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. 116, No. 21 ( 2010-11-19), p. 318-318
    Abstract: Abstract 318 The classical myeloproliferative neoplasms (MPNs) comprise of three entities: polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). Despite distinct phenotypic features of MPN entities they share characteristics like clonal hematopoiesis, risk for thrombosis and bleeding and tendency to transform to secondary acute myeloid leukemia (post-MPN AML). In order to investigate the genetic lesions associated with MPN a large single-center cohort of 311 MPN patients was analyzed for chromosomal aberrations using high resolution Affymetrix SNP 6.0 arrays. The cohort included 150 patients with PV, 90 with ET, 68 with PMF and 3 with post-MPN AML. Of the 311 patients, 144 (46%) had a normal karyotype and 167 (54%) harbored 1 to 8 detectable chromosomal aberrations. We found 51 gains, 102 deletions and 143 uniparental disomies (UPDs). A total of 13 recurrent chromosomal defects (more than three events) were detected. We investigated if either the number of chromosomal aberrations in a patient or specific types of lesions associate with a certain patient group defined by clinical criteria. Chromosomal aberrations were equally distributed among the three MPN entities and only 9pUPD showed significant clustering with PV. We did not detect an association between the number of chromosomal aberrations and disease duration. Patients positive or negative for JAK2 mutations did not differ significantly in the frequency of chromosomal aberrations (except of the association of 9pUPD with JAK2 positive MPN). Patients with complex karyotype were significantly older than patients with normal karyotype (P 〈 0.001). Transformation to post-MPN AML is an important complication in MPN. To investigate associations between chromosomal changes and transformation, we included additional 19 post-MPN AML patients from another center into the study (total N=22). Patients in the post-MPN AML group harbored significantly more chromosomal lesions (P 〈 0.001). Recurrent aberrations of chromosomes 1q, 7p, 7q, 5q, and 3q strongly associated with post-MPN AML. When we reviewed the clinical data of patients in chronic phase MPN harboring the leukemia-associated aberrations, they showed features of disease progression, and some transformed to AML at a later follow-up. We were able to map a common deleted region (CDR) on chromosome 4 to the tet oncogene family member 2 (TET2), a gene frequently deleted in myeloid disorders. On chromosome 7p we mapped a CDR to the Ikaros transcription factor (IKZF1) and a 7q CDR mapped to a novel putative tumor suppressor, the cut-like homeobox 1 gene (CUX1). Interestingly, in one patient who carried a UPD of chromosome 7q we did not detect a mutation in the CUX1 gene but an R288Q mutation was found in the EZH2 gene. Chromosome 7 aberrations in our cohort were strongly linked to post-MPN AML. Our results show that at least three chromosome 7 genes (IKZF1, CUX1, and EZH2) are relevant in leukemic transformation. In addition to chromosome 7, we found gains of chromosome 1q equally relevant in post-MPN AML. We mapped the common 1q amplification to a 3.5 Mbp region that contained the MDM4 gene. Mdm4 is a known negative regulator of p53 and was frequently shown amplified in various cancers. This result prompted us to investigate the relevance of the p53 pathway in post-MPN AML and we sequenced TP53 in all 22 leukemic patients and found mutations in 6 cases (27.3%). Interestingly, none of the patients with TP53 mutation carried an MDM4 amplification. Taken together, 10 out of 22 post-MPN AML cases (45.5%) had evidence of a p53-related defect. To gain deeper insight into the pathways involved in transformation to post-MPN AML we sequenced genes commonly affected in de novo AML, and found two patients with mutations in FLT3, two patients with RUNX1 mutations, two patients with either IDH1 or IDH2 mutations. We conclude that lesions known to play an important role in de novo AML are present only in a fraction of post-MPN AML patients. In this study we show that aberrations of the p53 pathway together with the chromosome 7 lesions affecting IKZF1 and CUX1 are present in 64% of all post-MPN AML patients. Our data give insight into the genetic complexity and heterogeneity of MPN patients in chronic phase as well as in post-MPN AML. The marked genetic heterogeneity of MPN patients will render targeted therapies challenging and underlines the requirement of personalized treatments. 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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 127, No. 3 ( 2016-01-21), p. 362-365
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 998-998
    Abstract: Abstract 998 Chromosomal deletions are frequent cytogenetic defects in hematological malignancies. Common deleted regions (CDRs) defined by the minimal physical overlap of all deletion events are thought to harbor tumor suppressors relevant for pathogenesis. Haploinsufficiency is likely to be a common consequence of chromosomal deletions affecting the function of tumor suppressors within the deleted locus. The use of RNA interference (RNAi) offers a unique opportunity to mimick haploinsufficiency by partial knock-down of the gene transcripts. Since deletions are usually large containing a high number of candidate genes, we aimed to develop a screening method targeting several candidates at once and assaying for tumor suppressor features in a pool of knock-downs. As a model for our approach we selected a CDR on chromosome 20q frequently found clonal in myeloid diseases. We established a screen for knock-downs causing cytokine hypersensitivity. This CDR (physical position chr20:38.7- 42.2) spans 3.5Mb and contains 16 genes (MAFB-JPH2). We cloned 3 short hairpin RNAs (shRNAs) for each mouse homologue of the 16 target genes into the pLKO.2 lentiviral vector. The 48 shRNA constructs were “bar-coded” with different, unique 24bp DNA bar-codes, that can be identified and quantified using the microbead-based xMAP technology (Luminex). We lentivirally delivered the constructs independently into the erythropoietin (Epo) dependent murine cell line Baf3/EpoR, pooled the individual knock-downs in equal amounts and assayed for cytokine hypersensitivity based proliferation advantage under stringent Epo concentrations. Applying a scoring system based on the relative increase/decrease of the individual bar-codes in the pool over time, we could identify the knock-down of topoisomerase 1 (Top1) to induce Epo-concentration dependent outgrowth. Two different Top1 shRNA constructs succeeded in consistently mediating proliferative advantage in three biological replicates. We set up a validation experiment pooling Top1 knock-down cells with control cells (bar-coded, no shRNA) in a 1:1 ratio and could observe significant dominance of the Top1 knock-down cells establishing after 10–15 days in culture and increasing over time (p 〈 0.0001). Knock-down efficiency of both successful constructs measured by qPCR and was consistently between 30–60% of control cell mRNA expression. Based on the convincing cell line data we set up an in vivo validation in a competitive repopulation mouse model. We established a protocol for lentiviral transduction of murine lineage depleted (lin-) bone marrow progenitor cells. Transplanting in a 1:1 ratio of Top1 knock-down and control progenitors into three lethally irradiated mice, we surprisingly observed an outcome opposite to the cell line results. Top1 knock-down progenitors showed a clear disadvantage in repopulation capacity, being underrepresented in the peripheral blood at week 3 post-transplantation and furthermore fully outcompeted by the control cells around week 15 post-transplantation. Based on the unreliability of cell line models in our setup we repeated the screen with the full library (48 shRNAs) in vivo. Two different constructs targeting phospholipase C gamma 1 (Plcg1) had the top score in 3 out of 5 transplanted mice, resulting in an impressive cumulative score. Showing equal representation with the other knock-downs (approximately 2%) in the donor pool, Plcg1 knock-down cells represented up to 13% of the peripheral blood cells at week 3 post- transplant, increasing to up to 29% at week 7 post-transplant. Our results suggest that cell lines often might not be the proper model for studying growth regulation. Further, our in vivo screen revealed Plcg1 as a promising candidate with a tumor suppressor function in hematopoiesis. 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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 435-435
    Abstract: Abstract 435 Transformation to acute myeloid leukemia (AML) is a major complication of myeloproliferative neoplasms (MPNs). Although clinical prognostic factors have been defined such as age, severe anemia, elevated white blood cell count with a high proportion of immature myeloid cells and a high number of circulating CD34+ cells, there is little knowledge of the genetic changes leading to leukemic transformation in MPN. To gain insight into these genetic changes, we studied 12 patients with post-MPN leukemia using Affymetrix SNP 6.0 microarrays. Acquired genetic lesions were detected in 10 patients. Beyond well described frequent events like 9pUDP, chromosome 7p deletions (del7p) emerged as a novel recurrent defect in this patient group. Mapping of the minimal deleted region of del7p by combining microarray, copy number and loss of heterozygosity analysis resulted in a minimal deleted region of 0.25 Mb in size, restricted to the IKZF1 gene. IKZF1 is encoding the transcription factor Ikaros, which is known to have a pleiotropic function in the regulation of hematopoiesis, as well as crucial involvement in hematopoietic malignancies, primarily of the lymphoid lineages. Based on the observation of 16.7% (2 out of 12) of post-MPN leukemia patients carrying an IKZF1 deletion, we further screened a chronic phase MPN patient cohort from Vienna for the defect and found 1 out of 235 informative patients carrying an IKZF1 deletion. Thus, deletion of IKZF1 exhibited statistically significant clustering with post-MPN leukemia when compared to chronic phase MPN (P=0.0063). To validate these results in an independent patient cohort, we screened 20 post-MPN leukemic and 156 chronic phase MPN patients from Pavia for loss of IKZF1, and found 4 and 1 positive patients, respectively (P=0.0006). Taking these two cohorts together, IKZF1 deletions could be found in 0.5% of chronic MPN patients and 18.8% of post-MPN leukemic patients, showing statistically significant association with post-MPN leukemia (P 〈 0.0001). We could define haploinsufficiency as mechanism of IKZF1 tumor suppressor inactivation, since IKZF1 exon sequencing of del7p patients did not show any mutations on the non-deleted allele of IKZF1 and IKZF1 mRNA could be detected in del7p patients. In order to characterize the role of IKZF1 deletion in clonal expansion, we monitored oncogenic mutation and deletion burdens in granulocytes of two del7p patients in serial samples over time. In both patients, one being positive for JAK2-V617F and the other for MPL-W515L, increased IKZF1 deletion burden was detected as a late event after the acquisition of oncogenic mutations. We also examined the hematopoietic progenitors of a del7p patient by genotyping individual BFU-E and CFU-GM colonies for defects detected in granulocytes. IKZF1 deletion was found to be a late defect in the clonal evolution of MPN, occurring after oncogenic mutations such as JAK2-V617F and chromosomal aberrations such as del13q. In murine hematopoietic progenitor cells, sh-RNA induced deficiency for Ikzf1 resulted in cytokine hypersensitivity in vitro. Based on these data we can conclude that IKZF1 deletion does not induce arrest in myeloid differentiation, it is a late genetic event in the clonal evolution of MPN and represents an important step in the leukemic transformation of a subpopulation of MPN 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: 2009
    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: American Journal of Hematology, Wiley, Vol. 87, No. 11 ( 2012-11), p. 1010-1016
    Type of Medium: Online Resource
    ISSN: 0361-8609
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 1492749-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3559-3559
    Abstract: Abstract 3559 Secondary acute myeloid leukemia (sAML) evolves from different types of chronic myeloproliferative neoplasms (MPN) and myelodysplastic syndromes (MDS). However, acute myeloid leukemia frequently arises de novo (dnAML) without a previous hematological phenotype. Since the genetic basis of sAML is poorly understood our objective was to define genetic aberration profiles of sAML patients and compare the distribution of these aberrations to the ones found in dnAML. We studied a total of 195 patients of which 122 were diagnosed with dnAML and 73 with either post-MDS (n=27) or post-MPN (n=46) sAML. In order to obtain high-resolution karyotypes we genotyped DNA samples using Affymetrix SNP 6.0 arrays enabling analysis of 1.8 million data points per genome. In addition to copy number data, loss of heterozygosity associated with acquired uniparental disomy (UPD) was evaluated. Furthermore, the cytogenetic data was complemented with mutational analyses of commonly mutated genes in myeloid malignancies: IDH1, IDH2, NPM1, CBL, FLT3 and TP53. With the arrays used we could not detect any cytogenetic lesion in 32% of dnAML and 18% of sAML. Furthermore, dnAML showed karyotypes with lower complexity compared to sAML. We observed 31 recurrent cytogenetic aberrations ( 〉 4 events). Of these 12 showed significant bias towards sAML. The most prominent were 9pUPD (P=0.0001), 1q gain (P=0.0023), del7q (P=0.0039), 11qUPD (P=0.0045) and del4q (P=0.006), targeting known genes JAK2, MDM4, CUX1, CBL and TET2, respectively. In our series gains of 3q involving EVI1 were exclusively found in sAML (P=0.018) and the 3q amplicon contained EVI1 and 10 other genes. The most common cytogenetic aberration observed overall was del5q (34 events) in our patient cohort, significantly associated with sAML (P=0.017). As frequent 1q gains targeting MDM4 in sAML previously implicated the p53 pathway in sAML leukemogenesis, we performed exon sequencing of TP53 in the entire patient cohort. The results clearly showed that TP53 is significantly more mutated in sAML compared to dnAML (P=0.0052), with 16.4% of sAML patients carrying the mutation, comparing to only 4% of dnAML. We also confirmed the previously described co-occurrence of del5q with TP53 mutations (P=0.0031). Another gene showing significantly higher frequency of mutations in sAML is CBL (P=0.0035), found mutated in 10.3% of sAML and 0.8% of dnAML, respectively. On the other hand mutations in FLT3 and NPM1 were more common in dnAMLs (P=0.0090 and P=0.0001, respectively). FLT3 was found mutated in 22.6% of dnAML and 7.6% in sAML. Mutations of NPM1 were present in 24.4% of dnAML and in only 3% of sAML. Mutations of IDH1 and IDH2 were found to be present at similar frequencies in both AML types. Despite the genetic and phenotypic diversity of MPN and MDS in the chronic phase of the diseases we observed no significant genetic differences in the leukemic phases of these disorders within our sAML cohort. Of all the aberrations, deletions were most common representing 55% of all lesions (361 total events). When we mapped all the detected deletion events we obtained a high-resolution deletion map of AML. A number of known leukemia-associated tumor suppressor genes were found within the common deleted regions such as TET2, CUX1, IKZF1, FOXP1, ETV6, NF1 and DNMT3A. We also identified a number of new tumor suppressor candidates distinct or common for both AML groups, such as ASXL2, BAI3 and others. The higher cytogenetic complexity of sAML in contrast to dnAML and the differences of aberration frequencies between the two AML types are likely attributed to a longer disease duration and clonal evolution in sAML. Furthermore, clinical management of the chronic disease phase might influence the cancer genome evolution in sAML whereas such influences are absent in dnAML. Despite these differences, certain defects appear to be universally contributing to leukemogenesis such as IDH1/2 mutations. Our data indicate that leukemogenesis in MPN and MDS is not predominantly driven by lesions typical for dnAML. This study also provides a number of new potential markers for genetic stratification of patients with acute myeloid leukemia. 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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3755-3755
    Abstract: Abstract 3755 Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder characterized by excessive production of myeloid cells and the presence of the BCR-ABL fusion oncogene resulting from the t(9;22) reciprocal translocation. The leukemic clone in CML often accumulates other somatic lesions that may collaborate with BCR-ABL oncogenicity or confer resistance to tyrosine kinase inhibitors. The chromosomal instability in CML is believed to be caused by the BCR-ABL oncogene. We hypothesized that the accumulation of cytogenetic lesions in CML are acquired after the t(9;22). Thus, imatinib therapy should not only cause the molecular remission of BCR-ABL positivity but also the remission of most cytogenetic lesions. If cytogenetic lesions preceded t(9;22) acquisition, a residual clone should be detectable after remission of BCR-ABL positivity. In order to test these predictions, we determined high-resolution karyotypes of CML patients at diagnosis and at time points with variable durations of imatinib therapy. At the same time, we aimed to characterize the overall cytogenetic aberration profile of CML at different disease stages. The copy number abnormalities and loss of heterozygosity coupled with acquired uniparental disomy (UPD) were determined using Affymetrix SNP 6.0 arrays. Overall 62 patients were included in the study. For 13 patients paired DNA samples were available, the first one taken at diagnosis and the second after treatment with imatinib. These samples were used to evaluate the cytogenetic remission after treatment. In 7 out of 13 patients we could detect an additional chromosomal aberration at diagnosis. Interestingly, the clone size assessment based on copy number signal intensity data was concordant with BCR-ABL burden. Analysis of the follow-up samples revealed that 6 patients with additional cytogenetic aberrations at diagnosis showed complete cytogenetic remission. In one patient imatinib therapy led to significant reduction of the BCR-ABL burden but a residual BCR-ABL negative clone persisted. This residual clone exhibiting del6p, del10q and del13q was detectable as a minor clone at diagnosis and it fully replaced the BCR-ABL positive clone after 8 months of imatinib therapy. This data indicates that the BCR-ABL fusion was either acquired through the clonal progression of the initial clone carrying described cytogenetic aberrations, or two distinct clones were present at diagnosis. In order to define the cytogenetic profile of CML samples we analyzed an additional number of 12 patient samples taken at diagnosis, as well as 37 samples taken after treatment with tyrosine kinase inhibitors. In 35.5% of the patients we could detect cytogenetic lesions, consisting of 28 deletions, 9 gains and 1 UPD. The most recurrent deletions were in the breakpoint region of the BCR (n=3) and ABL (n=5). Deletions of 13q (4 events) defined two common deleted regions (CDR) overlapping with previously defined CDR in other myeloproliferative neoplasms and chronic lymphoid leukemia. Other set of deletions clustered on chromosome 7p targeting 5 genes, among which was ABCB5, a member of ATP-binding cassette transporter family. One of the patients was also found to be a carrier of 11pUPD. In order to characterize the target gene within the UPD region we performed whole exome next generation sequencing of this sample and found 9 candidate mutations which are currently evaluated. Compared to other myeloproliferative neoplasms, CML exhibits low cytogenetic complexity. This is demonstrated by the fact that only 35.5% of patients have chromosomal aberrations in addition to t(9;22). Our data indicates that the majority of aberrations in CML occur after t(9;22) during the clonal evolution of the leukemic cells. Most of these aberrations disappear after treatment with tyrosine kinase inhibitors. However in a proportion of patients clonal hematopoiesis persists, even after successful targeting of BCR-ABL positive cells. In these patients imatinib therapy does not restore polyclonal hematopoiesis despite the BCR-ABL remission. The persistence of a clone prone to accumulation of other mutations can potentially lead to induction of other hematological phenotypes. 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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 123, No. 10 ( 2014-03-06), p. 1544-1551
    Abstract: JAK2 (V617F)-mutated essential thrombocythemia and polycythemia vera are different phenotypes in the evolution of a single neoplasm. CALR-mutated essential thrombocythemia is a distinct disease entity not only at the molecular level, but also with respect to clinical outcomes.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 141, No. 15 ( 2023-04-13), p. 1831-1845
    Abstract: Gain-of-function mutations in the signal transducer and activator of transcription 3 (STAT3) gene are recurrently identified in patients with large granular lymphocytic leukemia (LGLL) and in some cases of natural killer (NK)/T-cell and adult T-cell leukemia/lymphoma. To understand the consequences and molecular mechanisms contributing to disease development and oncogenic transformation, we developed murine hematopoietic stem and progenitor cell models that express mutated STAT3Y640F. These cells show accelerated proliferation and enhanced self-renewal potential. We integrated gene expression analyses and chromatin occupancy profiling of STAT3Y640F-transformed cells with data from patients with T-LGLL. This approach uncovered a conserved set of direct transcriptional targets of STAT3Y640F. Among these, strawberry notch homolog 2 (SBNO2) represents an essential transcriptional target, which was identified by a comparative genome-wide CRISPR/Cas9-based loss-of-function screen. The STAT3-SBNO2 axis is also present in NK-cell leukemia, T-cell non-Hodgkin lymphoma, and NPM-ALK-rearranged T-cell anaplastic large cell lymphoma (T-ALCL), which are driven by STAT3-hyperactivation/mutation. In patients with NPM-ALK+ T-ALCL, high SBNO2 expression correlates with shorter relapse-free and overall survival. Our findings identify SBNO2 as a potential therapeutic intervention site for STAT3-driven hematopoietic malignancies.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 131, No. 6 ( 2018-02-08), p. 649-661
    Abstract: Mutant CALR drives ET and MF in knockin mice. Mutant CALR expression results in expansion of phenotypic HSCs without a self-renewal advantage.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...