Acessibilidade / Reportar erro

Extracellular vesicles as modulators of monocyte and macrophage function in tumors

Abstract

The tumor microenvironment (TME) harbors several cell types, such as tumor cells, immune cells, and non-immune cells. These cells communicate through several mechanisms, such as cell-cell contact, cytokines, chemokines, and extracellular vesicles (EVs). Tumor-derived vesicles are known to have the ability to modulate the immune response. Monocytes are a subset of circulating innate immune cells and play a crucial role in immune surveillance, being recruited to tissues where they differentiate into macrophages. In the context of tumors, it has been observed that tumor cells can attract monocytes to the TME and induce their differentiation into tumor-associated macrophages with a pro-tumor phenotype. Tumor–derived EVs have emerged as essential structures mediating this process. Through the transfer of specific molecules and signaling factors, tumor-derived EVs can shape the phenotype and function of monocytes, inducing the expression of cytokines and molecules by these cells, thus modulating the TME towards an immunosuppressive environment.

Key words
Monocytes; extracellular vesicles; immunophenotype; immunomodulation; tumor-associated macrophages

INTRODUCTION

The TME is a complex system that harbors cancer cells, stromal cells, immune cells, and extracellular components. During tumor development, leukocyte recruitment, angiogenesis, and tissue remodeling occur (Baghban et al. 2020BAGHBAN R, ROSHANGAR L, JAHANBAN-ESFAHLAN R, SEIDI K, EBRAHIMI-KALAN A, JAYMAND M, KOLAHIAN S, JAVAHERI T ZARE P. 2020. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal 18: 59.). Among the cells participating in TME, one may highlight monocytes, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs). These cells interact with cancer cells, leading to the release of extracellular matrix (ECM), matrix metalloproteinase (MMPs), chemokines, EVs, and growth factors, which sustain the microenvironment and contribute to cancer progression (Ren et al. 2018REN B, CUI M, YANG G, WANG H, FENG M, YOU L ZHAO Y. 2018. Tumor microenvironment participates in metastasis of pancreatic cancer. Mol Cancer 17: 108.). Monocytes are cells of the innate immune system belonging to the mononuclear phagocytic system with high plasticity (van Furth & Cohn 1968VAN FURTH R COHN ZA. 1968. The origin and kinetics of mononuclear phagocytes. J Exp Med 128: 415-435.), participating in various mechanisms such as immunotolerance, angiogenesis, and the establishment of TAMs (Ugel et al. 2021UGEL S, CANEGRAVE S, DE SANCTIS F BRONTE V. 2021. Monocytes in the Tumor Microenvironment. Annu Rev Pathol 16: 93-122., Ding et al. 2016DING J, GUO C, HU P, CHEN J, LIU Q, WU X, CAO Y WU J. 2016. CSF1 is involved in breast cancer progression through inducing monocyte differentiation and homing. Int J Oncol 49: 2064-2074.). TAMs are the most abundant immune population in TME, whose density is related to a worse prognosis in different types of cancer (Medrek et al. 2012MEDREK C, PONTÉN F, JIRSTRÖM K LEANDERSSON K. 2012. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer 12: 1-9., Fan et al. 2014FAN QM ET AL. 2014. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett 352: 160-168., Fridman et al. 2017FRIDMAN WH, ZITVOGEL L, SAUTÈS–FRIDMAN C KROEMER G. 2017. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 14: 717-734.). The ability of monocytes and macrophages to mediate interactions between immune and tumor cells is critical for tumor progression. In addition, monocytes and TAMs can also uptake EVs, influencing cellular response and tumor progression(Nielsen & Schmid 2017NIELSEN SR SCHMID MC. 2017. Macrophages as Key Drivers of Cancer Progression and Metastasis. Mediators Inflamm 2017: 1-11.). Therefore, the presence of monocytes and TAMs in the TME is an essential indication for tumor progression and can be used as a prognostic factor.

Abbreviations

BC Breast cancer

ECM Extracellular matrix

EMT Epithelial-mesenchymal transition

ESCRT Endosomal sorting complexes required for transport

EVs Extracellular vesicles

GC Gastric cancer

HCC Hepatocellular carcinoma

HSPs Heat shock proteins

ILVs Intraluminal vesicles

lncRNAs Long non-coding RNAs

lVEs Large vesicles

MDSCs Myeloid-derived suppressor cells

MMPs Matrix metalloproteinase

MVB Multivesicular bodies

NBL Neuroblastoma

ncRNAs Non-coding RNAs

PC Pancreatic cancer

scRNA-Seq Single-cell RNA sequencing

sVEs Small vesicles

TAMs Tumor-associated macrophages

TEMs Tie2-expressing monocytes

TME Tumor microenvironment

TNBC Triple-negative breast cancer

Phenotypes of monocytes and macrophage

Two monocyte populations were initially described in humans according to their surface markers, CD14 and CD16, described as receptors for LPS and Fcγ-III, respectively(Geissmann et al. 2003GEISSMANN F, JUNG S LITTMAN DR. 2003. Blood Monocytes Consist of Two Principal Subsets with Distinct Migratory Properties. Immunity 19: 71-82.). This statement was revised, and three subsets of monocytes are currently described. The classical CD14++CD16- monocytes’ subset express high levels of CCR2 and migrate to tissues, where they differentiate into macrophages or dendritic cells. This subset comprises approximately 80-90% of the total monocyte population. The CD14+CD16+ intermediate subset, accounting for about 5% of monocytes, exhibits a strong TNF-α response when stimulated with LPS and expresses high levels of CCR5 (Hijdra et al. 2013HIJDRA D, VORSELAARS ADM, GRUTTERS JC, CLAESSEN AME RIJKERS GT. 2013. Phenotypic Characterization of Human Intermediate Monocytes. Front Immunol 4: 339.). The non-classical CD14dimCD16+ subset expresses CX3CR1 (Geissmann et al. 2003GEISSMANN F, JUNG S LITTMAN DR. 2003. Blood Monocytes Consist of Two Principal Subsets with Distinct Migratory Properties. Immunity 19: 71-82.) and patrols the vascular endothelium, resolving inflammation (Devêvre et al. 2015DEVÊVRE EF, RENOVATO-MARTINS M, CLÉMENT K, SAUTÈS-FRIDMAN C, CREMER I POITOU C. 2015. Subpopulations in Human Obesity Profiling of the Three Circulating Monocyte. J Immunol 194: 3917-3923., Kapellos et al. 2019KAPELLOS TS, BONAGURO L, GEMÜND I, REUSCH N, SAGLAM A, HINKLEY ER SCHULTZE JL. 2019. Human Monocyte Subsets and Phenotypes in Major Chronic Inflammatory Diseases. Front Immunol 10: 2035.). This subset represents approximately 7% of monocyte subtypes. Additionally, Tie2-expressing monocytes (TEMs), a distinct CD14+CD16+ subpopulation, constitute around 2% of circulating monocytes (De Palma et al. 2005DE PALMA M, VENNERI MA, GALLI R, SERGI LS, POLITI LS, SAMPAOLESI M NALDINI L. 2005. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 8: 211-226.). TEMs do not express CCR2, suggesting that different mechanisms govern their recruitment than monocytes dependent on CCL2. This subpopulation is associated with angiogenesis induction and tumor growth (Lewis et al. 2007LEWIS CE, DE PALMA M NALDINI L. 2007. Tie2-expressing monocytes and tumor angiogenesis: Regulation by hypoxia and angiopoietin-2. Cancer Res 67: 8429-8432., Murdoch et al. 2007MURDOCH C, TAZZYMAN S, WEBSTER S LEWIS CE. 2007. Expression of Tie-2 by Human Monocytes and Their Responses to Angiopoietin-2. J Immunol 178: 7405-7411.). In mice, peripheral blood monocytes can be divided into classic Ly6Chigh/+CD43+ and non-classical patrolling monocytes Ly6Clow/-CD43high/+ (Ziegler-Heitbrock et al. 2010ZIEGLER-HEITBROCK L ET AL. 2010. Nomenclature of monocytes and dendritic cells in blood. Blood 116: e74-e80.). It has been shown that at steady state, LyC6high/+ monocytes differentiate into Ly6Clow/- monocytes, responsible for patrolling the endothelium of small blood vessels by binding to it through CX3CR1 receptor depending on LAF-1/ICAM1(Yang et al. 2014YANG J, ZHANG L, YU C, YANG X-F WANG H. 2014. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res 2: 1.). Through fate mapping, Patel et al. (2017)PATEL AA ET AL. 2017. The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J Exp Med 214: 1913-1923. demonstrated that human classical monocytes grafted onto humanized mice differentiate into intermediate monocytes after 24 hours and non-classical monocytes after 96 hours (Patel et al. 2017PATEL AA ET AL. 2017. The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J Exp Med 214: 1913-1923.). Classic murine monocytes recruited by CCL2 have been linked to a role in promoting breast cancer metastasis, while non-classical monocytes trigger NK cell anti-metastatic activity after detecting circulating metastatic cells and eliminating their debris within blood vessels(Qian et al. 2011QIAN BZ, LI J, ZHANG H, KITAMURA T, ZHANG J, CAMPION LR, KAISER EA, SNYDER LA POLLARD JW. 2011a. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nat 475: 222-225., Hanna et al. 2015HANNA RN ET AL. 2015. Patrolling monocytes control tumor metastasis to the lung. Science 350: 985-990.).

Macrophages are traditionally divided into two activation profiles: the classically activated, pro-inflammatory M1 - with markers such as HLA-DR, CD80/CD86 - and the alternatively activated, anti-inflammatory type M2 -with features such as CD206 and CD163(Zhou et al. 2020ZHOU J, TANG Z, GAO S, LI C, FENG Y ZHOU X. 2020. Tumor-Associated Macrophages: Recent Insights and Therapies. Front Oncol 10: 188.). Evidence from the literature suggests that TME is enriched with TAMs resembling the M2 macrophage profile, responsible for releasing chemokines such as CCL2, capable of recruiting more monocytes for TME (Mantovani et al. 2010MANTOVANI A, SAVINO B, LOCATI M, ZAMMATARO L, ALLAVENA P BONECCHI R. 2010. The chemokine system in cancer biology and therapy. Cytokine Growth Factor Rev 21: 27-39.). This crosstalk is essential for maintaining this cell type and supporting tumor progression, thus promoting tumor growth, invasion, metastasis, and drug resistance (DeNardo & Ruffell 2019DENARDO DG RUFFELL B. 2019. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol 19: 369-382.). Myeloid-derived suppressor cells (MDSCs), related to TAMs, were the first cell population described in mice to inhibit the host immune response (Gabrilovich et al. 1998GABRILOVICH D, ISHIDA T, OYAMA T, RAN S, KRAVTSOV V, NADAF S CARBONE DP. 1998. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood 92: 4150-4166.). In mice, they can be classified into two subpopulations: polymorphonuclear MDSCs (PMN-MDSCs) with CD11b+Ly6GhiLy6C−/low and monocyte MDSCs (M-MDSCs) with CD11b+Ly6G−/lowLy6Chi (Bronte et al. 2016BRONTE V ET AL. 2016. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 7: 12150.). In humans, M-MDSCs are defined as CD14+CD33+CD11b+ HLA-DR− (Hegde et al. 2021HEGDE S, LEADER AM MERAD M. 2021. MDSC: Markers, development, states, and unaddressed complexity. Immunity 54: 875-884.). MDSCs play a crucial role in promoting tumor immune evasion. They consume amino acids necessary for T cell proliferation, leading to cell cycle arrest and decreased TCR expression. MDSCs also limit cysteine uptake by T cells, restricting their activation (Srivastava et al. 2010SRIVASTAVA MK, SINHA P, CLEMENTS VK, RODRIGUEZ P OSTRAND-ROSENBERG S. 2010. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 70: 68-77.). Additionally, overexpression of indoleamine 2,3-dioxygenase (IDO) within the tumor microenvironment supports MDSCs and contributes to immunosuppression(Prendergast et al. 2018PRENDERGAST GC, MALACHOWSKI WJ, MONDAL A, SCHERLE P MULLER AJ. 2018. Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer. In: Int Rev Cell Mol Biol 336: 175-203.).

Single-cell gene expression analysis showed that CD14++CD16- monocytes expressed more genes associated with phagocytosis and migration functions. In contrast, intermediate monocytes were related to antigen presentation, co-stimulation, and activation of NK and CD8+ T cells(Gren et al. 2015GREN ST, RASMUSSEN TB, JANCIAUSKIENE S, HAKANSSON K, GERWIEN JG GRIP O. 2015. A Single-Cell Gene-Expression Profile Reveals Inter-Cellular Heterogeneity within Human Monocyte Subsets. PLoS ONE 10: e0144351., Kapellos et al. 2019KAPELLOS TS, BONAGURO L, GEMÜND I, REUSCH N, SAGLAM A, HINKLEY ER SCHULTZE JL. 2019. Human Monocyte Subsets and Phenotypes in Major Chronic Inflammatory Diseases. Front Immunol 10: 2035.). In a single-cell RNA sequencing (scRNA-Seq) study conducted by Villani et al. (2017)VILLANI AC ET AL. 2017. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes and progenitors. Science 356: eaah4573., 372 monocytes were profiled, demonstrating four distinct clusters. The two largest groups represented classical and non-classical monocytes. In contrast, the remaining two represented monocytes derived from the intermediate subtype, indicating that this population may be less homogeneous, thus revealing potential new monocyte subtypes(Villani et al. 2017VILLANI AC ET AL. 2017. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes and progenitors. Science 356: eaah4573.). On the other hand, Zilionis et al. (2019)ZILIONIS R ET AL. 2019. Single cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50: 1317. identified only three monocyte gene signatures conserved between human and mouse species(Zilionis et al. 2019ZILIONIS R ET AL. 2019. Single cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50: 1317.). A study analyzing 175 immune cells from 11 patients with breast cancer identified a macrophage subset exhibiting immunosuppressive characteristics, presenting an M2 phenotype (Chung et al. 2017CHUNG W ET AL. 2017. Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer. Nat Commun 8: 15081.). However, scRNAseq studies have shown that the behavior of TAMs does not correspond to the classic M1/M2 polarization axis. Wagner et al. (2019)WAGNER J ET AL. 2019. A Single-Cell Atlas of the Tumor and Immune Ecosystem of Human Breast Cancer. Cell 177: 1330-1345. identified 19 myeloid clusters within the TME of breast cancer through scRNAseq analysis. Of these, two groups of CD14++CD16- monocytes, two groups of CD14intCD16+ monocytes, four groups of migrant macrophages, four groups of resident macrophages, and six groups of TAMs were identified, indicating heterogeneity between the macrophage populations present in this cancer(Wagner et al. 2019WAGNER J ET AL. 2019. A Single-Cell Atlas of the Tumor and Immune Ecosystem of Human Breast Cancer. Cell 177: 1330-1345.). In another study, it was determined that all three clusters of TAMs identified in scRNAseq were among the monocytic clusters that displayed the highest expression of the M2 canonical signature but also showed increased expression of the M1 gene signature. Hence, these single-cell studies have demonstrated divergences between establishing the M1/M2 axis to classify TAMs.

In the TME, monocytes, TAMs, and MDSCs are present in varying proportions depending on the type of carcinoma and degree of progression (Davidov et al. 2020DAVIDOV V, JENSEN G, MAI S, CHEN SH PAN PY. 2020. Analyzing One Cell at a TIME: Analysis of Myeloid Cell Contributions in the Tumor Immune Microenvironment. Front Immunol 11: 1842.). These studies have highlighted the importance of identifying and accurately characterizing the monocyte and macrophage populations present within TME since each subpopulation can have distinct functions and behaviors, thus impacting tumor progression differently(Larionova et al. 2020LARIONOVA I, TUGUZBAEVA G, PONOMARYOVA A, STAKHEYEVA M, CHERDYNTSEVA N, PAVLOV V, CHOINZONOV E KZHYSHKOWSKA J. 2020. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front Oncol 10: 566511., Mantovani et al. 2017MANTOVANI A, MARCHESI F, MALESCI A, LAGHI L ALLAVENA P. 2017. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 14: 399-416., Zhu et al. 2017ZHU Y ET AL. 2017. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 47: 323-338.).

Extracellular vesicles

Extracellular vesicles (EVs) are nanostructures delimited by a lipid bilayer known to deliver functional mRNAs, non-coding RNAs (ncRNAs) such as microRNAs (miRNAs), and long non-coding RNAs (lncRNAs); proteins, receptors, and growth factors from diverse cell types to target cells participating in cell signaling by mediating intercellular communication (van Niel et al. 2018VAN NIEL G, D’ANGELO G RAPOSO G. 2018. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 19: 213-228., Pathan et al. 2019PATHAN M, FONSEKA P, CHITTI SV, KANG T, SANWLANI R, VAN DEUN J, HENDRIX A MATHIVANAN S. 2019. Vesiclepedia 2019: A compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res 47: D516-D519.). EVs are heterogeneous regarding their biogenesis, size, and content (Cocucci & Meldolesi 2015COCUCCI E MELDOLESI J. 2015. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol 25: 364-372.). Large vesicles (lVEs) are formed through plasma membrane budding and subsequent fission. They are commonly referred to in the literature as microvesicles, ectosomes, microparticles, and exovesicles (Théry et al. 2009THÉRY C, OSTROWSKI M SEGURA E. 2009. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 9: 581-593.). They were firstly described as derived from the membrane of platelets (Heijnen et al. 1999HEIJNEN HFG, SCHIEL AE, FIJNHEER R, GEUZE HJ SIXMA JJ. 1999. Activated platelets release two types of membrane vesicles: Microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and α-granules. Blood 94: 3791-3799.), tumors (Al-Nedawi et al. 2008AL-NEDAWI K, MEEHAN B, MICALLEF J, LHOTAK V, MAY L, GUHA A RAK J. 2008. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol 10: 619-624.), neutrophils (Hess et al. 1999HESS C, SADALLAH S, HEFTI A, LANDMANN R SCHIFFERLI J-A. 1999. Ectosomes Released by Human Neutrophils Are Specialized Functional Units. J Immunol 163: 4564-4573.), and dendritic cells (Obregon et al. 2006OBREGON C, ROTHEN-RUTISHAUSER B, GITAHI SK, GEHR P NICOD LP. 2006. Exovesicles from human activated dendritic cells fuse with resting dendritic cells, allowing them to present alloantigens. Am J Pathol 169: 2127-2136.). On the other hand, small vesicles (sVEs) are secreted after the fusion of the internal compartments containing intraluminal vesicles (ILVs) with the plasma membrane. They are called exosomes, nanoparticles, or vesicle-type exosomes (Théry et al. 2009THÉRY C, OSTROWSKI M SEGURA E. 2009. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 9: 581-593.). The release of tumor-derived EVs occurs both in vitro and in vivo and is linked to various processes such as proliferation, activation, apoptosis, and migration of tumor cells (Sung et al. 2021SUNG BH, PARENT CA WEAVER AM. 2021. Extracellular vesicles: Critical players during cell migration. Dev Cell 56: 1861-1874., Phetfong et al. 2022PHETFONG J, TAWONSAWATRUK T, KAMPROM W, ONTONG P, TANYONG D, BORWORNPINYO S SUPOKAWEJ A. 2022. Bone marrow-mesenchymal stem cell-derived extracellular vesicles affect proliferation and apoptosis of leukemia cells in vitro. FEBS Open Bio 12: 470-479., Andreola et al. 2002ANDREOLA G ET AL. 2002. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J Exp Med 195 1303-1316.). This release is also associated with tumor progression, where highly metastatic tumors release more EVS than those with low metastatic ability (Friedl et al. 1997FRIEDL P, MAASER K, KLEIN CE, NIGGEMANN B, KROHNE G ZÄNKER KS. 1997. Migration of highly aggressive MV3 melanoma cells in 3-dimensional collagen lattices results in local matrix reorganization and shedding of α2 and β1 integrins and CD44. Cancer Res 57: 2061-2070., Poste & Nicolson 1980, Kalluri 2016KALLURI R. 2016. The biology and function of exosomes in cancer. J Clin Invest 126: 1208-1215.).

Large vesicles

The lVEs are produced through plasma membrane protrusion followed by fission. This process requires rearrangements of membrane lipids and is initiated by increasing intracellular calcium levels and activating calpain, the protease responsible for separating membrane proteins from the cytoskeleton (Pasquet et al. 1996PASQUET JM, DACHARY-PRIGENT J NURDEN AT. 1996. Calcium influx is a determining factor of calpain activation and microparticle formation in platelets. Eur J Biochem 239: 647-654.). Consequently, there is a remodeling of the actin filaments, allowing the occurrence of bubbles, with the release of vesicles through microdomains of the plasma membrane known as lipid rafts (Ståhl et al. 2019STÅHL A, JOHANSSON K, MOSSBERG M, KAHN R KARPMAN D. 2019. Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases. Pediatr Nephrol 34: 11-30., Del Conde et al. 2005DEL CONDE I, SHRIMPTON CN, THIAGARAJAN P LÓPEZ JA. 2005. Tissue-factor-bearing microvesicles arise from lipid rafts and fuse with activated platelets to initiate coagulation. Blood 106: 1604-1611.). Muralidharan-Chari et al. (2009)MURALIDHARAN-CHARI V, CLANCY J, PLOU C, ROMAO M, CHAVRIER P, RAPOSO G D’SOUZA-SCHOREY C. 2009. ARF6-Regulated Shedding of Tumor Cell-Derived Plasma Membrane Microvesicles. Curr Biol 19: 1875-1885. demonstrated the importance of the ARF6 protein in the release of microvesicles once the endosomal complex regulated by ARF6 is crucial in the selective incorporation of molecular charge in these structures(D’Souza-Schorey & Chavrier 2006D’SOUZA-SCHOREY C CHAVRIER P. 2006. ARF proteins: roles in membrane traffic and beyond. Nat Rev Mol Cell Biol 7: 347-358., Muralidharan-Chari et al. 2009MURALIDHARAN-CHARI V, CLANCY J, PLOU C, ROMAO M, CHAVRIER P, RAPOSO G D’SOUZA-SCHOREY C. 2009. ARF6-Regulated Shedding of Tumor Cell-Derived Plasma Membrane Microvesicles. Curr Biol 19: 1875-1885.). ARF6 targets include ERK and MLCK, important regulators of actin polymerization and myosin activity, both essential for releasing microvesicles (Minciacchi et al. 2015MINCIACCHI VR, FREEMAN MR DI VIZIO D. 2015. Extracellular Vesicles in Cancer: Exosomes, Microvesicles and the Emerging Role of Large Oncosomes. Semin Cell Dev Biol 40: 41-51., Muralidharan-Chari et al. 2009MURALIDHARAN-CHARI V, CLANCY J, PLOU C, ROMAO M, CHAVRIER P, RAPOSO G D’SOUZA-SCHOREY C. 2009. ARF6-Regulated Shedding of Tumor Cell-Derived Plasma Membrane Microvesicles. Curr Biol 19: 1875-1885.).

Small vesicles

The characterization and study of exosome origin are widely discussed in the literature. The process of exosome formation begins with the invagination of the plasma membrane to form endosomes, which are intracellular compartments involved in the screening and degradation of internalized molecules of the extracellular environment. As the endosomes mature, they fuse to form multivesicular bodies (MVB), intermediates of the endosomal system. The formation of exosomes requires the action of endosomal sorting complexes required for transport (ESCRT), which are composed of four proteins that work together to promote the formation of MVB (James 2015JAMES HH. 2015. ESCRTs are everywhere. Embo J 34: 2398-2407.). The mechanism of ESCRT is initiated through the recognition and sequestration of ubiquitinated proteins for specific endosomal domains. After cleavage of the ILVs, the ESCRT-III complex separates from the MVB membrane (Zhang et al. 2019ZHANG Y, LIU Y, LIU H TANG WH. 2019. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci 9: 19.), resulting in the secretion of exosomes through the fusion of these with the plasma membrane.

Exosomes were identified in the 1980s and were initially considered residues resulting from cell damage (Johnstone et al. 1987JOHNSTONE RM, ADAM M, HAMMOND JR, ORR L TURBIDE C. 1987. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem 262: 9412-9420.). Later, the importance of these for intercellular communication in different cellular processes was observed. They participate in various critical biological processes, including immunomodulation, inflammation, angiogenesis, and tumor progression (Kurywchak et al. 2018KURYWCHAK P, TAVORMINA J KALLURI R. 2018. The emerging roles of exosomes in the modulation of immune responses in cancer. Genome Med 10: 23., Song et al. 2018SONG W, YAN D, WEI T, LIU Q, ZHOU X LIU J. 2018. Tumor-derived extracellular vesicles in angiogenesis. Biomed Pharmacother 102: 1203-1208.). They have been studied as potential biomarkers in neurodegenerative diseases, cardiovascular diseases, and cancer, as they may contain valuable information about cell function (Raposo & Stoorvogel 2013RAPOSO G STOORVOGEL W. 2013. Extracellular vesicles: Exosomes, microvesicles, and friends. J Cell Biol 200: 373-383.). Exosomes have a distinct structure, including proteins, lipids, and RNAs of cellular origin, making them a potential source for biomarker identification and disease therapies.

EVs cargo

Although lVEs and sVEs are structurally similar, they differ in composition. The membrane of the lEVs is enriched in cholesterol and diacylglycerol, exposed phosphatidylserine, and lipid rafts, with membrane-derived receptors, cytokines, chemokines, and lipids (Ståhl et al. 2019STÅHL A, JOHANSSON K, MOSSBERG M, KAHN R KARPMAN D. 2019. Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases. Pediatr Nephrol 34: 11-30., Camussi et al. 2010CAMUSSI G, DEREGIBUS MC, BRUNO S, CANTALUPPI V BIANCONE L. 2010. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int 78: 838-848.). Dolo et al. (1998)DOLO V, GINESTRA A, CASSARÀ D, VIOLINI S, LUCANIA G, TORRISI MR, NAGASE H, CANEVARI S, PAVAN A VITTORELLI ML. 1998. Selective localization of matrix metalloproteinase 9, β1 integrins, and human lymphocyte antigen class I molecules on membrane vesicles shed by 8701-BC breast carcinoma cells. Cancer Res 58: 4468-4474. observed that microvesicles released by breast cancer cells contained proteases such as MMP-2 and MMP-9, and β1 integrin, indicating the importance of EVs release in extracellular matrix proteolysis during cell migration (Dolo et al. 1998DOLO V, GINESTRA A, CASSARÀ D, VIOLINI S, LUCANIA G, TORRISI MR, NAGASE H, CANEVARI S, PAVAN A VITTORELLI ML. 1998. Selective localization of matrix metalloproteinase 9, β1 integrins, and human lymphocyte antigen class I molecules on membrane vesicles shed by 8701-BC breast carcinoma cells. Cancer Res 58: 4468-4474.). Lischnig et al. (2022)LISCHNIG A, BERGQVIST M, OCHIYA T LÄSSER C. 2022. Quantitative Proteomics Identifies Proteins Enriched in Large and Small Extracellular Vesicles. Mol Cell Proteomics 21. identified that the primary biological functions of lVEs are related to translation initiation, cell-to-cell adhesion, and mitochondrial electron transport, while the main functions of sVEs are associated with the reorganization of the extracellular matrix, cell adhesion, endosomal transport, and nuclear organization. Additionally, lVEs are enriched in ribosomal, mitochondrial, and nuclear proteins and proteins involved in cytokinesis (Lischnig et al. 2022LISCHNIG A, BERGQVIST M, OCHIYA T LÄSSER C. 2022. Quantitative Proteomics Identifies Proteins Enriched in Large and Small Extracellular Vesicles. Mol Cell Proteomics 21.). On the other hand, sVEs are enriched in tetraspanins, ADAMs, and ESCRT-III complex proteins, as well as SNAREs and endosome-associated Rab proteins. Exosomes are rich in proteins such as tetraspanins CD9, CD63, CD81, heat shock proteins (HSP70, HSP90), and MVB-forming proteins that are involved in the release of exosomes such as Alix and TSG101. Furthermore, they have abundant miRNA, lncRNA, and circRNAs (Zhang et al. 2019ZHANG Y, LIU Y, LIU H TANG WH. 2019. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci 9: 19.). Additionally, the content of lVEs can be a source of information for the cellular microenvironment, acting as signals of cellular damage, inflammation, and other processes necessary for a cellular response. The lVEs are also considered essential for cancer progression since they may contain molecules that promote angiogenesis and inhibition of the immune response, among other effects (Loyer et al. 2014LOYER X, VION A-C, TEDGUI A BOULANGER CM. 2014. Microvesicles as Cell–Cell Messengers in Cardiovascular Diseases. Circ Res 114: 345-353., Tricarico et al. 2017TRICARICO C, CLANCY J D’SOUZA-SCHOREY C. 2017. Biology and biogenesis of shed microvesicles. Small GTPases 8: 220-232.)

Extracellular vesicles within the tumor microenvironment: actions on monocyte and macrophage functions

The interaction of monocytes with the microenvironment is crucial for their migration, differentiation, and action in healthy individuals or under pathophysiological conditions. Cytokines, chemokines, and EVs play a pivotal role in this process. Classic monocytes produce TNFα, IL-6, and IL-1β in response to TLR2 and TLR4 receptor agonists, while the non-classical subtype secretes INF-α in response to intracellular TLR3, TLR7, and TLR8 (Boyette et al. 2017BOYETTE LB, MACEDO C, HADI K, ELINOFF BD, WALTERS JT, RAMASWAMI B, CHALASANI G, TABOAS JM, LAKKIS FG METES DM. 2017. Phenotype, function, and differentiation potential of human monocyte subsets. PLoS ONE 12: e0176460., Geissmann et al. 2003GEISSMANN F, JUNG S LITTMAN DR. 2003. Blood Monocytes Consist of Two Principal Subsets with Distinct Migratory Properties. Immunity 19: 71-82., Devêvre et al. 2015DEVÊVRE EF, RENOVATO-MARTINS M, CLÉMENT K, SAUTÈS-FRIDMAN C, CREMER I POITOU C. 2015. Subpopulations in Human Obesity Profiling of the Three Circulating Monocyte. J Immunol 194: 3917-3923.). IL-6 in TME skew monocyte differentiation into TAM with M2 characteristics, exhibiting CD14highCD163highCD80low phenotype (Duluc et al. 2007DULUC D ET AL. 2007. Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood 110: 4319-4330.). The migration of monocytes to TME is crucial for establishing TAMS depending on the CCR2/CCL2 axis (Kadomoto et al. 2021KADOMOTO S, IZUMI K MIZOKAMI A. 2021. Roles of CCL2-CCR2 axis in the tumor microenvironment. Int J Mol Sci 22: 8530.). Macrophage infiltration into cancer tissues is positively correlated with increased expression of CCL2 (Mizutani et al. 2009MIZUTANI K, SUD S, MCGREGOR NA, MARTINOVSKI G, RICE BT, CRAIG MJ, VARSOS ZS, ROCA H PIENTA KJ. 2009. The chemokine CCL2 increases prostate tumor growth and bone metastasis through macrophage and osteoclast recruitment. Neoplasia 11: 1235-1242.). Additionally, CCL2 also has been linked to the recruitment of CCR2+ monocytes to facilitate metastasis in breast cancer (BC) (Qian et al. 2011QIAN BZ, LI J, ZHANG H, KITAMURA T, ZHANG J, CAMPION LR, KAISER EA, SNYDER LA POLLARD JW. 2011a. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nat 475: 222-225.).

Tumor-derived EVs induce the secretion of TNFα and IL-1β by CD14+ monocytes, activating the NF-kB pathway (Gärtner et al. 2018GÄRTNER K, BATTKE C, DÜNZKOFER J, HÜLS C, VON NEUBECK B, KELLNER M-K, FIESTAS E, FACKLER S, LANG S ZEIDLER R. 2018. Tumor-derived extracellular vesicles activate primary monocytes. Cancer Med 7: 2013-2020.). Popěna et al. (2018)POPĚNA I, ABOLS A, SAULITE L, PLEIKO K, ZANDBERGA E, JĚKABSONS K, ENDZELIŅŠ E, LLORENTE A, LINĚ A RIEKSTIŅA U. 2018. Effect of colorectal cancer-derived extracellular vesicles on the immunophenotype and cytokine secretion profile of monocytes and macrophages. Cell Commun Signal 16: 1-12. demonstrated that colorectal cancer-derived EVs can promote changes in the immunophenotype of monocytes and macrophages, increasing CD14+ in M0 and HLA-DR in M1 and M2 macrophages. In addition, the cytokine secretion profile of these cells was altered, resembling the M1 profile in monocytes (Popěna et al. 2018POPĚNA I, ABOLS A, SAULITE L, PLEIKO K, ZANDBERGA E, JĚKABSONS K, ENDZELIŅŠ E, LLORENTE A, LINĚ A RIEKSTIŅA U. 2018. Effect of colorectal cancer-derived extracellular vesicles on the immunophenotype and cytokine secretion profile of monocytes and macrophages. Cell Commun Signal 16: 1-12.). NcRNAs have been studied in cancer as promoters of invasion, metastasis, and initiation of epithelial-mesenchymal transition (EMT) (Beltran et al. 2008BELTRAN M, PUIG I, PEÑA C, GARCÍA JM, ÁLVAREZ AB, PEÑA R, BONILLA F DE HERREROS AG. 2008. A natural antisense transcript regulates Zeb2/Sip1 gene expression during Snail1-induced epithelial-mesenchymal transition. Genes Dev 22: 756-769., Xiu et al. 2019XIU B ET AL. 2019. LINC02273 drives breast cancer metastasis by epigenetically increasing AGR2 transcription. Mol Cancer 18: 1-20., Gupta et al. 2010GUPTA RA ET AL. 2010. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464: 1071-1076.). In a study analyzing scRNAseq data, colorectal cancer-derived EVs enriched with miR-21-5p and miR-200a promoted immune evasion, one of the hallmarks of cancer, through upregulation of PD-L1 in CD206+ TAMs, being associated with a worse prognosis (Yin et al. 2022YIN Y ET AL. 2022. Colorectal Cancer-Derived Small Extracellular Vesicles Promote Tumor Immune Evasion by Upregulating PD-L1 Expression in Tumor-Associated Macrophages. Adv Sci 9: 2102620.). PD-1 is an immunoinhibitory receptor highly expressed on tumor-specific T cells (Ahmadzadeh et al. 2009AHMADZADEH M, JOHNSON LA, HEEMSKERK B, WUNDERLICH JR, DUDLEY ME, WHITE DE ROSENBERG SA. 2009. Tumor antigen–specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114: 1537-1544.), which is critical in tumorigenesis. Tumor cells express PD-L1 to evade anti-tumor responses (Juneja et al. 2017JUNEJA VR, MCGUIRE KA, MANGUSO RT, LAFLEUR MW, COLLINS N, NICHOLAS HAINING W, FREEMAN GJ SHARPE AH. 2017. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med 214: 895-904.) since PD-1/PD-L1 interactions are implicated in T-cell exhaustion and reduced immune response (Veluswamy & Bruder 2018VELUSWAMY P BRUDER D. 2018. PD-1/PD-L1 pathway inhibition to restore effector functions in exhausted CD8+ T cells: Chances, limitations and potential risks. Transl Cancer Res 7.). Glioblastoma (GB)-derived EVs containing PD-L1 induce the formation of immunosuppressive non-classical monocytes that inhibit T cell proliferation in vitro (Hines et al. 2020HINES BT ET AL. 2020. The role of extracellular vesicles and PD-L1 in glioblastoma-mediated immunosuppressive monocyte induction. Neuro Oncol 22: 967-978.). Moreover, BC cells produce exosomes under endoplasmic reticulum (ER) stress, which contain miR-27a-3p, promoting immune escape by upregulating PD-L1 expression in macrophages by activating the PTIEN/AKT/PI3K pathway (Yao et al. 2020YAO X, TU Y, XU Y, GUO Y, YAO F ZHANG X. 2020. Endoplasmic reticulum stress-induced exosomal miR-27a-3p promotes immune escape in breast cancer via regulating PD-L1 expression in macrophages. J Cell Mol Med 24: 9560-9573.).

It has been shown that GB-derived EVs skew the differentiation of monocytes to M2 macrophages, which acquired characteristics that resemble tumor-supportive phenotype observed in patients. The macrophages presented a decrease in HLA-DR and an increased phagocytic capacity, with an increase in the secretion of IL-6, MCP-1, and VEGF. Glioma with a stem cell-like phenotype also presents a most pronounced modulation of the monocyte to macrophage differentiation (De Vrij et al. 2015DE VRIJ J ET AL. 2015. Glioblastoma-derived extracellular vesicles modify the phenotype of monocytic cells. Int J Cancer 137.). GB-derived exosomes significantly increased the expression of arginase-1, IL-10, and CD206 in macrophages. The reprogrammed macrophages arginase+ produce exosomes that promote glioblastoma progression(Azambuja et al. 2020AZAMBUJA JH, LUDWIG N, YERNENI SS, BRAGANHOL E WHITESIDE TL. 2020. Arginase-1+ exosomes from reprogrammed macrophages promote glioblastoma progression. Int J Mol Sci 21: 3990.). PD-L1 expression in gastric cancer (GC) is a decisive factor in evaluating prognosis (Wu et al. 2006WU C, ZHU Y, JIANG J, ZHAO J, ZHANG XG XU N. 2006. Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochem 108: 19-24.). A subset of tumor-associated macrophages, PD1+ macrophages, accumulate in advanced-stage GC and exhibit an M2-like surface profile, with a significant increase in CD206, IL-10, and CCL1 expression, which promotes disease progression. These macrophages have a robust immunosuppressive activity on CD8+ T cells and are associated with tumor progression and early recurrence in GC patients (Wang et al. 2018WANG F ET AL. 2018. Tumor-derived exosomes induce PD1+ macrophage population in human gastric cancer that promotes disease progression. Oncogenesis 7: 41.).

Effects of EVs on M-MDSC

Monocytes exposed to melanoma-derived EVs are converted to M-MDSC, presented decreased HLA-DR expression, and increased IL-6 and CCL2 transcription and secretion, exhibiting suppressive activity on activated T cells. Genome-wide transcriptional analysis revealed that regulation of these cells involves miRNA modulation and the upregulation of the CD274/PD-L1 gene. MDSC-miRs are enriched in the plasma of melanoma patients and correlated with resistance to immunotherapy (Huber et al. 2018HUBER V ET AL. 2018. Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J Clin Invest 128: 5505-5516.). In another study, EVs uptake by immature myeloid cells from murine melanoma leads to increased PD-L1 expression both in mRNA and protein levels, accompanied by upregulation of inflammatory and immunosuppressive mediators such as IL-1β, IL-6, IL-10, TNF-α, and COX-2. Melanoma-EV-mediated upregulation of PD-L1 involves TLR signaling through the HSP86/TLR4 axis (Fleming et al. 2019FLEMING V ET AL. 2019. Melanoma extracellular vesicles generate immunosuppressive myeloid cells by upregulating PD-L1 via TLR4 signaling. Cancer Res 79: 4715-4728.). Activation of TLR2 and -4 after EVs uptaken by macrophage cell lines can stimulate the production of cytokines in vitro (Chow et al. 2014CHOW A ET AL. 2014. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-κ B. Sci Rep 4: 5750., Bretz et al. 2013BRETZ NP ET AL. 2013. Body fluid exosomes promote secretion of inflammatory cytokines in monocytic cells via Toll-Like receptor signaling. J Biol Chem 288: 36691-36702.). Additionally, the BAL fluid obtained from wild-type mice with melanoma was enriched in macrophage compared to TLR4-deficient mice, indicating a possible involvement of TLR4 on macrophage migration(Lee et al. 2010LEE CH, WU CL SHIAU AL. 2010. Toll-like receptor 4 signaling promotes tumor growth. J Immunother 33: 73-82.). Furthermore, melanoma exosomes educate bone marrow (BM) progenitors, increasing the frequency of pro-angiogenic c-Kit+Tie2+ cells through MET, which enhances metastasis (Peinado et al. 2012PEINADO H ET AL. 2012. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 18: 883-891.). BC-derived exosomes containing miR-9 and miR-181a promote the development of early-stage MDSCs via inhibition of SOCS3 and PIAS3, respectively. High IL-6 expression is correlated with SOCS3 deficiency-dependent hyperactivation of the JAK/STAT signaling pathway in these cells (Jiang et al. 2020).

Heat shock proteins (HSPs) are found to be highly expressed in human cancers and play a significant role in the proliferation, differentiation, and immune recognition of tumor cells (Ciocca & Calderwood 2005CIOCCA DR CALDERWOOD SK. 2005. Heat shock proteins in cancer: Diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones 10: 86.). Tumor-derived exosome-associated HSP72 determines the suppressive activity of the mouse and human MDSCs via activation of STAT3. The TLR2/MyD88 and the STAT3 pathways play a role in MDSC activation by triggering the production of IL-6 and the subsequent activation of STAT3. The ERK pathway, on the other hand, triggers the expansion of MDSCs (Chalmin et al. 2010CHALMIN F ET AL. 2010. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest 120: 457-471.). Gao et al. (2020)GAO Y, XU H, LI N, WANG H, MA L, CHEN S, LIU J, ZHENG Y ZHANG Y. 2020. Renal cancer-derived exosomes induce tumor immune tolerance by MDSCs-mediated antigen-specific immunosuppression. Cell Commun Signal 18: 106. identified that tumor specific-antigen and HSP70 were enriched in renal cancer-derived exosomes, which is responsible for the expansion and activation of MDSCs, leading to the production of ROS and NO, secretion of IL-10 and TGF-β and intense arginase activity. The MDCS treated with these exosomes suppressed the cytotoxic activity of CD8+ T cells (Gao et al. 2020GAO Y, XU H, LI N, WANG H, MA L, CHEN S, LIU J, ZHENG Y ZHANG Y. 2020. Renal cancer-derived exosomes induce tumor immune tolerance by MDSCs-mediated antigen-specific immunosuppression. Cell Commun Signal 18: 106.). Tumor-derived exosomes harboring mutp53 shed miR-1246-enriched EVs. The uptake of these exosomes by macrophages induced their reprogramming to a tumor-supportive phenotype with increased secretion of IL-10, TNF-α, and CCL2 (Cooks et al. 2018COOKS T, PATERAS IS, JENKINS LM, PATEL KM, ROBLES AI, MORRIS J, FORSHEW T, APPELLA E, GORGOULIS VG HARRIS CC. 2018. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat Commun 9: 771.).

Crosstalk between cytokines signaling and EVs immunomodulation

Breast cancer (BC) is the most common cancer in women, after skin cancer. Studies conducted in BC models have indicated that CCL5 is crucial in attracting TAMs to tumors (Walens et al. 2019WALENS A, DIMARCO A V, LUPO R, KROGER BR, DAMRAUER JS ALVAREZ J V. 2019. CCL5 promotes breast cancer recurrence through macrophage recruitment in residual tumors. Elife 8: e43653.). In a syngeneic 4T1 mouse model, antagonists of CCL5 suppressed TAM recruitment (Robinson et al. 2003ROBINSON SC, SCOTT KA, WILSON JL, THOMPSON RG, PROUDFOOT AEI BALKWILL FR. 2003. A Chemokine Receptor Antagonist Inhibits Experimental Breast Tumor Growth. Cancer Res 63: 8360-8365.). EVs from triple-negative breast cancer (TNBC) reprogrammed macrophage towards TAM with a pro-tumor phenotype through an indirect mechanism involving autocrine stimulation of tumor cells by CCL5. Tumor-educated macrophages promote TNBC through TLR2 and -3 signaling (Rabe et al. 2018RABE DC, RUSTANDY FD, LEE J ROSNER MR. 2018. Tumor extracellular vesicles are required for tumor-associated macrophage programming. BioRxiv: 375022.). Accordingly, studies in vitro demonstrate that BC-derived exosomes stimulate the production of G-CSF, IL6, CCL2, and TNFα in macrophages, in a TLR2-dependent manner, inducing NF-kB activation (Chow et al. 2014CHOW A ET AL. 2014. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-κ B. Sci Rep 4: 5750.). Additionally, both TNF-α and TLR2 were found to be required for Lewis lung carcinoma metastasis, where the secreted factors from carcinoma cells induced macrophage production of TNF-α under versican stimulation through a process involving TLR2-TLR6 axis (Kim et al. 2009KIM S, TAKAHASHI H, LIN WW, DESCARGUES P, GRIVENNIKOV S, KIM Y, LUO JL KARIN M. 2009. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 457: 102-106.). Interestingly, Hartley et al. (2017)HARTLEY G, REGAN D, GUTH A DOW S. 2017. Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol Immunother 66: 523-535. identified that bone marrow-derived monocytes exhibit increased PD-L1 expression when exposed to tumor-conditioned media. TNF-α was identified as a critical cytokine responsible for this upregulation. The TNF-α production by monocytes was stimulated through the activation of TLR2 in response to versican secreted by tumor cells (Hartley et al. 2017HARTLEY G, REGAN D, GUTH A DOW S. 2017. Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol Immunother 66: 523-535.).

Tumor-derived EVs can modulate monocyte survival through antiapoptotic mechanisms and transport CD44v7/8 and CCR6 molecules that they take up (Baj-Krzyworzeka et al. 2006BAJ-KRZYWORZEKA M, SZATANEK R, WĘGLARCZYK K, BARAN J, URBANOWICZ B, BRAŃSKI P, RATAJCZAK MZ ZEMBALA M. 2006. Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes. Cancer Immunol Immunother 55: 808-818.). CCL20, the ligand of CCR6, induces monocyte migration in vitro and triggers macrophage accumulation in vivo in a model of colon cancer (Nandi et al. 2016NANDI B, SHAPIRO M, SAMUR MK, PAI C, FRANK NY, YOON C, PRABHALA RH, MUNSHI NC GOLD JS. 2016. Stromal CCR6 drives tumor growth in a murine transplantable colon cancer through recruitment of tumor-promoting macrophages. Oncoimmunology 5: e1189052.). High expression of CCL20 in tumor stroma has been identified as an adverse prognostic factor (Samaniego et al. 2018SAMANIEGO R, GUTIERREZ-GONZ ALEZ A, GUTIERREZ-SEIJO A, SANCHEZ-GREGORIO S, GARCÍA-GIMENEZ J, MERCADER E, MARQUEZ-RODAS I, AVILES JA, RELLOSO M SANCHEZ-MATEOS P. 2018. CCL20 Expression by tumor-associated macrophages predicts progression of human primary cutaneous melanoma. Cancer Immunol Res 6: 267-275.). Additionally, BC-derived exosomes promote monocyte survival due to the inhibition of caspase-8 activation through the MAPK pathway in monocytes (Song et al. 2016SONG X, DING Y, LIU G, YANG X, ZHAO R, ZHANG Y, ZHAO X, ANDERSON GJ NIE G. 2016. Cancer cell-derived exosomes induce mitogen-activated protein kinase-dependent monocyte survival by transport of functional receptor tyrosine kinases. J Biol Chem 291 8453-8464.). Lysine demethylase 3B KDM6B (JDJM1A) is related to reduced TAMs (Osawa et al. 2013OSAWA T ET AL. 2013. Inhibition of histone demethylase JMJD1A improves anti-angiogenic therapy and reduces tumor-associated macrophages. Cancer Res 73: 3019-3028.). Exosomal miR-138-5p produced by TNBC cells downregulates KDM6B expression in macrophages, regulating their polarization to an M2 phenotype and promoting the metastasis of BC to the lung (Xun et al. 2021XUN J ET AL. 2021. Cancer-derived exosomal miR-138-5p modulates polarization of tumor-Associated macrophages through inhibition of KDM6B. Theranostics 11: 6847.).

The neuroblastoma (NBL) exosomes increased miR-21 and miR-155 in both M1- and M2-polarized cells, with concomitant downregulation of TERF1. This circuit was associated with chemotherapy resistance in NBL (Challagundla et al. 2015CHALLAGUNDLA KB ET AL. 2015. Exosome-Mediated Transfer of microRNAs Within the Tumor Microenvironment and Neuroblastoma Resistance to Chemotherapy. JNCI J Natl Cancer Inst 107: djv135.). Snail, an EMT transcriptional factor, directly activates the transcription of miR-21. The miR-21-containing exosomes were engulfed by CD14+ human monocytes, suppressing the expression of M1 markers and increasing M2 markers, leading to M2-like polarization of TAMs. Inhibition of miR-21 suppresses snail-induced M2-like macrophage polarization and tumor progression in animal models (Hsieh et al. 2018HSIEH CH, TAI SK YANG MH. 2018. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia 20: 775-788.). Furthermore, melanoma-derived EVs containing miR-125b-5p have the potential to induce tumor-associated inflammation and angiogenesis as well as macrophage recruitment and survival, inducing the expression of IL1β, CCL1, CCL2, and CD80 in M1 macrophages by targeting Lysosomal Acid Lipase A (LIPA) (Gerloff et al. 2020GERLOFF D, LÜTZKENDORF J, MORITZ RKC, WERSIG T, MÄDER K, MÜLLER LP SUNDERKÖTTER C. 2020. Melanoma-derived exosomal mir-125b-5p educates tumor associated macrophages (TAMs) by targeting lysosomal acid lipase A (LIPA). Cancers (Basel) 12: 464.).

While cytotoxic chemotherapy is known to be an effective treatment for invasive BC, there is evidence from experimental studies in mice suggesting that it may also have pro-metastatic effects. EVs released after chemotherapy contain high levels of annexin A6, a protein that promotes NF-κB-dependent activation of endothelial cells. This activation leads to the induction of CCL2, resulting in the expansion of Ly6C+CCR2+ monocytes in the pulmonary pre-metastatic niche, ultimately facilitating the establishment of lung metastasis (Keklikoglou et al. 2019KEKLIKOGLOU I ET AL. 2019. Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nat Cell Biol 21: 190-202.). However, in a study by Plebanek and collaborators (2017), exosomes derived from non-metastatic melanoma cell lines migrate towards the lungs in tumor-bearing mice, preceding a significant increase in the Ly6Clow subpopulation. Non-metastatic exosomes were enriched with PEDF, which promotes the differentiation of patrolling monocytes in macrophages, which polarize to an M1 profile associated with the killing and phagocytosis of melanoma cells. (Plebanek et al. 2017PLEBANEK MP ET AL. 2017. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun 8: 1319.).

EVs carrying CSF-1 released by TNBC promote a tumor immune microenvironment associated with a better prognosis in TNBC patients through the induction of monocyte differentiation into macrophages; this leads to high levels of expression of CD163, MERKT, CD88, CD204, and PD-L1. The TNBC EVs promote an interferon response in macrophages, with the expression of immunostimulatory genes associated with the M1 phenotype, such as CXCL9 and CXCL10 (Tkach et al. 2022TKACH M ET AL. 2022. Extracellular vesicles from triple negative breast cancer promote pro-inflammatory macrophages associated with better clinical outcome. PNAS 119: e2107394119.). EVs present in the plasma of patients diagnosed with colorectal cancer are internalized by primary human monocytes cultured in vitro and lead to responses related to disease progression from a non-invasive state to an invasive state (Bjørnetrø et al. 2021BJØRNETRØ T ET AL. 2021. Uptake of circulating extracellular vesicles from rectal cancer patients and differential responses by human monocyte cultures. FEBS Open Bio 11: 724-740.). Results from a study conducted by Momen-Heravi and Bala (2018) suggest that EVs derived from head and neck squamous cell carcinoma (OSCC) are captured by monocytes and stimulate the activation of the NF-kB signaling pathway, which leads to an increase in MMP9 production and higher levels of COX2, PEG2 and VEGF mRNA after 24 hours of stimulation of THP-1 cells (Momen-Heravi & Bala 2018MOMEN-HERAVI F BALA S. 2018. Extracellular vesicles in oral squamous carcinoma carry oncogenic miRNA profile and reprogramme monocytes via NF-κB pathway. Oncotarget 9: 34838.). Interestingly, this expression profile is also seen in TEMs recruited to TME via Ang-2, whose upregulation results from hypoxia, leading to the destabilization of blood vessels (Murdoch et al. 2007MURDOCH C, TAZZYMAN S, WEBSTER S LEWIS CE. 2007. Expression of Tie-2 by Human Monocytes and Their Responses to Angiopoietin-2. J Immunol 178: 7405-7411.). In a VEGF-enriched TME, these blood vessels undergo angiogenic changes and sprout, ultimately forming new vasculature (Tait & Jones 2004TAIT CR JONES PF. 2004. Angiopoietins in tumours: The angiogenic switch. J Pathol 204: 1-10.).

In the TME, PKM2-containing ectosomes were found to accelerate the differentiation of monocytes into macrophages by activating glycolysis to provide acetyl-CoA, thus leading to the release of cytokines and chemokines promoting the progression of hepatocellular carcinoma (HCC). CCL1 was identified as one of the factors involved in a feedforward regulatory loop that further enhanced the excretion of PKM2 via ectosomes (Hou et al. 2020HOU P-P ET AL. 2020. Ectosomal PKM2 Promotes HCC by Inducing Macrophage Differentiation and Remodeling the Tumor Microenvironment. Mol Cell 78: 1192-1206.). PKM2, which catalyzes the final and irreversible step of glycolysis, is expressed at high levels in cancer cells. Studies have demonstrated that PKM2 promotes anabolic metabolism, cell survival, and tumor proliferation, mainly due to its intrinsic lower pyruvate kinase activity in the cytoplasm (Ward & Thompson 2012WARD PS THOMPSON CB. 2012. Metabolic Reprogramming: A Cancer Hallmark Even Warburg Did Not Anticipate. Cancer Cell 21: 297-308.). HCC-derived exosomes upregulate the expression of PD-L1 through STAT3 signaling and increase the secretion of IL-6, IL-10, IL-1β, and TNF-α in macrophages both in vitro and in vivo (Cheng et al. 2017CHENG L ET AL. 2017. Exosomes from melatonin treated hepatocellularcarcinoma cells alter the immunosupression status through STAT3 pathway in macrophages. Int J Biol Sci 13: 723.). IL-6/STAT3 signaling has been identified in numerous human cancers, including liver cancer, and is recognized as a crucial contributor to cancer initiation, growth, and advancement (Yu et al. 2007YU H, KORTYLEWSKI M PARDOLL D. 2007. Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat Rev Immunol 7: 41-51., 2009). Monocytes are reported to promote HCC growth via the IL-6/STAT3 axis, where the expression of STAT3 in these cells is correlated to a poor prognosis (Wu et al. 2011WU W-Y, LI J, WU Z-S, ZHANG C-L MENG X-L. 2011. STAT3 activation in monocytes accelerates liver cancer progression. BMC Cancer 11: 506.).

Chronic lymphocytic leukemia (CLL) is a type of cancer affecting mature B lymphocytes characterized by CD5+ and CD19+ cells (Nicholas et al. 2016NICHOLAS NS, APOLLONIO B RAMSAY AG. 2016. Tumor microenvironment (TME)-driven immune suppression in B cell malignancy. Biochim Biophys Acta - Mol Cell Res 1863: 471-482.). CCL-exosomes are reported to be enriched in Y RNAs that are uptaken by monocytes, leading to an upregulation of genes including CCL2, CCL4, CCL5, CXCL9, CXCL10, CXCL11, IL6, PD-L1, IDO1, and PD-1 (Haderk et al. 2017HADERK F ET AL. 2017. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci Immunol 2: eaah5509.). Y RNAs are conserved molecules (Perreault et al. 2007PERREAULT J, PERREAULT JP BOIRE G. 2007. Ro-associated Y RNAs in metazoans: Evolution and diversification. Mol Biol Evol 24: 1678-1689.) that bind with proteins. The expression of Y RNA is altered in human tumors, including breast (Guo et al. 2018bGUO Y, YU H, WANG J, SHENG Q, ZHAO S, ZHAO YY LEHMANN BD. 2018b. The landscape of small non-coding RNAs in triple-negative breast cancer. Genes (Basel) 9: 29.), colon, liver, pancreatic (Meiri et al. 2010MEIRI E ET AL. 2010. Discovery of microRNAs and other small RNAs in solid tumors. Nucleic Acids Res 38: 6234-6246.), and lung cancers (Li et al. 2018LI C, QIN F, HU F, XU H, SUN G, HAN G, WANG T GUO M. 2018. Characterization and selective incorporation of small non-coding RNAs in non-small cell lung cancer extracellular vesicles. Cell Biosci 8: 1-21.). In cultured monocytes/macrophages, Y RNAs play a role in caspase-dependent cell death and NF-κB-dependent inflammation via TLR7activation (Hizir et al. 2017HIZIR Z, BOTTINI S, GRANDJEAN V, TRABUCCHI M REPETTO E. 2017. RNY (YRNA)-derived small RNAs regulate cell death and inflammation in monocytes/macrophages. Cell Death Dis 8: e2530-e2530.).

Pancreatic cancer (PC)-derived exosomes downregulate HLA-DR expression on CD14+ monocytes, leading to tumor-induced immunosuppression in PC patients. Additionally, the exosomes can increase ROS production, arginase metabolism, and STAT3 signaling, leading to monocyte survival and immunosuppression (Javeed et al. 2017). Macrophages treated with PC cell-derived EVs and then co-culture with T cells arrest T cell function through the expression of T cell surface inhibitory receptors PD-1, TIGIT, and CTLA4, and reducing the secretion of IL-2, IFN-γ, and TNF-α by these cells; this seems to occur through the transfer of miR-155-5p to macrophages, thus promoting their polarization to the M2 phenotype (Wang & Gao 2021WANG S GAO Y. 2021. Pancreatic cancer cell-derived microRNA-155-5p-containing extracellular vesicles promote immune evasion by triggering EHF-dependent activation of Akt/NF-κB signaling pathway. Int Immunopharmacol 100: 107990.). Maia et al. (2020)MAIA J, OTAKE AH, POÇAS J, CARVALHO AS, BECK HC, MAGALHÃES A, MATTHIESEN R, STRANO MORAES MC COSTA-SILVA B. 2020. Transcriptome Reprogramming of CD11b+ Bone Marrow Cells by Pancreatic Cancer Extracellular Vesicles. Front Cell Dev Biol 8: 592518. identified that PC-EVs mediate a pancreatic cancer-BM communication axis by reprogramming gene expression in BM CD11b+ cells. Additionally, they found downregulated genes linked to monocyte/macrophage activation and trafficking, including transcription factors associated with monocyte and macrophage differentiation, macrophage polarization, cell-cell communication, and chemotactic response, such as Egr2, Nr4a1, Ccl2, Ccl3, and Rgs1 (Maia et al. 2020MAIA J, OTAKE AH, POÇAS J, CARVALHO AS, BECK HC, MAGALHÃES A, MATTHIESEN R, STRANO MORAES MC COSTA-SILVA B. 2020. Transcriptome Reprogramming of CD11b+ Bone Marrow Cells by Pancreatic Cancer Extracellular Vesicles. Front Cell Dev Biol 8: 592518.). PC-derived exosomes show significantly increased levels of the M2 markers CD163 and CD206 and induced an increased secretion of cytokines, including VEGF, MCP-1, IL-6, IL-1β, MMP-9, and TNFα in macrophages (Linton et al. 2018LINTON SS, ABRAHAM T, LIAO J, CLAWSON GA, BUTLER PJ, FOX T, KESTER M MATTERS GL. 2018. Tumor-promoting effects of pancreatic cancer cell exosomes on THP-1-derived macrophages. PLoS ONE 13: e0206759.). Furthermore, exosomes from PC initiate the formation of a pre-metastatic niche in the liver by inducing fibronectin (FN) expression and the recruitment of macrophages through the transfer of macrophage migration inhibitory factor (MIF) from the exosomes to the liver (Costa-Silva et al. 2015COSTA-SILVA B ET AL. 2015. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 17: 816-826.).

IL-10, TGF-β2, and CCL22 production were significantly elevated in murine alveolar macrophages following exposure to exosomes from metastatic osteosarcoma cells. Also, CXCL9 and CXCL10, markers of the M1 phenotype, were decreased in these cells (Wolf-Dennen et al. 2020WOLF-DENNEN K, GORDON N KLEINERMAN ES. 2020. Exosomal communication by metastatic osteosarcoma cells modulates alveolar macrophages to an M2 tumor-promoting phenotype and inhibits tumoricidal functions. Oncoimmunology 9.). Tumor-derived TGF-β-containing exomes induce the differentiation of myeloid cells with a decrease in CD14+ and HLA-DR expression and upregulate the secretion of IL-6, TNF-α, and TGF-β. When treated with these exosomes, monocytes significantly inhibited T-cell proliferation in a TGF-β-dependent manner (Valenti et al. 2006VALENTI R, HUBER V, FILIPAZZI P, PILLA L, SOVENA G, VILLA A, CORBELLI A, FAIS S, PARMIANI G RIVOLTINI L. 2006. Human Tumor-Released Microvesicles Promote the Differentiation of Myeloid Cells with Transforming Growth Factor-β–Mediated Suppressive Activity on T Lymphocytes. Cancer Res 66: 9290-9298.). In monocytes, EVs produced by osteosarcoma cells led to the upregulation of suppressive cytokines and effector molecules, such as IL-10 and arginase. Additionally, EVs caused a decrease in the expression of MHC class II and CD80 on monocytes, which was accompanied by an increase in the expression of PD-L1 on the monocyte membrane. (Luong et al. 2021LUONG N, LENZ JA, MODIANO JF OLSON JK. 2021. Extracellular Vesicles Secreted by Tumor Cells Promote the Generation of Suppressive Monocytes. Immunohorizons 5: 647-658.). Tumor-derived exosomes containing PGE2 and TGF-β induce the accumulation of MDSCs in the tumor, increasing IL-6 and VEGF production, facilitating tumor growth and suppressing immune function (Xiang et al. 2009XIANG X ET AL. 2009. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer 124: 2621-2633.).

Effects of EVs from hypoxic tumors on monocytes and macrophages

Studies have shown hypoxia as a significant driving force for tumor remodeling, promoting cancer progression and chemoresistance (Rohwer et al. 2009ROHWER N, LOBITZ S, DASKALOW K, JÖNS T, VIETH M, SCHLAG PM, KEMMNER W, WIEDENMANN B, CRAMER T HÖCKER M. 2009. HIF-1α determines the metastatic potential of gastric cancer cells. Br J Cancer 100: 772-781., Semenza 2010SEMENZA GL. 2010. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 29: 625-634., Rofstad et al. 2010ROFSTAD EK, GAUSTAD JV, EGELAND TAM, MATHIESEN B GALAPPATHI K. 2010. Tumors exposed to acute cyclic hypoxic stress show enhanced angiogenesis, perfusion and metastatic dissemination. Int J Cancer 127: 1535-1546.). Moreover, emerging evidence suggests a strong link between tumor hypoxia and immune suppression (Fu et al. 2021FU Z, MOWDAY AM, SMAILL JB, HERMANS IF PATTERSON AV. 2021. Tumour hypoxia-mediated immunosuppression: Mechanisms and therapeutic approaches to improve cancer immunotherapy. Cells 10: 1006.). Hypoxic lung-cancer-derived extracellular vesicles containing miRNA-103a from lung cancer cells can increase the expression of IL-10, CCL18, and VEGF-A in macrophages by directly targeting PTEN and causing activation of the PI3K/Akt and STAT3 signaling pathways. CD14+ monocytes with reduced expression of PTEN exhibited a CD163+CD206highHLA-DRlow phenotype (Hsu et al. 2018HSU YL, HUNG JY, CHANG WA, JIAN SF, LIN YS, PAN YC, WU CY KUO PL. 2018. Hypoxic Lung-Cancer-Derived Extracellular Vesicle MicroRNA-103a Increases the Oncogenic Effects of Macrophages by Targeting PTEN. Mol Ther 26: 568-581.). Hypoxia-induced tumor exosomes are highly enriched in immunomodulatory proteins and chemokines, including CSF-1, CCL2, TGF-β, and FTH. They drive pro-tumoral M2-like macrophage polarization in vivo and in vitro. Also, exposure to tumor exosomes significantly decreased the expression levels of let-7a target genes such as IRS-1, IRS-2, INSR, and IGF1R in bone marrow-derived macrophages. These findings point to let-7a miRNA as a potential suppressor of the insulin-mediated mTOR signaling pathway in macrophages (Park et al. 2019PARK JE, DUTTA B, TSE SW, GUPTA N, TAN CF, LOW JK, YEOH KW, KON OL, TAM JP SZE SK. 2019. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene 38: 5158-5173.). Hypoxic exosomes derived from ovarian cancer cells increased the expression of M2 markers genes, CD206, arginase-1, and IL-10. These exosomes seem to deliver miR-21-3p, miR-125b-5p, and miR-181d-5p to macrophages, modulating the SOCS4/5/STAT3 pathway inducing M2 polarization that supports tumor progression and metastasis (Chen et al. 2018CHEN X, ZHOU J, LI X, WANG X, LIN Y WANG X. 2018. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett 435: 80-91.). Glioma exosomes derived from hypoxic tumors exert an immunosuppressive effect through the induction of MDSCs through miR-10a and miR-21, leading to the expression of TGF-β and IL-10; this probably occurs by targeting RORA and PTEN pathways in glioma-bearing mice (Guo et al. 2018aGUO X, QIU W, LIU Q, QIAN M, WANG S, ZHANG Z, GAO X, CHEN Z, XUE H LI G. 2018a. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten Pathways. Oncogene 37: 4239-4259.).

Conclusion and future perspectives

As the frequency and occurrence of cancers continue to rise, there is an urgent demand for the creation of therapeutic approaches. This necessity arises as our comprehension of cancer’s intricate nature as a disease evolves. Despite the significant biological functions that EVs play in both standard and pathological conditions and their capacity to better encompass the dynamic diversity of cancer, the limited understanding and technical obstacles have hindered their effective application in clinical settings. The functional relevance of human tumor-derived EVs’ effects on monocytes and macrophages requires further characterization.

Future studies focusing on the production, composition, and physiology of different vesicles will add more knowledge about these, thus contributing to the development of possible therapeutic applications and a better understanding of tumor pathology, focused on understanding how the tumor microenvironment influences prognosis and development of the disease, allowing the development of engineered EVs based on these findings. Numerous preliminary investigations have unveiled EVs’ as a diagnostic tool and their immunotherapeutic capabilities across diverse cancer types. However, clinical trials exploring the therapeutic aspects of engineered EVs are currently in their early stages of development. Furthermore, conducting pertinent clinical studies with a substantial sample size is desirable. Additionally, there is a need to enhance the standardized techniques for isolating and purifying EVs prior to their clinical implementation to better evaluate their effects on cells.

EVs in the extracellular environment play a crucial role in regulating monocyte and macrophage functions in the TME, significantly impacting cancer progression and immune evasion. The main related studies are summarized in Table I. Monocytes stimulated by EVs can migrate to cancer tissue, where they differentiate into TAMs or M2 macrophages, and produce several cytokines, such as IL-6, IL-1β, TNF-α, and IL-10, enhance the membrane expression of PD-L1, CD14 e CD206, exhibiting a pro-tumoral profile and supporting tumor growth (Figure 1). Therefore, EVs can modulate the immune response and promote tumor progression by altering the phenotype and function of these cells. Overall, EVs provide insights into the complex interplay between cancer cells and the immune system.

Figure 1
Tumor-derived extracellular vesicles affect the function and phenotype of monocytes. The release of EVs under different conditions can induce monocyte differentiation into macrophages, which are polarized towards a profile that supports tumor growth and progression. This process also leads to the upregulation of the main cytokines and chemokines released by monocytes within the TME, such as IL-6, VEGF, TNF-α, and IL-10. Moreover, EVs upregulate CD14, PD-L1, and CD206 expression on monocytes, thus preventing apoptosis and enhancing their migration.
Table I
Main findings of the effects of extracellular vesicles on monocytes, macrophages, and MDSC cells related to the immunomodulation of functions and phenotypes.

REFERENCES

  • AHMADZADEH M, JOHNSON LA, HEEMSKERK B, WUNDERLICH JR, DUDLEY ME, WHITE DE ROSENBERG SA. 2009. Tumor antigen–specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114: 1537-1544.
  • AL-NEDAWI K, MEEHAN B, MICALLEF J, LHOTAK V, MAY L, GUHA A RAK J. 2008. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol 10: 619-624.
  • ANDREOLA G ET AL. 2002. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J Exp Med 195 1303-1316.
  • AZAMBUJA JH, LUDWIG N, YERNENI SS, BRAGANHOL E WHITESIDE TL. 2020. Arginase-1+ exosomes from reprogrammed macrophages promote glioblastoma progression. Int J Mol Sci 21: 3990.
  • BAGHBAN R, ROSHANGAR L, JAHANBAN-ESFAHLAN R, SEIDI K, EBRAHIMI-KALAN A, JAYMAND M, KOLAHIAN S, JAVAHERI T ZARE P. 2020. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal 18: 59.
  • BAJ-KRZYWORZEKA M, SZATANEK R, WĘGLARCZYK K, BARAN J, URBANOWICZ B, BRAŃSKI P, RATAJCZAK MZ ZEMBALA M. 2006. Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes. Cancer Immunol Immunother 55: 808-818.
  • BELTRAN M, PUIG I, PEÑA C, GARCÍA JM, ÁLVAREZ AB, PEÑA R, BONILLA F DE HERREROS AG. 2008. A natural antisense transcript regulates Zeb2/Sip1 gene expression during Snail1-induced epithelial-mesenchymal transition. Genes Dev 22: 756-769.
  • BJØRNETRØ T ET AL. 2021. Uptake of circulating extracellular vesicles from rectal cancer patients and differential responses by human monocyte cultures. FEBS Open Bio 11: 724-740.
  • BOYETTE LB, MACEDO C, HADI K, ELINOFF BD, WALTERS JT, RAMASWAMI B, CHALASANI G, TABOAS JM, LAKKIS FG METES DM. 2017. Phenotype, function, and differentiation potential of human monocyte subsets. PLoS ONE 12: e0176460.
  • BRETZ NP ET AL. 2013. Body fluid exosomes promote secretion of inflammatory cytokines in monocytic cells via Toll-Like receptor signaling. J Biol Chem 288: 36691-36702.
  • BRONTE V ET AL. 2016. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 7: 12150.
  • CAMUSSI G, DEREGIBUS MC, BRUNO S, CANTALUPPI V BIANCONE L. 2010. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int 78: 838-848.
  • CHALLAGUNDLA KB ET AL. 2015. Exosome-Mediated Transfer of microRNAs Within the Tumor Microenvironment and Neuroblastoma Resistance to Chemotherapy. JNCI J Natl Cancer Inst 107: djv135.
  • CHALMIN F ET AL. 2010. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest 120: 457-471.
  • CHEN X, ZHOU J, LI X, WANG X, LIN Y WANG X. 2018. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett 435: 80-91.
  • CHENG L ET AL. 2017. Exosomes from melatonin treated hepatocellularcarcinoma cells alter the immunosupression status through STAT3 pathway in macrophages. Int J Biol Sci 13: 723.
  • CHOW A ET AL. 2014. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-κ B. Sci Rep 4: 5750.
  • CHUNG W ET AL. 2017. Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer. Nat Commun 8: 15081.
  • CIOCCA DR CALDERWOOD SK. 2005. Heat shock proteins in cancer: Diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones 10: 86.
  • COCUCCI E MELDOLESI J. 2015. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol 25: 364-372.
  • COOKS T, PATERAS IS, JENKINS LM, PATEL KM, ROBLES AI, MORRIS J, FORSHEW T, APPELLA E, GORGOULIS VG HARRIS CC. 2018. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat Commun 9: 771.
  • COSTA-SILVA B ET AL. 2015. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 17: 816-826.
  • DAVIDOV V, JENSEN G, MAI S, CHEN SH PAN PY. 2020. Analyzing One Cell at a TIME: Analysis of Myeloid Cell Contributions in the Tumor Immune Microenvironment. Front Immunol 11: 1842.
  • DE PALMA M, VENNERI MA, GALLI R, SERGI LS, POLITI LS, SAMPAOLESI M NALDINI L. 2005. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 8: 211-226.
  • DE VRIJ J ET AL. 2015. Glioblastoma-derived extracellular vesicles modify the phenotype of monocytic cells. Int J Cancer 137.
  • DEL CONDE I, SHRIMPTON CN, THIAGARAJAN P LÓPEZ JA. 2005. Tissue-factor-bearing microvesicles arise from lipid rafts and fuse with activated platelets to initiate coagulation. Blood 106: 1604-1611.
  • DENARDO DG RUFFELL B. 2019. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol 19: 369-382.
  • DEVÊVRE EF, RENOVATO-MARTINS M, CLÉMENT K, SAUTÈS-FRIDMAN C, CREMER I POITOU C. 2015. Subpopulations in Human Obesity Profiling of the Three Circulating Monocyte. J Immunol 194: 3917-3923.
  • DING J, GUO C, HU P, CHEN J, LIU Q, WU X, CAO Y WU J. 2016. CSF1 is involved in breast cancer progression through inducing monocyte differentiation and homing. Int J Oncol 49: 2064-2074.
  • DOLO V, GINESTRA A, CASSARÀ D, VIOLINI S, LUCANIA G, TORRISI MR, NAGASE H, CANEVARI S, PAVAN A VITTORELLI ML. 1998. Selective localization of matrix metalloproteinase 9, β1 integrins, and human lymphocyte antigen class I molecules on membrane vesicles shed by 8701-BC breast carcinoma cells. Cancer Res 58: 4468-4474.
  • D’SOUZA-SCHOREY C CHAVRIER P. 2006. ARF proteins: roles in membrane traffic and beyond. Nat Rev Mol Cell Biol 7: 347-358.
  • DULUC D ET AL. 2007. Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood 110: 4319-4330.
  • FAN QM ET AL. 2014. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett 352: 160-168.
  • FLEMING V ET AL. 2019. Melanoma extracellular vesicles generate immunosuppressive myeloid cells by upregulating PD-L1 via TLR4 signaling. Cancer Res 79: 4715-4728.
  • FRIDMAN WH, ZITVOGEL L, SAUTÈS–FRIDMAN C KROEMER G. 2017. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 14: 717-734.
  • FRIEDL P, MAASER K, KLEIN CE, NIGGEMANN B, KROHNE G ZÄNKER KS. 1997. Migration of highly aggressive MV3 melanoma cells in 3-dimensional collagen lattices results in local matrix reorganization and shedding of α2 and β1 integrins and CD44. Cancer Res 57: 2061-2070.
  • FU Z, MOWDAY AM, SMAILL JB, HERMANS IF PATTERSON AV. 2021. Tumour hypoxia-mediated immunosuppression: Mechanisms and therapeutic approaches to improve cancer immunotherapy. Cells 10: 1006.
  • GABRILOVICH D, ISHIDA T, OYAMA T, RAN S, KRAVTSOV V, NADAF S CARBONE DP. 1998. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood 92: 4150-4166.
  • GAO Y, XU H, LI N, WANG H, MA L, CHEN S, LIU J, ZHENG Y ZHANG Y. 2020. Renal cancer-derived exosomes induce tumor immune tolerance by MDSCs-mediated antigen-specific immunosuppression. Cell Commun Signal 18: 106.
  • GÄRTNER K, BATTKE C, DÜNZKOFER J, HÜLS C, VON NEUBECK B, KELLNER M-K, FIESTAS E, FACKLER S, LANG S ZEIDLER R. 2018. Tumor-derived extracellular vesicles activate primary monocytes. Cancer Med 7: 2013-2020.
  • GEISSMANN F, JUNG S LITTMAN DR. 2003. Blood Monocytes Consist of Two Principal Subsets with Distinct Migratory Properties. Immunity 19: 71-82.
  • GERLOFF D, LÜTZKENDORF J, MORITZ RKC, WERSIG T, MÄDER K, MÜLLER LP SUNDERKÖTTER C. 2020. Melanoma-derived exosomal mir-125b-5p educates tumor associated macrophages (TAMs) by targeting lysosomal acid lipase A (LIPA). Cancers (Basel) 12: 464.
  • GREN ST, RASMUSSEN TB, JANCIAUSKIENE S, HAKANSSON K, GERWIEN JG GRIP O. 2015. A Single-Cell Gene-Expression Profile Reveals Inter-Cellular Heterogeneity within Human Monocyte Subsets. PLoS ONE 10: e0144351.
  • GUO X, QIU W, LIU Q, QIAN M, WANG S, ZHANG Z, GAO X, CHEN Z, XUE H LI G. 2018a. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten Pathways. Oncogene 37: 4239-4259.
  • GUO Y, YU H, WANG J, SHENG Q, ZHAO S, ZHAO YY LEHMANN BD. 2018b. The landscape of small non-coding RNAs in triple-negative breast cancer. Genes (Basel) 9: 29.
  • GUPTA RA ET AL. 2010. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464: 1071-1076.
  • HADERK F ET AL. 2017. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci Immunol 2: eaah5509.
  • HANNA RN ET AL. 2015. Patrolling monocytes control tumor metastasis to the lung. Science 350: 985-990.
  • HARTLEY G, REGAN D, GUTH A DOW S. 2017. Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol Immunother 66: 523-535.
  • HEGDE S, LEADER AM MERAD M. 2021. MDSC: Markers, development, states, and unaddressed complexity. Immunity 54: 875-884.
  • HEIJNEN HFG, SCHIEL AE, FIJNHEER R, GEUZE HJ SIXMA JJ. 1999. Activated platelets release two types of membrane vesicles: Microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and α-granules. Blood 94: 3791-3799.
  • HESS C, SADALLAH S, HEFTI A, LANDMANN R SCHIFFERLI J-A. 1999. Ectosomes Released by Human Neutrophils Are Specialized Functional Units. J Immunol 163: 4564-4573.
  • HIJDRA D, VORSELAARS ADM, GRUTTERS JC, CLAESSEN AME RIJKERS GT. 2013. Phenotypic Characterization of Human Intermediate Monocytes. Front Immunol 4: 339.
  • HINES BT ET AL. 2020. The role of extracellular vesicles and PD-L1 in glioblastoma-mediated immunosuppressive monocyte induction. Neuro Oncol 22: 967-978.
  • HIZIR Z, BOTTINI S, GRANDJEAN V, TRABUCCHI M REPETTO E. 2017. RNY (YRNA)-derived small RNAs regulate cell death and inflammation in monocytes/macrophages. Cell Death Dis 8: e2530-e2530.
  • HOU P-P ET AL. 2020. Ectosomal PKM2 Promotes HCC by Inducing Macrophage Differentiation and Remodeling the Tumor Microenvironment. Mol Cell 78: 1192-1206.
  • HSIEH CH, TAI SK YANG MH. 2018. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia 20: 775-788.
  • HSU YL, HUNG JY, CHANG WA, JIAN SF, LIN YS, PAN YC, WU CY KUO PL. 2018. Hypoxic Lung-Cancer-Derived Extracellular Vesicle MicroRNA-103a Increases the Oncogenic Effects of Macrophages by Targeting PTEN. Mol Ther 26: 568-581.
  • HUBER V ET AL. 2018. Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J Clin Invest 128: 5505-5516.
  • JAMES HH. 2015. ESCRTs are everywhere. Embo J 34: 2398-2407.
  • JAVEED N ET AL. 2020. Cancer exosome-derived miR-9 and miR-181a promote the development of early-stage MDSCs via interfering with SOCS3 and PIAS3 respectively in breast cancer. Oncogene 39: 4681-4694.
  • JOHNSTONE RM, ADAM M, HAMMOND JR, ORR L TURBIDE C. 1987. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem 262: 9412-9420.
  • JUNEJA VR, MCGUIRE KA, MANGUSO RT, LAFLEUR MW, COLLINS N, NICHOLAS HAINING W, FREEMAN GJ SHARPE AH. 2017. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med 214: 895-904.
  • KADOMOTO S, IZUMI K MIZOKAMI A. 2021. Roles of CCL2-CCR2 axis in the tumor microenvironment. Int J Mol Sci 22: 8530.
  • KALLURI R. 2016. The biology and function of exosomes in cancer. J Clin Invest 126: 1208-1215.
  • KAPELLOS TS, BONAGURO L, GEMÜND I, REUSCH N, SAGLAM A, HINKLEY ER SCHULTZE JL. 2019. Human Monocyte Subsets and Phenotypes in Major Chronic Inflammatory Diseases. Front Immunol 10: 2035.
  • KEKLIKOGLOU I ET AL. 2019. Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nat Cell Biol 21: 190-202.
  • KIM S, TAKAHASHI H, LIN WW, DESCARGUES P, GRIVENNIKOV S, KIM Y, LUO JL KARIN M. 2009. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 457: 102-106.
  • KURYWCHAK P, TAVORMINA J KALLURI R. 2018. The emerging roles of exosomes in the modulation of immune responses in cancer. Genome Med 10: 23.
  • LARIONOVA I, TUGUZBAEVA G, PONOMARYOVA A, STAKHEYEVA M, CHERDYNTSEVA N, PAVLOV V, CHOINZONOV E KZHYSHKOWSKA J. 2020. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front Oncol 10: 566511.
  • LEE CH, WU CL SHIAU AL. 2010. Toll-like receptor 4 signaling promotes tumor growth. J Immunother 33: 73-82.
  • LEWIS CE, DE PALMA M NALDINI L. 2007. Tie2-expressing monocytes and tumor angiogenesis: Regulation by hypoxia and angiopoietin-2. Cancer Res 67: 8429-8432.
  • LI C, QIN F, HU F, XU H, SUN G, HAN G, WANG T GUO M. 2018. Characterization and selective incorporation of small non-coding RNAs in non-small cell lung cancer extracellular vesicles. Cell Biosci 8: 1-21.
  • LINTON SS, ABRAHAM T, LIAO J, CLAWSON GA, BUTLER PJ, FOX T, KESTER M MATTERS GL. 2018. Tumor-promoting effects of pancreatic cancer cell exosomes on THP-1-derived macrophages. PLoS ONE 13: e0206759.
  • LISCHNIG A, BERGQVIST M, OCHIYA T LÄSSER C. 2022. Quantitative Proteomics Identifies Proteins Enriched in Large and Small Extracellular Vesicles. Mol Cell Proteomics 21.
  • LOYER X, VION A-C, TEDGUI A BOULANGER CM. 2014. Microvesicles as Cell–Cell Messengers in Cardiovascular Diseases. Circ Res 114: 345-353.
  • LUONG N, LENZ JA, MODIANO JF OLSON JK. 2021. Extracellular Vesicles Secreted by Tumor Cells Promote the Generation of Suppressive Monocytes. Immunohorizons 5: 647-658.
  • MAIA J, OTAKE AH, POÇAS J, CARVALHO AS, BECK HC, MAGALHÃES A, MATTHIESEN R, STRANO MORAES MC COSTA-SILVA B. 2020. Transcriptome Reprogramming of CD11b+ Bone Marrow Cells by Pancreatic Cancer Extracellular Vesicles. Front Cell Dev Biol 8: 592518.
  • MANTOVANI A, MARCHESI F, MALESCI A, LAGHI L ALLAVENA P. 2017. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 14: 399-416.
  • MANTOVANI A, SAVINO B, LOCATI M, ZAMMATARO L, ALLAVENA P BONECCHI R. 2010. The chemokine system in cancer biology and therapy. Cytokine Growth Factor Rev 21: 27-39.
  • MEDREK C, PONTÉN F, JIRSTRÖM K LEANDERSSON K. 2012. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer 12: 1-9.
  • MEIRI E ET AL. 2010. Discovery of microRNAs and other small RNAs in solid tumors. Nucleic Acids Res 38: 6234-6246.
  • MINCIACCHI VR, FREEMAN MR DI VIZIO D. 2015. Extracellular Vesicles in Cancer: Exosomes, Microvesicles and the Emerging Role of Large Oncosomes. Semin Cell Dev Biol 40: 41-51.
  • MIZUTANI K, SUD S, MCGREGOR NA, MARTINOVSKI G, RICE BT, CRAIG MJ, VARSOS ZS, ROCA H PIENTA KJ. 2009. The chemokine CCL2 increases prostate tumor growth and bone metastasis through macrophage and osteoclast recruitment. Neoplasia 11: 1235-1242.
  • MOMEN-HERAVI F BALA S. 2018. Extracellular vesicles in oral squamous carcinoma carry oncogenic miRNA profile and reprogramme monocytes via NF-κB pathway. Oncotarget 9: 34838.
  • MURALIDHARAN-CHARI V, CLANCY J, PLOU C, ROMAO M, CHAVRIER P, RAPOSO G D’SOUZA-SCHOREY C. 2009. ARF6-Regulated Shedding of Tumor Cell-Derived Plasma Membrane Microvesicles. Curr Biol 19: 1875-1885.
  • MURDOCH C, TAZZYMAN S, WEBSTER S LEWIS CE. 2007. Expression of Tie-2 by Human Monocytes and Their Responses to Angiopoietin-2. J Immunol 178: 7405-7411.
  • NANDI B, SHAPIRO M, SAMUR MK, PAI C, FRANK NY, YOON C, PRABHALA RH, MUNSHI NC GOLD JS. 2016. Stromal CCR6 drives tumor growth in a murine transplantable colon cancer through recruitment of tumor-promoting macrophages. Oncoimmunology 5: e1189052.
  • NICHOLAS NS, APOLLONIO B RAMSAY AG. 2016. Tumor microenvironment (TME)-driven immune suppression in B cell malignancy. Biochim Biophys Acta - Mol Cell Res 1863: 471-482.
  • NIELSEN SR SCHMID MC. 2017. Macrophages as Key Drivers of Cancer Progression and Metastasis. Mediators Inflamm 2017: 1-11.
  • OBREGON C, ROTHEN-RUTISHAUSER B, GITAHI SK, GEHR P NICOD LP. 2006. Exovesicles from human activated dendritic cells fuse with resting dendritic cells, allowing them to present alloantigens. Am J Pathol 169: 2127-2136.
  • OSAWA T ET AL. 2013. Inhibition of histone demethylase JMJD1A improves anti-angiogenic therapy and reduces tumor-associated macrophages. Cancer Res 73: 3019-3028.
  • PARK JE, DUTTA B, TSE SW, GUPTA N, TAN CF, LOW JK, YEOH KW, KON OL, TAM JP SZE SK. 2019. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene 38: 5158-5173.
  • PASQUET JM, DACHARY-PRIGENT J NURDEN AT. 1996. Calcium influx is a determining factor of calpain activation and microparticle formation in platelets. Eur J Biochem 239: 647-654.
  • PATEL AA ET AL. 2017. The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J Exp Med 214: 1913-1923.
  • PATHAN M, FONSEKA P, CHITTI SV, KANG T, SANWLANI R, VAN DEUN J, HENDRIX A MATHIVANAN S. 2019. Vesiclepedia 2019: A compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res 47: D516-D519.
  • PEINADO H ET AL. 2012. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 18: 883-891.
  • PERREAULT J, PERREAULT JP BOIRE G. 2007. Ro-associated Y RNAs in metazoans: Evolution and diversification. Mol Biol Evol 24: 1678-1689.
  • PETERSEN GM, CHARI ST, DIETZ AB MUKHOPADHYAY D. 2017. Immunosuppressive CD14+HLA-DRlo/neg monocytes are elevated in pancreatic cancer and “primed” by tumor-derived exosomes. Oncoimmunology 6: e1252013.
  • PHETFONG J, TAWONSAWATRUK T, KAMPROM W, ONTONG P, TANYONG D, BORWORNPINYO S SUPOKAWEJ A. 2022. Bone marrow-mesenchymal stem cell-derived extracellular vesicles affect proliferation and apoptosis of leukemia cells in vitro. FEBS Open Bio 12: 470-479.
  • PLEBANEK MP ET AL. 2017. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun 8: 1319.
  • POPĚNA I, ABOLS A, SAULITE L, PLEIKO K, ZANDBERGA E, JĚKABSONS K, ENDZELIŅŠ E, LLORENTE A, LINĚ A RIEKSTIŅA U. 2018. Effect of colorectal cancer-derived extracellular vesicles on the immunophenotype and cytokine secretion profile of monocytes and macrophages. Cell Commun Signal 16: 1-12.
  • POSTE G NICOLSON G l. 1980. Arrest and metastasis of blood-borne tumor cells are modified by fusion of plasma membrane vesicles from highly metastatic cells. Proc Natl Acad Sci 77: 399-403.
  • PRENDERGAST GC, MALACHOWSKI WJ, MONDAL A, SCHERLE P MULLER AJ. 2018. Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer. In: Int Rev Cell Mol Biol 336: 175-203.
  • QIAN BZ, LI J, ZHANG H, KITAMURA T, ZHANG J, CAMPION LR, KAISER EA, SNYDER LA POLLARD JW. 2011a. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nat 475: 222-225.
  • RABE DC, RUSTANDY FD, LEE J ROSNER MR. 2018. Tumor extracellular vesicles are required for tumor-associated macrophage programming. BioRxiv: 375022.
  • RAPOSO G STOORVOGEL W. 2013. Extracellular vesicles: Exosomes, microvesicles, and friends. J Cell Biol 200: 373-383.
  • REN B, CUI M, YANG G, WANG H, FENG M, YOU L ZHAO Y. 2018. Tumor microenvironment participates in metastasis of pancreatic cancer. Mol Cancer 17: 108.
  • ROBINSON SC, SCOTT KA, WILSON JL, THOMPSON RG, PROUDFOOT AEI BALKWILL FR. 2003. A Chemokine Receptor Antagonist Inhibits Experimental Breast Tumor Growth. Cancer Res 63: 8360-8365.
  • ROFSTAD EK, GAUSTAD JV, EGELAND TAM, MATHIESEN B GALAPPATHI K. 2010. Tumors exposed to acute cyclic hypoxic stress show enhanced angiogenesis, perfusion and metastatic dissemination. Int J Cancer 127: 1535-1546.
  • ROHWER N, LOBITZ S, DASKALOW K, JÖNS T, VIETH M, SCHLAG PM, KEMMNER W, WIEDENMANN B, CRAMER T HÖCKER M. 2009. HIF-1α determines the metastatic potential of gastric cancer cells. Br J Cancer 100: 772-781.
  • SAMANIEGO R, GUTIERREZ-GONZ ALEZ A, GUTIERREZ-SEIJO A, SANCHEZ-GREGORIO S, GARCÍA-GIMENEZ J, MERCADER E, MARQUEZ-RODAS I, AVILES JA, RELLOSO M SANCHEZ-MATEOS P. 2018. CCL20 Expression by tumor-associated macrophages predicts progression of human primary cutaneous melanoma. Cancer Immunol Res 6: 267-275.
  • SEMENZA GL. 2010. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 29: 625-634.
  • SONG W, YAN D, WEI T, LIU Q, ZHOU X LIU J. 2018. Tumor-derived extracellular vesicles in angiogenesis. Biomed Pharmacother 102: 1203-1208.
  • SONG X, DING Y, LIU G, YANG X, ZHAO R, ZHANG Y, ZHAO X, ANDERSON GJ NIE G. 2016. Cancer cell-derived exosomes induce mitogen-activated protein kinase-dependent monocyte survival by transport of functional receptor tyrosine kinases. J Biol Chem 291 8453-8464.
  • SRIVASTAVA MK, SINHA P, CLEMENTS VK, RODRIGUEZ P OSTRAND-ROSENBERG S. 2010. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 70: 68-77.
  • STÅHL A, JOHANSSON K, MOSSBERG M, KAHN R KARPMAN D. 2019. Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases. Pediatr Nephrol 34: 11-30.
  • SUNG BH, PARENT CA WEAVER AM. 2021. Extracellular vesicles: Critical players during cell migration. Dev Cell 56: 1861-1874.
  • TAIT CR JONES PF. 2004. Angiopoietins in tumours: The angiogenic switch. J Pathol 204: 1-10.
  • THÉRY C, OSTROWSKI M SEGURA E. 2009. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 9: 581-593.
  • TKACH M ET AL. 2022. Extracellular vesicles from triple negative breast cancer promote pro-inflammatory macrophages associated with better clinical outcome. PNAS 119: e2107394119.
  • TRICARICO C, CLANCY J D’SOUZA-SCHOREY C. 2017. Biology and biogenesis of shed microvesicles. Small GTPases 8: 220-232.
  • UGEL S, CANEGRAVE S, DE SANCTIS F BRONTE V. 2021. Monocytes in the Tumor Microenvironment. Annu Rev Pathol 16: 93-122.
  • VALENTI R, HUBER V, FILIPAZZI P, PILLA L, SOVENA G, VILLA A, CORBELLI A, FAIS S, PARMIANI G RIVOLTINI L. 2006. Human Tumor-Released Microvesicles Promote the Differentiation of Myeloid Cells with Transforming Growth Factor-β–Mediated Suppressive Activity on T Lymphocytes. Cancer Res 66: 9290-9298.
  • VAN FURTH R COHN ZA. 1968. The origin and kinetics of mononuclear phagocytes. J Exp Med 128: 415-435.
  • VAN NIEL G, D’ANGELO G RAPOSO G. 2018. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 19: 213-228.
  • VELUSWAMY P BRUDER D. 2018. PD-1/PD-L1 pathway inhibition to restore effector functions in exhausted CD8+ T cells: Chances, limitations and potential risks. Transl Cancer Res 7.
  • VILLANI AC ET AL. 2017. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes and progenitors. Science 356: eaah4573.
  • WAGNER J ET AL. 2019. A Single-Cell Atlas of the Tumor and Immune Ecosystem of Human Breast Cancer. Cell 177: 1330-1345.
  • WALENS A, DIMARCO A V, LUPO R, KROGER BR, DAMRAUER JS ALVAREZ J V. 2019. CCL5 promotes breast cancer recurrence through macrophage recruitment in residual tumors. Elife 8: e43653.
  • WANG F ET AL. 2018. Tumor-derived exosomes induce PD1+ macrophage population in human gastric cancer that promotes disease progression. Oncogenesis 7: 41.
  • WANG S GAO Y. 2021. Pancreatic cancer cell-derived microRNA-155-5p-containing extracellular vesicles promote immune evasion by triggering EHF-dependent activation of Akt/NF-κB signaling pathway. Int Immunopharmacol 100: 107990.
  • WARD PS THOMPSON CB. 2012. Metabolic Reprogramming: A Cancer Hallmark Even Warburg Did Not Anticipate. Cancer Cell 21: 297-308.
  • WOLF-DENNEN K, GORDON N KLEINERMAN ES. 2020. Exosomal communication by metastatic osteosarcoma cells modulates alveolar macrophages to an M2 tumor-promoting phenotype and inhibits tumoricidal functions. Oncoimmunology 9.
  • WU C, ZHU Y, JIANG J, ZHAO J, ZHANG XG XU N. 2006. Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochem 108: 19-24.
  • WU W-Y, LI J, WU Z-S, ZHANG C-L MENG X-L. 2011. STAT3 activation in monocytes accelerates liver cancer progression. BMC Cancer 11: 506.
  • XIANG X ET AL. 2009. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer 124: 2621-2633.
  • XIU B ET AL. 2019. LINC02273 drives breast cancer metastasis by epigenetically increasing AGR2 transcription. Mol Cancer 18: 1-20.
  • XUN J ET AL. 2021. Cancer-derived exosomal miR-138-5p modulates polarization of tumor-Associated macrophages through inhibition of KDM6B. Theranostics 11: 6847.
  • YANG J, ZHANG L, YU C, YANG X-F WANG H. 2014. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res 2: 1.
  • YAO X, TU Y, XU Y, GUO Y, YAO F ZHANG X. 2020. Endoplasmic reticulum stress-induced exosomal miR-27a-3p promotes immune escape in breast cancer via regulating PD-L1 expression in macrophages. J Cell Mol Med 24: 9560-9573.
  • YIN Y ET AL. 2022. Colorectal Cancer-Derived Small Extracellular Vesicles Promote Tumor Immune Evasion by Upregulating PD-L1 Expression in Tumor-Associated Macrophages. Adv Sci 9: 2102620.
  • YU H, KORTYLEWSKI M PARDOLL D. 2007. Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat Rev Immunol 7: 41-51.
  • YU H, PARDOLL D JOVE R. 2009. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 9: 798-809.
  • ZHANG Y, LIU Y, LIU H TANG WH. 2019. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci 9: 19.
  • ZHOU J, TANG Z, GAO S, LI C, FENG Y ZHOU X. 2020. Tumor-Associated Macrophages: Recent Insights and Therapies. Front Oncol 10: 188.
  • ZHU Y ET AL. 2017. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 47: 323-338.
  • ZIEGLER-HEITBROCK L ET AL. 2010. Nomenclature of monocytes and dendritic cells in blood. Blood 116: e74-e80.
  • ZILIONIS R ET AL. 2019. Single cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50: 1317.

Publication Dates

  • Publication in this collection
    24 June 2024
  • Date of issue
    2024

History

  • Received
    07 Nov 2023
  • Accepted
    17 Feb 2024
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100, CLOCKSS system has permission to ingest, preserve, and serve this Archival Unit - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br