The antiparasitic agent ivermectin induces chloride-dependent membrane hyperpolarization and cell death in leukemia cells

Blood ◽  
2010 ◽  
Vol 116 (18) ◽  
pp. 3593-3603 ◽  
Author(s):  
Sumaiya Sharmeen ◽  
Marko Skrtic ◽  
Mahadeo A. Sukhai ◽  
Rose Hurren ◽  
Marcela Gronda ◽  
...  

Abstract To identify known drugs with previously unrecognized anticancer activity, we compiled and screened a library of such compounds to identify agents cytotoxic to leukemia cells. From these screens, we identified ivermectin, a derivative of avermectin B1 that is licensed for the treatment of the parasitic infections, strongyloidiasis and onchocerciasis, but is also effective against other worm infestations. As a potential antileukemic agent, ivermectin induced cell death at low micromolar concentrations in acute myeloid leukemia cell lines and primary patient samples preferentially over normal hematopoietic cells. Ivermectin also delayed tumor growth in 3 independent mouse models of leukemia at concentrations that appear pharmacologically achievable. As an antiparasitic, ivermectin binds and activates chloride ion channels in nematodes, so we tested the effects of ivermectin on chloride flux in leukemia cells. Ivermectin increased intracellular chloride ion concentrations and cell size in leukemia cells. Chloride influx was accompanied by plasma membrane hyperpolarization, but did not change mitochondrial membrane potential. Ivermectin also increased reactive oxygen species generation that was functionally important for ivermectin-induced cell death. Finally, ivermectin synergized with cytarabine and daunorubicin that also increase reactive oxygen species production. Thus, given its known toxicology and pharmacology, ivermectin could be rapidly advanced into clinical trial for leukemia.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4370-4370
Author(s):  
Guo Kunyuan ◽  
Miaorong She ◽  
Haiyan Hu ◽  
Xinqing Niu ◽  
Sanfang Tu ◽  
...  

Abstract 2-Methoxyestradiol (2-ME) is a new anticancer agent currently under investigation for treatment of leukemia. We evaluated the effects of 2-ME-induced apoptosis in two myeloid leukemia cell lines (U937 and HL-60) in association with reactive oxygen species (ROS) generation. We found that 2-ME resulted in viability decrease in a dose-dependent manner, generated ROS: nitric oxide and superoxide anions, and mitochondria damage. 2-ME-induced apoptosis correlated with increase in ROS. Quenching of ROS with N-acetyl-L-cysteine protected leukemia cells from the cytotoxicity of 2-ME and prevented apoptosis induction by 2-ME. Furthermore, addition of manumycin, a farnesyltransferase inhibitor, demonstrated by our previous studies that induced apoptosis of leukemic cells and induced ROS, significantly enhanced the apoptosis-induced by 2-ME. In conclusion, cellular ROS generation play an important role in the cytotoxic effect of 2-ME. It is possible to use ROS-generation agents such as manumycin to enhance the antileukemic effect. Such a combination strategy need the further in vivo justify and may have potential clinical application.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 410-410 ◽  
Author(s):  
Sumaiya Sharmeen ◽  
Marko Skrtic ◽  
Mahadeo Sukhai ◽  
Joyce Sun ◽  
Rose Hurren ◽  
...  

Abstract Abstract 410 FDA-approved drugs with previously unrecognized anti-cancer activity could be rapidly repurposed for this new indication. To identify such compounds, we compiled a library of known drugs with high maximal tolerated doses and well-known toxicity profiles. We screened this library in a dose-response manner in 4 leukemia cell lines to identify cytotoxic compounds as measured by the MTS assay. From these screens, we identified the anti-parasitic agent ivermectin (IVM) that induced cell death at low micromolar concentrations in all four leukemia lines tested. IVM is a derivative of avermecin B1 and licensed for the treatment of strongyloidiasis and onchocerciasis parasitic infections, but also effective against other worm infestations (e.g., ascariasis, trichuriasis and enterobiasis). To evaluate the effects of IVM as a potential anti-cancer agent, leukemia and myeloma (n = 9) cell lines were treated with increasing concentrations of IVM. 72 hours after incubation, cell viability was determined by the MTS assay. IVM decreased the viability of 3/5 leukemia cell lines with an LD50 < 5uM and all other tested malignant cell lines with an LD50 < 10uM. Cell death was confirmed by Trypan blue staining and Annexin V staining. In clonogenic growth assays, 6uM IVM reduced clonogenic growth by ≥ 40% in 3/6 primary AML samples, but <15% in 3/3 samples of normal hematopoietic mononuclear cells. Given the effects of IVM in cell lines, we evaluated the drug in mouse models of leukemia. Here, K562, OCI-AML2, and MDAY-D2 leukemia cells were implanted subcutaneously into the flanks of sublethally irradiated NOD/SCID mice. One week after implantation, when the tumors were palpable, mice were treated with IVM at 3mg/kg/day by oral gavage or buffer control. Compared to control, IVM decreased tumor volume and weight in all 3 xenograft models by up to 72% without observable toxicity. Of note, a dose of 3mg/kg in mice translates to a dose of 0.24 mg/kg in humans based on scaling of body weight and surface area. This dose is readily achievable in human as patients routinely receive 0.2mg/kg for the treatment of parasitic disease and overdoses of up to 6g were not toxic. As an anti-parasitic, IVM binds and activates chloride ion channels, so we tested the effects of IVM on chloride flux in leukemia cells. OCI AML2 cells were treated with increasing concentrations of IVM and changes in intracellular chloride were measured using the fluorescent dye 6-methoxy-N-(3-sulfopropyl)quinolinium and flow cytometry. IVM increased intracellular chloride ion concentrations within 30 minutes of treatment. Chloride influx was accompanied by plasma membrane hyperpolarization within 1 hr of treatment, but no change in mitochondrial membrane potential was noted up to 24 hours after treatment. Plasma membrane hyperpolarization was dependent on chloride influx, as treatment with IVM in chloride-free media did not induce membrane hyperpolarization. Alternations in intracellular chloride and membrane hyperpolarization can lead to increased reactive oxygen species (ROS) generation. Therefore, we measured changes in ROS after treatment with IVM. IVM (6uM) increased ROS generation in OCI-AML2 cells up to 2 ± 0.2 fold within 4 hours of treatment. Increased ROS generation appeared functionally important for IVM-induced cell death as pre-treatment with the antioxidant N-acetyl-L-cysteine (NAC) inhibited IVM-induced cell death. Further supporting a mechanism of cell death related to increased ROS, IVM treatment increased expression of STAT1, IFIT3, OAS1, and TRIM22, members of the STAT1 signaling pathway that are known to be upregulated upon ROS generation. Cytarabine and daunorubicin are used in the treatment of AML and increase ROS production through mechanisms related to DNA damage. Therefore, we evaluated the combination of IVM and cytarabine and daunorubicin. By isobologram analysis, IVM synergized with cyatarabine (CI=0.51, 0.58, 0.65 at ED25, ED50, ED75, respectively) and daunorubicin (CI=0.48, 0.51, 0.54 at ED25, ED50, ED75, respectively). Thus, in summary, IVM activates chloride channels in leukemia cells leading to membrane hyperpolarization and increased ROS generation. In addition, it demonstrated preclinical activity in this disease at pharmacologically achievable concentrations. Therefore, IVM could be rapidly repurposed for the treatment of leukemia and highlights a potential new therapeutic strategy for this disease. Disclosures: Off Label Use: Ivermectin is an antiparasitic agent, licensed for the treatment of strongyloidiasis and onchocerciasis parasitic infections, but also effective against other worm infestations (e.g., ascariasis, trichuriasis and enterobiasis).


2003 ◽  
Vol 78 (3) ◽  
pp. 219-225 ◽  
Author(s):  
Hiroaki Goto ◽  
Hiroyuki Takahashi ◽  
Hisaki Fujii ◽  
Koichiro Ikuta ◽  
Shumpei Yokota

2021 ◽  
pp. 105252
Author(s):  
Carlos Angelé-Martínez ◽  
Fathima S. Ameer ◽  
Yash Raval ◽  
Guohui Huang ◽  
Tzuen-Rong J. Tzeng ◽  
...  

2009 ◽  
Vol 255 (1-2) ◽  
pp. 61-68 ◽  
Author(s):  
Kengo Yoshida ◽  
Yoshiko Kubo ◽  
Yoichiro Kusunoki ◽  
Yukari Morishita ◽  
Hiroko Nagamura ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document