Elsevier

Leukemia Research

Volume 64, January 2018, Pages 52-60
Leukemia Research

Invited review
Extracellular vesicles in leukemia

https://doi.org/10.1016/j.leukres.2017.11.011Get rights and content

Highlights

  • Comprehensive Review of Leukemia extracellular vesicles.

  • Leukemia extracellular vesicle characterization.

  • Leukemic extracellular vesicles alter their microenvironment.

  • Cancer extracellular vesicles act as immunomodulators.

  • Extracellular vesicle clinical, diagnostic, and therapeutic potential.

Abstract

Extracellular vesicles (EV) are nano-sized membrane enclosed vehicles that are involved in cell-to-cell communication and carry cargo that is representative of the parent cell. Recent studies have highlighted the significant roles leukemia EVs play in tumor progression, and ways in which they can lead to treatment evasion, thus meriting further investigation. Leukemia EVs are involved in crosstalk between the leukemia cell and its surroundings, transforming it into a cancer favorable microenvironment. Due to the diverse biological content found in leukemia EVs, they have an assortment of effects on the cells they interact with and can be harnessed as candidates for diagnostic and therapeutic treatments. This review focuses on EVs in the context of leukemia and the means by which they modulate their microenvironment, hematopoiesis, and the immune system to facilitate malignancy. We will also address current and prospective EV-based therapeutics.

Introduction

EVs are small, membrane-enclosed heterogeneous spheres of varying size that are directly secreted by all cells and carry a select cargo of cellular RNA, bioactive lipids, DNA and proteins that is usually representative of the parent cell [1], [2]. EVs are present in physiological and pathological circumstances, and their numbers, cellular origin, composition, and function are cell type and disease dependent [3]. They are known for their coagulant phenotype but can also affect inflammation, angiogenesis, and intercellular signaling [4]. There is increasing evidence suggesting that these vesicles have an important role in the regulation of immune stimulation or suppression that can drive inflammatory, autoimmune, and infectious disease pathology [5]. Furthermore, EVs have the potential to alter the fate of their target cells by regulating gene expression, partially through epigenetic changes in the recipient cells [6].

The first line of evidence that tumor cells shed membrane-vesicles came in 1978 from studies performed by Friend and colleagues on samples obtained from patients with Hodgkin’s disease [7]. In 1979, an independent study identified plasma-derived vesicles released by murine leukemia cells [8]. In the early 1980s, studies reported that EVs from pig hepatocellular carcinoma and mouse breast carcinoma cells were carriers of procoagulant activity. However, it was not until twenty years later that vesicles were acknowledged not to be artifacts but an active component of tumor microenvironment [9]. As in solid tumor malignancies, patients with hematologic malignancies have high levels of EVs in their biological fluids compared to normal controls that include cases of acute myeloid leukemia (AML) and chronic lymphocytic leukemia (CLL) [10], [11]. Moreover, EV cargo also increases in patients with hematological malignancies [12], [13].

EVs can be categorized based on their size. Microvesicles are 200–1000 nm in size and are formed by outward budding from the plasma membrane. Apoptotic bodies are 800–5000 nm in diameter and are released by cells undergoing programmed cell death [14]. More recently another category of EVs has been identified known as oncosomes that are much larger (1–10 μm) and are released as pinched membrane blebs from amoeboid cancer cells [15]. Exosomes are a subset of EV’s reported to vary between 30 and 100 nm, although the exact size varies with isolation approach and cellular conditions. Whereas microvesicles form by budding from the plasma membrane followed by fission of their connecting membrane stalks, exosomes are formed by a combination of processes starting with the inward invagination of clathrin-coated microdomains on the plasma membrane, suggesting a more active sorting mechanism. The Endosomal Sorting Complex Required for Transport (ESCRT) System facilitates the development of the invaginated vacuoles carrying ubiquitinated cargos into early endosomes. A second invagination of vesicles termed intraluminal vesicles (ILVs) occurs into the endosomes where they accumulate into large multivesicular bodies [16]. From this point they may be trafficked to lysosomes or fused with the plasma membrane for the release of ILVs at which point they are referred to as exosomes [17]. Sorting of exosomes by the ESCRT involves several proteins that are subsequently used to identify them as exosomes including ALG-2-interacting protein X (ALIX) and tumor susceptibility gene 101 protein (TSG101) [18]. Other proteins most frequently associated with identification of exosomes are tetraspanins (e.g., CD9, CD63, and CD81) membrane transporters and fusion proteins (e.g., GTPases, annexins, and flotillin), heat shock proteins, lipid-related proteins, and phospholipids [19]. Exosomes are involved in numerous biological functions such as intercellular communication, antigen presentation, protein secretion, and RNA shuttling [20]. Exosomes can play a key role in physiological and pathological processes depending on the cell of origin that induce proliferation, differentiation, inhibition, quiescence or cellular death.

To isolate EV populations various methodologies have been implemented including ultracentrifugation, size exclusion chromatography, microfluidics, immunoaffinity capture based techniques, size-based techniques, precipitation, and more [21]. Each methodology exploits a particular trait of EVs including size, density or immunophenotype [21]. Some techniques combine several different methodologies in an effort to isolate purer populations [22]. Due to the specific advantages and disadvantages each method provides, there is no standardized method for EV isolation [23]. For the purpose of this review, exosomes and microvesicles are referred to as EVs because of the potential heterogeneity that results from the different isolation procedures [23].

Due to EV ability to contain a subset of specific molecules and antigens from the parent cell, there is increasing evidence suggesting that EVs can serve as biomarkers [24]. Tumor EVs contain unique genetic information about the tumor related to its state of malignancy, cellular type, and susceptibility to therapeutic treatment. As such, they may be used for early diagnosis and monitoring of cancer. Importantly, EVs are used by tumors to evade or influence their environment to favor tumor progression. These tumor derived EVs are implicated in modulating the tumor microenvironment [25], acting as immunomodulators [26], and contributing to inhibition of anti-tumor activity [27]. The complexity of the biology of EVs is just being discovered (Fig. 1). This review focuses on the role of EVs in the pathogenesis of hematologic malignancies and their potential utilization in therapies (Table 1).

Section snippets

Leukemia EV content

AML EV content has been proposed to vary depending on the stage of disease and disease specific cell type [26]. Newly diagnosed AML patient plasma derived EVs, were shown to contain MICA/MICB, TGF-β1, myeloid blast markers (CD34, CD33, CD117), and a variety of microRNAs [10]. Other studies found that plasma-derived EVs in patients with AML carry the leukemia blast-relevant proteins CD34, CD44, CD96, CD123 and CLL-1 [13]. Hong and colleagues observed that TGF-β1 is found at higher concentrations

Leukemia EVs in the bone marrow microenvironment

Leukemia cells utilize EVs to transfer functional information to their microenvironment in amounts sufficient to alter intrinsic levels of respective molecules. The bone marrow niche is generally composed of osteocytes/osteoblasts/osteoclasts, the bone matrix, perivascular cells, quiescent hematopoietic stem cells (HSCs), sinusoidal endothelium, mesenchymal stem cells (MSCs), actively dividing HSC’s, different stroma cells, and immune cells [38]. EVs exert pleiotropic biological functions via

EVs act as immunomodulators

Emerging evidence suggests that tumor EVs play an important role as immune suppressors and modulators [10]. The molecular mechanisms underlying the modulation of the immune microenvironment by receptors, proteins, RNA, and DNA carried in leukemic EVs are still largely unknown [10]. Reported immune suppressing effects of EVs range from apoptosis of activated T-cells [69], [70] to impairment of monocyte differentiation into dendritic cells (DCs) [71].

EV-based clinical, diagnostic, and therapeutic potential

EVs ability to simultaneously contain tumor-specific cargo, play an important role in malignancy progression, and contain natural delivery characteristics makes them an attractive option for therapeutic use [35]. Investigators have proposed various avenues for their clinical use including diagnostics [26], drug delivery vehicles [83], and therapeutic targeting [84]. Many challenges before clinical implementation remain, but EVs ability to serve as micro tumor surrogates poses promise [85].

Conclusion

EVs are important bioactive vehicles that leukemic cells use to transform their surroundings to a microenvironment supporting their growth and survival, while inhibiting the immune system and normal hematopoiesis. Furthermore, leukemia EVs and their biological cargo provide unique information about pathological disease that can be harnessed for not just diagnostic or prognostic purposes but also as anti-leukemic treatment modalities. Although limited, EV use in the clinic has been well

Contributions

Alejandro Pando prepared the manuscript, John Reagan, Peter Quesenberry and Loren Fast reviewed and edited the manuscript.

Acknowledgements

Alejandro Pando is a recipient of a Brown University Presidential Graduate Fellowship. The authors would like to thank Patrycja Dubielecka-Szczerba for her proofreading of the manuscript.

References (100)

  • J. Paggetti et al.

    Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts

    Blood

    (2015)
  • T. Umezu et al.

    Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF -1

    Blood

    (2014)
  • A.K. Ghosh et al.

    Circulating microvesicles in B-cell chronic lymphocytic leukemia can stimulate marrow stromal cells: implications for disease progression

    Blood

    (2010)
  • J. Wang et al.

    Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells

    Blood

    (2014)
  • H. Tadokoro et al.

    Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells

    J. Biol. Chem.

    (2013)
  • A.L. Menke et al.

    The Wilms' tumor 1 gene: oncogene or tumor suppressor gene?

    Int. Rev. Cytol.

    (1998)
  • M. Mraz et al.

    Kipps, miR-150 influences B-cell receptor signaling in chronic lymphocytic leukemia by regulating expression of GAB1 and FOXP1

    Blood

    (2014)
  • K.S. Reiners et al.

    Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity

    Blood

    (2013)
  • E. Jaworski et al.

    Human T-lymphotropic virus type 1-infected cells secrete exosomes that contain tax protein

    J. Biol. Chem.

    (2014)
  • R. Koch et al.

    Populational equilibrium through exosome-mediated Wnt signaling in tumor progression of diffuse large B-cell lymphoma

    Blood

    (2014)
  • C. Bouvy et al.

    Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their microRNA cargo

    Leuk. Res.

    (2017)
  • P.J. Quesenberry et al.

    Role of extracellular RNA-carrying vesicles in cell differentiation and reprogramming

    Stem Cell Res. Therapy

    (2015)
  • A. Janowska-Wieczorek et al.

    Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer

    Int. J. Cancer

    (2005)
  • P.D. Robbins et al.

    Regulation of immune responses by extracellular vesicles

    Nat. Rev. Immunol.

    (2014)
  • J. Ratajczak et al.

    Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery

    Leukemia

    (2006)
  • C. Friend et al.

    Observations on cell lines derived from a patient with Hodgkin’s disease

    Cancer Res.

    (1978)
  • W.J. van Blitterswijk et al.

    Rigid plasma-membrane-derived vesicles, enriched in tumour-associated surface antigens (MLr), occurring in the ascites fluid of a murine leukaemia (GRSL)

    Int. J. Cancer

    (1979)
  • C.W. Kim et al.

    Extracellular membrane vesicles from tumor cells promote angiogenesis via sphingomyelin

    Cancer Res.

    (2002)
  • M.J. Szczepanski et al.

    Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1

    Haematologica

    (2011)
  • A. Caivano et al.

    High serum levels of extracellular vesicles expressing malignancy-related markers are released in patients with various types of hematological neoplastic disorders

    Tumour Biol.

    (2015)
  • A. Caivano et al.

    MicroRNA-155 in serum-derived extracellular vesicles as a potential biomarker for hematologic malignancies—a short report

    Cellular oncology (Dordrecht)

    (2017)
  • G. Raposo et al.

    Extracellular vesicles: exosomes, microvesicles, and friends

    J. Cell Biol.

    (2013)
  • D. Di Vizio et al.

    Oncosome formation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease

    Cancer Res.

    (2009)
  • K. Denzer et al.

    Exosome: from internal vesicle of the multivesicular body to intercellular signaling device

    J. Cell Sci.

    (2000)
  • C. Raiborg et al.

    The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins

    Nature

    (2009)
  • N.M. Mahaweni et al.

    Tumour-derived exosomes as antigen delivery carriers in dendritic cell-based immunotherapy for malignant mesothelioma

    J. Extracell Vesicles

    (2013)
  • P. Li et al.

    Progress in exosome isolation techniques

    Theranostics

    (2017)
  • C.S. Hong et al.

    Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer

    J. Extracell Vesicles

    (2016)
  • J. Lötvall et al.

    Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles

    J. Extracell Vesicles

    (2014)
  • Y.H. Soung et al.

    Exosomes in cancer diagnostics

    Cancers

    (2017)
  • H. Peinado et al.

    Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET

    Nat. Med.

    (2012)
  • C.S. Hong et al.

    Plasma exosomes as markers of therapeutic response in patients with acute myeloid leukemia

    Front. Immunol.

    (2014)
  • J. Huan et al.

    RNA trafficking by acute myelogenous leukemia exosomes

    Cancer Res.

    (2013)
  • J. Boysen et al.

    Dynamics of microvesicle generation in B-cell chronic lymphocytic leukemia: implication in disease progression

    Leukemia

    (2017)
  • L. Belov et al.

    Extensive surface protein profiles of extracellular vesicles from cancer cells may provide diagnostic signatures from blood samples

    J. Extracell Vesicles

    (2016)
  • L. De Luca et al.

    Characterization and prognostic relevance of circulating microvesicles in chronic lymphocytic leukemia

    Leukemia Lymphoma

    (2017)
  • K.S. Reiners et al.

    Extracellular vesicles released from chronic lymphocytic leukemia cells exhibit a disease relevant mRNA signature and transfer mRNA to bystander cells

    Haematologica Italy

    (2017)
  • S.M. Johnson et al.

    Acute lymphoblastic leukaemia cells produce large extracellular vesicles containing organelles and an active cytoskeleton

    J. Extracell Vesicles

    (2017)
  • T.L. Whiteside

    Extracellular vesicles isolation and their biomarker potential: are we ready for testing?

    Ann. Transl. Med. China

    (2017)
  • G. Marcucci et al.

    Clinical role of microRNAs in cytogenetically normal acute myeloid leukemia: miR-155 upregulation independently identifies high-risk patients

    J. Clin. Oncol.

    (2013)
  • Cited by (34)

    • Tumor Derived Extracellular Vesicles Modulate Gene Expression in T cells

      2023, Gene
      Citation Excerpt :

      Initially proposed to be a mechanism of discarding cellular waste, EVs have emerged as a significant instrument of intercellular communication (Admyre et al., 2006; Ali et al., 2010; Meckes and Raab-Traub, 2011). EVs have been associated with physiological and pathological circumstances, and their composition, amount, and function are cell type and disease dependent (Pando et al., 2018b). Tumor cells secrete EVs in higher quantities then normal cells and have been associated with immunomodulation, microenvironment restructuring, anti-therapeutic evasion, and more (Pando et al., 2018b; Pando et al., 2021).

    • AML-derived extracellular vesicles negatively regulate stem cell pool size: A step toward bone marrow failure

      2023, Current Research in Translational Medicine
      Citation Excerpt :

      Emerging evidence suggests that leukemic clones co-opt and modify the normal hematopoietic niche into a permissive milieu for leukemia growth, commonly referred to as tumor microenvironment (TME) [3,4]. In fact, cancer cells are just parts of a complex network made up not only of leukemic blasts but also of surrounding stromal cells, which provide favorable support to ensure the life-sustaining benefits as well as drug resistance of resident cells [5]. However, the precise molecular mechanism governing the induction of abnormalities in the unique cellular interactions of the hematopoietic microenvironment and its functional significance has remained unclear.

    • Extracellular vesicles (EVs): What we know of the mesmerizing roles of these tiny vesicles in hematological malignancies?

      2021, Life Sciences
      Citation Excerpt :

      Interestingly, CLL patients-derived EVs containing phosphorylated RTK could activate AKT pathway in BMSCs, and lead to the production of VEGF and expression of cyclin D1 and c-Myc which are necessary for the survival and progression of B-CLL [223–225]. Also, it has been demonstrated that EVs-mediated activation of AKT regulates β-catenin pathway and increases the expression of cyclin D1 and c-Myc in BM-MSCs, providing a tumor microenvironment that helps the progression of the disease [174,204,215,224]. Previous studies also reported that CLL EVs may reprogram stromal cells to support leukemic cell survival.

    • High-throughput analysis and functional interpretation of extracellular vesicle content in hematological malignancies

      2020, Computational and Structural Biotechnology Journal
      Citation Excerpt :

      Altogether, these data suggest that EVs are markedly implicated in the cross-talk between CLL cells and their microenvironment; in particular, EV protein and miR content seems to play an essential role in promoting tumor survival, proliferation, and eventually progression of CLL (Fig. 1). Acute leukemia-derived EVs contain a variety of non-coding RNAs supporting leukemogenesis and influencing the outcome and response to therapy through the regulation of several genes involved in the pathogenesis of this disease [56]. In addition, several studies have highlighted the promising role of circulating miRs as biomarkers in acute myeloid leukemia (AML) (Table 1) [4,34,74,82,85].

    View all citing articles on Scopus
    View full text