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: International Journal Of Community Medicine And Public Health, Medip Academy, Vol. 9, No. 2 ( 2022-01-28), p. 1113-
    Abstract: Back pain has been reported as a common cause for various patients to present in an emergency or primary care settings. Besides, the management of back pain has been associated with a huge economic burden and remarkably impacts the quality of life of the affected patients. The diagnosis of acute low-back pain can be adequately achieved by conducting proper clinical evaluation and knowing the characteristics of each condition. The present review discusses the clinical evaluation and red flags for diagnosing patients presenting with acute low-back pain. An adequate examination of patients is conducted by obtaining a thorough history and successful physical examination. It should be noted that obtaining an adequate history might not be enough in some cases, and physical examination might not show any diagnostic clues. However, we also reported various red flags for detecting serious conditions, including malignancy, infections, inflammation, and others. These might help establish a further assessment of these patients, including imaging and laboratory studies. Therefore, these cases should be managed as early as possible to enhance the prognosis and intervene against any potential complications. 
    Type of Medium: Online Resource
    ISSN: 2394-6040 , 2394-6032
    Language: Unknown
    Publisher: Medip Academy
    Publication Date: 2022
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Nanomaterials, MDPI AG, Vol. 12, No. 22 ( 2022-11-16), p. 4029-
    Abstract: 6-Mercaptopurine (6-MP) is a well-known immunosuppressive medication with proven anti-proliferative activities. 6-MP possesses incomplete and highly variable oral absorption due to its poor water solubility, which might reduce its anti-cancer properties. To overcome these negative effects, we developed neutral and positively charged drug-loaded liposomal formulations utilizing the thin-film hydration technique. The prepared liposomal formulations were characterized for their size, polydispersity index (PDI), zeta potential, and entrapment efficiency. The average size of the prepared liposomes was between 574.67 ± 37.29 and 660.47 ± 44.32 nm. Positively charged liposomes (F1 and F3) exhibited a lower PDI than the corresponding neutrally charged ones (F2 and F4). Entrapment efficiency was higher in the neutral liposomes when compared to the charged formulation. F1 showed the lowest IC50 against HepG2, HCT116, and MCF-7 cancer cells. HepG2 cells treated with F1 showed the highest level of inhibition of cell proliferation with no evidence of apoptosis. Cell cycle analysis showed an increase in the G1/G0 and S phases, along with a decrease in the G2/M phases in the cell lines treated with drug loaded positively charged liposomes when compared to free positive liposomes, indicating arrest of cells in the S phase due to the stoppage of priming and DNA synthesis outside the mitotic phase. As a result, liposomes could be considered as an effective drug delivery system for treatment of a variety of cancers; they provide a chance that a nanoformulation of 6-MP will boost the cytotoxicity of the drug in a small pharmacological dose which provides a dosage advantage.
    Type of Medium: Online Resource
    ISSN: 2079-4991
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2662255-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Medip Academy ; 2018
    In:  International Journal Of Community Medicine And Public Health Vol. 5, No. 7 ( 2018-06-22), p. 2622-
    In: International Journal Of Community Medicine And Public Health, Medip Academy, Vol. 5, No. 7 ( 2018-06-22), p. 2622-
    Abstract: Diabetes has been theorized to build the danger of gallbladder complaint in light of the perception that insulin resistance and obesity are related with gallbladder. We conducted this meta-analysis using a comprehensive search of MEDLINE, PubMed, EMBASE, Cochrane Database of Systematic Reviews, and Cochrane Central Register of Controlled Trials till 15 January 2017 for prospective observational studies that assessed the relationship of the effects of diabetes mellitus on gallbladder. We identified 8 prospective studies that could be included in the meta-analysis which included 6,089,807 participants. The summary RR for diabetes patients was 1.42 (95% CI: 1.32–1.87, I2=99.4%, p 〈 0.0001). Although heterogeneity in general was very high, there was no heterogeneity between the studies with longer duration of follow-up. There was no evidence of publication bias. Our study shows additional support for an increased risk of gallbladder disease between diabetes patients.
    Type of Medium: Online Resource
    ISSN: 2394-6040 , 2394-6032
    Language: Unknown
    Publisher: Medip Academy
    Publication Date: 2018
    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. 2022-2022
    Abstract: Background Chemo-immunotherapy (CIT) with fludarabine, cyclophosphamide, and rituximab (FCR) is the standard of care in frontline treatment of CLL. With this approach, 25% of patients relapse within 24 months, whereas approximately one third of patients with hypermutated immunoglobulin heavy chains (IgHV) achieve a functional cure (Hallek et al. Lancet. 2010; Tam et al. Blood, 2014, Fischer et al, Blood 2015; Philip A. Thompson et al. Blood 2016). So far, mutations and/or deletions of TP53 remain the only predictive marker screened for in routine clinical practice, accounting for only one third of patients relapsing early after CIT. Recent next-generation sequencing (NGS) studies have revealed novel candidate predictors of early relapse including somatic mutations in RPS15 (Landau et al. Nature, 2015) and SAMHD1 (Clifford et al., submitted). Taken together with TP53disruption, these only occur in a subset of high-risk patients. Here, we present a comprehensive analysis of high-risk patients using Whole Genome Sequencing (WGS). Patients and Methods Using WGS we investigated 149 CLL patients from 5 national UK clinical trials: CLEAR (n=8), RIAltO (n=45), CLL 210 (n=22), ARCTIC (n=32) and AdMIRe (n=42). The two first line FCR-based clinical trials (ARCTIC and AdMIRe) were studied in most detail: 56 patients relapsed within 24 months; this group of patients will be referred to as high risk patients. Leukemia samples (peripheral blood) and germline samples (saliva) were collected for each patient. We performed WGS on the HiSeqX (Illumina). After read alignment, we detected somatic variants using Strelka 2.4.7 for small variants detection (SNV and InDels), Manta 0.28.0 for Structural variant (SV) detection, and Canvas 1.3.1 for Copy number variant (CNV) detection (Illumina). Non-coding regions were annotated with information from primary CLL, CLL cell lines and B-cell ENCODE databases. We interrogated the data at a gene scale and global level in order to identify patterns of early relapsing patients. Operative mutational signatures were analysed according to Alexandrov et al. (Nature, 2013). Putative regions of kataegis were calculated based on Lawrence et al. (Nature, 2013) and Alexandrov et al. (Nature, 2013). Results The mean coverage for CLL tumour and germline samples was 105.2X and 33.7X, respectively. The analysis of the whole cohort highlighted 1,723,603 somatic SNVs (mean= 11,570/sample) and 555,179 InDels (mean= 3,726/sample). Somatic SNVs spectrum consisted mainly of C 〉 T/G 〉 A mutations (30% of total SNVs reported) as previously described. The analysis of 13,490 somatic functional SNVs and InDels revealed novel candidate genes as most commonly mutated in the cohort. In high-risk patients, we noticed an enrichment of mutations in known genes such as TP53, genes of the NF-κB pathway and novel candidate genes previously reported in other cancers. A specific analysis of the functional coding mutations of known CLL driver genes revealed ATM, SF3B1 and IGLL5 as most commonly mutated genes in FCR responders compared to TP53, RPS15 and EGR2 in high risk patients. In depth analysis of somatic non-coding regions also identified potential new candidate regions associated with early relapse. Next, we investigated 52,871 CNAs (mean= 380/sample) and 29,080 SVs (mean= 195/sample) and identified as expected del13q, del17p, del11q and tri12 as the most frequent aberrations. In addition, we identified SVs across genes of interest in CLL, for instance TP53, ATM and BIRC3. Finally, we performed global genome analyses with investigation of mutational signatures and kataegis analyses highlighting hypermutated candidate regions, including the previously described IGLL5gene. Conclusion Here we present initial analysis of WGS data on 149 CLL patients from 5 UK clinical trials. Different patterns of mutations between low and high risk clinical groups are suggested. More detailed analysis with greater numbers of samples is ongoing and will determine the true clinical significance of these preliminary findings. The possibility of using WGS to aid clinical decision-making is becoming a realistic goal. Disclosures Becq: Illumina: Employment. He:Illumina: Employment. Pettitt:Celgene: Speakers Bureau; Gilead: Research Funding, Speakers Bureau; Roche: Research Funding, Speakers Bureau; Infinity: Research Funding. Hillmen:Pharmacyclics: Research Funding; Janssen: Honoraria, Research Funding; Roche: Honoraria, Research Funding; Gilead: Honoraria, Research Funding; Abbvie: Research Funding. Bentley:Illumina: Employment. Schuh:Gilead: Consultancy, Honoraria, Research Funding; Roche, Janssen, Novartis, Celgene, Abbvie: Consultancy, Honoraria.
    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. 124, No. 21 ( 2014-12-06), p. 1958-1958
    Abstract: Background Chronic lymphocytic leukaemia (CLL) is characterised by clinical and biological heterogeneity. Despite significant advances in therapeutic management, CLL remains largely incurable. Current risk stratification is based on cytogenetic features (del(17p), del(11q), del(13q), +12). So far, sequencing studies in CLL have focussed predominantly on the exome. These have identified a number of genes that are recurrently mutated at low frequency such as TP53, SF3B1, ATM, NOTCH1, MYD88, and BIRC3. Apart from TP53 abnormalities, none of these are currently used to guide clinical decisions and it is unclear how they are implicated in disease pathogenesis. Methods In this study, we sought to further refine the molecular landscape of CLL using whole genome sequencing (WGS) of paired tumour and germline DNA samples from a cohort of clinically annotated patients with CLL. We sequenced a heterogeneous cohort of 41 samples (25 males, 16 females, median age 69 (range 49-94)) with a range of clinical features (49% fludarabine refractory, 61% unmutated IgVH). Whole genome sequencing libraries were generated using the Illumina TruSeq PCR-free sample preparation kit, with a median insert size of 400bp, and subjected to 100bp paired-end sequencing on an Illumina HiSeq 2500 platform. Both tumour and germline libraries were sequenced to an average depth of 38x. Sequencing reads were aligned using the Isaac algorithm and the Starling and Strelka algorithms were used for SNV and Indel calling in germline and tumour samples respectively. All variants with a read depth 〈 10x or a quality score 〈 Q30 were excluded using Illumina VariantStudio software. For validation, selected mutations were verified using a combination of a targeted deep sequencing panel on the Illumina MiSeq platform and conventional Sanger sequencing. Copy number alterations were identified from the whole genome sequencing data using Nexus 7.5 (Biodiscovery), with findings validated on Illumina OmniExpress24 arrays. Results Whole genome sequencing revealed a total of 95,305 somatic indels and base substitutions, averaging 30.8 per patient (range 7-57) or 0.3 mutations per megabase. Of these mutations, 1266 occur in protein coding regions across 1108 genes, including 556 in 3’ and 5’ untranslated regions. Of these 1108 genes, we identified 93 as recurrently mutated (mutations present in more than one sample), including the previously described SF3B1 (12/41, 29.3%), TP53 (9/41, 22%), ATM (6/41, 14.6%), NOTCH1 (6/41, 14.6%), FAT1 (4/41, 9.8%) and BIRC3 (2/41, 4.9%). In addition to FAT1, we also identified two missense mutations in another cadherin superfamily member, FAT4(2/41, 4.9%), both occurring within the extracellular cadherin domains. Missense mutations were the most frequent (42.7%) followed by those in 3’ UTRs (36.1%), 5’ UTRs (7.7%), splice sites (6.1%), small indels (4.3%) and nonsense mutations (3.1%). In addition, 61.5% of missense mutations were identified as either deleterious or damaging by the SIFT or PolyPhen-2 algorithms. We used a modified version of the MutSigCV algorithm to identify genes with significantly higher mutation rates in the coding sequence. A similar statistical approach was used to identify significant mutations in untranslated regions. Importantly, a number of interesting candidate genes carried mutations in non-coding regions, including NFKBIZ (3/41, 7.3%), IGLL5 (3/41, 7.3%) and BCL2(2/41, 4.9%). Conclusion To our knowledge, this is the largest whole genome sequencing study in CLL so far. We present a comprehensive catalogue of genomic alteration in CLL and associate genome-wide patterns, including the presence of subclones, with clinical outcome. In addition to demonstrating the heterogeneous nature of the CLL genome, our data highlights the variety of mutations present in the regulatory regions of genes as well as structural variations, thus providing new insights for hypothesis-driven biomarker and therapeutic discovery. Disclosures Humphray: Illumina Cambridge Ltd: Employment. Becq:Illumina Cambridge Ltd: Employment. Bentley:Illumina Cambridge Ltd: Employment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 126, No. 18 ( 2015-10-29), p. 2110-2117
    Abstract: Targeted NGS of relapsed/refractory CLL reveals a high incidence of concurrent mutations that mostly affect the TP53, ATM, and SF3B1 genes. Concurrent mutations of the TP53, ATM, and/or SF3B1 genes confer short survival in patients with relapsed/refractory CLL.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 5590-5590
    Abstract: Background During B-cell development, somatic mutations are introduced into the variable region (V) of Immunoglobulin Heavy (IGH) genes by activation-induced cytidine deaminase (AID). In CLL, the degree of mutation in these regions is tied to clinical outcome, with IgHV hypermutated status (IgHV+, 〈 98% homology to germline) strongly predicting increased survival rates over unmutated patients (IgHV-) (Gardiner et al., Blood, 1999). In addition to AID, APOBEC signatures have been found in many human cancers (Gordenin at al., Nature Genetics 2013). So far, WGS efforts have focused primarily on IgHV+ patients (Puente et al, Nature 2015; Kasar et al, Nature Com 2015). Here, we perform comparative analyses between IgHV+/- patients using Whole Genome Sequencing (WGS) to explore this link. Methods Whole genome sequencing was performed on matched tumour and germline DNA from a cohort of 46 CLL patients, divided into two groups; 16 IgHV+ and 30 IgHV-. Sequence data was generated using the Illumina HiSeq 2500 platform, and somatic variants were generated by Strelka 2.4.7. SNVs were annotated using ANNOVAR (version 2015 Dec 14) and supplemented with information from primary CLL cell lines and B-cell ENCODE databases for the non-coding regions. Kataegis was identified based on the methods of Lawrence et al. (Nature, 2013) and Alexandrov et al. (Nature, 2013). Mutation signatures were analysed according to Alexandrov et al. (Nature, 2013). Results We identified a total of 64,420 high confidence somatic SNVs from 46 samples (mean=1400), of which 44% were from the IgHV+ cohort (mean=1680) and 56% from IgHV- (mean=1237). Of these; SNVs in coding regions (exons, introns, UTRs) occurred at significantly higher proportions in IgHV- patients (P=0.0004, Fishers Exact test). Mutations in predicted active DNAse hypersensitivity regions and H3k27 acetylated regions, however, were significantly more likely to occur in IgHV+samples (P 〈 0.0001). Mutational signature analysis revealed three distinct signatures shared between the two cohorts. Two of these (Tsig1 and Tsig2) clustered with Alexandrov signature 1A, and the third to signature 1B (Tsig3), both of which were designated as ageing signatures. Despite this, our signatures significantly correlated with the proportion of mutated AID (P 〈 0.03; P 〈 0.03; Tsig1 and Tsig3 respectively), and APOBEC sites (P 〈 0.001; P 〈 0.001; Tsig1 and Tsig3 respectively), and not with age. These signatures were found to differ significantly between cohorts (P 〈 0.001), regardless of treatment. Tsig2 was not found to correlate with either patient age, AID signature or APOBEC signature, suggesting that it may be a novel signature. A total of 53 kataegis regions were identified across all patients, of which three were found on chromosomes 2, 14 and 22, corresponding to the IG loci. Coding mutation hotspots were located in known CLL driver genes, including TP53, ATM, IKZF3 and SF3B1. Non-coding recurrent hotspots caused by AID were found to predominately affect promoter and enhancer regions of key B-cell pathways, including BCL6, BCL2, BTG2, IGLL5, and PAX5. This observation is closely linked to the IgHV status; the IgHV+ cases frequently harboured mutated non-coding variants in genes involved in B cell signalling, whilst IgHV- cases were more likely to contain exonic driver mutations. Kataegis analysis also revealed novel non-coding mutations in recurrently mutated genes that were common in IgHV- cases, including CDK6 and BIRC3, and a non-coding RNA region on chromosome 9 that was hypermutated only in IgHV-cases. Conclusion Here, we present a whole genome sequencing study on 46 patients divided into two cohorts of IgHV+ and IgHV-. WGS revealed distinct changes in mutation distribution and signatures between these cohorts; differences that are mirrored in both the recurrently mutated gene profiles, and the regions of somatic hypermutation. We demonstrate that mutational differences in IgHV+ and IgHV- patients extend far beyond the IgHV regions and the 1% of the coding genome. This study paves the way for future work into understanding the genomic differences between these cohorts and thus, contribute to increasing our understanding of the molecular mechanisms underlying the different clinical outcomes. Disclosures Hillmen: Pharmacyclics: Research Funding; Janssen: Honoraria, Research Funding; Roche: Honoraria, Research Funding; Gilead: Honoraria, 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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 3115-3115
    Abstract: Background Chronic Lymphocytic Leukemia (CLL) is characterised by a highly heterogeneous natural history and treatment response. Indeed, 50% of immunoglobulin heavy chain variable region (IgHV) hypermutated patients have an excellent progression free survival (PFS) after chemoimmunotherapy. Conversely, 25% of FCR treated patients relapse within 24 months (high risk CLL). Recent studies have shown that complex karyotype with or without TP53 disruption predicts for relapse after BCL2 therapy and BTK inhibitors. However, TP53 is the only marker for which routine testing is available. Overall, nearly 80% of patients relapsing after frontline FCR do not present a known poor risk genomic marker. Additional candidate genomic predictors of poor outcome including mutations in coding regions of NOTCH1, SF3B1 and RPS15, non-coding regions of NOTCH1 and enhancer regions of PAX5, telomere length, IgHV status, and DNA Damage Repair (DDR) germline mutations including TP53 and ATM have been reported in CLL. Further, the role of mutational signatures and regions of kataegis also merit additional investigation in progressive CLL. Evaluating all candidate predictors requires complex time consuming, multi-modality testing outside the scope of routine clinical diagnostic practice, however, in isolation, each has low predictive value. Here, we show preliminary data on a novel patient stratification method based on whole genome sequencing (WGS) data incorporating multiple genomic features in a single test. Patients and Methods Tumor (peripheral blood) and germline (saliva) samples were collected from 321 patients from 6 UK trials via the Genomics England CLL pilot: ARCTIC (n=61), AdMIRe (n=64), CLL 210 (n=30), CLEAR (n=12), RIAltO (n=88) and FLAIR (n=66). We performed WGS on the HiSeqX (Illumina). After read alignment, we detected somatic variants using Strelka 2.4.7 for small variants detection (SNV and InDels), Manta 0.28.0 for structural variant (SV) detection, and Canvas 1.3.1 for copy number variant (CNV) detection (Illumina). Non-coding regions were annotated with information from primary CLL, CLL cell lines and B-cell ENCODE databases. Mutational signatures and putative regions of kataegis were calculated based on Alexandrov et al. (Nature, 2013) and Lawrence et al. (Nature, 2013). Telomere lengths were assessed using Telomerecat. Data aggregation was performed using contingency tables combined with non-negative matrix factorization. Results Mean coverage was 94.2X for tumor and 28.5X for germline samples. We found a median of 9172 SNPs/sample after filtering and 2348 indels/sample across 321 patients. High risk CLL was enriched for genomic complexity and poor prognostic mutations. The most frequently mutated genes were SF3B1 (17%), TP53 (13%), NOTCH1 (12%), IGLL5 (12%), and ATM (11%). Analysis of non-coding regions using DNA methylation markers, ATAC-seq and Hi-C revealed potential candidate regions associated with early relapse. Using CNA and SV data, we identified interesting patterns of genomic complexity and structural variants, including a trend towards enrichment of del8p in Relapse/Refractory and FCR non-responders. Additionally, we investigated mutation signatures and kataegis across coding and non-coding regions of the genome. We correlated exonic regions of DDR genes in germline data with clinical outcomes and extended this to genes mutated in both tumor and germline data, termed germline-tumor double-hits. We examined the relationship between the Alexandrov hypermutation signature, IgHV status (determined by % homology to the reference genome) and PFS, and combined mutational density at the Ig locus with mutation signature aiming to predict IgHV status. Finally, we produced a binary contingency matrix, using non-negative matrix factorization to cluster the samples. This method highlighted patient groups with shared genomic profiles. Conclusion We present preliminary data on a patient stratification method derived from WGS of 321 paired germline and CLL trial samples. Our predictive signature includes driver gene mutations, CNAs, IgHV status, genomic complexity, telomere length, overall mutation burden and genes with germline-tumor double-hits. Our comprehensive, NGS-based patient stratification attempts to predict patient outcome in a single sequencing run. Disclosures Becq: Illumina: Employment. He:Illumina: Employment. Ross:Illumina: Employment. Bentley:Illumina: Employment. Pettitt:Celgene: Research Funding; Gilead: Research Funding; Roche: Research Funding; GSK/Novartis: Research Funding; Napp: Research Funding; AstraZeneca: Research Funding; Chugai: Research Funding. Hillmen:Novartis: Research Funding; Gilead Sciences, Inc.: Honoraria, Research Funding; Alexion Pharmaceuticals, Inc: Consultancy, Honoraria; F. Hoffmann-La Roche Ltd: Research Funding; Celgene: Research Funding; Acerta: Membership on an entity's Board of Directors or advisory committees; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Schuh:Giles, Roche, Janssen, AbbVie: Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1974-1974
    Abstract: Background: Previous studies using next-generation sequencing (NGS) have led to the identification of a number of genes mutated frequently in CLL. Recent publications focus on the most recurrently mutated genes (TP53, SF3B1 and NOTCH1) which tend to be mutually exclusive. Large series of untreated patients have shown that these mutations have a prognostic impact. Relapse may be associated with more frequent mutational events. Further investigation of relapsed CLL genomes within a clinical trial setting using a comprehensive NGS gene panel is required. Methods: Using targeted NGS we determined the mutational spectrum of 118 refractory/relapsing CLL patients enrolled in one French and two UK prospective trials (ICLL01 from the French intergroup GCFLLC/MW-GOELAMS, NCRNCLL201, NCRNCLL202 respectively). Eighty percent of patients had an unmutated IGHV status and 21 (18%) patients carried a 17p deletion. Sequencing libraries were composed of a panel of nine recurrently mutated genes in CLL (i.e.TP53, SF3B1, ATM, NOTCH1, XPO1, SAMHD1, MED12, BIRC3 and MYD88) and run on the Illumina MiSeq instrument (Illumina Inc). On average 14.1 M reads were obtained per run of which 96.8% were identified reflecting an acceptable signal to noise ratio. Yield was 4.1 Gb and 95.9% of reads were above Q30 across 6 MiSeq runs. Data was analysed using our in-house bioinformatics pipeline consisting of a combination of two different aligners (Custom Amplicon Alignment, Illumina Inc and Stampy, Wellcome Trust centre for Human Genetics), two variant callers (GATK, Broad Institute and Platypus, Wellcome Trust centre for Human Genetics) and a stringent filtering process in order to detect SNVs and indels with a variant allele frequency down to 7%. Results: We identified a total of 196 mutations (mean=1.7/sample) in 95 (80%) patients: 138 missense mutations, 41 substitutions/indels, 12 nonsense and 5 splicing mutations. TP53, SF3B1 and ATM mutations occurred frequently in 29 (24.6%), 33 (28%) and 29 (24.6%) patients, respectively. Eighteen (15.3%) patients harbored a NOTCH1 mutation matching the range of reported frequency. Mutations in the other genes sequenced were distributed as follows: XPO1 mutations in 17 (14.4%), SAMHD1 mutations in 12 (10.2%), MED12 mutations in 10 (8.5%), BIRC3 mutations in 6 (5.1%) and MYD88 mutations in 3 (2.5%) patients. Twenty-three (20%) patients did not have any mutations present (Figure 1, cluster #1). A total of 51 (43%) patients had one gene mutated (Figure 1, cluster #2) and the remaining 44 (37%) patients had two or more genes mutated (Figure 1, clusters #3 & #4). Recurrent combinations of mutations (affecting more than 5% of patients) were found in a group of 23 (20%) patients. These combinations of mutations comprised of at least two of the following genes: TP53, SF3B1 and ATM (Figure 1, cluster #3, so called multiple-hit (MH) profile). Remarkably, mutations in these 3 genes were found significantly more frequently associated than in isolation. We then investigated the potential clinical relevance of the MH profile. This profile was associated with poorer ORR than the remaining cohort (43% vs 80%, P 〈 .0001). None of the patients with a MH profile achieved CR compared to 24% for the remaining patients (P=.006). MH patients have also shorter median PFS of 12 months compared to 19 months in cluster #1, 23 months in cluster #2 and 18 months in cluster #4 (P=.03). Multivariate analysis for PFS including relevant factors such as fludarabine-refractory' and TP53 disruption confirmed the adverse prognostic value related to the MH profile (HR=3.194 [95%CI=1.493-6.835], P=.003). Interestingly, among the TP53-disrupted patients, the MH profile retained its prognostic impact with a median PFS of 11 months for those with mut-SF3B1 and/or mut-ATM versus 22 months for those with wt-SF3B1 and wt-ATM (P=.022). Conclusion: The mutational landscape of relapsing CLL is marked by a group of patients with combined mutations of the TP53, ATM and SF3B1 genes (multiple-hit profile) and is associated with an adverse prognostic impact. In addition to TP53 and SF3B1, ATM should be sequenced at relapse to predict outcome and guide subsequent therapeutic intervention. Further studies are required to confirm these findings and to understand the subclonal distribution of these mutations. Figure 1 Figure 1. Disclosures Hillmen: Pharmacyclics, Janssen, Gilead, Roche: Honoraria, Research Funding. Tournilhac:mundipharma: Honoraria, Other, Research Funding; GSK: Honoraria, Other, Research Funding; Roche: Honoraria, Other, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Nature Genetics, Springer Science and Business Media LLC, Vol. 54, No. 11 ( 2022-11), p. 1675-1689
    Abstract: The value of genome-wide over targeted driver analyses for predicting clinical outcomes of cancer patients is debated. Here, we report the whole-genome sequencing of 485 chronic lymphocytic leukemia patients enrolled in clinical trials as part of the United Kingdom’s 100,000 Genomes Project. We identify an extended catalog of recurrent coding and noncoding genetic mutations that represents a source for future studies and provide the most complete high-resolution map of structural variants, copy number changes and global genome features including telomere length, mutational signatures and genomic complexity. We demonstrate the relationship of these features with clinical outcome and show that integration of 186 distinct recurrent genomic alterations defines five genomic subgroups that associate with response to therapy, refining conventional outcome prediction. While requiring independent validation, our findings highlight the potential of whole-genome sequencing to inform future risk stratification in chronic lymphocytic leukemia.
    Type of Medium: Online Resource
    ISSN: 1061-4036 , 1546-1718
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 1494946-5
    SSG: 12
    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...