Cargando…

Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells

BACKGROUND: Fms-related tyrosine kinase 3 (FLT3) receptor serves as a prognostic marker and therapeutic target in acute myeloid leukemia (AML). Approximately one-third of AML patients carry mutation in FLT3, associated with unfavourable prognosis and high relapse rate. The multitargeted kinase inhib...

Descripción completa

Detalles Bibliográficos
Autores principales: Rörby, Emma, Adolfsson, Jörgen, Hultin, Erik, Gustafsson, Thomas, Lotfi, Kourosh, Cammenga, Jörg, Jönsson, Jan-Ingvar
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7852110/
https://www.ncbi.nlm.nih.gov/pubmed/33531064
http://dx.doi.org/10.1186/s40164-021-00201-w
_version_ 1783645754825375744
author Rörby, Emma
Adolfsson, Jörgen
Hultin, Erik
Gustafsson, Thomas
Lotfi, Kourosh
Cammenga, Jörg
Jönsson, Jan-Ingvar
author_facet Rörby, Emma
Adolfsson, Jörgen
Hultin, Erik
Gustafsson, Thomas
Lotfi, Kourosh
Cammenga, Jörg
Jönsson, Jan-Ingvar
author_sort Rörby, Emma
collection PubMed
description BACKGROUND: Fms-related tyrosine kinase 3 (FLT3) receptor serves as a prognostic marker and therapeutic target in acute myeloid leukemia (AML). Approximately one-third of AML patients carry mutation in FLT3, associated with unfavourable prognosis and high relapse rate. The multitargeted kinase inhibitor midostaurin (PKC412) in combination with standard chemotherapy (daunorubicin and cytarabine) was recently shown to increase overall survival of AML patients. For that reason, PKC412 has been approved for treatment of AML patients with FLT3-mutation. PKC412 synergizes with standard chemotherapy, but the mechanism involved is not fully understood and the risk of relapse is still highly problematic. METHODS: By utilizing the unique nature of mass cytometry for single cell multiparameter analysis, we have explored the proteomic effect and intracellular signaling response in individual leukemic cells with internal tandem duplication of FLT3 (FLT3-ITD) after midostaurin treatment in combination with daunorubicin or cytarabine. RESULTS: We have identified a synergistic inhibition of intracellular signaling proteins after PKC412 treatment in combination with daunorubicin. In contrast, cytarabine antagonized phosphorylation inhibition of PKC412. Moreover, we found elevated levels of FLT3 surface expression after cytarabine treatment. Interestingly, the surface localization of FLT3 receptor increased in vivo on the blast cell population of two AML patients during day 3 of induction therapy (daunorubicin; once/day from day 1–3 and cytarabine; twice/day from day 1–7). We found FLT3 receptor expression to correlate with intracellular cytarabine (AraC) response. AML cell line cultured with AraC with or without PKC412 had an antagonizing phosphorylation inhibition of pAKT (p = 0.042 and 0.0261, respectively) and pERK1/2 (0.0134 and 0.0096, respectively) in FLT3(high) compared to FLT3(low) expressing cell populations. CONCLUSIONS: Our study provides insights into how conventional chemotherapy affects protein phosphorylation of vital signaling proteins in human leukemia cells. The results presented here support further investigation of novel strategies to treat FLT3-mutated AML patients with PKC412 in combination with chemotherapy agents and the potential development of novel treatment strategies.
format Online
Article
Text
id pubmed-7852110
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-78521102021-02-03 Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells Rörby, Emma Adolfsson, Jörgen Hultin, Erik Gustafsson, Thomas Lotfi, Kourosh Cammenga, Jörg Jönsson, Jan-Ingvar Exp Hematol Oncol Research BACKGROUND: Fms-related tyrosine kinase 3 (FLT3) receptor serves as a prognostic marker and therapeutic target in acute myeloid leukemia (AML). Approximately one-third of AML patients carry mutation in FLT3, associated with unfavourable prognosis and high relapse rate. The multitargeted kinase inhibitor midostaurin (PKC412) in combination with standard chemotherapy (daunorubicin and cytarabine) was recently shown to increase overall survival of AML patients. For that reason, PKC412 has been approved for treatment of AML patients with FLT3-mutation. PKC412 synergizes with standard chemotherapy, but the mechanism involved is not fully understood and the risk of relapse is still highly problematic. METHODS: By utilizing the unique nature of mass cytometry for single cell multiparameter analysis, we have explored the proteomic effect and intracellular signaling response in individual leukemic cells with internal tandem duplication of FLT3 (FLT3-ITD) after midostaurin treatment in combination with daunorubicin or cytarabine. RESULTS: We have identified a synergistic inhibition of intracellular signaling proteins after PKC412 treatment in combination with daunorubicin. In contrast, cytarabine antagonized phosphorylation inhibition of PKC412. Moreover, we found elevated levels of FLT3 surface expression after cytarabine treatment. Interestingly, the surface localization of FLT3 receptor increased in vivo on the blast cell population of two AML patients during day 3 of induction therapy (daunorubicin; once/day from day 1–3 and cytarabine; twice/day from day 1–7). We found FLT3 receptor expression to correlate with intracellular cytarabine (AraC) response. AML cell line cultured with AraC with or without PKC412 had an antagonizing phosphorylation inhibition of pAKT (p = 0.042 and 0.0261, respectively) and pERK1/2 (0.0134 and 0.0096, respectively) in FLT3(high) compared to FLT3(low) expressing cell populations. CONCLUSIONS: Our study provides insights into how conventional chemotherapy affects protein phosphorylation of vital signaling proteins in human leukemia cells. The results presented here support further investigation of novel strategies to treat FLT3-mutated AML patients with PKC412 in combination with chemotherapy agents and the potential development of novel treatment strategies. BioMed Central 2021-02-02 /pmc/articles/PMC7852110/ /pubmed/33531064 http://dx.doi.org/10.1186/s40164-021-00201-w Text en © The Author(s) 2021 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.
spellingShingle Research
Rörby, Emma
Adolfsson, Jörgen
Hultin, Erik
Gustafsson, Thomas
Lotfi, Kourosh
Cammenga, Jörg
Jönsson, Jan-Ingvar
Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells
title Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells
title_full Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells
title_fullStr Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells
title_full_unstemmed Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells
title_short Multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in AML cells
title_sort multiplexed single‐cell mass cytometry reveals distinct inhibitory effects on intracellular phosphoproteins by midostaurin in combination with chemotherapy in aml cells
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7852110/
https://www.ncbi.nlm.nih.gov/pubmed/33531064
http://dx.doi.org/10.1186/s40164-021-00201-w
work_keys_str_mv AT rorbyemma multiplexedsinglecellmasscytometryrevealsdistinctinhibitoryeffectsonintracellularphosphoproteinsbymidostaurinincombinationwithchemotherapyinamlcells
AT adolfssonjorgen multiplexedsinglecellmasscytometryrevealsdistinctinhibitoryeffectsonintracellularphosphoproteinsbymidostaurinincombinationwithchemotherapyinamlcells
AT hultinerik multiplexedsinglecellmasscytometryrevealsdistinctinhibitoryeffectsonintracellularphosphoproteinsbymidostaurinincombinationwithchemotherapyinamlcells
AT gustafssonthomas multiplexedsinglecellmasscytometryrevealsdistinctinhibitoryeffectsonintracellularphosphoproteinsbymidostaurinincombinationwithchemotherapyinamlcells
AT lotfikourosh multiplexedsinglecellmasscytometryrevealsdistinctinhibitoryeffectsonintracellularphosphoproteinsbymidostaurinincombinationwithchemotherapyinamlcells
AT cammengajorg multiplexedsinglecellmasscytometryrevealsdistinctinhibitoryeffectsonintracellularphosphoproteinsbymidostaurinincombinationwithchemotherapyinamlcells
AT jonssonjaningvar multiplexedsinglecellmasscytometryrevealsdistinctinhibitoryeffectsonintracellularphosphoproteinsbymidostaurinincombinationwithchemotherapyinamlcells