Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2020

Open Access 01.12.2020 | Research

The SMAC mimetic LCL-161 selectively targets JAK2V617F mutant cells

verfasst von: Brianna M. Craver, Thanh Kim Nguyen, Jenny Nguyen, Hellen Nguyen, Christy Huynh, Sarah J. Morse, Angela G. Fleischman

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2020

Abstract

Background

Evasion from programmed cell death is a hallmark of cancer and can be achieved in cancer cells by overexpression of inhibitor of apoptosis proteins (IAPs). Second mitochondria-derived activator of caspases (SMAC) directly bind to IAPs and promote apoptosis; thus, SMAC mimetics have been investigated in a variety of cancer types. particularly in diseases with high inflammation and NFĸB activation. Given that elevated TNFα levels and NFĸB activation is a characteristic feature of myeloproliferative neoplasms (MPN), we investigated the effect of the SMAC mimetic LCL-161 on MPN cell survival in vitro and disease development in vivo.

Methods

To investigate the effect of the SMAC mimetic LCL-161 in vitro, we utilized murine and human cell lines to perform cell viability assays as well as primary bone marrow from mice or humans with JAK2V617F–driven MPN to interrogate myeloid colony formation. To elucidate the effect of the SMAC mimetic LCL-161 in vivo, we treated a JAK2V617F–driven mouse model of MPN with LCL-161 then assessed blood counts, splenomegaly, and myelofibrosis.

Results

We found that JAK2V617F-mutated cells are hypersensitive to the SMAC mimetic LCL-161 in the absence of exogenous TNFα. JAK2 kinase activity and NFĸB activation is required for JAK2V617F-mediated sensitivity to LCL-161, as JAK or NFĸB inhibitors diminished the differential sensitivity of JAK2V617F mutant cells to IAP inhibition. Finally, LCL-161 reduces splenomegaly and may reduce fibrosis in a mouse model of JAK2V617F-driven MPN.

Conclusion

LCL-161 may be therapeutically useful in MPN, in particular when exogenous TNFα signaling is blocked. NFĸB activation is a characteristic feature of JAK2V617F mutant cells and this sensitizes them to SMAC mimetic induced killing even in the absence of TNFα. However, when exogenous TNFα is added, NFĸB is activated in both mutant and wild-type cells, abolishing the differential sensitivity. Moreover, JAK kinase activity is required for the differential sensitivity of JAK2V617F mutant cells, suggesting that the addition of JAK2 inhibitors to SMAC mimetics would detract from the ability of SMAC mimetics to selectively target JAK2V617F mutant cells. Instead, combination therapy with other agents that reduce inflammatory cytokines but preserve JAK2 signaling in mutant cells may be a more beneficial combination therapy in MPN.
Begleitmaterial
Additional file 4: Figure S4. Colony formation shown in Fig. 3 separated by erythroid and G/M colonies. (A) Erythroid and (B) G/M colony formation from MPN patients and normal controls with increasing concentrations of LCL-161. (C) Erythroid and (D) G/M colony formation from MPN patients and normal controls with 10 ng/ml TNFα + increasing concentrations of LCL-161.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s40164-019-0157-6.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BCA
bicinchoninic acid assay
BSA
bovine serum albumin
CALR
calreticulin
EPO
erythropoietin
FBS
fetal bovine serum
GFP
green fluorescent protein
G/M
granulocyte/monocyte
IAP
inhibitor of apoptosis proteins
IL-3
interleukin-3
JAK
Janus kinase
MPL
thrombopoietin receptor
MPN
myeloproliferative neoplasm
NFKB
nuclear factor kappa-light-chain-enhancer of activated B cells
PI
propidium iodide
PBMNC
peripheral blood mononuclear cells
SCF
stem cell factor
SMAC
second mitochondria-derived activator of caspases
TNFα
tumor necrosis factor-alpha
PBS
phosphate buffered saline

Background

Evasion of programmed cell death is a common survival technique utilized by cancer cells and one way these cells can evade apoptosis is by upregulating anti-apoptotic proteins such as inhibitor of apoptosis proteins (IAPs) [1]. IAPs inhibit apoptosis by directly binding to and inhibiting pro-apoptotic caspases. However, when a cell is triggered to undergo apoptosis, second mitochondria-derived activator of caspases (SMAC) is released into the cytosol and bind IAP proteins, leading to their degradation. Degradation of IAP proteins induces apoptosis by freeing pro-apoptotic caspases and allowing them to induce cell death. Small molecules which mimic the antagonistic effects of SMAC and lead to degradation of IAP proteins, termed SMAC mimetics, are actively being investigated in both hematologic malignancies and solid tumors [2, 3]. In preclinical studies, SMAC mimetics induce apoptosis in cancer cells directly or indirectly by priming these cells for killing by other cytotoxic agents. SMAC mimetics sensitize cells to TNFα-induced cell death [4]. TNFα is a master regulator of inflammation and induces NFĸB activation among other signaling pathways. However, NFĸB activation is critical for SMAC mimetic-induced apoptosis [5]. In a study involving primary AML samples, sensitivity to a SMAC mimetic correlated with activation of TNF signaling and low XIAP expression [6]. Therefore, SMAC mimetics may be particularly useful in cancers associated with high inflammation and NFĸB activation.
Myeloproliferative neoplasm (MPN) is a chronic hematologic malignancy characterized by chronic inflammation [79], high TNFα levels in serum [10], activation of NFĸB [11] and reduced expression of XIAP [12]. MPN is characterized by the somatic acquisition of a mutation in either JAK2 (JAK2V617F) [1317] or calreticulin (CALR) [18, 19] in a hematopoietic stem cell. This mutant clone expands, producing excessive numbers of mature myeloid cells that also carry the driver mutation. The current goals of therapy in MPN are reduction of thrombotic risk, reduction in spleen volume, and alleviation of symptoms. There are no pharmacologic therapies currently in use apart from interferon-alpha that specifically deplete the neoplastic clone and lead to molecular remission [20]. Thus, there is an unmet need to identify therapeutics to successfully target JAK2V617F cells in MPN. The SMAC mimetic LCL-161 is currently in a Phase 2 clinical trials for the MPN subtype myelofibrosis (NCT02098161) [21]. However, it still remains unclear how LCL-161 impacts JAK2V617F or CALR mutant cells compared to wildtype counterparts.
Based upon the fact that there is hyperactivation of NFĸB signaling pathway [22] we reasoned that MPN cells would be more susceptible to killing by SMAC mimetics. To test this, we performed cell viability assays using murine and human cell lines, myeloid colony formation assays using primary MPN patient cells and a mouse model to test the effect of the SMAC mimetic LCL-161 on JAK2V617F cells in the presence and absence of TNFα. Here, we find that murine and human JAK2V617F cell lines are hypersensitive to the SMAC mimetic LCL-161 in vitro in the absence but not presence of TNFα. Furthermore, LCL-161 reduced myeloid colony formation to a greater extent in MPN mice and patients and reduced spleen size and possibly myelofibrosis in vivo.

Methods

Drug formulation

LCL-161 was provided by Novartis Pharmaceutical Corporation. For in vitro assays, LCL-161 was dissolved in DMSO at a concentration of 10 mM, stored in single use aliquots at − 80 °C, and dissolved directly in culture media fresh for each experiment. LCL-161 was formulated for oral gavage by dissolving in 0.1 N HCl to a final concentration of 10 mg/ml in sodium acetate buffer (100 mM, pH 4.6). LCL-161 was prepared by first wetting 10 mg powder stock with 30 μl water, and then dissolving in two equivalents (0.73 μl/mg of compound) of 6.0 N HCl. The resulting (clear) solution was brought up to 1 ml in pH 4.6 acetate buffer, and the resulting stock was stored frozen at − 20 °C until used for gavage treatment of mice.

Creation of L929 cell lines

Parental L929 cell line (ATCC® CCL-1™, purchased from ATCC) were infected with retrovirus containing JAK2WT, JAK2V617F, CALRDEL, CALRWT, MPL, or empty vector containing green fluorescent protein (GFP) or human CD4 (hCD4) tags by spinoculation. GFP+ and/or hCD4+ cells were sorted on an Aria flow cytometer (BD) and expanded in culture.

Protein extraction and Western Blot

Following washes with PBS, cells were lysed in 1× Immunoprecipitation buffer (20 mM Tris–HCl pH 7.5, 150 mM NaCl, 1 mM EDTA pH 8.0, 1 mM EGTA, 1% Triton X-100, 10 mM sodium pyrophosphate, 10 mM α-glycerophosphate, 1 mM Na3VO4, and 50 mM NaF) by vortexing for 10 s and placing cells on ice for 10 min. Next, samples were centrifuged at 12,000×g for 15 min at 4° C. Supernatants were collected and 4× Lamelli buffer (Bio-Rad) was added to each sample prior to storage at − 20 °C. Protein concentration was determined by Bicinchoninic acid (BCA) assay at absorbance at 532 nm. Western blot analysis was performed using 30 µg for protein boiled for 10 min at 95 °C and electrophoresis was carried out on a 10% acrylamide gel. The protein was transferred onto a nitrocellulose membrane and blocked with 5% BSA in 0.1% PBS-T at 25 °C. The membrane was incubated for 1 h at room temperature or overnight at 4 °C with the primary antibodies for cIAP1 (ab2399, Abcam), cIAP2 (ab23423, Abcam), XIAP (ab28151, Abcam), and β-actin (ab8227, Abcam). Following three 15 min wash steps with 0.1% PBS-T, membranes were incubated with secondary antibody goat-anti-rabbit IgG for 1 h. Following an additional wash step, membranes were imaged on a Chemdoc luminescence reader (Bio-Rad).

Human cell lines and apoptosis assays

K562 and HEL cells (gift from Brian Druker Lab, Oregon Health & Science University) were maintained in RPMI 1640 (Corning) containing phenol red supplemented 100 U penicillin, 100 µg/ml streptomycin, 2 mM glutamine, and 10% FBS. Cells were cultured in a humidified tissue culture incubator at 37 °C and 5% CO2. For apoptosis assays, K562 and HEL cells were plated into 12-well plates at a density of 1 × 106 cells per well in RPMI+ 10% FBS + Pen/Strep/l-glutamine containing vehicle, LCL-161, or LCL-161 + TNFα. Following 16 h, flow cytometry for annexin-V and propidium iodide (PI) was performed.

Murine colony formation assays

Bone marrow was harvested from JAK2V617F knock-in mouse model of MPN and cells were plated at a density of 10,000 cells/ml in Methocult M3230 (stem cell) supplemented 10 ng/ml mouse interleukin-3 (mIL-3), 3 U/ml human erythropoietin (hEPO), and 50 ng/ml mouse stem cells factor (mSCF), all purchased from Peprotech. The cells were plated in triplicate at a density of 10,000 cells/ml and incubated at 37 °C in a 5% carbon dioxide humidified incubator for 5–7 days. Standard morphological criteria were used to score hematopoietic colonies using a light microscope.

Collection, isolation and culture of primary human samples

All participants gave their informed consent for the studies conducted in accordance with the Declaration of Helsinki. This study was approved by the Institutional Review Board of the University of California, Irvine (IRB HS #2014-9995). Peripheral blood samples were collected from MPN patients with Polycythemia Vera, Essential Thrombocytopenia, and Primary Myelofibrosis or normal controls. Density gradient centrifugation with Ficoll was performed to isolate blood mononuclear cells (PBMCs). Following PBMC isolation, a red blood cell lysis step was performed with ammonium chloride buffer (ACK).

Human hematopoietic colony assays

Isolated PBMCs were plated in Methocult H4320 (Stem Cell Technologies, Vancouver Canada) supplemented 10 ng/ml human interleukin-3 (hIL-3), 3 U/ml human erythropoietin (hEPO), and 50 ng/ml human stem cells factor (hSCF). The cells were plated in triplicate at a density of 100,000 cells/ml and incubated at 37 °C in a 5% CO2 humidified incubator for 7–10 days. Standard morphological criteria were used to score hematopoietic colonies using a light microscope.

Mice

All mouse work was performed with approval from the IACUC committee at University of California, Irvine. Equal numbers of male and female mice were used for all experiments. The Jak2V617F was a gift from Ann Mullally, Brigham and Women’s Hospital. The transduction–transplantation mouse model of JAK2V617F driven-MPN was established as previously described [10, 23].

Statistics

All statistical analyses were performed using Graphpad Prism version 7.0 (Graphpad, La Jolla, CA, USA). Statistical analyses were performed using an unpaired t-test (Holm–Sidak) or 2way ANOVA (Tukey’s test). Significant differences were indicated as *P < 0.05, **P < 0.01, ***P < 0.001, ***P < 0.0001. Error bars represent mean values ± SEM. All experiments were performed at least three independent times unless otherwise stated.

Results

JAK2V617F mutant cells are hyper-sensitive to the SMAC mimetic LCL-161 in the absence but not presence of TNFα

To investigate the sensitivity of JAK2V617F mutant cells to LCL-161, we created cell lines with stable expression of JAK2V617F, JAK2WT, or empty GFP vector by retroviral transduction. In this assay, we utilized the mouse fibroblast L929 line which are extremely sensitive to TNFα-induced death. We measured the impact of increasing concentrations of LCL-161 on the viability of these cells using a resazurin based assay in the presence or absence of TNFα (Fig. 1a–c). Sub-micromolar concentrations of LCL-161 significantly reduced the viability of L929 JAK2V617F cells compared to JAK2WT or empty vector cells (Fig. 1a). We observed 50% cell viability of L929 JAK2V617F cells with 0.45 µM LCL-161 as compared to similar cell viabilities with 5.5 µM and 9.5 µM LCL-161 for empty vector or JAK2WT cells, respectively (P < 0.0001, 2way ANOVA; Fig. 1a). However, this differential sensitivity was lost when TNFα was added to culture (Fig. 1b). Interestingly, overexpression of JAK2WT rendered L929 cells resistant to killing by higher concentrations of LCL-161 in the presence of TNFα, as these cells were 50% viable in the presence of 19 µM LCL-161 (P < 0.0001, 2way ANOVA; Fig. 1b).
Next, we assessed whether autonomous production of TNFα could explain the increased sensitivity of JAK2V617F cells to LCL-161. To do this, we tested whether TNFα neutralizing antibody abrogates the increased sensitivity of JAK2V617F mutant L929 cells to LCL-161. Addition of TNFα neutralizing antibody did not rescue the increased sensitivity of JAK2V617F cells to LCL-161 (Fig. 1c) suggesting that the sensitivity of JAK2V617F cells was not due to autonomous production of TNFα. Moreover, we could not detect any TNFα in the supernatant of L929 JAK2V617F cells, either at baseline or with the addition of LCL-161 by ELISA (data not shown). As expected, treatment with LCL-161 decreased both cIAP1 and cIAP2 (Fig. 1d). We found no marked differences in expression of cIAP1, cIAP2, or XIAP in JAK2V617F versus JAK2WT or parental L929 cells under any conditions tested (Fig. 1d). However, we did find evidence that sensitivity to LCL-161 is mediated at least in part to NFĸB activation, the NFĸB inhibitor QNZ had a protective effect on empty vector cells in the presence but not absence of TNF, whereas QNZ had a protective effect on JAK2V617F mutant cells only in the absence of TNF (Additional file 1: Figure S1). This suggests that JAK2V617F induces NFĸB activation in L929 cells and this mediates the sensitivity to LCL-161.
We also compared the ability of LCL-161 to induce apoptosis in the HEL human erythroleukemia cell line which harbors the JAK2V617F mutation versus the K562 human erythroleukemia cell line which harbors the BCR-ABL translocation. LCL-161 induced apoptosis significantly more in HEL cells as compared to K562 cells in the absence (P < 0.001, unpaired t test; Fig. 1e) but not presence of TNFα (Fig. 1f, g). Likewise, expression levels of cIAP1, cIAP2, or XIAP did not correlate with sensitivity to LCL-161 in HEL human cells expressing JAK2V617F as compared to K562 cells (Additional file 1: Figure S2). These data in human cell lines corroborate our findings in L929 cells that JAK2V617F mutant cells are more sensitive to killing by LCL-161 alone, but when TNFα is added this differential sensitivity is lost.

Calreticulin mutated cells are not hyper-sensitive to LCL-161

To investigate the effect of LCL-161 on calreticulin mutant cells, we also created L929 cell ectopically expressing MPN associated calreticulin (CALR) mutations with and without its obligate cytokine receptor scaffold thrombopoietin receptor (MPL). CALR mutant L929 cells were not more sensitive to LCL-161 as compared to empty vector either with or without co-expression of MPL (Additional file 1: Figure S3). Interestingly, overexpression of wild-type CALR led to increased sensitivity to LCL-161 when co-expressed with MPL as compared to cells with empty vector, CALR deletion or CALR insertion (Additional file 1: Figure S3).

Sensitivity of JAK2V617F cells to LCL-161 is dependent upon Janus kinase activity

To determine whether the increased sensitivity of JAK2V617F cells to LCL-161 is dependent upon Janus kinase (JAK) activity, we utilized the JAK inhibitors ruxolitinib (JAK1/2 inhibitor) and pacritinib (JAK2/Flt3 inhibitor) in a resazurin based cell viability assay. Sensitivity of JAK2V617F L929 cells was dependent upon activation of JAK2 activity, as treatment with either ruxolitinib or pacritinib reversed the increased sensitivity of JAK2V617F cells to LCL-161 (Fig. 2). This data suggests that the hyper-sensitivity of JAK2V617F cells to LCL-161 is due to constitutive JAK2 activation.

LCL-161 reduces myeloid colony formation in JAK2V617F knock-in mice and MPN patient cells in the absence of TNFα

To investigate the effect of LCL-161 on myeloid colony formation in primary mouse bone marrow cells, we performed colony formation assays from JAK2V617F knock-in and wild-type mice with increasing concentrations of LCL-161. LCL-161 reduced colony formation in JAK2V617F cells compared to wild-type cells in the presence of all doses tested, although only the 1 µM LCL-161 dose reached statistical significance (P < 0.001, unpaired t-test; Fig. 3a). However, the differential sensitivity of JAK2V617F cells was lost in the presence of TNFα (Fig. 3b). This data further supports the model that JAK2V617F cells have increased sensitivity to SMAC mimetics in the absence but not presence of TNFα.
We next investigated the effect of LCL-161 on colony formation using primary cells from MPN patients compared to unaffected individuals. In the presence of LCL-161 alone, there was a reduction in colony formation from JAK2V617F mutated MPN patients versus normal controls at 1 µM and 2.5 µM doses, though this trend did not reach significance (P = 0.05 and 0.07, respectively, unpaired t test; Fig. 3c). These same trends were observed when erythroid and G/M colonies were enumerated separately (Additional file 1: Figure S4). When TNFα was added to the culture, this diminished the apparent difference between MPN and normal controls in response to LCL-161 at all but the highest concentration (5 µM) of LCL-161 (P < 0.05, unpaired t test; Fig. 3d).

LCL-161 reduces spleen size and ameliorates fibrosis in a JAK2V617F transduction–transplantation model

To test the efficacy of LCL-161 in vivo, we utilized a JAK2V617F transduction–transplantation MPN mouse model that develops myelofibrosis around 1 year of age [2326]. Treatment with LCL-161 was initiated 15 weeks after transplant with twice weekly oral administration. We did not detect changes in peripheral blood counts nor percentage of GFP+ cells in the peripheral blood in LCL-treated mice compared to untreated mice (n = 3–4 mice/group, Fig. 4a–e). However, at sacrifice we found that LCL-161 treatment significantly reduced spleen size (P < 0.05, unpaired t test; Fig. 4f) and modestly reduced the percentage of GFP+ cells in the bone marrow and spleen of JAK2V617F mice (n = 3–4 mice/group, Fig. 4g). Upon analysis of reticulin-stained bone marrows, we observed that fibrosis was reduced in mice that had been treated with LCL-161 (Fig. 4h). These data demonstrate that LCL-161 may have a beneficial effect on an in vivo model of JAK2V617F mutated MPN.

Discussion

Here, we found that mouse cell lines ectopically expressing JAK2V617F, the JAK2V617F mutated human cell line HEL, JAK2V617F knock-in mice, and primary MPN samples were more sensitive to killing by the SMAC mimetic LCL-161 compared to their JAK2WT counterparts. Although addition of exogenous TNFα increased the sensitivity of both mutant and wild-type cells to LCL-161, the differential sensitivity of mutant cells was abolished when TNFα was added to culture. The increased sensitivity to LCL-161 was not due to autocrine production of TNFα, as pharmacologic blockade of TNFα did not rescue JAK2V617F mutant cells from LCL-161.
Activation of JAK2 signaling is critical for the increased sensitivity of JAK2V617F mutant cells to LCL-161. Addition of JAK2 inhibitors ruxolitinib or pacritinib restored LCL-161 sensitivity of JAK2V617F mutant to that of wild-type cells. This observation suggests JAK2 inhibitors may reduce the vulnerability of JAK2 mutant cells to be preferentially killed by SMAC mimetics and that therapeutic combinations of JAK2 inhibitors and SMAC mimetic in patients may not be an ideal combination. This is an important point given that it is common for new investigational agents in myelofibrosis to be tested in combination with ruxolitinib.
The ex vivo experiments with mouse and primary human myeloid colony formation corroborate our findings in cell lines that MPN mutant cells are more sensitive than wild-type cells in the absence but not presence of TNFα. Human MPN patients have variable frequencies of JAK2V617F and so our methylcellulose colony formation assays with primary MPN patient cells assess both mutant and wild-type colonies. This may explain why the impact of LCL-161 on MPN patient colony formation was less robust than our observations in cell lines bearing the JAK2V617F mutation.
Cells bearing the MPN associated CALRDEL mutation were not more sensitive to LCL-161 when expressed either with or without its obligate cytokine receptor scaffold MPL. This suggests that SMAC mimetics may be most beneficial in JAK2V617F mutated MPN patients. CALR binds to and specifically activates MPL, whereas JAK2V617F activates signaling of multiple cytokine receptor pathways. The sensitivity of JAK2V617F to SMAC mimetics is likely mediated via activation of pathways distinct from MPL. Therefore, combination therapy with SMAC mimetic and drugs which target MPL signaling may be additive or even synergistic in JAK2V617F mutated MPN.
In our in vivo mouse model, we found that LCL-161 had modest effects on peripheral blood counts. Although LCL-161 did reduce the percentage of GFP+ cells in the spleen and bone marrow, these effects were modest and did not reach significance. The primary benefit of LCL-161 in this mouse model was reduction in splenomegaly and possible reduction of fibrosis. Future studies would require increasing numbers of mice to validate the effect of LCL-161 on splenomegaly and myelofibrosis in vivo. Regardless, our findings suggest that LCL-161 may not be an ideal single agent in MPN but may be most useful as an adjunct to other MPN therapies.
Approaches that reduce TNFα but maintain constitutive activation of JAK2 in mutant cells may be an ideal combination with SMAC mimetics in MPN. The TNFα decoy receptor etanercept has been investigated as a single agent in an open label pilot study in myelofibrosis and was found to alleviate symptoms [27]. Another potential approach as a combinatorial study with SMAC mimetics would be to combine with a JAK inhibitor that is devoid of JAK2 inhibitory activity, such as itacitinib (INCB039110). This would result in reduction of inflammatory cytokines including TNFα while preserving the vulnerability of JAK2V617F cells to killing by SMAC mimetic.

Conclusion

SMAC mimetics may be useful in patients with JAK2V617F mutated MPN, as JAK2V617F mutant cells are more readily killed by SMAC mimetics as compared to wild-type cells. This vulnerability to SMAC mimetics is mediated via JAK2 activation, therefore addition of a JAK2 inhibitor to SMAC mimetics may detract from the ability of SMAC mimetics to preferentially kill JAK2V617F mutant cells. However, therapy with agents that reduce or block TNFα may augment the ability of SMAC mimetics to specifically target JAK2V617F mutant cells.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s40164-019-0157-6.

Acknowledgements

The authors would like to thank the Experimental Tissue Resource of University of California, Irvine for preparation of histology slides.
All primary human samples used in this study were derived from participants who consented to an IRB approved study “Role of Inflammation in Myeloproliferative Neoplasm” at University of California, Irvine. All animal use in this protocol was in accordance with an approved IACUC protocol.
All authors have reviewed the manuscript and consent for publication.

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.
Anhänge

Supplementary information

Additional file 4: Figure S4. Colony formation shown in Fig. 3 separated by erythroid and G/M colonies. (A) Erythroid and (B) G/M colony formation from MPN patients and normal controls with increasing concentrations of LCL-161. (C) Erythroid and (D) G/M colony formation from MPN patients and normal controls with 10 ng/ml TNFα + increasing concentrations of LCL-161.
Literatur
2.
Zurück zum Zitat Chen DJ, Huerta S. Smac mimetics as new cancer therapeutics. Anticancer Drugs. 2009;20(8):646–58.CrossRef Chen DJ, Huerta S. Smac mimetics as new cancer therapeutics. Anticancer Drugs. 2009;20(8):646–58.CrossRef
3.
Zurück zum Zitat Fulda S. Smac mimetics as IAP antagonists. Semin Cell Dev Biol. 2015;39:132–8.CrossRef Fulda S. Smac mimetics as IAP antagonists. Semin Cell Dev Biol. 2015;39:132–8.CrossRef
4.
Zurück zum Zitat Welsh K, Milutinovic S, Ardecky RJ, et al. Characterization of potent SMAC mimetics that sensitize cancer cells to TNF family-induced apoptosis. PLoS ONE. 2016;11(9):e0161952.CrossRef Welsh K, Milutinovic S, Ardecky RJ, et al. Characterization of potent SMAC mimetics that sensitize cancer cells to TNF family-induced apoptosis. PLoS ONE. 2016;11(9):e0161952.CrossRef
5.
Zurück zum Zitat Berger R, Jennewein C, Marschall V, et al. NF-kappaB is required for Smac mimetic-mediated sensitization of glioblastoma cells for gamma-irradiation-induced apoptosis. Mol Cancer Ther. 2011;10(10):1867–75.CrossRef Berger R, Jennewein C, Marschall V, et al. NF-kappaB is required for Smac mimetic-mediated sensitization of glioblastoma cells for gamma-irradiation-induced apoptosis. Mol Cancer Ther. 2011;10(10):1867–75.CrossRef
6.
Zurück zum Zitat Lueck SC, Russ AC, Botzenhardt U, et al. Smac mimetic induces cell death in a large proportion of primary acute myeloid leukemia samples, which correlates with defined molecular markers. Oncotarget. 2016;7(31):49539–51.CrossRef Lueck SC, Russ AC, Botzenhardt U, et al. Smac mimetic induces cell death in a large proportion of primary acute myeloid leukemia samples, which correlates with defined molecular markers. Oncotarget. 2016;7(31):49539–51.CrossRef
7.
Zurück zum Zitat Mondet J, Hussein K, Mossuz P. Circulating cytokine levels as markers of inflammation in philadelphia negative myeloproliferative neoplasms: diagnostic and prognostic interest. Mediat Inflamm. 2015;2015:670580.CrossRef Mondet J, Hussein K, Mossuz P. Circulating cytokine levels as markers of inflammation in philadelphia negative myeloproliferative neoplasms: diagnostic and prognostic interest. Mediat Inflamm. 2015;2015:670580.CrossRef
8.
Zurück zum Zitat Hermouet S, Bigot-Corbel E, Gardie B. Pathogenesis of myeloproliferative neoplasms: role and mechanisms of chronic inflammation. Mediat Inflamm. 2015;2015:145293. Hermouet S, Bigot-Corbel E, Gardie B. Pathogenesis of myeloproliferative neoplasms: role and mechanisms of chronic inflammation. Mediat Inflamm. 2015;2015:145293.
9.
Zurück zum Zitat Fleischman AG. Inflammation as a driver of clonal evolution in myeloproliferative neoplasm. Mediat Inflamm. 2015;2015:606819.CrossRef Fleischman AG. Inflammation as a driver of clonal evolution in myeloproliferative neoplasm. Mediat Inflamm. 2015;2015:606819.CrossRef
10.
Zurück zum Zitat Fleischman AG, Aichberger KJ, Luty SB, et al. TNFalpha facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood. 2011;118(24):6392–8.CrossRef Fleischman AG, Aichberger KJ, Luty SB, et al. TNFalpha facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood. 2011;118(24):6392–8.CrossRef
11.
Zurück zum Zitat Fisher DAC, Malkova O, Engle EK, et al. Mass cytometry analysis reveals hyperactive NF Kappa B signaling in myelofibrosis and secondary acute myeloid leukemia. Leukemia. 2017;31(9):1962–74.CrossRef Fisher DAC, Malkova O, Engle EK, et al. Mass cytometry analysis reveals hyperactive NF Kappa B signaling in myelofibrosis and secondary acute myeloid leukemia. Leukemia. 2017;31(9):1962–74.CrossRef
12.
Zurück zum Zitat Heaton WL, Senina AV, Pomicter AD, et al. Autocrine Tnf signaling favors malignant cells in myelofibrosis in a Tnfr2-dependent fashion. Leukemia. 2018;32(11):2399–411.CrossRef Heaton WL, Senina AV, Pomicter AD, et al. Autocrine Tnf signaling favors malignant cells in myelofibrosis in a Tnfr2-dependent fashion. Leukemia. 2018;32(11):2399–411.CrossRef
13.
Zurück zum Zitat Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365(9464):1054–61.CrossRef Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365(9464):1054–61.CrossRef
14.
Zurück zum Zitat Campbell PJ, Baxter EJ, Beer PA, et al. Mutation of JAK2 in the myeloproliferative disorders: timing, clonality studies, cytogenetic associations, and role in leukemic transformation. Blood. 2006;108(10):3548–55.CrossRef Campbell PJ, Baxter EJ, Beer PA, et al. Mutation of JAK2 in the myeloproliferative disorders: timing, clonality studies, cytogenetic associations, and role in leukemic transformation. Blood. 2006;108(10):3548–55.CrossRef
15.
Zurück zum Zitat James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144–8.CrossRef James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144–8.CrossRef
16.
Zurück zum Zitat Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779–90.CrossRef Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779–90.CrossRef
17.
Zurück zum Zitat Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387–97.CrossRef Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387–97.CrossRef
18.
Zurück zum Zitat Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379–90.CrossRef Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379–90.CrossRef
19.
Zurück zum Zitat Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405.CrossRef Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405.CrossRef
20.
Zurück zum Zitat Silver RT, Barel AC, Lascu E, et al. The effect of initial molecular profile on response to recombinant interferon-alpha (rIFNalpha) treatment in early myelofibrosis. Cancer. 2017;123(14):2680–7.CrossRef Silver RT, Barel AC, Lascu E, et al. The effect of initial molecular profile on response to recombinant interferon-alpha (rIFNalpha) treatment in early myelofibrosis. Cancer. 2017;123(14):2680–7.CrossRef
21.
Zurück zum Zitat Boddu P, Carter BZ, Verstovsek S, Pemmaraju N. SMAC mimetics as potential cancer therapeutics in myeloid malignancies. Br J Haematol. 2019;185(2):219–31.CrossRef Boddu P, Carter BZ, Verstovsek S, Pemmaraju N. SMAC mimetics as potential cancer therapeutics in myeloid malignancies. Br J Haematol. 2019;185(2):219–31.CrossRef
22.
Zurück zum Zitat Fisher DA EE, Malkova O, Fulbright MC, Zhou A, Oh ST. NF Kappa B signaling hyperactivation in myelofibrosis and secondary acute myeloid leukemia. In: American Society of Hematology annual meeting oral abstract. 2015. Fisher DA EE, Malkova O, Fulbright MC, Zhou A, Oh ST. NF Kappa B signaling hyperactivation in myelofibrosis and secondary acute myeloid leukemia. In: American Society of Hematology annual meeting oral abstract. 2015.
23.
Zurück zum Zitat Bumm TG, Elsea C, Corbin AS, et al. Characterization of murine JAK2V617F-positive myeloproliferative disease. Cancer Res. 2006;66(23):11156–65.CrossRef Bumm TG, Elsea C, Corbin AS, et al. Characterization of murine JAK2V617F-positive myeloproliferative disease. Cancer Res. 2006;66(23):11156–65.CrossRef
24.
Zurück zum Zitat Lacout C, Pisani DF, Tulliez M, Gachelin FM, Vainchenker W, Villeval J-L. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006;108(5):1652–60.CrossRef Lacout C, Pisani DF, Tulliez M, Gachelin FM, Vainchenker W, Villeval J-L. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006;108(5):1652–60.CrossRef
25.
Zurück zum Zitat Zaleskas VM, Krause DS, Lazarides K, et al. Molecular pathogenesis and therapy of polycythemia induced in mice by JAK2 V617F. PLoS ONE. 2006;1:e18.CrossRef Zaleskas VM, Krause DS, Lazarides K, et al. Molecular pathogenesis and therapy of polycythemia induced in mice by JAK2 V617F. PLoS ONE. 2006;1:e18.CrossRef
26.
Zurück zum Zitat Wernig G, Mercher T, Okabe R, Levine RL, Lee BH, Gilliland DG. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107(11):4274–81.CrossRef Wernig G, Mercher T, Okabe R, Levine RL, Lee BH, Gilliland DG. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107(11):4274–81.CrossRef
27.
Zurück zum Zitat Steensma DP, Mesa RA, Li CY, Gray L, Tefferi A. Etanercept, a soluble tumor necrosis factor receptor, palliates constitutional symptoms in patients with myelofibrosis with myeloid metaplasia: results of a pilot study. Blood. 2002;99(6):2252–4.CrossRef Steensma DP, Mesa RA, Li CY, Gray L, Tefferi A. Etanercept, a soluble tumor necrosis factor receptor, palliates constitutional symptoms in patients with myelofibrosis with myeloid metaplasia: results of a pilot study. Blood. 2002;99(6):2252–4.CrossRef
Metadaten
Titel
The SMAC mimetic LCL-161 selectively targets JAK2V617F mutant cells
verfasst von
Brianna M. Craver
Thanh Kim Nguyen
Jenny Nguyen
Hellen Nguyen
Christy Huynh
Sarah J. Morse
Angela G. Fleischman
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2020
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-019-0157-6

Weitere Artikel der Ausgabe 1/2020

Experimental Hematology & Oncology 1/2020 Zur Ausgabe

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Update Onkologie

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