1
|
Van Morckhoven D, Dubois N, Bron D, Meuleman N, Lagneaux L, Stamatopoulos B. Extracellular vesicles in hematological malignancies: EV-dence for reshaping the tumoral microenvironment. Front Immunol 2023; 14:1265969. [PMID: 37822925 PMCID: PMC10562589 DOI: 10.3389/fimmu.2023.1265969] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/04/2023] [Indexed: 10/13/2023] Open
Abstract
Following their discovery at the end of the 20th century, extracellular vesicles (EVs) ranging from 50-1,000 nm have proven to be paramount in the progression of many cancers, including hematological malignancies. EVs are a heterogeneous group of cell-derived membranous structures that include small EVs (commonly called exosomes) and large EVs (microparticles). They have been demonstrated to participate in multiple physiological and pathological processes by allowing exchange of biological material (including among others proteins, DNA and RNA) between cells. They are therefore a crucial way of intercellular communication. In this context, malignant cells can release these extracellular vesicles that can influence their microenvironment, induce the formation of a tumorigenic niche, and prepare and establish distant niches facilitating metastasis by significantly impacting the phenotypes of surrounding cells and turning them toward supportive roles. In addition, EVs are also able to manipulate the immune response and to establish an immunosuppressive microenvironment. This in turn allows for ideal conditions for heightened chemoresistance and increased disease burden. Here, we review the latest findings and reports studying the effects and therapeutic potential of extracellular vesicles in various hematological malignancies. The study of extracellular vesicles remains in its infancy; however, rapid advances in the analysis of these vesicles in the context of disease allow us to envision prospects to improve the detection and treatment of hematological malignancies.
Collapse
Affiliation(s)
- David Van Morckhoven
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nathan Dubois
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Departement of Hematology, Jules Bordet Institute, Brussels, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Departement of Hematology, Jules Bordet Institute, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Basile Stamatopoulos
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
2
|
Allam S, Nasr K, Khalid F, Shah Z, Khan Suheb MZ, Mulla S, Vikash S, Bou Zerdan M, Anwer F, Chaulagain CP. Liquid biopsies and minimal residual disease in myeloid malignancies. Front Oncol 2023; 13:1164017. [PMID: 37213280 PMCID: PMC10196237 DOI: 10.3389/fonc.2023.1164017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/25/2023] [Indexed: 05/23/2023] Open
Abstract
Minimal residual disease (MRD) assessment through blood component sampling by liquid biopsies (LBs) is increasingly being investigated in myeloid malignancies. Blood components then undergo molecular analysis by flow cytometry or sequencing techniques and can be used as a powerful tool for prognostic and predictive purposes in myeloid malignancies. There is evidence and more is evolving about the quantification and identification of cell-based and gene-based biomarkers in myeloid malignancies to monitor treatment response. MRD based acute myeloid leukemia protocol and clinical trials are currently incorporating LB testing and preliminary results are encouraging for potential widespread use in clinic in the near future. MRD monitoring using LBs are not standard in myelodysplastic syndrome (MDS) but this is an area of active investigation. In the future, LBs can replace more invasive techniques such as bone marrow biopsies. However, the routine clinical application of these markers continues to be an issue due to lack of standardization and limited number of studies investigating their specificities. Integrating artificial intelligence (AI) could help simplify the complex interpretation of molecular testing and reduce errors related to operator dependency. Though the field is rapidly evolving, the applicability of MRD testing using LB is mostly limited to research setting at this time due to the need for validation, regulatory approval, payer coverage, and cost issues. This review focuses on the types of biomarkers, most recent research exploring MRD and LB in myeloid malignancies, ongoing clinical trials, and the future of LB in the setting of AI.
Collapse
Affiliation(s)
- Sabine Allam
- Department of Medicine and Medical Sciences, University of Balamand, Dekwaneh, Lebanon
| | - Kristina Nasr
- Department of Medicine and Medical Sciences, University of Balamand, Dekwaneh, Lebanon
| | - Farhan Khalid
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ, United States
| | - Zunairah Shah
- Department of Internal Medicine, Weiss Memorial Hospital, Chicago, IL, United States
| | | | - Sana Mulla
- Department of Internal Medicine, St Mary’s Medical Center, Apple Valley, CA, United States
| | - Sindhu Vikash
- Department of Medicine, Jacobi Medical center/AECOM Bronx, Bronx, NY, United States
| | - Maroun Bou Zerdan
- Department of Internal Medicine, SUNY Upstate Medical University, New York, NY, United States
| | - Faiz Anwer
- Department of Hematology and Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, United States
| | - Chakra P. Chaulagain
- Department of Hematology and Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, United States
- *Correspondence: Chakra P. Chaulagain,
| |
Collapse
|
3
|
Extracellular Vesicles in Haematological Disorders: A Friend or a Foe? Int J Mol Sci 2022; 23:ijms231710118. [PMID: 36077514 PMCID: PMC9455998 DOI: 10.3390/ijms231710118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Extracellular vesicles (EVs) have emerged as important mediators of homeostasis, immune modulation and intercellular communication. They are released by every cell of the human body and accordingly detected in a variety of body fluids. Interestingly, their expression can be upregulated under various conditions, such as stress, hypoxia, irradiation, inflammation, etc. Their cargo, which is variable and may include lipids, proteins, RNAs and DNA, reflects that of the parental cell, which offers a significant diagnostic potential to EVs. In line with this, an increasing number of studies have reported the important contribution of cancer-derived EVs in altering the tumour microenvironment and allowing for cancer progression and metastasis. As such, cancer-derived EVs may be used to monitor the development and progression of disease and to evaluate the potential response to treatment, which has generated much excitement in the field of oncology and particularly in haemato-oncology. Finally, EVs are able to transfer their cargo to target cells, modifying the properties of the recipient cell, which offers great therapeutic potential for EVs (either by specific drug delivery or by delivery of siRNAs and other inhibitory proteins). In this manuscript, we review the potential diagnostic use and therapeutic options of EVs in the context of haematological malignancies.
Collapse
|
4
|
Karantanou C, Minciacchi VR, Karantanos T. Extracellular Vesicles in Myeloid Neoplasms. Int J Mol Sci 2022; 23:ijms23158827. [PMID: 35955960 PMCID: PMC9369333 DOI: 10.3390/ijms23158827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Myeloid neoplasms arise from malignant primitive cells, which exhibit growth advantage within the bone marrow microenvironment (BMM). The interaction between these malignant cells and BMM cells is critical for the progression of these diseases. Extracellular vesicles (EVs) are lipid bound vesicles secreted into the extracellular space and involved in intercellular communication. Recent studies have described RNA and protein alterations in EVs isolated from myeloid neoplasm patients compared to healthy controls. The altered expression of various micro-RNAs is the best-described feature of EVs of these patients. Some of these micro-RNAs induce growth-related pathways such as AKT/mTOR and promote the acquisition of stem cell-like features by malignant cells. Another well-described characteristic of EVs in myeloid neoplasms is their ability to suppress healthy hematopoiesis either via direct effect on healthy CD34+ cells or via alteration of the differentiation of BMM cells. These results support a role of EVs in the pathogenesis of myeloid neoplasms. mainly through mediating the interaction between malignant and BMM cells, and warrant further study to better understand their biology. In this review, we describe the reported alterations of EV composition in myeloid neoplasms and the recent discoveries supporting their involvement in the development and progression of these diseases.
Collapse
Affiliation(s)
- Christina Karantanou
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
| | - Valentina René Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
| | - Theodoros Karantanos
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21218, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, The Bunting-Blaustein Cancer Research Building, 1650 Orleans Street, Baltimore, MD 21218, USA
- Correspondence:
| |
Collapse
|
5
|
Stone AP, Nikols E, Freire D, Machlus KR. The pathobiology of platelet and megakaryocyte extracellular vesicles: A (c)lot has changed. J Thromb Haemost 2022; 20:1550-1558. [PMID: 35506218 DOI: 10.1111/jth.15750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022]
Abstract
Platelet-derived extracellular vesicles (PEVs) were originally studied for their potential as regulators of coagulation, a function redundant with that of their parent cells. However, as the understanding of the diverse roles of platelets in hemostasis and disease has developed, so has the understanding of PEVs. In addition, the more recent revelation of constitutively released megakaryocyte-derived extracellular vesicles (MKEVs) in circulation provides an interesting counterpoint and avenue for investigation. In this review, we highlight the historical link of PEVs to thrombosis and hemostasis and provide critical updates. We also expand our discussion to encompass the roles that distinguish PEVs and MKEVs from their parent cells. Furthermore, the role of extracellular vesicles in disease pathology, both as biomarkers and as exacerbators, has been of great interest in recent years. We highlight some of the key roles that PEVs and MKEVs play in autoimmune blood cell disorders, liver pathology, and cardiovascular disease. We then look at the future of PEVs and MKEVs as candidates for novel therapeutics.
Collapse
Affiliation(s)
- Andrew P Stone
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Emma Nikols
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Daniela Freire
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Kellie R Machlus
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Barone M, Barone M, Ricci F, Auteri G, Corradi G, Fabbri F, Papa V, Bandini E, Cenacchi G, Tazzari PL, Vianelli N, Turroni S, Cavo M, Palandri F, Candela M, Catani L. An Abnormal Host/Microbiomes Signature of Plasma-Derived Extracellular Vesicles Is Associated to Polycythemia Vera. Front Oncol 2021; 11:715217. [PMID: 34900671 PMCID: PMC8657945 DOI: 10.3389/fonc.2021.715217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/08/2021] [Indexed: 01/04/2023] Open
Abstract
Polycythemia Vera (PV) is a myeloproliferative neoplasm with increased risk of thrombosis and progression to myelofibrosis. Chronic inflammation is commonly observed in myeloproliferative neoplasms including PV. The inflammatory network includes the extracellular vesicles (EVs), which play a role in cell-cell communication. Recent evidence points to circulating microbial components/microbes as potential players in hemopoiesis regulation. To address the role of EVs in PV, here we investigated phenotype and microbial DNA cargo of circulating EVs through multidimensional analysis. Peripheral blood and feces were collected from PV patients (n=38) and healthy donors (n=30). Circulating megakaryocyte (MK)- and platelet (PLT)-derived EVs were analyzed by flow cytometry. After microbial DNA extraction from feces and isolated EVs, the 16S rDNA V3-V4 region was sequenced. We found that the proportion of circulating MK-derived EVs was significantly decreased in PV patients as compared with the healthy donors. By contrast, the proportion of the PLT-derived EVs was increased. Interestingly, PV was also associated with a microbial DNA signature of the isolated EVs with higher diversity and distinct microbial composition than the healthy counterparts. Of note, increased proportion of isolated lipopolysaccharide-associated EVs has been demonstrated in PV patients. Conversely, the gut microbiome profile failed to identify a distinct layout between PV patients and healthy donors. In conclusion, PV is associated with circulating EVs harbouring abnormal phenotype and dysbiosis signature with a potential role in the (inflammatory) pathogenesis of the disease.
Collapse
Affiliation(s)
- Monica Barone
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Martina Barone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Francesca Ricci
- Servizio di Immunoematologia e Trasfusionale, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giuseppe Auteri
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Giulia Corradi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Papa
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Erika Bandini
- Biosciences Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Pier Luigi Tazzari
- Servizio di Immunoematologia e Trasfusionale, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Nicola Vianelli
- Istituto di Ematologia "Seràgnoli", Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Michele Cavo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Francesca Palandri
- Istituto di Ematologia "Seràgnoli", Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Lucia Catani
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| |
Collapse
|
7
|
A Specific Host/Microbial Signature of Plasma-Derived Extracellular Vesicles Is Associated to Thrombosis and Marrow Fibrosis in Polycythemia Vera. Cancers (Basel) 2021; 13:cancers13194968. [PMID: 34638452 PMCID: PMC8507916 DOI: 10.3390/cancers13194968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Patients with polycythemia vera, a myeloproliferative neoplasm, are at increased risk of thrombosis and progression to myelofibrosis. However, no disease-specific risk factors have been identified so far. Extracellular vesicles, released from a broad variety of cells, are receiving increasing attention for their effects on cell-to-cell communication. In addition, they play a role in cancer and thrombosis. Interestingly, circulating microbial components/microbes have been recently indicated as potential modifiers of inflammation and coagulation. Here, we identified a signature of thrombosis history and marrow fibrosis by analyzing the phenotype and the microbial DNA cargo of the circulating extracellular vesicles after isolation from the plasma of patients with polycythemia vera. These data may support the role of extracellular vesicles as liquid biomarkers of aggressive disease, thus contributing to refining the prognosis of polycythemia vera. Abstract Polycythemia vera is a myeloproliferative neoplasm with increased risk of thrombosis and progression to myelofibrosis. However, no disease-specific risk factors have been identified so far. Circulating extracellular vesicles (EVs) are mostly of megakaryocyte (MK-EVs) and platelet (PLT-EVs) origin and, along with phosphatidylethanolamine (PE)-EVs, play a role in cancer and thrombosis. Interestingly, circulating microbial components/microbes have been recently indicated as potential modifiers of inflammation and coagulation. Here, we investigated phenotype and microbial DNA cargo of EVs after isolation from the plasma of 38 patients with polycythemia vera. Increased proportion of MK-EVs and reduced proportion of PLT-EVs identify patients with thrombosis history. Interestingly, EVs from patients with thrombosis history were depleted in Staphylococcus DNA but enriched in DNA from Actinobacteria members as well as Anaerococcus. In addition, patients with thrombosis history had also lower levels of lipopolysaccharide-associated EVs. In regard to fibrosis, along with increased proportion of PE-EVs, the EVs of patients with marrow fibrosis were enriched in DNA from Collinsella and Flavobacterium. Here, we identified a polycythemia-vera-specific host/microbial EV-based signature associated to thrombosis history and marrow fibrosis. These data may contribute to refining PV prognosis and to identifying novel druggable targets.
Collapse
|
8
|
Noh JY. Megakaryopoiesis and Platelet Biology: Roles of Transcription Factors and Emerging Clinical Implications. Int J Mol Sci 2021; 22:ijms22179615. [PMID: 34502524 PMCID: PMC8431765 DOI: 10.3390/ijms22179615] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
Platelets play a critical role in hemostasis and thrombus formation. Platelets are small, anucleate, and short-lived blood cells that are produced by the large, polyploid, and hematopoietic stem cell (HSC)-derived megakaryocytes in bone marrow. Approximately 3000 platelets are released from one megakaryocyte, and thus, it is important to understand the physiologically relevant mechanism of development of mature megakaryocytes. Many genes, including several key transcription factors, have been shown to be crucial for platelet biogenesis. Mutations in these genes can perturb megakaryopoiesis or thrombopoiesis, resulting in thrombocytopenia. Metabolic changes owing to inflammation, ageing, or diseases such as cancer, in which platelets play crucial roles in disease development, can also affect platelet biogenesis. In this review, I describe the characteristics of platelets and megakaryocytes in terms of their differentiation processes. The role of several critical transcription factors have been discussed to better understand the changes in platelet biogenesis that occur during disease or ageing.
Collapse
Affiliation(s)
- Ji-Yoon Noh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| |
Collapse
|
9
|
The Power of Extracellular Vesicles in Myeloproliferative Neoplasms: "Crafting" a Microenvironment That Matters. Cells 2021; 10:cells10092316. [PMID: 34571965 PMCID: PMC8464728 DOI: 10.3390/cells10092316] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Myeloproliferative Neoplasms (MPN) are acquired clonal disorders of the hematopoietic stem cells and include Essential Thrombocythemia, Polycythemia Vera and Myelofibrosis. MPN are characterized by mutations in three driver genes (JAK2, CALR and MPL) and by a state of chronic inflammation. Notably, MPN patients experience increased risk of thrombosis, disease progression, second neoplasia and evolution to acute leukemia. Extracellular vesicles (EVs) are a heterogeneous population of microparticles with a role in cell-cell communication. The EV-mediated cross-talk occurs via the trafficking of bioactive molecules such as nucleic acids, proteins, metabolites and lipids. Growing interest is focused on EVs and their potential impact on the regulation of blood cancers. Overall, EVs have been suggested to orchestrate the complex interplay between tumor cells and the microenvironment with a pivotal role in "education" and "crafting" of the microenvironment by regulating angiogenesis, coagulation, immune escape and drug resistance of tumors. This review is focused on the role of EVs in MPN. Specifically, we will provide an overview of recent findings on the involvement of EVs in MPN pathogenesis and discuss opportunities for their potential application as diagnostic and prognostic biomarkers.
Collapse
|
10
|
Găman MA, Cozma MA, Dobrică EC, Crețoiu SM, Găman AM, Diaconu CC. Liquid Biopsy and Potential Liquid Biopsy-Based Biomarkers in Philadelphia-Negative Classical Myeloproliferative Neoplasms: A Systematic Review. Life (Basel) 2021; 11:life11070677. [PMID: 34357048 PMCID: PMC8304270 DOI: 10.3390/life11070677] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are rare, clonal disorders of the hematopoietic stem cell in which an uncontrolled proliferation of terminally differentiated myeloid cells is noted. Polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) are included in the category of Philadelphia-negative, so-called classical MPNs. The potential applications of liquid biopsy and liquid biopsy-based biomarkers have not been explored in MPNs until now. Thus, a systematic search was computed in PubMed/MEDLINE, Web of Science and The Cochrane Library and, in total, 198 potentially relevant papers were detected. Following the removal of duplicates (n = 85), 113 records were screened. After the exclusion of irrelevant manuscripts based on the screening of their titles and abstracts (n = 81), we examined the full texts of 33 manuscripts. Finally, after we applied the exclusion and inclusion criteria, 27 original articles were included in this review. Overall, the data analyzed in this review point out that liquid biopsy and liquid biopsy-based biomarkers (cell-free DNA, extracellular vesicles, microparticles, circulating endothelial cells) could be used in MPNs for diagnostic and prognostic purposes. Future research is needed to clarify whether this technique can be employed to differentiate between MPN subtypes and secondary causes of erythrocytosis, thrombocytosis and myelofibrosis, as well as to predict the development of thrombosis.
Collapse
Affiliation(s)
- Mihnea-Alexandru Găman
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Correspondence: (M.-A.G.); (S.M.C.)
| | - Matei-Alexandru Cozma
- Department of Gastroenterology, Colentina Clinical Hospital, 020125 Bucharest, Romania;
| | - Elena-Codruța Dobrică
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (E.-C.D.); (A.M.G.)
- Department of Dermatology, “Elias” University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Crețoiu
- Department of Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Correspondence: (M.-A.G.); (S.M.C.)
| | - Amelia Maria Găman
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (E.-C.D.); (A.M.G.)
- Clinic of Hematology, Filantropia City Hospital, 200143 Craiova, Romania
| | - Camelia Cristina Diaconu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| |
Collapse
|
11
|
Forte D, Barone M, Palandri F, Catani L. The "Vesicular Intelligence" Strategy of Blood Cancers. Genes (Basel) 2021; 12:genes12030416. [PMID: 33805807 PMCID: PMC7999060 DOI: 10.3390/genes12030416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Blood cancers are a heterogeneous group of disorders including leukemia, multiple myeloma, and lymphoma. They may derive from the clonal evolution of the hemopoietic stem cell compartment or from the transformation of progenitors with immune potential. Extracellular vesicles (EVs) are membrane-bound nanovesicles which are released by cells into body fluids with a role in intercellular communication in physiology and pathology, including cancer. EV cargos are enriched in nucleic acids, proteins, and lipids, and these molecules can be delivered to target cells to influence their biological properties and modify surrounding or distant targets. In this review, we will describe the “smart strategy” on how blood cancer-derived EVs modulate tumor cell development and maintenance. Moreover, we will also depict the function of microenvironment-derived EVs in blood cancers and discuss how the interplay between tumor and microenvironment affects blood cancer cell growth and spreading, immune response, angiogenesis, thrombogenicity, and drug resistance. The potential of EVs as non-invasive biomarkers will be also discussed. Lastly, we discuss the clinical application viewpoint of EVs in blood cancers. Overall, blood cancers apply a ‘vesicular intelligence’ strategy to spread signals over their microenvironment, promoting the development and/or maintenance of the malignant clone.
Collapse
Affiliation(s)
- Dorian Forte
- IRCCS Azienda Ospedaliero—Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy; (D.F.); (M.B.)
| | - Martina Barone
- IRCCS Azienda Ospedaliero—Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy; (D.F.); (M.B.)
| | - Francesca Palandri
- IRCCS Azienda Ospedaliero—Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy
- Correspondence: (F.P.); (L.C.); Tel.: +39-5121-43044 (F.P.); +39-5121-43837 (L.C.)
| | - Lucia Catani
- IRCCS Azienda Ospedaliero—Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy; (D.F.); (M.B.)
- IRCCS Azienda Ospedaliero—Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy
- Correspondence: (F.P.); (L.C.); Tel.: +39-5121-43044 (F.P.); +39-5121-43837 (L.C.)
| |
Collapse
|
12
|
Bernardi S, Farina M. Exosomes and Extracellular Vesicles in Myeloid Neoplasia: The Multiple and Complex Roles Played by These " Magic Bullets". BIOLOGY 2021; 10:biology10020105. [PMID: 33540594 PMCID: PMC7912829 DOI: 10.3390/biology10020105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Extracellular vesicles (EVs) are released by the majority of cell types and can be isolated from both cell cultures and body fluids. They are involved in cell-to-cell communication and may shuttle different messages (RNA, DNA, and proteins). These messages are known to influence the microenvironment of cells and their behavior. In recent years, some evidence about the involvement of EVs and exosomes, an EV subgroup, in immunomodulation, the transfer of disease markers, and the treatment of myeloid malignancies have been reported. Little is known about these vesicles in this particular setting of hematologic neoplasia; here, we summarize and critically review the available results, aiming to encourage further investigations. Abstract Extracellular vesicles (exosomes, in particular) are essential in multicellular organisms because they mediate cell-to-cell communication via the transfer of secreted molecules. They are able to shuttle different cargo, from nucleic acids to proteins. The role of exosomes has been widely investigated in solid tumors, which gave us surprising results about their potential involvement in pathogenesis and created an opening for liquid biopsies. Less is known about exosomes in oncohematology, particularly concerning the malignancies deriving from myeloid lineage. In this review, we aim to present an overview of immunomodulation and the microenvironment alteration mediated by exosomes released by malicious myeloid cells. Afterwards, we review the studies reporting the use of exosomes as disease biomarkers and their influence in response to treatment, together with the recent experiences that have focused on the use of exosomes as therapeutic tools. The further development of new technologies and the increased knowledge of biological (exosomes) and clinical (myeloid neoplasia) aspects are expected to change the future approaches to these malignancies.
Collapse
Affiliation(s)
- Simona Bernardi
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- Correspondence: or ; Tel.: +39-0303998464
| | - Mirko Farina
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| |
Collapse
|
13
|
Forte D, Barone M, Morsiani C, Simonetti G, Fabbri F, Bruno S, Bandini E, Sollazzo D, Collura S, Deregibus MC, Auteri G, Ottaviani E, Vianelli N, Camussi G, Franceschi C, Capri M, Palandri F, Cavo M, Catani L. Distinct profile of CD34 + cells and plasma-derived extracellular vesicles from triple-negative patients with Myelofibrosis reveals potential markers of aggressive disease. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:49. [PMID: 33522952 PMCID: PMC7849077 DOI: 10.1186/s13046-020-01776-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
Background Myelofibrosis (MF) is a clonal disorder of hemopoietic stem/progenitor cells (HSPCs) with high prevalence in elderly patients and mutations in three driver genes (JAK2, MPL, or CALR). Around 10–15% of patients are triple-negative (TN) for the three driver mutations and display significantly worse survival. Circulating extracellular vesicles (EVs) play a role in intercellular signaling and are increased in inflammation and cancer. To identify a biomolecular signature of TN patients, we comparatively evaluated the circulating HSPCs and their functional interplay with the microenvironment focusing on EV analysis. Methods Peripheral blood was collected from MF patients (n = 29; JAK2V617F mutation, n = 23; TN, n = 6) and healthy donors (HD, n = 10). Immunomagnetically isolated CD34+ cells were characterized by gene expression profiling analysis (GEP), survival, migration, and clonogenic ability. EVs were purified from platelet-poor plasma by ultracentrifugation, quantified using the Nanosight technology and phenotypically characterized by flow cytometry together with microRNA expression. Migration and survival of CD34+ cells from patients were also analyzed after in vitro treatments with selected inflammatory factors, i.e. (Interleukin (IL)-1β, Tumor Necrosis Factor (TNF)-α, IL6) or after co-culture with EVs from MF patients/HD. Results The absolute numbers of circulating CD34+ cells were massively increased in TN patients. We found that TN CD34+ cells show in vitro defective functions and are unresponsive to the inflammatory microenvironment. Of note, the plasma levels of crucial inflammatory cytokines are mostly within the normal range in TN patients. Compared to JAK2V617F-mutated patients, the GEP of TN CD34+ cells revealed distinct signatures in key pathways such as survival, cell adhesion, and inflammation. Importantly, we observed the presence of mitochondrial components within plasma EVs and a distinct phenotype in TN-derived EVs compared to the JAK2V617F-mutated MF patients and HD counterparts. Notably, TN EVs promoted the survival of TN CD34+ cells. Along with a specific microRNA signature, the circulating EVs from TN patients are enriched with miR-361-5p. Conclusions Distinct EV-driven signals from the microenvironment are capable to promote the TN malignant hemopoiesis and their further investigation paves the way toward novel therapeutic approaches for rare MF. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-020-01776-8.
Collapse
Affiliation(s)
- Dorian Forte
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy. .,Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italy.
| | - Martina Barone
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy.,Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italy
| | - Cristina Morsiani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Giorgia Simonetti
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Francesco Fabbri
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Samantha Bruno
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy.,Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italy
| | - Erika Bandini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Daria Sollazzo
- Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italy
| | - Salvatore Collura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Maria Chiara Deregibus
- Department of Internal Medicine, Centre for Molecular Biotechnology and Centre for Research in Experimental Medicine, Torino, Italy
| | - Giuseppe Auteri
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy.,Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italy
| | - Emanuela Ottaviani
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Nicola Vianelli
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Giovanni Camussi
- Department of Internal Medicine, Centre for Molecular Biotechnology and Centre for Research in Experimental Medicine, Torino, Italy
| | - Claudio Franceschi
- Laboratory of Systems Medicine of Healthy Aging and Department of Applied Mathematics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Francesca Palandri
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Michele Cavo
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy.,Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italy
| | - Lucia Catani
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy.,Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italy
| |
Collapse
|
14
|
Polverelli N, Elli EM, Abruzzese E, Palumbo GA, Benevolo G, Tiribelli M, Bonifacio M, Tieghi A, Caocci G, D'Adda M, Bergamaschi M, Binotto G, Heidel FH, Cavazzini F, Crugnola M, Pugliese N, Bosi C, Isidori A, Bartoletti D, Auteri G, Latagliata R, Gandolfi L, Martino B, Scaffidi L, Cattaneo D, D'Amore F, Trawinska MM, Stella R, Markovic U, Catani L, Pane F, Cuneo A, Krampera M, Semenzato G, Lemoli RM, Vianelli N, Breccia M, Russo D, Cavo M, Iurlo A, Palandri F. Second primary malignancy in myelofibrosis patients treated with ruxolitinib. Br J Haematol 2020; 193:356-368. [PMID: 33222197 DOI: 10.1111/bjh.17192] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022]
Abstract
Ruxolitinib (RUX), the first JAK1/JAK2 inhibitor approved for myelofibrosis (MF) therapy, has recently been associated with the occurrence of second primary malignancies (SPMs), mainly lymphomas and non-melanoma skin cancers (NMSCs). We analyzed the incidence, risk factors and outcome of SPMs in 700 MF patients treated with RUX in a real-world context. Median follow-up from starting RUX was 2·9 years. Overall, 80 (11·4%) patients developed 87 SPMs after RUX start. NMSCs were the most common SPMs (50·6% of the cases). Multivariate analysis demonstrated that male sex [hazard ratio (HR): 2·37, 95% confidence interval (95%CI): 1·22-4·60, P = 0·01] and thrombocytosis> 400 × 109 /l at RUX start (HR:1·98, 95%CI: 1·10-4·60, P = 0·02) were associated with increased risk for SPMs. Risk factors for NMSC alone were male sex (HR: 3·14, 95%CI: 1·24-7·92, P = 0·02) and duration of hydroxycarbamide and RUX therapy > 5 years (HR: 3·20, 95%CI: 1·17-8·75, P = 0·02 and HR: 2·93, 95%CI: 1·39-6·17, P = 0·005 respectively). In SPMs excluding NMSCs, male sex (HR: 2·41, 95%CI: 1·11-5·25, P = 0·03), platelet > 400 × 109 /l (HR: 3·30, 95%CI: 1·67-6·50, P = 0·001) and previous arterial thromboses (HR: 3·47, 95%CI: 1·48-8·14, P = 0·004) were shown to be associated with higher risk of SPMs. While it is reassuring that no aggressive lymphoma was documented, active skin surveillance is recommended in all patients and particularly after prolonged hydroxycaramide therapy; oncological screening should be triggered by thrombocytosis and arterial thrombosis, particularly in males.
Collapse
Affiliation(s)
- Nicola Polverelli
- Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Elena M Elli
- Haematology Division, San Gerardo Hospital, ASST Monza, Monza, Italy
| | | | - Giuseppe A Palumbo
- Department of Scienze Mediche, Chirurgiche e Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Giulia Benevolo
- Division of Haematology, Città della Salute e della Scienza Hospital, Torino, Italy
| | - Mario Tiribelli
- Division of Haematology and BMT, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | | | - Alessia Tieghi
- Department of Haematology, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giovanni Caocci
- Department of Medical Sciences and Public Health, Haematology Unit, University of Cagliari, Cagliari, Italy
| | - Mariella D'Adda
- Division of Haematology, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Micaela Bergamaschi
- Department of Internal Medicine (DiMI), Clinic of Haematology, IRCCS AOU San Martino-IST, Genova, Italy
| | - Gianni Binotto
- Unit of Haematology and Clinical Immunology, University of Padova, Padova, Italy
| | - Florian H Heidel
- Internal Medicine II, Haematology and Oncology, Friedrich-Schiller-University Medical Center, Jena, Germany
| | | | - Monica Crugnola
- Division of Haematology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Novella Pugliese
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Costanza Bosi
- Division of Haematology, AUSL di Piacenza, Piacenza, Italy
| | - Alessandro Isidori
- Haematology and Stem Cell Transplant Center Marche Nord Hospital, Pesaro, Italy
| | - Daniela Bartoletti
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Giuseppe Auteri
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Roberto Latagliata
- Division of Cellular Biotechnologies and Haematology, University Sapienza, Roma, Italy
| | - Lisa Gandolfi
- Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Bruno Martino
- Division of Haematology, Azienda Ospedaliera "Bianchi Melacrino Morelli", Reggio Calabria, Italy
| | - Luigi Scaffidi
- Department of Medicine, Section of Haematology, University of Verona, Verona, Italy
| | - Daniele Cattaneo
- Haematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio D'Amore
- Unit of Haematology and Clinical Immunology, University of Padova, Padova, Italy
| | | | - Rossella Stella
- Division of Haematology and BMT, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Uros Markovic
- Division of Haematology, AOU Policlinico-Vittorio Emanuele, University of Catania, Catania, Italy
| | - Lucia Catani
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Fabrizio Pane
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Antonio Cuneo
- Division of Haematology, University of Ferrara, Ferrara, Italy
| | - Mauro Krampera
- Department of Medicine, Section of Haematology, University of Verona, Verona, Italy
| | - Gianpietro Semenzato
- Unit of Haematology and Clinical Immunology, University of Padova, Padova, Italy
| | - Roberto M Lemoli
- Department of Internal Medicine (DiMI), Clinic of Haematology, IRCCS AOU San Martino-IST, Genova, Italy
| | - Nicola Vianelli
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Massimo Breccia
- Division of Cellular Biotechnologies and Haematology, University Sapienza, Roma, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Michele Cavo
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| | - Alessandra Iurlo
- Haematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Palandri
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, Bologna, Italy
| |
Collapse
|
15
|
Barone M, Catani L, Ricci F, Romano M, Forte D, Auteri G, Bartoletti D, Ottaviani E, Tazzari PL, Vianelli N, Cavo M, Palandri F. The role of circulating monocytes and JAK inhibition in the infectious-driven inflammatory response of myelofibrosis. Oncoimmunology 2020; 9:1782575. [PMID: 32923146 PMCID: PMC7458658 DOI: 10.1080/2162402x.2020.1782575] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myelofibrosis (MF) is characterized by chronic inflammation and hyper-activation of the JAK-STAT pathway. Infections are one of the main causes of morbidity/mortality. Therapy with Ruxolitinib (RUX), a JAK1/2 inhibitor, may further increase the infectious risk. Monocytes are critical players in inflammation/immunity through cytokine production and release of bioactive extracellular vesicles. However, the functional behavior of MF monocytes, particularly during RUX therapy, is still unclear. In this study, we found that monocytes from JAK2V617F-mutated MF patients show an altered expression of chemokine (CCR2, CXCR3, CCR5) and cytokine (TNF-α-R, IL10-R, IL1β-R, IL6-R) receptors. Furthermore, their ability to produce and secrete free and extracellular vesicles-linked cytokines (IL1β, TNF-α, IL6, IL10) under lipopolysaccharides (LPS) stimulation is severely impaired. Interestingly, monocytes from RUX-treated patients show normal level of chemokine, IL10, IL1β, and IL6 receptors together with a restored ability to produce intracellular and to secrete extracellular vesicles-linked cytokines after LPS stimulation. Conversely, RUX therapy does not normalize TNF-R1/2 receptors expression and the LPS-driven secretion of free pro/anti-inflammatory cytokines. Accordingly, upon LPS stimulation, in vitro RUX treatment of monocytes from MF patients increases their secretion of extracellular vesicles-linked cytokines but inhibits the secretion of free pro/anti-inflammatory cytokines. In conclusion, we demonstrated that in MF the infection-driven response of circulating monocytes is defective. Importantly, RUX promotes their infection-driven cytokine production suggesting that infections following RUX therapy may not be due to monocyte failure. These findings contribute to better interpreting the immune vulnerability of MF and to envisaging strategies to improve the infection-driven immune response.
Collapse
Affiliation(s)
- Martina Barone
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Lucia Catani
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Ricci
- Immunohematology and Blood Bank, Azienda Ospedaliero-Universitaria S. Orsola-Malpighi di Bologna, Bologna, Italy
| | - Marco Romano
- School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Dorian Forte
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giuseppe Auteri
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Daniela Bartoletti
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Emanuela Ottaviani
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pier Luigi Tazzari
- Immunohematology and Blood Bank, Azienda Ospedaliero-Universitaria S. Orsola-Malpighi di Bologna, Bologna, Italy
| | - Nicola Vianelli
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Michele Cavo
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Palandri
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
16
|
Marin Oyarzún CP, Heller PG. Platelets as Mediators of Thromboinflammation in Chronic Myeloproliferative Neoplasms. Front Immunol 2019; 10:1373. [PMID: 31258539 PMCID: PMC6587101 DOI: 10.3389/fimmu.2019.01373] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic myeloproliferative neoplasms (MPN) are stem cell disorders driven by mutations in JAK2, CALR, or MPL genes and characterized by myeloid proliferation and increased blood cell counts. They encompass three closely related conditions, including essential thrombocythemia, polycythemia vera, and primary myelofibrosis. Elevated levels of cytokines released by clonal and non-clonal cells generate a chronic proinflammatory state that contributes to disease pathogenesis. Thrombosis represents the most common cause of morbidity and mortality in MPN, although paradoxically, patients may also present with a bleeding diathesis. The mechanisms leading to thrombosis are complex and multiple and include increased blood cells together with qualitative abnormalities of red cells, leukocytes, and platelets that favor a prothrombotic activated phenotype. The functional interplay between blood cells, the clotting cascade, and dysfunctional endothelium contributes to hypercoagulability and this process is perpetuated by the effect of inflammatory cytokines. In addition to their well-known function in hemostasis, platelets contribute to innate immunity and inflammation and play a key role in MPN thromboinflammatory state. In vivo platelet activation leads to platelet aggregate formation and exposure of adhesion molecules which favor their interaction with activated neutrophils and monocytes leading to circulating platelet-leukocyte heterotypic aggregates. Platelets are recruited to the activated endothelium further enhancing the reciprocal activation of both cell types. Crosstalk between activated cells drives cytokine production, further fuelling the self-reinforcing thromboinflammatory loop. In addition, MPN platelets provide a procoagulant scaffold which triggers the coagulation cascade and platelet-derived microparticles amplify this response. Markers of platelet, leukocyte, endothelial and coagulation activation are increased in MPN patients although prospective studies are required to determine the potential value of these parameters for identifying patients at increased thrombotic risk. Thrombosis remains the main complication of MPN patients, with a high risk of recurrence despite adequate cytoreductive and antithrombotic treatment. Deeper insight into the mechanism favoring thrombosis development in this setting may lead to novel therapeutic approaches for MPN thrombosis. Considering the critical role of inflammation in the vascular risk, concomitant targeting of inflammatory pathways could potentially impact on primary or secondary prevention strategies.
Collapse
Affiliation(s)
- Cecilia P Marin Oyarzún
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), Institute of Medical Research (IDIM) "Dr. Alfredo Lanari", University of Buenos Aires, Buenos Aires, Argentina
| | - Paula G Heller
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), Institute of Medical Research (IDIM) "Dr. Alfredo Lanari", University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|