scholarly journals All-trans retinoic acid (ATRA) in patients with chronic myeloid leukemia in the chronic phase

Leukemia ◽  
1998 ◽  
Vol 12 (4) ◽  
pp. 449-454 ◽  
Author(s):  
D Russo ◽  
M Regazzi ◽  
S Sacchi ◽  
G Visani ◽  
M Lazzarino ◽  
...  
Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2395-2395
Author(s):  
Wenli Liu ◽  
Hyun W Lee ◽  
Griffin P Rodgers

Abstract Abstract 2395 Poster Board II-372 Introduction: All-trans retinoic acid (ATRA) has been shown to induce cellular differentiation and growth inhibition of acute promyelocytic leukemia cells. Clinical application of ATRA has proved extremely successful in inducing clinical remission in most acute promyelocytic leukemia patients. Although the mechanisms of retinoid-dependent gene transcription regulation are well understood, the target genes that mediate retinoid-induced biological responses still remain to be defined. Olfactomedin 4 (OLFM4, also called hGC-1 and GW112) is a member of olfactomedin-related glycoprotein family. It is constitutively expressed in myeloid cells and gastrointestinal tract. It has been recently reported that OLFM4 expression is up-regulated in stomach and colon cancer patients. The purpose of this study is to examine its expression in myeloid leukemia patients, its regulation by ATRA and potential biological functions in myeloid leukemia. Results: 1) In this study, we found that OLFM4 expression was up-regulated in the peripheral leukocytes of chronic myeloid leukemia patients (91%, 22 cases) and acute myeloid leukemia patients (30%, 10 cases). OLFM4 expression in accelerated phase of chronic myeloid leukemia patients was significantly higher than that in chronic phase. 2) We identified that OLFM4 is a novel target gene of retinoic acids in myeloid leukemia cells. Treatment of HL-60 cells with ATRA and 9-cis-RA induced OLFM4 expression. The expression level of OLFM4 is correlated with the myeloid cell differentiation stage. Deletion analysis led to the identification of a positive retinoic acid response element (DR5) and a negative response element (DR1) within OLFM4 promoter. Furthermore, electrophoretic mobility-shift assays and transfection study in COS-7 cells demonstrated that RARα/RXRα binds to the DR5 site and mediates ATRA induced transactivation of OLFM4 promoter. 3) We showed that OLFM4 over-expression in HL-60 cells lead to growth inhibition, differentiation and apoptosis and potentates ATRA mediated these effects. Conversely, silencing of endogenous OLFM4 by lentiviral shRNA against OLFM4 in AML-193 cells reduces ATRA induced growth inhibition, differentiation and apoptosis. 4) We further investigated the molecular mechanism that OLFM4 is involved in leukemia cell growth and differentiation. We found that over-expression of OLFM4 in HL60 cells inhibited ATRA induced phosphorylation of translation repressor 4E-BP1. This inhibitory effect was further confirmed in 293T cells, in which over-expression of OLFM4 inhibits 4E-BP1 phosphorylation at all four phosphorylation sites (Thr37/46, Thr70 and Ser65). Application of lentiviral shRNA against OLFM4 in AML-193 increased phosphorylation of 4E-BP1 compared with control shRNA. Over-expression of OLFM4 in HL60 cells does not affect phosphorylation of Akt, p70S6 kinase, GSK3β and ERK1/2 . Conclusion: 1) OLFM4 expression is up-regulated in chronic and acute myeloid leukemia patients and is correlated with the stage of chronic myeloid leukemia. 2) OLFM4 is a novel target gene of retinoic acids. RARα/RXRα binds to the DR5 site of OLFM4 promoter and mediates the ATRA induced transactivation of OLFM4. 3) OLFM4 mediates ATRA induced growth inhibition, differentiation and apoptosis of myeloid leukemia cells. 4) OLFM4 acts an inhibitor of 4E-BP1 phosphorylation down stream of Akt and mTOR, suggesting OLFM4 may inhibit protein synthesis that is observed with the differentiation of myeloid leukemia cells. Our results suggest that OLFM4 up-regulation in the leukocytes of leukemia patients might have a feedback effect to restrain cell growth, and induce differentiation and apoptosis rather than act as an initiator of leukemiagenesis. As OLFM4 is a secreted glycoprotein, it could represent a promising therapeutic agent in the treatment of myeloid leukemia patients. The effect of purified OLFM4 on leukemia cells is currently under investigation. Disclosures: No relevant conflicts of interest to declare.


2000 ◽  
Vol 104 (2-3) ◽  
pp. 57-64 ◽  
Author(s):  
Fabio Stagno ◽  
Patrizia Guglielmo ◽  
Ugo Consoli ◽  
Giovanna Inghilterra ◽  
Giada Maria Giustolisi ◽  
...  

2008 ◽  
Vol 149 (32) ◽  
pp. 1509-1512
Author(s):  
Miklós Egyed ◽  
Balázs Kollár ◽  
Péter Rajnics ◽  
Éva Karádi ◽  
András Matolcsi

A krónikus myeloid leukaemia a pluripotens haemopoeticus őssejt malignus klonális betegsége. A kórkép genetikai háttere a t(9;22) (Philadelphia kromoszóma), amely konvencionális citogenetikával a krónikus myeloid leukaemiás esetek több mint 90%-ában kimutatható, illetve a bcr-abl fúziós gén, amely tirozin-kináz-aktivitású fúziós fehérje szintézisét kódolja. Mai ismereteink alapján ez a folyamatosan aktív tirozin-kináz a krónikus myeloid leukaemia oka. Az imatinib alkalmazása jelentősen megváltoztatta a krónikus myeloid leukaemia kezelését. Az eddigi eredmények a betegség minden fázisában magas arányú citogenetikai választ igazoltak. Módszerek: A szerzők az általuk kezelt 21, krónikus fázisú krónikus myeloid leukaemiás beteg citogenetikai és molekuláris válaszait mutatják be. Közülük 13 esetében az imatinib első vonalbeli kezelés volt, a 7 másik beteg esetében több szerrel (interferon-alfa, citarabin, all-trans-retinoic acid) történt előkezelést követően alkalmazták. Minden krónikus myeloid leukaemiás beteg kezelését hidroxi-ureával kezdték a hematológiai remisszió eléréséig. Háromhavonta perifériás vérből kvantitatív valós idejű polimeráz láncreakciós vizsgálatot, 6 havonta csontvelő-aspirátumból konvencionális citogenetikai vizsgálatot végeztek. Eredmények: Hidroxi-urea-kezeléssel minden betegüknél hematológiai remissziót értek el. A továbbiakban minden betegük a 6. hónapra komplett citogenetikai és a 12. hónapra major molekuláris remisszióba jutott. Következtetések: Az imatinibkezelés mindkét csoportban az összes beteg esetében komplett citogenetikai és major molekuláris remissziót okozott. Az elsődlegesen imatinibkezelt betegek esetén észlelt gyors és mély citogenetikai válasz felveti a hidroxi-urea által elért jelentős sejtredukció esetleges szerepét.


Cancers ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 2143
Author(s):  
Maria Hernandez-Valladares ◽  
Rebecca Wangen ◽  
Elise Aasebø ◽  
Håkon Reikvam ◽  
Frode S. Berven ◽  
...  

All-trans retinoic acid (ATRA) and valproic acid (VP) have been tried in the treatment of non-promyelocytic variants of acute myeloid leukemia (AML). Non-randomized studies suggest that the two drugs can stabilize AML and improve normal peripheral blood cell counts. In this context, we used a proteomic/phosphoproteomic strategy to investigate the in vivo effects of ATRA/VP on human AML cells. Before starting the combined treatment, AML responders showed increased levels of several proteins, especially those involved in neutrophil degranulation/differentiation, M phase regulation and the interconversion of nucleotide di- and triphosphates (i.e., DNA synthesis and binding). Several among the differentially regulated phosphorylation sites reflected differences in the regulation of RNA metabolism and apoptotic events at the same time point. These effects were mainly caused by increased cyclin dependent kinase 1 and 2 (CDK1/2), LIM domain kinase 1 and 2 (LIMK1/2), mitogen-activated protein kinase 7 (MAPK7) and protein kinase C delta (PRKCD) activity in responder cells. An extensive effect of in vivo treatment with ATRA/VP was the altered level and phosphorylation of proteins involved in the regulation of transcription/translation/RNA metabolism, especially in non-responders, but the regulation of cell metabolism, immune system and cytoskeletal functions were also affected. Our analysis of serial samples during the first week of treatment suggest that proteomic and phosphoproteomic profiling can be used for the early identification of responders to ATRA/VP-based treatment.


2021 ◽  
Vol 14 (5) ◽  
pp. 423
Author(s):  
Øystein Bruserud ◽  
Galina Tsykunova ◽  
Maria Hernandez-Valladares ◽  
Hakon Reikvam ◽  
Tor Henrik Anderson Tvedt

Even though allogeneic stem cell transplantation is the most intensive treatment for acute myeloid leukemia (AML), chemo-resistant leukemia relapse is still one of the most common causes of death for these patients, as is transplant-related mortality, i.e., graft versus host disease, infections, and organ damage. These relapse patients are not always candidates for additional intensive therapy or re-transplantation, and many of them have decreased quality of life and shortened expected survival. The efficiency of azacitidine for treatment of posttransplant AML relapse has been documented in several clinical trials. Valproic acid is an antiepileptic fatty acid that exerts antileukemic activity through histone deacetylase inhibition. The combination of valproic acid and all-trans retinoic acid (ATRA) is well tolerated even by unfit or elderly AML patients, and low-toxicity chemotherapy (e.g., azacitidine) can be added to this combination. The triple combination of azacitidine, valproic acid, and ATRA may therefore represent a low-intensity and low-toxicity alternative for these patients. In the present review, we review and discuss the general experience with valproic acid/ATRA in AML therapy and we discuss its possible use in low-intensity/toxicity treatment of post-allotransplant AML relapse. Our discussion is further illustrated by four case reports where combined treatments with sequential azacitidine/hydroxyurea, valproic acid, and ATRA were used.


Sign in / Sign up

Export Citation Format

Share Document