Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2021

Open Access 01.12.2021 | Research

Treatment-induced arteriolar revascularization and miR-126 enhancement in bone marrow niche protect leukemic stem cells in AML

verfasst von: Bin Zhang, Le Xuan Truong Nguyen, Dandan Zhao, David E. Frankhouser, Huafeng Wang, Dinh Hoa Hoang, Junjing Qiao, Christina Abundis, Matthew Brehove, Yu-Lin Su, Yuxin Feng, Anthony Stein, Lucy Ghoda, Adrianne Dorrance, Danilo Perrotti, Zhen Chen, Anjia Han, Flavia Pichiorri, Jie Jin, Tijana Jovanovic-Talisman, Michael A. Caligiuri, Calvin J. Kuo, Akihiko Yoshimura, Ling Li, Russell C. Rockne, Marcin Kortylewski, Yi Zheng, Nadia Carlesso, Ya-Huei Kuo, Guido Marcucci

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2021

Abstract

Background

During acute myeloid leukemia (AML) growth, the bone marrow (BM) niche acquires significant vascular changes that can be offset by therapeutic blast cytoreduction. The molecular mechanisms of this vascular plasticity remain to be fully elucidated. Herein, we report on the changes that occur in the vascular compartment of the FLT3-ITD+ AML BM niche pre and post treatment and their impact on leukemic stem cells (LSCs).

Methods

BM vasculature was evaluated in FLT3-ITD+ AML models (MllPTD/WT/Flt3ITD/ITD mouse and patient-derived xenograft) by 3D confocal imaging of long bones, calvarium vascular permeability assays, and flow cytometry analysis. Cytokine levels were measured by Luminex assay and miR-126 levels evaluated by Q-RT-PCR and miRNA staining. Wild-type (wt) and MllPTD/WT/Flt3ITD/ITD mice with endothelial cell (EC) miR-126 knockout or overexpression served as controls. The impact of treatment-induced BM vascular changes on LSC activity was evaluated by secondary transplantation of BM cells after administration of tyrosine kinase inhibitors (TKIs) to MllPTD/WT/Flt3ITD/ITD mice with/without either EC miR-126 KO or co-treatment with tumor necrosis factor alpha (TNFα) or anti-miR-126 miRisten.

Results

In the normal BM niche, CD31+Sca-1high ECs lining arterioles have miR-126 levels higher than CD31+Sca-1low ECs lining sinusoids. We noted that during FLT3-ITD+ AML growth, the BM niche lost arterioles and gained sinusoids. These changes were mediated by TNFα, a cytokine produced by AML blasts, which induced EC miR-126 downregulation and caused depletion of CD31+Sca-1high ECs and gain in CD31+Sca-1low ECs. Loss of miR-126high ECs led to a decreased EC miR-126 supply to LSCs, which then entered the cell cycle and promoted leukemia growth. Accordingly, antileukemic treatment with TKI decreased the BM blast-produced TNFα and increased miR-126high ECs and the EC miR-126 supply to LSCs. High miR-126 levels safeguarded LSCs, as shown by more severe disease in secondary transplanted mice. Conversely, EC miR-126 deprivation via genetic or pharmacological EC miR-126 knock-down prevented treatment-induced BM miR-126high EC expansion and in turn LSC protection.

Conclusions

Treatment-induced CD31+Sca-1high EC re-vascularization of the leukemic BM niche may represent a LSC extrinsic mechanism of treatment resistance that can be overcome with therapeutic EC miR-126 deprivation.

Graphic abstract

Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13045-021-01133-y.
Bin Zhang, Le Xuan Truong Nguyen, and Dandan Zhao have contributed equally

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AML
Acute myeloid leukemia
BM
Bone marrow
HSC
Hematopoietic stem cell
LSC
Leukemic stem cell
EC
Endothelial cell
TNFα
Tumor necrosis factor alpha
PB
Peripheral blood
MNC
Mononuclear cell
COHNMC
City of Hope National Medical Center
FLT3
FMS-like tyrosine kinase 3
ITD
Internal tandem duplication
TK
Tyrosine kinase
TKI
Tyrosine kinase inhibitor
wt
Wild type
CB
Cord blood
PDX
Patient-derived xenograft
LSK
LinSca-1c-Kit
LSK
LinSca-1+c-Kit+
LT-HSC
Long-term hematopoietic stem cell
mr
Murine recombinant
Ab
Antibody
OB
Osteoblast
MSP
Mesenchymal stromal progenitor
KO
Knockout
KD
Knock-down
HUVEC
Human umbilical vein endothelial cells
2nd
Secondary
Ang-1
Angiopoietin-1

Background

Acute myeloid leukemia (AML) is a hematopoietic malignancy characterized by somatic mutations occurring in the hematopoietic stem cells (HSCs) and progenitor cells that block hematopoietic differentiation and promote accumulation of leukemic “blasts” in the bone marrow (BM) and/or other extramedullary organs [1]. To date, despite a deep molecular understanding of the pathogenesis, the development of molecular targeting therapeutics and the broadened use of allogeneic HSC transplantation, the overall outcome of AML patients remains poor. Disease refractoriness to initial therapy or post-remission disease relapse [2] are likely due to persistence of treatment-resistant leukemic stem cells (LSCs) [3]. These are primitive leukemic cells capable of unlimited self-renewal and disease initiation [4, 5] and reside in a leukemic BM niche that also comprises several types of non-hematopoietic cells and that preferentially supports homeostasis and competitive growth of LSCs over those of HSCs [6, 7].
Mechanisms of treatment resistance in cancer are multifaceted and often result from the acquisition of genetic mutations that enable malignant cells to escape the therapeutic pressure. Recently, other non-genetic mechanisms of treatment resistance have been also described [8]. While these reported mechanisms have been mainly reported as intrinsic to malignant cells, it is possible that they also include mechanisms that are extrinsic to malignant cells, such as those involving the microenvironment and that protect malignant cells during treatment exposure [7].
Utilizing the FMS-like tyrosine kinase 3 (FLT3) gene internal tandem duplication (ITD) (FLT3-ITD) knock-in mouse and FLT3-ITD+ AML patient-derived xenograft (PDX) models that recapitulate features of human FLT3-ITD+ AML, we report here on previously unrecognized non-genetic, extrinsic mechanisms of treatment resistance in LSCs that involve the vascular compartment of the leukemic BM niche and that are mediated by a TNFα-miR-126 axis in the BM endothelial cells (ECs). FLT3-ITD occurs in approximately 25% of AML patients and the mutated gene encodes a mutant receptor with aberrant, ligand-independent tyrosine kinase (TK) activity that confers growth and survival advantages to leukemic blasts [9]. FLT3-ITD+ AML patients are treated with TK inhibitors (TKIs) in combination with chemotherapy. Although the addition of TKIs to chemotherapy confers a clinical advantage compared to chemotherapy alone, it is not curative in the majority of cases, suggesting treatment resistance arising over time [10].

Methods

An extended description of the methods is in the Additional file 1.

Human samples

Normal peripheral blood (PB) and BM samples were obtained from healthy donors at the City of Hope National Medical Center (COHNMC). AML samples were obtained from patients from the COHNMC (Additional file 1: Table S1). Mononuclear cells (MNCs) were isolated using Ficoll separation. When necessary, CD34+ cells were then isolated using a positive magnetic bead selection protocol (Miltenyi Biotech, Germany). Sample acquisition was approved by the Institutional Review Boards at the COHNMC, in accordance with an assurance filed with and approved by the Department of Health and Human Services and met all requirements of the Declaration of Helsinki. Healthy donors and AML patients were consented on the IRB # 06229 and IRB# 18067 protocols, respectively.

Animal studies

The MllPTD/WT/Flt3ITD/ITD mouse, an AML model in C57Bl/6J (B6) background (CD45.2), was generated and genotyped as previously described [11]. The MllPTD/WT/Flt3ITD/ITD mice (CD45.2, B6) were also bred with CD45.1 B6 mice to produce CD45.1/CD45.2 MllPTD/WT/Flt3ITD/ITD mice as donors for transplant experiments. To obtain conditional miR-126 knock-out (KO) or Spred1 KO in ECs, we bred miR-126flox/flox (CD45.2 B6) [12] and Spred1flox/flox (CD45.2 B6) [13] mice with Tie2-cre (CD45.2 B6, from The Jackson Laboratory, 008863) mice and obtained miR-126flox/floxTie2-cre+ (EC miR-126 KO, also called miR-126ECΔ/Δ) and Spred1flox/floxTie2-cre+ (EC Spred1 KO, also called Spred1ECΔ/Δ, representing a functional EC miR-126 overexpressing model) mice. Tie2-CreER/TdTomato/Tg(Ly6a-GFP) double fluorescent EC/Sca-1 reporter mice were generated by crossing Tie2-CreER/TdTomato mice (CD45.2 B6), in which BM ECs bearing the Tie2-promoter driven TdTomato color upon tamoxifen administration (EC-tdTomato+, EC reporter mice) [14, 15], with Tg(Ly6a-EGFP) (Sca-1-GFP+, Sca-1 reporter mice, from The Jackson Laboratory, 012643), which allowed us to visualize Sca-1high EC (tdTomato+GFPhigh) and Sca-1low EC (tdTomato+GFPlow) lined vessels based on the combined expression of these two endogenous reporters. To obtain conditional EC miR-126 KO reporter mice, we also bred Tie2-CreER/TdTomato/Tg(Ly6a-GFP) double fluorescent reporter mice with miR-126flox/flox mice and obtained inducible EC miR-126 KO reporter mice [i.e., miR-126flox/flox/Tie2-CreER/TdTomato/Tg(Ly6a-GFP), miR-126 KO in ECs upon tamoxifen administration].
To evaluate the impact of EC miR-126 on leukemia-induced vascular changes and in turn on LSC burden, BM MNCs from AML (i.e., MllPTD/WT/Flt3ITD/ITD, CD45.1/CD45.2 B6) or from normal wild-type (wt, CD45.1 B6) mice were transplanted into Cre+ or Cre- miR-126flox/flox/Tie2-cre, Spred1flox/flox/Tie2-cre, and tamoxifen-treated miR-126flox/flox/Tie2-CreER/TdTomato/Tg(Ly6a-GFP) reporter mice (all CD45.2 B6). To study the vascular changes induced by human FLT3-ITD+ AML blast growth, NOD.Cg-Prkdcscid Il2rgtm1Wjl Tg(CMV-IL3, CSF2, KITLG)1Eav/MloySzJ mice (NSG-SGM3 or NSGS, from The Jackson Laboratory, 013062) were transplanted with normal cord blood (CB) CD34+ cells or with human FLT3-ITD+ AML blasts (patient-derived xenograft, PDX).

Immunofluorescent staining and 3D confocal imaging of long bones

Long bones (femurs and/or tibias) from the mice were processed, sectioned and imaged as described previously [16] with ad-hoc modifications (see Additional file 1 for details).

Intravital imaging

Intravital confocal microscopy was used to image the calvarium BM vasculature to study the vascular permeability, as previously described [17] (see Additional file 1 for details).

Statistical analysis

Comparison between groups was performed by two-tailed, paired or unpaired Student's t-test, adjusting for multiple comparisons as appropriate. The log-rank test was used to assess significant differences between survival curves. All statistical analyses were performed using Prism version 8.0 software (GraphPad Software). Sample sizes chosen are indicated in the individual figure legends and were not based on formal power calculations to detect prespecified effect sizes but were based on previous experience with similar models. All of the in vitro experiments were performed 3–6 times using biologically independent samples; the in vivo experiments were performed using 6–16 mice in each group. p values < 0.05 were considered significant. Results shown represent mean ± SEM. ∗p ≤ 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001.

Results

Bone marrow vasculature of normal and leukemic mice

To determine how the vascular compartment of the BM niche adapts to leukemia growth and subsequently to antileukemic treatments, we performed immunofluorescence staining and 3D confocal imaging of vessels in the long bones of normal versus (vs) leukemic mice. To this end, we first analyzed normal mice and identified CD31+α-SMA+Sca-1highEndomucin(Emcn) vessels as arteries (Additional file 1: Fig. S1a–c, blue arrow), CD31+α-SMASca-1highEmcn vessels as arterioles (Additional file 1: Fig. S1a–e, yellow arrow), and CD31+α-SMASca-1lowEmcn+ vessels as sinusoids (Additional file 1: Fig. S1a–e, white arrow) [1821]. In a double fluorescent reporter mouse [i.e., tamoxifen-treated Tie2-CreER/TdTomato/Tg(Ly6a-GFP)], Sca-1high EC (i.e., tdTomato+GFPhigh) and Sca-1low EC (i.e., tdTomato+GFPlow) lined vessels were also morphologically consistent respectively with arteries and arterioles (both are tdTomato+GFPhigh but with different size; Additional file 1: Fig. S1f, blue arrow indicates artery and yellow arrows indicate arteriole) and sinusoids (Additional file 1: Fig. S1f, white arrow). Since Sca-1high expression appeared to be restricted to arteries and arterioles, we then utilized a simplified CD31+Sca-1high and CD31+Sca-1low staining along with morphology examination to mark respectively arterioles and sinusoids in the BM niche [19, 22].
To study changes of the vascular compartment of the BM niche during leukemia growth, we then utilized the MllPTD/WT/Flt3ITD/ITD mouse, a model that recapitulates phenotypic, cytogenetic, molecular and pathological features of human FLT3-ITD+ AML, a relatively frequent molecular subset of the disease [11]. Of note, we will refer hereafter to “wt” or “MllPTD/WT/Flt3ITD/ITD” to indicate the mouse genotype and to “normal” or “leukemic” to indicate the disease status.
Firstly, we noticed a significant decrease in CD31+Sca-1high EC lined vessels (i.e., arterioles) in the leukemic MllPTD/WT/Flt3ITD/ITD mice compared with normal wt mice (Fig. 1a; Additional file 1: Fig. S2a). These results were corroborated by a flow cytometry analysis showing an overall increase in total BM ECs (CD45Ter119CD31+) in the leukemic mice (Additional file 1: Fig. S2b, c) but with a lower frequency of CD31+Sca-1high ECs and a higher frequency of CD31+Sca-1low ECs compared with normal wt mice (Fig. 1b; Additional file 1: Fig. S2d). Similar results were also obtained when normal wt mice were engrafted with BM MNCs from congenic leukemic MllPTD/WT/Flt3ITD/ITD donors and compared with controls engrafted with BM MNCs from congenic normal wt donors (Fig. 1c, d; Additional file 1: Fig. S2e).
To validate these findings, we then transplanted BM MNCs from leukemic MllPTD/WT/Flt3ITD/ITD mice or from normal wt mice into the tamoxifen-induced Tie2-CreER/TdTomato/Tg(Ly6a-GFP) double reporter mice. At 4 weeks after transplantation, we observed reduced Sca-1high EC-lined vessels (i.e., tdTomato+ GFPhigh) in the recipients of AML BM MNCs compared with recipients of normal BM MNCs (Fig. 1e; Additional file 1: Fig. S2f). Consistent with these results, flow cytometry showed reduced Sca-1high ECs (i.e., tdTomato+ GFPhigh) and increased Sca-1low ECs (i.e., tdTomato+ GFPlow) in the double reporter mice engrafted with AML BM MNCs compared with those engrafted with normal BM MNCs (Fig. 1f; Additional file 1: Fig. S2g).
Next, to assess the relevance of these changes to the human disease, we transplanted human primary FLT3-ITD+ AML blasts into NSG-SGM3 (NSGS) mice and generated a patient-derived xenograft (PDX) model. Similar to the murine AML models, the FLT3-ITD+ PDX showed fewer BM CD31+Sca1high ECs and arterioles as compared with NSGS mice engrafted with normal CB CD34+ cells (Fig. 1g, h; Additional file 1: Fig. S3).
In the BM niche, CD31+Sca-1high ECs reportedly line impermeable vessels such as arteries and arterioles, and CD31+Sca-1low ECs border permeable vessels such as sinusoids [21]. To this end, we imaged the calvarium of normal Sca-1 reporter [i.e., Tg(Ly6a-GFP)] mice with intravital confocal microscopy. Prior to imaging, mice were administered TRITC-dextran (150 kDa, red) intravenously to identify the vasculature [23, 24]. As expected, dextran leakage was visible as diffuse staining preferentially around Sca-1-GFPlow vessels, rather than Sca-1-GFPhigh vessels (Additional file 1: Fig. S4). Consistent with a decrease in CD31+Sca-1high vessels (i.e., arterioles) (Fig. 1a, g), we observed increased vessel permeability in the MllPTD/WT/Flt3ITD/ITD AML mouse (TRITC-150 kDa dextran, red) and FLT3-ITD+ PDX (FITC-150 kDa dextran, green) (Additional file 1: Fig. S5a, b). Similar results were obtained when we transplanted BM MNCs from leukemic MllPTD/WT/Flt3ITD/ITD mice or from normal wt mice into wt (FITC-150 kDa dextran, green; Additional file 1: Fig. S5c), EC reporter (i.e., tamoxifen-treated Tie2-CreER/TdTomato; FITC-150 kDa dextran, green; Additional file 1: Fig. S5d), or EC/Sca-1 double reporter [i.e., tamoxifen-treated Tie2-CreER/TdTomato/Tg(Ly6a-EGFP); Alex647-150 kDa dextran, blue; Additional file 1: Fig. S5e] recipient mice.
Thus, using different AML models and imaging techniques, flow cytometric analyses and permeability studies of the BM niche, we showed that FLT3-ITD+ AML growth led to a decrease of CD31+Sca-1high vessels (i.e., arterioles) in the BM niche.

TNFα mediates loss of CD31+Sca-1high vessels in the leukemic BM niche

To gain insights into the mechanisms leading to CD31+Sca-1high EC and arteriole depletion in the BM niche during leukemia growth, we hypothesized that these effects could be associated with certain secretory features of the proliferating leukemic blasts. We therefore measured levels of cytokines and chemokines in the blood and BM of age- and gender-matched leukemic MllPTD/WT/Flt3ITD/ITD and normal wt mice. TNFα was the only cytokine significantly elevated in the BM of leukemic mice (Fig. 2a, left; Additional file 1: Fig. S6a–m). Higher TNFα mRNA levels were observed in myeloid cells and CD45+Lin progenitors [i.e., LinSca-1c-Kit (LSK) and LinSca-1+c-Kit+ (LSK)] from the leukemic MllPTD/WT/Flt3ITD/ITD mice compared to the counterparts from normal wt mice (Fig. 2a, right), suggesting an overproduction of this cytokine by the clonal myeloid subpopulations in the leukemic mice. In vitro treatment of BM ECs from normal wt mice with murine recombinant (mr) TNFα (mrTNFα: 1 ng/ml) for 96 h (h) recapitulated the observations in the leukemic mice, with an expansion of ECs (Fig. 2b), a decrease of CD31+Sca-1high EC subfraction and an increase of CD31+Sca-1low EC subfraction (Fig. 2c; Additional file 1: Fig. S7a). Both TNFα receptor type 1 and 2 (TNFαR1 and TNFαR2) were found to be co-expressed on the surface of CD31+Sca-1high ECs (Additional file 1: Fig. S7b) and TNFαR1 and TNFαR2 blocking antibodies (Abs) reversed the mrTNFα effect (Additional file 1: Fig. S7c).
Consistent with these results, normal wt mice given mrTNFα [1 µg/day, intraperitoneal (ip) injection, 3 weeks] [25, 26] showed a significant increase of BM ECs (Fig. 2d), and loss of CD31+Sca-1high ECs and arterioles compared with vehicle-treated controls (Fig. 2e, f). Accordingly, normal wt mice engrafted with BM from leukemic MllPTD/WT/Flt3ITD/ITD donors had higher levels of BM TNFα (Additional file 1: Fig. S7d) and loss of CD31+Sca-1high ECs and arterioles (Fig. 1c, d; Additional file 1: Fig. S2e), compared with mice engrafted with BM from normal wt donors. Similar results were obtained in NSGS mice engrafted with human FLT3-ITD+ AML blasts compared with NSGS mice engrafted with normal CB CD34+ cells (Fig. 1g, h; Additional file 1: Fig. S3, S7e, f). In vivo treatment of MllPTD/WT/Flt3ITD/ITD AML mice with TNFα-neutralizing Ab (anti-TNFα; MP6-XT22, Biolegend; 1 mg/day, ip, 4 times/week for 3 weeks) [27] rescued the loss of CD31+Sca-1high ECs and arterioles (Fig. 2g, h) otherwise observed in the leukemic BM niche.
Taken altogether, these results support a TNFα-induced depletion of CD31+Sca-1high vessels in the leukemic BM niche during FLT3-ITD+ AML growth.

TNFα depletes CD31+Sca-1high ECs via miR-126 downregulation

The vascular changes induced by TNFα in BM niche were strikingly similar to what we observed in the mice with miR-126 KO in ECs (miR-126flox/floxTie2-cre+, hereafter called miR-126ECΔ/Δ) obtained by crossing miR-126flox/flox with Tie2-cre mice. This led us to hypothesize that TNFα induces vascular changes during leukemia growth partly via miR-126 downregulation. To this end, it has been reported that in mature ECs, miR-126 contributes to the maintenance of vascular integrity and inhibition of endothelial permeability, which are main features of arterial and arteriolar vessels [12, 28, 29]. In the normal mouse BM niche, we found that ECs expressed at least a log-fold higher level of miR-126 than normal long-term (LT) HSCs (LSK Fit3CD150+CD48) and other non-hematopoietic stromal cells, including osteoblasts (OBs; CD45Ter119CD31CD166+Sca-1) and mesenchymal stromal progenitors (MSPs; CD45Ter119CD31CD166Sca-1+) [30] (Additional file 1: Fig. S8a, b). Within the ECs, Sca-1high ECs expressed significantly higher levels of miR-126 (Additional file 1: Fig. S8c, d) and lower levels of miR-126 verified targets (i.e., Vcam1 and Spred1) [12, 28, 29] (Additional file 1: Fig. S8e) than Sca-1low ECs. In addition, transcriptomes of Sca-1high and Sca-1low ECs from normal wt mice showed different gene expression profiles (Additional file 1: Fig. S8f), with pathways regulating angiogenesis, cell migration and vasculature development being activated in Sca-1high ECs (Additional file 1: Table S2). Q-RT-PCR assays (Additional file 1: Table S3) validated these findings (Additional file 1: Fig. S8g, right panel).
Interestingly, we noticed that miR-126 knock-down (KD) by miRZip anti-miR-126 lentiviral transduction (Fig. 3a, Additional file 1: Fig. S9a) or treatment with a CpG-miR-126 inhibitor [31] (an anti-miR-126 oligonucleotide hereafter called miRisten; Additional file 1: Fig. S9b, c) depleted the Sca-1high fraction in CD31+Sca-1high ECs from normal wt mice. Conversely, miR-126 over-expression (OE) in CD31+Sca-1low ECs obtained by transduction with miR-126 precursor lentivirus (Fig. 3b) or treatment with CpG-miR-126 mimics (Additional file 1: Fig. S9d, e) enriched the Sca-1high fraction. In vivo, mice with EC miR-126 KO (i.e., miR-126ECΔ/Δ; Fig. 3c, middle box) [12, 31] had fewer CD31+Sca-1high ECs and arterioles than miR-126flox/floxTie2-cre- controls (Fig. 3d–g, middle panel). Fluorescent reporter mice with tamoxifen-induced miR-126 KO in ECs [i.e., tamoxifen-treated miR-126flox/flox/Tie2-CreER/TdTomato/Tg(Ly6a-GFP)] also showed fewer Sca-1high ECs (i.e., tdTomato+GFPhigh) and arterioles than wt reporter mice [i.e., tamoxifen-treated miR-126wt/wt/Tie2-CreER/TdTomato/Tg(Ly6a-GFP)] (Additional file 1: Fig. S9f, g). Furthermore, normal wt mice treated in vivo with miRisten (20 mg/kg/day, iv) for 3 weeks showed fewer CD31+Sca-1high ECs and vessels (i.e., arterioles) than control mice treated with CpG-scramble (SCR) (Fig. 3h, i). In addition, similar to the leukemic mice, miR-126ECΔ/Δ mice had an increase of the BM vessel permeability compared to wt controls (Additional file 1: Fig. S9h).
We previously reported that Spred1, a member of the Sprouty family of proteins and an inhibitor of RAS small GTPases, is both a miR-126 target and a regulator of miR-126 biogenesis [31]. EC Spred1 KO mice (i.e., Spred1f/fTie2-cre+, hereafter called Spred1ECΔ/Δ) [13, 32, 33] therefore express constitutively higher levels of EC miR-126 than Spred1f/fTie2-cre- and miR-126ECΔ/Δ mice (Fig. 3c, right box) and represent a functional model for EC miR-126 OE. Accordingly, Spred1ECΔ/Δ mice had more BM CD31+Sca-1high ECs and vessels (i.e., arterioles) than wt controls (Fig. 3d–g, right panel).
Taken altogether, these results support a role of miR-126 in determining enrichment of the Sca-1high EC fraction and in turn arteriolar density in the BM niche and a role of TNFα-dependent miR-126 downregulation in the loss of arterioles during leukemia growth.
To this end, we also observed significantly lower BM EC miR-126 levels in the leukemic MllPTD/WT/Flt3ITD/ITD mice (Fig. 4a) in addition to fewer BM CD31+Sca-1high ECs and vessels (i.e., arterioles; Fig. 1a, b; Additional file 1: Fig. S2a) compared with normal wt controls. Similar changes were observed in normal wt mice engrafted with AML BM MNCs compared with those engrafted with normal BM MNCs (Figs. 1c, d and 4b; Additional file 1: Fig. S2e). Furthermore, murine BM ECs obtained from normal wt mice treated in vitro with mrTNFα (1 ng/ml, 24 h) showed reduced levels of pri/pre- and mature miR-126 (Fig. 4c) and increased levels of miR-126 targets (i.e., Vcam1 and Spred1) (Fig. 4d). Co-treatment with miR-126 mimics rescued the loss of the CD31+Sca-1high fraction induced by mrTNFα (Additional file 1: Fig. S10a, b, right panel). TNFαR1 and TNFαR2 blocking Abs also rescued the mrTNFα-induced miR-126 downregulation (Additional file 1: Fig. S10c) and the reduction of the CD31+Sca-1high EC fraction (Additional file 1: Fig. S7c).
Consistent with the in vitro results, normal wt mice that were given mrTNFα (1 µg/day, ip injection, 3 weeks) [25, 26] had a significant in vivo reduction of BM EC pri/pre- and mature miR-126 levels (Fig. 4e). As expected, these mice also showed a decrease in CD31+Sca-1high ECs and vessels (i.e., arterioles) compared with vehicle-treated controls (Fig. 4f, middle panel; Additional file 1: Fig. S10d, e, middle panel). In vivo co-treatment with miR-126 mimics (20 mg/kg/day, iv. 3 weeks) rescued these changes (Fig. 4f, bottom panel; Additional file 1: Fig. S10d, e, right panel). Accordingly, normal wt mice engrafted with BM MNCs from MllPTD/WT/Flt3ITD/ITD AML donors had higher levels of BM TNFα (Additional file 1: Fig. S7d) and reduced levels of BM EC miR-126 (Additional file 1: Fig. S10f), in addition to a decrease in CD31+Sca-1high ECs and arterioles (Fig. 1c, d; Additional file 1: Fig. S2e), compared with mice engrafted with BM MNCs from normal wt donors. These changes were rescued in vivo by blocking TNFα with anti-TNFα Ab (1 mg/day, ip, 4 times/week for 3 weeks) (Additional file 1: Fig. S10g–i). Similar results were observed in NSGS mice engrafted with human FLT3-ITD+ AML blasts compared with those engrafted with normal CB CD34+ cells (Fig. 1g, h; Additional file 1: Fig. S3, S7e and S10j).
Of note, Spred1ECΔ/Δ mice (EC miR-126 OE, Fig. 3c) treated in vivo with mrTNFα (1 µg/day, ip, 3 weeks) showed no significant difference in vascularization compared with vehicle-treated Spred1ECΔ/Δ controls (Additional file 1: Fig. S10k–l). Transplantation of AML BM MNCs resulted in a lesser degree of leukemia-induced BM vascular changes in Spred1ECΔ/Δ recipients compared with wt recipients (Fig. 4g, right panel; Additional file 1: Fig. S10m, right panel), suggesting that constitutively higher expression of miR-126, as occurred in ECs from Spred1ECΔ/Δ mice, may rescue in vivo TNFα-induced miR-126 depletion and loss of CD31+Sca-1high ECs.
While the molecular mechanisms through which TNFα induces endothelial miR-126 downregulation are likely to be multifaceted, we focused on GATA2 since this protein is a verified miR-126 transcription factor [3436] and is reportedly downregulated by TNFα [34, 37]. We first noticed that Gata2 levels were reduced in BM ECs from AML mice compared with normal mice (Additional file 1: Fig. S11a) and that GATA2 KD by siRNA in human umbilical vein ECs (HUVECs) decreased miR-126 levels (Additional file 1: Fig. S11b, c). We then demonstrated that in vitro, TNFα treatment (1 ng/ml) reduced Gata2 levels in both murine BM ECs and HUVECs (Additional file 1: Fig. S11d–f) as compared with vehicle-treated controls. Using chromatin immunoprecipitation assay, we showed a reduced GATA2 enrichment on the EGFL7/miR-126 promoter [34] in HUVECs exposed to TNFα (Additional file 1: Fig. S11g, h).
Taken altogether, these results support the notion that loss of CD31+Sca-1high ECs and associated vessels (i.e., arterioles) observed during AML growth is at least partly mediated by EC miR-126 downregulation via TNFα-dependent decrease of GATA2 transcriptional activity.

Antileukemic treatment restores CD31+Sca-1high vessels that safeguard LSCs

Having demonstrated that TNFα secreted by the AML blasts contributes to loss of CD31+Sca-1high ECs, we then reasoned that cytoreductive therapy that eliminates TNFα-secreting blasts could reverse the depletion of CD31+Sca-1high EC-lined vessels (i.e., arterioles) as observed in the leukemic BM niche. To this end, we transplanted BM MNCs from MllPTD/wt/Flt3ITD/ITD AML mice (CD45.2) into congenic B6 mice (CD45.1) and generated a cohort of AML mice that developed disease at a similar time. We elected to treat these mice with TKIs rather than chemotherapy to restrict the observed BM niche changes to a direct blast cytoreduction rather than to non-specific chemotherapy cytotoxicity to other non-hematopoietic cells.
Upon confirmation of AML development at 4 weeks after transplantation, we treated these mice with the TKI AC220 [38] to target FLT3-ITD and cytoreduce AML blasts (20 mg/kg/day, oral gavage, Fig. 5a). After 3 weeks treatment, we observed leukemic cytoreduction (Fig. 5b) and decreased BM TNFα levels (Fig. 5c) in AC220-treated AML mice compared with vehicle-treated controls. In AC220-treated mice, we noticed a gain in BM CD31+Sca-1high ECs and vessels (i.e., arterioles) (Fig. 5d, e), an increase of Gata2 levels in ECs (Fig. 5f) but not in LSKs (Additional file 1: Fig. S12a), an increase of miR-126 levels in both ECs and LSCs (i.e., AML LSK; Fig. 5g, Additional file 1: Fig. S12b), and a higher frequency of quiescent LSK cells (LSKG0, Fig. 5h; Additional file 1: Fig. S12c) compared with vehicle-treated controls.
In MllPTD/WT/Flt3ITD/ITD AML mice, BM LSKs are the subpopulation mostly enriched in leukemia-initiating cells (LICs; hereafter called LSCs) as shown by limiting-dilution transplantation of immunophenotypically defined BM cell subpopulations (Additional file 1: Fig. S12d). To this end, we showed that while AC220 effectively cytoreduced AML blasts and increased survival of the primary treated mice (Fig. 5i), it also unexpectedly increased LSC burden and/or activity. In fact, in secondary (2nd) transplant experiments, recipients of BM MNCs from AC220-treated donors had a higher disease burden (Additional file 1: Fig. S12e) and shorter survival (Fig. 5j) than recipients of BM MNCs from vehicle-treated donors, indicating not only persistence but also an expansion of LSCs.
In contrast, AC220-treated mice that also received mrTNFα (1 µg/day, ip, 3 weeks) to prevent the treatment-related TNFα decrease (Fig. 6a) maintained lower levels of BM EC miR-126 (Additional file 1: Fig. S12f) and had fewer CD31+Sca-1high ECs and arterioles (Fig. 6b, c) and less LSCs (i.e., CD45.1/CD45.2 AML LSKs; Fig. 6d; Additional file 1: Fig. S12b) than mice that received AC220 alone. In 2nd transplant experiments, the recipients of BM MNCs from AC220+ mrTNFα-treated donors had a significantly longer survival compared with the recipients of BM MNCs from AC220-treated donors (Fig. 6e, right; median survival: 84 vs. 24 days, p < 0.0001), supporting the hypothesis that prevention of treatment-related CD31+Sca-1high EC enrichment and arteriolar re-vascularization prevents LSC expansion. We confirmed the relevance of these results to human disease using the FLT3-ITD+ PDX model (Additional file 1: Fig. S12g–k).
Next, as TNFα acts on the vasculature of the BM niche via miR-126, we reasoned that a direct EC miR-126 deprivation of the BM niche could prevent treatment-induced CD31+Sca-1high EC enrichment, arteriolar re-vascularization and LSC protection. To this end, we transplanted BM MNCs from MllPTD/wt/Flt3ITD/ITD AML mice into miR-126ECwt/wt and miR-126ECΔ/Δ recipient mice and treated them with AC220 (20 mg/kg/day, oral gavage; Fig. 6f). After 3 weeks treatment, we observed reduced CD31+Sca-1high ECs and arterioles (Additional file 1: Fig. S13a–d), a significant reduction of leukemia burden and LSCs, reduced miR-126 levels in AML LSCs (Fig. 6g) likely due to reduced EC supply, and increased survival (Fig. 6h; left) in the AC220-treated miR-126ECΔ/Δ primary mice compared with the AC220-treated miR-126ECwt/wt controls. BM MNCs from AC220-treated miR-126ECΔ/Δ or miR-126ECwt/wt AML donors were then transplanted into 2nd wt recipient mice. Recipients of BM MNCs from the AC220-treated miR-126ECΔ/Δ donors had a significantly longer survival than recipients of BM MNCs from the AC220-treated miR-126ECwt/wt donors (Fig. 6h, right; median survival: 53 days vs. not reached at day 80 post transplantation, p < 0.0001).
Similar results were obtained by pharmacologic miR-126 downregulation with miRisten. A cohort of MllPTD/WT/Flt3ITD/ITD leukemic mice were generated as described above and then treated with miRisten (20 mg/kg, iv, daily), SCR, miRisten+ AC220 (10 mg/kg, oral gavage, daily), or SCR+ AC220 for 3 weeks, followed by assessment of donor AML cell engraftment in PB, BM and spleen (Additional file 1: Fig. S14a). We confirmed miR-126 KD in ECs from miRisten-treated mice compared with SCR-treated mice (Additional file 1: Fig. S14b). Mice receiving the combination of miRisten+ AC220 had a significant reduction in the percentage of AML cells in PB, BM and spleen (Additional file 1: Fig. S14c, d), a significant decrease in the frequency of BM AML LSKs (Additional file 1: Fig. S14e) and increased survival (Additional file 1: Fig. S14f) compared to mice treated with SCR+ AC220. Similar to the mrTNFα+ AC220 combination, the miRisten+ AC220 combination also significantly prolonged survival in 2nd transplant experiments (Additional file 1: Fig. S14g; median survival: 37 days vs. not reached at day 65 post transplantation, p < 0.0001), suggesting reduced post-treatment LSC burden.
These results were corroborated in NSGS mice transplanted with FLT3-ITD+ AML blasts and then treated with miRisten+ AC220 or SCR+ AC220 for 3 weeks (Additional file 1: Fig. S15a). Treatment with miRisten+ AC220 significantly reduced human CD45+CD33+ cell engraftment (Additional file 1: Fig. S15b) and prolonged survival (Additional file 1: Fig. S15c, left) in the primary treated mice. Recipient mice receiving BM cells from miRisten+ AC220-treated donors lived significantly longer than the mice receiving BM cells from SCR+ AC220-treated donors (Additional file 1: Fig. S15c, right; median survival: 167 vs. 126 days, p < 0.0001), suggesting that the combination of miRisten+ AC220 had decreased the LSC burden.

Discussion

AML cells reportedly produce inflammatory cytokines that profoundly remodel the BM vascular niche and create a microenvironment supportive of competitive leukemia growth over normal hematopoiesis [31, 39, 40]. However, how the leukemic BM niche adapts to the changing conditions caused by treatment and ultimately impacts on clinical outcome remains to be fully elucidated. Herein, we utilized FLT3-ITD+ AML murine and PDX models to study the vascular changes of the leukemic BM niche that occur upon molecular targeting of the AML blasts. During AML growth, in the leukemic BM niche, we observed a loss in CD31+Sca-1high ECs, which line mainly non-permeable arterioles, and a gain in CD31+Sca-1low ECs, which line mainly fenestrated, permeable sinusoids. These vascular changes were caused partly by high levels of TNFα produced by the AML blasts, causing downregulation of miR-126 in CD31+Sca-1high ECs, which became depleted while an enrichment in CD31+Sca-1low ECs was observed.
TNFα has been intensively studied for its role in normal and malignant hematopoiesis [41]. Yamashita and Passegue recently reported on the complex role of TNFα in normal and clonal hematopoiesis, showing that while inducing myeloid progenitor apoptosis, TNFα promotes HSC survival [42]. AML blasts express high levels of TNFα [43], and likely hijack TNFα-driven mechanisms of normal hematopoiesis to support leukemia growth [42]. Herein, we show a novel pro-leukemogenic role of TNFα that is extrinsic to AML cells and involves downregulation of miR-126 in the vascular compartment of the BM niche. While TNFα has been implicated in the remodeling of blood vessels and shown to promote angiogenesis during inflammation [44, 45], to our knowledge the TNFα-induced switch from Sca-1high ECs to Sca-1low ECs, and in turn from an arteriole- to a sinusoid-enriched BM niche as observed during leukemia growth has not been previously reported. As these changes were a phenocopy of genetic (EC miR-126 KO) and pharmacologic (i.e., miRisten) EC miR-126 deprivation, we postulated and proved that loss of CD31+Sca-1high ECs and arterioles were due to TNFα-induced miR-126 downregulation.
MiR-126 is one of the most highly expressed microRNAs in ECs, where it acts as a master regulator of angiogenesis [12, 28, 29]. In developmental vasculogenesis, miR-126 supports differentiation of embryonic stem cells into endothelial precursor cells and mature ECs [46]. In mature ECs, miR-126 contributes to the maintenance of quiescence and vascular integrity and inhibition of endothelial permeability and apoptosis [12, 28, 29]. Furthermore, miR-126 enhances the activity of angiopoietin-1 (Ang-1), a glycoprotein that regulates vessel stabilization, maturation and permeability [47, 48]. Of note, the molecular mechanisms through which TNFα induces endothelial miR-126 downregulation are likely to be multifaceted and remain to be fully elucidated. Herein, we showed that they likely involve GATA2 [34, 35], a miR-126 transcription factor, which is reportedly silenced by TNFα [37]. But how do the TNFα-induced vascular changes in the leukemic BM niche ultimately promote leukemia growth? We previously showed that CD31+Sca-1high ECs expressed the highest levels of miR-126 in the BM niche and supply miR-126 to maintain LSC quiescence [31]. Thus, by inducing loss of CD31+Sca-1high ECs, TNFα decreases the endothelial supply of miR-126 to LSCs and enables them to enter the cell cycle and partially differentiate into proliferating “bulk” AML blasts [31, 49].
Of note, these observations may have direct translational and clinical relevance. In fact, under these conditions, therapeutic cytoreduction of AML blasts can cause a drop in the BM levels of TNFα and in turn lead to a post-treatment enrichment of BM CD31+Sca-1high ECs and arterioles with a consequent increase in the endothelial miR-126 supply to LSCs, which, once enriched in miR-126, are more resistant to therapy[31, 49]. We proved this model in FLT3-ITD+ AML murine and PDX mice treated with TKIs. We showed that after TKI treatment, an increase in CD31+Sca-1high ECs and arterioles occurred and LSCs persisted as demonstrated using secondary transplant experiments. Of note, LSC persistence could be prevented by blocking the gain in CD31+Sca-1high ECs and the arteriolar “re-vascularization” of the BM niche with administration of recombinant TNFα or with deprivation of endothelial miR-126 via genetic EC miR-126 KO or pharmacological treatment with the anti-miR-126 miRisten.
In comparing our results with recent reports on the BM vascular changes occurring during AML growth and treatment, we found interesting similarities between these studies and our work [17, 39]. While Passaro et al. showed that engraftment of AML blasts in mouse BM increased CD31+Sca-1+ ECs [17], in line with our results, Duarte et al. reported that AML growth caused loss of CD31+Sca-1+ ECs in the BM niche [39]. Similar to our observation, both studies demonstrated an increase in BM vascular permeability during leukemia growth, which associates with CD31+Sca-1low vessels. While these studies showed that the post-treatment changes rescued AML-induced BM permeability and enhanced sensitivity of the proliferating blasts to chemotherapy, they did not test the effects of these changes on LSC activity as done in our study. To our knowledge, we are the first to show that, in the BM niche, therapeutic cytoreduction of proliferating blasts decreases TNFα levels and leads to enrichment in CD31+Sca-1high arterioles that safeguard LSCs. Of note, using PDX model transplanted with human FLT3-ITD+ AML cells, Maifrede et al. showed that AC220 (10 mg/kg/day, 7 days) prolonged survival of primary treated mice, but did not change survival of 2nd recipient mice [50]. Herein, using FLT3-ITD+ AML murine model, we showed that, treatment with AC220 (20 mg/kg/day, 21 days) prolonged survival of primary treated mice and resulted in a more severe disease in 2nd recipient mice (i.e., shorter survival). These results are not in conflict as in both studies, TKI treatment failed to reduce LSC burden. The difference in survival of secondary transplanted mice observed in these two studies could be related to technical differences including the used models and the schedule (7 vs. 21 days) and dosage (10 vs. 20 mg/kg/day) of the drug administration.
Thus, taken altogether, these results provide the evidence for non-genetic, LSC extrinsic mechanisms of treatment resistance that depend on the vascular plasticity of the leukemic BM niche and likely exemplify a treatment-related Janus phenomenon in AML [51]. Janus was an ancient Roman god with two faces looking in opposite directions. The two opposing “faces” of the TKI treatments in FLT3-ITD+ AML models as described here are the initially beneficial cytoreduction of proliferating blasts followed by post-treatment CD31+Sca-1high EC gain and arteriolar re-vascularization of the BM niche that safeguard LSCs thereby creating the condition for disease relapse. Of note, it is likely that the “Janus” phenomenon that we report here for TKIs may also apply to other antileukemic therapies that target proliferating blasts but do not kill LSCs. To this end, prevention of post-treatment CD31+Sca-1high EC expansion, arteriolar re-vascularization and LSC protection is achievable with pharmacological deprivation of BM endothelial miR-126 (i.e., miRisten), which may represent a novel strategy to overcome non-genetically driven, extrinsic mechanisms of LSC resistance in AML.

Conclusions

The TNFα-miR-126 axis plays a key role in the BM vascular changes induced by antileukemic treatments and mediates non-genetic, extrinsic mechanisms of LSC treatment-resistance that can be overcome with pre-emptive therapeutic deprivation of EC miR-126.

Acknowledgements

We are grateful to Marjorie Robbins and David Alexander for editing the manuscript. We acknowledge the support of the Animal Resources Center, Analytical Cytometry, Pathology (Hematopoietic Tissue Biorepository), Bioinformatics, Light Microscopy, Integrative Genomics and DNA/RNA Cores at City of Hope Comprehensive Cancer Center supported by the National Cancer Institute of the National Institutes of Health under award number P30CA33572. We are grateful to COH Comprehensive Cancer Center, the patients and their physicians for providing primary patient material for this study.

Declarations

Patient sample acquisition was approved by the Institutional Review Boards (IRB) at the COHNMC, in accordance with an assurance filed with and approved by the Department of Health and Human Services and met all requirements of the Declaration of Helsinki. CML patients were consented on the IRB #18067 protocol.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Khwaja A, Bjorkholm M, Gale RE, Levine RL, Jordan CT, Ehninger G, et al. Acute myeloid leukaemia. Nat Rev Dis Primers. 2016;2:16010.PubMedCrossRef Khwaja A, Bjorkholm M, Gale RE, Levine RL, Jordan CT, Ehninger G, et al. Acute myeloid leukaemia. Nat Rev Dis Primers. 2016;2:16010.PubMedCrossRef
2.
Zurück zum Zitat SEER Cancer Statistics Review (CSR) 1975–2017, updated April, 2020 [Internet]. National Cancer Institute, DCCPS, Surveillance Research Program, Surveillance Systems Branch. 2020. Available from: https://seer.cancer.gov/. SEER Cancer Statistics Review (CSR) 1975–2017, updated April, 2020 [Internet]. National Cancer Institute, DCCPS, Surveillance Research Program, Surveillance Systems Branch. 2020. Available from: https://​seer.​cancer.​gov/​.
3.
Zurück zum Zitat Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3(7):730–7.PubMedCrossRef Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3(7):730–7.PubMedCrossRef
5.
Zurück zum Zitat Shafat MS, Gnaneswaran B, Bowles KM, Rushworth SA. The bone marrow microenvironment—home of the leukemic blasts. Blood Rev. 2017;31(5):277–86.PubMedCrossRef Shafat MS, Gnaneswaran B, Bowles KM, Rushworth SA. The bone marrow microenvironment—home of the leukemic blasts. Blood Rev. 2017;31(5):277–86.PubMedCrossRef
6.
Zurück zum Zitat Mendez-Ferrer S, Bonnet D, Steensma DP, Hasserjian RP, Ghobrial IM, Gribben JG, et al. Bone marrow niches in haematological malignancies. Nat Rev Cancer. 2020;20(5):285–98.PubMedCrossRef Mendez-Ferrer S, Bonnet D, Steensma DP, Hasserjian RP, Ghobrial IM, Gribben JG, et al. Bone marrow niches in haematological malignancies. Nat Rev Cancer. 2020;20(5):285–98.PubMedCrossRef
7.
Zurück zum Zitat Zhang B, Ho YW, Huang Q, Maeda T, Lin A, Lee SU, et al. Altered microenvironmental regulation of leukemic and normal stem cells in chronic myelogenous leukemia. Cancer Cell. 2012;21(4):577–92.PubMedPubMedCentralCrossRef Zhang B, Ho YW, Huang Q, Maeda T, Lin A, Lee SU, et al. Altered microenvironmental regulation of leukemic and normal stem cells in chronic myelogenous leukemia. Cancer Cell. 2012;21(4):577–92.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Marine JC, Dawson SJ, Dawson MA. Non-genetic mechanisms of therapeutic resistance in cancer. Nat Rev Cancer. 2020;20(12):743–56.PubMedCrossRef Marine JC, Dawson SJ, Dawson MA. Non-genetic mechanisms of therapeutic resistance in cancer. Nat Rev Cancer. 2020;20(12):743–56.PubMedCrossRef
9.
Zurück zum Zitat Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–47.PubMedPubMedCentralCrossRef Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–47.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454–64.PubMedPubMedCentralCrossRef Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454–64.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Zorko NA, Bernot KM, Whitman SP, Siebenaler RF, Ahmed EH, Marcucci GG, et al. Mll partial tandem duplication and Flt3 internal tandem duplication in a double knock-in mouse recapitulates features of counterpart human acute myeloid leukemias. Blood. 2012;120(5):1130–6.PubMedPubMedCentralCrossRef Zorko NA, Bernot KM, Whitman SP, Siebenaler RF, Ahmed EH, Marcucci GG, et al. Mll partial tandem duplication and Flt3 internal tandem duplication in a double knock-in mouse recapitulates features of counterpart human acute myeloid leukemias. Blood. 2012;120(5):1130–6.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Kuhnert F, Mancuso MR, Hampton J, Stankunas K, Asano T, Chen CZ, et al. Attribution of vascular phenotypes of the murine Egfl7 locus to the microRNA miR-126. Development. 2008;135(24):3989–93.PubMedCrossRef Kuhnert F, Mancuso MR, Hampton J, Stankunas K, Asano T, Chen CZ, et al. Attribution of vascular phenotypes of the murine Egfl7 locus to the microRNA miR-126. Development. 2008;135(24):3989–93.PubMedCrossRef
13.
Zurück zum Zitat Inoue H, Kato R, Fukuyama S, Nonami A, Taniguchi K, Matsumoto K, et al. Spred-1 negatively regulates allergen-induced airway eosinophilia and hyperresponsiveness. J Exp Med. 2005;201(1):73–82.PubMedPubMedCentralCrossRef Inoue H, Kato R, Fukuyama S, Nonami A, Taniguchi K, Matsumoto K, et al. Spred-1 negatively regulates allergen-induced airway eosinophilia and hyperresponsiveness. J Exp Med. 2005;201(1):73–82.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Hochstetler CL, Feng Y, Sacma M, Davis AK, Rao M, Kuan CY, et al. KRas(G12D) expression in the bone marrow vascular niche affects hematopoiesis with inflammatory signals. Exp Hematol. 2019;79:3–15.PubMedPubMedCentralCrossRef Hochstetler CL, Feng Y, Sacma M, Davis AK, Rao M, Kuan CY, et al. KRas(G12D) expression in the bone marrow vascular niche affects hematopoiesis with inflammatory signals. Exp Hematol. 2019;79:3–15.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Feng Y, Liu M, Guo F, Liu W, Sampson L, You L-R, et al. Novel method to study mouse bone marrow endothelial cells in vivo and in vitro. Blood. 2012;120(21):617.CrossRef Feng Y, Liu M, Guo F, Liu W, Sampson L, You L-R, et al. Novel method to study mouse bone marrow endothelial cells in vivo and in vitro. Blood. 2012;120(21):617.CrossRef
16.
Zurück zum Zitat Kusumbe AP, Ramasamy SK, Starsichova A, Adams RH. Sample preparation for high-resolution 3D confocal imaging of mouse skeletal tissue. Nat Protoc. 2015;10(12):1904–14.PubMedCrossRef Kusumbe AP, Ramasamy SK, Starsichova A, Adams RH. Sample preparation for high-resolution 3D confocal imaging of mouse skeletal tissue. Nat Protoc. 2015;10(12):1904–14.PubMedCrossRef
17.
Zurück zum Zitat Passaro D, Di Tullio A, Abarrategi A, Rouault-Pierre K, Foster K, Ariza-McNaughton L, et al. Increased vascular permeability in the bone marrow microenvironment contributes to disease progression and drug response in acute myeloid leukemia. Cancer Cell. 2017;32(3):324–41.PubMedPubMedCentralCrossRef Passaro D, Di Tullio A, Abarrategi A, Rouault-Pierre K, Foster K, Ariza-McNaughton L, et al. Increased vascular permeability in the bone marrow microenvironment contributes to disease progression and drug response in acute myeloid leukemia. Cancer Cell. 2017;32(3):324–41.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Hooper AT, Butler JM, Nolan DJ, Kranz A, Iida K, Kobayashi M, et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell. 2009;4(3):263–74.PubMedPubMedCentralCrossRef Hooper AT, Butler JM, Nolan DJ, Kranz A, Iida K, Kobayashi M, et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell. 2009;4(3):263–74.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Kunisaki Y, Bruns I, Scheiermann C, Ahmed J, Pinho S, Zhang D, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502(7473):637–43.PubMedPubMedCentralCrossRef Kunisaki Y, Bruns I, Scheiermann C, Ahmed J, Pinho S, Zhang D, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502(7473):637–43.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Kusumbe AP, Ramasamy SK, Adams RH. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature. 2014;507(7492):323–8.PubMedPubMedCentralCrossRef Kusumbe AP, Ramasamy SK, Adams RH. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature. 2014;507(7492):323–8.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Itkin T, Gur-Cohen S, Spencer JA, Schajnovitz A, Ramasamy SK, Kusumbe AP, et al. Distinct bone marrow blood vessels differentially regulate haematopoiesis. Nature. 2016;532(7599):323–8.PubMedPubMedCentralCrossRef Itkin T, Gur-Cohen S, Spencer JA, Schajnovitz A, Ramasamy SK, Kusumbe AP, et al. Distinct bone marrow blood vessels differentially regulate haematopoiesis. Nature. 2016;532(7599):323–8.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Egawa G, Nakamizo S, Natsuaki Y, Doi H, Miyachi Y, Kabashima K. Intravital analysis of vascular permeability in mice using two-photon microscopy. Sci Rep. 2013;3:1932.PubMedPubMedCentralCrossRef Egawa G, Nakamizo S, Natsuaki Y, Doi H, Miyachi Y, Kabashima K. Intravital analysis of vascular permeability in mice using two-photon microscopy. Sci Rep. 2013;3:1932.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Biesmans S, Bouwknecht JA, Ver Donck L, Langlois X, Acton PD, De Haes P, et al. Peripheral administration of tumor necrosis factor-alpha induces neuroinflammation and sickness but not depressive-like behavior in mice. Biomed Res Int. 2015;2015:716920.PubMedPubMedCentralCrossRef Biesmans S, Bouwknecht JA, Ver Donck L, Langlois X, Acton PD, De Haes P, et al. Peripheral administration of tumor necrosis factor-alpha induces neuroinflammation and sickness but not depressive-like behavior in mice. Biomed Res Int. 2015;2015:716920.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Lee LF, Lih CJ, Huang CJ, Cao T, Cohen SN, McDevitt HO. Genomic expression profiling of TNF-alpha-treated BDC2.5 diabetogenic CD4+ T cells. Proc Natl Acad Sci USA. 2008;105(29):10107–12.PubMedPubMedCentralCrossRef Lee LF, Lih CJ, Huang CJ, Cao T, Cohen SN, McDevitt HO. Genomic expression profiling of TNF-alpha-treated BDC2.5 diabetogenic CD4+ T cells. Proc Natl Acad Sci USA. 2008;105(29):10107–12.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Via CS, Shustov A, Rus V, Lang T, Nguyen P, Finkelman FD. In vivo neutralization of TNF-alpha promotes humoral autoimmunity by preventing the induction of CTL. J Immunol. 2001;167(12):6821–6.PubMedCrossRef Via CS, Shustov A, Rus V, Lang T, Nguyen P, Finkelman FD. In vivo neutralization of TNF-alpha promotes humoral autoimmunity by preventing the induction of CTL. J Immunol. 2001;167(12):6821–6.PubMedCrossRef
28.
Zurück zum Zitat Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, et al. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15(2):261–71.PubMedPubMedCentralCrossRef Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, et al. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15(2):261–71.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, et al. miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 2008;15(2):272–84.PubMedPubMedCentralCrossRef Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, et al. miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 2008;15(2):272–84.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Hu X, Garcia M, Weng L, Jung X, Murakami JL, Kumar B, et al. Identification of a common mesenchymal stromal progenitor for the adult haematopoietic niche. Nat Commun. 2016;7:13095.PubMedPubMedCentralCrossRef Hu X, Garcia M, Weng L, Jung X, Murakami JL, Kumar B, et al. Identification of a common mesenchymal stromal progenitor for the adult haematopoietic niche. Nat Commun. 2016;7:13095.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Zhang B, Nguyen LXT, Li L, Zhao D, Kumar B, Wu H, et al. Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia. Nat Med. 2018;24(4):450–62.PubMedPubMedCentralCrossRef Zhang B, Nguyen LXT, Li L, Zhao D, Kumar B, Wu H, et al. Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia. Nat Med. 2018;24(4):450–62.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Ishizaki T, Tamiya T, Taniguchi K, Morita R, Kato R, Okamoto F, et al. miR126 positively regulates mast cell proliferation and cytokine production through suppressing Spred1. Genes Cells. 2011;16(7):803–14.PubMedCrossRef Ishizaki T, Tamiya T, Taniguchi K, Morita R, Kato R, Okamoto F, et al. miR126 positively regulates mast cell proliferation and cytokine production through suppressing Spred1. Genes Cells. 2011;16(7):803–14.PubMedCrossRef
33.
Zurück zum Zitat Tadokoro Y, Hoshii T, Yamazaki S, Eto K, Ema H, Kobayashi M, et al. Spred1 safeguards hematopoietic homeostasis against diet-induced systemic stress. Cell Stem Cell. 2018;22(5):713–25.PubMedCrossRef Tadokoro Y, Hoshii T, Yamazaki S, Eto K, Ema H, Kobayashi M, et al. Spred1 safeguards hematopoietic homeostasis against diet-induced systemic stress. Cell Stem Cell. 2018;22(5):713–25.PubMedCrossRef
34.
Zurück zum Zitat Hartmann D, Fiedler J, Sonnenschein K, Just A, Pfanne A, Zimmer K, et al. MicroRNA-based therapy of GATA2-deficient vascular disease. Circulation. 2016;134(24):1973–90.PubMedCrossRef Hartmann D, Fiedler J, Sonnenschein K, Just A, Pfanne A, Zimmer K, et al. MicroRNA-based therapy of GATA2-deficient vascular disease. Circulation. 2016;134(24):1973–90.PubMedCrossRef
35.
Zurück zum Zitat Le Bras A, Samson C, Trentini M, Caetano B, Lelievre E, Mattot V, et al. VE-statin/egfl7 expression in endothelial cells is regulated by a distal enhancer and a proximal promoter under the direct control of Erg and GATA-2. PLoS ONE. 2010;5(8):e12156.PubMedPubMedCentralCrossRef Le Bras A, Samson C, Trentini M, Caetano B, Lelievre E, Mattot V, et al. VE-statin/egfl7 expression in endothelial cells is regulated by a distal enhancer and a proximal promoter under the direct control of Erg and GATA-2. PLoS ONE. 2010;5(8):e12156.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Mahamud MR, Geng X, Ho YC, Cha B, Kim Y, Ma J, et al. GATA2 controls lymphatic endothelial cell junctional integrity and lymphovenous valve morphogenesis through miR-126. Development. 2019;146(21):1039. Mahamud MR, Geng X, Ho YC, Cha B, Kim Y, Ma J, et al. GATA2 controls lymphatic endothelial cell junctional integrity and lymphovenous valve morphogenesis through miR-126. Development. 2019;146(21):1039.
37.
Zurück zum Zitat Chan YC, Roy S, Khanna S, Sen CK. Downregulation of endothelial microRNA-200b supports cutaneous wound angiogenesis by desilencing GATA binding protein 2 and vascular endothelial growth factor receptor 2. Arterioscler Thromb Vasc Biol. 2012;32(6):1372–82.PubMedPubMedCentralCrossRef Chan YC, Roy S, Khanna S, Sen CK. Downregulation of endothelial microRNA-200b supports cutaneous wound angiogenesis by desilencing GATA binding protein 2 and vascular endothelial growth factor receptor 2. Arterioscler Thromb Vasc Biol. 2012;32(6):1372–82.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Zarrinkar PP, Gunawardane RN, Cramer MD, Gardner MF, Brigham D, Belli B, et al. AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML). Blood. 2009;114(14):2984–92.PubMedPubMedCentralCrossRef Zarrinkar PP, Gunawardane RN, Cramer MD, Gardner MF, Brigham D, Belli B, et al. AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML). Blood. 2009;114(14):2984–92.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Duarte D, Hawkins ED, Akinduro O, Ang H, De Filippo K, Kong IY, et al. Inhibition of endosteal vascular niche remodeling rescues hematopoietic stem cell loss in AML. Cell Stem Cell. 2018;22(1):64–77.PubMedPubMedCentralCrossRef Duarte D, Hawkins ED, Akinduro O, Ang H, De Filippo K, Kong IY, et al. Inhibition of endosteal vascular niche remodeling rescues hematopoietic stem cell loss in AML. Cell Stem Cell. 2018;22(1):64–77.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Batsivari A, Haltalli MLR, Passaro D, Pospori C, Lo Celso C, Bonnet D. Dynamic responses of the haematopoietic stem cell niche to diverse stresses. Nat Cell Biol. 2020;22(1):7–17.PubMedCrossRef Batsivari A, Haltalli MLR, Passaro D, Pospori C, Lo Celso C, Bonnet D. Dynamic responses of the haematopoietic stem cell niche to diverse stresses. Nat Cell Biol. 2020;22(1):7–17.PubMedCrossRef
41.
Zurück zum Zitat Silke J, Hartland EL. Masters, marionettes and modulators: intersection of pathogen virulence factors and mammalian death receptor signaling. Curr Opin Immunol. 2013;25(4):436–40.PubMedCrossRef Silke J, Hartland EL. Masters, marionettes and modulators: intersection of pathogen virulence factors and mammalian death receptor signaling. Curr Opin Immunol. 2013;25(4):436–40.PubMedCrossRef
42.
43.
Zurück zum Zitat Kovtonyuk LV, Fritsch K, Feng X, Manz MG, Takizawa H. Inflamm-aging of hematopoiesis, hematopoietic stem cells, and the bone marrow microenvironment. Front Immunol. 2016;7:502.PubMedPubMedCentralCrossRef Kovtonyuk LV, Fritsch K, Feng X, Manz MG, Takizawa H. Inflamm-aging of hematopoiesis, hematopoietic stem cells, and the bone marrow microenvironment. Front Immunol. 2016;7:502.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Baluk P, Yao LC, Feng J, Romano T, Jung SS, Schreiter JL, et al. TNF-alpha drives remodeling of blood vessels and lymphatics in sustained airway inflammation in mice. J Clin Investig. 2009;119(10):2954–64.PubMedPubMedCentral Baluk P, Yao LC, Feng J, Romano T, Jung SS, Schreiter JL, et al. TNF-alpha drives remodeling of blood vessels and lymphatics in sustained airway inflammation in mice. J Clin Investig. 2009;119(10):2954–64.PubMedPubMedCentral
45.
Zurück zum Zitat Chen JX, Chen Y, DeBusk L, Lin W, Lin PC. Dual functional roles of Tie-2/angiopoietin in TNF-alpha-mediated angiogenesis. Am J Physiol Heart Circ Physiol. 2004;287(1):H187–95.PubMedCrossRef Chen JX, Chen Y, DeBusk L, Lin W, Lin PC. Dual functional roles of Tie-2/angiopoietin in TNF-alpha-mediated angiogenesis. Am J Physiol Heart Circ Physiol. 2004;287(1):H187–95.PubMedCrossRef
46.
Zurück zum Zitat Chistiakov DA, Orekhov AN, Bobryshev YV. The role of miR-126 in embryonic angiogenesis, adult vascular homeostasis, and vascular repair and its alterations in atherosclerotic disease. J Mol Cell Cardiol. 2016;97:47–55.PubMedCrossRef Chistiakov DA, Orekhov AN, Bobryshev YV. The role of miR-126 in embryonic angiogenesis, adult vascular homeostasis, and vascular repair and its alterations in atherosclerotic disease. J Mol Cell Cardiol. 2016;97:47–55.PubMedCrossRef
47.
Zurück zum Zitat Sessa R, Seano G, di Blasio L, Gagliardi PA, Isella C, Medico E, et al. The miR-126 regulates angiopoietin-1 signaling and vessel maturation by targeting p85beta. Biochim Biophys Acta. 2012;1823(10):1925–35.PubMedCrossRef Sessa R, Seano G, di Blasio L, Gagliardi PA, Isella C, Medico E, et al. The miR-126 regulates angiopoietin-1 signaling and vessel maturation by targeting p85beta. Biochim Biophys Acta. 2012;1823(10):1925–35.PubMedCrossRef
48.
Zurück zum Zitat Pizurki L, Zhou Z, Glynos K, Roussos C, Papapetropoulos A. Angiopoietin-1 inhibits endothelial permeability, neutrophil adherence and IL-8 production. Br J Pharmacol. 2003;139(2):329–36.PubMedPubMedCentralCrossRef Pizurki L, Zhou Z, Glynos K, Roussos C, Papapetropoulos A. Angiopoietin-1 inhibits endothelial permeability, neutrophil adherence and IL-8 production. Br J Pharmacol. 2003;139(2):329–36.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Lechman ER, Gentner B, Ng SW, Schoof EM, van Galen P, Kennedy JA, et al. miR-126 regulates distinct self-renewal outcomes in normal and malignant hematopoietic stem cells. Cancer Cell. 2016;29(2):214–28.PubMedPubMedCentralCrossRef Lechman ER, Gentner B, Ng SW, Schoof EM, van Galen P, Kennedy JA, et al. miR-126 regulates distinct self-renewal outcomes in normal and malignant hematopoietic stem cells. Cancer Cell. 2016;29(2):214–28.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Maifrede S, Nieborowska-Skorska M, Sullivan-Reed K, Dasgupta Y, Podszywalow-Bartnicka P, Le BV, et al. Tyrosine kinase inhibitor-induced defects in DNA repair sensitize FLT3(ITD)-positive leukemia cells to PARP1 inhibitors. Blood. 2018;132(1):67–77.PubMedPubMedCentralCrossRef Maifrede S, Nieborowska-Skorska M, Sullivan-Reed K, Dasgupta Y, Podszywalow-Bartnicka P, Le BV, et al. Tyrosine kinase inhibitor-induced defects in DNA repair sensitize FLT3(ITD)-positive leukemia cells to PARP1 inhibitors. Blood. 2018;132(1):67–77.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Epstein SE, Stabile E, Kinnaird T, Lee CW, Clavijo L, Burnett MS. Janus phenomenon: the interrelated tradeoffs inherent in therapies designed to enhance collateral formation and those designed to inhibit atherogenesis. Circulation. 2004;109(23):2826–31.PubMedCrossRef Epstein SE, Stabile E, Kinnaird T, Lee CW, Clavijo L, Burnett MS. Janus phenomenon: the interrelated tradeoffs inherent in therapies designed to enhance collateral formation and those designed to inhibit atherogenesis. Circulation. 2004;109(23):2826–31.PubMedCrossRef
Metadaten
Titel
Treatment-induced arteriolar revascularization and miR-126 enhancement in bone marrow niche protect leukemic stem cells in AML
verfasst von
Bin Zhang
Le Xuan Truong Nguyen
Dandan Zhao
David E. Frankhouser
Huafeng Wang
Dinh Hoa Hoang
Junjing Qiao
Christina Abundis
Matthew Brehove
Yu-Lin Su
Yuxin Feng
Anthony Stein
Lucy Ghoda
Adrianne Dorrance
Danilo Perrotti
Zhen Chen
Anjia Han
Flavia Pichiorri
Jie Jin
Tijana Jovanovic-Talisman
Michael A. Caligiuri
Calvin J. Kuo
Akihiko Yoshimura
Ling Li
Russell C. Rockne
Marcin Kortylewski
Yi Zheng
Nadia Carlesso
Ya-Huei Kuo
Guido Marcucci
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2021
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-021-01133-y

Weitere Artikel der Ausgabe 1/2021

Journal of Hematology & Oncology 1/2021 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.