Pan RAF inhibitor LY3009120 is highly synergistic with low dose cytarabine, but not azacitidine, in acute myeloid leukemia with RAS mutations

 
CONTINUE READING
ONCOLOGY LETTERS 22: 745, 2021

                Pan‑RAF inhibitor LY3009120 is highly synergistic
                 with low‑dose cytarabine, but not azacitidine, in
                   acute myeloid leukemia with RAS mutations
                        JIHYUN PARK1,2, HYEJOO PARK1,2, JA MIN BYUN3, JUNSHIK HONG1‑3,
                           DONG‑YEOP SHIN2,3, YOUNGIL KOH1‑3 and SUNG‑SOO YOON1‑3

                    1
                 Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080;
    2
     Hematology Oncology Department, Center for Medical Innovation, Seoul National University Hospital, Seoul 03082;
                   3
                     Department of Internal Medicine, Division of Hematology and Medical Oncology,
                          Seoul National University Hospital, Seoul 03080, Republic of Korea

                                         Received April 15, 2021; Accepted July 26, 2021

                                                   DOI: 10.3892/ol.2021.13006

Abstract. Alterations in RAS oncogenes have been implicated       Introduction
in various types of cancer, including acute myeloid leukemia
(AML). Considering that currently, there are no targeted          Acute myeloid leukemia (AML) is a genetically heterogeneous
therapies for patients with RAS‑mutated AML despite the           and highly aggressive hematological malignancy with an
poor outcomes, RAF may be a potential target for AML. In          average onset age of 67 years (1). Patients with AML harbor
this study, we first analyzed the efficacy of different MAPK      distinct genetic and molecular abnormalities caused by various
inhibitors in AML cell lines. We found that LY3009120,            chromosomal aberrations in about 50‑60% of de novo AML
a pan‑RAF inhibitor, significantly decreased cell survival        cases and 80‑95% of secondary cases (2). Classically, mutated
in RAS‑mutated AML cell lines. We then investigated the           genes in AML are classified according to their characteristics,
synergistic effects of LY3009120 with either cytarabine           and the gene group that affects cell proliferation includes KIT,
or azacitidine. We found that the combination of low‑dose         FLT3, and RAS (3,4). MAPK, induced Ras signaling, is a major
cytarabine and LY3009120 showed a synergistic effect in           oncogenic pathway in AML.
NRAS‑mutated HL‑60 cells and KRAS‑mutated NB4 cells.                  Alterations in RAS are involved in the progression of various
This effect was caused by a decrease in proliferation, induc‑     cancers. Missense RAS mutations causing a gain‑of‑function
tion of apoptosis, and cell growth arrest through a decrease      phenotype occur in 25% of human cancers (5) and 20‑25% of
in phosphorylated MEK and ERK along with a cytotoxic              AMLs. KRAS and NRAS mutations are present in 5 and 11% of
response occurring specifically for the RAS mutation of the       AMLs, respectively (6). Patients with RAS‑mutated AML have
pan‑RAF inhibitor LY3009120. In addition, we confirmed            shorter overall survival and AML‑free survival, higher median
that combination treatment with low‑dose cytarabine and           age, and lower complete remission rates (7). In addition,
LY3009120 led to an increase in apoptosis in primary AML          abnormalities in Ras proteins that regulate various functions,
cells. Our findings indicate that combination therapy with        including growth, migration, adhesion, survival, and differen‑
pan‑RAF inhibitor LY3009120 and low‑dose cytarabine               tiation in normal cells, affect leukemia primarily and result in
may be a promising treatment strategy for RAS‑mutated             secondary effects such as chemotherapy resistance and cancer
AML.                                                              recurrence (8‑11). Therefore, the RAS status of a patient has
                                                                  prognostic value and is a potent cancer biomarker (12‑15).
                                                                  However, it has been challenging to target Ras directly. While
                                                                  drugs targeting the downstream signaling pathway, including
                                                                  RAF, mitogen‑activated protein kinase/ERK (extracellular
                                                                  signal‑regulated kinase) kinase (MEK), are being developed
Correspondence to: Professor Sung‑Soo Yoon or Professor
                                                                  mainly for solid tumors (12,16‑19), none of them are proven
Youngil Koh, Department of Internal Medicine, Division of
Hematology and Medical Oncology, Seoul National University
                                                                  successful (20‑22). Developed as a pan‑RAF inhibitor of RAF,
Hospital, 101 Daehak‑ro, Jongno, Seoul 03080, Republic of Korea   downstream of Ras, LY3009120 inhibits the activity of all
E‑mail: ssysmc@snu.ac.kr                                          RAF isoforms (A‑RAF, B‑RAF, and c‑RAF) and RAF dimers,
E‑mail: snuhgo01@snu.ac.kr                                        and it is currently in the first phase of clinical trials for solid
                                                                  tumors (23).
Key words: acute myeloid leukemia, RAS, mitogen‑activated             Therefore, in this study, using RAS mutant AML cells,
protein kinase, cytarabine, combined therapy                      we investigated the synergistic effects of MAPK inhibitors in
                                                                  combination with low‑dose cytarabine or azacytidine, currently
                                                                  used as first‑line treatments for AML in elderly patients.
2                              PARK et al: INHIBITION OF THE MAPK PATHWAY IN RAS MUTANT AML

Materials and methods                                              HRP‑conjugated secondary antibody for 1 h at 37˚C. GAPDH
                                                                   was used as a control marker in all the blots.
Cell culture and treatment. The three AML cell lines (HL‑60,
NB4, and KG‑1) used in the study were obtained from the            Cell cycle analysis. To investigate the cell cycle profiles of the
American Type Culture Collection (ATCC) and the German             treated and untreated cells after the treatment, the cells were
Collection of Microorganisms and Cell Cultures GmbH                collected, washed with DPBS, and fixed with 70% ethanol. The
(DSMZ). KG‑1 and HL‑60 cell lines were authenticated               fixed cells were treated with 0.4 mg/ml RNase (Promega) and
using the AmpFISTR® Identifiler ® PCR Amplification Kit            5 µl of 50 µg/ml propidium iodide (Invitrogen) (25). The stained
and the NB4 cell line using the PowerPlex®21 System Kit.           cells were analyzed using FACSCanto II (BD Biosciences) and
Cells were cultured in RPMI‑1640 medium supplemented               the apoptotic cells were detected using flow cytometry. For
with 10% heat‑inactivated fetal bovine serum (Gibco) and           cell cycle analysis, flow cytometry data was analyzed using
1% penicillin/streptomycin (Gibco) and incubated overnight         ‘ModFit’ software. Each experiment was repeated three times.
at 37˚C in 5% CO2. Cells were seeded for 24 h and treated with
cytarabine, azacytidine, LY3009120, LXH254, dabrafenib,            Apoptosis assay. Apoptosis induction after single or combined treat‑
and trametinib. For single treatment, each inhibitor was treated   ment with cytarabine and LY3009120 was measured using the Dead
at indicated concentrations ranging from 0.001 to 10 µM for        Cell Apoptosis Kit with Annexin V/FITC and PI (Thermo Fisher
72 h. After that, the combined treatment was treated with          Scientific), and DMSO was used as a control. Cells were collected
cytarabine 50 nM or/and LY3009120 6/50 nM concentration            24 or 72 h after their respective treatment and stained with
for 72 h. Each experiment was repeated five times.                 Annexin V/PI dye (26). Each experiment was repeated three times.

Patient samples. Bone marrow mononuclear cells (MNCs)              Statistical analysis. Statistical significance among the groups
from patients with AML were isolated using the Ficoll gradient     was determined using GraphPad Prism version 5.0 (GraphPad
method and cryopreserved in Cell banker 2, a serum‑free            Software Inc.). Statistical analyses for the efficacy of the
medium. The MNCs of the patients were thawed at 37˚C in            drugs were performed using the two‑tailed unpaired Student's
Iscove's modified Dulbecco's media containing 10% fetal            t‑test. One‑way ANOVA followed by Dunnett's post hoc test
bovine serum and incubated for 24 h.                               were used for multiple comparisons. Combination index (CI)
                                                                   values were calculated using the Chou‑Talalay equation (27).
Agents and antibodies. Cytarabine (Cytosar‑U®, Ara‑C,              CI values were used to validate combinatorial effects between
Arabinosylcytosine); the pan‑RAF inhibitors, LY3009120             the two drugs; CI>1.00 indicates antagonism, CI=1.00 indicates
and LXH254; the B‑RAF inhibitor, dabrafenib; and the MEK           additivity, and CI
ONCOLOGY LETTERS 22: 745, 2021                                                                   3

Figure 1. Effect of cytarabine and MAPK inhibitors on the proliferation of AML cells with wild‑type (KG‑1) or mutated RAS (HL‑60 and NB4). To confirm the
individual effect of (A) cytarabine, (B) azacitidine, (C) LY3009120, (D) LXH254, (E) dabrafenib and (F) trametinib on AML cell lines, the cells were treated
with each inhibitor at the indicated concentrations ranging from 0.001 to 10 µM for 72 h, and cell proliferation assays were performed. Following treatment
with 0.1 µM LY3009120 and LXH254, HL‑60 and NB4 cells showed significantly lower cell proliferation compared to that in KG‑1 cells. ****P
4                                PARK et al: INHIBITION OF THE MAPK PATHWAY IN RAS MUTANT AML

Table I. Evaluation of the synergistic effect of low‑dose cyta‑         showed a slight increase in the percentage G0G1 phase cells
rabine and LY3009120.                                                   and decrease in the S phase cells after combined treatment for
                                                                        24 h (Fig. 3A) (*P
ONCOLOGY LETTERS 22: 745, 2021                                                             5

Figure 2. Inhibition of the MAPK pathway protein in RAS mutant cells following combined treatment with LY3009120 and low‑dose cytarabine.
(A) NRAS‑mutated HL‑60 and KRAS‑mutated NB4 cells were treated with cytarabine and/or LY3009120 for 24 h, and the levels of the MAPK pathway mole‑
cules, including p‑c‑RAF, c‑RAF, B‑RAF, p‑MEK1/2, MEK1/2, p‑ERK1/2 and ERK1/2, were analyzed via western blotting. The HL‑60 cell line was treated
with 50 nM cytarabine, 6 nM LY3009120 or 50 nM cytarabine + 6 nM LY3009120. The NB4 cell line was treated with 50 nM cytarabine, 50 nM LY3009120
or 50 nM cytarabine + 50 nM LY3009120. (B) Semi‑quantification of protein phosphorylation in HL‑60 and NB4 cells. *P
6                                    PARK et al: INHIBITION OF THE MAPK PATHWAY IN RAS MUTANT AML

Figure 3. Cell cycle arrest of RAS mutant acute myeloid leukemia cells following combined treatment for 24 h. (A) Cells were treated with drugs alone or in
combination for 24 h. The cell cycle profiles of HL‑60 and NB4 cells were analyzed using PI staining and flow cytometry. The HL‑60 cell line was treated with
50 nM cytarabine, 6 nM LY3009120 or 50 nM cytarabine + 6 nM LY3009120. The NB4 cell line was treated with 50 nM cytarabine, 50 nM LY3009120 or
50 nM cytarabine + 50 nM LY3009120. (B) The changes in the number of cells in different phases of the cell cycle were statistically analyzed for both HL‑60
and NB4 cells. *P
ONCOLOGY LETTERS 22: 745, 2021                                                                7

Figure 4. Cell cycle arrest of RAS mutant acute myeloid leukemia cells following combined treatment for 72 h. (A and B) Cells were treated with the
indicated doses for 72 h. Cell cycle profiles of HL‑60 and NB4 were analyzed by flow cytometry and changes in the number of cells in different phases of
the cell cycle were statistically analyzed for both HL‑60 and NB4 cells. The HL‑60 cell line was treated with 50 nM cytarabine, 6 nM LY3009120 or 50 nM
cytarabine + 6 nM LY3009120. The NB4 cell line was treated with 50 nM cytarabine, 50 nM LY3009120 or 50 nM cytarabine + 50 nM LY3009120. *P
8                                   PARK et al: INHIBITION OF THE MAPK PATHWAY IN RAS MUTANT AML

Figure 5. Induction of apoptosis in RAS mutant acute myeloid leukemia cells by low dose cytarabine combined with LY3009120. Cells were treated with the
inhibitors at the indicated concentrations, and the apoptotic cells were analyzed by Annexin V and PI staining. (A) HL‑60 and NB4 cells treated for 24 h.
(B) HL‑60 and NB4 cells treated for 72 h. The HL‑60 cell line was treated with 50 nM cytarabine, 6 nM LY3009120 or 50 nM cytarabine + 6 nM LY3009120.
The NB4 cell line was treated with 50 nM cytarabine, 50 nM LY3009120 or 50 nM cytarabine + 50 nM LY3009120. ****P
ONCOLOGY LETTERS 22: 745, 2021                                                                 9

Figure 6. Identification of apoptosis pathway after treatment of RAS mutant acute myeloid leukemia cells with low‑dose cytarabine combined with LY3009120.
Cells were treated with the inhibitors at the indicated concentrations for 72 h to ensure cell death. Western blotting was performed using antibodies against
caspase‑3, caspase‑9 and PARP. The HL‑60 cell line was treated with 50 nM cytarabine, 6 nM LY3009120 or 50 nM cytarabine + 6 nM LY3009120. The NB4
cell line was treated with 50 nM cytarabine, 50 nM LY3009120 or 50 nM cytarabine + 50 nM LY3009120.

                                                                                  could be attributed to the mechanism of action of these inhibi‑
                                                                                  tors.
                                                                                      Although synergistic effects were observed in both the
                                                                                  AML cell lines studied, the effects were prominent in the
                                                                                  NRAS‑mutated HL‑60 cells, compared to KRAS‑mutated
                                                                                  NB4 cells. Therefore, we further investigated the response
                                                                                  of the two cell lines to the RAF inhibitor LY3009120. In
                                                                                  HL‑60 cells, the expression of phosphorylated ERK decreased
                                                                                  in the LY3009120‑treated cells compared to the control cells.
                                                                                  Conversely, in NB4 cells, the expression of phosphorylated
                                                                                  ERK and phosphorylated c‑RAF increased after LY3009120
                                                                                  treatment. Such paradoxical activity of ERK owing to a RAF
                                                                                  inhibitor is well known (35‑37). Therefore, we suggest that these
                                                                                  paradoxical activities of LY3009120 are reflected in the differ‑
                                                                                  ences observed in the response of HL‑60 and NB4 cells to the
Figure 7. Induction of apoptosis in primary cells of RAS mutant AML by low‑dose   combination therapy.
cytarabine combined with LY3009120. Cells were treated for 72 h to ensure             Despite this complex physiological phenomenon, our
apoptosis. The apoptotic cells were analyzed by Annexin V and PI staining. The    immunoblotting data show that the combination of cytarabine
HL‑60 cell line was treated with 50 nM cytarabine, 6 nM LY3009120 or 50 nM
                                                                                  and LY3009120 has shown synergy in RAS mutant AML,
cytarabine + 6 nM LY3009120. The NB4 cell line was treated with 50 nM cyta‑
rabine, 50 nM LY3009120 or 50 nM cytarabine + 50 nM LY3009120. *P
10                               PARK et al: INHIBITION OF THE MAPK PATHWAY IN RAS MUTANT AML

inhibitors and venetoclax, either with or without low‑dose               7. Kadia TM, Kantarjian H, Kornblau S, Borthakur G, Faderl S,
                                                                            Freireich EJ, Luthra R, Garcia‑Manero G, Pierce S, Cortes J
cytarabine (38) in RAS‑mutated AML.                                         and Ravandi F: Clinical and proteomic characterization of acute
                                                                            myeloid leukemia with mutated RAS. Cancer 118: 5550‑5559,
Acknowledgements                                                            2012.
                                                                         8. Steelman LS, Franklin RA, Abrams SL, Chappell W, Kempf CR,
                                                                            Bäsecke J, Stivala F, Donia M, Fagone P, Nicoletti F, et al:
Not applicable.                                                             Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy.
                                                                            Leukemia 25: 1080‑1094, 2011.
                                                                         9. Downward J: Targeting RAS signalling pathways in cancer
Funding                                                                     therapy. Nat Rev Cancer 3: 11‑22, 2003.
                                                                        10. Kornblau SM, Womble M, Qiu YH, Jackson CE, Chen W,
This work was supported by the National Research Foundation of              Konopleva M, Estey EH and Andreeff M: Simultaneous activation
                                                                            of multiple signal transduction pathways confers poor prognosis
Korea (NRF; grant no. NRF‑2014M3A6A4074727), Korea Health                   in acute myelogenous leukemia. Blood 108: 2358‑2365, 2006.
Industry Development Institute (KHIDI; grant no. HI18C1876)             11. Steelman LS, Abrams SL, Whelan J, Bertrand FE, Ludwig DE,
and Yuhan Corporation (grant no. 800‑20180097).                             Bäsecke J, Libra M, Stivala F, Milella M, Tafuri A, et al:
                                                                            Contributions of the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and
                                                                            Jak/STAT pathways to leukemia. Leukemia 22: 686‑707, 2008.
Availability of data and materials                                      12. Cox AD, Fesik SW, Kimmelman AC, Luo J and Der CJ:
                                                                            Drugging the undruggable RAS: Mission possible? Nat Rev
                                                                            Drug Discov 13: 828‑851, 2014.
The datasets used and/or analyzed during the current study are          13. Coombs CC, Tallman MS and Levine RL: Molecular therapy for
available from the corresponding author on reasonable request.              acute myeloid leukaemia. Nat Rev Clin Oncol 13: 305‑318, 2016.
                                                                        14. Kavanagh S, Murphy T, Law A, Yehudai D, Ho JM, Chan S and
                                                                            Schimmer AD: Emerging therapies for acute myeloid leukemia:
Authors' contributions                                                      Translating biology into the clinic. JCI Insight 2: e95679, 2017.
                                                                        15. Lu S, Jang H, Gu S, Zhang J and Nussinov R: Drugging Ras
JP acquired, analyzed and interpreted the data, and drafted                 GTPase: A comprehensive mechanistic and signaling structural
                                                                            view. Chem Soc Rev 45: 4929‑4952, 2016.
the manuscript. YK, HP, JMB, JH, DYS and SSY devised the                16. Schmieder R, Puehler F, Neuhaus R, Kissel M, Adjei AA,
study concept and design, and provided technical guidance                   Miner JN, Mumberg D, Ziegelbauer K and Scholz A: Allosteric
for all aspects of the project. All authors discussed the results           MEK1/2 inhibitor refametinib (BAY 86‑9766) in combination
                                                                            with sorafenib exhibits antitumor activity in preclinical murine
and contributed to the final manuscript. SSY and YK confirm                 and rat models of hepatocellular carcinoma. Neoplasia 15:
the authenticity of all the raw data. All authors have read and             1161‑1171, 2013.
approved the final manuscript.                                          17. Dickson MA, Gordon MS, Edelman G, Bendell JC,
                                                                            Kudchadkar RR, LoRusso PM, Johnston SH, Clary DO and
                                                                            Schwartz GK: Phase I study of XL281 (BMS‑908662), a potent
Ethics approval and consent to participate                                  oral RAF kinase inhibitor, in patients with advanced solid
                                                                            tumors. Invest New Drugs 33: 349‑356, 2015.
                                                                        18. Mendzelevski B, Ferber G, Janku F, Li BT, Sullivan RJ, Welsch D,
The study was reviewed and approved by the Institutional                    Chi W, Jackson J, Weng O and Sager PT: Effect of ulixertinib,
Review Board of Seoul National University Hospital                          a novel ERK1/2 inhibitor, on the QT/QTc interval in patients
(approval no. IRB‑1201‑099‑396; Seoul). Primary samples                     with advanced solid tumor malignancies. Cancer Chemother
                                                                            Pharmacol 81: 1129‑1141, 2018.
from patients with acute myeloid leukemia were obtained with            19. Ryan MB and Corcoran RB: Therapeutic strategies to target
written informed consent from all patients.                                 RAS‑mutant cancers. Nat Rev Clin Oncol 15: 709‑720, 2018.
                                                                        20. Borthakur G, Popplewell L, Boyiadzis M, Foran J, Platzbecker U,
                                                                            Vey N, Walter RB, Olin R, Raza A, Giagounidis A, et al: Activity
Patient consent for publication                                             of the oral mitogen‑activated protein kinase kinase inhibitor
                                                                            trametinib in RAS‑mutant relapsed or refractory myeloid malig‑
Not applicable.                                                             nancies. Cancer 122: 1871‑1879, 2016.
                                                                        21. Jain N, Curran E, Iyengar NM, Diaz‑Flores E, Kunnavakkam R,
                                                                            Popplewell L, Kirschbaum MH, Karrison T, Erba HP,
Competing interests                                                         Green M, et al: Phase II study of the oral MEK inhibitor selu‑
                                                                            metinib in advanced acute myelogenous leukemia: A University
                                                                            of Chicago phase II consortium trial. Clin Cancer Res 20:
The authors declare that they have no competing interests.                  490‑498, 2014.
                                                                        22. Maiti A, Naqvi K, Kadia TM, Borthakur G, Takahashi K, Bose P,
                                                                            Daver NG, Patel A, Alvarado Y, Ohanian M, et al: Phase II trial
References                                                                  of MEK inhibitor binimetinib (MEK162) in RAS‑mutant acute
                                                                            myeloid leukemia. Clin Lymphoma Myeloma Leuk 19: 142‑148,
 1. Almeida AM and Ramos F: Acute myeloid leukemia in the older             2019.
    adults. Leuk Res Rep 6: 1‑7, 2016.                                  23. Peng SB, Henry JR, Kaufman MD, Lu WP, Smith BD, Vogeti S,
 2. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI,                    Rutkoski TJ, Wise S, Chun L, Zhang Y, et al: Inhibition of RAF
    Paschka P, Roberts ND, Potter NE, Heuser M, Thol F,                     isoforms and active dimers by LY3009120 leads to anti‑tumor
    Bolli N, et al: Genomic classification and prognosis in acute           activities in RAS or BRAF mutant cancers. Cancer Cell 28:
    myeloid leukemia. N Engl J Med 374: 2209‑2221, 2016.                    384‑398, 2015.
 3. Lagunas‑Rangel FA, Chavez‑Valencia V, Gomez‑Guijosa MA              24. Keum H, Kim TW, Kim Y, Seo C, Son Y, Kim J, Kim D, Jung W,
    and Cortes‑Penagos C: Acute myeloid leukemia‑genetic altera‑            Whang CH and Jon S: Bilirubin nanomedicine alleviates psoriatic
    tions and their clinical prognosis. Int J Hematol Oncol Stem Cell       skin inflammation by reducing oxidative stress and suppressing
    Res 11: 328‑339, 2017.                                                  pathogenic signaling. J Control Release 325: 359‑369, 2020.
 4. Short NJ, Rytting ME and Cortes JE: Acute myeloid leukaemia.        25. Lee SH, Kim DK, Seo YR, Woo KM, Kim CS and Cho MH:
    Lancet 392: 593‑606, 2018.                                              Nickel (II)‑induced apoptosis and G2/M enrichment. Exp Mol
 5. Hobbs GA, Der CJ and Rossman KL: RAS isoforms and muta‑                 Med 30: 171‑176, 1998.
    tions in cancer at a glance. J Cell Sci 129: 1287‑1292, 2016.       26. Wang HM, Chen CY and Wu PF: Isophilippinolide A arrests cell
 6. Ward AF, Braun BS and Shannon KM: Targeting oncogenic Ras               cycle progression and induces apoptosis for anticancer inhibi‑
    signaling in hematologic malignancies. Blood 120: 3397‑3406,            tory agents in human melanoma cells. J Agric Food Chem 62:
    2012.                                                                   1057‑1065, 2014.
ONCOLOGY LETTERS 22: 745, 2021                                                       11

27. Chou TC: Drug combination studies and their synergy quantifica‑     34. Tambe M, Karjalainen E, Vähä‑Koskela M, Bulanova D,
    tion using the Chou‑Talalay method. Cancer Res 70: 440‑446, 2010.       Gjertsen BT, Kontro M, Porkka K, Heckman CA and
28. Zhao L, Au JL and Wientjes MG: Comparison of methods for                Wennerberg K: Pan‑RAF inhibition induces apoptosis in acute
    evaluating drug‑drug interaction. Front Biosci (Elite Ed) 2:            myeloid leukemia cells and synergizes with BCL2 inhibition.
    241‑249, 2010.                                                          Leukemia 34: 3186‑3196, 2020.
29. Kufe DW, Griffin JD and Spriggs DR: Cellular and clinical           35. Hall‑Jackson CA, Eyers PA, Cohen P, Goedert M, Boyle FT,
    pharmacology of low‑dose ara‑C. Semin Oncol 12 (Suppl 3):               Hewitt N, Plant H and Hedge P: Paradoxical activation of Raf by
    S200‑S207, 1985.                                                        a novel Raf inhibitor. Chem Biol 6: 559‑568, 1999.
30. Spriggs D, Griffin J, Wisch J and Kufe D: Clinical pharmacology     36. Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ,
    of low‑dose cytosine arabinoside. Blood 65: 1087‑1089, 1985.            Alvarado R, Ludlam MJ, Stokoe D, Gloor SL, Vigers G, et al:
31. Chen L, Guo P, Zhang Y, Li X, Jia P, Tong J and Li J: Autophagy         RAF inhibitors prime wild‑type RAF to activate the MAPK
    is an important event for low‑dose cytarabine treatment in acute        pathway and enhance growth. Nature 464: 431‑435, 2010.
    myeloid leukemia cells. Leuk Res 60: 44‑52, 2017                    37. King AJ, Patrick DR, Batorsky RS, Ho ML, Do HT, Zhang SY,
32. Liu Z, Chen J, Wang H, Ding K, Li Y, de Silva A, Sehgal V,              Kumar R, Rusnak DW, Takle AK, Wilson DM, et al:
    Burbano JL, Sundararaj R, Gamage J, et al: Chidamide shows              Demonstration of a genetic therapeutic index for tumors
    synergistic cytotoxicity with cytarabine via inducing G0/G1             expressing oncogenic BRAF by the kinase inhibitor SB‑590885.
    arrest and apoptosis in myelodysplastic syndromes. Am J Transl          Cancer Res 66: 11100‑11105, 2006.
    Res 9: 5631‑5642, 2017.                                             38. Wei AH, Strickland SA Jr, Hou JZ, Fiedler W, Lin TL, Walter RB,
33. Vakana E, Pratt S, Blosser W, Dowless M, Simpson N, Yuan XJ,            Enjeti A, Tiong IS, Savona M, Lee S, et al: Venetoclax combined
    Jaken S, Manro J, Stephens J, Zhang Y, et al: LY3009120, a              with low‑dose cytarabine for previously untreated patients with
    panRAF inhibitor, has significant anti‑tumor activity in BRAF           acute myeloid leukemia: Results from a phase Ib/II study. J Clin
    and KRAS mutant preclinical models of colorectal cancer.                Oncol 37: 1277‑1284, 2019.
    Oncotarget 8: 9251‑9266, 2017.
You can also read