You are on page 1of 38

www.clinical.proteomics-journal.

com Page 1 Proteomics - Clinical Applications

Proteomics in drug development: the dawn of a new era?

Maria Frantzi1*, Agnieszka Latosinska1* and Harald Mischak1,2

1
Mosaiques Diagnostics GmbH, Hannover, Germany

2
BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United

Kingdom

*equally contributing first authors

Corresponding Author

Prof. Harald Mischak

Mosaiques Diagnostics GmbH

Rotenburger Straße 20

D-30659 Hannover, Germany

Email: mischak@mosaiques-diagnostics.com

Phone: +49 (0)511 55 47 44 13

Fax: +49 (0)511 55 47 44 31

Keywords: Drug discovery, Mass Spectrometry, Proteomics

Total number of words: 7.896 (10.288 incl. references and figure legends)

List of Abbreviations (excl. standard abbreviations):

ABPP- activity-based protein profiling; ALK- anaplastic lymphoma kinase; ANGPTL2-

Angiopoietin-related protein 2; ANXA- annexin A BI-TK/GCV- Bifidobacterium infantis

thymidine kinase/nucleoside analogue ganciclovir; BTK- Bruton’s tyrosine kinase; CALU-

Calumenin; CETSA- Cellular thermal shift assay; CDH18- Cadherin-18; CDK- cyclin-

dependent kinases; CMTM6- CKLF-like MARVEL transmembrane domain containing protein

6; CNPY3- Protein canopy homolog 3; CPSN1- calpain small subunit 1; CRSwNP- Chronic

Received: 10 18, 2018; Revised: 01 13, 2019; Accepted: 02 04, 2019

This article has been accepted for publication and undergone full peer review but has not been
through the copyediting, typesetting, pagination and proofreading process, which may lead to
differences between this version and the Version of Record. Please cite this article as doi:
10.1002/prca.201800087.

This article is protected by copyright. All rights reserved.


www.clinical.proteomics-journal.com Page 2 Proteomics - Clinical Applications

rhinosinusitis with nasal polyps; EGFR- Epidermal growth factor receptor; EIF3D- Eukaryotic

translation initiation factor 3 subunit D; EIF5A- Eukaryotic translation initiation factor 5A;

EPHA2- Ephrin type-A receptor 2; ESCC- Esophageal squamous cell carcinoma; FGB-

fibrinogen B; FN1- Fibronectin 1; HSP- Heat shock protein; ICAM1- Adhesion Molecule 1;

IL1RAP- Interleukin 1 Receptor Accessory Protein; IL13RA2- Interleukin-13 receptor subunit

alpha-2; IL18- Interleukin 18; ITD- Internal tandem duplication; ITGB8- Integrin Subunit Beta

8; KRT17- Keratin 17; LGALS3BP- Galectin-3-binding protein; MCAM- Cell surface

glycoprotein MUC18; MCM2- minichromosome maintenance protein 2; MFAP5-

Microfibrillar-associated protein 5; MIBC- muscle invasive bladder cancer; NAP1L1-

Nucleosome assembly protein 1-like 1; NID-1- Nidogen-1; NKCC1- Sodium-potassium-

chloride cotransporter 1; NMIBC- non-muscle invasive bladder cancer; NSCLC- Non–small

cell lung cancer; PAL- photoaffinity labeling; PD-1- Programmed cell death protein 1;

PDLIM2- PDZ and LIM domain protein 2; PGRMC1- Progesterone receptor membrane

component 1; PHAP1- Putative HLA-DR-associated protein 1; PHB- prohibitin; PKM2 -

Pyruvate kinase M2; Plk1- Polo-like kinase 1; POSTN- Periostin; Prx- Peroxiredoxin;

PSMA3- proteasome subunit a3; PTPRZ1- Receptor-type tyrosine-protein phosphatase

zeta; TKI- tyrosine kinase inhibitors; RanBP1- GTPase RAN binding protein 1; RIOK-

Serine/threonine-protein kinase RIO1; S100A2- S100 calcium binding protein A2; SAFB-

Scaffold attachment factor B; SAP- Signalling lymphocytic activation molecule associated

protein; STMN1-Stathmin; TAGLN2- Transgelin-2; TGF‐β1- Transforming growth factor beta

1; TGF‐β3- Transforming growth factor beta 1; UBA5- Ubiquitin-like modifier activating

enzyme 5; UQCRC2- Cytochrome b-c1 complex subunit 2, mitochondrial

Abstract

Mass spectrometry offers the potential of acquiring high resolution data depicting the

functional status of a group of healthy or disease individuals, according to the different

conditions. As most of the drugs are currently targeting proteins, proteomics has a dual

value, both in the discovery of new molecules as therapeutic targets, but also as a

This article is protected by copyright. All rights reserved.


2
www.clinical.proteomics-journal.com Page 3 Proteomics - Clinical Applications

methodology to perform high throughput drug profiling. As there is an evident need for drugs

to be improved in terms of efficacy, a mechanistic insight for downstream effectors can be

valuable in order to predict side effects and resistance mechanisms. Recently developed

assays, like thermal proteome profiling enables comprehensive drug target profiling and is,

therefore, of high value in drug discovery. In this review, we have conducted a systematic

literature search and present the most prominent proteomics studies as implicated in

assisting drug discovery and development. Focus is placed on investigations that are closer

to implementation, therefore particular emphasis is given in studies conducted in human

diseased population and further verified in vitro or in vivo.

A. Concept of improving drug discovery and development by proteomics

Proteomic methodologies have proven to be valuable tools to provide novel insights into

disease processes. Proteins are the principal targets of drug discovery[1] and as such,

proteomics is a key technology to support development of specific drugs to stop or reverse

effects of the disease.[2]

In principle, proteomic analysis by mass spectrometry is based on the identification of

ionised compounds in a sample mixture by recording their mass-to-charge ratio.[3] The

relative abundance of the ionised compounds is measured and information on the amino

acid composition of the compounds is acquired, mainly via tandem MS or peptide mass

fingerprinting. Hence, depending on the focus, proteomics analysis can provide insights not

only for the peptide sequence, but also quantification of protein abundance, post

translational modifications (PTMs) and protein interactions. Compared to genome, which is

constant and depicts more the potential of translation, proteome is highly dynamic,

fundamentally the result of the translation process and its regulation. As a result, proteomics

represents a functional snapshot of the cell, in a much more dynamic perspective. Yet,

considering the complexity of the proteome,[4] interpretation of proteomics data is certainly

This article is protected by copyright. All rights reserved.


3
www.clinical.proteomics-journal.com Page 4 Proteomics - Clinical Applications

more challenging (for example, the definition of ion signal threshold over instrument noise is

crucial).[5] Moreover, the analytical sensitivity is lower, mainly because of the inability to

amplify the signal as in the case of nucleic acid amplification.[6] Still, proteomics has a great

potential in drug discovery, as can reflect protein activity, structure and kinetics.

Drug development is a long and costly process, frequently hampered by the fact that the

developed drugs have low response rates due to resistance mechanisms and/or

downstream pathways that are subsequently altered. Mass spectrometry techniques have

been advanced by now and are combined with new assays (e.g. thermal shift assay for drug

screening)[7, 8]
that enable high-throughput profiling. Due to the high-throughput potential,

proteins can be measured simultaneously in complex samples, in an unbiased, untargeted

manner. The approach finds applicability in practically every step of the drug development,

covering from drug target discovery to the clinical trial phases, with common applications of

proteomics in the pharmaceutical industry, which can be categorised to: a) discovery of

potential drug targets, b) investigations into mechanisms of drug action and protein-protein

interactions, and c) biomarkers from readily accessible biological fluids (Figure 1).

The challenge in the drug discovery process is to identify the underlying causes of disease

and to reverse them to a normal condition. Therefore, gaining a holistic understanding of

disease’s underlying pathology at the molecular level is essential for moving towards

personalised medicine. While the pathophysiology and causes of many diseases, like cancer

and other chronic conditions vary greatly in their nature and origin, in some cases, the cause

is found at the protein level, involving protein function, protein regulation, or protein-protein

interactions. Discovery investigations involve the identification of proteins whose expression

levels or activities change in disease state. In parallel, profiling of disease phenotype and

correlation with the drug mechanism of action and/or potential pathways controlling

resistance, provides with a mechanistic link which can be further explored towards

identification of systemic biomarkers.[9] As a result of our increased understanding at the

molecular level proteomics biomarkers have been additionally introduced to stratify the

This article is protected by copyright. All rights reserved.


4
www.clinical.proteomics-journal.com Page 5 Proteomics - Clinical Applications

patients for receiving an intervention and predict response- a topic which is as widely

covered in recent reviews.[10, 11]

B. New technological advancements

Latest technological breakthroughs have revolutionized drug development, particularly

regarding screening of ligands and/or inhibitors. Thermal proteome profiling has been

recently introduced,[12-15] as an evolution to the traditional fluorometric/ calorimetric thermal

shift assay, that now, when combined with mass spectrometry, can provide insights on

drug’s mechanism of action through comprehensive screening in living cells.[16] The

conventional thermal shift assay is based on the investigation of protein stabilization when

interacting with other biomolecules like drugs, DNA and co-factors. Changes in thermal

denaturation temperature of a protein under different conditions is representative of protein-

ligand interactions, thus appropriate for high throughput screening of small molecules/

inhibitors to develop ligand libraries.[16] On this basis, thermal proteome profiling assays were

further introduced in seminal studies, reporting new technologies like cellular thermal shift

assay (CETSA), activity-based protein profiling (ABPP),[17] photoaffinity labeling (PAL),[18]

and are comparatively summarized in recent reviews.[16, 19] Both PAL and ABPP assays are

based on labeling of proteins- in PAL assay through photoaffinity labeling with the ligand to

be covalently linked through UV irradiation[18] and in ABPP methodology through biotinylated

probes and affinity enriched on streptavidin beads.[17] Both approaches are subsequently

linked to downstream MS analysis and can provide insights in the structural proteome (in

case of PAL assay)[18] and enzyme functionality (ABPP assay).[17] Probably, the most recent

greatest breakthrough technology, however, is cellular thermal shift assay (CETSA),[7] as

when coupled with MS, enables untargeted label-free proteomics profiling to monitor drug/

target interactions in living cells. After its first introduction in 2013,[7] several reports have

made use of the technology in cell lines to monitor kinases inhibitors,[13] transmembrane cell

This article is protected by copyright. All rights reserved.


5
www.clinical.proteomics-journal.com Page 6 Proteomics - Clinical Applications

surface proteins[12] and even on- and off-target effect of chemotherapeutic agents, like

Methotrexate.[20] The potential of such applications is of high impact in the years to come.

C. From proteomics to personalized medicine: Literature review

Because of the wide area of applications, in this article, we focus on the application of

proteomics, as directly applied for discovery of novel targets and the use of mass

spectrometry (MS) techniques in the profiling of drug effects or mechanisms of action and

downstream effectors. For this purpose, a literature search was conducted using the Web of

Science database (22/08/18), as follows: TOPIC: "drug discover*" OR "drug target" OR

"therapeutic target" AND TOPIC: proteom*. Only manuscripts defined as “ARTICLE” OR

“REVIEW” types published within the last 5 years (2014 -2018) were considered. A total of

1625 papers were retrieved (excluding replicates) and those were further screened

independently by two co-authors (Supplementary Table 1). The manuscripts were further

shortlisted by including only research articles employing mass spectrometry platforms. For

the discovery studies, we considered only those, where association of proteomics results

was further verified in vitro and/or in vivo. Due to the broad spectrum of the field, in this

article we focused on chronic diseases. Therefore, biomarker studies, any investigations in

infectious diseases as well as technical papers on method development were excluded.

Forty-nine studies on chronic diseases were selected and are described in this review,

separated in the four following categories, according to the context of research: 1) discovery

studies for identification of potential drug targets and downstream pathways, 2) profiling

studies for drug effects on proteome, 3) chemical proteomic studies for drug selectivity and

inhibitor binding and 4) structural proteomics studies.

D. Discovery of potential drug targets

i. Tissue proteomics for drug discovery in oncology

This article is protected by copyright. All rights reserved.


6
www.clinical.proteomics-journal.com Page 7 Proteomics - Clinical Applications

Cancer includes a group of malignancies that presents high heterogeneity.[21] Cancer at its

advanced stage is still lethal mainly because of the variability that is observed among the

patients, resulting in a very low success rate for the proposed drugs.[21] Nevertheless, one

advantage compared to other chronic diseases, particularly for drug research is the

availability of tissue specimens, which can be rich in information. In this context, mass

spectrometry has been employed for large scale profiling to improve on the discovery, but

also in characterization of drugs’ action.[22] Several cancer discovery studies involving high

resolution proteomics, in which patient-derived tumour specimens were investigated, were

retrieved and are described below.

As many therapeutic targets for high grade glioma include immunotoxins targeting surface

sialylated glycoproteins, Autelitano et al.[23] have attempted to profile sialo-glycoproteins

implicated in glioma, through metabolic labeling with Ac4ManNAz and biorthogonal chemical

reporting with biotin-linked phopshine to bind the labelled glycans.[23] The enrichment was

followed by label-free MS to identify potential drug targets in human tissues from four cancer

patients. Although the number of patients was low, the findings were compared with fetal

astrocytes and human neural progenitor cells and additional verification was performed by

immunohistochemical staining (IHC).[23] Fifty-two proteins were classified as glioblastoma-

specific, including among others Angiopoietin-related protein 2 (ANGPTL2), Intercellular

Adhesion Molecule 1 (ICAM1), Interleukin 1 Receptor Accessory Protein (IL1RAP), IL13RA2

(Interleukin-13 receptor subunit alpha-2), Integrin Subunit Beta 8 (ITGB8), Galectin-3-binding

protein (LGALS3BP), Receptor-type tyrosine-protein phosphatase zeta (PTPRZ1). The idea

of employing an enrichment protocol to target glycoproteins is of particular interest,

considering that alterations of glycosylation are linked to tumour development, progression

as well as formation of metastasis.[24] However, these results are still at a preliminary phase,

as did not yet reach pre-clinical testing. Similarly, Collet et al.[25] analysed 23 primary cell

lines from 16 glioma patients, for which treatment and follow-up information was available.

As primary glioma cultures present different phenotypes (either adherent or as

This article is protected by copyright. All rights reserved.


7
www.clinical.proteomics-journal.com Page 8 Proteomics - Clinical Applications

neurospheres), the aim of this study was to profile both and identify common glioma specific

proteins as potential therapeutic targets.[25] Proteomic analysis by nano-LC/Q-TOF MS, was

performed in 11 adherent and 12 neurosphere- cell lines. 342 proteins indicative of cell cycle

processes were specifically identified in neurosphere lines, while 112 proteins indicative of

cell adhesion were solely found in adherent cultures. This is an interesting observation

considering the different adherent properties of the two types of cell lines. Forty-nine proteins

were significantly correlated with patients’ survival, among those, Calumenin (CALU),

Eukaryotic translation initiation factor 5A (EIF5A) and Transgelin-2 (TAGLN2) with increased

expression, while Nucleosome assembly protein 1-like 1 (NAP1L1) with decreased.[25] As in

the previous evaluation, the above proteins need to be further tested in vivo. Similarly, in a

study for proteomics characterization of melanoma,[26] isobaric tags for relative and absolute

quantitation (iTRAQ) was applied in melanoma tissues and matched non-malignant tissue.

Thirty differentially expressed proteins with more than 15‐fold were identified, while 67 were

decreased with more than 0.25‐fold. Among those, Periostin (POSTN) was highly

upregulated and selected to be further evaluated in melanoma cell lines. Co-cultures of

melanoma with human dermal fibroblasts revealed that POSTN expression was induced in

normal cells through induced cytokines, particularly Transforming growth factor beta 1 (TGF‐

β1) and TGF‐β3.[26]

Along these lines, Latosinska et al.[27] performed a comparative proteomic analysis of muscle

invasive (MIBC, n=6) and non-muscle invasive bladder cancer (NMIBC, n=5) tissue samples

using LC-MS/MS. Interestingly, the proteomic profiles were integrated in a pathway and

interactome analysis, which revealed involvement and functional significance of eukaryotic

translation initiation factor 3 subunit D (EIF3D) in MIBC. The validity of the EIF3D as a

potential target was further investigated by silencing of EIF3D which decreased cell

proliferation, colony formation as well as migration capacity in vitro. Additionally, knock-down

of EIF3D decreased tumor growth in xenograft models, highlighting this study as a pure

proteomics-driven investigation of a potential drug target that reached pre-clinical testing in

This article is protected by copyright. All rights reserved.


8
www.clinical.proteomics-journal.com Page 9 Proteomics - Clinical Applications

animal models.[27] In another bladder cancer investigation, iTRAQ-based quantitative

proteomics analysis on tissue samples and adjacent non-malignant tissue was conducted by
[28]
Zhang et al. Thirty five proteins were significantly altered (p<0.05), with up-regulation of

Scaffold attachment factor B (SAFB) and GTPase RAN binding protein 1 (RanBP1) further

confirmed by IHC. However, further assessment of these findings in disease models is

required. Additionally, Zha et al.[29] applied iTRAQ labeling coupled to MS analysis for the

comparative assessment of 15 paired cancer and adjacent non-cancerous tissue from

patients with laryngeal carcinoma, revealing 100 differentially expressed proteins. Among

them, S100 calcium binding protein A2 (S100A2), Keratin 17 (KRT17), Fibrinogen B (FGB),

Fibronectin 1 (FN1) and POSTN1 were verified by IHC in 60 laryngeal carcinoma and 20

non-malignant tissues. Although the identified proteins seem to be more related to

inflammation, interference in vitro assays for S100A2, suggested this protein as potential

therapeutic target.[29]

In another study, investigating osteosarcoma, Hypoxia-induced E3 ubiquitin ligase subunit

(WSB1) was highly expressed in tumor tissues, associated with progression and

metastasis, as revealed by IHC analysis in 40 primary osteosarcoma tissues.[30] The invasive

and metastatic potential increased in animal models, where WSB1 was overexpressed to

induce hypoxia. Downstream effectors were investigated by stable isotope labeling with

amino acids in cell culture (SILAC)/ MS. A total of 1,078 proteins were significantly altered

upon WSB1 overexpression (560 upregulated and 518 downregulated; 3.0-fold).[30] RhoGDI2

(inhibitor of Rho proteins) was identified downstream of WSB1 signalling, in decreased

levels. A negative correlation between WSB1 and RhoGDI2 was also confirmed in the

patients’ tissue, suggesting RhoGDI2 as downstream target of WSB1.[30] Zhang et al.[31] also

applied SILAC-based quantitative proteomic approach to investigate changes in human

renal carcinoma tissues and autologous para-cancerous kidney tissues. Eighty-two proteins

were defined as differentially regulated (>1.34 or <0.66-fold), including proteins previously

described in the context of renal cell carcinoma (e.g. nexin family proteins, vimentin, heat

This article is protected by copyright. All rights reserved.


9
www.clinical.proteomics-journal.com Page 10 Proteomics - Clinical Applications

shock proteins) and novel findings. Progesterone receptor membrane components 1

(PGRMC1), of which upregulation was reported in other types of cancer, was further verified

by immunohistochemistry (p<0.05, n=135 paired tissues). Its overexpression significantly

enhanced proliferation of renal cancer cells (p<0.05). Opposite effect was observed after

PGRMC1 silencing.

As evident by the retrieved studies in human cancerous specimens, patient excised tissue is

a valuable source for identification of a large number of cancer-associated proteins, several

of those can be positively verified either by IHC and/or correlated with disease outcome, and

of which functional relevance in cancer is supported by in vitro investigation. Yet, none of the

studies, except for the study by Latosinska et al. has introduced a proteomics- driven target

that has been initially investigated in a pre-clinical setting including animal models.

ii. Cell line proteomics for drug discovery in oncology

A considerable number of studies involve cell-line based investigations to identify potential

drug targets, of which validity/ relevance was supported by in vitro/ in vivo experiments. In

the study by Hue et al.[32] SILAC was employed to assess changes between HCT116 cell

line and its highly invasive subline (HCT116-I8). 3,161 proteins were commonly identified in

three experiments (FDR of 1%, ≥2 peptides)[32] including 128 differentially abundant proteins

(fold ≥1.5).[32] Cdc42BPA (up-regulated, downstream regulator of Cdc42 signalling pathway)

was selected for subsequent analysis in vitro and in human samples. Knock-down of

Cdc42BPA in two highly invasive cell lines resulted in a reduction of invasive potential. IHC

analysis of Cdc42BPA (n=100 primary colon cancer tissues and n=80 adjacent normal

tissues) revealed stronger staining in tumor vs adjacent normal tissues. High Cdc42BPA

expression was associated with lymph node (p=0.002) and distant metastasis (p=0.026), as

well as shorter survival time (p<0.05). Nevertheless, further in vivo experiments are required

to demonstrate its therapeutic potential. In another study, Zhou et al.[33] analysed by SILAC

the membrane proteome of hepatocellular carcinoma cell lines that differs in metastatic

This article is protected by copyright. All rights reserved.


10
www.clinical.proteomics-journal.com Page 11 Proteomics - Clinical Applications

potential. A total of 1,181 proteins were quantified, including 190 differentially abundant

proteins. From the latter, 90 proteins were annotated to plasma membrane. Subsequent

investigation was focused on sodium-potassium-chloride cotransporter 1 (NKCC1), of which

upregulation in cell lines with higher metastatic potential was confirmed by Western blot.

Immunohistochemistry analysis (n=67) showed that NKCC1 is significantly associated

(p<0.05) with tumor differentiation and microvascular invasion. In this study both in vitro and

in vivo experiments were conducted, demonstrating reduction of cell proliferation and

invasion after NKCC1 knockdown, while an opposite effect was observed after NKCC1 over-

expression.[33]

Bassiri et al.[34] aimed at identification of novel drug targets for Merlin-deficient meningioma

and schwannoma through parallel analysis of proteome and phosphoproteome in

schwannoma/ Schwann cells and meningioma/ Meningeal cells. This is a particularly

interesting approach, considering that PTMs play a major role in protein functionality, and

phosphorylation is one of the most intensively studied modification.[35] Importantly, aberrant

phosphorylation status have been implicated in cancer, and therapeutics targeting

phosphorylation pathways are emerging in drug discovery.[35] Upon characterisation of

individual types of cancer, the analysis was focused on 11 commonly deregulated

phosphoproteins between schwannoma and meningioma. Among those, overexpression of

PDZ and LIM domain protein 2 (PDLIM2) was validated through western blot in tissue and

cell lines from both cancers. Knock-down of PDLIM2 significantly reduced cell proliferation in

both meningioma and schwannoma cells, although no further in vivo experiments were

performed.

In a quite different setting, Zheng et al.[36] performed two-dimensional gel electrophoresis

followed by MS to investigate differences in the proteome of squamous cell carcinoma of

tongue cell lines (SCC-15) with acquired resistance to pingyangmycin in comparison to

parental cell lines. A total of 19 differential protein spots were identified (>2-fold change;

triplicate experiments). Western blot and ELISA confirmed significant increase of HSP27 in

This article is protected by copyright. All rights reserved.


11
www.clinical.proteomics-journal.com Page 12 Proteomics - Clinical Applications

cell extract and secretome of resistant cell lines in comparison to other SCCT cell lines,

respectively. Knock-down of HSP27 and treatment with anti-HSP27 antibody were able to

reverse the resistance. Further evaluation of the expression of HSP27 in tumor tissues in

two independent cohorts of patients with squamous cell carcinoma of tongue who received

pingyangmycin and/ or chemotherapy (cohort 1, n=84; cohort 2, n=81) revealed that high

level of HSP27 was associated with significantly shorter overall survival (p=0.0081 for cohort

1, and p=0.0024 for cohort 2).

From the above published reports, it is evident that proteomics has been extensively applied

in cell lines to investigate potential oncological targets. Even though, cell lines might not fully

reflect disease phenotype and also interactions of cancer cells with extracellular matrix, it is

frequently applied as it easily available, technically less challenging and better suited for

investigating secreted proteins.

iii. Cell line proteomics for discovery of downstream effectors

Apart from the application of proteomics for untargeted discovery of drug targets, global

analysis of the proteome was utilised to better understand and characterise protein partners/

effectors downstream of the targeted protein. The concept is of high interest, when

considering that downstream effectors can affect drug efficiency. Numerous studies take

advantage of MS (in many cases iTRAQ technology is applied) to profile downstream effects

of targets, targeting membrane proteins, known kinases and promising targets downstream

of the recently applied immunotherapy (PD1/ PDL1 axis).

In the study by Xie et al.[37] focusing on assessment of Putative HLA-DR-associated protein 1

(PHAP1) in glioma, immuno-based assays (n=30 human glioma tissues and n=12

specimens of non-tumour brain tissues) revealed significant increase of PHAP1 abundance

in tumoral vs non-tumoral tissue. In vitro investigation demonstrated that knock-down of

PHAP1 inhibited proliferation of glioma cells, while its overexpression had an opposite effect.

To investigate how PHAP1 regulates cell proliferation, U251 and U87 cells carrying PHAP1

This article is protected by copyright. All rights reserved.


12
www.clinical.proteomics-journal.com Page 13 Proteomics - Clinical Applications

knock-down and control cells were analysed using iTRAQ. 57 and 71 differentially abundant

proteins were identified through analysis of U251 and U87 cells, respectively; while 15

proteins were overlapping between two analyses. Western blot analysis confirmed that by

PHAP1 knock-down Stathmin (STMN1) was significant decreased. Further investigation of

p27- stathmin inhibitor, and its upstream regulator - phosphorylated Akt (S473) supported

the hypothesis that PHAP1 promotes proliferation of glioma cells through regulation of the

Akt/p27/stathmin pathway.

Along these lines, Programmed cell death protein 1 (PD-1) / ligand (PD-L1) inhibitors have

emerged as a promising strategy to treat various cancers. Considering the low response rate

to therapies targeting these proteins, there is an unmet need to better understand regulation

of its expression at the molecular level. Towards that end, Burr et al.[38] conducted a

genome-wide CRISPR-Cas9 screen in pancreatic cancer cell line to identify regulators of

PD-L1. Analysis resulted in identification of CKLF-like MARVEL transmembrane domain

containing protein 6 (CMTM6), of which depletion in resulted in reduction of PD-L1 level. Co-

immunoprecipitation experiments supported interaction between PD-L1 and CMTM6.

Tandem mass tag quantitative proteomics approach was applied to investigate membrane

proteome of wild type and CMTM6 depleted breast cancer cells. 1424 membrane proteins

were identified, and >2- fold decrease in PD-L1 abundance was observed. Further

experiments revealed that depletion of CMTM6 co-localises with PD-L1 at the plasma

membrane and prevents it from degradation in lysosomes. Moreover, in vitro and in vivo

experiments supported that CMTM6 acts as a regulator of T-lymphocyte-mediated anti-

tumour immunity.

In another study, Bai et al.[39] investigated the role of cadherin-18 (CADH18) in glioma.

Decrease in cadherin-18 from normal to stage IV gliomas was supported by publicly

available data and immunohistochemistry (n=453). Overexpression of CADH18 in glioma

cell lines (U87, U251) significantly decreased cell migration/ invasion (p<0.0001) in

comparison to controls. Overexpressing CADH18 in nude mice exhibited a slower rate of

This article is protected by copyright. All rights reserved.


13
www.clinical.proteomics-journal.com Page 14 Proteomics - Clinical Applications

growth and smaller volume. iTRAQ analysis resulted in identification of 201 and 331

differentially expressed proteins (p≤0.05, ≥1.5-fold change). Cytochrome b-c1 complex

subunit 2, mitochondrial (UQCRC2) was selected as a potential downstream target of

CADH18. Down-regulation of UQCRC2 in CADH18 overexpressed glioma cell lines, partly

reversed the inhibition of invasion/migration ability and chemoresistance. [39] Xu et al.[40]

investigated the impact of human antigen R on the progression of esophageal squamous cell

carcinoma (ESCC). Human antigen R showed to be overexpressed in tumor tissues (n=112)

in comparison to the matched adjacent tissues (n=52) using IHC. Knock-down of human

antigen R decreased proliferation and clonogenicity of ESCC cell lines (TE-1 and Eca-109),

along with a significant reduction of cell invasion and migration. Tandem mass tags- based

proteomics allowed for identification of 100 up-regulated and 122 down-regulated proteins

after human antigen R knock-down. Interleukin-18 (IL18) was identified among the up-

regulated proteins. Treatment of ESCC cells with IL18 led to inhibition of cell proliferation

and metastasis; while in vitro luciferase assay revealed that 3'-UTR of IL18 have a binding

site for human antigen R. IHC supported significant decrease of IL18 in ESCC vs tumor-

adjacent tissues.

Cheung et al.[41] investigated functional network of minichromosome maintenance protein 2

(MCM2) in lung cancer cell lines. Phosphoproteomics analysis resulted in identification of

215 and 107 phosphosites that were significantly regulated (>1.5-fold change) in response to

MCM2 overexpression and silencing, respectively; while proteomics analysis of MCM2

silenced cells led to identification of 46 differentially abundant proteins in comparison to

controls. Moreover, phosphoproteomics analysis revealed that MCM2 promotes cell

proliferation via regulation of high mobility group protein HMG-I/HMG-Y phosphorylation and

the impact of phosphorylation of HMGA1 was supported by Western blot and in vitro
[42]
experiments. Weinberg et al. focused on the characterisation of RIO kinases (RIOK) in

cancer. In vitro studies of isogenic colon-, breast- and lung cancer cell lines revealed that

silencing of RIOK1 inhibited cancer cells proliferation and invasiveness in 2D and 3D culture

This article is protected by copyright. All rights reserved.


14
www.clinical.proteomics-journal.com Page 15 Proteomics - Clinical Applications

system. To further investigate the protein network controlled by RIOK1, proteome of NCI-

H1299 cells expressing and depleted in RIOK1 was analysed using SILAC. Depletion of

RIOK1 affected expression and phosphorylation of proteins involved in multiple biological

processes including ribosomal biogenesis, cell cycle progression, metabolic activity, NF-κB

signaling and metastases. Moreover, in vivo studies demonstrated that mice inoculated with

NCI-H1299 cells expressing RIOK1 exhibited colony formation in lungs, which was not

observed after inoculation of cells depleted in RIOK1. IHC analysis of lung (n=30) and breast

cancer (n=22) tissue samples supported differential expression of RIOK1.

[43]
Li et al. employed immunoprecipitation combined with MS to investigate mechanisms of

anaplastic lymphoma kinase (ALK) signalling in neuroblastoma. Analysis of Ba/F3 cells

expressing FLAG-tagged wildtype ALK (ALKWT-FLAG) and neuroblastoma-associated

F1174L ALK (ALKF1174L-FLAG) resulted in identification of tyrosine kinase Bruton’s tyrosine

kinase (BTK) as a novel ALK interaction partner, further confirmed by co-

immunoprecipitation. BTK expression was detected in both neuroblastoma cell lines and

tumour tissue samples (n=4). Importantly, treatment of neuroblastoma cell lines with BTK

inhibitor (ibrutinib) inhibited cell proliferation, and growth of neuroblastoma xenograft in nude

mice; whereas combination of ibrutinib with ALK inhibitor (crizotinib) further enhance the

inhibitory effect.

In another study, the interaction partners of 14-3-3ζ, a known protein for its overexpression

in non–small cell lung cancer (NSCLC) were investigated by proteomics analysis,[44] after

immunoprecipitation in three different cells lines. 16 proteins that overlapped between all cell

lines, were considered, among those, heat shock protein 27 (Hsp27) was investigated with

interference experiments in vitro. Importantly, validation in human tissue specimens revealed

that combination of 14‐3‐3ζ and Hsp27 expression was an independent prognostic indicator

of OS (p=0.036).[44]

This article is protected by copyright. All rights reserved.


15
www.clinical.proteomics-journal.com Page 16 Proteomics - Clinical Applications

[45]
Additional efforts have been made to analyse the cancer stromal cells. Principe et al.

performed proteomics profiling of fibroblasts isolated from tumor and adjacent tissue from

patients with oral tongue squamous cell carcinoma, focusing on identification of stromal-

derived secreted factors. LC-MS/MS analysis was performed on cell lysates, conditioned

media and exosomes in four matched pairs of cancer associated fibroblast and matched

adjacent fibroblast. Further analysis of proteins identified in conditioned media and

exosomes, revealed 43 and 47 proteins consistently up-regulated (≥1.5-fold, p<0.05) in

fibroblasts derived from tumors. Microfibrillar-associated protein 5 (MFAP5) and Protein

canopy homolog 3 (CNPY3) overlapped between exosomes and conditioned media

analyses, with MFAP5 found more abundant in secretome (conditioned medium, exosomes)

in comparison to total cell lysate. Overexpression of MFAP5 in cancer-associated stroma

was confirmed by IHC (n=69 tongue cancer). Treatment of cancer cells with human

recombinant MFAP5 resulted in increased cell proliferation and migration, likely via induction

of MAPK and AKT pathways.


[46]
In the study by Ale kovi et al., SILAC was applied to investigate secretome from lung

metastatic breast and melanoma cancer cell lines. Up- and down-regulated cancer-specific

lung metastasis secretome signatures were established through the inclusion of proteins

found to be 2-fold up/ down regulated in at least one of the two sublines in comparison to

parental cell line. A signature of 149 and 210 up-regulated as well as 243 and 92 down-

regulated proteins in breast cancer and melanoma were established, respectively. 20 up-

regulated and 16 down-regulated proteins were common in both analyses. Overexpression

of Nidogen 1 (NID-1) enhanced cell migration, invasion, adhesion to the endothelium,

vascular permeability, and tube formation. NID-1 was also found to be upregulated in

patients with metastatic breast cancer and melanoma in comparison to patients without

metastases and its expression was correlated with poor outcome.

Altogether the above studies (as evident from the number of the identified proteins

downstream of a protein target) demonstrate the complexity of targeting one protein and the

This article is protected by copyright. All rights reserved.


16
www.clinical.proteomics-journal.com Page 17 Proteomics - Clinical Applications

potential of proteomics in identifying several downstream effectors/ pathways, moving from a

targeted one- pathway view to a more holistic approach of overlapping molecular pathways.

iv. Proteomics-driven drug discovery in other diseases

Oncology evidently covers the majority of the discovery studies, as tumorous tissue can be

excised from patients. Apart from cancer research though, several groups have employed

high resolution proteomics to investigate the pathophysiology of other chronic implications,

with very promising results in the pre-clinical setting.

Likely, the most successful example is the study of Qi et al.[47] attempting to introduce novel

drug targets for diabetic nephropathy. For this purpose, the authors analysed proteome of

glomeruli from two groups of individuals: 1) no or mild Diabetic nephropathy, Diabetic

nephropathy class 0–I and 2) severe Diabetic nephropathy, Diabetic nephropathy class IIb–

III. Comparative analysis indicated that 88 proteins were found to be up-regulated in group 1

(≥1.5-fold; p ≤ 0.05). Pathway enrichment analysis of these proteins revealed that most of

the top 12 top-ranked pathways were related to glucose metabolism and glycolysis. Pyruvate

kinase M2 (PKM2), up-regulated in group 1, was considered for further investigation

considering the prominent fold change, and possibility to modulate its action either via

pharmacological and genetic approaches.[47] Further in vitro and in vivo experiments

supported that activation of PKM2 likely protect against Diabetic nephropathy through

increase of glucose metabolic flux, inhibition of the production of toxic glucose metabolites

and induction of mitochondrial biogenesis, suggesting the potential of the protein as a drug

target.[47, 48]
The above studies have formed the basis for molecular treatment of diabetic

nephropathy through targeting of PKM2 activators.

Mulligan et al.[49] investigated novel therapeutic targets for Chronic rhinosinusitis with nasal

polyps (CRSwNP), focusing on complement component C3. Proteomics analysis of nasal

mucus samples (n=17) resulted in identification of 466 proteins, including 146 differentially

This article is protected by copyright. All rights reserved.


17
www.clinical.proteomics-journal.com Page 18 Proteomics - Clinical Applications

expressed proteins. 24 and 122 proteins exhibited an increase and decrease in the

abundance between CRSwNP and control group, respectively. Pathway analysis of the

proteins with increased abundance in CRSwNP identified initial triggering of complement

and complement cascade as top hit. Moreover, sinonasal mucus levels of complement C3

were correlated with disease severity. Investigation of the sinonasal tissue showed also an

increase levels of C3 and C3a in CRSwNP in comparison to controls. Further in vitro and in

vivo studies demonstrated that inhibition of the complement C3a receptor reduces

inflammation and CRS development in vivo, supporting its potential as novel therapeutic

target.

In the cardiovascular area, subendothelial retention of LDL is the early stage of

atherosclerosis and calcific aortic valve disease. In an effort to characterise LDL interaction
[50]
with proteoglycans, quantitative proteomics via iTRAQ coupled to MS was performed in

animal derived valve and diverter tissues (n=6 aortic valve leaflets and n=6 renal ostia) after

enrichment with LDL affinity columns. Matrix Proteoglycans and collagens were enriched
[50]
(decorin: 3.3 fold-change; biglycan: 4.4 fold-change . Specificity and affinity of decorin to

LDL was further verified, demonstrating a potential as therapeutic target [50].


[51]
In an investigation involving X-linked lymphoproliferative disease, Peled et al. analysed

molecular alteration downstream of PD-1. Using affinity purification and MS, signalling

lymphocytic activation molecule associated protein (SAP) was identified among proteins

interacting with PD-1. Knock-down of SAP in Jurkat T cells enhanced PD-1 function, while

its overexpression had an opposite effect on PD-1. Furthermore, evaluation of T cells from

patients lacking functional SAP – harbouring X-linked lymphoproliferative disease, indicated

a high hypersensitivity to PD-1 signalling, supporting role of SAP as an inhibitor of PD-1.

E. Profiling drug effects on proteome

i. Drug effects in cancer studies

This article is protected by copyright. All rights reserved.


18
www.clinical.proteomics-journal.com Page 19 Proteomics - Clinical Applications

Closer to drug development process, there are proteomics studies that investigate

mechanisms of drug action through characterisation of drug- affected proteins. This

information contributes to gain a better understanding of therapeutic potential of drugs, and

off-target effects. As concluded by the selected studies below, the majority of the proteomics

applications up to date, involve proteomic profiling upon treatment with kinase inhibitors to

therapeutically target malignancies.

In an effort to profile the alterations upon treatment with cyclin-dependent kinases (CDK)
[52]
inhibitors in neuroblastoma , Delehouse and collaborators applied roscovitine and CR8 in

neuroblastoma cell lines, both with amplified (IMR32) and not amplified MYCN (SH-SY5Y) .

iTRAQ labeling in combination LC-MS analysis, in the treated cells, resulted in the

quantification of 1320 proteins, out of those, expression of 25 proteins was altered by both

compounds. Of those, c-MYC and p27Kip1 were verified by western blot, suggesting a

downregulation of MYC signalling pathway downstream of CDK inhibition. To further confirm

this finding in vivo, IMR32 bearing mice (where MYC is amplified), were treated with CR8

and the tumor size and growth was decreased massively.[52] In another study, the proteomics

alterations downstream of BI-TK/GCV (Bifidobacterium infantis thymidine kinase/nucleoside

analogue ganciclovir) treatment system were investigated by iTRAQ- followed by MS

analysis for bladder cancer.[53] Sixty tumor- bearing animals (rats) were treated and further

proteomics analysis resulted in 402 differentially expressed proteins (1.3-fold change; 192

down-regulated and 210 up-regulated) after BI-TK/GCV treatment. Of the most

downregulated proteins, Peroxiredoxin-I (Prx-I) was verified by Western blotting and IHC,

while the effect of knocking down of the protein was also investigated in a human cell line

(T24), resulting in decrease of phospho-NF-κB p50 and p65. The results showed a

mechanistic insight of treatment with BI-TK/GCV, likely through Prx-I and NF-kB apoptotic

pathway.[53]

This article is protected by copyright. All rights reserved.


19
www.clinical.proteomics-journal.com Page 20 Proteomics - Clinical Applications

Similarly, the effect of Polo-like kinase 1 (Plk1) inhibition was investigated in melanoma.

Cholewa et al.[54] treated BRAFV600E mutant melanoma cell lines with a Plk1

(serine/threonine kinase) inhibitor, namely BI 6727. Label-free nano-LC–MS/MS analysis

was performed on a Q-Exactive instrument, revealing alterations on cellular metabolism,

proteasome degradation, and p53 translation, directing towards apoptosis and cell survival.

Particularly, proteasome subunits, a3, β2, β5, and β1 (PSMA3, PSMB2, PSMB5, and

PSMB1) were identified as downregulated upon MS analysis. For the catalytic subunits, β2,

β5, and β1, the result was verified by western blot following Plk1 inhibition.[54] Ferruci et al[55]

investigated angiogenesis in myeloma progression,[55] focusing on HGF/cMET pathway.

SU11274, a cMET inhibitor was applied together with other anti-myeloma drugs, Bortezomib

and lenalidomide. Upon SU11274 inhibition, 14 proteins were significantly altered (2-fold

change) among those four angiogenic proteins, like annexin A4 (ANXA4), prohibitin (PHB),

peroxiredoxin-6 (PRDX6), and annexin A2 (ANXA2) were downregulated after gel-based

proteomics analysis (2D coupled to MS), while calpain small subunit 1 (CPNS1) was

upregulated. Verification of these 5 proteins was further performed with real-time RT-PCR.[55]

A novel small-molecule inhibitor SKLB-163 (benzothiazole-2-thiol derivative) was developed

based on in silico design for cancer treatment.[56] Apart from the anti-tumor benefit of SKLB-

163 in vivo, proteomics analysis was performed to profile drug’s action in treated human

melanoma cells (A375). 21 proteins were identified with significantly altered expression, with

Rho GDP-dissociation inhibitor 1 (RhoGDI) to be shown with very significant decrease (3.6-

fold), was further validated by western blot, while overexpression rescued tumor

phenotype.[56]

Armstrong et al.[57] combined proteomics and transcriptomics analysis to investigate the

mechanism of action for AUY922 (next generation Hsp90 inhibitor) in prostate cancer in

explants from prostate cancer patients (n=6 for RNA Seq and n=12 for 2D-DIGE). Pathway

analysis of differentially expressed genes and proteins pointed towards enrichment in

regulation of actin cytoskeleton in AUY922 treated explants. AUY922 treatment notably

This article is protected by copyright. All rights reserved.


20
www.clinical.proteomics-journal.com Page 21 Proteomics - Clinical Applications

affected cell morphology and inhibited cell motility and cell invasion, in comparison to vehicle

or 17-AAG (first-generation inhibitor). Assessment of cytoskeletal and ECM-related proteins

revealed a most notable induction of fibronectin by AUY922. Interference with fibronectin

decreased the invasive potential of prostate cancer cell lines, similarly to AUY922. Jain et

al.[58] investigated changes in the proteome of U87 glioma cells after treatment with S3I201

(STAT3 inhibitor) using two dimensional gel electrophoresis and iTRAQ, resulting in

identification of six differentially expressed protein spots between treated and untreated

cells. Cytochrome C oxidase subunit 5A, peroxiredoxin-6, Prot DJ-1, chloride intra-cellular

channel protein 1 were identified via MS. iTRAQ approach enabled identification of 134

significantly altered proteins expressed (1% FDR and fold≥1.5). Differential expression of

two proteins involved in cellular metabolism i.e. PGAM1 and TPI1 was investigated by

Western blot. An increased expression of PGAM1 was observed in glioma cells treated with

S3I201, in line with proteomics analysis.


[59]
Roolf et al. investigated effect of sorafenib, inhibitor targeting receptor tyrosine kinase

FLT3, on a panel of thirteen Acute Myeloid Leukemia cell lines that differs in FLT3 status [i.e.

FLT3 wild-type and internal tandem duplication (ITD) mutated cell lines]. Treatment of cells

with increased concentration of Sorafenib revealed that FLT3-ITD+ cells are generally more

sensitive to sorafenib than FLT3 wild-type cells. Phosphoproteomics was applied to

investigate mechanisms underlying sensitivity to sorafenib in both FTL3 wild-type and

mutated cell lines. For that purpose, SILAC labelled MV4-11 (Sorafenib sensitive and FLT3-

ITD+), MONO-MAC-1 (Sorafenib sensitive and FLT3 wild-type), and SKM-1 (Sorafenib

insensitive and FLT3 wild-type) cell lines were treated with 0.01 M sorafenib, and 0.2 M

sorafenib. Phosphoproteomics analysis and in silico pathway analysis indicated different

pathways being affected in Sorafenib sensitive FLT3 wild-type and FLT3-ITD+ cells. In a

FLT3 wildtype cells Sorafenib inhibited MEK/ERK signaling pathway, while in the FLT3-ITD+

cell line, Sorafenib treatment supressed mTOR signalling.

This article is protected by copyright. All rights reserved.


21
www.clinical.proteomics-journal.com Page 22 Proteomics - Clinical Applications

[60]
On a study investigating resistance to chemotherapy, Tripathi et al. studied mechanisms

of chemoresistance of small-cell lung cancer (SCLC) cell lines via LC-MS/MS analysis. Five

cell surface receptors exhibiting the most robust difference in abundance between

chemoresistant and chemosensitive cells were further validated by WB and its expression

was assessed in patients’ derived xenografts. Among them, upregulation of Cell surface

glycoprotein MUC18 (MCAM) in chemoresistant tumors was supported by IHC. Knock-down

of MCAM in chemoresistant cells significantly decreased cell proliferation (p<0.001) and

colony-formation (p<0.01). Additional experiments suggested that the MCAM-mediated

sensitization to chemotcherapeuic agents occurred through SOX2-dependent up-regulation

of mitochondrial 37S ribosomal protein 1/ATP-binding cassette subfamily C member 1

(MRP1/ABCC1) and the PI3/AKT pathway.

ii. Proteomics drug effects’ investigation in other pathological conditions

Filipovic´ et al.[61] investigated mechanisms of Fluoxetine action, an anti-depressive drug,

using GeLC-MS/MS analysis of hippocampal tissue excised from treated and untreated rats.

83 proteins were identified as differentially expressed in cytosolic hippocampal proteome

after Fluoxetine treatment, with majority of proteins being down-regulated in comparison to

vehicle-treated controls; 63 proteins were identified as differentially expressed in

mitochondrial proteome, with most of them being up-regulated after treatment. Differential

expression of Calretinin was confirmed by Western Blot, while the change in Parvalbumin

expression was supported by immunofluorescence.

Bowen et al.[62] focused on identification of compounds able to inhibit Muscle ring finger 1 –

titin interaction, previously shown to be activated in numerous conditions associated with

skeletal muscle wasting. Using AlphaScreen assay, 9 compounds have shown to have a

high Muscle ring finger 1- titin specificity. From these, three compounds inhibited also

Muscle ring finger 1 activity, with ID#704946 compound characterised by low cytotoxicity on

This article is protected by copyright. All rights reserved.


22
www.clinical.proteomics-journal.com Page 23 Proteomics - Clinical Applications

myoblasts or myotubes. Animal experiments with ID#704946, showed that the compound

prevented atrophy in myotubes and attenuated fibre atrophy and contractile dysfunction in

mice during cardiac cachexia. Proteomics analysis of muscle tissue was applied to

investigate proteins/ pathways affected by this compound, and 51 proteins were found to be

differentially expressed between MCT group (cachexia model) vs sham (p<0.01), and 70

proteins were found as differentially expressed between MCT group treated with ID#704946

vs MCT group (p<0.01). Analysis revealed that compound attenuated proteins involved in

stress pathways (e.g. Muscle ring finger 1), apoptosis (BAX), protein synthesis (elF2B-delta),

along with inhibition actin ubiquitinylation and proteasome activity.[62]

F. Chemical proteomics for drug selectivity and inhibitor binding

Chemical proteomics platforms have been widely applied to support drug discovery process,

with activity-based protein profiling (ABPP) in combination with MS to be the most common

technique to discover selective and in vivo-active inhibitors for enzymes.[63] After evaluating

the proteomics studies (that are described in detail below), a popular field of research

involves quantitative MS protocols to profile drug targets of covalent kinase inhibitors, based

on conserved active cysteines.[63] A common approach is based on Kinobead technology,[64]

through which, a broad-range of kinase inhibitors is immobilized to beads for purification of

kinases from tissue samples or cells. In combination with MS analysis, this approach allows

for simulations measurement of multiple protein targets.[65] Along these lines, combination of

chemical proteomics with fragment-based ligand discovery was established as a platform to

map interactions of small molecules and proteins in human cells.[66] Last but not least,

chemical proteomics approach has been used to assess lipid-protein interactions.[67]

i. Chemical proteomics coupled to mass spectrometry in oncology

This article is protected by copyright. All rights reserved.


23
www.clinical.proteomics-journal.com Page 24 Proteomics - Clinical Applications

Kinase inhibitors represent a major class of drug targets. Recently, efforts have been

undertaken to characterize target space of 243 clinically evaluated kinase inhibitors. For that

purpose, Klaeger et al.[65] applied kinobead technology in combination with MS on cancer

lysate cells.[65] As a result, the largest resource for clinical drug-target interactions has been

established[65]. Along these lines, tyrosine kinase inhibitors (TKIs) have become an important

therapeutic option for treating several forms of cancer. Gefitinib, an inhibitor of the epidermal

growth factor receptor (EGFR), is in clinical use for treating NSCLC harboring activating

EGFR mutations. A chemical proteomic approach[68] comprising from kinobeads and

quantitative MS was applied for the identification of kinase inhibitor resistance mechanisms

in cancer cells. Previously described amplification of MET and found Ephrin type-A receptor

2 (EPHA2) was confirmed to be more than 10-fold overexpressed (p <0.001) in gefitinib-

resistant HCC827 cells suggesting a potential role in developing resistance. siRNA-mediated

EPHA2 knock-down or treating cells with the multikinase inhibitor dasatinib restored

sensitivity to gefitinib. Of all dasatinib targets, EPHA2 exhibited the most drastic effect (p

<0.001). In addition, EPHA2 knock-down or ephrin-A1 treatment of resistant cells decreased

FAK phosphorylation and cell migration.[68] In another study, Heinzlmeir et al. [69]
aimed at

development of EPHA2 inhibitors with improved target profile, with dasatinib used as a basis

of a lead structure. For that purpose, chemoproteomics-aided medicinal chemistry approach

was employed. Kinobeads pulldown assays in combination with MS analysis was used to

assess the affinity of identified inhibitors as well as investigate their selectivity in comparison

to parent lead compound. Furthermore, the effect of the inhibitor candidates on proliferation

of SF-268 glioblastoma cell line was assessed, and one compound showed a strong anti-

proliferative potential and warrants further investigation.


[70]
Along the same lines , Wang et al. profiled the protein targets of andrographolide (Andro)

a natural product with anti-inflammation and anti-cancer effects. The authors introduced a

combination of state-of-art chemical and proteomics techniques, combining ABPP with bio-

orthogonal click chemistry and iTRAQ to identify drug targets in vitro and in vivo. 75 potential

This article is protected by copyright. All rights reserved.


24
www.clinical.proteomics-journal.com Page 25 Proteomics - Clinical Applications

Andro targets were identified with high confidence and two targets, NF-κB and β-actin, were

validated by pull-down assays and WB, while NF-κB p50 protein, was investigated for the

PTM site after Andro treatment, by MS/MS.[70] Roberts et al.[71] combined chemical genetic

screening with chemoproteomics platform to identify novel therapeutic compounds for

pancreatic cancer and identify its targets. Considering the ability of compounds to impair

pancreatic cancer cell survival or proliferation as well as nonspecific toxicity, chemical

genetic screening pointed out three compounds (DKM 2−67, DKM2−83, and DKM 2−93),

and DKM 2−93 was selected for further investigations in vivo and for targets identification

using chemoproteomics. It has been shown that DKM 2−93 impairs tumor growth in vivo,

while thorough chemoproteomics approach cysteine 250 on ubiquitin-like modifier activating

enzyme 5 (UBA5) was identified as a druggable hotspot targeted by DKM 2−93. Moreover,

knock-down of UBA5 in pancreatic cancer cell lines mimicked the effect of DKM 2−93

treatment. In a similar way, Bateman et al.[72] identified compound that impairs colorectal

cancer cell pathogenicity via targeting cystein 1101 on Reticulon 4. Knock-down of Reticulon

4 or treatment of cells with DKM 3-30 affected also morphology of endoplasmic reticulum

and nuclear envelope. Along these lines, Grossman et al.[73] applied the same

chemoproteomic platform to discover drugabble hotspots targeted by withaferin A, an anti-

cancer natural product, followed by covalent ligand screening to support the translation of

natural products into therapeutics, through development of more synthetically tractable

compounds. Chemoproteomics analysis showed that withaferin A specifically targets

cysteine 377 of a regulatory subunit within the tumor-suppressor protein phosphatase 2A,

leading to its activation, and inactivation of AKT signalling. This impairs breast cancer

pathogenicity, as supported by in vitro and in vivo investigation. JNS 1-40 was identified as a

ligand interacting with C377 of PPP2R1A through covalent ligand screening, having similar

effects to withaferin A.

ii. Proteomics drug profiling in other conditions

This article is protected by copyright. All rights reserved.


25
www.clinical.proteomics-journal.com Page 26 Proteomics - Clinical Applications

Excepting the cancer studies, Sulaiman et al. employed chemical proteomics to identify

protein targets of SH-11037, an homoisoflavonoid derivative, with antiangiogenic activity in

mouse model of wet age-related macular degeneration. Pull-down proteins were separated

by SDS-PAGE, followed by MS analysis used for protein identification and Soluble epoxide

hydrolase was identified as a target of SH-1103. In silico docking analysis showed that SH-

1103 occupies the entire active site of Soluble epoxide hydrolase. Soluble epoxide hydrolase

was inhibited by SH-11037 in vitro and in vivo, although the effect was not that prominent as

for known inhibitors of Soluble epoxide hydrolase. Activity and level of Soluble epoxide

hydrolase in the eyes of a choroidal neovascularization mouse model significantly was up-

regulated as well as the enzyme was also overexpressed in human wet age-related macular

degeneration eyes.
[74]
Tian et al. investigated targets of calenduloside E, being triterpenoid saponin compound,

and signaling pathways underlying its protective activity on endothelial cells using chemical

proteomics. 587 proteins were identified (at least 1 unique peptide, score >0) through biotin–

neutravidin pull-down assay followed by geLC-MS/MS. Numerous proteins were involved in

cell survival signaling, with some of these pathways being associated with anti-apoptotic

activity such as PI3K-AKT, VEGF, MAPK and ER stress. In vitro assay supported that biotin

conjugated calenduloside E analogue protects HUVEC cells against ox-LDL-induced injury.

G. Structural insights by using mass spectrometry

Mass spectrometry analysis has been also employed to confirm the chemical structure of

synthesized compounds/ ligands targeting proteins of interest. Kadasi et al. [75] performed a

structure based design of benzofuran-3(2H)-one analogues targeting HSP90. In silico

docking study was conducted followed by chemical synthesis of the ligands. Chemical

structure of the compounds was confirmed by IR, NMR and MS. Subsequent in vitro

investigation in breast cancer and melanoma cells highlighted 2-(4-(4-fluorobenzyloxy)-

This article is protected by copyright. All rights reserved.


26
www.clinical.proteomics-journal.com Page 27 Proteomics - Clinical Applications

benzylidene)-5-chloro-6-hydroxybenzofuran-3(2H)-one (SY3) as the promising anticancer

agent that enabled significant decrease of cell proliferation, when comparing to first-class

HSP90 inhibitor, 17-AAG.

H. Outlook

Through a systematic literature search, in this article, we attempted to provide an overview

on the most promising proteomics studies categorized according to their application in

different fields of drug discovery and development, covering the following areas: target

identification for drug discovery, proteome profiling for drug effects, chemical proteomics for

drug selectivity and inhibitor binding and structural proteomics studies for assisting in silico

docking. The aim of the evaluation was to provide a critical view on how close the

proteomics results are to being implemented in drug development pipeline. Collective

elements based on the literature search showed that high-resolution MS-based proteomics

has been employed in a plethora of studies, involving patient-derived patients, cell lines and

already developed drugs. As evident from the number of the retrieved studies, most of the

proteomics investigations are in the context of oncology. This observation is, to certain

extent, expected considering that access to tissue biospecimens is generally more

permissive in tumor affected areas and after the surgical removal of the malignant tissue.

Currently, most of the cancer drugs that are developed show low response rates and are

associated with side-effects and/or mechanisms of resistance, which are frequently adopted.

Considering this aspect, global proteomic profiling, particularly for identifying downstream

effectors and drug’s mechanism of action are of vital importance, as they enable a more

comprehensive description of molecular alterations, not only at the level of individual

proteins, but also at the level of pathways and their relationship. Moreover, combination of

knowledge at the molecular level with pathology can improve on the drug discovery and

development process, as also schematically depicted in Figure 2.

This article is protected by copyright. All rights reserved.


27
www.clinical.proteomics-journal.com Page 28 Proteomics - Clinical Applications

Nevertheless, the proteomic studies, at least as derived by this systematic search are not

free of limitations. Frequently the number of samples (particularly the patient-derived tissue

specimens) is low, as the availability of the tissue may be also limited. This has an impact on

the analysis in terms of statistical power, and application of correction for multiple testing is

challenging. Moreover, cell line models cannot always accurately reflect the human

physiological conditions, as diseases like cancer can be highly heterogeneous among

different patients. More importantly, apart from the technical limitations, the field is yet

noticeably not that advanced in terms of proteomics-derived drugs that have reached the

phase of clinical trials, and ultimately clinical implementation. On a positive note, certain very

promising studies, as in the case of diabetic nephropathy through the targeting of PKM2 and

bladder cancer, through EIF3D pave the way towards molecular therapy based on the

proteomics profiling. Certainly, recent breakthroughs in thermal proteome profiling assays

are expected to change drug development landscape, particularly regarding drug screening.

In an effort to move from a more static observation (likely this is mostly the challenge in

genomics analysis) to a more dynamic profiling and in order to improve real time monitoring,

novel innovative proteomics platforms have been recently introduced. Along these lines, the

advantages of chemical proteomics (i.e. ABPP) and cellular thermal shift assay (CETSA) are

now combined with high resolution MS, allowing for characterization of drug’s action in an in

vitro environment that is closer to the physiological conditions in the cell.

Ideally, in a future clinical scenario (as depicted in Figure 3) where personalised medicine is

followed, proteomics is envisioned to have an important role in all parts from drug targeting

till section of appropriate intervention. Proteomics profiling can at first indicate altered

pathways, providing insights at the molecular level, beyond the clinical and/ histological

phenotype. Then, drugs can be directly predicted based on molecular profiling data and can

be suggested considering the probability to reverse the clinical phenotype. As through

proteomics and drug mechanism networks, drug targets can be linked with biomarkers,

especially markers to predict and monitor treatment response can be subsequently applied.

This article is protected by copyright. All rights reserved.


28
www.clinical.proteomics-journal.com Page 29 Proteomics - Clinical Applications

Collectively, by this article we provide an overview on the status of proteomics in the field of

drug discovery, exemplified by the most prominent studies in the field of chronic diseases.

Acknowledgements

The work was supported by the BioMedBC Project (Project ID: 752755) and PCaProTreat

Project (Project ID: 800048), funded by the EU Commission, under the MSCA-IF-2016

Individual Fellowships.

Conflicts of interest

Harald Mischak is the founder and co-owner of Mosaiques Diagnostics GmbH. Maria Frantzi

and Agnieszka Latosinska are employed by Mosaiques Diagnostics GmbH.

References

[1] A. L. Hopkins, C. R. Groom, Nature reviews. Drug discovery 2002, 1, 727.


[2] J. L. Walgren, D. C. Thompson, Toxicology letters 2004, 149, 377.
[3] R. Aebersold, M. Mann, Nature 2003, 422, 198.
[4] J. W. Harper, E. J. Bennett, Nature 2016, 537, 328.
[5] C. C. Wong, D. Cociorva, J. D. Venable, T. Xu, J. R. Yates, 3rd, Journal of the
American Society for Mass Spectrometry 2009, 20, 1405.
[6] T. Nilsson, M. Mann, R. Aebersold, J. R. Yates, 3rd, A. Bairoch, J. J. Bergeron,
Nature methods 2010, 7, 681.
[7] D. Martinez Molina, R. Jafari, M. Ignatushchenko, T. Seki, E. A. Larsson, C. Dan, L.
Sreekumar, Y. Cao, P. Nordlund, Science 2013, 341, 84.
[8] R. Jafari, H. Almqvist, H. Axelsson, M. Ignatushchenko, T. Lundback, P. Nordlund, D.
Martinez Molina, Nature protocols 2014, 9, 2100.
[9] B. Mayer, A. Heinzel, A. Lukas, P. Perco, Current pharmaceutical design 2017, 23,
29.
[10] I. Belczacka, A. Latosinska, J. Metzger, D. Marx, A. Vlahou, H. Mischak, M. Frantzi,
Mass spectrometry reviews 2018.
[11] M. Frantzi, A. Latosinska, G. Kontostathi, H. Mischak, Proteomics 2018, 18,
e1700463.
[12] F. B. Reinhard, D. Eberhard, T. Werner, H. Franken, D. Childs, C. Doce, M. F.
Savitski, W. Huber, M. Bantscheff, M. M. Savitski, G. Drewes, Nature methods 2015, 12,
1129.
[13] M. M. Savitski, F. B. Reinhard, H. Franken, T. Werner, M. F. Savitski, D. Eberhard,
D. Martinez Molina, R. Jafari, R. B. Dovega, S. Klaeger, B. Kuster, P. Nordlund, M.
Bantscheff, G. Drewes, Science 2014, 346, 1255784.
[14] G. M. West, J. W. Thompson, E. J. Soderblom, L. G. Dubois, P. D. Dearmond, M. A.
Moseley, M. C. Fitzgerald, Analytical chemistry 2010, 82, 5573.
[15] J. R. Thomas, S. M. Brittain, J. Lipps, L. Llamas, R. K. Jain, M. Schirle, Methods in
molecular biology 2017, 1647, 1.
[16] A. Mateus, T. A. Maatta, M. M. Savitski, Proteome science 2016, 15, 13.

This article is protected by copyright. All rights reserved.


29
www.clinical.proteomics-journal.com Page 30 Proteomics - Clinical Applications

[17] B. F. Cravatt, A. T. Wright, J. W. Kozarich, Annual review of biochemistry 2008, 77,


383.
[18] D. Robinette, N. Neamati, K. B. Tomer, C. H. Borchers, Expert review of proteomics
2006, 3, 399.
[19] K. Kubota, M. Funabashi, Y. Ogura, Biochimica et biophysica acta. Proteins and
proteomics 2019, 1867, 22.
[20] K. V. Huber, K. M. Olek, A. C. Muller, C. S. Tan, K. L. Bennett, J. Colinge, G. Superti-
Furga, Nature methods 2015, 12, 1055.
[21] I. Dagogo-Jack, A. T. Shaw, Nature reviews. Clinical oncology 2018, 15, 81.
[22] M. Schirle, M. Bantscheff, B. Kuster, Chemistry & biology 2012, 19, 72.
[23] F. Autelitano, D. Loyaux, S. Roudieres, C. Deon, F. Guette, P. Fabre, Q. Ping, S.
Wang, R. Auvergne, V. Badarinarayana, M. Smith, J.-C. Guillemot, S. A. Goldman, S.
Natesan, P. Ferrara, P. August, Plos One 2014, 9.
[24] J. G. Rodrigues, M. Balmana, J. A. Macedo, J. Pocas, A. Fernandes, J. C. M. de-
Freitas-Junior, S. S. Pinho, J. Gomes, A. Magalhaes, C. Gomes, S. Mereiter, C. A. Reis,
Cellular immunology 2018, 333, 46.
[25] B. Collet, T. Avril, M. Aubry, A. Hamlat, P.-J. Le Reste, D. Chiforeanu, E. Vauleon, J.
Mosser, V. Quillien, Journal of Proteomics 2014, 110, 7.
[26] Y. Kotobuki, L. Yang, S. Serada, A. Tanemura, F. Yang, S. Nomura, A. Kudo, K.
Izuhara, H. Murota, M. Fujimoto, I. Katayama, T. Naka, Pigment Cell & Melanoma Research
2014, 27.
[27] A. Latosinska, M. Mokou, M. Makridakis, W. Mullen, J. Zoidakis, V. Lygirou, M.
Frantzi, I. Katafigiotis, K. Stravodimos, M. C. Hupe, M. Dobrzynski, W. Kolch, A. S.
Merseburger, H. Mischak, M. G. Roubelakis, A. Vlahou, Oncotarget 2017.
[28] Q. Zhang, S. Huang, H. Luo, X. Zhao, G. Wu, D. Wu, American Journal of Cancer
Research 2017, 7, 935.
[29] C. Zha, X. H. Jiang, S. F. Peng, Plos One 2015, 10.
[30] J. Cao, Y. Wang, R. Dong, G. Lin, N. Zhang, J. Wang, N. Lin, Y. Gu, L. Ding, M.
Ying, Q. He, B. Yang, Cancer Research 2015, 75, 4839.
[31] D. Zhang, X. Xia, X. Wang, P. Zhang, W. Lu, Y. Yu, S. Deng, H. Yang, H. Zhu, N. Xu,
S. Liang, Plos One 2017, 12.
[32] H.-F. Hu, W. W. Xu, Y. Wang, C.-C. Zheng, W.-X. Zhang, B. Li, Q.-Y. He, Journal of
Proteome Research 2018, 17, 265.
[33] Y. Zhou, W. Sun, N. Chen, C. Xu, X. Wang, K. Dong, B. Zhang, J. Zhang, N. Hao, A.
Sun, H. Wei, F. He, Y. Jiang, Oncotarget 2017, 8, 66328.
[34] K. Bassiri, S. Ferluga, V. Sharma, N. Syed, C. L. Adams, E. Lasonder, C. O.
Hanemann, Ebiomedicine 2017, 16, 76.
[35] V. Singh, M. Ram, R. Kumar, R. Prasad, B. K. Roy, K. K. Singh, The protein journal
2017, 36, 1.
[36] G. Zheng, Z. Zhang, H. Liu, Y. Xiong, L. Luo, X. Jia, C. Peng, Q. Zhang, N. Li, Y. Gu,
M. Lu, Y. Song, H. Pan, J. Liu, W. Liu, Z. He, Clinical Cancer Research 2018, 24, 1163.
[37] M. Xie, Z. Ji, Y. Bao, Y. Zhu, Y. Xu, L. Wang, S. Gao, Z. Liu, Z. Tian, Q. Meng, H.
Shi, R. Yu, Journal of Cellular and Molecular Medicine 2018, 22, 3595.
[38] M. L. Burr, C. E. Sparbier, Y.-C. Chan, J. C. Williamson, K. Woods, P. A. Beavis, E.
Y. N. Lam, M. A. Henderson, C. C. Bell, S. Stolzenburg, O. Gilan, S. Bloor, T. Noori, D. W.
Morgens, M. C. Bassik, P. J. Neeson, A. Behren, P. K. Darcy, S.-J. Dawson, I. Voskoboinik,
J. A. Trapani, J. Cebon, P. J. Lehner, M. A. Dawson, Nature 2017, 549, 101.
[39] Y.-H. Bai, Y.-B. Zhan, B. Yu, W.-W. Wang, L. Wang, J.-Q. Zhou, R.-K. Chen, F.-J.
Zhang, X.-W. Zhao, W.-C. Duan, Y.-M. Wang, J. Liu, J.-J. Bao, Z.-Y. Zhang, X.-Z. Liu,
Cellular physiology and biochemistry : international journal of experimental cellular
physiology, biochemistry, and pharmacology 2018, 48, 1755.
[40] X. Xu, C. Song, Z. Chen, C. Yu, Y. Wang, Y. Tang, J. Luo, Cancer Research and
Treatment 2018, 50, 71.

This article is protected by copyright. All rights reserved.


30
www.clinical.proteomics-journal.com Page 31 Proteomics - Clinical Applications

[41] C. H. Y. Cheung, C.-L. Hsu, K.-P. Chen, S.-T. Chong, C.-H. Wu, H.-C. Huang, H.-F.
Juan, Scientific Reports 2017, 7.
[42] F. Weinberg, N. Reischmann, L. Fauth, S. Taromi, J. Mastroianni, M. Koehler, S.
Halbach, A. C. Becker, N. Deng, T. Schmitz, F. M. Uhl, N. Herbener, B. Riedel, F. Beier, A.
Swarbrick, S. Lassmann, J. Dengjel, R. Zeiser, T. Brummer, Ebiomedicine 2017, 20, 79.
[43] T. Li, Y. Deng, Y. Shi, R. Tian, Y. Chen, L. Zou, J. U. Kazi, L. Ronnstrand, B. Feng,
S. O. Chan, W. Y. Chan, J. Sun, H. Zhao, Oncogene 2018.
[44] G.-Y. Zhao, J.-Y. Ding, J. Gu, C.-L. Lu, Z.-W. Lin, J. Guo, D. Ge, Cancer 2014, 120,
652.
[45] S. Principe, S. Mejia-Guerrero, V. Ignatchenko, A. Sinha, A. Ignatchenko, W. Shi, K.
Pereira, S. Su, S. H. Huang, B. O'Sullivan, W. Xu, D. P. Goldstein, I. Weinreb, L. Ailles, F.-F.
Liu, T. Kislinger, Journal of Proteome Research 2018, 17, 2045.
[46] M. Aleckovic, Y. Wei, G. LeRoy, S. Sidoli, D. D. Liu, B. A. Garcia, Y. Kang, Genes &
Development 2017, 31, 1439.
[47] W. Qi, H. A. Keenan, Q. Li, A. Ishikado, A. Kannt, T. Sadowski, M. A. Yorek, I. H.
Wu, S. Lockhart, L. J. Coppey, A. Pfenninger, C. W. Liew, G. Qiang, A. M. Burkart, S.
Hastings, D. Pober, C. Cahill, M. A. Niewczas, W. J. Israelsen, L. Tinsley, I. E. Stillman, P.
S. Amenta, E. P. Feener, M. G. Vander Heiden, R. C. Stanton, G. L. King, Nature medicine
2017, 23, 753.
[48] N. Chun, C. M. Wyatt, J. C. He, Kidney International 2017, 92, 780.
[49] J. K. Mulligan, K. Patel, T. Williamson, N. Reaves, W. Carroll, S. E. Stephenson, P.
Gao, R. R. Drake, B. A. Neely, S. Tomlinson, R. J. Schlosser, C. Atkinson, Mucosal
immunology 2018.
[50] E. B. Neufeld, L. M. Zadrozny, D. Phillips, A. Aponte, Z.-X. Yu, R. S. Balaban,
Atherosclerosis 2014, 233, 113.
[51] M. Peled, A. S. Tocheva, S. Sandigursky, S. Nayak, E. A. Philips, K. E. Nichols, M.
Strazza, I. Azoulay-Alfaguter, M. Askenazi, B. G. Neel, A. J. Pelzek, B. Ueberheide, A. Mor,
Proceedings of the National Academy of Sciences of the United States of America 2018,
115, E468.
[52] C. Delehouze, K. Godl, N. Loaec, C. Bruyere, N. Desban, N. Oumata, H. Galons, T.
I. Roumeliotis, E. G. Giannopoulou, J. Grenet, D. Twitchell, J. Lahti, N. Mouchet, M. D.
Galibert, S. D. Garbis, L. Meijer, Oncogene 2014, 33, 5675.
[53] L. Jiang, X. Xiao, J. Ren, Y. Tang, H. Weng, Q. Yang, M. Wu, W. Tang, Plos One
2014, 9.
[54] B. D. Cholewa, M. C. Pellitteri-Hahn, C. O. Scarlett, N. Ahmad, Journal of Proteome
Research 2014, 13, 5041.
[55] A. Ferrucci, M. Moschetta, M. A. Frassanito, S. Berardi, I. Catacchio, R. Ria, V.
Racanelli, A. Caivano, A. G. Solimando, D. Vergara, M. Maffia, D. Latorre, A. Rizzello, A.
Zito, P. Ditonno, E. Maiorano, D. Ribatti, A. Vacca, Clinical Cancer Research 2014, 20,
5796.
[56] X. Peng, G. Xie, Z. Wang, H. Lin, T. Zhou, P. Xiang, Y. Jiang, S. Yang, Y. Wei, L. Yu,
Y. Zhao, Cell Death & Disease 2014, 5.
[57] H. K. Armstrong, J. L. Gillis, I. R. D. Johnson, Z. D. Nassar, M. Moldovan, C. Levrier,
M. C. Sadowski, M. Y. Chin, E. S. T. Guns, G. Tarulli, D. J. Lynn, D. A. Brooks, L. A. Selth,
M. M. Centenera, L. M. Butler, Scientific Reports 2018, 8.
[58] R. Jain, A. Atak, A. Yeola, S. Srivastava, Journal of Proteomics 2017, 150, 341.
[59] C. Roolf, N. Dybowski, A. Sekora, S. Mueller, G. Knuebel, A. Tebbe, H. M. Escobar,
K. Godl, C. Junghanss, C. Schaab, Molecular & Cellular Proteomics 2017, 16, 1365.
[60] S. C. Tripathi, J. F. Fahrmann, M. Celiktas, M. Aguilar, K. D. Marini, M. K. Jolly, H.
Katayama, H. Wang, E. N. Murage, J. B. Dennison, D. N. Watkins, H. Levine, E. J. Ostrin, A.
Taguchi, S. M. Hanash, Cancer Research 2017, 77, 4414.
[61] D. Filipovic, V. Costina, I. Peric, A. Stanisavljevic, P. Findeisen, Brain Research
2017, 1659, 41.

This article is protected by copyright. All rights reserved.


31
www.clinical.proteomics-journal.com Page 32 Proteomics - Clinical Applications

[62] T. S. Bowen, V. Adams, S. Werner, T. Fischer, P. Vinke, M. N. Brogger, N. Mangner,


A. Linke, P. Sehr, J. Lewis, D. Labeit, A. Gasch, S. Labeit, Journal of Cachexia Sarcopenia
and Muscle 2017, 8, 939.
[63] B. R. Lannine, L. R. Whitby, M. M. Dix, J. Douhan, A. M. Gilbert, E. C. Hett, T.
Johnson, C. Joslynl, J. C. Kath, S. Niessen, L. R. Roberts, M. E. Schnute, C. Wang, J. J.
Hulce, B. Wei, L. O. Whiteley, M. M. Hayward, B. F. Cravatt, Nature Chemical Biology 2014,
10, 760.
[64] F. M. Deane, A. J. S. Lin, P. G. Hains, S. L. Pilgrim, P. J. Robinson, A. McCluskey,
Acs Omega 2017, 2, 3828.
[65] S. Klaeger, S. Heinzlmeir, M. Wilhelm, H. Polzer, B. Vick, P.-A. Koenig, M. Reinecke,
B. Ruprecht, S. Petzoldt, C. Meng, J. Zecha, K. Reiter, H. Qiao, D. Helm, H. Koch, M.
Schoof, G. Canevari, E. Casale, S. R. Depaolini, A. Feuchtinger, Z. Wu, T. Schmidt, L.
Rueckert, W. Becker, J. Huenges, A.-K. Garz, B.-O. Gohlke, D. P. Zolg, G. Kayser, T.
Vooder, R. Preissner, H. Hahne, N. Tonisson, K. Kramer, K. Goetze, F. Bassermann, J.
Schlegl, H.-C. Ehrlich, S. Aiche, A. Walch, P. A. Greif, S. Schneider, E. R. Felder, J. Ruland,
G. Medard, I. Jeremias, K. Spiekermann, B. Kuster, Science 2017, 358.
[66] C. G. Parker, A. Galmozzi, Y. Wang, B. E. Correia, K. Sasaki, C. M. Joslyn, A. S.
Kim, C. L. Cavallaro, R. M. Lawrence, S. R. Johnson, I. Narvaiza, E. Saez, B. F. Cravatt,
Cell 2017, 168, 527.
[67] K. M. Lum, Y. Sato, B. A. Beyer, W. C. Plaisted, J. L. Anglin, L. L. Lairson, B. F.
Cravatt, Acs Chemical Biology 2017, 12, 2671.
[68] H. Koch, M. E. D. C. Busto, K. Kramer, G. Medard, B. Kuster, Journal of Proteome
Research 2015, 14, 2617.
[69] S. Heinzlmeir, J. Lohse, T. Treiber, D. Kudlinzki, V. Linhard, S. L. Gande, S.
Sreeramulu, K. Saxena, X. Liu, M. Wilhelm, H. Schwalbe, B. Kuster, G. Medard,
Chemmedchem 2017, 12, 999.
[70] J. Wang, X. F. Tan, N. Van Sang, P. Yang, J. Zhou, M. Gao, Z. Li, T. K. Lim, Y. He,
C. S. Ong, Y. Lay, J. Zhang, G. Zhu, S.-L. Lai, D. Ghosh, Y. K. Mok, H.-M. Shen, Q. Lin,
Molecular & Cellular Proteomics 2014, 13, 876.
[71] A. M. Roberts, D. K. Miyamoto, T. R. Huffman, L. A. Bateman, A. N. Ives, D.
Akopian, M. J. Heslin, C. M. Contreras, M. Rape, C. F. Skibola, D. K. Nomura, Acs Chemical
Biology 2017, 12, 899.
[72] L. A. Bateman, T. B. Nguyen, A. M. Roberts, D. K. Miyamoto, W. M. Ku, T. R.
Huffman, Y. Petri, M. J. Heslin, C. M. Contreras, C. F. Skibola, J. A. Olzmann, D. K. Nomura,
Chemical Communications 2017, 53, 7234.
[73] E. A. Grossman, C. C. Ward, J. N. Spradlin, L. A. Bateman, T. R. Huffman, D. K.
Miyamoto, J. I. Kleinman, D. K. Nomura, Cell Chemical Biology 2017, 24, 1368.
[74] Y. Tian, S. Wang, H. Shang, M. Wang, G. Sun, X. Xu, X. Sun, Rsc Advances 2017,
7, 6259.
[75] S. Kadasi, T. E. M. M. Costa, N. Arukala, M. Toshakani, C. Duggineti, S. Thota, S. D.
Gupta, S. Raj, C. Penido, M. G. Henriques, N. M. Raghavendra, Medicinal Chemistry 2018,
14, 44.

This article is protected by copyright. All rights reserved.


32
www.clinical.proteomics-journal.com Page 33 Proteomics - Clinical Applications

Figure Legends

Figure 1. Areas of application for proteomics generally cover the discovery of potential drug

targets, investigations into mechanisms of drug action and protein-protein interactions, and

the discovery and application of stratification biomarkers.

This article is protected by copyright. All rights reserved.


33
www.clinical.proteomics-journal.com Page 34 Proteomics - Clinical Applications

Figure 2. Knowledge at the molecular level and pathology can improve on the drug

discovery and development process.

This article is protected by copyright. All rights reserved.


34
www.clinical.proteomics-journal.com Page 35 Proteomics - Clinical Applications

Figure 3. Envisioned clinical setting where proteomics can guide drug selection and

intervention.

This article is protected by copyright. All rights reserved.


35
www.clinical.proteomics-journal.com Page 36 Proteomics - Clinical Applications

Supplementary Table 1. List of the 1625 studies, as retrieved by the literature search.

Harald Mischak

Harald Mischak is the director of Mosaiques diagnostics, which he founded in 2002. In


addition, he is a Professor at the University of Glasgow. He received his PhD from the
Technical University of Vienna and has been involved in proteomics during the last 30 years,
with focus on deciphering signaling processes and identifying biomarkers and therapeutic
targets. He initiated the use of urinary proteomics and capillary electrophoresis coupled
mass spectrometry for clinical application, and is a leading authority in clinical proteomics
and biomarker identification, with over 300 scientific publications on signaling and
proteomics that have been cited over 20000 times. Among his achievements in this field are
the development of guidelines for clinical proteome analysis, the demonstration of successful
application of clinical proteomics in the diagnosis and prognosis of several diseases, and the
initiation of proteomics-guided randomized controlled clinical trials.

This article is protected by copyright. All rights reserved.


36
www.clinical.proteomics-journal.com Page 37 Proteomics - Clinical Applications

Maria Frantzi

Maria Frantzi received a MSc (2012) and a PhD (2016) in Molecular Medicine from Athens
Medical School. During her PhD studies, she received a Marie Skłodowska-Curie fellowship,
during which she shared her time between University of Glasgow (UK), Biomedical
Research Foundation of Academy of Athens (BRFAA, Greece) and Mosaiques diagnostics
GmbH (Germany). She currently works in Mosaiques diagnostics GmbH with main research
focus on proteomics, cancer diagnostic and stratification biomarkers. Through a second
post-doctoral Marie Skłodowska-Curie fellowship, she has expanded her research towards
personalized medicine and -omics data integration, through the application of proteomics
technologies and system biology approaches for investigating disease pathophysiology and
molecularly driven drug targets.

This article is protected by copyright. All rights reserved.


37
www.clinical.proteomics-journal.com Page 38 Proteomics - Clinical Applications

Agnieszka Latosinska

Agnieszka Latosinska graduated from Jagiellonian University, Poland in 2012, and received
a PhD from the Charité-Universitätsmedizin Berlin, Germany in 2016. In parallel, she worked
as an Early Stage Researcher in the Marie Curie BCMolMed (Molecular Medicine for
Bladder Cancer) EID Program. During the programme she was performing research in the
proteomic laboratory in the Biomedical Research Foundation of the Academy of Athens
(Greece) and in Mosaiques Diagnostics in Hannover (Germany). She is currently holding a
Marie Skłodowska-Curie Individual Fellowship at Mosaiques Diagnostics, with the main line
of research focused on the identification of therapeutic agents and drug targets through the
knowledge gained based on molecular characterization of Prostate Cancer.

This article is protected by copyright. All rights reserved.


38

You might also like