scholarly journals Inherited Blood Cancer Predisposition through Altered Transcription Elongation

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 629-629
Author(s):  
Liam Cato ◽  
Jiawei Zhao ◽  
Erik L. Bao ◽  
Samuel Bryant ◽  
Nicholas Williams ◽  
...  

Abstract Despite considerable advances in defining the somatic driver mutations underlying myeloid malignancies, including the myeloproliferative neoplasms (MPNs), a significant heritable component for these diseases remains poorly understood. While common genetic variant association studies have been valuable, they fail to explain the majority of heritable variation. We reasoned that rare variant association studies could provide a valuable complementary approach to identify additional inherited risk factors. We therefore utilized exome sequencing data from 166,953 UK Biobank participants and performed a gene-based burden analysis for germline genetic variants conferring risk for acquiring a myeloid malignancy. CTR9, which encodes a key component of the PAF1 transcription elongation complex, was among the significant genes identified (SKAT-O p-value = 5.47x10 -7). The deleterious variants in CTR9 collectively exhibit a 9.6 (95%CI = 4.86-19.04) increased odds of acquiring a myeloid malignancy and this risk was largely driven by the MPNs. We replicated this association in an independent cohort of 211 MPN patients using external controls. We could show through structural and biochemical analyses that the identified deleterious variants perturbed assembly of the PAF1 complex but did not display dominant negative activity. Given that increased hematopoietic stem cell (HSC) self-renewal has been shown to predispose to the risk of acquiring MPNs, we sought to define whether CTR9 perturbation could alter HSC self-renewal or function. We achieved predominantly heterozygous loss-of-function in human hematopoietic stem and progenitor cells (HSPCs) by titrating Cas9 ribonucleoprotein delivery with several independent guide RNAs. Partial loss of CTR9 in HSPCs resulted in expansion of phenotypic long-term HSCs (LT-HSCs) and more differentiated short-term HSCs (ST-HSCs). We additionally could show through single cell RNA-sequencing (scRNA-seq) that there was an expansion of molecularly defined HSCs upon partial loss of CTR9. The observed increase in HSCs appeared paradoxical, given that the PAF1 complex has been suggested to be crucial for HSC maintenance. To explore how the observed HSC expansion with CTR9 perturbation may arise, as well as given known interactions between the PAF1 complex and the competing transcriptional super elongation complex (SEC), we examined whether SEC target genes in HSCs, such as mid to posterior HOXA genes, may be activated with partial CTR9 loss. Remarkably, we observed a significant enrichment for hematopoietic SEC target genes upon CTR9 perturbation in HSCs by gene set enrichment analysis (normalized enrichment score = 3.29, p-value < 0.001). In light of these findings suggesting that SEC activity may be increased with partial CTR9 loss-of-function, as occurs in individuals harboring myeloid malignancy variants, we sought to functionally validate these observations. Using the inhibitors of the SEC, including SR-0813 that targets MLLT3 or with an inhibitor of CDK9, we noted rescue of the CTR9-mediated expansion of phenotypic LT- and ST-HSCs without a significant impact on the bulk HSPC population. To further elucidate underlying mechanisms, we performed immunoprecipitation of PAF1 or SEC component MLLT3 in HSPCs with control or CTR9 editing. While we continued to pull down all PAF1 complex components with PAF1, we also noted pulldown of MLLT3, which increased with CTR9 editing. MLLT3 immunoprecipitation revealed selective pulldowns of PAF1 and CDC73, which also increased with CTR9 editing. These findings show how PAF1 complex components PAF1 and CDC73 interact with and stimulate SEC activity. Our findings reveal how CTR9 usually restricts this activity and constrains transcriptional elongation to limit HSC self-renewal. We functionally validated these findings through selective editing of different PAF1 complex components in HSPCs: we observed reduced HSCs upon editing of PAF1 and CDC73, but increases with editing of other PAF1 complex components. Our findings collectively demonstrate a mechanism by which a previously undefined myeloid malignancy predisposition occurs. We demonstrate that CTR9 loss-of-function stimulates SEC activity and thereby results in HSC expansion to confer risk for acquiring MPNs and other myeloid malignancies. Disclosures Armstrong: Neomorph Inc: Consultancy, Current holder of individual stocks in a privately-held company; Imago Biosciences: Consultancy; Vitae/Allergan Pharma: Consultancy; Cyteir Therapeutics: Consultancy; C4 Therapeutics: Consultancy; OxStem Oncology: Consultancy; Accent Therapeutics: Consultancy; Mana Therapeutics: Consultancy; Janssen: Research Funding; Novartis: Research Funding; Syndax: Research Funding; AstraZeneca: Research Funding. Sankaran: Ensoma: Consultancy; Forma: Consultancy; Cellarity: Consultancy; Novartis: Consultancy; Branch Biosciences: Consultancy.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 386-386 ◽  
Author(s):  
Grant A. Challen ◽  
Deqiang Sun ◽  
Mira Jeong ◽  
Min Luo ◽  
Jaroslav Jelinek ◽  
...  

Abstract Abstract 386 Aberrant genomic DNA methylation patterns are widely reported in human cancers but the prognostic value and pathological consequences of these marks remain uncertain. CpG methylation is catalyzed by a family of DNA methyltransferase enzymes comprised of three members – Dnmt1, Dnmt3a and Dnmt3b. Mutations in the de novo DNA methyltransferase enzyme DNMT3A have now been reported in over 20% of adult acute myeloid leukemia (AML) and 10–15% of myelodysplastic syndrome (MDS) patients. However, analysis of promoter methylation and gene expression in these patients has thus far failed to yield any mechanistic insight into the pathology of DNMT3A mutation-driven leukemia. In this study, we have used a conditional knockout mouse model to study the role of Dnmt3a in normal hematopoiesis. Hematopoietic stem cells (HSCs) from Mx1-Cre:Dnmt3afl/fl mice were serially transplanted into lethally irradiated recipient mice to study the effect of loss of Dnmt3a on HSC self-renewal and differentiation. We show that loss of Dnmt3a progressively impedes HSC differentiation over four-rounds of serial transplantation, while simultaneously expanding HSC numbers in the bone marrow. Examination of the bone marrow post-transplant revealed that control HSCs showed a gradual decline in their ability to regenerate the HSC pool at each successive round of transplantation, while in contrast Dnmt3a-KO HSCs show a remarkably robust capacity for amplification, generating 40,000 – 100,000 HSCs per mouse. Quantification of peripheral blood differentiation on a per HSC basis demonstrated in the absence of Dnmt3a, a cell division is more likely to result in a self-renewal rather than differentiation fate (Figure 1). Using semi-global reduced representation bisulfite sequencing (RRBS), we show that Dnmt3a-KO HSCs manifest both increased and decreased methylation at distinct loci, including dramatic CpG island hypermethylation. Global transcriptional analysis by microarray revealed that Dnmt3a-KO HSCs show upregulation of HSC multipotency genes coupled with simultaneous downregulation of early differentiation factors (e.g. Flt3, PU.1, Mef2c), likely inhibiting the initial stages of HSC differentiation. Upregulation of key HSC regulators including Runx1, Gata3 and Nr4a2 was associated with gene-body hypomethylation and activated chromatin marks (H3K4me3) in Dnmt3a-KO HSCs. Finally, we show that Dnmt3a-KO HSCs are unable to methylate and transcriptionally repress these key HSC multipotency genes in response to chemotherapeutic ablation of the hematopoietic system, leading to inefficient differentiation and manifesting hypomethylation and incomplete repression of HSC-specific genes in their limited differentiated progeny. In conclusion, we show that Dnmt3a plays a specific role in permitting HSC differentiation, as in its absence, phenotypically normal but impotent stem cells accumulate and differentiation capacity is progressively lost. This differentiation-deficit phenotype is reminiscent of Dnmt3a/Dnmt3b-null embryonic stem (ES) cells while markedly distinct from that of Dnmt1-KO HSCs which show premature HSC exhaustion and lymphoid-deficient differentiation, demonstrating distinct roles for the different DNA methyltransferase enzymes in HSCs. In light of the recently-identified DNMT3A mutations in AML and MDS patients, these studies are the first biological models linking mutation of Dnmt3a with inhibition of HSC differentiation which may be one of the first pathogenic steps occuring in such patients.Figure 1Dnmt3a-KO HSCs become biased towards self-renewal as opposed to differentiation. At each transplant round, the self-renewal quotient was calculated as the number of donor-derived HSCs recovered at the end of the transplant divided by 250 (the number of HSC initially transplanted). The differentiation quotient was calculated as (the white blood cell count per μl of blood at 16 weeks) X (percentage of donor-cell chimerism)/number of donor HSC at the end of the transplant. Over serial transfer, Dnmt3a-KO HSCs more rapidly lose their differentiation capacity compared to control HSCs, while sustaining robust self-renewal.Figure 1. Dnmt3a-KO HSCs become biased towards self-renewal as opposed to differentiation. At each transplant round, the self-renewal quotient was calculated as the number of donor-derived HSCs recovered at the end of the transplant divided by 250 (the number of HSC initially transplanted). The differentiation quotient was calculated as (the white blood cell count per μl of blood at 16 weeks) X (percentage of donor-cell chimerism)/number of donor HSC at the end of the transplant. Over serial transfer, Dnmt3a-KO HSCs more rapidly lose their differentiation capacity compared to control HSCs, while sustaining robust self-renewal. Disclosures: Issa: Novartis: Honoraria; GSK: Consultancy; SYNDAX: Consultancy; Merck: Research Funding; Eisai: Research Funding; Celgene: Research Funding; Celgene: Honoraria; J&J: Honoraria.


2008 ◽  
Vol 28 (21) ◽  
pp. 6668-6680 ◽  
Author(s):  
Albertus T. J. Wierenga ◽  
Edo Vellenga ◽  
Jan Jacob Schuringa

ABSTRACT The level of transcription factor activity critically regulates cell fate decisions, such as hematopoietic stem cell (HSC) self-renewal and differentiation. We introduced STAT5A transcriptional activity into human HSCs/progenitor cells in a dose-dependent manner by overexpression of a tamoxifen-inducible STAT5A(1*6)-estrogen receptor fusion protein. Induction of STAT5A activity in CD34+ cells resulted in impaired myelopoiesis and induction of erythropoiesis, which was most pronounced at the highest STAT5A transactivation levels. In contrast, intermediate STAT5A activity levels resulted in the most pronounced proliferative advantage of CD34+ cells. This coincided with increased cobblestone area-forming cell and long-term-culture-initiating cell frequencies, which were predominantly elevated at intermediate STAT5A activity levels but not at high STAT5A levels. Self-renewal of progenitors was addressed by serial replating of CFU, and only progenitors containing intermediate STAT5A activity levels contained self-renewal capacity. By extensive gene expression profiling we could identify gene expression patterns of STAT5 target genes that predominantly associated with a self-renewal and long-term expansion phenotype versus those that identified a predominant differentiation phenotype.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1690-1690
Author(s):  
Rebecca J. Chan ◽  
Yanjun Li ◽  
Chris Shelley ◽  
Mervin C. Yoder

Abstract The protein tyrosine phosphatase, Shp-2, has been shown to be necessary for normal hematopoiesis based on embryonic stem (ES) cell-based assays; however, due to the early lethality of the homozygous Shp-2 mutant mice (Shp-2−/−) the role of Shp-2 in adult hematopoietic stem cell (HSC) function has never been examined. The Shp-2 heterozygous mice (Shp-2+/−) bear a mutant allele of the Shp-2 gene resulting in the production of a mutant protein lacking amino acids 46–110, which confers a loss of function. To test the hypothesis that Shp-2 is required for normal HSC activity, we compared the competitive repopulating ability of Shp-2+/− bone marrow-derived cells with WT cells. Total adult bone marrow low density mononuclear cells were isolated from Shp-2+/− and WT littermate controls (test cells, C57Bl/6 background, CD45.2+), mixed with a common pool of competitor (comp) cells (BoyJ background, CD45.1+), and administered to lethally irradiated (1100 cGy split dose) Gpi/BoyJ recipients. Based on peripheral blood chimerism, the repopulating ability of the Shp-2+/− cells was significantly lower than that of the WT cells (Figure 1, *p<0.0001 Shp-2+/− v. WT at ratio 1:2; **p=0.001 Shp-2+/− v. WT at ratio 1:1). We next converted the chimerism to repopulating units using the formula [competitor number x 105] X [% 45.2]/100 − [% 45.2] to quantitatively asses the repopulating defect in Shp-2+/− HSCs. We observed that the repopulating units of the Shp-2+/− cells was approximately 3-fold lower than that of the WT cells at both cell doses administered (Figure 2, *p=0.003 Shp-2+/− v. WT at ratio 1:2; **p=0.03 comparing Shp-2+/− v. WT at ratio 1:1). Multi-lineage analysis using two color fluorescence cytometry revealed a significantly lower contribution of Shp-2+/− cells to all lineages tested (B220, GR1, Mac, and CD4/8) compared to WT cells. As Shp-2 has been shown to participate in cell migration, we sought to rule out a homing deficiency of the Shp-2+/− HSCs. We performed short term homing assays and observed no difference in spleen-homed or bone marrow-homed Shp-2+/− and WT lin- cells twenty hours following transplantation. To evaluate self-renewal potential, we conducted serial transplantation experiments. Total bone marrow low density mononuclear cells were isolated from primary or seconary recipient mice with equal chimerism and transplanted into lethally irradiated (1100 cGy split dose) Gpi/BoyJ recipients. While no significant difference was observed between Shp-2+/− and WT engraftement in secondary transplants, eight weeks following tertiary transplantation, engraftment of the Shp-2+/− cells is significantly lower than that of the WT cells (WT 68.9% +/− 9.5 v. Shp-2+/− 26.1% +/− 11.7, n=6, p<0.0001) suggesting that a self-renewal defect contributes to the decreased HSC activity of the Shp-2+/− cells. These data demonstrate that Shp-2 function is not only necessary within the progenitor compartment to support proficient hematopoiesis, but is also needed within the HSC compartment to support normal HSC self-renewal. These findings provide insight into how oncogenic Shp-2 potentially may contribute to the dysregulation of hematopoiesis and the pathogenesis of childhood leukemias.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. SCI-32-SCI-32
Author(s):  
Kara A. Scheibner ◽  
Diane Heiser ◽  
Ian M Kaplan ◽  
Wen-Chih Cheng ◽  
MinJung Kim ◽  
...  

Abstract Abstract SCI-32 MicroRNAs (miRs) inhibit stability and/or translation of mRNAs, usually by binding to specific sites in the 32′UTRs of their target mRNAs. Due to imperfect (i.e. partially complementary) miR:mRNA base-pairing, miRs can block translation of many mRNAs and serve as powerful master switches to regulate cell functions. Therefore, we profiled miR expression in human CD34+ hematopoietic stem-progenitor cells (HSPCs) and combined human HSPC miR expression, mRNA expression, and miR-mRNA target predictions to hypothesize that certain HSPC-expressed miRs (HE-miRs) target several mRNAs critical to hematopoiesis. On this informatic basis, we formulated a model of hematopoietic differentation in which many genes specifying hematopoietic differentiation are expressed by early HSPCs, but held in check by miRs [1]. In addition, we noted that the miR-23a cluster (i.e. adjacent, co-transcribed miR-23a, miR-27a, and miR-24-2) is not expressed or is expressed at levels >2-fold lower in 50% of acute myeloid leukemias and 80% of acute lymphoid leukemias tested compared to normal human HSPCs. ‘Re-expressing’ 1 or more of these miR-23a cluster members in leukemia cells promotes their apoptosis and reduces their proliferation, thus suggesting that these miRs have a tumor suppressive role. We have identified YWHAQ (14-3-3q) and several other 14-3-3 isoforms, which are anti-apoptotic and have established roles as oncogenes, as miR-23a cluster target molecules. Artificial manipulation of these HE-miRs and their target genes may lead to novel strategies for leukemia treatment and/or for expansion of normal HSPCs. Since the CD34+ HSPCs that we studied initially include rare stem cells and various stages of progenitors, we have expanded our miR profiling to more highly purified subsets of mouse HSPCs. Several previously described (e.g. miR-155 [1], miR-451 [2], miR-146 [3]) and novel HE-miRs are expressed differentially in lineages/stages of HSPCs, and their selective expression has been confirmed in human HSPC subsets. We are using cellular gain- and loss-of-function approaches with hematopoietic functional assays to determine whether these HE-miRs control human hematopoiesis. Understanding the effects of HE-miRs in hematopoiesis may elucidate hematopoietic and general stem cell biologic mechanisms. 1. Georgantas RW, 3rd, Hildreth R, Morisot S, Alder J, Liu CG, Heimfeld S, Calin GA, Croce CM, Civin CI. CD34+ hematopoietic stem-progenitor cell microRNA expression and function. A circuit diagram of differentiation control. Proc Natl Acad Sci USA. 2007;104:2750–2755. 2. Dore LC, Amigo JD, Dos Santos CO, Zhang Z, Gai X, Tobias JW, Yu D, Klein AM, Dorman C, Wu W, Hardison RC, Paw BH, Weiss MJ. A GATA-1-regulated microRNA locus essential for erythropoiesis. Proc Natl Acad Sci USA. 2008;105:3333–3338. 3. Starczynowski DT, Kuchenbauer F, Argiropoulos B, Sung S, Morin R, Muranyi A, Hirst M, Hogge D, Marra M, Wells RA, Buckstein R, Lam W, Humphries RK, Karsan A. Identification of miR-145 and miR-146a as mediators of the 5q- syndrome phenotype. Nat Med. 2010;16:49–58. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1190-1190
Author(s):  
Wenhuo Hu ◽  
James Dooley ◽  
Stephen S. Chung ◽  
Safak Yalcin ◽  
Yu Sup Shin ◽  
...  

Abstract microRNAs (miRNAs) are important regulators of both embryonic and adult tissue stem cell self-renewal. We previously showed that ectopic expression of miR-29a, a miRNA highly expressed in HSCs as well as in human acute myeloid leukemia (AML) stem cells, in immature mouse hematopoietic cells is sufficient to induce a myeloproliferative disorder that progresses to AML. During the early phase of this disease, miR-29a induces aberrant self-renewal of committed myeloid progenitors, strongly suggesting a role for miR-29a in regulating HSC self-renewal. In order to determine the role of miR-29a in HSC function, we have evaluated our recently described miR-29a/b1 null mouse. Homozygous deletion of miR-29a/b1 resulted in reduced bone marrow cellularity and reduced colony forming capacity of hematopoietic stem and progenitor cells (HSPCs). The phenotype was mediated specifically by miR-29a since miR-29b expression was not significantly altered in HSCs and reconstitution of miR-29a/b1 null HSPCs with miR-29a, but not miR-29b, rescued in vitro colony formation defects. Self-renewal defects were observed in miR-29a deficient HSCs in both competitive and non-competitive transplantation assays, and these deficits were associated with increased HSC cell cycling and apoptosis. Gene expression studies of miR-29a deficient HSCs demonstrated widespread gene dysregulation including a number of up-regulated miR-29a target genes including DNA methylation enzymes (Dnmt3a, -3b) and cell cycle regulators (e.g. Cdk6, Tcl1, Hbp1, Pten). Knockdown of one of these targets, Dnmt3a, in miR-29a deficient HSCs resulted in partial restoration of colony formation, providing functional validation that Dnmt3a mediates part of miR-29a null HSPCs functional defects. miR-29a loss also abrogated leukemogenesis in the MLL-AF9 retroviral AML model. Together, our results demonstrate that miR-29a positively regulates HSC self-renewal and is required for myeloid leukemogenesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3754-3754
Author(s):  
Hee-Don Chae ◽  
Kathleen Sakamoto

Abstract CREB (cAMP Response Element Binding protein) promotes cellular transformation of hematopoietic cells and proliferation of myeloid leukemia cells. However, the underlying mechanisms of CREB function in leukemic transformation and hematopoiesis are not fully understood. To address this, we have investigated the downstream pathways of CREB activation in proliferation using a human acute myeloid leukemia (AML) cell line KG1 cells knocked-down for CREB with specific shRNAs. The CREB-knockdown KG1 cells were significantly defective in proliferative capability compared to control cells [cell number after 4d (X105), seeding (1X105), control vs. CREB-knockdown: 34.18 +/– 1.27 vs. 14.52 +/– 0.46, n=3, p< 0.01, mean +/– SEM]. In order to characterize the specific role of CREB in cell proliferation, we analyzed cell cycle progression patterns of CREB-knockdown and control KG1 cells after release from mitotic arrest. Our results indicated that G1 to S phase transition as assessed by % S phase was impeded by CREB-knockdown [S phase (%), control vs. CREB-knockdown cells, 8h after release: 53.29 +/– 0.54 vs. 23.57 +/– 1.69; 12h: 66.92 +/– 0.63 vs. 45.16 +/– 0.50, n=3, p< 0.01, mean +/– SEM]. To identify potential CREB target genes, we chose several cell cycle related genes such as CCNE1, CCNA1, CCNB1 and PCNA and compared their RNA expression levels in the CREB-knockdown with those in control KG1 cells after release from mitotic arrest. To our surprise, we failed to detect any noticeable differences in the mRNA expression levels of those genes between CREB-knockdown and control KG1 cells. In an effort to search for CREB responsive target genes, we analyzed additional CREB targets previously identified from microarray data (Pellegrini et al BMC Cancer 2008). We found that expression of replication factor C3 (RFC3), a 38kDa subunit of the RFC complex involved in DNA replication and repair processes, was significantly reduced in CREB-knockdown cells compared to control cells [38 +/– 1% of control, n=3, p<0.01]. CREB-knockdown also inhibited RFC3 mRNA expression in U937 and HL60 AML cell lines. Consistent with these results, mRNA expression levels of RFC3 appeared to be closely correlated with those of CREB when we examined bone marrow samples obtained from AML patients [n = 16, Pearson coefficient = 0.6366, p = 0.0008]. Moreover, we found that CREB directly interacted with the CRE site in the RFC3 promoter region in vivo, as assessed by chromatin immunoprecipitation assays. Exogenous overexpression of RFC3 in CREB-knockdown KG1 cells restored the defective G1/S progression [S phase (%), CREB-knockdown vs. CREB-knockdown with RFC3 overexpression, 9h after release: 38.97 +/– 0.45 vs. 62.24 +/– 1.06; 12h: 48.12 +/– 0.60 vs. 67.70 +/– 1.15, n=3, p< 0.01, mean +/– SEM]. Taken together, these results suggest that RFC3 may act as a novel downstream oncogenic target of activated CREB in AML cells. We previously reported that CREB is a critical regulator of normal myelopoiesis (Cheng et al Blood 2008). To determine whether RFC3 could exert similar effects on normal hematopoiesis, we compared human umbilical cord blood derived CD34-positive cells with and without RFC3 overexpression for the capacity to form hematopoietic colonies. Overexpression of RFC3 in the CD34-positive cells resulted in significant increases of multi-potential CFU-GEMM colony numbers [without vs. with overexpression of RFC3 (per 1000 cells): 3.2 +/– 1.3 vs. 22.3 +/– 3.3, n=3, p< 0.01, mean +/– SEM]. The RFC3 effect on stimulating colony formation was magnified in secondary colony forming assays [without vs. with overexpression of RFC3 (per 100,000 cells): 10.7 +/– 3.5 vs. 180.2 +/– 44.4, n=3, p< 0.05, mean +/– SEM]. Since the formation of secondary colonies was derived mainly from residual stem/progenitor cell populations after long-term culture, RFC3 overexpression may enhance self-renewal of stem/progenitor cells. In conclusion, our results suggest that RFC3 is able to promote G1/S transition in a human AML cell line downstream of CREB activation. In addition, we provide evidence that RFC3 is involved in normal hematopoiesis and contributes to increased self-renewal potential of hematopoietic stem/progenitor cells. Our data demonstrate that RFC3 plays multiple roles in promoting AML cells proliferation as well as normal myelopoiesis through increasing the self-renewal potential of hematopoietic stem/progenitor cells in response to CREB activation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 390-390 ◽  
Author(s):  
Michael J Dugan ◽  
Drew Winston ◽  
Michele I Morris ◽  
James E Williams ◽  
Natasha Broyde ◽  
...  

Abstract Introduction: Clostridium difficile-associated diarrhea (CDAD) is a common complication during HSCT that has been associated with increased morbidity and mortality. CDAD has been reported to occur in 6 to 30% of HSCT patients. An ideal agent for prevention of CDAD has not been established. Fidaxomicin (FDX) is bactericidal against C. difficile, has low systemic absorption, provides a prolonged post-antibiotic effect, and is currently approved for the treatment of CDAD. The efficacy and safety of low dose FDX (200mg once daily) for prevention of CDAD were evaluated in both autologous (AUTO) and allogenic (ALLO) HSCT patients receiving fluoroquinolone (FQ) prophylaxis. Methods: HSCT patients were randomized to receive FDX 200 mg once daily (QD) or placebo (PLA) from start of conditioning therapy or FQ prophylaxis until 7 days after post-transplant engraftment (ANC >500/mm3) or completion of FQ prophylaxis. Randomization was stratified by type of HSCT (AUTO vs. ALLO). Patients were contacted twice weekly for 30 days and then weekly through 60 days post-prophylaxis for symptoms; CDAD was confirmed by toxin testing or PCR. The incidence of CDAD from start of prophylaxis up to 30 days and 60 days post-treatment were primary and secondary endpoints, respectively. For the primary analysis, failures were defined as patients with confirmed CDAD, as well as those who had missing data for the primary endpoint assessment (due to death or discontinuations resulting from adverse events, protocol non-compliance, lost follow-up, or any other reasons) and those who received any CDAD-effective medication (without confirmed CDAD). Sensitivity analyses, including an analysis that defined failure as confirmed CDAD, were prospectively defined for the overall population. These same analyses were performed for the ALLO and AUTO subgroups. Results: Of 611 enrolled patients, 600 were evaluable: 352 (59%) in AUTO and 248 (41%) in ALLO subgroups. The primary analysis for the primary endpoint did not show a significant reduction in incidence of CDAD with FDX prophylaxis (28.6% FDX vs. 30.8% PLA, p = 0.28). Secondary endpoint results were similar. However, the incidence of confirmed CDAD for up to 30 days was significantly lower in AUTO, ALLO, and overall in HSCT patients receiving prophylactic FDX versus PLA (see table). There were similar trends for 60 day post-treatment results. Overall, serious adverse events (AEs) and AEs resulting in death were more frequent in the ALLO vs. AUTO subgroup. In both subgroups, no differences in drug-related serious and non-serious AEs were seen between FDX and PLA. Table. Incidence of Confirmed CDAD, 30d Post-treatment AUTO ALLO Total FDX (%) 5/176 (2.8%) 8/125 (6.4%) 13/301 (4.3%) PLA (%) 14/176 (8.0%) 18/123 (14.6%) 32/299 (10.7%) PLA-FDX, % 5.1 8.2 6.4 Wald p-value 0.0163 0.0166 0.0014 Incidence of Confirmed CDAD, 60d Post-treatment FDX (%) 6/176 (3.4%) 11/125 (8.8%) 17/301 (5.6%) PLA (%) 14/176 (8.0%) 18/123 (14.6%) 32/299 (10.7%) PLA-FDX, % 4.5 5.8 5.1 Wald p-value 0.0321 0.0759 0.0117 Conclusions: For the primary endpoint (failure of prophylaxis due to confirmed CDAD or non-CDAD events), the efficacy of FDX and PLA were similar. However, in both AUTO and ALLO HSCT patients, prophylactic FDX significantly reduced the incidence of confirmed CDAD and was not associated with drug-related adverse events. Disclosures Dugan: Cubist/Merck & Co., Inc.: Research Funding. Winston:Cubist/Merck & Co., Inc.: Research Funding. Morris:Cubist/ Merck & Co., Inc.: Research Funding. Williams:Merck & Co., Inc.: Employment. Broyde:Merck & Co., Inc.: Employment. Sears:Merck & Co., Inc.: Employment.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 543-543 ◽  
Author(s):  
Maurice Labuhn ◽  
Kelly Perkins ◽  
Elli Papaemmanuil ◽  
Catherine Garnett ◽  
Soeren Matzk ◽  
...  

Abstract Myeloid leukemia of Down syndrome (ML-DS) is a tractable human model of acute myeloid leukemia. A preleukemia phase, transient abnormal myelopoiesis (TAM) and silent TAM, occurs in 28% of neonates with Down Syndrome (Roberts et al. Blood 2013). TAM is caused by trisomy 21 and acquired mutations in GATA1 that result in a N-terminal truncated protein, GATA1s, in hematopoietic stem and progenitor cells (HSPCs) of fetal origin. ML-DS evolves from TAM by acquisition of additional genetic lesions. The nature of these lesions and the mechanism of transformation are incompletely understood. We performed exome sequencing and targeted resequencing of 141 ML-DS and 111 TAM patients to characterize the evolving mutational landscape from TAM to ML-DS. On average 1.6 acquired mutations were detected in ML-DS (in addition to GATA1 mutations), significantly more than in TAM (0.4 mutations per sample). Additional anticipated loss-of-function mutations acquired in ML-DS mainly affected cohesin components including CTCF (43% of patients), PRC2 components (13%), KANSL1 and other epigenetic regulators (14%). Conversely, anticipated gain-of-function mutations were most prevalent in signaling pathways, e.g. JAK kinases, MPL, KIT and RAS family members (40%). Importantly, we detected a novel recurrent hotspot mutation in 4% of patients (6/141 cases) in CSF2RB encoding the IL3-, IL5-, GM-CSF-receptor common beta chain. To test if the A455D/T variant in the CSF2RB transmembrane domain is a putative oncogenic driver, we ectopically expressed CSF2RBA455D in TF1 cells. Cells expressing CSF2RBA455D exhibited cytokine independent growth and STAT5 autonomous phosphorylation. In a CD34+-HSPC megakaryocytic differentiation assay, CSF2RBA455D blocked terminal megakaryocytic differentiation whilst increasing proliferation by 30-fold (P=0.046). Moreover, the median survival of NSG mice transplanted with CSF2RBA455DTF1 cells was shortened by 30 days compared to wild type TF1 cells (23 days compared to 53 days, P=0.0097). To experimentally test the potential of loss-of-function mutations to transform TAM to ML-DS, we performed an in vivo murine isogenic transplantation screen using Gata1s expressing fetal hematopoietic cells from Cas9-knockin mice. We tested variants in 22 genes, recurrently detected in ML-DS, with a pool of prevalidated gRNAs. This resulted in short latency (n=18 mice; median survival 36 days) and high penetrance (100%) leukemia. Leukemia was not detected in mice infected with control gRNAs. Leukemias had a typical ML-DS megakaryoblastic phenotype (CD117+ and CD41a+). Amplicon sequencing revealed on average 2.9 gRNAs per leukemia and high representation (61% of all leukemias) of gRNAs directed to the tumor suppressor Trp53, which was alone sufficient to induce leukemia with 100% penetrance. When excluding the Trp53 gRNA from pools, leukemic cells from moribund mice contained gRNAs against negative regulators of the RAS and JAK-STAT signaling cascade, such as Nf1, Cbl and Sh2b3 (70% of the mice), Ezh2, Asxl1, Kdm6a,Bcor and other epigenetic modifiers (85%) or Ctcf (15%), closely resembling the mutational landscape of ML-DS. In contrast to ML-DS, gRNAs targeting cohesion components, such as Rad21 and Stag2, were not present in any of the leukemias. In summary, we performed the largest genetic analysis of transforming events in ML-DS that cooperate with trisomy 21 and GATA1s and uncovered a previously undescribed activating mutation in CSR2B. We experimentally validated many of the loss-of-function mutations in a novel murine fetal leukemia assay for ML-DS. The field is now well-placed to study mechanisms of oncogenic cooperativity and identify novel therapeutic approaches for this leukemia. Disclosures Crispino: Scholar Rock: Research Funding; Forma Therapeutics: Research Funding.


2018 ◽  
Author(s):  
Satish K Nandakumar ◽  
Sean K McFarland ◽  
Laura Marlene Mateyka ◽  
Caleb A Lareau ◽  
Jacob C Ulirsch ◽  
...  

Genome-wide association studies (GWAS) have identified thousands of variants associated with human diseases and traits. However, the majority of GWAS-implicated variants are in non-coding genomic regions and require in depth follow-up to identify target genes and decipher biological mechanisms. Here, rather than focusing on causal variants, we have undertaken a pooled loss-of-function screen in primary hematopoietic cells to interrogate 389 candidate genes contained in 75 loci associated with red blood cell traits. Using this approach, we identify 77 genes at 38 GWAS loci, with most loci harboring 1-2 candidate genes. Importantly, the hit set was strongly enriched for genes validated through orthogonal genetic approaches. Genes identified by this approach are enriched in relevant biological pathways, allowing regulators of human erythropoiesis and blood disease modifiers to be defined. More generally, this functional screen provides a paradigm for gene-centric follow up of GWAS for a variety of human diseases and traits.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1391-1391
Author(s):  
Costakis Frangou ◽  
Jason Den Haese ◽  
Jordan Warunek ◽  
Scott Portwood ◽  
Norma J Nowak ◽  
...  

Abstract Chemotherapy or targeted cancer therapies have greatly improved the treatment outcome of patients with leukemia; however, many will ultimately die because of disease relapse and development of drug resistance. Leukemias are cancers of the blood cells that result from alteration of the normal physiological constraints that regulate hematopoietic stem cells (HSCs). General characteristics of leukemia stem cells (LSCs) such as self-renewal, self-protection and proliferative quiescence represent inherent mechanisms that at least partially explain drug resistance and recurrence in post-therapy leukemia patients. Acute myeloid leukemia (AML) is a heterogeneous disease, both biologically and clinically, in which a number of distinct genetic abnormalities have been described. Several recent studies suggest that this heterogeneity extends to LSCs and can vary between patient subgroups, and even within individual patients. Moreover, the complexity of AML is further complicated by the existence of functionally diverse leukemic and preleukemic clones. Accordingly, the hierarchical organization of AML suggests that this may be relevant to current therapies that primarily target proliferating progenitors/blast cells, which lack self-renewal capacity, and not LSCs. In the current study, we rationalized that understanding how LSCs differ from normal HSCs at the molecular level, is an essential first step towards developing novel targeted therapies and achieving permanent disease remission. Despite the identification of novel LSC-specific markers, there is considerable heterogeneity in expression of these markers amongst AML patients. However, in addition to marker-enrichment strategies, LSCs can be identified by virtue of their quiescent and slow-cycling properties. For example, label-retaining cells can be isolated and used in functional assays but significant technical limitations impede broad utility of this approach. To this end, we describe the development and use of novel multi-fluorescent protein markers and DNA bar codes integrated into the cellular genomes by lentivirus, as single-cell tracking devices for monitoring LSCs in vivo. We demonstrate how LSCs can transition between a "proliferation phase" and a "quiescence phase" in vivo. Furthermore, using high-throughput quantitative transcriptome sequencing (Q-RNA-Seq) and RNAi genetic perturbation's focusing on well-defined self-renewal signaling pathways, we develop a differential network-based model to identify LSC-specific genes and subsequently prioritize/rank candidates as potential drug targets. In the current study, we identify several molecular targets deregulated in quiescent versus proliferating LSCs and a mutual set of signaling pathways that facilitate leukemic transformation downstream of diverse initiating mutations/lesions. Remarkably, both quiescent and dividing LSCs but not HSCs, were 'addicted' to SSRP1 - an essential component of the ubiquitous FACT chromatin remodeling complex. Two orally available quinacrine-related DNA-intercalating compounds inhibiting function of FACT (CBL0100 and CBL0175, respectively) suppressed LSC proliferation in vitro and in vivo, as demonstrated by production of leukemic clonogenic cells (CFU) and long-term engraftment of immunodeficient NSG mice, by simultaneous inhibition of NF-kB (stimulated and basal forms) and activation of p53. Furthermore, in a secondary transplantation experiment, leukemic cells obtained from CBL0175 treated mice (primary) failed to engraft into secondary NSG mice in a serial transplantation model by selectively targeting the LSC compartment. Collectively, we present a novel network-based polypharmacology approach that provides unique opportunities to preferentially ablate LSCs (quiescent and dividing types), with potentially profound clinical implications. Disclosures Frangou: Cellecta: Employment. Portwood:ImmunoGen: Research Funding. Wang:ImmunoGen: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document