1
|
Delshad M, Sanaei MJ, Mohammadi MH, Sadeghi A, Bashash D. Exosomal Biomarkers: A Comprehensive Overview of Diagnostic and Prognostic Applications in Malignant and Non-Malignant Disorders. Biomolecules 2025; 15:587. [PMID: 40305328 PMCID: PMC12024574 DOI: 10.3390/biom15040587] [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: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Exosomes are small extracellular vesicles, ranging from 30 to 150 nm, that are essential in cell biology, mediating intercellular communication and serving as biomarkers due to their origin from cells. Exosomes as biomarkers for diagnosing various illnesses have gained significant investigation due to the high cost and invasive nature of current diagnostic procedures. Exosomes have a clear advantage in the diagnosis of diseases because they include certain signals that are indicative of the genetic and proteomic profile of the ailment. This feature gives them the potential to be useful liquid biopsies for real-time, noninvasive monitoring, enabling early cancer identification for the creation of individualized treatment plans. According to our analysis, the trend toward utilizing exosomes as diagnostic and prognostic tools has raised since 2012. In this regard, the proportion of malignant indications is higher compared with non-malignant ones. To be precise, exosomes have been used the most in gastrointestinal, thoracic, and urogenital cancers, along with cardiovascular, diabetic, breathing, infectious, and brain disorders. To the best of our knowledge, this is the first research to examine all registered clinical trials that look at exosomes as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan 1411718541, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| |
Collapse
|
2
|
Wylie C, Rowan R, Malinova D, Crawford L. Extracellular vesicles in multiple myeloma: pathogenesis and therapeutic application. FEBS J 2025. [PMID: 40205752 DOI: 10.1111/febs.70093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Multiple myeloma (MM), characterised by the clonal proliferation of plasma cells in the bone marrow, is the second most common haematological malignancy worldwide. Although there is now an impressive artillery of therapeutics to tackle this condition, resistance remains a prevalent issue. The bone marrow microenvironment performs a crucial role in supporting MM pathogenesis and promoting the development of therapeutic resistance. Extracellular vesicles (EVs), small vesicles that carry bioactive molecules, are a key component of cell-to-cell communication within the bone marrow microenvironment. In this review, we summarise the contribution of EVs to disease progression and anticancer treatment resistance and discuss the potential therapeutic applications of EVs in MM.
Collapse
Affiliation(s)
- Chloe Wylie
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, UK
| | - Rebecca Rowan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, UK
| | - Dessi Malinova
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, UK
| | - Lisa Crawford
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, UK
| |
Collapse
|
3
|
Mamgain G, Yadav SRM. Potential Effect of Extracellular Vesicles in Clinical Settings of Lymphoma. Indian J Clin Biochem 2025; 40:12-24. [PMID: 39835236 PMCID: PMC11741971 DOI: 10.1007/s12291-023-01156-x] [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: 12/26/2022] [Accepted: 09/21/2023] [Indexed: 01/22/2025]
Abstract
Liquid biopsy is gaining importance in oncology in the age of precision medicine. Extracellular vesicles (EVs), among other tumor-derived indicators, are isolated and analysed from bodily fluids. EVs are secreted by both healthy and cancerous cells and are lipid bilayer-enclosed particles that are diverse in size and molecular makeup. Since their quantity, phenotype, and molecular payload, which includes proteins, lipids, metabolites, and nucleic acids, mirror the nature and origin of parental cells, EVs are valuable transporters of cancer information in tumour context. This makes them interesting candidates for new biomarkers. Being closely linked to the parental cells in terms of composition, quantity, and roles is a crucial aspect of EVs. Multiple studies have shown the crucial part tumor-derived EVs plays in the development of cancer, and this subject is currently a hot one in the field of oncology. The clinical applications of EVs-based technology that are currently being tested in the areas of biomarkers, therapeutic targets, immune evasion tools, biologically designed immunotherapies, vaccines, neutralising approaches, targeting biogenesis, and extracorporeal removal were the main focus of this review. However, more bioengineering refinement is needed to address clinical and commercial limitations. The introduction of these new potential diagnostic tools into clinical practise has the potential to profoundly revolutionise the cancer field, primarily for solid tumours but also for haematological neoplasms. The development of EV-based therapies will be facilitated by improvements in EV engineering methodology and design, transforming the current pharmaceutical environment.
Collapse
Affiliation(s)
- Garima Mamgain
- Department of Medical Oncology, All India Institute of Medical Sciences Rishikesh, Rishikesh, 249203 India
| | - Shashi Ranjan Mani Yadav
- Department of Biochemistry, All India Institute of Medical Sciences Rishikesh, Rishikesh, 249203 India
| |
Collapse
|
4
|
Kale V. Extracellular vesicles as standard-of-care therapy: will fast-tracking the regulatory processes help achieve the goal? Regen Med 2024; 19:617-635. [PMID: 39688586 PMCID: PMC11730413 DOI: 10.1080/17460751.2024.2442847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024] Open
Abstract
Extracellular Vesicles (EVs) became a focus of clinical research when experimental and pre-clinical studies showed that they mimic their parent cells' regenerative and therapeutic effects and their cargo carries disease-specific diagnostic and prognostic biomarkers. Since the publication of data forms an endpoint of the study, this review specifically focused on the published clinical trials done with EVs. For brevity, this review was restricted to the last 10 years. Unexpectedly, the literature search showed that very few clinical trials assessing the therapeutic applications of EVs were published in this period indicating that they have not reached their desired endpoint. Conversely, most studies showed the potential of EVs present in various biofluids as a promising source of diagnostic and prognostic biomarkers for various diseases, and predictive markers to assess the effectiveness of therapy. This stark difference in the numbers could perhaps be due to the time-consuming regulatory processes involved in the clinical-grade preparation and characterization of EVs, and the determination of their safety and effective dose regimens. One wonders whether fast-tracking regulatory affairs could help accelerate the therapeutic use of EVs. This aspect needs urgent attention.
Collapse
Affiliation(s)
- Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, India
| |
Collapse
|
5
|
Ghosh S, Dey A, Chakrabarti A, Bhuniya T, Indu N, Hait A, Chowdhury A, Paul A, Mahajan AA, Papadakis M, Alexiou A, Jha SK. The theragnostic advances of exosomes in managing leukaemia. J Cell Mol Med 2024; 28:e70052. [PMID: 39659020 PMCID: PMC11632122 DOI: 10.1111/jcmm.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/21/2024] [Accepted: 08/20/2024] [Indexed: 12/12/2024] Open
Abstract
Leukaemia, a group of haematological malignancies, presents ongoing diagnosis, prognosis, and treatment challenges. A major obstacle in treating this disease is the development of drug resistance. Overcoming drug resistance poses a significant barrier to effective leukaemia treatment. The emergence of exosome research has unveiled new insights into the probable theragnostic implementations in leukaemia. Various research has exhibited the diagnostic possibilities of exosomes in identifying leukaemia-specific biomarkers, including genetic mutations and fusion transcripts. Additionally, exosomes have been implicated in disease progression and treatment response, rendering them appealing targets for therapeutics. Exosomes, originating from diverse cell types, are instrumental in intercellular communication as they participate in the functional transportation of molecules like proteins, nucleic acids and lipids across space. Exosomes have a dual role in immune regulation, mediating immune suppression and modulating anti-leukaemia immune responses. Interestingly, exosomes can even act as drug transport vehicles. This review delves into the intricate process of exosome biogenesis, shedding light on their formation and release from donor cells. The key mechanisms engaged in exosome biogenesis, for instance, the endosomal sorting complexes required for transport (ESCRT) machinery and ESCRT-independent pathways, are thoroughly discussed. Looking ahead, future approaches that leverage innovative technologies hold the promise of revolutionizing disease management and improving patient outcomes.
Collapse
Affiliation(s)
- Subhrojyoti Ghosh
- Department of BiotechnologyIndian Institute of Technology MadrasChennaiTamil NaduIndia
| | - Anuvab Dey
- Department of Biosciences and BioengineeringIndian Institute of Technology GuwahatiGuwahatiAssamIndia
| | - Aneshwa Chakrabarti
- Department of Chemistry and Chemical BiologyIndian Institute of Technology, Indian School of Mines DhanbadDhanbadIndia
| | - Tiyasa Bhuniya
- Department of BiotechnologyNIT DurgapurDurgapurWest BengalIndia
| | - Neelparna Indu
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | - Anirban Hait
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | - Ankita Chowdhury
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | - Aritra Paul
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | | | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐HerdeckeWuppertalGermany
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Research & DevelopmentFunogenAthensGreece
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | | |
Collapse
|
6
|
Xiao S, Chen L, Chen Z, Li Q. Therapeutically Harnessing Tumor Cell-Derived Extracellular Vesicles for Multiple Myeloma: Recent Advances and Future Perspectives. Pharmaceutics 2024; 16:1439. [PMID: 39598562 PMCID: PMC11597712 DOI: 10.3390/pharmaceutics16111439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as pivotal regulators for extensive intercellular crosstalk owing to capsuled diverse bioactive substances such as proteins, nucleic acids, and lipids. Recent studies have shown that tumor-derived EVs significantly influence the bone marrow microenvironment, contributing to the progression of multiple myeloma (MM). This highlights the robust potential of EVs as a promising avenue for developing more effective and precise diagnostic and therapeutic strategies for MM. In this review, we briefly discuss the multifaceted roles of EVs in MM progression, as well as the diagnostic and therapeutic value in MM management. Specifically, we focus on the latest research progress regarding the therapeutic potential of EVs for MM, particularly tumor cell-derived EVs, as we elaborate on three main aspects: (i) EVs as therapeutic targets, including the targeted inhibition of EV biogenesis and uptake, and the possibility of eliminating tumor-derived EVs; (ii) EVs as delivery nanovectors, where we discuss the latest anti-MM candidates and potential ways to optimize therapeutic efficiency; and (iii) engineered EVs as antitumor vaccines, focusing on the use of tumor cell-derived EVs in immunotherapy. Finally, we address the prospects and challenges of harnessing the therapeutic potential of EVs in clinical transformation.
Collapse
Affiliation(s)
- Shumei Xiao
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Lei Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (L.C.); (Z.C.)
| | - Zhichao Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (L.C.); (Z.C.)
| | - Qiubai Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Hubei Engineering Research Center for Application of Extracellular Vesicles, Hubei University of Science and Technology, Xianning 437100, China
| |
Collapse
|
7
|
Fathi E, Valipour B, Jafari S, Kazemi A, Montazersaheb S, Farahzadi R. The role of the hematopoietic stem/progenitor cells-derived extracellular vesicles in hematopoiesis. Heliyon 2024; 10:e35051. [PMID: 39157371 PMCID: PMC11327835 DOI: 10.1016/j.heliyon.2024.e35051] [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: 10/12/2023] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are tightly regulated by specific microenvironments called niches to produce an appropriate number of mature blood cell types. Self-renewal and differentiation are two hallmarks of hematopoietic stem and progenitor cells, and their balance is critical for proper functioning of blood and immune cells throughout life. In addition to cell-intrinsic regulation, extrinsic cues within the bone marrow niche and systemic factors also affect the fate of HSCs. Despite this, many paracrine and endocrine factors that influence the function of hematopoietic cells remain unknown. In hematological malignancies, malignant cells remodel their niche into a permissive environment to enhance the survival of leukemic cells. These events are accompanied by loss of normal hematopoiesis. It is well known that extracellular vehicles (EVs) mediate intracellular interactions under physiological and pathological conditions. In other words, EVs transfer biological information to surrounding cells and contribute not only to physiological functions but also to the pathogenesis of some diseases, such as cancers. Therefore, a better understanding of cell-to-cell interactions may lead to identification of potential therapeutic targets. Recent reports have suggested that EVs are evolutionarily conserved constitutive mediators that regulate hematopoiesis. Here, we focus on the emerging roles of EVs in normal and pathological conditions, particularly in hematological malignancies. Owing to the high abundance of EVs in biological fluids, their potential use as biomarkers and therapeutic tools is discussed.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Behnaz Valipour
- Department of Basic Sciences and Health, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abdolhassan Kazemi
- Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Sousa GC, Carvalho MG, Fonseca-Alves CE, Souza FF. Serum Extracellular Vesicles Cargo Approach in Bitches with Mammary Tumors. Curr Issues Mol Biol 2024; 46:7745-7768. [PMID: 39057100 PMCID: PMC11275879 DOI: 10.3390/cimb46070459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
This study investigated serum extracellular vesicles (EVs) in bitches with mammary neoplasms, in order to understand their size, shape, and concentration, as well as their association with tumor malignancy. Thirty bitches were categorized into control (n = 10), mammary tumor grades I and II (GI, n = 13), and grade III (GII, n = 7). Serum was separated from blood collected during mastectomy, and EVs were isolated using size exclusion chromatography. The analysis revealed no significant differences in EV concentrations among groups, with similar concentrations for control, GI, and GII. Ninety-one proteins were identified in EV-enriched samples, with six showing varied abundance across groups. Notably, keratin 18 was highly abundant in GI, while sushi domain-containing protein, EvC ciliary subunit 2, and the joining chain of multimeric IgM and IgA were increased in GII. Additionally, protocadherin 17 and albumin were upregulated in both GI and GII. ROC curves identified potential biomarkers for differentiating tumor grades. Enrichment pathway analysis revealed AFP gene upregulation in the GI. Mass spectrometry proteomics data were deposited in Mendeley Data. The study provides valuable insights into serum EV characterization in bitches, suggesting keratin 18 and protocadherin 17 as potential biomarkers for canine mammary neoplasia, with implications for future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Gabriela C. Sousa
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University, Unesp, Botucatu 18618-687, São Paulo, Brazil; (G.C.S.); (M.G.C.); (C.E.F.-A.)
- Department of Small Animal Clinical Sciences, Virginia Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Marcos G. Carvalho
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University, Unesp, Botucatu 18618-687, São Paulo, Brazil; (G.C.S.); (M.G.C.); (C.E.F.-A.)
| | - Carlos E. Fonseca-Alves
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University, Unesp, Botucatu 18618-687, São Paulo, Brazil; (G.C.S.); (M.G.C.); (C.E.F.-A.)
| | - Fabiana F. Souza
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University, Unesp, Botucatu 18618-687, São Paulo, Brazil; (G.C.S.); (M.G.C.); (C.E.F.-A.)
| |
Collapse
|
9
|
Abduh MS. An overview of multiple myeloma: A monoclonal plasma cell malignancy's diagnosis, management, and treatment modalities. Saudi J Biol Sci 2024; 31:103920. [PMID: 38283805 PMCID: PMC10818257 DOI: 10.1016/j.sjbs.2023.103920] [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: 07/25/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Multiple Myeloma (MM) is a plasma cell cancer with high mortality and morbidity rates. Its incidence rate has increased by 143% since 1975. Adipokines, cytokines, chemokines, and genetic variations influence the development and progression of MM. Chromosomal translocations cause mutations associated with MM. The pathogenesis of MM is complicated by novel issues like miRNAs, RANKL, Wnt/DKK1, Wnt, and OPG. Conventional diagnosis methods include bone marrow biopsy, sPEP or uPEP, sIFE and uIFE, and sFLC assay, along with advanced techniques such as FISH, SNPA, and gene expression technologies. A novel therapeutic strategy has been developed recently. Chemotherapy, hematopoietic stem cell transplantation, and a variety of drug classes in combination are used to treat patients with high-risk diseases. Alkylating agents, PIs, and IMiDs have all been developed as effective treatment options for MM in recent years. This review overviews the current recommendations for managing MGUS, SMM, MM, SP and NSMM and discusses practices in diagnosing and treating MM.
Collapse
Affiliation(s)
- Maisa Siddiq Abduh
- Immune Responses in Different Diseases Research Group, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
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
|
11
|
Ismail NH, Mussa A, Al-Khreisat MJ, Mohamed Yusoff S, Husin A, Johan MF. Proteomic Alteration in the Progression of Multiple Myeloma: A Comprehensive Review. Diagnostics (Basel) 2023; 13:2328. [PMID: 37510072 PMCID: PMC10378430 DOI: 10.3390/diagnostics13142328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematologic malignancy. Most MM patients are diagnosed at a late stage because the early symptoms of the disease can be uncertain and nonspecific, often resembling other, more common conditions. Additionally, MM patients are commonly associated with rapid relapse and an inevitable refractory phase. MM is characterized by the abnormal proliferation of monoclonal plasma cells in the bone marrow. During the progression of MM, massive genomic alterations occur that target multiple signaling pathways and are accompanied by a multistep process involving differentiation, proliferation, and invasion. Moreover, the transformation of healthy plasma cell biology into genetically heterogeneous MM clones is driven by a variety of post-translational protein modifications (PTMs), which has complicated the discovery of effective treatments. PTMs have been identified as the most promising candidates for biomarker detection, and further research has been recommended to develop promising surrogate markers. Proteomics research has begun in MM, and a comprehensive literature review is available. However, proteomics applications in MM have yet to make significant progress. Exploration of proteomic alterations in MM is worthwhile to improve understanding of the pathophysiology of MM and to search for new treatment targets. Proteomics studies using mass spectrometry (MS) in conjunction with robust bioinformatics tools are an excellent way to learn more about protein changes and modifications during disease progression MM. This article addresses in depth the proteomic changes associated with MM disease transformation.
Collapse
Affiliation(s)
- Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, Omdurman P.O. Box 382, Sudan
| | - Mutaz Jamal Al-Khreisat
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Shafini Mohamed Yusoff
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Azlan Husin
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Muhammad Farid Johan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| |
Collapse
|
12
|
Das K, Mukherjee T, Shankar P. The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy. Biomolecules 2023; 13:897. [PMID: 37371477 DOI: 10.3390/biom13060897] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The tumor microenvironment (TME) plays an important role in the development and progression of hematological malignancies. In recent years, studies have focused on understanding how tumor cells communicate within the TME. In addition to several factors, such as growth factors, cytokines, extracellular matrix (ECM) molecules, etc., a growing body of evidence has indicated that extracellular vesicles (EVs) play a crucial role in the communication of tumor cells within the TME, thereby contributing to the pathogenesis of hematological malignancies. The present review focuses on how EVs derived from tumor cells interact with the cells in the TME, such as immune cells, stromal cells, endothelial cells, and ECM components, and vice versa, in the context of various hematological malignancies. EVs recovered from the body fluids of cancer patients often carry the bioactive molecules of the originating cells and hence can be considered new predictive biomarkers for specific types of cancer, thereby also acting as potential therapeutic targets. Here, we discuss how EVs influence hematological tumor progression via tumor-host crosstalk and their use as biomarkers for hematological malignancies, thereby benefiting the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Tanmoy Mukherjee
- Department of Pulmonary Immunology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Prem Shankar
- Department of Pulmonary Immunology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| |
Collapse
|
13
|
Punetha A, Kotiya D. Advancements in Oncoproteomics Technologies: Treading toward Translation into Clinical Practice. Proteomes 2023; 11:2. [PMID: 36648960 PMCID: PMC9844371 DOI: 10.3390/proteomes11010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Proteomics continues to forge significant strides in the discovery of essential biological processes, uncovering valuable information on the identity, global protein abundance, protein modifications, proteoform levels, and signal transduction pathways. Cancer is a complicated and heterogeneous disease, and the onset and progression involve multiple dysregulated proteoforms and their downstream signaling pathways. These are modulated by various factors such as molecular, genetic, tissue, cellular, ethnic/racial, socioeconomic status, environmental, and demographic differences that vary with time. The knowledge of cancer has improved the treatment and clinical management; however, the survival rates have not increased significantly, and cancer remains a major cause of mortality. Oncoproteomics studies help to develop and validate proteomics technologies for routine application in clinical laboratories for (1) diagnostic and prognostic categorization of cancer, (2) real-time monitoring of treatment, (3) assessing drug efficacy and toxicity, (4) therapeutic modulations based on the changes with prognosis and drug resistance, and (5) personalized medication. Investigation of tumor-specific proteomic profiles in conjunction with healthy controls provides crucial information in mechanistic studies on tumorigenesis, metastasis, and drug resistance. This review provides an overview of proteomics technologies that assist the discovery of novel drug targets, biomarkers for early detection, surveillance, prognosis, drug monitoring, and tailoring therapy to the cancer patient. The information gained from such technologies has drastically improved cancer research. We further provide exemplars from recent oncoproteomics applications in the discovery of biomarkers in various cancers, drug discovery, and clinical treatment. Overall, the future of oncoproteomics holds enormous potential for translating technologies from the bench to the bedside.
Collapse
Affiliation(s)
- Ankita Punetha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers University, 225 Warren St., Newark, NJ 07103, USA
| | - Deepak Kotiya
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 South Limestone St., Lexington, KY 40536, USA
| |
Collapse
|
14
|
Datta B, Dutta N, Ashish A, Mandal M, Shukla J, Suresh R, Choudhury P, Chaudhury K, Dutta G. Electrochemical Detection of Cancer Fingerprint: A Systematic Review on Recent Progress in Extracellular Vesicle Research from Lab to Market. NEXT-GENERATION NANOBIOSENSOR DEVICES FOR POINT-OF-CARE DIAGNOSTICS 2023:47-77. [DOI: 10.1007/978-981-19-7130-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
|
15
|
Menu E, Vanderkerken K. Exosomes in multiple myeloma: from bench to bedside. Blood 2022; 140:2429-2442. [PMID: 35271699 PMCID: PMC10653045 DOI: 10.1182/blood.2021014749] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/10/2022] [Accepted: 02/23/2022] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable plasma cell malignancy that develops in the bone marrow (BM). This BM is partially responsible for protecting the MM cells against current standard-of-care therapies and for accommodating MM-related symptoms such as bone resorption and immune suppression. Increasing evidence has implicated extracellular vesicles (EV), including exosomes in the different processes within the BM. Exosomes are <150-nm-sized vesicles secreted by different cell types including MM cells. These vesicles contain protein and RNA cargo that they deliver to the recipient cell. In this way, they have been implicated in MM-related processes including osteolysis, angiogenesis, immune suppression, and drug resistance. Targeting exosome secretion could therefore potentially block these different processes. In this review, we will summarize the current findings of exosome-related processes in the BM and describe not only the current treatment strategies to counter them but also how exosomes can be harnessed to deliver toxic payloads. Finally, an overview of the different clinical studies that investigate EV cargo as potential MM biomarkers in liquid biopsies will be discussed.
Collapse
Affiliation(s)
- Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
16
|
Bergantim R, Peixoto da Silva S, Polónia B, Barbosa MAG, Albergaria A, Lima J, Caires HR, Guimarães JE, Vasconcelos MH. Detection of Measurable Residual Disease Biomarkers in Extracellular Vesicles from Liquid Biopsies of Multiple Myeloma Patients-A Proof of Concept. Int J Mol Sci 2022; 23:13686. [PMID: 36430163 PMCID: PMC9690807 DOI: 10.3390/ijms232213686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Monitoring measurable residual disease (MRD) is crucial to assess treatment response in Multiple Myeloma (MM). Detection of MRD in peripheral blood (PB) by exploring Extracellular Vesicles (EVs), and their cargo, would allow frequent and minimally invasive monitoring of MM. This work aims to detect biomarkers of MRD in EVs isolated from MM patient samples at diagnosis and remission and compare the MRD-associated content between BM and PB EVs. EVs were isolated by size-exclusion chromatography, concentrated by ultrafiltration, and characterized according to their size and concentration, morphology, protein concentration, and the presence of EV-associated protein markers. EVs from healthy blood donors were used as controls. It was possible to isolate EVs from PB and BM carrying MM markers. Diagnostic samples had different levels of MM markers between PB and BM paired samples, but no differences between PB and BM were found at remission. EVs concentration was lower in the PB of healthy controls than of patients, and MM markers were mostly not detected in EVs from controls. This study pinpoints the potential of PB EVs from MM remission patients as a source of MM biomarkers and as a non-invasive approach for monitoring MRD.
Collapse
Affiliation(s)
- Rui Bergantim
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital Center of São João, 4200-319 Porto, Portugal
- Clinical Hematology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| | - Sara Peixoto da Silva
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Bárbara Polónia
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Mélanie A. G. Barbosa
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - André Albergaria
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Research Innovation Unit, Translational Research & Industry Partnerships Office, i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Jorge Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Research Innovation Unit, Translational Research & Industry Partnerships Office, i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Hugo R. Caires
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital Center of São João, 4200-319 Porto, Portugal
- Clinical Hematology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário IUCSESPU, 4585-116 Gandra-Paredes, Portugal
| | - M. Helena Vasconcelos
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
17
|
Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, Chen Y, Han B. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther 2022; 7:286. [PMID: 35963853 PMCID: PMC9376115 DOI: 10.1038/s41392-022-01110-y] [Citation(s) in RCA: 407] [Impact Index Per Article: 135.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023] Open
Abstract
Regulated cell death (RCD), also well-known as programmed cell death (PCD), refers to the form of cell death that can be regulated by a variety of biomacromolecules, which is distinctive from accidental cell death (ACD). Accumulating evidence has revealed that RCD subroutines are the key features of tumorigenesis, which may ultimately lead to the establishment of different potential therapeutic strategies. Hitherto, targeting the subroutines of RCD with pharmacological small-molecule compounds has been emerging as a promising therapeutic avenue, which has rapidly progressed in many types of human cancers. Thus, in this review, we focus on summarizing not only the key apoptotic and autophagy-dependent cell death signaling pathways, but the crucial pathways of other RCD subroutines, including necroptosis, pyroptosis, ferroptosis, parthanatos, entosis, NETosis and lysosome-dependent cell death (LCD) in cancer. Moreover, we further discuss the current situation of several small-molecule compounds targeting the different RCD subroutines to improve cancer treatment, such as single-target, dual or multiple-target small-molecule compounds, drug combinations, and some new emerging therapeutic strategies that would together shed new light on future directions to attack cancer cell vulnerabilities with small-molecule drugs targeting RCD for therapeutic purposes.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minru Liao
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shiou Zhu
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Yi Chen
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
18
|
Lopes R, Caetano J, Barahona F, Pestana C, Ferreira BV, Lourenço D, Queirós AC, Bilreiro C, Shemesh N, Beck HC, Carvalho AS, Matthiesen R, Bogen B, Costa-Silva B, Serre K, Carneiro EA, João C. Multiple Myeloma-Derived Extracellular Vesicles Modulate the Bone Marrow Immune Microenvironment. Front Immunol 2022; 13:909880. [PMID: 35874665 PMCID: PMC9302002 DOI: 10.3389/fimmu.2022.909880] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/23/2022] [Indexed: 01/10/2023] Open
Abstract
Multiple myeloma (MM), the third most frequent hematological cancer worldwide, is characterized by the proliferation of neoplastic plasma cells in the bone marrow (BM). One of the hallmarks of MM is a permissive BM microenvironment. Increasing evidence suggests that cell-to-cell communication between myeloma and immune cells via tumor cell-derived extracellular vesicles (EV) plays a key role in the pathogenesis of MM. Hence, we aimed to explore BM immune alterations induced by MM-derived EV. For this, we inoculated immunocompetent BALB/cByJ mice with a myeloma cell line, MOPC315.BM, inducing a MM phenotype. Upon tumor establishment, characterization of the BM microenvironment revealed the expression of both activation and suppressive markers by lymphocytes, such as granzyme B and PD-1, respectively. In addition, conditioning of the animals with MOPC315.BM-derived EV, before transplantation of the MOPC315.BM tumor cells, did not anticipate the disease phenotype. However, it induced features of suppression in the BM milieu, such as an increase in PD-1 expression by CD4+ T cells. Overall, our findings reveal the involvement of MOPC315.BM-derived EV protein content as promoters of immune niche remodeling, strengthening the importance of assessing the mechanisms by which MM may impact the immune microenvironment.
Collapse
Affiliation(s)
- Raquel Lopes
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Joana Caetano
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Foundation, Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School (NMS), Lisbon, Portugal
| | - Filipa Barahona
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School (NMS), Lisbon, Portugal
| | - Carolina Pestana
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Centre of Statistics and Its Applications, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Bruna Velosa Ferreira
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Foundation, Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School (NMS), Lisbon, Portugal
| | - Diana Lourenço
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana C. Queirós
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
| | - Carlos Bilreiro
- Faculty of Medical Sciences, NOVA Medical School (NMS), Lisbon, Portugal
- Neural Plasticity and Neural Activity Laboratory, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
| | - Noam Shemesh
- Neural Plasticity and Neural Activity Laboratory, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Ana Sofia Carvalho
- Computational and Experimental Biology, Chronic Diseases Research Centre (CEDOC); NOVA Medical School (NMS), Lisbon, Portugal
| | - Rune Matthiesen
- Computational and Experimental Biology, Chronic Diseases Research Centre (CEDOC); NOVA Medical School (NMS), Lisbon, Portugal
| | - Bjarne Bogen
- Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Bruno Costa-Silva
- Systems Oncology, Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, Lisbon, Portugal
| | - Karine Serre
- Molecular Medicine Institute-Laço Hub, Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
| | - Cristina João
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Foundation, Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School (NMS), Lisbon, Portugal
- *Correspondence: Cristina João,
| |
Collapse
|
19
|
Ferreira BV, Carneiro EA, Pestana C, Barahona F, Caetano J, Lopes R, Lúcio P, Neves M, Beck HC, Carvalho AS, Matthiesen R, Costa-Silva B, João C. Patient-Derived Extracellular Vesicles Proteins as New Biomarkers in Multiple Myeloma - A Real-World Study. Front Oncol 2022; 12:860849. [PMID: 35800053 PMCID: PMC9254863 DOI: 10.3389/fonc.2022.860849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/16/2022] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of clonal antibody–secreting plasma cells (PCs). MM diagnosis and risk stratification rely on bone marrow (BM) biopsy, an invasive procedure prone to sample bias. Liquid biopsies, such as extracellular vesicles (EV) in peripheral blood (PB), hold promise as new minimally invasive tools. Real-world studies analyzing patient-derived EV proteome are rare. Here, we characterized a small EV protein content from PB and BM samples in a cohort of 102 monoclonal gammopathies patients routinely followed in the clinic and 223 PB and 111 BM samples were included. We investigated whether EV protein and particle concentration could predict an MM patient prognosis. We found that a high EV protein/particle ratio, or EV cargo >0.6 µg/108 particles, is related to poorer survival and immune dysfunction. These results were supported at the protein level by mass spectrometry. We report a set of PB EV-proteins (PDIA3, C4BPA, BTN1A1, and TNFSF13) with a new biomarker potential for myeloma patient outcomes. The high proteomic similarity between PB and BM matched pairs supports the use of circulating EV as a counterpart of the BM EV proteome. Overall, we found that the EV protein content is related to patient outcomes, such as survival, immune dysfunction, and possibly treatment response.
Collapse
Affiliation(s)
- Bruna Velosa Ferreira
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- NOVA Medical School (NMS), NOVA University Lisbon, Lisbon, Portugal
- Hemato-Oncology Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
| | - Carolina Pestana
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Filipa Barahona
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- NOVA Medical School (NMS), NOVA University Lisbon, Lisbon, Portugal
| | - Joana Caetano
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- NOVA Medical School (NMS), NOVA University Lisbon, Lisbon, Portugal
- Hemato-Oncology Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Lisbon, Portugal
| | - Raquel Lopes
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Paulo Lúcio
- Hemato-Oncology Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Lisbon, Portugal
| | - Manuel Neves
- Hemato-Oncology Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Ana Sofia Carvalho
- NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Rune Matthiesen
- NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, Lisbon, Portugal
- *Correspondence: Bruno Costa-Silva, ; Cristina João,
| | - Cristina João
- Myeloma Lymphoma Research Group, Champalimaud Experimental Clinical Research Programme, Champalimaud Foundation, Lisbon, Portugal
- NOVA Medical School (NMS), NOVA University Lisbon, Lisbon, Portugal
- Hemato-Oncology Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Lisbon, Portugal
- *Correspondence: Bruno Costa-Silva, ; Cristina João,
| |
Collapse
|
20
|
Chen K, Li Y, Xu L, Qian Y, Liu N, Zhou C, Liu J, Zhou L, Xu Z, Jia R, Ge YZ. Comprehensive insight into endothelial progenitor cell-derived extracellular vesicles as a promising candidate for disease treatment. Stem Cell Res Ther 2022; 13:238. [PMID: 35672766 PMCID: PMC9172199 DOI: 10.1186/s13287-022-02921-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/29/2022] [Indexed: 12/21/2022] Open
Abstract
Endothelial progenitor cells (EPCs), which are a type of stem cell, have been found to have strong angiogenic and tissue repair capabilities. Extracellular vesicles (EVs) contain many effective components, such as cellular proteins, microRNAs, messenger RNAs, and long noncoding RNAs, and can be secreted by different cell types. The functions of EVs depend mainly on their parent cells. Many researchers have conducted functional studies of EPC-derived EVs (EPC-EVs) and showed that they exhibit therapeutic effects on many diseases, such as cardiovascular disease, acute kidney injury, acute lung injury, and sepsis. In this review article, we comprehensively summarized the biogenesis and functions of EPCs and EVs and the potent role of EPC-EVs in the treatment of various diseases. Furthermore, the current problems and future prospects have been discussed, and further studies are needed to compare the therapeutic effects of EVs derived from various stem cells, which will contribute to the accelerated translation of these applications in a clinical setting.
Collapse
Affiliation(s)
- Ke Chen
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Yiguan Qian
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Ning Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| | - Yu-Zheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| |
Collapse
|
21
|
Ben-Nissan G, Katzir N, Füzesi-Levi MG, Sharon M. Biology of the Extracellular Proteasome. Biomolecules 2022; 12:619. [PMID: 35625547 PMCID: PMC9139032 DOI: 10.3390/biom12050619] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
Proteasomes are traditionally considered intracellular complexes that play a critical role in maintaining proteostasis by degrading short-lived regulatory proteins and removing damaged proteins. Remarkably, in addition to these well-studied intracellular roles, accumulating data indicate that proteasomes are also present in extracellular body fluids. Not much is known about the origin, biological role, mode(s) of regulation or mechanisms of extracellular transport of these complexes. Nevertheless, emerging evidence indicates that the presence of proteasomes in the extracellular milieu is not a random phenomenon, but rather a regulated, coordinated physiological process. In this review, we provide an overview of the current understanding of extracellular proteasomes. To this end, we examine 143 proteomic datasets, leading us to the realization that 20S proteasome subunits are present in at least 25 different body fluids. Our analysis also indicates that while 19S subunits exist in some of those fluids, the dominant proteasome activator in these compartments is the PA28α/β complex. We also elaborate on the positive correlations that have been identified in plasma and extracellular vesicles, between 20S proteasome and activity levels to disease severity and treatment efficacy, suggesting the involvement of this understudied complex in pathophysiology. In addition, we address the considerations and practical experimental methods that should be taken when investigating extracellular proteasomes. Overall, we hope this review will stimulate new opportunities for investigation and thoughtful discussions on this exciting topic that will contribute to the maturation of the field.
Collapse
Affiliation(s)
| | | | | | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel; (G.B.-N.); (N.K.); (M.G.F.-L.)
| |
Collapse
|
22
|
Extracellular vesicle proteomic analysis leads to the discovery of HDGF as a new factor in multiple myeloma biology. Blood Adv 2022; 6:3458-3471. [PMID: 35395072 PMCID: PMC9198912 DOI: 10.1182/bloodadvances.2021006187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/15/2022] [Indexed: 11/20/2022] Open
Abstract
HDGF is secreted by and found in multiple myeloma cell extracellular vesicles; it activates AKT and sustains multiple myeloma cell growth. HDGF polarizes naïve macrophages to an M1 phenotype and generates immunosuppressive M-MDSC.
Identifying factors secreted by multiple myeloma (MM) cells that may contribute to MM tumor biology and progression is of the utmost importance. In this study, hepatoma-derived growth factor (HDGF) was identified as a protein present in extracellular vesicles (EVs) released from human MM cell lines (HMCLs). Investigation of the role of HDGF in MM cell biology revealed lower proliferation of HMCLs following HDGF knockdown and AKT phosphorylation following the addition of exogenous HDGF. Metabolic analysis demonstrated that HDGF enhances the already high glycolytic levels of HMCLs and significantly lowers mitochondrial respiration, indicating that HDGF may play a role in myeloma cell survival and/or act in a paracrine manner on cells in the bone marrow (BM) tumor microenvironment (ME). Indeed, HDGF polarizes macrophages to an M1-like phenotype and phenotypically alters naïve CD14+ monocytes to resemble myeloid-derived suppressor cells which are functionally suppressive. In summary, HDGF is a novel factor in MM biology and may function to both maintain MM cell viability as well as modify the tumor ME.
Collapse
|
23
|
Mann Z, Sengar M, Verma YK, Rajalingam R, Raghav PK. Hematopoietic Stem Cell Factors: Their Functional Role in Self-Renewal and Clinical Aspects. Front Cell Dev Biol 2022; 10:664261. [PMID: 35399522 PMCID: PMC8987924 DOI: 10.3389/fcell.2022.664261] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/14/2022] [Indexed: 01/29/2023] Open
Abstract
Hematopoietic stem cells (HSCs) possess two important properties such as self-renewal and differentiation. These properties of HSCs are maintained through hematopoiesis. This process gives rise to two subpopulations, long-term and short-term HSCs, which have become a popular convention for treating various hematological disorders. The clinical application of HSCs is bone marrow transplant in patients with aplastic anemia, congenital neutropenia, sickle cell anemia, thalassemia, or replacement of damaged bone marrow in case of chemotherapy. The self-renewal attribute of HSCs ensures long-term hematopoiesis post-transplantation. However, HSCs need to be infused in large numbers to reach their target site and meet the demands since they lose their self-renewal capacity after a few passages. Therefore, a more in-depth understanding of ex vivo HSCs expansion needs to be developed to delineate ways to enhance the self-renewability of isolated HSCs. The multifaceted self-renewal process is regulated by factors, including transcription factors, miRNAs, and the bone marrow niche. A developed classical hierarchical model that outlines the hematopoiesis in a lineage-specific manner through in vivo fate mapping, barcoding, and determination of self-renewal regulatory factors are still to be explored in more detail. Thus, an in-depth study of the self-renewal property of HSCs is essentially required to be utilized for ex vivo expansion. This review primarily focuses on the Hematopoietic stem cell self-renewal pathway and evaluates the regulatory molecular factors involved in considering a targeted clinical approach in numerous malignancies and outlining gaps in the current knowledge.
Collapse
Affiliation(s)
- Zoya Mann
- Independent Researcher, New Delhi, India
| | - Manisha Sengar
- Department of Zoology, Deshbandhu College, University of Delhi, Delhi, India
| | - Yogesh Kumar Verma
- Stem Cell and Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), Delhi, India
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Pawan Kumar Raghav
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
24
|
Jin Y, Liang Y, Su Y, Hui L, Liu H, Ding L, Zhou F. Identification of novel combined biomarkers in the diagnosis of multiple myeloma. Hematology 2021; 26:964-969. [PMID: 34871540 DOI: 10.1080/16078454.2021.2003065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Multiple myeloma (MM) is a haematological malignant disease with a clonal proliferation of plasma cells, and timely surveillance is helpful to improve the survival rate of patients with MM. However, there is a lack of simple and effective biomarkers for the diagnosis, prognosis, and residual disease evaluation of MM. MATERIAL & METHODS In the detection cohort, we used the samples from six newly diagnosed MM patients and six control subjects. Plasma proteins were labelled with dimethyl reagents and enriched by lectin AANL6, then the deglycosylated peptides were identified by LC-MS/MS. Differentially expressed proteins were used for further exploration. In the validation cohort, we used 90 newly diagnosed patients with MM and 70 cases of unrelated diseases as controls. The diagnosis performance was analysed by ROC analysis using SPSS. RESULTS In this study, we show, using lectin blots with AANL6, that glycosylation levels were higher in MM patients than in controls. After AANL6 enrichment, we detected 58 differentially expressed proteins using quantitative proteomics. We further validated one candidate Fibulin-1 (FBLN1). Using an Elisa assay, we showed that FBLN1 expression was increased in plasma of 90 cases of MM, and which was significantly correlated with DKK1 expression. ROC analysis showed that these two markers had a 95.7% specificity for determining the diagnosis of MM. CONCLUSION These data suggest that the MM cases display increased glycosylation after AANL6 enrichment and that the combined expression of FBLN1 and DKK1 can be used as an effective diagnostic biomarker.
Collapse
Affiliation(s)
- Yanxia Jin
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, Hubei Normal University, Huangshi, People's Republic of China
| | - Yuxing Liang
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yanting Su
- College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Lingyun Hui
- Department of Laboratory Medicine, First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Hailing Liu
- Department of Clinical Haematology, Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Lu Ding
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Fuling Zhou
- Department of Haematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
25
|
Allegra A, Di Gioacchino M, Tonacci A, Petrarca C, Musolino C, Gangemi S. Multiple Myeloma Cell-Derived Exosomes: Implications on Tumorigenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cells 2021; 10:2865. [PMID: 34831088 PMCID: PMC8616233 DOI: 10.3390/cells10112865] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a hematological disease that is still not curable. The bone marrow milieu, with cellular and non-cellular elements, participate in the creation of a pro-tumoral environment enhancing growth and survival of MM plasma cells. Exosomes are vesicles oscillating in dimension between 50 nm and 100 nm in size that can be released by various cells and contribute to the pathogenesis and progression of MM. Exosomes enclose proteins, cytokines, lipids, microRNAs, long noncoding RNAs, and circular RNAs able to regulate interactions between MM plasma cells and adjacent cells. Through exosomes, mesenchymal stem cells confer chemoresistance to MM cells, while myeloma cells promote angiogenesis, influence immune response, cause bone lesions, and have an impact on the outcome of MM patients. In this review, we analyze the role played by exosomes in the progression of monoclonal gammopathies and the effects on the proliferation of neoplastic plasma cells, and discuss the possible employment of exosomes as potential targets for the treatment of MM patients.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
| | - Alessandro Tonacci
- National Research Council of Italy (IFC-CNR), Clinical Physiology Institute, 56124 Pisa, Italy;
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- National Research Council of Italy (IFC-CNR), Clinical Physiology Institute, 56124 Pisa, Italy;
- Department of Medicine and Science of Ageing, G. D’Annunzio University, 66100 Chieti, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
26
|
The role of CD44 in the assessment of minimal residual disease of multiple myeloma by flow cytometry. J Hematop 2021. [DOI: 10.1007/s12308-021-00468-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
27
|
Liquid biopsy: an evolving paradigm for the biological characterisation of plasma cell disorders. Leukemia 2021; 35:2771-2783. [PMID: 34262132 DOI: 10.1038/s41375-021-01339-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/14/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Liquid biopsies-a source of circulating cell-free nucleic acids, proteins and extracellular vesicles-are currently being explored for the quantitative and qualitative characterisation of the tumour genome and as a mode of non-invasive therapeutic monitoring in cancer. Emerging data suggest that liquid biopsies might offer a potentially simple, non-invasive, repeatable strategy for diagnosis, prognostication and therapeutic decision making in a genetically heterogeneous disease like multiple myeloma (MM), with particular applicability in subsets of patients where conventional markers of disease burden may be less informative. In this review, we describe the emerging utility of the evaluation of circulating tumour DNA, extracellular RNA, cell-free proteins and metabolites and extracellular vesicles in MM.
Collapse
|
28
|
Reale A, Khong T, Mithraprabhu S, Spencer A. Translational Potential of RNA Derived From Extracellular Vesicles in Multiple Myeloma. Front Oncol 2021; 11:718502. [PMID: 34513695 PMCID: PMC8429596 DOI: 10.3389/fonc.2021.718502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
The cross-talk between tumour cells and stromal cells is a hallmark of multiple myeloma (MM), a blood cancer that still remains incurable despite increased knowledge of its biology and advances in its treatment. Extracellular vesicles (EVs) derived from both tumour and stromal cells have been shown to play an important role in mediating this cross-talk ultimately favouring MM progression and drug resistance. Furthermore, EVs and their content including RNA (EV-RNA) have been successfully isolated from blood and are being explored as liquid biomarkers in MM with the potential to improve diagnosis and monitoring modalities with a minimally-invasive and repeatable analysis, i.e. liquid biopsy. In this review, we describe both the role of EV-RNA in defining the biological features of MM and their potential translational relevance as liquid biomarkers, therapeutic targets and delivery systems. We also discuss the limitations and technical challenges related to the isolation and characterization of EVs and provide a perspective on the future of MM-derived EV-RNA in translational research.
Collapse
Affiliation(s)
- Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia
| | - Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia.,Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, and Department of Clinical Haematology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
29
|
Dunphy K, O’Mahoney K, Dowling P, O’Gorman P, Bazou D. Clinical Proteomics of Biofluids in Haematological Malignancies. Int J Mol Sci 2021; 22:ijms22158021. [PMID: 34360786 PMCID: PMC8348619 DOI: 10.3390/ijms22158021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Since the emergence of high-throughput proteomic techniques and advances in clinical technologies, there has been a steady rise in the number of cancer-associated diagnostic, prognostic, and predictive biomarkers being identified and translated into clinical use. The characterisation of biofluids has become a core objective for many proteomic researchers in order to detect disease-associated protein biomarkers in a minimally invasive manner. The proteomes of biofluids, including serum, saliva, cerebrospinal fluid, and urine, are highly dynamic with protein abundance fluctuating depending on the physiological and/or pathophysiological context. Improvements in mass-spectrometric technologies have facilitated the in-depth characterisation of biofluid proteomes which are now considered hosts of a wide array of clinically relevant biomarkers. Promising efforts are being made in the field of biomarker diagnostics for haematologic malignancies. Several serum and urine-based biomarkers such as free light chains, β-microglobulin, and lactate dehydrogenase are quantified as part of the clinical assessment of haematological malignancies. However, novel, minimally invasive proteomic markers are required to aid diagnosis and prognosis and to monitor therapeutic response and minimal residual disease. This review focuses on biofluids as a promising source of proteomic biomarkers in haematologic malignancies and a key component of future diagnostic, prognostic, and disease-monitoring applications.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Kelly O’Mahoney
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
- Correspondence:
| |
Collapse
|
30
|
Khalife J, Sanchez JF, Pichiorri F. The Emerging Role of Extracellular Vesicle-Associated RNAs in the Multiple Myeloma Microenvironment. Front Oncol 2021; 11:689538. [PMID: 34235082 PMCID: PMC8255802 DOI: 10.3389/fonc.2021.689538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a cancer of terminally differentiated plasma cells (PCs) that develop at multiple sites within the bone marrow (BM). MM is treatable but rarely curable because of the frequent emergence of drug resistance and relapse. Increasing evidence indicates that the BM microenvironment plays a major role in supporting MM-PC survival and resistance to therapy. The BM microenvironment is a complex milieu containing hematopoietic cells, stromal cells, endothelial cells, immune cells, osteoclasts and osteoblasts, all contributing to the pathobiology of MM, including PC proliferation, escape from immune surveillance, angiogenesis and bone disease development. Small extracellular vesicles (EVs) are heterogenous lipid structures released by all cell types and mediate local and distal cellular communication. In MM, EVs are key mediators of the cross-talk between PCs and the surrounding microenvironment because of their ability to deliver bioactive cargo molecules such as lipids, mRNAs, non-coding regulatory RNA and proteins. Hence, MM-EVs highly contribute to establish a tumor-supportive BM niche that impacts MM pathogenesis and disease progression. In this review, we will first highlight the effects of RNA-containing, MM-derived EVs on the several cellular compartments within the BM microenvironment that play a role in the different aspects of MM pathology. We will also touch on the prospective use of MM-EV-associated non-coding RNAs as clinical biomarkers in the context of “liquid biopsy” in light of their importance as a promising tool in MM diagnosis, prognosis and prediction of drug resistance.
Collapse
Affiliation(s)
- Jihane Khalife
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA, United States.,Department of Hematologic Malignancies Translational Science, City of Hope, Duarte, CA, United States
| | - James F Sanchez
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA, United States
| | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA, United States.,Department of Hematologic Malignancies Translational Science, City of Hope, Duarte, CA, United States
| |
Collapse
|
31
|
Human Plasma Extracellular Vesicle Isolation and Proteomic Characterization for the Optimization of Liquid Biopsy in Multiple Myeloma. Methods Mol Biol 2021; 2261:151-191. [PMID: 33420989 DOI: 10.1007/978-1-0716-1186-9_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer cells secrete membranous extracellular vesicles (EVs) which contain specific oncogenic molecular cargo (including oncoproteins, oncopeptides, and RNA) into their microenvironment and the circulation. As such, EVs including exosomes (small EVs) and microvesicles (large EVs) represent important circulating biomarkers for various diseases, including cancer and its progression. These circulating biomarkers offer a potentially minimally invasive and repeatable targets for analysis (liquid biopsy) that could aid in the diagnosis, risk stratification, and monitoring of cancer. Although their potential as cancer biomarkers has been promising, the identification and quantification of EVs in clinical samples remain challenging. Like EVs, other types of circulating biomarkers (including cell-free nucleic acids, cf-NAs; or circulating tumor cells, CTCs) may represent a complementary or alternative approach to cancer diagnosis. In the context of multiple myeloma (MM), a systemic cancer type that causes cancer cells to accumulate in the bone marrow, the specific role for EVs as biomarkers for diagnosis and monitoring remains undefined. Tumor heterogeneity along with the various subtypes of MM (such as non-secretory MM) that cannot be monitored using conventional testing (e.g. sequential serological testing and bone marrow biopsies) render liquid biopsy and circulating tumor-derived EVs a promising approach. In this protocol, we describe the isolation and purification of EVs from peripheral blood plasma (PBPL) collected from healthy donors and patients with MM for a biomarker discovery strategy. Our results demonstrate detection of circulating EVs from as little as 1 mL of MM patients' PBPL. High-resolution mass spectrometry (MS)-based proteomics promises to provide new avenues in identifying novel markers for detection, monitoring, and therapeutic intervention of disease. We describe biophysical characterization and quantitative proteomic profiling of disease-specific circulating EVs which may provide important implications for the development of cancer diagnostics in MM.
Collapse
|
32
|
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
|
33
|
Reale A, Carmichael I, Xu R, Mithraprabhu S, Khong T, Chen M, Fang H, Savvidou I, Ramachandran M, Bingham N, Simpson RJ, Greening DW, Spencer A. Human myeloma cell- and plasma-derived extracellular vesicles contribute to functional regulation of stromal cells. Proteomics 2021; 21:e2000119. [PMID: 33580572 DOI: 10.1002/pmic.202000119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Circulating small extracellular vesicles (sEV) represent promising non-invasive biomarkers that may aid in the diagnosis and risk-stratification of multiple myeloma (MM), an incurable blood cancer. Here, we comprehensively isolated and characterized sEV from human MM cell lines (HMCL) and patient-derived plasma (psEV) by specific EV-marker enrichment and morphology. Importantly, we demonstrate that HMCL-sEV are readily internalised by stromal cells to functionally modulate proliferation. psEV were isolated using various commercial approaches and pre-analytical conditions (collection tube types, storage conditions) assessed for sEV yield and marker enrichment. Functionally, MM-psEV was shown to regulate stromal cell proliferation and migration. In turn, pre-educated stromal cells favour HMCL adhesion. psEV isolated from patients with both pre-malignant plasma cell disorders (monoclonal gammopathy of undetermined significance [MGUS]; smouldering MM [SMM]) and MM have a similar ability to promote cell migration and adhesion, suggesting a role for both malignant and pre-malignant sEV in disease progression. Proteomic profiling of MM-psEV (305 proteins) revealed enrichment of oncogenic factors implicated in cell migration and adhesion, in comparison to non-disease psEV. This study describes a protocol to generate morphologically-intact and biologically functional sEV capable of mediating the regulation of stromal cells, and a model for the characterization of tumour-stromal cross-talk by sEV in MM.
Collapse
Affiliation(s)
- Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Irena Carmichael
- Monash Micro Imaging-AMREP, Monash University, Melbourne, Victoria, Australia
| | - Rong Xu
- Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Maoshan Chen
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia
| | - Haoyun Fang
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ioanna Savvidou
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Malarmathy Ramachandran
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Nicholas Bingham
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - David W Greening
- Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.,Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, and Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Trino S, Lamorte D, Caivano A, De Luca L, Sgambato A, Laurenzana I. Clinical relevance of extracellular vesicles in hematological neoplasms: from liquid biopsy to cell biopsy. Leukemia 2021; 35:661-678. [PMID: 33299143 PMCID: PMC7932927 DOI: 10.1038/s41375-020-01104-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
In the era of precision medicine, liquid biopsy is becoming increasingly important in oncology. It consists in the isolation and analysis of tumor-derived biomarkers, including extracellular vesicles (EVs), in body fluids. EVs are lipid bilayer-enclosed particles, heterogeneous in size and molecular composition, released from both normal and neoplastic cells. In tumor context, EVs are valuable carriers of cancer information; in fact, their amount, phenotype and molecular cargo, including proteins, lipids, metabolites and nucleic acids, mirror nature and origin of parental cells rendering EVs appealing candidates as novel biomarkers. Translation of these new potential diagnostic tools into clinical practice could deeply revolutionize the cancer field mainly for solid tumors but for hematological neoplasms, too.
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy.
| | - Antonella Caivano
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Luciana De Luca
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Alessandro Sgambato
- Scientific Direction, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy.
| |
Collapse
|
35
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021. [DOI: 10.37349/etat.2020.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3University of Montpellier, UFR Medicine, 34093 Montpellier, France 4 Institut Universitaire de France (IUF), 75000 Paris France
| |
Collapse
|
36
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:65-106. [PMID: 36046090 PMCID: PMC9400753 DOI: 10.37349/etat.2021.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/06/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3UFR Medicine, University of Montpellier, 34093 Montpellier, France 4Institut Universitaire de France (IUF), 75000 Paris, France
| |
Collapse
|
37
|
López-Pacheco C, Bedoya-López A, Olguín-Alor R, Soldevila G. Analysis of Tumor-Derived Exosomes by Nanoscale Flow Cytometry. Methods Mol Biol 2021; 2174:171-191. [PMID: 32813250 DOI: 10.1007/978-1-0716-0759-6_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The study of tumor exosomes has gained relevance in the last decades due to their potential use for therapeutic and diagnostic application. Although there is extensive knowledge of exosome biology, some biological samples like tumor-derived exosomes have been difficult to characterize due to their complexity and heterogeneity. This distinctive feature makes difficult the identification of specific exosome subpopulations with a shared molecular signature that could allow for targeting of exosomes with therapeutic and diagnostic potential use in cancer patients. Nanoscale flow cytometry has lately emerged as an alternative tool that can be adapted to the study of nanoparticles, such as exosomes. However, the physicochemical properties of these particles are an important issue to consider as nanoparticles need the application of specific settings which differ from those used in conventional flow cytometry of cells. Therefore, in the last few years, one of the main aims has been the optimization of technical and experimental protocols to improve exosome analysis. In this chapter, we discuss several aspects of cytometric systems with a special emphasis in technical considerations of samples and equipment.
Collapse
Affiliation(s)
- Cynthia López-Pacheco
- Departamento de Inmunología and Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Andrea Bedoya-López
- Departamento de Inmunología and Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Roxana Olguín-Alor
- Departamento de Inmunología and Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gloria Soldevila
- Departamento de Inmunología and Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
38
|
Extracellular Vesicles in Hematological Malignancies: From Biomarkers to Therapeutic Tools. Diagnostics (Basel) 2020; 10:diagnostics10121065. [PMID: 33316884 PMCID: PMC7763630 DOI: 10.3390/diagnostics10121065] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/06/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023] Open
Abstract
Small extracellular vesicles (EVs) are a heterogenous group of lipid particles released by all cell types in physiological and pathological states. In hematological malignancies, tumor-derived EVs are critical players in mediating intercellular communications through the transfer of genetic materials and proteins between neoplastic cells themselves and to several components of the bone marrow microenvironment, rendering the latter a “stronger” niche supporting cancer cell proliferation, drug resistance, and escape from immune surveillance. In this context, the molecular cargoes of tumor-derived EVs reflect the nature and status of the cells of origin, making them specific therapeutic targets. Another important characteristic of EVs in hematological malignancies is their use as a potential “liquid biopsy” because of their high abundance in biofluids and their ability to protect their molecular cargoes from nuclease and protease degradation. Liquid biopsies are non-invasive blood tests that provide a molecular profiling clinical tool as an alternative method of disease stratification, especially in cancer patients where solid biopsies have limited accessibility. They offer accurate diagnoses and identify specific biomarkers for monitoring of disease progression and response to treatment. In this review, we will focus on the role of EVs in the most prevalent hematological malignancies, particularly on their prospective use as biomarkers in the context of liquid biopsies, as well as their molecular signature that identifies them as specific therapeutic targets for inhibiting cancer progression. We will also highlight their roles in modulating the immune response by acting as both immunosuppressors and activators of anti-tumor immunity.
Collapse
|
39
|
Barreiro K, Dwivedi OP, Leparc G, Rolser M, Delic D, Forsblom C, Groop P, Groop L, Huber TB, Puhka M, Holthofer H. Comparison of urinary extracellular vesicle isolation methods for transcriptomic biomarker research in diabetic kidney disease. J Extracell Vesicles 2020; 10:e12038. [PMID: 33437407 PMCID: PMC7789228 DOI: 10.1002/jev2.12038] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/20/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
Urinary Extracellular Vesicles (uEV) have emerged as a source for biomarkers of kidney damage, holding potential to replace the conventional invasive techniques including kidney biopsy. However, comprehensive studies characterizing uEV isolation methods with patient samples are rare. Here we compared performance of three established uEV isolation workflows for their subsequent use in transcriptomics analysis for biomarker discovery in diabetic kidney disease. We collected urine samples from individuals with type 1 diabetes with macroalbuminuria and healthy controls. We isolated uEV by Hydrostatic Filtration Dialysis (HFD), ultracentrifugation (UC), and a commercial kit- based isolation method (NG), each with different established urine clearing steps. Purified EVs were analysed by electron microscopy, nanoparticle tracking analysis, and Western blotting. Isolated RNAs were subjected to miRNA and RNA sequencing. HFD and UC samples showed close similarities based on mRNA sequencing data. NG samples had a lower number of reads and different mRNA content compared to HFD or UC. For miRNA sequencing data, satisfactory miRNA counts were obtained by all methods, but miRNA contents differed slightly. This suggests that the isolation workflows enrich specific subpopulations of miRNA-rich uEV preparation components. Our data shows that HFD,UC and the kit-based method are suitable methods to isolate uEV for miRNA-seq. However, only HFD and UC were suitable for mRNA-seq in our settings.
Collapse
Affiliation(s)
- Karina Barreiro
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - Om Prakash Dwivedi
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - German Leparc
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Marcel Rolser
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Denis Delic
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre MannheimUniversity of HeidelbergHeidelbergGermany
| | - Carol Forsblom
- Folkhälsan Institute of GeneticsFolkhälsan Research CenterHelsinkiFinland
- Abdominal Center, NephrologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Research Program for Clinical and Molecular Metabolism, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Per‐Henrik Groop
- Folkhälsan Institute of GeneticsFolkhälsan Research CenterHelsinkiFinland
- Abdominal Center, NephrologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Research Program for Clinical and Molecular Metabolism, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Diabetes, Central Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Leif Groop
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - Tobias B. Huber
- III Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Maija Puhka
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - Harry Holthofer
- Institute for Molecular Medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
- III Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
40
|
Luo M, Liu Y, Shao R, Bhyan SB, Wee Y, Zhao M. Mutational analysis revealed 97 key cancer metastasis genes from extracellular vesicles associated with patient survival. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
41
|
Mallia A, Gianazza E, Zoanni B, Brioschi M, Barbieri SS, Banfi C. Proteomics of Extracellular Vesicles: Update on Their Composition, Biological Roles and Potential Use as Diagnostic Tools in Atherosclerotic Cardiovascular Diseases. Diagnostics (Basel) 2020; 10:diagnostics10100843. [PMID: 33086718 PMCID: PMC7588996 DOI: 10.3390/diagnostics10100843] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bound vesicles released from cells under physiological and pathological conditions. Basing on biogenesis, dimension, content and route of secretion, they can be classified into exosomes, microvesicles (MVs) and apoptotic bodies. EVs have a key role as bioactive mediators in intercellular communication, but they are also involved in other physiological processes like immune response, blood coagulation, and tissue repair. The interest in studying EVs has increased over the years due to their involvement in several diseases, such as cardiovascular diseases (CVDs), and their potential role as biomarkers in diagnosis, therapy, and in drug delivery system development. Nowadays, the improvement of mass spectrometry (MS)-based techniques allows the characterization of the EV protein composition to deeply understand their role in several diseases. In this review, a critical overview is provided on the EV’s origin and physical properties, as well as their emerging functional role in both physiological and disease conditions, focusing attention on the role of exosomes in CVDs. The most important cardiac exosome proteomic studies will be discussed giving a qualitative and quantitative characterization of the exosomal proteins that could be used in future as new potential diagnostic markers or targets for specific therapies.
Collapse
|
42
|
Gargiulo E, Morande PE, Largeot A, Moussay E, Paggetti J. Diagnostic and Therapeutic Potential of Extracellular Vesicles in B-Cell Malignancies. Front Oncol 2020; 10:580874. [PMID: 33117718 PMCID: PMC7550802 DOI: 10.3389/fonc.2020.580874] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EV), comprising microvesicles and exosomes, are particles released by every cell of an organism, found in all biological fluids, and commonly involved in cell-to-cell communication through the transfer of cargo materials such as miRNA, proteins, and immune-related ligands (e.g., FasL and PD-L1). An important characteristic of EV is that their composition, abundance, and roles are tightly related to the parental cells. This translates into a higher release of characteristic pro-tumor EV by cancer cells that leads to harming signals toward healthy microenvironment cells. In line with this, the key role of tumor-derived EV in cancer progression was demonstrated in multiple studies and is considered a hot topic in the field of oncology. Given their characteristics, tumor-derived EV carry important information concerning the state of tumor cells. This can be used to follow the outset, development, and progression of the neoplasia and to evaluate the design of appropriate therapeutic strategies. In keeping with this, the present brief review will focus on B-cell malignancies and how EV can be used as potential biomarkers to follow disease progression and stage. Furthermore, we will explore several proposed strategies aimed at using biologically engineered EV for treatment (e.g., drug delivery mechanisms) as well as for impairing the biogenesis, release, and internalization of cancer-derived EV, with the final objective to disrupt tumor–microenvironment communication.
Collapse
Affiliation(s)
- Ernesto Gargiulo
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Pablo Elías Morande
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Anne Largeot
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Etienne Moussay
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Jérôme Paggetti
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
43
|
Tanasi I, Adamo A, Kamga PT, Bazzoni R, Krampera M. High-throughput analysis and functional interpretation of extracellular vesicle content in hematological malignancies. Comput Struct Biotechnol J 2020; 18:2670-2677. [PMID: 33101605 PMCID: PMC7554250 DOI: 10.1016/j.csbj.2020.09.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane-coated particles secreted by virtually all cell types in response to different stimuli, both in physiological and pathological conditions. Their content generally reflects their biological functions and includes a variety of molecules, such as nucleic acids, proteins and cellular components. The role of EVs as signaling vehicles has been widely demonstrated. In particular, they are actively involved in the pathogenesis of several hematological malignancies (HM), mainly interacting with a number of target cells and inducing functional and epigenetic changes. In this regard, by releasing their cargo, EVs play a pivotal role in the bilateral cross-talk between tumor microenvironment and cancer cells, thus facilitating mechanisms of immune escape and supporting tumor growth and progression. Recent advances in high-throughput technologies have allowed the deep characterization and functional interpretation of EV content. In this review, the current knowledge on the high-throughput technology-based characterization of EV cargo in HM is summarized.
Collapse
Affiliation(s)
- Ilaria Tanasi
- Department of Medicine, Hematology Section, University of Verona, Italy
| | - Annalisa Adamo
- Department of Medicine, Immunology Section, University of Verona, Italy
| | - Paul Takam Kamga
- Department of Medicine, Hematology Section, University of Verona, Italy
| | - Riccardo Bazzoni
- Department of Medicine, Hematology Section, University of Verona, Italy
| | - Mauro Krampera
- Department of Medicine, Hematology Section, University of Verona, Italy
| |
Collapse
|
44
|
Challenges and Opportunities in Clinical Applications of Blood-Based Proteomics in Cancer. Cancers (Basel) 2020; 12:cancers12092428. [PMID: 32867043 PMCID: PMC7564506 DOI: 10.3390/cancers12092428] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The traditional approach in identifying cancer related protein biomarkers has focused on evaluation of a single peptide/protein in tissue or circulation. At best, this approach has had limited success for clinical applications, since multiple pathological tumor pathways may be involved during initiation or progression of cancer which diminishes the significance of a single candidate protein/peptide. Emerging sensitive proteomic based technologies like liquid chromatography mass spectrometry (LC-MS)-based quantitative proteomics can provide a platform for evaluating serial serum or plasma samples to interrogate secreted products of tumor–host interactions, thereby revealing a more “complete” repertoire of biological variables encompassing heterogeneous tumor biology. However, several challenges need to be met for successful application of serum/plasma based proteomics. These include uniform pre-analyte processing of specimens, sensitive and specific proteomic analytical platforms and adequate attention to study design during discovery phase followed by validation of discovery-level signatures for prognostic, predictive, and diagnostic cancer biomarker applications. Abstract Blood is a readily accessible biofluid containing a plethora of important proteins, nucleic acids, and metabolites that can be used as clinical diagnostic tools in diseases, including cancer. Like the on-going efforts for cancer biomarker discovery using the liquid biopsy detection of circulating cell-free and cell-based tumor nucleic acids, the circulatory proteome has been underexplored for clinical cancer biomarker applications. A comprehensive proteome analysis of human serum/plasma with high-quality data and compelling interpretation can potentially provide opportunities for understanding disease mechanisms, although several challenges will have to be met. Serum/plasma proteome biomarkers are present in very low abundance, and there is high complexity involved due to the heterogeneity of cancers, for which there is a compelling need to develop sensitive and specific proteomic technologies and analytical platforms. To date, liquid chromatography mass spectrometry (LC-MS)-based quantitative proteomics has been a dominant analytical workflow to discover new potential cancer biomarkers in serum/plasma. This review will summarize the opportunities of serum proteomics for clinical applications; the challenges in the discovery of novel biomarkers in serum/plasma; and current proteomic strategies in cancer research for the application of serum/plasma proteomics for clinical prognostic, predictive, and diagnostic applications, as well as for monitoring minimal residual disease after treatments. We will highlight some of the recent advances in MS-based proteomics technologies with appropriate sample collection, processing uniformity, study design, and data analysis, focusing on how these integrated workflows can identify novel potential cancer biomarkers for clinical applications.
Collapse
|
45
|
Deng W, Wang L, Pan M, Zheng J. The regulatory role of exosomes in leukemia and their clinical significance. J Int Med Res 2020; 48:300060520950135. [PMID: 32840158 PMCID: PMC7450464 DOI: 10.1177/0300060520950135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Recurrence is a primary cause of death in patients with leukemia. The
interactions of tumor cells with the microenvironment and tumor stem cells
hidden in bone marrow promote the recurrence and metastasis of leukemia to
lymphoid tissue. Exosomes, membrane-coated nanovesicles secreted by living
cells, perform biomaterial transfer and information exchange between cells.
Exosomes contain various other biological components derived from parental
cells, and they remotely regulate the function of target cells through body
fluid flow. Recent studies revealed that exosomes participate in the development
of leukemia and play important roles in its diagnosis and treatment by
influencing cell proliferation and apoptosis, regulating bone marrow
microenvironment, promoting angiogenesis, and inhibiting hematopoiesis. Exosomes
are potential biomarkers and therapeutic targets for leukemia, and they can
influence drug resistance. Leukemia-derived exosomes present leukemia-related
antigens to target cells, promote the proliferation of leukemic cells, help
these cells escape immunity, protect them from the cytotoxic effects of
chemotherapeutics, and promote angiogenesis and tumor migration. Therefore,
exosomes are closely related to the metastasis, treatment, and prognosis of
leukemia, and they can be used to detect and monitor the progression of
leukemia. This paper reviews the regulatory roles of exosomes in leukemia and
their clinical significance.
Collapse
Affiliation(s)
- Wei Deng
- Department of Pediatric General Internal Medicine, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, Gansu, China
| | - Li Wang
- Department of Pediatric General Internal Medicine, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, Gansu, China
| | - Ming Pan
- Department of Hematology, Wuwei People's Hospital, Wuwei, Gansu, China
| | - Jianping Zheng
- Department of Orthopedic Surgery, Xiangyang Central Hospital, the Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
46
|
Wang X, Cheng K, Zhang G, Jia Z, Yu Y, Guo J, Hua Y, Guo F, Li X, Zou W, Sun H, Dong J, Yang Z. Enrichment of CD44 in Exosomes From Breast Cancer Cells Treated With Doxorubicin Promotes Chemoresistance. Front Oncol 2020; 10:960. [PMID: 32760666 PMCID: PMC7373100 DOI: 10.3389/fonc.2020.00960] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Exosomes secreted from tumor cells can remodel the tumor environment by promoting tumor metastasis and multidrug resistance. The aim of this study was to analyze the proteome profile of the breast cancer line resistant to doxorubicin resistance (MCF-7/ADR) by liquid chromatography linked to tandem mass spectrometry assay (LC-MS/MS). Our results revealed that DOX increases the exosomes release from MCF-7/ADR cells and the exosome-mediated proteins intercellular transfer in breast cancer chemoresistance regulation. The expression of the candidate target exosomic CD44 in DOX-resistant cells (A/Exo) was higher than in parental breast cancer cells (S/Exo), and the increasing levels of exosomic CD44 (21.65-fold) were higher than those of cellular CD44 (6.55-fold) (all p < 0.05). Similar results were obtained in clinical samples; exosomal CD44 in the serum of nonresponders was significantly higher than that in the chemotherapy-responsive group (p < 0.05). Also, we modified the MCF-7-derived exosomes loaded with siRNA against CD44 to observe the effects of targeting reduced CD44 expression in luminal A breast cancer cells. Exosome-siRNA targeted CD44 (Exos-siCD44) could efficiently silence its expression. When cocultured on Exos-siCD44, breast cancer cells exhibited reduced cell proliferation and enhanced susceptibility to DOX. The same phenomenon was observed in mice. In conclusion, breast cancer cells could spread resistance capacity by the intercellular transfer of proteins, especially CD44, via exosomes.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Kai Cheng
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Guoqiang Zhang
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Zhongming Jia
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Yue Yu
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Jiwei Guo
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Yitong Hua
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Fengli Guo
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Xiaoqiang Li
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Weiwei Zou
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Hongguang Sun
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Jianli Dong
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Zhenlin Yang
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
47
|
Xing Z, Zhao C, Liu H, Fan Y. Endothelial Progenitor Cell-Derived Extracellular Vesicles: A Novel Candidate for Regenerative Medicine and Disease Treatment. Adv Healthc Mater 2020; 9:e2000255. [PMID: 32378361 DOI: 10.1002/adhm.202000255] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/12/2020] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membranous structures, which can be secreted by most cell types. As a product of paracrine secretion, EVs are considered to be a regulatory mediator for intercellular communication. There are many bioactive cargos in EVs, such as proteins, lipids, and nucleic acids. As the precursor cell of vascular endothelial cells (ECs), endothelial progenitor cells (EPCs) are first discovered in peripheral blood. With the development of studies about the functions of EPCs, an increasing number of researchers focus on EPC-derived EVs (EPC-EVs). EPC-EVs exert key functions for promoting angiogenesis in regenerative medicine and show significant therapeutic effects on a variety of diseases such as circulatory diseases, kidney diseases, diabetes, bone diseases, and tissue/organ damages. This article reviews the current knowledge on the role of EPC-EVs in regenerative medicine and disease treatment, discussing the main challenges and future directions in this field.
Collapse
Affiliation(s)
- Zheng Xing
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang University Beijing 100191 P. R. China
| | - Chen Zhao
- School of Pharmaceutical SciencesTsinghua University Beijing 100084 P. R. China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang University Beijing 100191 P. R. China
- Beijing Advanced Innovation Centre for Biomedical EngineeringBeihang University Beijing 100191 P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang University Beijing 100191 P. R. China
- Beijing Advanced Innovation Centre for Biomedical EngineeringBeihang University Beijing 100191 P. R. China
- National Research Center for Rehabilitation Technical Aids Beijing 100176 P. R. China
| |
Collapse
|
48
|
Ho M, Bianchi G, Anderson KC. Proteomics-inspired precision medicine for treating and understanding multiple myeloma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:67-85. [PMID: 34414281 DOI: 10.1080/23808993.2020.1732205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Introduction Remarkable progress in molecular characterization methods has led to significant improvements in how we manage multiple myeloma (MM). The introduction of novel therapies has led to significant improvements in overall survival over the past 10 years. However, MM remains incurable and treatment choice is largely based on outdated risk-adaptive strategies that do not factor in improved treatment outcomes in the context of modern therapies. Areas covered This review discusses current risk-adaptive strategies in MM and the clinical application of proteomics in the monitoring of treatment response, disease progression, and minimal residual disease (MRD). We also discuss promising biomarkers of disease progression, treatment response, and chemoresistance. Finally, we will discuss an immunomics-based approach to monoclonal antibody (mAb), vaccine, and CAR-T cell development. Expert opinion It is an exciting era in oncology with basic scientific knowledge translating in novel therapeutic approaches to improve patient outcomes. With the advent of effective immunotherapies and targeted therapies, it has become crucial to identify biomarkers to aid in the stratification of patients based on anticipated sensitivity to chemotherapy. As a paradigm of diseases highly dependent on protein homeostasis, multiple myeloma provides the perfect opportunity to investigate the use of proteomics to aid in precision medicine.
Collapse
Affiliation(s)
- Matthew Ho
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Dublin, Ireland
| | - Giada Bianchi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Pourhanifeh MH, Mahjoubin-Tehran M, Shafiee A, Hajighadimi S, Moradizarmehri S, Mirzaei H, Asemi Z. MicroRNAs and exosomes: Small molecules with big actions in multiple myeloma pathogenesis. IUBMB Life 2019; 72:314-333. [PMID: 31828868 DOI: 10.1002/iub.2211] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/22/2019] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM), an incurable hematologic malignancy of plasma cells increasing in the bone marrow (BM), has a complex microenvironment made to support proliferation, survival, and drug resistance of tumor cells. MicroRNAs (miRNAs), short non-coding RNAs regulating genes expression at posttranscriptional level, have been indicated to be functionally deregulated or abnormally expressed in MM cells. Moreover, by means of miRNAs, tumor microenvironment also modulates the function of MM cells. Consistently, it has been demonstrated that miRNA levels regulation impairs their interaction with the microenvironment of BM as well as create considerable antitumor feature even capable of overcoming the protective BM milieu. Communication between cancer stromal cells and cancer cells is a key factor in tumor progression. Finding out this interaction is important to develop effective approaches that reverse bone diseases. Exosomes, nano-vehicles having crucial roles in cell-to-cell communication, through targeting their cargos (i.e., miRNAs, mRNAs, DNAs, and proteins), are implicated in MM pathogenesis.
Collapse
Affiliation(s)
- Mohammad H Pourhanifeh
- Halal Research Center of IRI, FDA, Tehran, Iran.,Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alimohammad Shafiee
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Sarah Hajighadimi
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Sanaz Moradizarmehri
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
50
|
Khalife J, Ghose J, Martella M, Viola D, Rocci A, Troadec E, Terrazas C, Satoskar AR, Gunes EG, Dona A, Sanchez JF, Bergsagel PL, Chesi M, Pozhitkov A, Rosen S, Marcucci G, Keats JJ, Hofmeister CC, Krishnan A, Caserta E, Pichiorri F. MiR-16 regulates crosstalk in NF-κB tolerogenic inflammatory signaling between myeloma cells and bone marrow macrophages. JCI Insight 2019; 4:129348. [PMID: 31593552 DOI: 10.1172/jci.insight.129348] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 10/02/2019] [Indexed: 12/27/2022] Open
Abstract
High levels of circulating miR-16 in the serum of multiple myeloma (MM) patients are independently associated with longer survival. Although the tumor suppressor function of intracellular miR-16 in MM plasma cells (PCs) has been elucidated, its extracellular role in maintaining a nonsupportive cancer microenvironment has not been fully explored. Here, we show that miR-16 is abundantly released by MM cells through extracellular vesicles (EVs) and that differences in its intracellular expression as associated with chromosome 13 deletion (Del13) are correlated to extracellular miR-16 levels. We also demonstrate that EVs isolated from MM patients and from the conditioned media of MM-PCs carrying Del13 more strongly differentiate circulating monocytes to M2-tumor supportive macrophages (TAMs), compared with MM-PCs without this chromosomal aberration. Mechanistically, our data show that miR-16 directly targets the IKKα/β complex of the NF-κB canonical pathway, which is critical not only in supporting MM cell growth, but also in polarizing macrophages toward an M2 phenotype. By using a miR-15a-16-1-KO mouse model, we found that loss of the miR-16 cluster supports polarization to M2 macrophages. Finally, we demonstrate the therapeutic benefit of miR-16 overexpression in potentiating the anti-MM activity by a proteasome inhibitor in the presence of MM-resident bone marrow TAM.
Collapse
Affiliation(s)
- Jihane Khalife
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Jayeeta Ghose
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio, USA
| | - Marianna Martella
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Domenico Viola
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Alberto Rocci
- Faculty of Biology, Medicine and Health, School of Medical Science, Division of Cancer Science, University of Manchester, Manchester, United Kingdom
| | - Estelle Troadec
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Cesar Terrazas
- Division of Experimental Pathology, Department of Microbiology, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Abhay R Satoskar
- Division of Experimental Pathology, Department of Microbiology, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Emine Gulsen Gunes
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Ada Dona
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - James F Sanchez
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - P Leif Bergsagel
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Marta Chesi
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Alex Pozhitkov
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Steven Rosen
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA.,Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope, Duarte, California, USA
| | - Jonathan J Keats
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Amrita Krishnan
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Enrico Caserta
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|