Acessibilidade / Reportar erro

MicroRNAs and exosomes: promising new biomarkers in acute myeloid leukemias?

ABSTRACT

Despite advances in understanding of carcinogenesis and of treatment of acute myeloid leukemia, this neoplasm still has a lethality of at least 30%. The search for biomarkers that can predict the response to treatment in the early stages of the disease is still necessary. In recent years, a new form of cellular communication between tumor and non-neoplastic cells has been discovered: the exchange of information through extracellular vesicles. These are small vesicles released by membrane-coated cells that carry proteins, lipids, messenger RNAs, microRNA and DNA, which can be internalized and promote biological changes in target cells. Exosomes are qualified as a type of extracellular vesicle and, in tumors, carry immunoinhibitory signals that promote the escape of immune control. Recent studies have showed their involvement in communication with the cells of the tumor microenvironment and with chemoresistance in several tumors. To date, there is no information about immunoregulatory microRNAs transported by exosomes and their correlation with clinical evolution during chemotherapy for acute myeloid leukemia. Knowledge about immunomodulatory microRNAs obtained by leukemic cells and transported by exosomes can direct us towards the design of new diagnostic and treatment tools in this type of leukemia.

Leukemia, myeloid, acute; Exosomes; MicroRNAs; Immunomodulation; Biomarkers

INTRODUCTION

The overall survival rates for adult and childhood acute myeloid leukemia (AML), in addition to being suboptimal, have been stagnant for more than 20 years. This demonstrates the efficacy of chemotherapeutic regimens appears to have reached its upper limit. The need for the search for new therapeutic weapons against the disease is urgent. The way in which the tumor cell acts on the immune system and its ability to produce exosomes that influence the tumor niche has drawn attention of the scientific community. Thus, the transfer of non-coding RNAs by exosomes and the modulation of gene expression in target cells present new horizons to be explored.

ACUTE MYELOID LEUKEMIA

Acute myeloid leukemia is a hematologic malignancy of complex pathogenesis, characterized by abnormal proliferation and differentiation of clonal populations of myeloid stem cells in the bone marrow.(11. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. WHO classification of tumours. 4th Revised ed. Lyon: WHO; 2017. p. 586 [WHO series on histological and genetic typing of human tumours].)

The worldwide incidence of AML is 2.5 to three new cases per 100,000 individuals per year, and although it can occur at any age, it is most commonly diagnosed in individuals aged over 65 years.(11. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. WHO classification of tumours. 4th Revised ed. Lyon: WHO; 2017. p. 586 [WHO series on histological and genetic typing of human tumours].) In children under 15 years of age, it accounts for only 15% to 20% of acute leukemia cases.(11. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. WHO classification of tumours. 4th Revised ed. Lyon: WHO; 2017. p. 586 [WHO series on histological and genetic typing of human tumours].)

Some genetic and environmental risk factors seem to be associated with a higher frequency of AML. Among the constitutional genetic syndromes that may be correlated with AML, Down syndrome, Fanconi anemia, Blackfan-Diamond anemia, Shwachman-Diamond syndrome, and congenital neutropenia (Kostmann syndrome) stand out. Among the environmental factors, exposure to ionizing radiation, alkylating agents, and topoisomerase-II inhibitors are of epidemiological importance.(11. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. WHO classification of tumours. 4th Revised ed. Lyon: WHO; 2017. p. 586 [WHO series on histological and genetic typing of human tumours].)

The treatment protocol of the Berlin-Frankfurt-Münster (BFM) Group is the most widely used for treatment of AML in Brazil. The treatment is based on induction chemotherapy, with the infusion of cytarabine (ARA-C), idarubicin, and etoposide (VP-16), in addition to the administration of intrathecal cytarabine.(22. Rasche M, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, et al. Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia. 2018;32(10):2167-77.)Despite the high remission rate, 90% of patients relapse if they do not continue the protocol with consolidation and intensification. Consolidation is performed in patients classified as low risk on the 15th day of induction, using prednisone, thioguanine (Lanvis™), vincristine, doxorubicin (Adriamycin), cytarabine, intrathecal cytarabine, cyclophosphamide, and granulocyte colony-stimulating factor (G-CSF).(22. Rasche M, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, et al. Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia. 2018;32(10):2167-77.)The other patients start consolidation after high-dose cytarabine and etoposide intensification, while low-risk patients perform intensification after consolidation.(22. Rasche M, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, et al. Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia. 2018;32(10):2167-77.)

A considerable part of the increase in survival rates of these patients, however, is due to the improvement in supportive treatment for the complications of chemotherapy. Allogeneic hematopoietic stem cell transplantation in the first remission is also an option, especially for those who are poor responders to induction therapy or who have poor prognostic cytogenetic and molecular characteristics.(22. Rasche M, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, et al. Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia. 2018;32(10):2167-77.)

Despite the advances made in the last 40 years, the survival rates for AML are less than desirable, remaining around 70% at 5 years, even for good responders to induction therapy.(22. Rasche M, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, et al. Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia. 2018;32(10):2167-77.) New therapies are already being used for gene variants of AML, such as FLT3 (midostaurin and gilterinib), BCL2 (venetoclax), and IDH1/IDH2 (ivosidenib and enasidenib) inhibitors.(33. Short NJ, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD, et al. Advances in the treatment of acute myeloid leukemia: new drugs and new challenges. Cancer Discov. 2020;10(4):506-25. Review.) However, such alternative therapies are not yet widely used, either because of their cost or because they require approval for use in Brazil.

WHAT IS KNOWN ABOUT EXTRACELLULAR VESICLES, EXOSOMES, AND MICRORNAS IN ACUTE MYELOID LEUKEMIAS?

Extracellular vesicles are secreted by almost all cell types and have the function of performing intercellular communication, which can occur through ligand-receptor interaction, by fusion, and/or internalization.(44. Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Mol Oncol. 2021;15(1):3-26. Review.,55. Fleury A, Martinez MC, Le Lay S. Extracellular vesicles as therapeutic tools in cardiovascular diseases. Front Immunol. 2014;5:370. Review.) As mediators of intercellular communication, the extracellular vesicles can transmit proteins and lipids, besides different types of RNAs and DNA.(44. Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Mol Oncol. 2021;15(1):3-26. Review.)

Extracellular vesicles are classified according to their size into exosomes (30nm-150nm), microvesicles (0.1μm-2μm) and apoptotic bodies (0.1μm-5μm).(44. Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Mol Oncol. 2021;15(1):3-26. Review.,66. Zhou X, Xie F, Wang L, Zhang L, Zhang S, Fang M, et al. The function and clinical application of extracellular vesicles in innate immune regulation. Cell Mol Immunol. 2020;17(4):323-34. Review.) Recently, exosomes were subclassified into Exo-L (large exosome vesicles) and Exo-S (small exosome vesicles), with 90nm-150nm and 60nm-80nm, respectively. In addition, the term “exomere” was created for classification of non-membranous nanoparticles with approximately 35nm.(66. Zhou X, Xie F, Wang L, Zhang L, Zhang S, Fang M, et al. The function and clinical application of extracellular vesicles in innate immune regulation. Cell Mol Immunol. 2020;17(4):323-34. Review.)

Besides being differentiated by size, extracellular vesicles have different densities and origins. While microvesicles and apoptotic bodies are formed from the direct evagination of the plasma membrane, exosomes are formed from a more complex process, which involves the incorporation of protein or molecular content by invagination into cytoplasmic multivesicular endosomes.(44. Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Mol Oncol. 2021;15(1):3-26. Review.) Subsequently, these endosomes are fused to the plasma membrane, releasing the exosomes into the extracellular environment. As a consequence of their endosomal origin, exosomes contain proteins involved in membrane transport and fusion (e.g., Rab GTPases and annexin), as well as in the biogenesis of multivesicular bodies (e.g., TSG101), and integrins and tetraspanins (e.g., CD9, CD63, CD81, and CD82).(77. Simons M, Raposo G. Exosomes--vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21(4):575-81. Review.)

Various physiological and pathological stimuli can increase the production of extracellular vesicles, which can be detected in body fluids. This feature has opened the horizons for investment in research aimed to use these changes as possible biomarkers in various diseases.(77. Simons M, Raposo G. Exosomes--vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21(4):575-81. Review.,88. Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;2:20360.)

With respect to exosomes, tumor-derived exosomes are known to carry immunoinhibitory signals that promote escape from immune control. Evidence shows that tumor cell-derived exosomes affect proliferation, apoptosis, and cytokine production, and are able to reprogram cells of innate and adaptive immunity. It has been described, for example, that exosomes containing miRNA-21-3p, miRNA-185d-5p, and miRNA-1246 have the potential to reprogram macrophages into tumor support agents. Furthermore, exosomes expressing transforming growth factor β1 (TGF-β1) and binding to NKG2D may downregulate NKG2D expression, reducing natural killer (NK)-induced cytotoxicity.(66. Zhou X, Xie F, Wang L, Zhang L, Zhang S, Fang M, et al. The function and clinical application of extracellular vesicles in innate immune regulation. Cell Mol Immunol. 2020;17(4):323-34. Review.)Moreover, exosomes are involved in communication with cells in the tumor microenvironment and chemoresistance in several solid neoplasms.(77. Simons M, Raposo G. Exosomes--vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21(4):575-81. Review.,88. Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;2:20360.)

Exosomes can carry inside them microRNAs (miRNAs), small endogenous non-coding RNAs (19 to 25 nucleotides), originally identified as regulators of larval development in Caenorhabditis elegans.(99. Lee CR, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993; 75(5):843-54.) Later, they were also associated with several types of cancer and with various cellular processes, such as DNA methylation, cell growth, differentiation, and apoptosis.(1010. Ultimo S, Martelli AM, Zauli G, Vitale M, Calin GA, Neri LM. Roles and clinical implications of microRNAs in acute lymphoblastic leukemia. J Cell Physiol. 2018;233(8):5642-54. Review.) Abnormal levels of these miRNAs may play a role in carcinogenesis, since certain overexpressed miRNAs can lead to silencing of tumor suppressors.(1010. Ultimo S, Martelli AM, Zauli G, Vitale M, Calin GA, Neri LM. Roles and clinical implications of microRNAs in acute lymphoblastic leukemia. J Cell Physiol. 2018;233(8):5642-54. Review.)The imbalance of these miRNAs has been described in several types of cancers and in hematological neoplasms. Additionally, miRNAs can induce molecular alterations in target cells, affecting the regulation of specific genes, making it pathologically possible to alter processes, such as differentiation, proliferation, and apoptosis.(1111. Ehsanpour A, Saki N, Bagheri M, Maleki Behzad M, Abroun S. The expression of microvesicles in leukemia: prognostic approaches. Cell J. 2019;21(2):115-23.)

Recent studies suggest that extracellular vesicles carry an important amount of non-coding RNA that is relevant to the pathogenesis of leukemias. Some miRNAs have already been described as involved in the pathogenesis of AMLs, as reviewed by Trino et al.,(1212. Trino S, Lamorte D, Caivano A, Laurenzana I, Tagliaferri D, Falco G, et al. MicroRNAs as new biomarkers for diagnosis and prognosis, and as potential therapeutic targets in acute myeloid leukemia. Int J Mol Sci. 2018;19(2):460. Review.) and Li et al,.(1313. Li M, Cui X, Guan H. MicroRNAs: pivotal regulators in acute myeloid leukemia. Ann Hematol. 2020;99(3):399-412. Review.) as shown on table 1.

Table 1
MicroRNAs as prognostic biomarkers in acute myeloid leukemia

Exosomes originating from leukemic cells in AML patients have a distinct molecular profile when compared to healthy individuals.(1414. Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.) In addition to conventional markers for exosomes such as tetrapanins, AML exosomes contain TGF-β1, MICA/MICB, FasL, and markers of myeloid blast cells (CD33, CD34, and CD117).(1414. Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.)Ex vivo, these exosomes decrease NK cell cytotoxicity by decreasing expression of NKG2D.(1414. Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.) In addition, TGF-β1 is partially responsible for NK cell dysfunction in AML. Thus, it can be said that exosomes derived from leukemic cells in AML have the potential to modulate the immune system and may influence disease progression as well as response to therapy.(1414. Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.)A summary of functional studies demonstrating the relation between altered exosome production and AML is shown on table 2.(1515. Huan J, Hornick NI, Shurtleff MJ, Skinner AM, Goloviznina NA, Roberts CT Jr, et al. RNA trafficking by acute myelogenous leukemia exosomes. Cancer Res. 2013;73(2):918-29.

16. Huan J, Hornick NI, Goloviznina NA, Kamimae-Lanning AN, David LL, Wilmarth PA, et al. Coordinate regulation of residual bone marrow function by paracrine trafficking of AML exosomes. Leukemia. 2015;29(12):2285-95.

17. Kumar B, Garcia M, Weng L, Jung X, Murakami JL, Hu X, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575-87.

18. Viola S, Traer E, Huan J, Hornick NI, Tyner JW, Agarwal A, et al. Alterations in acute myeloid leukaemia bone marrow stromal cell exosome content coincide with gains in tyrosine kinase inhibitor resistance. Br J Haematol. 2016;172(6):983-6.
-1919. Hong CS, Sharma P, Yerneni SS, Simms P, Jackson EK, Whiteside TL, et al. Circulating exosomes carrying an immunosuppressive cargo interfere with cellular immunotherapy in acute myeloid leukemia. Sci Rep. 2017;7(1):14684.)

Table 2
Review on functional studies of exosomes in acute myeloid leukemia

The first study to show that AML cell lines release exosomes was published in 2013.(1515. Huan J, Hornick NI, Shurtleff MJ, Skinner AM, Goloviznina NA, Roberts CT Jr, et al. RNA trafficking by acute myelogenous leukemia exosomes. Cancer Res. 2013;73(2):918-29.) Exosomes appear to be involved in the deregulation of hematopoiesis during the conversion from a homeostatic microenvironment to a leukemic niche. In the in vivo model,(1616. Huan J, Hornick NI, Goloviznina NA, Kamimae-Lanning AN, David LL, Wilmarth PA, et al. Coordinate regulation of residual bone marrow function by paracrine trafficking of AML exosomes. Leukemia. 2015;29(12):2285-95.)exosomes of AML cell lines have been observed to migrate to the bone marrow, decreasing the colony-forming capacity of hematopoietic progenitor stem cells, and the expression of several transcription factors of genes involved in hematopoiesis.(1616. Huan J, Hornick NI, Goloviznina NA, Kamimae-Lanning AN, David LL, Wilmarth PA, et al. Coordinate regulation of residual bone marrow function by paracrine trafficking of AML exosomes. Leukemia. 2015;29(12):2285-95.) The influence of extracellular vesicles on the tumor niche was also observed by Kumar et al.,(1717. Kumar B, Garcia M, Weng L, Jung X, Murakami JL, Hu X, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575-87.) bone marrow preconditioning with exosomes from AML cells accelerated tumor engraftment and growth, promoted increased expression of DKK1 in bone marrow (a negative regulator of osteoblastic development), and caused reduced trabecular bone volume.(1717. Kumar B, Garcia M, Weng L, Jung X, Murakami JL, Hu X, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575-87.) In patients with AML, plasma levels of the bone formation indicator gene (OCN) are decreased.(1717. Kumar B, Garcia M, Weng L, Jung X, Murakami JL, Hu X, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575-87.) Interestingly, the number of circulating exosomes in plasma is significantly increased in patients with AML when compared to healthy individuals.(1717. Kumar B, Garcia M, Weng L, Jung X, Murakami JL, Hu X, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575-87.) In view of these findings, the authors emphasize the potential need to treat the induced changes in the bone marrow stroma as well, and not only to eradicate leukemic cells. Viola et al.,(1818. Viola S, Traer E, Huan J, Hornick NI, Tyner JW, Agarwal A, et al. Alterations in acute myeloid leukaemia bone marrow stromal cell exosome content coincide with gains in tyrosine kinase inhibitor resistance. Br J Haematol. 2016;172(6):983-6.) corroborated these observations and showed that monocyte-derived exosomes from patient stroma protect the AML cell line against chemotherapy treatment with cytarabine or FLT3 inhibitor.(1818. Viola S, Traer E, Huan J, Hornick NI, Tyner JW, Agarwal A, et al. Alterations in acute myeloid leukaemia bone marrow stromal cell exosome content coincide with gains in tyrosine kinase inhibitor resistance. Br J Haematol. 2016;172(6):983-6.)

Among the investigations that evaluated the content of extracellular vesicles in AML, miRNA in exosomes circulating in plasma(2020. Hornick NI, Huan J, Doron B, Goloviznina NA, Lapidus J, Chang BH, et al. Serum exosome MicroRNA as a minimally-invasive early biomarker of AML. Sci Rep. 2015;5:11295.) and released by bone marrow monocytes,(2121. Barrera-Ramirez J, Lavoie JR, Maganti HB, Stanford WL, Ito C, Sabloff M, et al. Micro-RNA profiling of exosomes from marrow-derived mesenchymal stromal cells in patients with acute myeloid leukemia: implications in leukemogenesis. Stem Cell Rev Rep. 2017;13(6):817-25.) and apoptotic proteins found in exosomes derived from bone marrow blasts have been described.(2222. Wojtuszkiewicz A, Schuurhuis GJ, Kessler FL, Piersma SR, Knol JC, Pham TV, et al. Exosomes secreted by apoptosis-resistant acute myeloid leukemia (AML) blasts harbor regulatory network proteins potentially involved in antagonism of apoptosis. Mol Cell Proteomics. 2016;15(4):1281-98.) In patients with AML refractory to treatment with adoptive cell therapy using NK-92 cells, exosomes isolated from plasma have been shown to reduce the cytolytic activity of NK-92 cells and to decrease their migration capacity.(1919. Hong CS, Sharma P, Yerneni SS, Simms P, Jackson EK, Whiteside TL, et al. Circulating exosomes carrying an immunosuppressive cargo interfere with cellular immunotherapy in acute myeloid leukemia. Sci Rep. 2017;7(1):14684.) Exosomes have also been found to carry TGF-β and PD-L1 (programmed cell death ligand 1), an important T cell inhibitor.(1919. Hong CS, Sharma P, Yerneni SS, Simms P, Jackson EK, Whiteside TL, et al. Circulating exosomes carrying an immunosuppressive cargo interfere with cellular immunotherapy in acute myeloid leukemia. Sci Rep. 2017;7(1):14684.)

In addition, AML blasts can secrete extracellular vesicles that express myeloid cell line-specific markers distinct from those present in normal cells. The expression of CD13, CD34, CD117, and CD33 on extracellular vesicles isolated from plasma of AML patients may indicate the presence of active leukemic cells.(2323. Pando A, Reagan JL, Quesenberry P, Fast LD. Extracellular vesicles in leukemia. Leuk Res. 2018;64:52-60. Review.) Thus, in clinical practice, flow cytometry assays may incorporate the search for extracellular vesicles as biomarkers of disease activity.

COMMENT

Although some studies have already evaluated some of the content of extracellular vesicles at specific times in acute myeloid leukemia, no research has shown whether there is modification of this content over the course of treatment. In addition, some immunoregulatory microRNAs have not yet been evaluated and, perhaps, they may have relevance in the pathogenesis of acute myeloid leukemia. Likewise, very little is known about the dynamics of extracellular vesicles in childhood leukemias.

The importance of circulating exosomes and cellular communication by microRNA in acute leukemias remains poorly elucidated, but recent studies(2424. Xu YC, Lin YS, Zhang L, Lu Y, Sun YL, Fang ZG, et al. MicroRNAs of bone marrow mesenchymal stem cell-derived exosomes regulate acute myeloid leukemia cell proliferation and apoptosis. Chin Med J (Engl). 2020;133(23):2829-39.) indicate the possibility that in the future, extracellular vesicles will be used as a substrate in laboratory assays for patient follow-up, prognosis determination, and therapeutic response.

REFERENCES

  • 1
    Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. WHO classification of tumours. 4th Revised ed. Lyon: WHO; 2017. p. 586 [WHO series on histological and genetic typing of human tumours].
  • 2
    Rasche M, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, et al. Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia. 2018;32(10):2167-77.
  • 3
    Short NJ, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD, et al. Advances in the treatment of acute myeloid leukemia: new drugs and new challenges. Cancer Discov. 2020;10(4):506-25. Review.
  • 4
    Schubert A, Boutros M. Extracellular vesicles and oncogenic signaling. Mol Oncol. 2021;15(1):3-26. Review.
  • 5
    Fleury A, Martinez MC, Le Lay S. Extracellular vesicles as therapeutic tools in cardiovascular diseases. Front Immunol. 2014;5:370. Review.
  • 6
    Zhou X, Xie F, Wang L, Zhang L, Zhang S, Fang M, et al. The function and clinical application of extracellular vesicles in innate immune regulation. Cell Mol Immunol. 2020;17(4):323-34. Review.
  • 7
    Simons M, Raposo G. Exosomes--vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21(4):575-81. Review.
  • 8
    Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;2:20360.
  • 9
    Lee CR, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993; 75(5):843-54.
  • 10
    Ultimo S, Martelli AM, Zauli G, Vitale M, Calin GA, Neri LM. Roles and clinical implications of microRNAs in acute lymphoblastic leukemia. J Cell Physiol. 2018;233(8):5642-54. Review.
  • 11
    Ehsanpour A, Saki N, Bagheri M, Maleki Behzad M, Abroun S. The expression of microvesicles in leukemia: prognostic approaches. Cell J. 2019;21(2):115-23.
  • 12
    Trino S, Lamorte D, Caivano A, Laurenzana I, Tagliaferri D, Falco G, et al. MicroRNAs as new biomarkers for diagnosis and prognosis, and as potential therapeutic targets in acute myeloid leukemia. Int J Mol Sci. 2018;19(2):460. Review.
  • 13
    Li M, Cui X, Guan H. MicroRNAs: pivotal regulators in acute myeloid leukemia. Ann Hematol. 2020;99(3):399-412. Review.
  • 14
    Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.
  • 15
    Huan J, Hornick NI, Shurtleff MJ, Skinner AM, Goloviznina NA, Roberts CT Jr, et al. RNA trafficking by acute myelogenous leukemia exosomes. Cancer Res. 2013;73(2):918-29.
  • 16
    Huan J, Hornick NI, Goloviznina NA, Kamimae-Lanning AN, David LL, Wilmarth PA, et al. Coordinate regulation of residual bone marrow function by paracrine trafficking of AML exosomes. Leukemia. 2015;29(12):2285-95.
  • 17
    Kumar B, Garcia M, Weng L, Jung X, Murakami JL, Hu X, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575-87.
  • 18
    Viola S, Traer E, Huan J, Hornick NI, Tyner JW, Agarwal A, et al. Alterations in acute myeloid leukaemia bone marrow stromal cell exosome content coincide with gains in tyrosine kinase inhibitor resistance. Br J Haematol. 2016;172(6):983-6.
  • 19
    Hong CS, Sharma P, Yerneni SS, Simms P, Jackson EK, Whiteside TL, et al. Circulating exosomes carrying an immunosuppressive cargo interfere with cellular immunotherapy in acute myeloid leukemia. Sci Rep. 2017;7(1):14684.
  • 20
    Hornick NI, Huan J, Doron B, Goloviznina NA, Lapidus J, Chang BH, et al. Serum exosome MicroRNA as a minimally-invasive early biomarker of AML. Sci Rep. 2015;5:11295.
  • 21
    Barrera-Ramirez J, Lavoie JR, Maganti HB, Stanford WL, Ito C, Sabloff M, et al. Micro-RNA profiling of exosomes from marrow-derived mesenchymal stromal cells in patients with acute myeloid leukemia: implications in leukemogenesis. Stem Cell Rev Rep. 2017;13(6):817-25.
  • 22
    Wojtuszkiewicz A, Schuurhuis GJ, Kessler FL, Piersma SR, Knol JC, Pham TV, et al. Exosomes secreted by apoptosis-resistant acute myeloid leukemia (AML) blasts harbor regulatory network proteins potentially involved in antagonism of apoptosis. Mol Cell Proteomics. 2016;15(4):1281-98.
  • 23
    Pando A, Reagan JL, Quesenberry P, Fast LD. Extracellular vesicles in leukemia. Leuk Res. 2018;64:52-60. Review.
  • 24
    Xu YC, Lin YS, Zhang L, Lu Y, Sun YL, Fang ZG, et al. MicroRNAs of bone marrow mesenchymal stem cell-derived exosomes regulate acute myeloid leukemia cell proliferation and apoptosis. Chin Med J (Engl). 2020;133(23):2829-39.

Publication Dates

  • Publication in this collection
    07 Mar 2022
  • Date of issue
    2022

History

  • Received
    30 Sept 2020
  • Accepted
    20 Aug 2021
Instituto Israelita de Ensino e Pesquisa Albert Einstein Avenida Albert Einstein, 627/701 , 05651-901 São Paulo - SP, Tel.: (55 11) 2151 0904 - São Paulo - SP - Brazil
E-mail: revista@einstein.br