduced the clonal evolution in the bone marrow in response to AZA therapy (by whole exome-sequencing, followed by targeted capture resequencing, and genotyping of individual CFU colonies). Our findings were validated in an independent cohort of 57 patients. We used flow cytometry to develop a clinically relevant prognostic assay for AZA resistance and developed a novel stromal co-culture based functional drug testing platform to rationally discover combinational drug therapies to overcome AZA resistance. Results: We hypothesised that primary AZA resistance would be driven by pre-existing molecular differences between responders and non-responders. Analysis of the pre-treatment RNA-seq data strikingly revealed the differential expression of 1148 genes between responders and non-responders (Figure B). Pathway analyses of these genes indicated that cell cycle and DNA damage response pathways were relatively up-regulated in responders compared to non-responders, indicating that HSPCs of non-responders are more quiescent compared to responders (Figure C). We validated these gene expression differences in independent patient cohorts from the U.K. and Sweden (n=57; 27 responders, 30 non-responders). We then adapted a flow cytometry based assay, amenable to prospective use in a clinical diagnostic setting, to directly detect the increased quiescence of CD34+ CD38+ haematopoietic progenitors in unsorted bone marrows of non-responders across all cohorts (Figure D). Finally, to reverse the quiescence of progenitor cells of non-responders, and make them more susceptive to AZA, we leveraged our RNA-seq discoveries to target pathways that were relatively up-regulated in non-responders. Using a stromal co-culture drug testing platform that we developed, we discovered that inhibiting integrin-linked signalling combinatorially with AZA improved the functionality of dysplastic cells (Figure E). Additionally, dysplastic cells were particularly sensitive to the inhibition of the mTOR pathway. To trace the fate of dysplastic cells as patients undergo AZA therapy, and thereby understand the basis of eventual relapse in responders, we performed whole exome sequencing of all patients (Figure F). Using the mutations as "molecular barcodes", we deduced the clonal architecture in each individual and observed the clonal evolution that occurred in response to AZA treatment. Combined with genotyping of CFU colonies grown in vitro, we have discovered that clonal haematopoiesis originating from resistant multipotent cells bearing mutations persists even upon complete response and forms the basis for eventual relapse (Figure G). Conclusions: Our findings, across independent cohorts and relevant to both MDS and CMML, have immediate clinical utility not simply to prospectively identify AZA non-responders but also by suggesting combinatorial therapies that could improve response. Finally, elucidating the in vivo effects of AZA therapy lay the foundation for developing more durable treatments. Figure 1. Figure 1. Disclosures Kulasekararaj: Alexion: Consultancy. Lynch:Celgene: Employment, Equity Ownership. Campbell:14M genomics: Other: Co-founder and consultant.
    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. 4195-4195
    Abstract: The popular concept that human cancers might be driven by rare self-renewing cancer stem cells (CSCs) has extensive implications for cancer biology and modelling, as well for development of more efficient and targeted therapies. However, experimental support for the existence of distinct and rare CSCs in human malignancies remain contentious, particularly in light of compelling evidence that cancer-propagating cells frequently fail to read out in existing human stem cell assays. Therefore, to unequivocally establish the existence and identity of human CSCs, the challenge is first to identify candidate CSCs, and to establish their unique ability to self-renew and replenish molecularly and functionally distinct non-tumorigenic progeny followed by functional in situ validation within the patients themselves. Methods We have in the hematological malignancy myelodysplastic syndromes (MDS) characterize candidate hematopoietic stem and progenitor stages in the bone marrow of low-intermediate risk MDS patients by flow cytometry. Distinct cell populations were functionally characterised for lineage commitment in standard colony forming cell (CFC) assays, and for self-renewal potential in long-term culture initiating cell (LTC-IC) assays and in immune-deficient (NSG) mice. Moreover, we tracked the cellular origin of all identified somatic genetic lesions identified in each patient by targeted next-generation sequencing of genomic DNA isolated from each purified MDS stem and progenitor cell population. Results In low-intermediate risk MDS patients, regardless whether they were del(5q) (n=19) or non-del(5q) (n=11), we could identify rare but distinct Lin-CD34+CD38-CD90+CD45RA- candidate stem cells, granuclocyte-monocyte progenitors (GMPs) and megakaryocyte-erythroid progenitors (MEPs) with frequencies within total BM similar to that of normal age-matched controls. Global gene expression analysis by RNA sequencing of MDS stem cells, GMPs and MEPs suggested that these are molecularly distinct populations. Myeloid and erythroid gene expression signatures were restricted to the GMPs and MEPs, respectively, whereas a transcriptional stem cell signature was restricted to the MDS stem cells. GMPs and MEPs isolated from del(5q) (n=12) and non-del(5q) (n=8) MDS patients displayed lineage-restricted myeloid and erythroid differentiation potentials, respectively. Self-renewal in LTC-IC assay was restricted exclusively to MDS Lin-CD34+CD38-CD90+CD45RA- stem cells in del(5q) (n=11) and non-del(5q) (n=8) MDS patients. Xenotransplantation into NSG mice also confirmed that only Lin-CD34+CD38-CD90+CD45RA- MDS stem cells have in vivo self-renewal potential, and these experiments also demonstrated their ability to replenish downstream CMPs, GMPs and MEPs, establishing the hierarchical relationship of MDS stem and progenitor cells. Targeted DNA sequencing of 88 genes recurrently mutated in MDS and other myeloid malignancies was pursued to identify somatic genetic lesions within the bulk bone marrow of MDS patients (n=13). In total we identified 30 presumed genetic driver lesions, including del(5q) and mutations in key transcription factors (RUNX1), signalling pathways (JAK2, CSF3R), epigenetic regulators (TET2, ASXL1), apoptosis regulators (TP53), and spliceosome components (SF3B1, SRSF2, U2AF2, SRSF6). Importantly, in support of their unique ability to self-renew and replenish lineage-restricted MDS progenitors, all stable somatic genetic lesions identified could in each MDS patient be backtracked to the rare stem cell population as defined phenotypically by flow cytometry and functionally by LTC-IC or xenograft potential, unequivocally establishing their unique stem cell identity within the malignant clone. Conclusions These findings provide definitive evidence for the existence of rare and distinct stem cells in MDS, a finding with extensive implications for therapeutic strategies in MDS and other cancers whose existence might also strictly depend on the persistence of rare CSCs. MDS stem cells typically acquire multiple driver mutations, together conferring a competitive advantage over normal stem cells, while even in combination failing to inflict self-renewal ability on MDS myelo-erythroid progenitor cells. 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: 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...