Next Article in Journal
A Scoping Review of Precision Medicine in Breast Reconstruction (2011–2025)
Previous Article in Journal
The Association of Assisted Reproductive Technology with Placental and Umbilical Abnormalities
Previous Article in Special Issue
Treatments of Interest in Male Breast Cancer: An Umbrella Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Proteomics in Diagnostic Evaluation and Treatment of Breast Cancer: A Scoping Review

by
Menelaos Zafrakas
1,2,*,
Ioannis Gavalas
2,
Panayiota Papasozomenou
1,
Christos Emmanouilides
2 and
Maria Chatzidimitriou
1
1
School of Health Science, International Hellenic University, 57400 Thessaloniki, Greece
2
European Interbalkan Medical Center, Department of Medical Oncology, 55535 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2025, 15(5), 177; https://doi.org/10.3390/jpm15050177
Submission received: 25 March 2025 / Revised: 20 April 2025 / Accepted: 24 April 2025 / Published: 27 April 2025

Abstract

:
Objectives: The aim of this scoping review was to delineate the current role and possible applications of proteomics in personalized breast cancer diagnostic evaluation and treatment. Methods: A comprehensive search in PubMed/MEDLINE and Scopus/EMBASE was conducted, according to the PRISMA–ScR guidelines. Inclusion criteria: proteomic studies of specimens from breast cancer patients, clinically relevant studies and clinical studies. Exclusion criteria: in silico, in vitro and studies in animal models, review articles, case reports, case series, comments, editorials, and articles in language other than English. The study protocol was registered in the Open Science Framework. Results: In total, 1093 records were identified, 170 papers were retrieved and 140 studies were selected for data extraction. Data analysis and synthesis of evidence showed that most proteomic analyses were conducted in breast tumor specimens (n = 77), followed by blood samples (n = 48), and less frequently in other biologic material taken from breast cancer patients (n = 19). The most commonly used methods were liquid chromatography–tandem mass spectrometry (LC–MS/MS), followed by Matrix-assisted laser desorption/ionization-time of flight (MALDI–TOF), Surface-Enhanced Laser Desorption/Ionization Time-of-Flight (SELDI–TOF) and Reverse Phase Protein Arrays (RPPA). Conclusions: The present review provides a thorough map of the published literature reporting clinically relevant results yielded from proteomic studies in various biological samples from different subgroups of breast cancer patients. This analysis shows that, although proteomic methods are not currently used in everyday practice to guide clinical decision-making, nevertheless numerous proteins identified by proteomics could be used as biomarkers for personalized diagnostic evaluation and treatment of breast cancer patients.

1. Introduction

Breast cancer is the most common malignancy in women worldwide, ranking first in 157 countries, and it is the leading cause of death from cancer in 112 countries [1]. Incidence rates are higher in transitioned countries as compared with transitioning countries (54.1 vs. 30.8 per 100,000), whereas mortality rates are lower in the former (11.3 vs. 15.3 per 100,000, respectively) [1]. In the USA, breast cancer alone accounts for 32% of new cancer diagnoses [2]; approximately one in eight women, or 12.5%, will be diagnosed with invasive breast cancer, and one in forty-three, or 2.3%, will die from the disease [3]. In many high-income countries, mortality rates have decreased since the early 1990s, following new developments in early detection by screening, and progress in treatment [1,4].
Since the dawn of the 21st century, intensive research in gene expression analysis has led to the identification of different multi-gene signatures and the characterization of four subtypes of breast cancer, luminal A, luminal B, ERBB-B2-overexpressing and basal-like, and their respective clinico-pathological surrogate definitions, based on immunohistochemistry, i.e., luminal A-like, luminal B-like, HER2 positive (non-luminal) and triple negative (ductal) [5,6,7]. In recent years, several multi-gene expression assays, have been increasingly used in clinical practice, playing a pivotal role in personalized decision-making on whether to administer, or not, chemotherapy to patients with early luminal breast cancer; these assays include the cDNA-microarray-based 70-gene assay (MammaPrint), and the RT–PCR-based methods 21-gene assay (Oncotype DX), 50-gene assay (PAM50), and 12-gene assay (Endopredict) [8,9]. Modern high-throughput proteomic analysis methods have emerged at the end of the last century [10,11], and since then they have been widely used in breast cancer research [12,13]. Still, in contrast to genomic analyses, it is not clear to clinicians whether proteomic-based methods have any clinical applications in breast cancer diagnosis and therapy. Hence, the aim of the present scoping review was to investigate and delineate the current role and possible emerging clinical applications of proteomic analyses in personalized diagnostic evaluation and treatment of breast cancer patients.

2. Materials and Methods

The present study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses extension for Scoping Reviews (PRISMA–ScR) [14]; the PRISMA–ScR Checklist is presented in Supplementary Table S1. The study protocol was registered in the Open Science Framework (registration DOI: https://doi.org/10.17605/OSF.IO/BE3WF, accessed on 23 March 2025).

2.1. Eligibility Criteria

The PCC (i.e., Population, Concept, Context) criteria [14,15] were used as follows: (1) Population: Breast cancer patients. (2) Concept: Proteomic(s) analyses applied in clinical and clinically relevant research in breast cancer. (3) Context: Breast cancer and normal breast tissue, lymph nodes, blood (serum and/or plasma) and any other biological material taken from breast cancer patients; in vitro studies in breast cancer cell lines would be excluded.
The following inclusion criteria were used: (1) Studies with proteomic analyses of specimens taken from breast cancer patients, i.e., primary and metastatic lesions, normal breast tissue, lymph nodes, blood (serum and plasma), other biological material; (2) clinically relevant studies for diagnostic and therapeutic target discovery and validation; (3) clinical studies. The following exclusion criteria were used: (1) in silico studies; (2) in vitro studies in breast cancer cell lines; (3) studies in animal models; (4) review articles; (5) case reports, case series and, in general, studies with a number of patient specimens lower than 10; (6) comments and editorials; (7) articles in language other than English.

2.2. Information Sources and Search

The present scoping review was conducted by searching the two primary online medical databases: the PubMed/MEDLINE database (PubMed) and the EMBASE database (Scopus) for articles published until 23 March 2025. The following search terms were used in both database searches: “proteomic” AND “breast cancer”, and “proteomics” AND “breast cancer”, all in title.

2.3. Selection of Sources of Evidence

For study selection, two authors (IG and PP) screened independently each study in order to exclude overlapping and duplicate publications, as well as studies not published in English, and then assessed the titles and/or abstracts of the remaining records according to the inclusion and exclusion criteria; after retrieval of eligible studies, the full-texts were further assessed according to the inclusion and exclusion criteria; any conflicts were resolved by discussion or referral to a third author (MZ), who also re-assessed and confirmed the inclusion of eligible publications.

2.4. Data Charting Process, Data Items, and Synthesis of Results

Data collection was carried out by two independent reviewers (IG and PP), by using Microsoft Excel filled out with data taken from the main text, the figures, and the tables of the included studies; any conflicts were resolved by discussion or referral to a third author (MZ), who also re-assessed and confirmed collection of data. Furthermore, two separate lists of excluded studies were constructed, i.e., in vitro and in silico studies.
The following data items were extracted from the included articles: (1) first author; (2) year of publication; (3) country of first author’s affiliation; (4) clinical setting, i.e., whether the possible clinical application of the study was diagnostic or relevant to treatment; (5) disease stage, i.e., ductal carcinoma in situ (stage 0), primary invasive breast cancer (stages I, II and III), and metastatic breast cancer (stage IV); (6) type of specimen analyzed; (7) number of specimens analyzed; (8) the primary proteomic method used; (9) other methods used; (10) main conclusions. Data items were not extracted from the excluded articles. Extracted data from included studies were further analyzed according to clinical relevance, i.e., according to (a) the diagnostic or therapeutic setting, (b) the type of tissue specimen analyzed, and (c) the disease stage.

3. Results

3.1. Search Results

The study selection process and results are presented diagrammatically as a flow chart in Figure 1, according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses extension for Scoping Reviews (PRISMA–ScR) [14]. In brief, 1093 studies (584 in PubMed and 509 in Scopus) were identified by using the search terms “proteomic” AND “breast cancer”, and “proteomics” AND “breast cancer”, all in title. After removal of duplicates, overlapping and studies not published in English (n = 515), the titles and/or abstracts of 578 records were screened and 401 articles were excluded, leaving 177 articles to be sought for retrieval. Next, 170 articles were retrieved and, after assessment of their full-text, 30 reports were excluded according to the eligibility criteria. Finally, 140 articles were included [16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155], and data extraction followed. Two hundred and ninety eight excluded studies, i.e., 243 in vitro studies, 23 in silico studies and 32 reviews, are presented in Supplementary File S1–S3, respectively.

3.2. Year of Publication and Geographical Area of Included Studies

Regarding the year of publication, the number of studies appeared to increase over time. In detail, six studies were published between 2001 and 2004 [82,83,104,112,113,118], 22 studies between 2005 and 2009 [26,46,48,61,68,72,80,84,96,97,98,99,100,101,105,109,116,119,126,128,146,147], 30 studies between 2010 and 2014 [25,31,32,36,37,38,39,40,42,43,49,57,58,59,62,63,69,78,90,92,94,102,111,115,124,140,141,148,150,151], 38 studies between 2015 and 2019 [16,18,28,29,33,34,35,41,45,51,52,53,54,55,66,70,71,76,81,88,93,95,106,107,108,110,114,127,131,132,135,138,139,142,144,145,149,153] and 44 studies were published between 2020 and March 23, 2025 [17,19,20,21,22,23,24,27,30,44,47,50,56,60,64,65,67,73,74,75,77,79,85,86,87,89,91,103,117,120,121,122,123,125,129,130,133,134,136,137,143,152,154,155] (see Supplementary Figure S1).
Regarding the country of first author’s affiliation (a) 47 studies came from Europe, i.e., nine from Italy [26,37,38,45,57,58,96,109,110], eight from Germany [29,31,85,88,98,99,114,121], six from Spain [42,53,54,55,56,114], five from France [32,43,61,66,132], five from the Netherlands [34,44,59,92,94], four from Sweden [64,67,77,100], three from Denmark [36,65,129], two from the UK [95,144], two from the Czech Republic [33,108], two from Russia [125,130], and one from Norway [30]; (b) 46 studies came from Asia, i.e., 18 from China [21,47,72,85,87,113,121,134,135,136,137,138,139,140,143,145,152,153], six from South Korea [76,79,81,82,122,141], six from India [51,52,60,103,127,154], three from Israel [107,120,142], three from Singapore [101,149,150], two from Japan [90,128], two from Malaysia [20,74], two from Thailand [40,75], one from Iran [91], one from Saudi Arabia [16], one from Turkey [18], and one from Taiwan [155]; (c) 45 studies came from North and South America, i.e., 35 from the USA [17,19,25,27,46,48,50,62,63,68,69,70,71,73,78,80,83,84,89,93,97,104,105,111,116,118,119,123,124,126,133,146,147,148,151], seven from Brazil [23,24,35,41,102,106,117], two from Canada [22,39], and one from Mexico [49]; and (d) two studies came from Australia [28,112] (Supplementary Figure S2). It is noteworthy that, in 17 studies, the authors came from more than one country [16,26,33,42,50,55,59,63,65,84,88,94,116,123,126,131,133].

3.3. Clinical Application, Disease Stage and Specimen Type of Included Studies

Regarding the possible clinical application of each study 105 studies were relevant to diagnosis [16,18,20,21,22,23,24,25,26,27,28,29,30,31,33,34,35,37,38,39,40,41,45,46,49,50,51,52,53,54,57,58,59,60,61,64,67,70,71,72,73,74,75,79,80,81,82,83,84,85,88,89,93,94,95,96,97,98,99,101,102,103,104,105,107,108,109,110,111,112,113,114,115,116,117,118,119,122,123,125,127,128,129,130,131,132,133,134,135,136,139,140,142,143,144,145,146,147,148,149,150,151,152,153,154], 25 were relevant to treatment [17,19,32,36,43,44,47,56,62,65,66,76,77,86,90,91,92,106,120,121,124,137,138,141,155] and 10 were relevant to both diagnosis and treatment [42,48,55,63,68,69,78,87,100,126] (Supplementary Figure S3).
Regarding the disease stage, specimens taken from patients with primary invasive breast cancer (stages I, II and III) were examined in 135 studies (in 114 studies primary invasive alone [16,18,19,20,21,22,23,24,26,27,29,30,31,32,33,34,35,36,37,38,39,40,41,42,44,45,46,47,49,50,51,52,53,54,55,56,57,58,59,61,62,64,65,66,67,68,69,70,74,75,76,77,78,81,82,83,85,86,87,88,89,91,93,94,95,96,97,98,99,100,101,103,104,105,106,107,108,109,110,112,114,115,116,117,118,120,121,123,124,126,127,128,129,130,131,133,134,135,136,137,138,139,140,142,144,145,146,148,149,150,151,152,153,155], in 10 studies together with metastatic breast cancer [48,72,80,84,102,113,122,141,143,154], in eight studies together with DCIS [28,71,73,79,90,111,132,147], in two studies together with DCIS and metastatic [63,119], and in one study together with DCIS and recurrent breast cancer [25]), while specimens taken from patients with metastatic breast cancer were examined in seventeen studies (in five metastatic alone [17,43,60,92,125], in ten together with primary invasive breast cancer [48,72,80,84,102,113,122,141,143,154] and in two together with primary invasive breast cancer and DCIS [63,119]) (Supplementary Figure S4).
Regarding the type of specimen analyzed, breast cancer tissue was analyzed in 77 studies (Table 1), blood samples were analyzed in 48 studies (in 33 studies in serum and in 15 studies in plasma) (Table 2), and other biologic materials, i.e., nipple aspiration fluid (NAF), urine, saliva, tear fluid, pleural effusions, tumor interstitial fluid, and lymph nodes, were analyzed in 19 studies (Table 3).

3.3.1. Proteomic Studies in Breast Cancer Tissue

For each proteomic study in breast cancer tissue, the number of specimens analyzed, the primary proteomic method used, other methods used if any and the main findings of each study are presented in Table 1. In brief, 52 studies were relevant to breast cancer diagnostic evaluation [16,18,20,22,23,24,25,29,30,33,34,37,38,39,40,45,46,49,53,54,67,73,74,79,82,93,94,96,97,98,99,101,102,107,108,109,110,112,113,114,116,122,128,131,132,139,140,142,145,149,150,153], 18 were relevant to breast cancer therapy [17,32,36,44,47,56,62,65,66,76,77,90,120,121,124,138,140,155], and 8 studies were relevant to both breast cancer diagnostic evaluation and treatment [42,55,63,68,69,87,100,126].
In sixty-seven studies proteomic analysis was conducted in primary invasive breast tumors only [16,18,20,22,23,24,29,30,32,33,34,36,37,38,39,40,42,44,45,46,47,49,53,54,55,56,62,65,66,67,68,69,74,76,77,82,87,93,94,96,97,98,99,100,101,107,108,109,110,112,114,116,120,121,124,126,128,131,138,139,140,142,145,149,150,153,155], in four studies in both primary invasive breast tumors and ductal carcinoma in situ (DCIS) [73,79,90,132], in one study in primary invasive, locally recurrent and DCIS [25], in one study in primary invasive, metastatic and DCIS [63], in four studies in both primary and metastatic lesions [102,113,122,141], and in one study only in breast cancer metastases [17]. The number of breast cancer specimens analyzed ranged between 10 and 990 (median = 60). The most common proteomic method used was liquid chromatography–tandem mass spectrometry (LC–MS/MS), followed by Matrix-assisted laser desorption/ionization-time of flight (MALDI–TOF) and Reverse Phase Protein Arrays (RPPA). The most common additional methods used were immunohistochemistry and Western blot.
Regarding the main findings, 29 studies identified possible biomarkers for breast cancer diagnosis, prognosis, staging and disease progression [17,18,20,22,24,25,29,33,37,38,39,45,46,54,73,82,97,102,107,108,109,113,116,122,128,132,145,149,153], 17 studies identified possible biomarkers for specific breast cancer subtypes [23,34,42,49,54,55,68,76,87,94,96,98,114,131,139,142,150], 9 studies identified possible biomarkers for breast cancer diagnosis and pathogenesis [16,30,40,67,74,79,101,110,112], 16 studies identified possible biomarkers for response to therapy [32,44,47,63,65,66,69,77,93,99,100,120,124,138,140,141], and 7 studies identified possible therapeutic targets [36,56,62,90,121,126,155]. More details regarding the main findings of each study are presented in Supplementary Table S2.

3.3.2. Proteomic Studies in Plasma and Serum from Breast Cancer Patients

An overview of proteomic studies in plasma and serum from breast cancer patients is presented in Table 2, showing the type and number of specimens analyzed, the primary proteomic method used, other methods used if any and the main findings of each study. In brief, serum was analyzed in 32 studies [26,27,43,48,50,51,57,58,59,61,70,71,72,79,83,87,88,90,94,111,113,119,123,125,134,135,136,137,144,148,149,154], and plasma was analyzed in 15 studies [19,21,41,64,77,81,85,106,117,127,130,133,143,150,152]. Of these, 39 studies were relevant to breast cancer diagnostic evaluation [21,26,27,41,50,51,57,58,59,61,64,70,71,72,79,81,83,87,88,94,111,113,117,119,123,125,127,130,133,134,135,136,143,144,148,149,150,152,154], 6 studies were relevant to breast cancer treatment [19,43,85,90,106,137], and 2 studies were relevant to both breast cancer diagnostic evaluation and treatment [48,77].
In 35, studies proteomic analysis was conducted in patients with primary invasive breast tumors (stage I-III) only [19,21,26,27,41,50,51,57,58,59,61,64,77,81,83,85,87,88,90,94,106,113,117,123,127,130,133,134,135,136,137,144,149,150,152], in 4 studies in patients with both primary invasive breast tumors and ductal carcinoma in situ (DCIS) [70,71,111,148], in 1 study in patients with primary invasive, metastatic and DCIS [119], in 5 studies in patients with primary and patients with metastatic disease [48,72,79,143,154], and in 2 studies only in patients with breast cancer metastases [43,125]. The number of breast cancer specimens analyzed ranged between 10 and 796 (median = 76). The most common proteomic method used was liquid chromatography–tandem mass spectrometry (LC–MS/MS), followed by Surface-Enhanced Laser Desorption/Ionization Time-of-Flight (SELDI–TOF) and Matrix-assisted laser desorption/ionization-time of flight (MALDI–TOF). The most common additional method used was Western blot.
Regarding the main findings, 14 studies identified possible biomarkers for early diagnosis of breast cancer [26,50,70,71,72,81,83,88,123,133,134,148,149,150], 16 studies identified possible biomarkers for diagnosis, prognosis, staging and disease progression [21,27,58,59,61,64,79,111,113,119,125,130,136,143,144,154], 6 studies identified possible biomarkers for specific breast cancer subtypes [41,51,57,117,127,152], 8 studies identified possible biomarkers for response to therapy [19,43,48,85,90,106,135,137], 1 study identified possible diagnostic and therapeutic targets [77], 1 study identified possible biomarkers for cancer related fatigue syndrome [94], and 1 study identified possible biomarkers for neuropathic pain [87]. More details regarding the main findings of each study are presented in Supplementary Table S3.

3.3.3. Proteomic Studies in Other Biologic Material from Breast Cancer Patients

An overview of proteomic studies in biologic material from breast cancer patients other than breast cancer tissue, plasma and serum, i.e., nipple aspiration fluid, urine, saliva, tear fluid, pleural effusions, tumor interstitial fluid and lymph nodes, is given in Table 3, which presents the type and number of specimens analyzed, the primary proteomic method used, other methods used if any, and the main findings of each study. In brief, proteomic studies were conducted in nipple aspiration fluid (NAF) in five studies [35,82,104,105,118], in urine in three studies [28,52,74], in saliva in three studies [60,123,151], in tear fluid in two studies [25,50], in pleural effusions in one study [91], in tumor interstitial fluid in one study [129], and in lymph nodes in four studies [103,107,145,153]; eighteen studies were relevant to breast cancer diagnostic evaluation [25,28,35,50,52,60,74,82,103,104,105,107,118,123,129,145,151,153], and only one study was relevant to breast cancer therapy [91]. The number of breast cancer specimens analyzed ranged between 10 and 54 (median = 25). The most common proteomic method used was liquid chromatography–tandem mass spectrometry (LC–MS/MS), followed by Surface-Enhanced Laser Desorption/Ionization Time-of-Flight (SELDI–TOF) and Isobaric Tag for Relative and Absolute Quantification (iTRAQ). The most common additional method used was Western blot. Regarding the main findings, 12 studies identified possible biomarkers for differentiating breast cancer patients from healthy individuals [28,31,35,52,60,74,84,104,105,118,123,151], four studies identified possible biomarkers for nodal status [82,103,145,153], one study identified possible biomarkers for specific breast cancer subtypes [129], and one study identified possible biomarkers for response to therapy [91]. More details regarding the main findings of each study are presented in Supplementary Table S4.

3.4. Possible Clinical Use of Biomarkers Identified by the Included Studies

Studies with at least 30 specimens from breast cancer patients analyzed were further categorized according to their possible clinical use, in studies that identified biomarkers for breast cancer prognosis (Table 4), early diagnosis (Table 5), characterization of breast cancer subtypes (Table 6), and biomarkers for response to treatment (Table 7).
In Table 4, Table 5, Table 6 and Table 7 studies are presented in descending order of the number of specimens analyzed in each study, as this order reflects the impact of studies. Table 4 presents 11 studies that identified biomarkers for breast cancer prognosis; the number of specimens analyzed ranged between 60 and 990; these studies were carried out in primary tumor specimens (10 studies) or blood samples (1 study) from patients with non-metastatic invasive breast cancer (stages I-III). Table 5 presents 13 studies that identified biomarkers for early diagnosis of breast cancer; the number of specimens from breast cancer patients analyzed ranged between 30 and 796; it is noteworthy that all 13 studies were conducted in blood samples (serum or plasma) from patients with stage 0-IV disease. Table 6 shows 17 studies that identified biomarkers for characterization of breast cancer subtypes; the number of specimens analyzed ranged between 32 and 429; these studies were carried out either in primary tumor specimens (12 studies) or blood samples (5 studies) from patients with invasive non-metastatic breast cancer (stages I-III). Finally, Table 7 presents 21 studies that identified biomarkers for response to treatment; the number of specimens analyzed was between 36 and 880; these studies were carried out in primary tumor specimens (16 studies), blood samples (4 studies), and pleural effusions (1 study) from patients with stage 0-IV disease.

3.5. Overview of the Most Important Findings from the Largest Studies and the Most Commonly Used Proteomic Platforms Across the Studies

Regarding prognosis and response to treatment, in the largest study to date, Cawthorn et al. [39] analyzed a tissue microarray consisting of 990 early breast cancer cases (590 stage I and 400 stage II) and found that high expression levels of Decorin and Endoplasmin (HSP90B1) were associated with increased metastasis-poorer survival and may guide the use of hormonal therapy. Gonzalez-Angulo et al. [48] analyzed 880 primary breast tumor specimens and 256 FNAs obtained from primary breast cancer and developed a 10-protein biomarker panel that classifies breast cancer into prognostic subgroups, predicting relapse-free survival (RFS) and pathologic Complete Response (pCR) to neoadjuvant therapy. Bernhardt et al. [29] analyzed breast cancer specimens from 801 consecutive patients and identified SHMT2 and ASCT2 protein expression as novel potential prognostic biomarkers for breast cancer, as their high protein expression was associated with poor outcome.
Regarding early diagnosis, in the largest study to date, Grassmann et al. [64] anayzed plasma samples from 1577 women (796 incident breast cancer cases and 781 controls) participating in the prospective KARMA mammographic screening cohort and concluded that the levels of the studied plasma proteins were unlikely to offer additional benefits for risk prediction of short-term overall breast cancer risk, but could provide interesting insights into the biological basis of breast cancer in the future. Kim et al. [81] quantified three peptides, apolipoprotein C-1, carbonic anhydrase 1, and neural cell adhesion molecule L1-like protein in human plasma from 575 breast cancer patients and 454 healthy controls and concluded that these three petides can be a useful tool for breast cancer screening.
The most commonly used proteomic methods across the studies were mass spectrometry-based methods in 61 studies [4,5,6,7,8,9,10,13,19,20,21,25,26,32,34,35,37,38,39,40,51,52,53,59,60,61,62,63,65,72,82,88,91,92,93,95,97,103,106,107,108,111,114,115,116,117,118,119,121,122,125,126,127,128,129,130,131,133,134,138,139], most commonly liquid chromatography–tandem mass spectrometry (LC–MS/MS), followed by Matrix-assisted laser desorption/ionization-time of flight (MALDI–TOF), in 20 studies [1,16,22,23,30,64,67,70,77,85,86,87,96,101,102,105,123,135,136,137], Surface-Enhanced Laser Desorption/Ionization Time-of-Flight (SELDI–TOF) in 15 studies [11,17,28,42,43,44,46,54,57,69,71,81,83,90,104], Reverse Phase Protein Arrays (RPPA) in 11 studies [2,14,27,47,48,50,79,80,110,112,120], and Isobaric tag for absolute and relative quantification (iTRAQ) in 9 studies [18,24,36,73,89,109,113,132,140].

4. Discussion

Although proteomic analyses have been widely used in breast cancer research over the last two decades, it remains unclear if these efforts have been translated into direct clinical applications. The present scoping review is the first structured synthesis of evidence, delineating the current state and the possible future clinical applications of proteomic analyses in breast cancer diagnosis and treatment. To this end, we conducted a thorough search of the literature and found that, so far, the majority of pertinent proteomic analyses have been carried out in breast cancer cell lines rather than clinical specimens from breast cancer patients (see Supplementary File S1). After the search of the literature, study selection and data extraction followed and our data analysis and synthesis of results from 140 clinically relevant studies [16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155] showed that the number of publications regarding proteomic analyses in clinical specimens from breast cancer patients has been steadily increasing over time (see Supplementary Figure S1), and that three geographical areas, i.e., Europe, Asia, and the Americas, have an almost equal share in these publications (see Supplementary Figure S2). We also found that most clinically relevant studies have possible applications in diagnosis rather than therapy (see Supplementary Figure S3), and that most studies were carried out in specimens taken from patients with primary invasive breast cancer (stages I-III), rather than patients with metastatic disease (stage IV) or DCIS (stage 0) (see Supplementary Figure S4). Furthermore, our data analysis and synthesis of evidence showed that most proteomic analyses were conducted in breast tumor specimens, followed by plasma and serum, and only rarely in other biologic material taken from breast cancer patients. Overall, the most commonly used method in clinically relevant studies were liquid chromatography–tandem mass spectrometry (LC–MS/MS), followed by Matrix-assisted laser desorption/ionization-time of flight (MALDI–TOF), Reverse Phase Protein Arrays (RPPA) and Surface-Enhanced Laser Desorption/Ionization Time-of-Flight (SELDI–TOF) (see Table 1, Table 2 and Table 3).
In the present scoping review, neither a critical appraisal nor a risk of bias assessment across studies was conducted, since the former is not mandatory and the latter is not applicable in scoping reviews [14,15]. However, certain clinically relevant points should be highlighted. First, the wide-spread use of neoadjuvant chemotherapy in recent years has been a paradigm shift in breast cancer treatment, with new clinically critical questions arising, such as response to treatment, and residual disease after neoadjuvant therapy. However, studies with proteomic analyses of specimens from patients treated with neoadjuvant chemotherapy are still scarce [32,47,63,120,124,141]. Also hard to find are studies with proteomic analyses of specimens from patients diagnosed with triple negative tumors, the most aggressive form of breast cancer, with currently finite treatment options [36,55,56,60,65,85,86,114,121,124]. Likewise, proteomic analyses in specimens obtained from patients with metastatic disease are also scarce [17,43,48,63,72,79,102,113,119,122,125,141,143]. Intriguingly, though blood samples are theoretically more readily available than tumor tissue, our analysis showed that more studies have been published regarding proteomic analysis of breast tumor specimens than studies in blood taken from breast cancer patients (see Table 1 and Table 2). Even more rare are studies in lymph nodes, and surprisingly studies with proteomic analysis of urine from breast cancer patients (see Table 3). In recent years, the extent of axillary surgery has been steadily receding, due to the ever increasing number of screen-detected breast cancers at earlier stages, the wide-spread use of sentinel lymph node biopsy and, more recently, due to the use of targeted axillary dissection (TAD) following neoadjuvant chemotherapy [156,157]. With such novel developments, new clinical questions arise in respect to nodal status of breast cancer patients, with implications regarding patient diagnosis, staging, prognosis and treatment, directing proteomic and basic research in general to be focused on these issues.
Another noteworthy clinically relevant point is that, although different studies claim the identification of new therapeutic targets by using proteomic analysis, in fact these targets may not be all that new; for example, the MAGE genes were identified as potential therapeutic targets in breast cancer by RT–PCR gene expression analysis [158] many years before proteomic studies came to the same conclusion [36,121]. A final critical issue worthy to be addressed is that the number of specimens analyzed in proteomic studies is relatively low as compared with that of genomic studies. In particular, as shown in Table 1 and Table 4, the highest numbers of breast tumor specimens analyzed by proteomics in the included studies were 990 [39], 880 [62] and 801 [29] and the findings of these studies did not have any direct impact on clinical decision-making. In contrast, much higher numbers of breast tumor specimens have been analyzed in studies using multigene assays, such as the 70-gene assay (Mammaprint) (n = 6600) [159], the 21-gene assay (OncotypeDX) (n = 10,273) [160], the 50-gene assay (PAM50) (n = 2558) [161], and the 12-gene assay (Endopredict) (n = 2185) [162], and these genomic assays are used in everyday practice to guide clinical decision-making on whether or not to administer chemotherapy in early luminal A and B (estrogen-receptor-positive) breast cancer. This is not surprising, since historically the development of high-throughput transcriptomics preceded that of proteomics, and gene expression signatures with prognostic significance were already identified at the eve of the 21st century [5,6], well before the widespread use of proteomics. Hence, it seems that so far the relatively low number of specimens analyzed by proteomics has been a barrier to translation of findings into clinical practice. Besides small sample sizes, other such possible barriers may include lack of assay standardization, lack of reproducibility of results, costs, validation challenges, and diversity of proteomic methods applied. In the future, the investigational use of proteomic analyses in the translational arm of large, adequately powered, multicenter randomized clinical trials, with centralized laboratory facilities using standardized, reproducible and validated assays, might lead to the integration of proteomics in clinical practice, especially since many clinically relevant questions remain open. In particular, the aforementioned multigene assays (Mammaprint, OncotypeDX, PAM50 and Endopredict) provide prognostic and in part predictive information in a subset of breast cancer patients with early disease (hormone receptor positive, HER2-negative tumors, up to 5 cm in maximal diameter, with up to three positive lymph nodes), while no such robust evidence-based information is available for more advanced breast cancer stages, neither with transcriptomic nor with proteomic approaches. Therefore, proteomic analyses may be used for the identification of biomarkers providing prognostic information, especially in advanced breast cancer stages, as well as for the identification of biomarkers for other applications, such as early detection, response to treatment and resistance to treatment.
The main strength of the present study is its originality, as this is the first scoping review on this topic. Most of the other published articles reviewing the role of proteomics in breast cancer research are narrative reviews and there is a paucity of relevant systematic reviews (see Supplementary File S3). Hence, it is possible that the present study may lead to primary studies and systematic reviews regarding applications of proteomics in identifying biomarkers for early diagnosis of breast cancer, characterization of breast cancer subtypes, potential therapeutic targets, and specific biomarkers for response to treatment. Another strong point of this review is that we have focused on proteomic analyses of clinical specimens taken from patients with breast cancer, by excluding in vitro and in silico studies. On the other hand, a weakness of this study is that both the variety of proteomic methods used and the diversity of clinical specimens analyzed hamper comparisons between studies and data synthesis leading to clinically applicable conclusions. Nevertheless, it is the vast heterogeneity of breast cancer at the molecular, histopathological and clinical level that necessitates this multiplicity of methodological approaches.

5. Conclusions

This is the first methodologically structured review investigating the current state of proteomic applications in breast cancer diagnostic evaluation and treatment. Given the variety of available proteomic methods and the vast heterogeneity of breast cancer at the molecular, histopathological and clinical level, conducting a scoping review was deemed the most appropriate methodological approach. Our data analysis and synthesis of evidence showed a relatively low number of studies in blood and a paucity of studies in specimens such as lymph nodes and urine obtained from breast cancer patients. Furthermore, the number of studies with proteomic analyses focusing on certain breast cancer subtypes, especially on triple negative tumors, and on monitoring response to treatment in the neoadjuvant and metastatic setting is relatively low. Currently, in contrast to transcriptomics, proteomic analyses are not used in everyday practice to guide clinical decision-making. On the other hand, multiple proteomic studies yielded clinically relevant results, which hold the promise of translating these findings to routine clinical applications. Hence, it seems that there are multiple perspectives for proteomic studies in breast cancer research in the future. The insights provided by the present study can be used to develop new approaches in research and novel strategies in personalized diagnostic evaluation and treatment of breast cancer, and other tumor entities.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jpm15050177/s1, Supplementary Table S1. PRISMA-ScR Checklist, Supplementary Table S2. Main findings and conclusions of proteomic studies in tumor tissue specimens from breast cancer patients, Supplementary Table S3. Main findings and conclusions of proteomic studies in plasma and serum from breast cancer patients. Supplementary Table S4. Main findings and conclusions of proteomic studies in nipple aspiration fluid (NAF), urine, saliva, tear fluid, pleural effusions, tumor interstitial fluid and lymph nodes from breast cancer patients. Supplementary Figure S1: Number of studies according to the year of publication. Supplementary Figure S2: Number of studies according to geographical area. Supplementary Figure S3: Number of studies according to clinical relevance. Supplementary Figure S4: Number of studies according to disease stage of analyzed clinical specimens. Supplementary File S1: List of In Vitro Studies. Supplementary File S2: List of In Silico Studies. Supplementary File S3: List of Reviews.

Author Contributions

Conceptualization: M.Z.; methodology: M.Z.; formal analysis and investigation: M.Z., I.G. and P.P.; writing, original draft preparation: M.Z., I.G. and P.P.; writing—review and editing: M.Z., C.E. and M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original data presented in the study are openly available inMedline/PubMed (https://pubmed.ncbi.nlm.nih.gov/, accessed on 23 March 2025), and EMBASE/Scopus (https://www.elsevier.com/products/scopus, accessed on 23 March 2025).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef] [PubMed]
  3. Giaquinto, A.N.; Sung, H.; Newman, L.A.; Freedman, R.A.; Smith, R.A.; Star, J.; Jemal, A.; Siegel, R.L. Breast cancer statistics 2024. CA Cancer J. Clin. 2024, 74, 477–495. [Google Scholar] [CrossRef] [PubMed]
  4. Bray, F.; McCarron, P.; Parkin, D.M. The changing global patterns of female breast cancer incidence and mortality. Breast Cancer Res. 2004, 6, 229–239. [Google Scholar] [CrossRef]
  5. van ‘t Veer, L.J.; Dai, H.; van de Vijver, M.J.; He, Y.D.; Hart, A.A.M.; Mao, M.; Peterse, H.L.; van der Kooy, K.; Marton, M.J.; Witteveen, A.T.; et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002, 415, 530–536. [Google Scholar] [CrossRef]
  6. van de Vijver, M.J.; He, Y.D.; van’t Veer, L.J.; Dai, H.; Hart, A.A.M.; Voskuil, D.W.; Schreiber, G.J.; Peterse, J.L.; Roberts, C.; Marton, M.J.; et al. A gene expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 2002, 347, 1999–2009. [Google Scholar] [CrossRef]
  7. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.-J.; Panel members. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223. [Google Scholar] [CrossRef]
  8. Zeng, C.; Zhang, J. A narrative review of five multigenetic assays in breast cancer. Transl. Cancer Res. 2022, 11, 897–907. [Google Scholar] [CrossRef]
  9. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Breast Cancer Screening and Diagnosis Version 6.2024—November 11, 2024. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1419 (accessed on 12 January 2025).
  10. Geisow, M.J. Proteomics: One small step for a digital computer, one giant leap for humankind. Nat. Biotechnol. 1998, 16, 206. [Google Scholar] [CrossRef]
  11. Anderson, N.L.; Anderson, N.G. Proteome and proteomics: New technologies, new concepts, and new words. Electrophoresis 1998, 19, 1853–1861. [Google Scholar] [CrossRef]
  12. Brožová, K.; Hantusch, B.; Kenner, L.; Kratochwill, K. Spatial Proteomics for the Molecular Characterization of Breast Cancer. Rev. Proteomes 2023, 11, 17. [Google Scholar] [CrossRef]
  13. Neagu, A.N.; Jayathirtha, M.; Whitham, D.; Mutsengi, P.; Sullivan, I.; Petre, B.A.; Darie, C.C. Proteomics-Based Identification of Dysregulated Proteins in Breast Cancer. Proteomes 2022, 10, 35. [Google Scholar] [CrossRef] [PubMed]
  14. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.J.; Horsley, T.; Weeks, L.; et al. PRISMA extension for scoping reviews (PRISMA-ScR): Checklist and explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef] [PubMed]
  15. Peters, M.D.; Godfrey, C.M.; Khalil, H.; McInerney, P.; Parker, D.; Soares, C.B. Guidance for conducting systematic scoping reviews. Int. J. Evid. Based Healthc. 2015, 13, 141–146. [Google Scholar] [CrossRef]
  16. Abdullah Al-Dhabi, N.; Srigopalram, S.; Ilavenil, S.; Kim, Y.O.; Agastian, P.; Baaru, R.; Balamurugan, K.; Choi, K.C.; Valan Arasu, M. Proteomic Analysis of Stage-II Breast Cancer from Formalin-Fixed Paraffin-Embedded Tissues. Biomed Res. Int. 2016, 2016, 3071013. [Google Scholar] [CrossRef] [PubMed]
  17. Akcakanat, A.; Zheng, X.; Cruz Pico, C.X.; Kim, T.B.; Chen, K.; Korkut, A.; Sahin, A.; Holla, V.; Tarco, E.; Singh, G.; et al. Genomic, Transcriptomic, and Proteomic Profiling of Metastatic Breast Cancer. Clin. Cancer Res. 2021, 27, 3243–3252. [Google Scholar] [CrossRef]
  18. Akpinar, G.; Simsek, T.; Guler, A.; Kasap, M.; Canturk, N.Z. Elucidation of a Conserved Proteomic Pattern of Breast Cancer Tissue and Metastatic Axillary Lymph Node. Chirurgia 2017, 112, 443–448. [Google Scholar] [CrossRef]
  19. Alvarez, F.A.; Kaddour, H.; Lyu, Y.; Preece, C.; Cohen, J.; Baer, L.; Stopeck, A.T.; Thompson, P.; Okeoma, C.M. Blood plasma derived extracellular vesicles (BEVs): Particle purification liquid chromatography (PPLC) and proteomic analysis reveals BEVs as a potential minimally invasive tool for predicting response to breast cancer treatment. Breast Cancer Res. Treat. 2022, 196, 423–437. [Google Scholar] [CrossRef]
  20. Al-Wajeeh, A.S.; Salhimi, S.M.; Al-Mansoub, M.A.; Khalid, I.A.; Harvey, T.M.; Latiff, A.; Ismail, M.N. Comparative proteomic analysis of different stages of breast cancer tissues using ultra high performance liquid chromatography tandem mass spectrometer. PLoS ONE 2020, 15, e0227404. [Google Scholar] [CrossRef]
  21. An, R.; Yu, H.; Wang, Y.; Lu, J.; Gao, Y.; Xie, X.; Zhang, J. Integrative analysis of plasma metabolomics and proteomics reveals the metabolic landscape of breast cancer. Cancer Metab. 2022, 10, 13. [Google Scholar] [CrossRef]
  22. Asleh, K.; Negri, G.L.; Spencer Miko, S.E.; Colborne, S.; Hughes, C.S.; Wang, X.Q.; Gao, D.; Gilks, C.B.; Chia, S.K.L.; Nielsen, T.O.; et al. Proteomic analysis of archival breast cancer clinical specimens identifies biological subtypes with distinct survival outcomes. Nat. Commun. 2022, 13, 896. [Google Scholar] [CrossRef] [PubMed]
  23. Azevedo, A.L.K.; Gomig, T.H.B.; Batista, M.; Marchini, F.K.; Spautz, C.C.; Rabinovich, I.; Sebastião, A.P.M.; Oliveira, J.C.; Gradia, D.F.; Cavalli, I.J.; et al. High-throughput proteomics of breast cancer subtypes: Biological characterization and multiple candidate biomarker panels to patients’ stratification. J. Proteom. 2023, 285, 104955. [Google Scholar] [CrossRef] [PubMed]
  24. Azevedo, A.L.K.; Gomig, T.H.B.; Giner, I.S.; Batista, M.; Marchini, F.K.; Lima, R.S.; Urban, C.A.; Sebastião, A.P.M.; Cavalli, I.J.; Ribeiro, E.M.S.F. Comprehensive analysis of the large and small ribosomal proteins in breast cancer: Insights on proteomic and transcriptomic expression patterns, regulation, mutational landscape, and prognostic significance. Comput. Biol. Chem. 2022, 100, 107746. [Google Scholar] [CrossRef] [PubMed]
  25. Bateman, N.W.; Sun, M.; Bhargava, R.; Hood, B.L.; Darfler, M.M.; Kovatich, A.J.; Hooke, J.A.; Krizman, D.B.; Conrads, T.P. Differential proteomic analysis of late-stage and recurrent breast cancer from formalin-fixed paraffin-embedded tissues. J. Proteome Res. 2011, 10, 1323–1332. [Google Scholar] [CrossRef]
  26. Belluco, C.; Petricoin, E.F.; Mammano, E.; Facchiano, F.; Ross-Rucker, S.; Nitti, D.; Di Maggio, C.; Liu, C.; Lise, M.; Liotta, L.A.; et al. Serum proteomic analysis identifies a highly sensitive and specific discriminatory pattern in stage 1 breast cancer. Ann. Surg. Oncol. 2007, 14, 2470–2476. [Google Scholar] [CrossRef]
  27. Bera, A.; Russ, E.; Manoharan, M.S.; Eidelman, O.; Eklund, M.; Hueman, M.; Pollard, H.B.; Hu, H.; Shriver, C.D.; Srivastava, M. Proteomic Analysis of Inflammatory Biomarkers Associated with Breast Cancer Recurrence. Mil. Med. 2020, 185, 669–675. [Google Scholar] [CrossRef]
  28. Beretov, J.; Wasinger, V.C.; Millar, E.K.; Schwartz, P.; Graham, P.H.; Li, Y. Proteomic Analysis of Urine to Identify Breast Cancer Biomarker Candidates Using a Label-Free LC-MS/MS Approach. PLoS ONE 2015, 10, e0141876. [Google Scholar] [CrossRef]
  29. Bernhardt, S.; Bayerlová, M.; Vetter, M.; Wachter, A.; Mitra, D.; Hanf, V.; Lantzsch, T.; Uleer, C.; Peschel, S.; John, J.; et al. Proteomic profiling of breast cancer metabolism identifies SHMT2 and ASCT2 as prognostic factors. Breast Cancer Res. 2017, 19, 112. [Google Scholar] [CrossRef]
  30. Bjørnstad, O.V.; Carrasco, M.; Finne, K.; Ardawatia, V.; Winge, I.; Askeland, C.; Arnes, J.B.; Knutsvik, G.; Kleftogiannis, D.; Paulo, J.A.; et al. Global and single-cell proteomics view of the co-evolution between neural progenitors and breast cancer cells in a co-culture model. EBioMedicine 2024, 108, 105325. [Google Scholar] [CrossRef]
  31. Böhm, D.; Keller, K.; Pieter, J.; Boehm, N.; Wolters, D.; Siggelkow, W.; Lebrecht, A.; Schmidt, M.; Kölbl, H.; Pfeiffer, N.; et al. Comparison of tear protein levels in breast cancer patients and healthy controls using a de novo proteomic approach. Oncol. Rep. 2012, 28, 429–438. [Google Scholar] [CrossRef]
  32. Bonneterre, J.; Révillion, F.; Desauw, C.; Blot, E.; Kramar, A.; Fournier, C.; Hornez, L.; Peyrat, J.P. Plasma and tissue proteomic prognostic factors of response in primary breast cancer patients receiving neoadjuvant chemotherapy. Oncol. Rep. 2013, 29, 355–361. [Google Scholar] [CrossRef] [PubMed]
  33. Bouchal, P.; Dvořáková, M.; Roumeliotis, T.; Bortlíček, Z.; Ihnatová, I.; Procházková, I.; Ho, J.T.; Maryáš, J.; Imrichová, H.; Budinská, E.; et al. Combined Proteomics and Transcriptomics Identifies Carboxypeptidase B1 and Nuclear Factor κB (NF-κB) Associated Proteins as Putative Biomarkers of Metastasis in Low Grade Breast Cancer. Mol. Cell Proteom. 2015, 14, 1814–1830. [Google Scholar] [CrossRef]
  34. Braakman, R.B.; Bezstarosti, K.; Sieuwerts, A.M.; de Weerd, V.; van Galen, A.M.; Stingl, C.; Luider, T.M.; Timmermans, M.A.; Smid, M.; Martens, J.W.; et al. Integrative analysis of genomics and proteomics data on clinical breast cancer tissue specimens extracted with acid guanidinium thiocyanate-phenol-chloroform. J. Proteome Res. 2015, 14, 1627–1636. [Google Scholar] [CrossRef]
  35. Brunoro, G.V.F.; Carvalho, P.C.; Barbosa, V.C.; Pagnoncelli, D.; De Moura Gallo, C.V.; Perales, J.; Zahedi, R.P.; Valente, R.H.; Neves-Ferreira, A.G.D.C. Differential proteomic comparison of breast cancer secretome using a quantitative paired analysis workflow. BMC Cancer 2019, 19, 365. [Google Scholar] [CrossRef]
  36. Cabezón, T.; Gromova, I.; Gromov, P.; Serizawa, R.; Timmermans Wielenga, V.; Kroman, N.; Celis, J.E.; Moreira, J.M. Proteomic profiling of triple-negative breast carcinomas in combination with a three-tier orthogonal technology approach identifies Mage-A4 as potential therapeutic target in estrogen receptor negative breast cancer. Mol. Cell Proteom. 2013, 12, 381–394. [Google Scholar] [CrossRef] [PubMed]
  37. Cancemi, P.; Di Cara, G.; Albanese, N.N.; Costantini, F.; Marabeti, M.R.; Musso, R.; Lupo, C.; Roz, E.; Pucci-Minafra, I. Large-scale proteomic identification of S100 proteins in breast cancer tissues. BMC Cancer 2010, 10, 476. [Google Scholar] [CrossRef] [PubMed]
  38. Cancemi, P.; Di Cara, G.; Albanese, N.N.; Costantini, F.; Marabeti, M.R.; Musso, R.; Riili, I.; Lupo, C.; Roz, E.; Pucci-Minafra, I. Differential occurrence of S100A7 in breast cancer tissues: A proteomic-based investigation. Proteom. Clin. Appl. 2012, 6, 364–373. [Google Scholar] [CrossRef]
  39. Cawthorn, T.R.; Moreno, J.C.; Dharsee, M.; Tran-Thanh, D.; Ackloo, S.; Zhu, P.H.; Sardana, G.; Chen, J.; Kupchak, P.; Jacks, L.M.; et al. Proteomic analyses reveal high expression of decorin and endoplasmin (HSP90B1) are associated with breast cancer metastasis and decreased survival. PLoS ONE 2012, 7, e30992. [Google Scholar] [CrossRef]
  40. Champattanachai, V.; Netsirisawan, P.; Chaiyawat, P.; Phueaouan, T.; Charoenwattanasatien, R.; Chokchaichamnankit, D.; Punyarit, P.; Srisomsap, C.; Svasti, J. Proteomic analysis and abrogated expression of O-GlcNAcylated proteins associated with primary breast cancer. Proteomics 2013, 13, 2088–2099. [Google Scholar] [CrossRef]
  41. Corrêa, S.; Panis, C.; Binato, R.; Herrera, A.C.; Pizzatti, L.; Abdelhay, E. Identifying potential markers in Breast Cancer subtypes using plasma label-free proteomics. J. Proteom. 2017, 151, 33–42. [Google Scholar] [CrossRef]
  42. Creighton, C.J.; Fu, X.; Hennessy, B.T.; Casa, A.J.; Zhang, Y.; Gonzalez-Angulo, A.M.; Lluch, A.; Gray, J.W.; Brown, P.H.; Hilsenbeck, S.G.; et al. Proteomic and transcriptomic profiling reveals a link between the PI3K pathway and lower estrogen-receptor (ER) levels and activity in ER+ breast cancer. Breast Cancer Res. 2010, 12, R40. [Google Scholar] [CrossRef] [PubMed]
  43. Dalenc, F.; Doisneau-Sixou, S.F.; Allal, B.C.; Marsili, S.; Lauwers-Cances, V.; Chaoui, K.; Schiltz, O.; Monsarrat, B.; Filleron, T.; Renée, N.; et al. Tipifarnib plus tamoxifen in tamoxifen-resistant metastatic breast cancer: A negative phase II and screening of potential therapeutic markers by proteomic analysis. Clin. Cancer Res. 2010, 16, 1264–1271. [Google Scholar] [CrossRef]
  44. Debets, D.O.; Stecker, K.E.; Piskopou, A.; Liefaard, M.C.; Wesseling, J.; Sonke, G.S.; Lips, E.H.; Altelaar, M. Deep (phospho)proteomics profiling of pre- treatment needle biopsies identifies signatures of treatment resistance in HER2+ breast cancer. Cell Rep. Med. 2023, 4, 101203. [Google Scholar] [CrossRef]
  45. Di Cara, G.; Marabeti, M.R.; Musso, R.; Riili, I.; Cancemi, P.; Pucci Minafra, I. New Insights into the Occurrence of Matrix Metalloproteases -2 and -9 in a Cohort of Breast Cancer Patients and Proteomic Correlations. Cells 2018, 7, 89. [Google Scholar] [CrossRef]
  46. Drukier, A.K.; Ossetrova, N.; Schors, E.; Krasik, G.; Grigoriev, I.; Koenig, C.; Sulkowski, M.; Holcman, J.; Brown, L.R.; Tomaszewski, J.E.; et al. High-sensitivity blood-based detection of breast cancer by multi photon detection diagnostic proteomics. J. Proteome Res. 2006, 5, 1906–1915. [Google Scholar] [CrossRef] [PubMed]
  47. Duan, J.; Zhao, Y.; Sun, Q.; Liang, D.; Liu, Z.; Chen, X.; Li, Z.C. Imaging-proteomic analysis for prediction of neoadjuvant chemotherapy responses in patients with breast cancer. Cancer Med. 2023, 12, 21256–21269. [Google Scholar] [CrossRef]
  48. Fernandez-Pol, J.A.; Hamilton, P.D.; Klos, D.J. Genomics, Proteomics and Cancer: Specific Ribosomal, Mitochondrial, and Tumor Reactive Proteins Can Be Used as Biomarkers for Early Detection of Breast Cancer in Serum. Cancer Genom. Proteom. 2005, 2, 1–24. [Google Scholar]
  49. Fonseca-Sánchez, M.A.; Rodríguez Cuevas, S.; Mendoza-Hernández, G.; Bautista-Piña, V.; Arechaga Ocampo, E.; Hidalgo Miranda, A.; Quintanar Jurado, V.; Marchat, L.A.; Alvarez-Sánchez, E.; Pérez Plasencia, C.; et al. Breast cancer proteomics reveals a positive correlation between glyoxalase 1 expression and high tumor grade. Int. J. Oncol. 2012, 41, 670–680. [Google Scholar] [CrossRef]
  50. Fredolini, C.; Pathak, K.V.; Paris, L.; Chapple, K.M.; Tsantilas, K.A.; Rosenow, M.; Tegeler, T.J.; Garcia-Mansfield, K.; Tamburro, D.; Zhou, W.; et al. Shotgun proteomics coupled to nanoparticle-based biomarker enrichment reveals a novel panel of extracellular matrix proteins as candidate serum protein biomarkers for early-stage breast cancer detection. Breast Cancer Res. 2020, 22, 135. [Google Scholar] [CrossRef]
  51. Gajbhiye, A.; Dabhi, R.; Taunk, K.; Jagadeeshaprasad, M.G.; RoyChoudhury, S.; Mane, A.; Bayatigeri, S.; Chaudhury, K.; Santra, M.K.; Rapole, S. Multipronged quantitative proteomics reveals serum proteome alterations in breast cancer intrinsic subtypes. J. Proteom. 2017, 163, 1–13. [Google Scholar] [CrossRef]
  52. Gajbhiye, A.; Dabhi, R.; Taunk, K.; Vannuruswamy, G.; RoyChoudhury, S.; Adhav, R.; Seal, S.; Mane, A.; Bayatigeri, S.; Santra, M.K.; et al. Urinary proteome alterations in HER2 enriched breast cancer revealed by multipronged quantitative proteomics. Proteomics 2016, 16, 2403–2418. [Google Scholar] [CrossRef] [PubMed]
  53. Gámez-Pozo, A.; Berges-Soria, J.; Arevalillo, J.M.; Nanni, P.; López-Vacas, R.; Navarro, H.; Grossmann, J.; Castaneda, C.A.; Main, P.; Díaz-Almirón, M.; et al. Combined Label-Free Quantitative Proteomics and microRNA Expression Analysis of Breast Cancer Unravel Molecular Differences with Clinical Implications. Cancer Res. 2015, 75, 2243–2253. [Google Scholar] [CrossRef]
  54. Gámez-Pozo, A.; Trilla-Fuertes, L.; Berges-Soria, J.; Selevsek, N.; López-Vacas, R.; Díaz-Almirón, M.; Nanni, P.; Arevalillo, J.M.; Navarro, H.; Grossmann, J.; et al. Functional proteomics outlines the complexity of breast cancer molecular subtypes. Sci. Rep. 2017, 7, 10100. [Google Scholar] [CrossRef]
  55. Gámez-Pozo, A.; Trilla-Fuertes, L.; Prado-Vázquez, G.; Chiva, C.; López-Vacas, R.; Nanni, P.; Berges-Soria, J.; Grossmann, J.; Díaz-Almirón, M.; Ciruelos, E.; et al. Prediction of adjuvant chemotherapy response in triple negative breast cancer with discovery and targeted proteomics. PLoS ONE 2017, 12, e0178296. [Google Scholar] [CrossRef] [PubMed]
  56. García-Adrián, S.; Trilla-Fuertes, L.; Gámez-Pozo, A.; Chiva, C.; López-Vacas, R.; López-Camacho, E.; Zapater-Moros, A.; Lumbreras-Herrera, M.I.; Hardisson, D.; Yébenes, L.; et al. Molecular characterization of triple negative breast cancer formaldehyde-fixed paraffin-embedded samples by data-independent acquisition proteomics. Proteomics 2022, 22, e2100110. [Google Scholar] [CrossRef]
  57. Garrisi, V.M.; Tommasi, S.; Facchiano, A.; Bongarzone, I.; De Bortoli, M.; Cremona, M.; Cafagna, V.; Abbate, I.; Tufaro, A.; Quaranta, M.; et al. Proteomic profile in familial breast cancer patients. Clin. Biochem. 2013, 46, 259–265. [Google Scholar] [CrossRef] [PubMed]
  58. Garrisi, V.M.; Tufaro, A.; Trerotoli, P.; Bongarzone, I.; Quaranta, M.; Ventrella, V.; Tommasi, S.; Giannelli, G.; Paradiso, A. Body mass index and serum proteomic profile in breast cancer and healthy women: A prospective study. PLoS ONE 2012, 7, e49631. [Google Scholar] [CrossRef]
  59. Gast, M.C.; Zapatka, M.; van Tinteren, H.; Bontenbal, M.; Span, P.N.; Tjan-Heijnen, V.C.; Knol, J.C.; Jimenez, C.R.; Schellens, J.H.; Beijnen, J.H. Postoperative serum proteomic profiles may predict recurrence-free survival in high-risk primary breast cancer. J. Cancer Res. Clin. Oncol. 2011, 137, 1773–1783. [Google Scholar] [CrossRef]
  60. Giri, K.; Maity, S.; Ambatipudi, K. Targeted proteomics using parallel reaction monitoring confirms salivary proteins indicative of metastatic triple-negative breast cancer. J. Proteom. 2022, 267, 104701. [Google Scholar] [CrossRef]
  61. Gonçalves, A.; Esterni, B.; Bertucci, F.; Sauvan, R.; Chabannon, C.; Cubizolles, M.; Bardou, V.J.; Houvenaegel, G.; Jacquemier, J.; Granjeaud, S.; et al. Postoperative serum proteomic profiles may predict metastatic relapse in high-risk primary breast cancer patients receiving adjuvant chemotherapy. Oncogene 2006, 25, 981–989. [Google Scholar] [CrossRef]
  62. Gonzalez-Angulo, A.M.; Hennessy, B.T.; Meric-Bernstam, F.; Sahin, A.; Liu, W.; Ju, Z.; Carey, M.S.; Myhre, S.; Speers, C.; Deng, L.; et al. Functional proteomics can define prognosis and predict pathologic complete response in patients with breast cancer. Clin. Proteom. 2011, 8, 11. [Google Scholar] [CrossRef] [PubMed]
  63. Gonzalez-Angulo, A.M.; Liu, S.; Chen, H.; Chavez-Macgregor, M.; Sahin, A.; Hortobagyi, G.N.; Mills, G.B.; Do, K.A.; Meric-Bernstam, F. Functional proteomics characterization of residual breast cancer after neoadjuvant systemic chemotherapy. Ann. Oncol. 2013, 24, 909–916. [Google Scholar] [CrossRef] [PubMed]
  64. Grassmann, F.; Mälarstig, A.; Dahl, L.; Bendes, A.; Dale, M.; Thomas, C.E.; Gabrielsson, M.; Hedman, Å.K.; Eriksson, M.; Margolin, S.; et al. The impact of circulating protein levels identified by affinity proteomics on short-term, overall breast cancer risk. Br. J. Cancer 2024, 130, 620–627. [Google Scholar] [CrossRef] [PubMed]
  65. Gromova, I.; Espinoza, J.A.; Grauslund, M.; Santoni-Rugiu, E.; Møller Talman, M.L.; van Oostrum, J.; Moreira, J.M.A. Functional Proteomic Profiling of Triple-Negative Breast Cancer. Cells 2021, 10, 2768. [Google Scholar] [CrossRef]
  66. Guerin, M.; Gonçalves, A.; Toiron, Y.; Baudelet, E.; Pophillat, M.; Granjeaud, S.; Fourquet, P.; Jacot, W.; Tarpin, C.; Sabatier, R.; et al. Development of parallel reaction monitoring (PRM)-based quantitative proteomics applied to HER2-Positive breast cancer. Oncotarget 2018, 9, 33762–33777. [Google Scholar] [CrossRef]
  67. Gustafsson, A.; Jonasson, E.; Ståhlberg, A.; Landberg, G. Proteomics of cell-free breast cancer scaffolds identify clinically relevant imprinted proteins and cancer-progressing properties. Cancer Commun. 2024, 44, 695–699. [Google Scholar] [CrossRef]
  68. He, J.; Shen, D.; Chung, D.U.; Saxton, R.E.; Whitelegge, J.P.; Faull, K.F.; Chang, H.R. Tumor proteomic profiling predicts the susceptibility of breast cancer to chemotherapy. Int. J. Oncol. 2009, 35, 683–692. [Google Scholar] [CrossRef]
  69. He, J.; Whelan, S.A.; Lu, M.; Shen, D.; Chung, D.U.; Saxton, R.E.; Faull, K.F.; Whitelegge, J.P.; Chang, H.R. Proteomic-based biosignatures in breast cancer classification and prediction of therapeutic response. Int. J. Proteom. 2011, 896476. [Google Scholar] [CrossRef]
  70. Henderson, M.C.; Hollingsworth, A.B.; Gordon, K.; Silver, M.; Mulpuri, R.; Letsios, E.; Reese, D.E. Integration of Serum Protein Biomarker and Tumor Associated Autoantibody Expression Data Increases the Ability of a Blood-Based Proteomic Assay to Identify Breast Cancer. PLoS ONE 2016, 11, e0157692. [Google Scholar] [CrossRef]
  71. Henderson, M.C.; Silver, M.; Tran, Q.; Letsios, E.E.; Mulpuri, R.; Reese, D.E.; Lourenco, A.P.; LaBaer, J.; Anderson, K.S.; Alpers, J.; et al. A Noninvasive Blood-based Combinatorial Proteomic Biomarker Assay to Detect Breast Cancer in Women over age 50 with BI-RADS 3, 4, or 5 Assessment. Clin. Cancer Res. 2019, 25, 142–149. [Google Scholar] [CrossRef]
  72. Hu, Y.; Zhang, S.; Yu, J.; Liu, J.; Zheng, S. SELDI-TOF-MS: The proteomics and bioinformatics approaches in the diagnosis of breast cancer. Breast 2005, 14, 250–255. [Google Scholar] [CrossRef]
  73. Hulahan, T.S.; Spruill, L.; Wallace, E.N.; Park, Y.; West, R.B.; Marks, J.R.; Hwang, E.S.; Drake, R.R.; Angel, P.M. Extracellular Microenvironment Alterations in Ductal Carcinoma In Situ and Invasive Breast Cancer Pathologies by Multiplexed Spatial Proteomics. Int. J. Mol. Sci. 2024, 25, 6748. [Google Scholar] [CrossRef]
  74. Jeanmard, N.; Bissanum, R.; Sriplung, H.; Charoenlappanit, S.; Roytrakul, S.; Navakanitworakul, R. Proteomic profiling of urinary extracellular vesicles differentiates breast cancer patients from healthy women. PLoS ONE 2023, 18, e0291574. [Google Scholar] [CrossRef] [PubMed]
  75. Jeon, Y.; Lee, G.; Jeong, H.; Gong, G.; Kim, J.; Kim, K.; Jeong, J.H.; Lee, H.J. Proteomic analysis of breast cancer based on immune subtypes. Clin. Proteom. 2024, 21, 17. [Google Scholar] [CrossRef]
  76. Johansson, H.J.; Sanchez, B.C.; Forshed, J.; Stål, O.; Fohlin, H.; Lewensohn, R.; Hall, P.; Bergh, J.; Lehtiö, J.; Linderholm, B.K. Proteomics profiling identify CAPS as a potential predictive marker of tamoxifen resistance in estrogen receptor positive breast cancer. Clin. Proteom. 2015, 12, 8. [Google Scholar] [CrossRef] [PubMed]
  77. Jordan, K.R.; Hall, J.K.; Schedin, T.; Borakove, M.; Xian, J.J.; Dzieciatkowska, M.; Lyons, T.R.; Schedin, P.; Hansen, K.C.; Borges, V.F. Extracellular vesicles from young women’s breast cancer patients drive increased invasion of non-malignant cells via the Focal Adhesion Kinase pathway: A proteomic approach. Breast Cancer Res. 2020, 22, 128. [Google Scholar] [CrossRef] [PubMed]
  78. Kang, S.; Kim, M.J.; An, H.; Kim, B.G.; Choi, Y.P.; Kang, K.S.; Gao, M.Q.; Park, H.; Na, H.J.; Kim, H.K.; et al. Proteomic molecular portrait of interface zone in breast cancer. J. Proteome Res. 2010, 9, 5638–5645. [Google Scholar] [CrossRef]
  79. Kaur, J.; Jung, S.Y.; Austdal, M.; Arun, A.K.; Helland, T.; Mellgren, G.; Lende, T.H.; Janssen, E.A.M.; Søiland, H.; Aneja, R. Quantitative proteomics reveals serum proteome alterations during metastatic disease progression in breast cancer patients. Clin. Proteom. 2024, 21, 52. [Google Scholar] [CrossRef]
  80. Kim, D.H.; Bae, J.; Lee, J.W.; Kim, S.Y.; Kim, Y.H.; Bae, J.Y.; Yi, J.K.; Yu, M.H.; Noh, D.Y.; Lee, C. Proteomic analysis of breast cancer tissue reveals upregulation of actin-remodeling proteins and its relevance to cancer invasiveness. Proteom. Clin. Appl. 2009, 3, 30–40. [Google Scholar] [CrossRef]
  81. Kim, Y.; Kang, U.B.; Kim, S.; Lee, H.B.; Moon, H.G.; Han, W.; Noh, D.Y. A Validation Study of a Multiple Reaction Monitoring-Based Proteomic Assay to Diagnose Breast Cancer. J. Breast Cancer 2019, 22, 579–586. [Google Scholar] [CrossRef]
  82. Kuerer, H.M.; Coombes, K.R.; Chen, J.N.; Xiao, L.; Clarke, C.; Fritsche, H.; Krishnamurthy, S.; Marcy, S.; Hung, M.C.; Hunt, K.K. Association between ductal fluid proteomic expression profiles and the presence of lymph node metastases in women with breast cancer. Surgery 2004, 136, 1061–1069. [Google Scholar] [CrossRef] [PubMed]
  83. Le Naour, F.; Misek, D.E.; Krause, M.C.; Deneux, L.; Giordano, T.J.; Scholl, S.; Hanash, S.M. Proteomics-based identification of RS/DJ-1 as a novel circulating tumor antigen in breast cancer. Clin. Cancer Res. 2001, 7, 3328–3335. [Google Scholar]
  84. Lebrecht, A.; Boehm, D.; Schmidt, M.; Koelbl, H.; Schwirz, R.L.; Grus, F.H. Diagnosis of breast cancer by tear proteomic pattern. Cancer Genom. Proteom. 2009, 6, 177–182. [Google Scholar]
  85. Li, Y.; Yue, L.; Zhang, S.; Wang, X.; Zhu, Y.N.; Liu, J.; Ren, H.; Jiang, W.; Wang, J.; Zhang, Z.; et al. Proteomic, single-cell and bulk transcriptomic analysis of plasma and tumor tissues unveil core proteins in response to anti-PD-L1 immunotherapy in triple negative breast cancer. Comput. Biol. Med. 2024, 176, 108537. [Google Scholar] [CrossRef]
  86. Lin, Y.; Lin, L.; Fu, F.; Wang, C.; Hu, A.; Xie, J.; Jiang, M.; Wang, Z.; Yang, L.; Guo, R.; et al. Quantitative proteomics reveals stage-specific protein regulation of triple negative breast cancer. Breast Cancer Res. Treat. 2021, 185, 39–52. [Google Scholar] [CrossRef] [PubMed]
  87. Lötsch, J.; Mustonen, L.; Harno, H.; Kalso, E. Machine-Learning Analysis of Serum Proteomics in Neuropathic Pain after Nerve Injury in Breast Cancer Surgery Points at Chemokine Signaling via SIRT2 Regulation. Int. J. Mol. Sci. 2022, 23, 3488. [Google Scholar] [CrossRef]
  88. Lourenco, A.P.; Benson, K.L.; Henderson, M.C.; Silver, M.; Letsios, E.; Tran, Q.; Gordon, K.J.; Borman, S.; Corn, C.; Mulpuri, R.; et al. A Noninvasive Blood-based Combinatorial Proteomic Biomarker Assay to Detect Breast Cancer in Women Under the Age of 50 Years. Clin. Breast Cancer 2017, 17, 516–525.e6. [Google Scholar] [CrossRef]
  89. Magara, K.; Takasawa, A.; Takasawa, K.; Aoyama, T.; Ota, M.; Kyuno, D.; Ono, Y.; Murakami, T.; Yamamoto, S.; Nakamori, Y.; et al. Multilayered proteomics reveals that JAM-A promotes breast cancer progression via regulation of amino acid transporter LAT1. Cancer Sci. 2024, 115, 3153–3168. [Google Scholar] [CrossRef]
  90. Majidzadeh, A.K.; Gharechahi, J. Plasma proteomics analysis of tamoxifen resistance in breast cancer. Med. Oncol. 2013, 30, 753. [Google Scholar] [CrossRef]
  91. Mayayo-Peralta, I.; Debets, D.O.; Prekovic, S.; Schuurman, K.; Beerthuijzen, S.; Almekinders, M.; Sanders, J.; Moelans, C.B.; Saleiro, S.; Wesseling, J.; et al. Proteomics on malignant pleural effusions reveals ERα loss in metastatic breast cancer associates with SGK1-NDRG1 deregulation. Mol. Oncol. 2024, 18, 156–169. [Google Scholar] [CrossRef]
  92. Meric-Bernstam, F.; Akcakanat, A.; Chen, H.; Sahin, A.; Tarco, E.; Carkaci, S.; Adrada, B.E.; Singh, G.; Do, K.A.; Garces, Z.M.; et al. Influence of biospecimen variables on proteomic biomarkers in breast cancer. Clin. Cancer Res. 2014, 20, 3870–3883. [Google Scholar] [CrossRef] [PubMed]
  93. Michaut, M.; Chin, S.F.; Majewski, I.; Severson, T.M.; Bismeijer, T.; de Koning, L.; Peeters, J.K.; Schouten, P.C.; Rueda, O.M.; Bosma, A.J.; et al. Integration of genomic, transcriptomic and proteomic data identifies two biologically distinct subtypes of invasive lobular breast cancer. Sci. Rep. 2016, 6, 18517. [Google Scholar] [CrossRef]
  94. Minton, O.; Stone, P.C. The identification of plasma proteins associated with cancer-related fatigue syndrome (CRFS) in disease-free breast cancer patients using proteomic analysis. J. Pain Symptom Manag. 2013, 45, 868–874. [Google Scholar] [CrossRef] [PubMed]
  95. Moriggi, M.; Giussani, M.; Torretta, E.; Capitanio, D.; Sandri, M.; Leone, R.; De Palma, S.; Vasso, M.; Vozzi, G.; Tagliabue, E.; et al. ECM Remodeling in Breast Cancer with Different Grade: Contribution of 2D-DIGE Proteomics. Proteomics 2018, 18, e1800278. [Google Scholar] [CrossRef] [PubMed]
  96. Nakagawa, T.; Huang, S.K.; Martinez, S.R.; Tran, A.N.; Elashoff, D.; Ye, X.; Turner, R.R.; Giuliano, A.E.; Hoon, D.S. Proteomic profiling of primarybreast cancer predicts axillary lymph node metastasis. Cancer Res. 2006, 66, 11825–11830. [Google Scholar] [CrossRef]
  97. Neubauer, H.; Clare, S.E.; Kurek, R.; Fehm, T.; Wallwiener, D.; Sotlar, K.; Nordheim, A.; Wozny, W.; Schwall, G.P.; Poznanović, S.; et al. Breast cancer proteomics by laser capture microdissection, sample pooling, 54-cm IPGIEF, and differential iodine radioisotope detection. Electrophoresis 2006, 27, 1840–1852. [Google Scholar] [CrossRef]
  98. Neubauer, H.; Clare, S.E.; Wozny, W.; Schwall, G.P.; Poznanovic, S.; Stegmann, W.; Vogel, U.; Sotlar, K.; Wallwiener, D.; Kurek, R.; et al. Breast cancer proteomics reveals correlation between estrogen receptor status and differential phosphorylation of PGRMC1. Breast Cancer Res. 2008, 10, R85. [Google Scholar] [CrossRef]
  99. Niméus, E.; Malmström, J.; Johnsson, A.; Marko-Varga, G.; Fernö, M. Proteomic analysis identifies candidate proteins associated with distant recurrences in breast cancer after adjuvant chemotherapy. J. Pharm. Biomed. Anal. 2007, 43, 1086–1093. [Google Scholar] [CrossRef]
  100. Othman, M.I.; Majid, M.I.; Singh, M.; Subathra, S.; Seng, L.; Gam, L.H. Proteomics of Grade 3 infiltrating ductal carcinoma in Malaysian Chinese breast cancer patients. Biotechnol. Appl. Biochem. 2009, 52, 209–219. [Google Scholar] [CrossRef]
  101. Ou, K.; Yu, K.; Kesuma, D.; Hooi, M.; Huang, N.; Chen, W.; Lee, S.Y.; Goh, X.P.; Tan, L.K.; Liu, J.; et al. Novel breast cancer biomarkers identified by integrative proteomic and gene expression mapping. J. Proteome Res. 2008, 7, 1518–1528. [Google Scholar] [CrossRef]
  102. Panis, C.; Pizzatti, L.; Herrera, A.C.; Cecchini, R.; Abdelhay, E. Putative circulating markers of the early and advanced stages of breast cancer identified by high-resolution label-free proteomics. Cancer Lett. 2013, 330, 57–66. [Google Scholar] [CrossRef] [PubMed]
  103. Pathania, S.; Khan, M.I.; Bandyopadhyay, S.; Singh, S.S.; Rani, K.; Parashar, T.R.; Jayaram, J.; Mishra, P.R.; Srivastava, A.; Mathur, S.; et al. iTRAQ proteomics of sentinel lymph nodes for identification of extracellular matrix proteins to flag metastasis in early breast cancer. Sci. Rep. 2022, 12, 8625. [Google Scholar] [CrossRef]
  104. Paweletz, C.P.; Trock, B.; Pennanen, M.; Tsangaris, T.; Magnant, C.; Liotta, L.A.; Petricoin, E.F., 3rd. Proteomic patterns of nipple aspirate fluids obtained by SELDI-TOF: Potential for new biomarkers to aid in the diagnosis of breast cancer. Dis. Markers 2001, 17, 301–307. [Google Scholar] [CrossRef]
  105. Pawlik, T.M.; Hawke, D.H.; Liu, Y.; Krishnamurthy, S.; Fritsche, H.; Hunt, K.K.; Kuerer, H.M. Proteomic analysis of nipple aspirate fluid from women with early-stage breast cancer using isotope-coded affinity tags and tandem mass spectrometry reveals differential expression of vitamin D binding protein. BMC Cancer 2006, 6, 68. [Google Scholar] [CrossRef]
  106. Pires, B.R.B.; Panis, C.; Alves, V.D.; Herrera, A.C.S.A.; Binato, R.; Pizzatti, L.; Cecchini, R.; Abdelhay, E. Label-Free Proteomics Revealed Oxidative Stress and Inflammation as Factors That Enhance Chemoresistance in Luminal Breast Cancer. Oxidative Med. Cell. Longev. 2019, 2019, 5357649. [Google Scholar] [CrossRef]
  107. Pozniak, Y.; Balint-Lahat, N.; Rudolph, J.D.; Lindskog, C.; Katzir, R.; Avivi, C.; Pontén, F.; Ruppin, E.; Barshack, I.; Geiger, T. System-wide Clinical Proteomics of Breast Cancer Reveals Global Remodeling of Tissue Homeostasis. Cell Syst. 2016, 2, 172–184. [Google Scholar] [CrossRef]
  108. Procházková, I.; Lenčo, J.; Bouchal, P. Targeted Proteomics Driven Verification of Biomarker Candidates Associated with Breast Cancer Aggressiveness. Methods Mol. Biol. 2018, 1788, 177–184. [Google Scholar] [CrossRef] [PubMed]
  109. Pucci-Minafra, I.; Cancemi, P.; Albanese, N.N.; Di Cara, G.; Marabeti, M.R.; Marrazzo, A.; Minafra, S. New protein clustering of breast cancer tissue proteomics using actin content as a cellularity indicator. J. Proteome Res. 2008, 7, 1412–1418. [Google Scholar] [CrossRef] [PubMed]
  110. Pucci-Minafra, I.; Di Cara, G.; Musso, R.; Cancemi, P.; Albanese, N.N.; Roz, E.; Minafra, S. Retrospective Proteomic Screening of 100 Breast Cancer Tissues. Proteomes 2017, 5, 15. [Google Scholar] [CrossRef]
  111. Riley, C.P.; Zhang, X.; Nakshatri, H.; Schneider, B.; Regnier, F.E.; Adamec, J.; Buck, C. A large, consistent plasma proteomics data set from prospectively collected breast cancer patient and healthy volunteer samples. J. Transl. Med. 2011, 9, 80. [Google Scholar] [CrossRef]
  112. Roberts, K.; Bhatia, K.; Stanton, P.; Lord, R. Proteomic analysis of selected prognostic factors of breast cancer. Proteomics 2004, 4, 784–792. [Google Scholar] [CrossRef] [PubMed]
  113. Rui, Z.; Jian-Guo, J.; Yuan-Peng, T.; Hai, P.; Bing-Gen, R. Use of serological proteomic methods to find biomarkers associated with breast cancer. Proteomics 2003, 3, 433–439. [Google Scholar] [CrossRef] [PubMed]
  114. Rojas, L.K.; Trilla-Fuertes, L.; Gámez-Pozo, A.; Chiva, C.; Sepúlveda, J.; Manso, L.; Prado-Vázquez, G.; Zapater-Moros, A.; López-Vacas, R.; Ferrer-Gómez, M.; et al. Proteomics characterisation of central nervous system metastasis biomarkers in triple negative breast cancer. Ecancermedicalscience 2019, 13, 891. [Google Scholar] [CrossRef]
  115. Ruckhäberle, E.; Karn, T.; Hanker, L.; Schwarz, J.; Schulz-Knappe, P.; Kuhn, K.; Böhm, G.; Selzer, S.; Erhard, N.; Engels, K.; et al. Breast Cancer Proteomics—Differences in Protein Expression between Estrogen Receptor-Positive and -Negative Tumors Identified by Tandem Mass Tag Technology. Breast Care 2010, 5, 7–10. [Google Scholar] [CrossRef] [PubMed]
  116. Sanders, M.E.; Dias, E.C.; Xu, B.J.; Mobley, J.A.; Billheimer, D.; Roder, H.; Grigorieva, J.; Dowsett, M.; Arteaga, C.L.; Caprioli, R.M. Differentiating proteomic biomarkers in breastcancer by laser capture microdissection and MALDI MS. J. Proteome Res. 2008, 7, 1500–1507. [Google Scholar] [CrossRef]
  117. Santana, M.F.M.; Sawada, M.I.B.A.C.; Junior, D.R.S.; Giacaglia, M.B.; Reis, M.; Xavier, J.; Côrrea-Giannella, M.L.; Soriano, F.G.; Gebrim, L.H.; Ronsein, G.E.; et al. Proteomic Profiling of HDL in Newly Diagnosed Breast Cancer Based on Tumor Molecular Classification and Clinical Stage of Disease. Cells 2024, 13, 1327. [Google Scholar] [CrossRef]
  118. Sauter, E.R.; Zhu, W.; Fan, X.J.; Wassell, R.P.; Chervoneva, I.; Du Bois, G.C. Proteomic analysis of nipple aspirate fluid to detect biologic markers of breast cancer. Br. J. Cancer 2002, 86, 1440–1443. [Google Scholar] [CrossRef]
  119. Schaub, N.P.; Jones, K.J.; Nyalwidhe, J.O.; Cazares, L.H.; Karbassi, I.D.; Semmes, O.J.; Feliberti, E.C.; Perry, R.R.; Drake, R.R. Serum proteomic biomarker discovery reflective of stage and obesity in breast cancer patients. J. Am. Coll. Surg. 2009, 208, 970–980. [Google Scholar] [CrossRef]
  120. Shenoy, A.; Belugali Nataraj, N.; Perry, G.; Loayza Puch, F.; Nagel, R.; Marin, I.; Balint, N.; Bossel, N.; Pavlovsky, A.; Barshack, I.; et al. Proteomic patterns associated with response to breast cancer neoadjuvant treatment. Mol. Syst. Biol. 2020, 16, e9443. [Google Scholar] [CrossRef]
  121. Shi, X.; Liu, C.; Zheng, W.; Cao, X.; Li, W.; Zhang, D.; Zhu, J.; Zhang, X.; Chen, Y. Proteomic Analysis Revealed the Potential Role of MAGE-D2 in the Therapeutic Targeting of Triple-Negative Breast Cancer. Mol. Cell. Proteom. 2024, 23, 100703. [Google Scholar] [CrossRef]
  122. Shin, D.; Park, J.; Han, D.; Moon, J.H.; Ryu, H.S.; Kim, Y. Identification of TUBB2A by quantitative proteomic analysis as a novel biomarker for the prediction of distant metastatic breast cancer. Clin. Proteom. 2020, 17, 16. [Google Scholar] [CrossRef] [PubMed]
  123. Sinha, I.; Fogle, R.L.; Gulfidan, G.; Stanley, A.E.; Walter, V.; Hollenbeak, C.S.; Arga, K.Y.; Sinha, R. Potential Early Markers for Breast Cancer: A Proteomic Approach Comparing Saliva and Serum Samples in a Pilot Study. Int. J. Mol. Sci. 2023, 24, 4164. [Google Scholar] [CrossRef] [PubMed]
  124. Sohn, J.; Do, K.A.; Liu, S.; Chen, H.; Mills, G.B.; Hortobagyi, G.N.; Meric-Bernstam, F.; Gonzalez-Angulo, A.M. Functional proteomics characterization of residual triple-negative breast cancer after standard neoadjuvant chemotherapy. Ann. Oncol. 2013, 24, 2522–2526. [Google Scholar] [CrossRef]
  125. Starodubtseva, N.L.; Tokareva, A.O.; Rodionov, V.V.; Brzhozovskiy, A.G.; Bugrova, A.E.; Chagovets, V.V.; Kometova, V.V.; Kukaev, E.N.; Soares, N.C.; Kovalev, G.I.; et al. Integrating Proteomics and Lipidomics for Evaluating the Risk of Breast Cancer Progression: A Pilot Study. Biomedicines 2023, 11, 1786. [Google Scholar] [CrossRef]
  126. Stemke-Hale, K.; Gonzalez-Angulo, A.M.; Lluch, A.; Neve, R.M.; Kuo, W.L.; Davies, M.; Carey, M.; Hu, Z.; Guan, Y.; Sahin, A.; et al. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 2008, 68, 6084–6091. [Google Scholar] [CrossRef] [PubMed]
  127. Suman, S.; Basak, T.; Gupta, P.; Mishra, S.; Kumar, V.; Sengupta, S.; Shukla, Y. Quantitative proteomics revealed novel proteins associated with molecular subtypes of breast cancer. J. Proteom. 2016, 148, 183–193. [Google Scholar] [CrossRef]
  128. Tamesa, M.S.; Kuramitsu, Y.; Fujimoto, M.; Maeda, N.; Nagashima, Y.; Tanaka, T.; Yamamoto, S.; Oka, M.; Nakamura, K. Detection of autoantibodies against cyclophilin A and triosephosphate isomerase in sera from breast cancer patients by proteomic analysis. Electrophoresis 2009, 30, 2168–2181. [Google Scholar] [CrossRef]
  129. Terkelsen, T.; Pernemalm, M.; Gromov, P.; Børresen-Dale, A.L.; Krogh, A.; Haakensen, V.D.; Lethiö, J.; Papaleo, E.; Gromova, I. High-throughput proteomics of breast cancer interstitial fluid: Identification of tumor subtype-specific serologically relevant biomarkers. Clin. Trial Mol. Oncol. 2021, 15, 429–461. [Google Scholar] [CrossRef]
  130. Tutanov, O.; Orlova, E.; Proskura, K.; Grigor’eva, A.; Yunusova, N.; Tsentalovich, Y.; Alexandrova, A.; Tamkovich, S. Proteomic Analysis of Blood Exosomes from Healthy Females and Breast Cancer Patients Reveals an Association between Different Exosomal Bioactivity on Non-tumorigenic Epithelial Cell and Breast Cancer Cell Migration in Vitro. Biomolecules 2020, 10, 495. [Google Scholar] [CrossRef]
  131. Tyanova, S.; Albrechtsen, R.; Kronqvist, P.; Cox, J.; Mann, M.; Geiger, T. Proteomic maps of breast cancer subtypes. Nat. Commun. 2016, 7, 10259. [Google Scholar] [CrossRef]
  132. Valo, I.; Raro, P.; Boissard, A.; Maarouf, A.; Jézéquel, P.; Verriele, V.; Campone, M.; Coqueret, O.; Guette, C. OLFM4 Expression in Ductal Carcinoma In Situ and in Invasive Breast Cancer Cohorts by a SWATH-Based Proteomic Approach. Proteomics 2019, 19, e1800446. [Google Scholar] [CrossRef]
  133. Vinik, Y.; Ortega, F.G.; Mills, G.B.; Lu, Y.; Jurkowicz, M.; Halperin, S.; Aharoni, M.; Gutman, M.; Lev, S. Proteomic analysis of circulating extracellular vesicles identifies potential markers of breast cancer progression, recurrence, and response. Sci. Adv. 2020, 6, eaba5714. [Google Scholar] [CrossRef]
  134. Xu, G.; Huang, R.; Wumaier, R.; Lyu, J.; Huang, M.; Zhang, Y.; Chen, Q.; Liu, W.; Tao, M.; Li, J.; et al. Proteomic Profiling of Serum Extracellular Vesicles Identifies Diagnostic Signatures and Therapeutic Targets in Breast Cancer. Cancer Res. 2024, 84, 3267–3285. [Google Scholar] [CrossRef]
  135. Xu, Q.; Zhu, M.; Yang, T.; Xu, F.; Liu, Y.; Chen, Y. Quantitative assessment of human serum transferrin receptor in breast cancer patients pre- and post-chemotherapy using peptide immunoaffinity enrichment coupled with targeted proteomics. Clin. Chim. Acta 2015, 448, 118–123. [Google Scholar] [CrossRef] [PubMed]
  136. Yan, Y.; Zhou, Y.; Wang, K.; Qiao, Y.; Zhao, L.; Chen, M. Apolipoprotein C1 (APOC1), A Candidate Diagnostic Serum Biomarker for Breast Cancer Identified by Serum Proteomics Study. Crit. Rev. Eukaryot. Gene Expr. 2022, 32, 1–9. [Google Scholar] [CrossRef] [PubMed]
  137. Yang, T.; Fu, Z.; Zhang, Y.; Wang, M.; Mao, C.; Ge, W. Serum proteomics analysis of candidate predictive biomarker panel for the diagnosis of trastuzumab-based therapy resistant breast cancer. Biomed. Pharmacother. 2020, 129, 110465. [Google Scholar] [CrossRef] [PubMed]
  138. Yang, T.; Xu, F.; Fang, D.; Chen, Y. Targeted Proteomics Enables Simultaneous Quantification of Folate Receptor Isoforms and Potential Isoform-based Diagnosis in Breast Cancer. Sci. Rep. 2015, 5, 16733. [Google Scholar] [CrossRef]
  139. Yang, T.; Xu, F.; Sheng, Y.; Zhang, W.; Chen, Y. A targeted proteomics approach to the quantitative analysis of ERK/Bcl-2-mediated anti-apoptosis and multi-drug resistance in breast cancer. Anal. Bioanal. Chem. 2016, 408, 7491–7503. [Google Scholar] [CrossRef]
  140. Yang, T.; Xu, F.; Zhao, Y.; Wang, S.; Yang, M.; Chen, Y. A liquid chromatography-tandem mass spectrometry-based targeted proteomics approach for the assessment of transferrin receptor levels in breast cancer. Proteom. Clin. Appl. 2014, 8, 773–782. [Google Scholar] [CrossRef]
  141. Yang, W.S.; Moon, H.G.; Kim, H.S.; Choi, E.J.; Yu, M.H.; Noh, D.Y.; Lee, C. Proteomic approach reveals FKBP4 and S100A9 as potential prediction markers of therapeutic response to neoadjuvant chemotherapy in patients with breast cancer. J. Proteome Res. 2012, 11, 1078–1088. [Google Scholar] [CrossRef]
  142. Yanovich, G.; Agmon, H.; Harel, M.; Sonnenblick, A.; Peretz, T.; Geiger, T. Clinical Proteomics of Breast Cancer Reveals a Novel Layer of Breast Cancer Classification. Cancer Res. 2018, 78, 6001–6010. [Google Scholar] [CrossRef] [PubMed]
  143. Ye, H.; Shen, X.; Li, Y.; Zou, W.; Hassan, S.S.U.; Feng, Y.; Wang, X.; Tian, J.; Shao, X.; Tao, Y.; et al. Proteomic and metabolomic characterization of bone, liver, and lung metastases in plasma of breast cancer patients. Proteom. Clin. Appl. 2024, 18, e2300136. [Google Scholar] [CrossRef]
  144. Zeidan, B.; Manousopoulou, A.; Garay-Baquero, D.J.; White, C.H.; Larkin, S.E.T.; Potter, K.N.; Roumeliotis, T.I.; Papachristou, E.K.; Copson, E.; Cutress, R.I.; et al. Increased circulating resistin levels in early-onset breast cancer patients of normal body mass index correlate with lymph node negative involvement and longer disease free survival: A multi-center POSH cohort serum proteomics study. Breast Cancer Res. 2018, 20, 19. [Google Scholar] [CrossRef] [PubMed]
  145. Zeng, L.; Zhong, J.; He, G.; Li, F.; Li, J.; Zhou, W.; Liu, W.; Zhang, Y.; Huang, S.; Liu, Z.; et al. Identification of Nucleobindin-2 as a Potential Biomarker for Breast Cancer Metastasis Using iTRAQ-based Quantitative Proteomic Analysis. J. Cancer 2017, 8, 3062–3069. [Google Scholar] [CrossRef]
  146. Zhang, D.; Tai, L.K.; Wong, L.L.; Putti, T.C.; Sethi, S.K.; Teh, M.; Koay, E.S. Proteomic characterization of differentially expressed proteins in breast cancer: Expression of hnRNP H1, RKIP and GRP78 is strongly associated with HER-2/neu status. Proteom. Clin. Appl. 2008, 2, 99–107. [Google Scholar] [CrossRef]
  147. Zhang, D.H.; Tai, L.K.; Wong, L.L.; Sethi, S.K.; Koay, E.S. Proteomics of breast cancer: Enhanced expression of cytokeratin19 in human epidermal growth factor receptor type 2 positive breast tumors. Proteomics 2005, 5, 1797–1805. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, F.; Chen, J.; Wang, M.; Drabier, R. A neural network approach to multi-biomarker panel discovery by high-throughput plasma proteomics profiling of breast cancer. BMC Proc. 2013, 7, S10. [Google Scholar] [CrossRef]
  149. Zhang, F.; Deng, Y.; Wang, M.; Cui, L.; Drabier, R. Pathway-based Biomarkers for Breast Cancer in Proteomics. Cancer Inform. 2015, 13, 101–108. [Google Scholar] [CrossRef]
  150. Zhang, F.; Wang, M.; Michael, T.; Drabier, R. Novel alternative splicing isoform biomarkers identification from high-throughput plasma proteomics profiling of breast cancer. BMC Syst. Biol. 2013, 7 (Suppl. S5), S8. [Google Scholar] [CrossRef]
  151. Zhang, L.; Xiao, H.; Karlan, S.; Zhou, H.; Gross, J.; Elashoff, D.; Akin, D.; Yan, X.; Chia, D.; Karlan, B.; et al. Discovery and preclinical validation of salivary transcriptomic and proteomic biomarkers for the non-invasive detection of breast cancer. PLoS ONE 2010, 5, e15573. [Google Scholar] [CrossRef]
  152. Zhao, M.; Jiang, Y.; Kong, X.; Liu, Y.; Gao, P.; Li, M.; Zhu, H.; Deng, G.; Feng, Z.; Cao, Y.; et al. The Analysis of Plasma Proteomics for Luminal A Breast Cancer. Cancer Med. 2024, 13, e70470. [Google Scholar] [CrossRef]
  153. Zhong, J.M.; Li, J.; Kang, A.D.; Huang, S.Q.; Liu, W.B.; Zhang, Y.; Liu, Z.H.; Zeng, L. Protein S100-A8: A potential metastasis-associated protein for breast cancer determined via iTRAQ quantitative proteomic and clinicopathological analysis. Oncol. Lett. 2018, 15, 5285–5293. [Google Scholar] [CrossRef] [PubMed]
  154. Tomar, A.K.; Thapliyal, A.; Mathur, S.R.; Parshad, R.; Suhani Yadav, S. Exploring Molecular Alterations in Breast Cancer Among Indian Women Using Label-Free Quantitative Serum Proteomics. Biochem. Res. Int. 2024, 2024, 5584607. [Google Scholar] [CrossRef] [PubMed]
  155. Ku, W.C.; Liu, C.Y.; Huang, C.J.; Liao, C.C.; Huang, Y.C.; Kong, P.H.; Chen-Chan, H.; Tseng, L.M.; Huang, C.C. Integrating functional proteomics and next generation sequencing reveals potential therapeutic targets for Taiwanese breast cancer. Clin. Proteom. 2025, 22, 4. [Google Scholar] [CrossRef] [PubMed]
  156. Morrow, M. Sentinel-Lymph-Node Biopsy in Early-Stage Breast Cancer—Is It Obsolete? N. Engl. J. Med. 2024, in press. [Google Scholar] [CrossRef]
  157. Reimer, T.; Stachs, A.; Veselinovic, K.; Kühn, T.; Heil, J.; Polata, S.; Marmé, F.; Müller, T.; Hildebrandt, G.; Krug, D.; et al. Axillary Surgery in Breast Cancer—Primary Results of the INSEMA Trial. N. Engl. J. Med. 2024, in press. [Google Scholar] [CrossRef]
  158. Otte, M.; Zafrakas, M.; Riethdorf, L.; Pichlmeier, U.; Löning, T.; Jänicke, F.; Pantel, K. MAGE-A gene expression pattern in primary breast cancer. Cancer Res. 2001, 61, 6682–6687. [Google Scholar]
  159. Rutgers, E.; Piccart-Gebhart, M.J.; Bogaerts, J.; Delaloge, S.; Veer, L.V.; Rubio, I.T.; Viale, G.; Thompson, A.M.; Passalacqua, R.; Nitz, U.; et al. The EORTC 10041/BIG 03-04 MINDACT trial is feasible: Results of the pilot phase. Eur. J. Cancer 2011, 47, 2742–2749. [Google Scholar] [CrossRef]
  160. Sparano, J.A.; Gray, R.J.; Makower, D.F.; Pritchard, K.I.; Albain, K.S.; Hayes, D.F.; Geyer, C.E., Jr.; Dees, E.C.; Goetz, M.P.; Olson, J.A.; et al. Adjuvant Chemotherapy Guided by a 21-Gene Expression Assay in Breast Cancer. N. Engl. J. Med. 2018, 379, 111–121. [Google Scholar] [CrossRef]
  161. Lænkholm, A.V.; Jensen, M.B.; Eriksen, J.O.; Rasmussen, B.B.; Knoop, A.S.; Buckingham, W.; Ferree, S.; Schaper, C.; Nielsen, T.O.; Haffner, T.; et al. PAM50 Risk of Recurrence Score Predicts 10-Year Distant Recurrence in a Comprehensive Danish Cohort of Postmenopausal Women Allocated to 5 Years of Endocrine Therapy for Hormone Receptor-Positive Early Breast Cancer. J. Clin. Oncol. 2018, 36, 735–740. [Google Scholar] [CrossRef]
  162. Soliman, H.; Flake, D.D., 2nd; Magliocco, A.; Robson, M.; Schwartzberg, L.; Sharma, P.; Brown, K.; Wehnelt, S.; Kronenwett, R.; Gutin, A.; et al. Predicting Expected Absolute Chemotherapy Treatment Benefit in Women with Early-Stage Breast Cancer Using EndoPredict, an Integrated 12-Gene Clinicomolecular Assay. JCO Precis. Oncol. 2019, 3, PO.18.00361. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-analyses extension for Scoping Reviews (PRISMA–ScR) flowchart.
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-analyses extension for Scoping Reviews (PRISMA–ScR) flowchart.
Jpm 15 00177 g001
Table 1. Proteomic studies in tumor tissue specimens from breast cancer patients.
Table 1. Proteomic studies in tumor tissue specimens from breast cancer patients.
StudyCountrySettingBC StageNumber of BC SpecimensPrimary
Proteomic
Method
Additional Laboratory MethodsMain Findings
Abdullah et al., 2016 [16]Saudi ArabiaDiagnosisPrimary20MALDI–TOFn.a.Biomarkers for diagnosis and pathogenesis
Akcakanat et al., 2021 [17]USATherapyMetastatic37RPPADNA and RNA sequencingBiomarkers differentiating primary from metastatic BC
Akpinar et al., 2017 [18]TurkeyDiagnosisPrimary102D–PAGEn.a.Biomarkers differentiating primary from metastatic BC
Al-Wajeeh et al., 2020 [20]MalaysiaDiagnosisPrimary80SDS–PAGE andLC–MS/MSn.a.Biomarkers for staging
Asleh et al., 2022 [22]CanadaDiagnosisPrimary300LC–MS/MS-based proteomicsIHCBiomarkers for diagnosis and prognosis
Azevedo et al., 2023 [23]BrazilDiagnosisPrimary19High-throughput MSIHCBiomarkers for BC subtypes
Azevedo et al., 2022 [24]BrazilDiagnosisPrimary19LC–MS/MSIn silico transcriptomic analysisBiomarkers for diagnosis and prognosis
Bateman et al., 2010 [25]USADiagnosisP, R, DCIS25LC–MS/MSIHCBiomarkers for disease progression and recurrence
Bernhardt et al., 2017 [29] GermanyDiagnosisPrimary801RPPAIHCBiomarkers for prognosis
Bjørnstad et al., 2024 [30]NorwayDiagnosisPrimary107MSMs in vitroBiomarkers for diagnosis and pathogenesis
Bonneterre et al., 2013 [32]FranceTherapyPrimary149SELDI–TOF MSn.a.Biomarkers for response to therapy
Bouchal et al., 2015 [33]Czech RepublicDiagnosisPrimary160iTRAQ-based proteomicsIHC, transcriptomicsBiomarkers for staging and nodal status
Braakman et al., 2015 [34]NedetherlandsDiagnosisPrimary11nano-LC–MS/MSDNA analysisBiomarkers for BC subtypes
Cabezón et al., 2012 [36]DenmarkTherapyPrimary782D-PAGE and MS2D Western Immunoblotting, IHCBiomarkers for therapy
Cancemi et al., 2012 [37]ItalyDiagnosisPrimary100MALDI–TOF MSIHC, Western blotBiomarkers for disease progression
Cancemi et al., 2010 [38]ItalyDiagnosisPrimary100MALDI–TOF MSWestern blot, n terminal microsequencingBiomarkers for prognosis
Cawthorn et al., 2012 [39]CanadaDiagnosisPrimary990iTRAQ and LC–MS/MSIHC, SRM–MSBiomarkers for prognosis
Champattanachai et al., 2013 [40]ThailandDiagnosisPrimary26LC–MS/MSin vitro assaysBiomarkers for diagnosis and pathogenesis
Creighton et al., 2010 [42]SpainD and TPrimary429RPPAIn vitro assays and quantitative real-time PCRBiomarkers for BC subtypes
Debets et al., 2023 [44]NetherlandsTherapyPrimary45Phosphoproteomicsn.a.Biomarkers for response to therapy
Di Cara et al., 2019 [45]ItalyDiagnosisPrimary80MALDI–TOF MSn.a.Biomarkers for disease progression and prognosis
Duan et al., 2023 [47]ChinaTherapyPrimary139MSn.a.Biomarkers for response to therapy
Fonseca-Sánchez et al., 2012 [49]MexicoDiagnosisPrimary105LC/ESI-MS/MSIHC, Western blotBiomarkers for BC subtypes
Gámez-Pozo et al., 2017 [53]SpainDiagnosisPrimary106LC–MSMicroRNA expression analysisBiomarkers for BC subtypes
Gámez-Pozo et al., 2017 [54]SpainDiagnosisPrimary60LC–MSParallel reaction monitoringBiomarkers for prognosis
Gámez-Pozo et al., 2015 [55]SpainD and TPrimary96LC–MS/MSMicroRNA expression and in vitro assaysBiomarkers for BC subtypes
García-Adrián et al., 2021 [56]SpainTherapyPrimary125MSn.a.Biomarkers for therapy
Gonzalez-Angulo et al., 2013 [62]USATherapyPrimary175RPPAIHCBiomarkers for therapy
Gonzalez-Angulo et al., 2011 [63]USAD and TP, M, DCIS880RPPAIHCBiomarkers for prognosis and response to therapy
Gromova et al., 2021 [65]DenmarkTherapyPrimary44RPPAIHC and PCRBiomarkers for response to therapy
Guerin et al., 2018 [66]FranceTherapyPrimary46MS RPLCWestern blotBiomarkers for response to therapy
Gustafsson et al., 2024 [67]SwedenDiagnosisPrimary63LC–MS/MSn.a.Biomarkers for diagnosis and pathogenesis
He et al., 2011 [68]USAD and TPrimary39LC–MSIHCBiomarkers for BC subtypes and response to therapy
He et al., 2009 [69]USAD and TPrimary52SELDI–TOF MSIHCBiomarkers for response to therapy
Hulahan et al., 2024 [73]USADiagnosisP and DCIS22Multiplexed spatial proteomicsn.a.Biomarkers differentiating invasive from DCIS
Izani Othman et al., 2009 [74]MalaysiaDiagnosisPrimary20LC–MS/MSWestern blot, MSBiomarkers for diagnosis and pathogenesis
Jeon et al., 2024 [76]South KoreaTherapyPrimary56Mass spectrometryn.a.Biomarkers for BC subtypes
Johansson et al., 2015 [77]SwedenTherapyPrimary24nanoLC–MS/MSWestern blot, ELISABiomarkers for response to therapy
Kang et al., 2010 [79]South KoreaDiagnosisP and DCIS164MALDI–TOF MSIHC, Western blotBiomarkers for diagnosis and pathogenesis
Kim et al., 2009 [82]South KoreaDiagnosisPrimary17MALDI–TOF MSn.a.Biomarkers for disease progression
Ku et al., 2025 [155]TaiwanTherapyPrimary61nanoLC–MS/MSNGSBiomarkers for therapy
Lin et al., 2021 [87]ChinaD and TPrimary24iTRAQ LC–MS/MSWestern blotBiomarkers for BC subtypes
Magara et al., 2024 [90]JapanTherapyP and DCIS133Multilayered proteomicsMultilayered proteomics in BC cellsBiomarkers for therapy
Meric-Bernstam et al., 2014 [93]USADiagnosisprimary53RPPAIHCResponse to surgical treatment
Michaut et al., 2016 [94]NetherlandsDiagnosisPrimary55RPPAcDNA-microarrays, DNA-sequencing, Western blotBiomarkers for BC subtypes
Moriggi et al., 2018 [96]ItalyDiagnosisPrimary262-DE, MALDI–MSGene expression microarrays, immunoblotting.Biomarkers for BC subtypes
Nakagawa et al., 2006 [97]USADiagnosisPrimary65SELDI–TOF MSn.a.Biomarkers for nodal status
Neubauer et al., 2008 [98]GermanyDiagnosisPrimary16Quantitative multiplex proteomicsWestern blot, immunofluoresenceBiomarkers for BC subtypes
Neubauer et al., 2006 [99]GermanyDiagnosisPrimary24MALDI–TOF MSn.a.Biomarkers for response to therapy
Niméus et al., 2007 [100]SwedenD and TPrimary20MALDI–TOF-TOF MStranscriptomicsBiomarkers for response to therapy
Ou et al., 2008 [101]SingaporeDiagnosisPrimary63MALDI–TOF MScDNA microarrays, IHC, Western blotBiomarkers for diagnosis and pathogenesis
Panis et al., 2013 [102]BrazilDiagnosisP and M135label-free MSWestern blot, ELISA, IHCBiomarkers for staging
Pozniak et al., 2016 [107]IsraelDiagnosisPrimary41/25MSPulsed-SILAC Assay, IHCHigh similarity in protein expression
Procházková et al., 2017 [108]Czech RepublicDiagnosisPrimary96mTRAQ labeling (mTRAQ–SRM)Transcriptomics, IHCBiomarkers for disease progression
Pucci-Minafra et al., 2017 [109]ItalyDiagnosisPrimary132D gel electrophoresis and MSWestern blotBiomarkers for prognosis
Pucci-Minafra et al., 2007 [110]ItalyDiagnosisPrimary37MALDI–TOFWestern blotBiomarkers for diagnosis and pathogenesis
Roberts et al., 2004 [112]AustraliaDiagnosisPrimary272-DEWestern blotBiomarkers for diagnosis and pathogenesis
Rojas et al., 2019 [113]SpainDiagnosisP and M51PRM targeted proteomicsn.a.Biomarkers for prediction of distant recurrence
Ruckhäberle et al., 2010 [114]GermanyDiagnosisPrimary19Isobaric TMT label-based proteomicsWestern blot, IHC, RNA expression microarray, LC–MSBiomarkers for BC subtypes
Sanders et al., 2008 [116]USADiagnosisprimary122MALDI–TOF MSLC–MS/MS, IHC, HPLCBiomarkers for disease progression, diagnosis and subtypes
Shenoy et al., 2020 [120]IsraelTherapyPrimary113LC–MS/MS-based proteomic analysisIHC, in vitro assaysBiomarkers for response to therapy
Shi et al., 2024 [121]ChinaTherapyPrimary50MS-Based Label-Free Proteomicsin vitro assaysBiomarkers for therapy
Shin et al., 2020 [122]South KoreaDiagnosisP and M36Reversed-phase (RP)-nano LC–ESI–MS/MSin vitro assaysBiomarkers for prediction of distant recurrence
Sohn et al., 2013 [124]USATherapyPrimary54RPPAn.a.Biomarkers for response to therapy
Stemke-Hale et al., 2008 [126]USAD and TPrimary547RPPALC–MS/MS, IHC, Western blot, transcriptomicsBiomarkers for therapy
Tamesa et al., 2009 [128]JapanDiagnosisPrimary40/30LC–MS/MSWestern blot, transcriptomicsBiomarkers for diagnosis
Tyanova et al., 2016 [131]GermanyDiagnosisPrimary40MS analysisn.a.Biomarkers for BC subtypes
Valo et al., 2019 [132]FranceDiagnosisP and DCIS72SWATH–MSIHC, ELISABiomarkers for diagnosis
Yang et al., 2016 [138]ChinaTherapyPrimary36LC–MS/MSn.a.Biomarkers for response to therapy
Yang et al., 2015 [139]ChinaDiagnosisPrimary60LC–MS/MSn.a.Biomarkers for BC subtypes
Yang et al., 2014 [140]ChinaDiagnosisPrimary36LC–MS/MSin vitro assaysBiomarkers for response to therapy
Yang et al., 2012 [141]South KoreaTherapyP and M83LC–MS/MSIHC Western blotBiomarkers for response to therapy
Yanovich et al., 2018 [142]IsraelDiagnosisPrimary109LC–MSn.a.Biomarkers for BC subtypes
Zeng et al., 2017 [145]ChinaDiagnosisPrimary23/23Quantitative iTRAQIHCBiomarkers for prediction of distant recurrence
Zhang et al., 2008 [149]SingaporeDiagnosisprimary94MALDI–TOF–TOF MSIHC Western blotBiomarkers for prognosis
Zhang et al., 2005 [150]SingaporeDiagnosisPrimary25MALDI–TOFIHC western blot, transcriptomicsBiomarkers for BC subtypes
Zhong et al., 2018 [153]ChinaDiagnosisPrimary54/54iTRAQIHCBiomarkers for nodal status
BC = breast cancer; CISH = Chromogenic in situ hybridization; D = Diagnosis; DCIS = Ductal carcinoma in situ; HPLC = High-Performance Liquid Chromatography; IA/MPD = Immunoassay using multiphoton-detection; IHC = Immunohistochemistry; iTRAQ = Isobaric Tag for Relative and Absolute Quantification; LC/ESI-MS/MS = Liquid Chromatography, Electrospray Ionization, Mass spectrometry; LC–MS/MS = Liquid chromatography–tandem mass spectrometry; M = Metastasis; MALDI–TOF = Matrix-assisted laser desorption/ionization-time of flight; MS = Mass spectrometry; n.a. = not available; nano-LC–MS/MS = nanoscale liquid chromatography–tandem mass spectrometry; P = Primary; PRM = Parallel reaction monitoring; RPLC = Reversed-phase liquid chromatography; RPPA = Reverse Phase Protein Arrays; SDS-PAGE = Sodium dodecyl sulfate polyacrylamide gel electrophoresis; SELDI–TOF= Surface-Enhanced Laser Desorption/Ionization Time-of-Flight; SRM-MS = Selected Reaction Monitoring Mass Spectrometry; SWATH = Sequential Window Acquisition of all Theoretical Mass Spectra; T = Therapy; TMT = Tandem Mass Tag; 2D-PAGE = Two-dimensional polyacrylamide gel electrophoresis.
Table 2. Proteomic studies in plasma and serum from breast cancer patients.
Table 2. Proteomic studies in plasma and serum from breast cancer patients.
StudyCountrySettingBC StageType of SpecimenNumber of BC SpecimensPrimary
Proteomic
Method
Additional Laboratory MethodsMain Findings
Alvarez et al., 2022 [19]USATherapyPrimaryPlasma Evs17PPLC and LC–MS/MSWestern blotBiomarkers for response to therapy
An et al., 2022 [21]ChinaDiagnosisPrimaryPlasma107Nano-LC–MS/MSMetabolomic analysisBiomarkers for diagnosis
Belluco et al., 2007 [26]ItalyDiagnosisPrimarySerum155SELDI–TOF MSn.a.Biomarkers for early diagnosis
Bera et al., 2020 [27]USADiagnosisPrimarySerum240Antibody Microarrays and MSD Multi-arrayn.a.Biomarkers for recurrence prediction
Corrêa et al., 2017 [41]BrazilDiagnosisPrimaryPlasma107Nano-LC–MS/MSIHC, FISH, Western blotBiomarkers for BC subtypes
Dalenc et al., 2010 [43]FranceTherapyMetastaticSerum57SELDI–TOF MSLC–MS/MSBiomarkers for response to therapy
Drukier et al., 2006 [46]USADiagnosisPrimarySerum264IA/MPDELISA, LuminexBiomarkers for early diagnosis
Fernandez-Pol et al., 2005 [48]USAD and TP and MSerum243HPLCMS, Western blot, RadioimmunoassayBiomarkers for diagnosis and response to therapy
Fredolini et al., 2020 [50]USADiagnosisPrimarySerum20Affinity hydrogel nanoparticles coupled with LC–MS/MSn.a.Biomarkers for early diagnosis
Gajbhiye et al., 2017 [51]IndiaDiagnosisPrimarySerum762D-DIGE, iTRAQ and SWATH–MSn.a.Biomarkers for BC subtypes
Garisi, Tommasi et al., 2012 [57]ItalyDiagnosisPrimarySerum192SELDI–TOF MSn.a.Biomarkers for BC subtypes
Garisi, Tufaro et al., 2012 [58]ItalyDiagnosisPrimarySerum138SELDI–TOF MSn.a.Biomarkers for diagnosis
Gast et al., 2011 [59]NetherlandsDiagnosisprimarySerum82SELDI–TOF MSMALDI–TOFBiomarkers for recurrence prediction
Goncalves et al., 2006 [61]FranceDiagnosisprimarySerum81SELDI–TOF MSImmunodepletionBiomarkers for recurrence prediction
Grassmann et al., 2024 [64]SwedenDiagnosisprimaryPlasma796Proximity Extension Assayn.a.No benefit for recurrence prediction
Henderson et al., 2019 [70]USADiagnosisP and DCISSerum123Modified ECL based ELISAn.a.Biomarkers for early diagnosis
Henderson et al., 2016 [71]USADiagnosisP and DCISSerum100modified ELISAn.a.Biomarkers for early diagnosis
Hu et al., 2005 [72]ChinaDiagnosisP and MSerum49SELDI–TOF MSn.a.Biomarkers for early diagnosis
Jordan et al., 2020 [77]USAD and TprimaryPlasma Evs20MSWestern blot, in vitro assays, Multiplex gene expression analysisBiomarkers for diagnosis and therapy
Kaur et al., 2024 [79]USADiagnosisP and MSerum73MSn.a.Biomarkers for disease progression
Kim et al., 2019 [81]South KoreaDiagnosisprimary Plasma575MRM MSn.a.Biomarkers for early diagnosis
Le Naour et al., 2001 [83]USADiagnosisprimary Serum30MALDI–TOFWestern blot, IHCBiomarkers for early diagnosis
Li et al., 2024 [85]ChinaTherapyprimary Plasma40MSProtein-protein interaction (PPI) analysis and Single-cell RNA sequencingBiomarkers for response to therapy
Lötsch et al., 2022 [87]GermanyDiagnosisprimary Serum27PEAn.a.Biomarkers for neuropathic pain
Lourenco et al., 2017 [88]USADiagnosisprimary Serum26modified ELISAn.a.Biomarkers for early diagnosis
Majidzadeh-A et al., 2013 [90] IranTherapyprimary Serum10MALDI–TOFn.a.Biomarkers for response to therapy
Minton et al., 2013 [94]UKDiagnosisprimary Serum45SELDI–TOF MSLC–MSBiomarkers for cancer Related Fatigue Syndrome
Pires et al., 2019 [106]BrazilTherapyprimary Plasma200MSOxidative Stress AnalysesBiomarkers for response to therapy
Riley et al., 2011 [111]USA DiagnosisP and DCISSerum216LC–MS/MSn.a.Biomarkers for diagnosis
Rui et al., 2003 [113]ChinaDiagnosisprimary Serum145MALDI–TOFN-terminal sequencingBiomarkers for diagnosis
Santana et al., 2024 [117]BrazilDiagnosisprimary Plasma143LC–MS/MSn.a.Biomarkers for BC subtypes
Schaub et al., 2009 [119]USADiagnosisP, M and DCISSerum125MALDI–TOF MSn.a.Biomarkers for staging and nodal status
Sinha et al., 2023 [123]USADiagnosisprimary Saliva and Serum15iTRAQ analysisn.a.Biomarkers for early diagnosis
Starodubtseva et al., 2023 [125]RussiaDiagnosismetastaticSerum25LC–MRM MSLipidomicsBiomarkers differentiating primary from metastatic BC
Suman et al., 2016 [127]IndiaDiagnosisprimary Plasma32iTRAQ analysisWestern blot, ELISA and IHCBiomarkers for BC subtypes
Tomar et al. [154]IndiaDiagnosisP and MSerum12MSn.aBiomarkers for diagnosis
Tutanova et al., 2020 [130]RussiaDiagnosisprimary Plasma, WBE23MSFlow CytometryBiomarkers for diagnosis
Vinik et al., 2020 [133]USADiagnosisprimary Plasma Evs52RPPAImmunoblottingBiomarkers for early diagnosis
Xu et al., 2015 [135]ChinaDiagnosisprimary Serum60LC–MS/MSELISABiomarkers for response to therapy
Xu et al., 2024 [134]ChinaDiagnosisprimary Serum Evs126MSIHC and in vitro cell assays Biomarkers for early diagnosis and staging
Yan et al., 2022 [136]ChinaDiagnosisprimary Serum64MB–IMAC-Cu and MALDI–TOF MSLC-ESI-MS/MS, ELISABiomarkers for diagnosis
Yang et al., 2020 [137]ChinaTherapyprimary Serum51Isobaric TMT label-based quantitative proteomicsLC–MS/MSBiomarkers for response to therapy
Ye et al., 2024 [143]ChinaDiagnosisP and MPlasma51UPLC–MS/MSELISA, metabolomicsBiomarkers differentiating different sites of metastasis
Zeidan et al., 2018 [144]UKDiagnosisprimary Serum399LC–MS/MSELISABiomarkers for staging and nodal status
Zhang et al., 2013 [148]USADiagnosisP and DCISSerum100LC–MS/MSn.a.Biomarkers for early diagnosis
Zhang et al., 2015 [149]USADiagnosisprimary Serum80LC–MS/MSn.a.Biomarkers for early diagnosis
Zhang et al., 2013 [150]USADiagnosisprimary Plasma80LC–ESI–MS/MSn.a.Biomarkers for early diagnosis
Zhao et al., 2024 [152]ChinaDiagnosisprimary Plasma10LC–MS/MSn.a.Biomarkers for BC subtypes
BC = breast cancer; ECL = electro-chemiluminescent; EVs = extracellular vesicles; iTRAQ = Isobaric Tag for Relative and Absolute Quantification; LC = Liquid chromatography; LC–ESI–MS/MS = Liquid Chromatography-Electrospray Ionization-Tandem Mass Spectrometry; LC–MS/MS = Liquid chromatography–tandem mass spectrometry; MALDI–TOF = Matrix-assisted laser desorption/ionization-time of flight; MB–IMAC = Magnetic beads based immobilized metal ion affinity chromatography; MRM = Multiple reaction monitoring; MS = Mass spectrometry; PEA = proximity extension assay; PPLC = Particle purification liquid chromatography; RPPA = Reverse Phase Protein Arrays; SELDI–TOF= Surface-Enhanced Laser Desorption/Ionization Time-of-Flight; SWATH = Sequential Window Acquisition of all Theoretical Mass Spectra; TMT = Tandem Mass Tag; 2D-DIGE = two-dimensional difference gel electrophoresis; UPLC = Ultra-performance liquid chromatography; WBE = whole blood exosomes.
Table 3. Proteomic studies in nipple aspiration fluid (NAF), urine, saliva, tear fluid, pleural effusions, tumor interstitial fluid and lymph nodes from breast cancer patients.
Table 3. Proteomic studies in nipple aspiration fluid (NAF), urine, saliva, tear fluid, pleural effusions, tumor interstitial fluid and lymph nodes from breast cancer patients.
StudyCountrySettingBC StageType of SpecimenNumber
of BC Specimens
Primary Proteomic
Method
Additional Laboratory MethodsMain Findings
Brunoro et al., 2019 [35]BrazilDiagnosisprimaryNAF10MSn.a.Biomarkers differentiating BC from healthy
Kuerer et al., 2004 [82]USADiagnosisprimaryNAF23SELDI–TOF MSn.a.Biomarkers for nodal status
Paweletz et al., 2001 [104]USADiagnosisprimaryNAF12SELDI–TOF MSn.a.Biomarkers differentiating BC from healthy
Pawlik et al., 2006 [105]USADiagnosisprimaryNAF18LC–MS/MSWestern blotBiomarkers differentiating BC from healthy
Sauter et al., 2002 [118]USADiagnosisprimaryNAF20SELDI–TOF MSn.a.Biomarkers differentiating BC from healthy
Beretov et al., 2015 [28]AustraliaDiagnosisP and DCISUrine20LC–MS/MSWestern blot, IHCBiomarkers differentiating BC from healthy
Gajbhiye et al., 2016 [52]IndiaDiagnosisprimaryUrine432D-DIGE, iTRAQ, SWATH MSWestern blotBiomarkers differentiating BC from healthy
Jeanmard et al., 2023 [74]ThailandDiagnosisprimaryUrinary EVs47LC–MS/MSWestern blotBiomarkers differentiating BC from healthy
Giri et al., 2022 [60]IndiaDiagnosismetastaticSaliva20PRM–MSWestern blotBiomarkers differentiating BC from healthy
Zhang et al., 2010 [151]USADiagnosisprimarysaliva40MALDI–TOFWestern blotBiomarkers differentiating BC from healthy
Sinha et al., 2023 [123]USADiagnosisprimarySaliva, Serum15iTRAQ proteomic analysisn.a.Biomarkers differentiating BC from healthy
Böhm et al., 2012 [31]GermanyDiagnosisprimaryTear fluid25MALDI–TOFn.a.Biomarkers differentiating BC from healthy
Lebrecht et al., 2009 [84]GermanyDiagnosisprimaryTear fluid50SELDI–TOF MSn.a.Biomarkers differentiating BC from healthy
Mayayo-Peralta et al., 2024 [91]NetherlandsTherapymetastaticPleural effusions47Phosphoproteomics analysisn.a.Biomarkers for response to therapy
Terkelsen et al., 2020 [129]DenmarkDiagnosisprimaryTIF35LC–MS/MSIHCBiomarkers for BC subtypes
Pathania et al., 2022 [103]IndiaDiagnosisprimarySLN13iTRAQ proteomic analysis and MSELISABiomarkers for nodal status
Pozniak et al., 2016 [107]IsraelDiagnosisprimaryBC, LN41+25MSPulsed-SILAC, IHCNo difference in protein expression
Zeng et al., 2017 [145]ChinaDiagnosisprimaryBC, LN23+23iTRAQ proteomic analysisIHCBiomarkers for nodal status
Zhong et al., 2018 [153]ChinaDiagnosisprimaryBC, LN54+54iTRAQ proteomic analysisIHCBiomarkers for nodal status
BC = breast cancer; EVs = extracellular vesicles; TIF = Tumor interstitial fluid; IHC = Immunohistochemistry; iTRAQ = Isobaric Tag for Relative and Absolute Quantification; LC–MS/MS = Liquid chromatography–tandem mass spectrometry; LN = Lymph nodes; MALDI–TOF = Matrix-assisted laser desorption/ionization-time of flight; MS = Mass spectrometry; NAF = Nipple aspiration fluid; PRM = Parallel reaction monitoring; SELDI–TOF = Surface-Enhanced Laser Desorption/Ionization Time-of-Flight; SLN = Sentinel lymph nodes; SWATH = Sequential Window Acquisition of all Theoretical Mass Spectra; 2D-DIGE = two-dimensional difference gel electrophoresis.
Table 4. Proteomic studies that identified biomarkers for prognosis of breast cancer patients.
Table 4. Proteomic studies that identified biomarkers for prognosis of breast cancer patients.
StudyBC StageType of SpecimenNumber of BC SpecimensPrimary
Proteomic
Method
Additional Laboratory MethodsMain Findings
Cawthorn et al., 2012 [39]primaryTumor990iTRAQ and LC–MS/MSIHC, SRM–MSHigh expression levels of Decorin and Endoplasmin (HSP90B1) are associated with increased metastasis-poorer survival and may guide the use of hormonal therapy.
Gonzalez-Angulo et al., 2011 [63]P, M, DCISTumor880RPPAIHCA 10-protein biomarker panel was developed that classifies breast cancer into prognostic groups that may have potential utility in the management of patients who receive anthracycline-taxane-based neoadjuvant systemic therapy.
Bernhardt et al., 2017 [29] primaryTumor801RPPAIHCSHMT2 and ASCT2 protein expression were identified as novel potential prognostic biomarkers for BC, as their high protein expression is associated with poor outcome.
Asleh et al., 2022 [22]primaryTumor300LC–MS/MS-based proteomicsIHCPotential diagnostic and prognostic biomarkers were identified.
Cancemi et al., 2012 [37]primaryTumor100MALDI–TOF MSIHC, Western blotDeregulation of proteins of S100 family is associated with breast cancer progression and may serve as potential prognostic biomarkers for patient stratification.
Cancemi et al., 2010 [38]primaryTumor100MALDI–TOF MSWestern blot, n terminal microsequencingS100A7 protein, with its two isoforms, serve a potential role in the progression and biological mechanisms of infiltrating ductal carcinoma.
Procházková et al., 2017 [108]primaryTumor96mTRAQ labeling (mTRAQ-SRM)Transcriptomics, IHCA panel of gene products that can contribute to breast cancer aggressiveness and metastasis was identified.
Zhang et al., 2008 [149]primaryTumor94MALDI–TOF-TOF MSIHC Western blotCK19 in HER-2+ breast cancer is associated with tumor aggressiveness, suggesting CK19’s potential role as a biomarker for identifying more aggressive BC subtypes.
Di Cara et al., 2019 [45]primaryTumor80MALDI–TOF MSn.a.MMP-2 and MMP-9 can be involved in the complicated scenario in which the mechanisms of tumor progression are correlated with unfavorable prognosis.
Kaur et al., 2024 [79]P and MSerum73MSn.a.A set of proteins that could be involved in breast cancer progression in serum was identified.
Gámez-Pozo et al., 2017 [54]primaryTumor60LC–MSParallel reaction monitoringSome ER+/PR+ samples had a protein expression profile similar to that of triple negative breast cancer (TNBC) and had a clinical outcome similar to those with TNBC.
BC = breast cancer; DCIS = Ductal carcinoma in situ; IHC = Immunohistochemistry; iTRAQ = Isobaric Tag for Relative and Absolute Quantification; LC–MS/MS = Liquid chromatography–tandem mass spectrometry; M = Metastasis; MALDI–TOF = Matrix-assisted laser desorption/ionization-time of flight; MS = Mass spectrometry; n.a. = not available; P = Primary; RPPA = Reverse Phase Protein Arrays; SRM–MS = Selected Reaction Monitoring Mass Spectrometry.
Table 5. Proteomic studies that identified biomarkers for early diagnosis of breast cancer.
Table 5. Proteomic studies that identified biomarkers for early diagnosis of breast cancer.
StudyBC StageType of SpecimenNumber of BC SpecimensPrimary
Proteomic
Method
Additional MethodsMain Findings
Grassmann et al., 2024 [64]primaryPlasma796Proximity Extension Assayn.a.No benefit for recurrence prediction
Kim et al., 2019 [81]primary Plasma575MRM MSn.a.Three specific peptides can be a useful tool for breast cancer screening and its accuracy is cancer-type specific.
Drukier et al., 2006 [46]primary Tumor264IA/MPDELISA, LuminexUltrasensitive, multi-biomarker immunoassays significantly improve early breast cancer detection accuracy.
Belluco et al., 2007 [26]primarySerum155SELDI–TOF MSn.a.A proteomic pattern consisting of 7 low-molecular-weight ion peaks is a highly sensitive and specific method for early detection of stage 1 breast cancer.
Xu et al., 2024 [134]primary Serum Evs126MSIHC and in vitro cell assays Proteins carried by breast cancer–derived EVs could be used as minimally invasive liquid biopsy tool for the early detection of breast cancer and for discriminating lymph node involvement and distant metastasis.
Henderson et al., 2019 [70]P and DCISSerum123Modified ECL based ELISAn.a.Serum biomarkers provide clinicians with additional information for patients with indeterminate breast imaging results, potentially reducing false-positive breast biopsies.
Zhang et al., 2013 [148]P and DCISSerum100LC–MS/MSn.a.Feed Forward Neural Network (FFNN) enhances the development of more accurate and reliable biomarker panels for the early diagnosis of breast cancer.
Henderson et al., 2016 [71]P and DCISSerum100modified ELISAn.a.SPB and TAAb combinatorial protein biomarker assays may aid in the detection of early BC and guide decisions between imaging and tissue biopsy.
Zhang et al., 2015 [149]primary Serum80LC–MS/MSn.a.Pathway-based biomarkers can significantly enhance the early detection and diagnostic accuracy of breast cancer.
Zhang et al., 2013 [150]primary Plasma80LC-ESI-MS/MSn.a.Identification of eight alternative splicing isoform biomarkers can assist the early diagnosis of breast cancer.
Vinik et al., 2020 [133]primary Plasma Evs52RPPAImmunoblottingSeveral potential markers that could contribute to early detection of BC were identified.
Hu et al., 2005 [72]P and MSerum49SELDI–TOF MSn.a.SELDI–TOF-MS combined with bioinformatics tools is a promising approach for the early detection of breast cancer, identifying four candidate biomarkers.
Le Naour et al., 2001 [83]primary Serum30MALDI–TOFWestern blot, IHCRS/DJ-1 is a novel circulating tumor antigen eliciting a humoral immune response in breast cancer patients and can be used for early detection and monitoring of BCr.
BC = breast cancer; DCIS = Ductal carcinoma in situ; EVs = extracellular vesicles; IA/MPD = Immunoassay using multiphoton-detection; IHC = Immunohistochemistry; iTRAQ = Isobaric Tag for Relative and Absolute Quantification; LC/ESI-MS/MS = Liquid Chromatography, Electrospray Ionization, Mass spectrometry; LC–MS/MS = Liquid chromatography–tandem mass spectrometry; M = Metastasis; MALDI–TOF = Matrix-assisted laser desorption/ionization-time of flight; MS = Mass spectrometry; n.a. = not available; P = Primary; RPPA = Reverse Phase Protein Arrays; SELDI–TOF = Surface-Enhanced Laser Desorption/Ionization Time-of-Flight.
Table 6. Proteomic studies that identified biomarkers for characterization of breast cancer subtypes.
Table 6. Proteomic studies that identified biomarkers for characterization of breast cancer subtypes.
StudyBC StageType of SpecimenNumber of BC SpecimensPrimary
Proteomic
Method
Additional Laboratory MethodsMain Findings
Creighton et al., 2010 [42]primaryTumor429RPPAIn vitro assays and quantitative real-time PCRHyperactive PI3K signaling is associated with low estrogen receptor (ER) levels and luminal B molecular subtype in ER-positive BC.
Garisi et al., 2012 [57]primarySerum192SELDI–TOF MSn.a.The serum profile of familial breast cancer patients was different when compared with that of sporadic breast cancer patients.
Santana et al., 2024 [117]primary Plasma143LC–MS/MSn.a.The HDL proteome showed discriminatory abilities across different clinical stages of breast cancer and a distinct profile in triple negative breast cancer.
Sanders et al., 2008 [116]primaryTumor122MALDI–TOF MSLC–MS/MS, IHC, HPLCS100A6 (calcyclin) and S100A8 (calgranulin A) highlight potential roles in cancer progression, diagnosis, and molecular classification.
Yanovich et al., 2018 [142]primaryTumor109LC–MSn.a.A novel luminal subtype characterized by increased PI3K signaling has been identified.
Corrêa et al., 2017 [41]primaryPlasma107Nano-LC–MS/MSIHC, FISH, Western blotThe plasma proteomic profile of breast cancer subtypes was determined.
Gámez-Pozo et al., 2017 [53]primaryTumor106LC–MSMicroRNA expression analysisER+ BC and triple negative breast cancer exhibit distinct molecular and metabolic profiles.
Fonseca-Sánchez et al., 2012 [49]primaryTumor105LC/ESI-MS/MSIHC, Western blotGlyoxalase 1 (GLO1) is overexpressed in breast cancer and correlates significantly with high tumor grade.
Gámez-Pozo et al., 2015 [55]primaryTumor96LC–MS/MSMicroRNA expression and in vitro assaysSome ER+/PR+ samples had a protein expression profile similar to that of triple negative breast cancer (TNBC) and had a clinical outcome similar to those with TNBC.
Gajbhiye et al., 2017 [51]primarySerum762D-DIGE, iTRAQ and SWATH-MSn.a.Serum proteome alterations may help to distinguish breast cancer subtypes (luminal A and, B, HER2-positive and triple negative BCr).
Jeon et al., 2024 [76]primaryTumor56Mass spectrometryn.a.Coronin-1A and titin were upregulated in the immune-inflamed subtype, and α-1-antitrypsin was upregulated in the immune-excluded/desert subtype.
Michaut et al., 2016 [94]primaryTumor55RPPAcDNA-microarrays, DNA-sequencing, Western blotTwo biologically distinct subtypes of invasive lobular breast cancer were identified.
He et al., 2009 [69]primaryTumor52SELDI–TOF MSIHCProtein biosignatures were identified as having potential utility in tumor classification and predicting therapeutic responses.
Tyanova et al., 2016 [131]primaryTumor40MS analysisn.a.Global profiling of breast cancer clinical samples allows the attribution of biological processes to the different breast cancer subtypes (ER/PR, HER2 positive and TNBC).
Yang et al., 2014 [140]primaryTumor36LC–MS/MSin vitro assaysThe level of FR isoforms was associated with several histopathological features and molecular subtypes.
Terkelsen et al., 2020 [129]primaryTIF35LC–MS/MSIHCTen proteins, AGR3, BCAM, CELSR1, MIEN1, NAT1, PIP4K2B, SEC23B, THTPA, TMEM51, and ULBP2 stratify the tumor subtype-specific TIFs.
Suman et al., 2016 [127]primary Plasma32iTRAQ analysisWestern blot, ELISA and IHCFour proteins (FN1, A2M, C4BPA and CFB) had strong association with molecular subtypes of breast cancer.
BC = breast cancer; DCIS = Ductal carcinoma in situ; HPLC = High-Performance Liquid Chromatography; IHC = Immunohistochemistry; iTRAQ = Isobaric Tag for Relative and Absolute Quantification; LC/ESI-MS/MS = Liquid Chromatography, Electrospray Ionization, Mass spectrometry; LC–MS/MS = Liquid chromatography–tandem mass spectrometry; M = Metastasis; MALDI–TOF = Matrix-assisted laser desorption/ionization-time of flight; MS = Mass spectrometry; n.a. = not available; nano-LC–MS/MS = nanoscale liquid chromatography–tandem mass spectrometry; P = Primary; RPPA = Reverse Phase Protein Arrays; SELDI–TOF = Surface-Enhanced Laser Desorption/Ionization Time-of-Flight; SWATH = Sequential Window Acquisition of all Theoretical Mass Spectra; TIF = Tissue Interstitial Fluid TMT = Tandem Mass Tag.
Table 7. Proteomic studies that identified biomarkers for response to treatment in breast cancer patients.
Table 7. Proteomic studies that identified biomarkers for response to treatment in breast cancer patients.
StudyBC StageType of SpecimenNumber of BC SpecimensPrimary
Proteomic
Method
Additional Laboratory MethodsMain Findings
Gonzalez-Angulo et al., 2011 [63]P, M, DCISTumor880RPPAIHCA 10-protein biomarker panel was developed that classifies breast cancer into prognostic groups that may have potential utility in the management of patients who receive anthracycline-taxane-based neoadjuvant systemic therapy.
Fernandez-Pol et al., 2005 [48]P and MSerum243HPLCMS, Western blot, RadioimmunoassayMPS-1 is useful for early detection, monitoring, and management of breast cancer, superior than CA-15-3 and CEA.
Pires et al., 2019 [106]primary Plasma200MSOxidative Stress AnalysesThe connection between inflammation, the complement and oxidative stress seems to be a pivotal axis in chemoresistance of luminal A breast cancer.
Bonneterre et al., 2013 [32]primaryTumor149SELDI–TOF MSn.a.A combined signature using the cytosol and plasma proteomic data may identify breast cancer patients like to achieve complete response in neoadjuvant chemotherapy.
Duan et al., 2023 [47]primaryTumor139MSn.a.115 proteins were differentially expressed between patients with pathologic Complete Response (pCR) and the non-pCR group.
Shenoy et al., 2020 [120]primaryTumor113LC–MS/MS-based proteomic analysisIHC, in vitro assaysTwo proteins of proline biosynthesis pathway, PYCR1 and ALDH18A1, were significantly associated with resistance to treatment.
Yang et al., 2012 [141]P and MTumor83LC–MS/MSIHC Western blotERK/Bcl-2-mediated anti-apoptosis was investigated in general and in the development of drug resistance.
Xu et al., 2015 [135]primary Serum60LC–MS/MSELISASISCAPA-targeted proteomics alllowed quatification of low-abundant serum transferrin receptor in breast cancer patients pre- and post-chemotherapy.
Yang et al., 2015 [139]primaryTumor60LC–MS/MSn.a.TfR levels in breast tissue can be measured precisely with LC–MS/MS and could possibly improve the diagnosis of breast cancer and assessment of drug resistance.
Dalenc et al., 2010 [43]metastaticSerum57SELDI–TOF MSLC–MS/MSFibrinogen α peptide could serve as a predictive biomarker for therapeutic response in ER+ breast cancer patients undergoing the tipifarnib and tamoxifen combination therapy.
Sohn et al., 2013 [124]primaryTumor54RPPAn.a.AKT, IGFBP2, LKB1, S6 and Stathmin predict relapse-free survival (RFS) in residual triple negative BC patients after neoadjuvant chemotherapy, while PI3K pathway may represent a potential therapeutic target.
Meric-Bernstam et al., 2014 [93]primaryTumor53RPPAIHCPI3K pathway activation is greater in core needle biopsy compared with postexcision surgical samples, suggesting a potential loss of phosphorylation during surgical manipulation, or with cold ischemia of surgical specimens.
He et al., 2009 [69]primaryTumor52SELDI–TOF MSIHCProtein biosignatures were identified as having potential utility in tumor classification and predicting therapeutic responses.
Yang et al., 2020 [137]primary Serum51Isobaric TMT quantitative proteomicsLC–MS/MSA serum-based protein signature that potentially predicts the therapeutic effects of trastuzumab-based therapy for HER2-positive breast cancer patients was developed.
Mayayo-Peralta et al., 2024 [91]metastaticPleural effusions47Phosphoproteomics analysisn.a.Evidence for decreased activity of several key kinases in ERα-converted metastases was found.
Guerin et al., 2018 [66]primaryTumor46MS RPLCWestern blotDifferential gene expression correlated with sensitivity to trastuzumab.
Debets et al., 2023 [44]primaryTumor45Phosphoproteomicsn.a.Treatment response to trastuzumab, pertuzumab may be predicted.
Gromova et al., 2021 [65]primaryTumor44RPPAIHC and PCRHigh-level c-Kit expression is a frequent event in triple negative BC (TNBC), and activating mutations were also present, suggesting a potential effect of c-Kit inhibitors on TNBCs.
Li et al., 2024 [85]primary Plasma40MSProtein-protein interaction (PPI) analysis and Single-cell RNA sequencingSpecific plasma proteins act as predictive biomarkers of response to immunotherapy.
He et al., 2011 [68]primaryTumor39LC–MSIHCProteomic profiling of breast cancer tissues can predict tumor response to neoadjuvant chemotherapy.
Yang et al., 2016 [138]primaryTumor36LC–MS/MSn.a.FKBP4 and S100A9 are promising predictive biomarkers for determining drug resistance in breast cancer patients undergoing neoadjuvant chemotherapy.
BC = breast cancer; DCIS = Ductal carcinoma in situ; HPLC = High-Performance Liquid Chromatography; IHC = Immunohistochemistry; LC–MS/MS = Liquid chromatography–tandem mass spectrometry; M = Metastasis; MALDI–TOF = Matrix-assisted laser desorption/ionization-time of flight; MS = Mass spectrometry; n.a. = not available; nano-LC–MS/MS = nanoscale liquid chromatography–tandem mass spectrometry; P = Primary; PRM = Parallel reaction monitoring; RPLC = Reversed-phase liquid chromatography; RPPA = Reverse Phase Protein Arrays; SELDI–TOF = Surface-Enhanced Laser Desorption/Ionization Time-of-Flight; TMT = Tandem Mass Tag.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zafrakas, M.; Gavalas, I.; Papasozomenou, P.; Emmanouilides, C.; Chatzidimitriou, M. Proteomics in Diagnostic Evaluation and Treatment of Breast Cancer: A Scoping Review. J. Pers. Med. 2025, 15, 177. https://doi.org/10.3390/jpm15050177

AMA Style

Zafrakas M, Gavalas I, Papasozomenou P, Emmanouilides C, Chatzidimitriou M. Proteomics in Diagnostic Evaluation and Treatment of Breast Cancer: A Scoping Review. Journal of Personalized Medicine. 2025; 15(5):177. https://doi.org/10.3390/jpm15050177

Chicago/Turabian Style

Zafrakas, Menelaos, Ioannis Gavalas, Panayiota Papasozomenou, Christos Emmanouilides, and Maria Chatzidimitriou. 2025. "Proteomics in Diagnostic Evaluation and Treatment of Breast Cancer: A Scoping Review" Journal of Personalized Medicine 15, no. 5: 177. https://doi.org/10.3390/jpm15050177

APA Style

Zafrakas, M., Gavalas, I., Papasozomenou, P., Emmanouilides, C., & Chatzidimitriou, M. (2025). Proteomics in Diagnostic Evaluation and Treatment of Breast Cancer: A Scoping Review. Journal of Personalized Medicine, 15(5), 177. https://doi.org/10.3390/jpm15050177

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop