Toll-like receptor 7 agonist induces apoptosis of human B-lineage acute lymphocytic leukemia cells in vitro and in vivo

2008 ◽  
Vol 26 (15_suppl) ◽  
pp. 3033-3033
Author(s):  
X. Liang ◽  
J. Tolar ◽  
J. S. Miller ◽  
T. W. LeBien ◽  
B. R. Blazar ◽  
...  
Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 706-706 ◽  
Author(s):  
Xueqing Liang ◽  
Jakub Tolar ◽  
Jeffery S. Miller ◽  
Tucker W. LeBien ◽  
Bruce R. Blazar ◽  
...  

Abstract Acute lymphocytic leukemia (ALL) is the most common childhood leukemia and remains a difficult disease with poor survival in patients who have failed standard therapy. New therapeutic strategies are needed to achieve longer survival and improved cure rates in both pediatric and adult ALL patients. In this study, we show human B-lineage acute lymphocytic leukemia (B-ALL) cell lines (6/6 tested) and CD19+CD10+ primary B-ALL cells from patients (8/8 tested) express TLR7 mRNA by real-time RT-PCR and TLR7 proteins by Western blot. Triggering TLR7 on B-ALL cells with a TLR7 agonist (imiquimod) significantly increases the cell surface expression of molecules essential for T cell activation (CD40, CD54, CD80, CD86, and HLA-DR), the ligands for NKG2D and ligands for natural cytotoxicity receptors (NKp30, NKp44, and NKp46) which regulate NK-mediate killing. Thus, TLR7 signaling enhances the immunogenicity of B-ALL cells and makes them more suitable targets for T cell and NK cell mediated attack. Most importantly, TLR7 agonists strongly suppress in vitro growth of B-ALL cell lines (RS4;11, BLIN-1) and induces profound apoptosis of primary B-ALL cells from patients in culture in a TLR7 agonist dose-dependent manner. Both t(4;11)-positive RS4;11 cells and t(4;11)-negative BLIN-1 cells proliferate rapidly in culture with a 30–40 fold increases of leukemia cell number in 7 days. The addition of TLR7 agonist at 10 ug/ml fully inhibit the growth of RS4;11 and BLIN-1 cells in culture. Furthermore, TLR7 agonist treatment dramatically induces apoptosis of primary B-ALL cells isolated from the patients (2/2 with t(9;22), 6/6 without t(9;22)) with 0.4%–13.3% leukemia cells left at day 5 of culture. The TLR7 agonist-mediated apoptotic death of B-ALL cells was conformed by viable cell counts, TMRE staining, and, Western blots of the activation and cleavage of caspases. To study the in vivo therapeutic effects of TLR7 agonist against human B-ALL, RS4;11 and BLIN-1 cells were luciferase labeled and injected into NOD/SCID mice. Both RS4;11 and BLIN-1 leukemia cells engrafted in multiple organs (BM, spleen, liver, lymph nodes, kidney) resulting in uniform lethality of RS4;11 mice in 8 weeks and BLIN-1 mice in 12 weeks, respectively. Flow cytometry and tissue staining results confirmed that these organs were massively infiltrated with human CD45+19+ leukemia cells. To determine whether TLR7 preincubation of RS4;11 or BLIN-1 cells would prolong survival due to an apoptotic effect, cohorts of mice were injected with a lethal dose of RS4;11 or BLIN-1 cells with or without pre-incubation with TLR7 agonist. Mice receiving TLR7 agonist pre-pretreated B-ALL cells had a significant increase in long-term survival rate and significant reduction in tumor burden at the time points evaluated. These in vivo results confirm previous in vitro findings and suggest that TLR agonist-treated B-ALL cells are programmed to die. The antitumor efficacy of systemic administration of TLR7 agonist in NOD/SCID mice with established B-ALL is being investigated using these xenograft mouse models. These results form the basis for a clinical trial of systemic TLR7 agonist administration for treating patients with B-ALL. In summary, we have shown that TLR7 targeting increases B-ALL immunogenicity and directly induces B-ALL apoptosis, providing new insights into the biology and therapy of B-ALL.


Blood ◽  
2016 ◽  
Vol 127 (5) ◽  
pp. 582-595 ◽  
Author(s):  
Marwan Kwok ◽  
Nicholas Davies ◽  
Angelo Agathanggelou ◽  
Edward Smith ◽  
Ceri Oldreive ◽  
...  

Key PointsATR inhibition is synthetically lethal to TP53- or ATM-defective CLL cells. ATR targeting induces selective cytotoxicity and chemosensitization in TP53- or ATM-defective CLL cells in vitro and in vivo.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 248-248
Author(s):  
Alice Bonato ◽  
Riccardo Bomben ◽  
Supriya Chakraborty ◽  
Giulia Felician ◽  
Claudio Martines ◽  
...  

Abstract Inactivating mutations in NF-kB pathway genes, such as the NF-kB inhibitor NFKBIE, are among the more frequent genetic lesions in chronic lymphocytic leukemia (CLL). However, the role of these genetic lesions in CLL pathogenesis and treatment resistance is still largely unknown and requires further study in in vivo models of the disease. To this end, we generated transplantable murine leukemias with inactivating NFKBIE mutations and investigated their impact on leukemia growth and response to ibrutinib (IBR) treatment. The NFKBIE mutations were introduced by CRISPR/Cas9 editing in two recently established autoreactive leukemia lines derived from the Eμ-TCL1 murine CLL model. These cell lines proliferate spontaneously in vitro in a BCR-dependent manner, but also respond with increased proliferation to certain microenvironmental signals, such as those generated by Toll-like receptor (TLR) stimulation (Chakraborty S et al, Blood 2021). To investigate whether NFKBIE mutations can affect the proliferation of these cell lines in vitro, we performed competition experiments with mixed cultures of cells with wild type and mutated NFKBIE. Analysis of the clonal composition after 2 weeks showed no change in the mutant allele frequency (MAF), suggesting that NFKBIE mutations do not affect the spontaneous in vitro growth of the immortalized leukemia cells. However, repeated TLR or BCR stimulation of these cells with CpG-DNA, LPS, anti-IgM or autoantigen resulted in a 2-3 fold increase in MAF, suggesting that NFKBIE mutations provide a growth advantage when the cells are exposed to certain microenvironmental signals (n=3 experiments/condition, P<0.05 for each condition). To investigate the impact of NFKBIE mutations on leukemia growth in vivo, the same cells were transplanted by intraperitoneal injection in wild type mouse recipients (n=8) and the clonal composition was determined 3 weeks later by MAF analysis of cells isolated from peritoneal cavity (PC), blood and spleen. A significant increase in MAF was observed only in leukemia cells isolated from the spleen (P<0.05), suggesting that microenvironmental signals that positively select NFKBIE-mutated cells are available only in certain tissue compartments. Because mutations in other NF-kB pathway genes have been associated with resistance to IBR in mantle cell lymphoma, we next investigated whether NFKBIE mutations can also affect the response to IBR treatment. In vitro BrdU-incorporation experiments showed that IBR inhibits the proliferation of cells with mutated NFKBIE to a significantly lesser extent compared to cells with wild type NFKBIE (% proliferating cells with wild type and mutated NFKBIE, respectively, cultured without IBR: 90% vs 88%, P=n.s., with 0.2 μM IBR: 57% vs 73%, P<0.001, with 1.0 μM IBR: 28% vs 53%, P<0.001). Consistent with this finding, positive selection of NFKBIE-mutated cells was observed in the presence of IBR after 14 days in mixed culture competition experiments (mean MAF without IBR 47%, with 0.2 μM IBR 61%, p=0.032, with 1.0 μM IBR 64%, p=0.034). The greater resistance of NFKBIE-mutated cells to IBR was further validated by in vivo competition experiments showing a significantly greater increase in MAF in mice treated with IBR compared to controls in all three investigated compartments (n=4 mice/group, PC: P=0.029, blood P=0.029, spleen: P=0.001). To validate these findings in the clinical setting, we investigated the presence of NFKBIE mutations in a cohort of 84 IBR-treated CLL patients. Mutations of NFKBIE were detected at pre-treatment in 10/84 patients, 7/10 with >10% VAF values. Kaplan Meier analysis showed a trend towards reduced progression-free and overall survival from the beginning of IBR treatment for NFKBIE-mutated cases (Figure 1A). Analysis of an extended cohort of over 200 cases is ongoing and will be presented at the meeting. Finally, to investigate whether leukemic cells with mutated NFKBIE remain sensitive to other BCR inhibitors, we tested their growth in the presence of the PI3K inhibitor idelalisib or SYK inhibitor fostamatinib (Figure 1B). In contrast to IBR, both drugs inhibited the proliferation of NFKBIE-mutated cells in vitro, with a greater effect observed with idelalisib. Collectively, these data demonstrate that NFKBIE mutations can reduce the response to IBR treatment and suggest that such cases may benefit more from treatment with a PI3K inhibitor. Figure 1 Figure 1. Disclosures Marasca: Janssen: Honoraria, Other: Travel grants; AstraZeneca: Honoraria; AbbVie: Honoraria, Other: Travel grants. Tafuri: Roche: Research Funding; Novartis: Research Funding; Celgene: Research Funding. Laurenti: Janssen: Consultancy, Honoraria; AstraZeneca: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria, Research Funding; Roche: Honoraria, Research Funding; Gilead: Honoraria; BeiGene: Honoraria. Gattei: abbVie: Research Funding; Janssen: Research Funding; Menarini: Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2040-2040 ◽  
Author(s):  
Marina Konopleva ◽  
Juliana Benito ◽  
Yue-Xi Shi ◽  
Sergej Konoplev ◽  
Steven M. Kornblau ◽  
...  

Abstract Abstract 2040 Poster Board II-17 The main challenge in the treatment of acute lymphocytic leukemia (ALL) is overcoming resistance to chemotherapy. Recent studies indicate that interactions between leukemia cells and bone marrow (BM) microenvironment promote leukemia cell survival and confer resistance to drugs commonly used to treat ALL. We investigated whether hypoxia was a contributing factor in the protective role of the BM microenvironment. We found that the Hypoxia-Inducible-Factor 1a (HIF-1a; a marker normally expressed by only a few hematopoietic cells) was expressed in 68% of BM biopsies from patients with B-lineage ALL (n=53). Expression of HIF-1α detected either histochemically (n=53, p=0.023) or by Reverse Phase Protein Arrays (RPPA, n=116, p=0.0013) inversely correlated with survival of patients with newly diagnosed B-lineage ALL. Silencing of HIF-1α with siRNA, or blockade of mTOR signaling with rapamycin derivatives, reduced expression of the glucose transporter Glut-1, diminished glucose flux, decreased glycolytic rate and ATP production and sensitized leukemic cells to the pro-apoptotic effects of chemotherapeutic agents under hypoxic conditions. In line with this findings, we observed a marked expansion of hypoxic BM areas in immunodeficient mice engrafted with the ALL cell line Nalm6 or with primary ALL cells, as detected by administration of the reductive 2-nitroimidazole compound pimonidazole (PIM), which forms stable adducts in hypoxic regions. Altogether, these findings provided a rationale for examining the effects of hypoxia-activated pro-drugs or HIF-1a inhibitors to eliminate ALL progenitor cells within hypoxic niches. To this end, we tested PR-104, a hypoxia-activated dinitrobenzamide mustard currently undergoing Phase II trials in solid tumors. Under hypoxic conditions, this agent is reduced to hydroxylamine and amine metabolites that in turn induce DNA cross-links and cell death (Patterson et al., Clin Can Res 2007). In vitro, 25μM PR-104 induced hypoxia-selective cell death in Nalm6 ALL cells with 80% Annexin V-positivity at 0.1% O2, 46% at 1%O2 compared to 13% at 21%O2. The anti-leukemic efficacy of PR-104 was next examined in the in vivo leukemia models. Administration of PR-104 (250 mg/kg IP TIW for two weeks) prolonged survival of NOD/Scid/IL2Rg-KO (NOG) mice injected with cells from primary refractory FLT3-mutated AML, and decreased leukemia burden as indicated by histopathological analyses of CD45 positive cells in the BM, spleen, lung and liver. Notably, analysis of PIM distribution indicated clearance of the hypoxic leukemic niches. In NOG mice injected with leukemic cells from an infant with MLL-rearranged B-lineage ALL, PR-104 at 200 mg/kg IP on days 1, 2 and 6 resulted in a dramatic decrease in the percentage of circulating leukemic CD45+ cells on day 15 (control, 92%±6%; treated, 9%±4%; n=7 mice/group). The therapeutic effect of the drug was also tested in a Nalm6-luciferase ALL model where PR-104 administration resulted in decreased tumor burden as determined by luciferase activity and prolonged survival of the PR-104 treated as compared to control mice (p=0.006). Similar to the models of human leukemia, analysis of BM sections of control mice showed extensive areas of hypoxia (PIM-positive) in close proximity to GFP-positive leukemia cells in contrast to the treated mice in which only discrete areas of PIM positivity were detectable. Altogether, these findings strongly suggest that targeting hypoxia is feasible and may increase the sensitivity of ALL cells to chemotherapy. If successful, this approach of targeting hypoxic microenvironment, alone or in combination with other chemotherapeutic or targeted agents, may significantly impact ALL therapy and ultimately improve patient survival. Figure 1. Co-localization of hypoxic PIM(+) areas with GFP positive HALMG tumor areas in bone marrow of control but not of PR-104 treated mice. Figure 1. Co-localization of hypoxic PIM(+) areas with GFP positive HALMG tumor areas in bone marrow of control but not of PR-104 treated mice. Disclosures: No relevant conflicts of interest to declare.


ACS Omega ◽  
2020 ◽  
Vol 5 (31) ◽  
pp. 19844-19852
Author(s):  
Li Xia ◽  
Dali Kang ◽  
Dan Wan ◽  
Chu Chu ◽  
Meizi Chen ◽  
...  

Neoplasia ◽  
2010 ◽  
Vol 12 (4) ◽  
pp. 326-IN6 ◽  
Author(s):  
Rajesh Kumar Gandhirajan ◽  
Peter Anton Staib ◽  
Katharina Minke ◽  
Iris Gehrke ◽  
Günther Plickert ◽  
...  

Marine Drugs ◽  
2021 ◽  
Vol 19 (10) ◽  
pp. 565
Author(s):  
Ye Yuan ◽  
Te Li ◽  
Tingting Wang ◽  
C. Benjamin Naman ◽  
Jing Ye ◽  
...  

LC-MS/MS-based molecular networking facilitated the targeted isolation of a new cyclic hexadepsipeptide, cymodepsipeptide (1), and two known analogues, RF–2691A (2) and RF–2691B (3), from the fungus Cymostachys sp. NBUF082 that was derived from a mesophotic zone Aaptos sponge collected near Apo Island. The constitution and configuration of 1 was elucidated through 1D and 2D NMR-spectroscopy, high resolution mass-spectrometry, and chemical degradations including Marfey’s analysis and chiral HPLC. It was observed that 1 was moderately cytotoxic against CCRF-CEM human acute lymphocytic leukemia cells in vitro with the IC50 value of 9.2 ± 1.1 μM.


Blood ◽  
2013 ◽  
Vol 121 (18) ◽  
pp. 3658-3665 ◽  
Author(s):  
Katrin S. Reiners ◽  
Daniela Topolar ◽  
Alexander Henke ◽  
Venkateswara R. Simhadri ◽  
Jörg Kessler ◽  
...  

Key Points Exosomal NKp30-ligand BAG6 is crucial for detection of tumor cells by NK cells in vitro and in vivo. Soluble plasma factors including BAG6 suppress NK cell cytotoxicity and promote evasion of CLL cells from NK cell anti-tumor activity.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5303-5303
Author(s):  
Suping Zhang ◽  
Hsien Lai ◽  
Grace Liu ◽  
Laura Rassenti ◽  
Michael Y. Choi ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) cells express high levels of CD44, a cell-surface glycoprotein receptor for hyaluronic acid (HA). We found that a mAb specific for CD44 was directly cytotoxic for leukemia B cells, but had little effect on normal B cells. Moreover, this anti-CD44 mAb could induce CLL cells that expressed the zeta-associated protein of 70 kDa (ZAP-70) to undergo caspase-dependent apoptosis, independent of complement or cytotoxic effector cells (Proc Natl Acad Sci, USA 2013, PMID: 23530247). The cytotoxic effect of this mAb was not mitigated when the CLL cells were co-cultured with mesenchymal stromal cells (MSCs) or hyaluronic acid or when they were stimulated via ligation of the B-cell receptor with anti-µ. A6 (Angstrom Pharmaceuticals) is an 8-amino acid peptide that has marked homology with a linear sequence of CD44. A6 can bind CD44 within a region of the ligand-binding domain, leading to inhibition of the migration and metastatic potential of CD44-expressing cancer cells in vitro and in vivo (Mol Cancer Ther, 2011 PMID: 21885863). We evaluated the cytotoxic activity of A6 against primary leukemia cells of patients with CLL (n = 22). We found that A6 peptide also was directly cytotoxic for CLL cells isolated from different patients in a dose-dependent manner at concentrations that may be achieved in vivo. The A6 peptide appeared less cytotoxic for CLL cells than the intact anti-CD44 mAb, but still had greater direct cytotoxicity for CLL cells that expressed ZAP-70 than for CLL cells that were ZAP-70 negative. Furthermore, the A6 peptide had negligible effect on the viability of lymphocytes isolated from the blood of healthy donors (n = 3). Because clinical studies have found the A6 peptide to be well-tolerated and without dose-limiting toxicity in patients with solid tumors who have been treated to date (N = 40), a clinical study is planned to evaluate the safety and activity of the A6 peptide in the treatment of patients with CLL. Disclosures: Howell: Angstrom Phamaceuticals: Membership on an entity’s Board of Directors or advisory committees. Finlayson:Angstrom Phamaceuticals: Employment.


Sign in / Sign up

Export Citation Format

Share Document