The Cancer Proteome

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Biomarkers".

Deadline for manuscript submissions: closed (30 September 2020) | Viewed by 71404

Special Issue Editor


E-Mail Website
Guest Editor
Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
Interests: cancer biomarkers; proteomics; cancer surfaceome; immunooncology
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Interest in the cancer proteome has accelerated—substantially driven by the need to elucidate altered protein expression at the tumor cell level on a compartment-by-compartment basis; this includes analyses involving the secretome, exosomes, surfaceome, immunopeptidome, phosphoproteome, glycoproteome, and other post-translational modifications. Likewise, there is a need to elucidate changes in the tumor microenvironment that drive tumor development and progression, and to identify prognostic and predictive markers as well as novel targets for therapy, particularly antigens relevant to immunotherapy and vaccine development. This Special Issue intends to showcase many of the numerous facets of the cancer proteome and related translational applications and innovative technologies.

Prof. Dr. Samir M. Hanash
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cancer secretome
  • exosomes
  • surfaceome
  • immunopeptidome
  • phosphoproteome
  • glycoproteome
  • post-translational modifications

Published Papers (17 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review, Other

3 pages, 185 KiB  
Editorial
Proteomic Dissection of Exosome Cargo: Progress and Future Perspectives
by Shahab Uddin and Aamir Ahmad
Cancers 2023, 15(17), 4292; https://doi.org/10.3390/cancers15174292 - 28 Aug 2023
Cited by 1 | Viewed by 717
Abstract
The interest in exosomes in cancer research and treatment has increased exponentially in the past few years [...] Full article
(This article belongs to the Special Issue The Cancer Proteome)

Research

Jump to: Editorial, Review, Other

18 pages, 7349 KiB  
Article
Temporal Quantitative Proteomics Reveals Proteomic and Phosphoproteomic Alterations Associated with Adaptive Response to Hypoxia in Melanoma Cells
by Keshava K. Datta, Parthiban Periasamy, Sonali V. Mohan, Rebekah Ziegman and Harsha Gowda
Cancers 2021, 13(9), 2175; https://doi.org/10.3390/cancers13092175 - 30 Apr 2021
Cited by 4 | Viewed by 2590
Abstract
Hypoxia is a common feature in various solid tumours, including melanoma. Cancer cells in hypoxic environments are resistant to both chemotherapy and radiation. Hypoxia is also associated with immune suppression. Identification of proteins and pathways that regulate cancer cell survival in hypoxic environments [...] Read more.
Hypoxia is a common feature in various solid tumours, including melanoma. Cancer cells in hypoxic environments are resistant to both chemotherapy and radiation. Hypoxia is also associated with immune suppression. Identification of proteins and pathways that regulate cancer cell survival in hypoxic environments can reveal potential vulnerabilities that can be exploited to improve the efficacy of anticancer therapies. We carried out temporal proteomic and phosphoproteomic profiling in melanoma cell lines to identify hypoxia-induced protein expression and phosphorylation changes. By employing a TMT-based quantitative proteomics strategy, we report the identification and quantitation of >7000 proteins and >10,000 phosphosites in melanoma cell lines grown in hypoxia. Proteomics data show metabolic reprogramming as one of the prominent adaptive responses in hypoxia. We identify several novel hypoxia-mediated phosphorylation changes that have not been reported before. They reveal kinase signalling pathways that are potentially involved in modulating cellular response to hypoxia. In addition to known protein expression changes, we identify several novel proteomic alterations associated with adaptive response to hypoxia. We show that cancer cells require the ubiquitin–proteasome system to survive in both normoxia and hypoxia. Inhibition of proteasome activity affects cell survival and may provide a novel therapeutic avenue to target cancer cells in hypoxia. Our study can serve as a valuable resource to pursue novel candidates to target hypoxia in cancers and improve the efficacy of anticancer therapies. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

21 pages, 3514 KiB  
Article
Interactome Mapping of eIF3A in a Colon Cancer and an Immortalized Embryonic Cell Line Using Proximity-Dependent Biotin Identification
by Diep-Khanh Vo, Alexander Engler, Darko Stoimenovski, Roland Hartig, Thilo Kaehne, Thomas Kalinski, Michael Naumann, Johannes Haybaeck and Norbert Nass
Cancers 2021, 13(6), 1293; https://doi.org/10.3390/cancers13061293 - 14 Mar 2021
Cited by 1 | Viewed by 3555
Abstract
Translation initiation comprises complex interactions of eukaryotic initiation factor (eIF) subunits and the structural elements of the mRNAs. Translation initiation is a key process for building the cell’s proteome. It not only determines the total amount of protein synthesized but also controls the [...] Read more.
Translation initiation comprises complex interactions of eukaryotic initiation factor (eIF) subunits and the structural elements of the mRNAs. Translation initiation is a key process for building the cell’s proteome. It not only determines the total amount of protein synthesized but also controls the translation efficiency for individual transcripts, which is important for cancer or ageing. Thus, understanding protein interactions during translation initiation is one key that contributes to understanding how the eIF subunit composition influences translation or other pathways not yet attributed to eIFs. We applied the BioID technique to two rapidly dividing cell lines (the immortalized embryonic cell line HEK-293T and the colon carcinoma cell line HCT-166) in order to identify interacting proteins of eIF3A, a core subunit of the eukaryotic initiation factor 3 complex. We identified a total of 84 interacting proteins, with very few proteins being specific to one cell line. When protein biosynthesis was blocked by thapsigargin-induced endoplasmic reticulum (ER) stress, the interacting proteins were considerably smaller in number. In terms of gene ontology, although eIF3A interactors are mainly part of the translation machinery, protein folding and RNA binding were also found. Cells suffering from ER-stress show a few remaining interactors which are mainly ribosomal proteins or involved in RNA-binding. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Graphical abstract

25 pages, 4691 KiB  
Article
Leucine-Rich Diet Modulates the Metabolomic and Proteomic Profile of Skeletal Muscle during Cancer Cachexia
by Bread Cruz, André Oliveira, Lais Rosa Viana, Leisa Lopes-Aguiar, Rafael Canevarolo, Maiara Caroline Colombera, Rafael Rossi Valentim, Fernanda Garcia-Fóssa, Lizandra Maia de Sousa, Bianca Gazieri Castelucci, Sílvio Roberto Consonni, Daniel Martins-de-Souza, Marcelo Bispo de Jesus, Steven Thomas Russell and Maria Cristina Cintra Gomes-Mardondes
Cancers 2020, 12(7), 1880; https://doi.org/10.3390/cancers12071880 - 13 Jul 2020
Cited by 15 | Viewed by 3933
Abstract
Background: Cancer-cachexia induces a variety of metabolic disorders, including skeletal muscle imbalance. Alternative therapy, as nutritional supplementation with leucine, shows a modulatory effect over tumour damage in vivo and in vitro. Method: Adult rats distributed into Control (C), Walker tumour-bearing (W), control fed [...] Read more.
Background: Cancer-cachexia induces a variety of metabolic disorders, including skeletal muscle imbalance. Alternative therapy, as nutritional supplementation with leucine, shows a modulatory effect over tumour damage in vivo and in vitro. Method: Adult rats distributed into Control (C), Walker tumour-bearing (W), control fed a leucine-rich diet (L), and tumour-bearing fed a leucine-rich diet (WL) groups had the gastrocnemius muscle metabolomic and proteomic assays performed in parallel to in vitro assays. Results: W group presented an affected muscle metabolomic and proteomic profile mainly related to energy generation and carbohydrates catabolic processes, but leucine-supplemented group (WL) recovered the energy production. In vitro assay showed that cell proliferation, mitochondria number and oxygen consumption were higher under leucine effect than the tumour influence. Muscle proteomics results showed that the main affected cell component was mitochondria, leading to an impacted energy generation, including impairment in proteins of the tricarboxylic cycle and carbohydrates catabolic processes, which were modulated and improved by leucine treatment. Conclusion: In summary, we showed a beneficial effect of leucine upon mitochondria, providing information about the muscle glycolytic pathways used by this amino acid, where it can be associated with the preservation of morphometric parameters and consequent protection against the effects of cachexia. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

15 pages, 668 KiB  
Article
Identification of a Blood-Based Protein Biomarker Panel for Lung Cancer Detection
by Victoria El-Khoury, Anna Schritz, Sang-Yoon Kim, Antoine Lesur, Katriina Sertamo, François Bernardin, Konstantinos Petritis, Patrick Pirrotte, Cheryl Selinsky, Jeffrey R. Whiteaker, Haizhen Zhang, Jacob J. Kennedy, Chenwei Lin, Lik Wee Lee, Ping Yan, Nhan L. Tran, Landon J. Inge, Khaled Chalabi, Georges Decker, Rolf Bjerkvig, Amanda G. Paulovich, Guy Berchem and Yeoun Jin Kimadd Show full author list remove Hide full author list
Cancers 2020, 12(6), 1629; https://doi.org/10.3390/cancers12061629 - 19 Jun 2020
Cited by 23 | Viewed by 4564
Abstract
Lung cancer is the deadliest cancer worldwide, mainly due to its advanced stage at the time of diagnosis. A non-invasive method for its early detection remains mandatory to improve patients’ survival. Plasma levels of 351 proteins were quantified by Liquid Chromatography-Parallel Reaction Monitoring [...] Read more.
Lung cancer is the deadliest cancer worldwide, mainly due to its advanced stage at the time of diagnosis. A non-invasive method for its early detection remains mandatory to improve patients’ survival. Plasma levels of 351 proteins were quantified by Liquid Chromatography-Parallel Reaction Monitoring (LC-PRM)-based mass spectrometry in 128 lung cancer patients and 93 healthy donors. Bootstrap sampling and least absolute shrinkage and selection operator (LASSO) penalization were used to find the best protein combination for outcome prediction. The PanelomiX platform was used to select the optimal biomarker thresholds. The panel was validated in 48 patients and 49 healthy volunteers. A 6-protein panel clearly distinguished lung cancer from healthy individuals. The panel displayed excellent performance: area under the receiver operating characteristic curve (AUC) = 0.999, positive predictive value (PPV) = 0.992, negative predictive value (NPV) = 0.989, specificity = 0.989 and sensitivity = 0.992. The panel detected lung cancer independently of the disease stage. The 6-protein panel and other sub-combinations displayed excellent results in the validation dataset. In conclusion, we identified a blood-based 6-protein panel as a diagnostic tool in lung cancer. Used as a routine test for high- and average-risk individuals, it may complement currently adopted techniques in lung cancer screening. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

30 pages, 5981 KiB  
Article
Proteomic Profiling of Retinoblastoma-Derived Exosomes Reveals Potential Biomarkers of Vitreous Seeding
by Angela Galardi, Marta Colletti, Chiara Lavarello, Virginia Di Paolo, Paolo Mascio, Ida Russo, Raffaele Cozza, Antonino Romanzo, Paola Valente, Rita De Vito, Luisa Pascucci, Hector Peinado, Angel M. Carcaboso, Andrea Petretto, Franco Locatelli and Angela Di Giannatale
Cancers 2020, 12(6), 1555; https://doi.org/10.3390/cancers12061555 - 12 Jun 2020
Cited by 34 | Viewed by 4248
Abstract
Retinoblastoma (RB) is the most common tumor of the eye in early childhood. Although recent advances in conservative treatment have greatly improved the visual outcome, local tumor control remains difficult in the presence of massive vitreous seeding. Traditional biopsy has long been considered [...] Read more.
Retinoblastoma (RB) is the most common tumor of the eye in early childhood. Although recent advances in conservative treatment have greatly improved the visual outcome, local tumor control remains difficult in the presence of massive vitreous seeding. Traditional biopsy has long been considered unsafe in RB, due to the risk of extraocular spread. Thus, the identification of new biomarkers is crucial to design safer diagnostic and more effective therapeutic approaches. Exosomes, membrane-derived nanovesicles that are secreted abundantly by aggressive tumor cells and that can be isolated from several biological fluids, represent an interesting alternative for the detection of tumor-associated biomarkers. In this study, we defined the protein signature of exosomes released by RB tumors (RBT) and vitreous seeding (RBVS) primary cell lines by high resolution mass spectrometry. A total of 5666 proteins were identified. Among these, 5223 and 3637 were expressed in exosomes RBT and one RBVS group, respectively. Gene enrichment analysis of exclusively and differentially expressed proteins and network analysis identified in RBVS exosomes upregulated proteins specifically related to invasion and metastasis, such as proteins involved in extracellular matrix (ECM) remodeling and interaction, resistance to anoikis and the metabolism/catabolism of glucose and amino acids. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

20 pages, 2564 KiB  
Article
Plasma-Derived Extracellular Vesicles Convey Protein Signatures That Reflect Pathophysiology in Lung and Pancreatic Adenocarcinomas
by Johannes F. Fahrmann, Xiangying Mao, Ehsan Irajizad, Hiroyuki Katayama, Michela Capello, Ichidai Tanaka, Taketo Kato, Ignacio I. Wistuba, Anirban Maitra, Edwin J. Ostrin, Samir M. Hanash and Jody Vykoukal
Cancers 2020, 12(5), 1147; https://doi.org/10.3390/cancers12051147 - 02 May 2020
Cited by 17 | Viewed by 3567
Abstract
Using a combination of mass-spectrometry and aptamer array-based proteomics, we characterized the protein features of circulating extracellular vesicles (EVs) in the context of lung (LUAD) and pancreatic ductal (PDAC) adenocarcinomas. We profiled EVs isolated from conditioned media of LUAD and PDAC cell lines [...] Read more.
Using a combination of mass-spectrometry and aptamer array-based proteomics, we characterized the protein features of circulating extracellular vesicles (EVs) in the context of lung (LUAD) and pancreatic ductal (PDAC) adenocarcinomas. We profiled EVs isolated from conditioned media of LUAD and PDAC cell lines to identify EV-associated protein cargoes released by these cancer cell types. Analysis of the resulting data identified LUAD and PDAC specific and pan-adenocarcinoma EV protein signatures. Bioinformatic analyses confirmed enrichment of proteins annotated to vesicle-associated processes and intracellular compartments, as well as representation of cancer hallmark functions and processes. Analysis of upstream regulator networks indicated significant enrichment of TP53, MYC, TGFB1 and KRAS-driven network effectors (p = 1.69 × 10−77–2.93 × 10−49) manifest in the adenocarcinoma sEV protein cargoes. We extended these findings by profiling the proteome of EVs isolated from lung (N = 15) and pancreatic ductal (N = 6) adenocarcinoma patient plasmas obtained at time of diagnosis, along with EVs derived from matched healthy controls (N = 21). Exploration of these proteomic data revealed abundant protein features in the plasma EVs with capacity to distinguish LUAD and PDAC cases from controls, including features yielding higher performance in the plasma EV isolates relative to unfractionated plasmas. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

22 pages, 6642 KiB  
Article
Mass Spectrometry Imaging Reveals Neutrophil Defensins as Additional Biomarkers for Anti-PD-(L)1 Immunotherapy Response in NSCLC Patients
by Eline Berghmans, Julie Jacobs, Christophe Deben, Christophe Hermans, Glenn Broeckx, Evelien Smits, Evelyne Maes, Jo Raskin, Patrick Pauwels and Geert Baggerman
Cancers 2020, 12(4), 863; https://doi.org/10.3390/cancers12040863 - 02 Apr 2020
Cited by 19 | Viewed by 4395
Abstract
(1) Background: Therapeutic blocking of the interaction between programmed death-1 (PD-1) with its ligand PD-L1, an immune checkpoint, is a promising approach to restore the antitumor immune response. Improved clinical outcomes have been shown in different human cancers, including non-small cell lung cancer [...] Read more.
(1) Background: Therapeutic blocking of the interaction between programmed death-1 (PD-1) with its ligand PD-L1, an immune checkpoint, is a promising approach to restore the antitumor immune response. Improved clinical outcomes have been shown in different human cancers, including non-small cell lung cancer (NSCLC). Unfortunately, still a high number of NSCLC patients are treated with immunotherapy without obtaining any clinical benefit, due to the limitations of PD-L1 protein expression as the currently sole predictive biomarker for clinical use; (2) Methods: In this study, we applied mass spectrometry imaging (MSI) to discover new protein biomarkers, and to assess the possible correlation between candidate biomarkers and a positive immunotherapy response by matrix-assisted laser desorption/ionization (MALDI) MSI in 25 formalin-fixed paraffin-embedded (FFPE) pretreatment tumor biopsies (Biobank@UZA); (3) Results: Using MALDI MSI, we revealed that the addition of neutrophil defensin 1, 2 and 3 as pretreatment biomarkers may more accurately predict the outcome of immunotherapy treatment in NSCLC. These results were verified and confirmed with immunohistochemical analyses. In addition, we provide in-vitro evidence of the immune stimulatory effect of neutrophil defensins towards cancer cells; and (4) Conclusions: With proteomic approaches, we have discovered neutrophil defensins as additional prospective biomarkers for an anti-PD-(L)1 immunotherapy response. Thereby, we also demonstrated that the neutrophil defensins contribute in the activation of the immune response towards cancer cells, which could provide a new lead towards an anticancer therapy. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Graphical abstract

16 pages, 2119 KiB  
Article
Proteomic Discovery of Biomarkers to Predict Prognosis of High-Grade Serous Ovarian Carcinoma
by Se Ik Kim, Minsun Jung, Kisoon Dan, Sungyoung Lee, Cheol Lee, Hee Seung Kim, Hyun Hoon Chung, Jae-Weon Kim, Noh Hyun Park, Yong-Sang Song, Dohyun Han and Maria Lee
Cancers 2020, 12(4), 790; https://doi.org/10.3390/cancers12040790 - 26 Mar 2020
Cited by 18 | Viewed by 4761
Abstract
Initial identification of biomarkers predicting the exact prognosis of high-grade serous ovarian carcinoma (HGSOC) is important in precision cancer medicine. This study aimed to investigate prognostic biomarkers of HGSOC through proteomic analysis. We conducted label-free liquid chromatography-mass spectrometry using chemotherapy-naïve, fresh-frozen primary HGSOC [...] Read more.
Initial identification of biomarkers predicting the exact prognosis of high-grade serous ovarian carcinoma (HGSOC) is important in precision cancer medicine. This study aimed to investigate prognostic biomarkers of HGSOC through proteomic analysis. We conducted label-free liquid chromatography-mass spectrometry using chemotherapy-naïve, fresh-frozen primary HGSOC specimens, and compared the results between a favorable prognosis group (progression-free survival (PFS) ≥ 18 months, n = 6) and a poor prognosis group (PFS < 18 months, n = 6). Among 658 differentially expressed proteins, 288 proteins were upregulated in the favorable prognosis group and 370 proteins were upregulated in the poor prognosis group. Using hierarchical clustering, we selected α1-antitrypsin (AAT), nuclear factor-κB (NFKB), phosphomevalonate kinase (PMVK), vascular adhesion protein 1 (VAP1), fatty acid-binding protein 4 (FABP4), platelet factor 4 (PF4), apolipoprotein A1 (APOA1), and α1-acid glycoprotein (AGP) for further validation via immunohistochemical (IHC) staining in an independent set of chemotherapy-naïve primary HGSOC samples (n = 107). Survival analyses revealed that high expression of AAT, NFKB, and PMVK were independent biomarkers for favorable PFS. Conversely, high expression of VAP1, FABP4, and PF4 were identified as independent biomarkers for poor PFS. Furthermore, we constructed models predicting the 18-month PFS by combining clinical variables and IHC results. Through leave-one-out cross-validation, the optimal model was based on initial serum CA-125, germline BRCA1/2 mutations, residual tumors after surgery, International Federation of Gynecology and Obstetrics (FIGO) stage, and expression levels of the six proteins. The present results elucidate the proteomic landscape of HGSOC and six protein biomarkers to predict the prognosis of HGSOC. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

14 pages, 3866 KiB  
Article
Immune-Complexome Analysis Identifies Immunoglobulin-Bound Biomarkers That Predict the Response to Chemotherapy of Pancreatic Cancer Patients
by Giorgia Mandili, Laura Follia, Giulio Ferrero, Hiroyuki Katayama, Wang Hong, Amin A. Momin, Michela Capello, Daniele Giordano, Rosella Spadi, Maria Antonietta Satolli, Andrea Evangelista, Samir M. Hanash, Francesca Cordero and Francesco Novelli
Cancers 2020, 12(3), 746; https://doi.org/10.3390/cancers12030746 - 21 Mar 2020
Cited by 8 | Viewed by 2972
Abstract
Pancreatic Ductal Adenocarcinoma (PDA) is an aggressive malignancy with a very poor outcome. Although chemotherapy (CT) treatment has poor efficacy, it can enhance tumor immunogenicity. Tumor-Associated Antigens (TAA) are self-proteins that are overexpressed in tumors that may induce antibody production and can be [...] Read more.
Pancreatic Ductal Adenocarcinoma (PDA) is an aggressive malignancy with a very poor outcome. Although chemotherapy (CT) treatment has poor efficacy, it can enhance tumor immunogenicity. Tumor-Associated Antigens (TAA) are self-proteins that are overexpressed in tumors that may induce antibody production and can be PDA theranostic targets. However, the prognostic value of TAA-antibody association as Circulating Immune Complexes (CIC) has not yet been elucidated, mainly due to the lack of techniques that lead to their identification. In this study, we show a novel method to separate IgG, IgM, and IgA CIC from sera to use them as prognostic biomarkers of CT response. The PDA Immune-Complexome (IC) was identified using a LTQ-Orbitrap mass spectrometer followed by computational analysis. The analysis of the IC of 37 PDA patients before and after CT revealed differential associated antigens (DAA) for each immunoglobulin class. Our method identified different PDA-specific CIC in patients that were associated with poor prognosis patients. Finally, CIC levels were significantly modified by CT suggesting that they can be used as effective prognostic biomarkers to follow CT response in PDA patients. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

16 pages, 1951 KiB  
Article
Proteome Profiling Uncovers an Autoimmune Response Signature That Reflects Ovarian Cancer Pathogenesis
by Makoto Kobayashi, Hiroyuki Katayama, Ehsan Irajizad, Jody V. Vykoukal, Johannes F. Fahrmann, Deepali L. Kundnani, Chuan-Yih Yu, Yining Cai, Fu Chung Hsiao, Wei-Lei Yang, Zhen Lu, Joseph Celestino, James P. Long, Kim-Ann Do, Karen H. Lu, Jon J. Ladd, Nicole Urban, Robert C. Bast Jr. and Samir M. Hanash
Cancers 2020, 12(2), 485; https://doi.org/10.3390/cancers12020485 - 19 Feb 2020
Cited by 8 | Viewed by 3479
Abstract
Harnessing the immune response to tumor antigens in the form of autoantibodies, which occurs early during tumor development, has relevance to the detection of cancer at early stages. We conducted an initial screen of antigens associated with an autoantibody response in serous ovarian [...] Read more.
Harnessing the immune response to tumor antigens in the form of autoantibodies, which occurs early during tumor development, has relevance to the detection of cancer at early stages. We conducted an initial screen of antigens associated with an autoantibody response in serous ovarian cancer using recombinant protein arrays. The top 25 recombinants that exhibited increased reactivity with cases compared to controls revealed TP53 and MYC, which are ovarian cancer driver genes, as major network nodes. A mass spectrometry based independent analysis of circulating immunoglobulin (Ig)-bound proteins in ovarian cancer and of ovarian cancer cell surface MHC-II bound peptides also revealed a TP53–MYC related network of antigens. Our findings support the occurrence of a humoral immune response to antigens linked to ovarian cancer driver genes that may have utility for early detection applications. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

15 pages, 1773 KiB  
Article
Kinase Inhibitor Treatment of Patients with Advanced Cancer Results in High Tumor Drug Concentrations and in Specific Alterations of the Tumor Phosphoproteome
by Mariette Labots, Thang V. Pham, Richard J. Honeywell, Jaco C. Knol, Robin Beekhof, Richard de Goeij-de Haas, Henk Dekker, Maarten Neerincx, Sander R. Piersma, Johannes C. van der Mijn, Donald L. van der Peet, Martijn R. Meijerink, Godefridus J. Peters, Nicole C.T. van Grieken, Connie R. Jiménez and Henk M.W. Verheul
Cancers 2020, 12(2), 330; https://doi.org/10.3390/cancers12020330 - 01 Feb 2020
Cited by 12 | Viewed by 3089
Abstract
Identification of predictive biomarkers for targeted therapies requires information on drug exposure at the target site as well as its effect on the signaling context of a tumor. To obtain more insight in the clinical mechanism of action of protein kinase inhibitors (PKIs), [...] Read more.
Identification of predictive biomarkers for targeted therapies requires information on drug exposure at the target site as well as its effect on the signaling context of a tumor. To obtain more insight in the clinical mechanism of action of protein kinase inhibitors (PKIs), we studied tumor drug concentrations of protein kinase inhibitors (PKIs) and their effect on the tyrosine-(pTyr)-phosphoproteome in patients with advanced cancer. Tumor biopsies were obtained from 31 patients with advanced cancer before and after 2 weeks of treatment with sorafenib (SOR), erlotinib (ERL), dasatinib (DAS), vemurafenib (VEM), sunitinib (SUN) or everolimus (EVE). Tumor concentrations were determined by LC-MS/MS. pTyr-phosphoproteomics was performed by pTyr-immunoprecipitation followed by LC-MS/MS. Median tumor concentrations were 2–10 µM for SOR, ERL, DAS, SUN, EVE and >1 mM for VEM. These were 2–178 × higher than median plasma concentrations. Unsupervised hierarchical clustering of pTyr-phosphopeptide intensities revealed patient-specific clustering of pre- and on-treatment profiles. Drug-specific alterations of peptide phosphorylation was demonstrated by marginal overlap of robustly up- and downregulated phosphopeptides. These findings demonstrate that tumor drug concentrations are higher than anticipated and result in drug specific alterations of the phosphoproteome. Further development of phosphoproteomics-based personalized medicine is warranted. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

22 pages, 2523 KiB  
Article
Proteogenomics of Colorectal Cancer Liver Metastases: Complementing Precision Oncology with Phenotypic Data
by Bernhard Blank-Landeshammer, Vincent R. Richard, Georgia Mitsa, Maud Marques, André LeBlanc, Laxmikanth Kollipara, Ingo Feldmann, Mathilde Couetoux du Tertre, Karen Gambaro, Suzan McNamara, Alan Spatz, René P. Zahedi, Albert Sickmann, Gerald Batist and Christoph H. Borchers
Cancers 2019, 11(12), 1907; https://doi.org/10.3390/cancers11121907 - 01 Dec 2019
Cited by 12 | Viewed by 4824
Abstract
Hotspot testing for activating KRAS mutations is used in precision oncology to select colorectal cancer (CRC) patients who are eligible for anti-EGFR treatment. However, even for KRASwildtype tumors anti-EGFR response rates are <30%, while mutated-KRAS does not entirely rule out response, [...] Read more.
Hotspot testing for activating KRAS mutations is used in precision oncology to select colorectal cancer (CRC) patients who are eligible for anti-EGFR treatment. However, even for KRASwildtype tumors anti-EGFR response rates are <30%, while mutated-KRAS does not entirely rule out response, indicating the need for improved patient stratification. We performed proteogenomic phenotyping of KRASwildtype and KRASG12V CRC liver metastases (mCRC). Among >9000 proteins we detected considerable expression changes including numerous proteins involved in progression and resistance in CRC. We identified peptides representing a number of predicted somatic mutations, including KRASG12V. For eight of these, we developed a multiplexed parallel reaction monitoring (PRM) mass spectrometry assay to precisely quantify the mutated and canonical protein variants. This allowed phenotyping of eight mCRC tumors and six paired healthy tissues, by determining mutation rates on the protein level. Total KRAS expression varied between tumors (0.47–1.01 fmol/µg total protein) and healthy tissues (0.13–0.64 fmol/µg). In KRASG12V-mCRC, G12V-mutation levels were 42–100%, while one patient had only 10% KRASG12V but 90% KRASwildtype. This might represent a missed therapeutic opportunity: based on hotspot sequencing, the patient was excluded from anti-EGFR treatment and instead received chemotherapy, while PRM-based tumor-phenotyping indicates the patient might have benefitted from anti-EGFR therapy. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

41 pages, 3264 KiB  
Review
Proteomics as a Complementary Technique to Characterize Bladder Cancer
by Rubén López-Cortés, Sergio Vázquez-Estévez, Javier Álvarez Fernández and Cristina Núñez
Cancers 2021, 13(21), 5537; https://doi.org/10.3390/cancers13215537 - 04 Nov 2021
Cited by 5 | Viewed by 2818
Abstract
Bladder cancer (BC) is the most common tumor of the urinary tract and is conventionally classified as either non-muscle invasive or muscle invasive. In addition, histological variants exist, as organized by the WHO-2016 classification. However, innovations in next-generation sequencing have led to molecular [...] Read more.
Bladder cancer (BC) is the most common tumor of the urinary tract and is conventionally classified as either non-muscle invasive or muscle invasive. In addition, histological variants exist, as organized by the WHO-2016 classification. However, innovations in next-generation sequencing have led to molecular classifications of BC. These innovations have also allowed for the tracing of major tumorigenic pathways and, therefore, are positioned as strong supporters of precision medicine. In parallel, immunohistochemistry is still the clinical reference to discriminate histological layers and to stage BC. Key contributions have been made to enlarge the panel of protein immunomarkers. Moreover, the analysis of proteins in liquid biopsy has also provided potential markers. Notwithstanding, their clinical adoption is still low, with very few approved tests. In this context, mass spectrometry-based proteomics has remained a step behind; hence, we aimed to develop them in the community. Herein, the authors introduce the epidemiology and the conventional classifications to review the molecular classification of BC, highlighting the contributions of proteomics. Then, the advances in mass spectrometry techniques focusing on maintaining the integrity of the biological structures are presented, a milestone for the emergence of histoproteomics. Within this field, the review then discusses selected proteins for the comprehension of the pathophysiological mechanisms of BC. Finally, because there is still insufficient knowledge, this review considers proteomics as an important source for the development of BC therapies. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

24 pages, 817 KiB  
Review
Challenges and Opportunities in Clinical Applications of Blood-Based Proteomics in Cancer
by Ruchika Bhawal, Ann L. Oberg, Sheng Zhang and Manish Kohli
Cancers 2020, 12(9), 2428; https://doi.org/10.3390/cancers12092428 - 27 Aug 2020
Cited by 45 | Viewed by 7094
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Graphical abstract

19 pages, 701 KiB  
Review
Proteomic Analysis of Exosomes for Discovery of Protein Biomarkers for Prostate and Bladder Cancer
by Yi-Ting Wang, Tujin Shi, Sudhir Srivastava, Jacob Kagan, Tao Liu and Karin D. Rodland
Cancers 2020, 12(9), 2335; https://doi.org/10.3390/cancers12092335 - 19 Aug 2020
Cited by 44 | Viewed by 9151
Abstract
Extracellular vesicles (EVs) are released by nearly all cell types as part of normal cell physiology, transporting biological cargo, including nucleic acids and proteins, across the cell membrane. In pathological states such as cancer, EV-derived cargo may mirror the altered state of the [...] Read more.
Extracellular vesicles (EVs) are released by nearly all cell types as part of normal cell physiology, transporting biological cargo, including nucleic acids and proteins, across the cell membrane. In pathological states such as cancer, EV-derived cargo may mirror the altered state of the cell of origin. Exosomes are the smaller, 50–150 nanometer-sized EVs released from fusion of multivesicular endosomes with the plasma membrane. Exosomes play important roles in cell-cell communication and participate in multiple cancer processes, including invasion and metastasis. Therefore, proteomic analysis of exosomes is a promising approach to discover potential cancer biomarkers, even though it is still at an early stage. Herein, we critically review the advances in exosome isolation methods and their compatibility with mass spectrometry (MS)-based proteomic analysis, as well as studies of exosomes in pathogenesis and progression of prostate and bladder cancer, two common urologic cancers whose incidence rates continue to rise annually. As urological tumors, both urine and blood samples are feasible for noninvasive or minimally invasive analysis. A better understanding of the biological cargo and functions of exosomes via high-throughput proteomics will help provide new insights into complex alterations in cancer and provide potential therapeutic targets and personalized treatment for patients. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

Other

23 pages, 3142 KiB  
Perspective
Protein Expression in Metastatic Melanoma and the Link to Disease Presentation in a Range of Tumor Phenotypes
by Yonghyo Kim, Jeovanis Gil, Indira Pla, Aniel Sanchez, Lazaro Hiram Betancourt, Boram Lee, Roger Appelqvist, Christian Ingvar, Lotta Lundgren, Håkan Olsson, Bo Baldetorp, Ho Jeong Kwon, Henriett Oskolás, Melinda Rezeli, Viktoria Doma, Sarolta Kárpáti, A. Marcell Szasz, István Balázs Németh, Johan Malm and György Marko-Varga
Cancers 2020, 12(3), 767; https://doi.org/10.3390/cancers12030767 - 24 Mar 2020
Cited by 2 | Viewed by 4647
Abstract
Malignant melanoma is among the most aggressive skin cancers and it has among the highest metastatic potentials. Although surgery to remove the primary tumor is the gold standard treatment, once melanoma progresses and metastasizes to the lymph nodes and distal organs, i.e., metastatic [...] Read more.
Malignant melanoma is among the most aggressive skin cancers and it has among the highest metastatic potentials. Although surgery to remove the primary tumor is the gold standard treatment, once melanoma progresses and metastasizes to the lymph nodes and distal organs, i.e., metastatic melanoma (MM), the usual outcome is decreased survival. To improve survival rates and life span, advanced treatments have focused on the success of targeted therapies in the MAPK pathway that are based on BRAF (BRAF V600E) and MEK. The majority of patients with tumors that have higher expression of BRAF V600E show poorer prognosis than patients with a lower level of the mutated protein. Based on the molecular basis of melanoma, these findings are supported by distinct tumor phenotypes determined from differences in tumor heterogeneity and protein expression profiles. With these aspects in mind, continued challenges are to: (1) deconvolute the complexity and heterogeneity of MM; (2) identify the signaling pathways involved; and (3) determine protein expression to develop targeted therapies. Here, we provide an overview of the results from protein expression in MM and the link to disease presentation in a variety of tumor phenotypes and how these will overcome the challenges of clinical problems and suggest new promising approaches in metastatic melanoma and cancer therapy. Full article
(This article belongs to the Special Issue The Cancer Proteome)
Show Figures

Figure 1

Back to TopTop