scholarly journals Anlotinib Potentially Eliminates Leukemia Stem Cells and Modulates the Chemosensitivity via Inhibiting JAK2-STAT3/5 Signaling

Author(s):  
Yuelong Jiang ◽  
Long Liu ◽  
Zhifeng Li ◽  
Liying Feng ◽  
Zhijuan Lin ◽  
...  

Abstract Leukemia stem cells (LSCs) remain as the critical barrier to cure of acute myeloid leukemia (AML) due to its chemoresistance. Here, we explore the role anlotinib -a multiple tyrosine kinases inhibitor in killing LSCs and regulating the chemoresistance. We found anlotinib could effectively induce apoptosis of LSC-like cells as well as primary CD34+ AML LSCs while sparing the normal mononuclear cells in vitro. The anti-leukemia activity of anlotinib was also confirmed in the mice model with Kasumi-1 cells; we further found that anlotinib could impair the regeneration capacity of LSCs in the patient-derived leukemia xenograft mouse model. Mechanistically, anlotinib could not only inhibit phosphorylation of c-kit and JAK2 /STAT3 and STAT5 but also downregulate STAT3 and STAT5 expression. In addition, anlotinib downregulated the anti-apoptotic protein Bcl-2 and Bcl-xl and upregulated Bax, thereby enhancing the sensitivity of LSCs to idarubicin in vitro. In conclusion, our results demonstrated anlotinib showed anti-LSCs activity and enhanced the chemosensitivity via inhibiting JAK2/STAT signaling in preclinical study and provided a rational basis for combinatory strategies that invoving anlotinib and idarubicin.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3065-3065
Author(s):  
Wenxue Ma ◽  
Cayla N Mason ◽  
Ping Chen ◽  
Nathaniel Delos Santos ◽  
Jiang Qingfei ◽  
...  

Abstract Introduction Leukemia stem cells (LSCs) in chronic myeloid leukemia (CML) are generated from progenitors that have aberrantly activated self-renewal pathways thereby resulting in tyrosine kinase inhibitor (TKI) resistance. The telomerase complex, consisting of a reverse transcriptase subunit (TERT), an RNA template subunit (TERC), and a protective shelterin scaffold, transcriptionally modulates the Wnt/b-catenin self-renewal pathway. Many malignancies, including BCR-ABL TKI resistant blast crisis CML (BC CML), exhibit robust telomerase activity thereby prompting the development of imetelstat, a competitive inhibitor of telomerase enzymatic activity. Imetelstat is a covalently lipidated 13-mer oligonucleotide that binds with high affinity to the TERC subunit. Recent clinical trials showed early signs of efficacy in myeloproliferative neoplasms. However, the role of imetelstat in selective self-renewing LSC inhibition in CML had not been elucidated. Thus, we performed progenitor RNA sequencing (RNA-seq), stromal co-cultures and humanized LSC primagraft studies to investigate the capacity of imetelstat to selectively inhibit LSC self-renewal and to determine the mechanism of action. Methods and Results Cytoscape analysis of RNA-seq data derived from FACS-purified progenitors from human blast crisis (BC; n=9) compared with chronic phase (CP; n=8) CML and primary normal (n=6) samples revealed transcriptional upregulation of b-catenin, LEF1, TCF7L1, ABL1 and other key genes within the TERT interactome suggesting a role for TERT activation in human BC LSC generation. Human progenitor LSC-supportive SL/M2 stromal co-culture experiments revealed that combined treatment with a potent BCR-ABL TKI, dasatinib at 1 nM, and imetelstat at 1 or 5 mM significantly inhibited (p<0.001, ANOVA) in vitro self-renewal of BC CML (n=5) compared with age-matched normal bone marrow progenitors. Treatment of primagraft mouse models of human BC CML with 30 mg/kg of imetelstat three times a week for four weeks resulted in a significant reduction in bone marrow progenitor LSC burden compared with mismatch treated controls (p=0.04). Furthermore, qRT-PCR showed decreased β-catenin transcript levels in imetelstat compared with vehicle-treated samples. Finally, FACS analysis revealed a significant reduction in activated b-catenin protein levels in engrafted human myeloid progenitors following imetelstat treatment in the TKI resistant bone marrow niche. Conclusions Niche responsive interactions between the telomerase complex and the Wnt/b-catenin self-renewal pathway sensitize b-catenin activated LSC to imetelstat in both in vitro and in vivo humanized pre-clinical BC CML models thereby providing a strong rationale for LSC eradication trials involving imetelstat. Disclosures Huang: Janssen Research & Development, LLC: Employment, Other: I am an employee of Janssen and a stock owner . Jamieson:UC San Diego: Other: I received funding from Janssen Research & Development, LLC.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 49-49 ◽  
Author(s):  
Ross Kinstrie ◽  
Gillian A. Horne ◽  
Heather Morrison ◽  
Hothri A. Moka ◽  
Jennifer Cassels ◽  
...  

Abstract The introduction of BCR-ABL tyrosine kinase inhibitors (TKIs) has revolutionized the treatment of chronic myeloid leukemia (CML). However, although the majority of patients with chronic phase (CP)-CML obtain durable complete cytogenetic and major molecular responses, there is low level disease persistence postulated to be due to a population of TKI-insensitive leukemia stem cells (LSC). The aims of this study were (1) to fully characterize differences in gene expression between normal hematopoietic stem cells (HSC) and CP-CML LSC and (2) identify potential novel therapeutic targets specific to CML LSC. Lin-CD34+CD38- CD45RA-CD90+ normal HSC (n=3) and CP-CML LSC (n=6 patients at diagnosis), populations were isolated using a FACSAria and applied to Affymetrix HuGene 1.0ST arrays. The raw data (.CEL files) was imported into Partek Genomics Suite and Ingenuity Pathway Analysis software and principal component analysis and gene ontology ANOVA performed. A total of 1217 genes were significantly deregulated between normal HSC and CP-CML LSC. The most significantly deregulated genes and pathways were involved with the molecular and cellular functions of cell cycle, cell assembly and organisation, cellular movement, cell death and DNA replication, recombination and repair. These results suggested that CML LSC were less quiescent than normal HSC. Importantly, complimentary functional studies indicated that CML LSC have significantly increased proliferation (14 fold expansion; P<0.001) compared to normal HSC (no expansion) after 5 days in vitro culture. In addition, equivalent numbers of CML LSC produce ~4-fold more colonies in colony forming cell (CFC) assays than normal HSC (329±56 versus 86±17 per 2,000 cells, respectively; P<0.05). Fluorescence in situ hybridisation (FISH) demonstrated that >90% of lin- CD34+ CD38- CD45RA- CD90+ CML LSC from all patient samples were BCR-ABL positive (+). In addition to these deregulated intracellular pathways, we sought to assess if there were differences in expression of cell surface molecules that may be amenable to therapeutic manipulation. Of particular interest, our microarray studies demonstrated that CD93 was highly upregulated in CP-CML LSC (6 fold, p = 2.5x10-6). Increased CD93 expression was validated by Fluidigm digital PCR (6 fold increase, p = 0.02; n=6). Furthermore, using flow cytometry, we demonstrated significant upregulation of CD93 protein expression on lin-CD34+ CD38- CD45RA- CD90+ CML LSC from peripheral blood and bone marrow of CP-CML patients (n= 17; mean = 63.8% CD93+) compared to normal HSC from healthy peripheral blood stem cell donors (n=7; mean = 0.8% CD93+) and bone marrow donors (n=4; mean = 0.2% CD93+; p < 0.0001). FISH confirmed that 100% of lin-CD34+ CD38- CD90+ CD93+ CML cells were BCR-ABL+ in all samples assessed. CD93 (also known as C1qRp) is a C-type lectin-like domain (CTLD)-containing glycoprotein which regulates phagocytosis, with roles in cell adhesion and leukocyte migration. It is normally expressed on endothelial cells, hematopoietic precursors and mature cells including neutrophils, monocytes and platelets. Previous studies have shown CD93 to be upregulated in a proportion of AML patients (Saito et al, Sci Transl Med, 2010. 2(17): p. 17ra9). Short term (24h) in vitro exposure of lin-CD34+CD38- CD45RA- CD90+ CML LSC to TKIs (Imatinib or Dasatinib; n=3) reduced, but did not fully eliminate CD93 expression (Imatinib, 48.5% to 22.9%; Dasatinib, 47.7% to 9.2%). Importantly, following long-term TKI treatment of patients, lin-CD34+CD38-CD45RA-CD90+ cells from CP-CML patient bone marrow samples (n=2) taken in major molecular response demonstrated a small, but persistent population of CD93+ LSC which were BCR-ABL+ by FISH. Furthermore, in xenograft transplantation experiments (n = 5), after 16 weeks, CD34+CD93+ CML LSC engrafted lethally irradiated NOD/SCID/IL-2Rg-/- (NSG) mice with BCR-ABL+ cells, whereas CD34+CD93- cells from the same patient samples failed to engraft to significant levels (3.5-30 fold increase in engraftment with CD34+CD93- cells; p < 0.03). FISH confirmed that engrafted human cells were BCR-ABL+. Taken together, our results identify CD93 as a potential novel biomarker of CML LSC, which may also be helpful in assessing minimal residual disease at the LSC level. Further studies are ongoing to assess the therapeutic potential of inhibiting CD93 in CML LSC. Disclosures Copland: Ariad: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 15-15
Author(s):  
Anna Sergeeva ◽  
Gheath Alatrash ◽  
Karen Clise-Dwyer ◽  
Kathryn E Quintanilla ◽  
Sijie Lu ◽  
...  

Abstract Abstract 15 PR1 (VLQELNVTV) is an HLA-A2-restricted peptide derived from endogenous myeloid leukemia-associated antigens (LAA) proteinase 3 and neutrophil elastase. PR1/HLA-A2 is targeted by PR1-specific cytotoxic T lymphocytes (PR1-CTL) that contribute to cytogenetic remission in patients with CML after allogeneic stem cell transplantation or interferon therapy, and PR1 peptide vaccination induces specific CD8 immunity in AML, MDS, and CML patients. Because clinical effects after PR1 vaccination are associated with a low amount of leukemia and because the effects on leukemia stem cells (LSCs) are unknown, we sought to develop a PR1/HLA-A2-specific MAB that could be more effective in high disease burden cases and that might also target LSC. We previously reported a mouse IgG2A antibody (8F4) with PR1/HLA-A2 specificity. We now used 8F4 to study PR1 expression by FACS and confocal mircroscopy on leukemia subsets and normal peripheral blood mononuclear cells (PBMC). Circulating blasts from peripheral blood (PB) or leukapheresis products (LP) from AML patients (5 HLA-A2+, 2 control HLA-A2-,) were studied (see Table 1). PR1 expression (MFI±SEM) was higher in AML (23.7±5.18) compared to control (4.0±2.42) and normal PBMC (13.6±0.23; p = 0.046). Confocal microscopy confirmed bright heterogeneous surface expression of PR1 on AML, but was nearly absent on healthy donor PB. Interestingly, PR1 expression was higher on LSC (CD34+CD38-lin-) (76±44) compared to healthy donor HSC (CD34+CD38-lin-) (13.4), which suggests that only differentiated myeloid cells might be more susceptible to 8F4 toxicity. To study whether 8F4 mediated specific lysis of PR1+ cells, we determined cell-mediated cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) of 8F4 compared to isotype control. 8F4 induced 51% specific lysis by CDC and 15% specific lysis by ADCC of PR1-pulsed T2 cells, which was dose-dependent on both 8F4 and PR1, with no lysis of target cells by isotype control. Moreover, 8F4 induced CDC-mediated lysis of blasts from 5/5 HLA-A2+ AML patients, but not blasts from an HLA-A2- AML patient (see Table 1), or mononuclear cells from HLA-A2+ healthy donors. To study the in vivo effects of 8F4 on AML, we used an established xenogeneic model of human AML in NOD/SCID mice. 1 × 106 blasts from 4 HLA-A2+ AML patients (including the HLA-A2+ patients susceptible to 8F4-mediated lysis in vitro) and 1 HLA-A2- patient were mixed for one hour under serum-free conditions with 8F4 or isotype control antibody and adoptively transferred to NOD/SCID mice. Engraftment of AML was compared on day 14 and AML growth was confirmed on day 28 by comparing FACS analysis and histopathology of BM from sacrificed animals. 8F4 treatment of AML cells, but not isotype treatment, abrogated engraftment of 4/4 HLA-A2+ AML patients, but not of 1 HLA-A2- AML patient on d14. By day 28, AML expanded by 150% in one surviving isotype-treated animal. Thus, 8F4-mediated specific lysis of AML and LSC by CDC and ADCC mechanisms in vitro were confirmed by in vivo results of AML engraftment. Taken together, these results show that 8F4, a novel TCR-mimic antibody that targets only PR1/HLA-A2+ cells, mediates lysis of AML blasts and LSC, but not total normal BM cells or HSC. Therefore, 8F4 may be useful as an immunotherapeutic agent in the treatment of HLA-A2+ myeloid leukemia, and as the first antibody therapy that may target and eliminate LSC. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4616-4616
Author(s):  
Rosy Dabas ◽  
Poonam Dharmani ◽  
Monica Modi ◽  
Tiffany Van Slyke ◽  
Joanne Luider ◽  
...  

Abstract Background: Although curative treatment for acute myeloid leukemia, the success of allogeneic hematopoietic cell transplantation (HCT) is limited due to leukemia relapse and graft-versus-host disease (GvHD). Rabbit anti-thymocyte globulin (ATG) used for GvHD prophylaxis does not increase relapse (Walker et al: Lancet Oncol 2016). The non-increase in relapse could be because ATG has a direct anti-leukemic effect (Dabas et al: BBMT 2016). Multiple studies have suggested that a high number of leukemic stem cells (LSCs, also called leukemia initiating cells) remaining after therapy is associated with relapse. Therefore, targeting LSCs may be important for treating or preventing relapse. We demonstrated ATG's cytotoxic effect against acute myeloid leukemia (AML) blasts (Dabas et al: BBMT 2016). However, ATG's effect on LSCs has not been evaluated. In this study, we investigated in vitro ATG-induced complement-independent cytotoxicity (CIC, presumably direct induction of apoptosis) and complement-dependent cytotoxicity (CDC) against LSCs. This was also compared to ATG-induced CIC and CDC against healthy hematopoietic stem cells (HSCs). Methods:Frozen peripheral blood mononuclear cells (PBMNCs) from 15 patients newly diagnosed with AML were used as the source of LSCs. Frozen mononuclear cell apheresis samples from 15 healthy stem cell donors were used as the source of HSCs. We measured by flow cytometry CIC and CDC induced by ATG at 10 mg/L (the median peak concentration achieved with 4.5 mg/kg ATG given over day -2, -1 and 0) and 50 mg/L. CIC was induced by incubating cells with ATG in the presence of heat-inactivated (complement-depleted) fetal bovine serum for 4 hours. As a negative control for the calculation of background, cells were cultured under the same conditions except without ATG. CDC was induced by incubating cells with ATG in the presence of human serum as a source of complement for 15 minutes. As a negative control (for background), cells were incubated with ATG in the presence of heat-inactivated human serum (no complement). The LSCs and HSCs were phenotypically defined as CD45dim/neg, side scatterlow, CD34+, CD38neg and negative for CD14, CD16, CD19, CD56, CD235a and CD41a. For CIC, dying/dead cells were identified as Annexin V positive (Annexin V+). For CDC, dead cells were identified as 7-amino-actinomycin D positive (7AAD+). Results: ATG induced both CIC and CDC of LSCs at the concentration of 50 mg/L but not 10 mg/L. For CIC, the median percent Annexin V+ LSCs after incubation with 50 mg/L ATG was 22.3% vs 18.4% for background (p=0.0043, Wilcoxon matched pairs test). For CDC, the median percent 7AAD+ LSCs after incubation with 50 mg/L ATG was 37.2% vs 2% background (p=0.0004, Wilcoxon matched pairs test). Next, we compared the cytotoxicity of 50 mg/L ATG against LSCs versus healthy HSCs. For CIC, a significantly greater percent of LSCs than HSCs was killed (P=0.0363, Mann-Whitney rank sum test) (Figure 1). Similarly, for CDC, there was a trend toward a greater percent of LSCs than HSCs killed (P=0.0971) (Figure 2). Conclusion: ATG kills LSCs in vitro via both CIC and CDC. However, the degree of the killing is minor and only observed at a higher ATG concentration than typically achieved in patients. Our data also suggest that if high dose ATG was used in patients (resulting in ≥50 mg/L concentration), LSCs could be killed to a greater degree than healthy HSCs. Figure 1 LSCs are more sensitive to ATG mediated CIC than healthy HSCs. Mononuclear cells from 15 patients newly diagnosed with AML and cells from leukapheresis products of 15 healthy stem cell donors were incubated with ATG (50 mg/L) in the absence of complement. After 4 hours, the percentage of dead or dying (Annexin V+) cells was measured in LSCs and HSCs. Adjusted percentage (background subtracted) is shown for each patient/donor. Medians are shown as horizontal lines. Figure 1. LSCs are more sensitive to ATG mediated CIC than healthy HSCs. Mononuclear cells from 15 patients newly diagnosed with AML and cells from leukapheresis products of 15 healthy stem cell donors were incubated with ATG (50 mg/L) in the absence of complement. After 4 hours, the percentage of dead or dying (Annexin V+) cells was measured in LSCs and HSCs. Adjusted percentage (background subtracted) is shown for each patient/donor. Medians are shown as horizontal lines. Figure 2 LSCs are more sensitive to ATG mediated CDC than healthy HSCs. Mononuclear cells from 15 patients newly diagnosed with AML and cells from leukapheresis products of 15 healthy stem cell donors were incubated with ATG (50 mg/L) in the presence of complement. After 15 minutes, the percentage of dead (7AAD+) cells was measured in LSCs and HSCs. Adjusted percentage (background subtracted) is shown for each patient/donor. Medians are shown as horizontal lines. Figure 2. LSCs are more sensitive to ATG mediated CDC than healthy HSCs. Mononuclear cells from 15 patients newly diagnosed with AML and cells from leukapheresis products of 15 healthy stem cell donors were incubated with ATG (50 mg/L) in the presence of complement. After 15 minutes, the percentage of dead (7AAD+) cells was measured in LSCs and HSCs. Adjusted percentage (background subtracted) is shown for each patient/donor. Medians are shown as horizontal lines. Disclosures Daly: Sanofi-Genzyme: Other: Advisory Board.


2016 ◽  
Vol 13 (2) ◽  
pp. 248-259 ◽  
Author(s):  
Hong-Sheng Zhou ◽  
Hong-Sheng Zhou ◽  
Bing Z. Carter ◽  
Michael Andreeff ◽  
Bing Z. Carter ◽  
...  

2021 ◽  
Author(s):  
Huixin Li ◽  
Shunichiro Yasuda ◽  
Satoru Aoyama ◽  
Chenyang Zhang ◽  
Yohei Kawano ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document