1
|
Lattmann E, Räss L, Tognetti M, Gómez JMM, Lapaire V, Bruderer R, Reiter L, Feng Y, Steinmetz LM, Levesque MP. Size-exclusion chromatography combined with DIA-MS enables deep proteome profiling of extracellular vesicles from melanoma plasma and serum. Cell Mol Life Sci 2024; 81:90. [PMID: 38353833 PMCID: PMC10867102 DOI: 10.1007/s00018-024-05137-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/16/2024]
Abstract
Extracellular vesicles (EVs) are important players in melanoma progression, but their use as clinical biomarkers has been limited by the difficulty of profiling blood-derived EV proteins with high depth of coverage, the requirement for large input amounts, and complex protocols. Here, we provide a streamlined and reproducible experimental workflow to identify plasma- and serum- derived EV proteins of healthy donors and melanoma patients using minimal amounts of sample input. SEC-DIA-MS couples size-exclusion chromatography to EV concentration and deep-proteomic profiling using data-independent acquisition. From as little as 200 µL of plasma per patient in a cohort of three healthy donors and six melanoma patients, we identified and quantified 2896 EV-associated proteins, achieving a 3.5-fold increase in depth compared to previously published melanoma studies. To compare the EV-proteome to unenriched blood, we employed an automated workflow to deplete the 14 most abundant proteins from plasma and serum and thereby approximately doubled protein group identifications versus native blood. The EV proteome diverged from corresponding unenriched plasma and serum, and unlike the latter, separated healthy donor and melanoma patient samples. Furthermore, known melanoma markers, such as MCAM, TNC, and TGFBI, were upregulated in melanoma EVs but not in depleted melanoma plasma, highlighting the specific information contained in EVs. Overall, EVs were significantly enriched in intact membrane proteins and proteins related to SNARE protein interactions and T-cell biology. Taken together, we demonstrated the increased sensitivity of an EV-based proteomic workflow that can be easily applied to larger melanoma cohorts and other indications.
Collapse
Affiliation(s)
- Evelyn Lattmann
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | - Luca Räss
- Biognosys AG, Schlieren, Switzerland
| | | | - Julia M Martínez Gómez
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | - Valérie Lapaire
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | | | | | | | - Lars M Steinmetz
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland.
| |
Collapse
|
2
|
Whiteside TL, Diergaarde B, Hong CS. Tumor-Derived Exosomes (TEX) and Their Role in Immuno-Oncology. Int J Mol Sci 2021; 22:ijms22126234. [PMID: 34207762 PMCID: PMC8229953 DOI: 10.3390/ijms22126234] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) play a key role in health and disease, including cancer. Tumors produce a mix of EVs differing in size, cellular origin, biogenesis and molecular content. Small EVs (sEV) or exosomes are a subset of 30-150 nm (virus-size) vesicles originating from the multivesicular bodies (MVBs) and carrying a cargo that in its content and topography approximates that of a parent cell. Tumor-derived exosomes (TEX) present in all body fluids of cancer patients, are considered promising candidates for a liquid tumor biopsy. TEX also mediate immunoregulatory activities: they maintain a crosstalk between the tumor and various non-malignant cells, including immunocytes. Effects that EVs exert on immune cells may be immunosuppressive or immunostimulatory. Here, we review the available data for TEX interactions with immunocytes, focusing on strategies that allow isolation from plasma and separation of TEX from sEV produced by non-malignant cells. Immune effects mediated by either of the subsets can now be distinguished and measured. The approach has allowed for the comparison of molecular and functional profiles of the two sEV fractions in plasma of cancer patients. While TEX carried an excess of immunosuppressive proteins and inhibited immune cell functions in vitro and in vivo, the sEV derived from non-malignant cells, including CD3(+)T cells, were variably enriched in immunostimulatory proteins and could promote functions of immunocytes. Thus, sEV in plasma of cancer patients are heterogenous, representing a complex molecular network which is not evident in healthy donors' plasma. Importantly, TEX appear to be able to reprogram functions of non-malignant CD3(+)T cells inducing them to produce CD3(+)sEV enriched in immunosuppressive proteins. Ratios of stimulatory/inhibitory proteins carried by TEX and by CD3(+)sEV derived from reprogrammed non-malignant cells vary broadly in patients and appear to negatively correlate with disease progression. Simultaneous capture from plasma and functional/molecular profiling of TEX and the CD3(+)sEV fractions allows for defining their role as cancer biomarkers and as monitors of cancer patients' immune competence, respectively.
Collapse
Affiliation(s)
- Theresa L. Whiteside
- Department of Pathology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- Correspondence: ; Tel.: +1-(412)-624-0096; Fax: +1-(412)-623-0264
| | - Brenda Diergaarde
- Department of Human Genetics and UPMC Hillman Cancer Center, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15213, USA;
| | - Chang-Sook Hong
- Department of Pathology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| |
Collapse
|
3
|
Immunoaffinity-Based Isolation of Melanoma Cell-Derived and T Cell-Derived Exosomes from Plasma of Melanoma Patients. Methods Mol Biol 2021; 2265:305-321. [PMID: 33704724 DOI: 10.1007/978-1-0716-1205-7_23] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tumor-derived exosomes (TEX), a subset of small extracellular vesicles (EVs) which originate from the endocytic compartment of tumor cells, are emerging as key players in cancer progression. TEX circulate freely in patients' body fluids and transfer bioactive cargos from tumor to various recipient cells. The molecular cargo of melanoma cell-derived exosomes (MTEX) mimics that of the tumor, and MTEX serve as a liquid biopsy that provides potentially useful information for cancer diagnosis, prognosis, or responses to therapy. Plasma of melanoma patients contains a mix of MTEX and exosomes produced by nonmalignant cells (NMTEX). Isolation of these exosome subtypes from the bulk of plasma exosomes is necessary to evaluate contributions of each as potential biomarkers of melanoma progression and outcome. Here, methods for separation of MTEX from T cell-derived exosomes from a single small volume of plasma and their subsequent molecular and functional characterization are described. Following size exclusion chromatography (SEC) to isolate total plasma exosomes, immune affinity-based capture of MTEX with anti-CSPG4 antibody and then of exosomes produced by T cells with anti-CD3 antibody is used to sequentially isolate the two subsets. This immune capture method enables the recovery of MTEX and CD3+ exosomes in quantities sufficient both for molecular profiling by flow cytometry or western blotting and for functional analyses.
Collapse
|
4
|
Leuci V, Donini C, Grignani G, Rotolo R, Mesiano G, Fiorino E, Gammaitoni L, D'Ambrosio L, Merlini A, Landoni E, Medico E, Capellero S, Giraudo L, Cattaneo G, Iaia I, Pignochino Y, Basiricò M, Vigna E, Pisacane A, Fagioli F, Ferrone S, Aglietta M, Dotti G, Sangiolo D. CSPG4-Specific CAR.CIK Lymphocytes as a Novel Therapy for the Treatment of Multiple Soft-Tissue Sarcoma Histotypes. Clin Cancer Res 2020; 26:6321-6334. [PMID: 32900797 DOI: 10.1158/1078-0432.ccr-20-0357] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/14/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE No effective therapy is available for unresectable soft-tissue sarcomas (STS). This unmet clinical need prompted us to test whether chondroitin sulfate proteoglycan 4 (CSPG4)-specific chimeric antigen receptor (CAR)-redirected cytokine-induced killer lymphocytes (CAR.CIK) are effective in eliminating tumor cells derived from multiple STS histotypes in vitro and in immunodeficient mice. EXPERIMENTAL DESIGN The experimental platform included patient-derived CAR.CIK and cell lines established from multiple STS histotypes. CAR.CIK were transduced with a retroviral vector encoding second-generation CSPG4-specific CAR (CSPG4-CAR) with 4-1BB costimulation. The functional activity of CSPG4-CAR.CIK was explored in vitro, in two- and three-dimensional STS cultures, and in three in vivo STS xenograft models. RESULTS CSPG4-CAR.CIK were efficiently generated from patients with STS. CSPG4 was highly expressed in multiple STS histotypes by in silico analysis and on all 16 STS cell lines tested by flow cytometry. CSPG4-CAR.CIK displayed superior in vitro cytolytic activity against multiple STS histotypes as compared with paired unmodified control CIK. CSPG4-CAR.CIK also showed strong antitumor activity against STS spheroids; this effect was associated with tumor recruitment, infiltration, and matrix penetration. CSPG4-CAR.CIK significantly delayed or reversed tumor growth in vivo in three STS xenograft models (leiomyosarcoma, undifferentiated pleomorphic sarcoma, and fibrosarcoma). Tumor growth inhibition persisted for up to 2 weeks following the last administration of CSPG4-CAR.CIK. CONCLUSIONS This study has shown that CSPG4-CAR.CIK effectively targets multiple STS histotypes in vitro and in immunodeficient mice. These results provide a strong rationale to translate the novel strategy we have developed into a clinical setting.
Collapse
Affiliation(s)
- Valeria Leuci
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Chiara Donini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Ramona Rotolo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Giulia Mesiano
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Erika Fiorino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | | | - Alessandra Merlini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Elisa Landoni
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Enzo Medico
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Sonia Capellero
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Lidia Giraudo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Giulia Cattaneo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Ilenia Iaia
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Ymera Pignochino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Marco Basiricò
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Elisa Vigna
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Franca Fagioli
- Pediatric Onco-Hematology, Division of Stem Cell Transplantation and Cellular Therapy, Regina Margherita Children's Hospital, University of Turin, Turin, Italy
| | - Soldano Ferrone
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Massimo Aglietta
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Dario Sangiolo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy. .,Department of Oncology, University of Torino, Turin, Italy
| |
Collapse
|
5
|
Theodoraki MN, Hong CS, Donnenberg VS, Donnenberg AD, Whiteside TL. Evaluation of Exosome Proteins by on-Bead Flow Cytometry. Cytometry A 2020; 99:372-381. [PMID: 33448645 DOI: 10.1002/cyto.a.24193] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022]
Abstract
Exosomes, recently re-named "small extracellular vesicles" or "sEV," are emerging as an intercellular communication system. Quantification of the molecular cargo exosomes carry by on-bead flow cytometry is needed for defining their role in information transfer and in human disease. Exosomes (sEV) isolated from cell supernatants or plasma of cancer patients by size-exclusion chromatography were captured by biotinylated antibodies specific for antigens in the exosome cargo (e.g., tetraspanins) and placed on streptavidin-labeled beads. Detection was performed with pretitered fluorochrome-labeled antibodies of desired specificity. The data were acquired in a conventional cytometer, and molecules of equivalent soluble fluorochrome (MESF) beads were used to quantify the number of fluorescent molecules bound per bead. Isotype antibody controls were obligatory. The mean fluorescence intensity (MFI) value of each sample was converted into MESF units, and the separation index (SI), which quantifies separation of stained and isotype control beads, was determined. Various proteins identified by labeled antibodies were quantified on the surface of tumor cell-derived exosomes. To identify intravesicular cargo, such as cytokines or chemokines, exosomes were lysed with 0.3% Triton-100, and the proteins in lysates were loaded on aldehyde/sulfate latex beads for flow cytometry. Examples of quantitative surface and/or intravesicular on-bead flow cytometry for exosomes produced by various cells or present in body fluids of cancer patients are provided. On-bead flow cytometry standardized for use with conventional cytometers is a useful method for protein detection and quantitation in exosomes isolated from supernatants of cell lines or plasma of patients with cancer. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Marie-Nicole Theodoraki
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Ulm, Ulm, Germany.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Chang-Sook Hong
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Vera S Donnenberg
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Albert D Donnenberg
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Chondroitin sulfate proteoglycan 4 enhanced melanoma motility and growth requires a cysteine in the core protein transmembrane domain. Melanoma Res 2020; 29:365-375. [PMID: 31140988 DOI: 10.1097/cmr.0000000000000574] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Chondroitin sulfate proteoglycan 4 (CSPG4) is a cell surface proteoglycan that enhances malignant potential in melanoma and several other tumor types. CSPG4 functions as a transmembrane scaffold in melanoma cells to activate oncogenic signaling pathways such as focal adhesion kinase (FAK) and extracellular signal regulated kinases 1,2, that control motility, invasion and anchorage independent growth. Here, we demonstrate that CSPG4 promotes directional motility and anchorage independent growth of melanoma cells by organizing and positioning a signaling complex containing activated FAK to lipid rafts within the plasma membrane of migrating cells. This FAK-containing signal transduction platform, which consists of syntenin-1, active Src and caveolin-1 requires the cytoplasmic domain of CSPG4 for assembly. Enhanced directional motility promoted by this complex also requires a CSPG4 transmembrane cysteine residue C2230. Substituting C2230 with alanine (CSPG4) still permits assembly of the signaling complex, however Src remains in an inactive state. CSPG4 also fails to promote anchorage independent growth and activation of extracellular signal regulated kinases 1,2. Therapies that target the transmembrane domain of CSPG4 could be a novel strategy for limiting progression by disrupting its function as a compartmentalized motogenic and growth-promoting oncogenic signaling node.
Collapse
|
7
|
Jing J, Du Z, Ji S, Han K. Urinary proteome analysis of acute hypercoagulable state in rat model induced by ε-aminocaproic acid. Biomed Pharmacother 2018; 110:275-284. [PMID: 30513505 DOI: 10.1016/j.biopha.2018.11.148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
The hypercoagulable state occurs in a group of prothrombotic disorders associated with an increased risk for thromboembolic events, but it is difficult to diagnose due to the lack of available biomarkers. This study aimed to investigate systematic changes of urinary proteome in acute hypercoagulable state induced by certain antifibrinolytics. To reduce the effects of both genetic and environmental factors on the urinary proteome, we used a rat model of acute hypercoagulable state induced by an antifibrinolytic agent ε-aminocaproic acid, resembling human hypercoagulable state. Urine samples were collected during acute hypercoagulable state for analysis by liquid chromatography-tandem mass spectrometry (LCMS/MS). Of 65 significantly changed proteins in acute hypercoagulable state, 38 proteins had human orthologs, and 18 proteins were identified as stable in normal human urine. None of the identified proteins have been found to be clotting factors, but 4 proteins are known to be involved in the regulation of blood coagulation factors. Two proteins were verified as the markers associated with acute hypercoagulable state by Western blot analysis. In addition, four common differential urinary proteins have been found in acute hypercoagulable state induced by another antifibrinolytics tranexamic acid. These four proteins are potential biomarkers for early diagnosis of hypercoagulable state to prevent the development of thrombotic diseases.
Collapse
Affiliation(s)
- Jian Jing
- Beijing Key Lab of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Xinwai St 19, Haidian District, Beijing, 100875, China.
| | - Zhenhuan Du
- Beijing Key Lab of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Xinwai St 19, Haidian District, Beijing, 100875, China
| | - Songyang Ji
- Beijing Key Lab of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Xinwai St 19, Haidian District, Beijing, 100875, China
| | - Keqiang Han
- Beijing Key Lab of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Xinwai St 19, Haidian District, Beijing, 100875, China
| |
Collapse
|
8
|
Whiteside TL. The potential of tumor-derived exosomes for noninvasive cancer monitoring: an update. Expert Rev Mol Diagn 2018; 18:1029-1040. [PMID: 30406709 DOI: 10.1080/14737159.2018.1544494] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Liquid biopsy platforms are being actively developed in the biomarker field. Extracellular vesicles (EVs), especially the tumor-derived exosome (TEX) subsets of EVs, represent a platform that allows for molecular and genetic profiling of parent tumor cells. TEX are ubiquitous in body fluids of cancer patients and are promising clinically relevant surrogates of cancer cells. Areas covered: Isolation from body fluids of cancer patients and subsetting of exosomes based on immunoaffinity capture offers a means of evaluating proteins, lipids, nucleic acids and other molecular contents that are a characteristic of TEX and exosomes produced by reprogrammed normal cells. The same liquid biopsy can inform about the status of a tumor and simultaneously evaluate the competency of immune cells to mediate anti-tumor activities. Expert commentary: TEX and reprogrammed non-TEX isolated from plasma of cancer patients have the potential to become non-invasive biomarkers of cancer diagnosis, prognosis and response to therapies.
Collapse
Affiliation(s)
- Theresa L Whiteside
- a Departments of Pathology, Immunology and Otolaryngology , University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center , Pittsburgh , PA , USA
| |
Collapse
|
9
|
Sharma P, Ludwig S, Muller L, Hong CS, Kirkwood JM, Ferrone S, Whiteside TL. Immunoaffinity-based isolation of melanoma cell-derived exosomes from plasma of patients with melanoma. J Extracell Vesicles 2018; 7:1435138. [PMID: 29511460 PMCID: PMC5827723 DOI: 10.1080/20013078.2018.1435138] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/26/2018] [Indexed: 12/12/2022] Open
Abstract
Tumour-derived exosomes (TEX) are a subset of extracellular vesicles (EVs) present in body fluids of patients with cancer. The role of this exosome subset in melanoma progression has been of interest ever since ex vivo studies of exosomes produced by melanoma cell lines were shown to suppress anti-melanoma immune responses. To study the impact of melanoma-derived exosomes (MTEX) present in patients’ plasma on melanoma progression, isolation of MTEX from total plasma exosomes is necessary. We have developed an immunoaffinity-based method for MTEX capture from plasma of melanoma patients. Using mAb 763.74 specific for the CSPG4 epitope uniquely expressed on melanoma cells, we separated MTEX from non-tumour cell-derived exosomes and evaluated the protein cargo of both fractions by quantitative flow cytometry. Melanoma-associated antigens were carried by MTEX but were not detectable in exosomes produced by normal cells. Separation of plasma-derived MTEX from non-MTEX provides an opportunity for future evaluation of MTEX as potential biomarkers of melanoma progression and as surrogates of melanoma in tumour liquid biopsy studies.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sonja Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Otolaryngology and Head & Neck Surgery, University of Duisburg-Essen, Essen, Germany
| | - Laurent Muller
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Head and Neck Surgery, University of Basel, Basel, Switzerland
| | - Chang Sook Hong
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - John M Kirkwood
- Department of Medicine, University of Pittsburgh School of Medicine and UPMC Hillman Cancer, Pittsburgh, PA, USA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
10
|
Fenton M, Whiteside TL, Ferrone S, Boyiadzis M. Chondroitin sulfate proteoglycan-4 (CSPG4)-specific monoclonal antibody 225.28 in detection of acute myeloid leukemia blasts. Oncol Res 2017; 22:117-21. [PMID: 25706398 PMCID: PMC7838444 DOI: 10.3727/096504014x14174484758503] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chondroitin sulfate proteoglycan-4 (CSPG4), a membrane-bound proteoglycan known to be expressed on the surface of malignant cells, has a restricted distribution in normal tissues. CSPG4 is a potential candidate tumor marker. We investigate CSPG4 expression on blasts in newly diagnosed acute myeloid leukemia (AML) patients and its relation with cytogenetic abnormalities and molecular markers known to have prognostic significance in this disease. Using hybridoma technology, we generated a specific monoclonal antibody (mAb), mAb 225.28, reactive with CSPG4. Blast samples obtained from the peripheral blood of newly diagnosed AML patients were analyzed for CSPG4 expression using the CSPG4-specific mAb and multiparameter flow cytometry. The results were correlated with cytogenetic and molecular characteristics of AML. CSPG4 was found to be expressed on a variable fraction of leukemic blasts in all AML patients with different leukemia morphology, including monoblastic cases. Reactivity of CSPG4-specific mAb with leukemic blasts was not limited to those with the rearranged MLL gene. CSPG4 was also expressed on AML blasts with a complex karyotype, FLT3 mutation, or NPM1 mutation. The results indicate that CSPG4 is expressed and detectable by flow cytometry using the mAb 225.28 on a proportion of blasts of all subtypes of AML irrespective of cytogenetic and molecular abnormalities. mAb 225.28 could be useful in detecting AML blasts by flow cytometry.
Collapse
Affiliation(s)
- Moon Fenton
- Division of Hematology and Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
11
|
Moreno-Traspas R, Vujic I, Sanlorenzo M, Ortiz-Urda S. New insights in melanoma biomarkers: long-noncoding RNAs. Melanoma Manag 2016; 3:195-205. [PMID: 30190889 DOI: 10.2217/mmt-2016-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/17/2016] [Indexed: 11/21/2022] Open
Abstract
Melanoma is one of the leading cancers worldwide, distinguished for its malignancy and low survival rates. Although the poor outcome could improve with an early diagnosis and a good monitoring of the disease, current melanoma biomarkers display several limitations which make them useless. Interestingly, long-noncoding RNAs are secreted into the bloodstream inside exosomes by a wide range of malignant cells, and several of them have been validated as promising circulating molecular signatures of other tumors, but not melanoma. In this review we propose to explore the booming field of long-noncoding RNAs in order to find potential candidates to be tested as novel melanoma biomarkers, with the ultimate goal of improving melanoma detection, diagnosis and prognosis.
Collapse
Affiliation(s)
- Ricardo Moreno-Traspas
- Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA.,Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA
| | - Igor Vujic
- Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA.,The Rudolfstiftung Hospital, Academic Teaching Hospital, Department of Dermatology, Medical University Vienna, Juchgasse 25, 1030 Vienna, Austria.,Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA.,The Rudolfstiftung Hospital, Academic Teaching Hospital, Department of Dermatology, Medical University Vienna, Juchgasse 25, 1030 Vienna, Austria
| | - Martina Sanlorenzo
- Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA.,Department of Medical Sciences, Section of Dermatology, University of Turin, Italy.,Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA.,Department of Medical Sciences, Section of Dermatology, University of Turin, Italy
| | - Susana Ortiz-Urda
- Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA.,Department of Dermatology, University of California San Francisco, Mt. Zion Cancer Research Center, 2340 Sutter Street N461, San Francisco, CA 94115, USA
| |
Collapse
|
12
|
Abstract
Tumor-derived exosomes (TEX) are emerging as a new type of cancer biomarker. TEX are membrane-bound, virus-size vesicles of endocytic origin present in all body fluids of cancer patients. Based on the expanding albeit incomplete knowledge of their biogenesis, secretion by tumor cells and cancer cell-specific molecular and genetic contents, TEX are viewed as promising, clinically-relevant surrogates of cancer progression and response to therapy. Preliminary proteomic, genetic and functional profiling of tumor cell-derived or cancer plasma-derived exosomes confirms their unique characteristics. Alterations in protein or nucleic acid profiles of exosomes in plasma of cancer patients responding to therapies appear to correlate with clinical endpoints. However, methods for TEX isolation and separation from the bulk of human plasma-derived exosomes are not yet established and their role as biomarkers remains to be confirmed. Further development and validation of TEX as noninvasive, liquid equivalents of tumor biopsies are necessary to move this effort forward.
Collapse
Affiliation(s)
- Theresa L. Whiteside
- University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, Departments of Pathology, Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, , Phone: 412-624-0096, FAX: 412-624-0264
| |
Collapse
|