scholarly journals Therapeutic Targeting of the Leukaemia Microenvironment

2021 ◽  
Vol 22 (13) ◽  
pp. 6888
Author(s):  
Vincent Kuek ◽  
Anastasia M. Hughes ◽  
Rishi S. Kotecha ◽  
Laurence C. Cheung

In recent decades, the conduct of uniform prospective clinical trials has led to improved remission rates and survival for patients with acute myeloid leukaemia and acute lymphoblastic leukaemia. However, high-risk patients continue to have inferior outcomes, where chemoresistance and relapse are common due to the survival mechanisms utilised by leukaemic cells. One such mechanism is through hijacking of the bone marrow microenvironment, where healthy haematopoietic machinery is transformed or remodelled into a hiding ground or “sanctuary” where leukaemic cells can escape chemotherapy-induced cytotoxicity. The bone marrow microenvironment, which consists of endosteal and vascular niches, can support leukaemogenesis through intercellular “crosstalk” with niche cells, including mesenchymal stem cells, endothelial cells, osteoblasts, and osteoclasts. Here, we summarise the regulatory mechanisms associated with leukaemia–bone marrow niche interaction and provide a comprehensive review of the key therapeutics that target CXCL12/CXCR4, Notch, Wnt/b-catenin, and hypoxia-related signalling pathways within the leukaemic niches and agents involved in remodelling of niche bone and vasculature. From a therapeutic perspective, targeting these cellular interactions is an exciting novel strategy for enhancing treatment efficacy, and further clinical application has significant potential to improve the outcome of patients with leukaemia.

Cancers ◽  
2021 ◽  
Vol 13 (16) ◽  
pp. 4116
Author(s):  
Syed A. Mian ◽  
Dominique Bonnet

Myelodysplastic syndrome (MDS) are clonal haematopoietic stem cell (HSC) disorders driven by a complex combination(s) of changes within the genome that result in heterogeneity in both clinical phenotype and disease outcomes. MDS is among the most common of the haematological cancers and its incidence markedly increases with age. Currently available treatments have limited success, with <5% of patients undergoing allogeneic HSC transplantation, a procedure that offers the only possible cure. Critical contributions of the bone marrow microenvironment to the MDS have recently been investigated. Although the better understanding of the underlying biology, particularly genetics of haematopoietic stem cells, has led to better disease and risk classification; however, the role that the bone marrow microenvironment plays in the development of MDS remains largely unclear. This review provides a comprehensive overview of the latest developments in understanding the aetiology of MDS, particularly focussing on understanding how HSCs and the surrounding immune/non-immune bone marrow niche interacts together.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5009-5009
Author(s):  
Yan Lin ◽  
Quan Gu ◽  
Hui Cheng ◽  
Zhaofeng Zheng ◽  
Sun Guohuan ◽  
...  

Severe acute graft-versus-host disease (aGVHD) indicates a poor prognosis after allogeneic hematopoietic stem cell transplantation. In our previous study, we found that hematopoiesis was progressively suppressed during aGVHD, while the hematopoietic regenerative potential of donor-derived hematopoietic stem cells remains intact. It prompts us to investigate whether bone marrow niche is a major target of GVHD. We addressed this issue by studying the critical components in bone marrow microenvironment, including mesenchymal stem cell (MSC), osteoblasts and adipocytes in haplo-MHC-matched murine bone marrow transplantation model. By comparing confocal images of femurs from control and aGVHD SCF-GFP mice or Col2.3-GFP mice, we found that both MSCs and osteoblasts were significantly reduced during aGVHD development. In addition, anti-perilipin staining showed that adipocytes were also decreased in aGVHD mice. We found a defect in the differentiation potential of MSCs from aGVHD niche by in vitro culture of both murine and human bone marrow niche cells. qRT-PCR showed decreased gene expressions of PPAR-gamma, Adipoq, Runx1 and Col2a1, suggesting the potential of MSCs differentiation into adipocytes and osteoblasts was blocked. These data provide new insights into the pathogenesis of aGVHD and may improve the clinical management of aGVHD. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Author(s):  
Chiara Baccin ◽  
Jude Al-Sabah ◽  
Lars Velten ◽  
Patrick M. Helbling ◽  
Florian Grünschläger ◽  
...  

SUMMARYThe bone marrow (BM) constitutes the primary site for life-long blood production and skeletal regeneration. However, its cellular composition and the spatial organization into distinct ‘niches’ remains controversial. Here, we combine single-cell and spatially resolved transcriptomics to systematically map the molecular and cellular composition of the endosteal, sinusoidal, and arteriolar BM niches. This allowed us to transcriptionally profile all major BM resident cell types, determine their localization, and clarify the cellular and spatial sources of key growth factors and cytokines. Our data demonstrate that previously unrecognized Cxcl12-abundant reticular (CAR) cell subsets (i.e. Adipo- and Osteo-CAR cells) differentially localize to sinusoidal or arteriolar surfaces, locally act as ‘professional cytokine secreting cells’, and thereby establish distinct peri-vascular micro-niches. Importantly, we also demonstrate that the 3-dimensional organization of the BM can be accurately inferred from single-cell gene expression data using the newly developed RNA-Magnet algorithm. Together, our study reveals the cellular and spatial organization of BM niches, and offers a novel strategy to dissect the complex organization of whole organs in a systematic manner.One Sentence SummaryIntegration of single-cell and spatial transcriptomics reveals the molecular, cellular and spatial organization of bone marrow niches


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1721-1721
Author(s):  
Amina M Abdul-Aziz ◽  
Manar S Shafat ◽  
Lyubov Zaitseva ◽  
Matthew J Lawes ◽  
Stuart A Rushworth ◽  
...  

Abstract Introduction Hypoxia is an important component of the bone marrow microenvironment and the hematopoietic stem cell niche. Studies have shown that hypoxia contributes to the development and maintenance of acute myeloid leukemia (AML) cells within the bone marrow microenvironment. Hypoxia is principally maintained by members of the hypoxia-inducible factor (HIF), in particular HIF1a and its target genes, including MIF. We have previously shown that AML cells express constitutively high macrophage migration inhibitory factor (MIF) which drives IL-8 expression by the BM-MSC which in turn supports AML cell survival and proliferation. The aim of the present study is to determine if there is a connection between the role of hypoxia in regulating AML survival and MIF survival signals. Furthermore we investigate the role BM-MSC in regulating the hypoxic response. Methods Primary AML and BM-MSC were isolated from AML patients following informed consent and under approval from the UK National Research Ethics Service (LRCEref07/H0310/146). AML cell lines and primary AML blasts were cultured under normoxic (20% oxygen) or hypoxic conditions (1% oxygen) for 4 - 24 hours, mRNA expression of MIF, HIF1a, VEGF and IL-8 were determined by RT-PCR. MIF and IL-8 protein was determined using target specific ELISA. HIF1a protein expression was determined by western blotting. Hypoxia-mimetic agents, cobalt chloride (CoCl2) and desferrioxamine (DFO) were used. Cell proliferation was determines using CellTiter Glo and trypan blue exclusion. CFU-assays were performed using complete methylcellulose media. Results To determine If MIF is regulated by HIF1a in AML cells, we mimicked hypoxic conditions using CoCl2 and DFO in AML cells. Both CoCl2 and DFO upregulate MIF transcription and protein expression in OCI-AML3 cell lines and in primary AML blasts. Moreover, hypoxia increases both MIF mRNA expression and MIF chemokine expression compared to normoxic conditions. Lentiviral mediated knockdown of MIF in AML cells show significantly reduced cell proliferation and colony formation in methylcellulose media. Recombinant MIF induced interleukin-8 in AML blasts and the MIF inhibitor blocked MIF induced IL-8 release. Lentiviral mediated KD of HIF1a decreased MIF expression in human AML cells and a significant reduced their proliferative capacity. Finally we found that hypoxia increased MIF in AML blasts which was further increased when in co-culture with BM-MSC. Conclusions The results reported here suggest that hypoxia significantly affects the expression of the survival cytokine MIF in AML blasts. Furthermore, we show that BM-MSC regulate HIF1a induced MIF expression in AML under hypoxic conditions. We propose this hypoxia regulated HIF1a/MIF axis is essential to blast survival in the bone marrow niche. Disclosures Rushworth: Infinity Pharmaceuticals: Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4372-4372 ◽  
Author(s):  
Ashley Hamilton ◽  
Katie Foster ◽  
Dominique Bonnet

Abstract The cell fate of the HSC to either self-renew or differentiate is controlled by a complex interplay between cell-intrinsic and -extrinsic regulatory signals generated by the surrounding bone marrow microenvironment called the HSC niche. A balance exists within the “cross talk” between HSCs and the niche, which allows HSC dormancy, activation and differentiation. Any alterations of this balance may lead to uncontrolled cellular proliferation and ultimately the promotion of leukemia. However, it remains to be determined exactly how the hematopoietic microenvironment contributes to the deregulation of normal hematopoiesis and/or promotes the maintenance of leukemia cells as a “leukemic niche”. To investigate this, we have now performed micro-array analysis of MS5 stromal cells that were co-cultured with a panel of leukemic cell lines and acute myeloid leukemia (AML) patient samples. The most significantly up-regulated pathways as compared to MS5 cells cultured alone involved cytoskeleton remodeling, cell cycle, cell adhesion and development through cytokine signaling. Since transcript and protein levels of number of effectors of the TGF-beta (TGF-β) signaling pathway were up-regulated in the stroma co-cultured with leukemic cells, we next investigated inhibition of this pathway using a specific inhibitor of TGF-β receptor kinase, SB-431542 (10µM). Treatment with the inhibitor significantly reduced the cell number and increased the levels of apoptosis in the AML cells co-cultured on stromal cells, whilst having mininal effect on normal cells. Treatment with SB-431542 (10mg/kg), also significantly reduced the level of AML cell engraftment on treatment in vivo (n=3) (untreated- 68.65±6.95; 56.15±22.85; 84.35±5.75 and SB-treated- 45.5±11.6; 30.5±19.6; 54.1±4.9). In order to inhibit TGF-β signaling more specifically within the stromal compartment, we next used shRNA against TGF-β Receptor II (TGFBR2) in MS5 stromal cells and co-cultured them with AML cells within 3D scaffold models (n=4), which were implanted in vivo. A significant reduction in engraftment was observed as compared to controls (shRNA control- 64.65±32.65; 87.7±7.2; 23.55±4.35; 49.65±33.65 and TGFBR2 knockdown- 20.2±3; 62.95±4.05; 15.7±1.5; 20.385±17.415). The co-culturing of normal cord blood CD34+ or mononuclear cells on the TGFBR2 knockdown stroma had no significant effect both in vitro and in vivo (n=3). To investigate whether TGF-β inhibition had an effect on the interaction of AML cells to the niche, we used intravital microscopy to track the cells live in vivo. HL60 (AML cell line) cells were labeled with 2µM CFSE and pretreated ±SB-431542 (10µM) on stroma, before being sorted and transplanted into immunodeficient mice. Distance to the calvaria was measured at 16 hours and we observed that the SB-431542-treated cells were positioned significantly further away from the bone surface as compared to untreated control (p=0.0001). Since the TGF-β inhibited cells appeared to have impaired ability to adhere to the bone marrow, we next investigated the relationship between extracellular matrix molecules and TGF-β signaling. We saw that stromal cells that were co-cultured with AML cells had a significantly increased expression of laminins A1, A5, B1 and G1. This effect could be recapitulated by treatment of naïve stromal cells with TGF-β2 and 3. We also observed a reciprocal decrease in expression of laminins following both treatment of AML-stromal co-cultures with SB-431542 and within TGFBR2 knockdown stroma. Furthermore, we saw an increase in the laminin receptor, integrin alpha-6 (CD49f), in AML cells treated with TGF-β 1, 2 and 3 and a reciprocal decrease following treatment with SB-431542, thereby, indicating that the abrogation of this signaling axis may be, at least, partially responsible for the impaired engraftment of AML cells to their niche following inhibition of the TGF-β pathway. These data thus highlight the potential for the development of therapies directed at modifying the bone marrow microenvironment. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2019 ◽  
Vol 11 (12) ◽  
pp. 1939 ◽  
Author(s):  
Severin ◽  
Frezzato ◽  
Visentin ◽  
Martini ◽  
Trimarco ◽  
...  

The bone marrow microenvironment promotes proliferation and drug resistance in chronic lymphocytic leukemia (CLL). Although ibrutinib is active in CLL, it is rarely able to clear leukemic cells protected by bone marrow mesenchymal stromal cells (BMSCs) within the marrow niche. We investigated the modulation of JAK2/STAT3 pathway in CLL by BMSCs and its targeting with AG490 (JAK2 inhibitor) or Stattic (STAT3 inhibitor). B cells collected from controls and CLL patients, were treated with medium alone, ibrutinib, JAK/Signal Transducer and Activator of Transcription (STAT) inhibitors, or both drugs, in the presence of absence of BMSCs. JAK2/STAT3 axis was evaluated by western blotting, flow cytometry, and confocal microscopy. We demonstrated that STAT3 was phosphorylated in Tyr705 in the majority of CLL patients at basal condition, and increased following co-cultures with BMSCs or IL-6. Treatment with AG490, but not Stattic, caused STAT3 and Lyn dephosphorylation, through re-activation of SHP-1, and triggered CLL apoptosis even when leukemic cells were cultured on BMSC layers. Moreover, while BMSCs hamper ibrutinib activity, the combination of ibrutinib+JAK/STAT inhibitors increase ibrutinib-mediated leukemic cell death, bypassing the pro-survival stimuli derived from BMSCs. We herein provide evidence that JAK2/STAT3 signaling might play a key role in the regulation of CLL-BMSC interactions and its inhibition enhances ibrutinib, counteracting the bone marrow niche.


2021 ◽  
Vol 10 ◽  
Author(s):  
Tyler Moser-Katz ◽  
Nisha S. Joseph ◽  
Madhav V. Dhodapkar ◽  
Kelvin P. Lee ◽  
Lawrence H. Boise

Multiple myeloma is a clonal disease of long-lived plasma cells and is the second most common hematological cancer behind Non-Hodgkin’s Lymphoma. Malignant transformation of plasma cells imparts the ability to proliferate, causing harmful lesions in patients. In advanced stages myeloma cells become independent of their bone marrow microenvironment and form extramedullary disease. Plasma cells depend on a rich array of signals from neighboring cells within the bone marrow for survival which myeloma cells exploit for growth and proliferation. Recent evidence suggests, however, that both the myeloma cells and the microenvironment have undergone alterations as early as during precursor stages of the disease. There are no current therapies routinely used for treating myeloma in early stages, and while recent therapeutic efforts have improved patients’ median survival, most will eventually relapse. This is due to mutations in myeloma cells that not only allow them to utilize its bone marrow niche but also facilitate autocrine pro-survival signaling loops for further progression. This review will discuss the stages of myeloma cell progression and how myeloma cells progress within and outside of the bone marrow microenvironment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 82-82
Author(s):  
Andre Larochelle ◽  
Jennifer Gillette ◽  
Amy Cantilena ◽  
Alexandra Cerf ◽  
Jennifer Lippincott-Schwartz ◽  
...  

Abstract Abstract 82 Hematopoietic stem/progenitor cells (HSPCs) reside in a bone marrow niche, where adhesive interactions with osteoblasts provide essential cues for their proliferation and survival. In co-cultures of osteoblasts with primary human normal CD34+ cells, CD34+CD38- cells, or the KG1a progenitor cell line, we previously showed, using live cell imaging approaches, that HSPCs made prolonged contact with the osteoblast surface via a specialized membrane domain enriched in prominin 1 (CD133), the very late antigen-4 (VLA-4), the phosphatidylethanolamine (PE) analogue rhodamine-PE, and the tetraspanins CD63 and CD81. At the contact site, portions of the specialized domain of the CD34+ cells containing these molecules were taken up by osteoblasts and internalized into signaling endosomes within the osteoblasts. This caused the osteoblasts to downregulate Smad signaling and to increase their production of stromal-derived factor-1 (SDF-1), a chemokine responsible for HSPC homing to bone marrow (Gillette J. et al, Nat. Cell Biol. 11(3): 303–311, 2009). We have now evaluated the functional significance of these specialized membrane domains for in vivo homing of normal HSPCs to the bone marrow microenvironment. G-CSF mobilized human peripheral blood CD34+ cells from two normal donors were treated for 30 minutes with the cholesterol sequestration agent methyl-β-cyclodextrin (MβCD). This treatment resulted in disruption of the CD34+ cell membrane domains but had no effect on cell viability, proliferation or colony forming capacity in vitro. However, in two independent experiments, we observed a three-fold decrease in homing of MβCD-treated CD34+ cells to the bone marrow of NOD/SCID IL2rψcnull mice 16 hours after transplantation as compared to mock-treated CD34+ cells (p=0.0002). In contrast to homing studies, long-term human cell engraftment determined by CD45 cell surface expression 2 months after transplantation in two independent experiments was not significantly different in mice transplanted with MβCD-treated CD34+ cells compared to mock-treated CD34+ cells (p=0.13). Rapid repolarization of the membrane domain after transplantation may have resulted in engraftment of MβCD-treated CD34+ cells at levels similar to those observed with mock-treated CD34+ cells. Given the known homing/engraftment defect of actively cycling HSPC, we compared membrane domains on quiescent and cycling CD34+ cells from two normal donors. At baseline, 55–68% of CD34+ cells were in the G0 phase of the cell cycle as measured by Hoechst/Pyronin Y staining and specialized membrane domains were detected on a similar percentage of CD34+ cells for both donors. After culture for 4 days in the presence of stimulatory cytokines (SCF, TPO and Flt3), less than 10% of the CD34+ cells remained in G0 and, similarly, less than 10% of the cells analyzed by microscopy had a specialized membrane domain. After 4 days, cells were continued in culture for 2 days under non-stimulatory conditions (SCF alone). Under these conditions, for one donor, the percentage of cells in G0 increased from 9% to 18% and the percentage of cells with membrane domains increased similarly from 6% to 11%. For the other donor, the percentage of cells in G0 and with membrane domains both remained unchanged after 2 days in non-stimulatory culture. Surprisingly, the polarized membrane domains detected on normal CD34+ cells were not found on peripheral blood blast cells from patients diagnosed with relapsed AML (n=2 patients) or CML (n=1 patient). Accordingly, in a preliminary experiment, no difference in homing was observed 16 hours after transplantation of MβCD- or mock-treated peripheral blood cells from an AML patient in NOD/SCID IL2rψcnull mice. Additional homing and engraftment studies using cells from leukemic patients are ongoing. In combination, these findings indicate that specialized membrane domains are found on normal but not leukemic HSPCs and that these domains are required for homing to the bone marrow microenvironment. Disruption of these domains in actively cycling progenitor cells may provide an explanation for the previously demonstrated homing/engraftment defect of cycling cells compared to quiescent cells in the G0 phase of the cell cycle. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Vol 38 (22) ◽  
Author(s):  
Yubin Luo ◽  
Bettina Grötsch ◽  
Nicole Hannemann ◽  
Maria Jimenez ◽  
Natacha Ipseiz ◽  
...  

ABSTRACT Inflammatory responses require mobilization of innate immune cells from the bone marrow. The functionality of this process depends on the state of the bone marrow microenvironment. We therefore hypothesized that molecular changes in osteoblasts, which are essential stromal cells of the bone marrow microenvironment, influence the inflammatory response. Here, we show that osteoblast-specific expression of the AP-1 transcription factor Fra-2 (Fra-2Ob-tet) induced a systemic inflammatory state with infiltration of neutrophils and proinflammatory macrophages into the spleen and liver as well as increased levels of proinflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, and granulocyte-macrophage colony-stimulating factor (GM-CSF). By in vivo inhibition of osteopontin (OPN) in Fra-2Ob-tet mice, we demonstrated that this process was dependent on OPN expression, which mediates alterations of the bone marrow niche. OPN expression was transcriptionally enhanced by Fra-2 and stimulated mesenchymal stem cell (MSC) expansion. Furthermore, in a murine lung injury model, Fra-2Ob-tet mice showed increased inflammatory responses and more severe disease features via an enhanced and sustained inflammatory response to lipopolysaccharide (LPS). Our findings demonstrate for the first time that molecular changes in osteoblasts influence the susceptibility to inflammation by altering evasion of innate immune cells from the bone marrow space.


Sign in / Sign up

Export Citation Format

Share Document