Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Osteoarthritis and Cartilage 31 (2023) 1437–1453

Osteoarthritis year in review 2023: metabolite and protein biomarkers


Ming Liu, Nafiza Haque, Jingyi Huang, Guangju Zhai
⁎ ]]
]]]]]]
]]

Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Canada

a r t i c l e i n f o s u m m a r y

Article history: Objective: To highlight the advances over the past year in metabolite/protein biomarkers for osteoarthritis
Received 23 May 2023 (OA).
Accepted 14 August 2023 Method: A literature search of five databases including PubMed, Web of Science, Scopus, Ovid Medline, and
Embase was performed for studies on metabolite/protein/peptide/biochemical markers for OA published
Keywords: between April 1st, 2022 and March 31st, 2023. Records were then screened to include only original research
Osteoarthritis
articles using directly collected human specimens, in English language, and with full text available. Data
Biomarker
from eligible studies were systematically extracted and summarized.
Metabolite
Protein
Results: A total of 1600 unique records were extracted, out of which 46 fulfilled the inclusion criteria
and were used for data extraction. Forty-one of these 46 studies focused on biomarkers for OA/OA
severity/progression, four on OA clustering, and one on OA treatment outcomes. Twenty-nine studied
protein markers for OA, thirteen studied metabolite markers, and four studied both. While many
studies were the validation of the previously reported biomarkers, a number of novel metabolite/
protein biomarkers and biomarker panels were reported in the past year. Biomarker panels might be
useful to subset OA patients.
Conclusion: The number of studies on OA clustering is rising. Although validation in larger cohorts is needed
in order to utilize reported biomarkers in clinical practice, these discoveries help better understand the
pathogenesis of OA, provide insights into possible mechanisms underlying poor treatment outcomes, and
aid in developing personalized treatment based on OA subtypes.
© 2023 The Author(s). Published by Elsevier Ltd on behalf of Osteoarthritis Research Society International.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-
nd/4.0/).

Introduction the molecular alterations preceding the structural changes or


provide therapeutic targets, thus, there is a pressing need for
Osteoarthritis (OA) is a heterogeneous disorder characterized meaningful diagnostic and prognostic biochemical markers that
by multi-tissue failure in diarthrodial joints, and has been pro­ can be easily measured and can help understand the pathogen­
posed to be a complex syndrome 1 and a whole person disease 2 esis of the disease and mechanisms underlying poor treatment
due to its complexity as seen in the variable etiologies and outcomes, and in the clinical setting, assist in early diagnosis,
clinical presentation, discordance between structural signs and prediction of disease progression, and stratification of patients
symptoms, 3 and different responses to treatment,4 all of which for clinical trials, which eventually aids in the development of
pose significant challenges for OA prevention, diagnosis, and disease modifying OA drugs and precision medicine in OA. In this
treatment development. Biomarkers are objective, quantifiable review, we aimed to highlight the advances over the past year in
characteristics of biological processes that can be measured re­ metabolite and protein biomarkers for OA risk and progression,
liability with sufficient precision and provide summary of bio­ OA clustering, and OA treatment outcomes.
logical, physiological, or pathological pathways associated with
the conditions examined.5,6 In the OA field, the only biomarkers
currently in use are the imaging markers which can not detect Methods

This review was conducted and reported in accordance with the


* Correspondence to: Division of Biomedical Sciences (Genetics), Faculty of Preferred Reporting Items for Systematic Review and Meta-
Medicine, Memorial University of Newfoundland, St. John’s A1B 3V6, Canada. Analyses7 (Supplementary Table 1).
E-mail address: guangju.zhai@med.mun.ca (G. Zhai).

https://doi.org/10.1016/j.joca.2023.08.005
1063-4584/© 2023 The Author(s). Published by Elsevier Ltd on behalf of Osteoarthritis Research Society International. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
1438 M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

Database Search query Filter/limit

PubMed ((osteoarthriti*) AND ((biomarker*) OR (proteomic*) OR (metabolom*) English;


OR ((( marker*) OR (predict*) OR (associat*)) AND ((biochemical) OR Full text;
(metaboli*) OR (peptide*) OR (protein))))) Date of publication=April 1st, 2022-March 31st, 2023
Web of Science TS=((osteoarthriti*) AND ((biomarker*) OR (proteomic*) OR English;
(metabolom*) OR (((marker*) OR (predict*) OR (associat*)) AND Document type=article/proceeding paper/early access;
((biochemical) OR (metaboli*) OR (peptide*) OR (protein))))) Date of publication=April 1st, 2022-March 31st, 2023
Scopus (TITLE-ABS-KEY ((osteoarthriti*) AND ((biomarker*) OR (proteomic*) OR English;
(metabolom*) OR (((marker*) OR (predict*) OR (associat*)) AND Document type=article/letter;
((biochemical) OR (metaboli*) OR (peptide*) OR (protein)))))) Subject=medicine/biochemistry, genetics and molecular biology/
immunology and microbiology/pharmacology, toxicology and
pharmaceutics/ health professions/ multidisciplinary;
Date of publication=April 1st, 2022-March 31st, 2023
Ovid Medline (exp osteoarthritis/ OR osteoarthrit*.mp.) AND (exp proteomics/ OR exp English;
metabolomics/ OR exp biomarker/ OR (proteomic* OR metabolom* OR Full text;
biomarker*).mp. OR ((exp peptides/ OR (peptide* OR protein* OR Year of publication=2022–2023#
biochemical OR metaboli*).mp.) AND (marker* OR predict* OR
associat*).mp.))
Embase ((’osteoarthritis’/exp OR osteoarthriti*:ab,ti) AND (’proteomics’/exp OR English;
’metabolomics’/exp OR ’biomarker’/exp OR proteomic*:ab,ti OR Document type=article/article in press/conference paper/data paper/letter
metabolom*:ab,ti OR biomarker*:ab,ti OR ((’peptides’/exp OR /preprint;
peptide*:ab,ti OR protein*:ab,ti OR biochemical:ab,ti OR metaboli*:ab,ti) Year of publication=2022–2023#
AND (marker*:ab,ti OR predict*:ab,ti OR associat*:ab,ti))))
#
: Date of publication filter does not apply, hence records prior to April 1st, 2022 or after March 31st, 2023 were removed after literature search.

Table 1

Literature search queries and filters.

Search strategy assessed using a narrative analysis11 (supplementary methods,


Supplementary Table 2).
Five databases including PubMed, Web of Science, Scopus, Ovid
Medline, and Embase were searched for publications on OA metabolite/ Results
protein biomarkers published between April 1st, 2022 and March 31st,
2023. The key phrases used for the search were: osteoarthritis/osteoar­ The aforementioned search strategy identified a total of 3421
thritic+biomarker(s)/proteomic(s)/metabolomic(s)/metabolome; os­ records, out of which 1134 were from PubMed, 594 from Web of
teoarthritis/osteoarthritic+metabolism/metabolic/metabolite(s)/bio­ Science, 738 from Scopus, 97 from Ovid Medline, and 858 from
chemical/peptide(s)/protein+marker(s)/predictor(s)/predict(s/ed/ing/ Embase. After duplicate removal and abstract screening, full text of
ion)/associat(s/ed/ing/ion). The search queries and filters are presented the remaining 187 studies were accessed, and 141 studies were
in Table 1. further excluded based on the exclusion criteria. 46 studies were
included in this review. The detailed screening process is presented
Study selection in Fig. 1.

After the removal of duplicated records, abstract and full text Study Characteristics
screening was conducted according to the following inclusion and
exclusion criteria: Characteristics of each study including article first author, the
approach(es) used to measure biomarker levels, phenotype(s) of
• Inclusion criterion: original research article focusing on meta­ study group(s), sample size(s), disease joint(s), and sample type(s)
bolite/protein/peptide/biochemical biomarkers for OA in any were extracted and summarized during full text review and are
joint in human. presented in Table 2. Among the 46 studies, 34 studied OA in knee
• Exclusion criteria: no full text; not in English; pure computa­ joint, one studied hand, three studied temporomandibular joint
tional studies; review articles or meta-analyses; not population- (TMJ), two studied spine, four studied multiple joints (knee, hip,
based study; not OA-focused; not metabolite/protein biomarker- hand), and two did not specify joint studied. Blood sample was used
focused; biomarker level did not differ between controls and OA in 29 studies, blood and urine were used in five studies, synovial
patients examined; sample size < 10 in any study group without fluid (SF)/synovial tissue (ST) was used in five studies, cartilage was
an independent replication; data inconsistency throughout the used in one study, and blood and SF/ST/cartilage were used in six
article (discrepancies between the results described in the text studies. The majority of the studies employed targeted approach
and the results presented in the table and/or figure). (n = 43), two studies applied untargeted approach, and one study
used both.
Study risk of bias assessment
Risk of bias in studies
The methodological quality of the included studies was assessed
using different tools according to study types (cross-sectional, The quality assessment results are presented in Supplementary
longitudinal, case-control),8–10 and OA clustering studies were results and Supplementary Table 3.
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453 1439

Fig. 1
PRISMA flow diagram - search strategy and selection process of the included studies. PRISMA: Preferred Reporting Items for Systematic
Review and Meta-Analyses.

OA biomarkers as a biomarker for OA, these new studies all found a small effect size
and did not add much convincing evidence.
Forty-one out of these 46 articles studied metabolite/protein C-terminal cross-linked telopeptide of type II collagen (CTX-II) is
biomarkers for OA/OA severity/progression, four studied biomarkers a byproduct of articular cartilage breakdown. Liem et al.16 found that
for OA clustering, and one studied biomarkers for OA treatment higher values of baseline urinary CTX-II were associated with an
outcomes. Results for individual studies are present in Tables 3 increased risk of OA disease progression. Lu et al.17 reported that
and 4. serum CTX-II level was higher in traumatic KOA patients than in
controls, and higher in Kellgren and Lawrence (KL) grade III+IV than
Discussion in KL grade I+II patients. Studies on CTX-II as an OA biomarker have
consistently shown that urinary CTX-II had a potential to be a pro­
Biomarkers for OA/OA severity/progression tein biomarker for OA and OA progression.18–20 The two new studies
provided further evidence, though the predictive power of this
Biomarkers for OA/OA severity/progression reported in the past protein needs to be evaluated in larger cohorts and within different
year mainly belong to at least one of the following categories: car­ subgroups as the performance of this biomarker differs between
tilage/bone structure/damage, inflammation, metabolism, or oxida­ different age and sex groups and different populations.19–21 CTX-I
tive stress markers. The majority of the identified biomarkers were was reported in one study15 to be upregulated in KOA, had higher
previously reported. levels in KL grade II than in KL grade I patients, and could distinguish
KOA patients from controls and between different severity groups.
Cartilage/bone structure/damage biomarkers This study had a small sample size and significantly different age and
Cartilage oligomeric matrix protein (COMP) is a large pentameric sex distribution between groups, its findings need to be validated.
glycoprotein that plays an important role in mediating the matrix CTX-I was not as well studied in regard to its association with OA and
molecule interactions needed to organize the cartilage matrix.12 there has not been any strong evidence suggesting its potential as an
Findings from three new studies13–15 showed that serum COMP level OA biomarker previously.
was higher in knee OA (KOA) than in controls and could distinguish Cartilage acidic protein 1 (CRTAC1) was highlighted in four stu­
these two groups; it was positively associated with overall OA dies to be positively associated with OA risk, severity, and overall OA
burden and negatively associated with medial femorotibial com­ burden, as well as a predictor for incident diagnosis and progression
partment cartilage thickness at 4-year follow-up. Although some of OA,13,22–24 but the effect sizes and predictive power for overall OA
evidence has shown that COMP has diagnostic and prognostic value burden, severity, and progression were all small. In a study with
1440

Reference Approach Phenotype and sample size Joint Tissue type

Abou-Raya et al.43 Targeted Early KOA (n = 40), severe KOA (n = 10), non-OA (n = 40) Knee Serum
Amirkhizi et al.38 Targeted Vitamin-D-sufficient KOA(n = 60), vitamin-D-insufficient KOA (n = 64), non-OA (n = 65) Knee Serum
Angelini et al.74 Targeted KOA (n = 297) Knee Serum, urine
Cambon-Binder et al.59 Targeted Erosive hand OA (n = 168), non-erosive hand OA (n = 218); OA PIPs (n = 7), non-OA PIPs (n = 7); OA MCPs (n = 5), non-OA MCPs Hand Serum, cartilage
(n = 9)
Cheng et al.51 Targeted KOA of different disease severities (n = 50) Knee Plasma, SF
Chiba et al.54 Targeted KOA with CS (n = 48), KOA without CS (n = 249) Knee Serum
Costello et al.4 Targeted Discovery: KOA with sustained pain post TKR (n = 35), KOA without sustained pain post TKR (n = 323); replication: KOA with Knee Plasma
sustained pain post TKR (n = 118), KOA without sustained pain post TKR (n = 118)
Drvar et al.67 Targeted KOA (n = 40), non-OA (n = 40) Knee PBMCs
Elamir et al.27 Targeted KOA (n = 100), non-OA (n = 80) Knee Serum
Feng et al.34 Targeted DDw/oR with early-stage OA (n = 15), DDw/oR with late-stage OA (n = 13), DDw/oR without OA (n = 14) TMJ SF
Goode et al.75 Targeted Patients with different lumbar spine radiographic features with and without LBP (n = 731) Spine Serum, urine
Hunter et al.32 Targeted KOA with radiographic and pain progression (n = 194) and KOA with only radiographic or pain progression or no progression at 48 Knee Serum, urine
months (n = 406); KOA with radiographic progression (n = 297) and KOA without radiographic progression (n = 303) at 48 months
Hussain et al.15 Targeted KOA of different severities (n = 100), non-OA (n = 50) Knee Serum
Jitjumnong et al.60 Targeted LDD of different severities (n = 50), non-LDD (n = 50) Spine Plasma
Kamel et al.44 Targeted KOA (n = 70), non-OA (n = 30) Knee Serum, SF
Liang et al.65 Targeted OA (n = 206), non-OA (n = 60) Not specified Plasma
Liem et al.16 Targeted KOA with different radiographic or clinical features (n = 600) Knee Serum, urine
Loef et al.53 Targeted/ untargeted KOA and hand OA with different severities (n = 216) Knee, hand Plasma
Lu et al.17 Targeted KL grade I+II traumatic KOA (n = 87), KL grade III+IV traumatic KOA (n = 83), non-OA (n = 80) Knee Serum
Nemet et al.57 Targeted KOA (n = 25), non-OA (n = 19) Knee Blood
Papaneophytou et al.33 Targeted Early OA (n = 44), late OA of different severities (n = 130), overweight non-OA (n = 38) Knee Serum
Perruccio et al.42 Targeted OA (n = 3049), non-OA (n = 3049); female OA (n = 1702), male OA (n = 1348); multisite OA (n = 25), single-site OA (n = 3024) knees, hips, hands Serum
Rifal et al.41 Targeted KOA of different severities with hyperuricemia (n = 38), KOA of different severities with normal uric acid levels (n = 36) Knee Serum
Rockel et al.76 Targeted KOA (n = 214) Knee Plasma
Rong et al.66 Targeted KOA (n = 41), meniscal injury patients (n = 20) Knee Synovium, SF
Sarkar et al.62 Untargeted KOA (n = 60), non-OA (n = 40) Knee Plasma, SF
Sasaki et al.70 Targeted KOA with different KL grades and synovitis scores (n = 597) Knee Plasma, serum
Selim et al.58 Targeted KOA (n = 40), non-OA (n = 20) Knee Serum
Shao et al.46 Targeted TMJOA (n = 30), TMJID (n = 30) TMJ SF
Shekhar et al.61 Targeted KOA (n = 49), non-OA (n = 49) Knee Serum, SF
Singh et al.14 Targeted Participants with data on MRI imaging markers and biochemical markers (n = 119 and 155 for baseline, n = 127 and 129 for Knee Serum
follow-up)
Singh et al.26 Targeted KOA (n = 80), non-OA (n = 80) Knee Serum
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

Sodhi et al.52 Targeted KOA with different synovial vascular pathology features (n = 97) Knee SF
Styrkarsdottir et al.22 Targeted Knee/hip/hand OA (n = 3330), non-OA (n = 50,935); incident knee/hip/hand OA (n = 6619) and non-OA during follow-up Knee, hip Plasma
(n = 44,316); knee/hip OA who progressed to having TJR (n = 607), knee/hip OA who did not progress to having TJR (n = 671)
Szilagyi et al.13 Targeted OA (n = 426), non-OA (n = 1847); OA of different severities (n = 3103); KOA progressor (n = 198) and non-progressor (n = 1767); HOA Knee, hip, hand Plasma
progressor (n = 127) and non-progressor (n = 1871)
23
Tardif et al. Untargeted Discovery: obese KOA (n = 10), non-obese KOA (n = 10), non-OA (n = 8); replication: obese OA (n = 10), non-obese KOA (n = 10), non- Knee Serum, plasma
OA (n = 20)
37
Torga et al. Targeted KOA of different OARSI grades (n = 20) Knee Cartilage
Trajerova et al.50 Targeted KOA patients with improved, unchanged, and worsened clinical trajectories (n = 119) Knee SF
Tudorachi et al.56 Targeted Mild KOA (n = 39), severe KOA (n = 17), non-OA (n = 29) Knee Blood
Ukibe et al.40 Targeted Premenopausal/HIV/OA (n = 25), premenopausal/HIV/non-OA (n = 21), postmenopausal/HIV/OA (n = 21), postmenopausal/HIV/ Not specified Serum
non-OA (n = 21)
Ungsudechachai et al.45 Targeted KOA without synovitis (n = 16), KOA with low-grade synovitis (n = 19), KOA with high-grade synovitis (n = 15) Knee Plasma, ST, SF
Watanabe et al.35 Targeted DJD-TMJ (n = 17), non-DJD-TMJ (n = 17); younger DJD-TMJ (n = 10) and non-DJD-TMJ (n = 11); older DJD-TMJ (n = 7) and non-DJD- TMJ Serum, urine
TMJ (n = 6)
Xie et al.68 Targeted Participants with different rates of cartilage volume reduction (n = 344) Knee Serum
Zertuche et al.69 Targeted Incident radiographic OA (n = 258), non-OA (n = 516); incident symptomatic OA (n = 260), non-OA (n = 518) Knee Plasma
Zheng et al.55 Targeted Participants who developed OA at 5-year follow-up (n = 1089), non-OA at 5-year follow-up (n = 9641) Knee Plasma, whole blood
(continued on next page)
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453 1441

small sample sizes, CRTAC1 was found to be the most significant

peripheral blood mononuclear cells; SF: synovial fluid; ST: synovial tissue; TMJ: temporomandibular joint; TJR: total joint replacement; TMJID: temporomandibular joint internal derangement; TMJOA: temporomandibular joint
CS: central sensitization; DDw/oR: disc displacement without reduction; DJD-TMJ: degenerative joint disease of the temporomandibular joints; HIV: human immunodeficiency virus; HOA: hip osteoarthritis; JSL: joint space loss;
KL: Kellgren and Lawrence; KOA: knee osteoarthritis; LBP: lower back pain; LDD: lumbar disc degeneration; MRI: magnetic resonance imaging; OA: osteoarthritis; OARSI: osteoarthritis research society international; PBMC:
contributor among a set of 9 proteins discriminating OA from con­
trols.29 CRTAC1 is a glycosylated extracellular matrix protein that
was initially identified as a marker of chondrocyte development
Tissue type

from mesenchymal stem cells.25 CRTAC1 as a potential OA biomarker


was only discovered recently and relevant studies are very limited.
Serum

Extended research on this protein is much needed before it can be


utilized as an OA biomarker.
Matrix metallopeptidases (MMPs) are also proteins playing im­
portant roles in cartilage integrity. Three studies focused on MMP-3
in KOA14,26,27 and found that serum MMP-3 level was higher in KOA
especially in patients of higher KL grades, could distinguish between
Discovery: KOA JSL and pain progressor (n = 192), composite comparator (JSL and pain non-progressor+JSL or pain progressor only, Knee
Joint

KOA and controls and different severity groups with moderate ac­
curacy. Its level was also positively correlated with Western Ontario
and McMaster Universities Osteoarthritis Index (WOMAC) score,
patellar cartilage volume loss and total bone area reduction after 4
n = 404); JSL progressor (n = 103), JSL and pain non-progressor (n = 199). Replication: KOA JSL progressor (n = 37), JSL non-

years. Levels of MMP-1 and -13 were also found to be higher in


KOA.34 MMPs are the major proteases involved in extracellular ma­
trix degradation,28 a process that plays a significant role in the pa­
thogenesis of OA. MMP-3 and -13 levels increase in response to axial
compression and degradation of cartilage matrix, MMP-3 then pro­
motes synovitis and synovitis promotes the expression of MMP-3 in
return, forming a vicious cycle in the progression of OA.29 MMP-13
preferentially digests type II collagen,28 the main component of
cartilage, also degrades the proteoglycan molecule aggrecan, giving
it a dual role in matrix destruction,30 therefore, it is thought to be
the most important MMP in OA pathogenesis and an attractive
target for inhibitor development in the treatment of OA. MMP-1
protein expression in OA is not well studied in humans, but there has
been evidence that MMP-1 level is upregulated in SF of OA pa­
tients.31 The three new studies validated that MMPs have potential
to be OA biomarkers.
A number of other previously discovered cartilage/bone struc­
ture/damage biomarkers were also reported in the past year in­
cluding N-telopeptide of type I collagen (NTX-I), procollagen type II
C-terminal propeptide (PIICP), C-C chemokine receptor type 5
(CCL5), cartilage intermediate layer protein 2 (CLIP-2), and vitamin
D. Hunter et al.32 reported that adding baseline serum NTX-I or
change of urine NTX-I level over 24 months to respective imaging
marker models slightly increased the predictive power for OA
radiographic and pain progression over 48 months. Papaneophytou
et al.33 found that serum PIICP level was decreased in late OA and
Phenotype and sample size

negatively correlated with OA severity and knee pain, and can dis­
criminate late OA patients from overweight individuals without OA.
In two studies on temporomandibular joint OA (TMJOA),34,35 SF CCL5
progressor (n = 49)

level was found to be increased in TMJ disc displacement without


reduction (DDw/oR) patients with early-stage OA than in patients
without OA or with late-stage OA, and can distinguish DDw/oR pa­
tients with early-stage OA from other DDw/oR patients. Serum CCL5
level was also higher in female patients with degenerative joint
disease of the TMJ (DJD-TMJ) than in female controls, and was po­
Characteristics of the included studies.

sitively associated with bone metabolism rate, especially in rela­


osteoarthritis; TKR: total knee replacement.

tively young DJD-TMJ patients without other systemic symptoms,


and this level could distinguish DJD-TMJ from non-DJD-TMJ in fe­
Approach

Targeted

males. However, the diagnostic performance of this protein in male


patients needs further investigation. CCL5 is a chemokine that plays
a role in the recruitment of T helper cells to the affected joint to help
trigger inflammation processes via the release of pro-inflammatory
cytokines.36 The results of these two studies suggested that serum or
Table 2 (continued)

SF CCL5 level has the potential to be a biomarker for early detection


Zhou et al.24

of TMJOA especially in younger patients. Torga et al.37 reported that


Table 2
Reference

in end-stage KOA patients, the immunohistochemistry staining in­


tensity of CILP-2 in cartilage was positively associated with Os­
teoarthritis Research Society International (OARSI) grading. Again,
the main drawback of these studies are the small sample sizes.
1442

Reference # of markers Identified Biomarker (s) Findings Estimates Biomarker Biomarker


tested application type
(identified)

Abou-Raya et al.43 2 (1) IL-8 Serum IL-8 ↑ in early KOA than in controls. OA vs. controls: 162.90 ± 145.50 vs. OA stage Protein
139.15 ± 39.64, p = 0.027.
Amirkhizi et al.38 1 (1) Vitamin D Vitamin D ↓ in KOA with insufficient Vitamin D OA vs. controls: 19.7 ± 5.5 vs. 42.7 ± 9.6 ng/ml, OA/OA severity Metabolite
and inversely correlated with inflammatory p < 0.0001. Correlation with IL-1β, TNF-α, and
factors. WOMAC scores ↑ in KOA with vitamin D hs-CRP: β = −0.51, −0.44, −0.50, all p < 0.0001.
insufficiency. WOMAC: p ≤ 0.011.
Angelini et al.74 16 (16) Combination of serum C2M, C3M, COMP, Three OA clusters: C1: low tissue turnover; F1 scores: discovery cohort: C1: 0.85, C2: 0.91, OA clustering Protein +
CRPM, ARGS, C10C, COLL2_1, COLL2_1NO2, C2: structural damage; C3: systemic C3: 0.88; validation cohort: no data. Metabolite
CTXI, hyaluronic acid, hsCRP, PRO_C2, NMID, inflammation. Validated in an external cohort.
RE_C1M, urine CTXI_ALPHA and CTXII.
Cambon-Binder 2 (1) CML Serum CML ↓ in erosive than in non-erosive Erosive vs. non-erosive hand OA: 178.7 OA/OA severity Metabolite
et al.59 hand OA. Cartilage CML ↑ in hand OA than in (157.1–208.8) vs. 194.7 (168.9–217.1) μmol/mol
non-OA. lysine, p = 0.01. OA vs. non-OA: 7 vs. 4 mmol/mol
lysine, p = 0.01.
Cheng et al.51 2 (2) C3-α and C3-β SF C3-α and C3-β levels were correlated with Correlation with severity: r = 0.39 and 0.47, both OA severity Protein
KOA severity and ↑ in OARSI grade ≥ 6 group. p < 0.01. SF level in OARSI ≥6 vs. < 6 groups:
C3-α: 1131 ± 69 vs. 939 ± 47 unit/ml; C3-β:
1011 ± 63 vs. 783 ± 51 unit/ml; both p < 0.05.
Chiba et al.54 1 (1) Glucose In KOA patients with CS, glucose was Association with KOOS pain: β = −0.448 OA severity Metabolite
negatively associated with changes in KOOS (−0.737,−0.158); p = 0.003; ADLs: β = −0.438
pain, ADLs, and sports scores from baseline to (−0.713, −0.163); p = 0.003; sports scores:
1-year follow-up. β = −0.706 (−1.21, −0.202); p = 0.007.
Costello et al.4 18,769 (4) PC aa C28:1, PC aa C32:0/PC aa C28:1, PC aa Plasma PC aa C28:1 and PC aa C28:1/PC aa PC aa C28:1: OR=0.66 (0.52–0.82); PC aa OA treatment Metabolite
C28:1/PC aa C32:0, and C14:2/SM C20:2 C32:0 were negatively associated with C28:1/PC aa C32:0: OR=0.6 (0.48–0.76); PC aa outcome
sustained pain after TKR, PC aa C32:0/PC aa C32:0/PC aa C28:1: OR=1.59 (1.29–1.96);
C28:1 and C14:2/SM C20:2 were positively C14:2/SM C20:2: OR=1.59 (1.29–1.96).
associated.
Drvar et al.67 1 (1) GNLY In female, GNLY labeling in PBL ↑ in OA than in OA vs. controls: p = 1 × 10-6. OA Protein
controls.
Elamir et al.27 3 (3) MMP-1, MMP-3, MMP-13 Serum MMP-1, MMP-3, and MMP-13 ↑ in KOA OA vs. controls: MMP-1: 2.89 ± 0.01 vs. OA Protein
than in controls and could diagnose OA. 1.15 ± 0.04; MMP-3: 2.37 ± 0.07 vs. 0.79 ± 0.01;
MMP-13:2.56 ± 0.02 vs. 1.02 ± 0.08; all p < 0.01.
Diagnostic performance: 100% specificity and a
sensitivity of 98%, 100%, and 100%, respectively,
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

from a model fit, no AUC data.


Feng et al.34 1 (1) RANTES (CCL5) RANTES ↑ in DDw/oR with early-stage TMJOA, DDw/oR with early-stage TMJOA vs. other DDw/oR OA stage Protein
can distinguish these patients from other patients: p < 0.05. AUC=0.798, from a model fit, no
DDw/oR patients. sensitivity/specificity data.
Goode et al.75 12 (10) Combination of N-cadherin, lumican, CTX-II, Three OA clusters: C1: degenerative changes; Variance explained by the principal OA clustering Protein +
OPG, RANTES, IL-6, CXCL6, hyaluronic acid, C2: ↓ biomarkers, pain, and degenerative components=48%, CCC=7.76. Metabolite
NPY, BDNF changes; C3: degenerative changes and
inflammation with pain.
Hunter et al.32 1 (1) NTX-1 For baseline and 24-month change in Imaging+NTX-1 vs. imaging: baseline markers: OA progression Protein
markers, adding NTX-I to imaging marker AUC=0.671 vs. 0.669; 24-month change in markers:
model increased the power for predicting AUC=0.724 vs. 0.713, from individual model fit.
progression.
(continued on next page)
Table 3 (continued)

Reference # of markers Identified Biomarker (s) Findings Estimates Biomarker Biomarker


tested application type
(identified)

Hussain et al.15 2 (2) COMP, CTX-I Serum COMP and CTX-I ↑ in KOA than in OA vs. controls: COMP: 17.38 ± 4.99 vs. OA/OA severity Protein
controls, and could distinguish KOA from 1.16 ± 0.39 U/L; CTX-I: 4.59 ± 1.69 ng/ml vs.
controls. 0.35 ± 0.14 ng/ml; both p = 0.001. AUC=1,
specificity=100%, sensitivity=99% and 100%,
respectively, p = 0.001 and 0.02, respectively,
from individual model fit.
Jitjumnong et al.60 2 (2) MDA, 8-OHdG Plasma MDA and 8-OHdG ↑ in LDD than in LDD vs. controls: MDA: p = 0.01; 8-OHdG: 179.28 OA/OA severity Metabolite
controls, and positively correlated with LDD (101.88–247.55) vs. 115.76 (77.12–164.79) ng/L,
severity. p = 0.002. Correlation with severity: MDA: r = 0.49,
p = 0.001; 8-OHdG: r = 0.60, p < 0.001.
Kamel et al.44 1 (1) IL-17A In KOA, serum IL-17A ↑ than in controls and OA vs. controls: 11.6 ± 1.6 vs. 10.5 ± 1.9 pg/ml, OA/OA severity Protein
positively correlated with SF IL-17A. Both IL- p < 0.05. Correlation between serum and SF IL-
17A levels positively correlated with VAS, 17A: r = 0.34, p = 0.04. Correlation with disease
WOMAC scores, Lequesne scores, and parameters: serum: r: 0.35–0.42, p: 0.01–0.02;
Lequesne index. SF IL-17A positively SF: r: 0.78–0.92, p < 0.001; KL grade: r = 0.93,
correlated with KL grade and OA duration. p < 0.001; OA duration: r = 0.66, p < 0.001.
Liang et al.65 2 (1) mCRP Plasma mCRP level ↑ in OA than in controls, OA vs. controls: 12.51 (7.78–24.81) vs. 5.04 OA/OA severity Protein
and ↑ in KL grade IV than in KL grade III KOA, (3.45–9.77) ng/ml, p = 3.72x10-5; KL grade IV vs.
can discriminate OA from controls. III: p < 0.05. AUC=0.76, from a model fit, no
sensitivity/specificity data.
Liem et al.16 13 (3) CTX-II Baseline uCTXII was associated with KL grade, KL grade: OR=1.15 (1.03–1.28); medial JSN: OA progression Protein
medial JSN, lateral osteophytes, joint space OR=1.06 (1.02–1.1); lateral osteophytes: OR=1.05
width, and WOMAC pain scores. (1.01–1.1); joint space width: β = −0.005
(−0.008,−0.001); WOMAC pain scores: OR=1.02
(1.01–1.04).
Loef et al.53 631 (5) Fatty acids 20:4, 16:1, 18:1, 18:2, 22:5 KOA and hand OA were associated with the Radiographic KOA: R2lipidomics = 0.03; radiographic OA severity Metabolite
lipidomic fraction of bound and free FA 20:4, hand OA: R2lipidomics = 0.02; hand function:
bound FA 16:1, FA 18:1, FA 18:2, and FA 22:5. R2lipidomics = 0.06; hand pain: R2lipidomics = 0.12.
Lu et al.17 3 (3) TLR4, CXCL12, CTX-II Serum TLR4, CXCL12, and CTX-II ↑ in OA vs. controls: TLR4: 13.87 ± 1.99 vs. OA/OA severity Protein
traumatic KOA than in controls, and ↑ in KL 9.01 ± 2.21 ng/ml; CXCL12: 9.27 ± 1.47 vs.
grade III+IV than in KL grade I+II group. 3.9 ± 0.96 ng/ml; CTX-II: 707.67 ± 119.17 vs.
341.18 ± 84.44 ng/L; all p < 0.001; KL III+IV vs. I+II
group: TLR4: 15.21 ± 1.54 vs. 12.58 ± 1.45 ng/ml;
CXCL12: 10.38 ± 1.05 vs. 8.21 ± 0.94 ng/ml, CTX-II:
801.45 ± 81.04 vs. 618.20 ± 71.12 ng/L; all
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

p < 0.001.
Nemet et al.57 5 (2) TC, LDL-C Prevalence of high TC and high LDL-C ↑ in OA vs. controls: high TC: 92% vs. 63.2%, p = 0.019; OA Metabolite
female KOA patients than in female controls. high LDL-C: 72% vs. 15.8%, p = 0.000.
Papaneophytou 3 (1) PIICP sPIICP ↓ in mild and severe LOA and negatively Mild-LOA:179.1 (148.4–278.9) pg/ml, severe-LOA: OA/OA severity Protein
et al.33 correlated with OA severity and knee pain at 177.6 (109.1–273.3) pg/ml, control: 703.4
rest and during walking, can discriminate LOA (593.0–922.8) pg/ml, p < 0.0001. Correlation:
from controls. severity: r = −0.68; pain: r = −0.44 for resting and
−0.42 for walking; all p < 0.0001. AUC=0.98
(0.945–0.995), sensitivity=96.2%, specificity=92.1%,
p < 0.0001, from a model fit.
(continued on next page)
1443
Table 3 (continued) 1444

Reference # of markers Identified Biomarker (s) Findings Estimates Biomarker Biomarker


tested application type
(identified)

Perruccio et al.42 5 (2) HbA1c, hsCRP Percentage of individuals with hsCRP and OA vs. controls: hsCRP: 66.4% vs. 61.9%; HbA1c: OA/OA severity Protein
HbA1c levels within published threshold for 22.5% vs. 17.7%, both p ≤ 0.001; multisite vs. single-
moderate/elevated CVD risk ↑ in OA and site OA: hsCRP: 72.1% vs. 64.5%; HbA1c: 25.2% vs.
multisite OA, with hsCRP being predominant 21.7%, both p < 0.05. Female OA with high hsCRP:
in women and HbA1c being predominant 68.6%, male OA with high HbA1c: 26.4%.
in men.
Rifal et al.41 1 (1) Uric acid Hyperuricemia group had ↑ percentage of Severe OA in hyperuricemia vs. normal uric OA severity Metabolite
severe KOA compared to normal uric acid acid group: 60.5% vs. 22.2%, p = 0.002.
group, had a ↑ risk of severe KOA. OR=5.36 (1.93–14.84).
Rockel et al.76 151 (24) Combination of C0, tyrosine, threonine, Two KOA clusters were identified based on AUC=0.921 (0.846–0.965), p < 0.0001, no OA clustering Metabolite
histidine, lysine, taurine, leucine, valine, profiles of 24 metabolites. sensitivity/specificity data, used internal
citurlline, spermidine, LPC 14:0, PC aa C36:0, validation, mean predicted accuracy=85.1%.
36:6, 40:1, 42:0, 42:2, PC ae C38:6, 42:0,
42:2, 42:3, 42:5, 44:3, 44:6, SM C24:0
Rong et al.66 1 (1) TDO2 In OA, synovium TDO2 ↑ and positively OA vs. controls: t = −4.78, p = 0.009. Correlation OA/OA severity Protein
correlated with IL-1β, TNF-α, and KL grades, with IL-1β: r = 0.367, p = 0.018; TNF-α: r = 0.519,
can discriminate OA from controls. p < 0.001; KL grades: r = 0.521, p < 0.001.
AUC=0.80 (0.689–0.911), sensitivity=63.4%,
specificity=85%, from a model fit.
Sarkar et al.62 192 (1) LRG1 Plasma LRG1 ↑ in OA patients than in controls, OA vs. controls: fold increase > 1.66, p ≤ 0.0001. OA Protein
↑ in TKR than in UKR. TKR vs. UKR: fold increase=1.66, p = 0.015.
Sasaki et al.70 82 (1) Cystine Cystine was associated with OA severity, Correlation: severity: r = 0.397, p < 0.001, OA severity Metabolite
effusion-synovitis, and radiographic OA. q < 0.001, effusion-synovitis: r = 0.151, p = 0.009,
q = 0.375. Low vs. high effusion-synovitis groups:
p = 0.039, q = 0.587. Association with radiographic
OA: AUC=0.714 (0.667, 0.761), p < 0.001,
from a model fit; OR=3.7.
Selim et al.58 1 (1) Adropin Serum adropin ↓ in KOA than in controls and ↓ OA vs. controls: 46.3 ± 27.9 vs. 76.5 ± 27.2 pg/ml, OA/OA severity Protein
in female than in male OA, negatively p < 0.0001; female vs. male OA: 40.2 ± 17.7 vs.
correlated with KL grading, could distinguish 75.2 ± 47.2 pg/ml, p = 0.0016. Correlation with KL
between KOA and controls. grades: r = −0.54, p = 0.0003. AUC=0.85,
sensitivity=80%, specificity=90%, p < 0.001, from a
model fit.
Shao et al.46 6 (1) HMGB1 SF HMGB1 ↑ in TMJOA than in TMJID, TMJOA vs. TMJID: p < 0.05. Correlation: VAS: OA/OA severity Protein
positively correlated with VAS, JFLS-8, and r = 0.5512, p = 0.0016; JFLS-8: r = 0.4684,
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

radiographic stage, could distinguish TMJID p = 0.0054; OA stage: r = 0.4923, p = 0.0057.


from TMJOA patients. AUC= 0.8344 (0.7186–0.9503), Youden’s
Index= 0.7667, p < 0.0001, from a model fit.
Shekhar et al.61 1 (1) MMP-17 Serum MMP-17 ↑ in OA than in controls and ↑ OA vs. controls: 15.43 ± 0.91 vs. OA/OA severity Protein
with disease severity, can distinguish OA from 14.71 ± 1.04 ng/ml, p < 0.001. KL grade IV vs. III
controls. Serum and SF MMP-17 were vs. II: serum: 16.06 ± 0.7 vs. 15.58 ± 0.27 vs.
associated with ↑ risk for advanced KOA. 14.76 ± 0.81 ng/ml, p < 0.001; SF: 5.96 ± 2.93
vs. 3.5 ± 1.79 vs. 2.3 ± 1.54 ng/ml, p = 0.001.
AUC=0.743 (0.646–0.841), sensitivity=75.5%,
specificity=71.4%, from a model fit. Association
with advanced KOA: serum: OR=11.333
(2.209–58.147), p = 0.004; SF: OR=27.556
(3.031–250.524), p = 0.003.
Singh et al.14 3 (2) COMP, MMP-3 Serum COMP was negatively associated with COMP for cartilage thickness: β = −0.070 mm OA severity/ Protein
medial femorotibial compartment cartilage (−0.138,−0.001), p = 0.045. MMP-3 for cartilage progression
thickness, and MMP-3 was negatively volume: β = −141.548 mm3 (−254.917,−28.179),
associated with patellar cartilage volume and p = 0.015; bone area: β = −0.729 mm2
total bone area after 4 years. (−1.340,−0.118), p = 0.02.
(continued on next page)
Table 3 (continued)

Reference # of markers Identified Biomarker (s) Findings Estimates Biomarker Biomarker


tested application type
(identified)

Singh et al.26 1 (1) MMP-3 Serum MMP-3 ↑ in OA than in controls, and OA vs. controls: 59.85 ± 39.92 vs. OA/OA severity Protein
positively correlated with KL grades, could 14.52 ± 6.09 ng/ml, p = 0.0001. Correlation with
distinguish between OA and control and KL grades: r = 0.68, p = 0.000. OA vs. controls:
different KL grades. AUC=73.04% (0.775–0.904), specificity 71.25%,
sensitivity 77.5%, p = 0.000; KL grade I vs. II:
AUC=70.69% (0.681–0.922), sensitivity 72.41%,
specificity 70.00%, p = 0.000; KL grade II vs. III:
AUC=54.42% (0.569–0.851), Specificity 44.83%,
Sensitivity 78.26%, p = 0.009; KL grade III vs.
IV: AUC=74.22% (0.524–0.991), specificity
73.91%, sensitivity 75%, p = 0.032; all from
individual model fit.
Sodhi et al.52 6 (1) C5 Among OA patients, ↑ SF C5 level was Vascularization score in males: β = −0.005 (−0.009, OA severity Protein
associated with ↓ vascularization scores in −0.0001) per 1 ng/ml C5 change, p = 0.04. sC5b-C9:
males. SF sC5b-C9 level ↑ in males than in male vs. female: p < 0.05; in males, high vs. low SF
females, and ↑ in males with high SF C5. C5: p < 0.05.
Styrkarsdottir et al.22 1462 (3) CRTAC1, ACAN, NCAN CRTAC1 was positively associated with CRTAC1: KOA: OR=1.34 (1.27–1.41), p = 5.5 × 10-29, OA/OA Protein
prevalent OA, predicted incident OA diagnosis HOA: OR=1.19 (1.11–1.28), p = 2.1 × 10-6, predict progression
and progression to joint replacement. KOA: HR=1.40 (1.35–1.46), p = 1.08 × 10-69, predict
Individuals in the highest quintile of risk HOA: HR=1.25 (1.19–1.31), p = 2.61 × 10-19,
based on CRTAC1 level, age, sex, and BMI had progression to knee replacement: HR= 1.20
a 9.8-fold risk of knee or hip OA. Adding ACAN (1.08–1.33), p = 5.2 × 10-4, hip replacement:
and NCAN to the model improved the HR= 1.22 (1.08–1.38), p = 1.4 × 10-3. CRTAC1 vs.
prediction of OA. CRTAC1+ACAN+NCAN model: AUCprevalent
KOA= 0.736 (0.724–0.749) vs. 0.742 (0.729–0.754),
AUCprevalent HOA= 0.738 (0.722–0.754) vs. 0.749
(0.733–0.765), AUCincident KOA= 0.718 (0.712–0.724)
vs. 0.722 (0.716–0.728), AUCincident HOA= 0.696
(0.689–0.703) vs. 0.705 (0.698–0.712). All from
individual model fit.
Szilagyi et al.13 171 (1) CRTAC1 CRTAC1 was positively associated with Overall OA: β = 0.09 (0.06–0.12), p = 3 × 10-8; hand OA/OA severity/ Protein
overall OA burden, hand OA and KOA OA severity: β = 0.03 (0.01–0.04), p = 1 × 10-3; KOA progression
severity and KOA progression. severity: β = 0.05 (0.03–0.07), p = 3 × 10-6; KOA
progression: β = 0.25 (0.09–0.41), p = 2 × 10-3.
Tardif et al.23 279 (4) CRTAC1, FBN1, VDBP, SERPINF1 A set of 9 proteins could discriminate OA from OA vs. controls: AUC > 95%, from a model fit. OA Protein
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

controls with CRTAC1 being the most CRTAC1: OA-obese vs. controls: p = 0.007,
significant contributor and ↑ in OA than in OA-non-obese vs. controls: p = 0.003. Protein
controls. FBN1, VDBP, SERPINF1 all ↑ in OA validation: FBN1: p = 0.044, VDPB: p = 0.022,
than in controls in the validation cohort. SERPINF1: p = 0.064.
Torga et al.37 2 (1) CILP-2 CILP-2 IHC staining intensity was positively Rho = 0.436, p = 0.005. OA severity Protein
associated with OARSI grading.
(continued on next page)
1445
Table 3 (continued) 1446

Reference # of markers Identified Biomarker (s) Findings Estimates Biomarker Biomarker


tested application type
(identified)

Trajerova et al.50 20 (3) CXCR4, CCR7, CXCL10 Low CXCR4 and CCR7 expression on Improved vs. worsened: Subset 1 (high expression OA clustering Protein
macrophages and high CXCL10 in SF were of CXCR4 and CCR7): 86.1% vs 95.5%; p = 0.009;
linked to improved clinical trajectory. Subset 2 and 3 (low expression of CXCR4 and
CCR7): 4.72% vs 1.70%; p = 0.024, 4.48% vs 0.24%;
p = 0.005. Improved vs. unchanged: Subset 1: 86.1%
vs 96.9%; p = 0.025; Subset 2: 4.72% vs 1.35%,
p = 0.042. CXCL10: improved vs. worsened: 339
(104–575) vs. 222 (2.96–441) pg/ml, unchanged vs.
worsened: 378 (115–640) vs. 222 (2.96–441) pg/
ml, both p = 0.002.
Tudorachi et al.56 5 (4) Glucose, TGs, LDL-C, and TC LDL, TC, TGs, and glucose ↑ in OA than in Severe OA vs. controls: TC: 207 vs. 193 mg/dl, OA/OA severity Metabolite
controls. Glucose level was associated with p = 0.026; OA vs. controls: TGs: 126 vs. 89 mg/dl,
KOA risk. p = 0.005; glucose: 105 vs. 97 mg/dl. LDL: ANOVA
p = 0.0427; glucose and KOA risk: OR=1.017
(1.001–1.035), p = 0.042.
Ukibe et al.40 2 (2) hsCRP, uric acid In premenopausal group, serum uric acid ↑ in Premenopausal OA vs. controls: uric acid: OA Protein +
OA than in non-OA. In postmenopausal group, 353.24 ± 67.15 vs. 256.76 ± 42.87, t = 5.68, p = 0.00. Metabolite
serum CRP and uric acid ↑ in OA than in Postmenopausal OA vs. controls: uric acid:
non-OA. 332.67 ± 73.15 vs. 261.62 ± 42.32, t = 3.85,
p = 0.00; CRP: 0.16 ± 0.09 vs. 0.03 ± 0.02, t = 6.03,
p = 0.00.
Ungsudechachai 1 (1) CLU In KOA, ST CLU ↑ in patients with synovitis ST CLU: data not reported. Plasma and SF CLU: OA severity Protein
et al.45 than in those without; plasma and SF CLU ↑ in high- vs. low-grade synovitis: p < 0.05 and
high-grade synovitis than in low-grade or no p < 0.01; high-grade vs. no synovitis: both
synovitis group and positively associated with p < 0.001; association with synovitis severity:
synovitis severity. plasma: rho = 0.56, p < 0.001, β = 0.004
(0.001–0.007), p = 0.017; SF: rho= 0.75, p < 0.001,
β = 0.005 (0.003–0.006), p < 0.001.
Watanabe et al.35 12 (1) CCL5 Serum CCL5 ↑ in DJD-TMJ than in controls, DJD-TMJ vs. controls: p < 0.05. Correlation with OA Protein
positively associated with bone metabolism bone metabolism markers NTX, CTX, BAP: DJD-TMJ
rate, especially in young DJD-TMJ subjects, group: r = 0.14, 0.52, 0.38; young DJD-TMJ: r = 0.41,
could distinguish DJD-TMJ from controls. 0.64, 0.64; controls: r = −0.56, 0.01, −0.28.
AUC=0.7093 (0.5172–0.9015), p = 0.0372, from a
model fit.
Xie et al.68 16,641 (2) C16:1/C14, C16:1/C12 C16:1/C14 and C16:1/C12 were associated Percentage of cartilage volume loss/year: C16:1/ OA progression Metabolite
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

with patellar cartilage volume change. C14: β = −0.12 (−0.10, −0.14), p = 8.80 × 10−7, C16:1/
C12: β = −0.12 (−0.10, −0.14), p = 2.66 × 10−6.
Zertuche et al.69 6 (1) AR Baseline AR was associated with a ↓ risk of RR (Q4 vs. Q1)=0.31 (0.15, 0.64). OA progression Metabolite
radiographic KOA at 30-month.
Zheng et al.55 3 (2) Glucose, HbA1c DM and poor glycemic control were Symptomatic KOA at 5-year follow-up: DM and OA Protein +
associated with symptomatic KOA at 5-year HbA1c≥7%: HR=1.41 (1.05–2.09), p = 0.036. Incident Metabolite
follow-up. ↑ HbA1c and glucose were symptomatic KOA was associated with
associated with ↑ risk of incident HbA1c > 7.7% and glucose > 186 mg/dl (HR > 1).
symptomatic KOA.
(continued on next page)
Table 3 (continued)

Reference # of markers Identified Biomarker (s) Findings Estimates Biomarker Biomarker


tested application type
(identified)

Zhou et al.24 107 (22) Combination of 13 peptide markers A set of 13 serum proteomic markers could JSL and pain progressor vs. non-progressor: OA progression Protein
CD44(30–38), ZPI(438–444), FA5(1506–1517), distinguish JSL and pain progressors from AUC=0.740 (0.669–0.772), from a model fit. JSL
HEP2(469–483), HEMO(198–208), FETUA(58–67), non-progressors. A set of 11 serum proteomic progressor vs. non-progressor: AUC=0.698
TSP1(217–228), DOPO(585–602), CRAC1(101–108), markers could distinguish JSL progressors (0.631–0.735), validation in an external cohort:
PHLD(800–808), KNG1(479–496), C1R(683–689), from non-progressors, and was validated in an AUC=0.697.
and VTDB(95–114). Combination of 11 peptide external cohort.
markers ZPI(438–444), VTDB(364–370),
IC1(287–298), CD14(192–210), RET4(185–195),
HRG(140–153), PLF4(82–92), CRAC1(170–178),
SHBG(170–183), VTDB(128–149), C1R(683–689).

8-OHdG: 8-hydroxy-2′-deoxyguanosine; aa: diacyl; ACAN: aggrecan core protein; ADL: activities of daily living; ae: acyl-alkyl; ALP: alkaline phosphatase; AR: alkylresorcinol; ARGS: ADAMTS-mediated aggrecan degradation
products; AUC: area under the curve; BDNF: brain-derived neurotrophic factor; C0: carnitine; C10C: cathepsin K-mediated type X collagen degradation fragment; C12: dodecanoylcarnitine; C14: tetradecanoylcarnitine; C14:2:
tetradecadienylcarnitine; C16:1: hexadecenoylcarnitine; C1R: complement C1r subcomponent; C2M: matrix metalloproteinase-mediated type II collagen degradation fragment; C3M: matrix metalloproteinase-mediated type III
collagen degradation fragment; CCC: cubic clustering criterion; CCL5: C-C chemokine receptor type 5; CCR7: C-C chemokine receptor type 7; CD14: monocyte differentiation antigen CD14; CD44: monocyte differentiation antigen
CD44; CILP-2: cartilage intermediate layer protein 2; CLU: clusterin; CML: carboxymethyllysine; COLL2_1: type II collagen degradation fragmen; COLL2_1NO2: inflammation-related (nitrated) type-II collagen degradation
fragment; COMP: cartilage oligomeric matrix protein; CRAC1: cartilage acidic protein 1; CRPM: matrix metalloproteinase-mediated c reactive protein degradation fragment; CRTAC1: cartilage acidic protein 1; CS: central
sensitization; CTXI: cross-linked, isomerised and cathepsin k-generated fragment of type I collagen C-terminal telopeptide; CTX-I: C-propeptide of type I collagen; CTXI_ALPHA: cathepsin K-generated fragment of type I collagen
C-terminal telopeptide; CTXII: matrix metalloproteinase- and cathepsin K-mediated type II collagen degradation fragment; CTX-II: C-propeptide of type II collagen; CVD: cardiovascular disease; CXCL: C-X-C motif chemokine
ligand; CXCR4: C-X-C chemokine receptor type 4; DDR2: discoidin domain receptor 2; DDw/oR: disc displacement without reduction; DJD-TMJ: degenerative joint disease of the temporomandibular joints; DM: diabetes mellitus;
DOPO: dopamine β-hydroxylase; FA 16:1: palmitoleic acid; FA 18:1: oleic acid; FA 18:2: linoleic acid; FA 20:4: arachidonic acid; FA 22:5: docosapentaenoic acid; FA5: coagulation factor V; FBN1: fibrillin-1; FETUA: alpha-2-HS
glycoprotein; GNLY: granulysin; HbA1c: glycosylated hemoglobin; HEMO: hemopexin; HEP2: heparin cofactor 2; HIV: human immunodeficiency virus; HMGB1: high mobility group box 1 protein; HR: hazard ratio; HRG:
histidine-rich glycoprotein; hsCRP: high-sensitive C-reactive protein; IC1: plasma protease C1 inhibitor; IHC: immunohistochemistry; IL: interleukin; JFLS-8: Jaw Functional Limitation Scale-8; JSL: joint space loss; JSN: joint space
narrowing; KL: Kellgren and Lawrence; KNG1: kininogen-1; KOA: knee osteoarthritis; KOOS: knee injury and osteoarthritis outcome score; LDD: lumbar disc degeneration; LDL-C: low density lipoprotein cholesterol; LOA: late
osteoarthritis; LPC: lysophosphatidylcholine; LRG1: leucine-rich alpha-2 glycoprotein; mCRP: monomeric C-reactive protein; MDA: malondialdehyde; MMP: matrix metallopeptidase; MRI: magnetic resonance imaging; NCAN:
neurocan core protein; NMID: bone gamma-carboxyglutamic acid-containing protein; NPY: neuropeptide-Y; NTX-1: N-telopeptide of type I collagen; OA: osteoarthritis; OARSI: Osteoarthritis Research Society International; OPG:
osteoprotegerin; OR: odds ratio; PBL: peripheral blood lymphocyte; PC: phosphatidylcholine; PGRN: progranulin; PHLD: phosphatidylinositol glycan–specific phospholipase; PIICP: procollagen type II C-terminal propeptide;
PLF4: platelet factor 4; PRO_C2: type IIB collagen propeptide; RANTES: regulated upon activation, normal T cell expressed and secreted; RE_C1M: matrix metalloproteinase-mediated type I collagen degradation; RET4: retinol-
binding protein 4; RR: relative risk; SERPINF1: pigment epithelium-derived factor; SF: synovial fluid; SHBG: sex hormone binding globulin; SM: sphingomyelin; TC: total cholesterol; TDO2: tryptophan 2,3-dioxygenase; TGs:
triglycerides; TKR: total knee replacement; TLR4: toll-like receptor-4; TMJOA: temporomandibular joint osteoarthritis; TNF-α: tumor necrosis factor-α; TSP1: thrombospondin-1; UKR: unicompartmental knee replacement; VAS:
visual analogue scale; VDBP: vitamin D-binding protein; VTDB: vitamin D binding protein; WOMAC: the Western Ontario and Mcmaster Universities Arthritis Index; ZPI: protein Z–dependent protease inhibitor.

Table 3
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

Results of individual studies.


1447
1448 M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

Identified Biomarker (s) Reference Findings

CCL5 Feng et al.34 CCL5 ↑ in DDw/oR with early-stage TMJOA, can distinguish these patients from other DDw/oR patients.
Goode et al.75 CCL5 was one of the proteins in the panel to cluster OA patients.
Watanabe et al.35 Serum CCL5 ↑ in DJD-TMJ than in controls, positively associated with bone metabolism rate, especially in young
DJD-TMJ subjects, could distinguish DJD-TMJ from controls.
COMP Angelini et al.74 COMP was one of the proteins in the panel to cluster OA patients.
Hussain et al.15 COMP ↑ in KOA than in controls, and could distinguish KOA from controls.
Singh et al.14 COMP was negatively associated with medial femorotibial compartment cartilage thickness.
CRTAC1 Styrkarsdottir et al.22 CRTAC1 was positively associated with prevalent OA, predicted incident OA diagnosis and progression to joint
replacement. Individuals in the highest quintile of risk based on CRTAC1 level, age, sex, and BMI had a 9.8-fold risk of
knee or hip OA.
Szilagyi et al.13 CRTAC1 was positively associated with overall OA burden, hand OA and KOA severity and KOA progression.
Tardif et al.23 CRTAC1 was the most significant contributor to the panel discriminating OA from controls and ↑ in OA than in
controls.
Zhou et al.24 CRTAC1 was one of the proteins in the panel discriminating JSL and pain progressors from non-progressors, and in the
panel discriminating JSL progressors from non-progressors, and was validated in an external cohort.
CTX-I Angelini et al.74 CTX-I was one of the proteins in the panel to cluster OA patients.
Hussain et al.15 CTX-I ↑ in KOA than in controls, and could distinguish KOA from controls.
CTX-II Angelini et al.74 CTX-II was one of the proteins in the panel to cluster OA patients.
Goode et al.75 CTX-II was one of the proteins in the panel to cluster OA patients.
Liem et al.16 Baseline uCTX-II was associated with KL grade, medial JSN, lateral osteophytes, joint space width, and WOMAC pain
scores.
Lu et al.17 CTX-II ↑ in traumatic KOA than in controls, and ↑ in KL grade III+IV than in KL grade I+II group.
Glucose Chiba et al.54 In KOA patients with CS, glucose was negatively associated with changes in KOOS pain, ADLs, and sports scores from
baseline to 1-year follow-up.
Tudorachi et al.56 Glucose ↑ in OA than in controls and was associated with KOA risk.
Zheng et al.55 Poor glycemic control were associated with symptomatic KOA at 5-year follow-up. ↑ glucose were associated with ↑
risk of incident symptomatic KOA.
HbA1c Perruccio et al.42 Percentage of individuals with hsCRP level within published threshold for moderate/elevated CVD risk ↑ in OA and
multisite OA, which was predominant in men.
Zheng et al.55 ↑ HbA1c were associated with ↑ risk of incident symptomatic KOA.
hsCRP Angelini et al.74 hsCRP was one of the proteins in the panel to cluster OA patients.
Perruccio et al.42 Percentage of individuals with hsCRP level within published threshold for moderate/elevated CVD risk ↑ in OA and
multisite OA, which was predominant in women.
Ukibe et al.40 In postmenopausal women, hsCRP ↑ in OA than in non-OA.
Hyaluronic acid Angelini et al.74 Hyaluronic acid was one of the proteins in the panel to cluster OA patients.
Goode et al.75 Hyaluronic acid was one of the proteins in the panel to cluster OA patients.
LDL-C Nemet et al.57 Prevalence of high LDL-C ↑ in female KOA patients than in female controls.
Tudorachi et al.56 LDL ↑ in OA than in controls.
MMP3 Elamir et al.27 MMP-3 ↑ in KOA than in controls and could diagnose OA.
Singh et al.14 MMP-3 was negatively associated with patellar cartilage volume and total bone area after 4 years.
Singh et al.26 MMP-3 ↑ in OA than in controls, and positively correlated with KL grades, could distinguish between OA and controls
and different KL grades.
TC Nemet et al.57 Prevalence of high TC ↑ in female KOA patients than in female controls.
Tudorachi et al.56 TC ↑ in OA than in controls.
Uric acid Rifal et al.41 Hyperuricemia group had ↑ percentage of severe KOA compared to normal uric acid group, had a ↑ risk of
severe KOA.
Ukibe et al.40 In women, uric acid ↑ in OA than in non-OA.
VDBP Tardif et al.23 VDBP was one of the proteins in the panel discriminating OA from controls and ↑ in OA than in controls, which was
validated in an external cohort.
Zhou et al.24 VDBP was one of the proteins in the panel discriminating JSL and pain progressors from non-progressors, and in the
panel discriminating JSL progressors from non-progressors, and was validated in an external cohort.

ADL: activities of daily living; CCL5: C-C chemokine receptor type 5; COMP: cartilage oligomeric matrix protein; CRTAC1: cartilage acidic protein 1; CS: central sensitization;
CTX-I: C-propeptide of type I collagen; CTX-II: C-propeptide of type II collagen; CVD: cardiovascular disease; DDw/oR: disc displacement without reduction; DJD-TMJ:
degenerative joint disease of the temporomandibular joints; DM: diabetes mellitus; HbA1c: glycosylated hemoglobin; hsCRP: high-sensitive C-reactive protein; JSL: joint
space loss; JSN: joint space narrowing; KL: Kellgren and Lawrence; KOA: knee osteoarthritis; KOOS: knee injury and osteoarthritis outcome score; LDL-C: low density
lipoprotein cholesterol; MMP: matrix metallopeptidase; OA: osteoarthritis; TC: total cholesterol; TMJOA: temporomandibular joint osteoarthritis; VDBP: vitamin D-binding
protein; WOMAC: the Western Ontario and Mcmaster Universities Arthritis Index.

Table 4

Overlapping findings between studies.

Amirkhizi et al.38 found that KOA patients had lower levels of vi­ cells leads to immunomodulatory events. In case of vitamin D defi­
tamin D than controls. In KOA, compared with patients with suffi­ ciency, cartilage repair could be weakened and inflammatory re­
cient vitamin D, vitamin D insufficiency group had higher serum sponses increase leading to more severe OA symptoms.38
levels of inflammatory biomarkers, which were inversely correlated
with serum vitamin D level, and higher WOMAC scores. Vitamin D Inflammation-related biomarkers
exerts metabolic functions through vitamin D receptors (VDRs) and Given the important role of inflammation in OA pathogenesis, in­
their activation could stimulate proteoglycan synthesis in mature flammation-related biomarkers are also the focus of some studies. High-
chondrocytes. In addition, binding of vitamin D to VDRs on immune sensitive C-reactive protein (hsCRP) is an established systematic
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453 1449

inflammatory marker and uric acid has been suggested to be a factor Metabolism-related biomarkers
promoting the pathological process of OA through activation of the in­ A number of metabolism-related biomarkers were reported in
flammasome.39 Ukibe et al.40 found that in human immunodeficiency the past year including the most commonly recognized glucose
virus infected women, serum uric acid level was higher in OA patients in and HbA1c, blood lipids total cholesterol (TC), low density lipo­
premenopausal groups, and both CRP and uric acid levels were higher in protein cholesterol (LDL-C), and triglycerides (TGs), obesity and
OA patients in postmenopausal groups compared with controls. Rifal energy homeostasis hormone adropin, and aging biomarker car­
et al.41 found that the percentage of KL grade I-II patients was higher boxymethyllysine (CML). Chiba et al.54 found that in OA patients
than that of grade III-IV patients in the normal uric acid group, while it with central sensitization, glucose level at baseline was negatively
was the opposite in hyperuricemia group, which had a greater risk of associated with changes in pain, activities, and sports scores from
experiencing severe KOA. Perruccio et al.42 reported that higher per­ baseline to one-year follow-up. Zheng et al.55 found that study
centage of OA patients especially multisite OA patients had hsCRP and participants with diabetes mellitus (DM) and HbA1c≥7% were
glycosylated hemoglobin (HbA1c) levels within published threshold for more likely to develop symptomatic KOA at 5-year follow-up
moderate/elevated cardiovascular disease (CVD) risk, pointing to a po­ compared to those without DM. Higher levels of HbA1c and
tentially important role for inflammation in OA over and above tradi­ fasting blood glucose were associated with higher risk of incident
tional CVD risk factors. In two studies on pro-inflammatory symptomatic KOA. Tudorachi et al.56 reported that levels of TC,
cytokines,43,44 it was found that serum interleukin (IL)-8 level was LDL, TGs, and glucose were higher in OA or severe OA patients, and
higher in early KOA patients, and serum IL-17A level was higher in KOA glucose level was associated with KOA risk, but the predictive
patients, specifically KL grade IV patients, than in controls, and positively power was low compared to other factors such as sex, body mass
correlated with SF IL-17A level in OA patients. Serum and SF IL-17A levels index, and environment of origin. Nemet et al.57 reported that in
had positive correlations with pain and functional deficit scores with the female, fasting glucose and TG levels and percentages of patients
correlations being stronger for SF IL-17A, which also had strong positive with high TC and LDL-C levels were all higher in KOA patients than
correlations with radiographic severity and duration of OA. Un­ in controls. These studies demonstrated that metabolic syndrome
gsudechachai et al.45 reported that the level of a multifunctional glyco­ components could contribute to OA development and progression.
protein clusterin was increased in KOA patients with synovitis than in Selim et al.58 studied adropin, a protein that participates in energy
those without, greater in those with high-grade synovitis than in those homeostasis and the control of fatty acid and glucose metabolism,
with low-grade or no synovitis, and positively associated with synovitis and found that serum adropin level in KOA patients was lower
severity, but the effect sizes were very small. Shao et al.46 found that SF than in controls, and lower in females than in males in OA pa­
level of high mobility group box 1 protein (HMGB1) was higher in tients. Serum adropin level was negatively correlated with KL
TMJOA than in TMJ internal derangement (TMJID) group, and positively grading and could distinguish between KOA patients and controls.
correlated with pain and functional deficit scores and radiographic stage, Cambon-Binder et al.59 discovered that serum CML concentration
could distinguish TMJID from TMJOA. HMGB1 is a non-histone nuclear was lower in erosive than in non-erosive hand OA, and cartilage
protein that can serve as an alarmin which is released into the extra­ CML concentration was higher in OA than in non-OA metacarpo­
cellular space upon tissue injury or cellular activation to trigger in­ phalangeal (MCP) joints. However, only five OA MCP joints were
flammation and immune responses,47 which could contribute to OA included in the analysis. Their findings seem to suggest that CML
development and progression. Lu et al.17 reported that serum levels of plays different roles in bone erosion and cartilage degeneration
toll-like receptor-4 (TLR4) and C-X-C motif chemokine ligand (CXCL)12 but need further validation.
were higher in traumatic KOA patients than in controls, and higher in KL
grade III+IV than in KL grade I+II group. The activation of TLR4 signaling Oxidative stress-related biomarkers
pathway leads to host responses in the form of de novo expression of Jitjumnong et al.60 reported that plasma levels of two previously
genes such as inflammatory cytokines, and is followed by the production discovered oxidative stress-related biomarkers, 8-hydroxy-2′-deox­
of chemokines and cytokines.48 CXCL12 has the ability of inducing yguanosine (8-OHdG) and malondialdehyde (MDA) were higher in
chondrocyte hypertrophy, and by binding to its receptors C-X-C che­ lumbar disc degeneration (LDD) patients than in controls, and po­
mokine receptor type 4 (CXCR4) on chondrocytes, it can regulate many sitively correlated with LDD severity. 8-OHdG is a biomarker of
homeostatic and pathological processes during the progression of OA via oxidative DNA damage and MDA is an end-product of lipid perox­
the activation of downstream signaling pathways.49 Interestingly, an­ idation. These evidences showed that higher level of oxidative stress
other study conducted by Trajerova et al.50 showed that elevated ex­ was present in LDD patients, and plasma 8-OHdG and MDA may
pression of CXCR4 was linked to persistent KOA symptoms. Cheng et al.51 have potential as noninvasive biomarkers for the assessment of LDD
and Sodhi52 et al. found that SF C3-α and C3-β levels were correlated severity.
with the KOA severity and higher in the OARSI grade ≥6 group. In
male patients, increased SF C5 level was associated with lower Novel biomarkers/biomarker panels
vascularization scores and higher SF level of terminal complement A number of novel biomarkers/biomarker panels were re­
complex sC5b-9. Vascularization scores indicate wound healing ported in the past year. Shekhar et al.61 reported that serum MMP-
and sC5b-9 level indicates complement activation. Their finding 17 level was higher in KOA and increased with disease severity,
raised the hypothesis that the positive association between com­ and could distinguish KOA patients from controls. Higher serum
plement signaling and OA outcomes might be due to the inhibition MMP-17 level was associated with higher risk for having advanced
of synovial vascularization in OA. However, the effect size was quite stage KOA. Sarkar et al.62 showed that in plasma, leucine-rich
small and the findings need further validation. In a lipidomics alpha-2 glycoprotein (LRG1) level was increased in OA patients
study, Loef et al.53 discovered that lipidomics accounted for a small compared to controls. In SF, LRG1 level was higher in OA patients
portion of the variance in OA severity, pain, and function, which who underwent total knee replacement (TKR) than in those who
were associated with the lipidomic fraction of bound and free underwent unicompartmental knee replacement. LRG1 was
arachidonic acid, bound palmitoleic acid, oleic acid, linoleic acid, identified as one of the important proteins that might play a
and docosapentaenoic acid (FA 22:5), with FA 22:5 being a newly central role in fibrosis in OA. In the previously mentioned study on
discovered lipid species associated with OA. This study only pre­ CRTAC123, the authors also found that serum levels of vitamin D
sented R2 not p values which made it difficult to evaluate the sig­ binding protein (VDBP), fibrillin-1 (FBN1), and pigment epithe­
nificance of the identified metabolites. lium-derived factor (SERPINF1) were all upregulated in OA
1450 M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

patients than in controls, and validated their findings in another Biomarkers for OA clustering
independent cohort except for SERPINF1. Three peptides of VDBP
were reported in another study to be associated with OA radio­ Trajerova et al.50 identified four immune-phenotypes in KOA, and
graphic and/or pain progression over 48 months.24 VDBP was also found that elevated CXCR4 and CCR7 expression on macrophages
found to be the second contributor in the component dis­ and low CXCL10 in SF were linked to the persistence of KOA symp­
criminating OA from controls in this study, and has been pre­ toms. All three of these proteins have been previously linked to OA
viously reported to be upregulated in skeletal muscles from end- with CXCR4 signaling being involved in the expression of matrix-
stage KOA patients.63 FBN1 is an extracellular matrix protein that degrading enzymes,71 CCR7 deficiency leading to delayed develop­
has been reported to regulate the bioavailability of the latent ment of joint damage and functional deficits,72 and CXCL10 playing
transforming growth factor beta-1 complex, a key factor involved an important role in tissue repair.73 Angelini et al.74 identified three
in OA cartilage and bone.64 Liang et al.65 found that plasma C- KOA clusters based on the concentrations of 16 baseline serum and
reactive protein monomer (mCRP) level was higher in OA patients urine biochemical markers reflecting joint tissue turnover. Cluster 1
than in controls and higher in KL grade IV than in KL grade III (C1) represented a low tissue turnover phenotype and had the
patients, and had a better predictive performance for diagnosing highest proportion of non-progressors; cluster 2 (C2) represented a
OA than CRP. mCRP is the monomeric form of CRP and the main structural damage phenotype and was mostly linked to longitudinal
active conformation of local tissues. Compared to CRP which is a structural progression; cluster 3 (C3) represented a systemic in­
non-specific inflammatory marker, mCRP is considered to play a flammation phenotype and was linked to sustained or progressive
role by depolymerizing pentamer CRP into monomer form in the pain. The authors also validated the clustering in an independent
local tissue of the lesion and may be a more sensitive indicator in a cohort. However, the cluster separation in the discovery cohort was
specific disease. Another newly discovered inflammation-related vague, and all markers showed differences in median cluster values
protein biomarker for OA was tryptophan 2,3-dioxygenase (TDO2). between the discovery and validation cohorts. Goode et al.75 iden­
Rong et al.66 reported that synovium level of TDO2 was higher in tified three OA clusters based on profiles of 10 serum and urine
OA and could discriminate OA patients from controls. In OA pa­ protein/metabolite biomarkers associated with lumbar spine struc­
tients, TDO2 level was positively correlated with pro-in­ tural changes, inflammation, or pain. C1 was a structural change
flammatory cytokine levels in synovium and SF, and synovium subgroup characterized by a higher percentage of lumbar spine
TDO2 level was positively correlated with KL scores. TDO2 is the structural changes; C2 was a reference subgroup with lower levels of
primary enzyme that catabolizes tryptophan to kynurenine and biomarkers, pain, and structural degenerative changes; C3 was a
has been suggested to be involved in inflammation-related dis­ pain and inflammation subgroup with higher levels of structure and
eases. However, the sample size of this study was small and the inflammation-related biomarkers and pain. These findings may have
controls were patients that had meniscal injury, which could have utility for differentiating particular subgroups of patients who may
affected the TDO2 level of the control group. Drvar et al.67 found have a nociception generating structure resulting in pain, which may
that in female, the intensity of granulysin (GNLY) labeling in help increase targeting specificity when considering interventions.
peripheral blood lymphocytes was higher in KOA than in controls. In addition to protein biomarkers, OA clustering was also explored
More investigations were done in this study using cells to de­ using metabolites. Rockel et al.76 identified two end-stage KOA
monstrate that GNLY-mediated apoptosis of K-562 targets was clusters based on plasma concentrations of 151 metabolites, and 24
higher in KOA patients alongside lymphocyte pro-inflammatory metabolites could predict cluster classification, highlighting en­
polarization. However, this study needs to be broadened with a riched tRNA acylation and B-vitamin metabolism pathways. This
larger cohort and different OA phenotypes in both sexes to reach a study was unable to examine the differences in OA clinical pheno­
definitive conclusion. In addition to these protein biomarkers, a types or trajectories of the two clusters as this was a cross-sectional
few new metabolite biomarkers for OA were reported in relatively study and all participants were end-stage OA patients under­
large cohorts. Xie et al.68 identified C16:1/C14 and C16:1/C12 to be going TKR.
positively associated with patellar cartilage volume loss, sug­
gesting the involvement of altered long chain fatty acid β-oxida­ Biomarkers for OA treatment outcomes
tion in this process. Zertuche et al.69 found that baseline level of
alkylresorcinol, a marker of whole grain intake, was associated The most effective treatment for end-stage KOA is TKR, yet a
with a reduced risk of radiographic KOA at 30-month, but this great portion of patients still report pain post-TKR. Using meta­
association did not hold for symptomatic KOA at 30-month or 60- bolomics profiling and meta-analysis, Costello et al. 4 found that
month cumulative outcomes. This study provided an uncertain plasma phosphatidylcholine (PC) diacyl (aa) C28:1 concentration
but suggestive result that high fiber intake might protect against and PC aa C28:1/PC aa C32:0 were negatively associated with
KOA. Sasaki et al.70 identified 42 plasma metabolites to be corre­ sustained pain post TKR, while PC aa C32:0/PC aa C28:1 and
lated with OA severity in female, indicating that urea cycle and C14:2/sphingomyelin C20:2 were positively associated, sug­
tricarboxylic acid (TCA) cycle were the key metabolic pathways in gesting potential roles of inflammation, oxidative stress, pain
OA, and the level of cysteine, an amino acid associated with an­ sensitization and altered lipid metabolism in sustained pain
tioxidant activity and glutamate homeostasis, was moderately in KOA.
associated with radiographic OA. Zhou et al.24 identified a set of 13
serum proteomic markers that could distinguish KOA joint space Conclusion
loss and pain progressors from non-progressors, better than tra­
ditional prediction based on baseline structural OA and pain se­ The majority of the studies published in the past year investigated
verity or urinary CTX-II. A set of 11 serum proteomic markers biomarkers for OA or OA severity/progression. However, the number of
could distinguish JSL progressors from non-progressors, and was studies exploring biomarkers for OA endotypes is rising, and three OA
confirmed in a validation cohort. These two biomarker panels had subtypes, low progression, structure change, and inflammation with
higher predictive power for clinically relevant KOA progression pain, have been consistently identified using protein and metabolite
than single protein markers, and provided a basis for future de­ biomarker panels in two separate studies on knee and spine, respec­
velopment of means of identifying individuals most in need of tively.74,75 A number of previously discovered biomarkers related to
surveillance and disease modifying therapies. cartilage/bone structure, inflammation, metabolism, and oxidative stress
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453 1451

were reported with MMPs, CTX-II, COMP, and CRTAC1 being identified in 2. Roos EM, Arden NK. Strategies for the prevention of knee os­
multiple studies, although the predictive powers of these biomarkers teoarthritis. Nat Rev Rheumatol 2016;12:92–101.
need to be carefully considered and further investigated as all these 3. Javaid MK, Kiran A, Guermazi A, Kwoh CK, Zaim S, Carbone L,
proteins had small effect sizes in most of the studies. Among the newly et al. Individual magnetic resonance imaging and radiographic
discovered biomarkers, increasing number of metabolites were reported features of knee osteoarthritis in subjects with unilateral knee
and they pointed out several pathways including urea cycle, TCA cycle, pain: the health, aging, and body composition study. Arthritis
tRNA acylation, B-vitamin metabolism, pain sensitization, and altered Rheum 2012;64:3246–55.
lipid metabolism to be associated with OA severity/progression, en­ 4. Costello CA, Rockel JS, Liu M, Gandhi R, Perruccio AV,
dotypes, and treatment outcomes,4,68,70,76 which may assist in eluci­ Rampersaud YR, et al. Individual participant data meta-analysis
dating pathogenesis of OA and developing new interventions. Several of metabolomics on sustained knee pain in primary osteoar­
studies explored biomarker panels for OA progression24 and clus­ thritis patients. Rheumatology 2022;62:1964–71.
tering74–76 and demonstrated that biomarker panels might have better 5. Califf RM. Biomarker definitions and their applications. Exp Biol
predictive powers compared to individual markers, at least for OA pro­ Med 2018;243:213–21.
gression.24 Combination of biochemical markers with other types of 6. FDA-NIH Biomarker Working Group. Contents of a Biomarker
makers (e.g., imaging markers, clinical factors, etc.) might also be a better Description. BEST (Biomarkers, EndpointS, and other Tools)
strategy to predict OA severity and progression.16,53 The major limitation Resource. Bethesda (MD): National Institutes of Health (US),; 2016.
of the studies published in the past year is that nearly half of these 7. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC,
studies had small (n < 50) or very small (n < 20) sample sizes in at least Mulrow CD, et al. The PRISMA 2020 statement: an updated
one study group, and most studies did not validate their findings or guideline for reporting systematic reviews. Rev Esp Cardiol
models in other independent cohorts, which could limit broader clinical 2021;74:790–9.
applicability of discovered markers. Moreover, in the real-life clinical 8. Ma LL, Wang YY, Yang ZH, Huang D, Weng H, Zeng XT.
practice, the most needed biomarkers are the ones that can help identify Methodological quality (risk of bias) assessment tools for pri­
OA patients among individuals with joint symptoms, whereas most of mary and secondary medical studies: what are they and which is
these studies compared OA patients with healthy controls or individuals better? Mil Med Res 2020;7:7.
without joint pain. OA biomarker studies should be taken one-step 9. Moola S, Munn Z, Tufanaru C, Aromataris E, Sears K, Sfetcu R,
further and the design of the studies should be carefully considered to et al. Chapter 7: Systematic reviews of etiology and risk. In:
connect the lab findings with clinical applications. Despite these lim­ Aromataris E, Munn Z, editors. JBI Manual for Evidence
itations, these discoveries may help better understand the pathogenesis Synthesis. JBI; 2020.
of OA, provide potential therapeutic targets and insights into possible 10. G.A. Wells, B. Shea, D. O’Connell, J. Peterson , V. Welch, M. Losos,
mechanisms underlying poor treatment outcomes, and aid in developing et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality
personalized or stratified interventions based on OA subtypes. of nonrandomised studies in meta-analyses. 〈https://www.ohri.
ca/programs/clinical_epidemiology/oxford.asp〉. [Accessed on
CRediT authorship contribution statement April 23, 2023].
11. Van Pevenage PM, Birchmier JT, June RK. Utilizing metabolomics
Ming Liu: Literature search and data acquisition, Data analysis to identify potential biomarkers and perturbed metabolic path­
and interpretation, Drafting original manuscript, Critical manuscript ways in osteoarthritis: a systematic review. Semin Arthritis
revision and approval of final manuscript. Nafiza Haque: Literature Rheum 2023;59, 152163.
search and data acquisition, Data analysis and interpretation, Critical 12. Posey KL, Coustry F, Hecht JT. Cartilage oligomeric matrix protein:
manuscript revision and approval of final manuscript. Jingyi Huang: COMPopathies and beyond. Matrix Biol 2018;71–72:161–73.
Literature search and data acquisition, Data analysis and inter­ 13. Szilagyi IA, Vallerga CL, Boer CG, Schiphof D, Ikram MA, Bierma-
pretation, Critical manuscript revision and approval of final manu­ Zeinstra SMA, et al. Plasma proteomics identifies CRTAC1 as a
script. Guangju Zhai: Conception and design, Literature search and biomarker for osteoarthritis severity and progression.
data acquisition, Data analysis and interpretation, Critical manu­ Rheumatology 2023;62:1286–95.
script revision and approval of final manuscript. 14. Singh A, Venn A, Blizzard L, Jones G, Burgess J, Parameswaran V,
et al. Association between osteoarthritis-related serum biochemical
Declaration of Competing Interest
markers over 11 years and knee MRI-based imaging biomarkers in
middle-aged adults. Osteoarthr Cartil 2022;30:756–64.
The authors declare no conflicts of interest.
15. Hussain A, Verma C, Kaushik LS, Singh S. Efficacy of sCOMP and
Acknowledgements sCTX-I in diagnosis of knee osteoarthritis. Indian J Orthop
2022;56:1565–71.
This study was supported by the Canadian Institutes of Health 16. Liem Y, Judge A, Li Y, Sharif M. Biochemical, clinical, demo­
Research (FRN#175015; 153298; 143058; 132178). The funder had no graphic and imaging biomarkers for disease progression in knee
role in the design of the study; in the collection, analyses, or inter­ osteoarthritis. Biomark Med 2022;16:633–45.
pretation of data; in the writing of the manuscript, or in the decision 17. Lu Z, Xu Z, Tao S, Dong Z, Li Z. Analysis on the changes of serum
to publication. TLR4, CXCL12 and CTX-II levels in patients with traumatic knee
osteoarthritis. Acta Medica Mediterranea 2023;39:505–8.
Appendix A. Supporting information 18. Dam EB, Byrjalsen I, Karsdal MA, Qvist P, Christiansen C.
Increased urinary excretion of C-telopeptides of type II collagen
Supplementary data associated with this article can be found in (CTX-II) predicts cartilage loss over 21 months by MRI.
the online version at doi:10.1016/j.joca.2023.08.005. Osteoarthr Cartil 2009;17:384–9.
19. Hao HQ, Zhang JF, He QQ, Wang Z. Cartilage oligomeric matrix
References protein, C-terminal cross-linking telopeptide of type II collagen,
and matrix metalloproteinase-3 as biomarkers for knee and hip
1. Deveza LA, Loeser RF. Is osteoarthritis one disease or a collection osteoarthritis (OA) diagnosis: a systematic review and meta-
of many? Rheumatology 2018;57:iv34–42. analysis. Osteoarthr Cartil 2019;27:726–36.
1452 M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453

20. Cheng H, Hao B, Sun J, Yin M. C-Terminal cross-linked telopep­ cartilage of patients with knee osteoarthritis undergoing total
tides of type II collagen as biomarker for radiological knee os­ knee arthroplasty: a pilot morphological study. Med Mol
teoarthritis: a meta-analysis. Cartilage 2020;11:512–20. Morphol 2023;56:46–57.
21. Tanishi N, Yamagiwa H, Hayami T, Mera H, Koga Y, Omori G, et al. 38. Amirkhizi F, Asoudeh F, Hamedi-Shahraki S, Asghari S. Vitamin
Relationship between radiological knee osteoarthritis and bio­ D status is associated with inflammatory biomarkers and clinical
chemical markers of cartilage and bone degradation (urine CTX- symptoms in patients with knee osteoarthritis. Knee 2022;36:
II and NTX-I): the Matsudai Knee Osteoarthritis Survey. J Bone 44–52.
Miner Metab 2009;27:605–12. 39. Denoble AE, Huffman KM, Stabler TV, Kelly SJ, Hershfield MS,
22. Styrkarsdottir U, Lund SH, Thorleifsson G, Saevarsdottir S, McDaniel GE, et al. Uric acid is a danger signal of increasing risk
Gudbjartsson DF, Thorsteinsdottir U, et al. Cartilage acidic pro­ for osteoarthritis through inflammasome activation. Proc Natl
tein 1 in plasma associates with prevalent osteoarthritis and Acad Sci USA 2011;108:2088–93.
predicts future risk as well as progression to joint replacements: 40. Ukibe NR, Kalu OA, Chineduihim A, Meniru OA, Ezinneukibe G,
results from the UK biobank resource. Arthritis Rheumatol Chinyereukibe B. Changes in serum uric acid and C-reactive
2022;75:544–52. protein in menopausal HIV seropositive with osteoartritisin
23. Tardif G, Pare F, Gotti C, Roux-Dalvai F, Droit A, Zhai G, et al. Mass NAUTH, Nnewi, Nigeria. NeuroQuantology 2022;20:8700–12.
spectrometry-based proteomics identify novel serum osteoar­ 41. Rifal M, Syahriani F, Adnan E, Bakri S, Halim R, Udaya W, et al.
thritis biomarkers. Arthritis Res Ther 2022;24:120. The relationship between uric acid levels and the severity of
24. Zhou K, Li YJ, Soderblom EJ, Reed A, Jain V, Sun S, et al. A "best- knee osteoarthritis. NeuroQuantology 2022;20:6937–41.
in-class" systemic biomarker predictor of clinically relevant 42. Perruccio AV, Zahid S, Yip C, Power JD, Canizares M, Heckman
knee osteoarthritis structural and pain progression. Sci Adv GA, et al. Cardiovascular risk profile and osteoarthritis-con­
2023;9, eabq5095. sidering sex and multisite joint involvement: a Canadian long­
25. Steck E, Benz K, Lorenz H, Loew M, Gress T, Richter W. Chondrocyte itudinal study on aging. Arthritis Care Res 2023;75:893–901.
expressed protein-68 (CEP-68), a novel human marker gene for 43. Abou-Raya A, Rizk M, AbdelGhani E, AbdelMegid N.
cultured chondrocytes. Biochem J 2001;353:169–74. Identification of serum micro-RNAs of early knee osteoarthritis
26. Singh S, Jindal D, Khanna R. Can serum MMP-3 diagnose early in a cohort of Egyptian patients. Alexandria J Med 2023;59:1–14.
knee osteoarthritis? J Orthop 2023;38:42–6. 44. Kamel S, Khalaf R, Moness H, Ahmed S. Serum and synovial fluid
27. Elamir AM, Zahra A, Senara SH, Ezzat EM, El Sayed HS. levels of interleukin-17A in primary knee osteoarthritis patients:
Diagnostic value of matrix metalloproteinases-1, -3 and -13 in correlations with functional status, pain, and disease severity.
patients with primary knee osteoarthritis: relation to radi­ Arch Rheumatol 2022;37:187–94.
ological severity. Egyptian Rheumatol 2022;45:17–20. 45. Ungsudechachai T, Honsawek S, Jittikoon J, Udomsinprasert W.
28. Mehana EE, Khafaga AF, El-Blehi SS. The role of matrix me­ Clusterin exacerbates interleukin-1beta-induced inflammation
talloproteinases in osteoarthritis pathogenesis: an updated re­ via suppressing PI3K/Akt pathway in human fibroblast-like sy­
view. Life Sci 2019;234, 116786. noviocytes of knee osteoarthritis. Sci Rep 2022;12:9963.
29. Wan J, Zhang G, Li X, Qiu X, Ouyang J, Dai J, et al. Matrix me­ 46. Shao B, Xu Y, Jia M, Li CX, Gong ZC. Association of HMGB1 levels
talloproteinase 3: a promoting and destabilizing factor in the in synovial fluid with the severity of temporomandibular joint
pathogenesis of disease and cell differentiation. Front Physiol osteoarthritis. BMC Musculoskelet Disord 2023;24:183.
2021;12, 663978. 47. Bertheloot D, Latz E. HMGB1, IL-1alpha, IL-33 and S100 proteins:
30. Burrage PS, Mix KS, Brinckerhoff CE. Matrix metalloproteinases: dual-function alarmins. Cell Mol Immunol 2017;14:43–64.
role in arthritis. Front Biosci 2006;11:529–43. 48. Barreto G, Manninen M. Osteoarthritis and toll-like receptors:
31. Sauerschnig M, Stolberg-Stolberg J, Schulze A, Salzmann GM, when innate immunity meets chondrocyte apoptosis. Biology
Perka C, Dynybil CJ. Diverse expression of selected cytokines and 2020;9:65.
proteinases in synovial fluid obtained from osteoarthritic and 49. Li J, Chen H, Zhang D, Xie J, Zhou X. The role of stromal cell-
healthy human knee joints. Eur J Med Res 2014;19:65. derived factor 1 on cartilage development and disease.
32. Hunter DJ, Deveza LA, Collins JE, Losina E, Katz JN, Nevitt MC, Osteoarthr Cartil 2021;29:313–22.
et al. Multivariable modeling of biomarker data from the phase I 50. Trajerova M, Kriegova E, Mikulkova Z, Savara J, Kudelka M, Gallo
foundation for the National Institutes of Health Osteoarthritis J. Knee osteoarthritis phenotypes based on synovial fluid im­
Biomarkers Consortium. Arthritis Care Res 2022;74:1142–53. mune cells correlate with clinical outcome trajectories.
33. Papaneophytou C, Alabajos-Cea A, Viosca-Herrero E, Calvis C, Osteoarthr Cartil 2022;30:1583–92.
Costa M, Christodoulides AE, et al. Associations between serum 51. Cheng TH, Zeng J, Dehghani A, Dimaculangan D, Zhang M,
biomarkers of cartilage metabolism and serum hyaluronic acid, Maheshwari AV. Complement C3-alpha and C3-beta levels in
with risk factors, pain categories, and disease severity in knee synovial fluid but not in blood correlate with the severity of
osteoarthritis: a pilot study. BMC Musculoskelet Disord osteoarthritis research society international histopathological
2022;23:195. grades in primary knee osteoarthritis. J Arthroplasty 2022;37:
34. Feng SY, Lei J, Li YX, Shi WG, Wang RR, Yap AU, et al. Increased 1541–1548 e1541.
joint loading induces subchondral bone loss of the tempor­ 52. Sodhi EU, Philpott HT, Carter MM, Birmingham TB, Appleton CT.
omandibular joint via the RANTES-CCRs-Akt2 axis. JCI Insight Sex-differences and associations between complement activa­
2022;7, e158874. tion and synovial vascularization in patients with late-stage
35. Watanabe H, Iori T, Lee JW, Kajii TS, Takakura A, Takao-Kawabata knee osteoarthritis. Front Immunol 2022;13, 890094.
R, et al. Association between an increased serum CCL5 level and 53. Loef M, van de Stadt L, Bohringer S, Bay-Jensen AC, Mobasheri A,
pathophysiology of degenerative joint disease in the tempor­ Larkin J, et al. The association of the lipid profile with knee and
omandibular joint in females. Int J Mol Sci 2023;24:2775. hand osteoarthritis severity: the IMI-APPROACH cohort.
36. Chow YY, Chin KY. The Role of Inflammation in the pathogenesis Osteoarthr Cartil 2022;30:1062–9.
of osteoarthritis. Med Inflamm 2020;2020, 8293921. 54. Chiba D, Ohyama T, Sasaki E, Daimon M, Nakaji S, Ishibashi Y.
37. Torga T, Suutre S, Kisand K, Aunapuu M, Arend A. Expression of Higher fasting blood glucose worsens knee symptoms in pa­
CILP-2 and DDR2 and ultrastructural changes in the articular tients with radiographic knee osteoarthritis and comorbid
M. Liu et al. / Osteoarthritis and Cartilage 31 (2023) 1437–1453 1453

central sensitization: an Iwaki cohort study. Arthritis Res Ther and synovial fluid of patients with osteoarthritis. BMC
2022;24:269. Musculoskelet Disord 2022;23:604.
55. Zheng J, Huang X, Huang J, Meng B, Li F, Liu H, et al. Association 67. Drvar V, Curko-Cofek B, Karleusa L, Aralica M, Rogoznica M,
of diabetes mellitus status and hyperglycemia with sympto­ Kehler T, et al. Granulysin expression and granulysin-mediated
matic knee osteoarthritis. Arthritis Care Res 2023;75:509–18. apoptosis in the peripheral blood of osteoarthritis patients.
56. Tudorachi NB, Totu T, Eva I, Barbieru B, Totu EE, Fifere A, et al. Biomed Rep 2022;16:44.
Knee osteoarthritis in relation to the risk factors of the meta­ 68. Xie Z, Aitken D, Liu M, Lei G, Jones G, Cicuttini F, et al. Serum
bolic syndrome components and environment of origin. J Clin metabolomic signatures for knee cartilage volume loss over
Med 2022;11:7302. 10 years in community-dwelling older adults. Life 2022;12:869.
57. Nemet M, Blazin T, Milutinovic S, Cebovic T, Stanojevic D, Zvekic 69. Zertuche JP, Rabasa G, Lichtenstein AH, Matthan NR, Nevitt M,
Svorcan J. Association between metabolic syndrome, its com­ Torner J, et al. Alkylresorcinol, a biomarker for whole grain in­
ponents, and knee osteoarthritis in premenopausal and meno­ take, and its association with osteoarthritis: the MOST study.
pausal women: a pilot study. Cureus 2022;14, e26726. Osteoarthr Cartil 2022;30:1337–43.
58. Selim F, Ahmed R, Eid M, ElSaadany H, Nada DW. Role of serum 70. Sasaki E, Yamamoto H, Asari T, Matsuta R, Ota S, Kimura Y, et al.
adropin as a novel biomarker in primary knee osteoarthritis Metabolomics with severity of radiographic knee osteoarthritis
patients: Relation to the functional status, radiographic grading and early phase synovitis in middle-aged women from the Iwaki
and ultrasonographic findings. Egypt Rheumatol 2022;44: Health Promotion Project: a cross-sectional study. Arthritis Res
295–299. Ther 2022;24:145.
59. Cambon-Binder A, Jaisson S, Tuffet S, Courties A, Eymard F, Okwieka 71. Wang K, Li Y, Han R, Cai G, He C, Wang G, et al. T140 blocks the
A, et al. Serum carboxymethyllysine concentration is associated with SDF-1/CXCR4 signaling pathway and prevents cartilage degen­
erosive hand osteoarthritis. Osteoarthr Cartil 2023;31:976–84. eration in an osteoarthritis disease model. PLoS One 2017;12,
60. Jitjumnong M, Chalermkitpanit P, Suantawee T, Dechsupa S, e0176048.
Vajarintarangoon L, Honsawek S. Telomere shortening and in­ 72. Sambamurthy N, Nguyen V, Smalley R, Xiao R, Hankenson K, Gan
creased oxidative stress in lumbar disc degeneration. Int J Mol J, et al. Chemokine receptor-7 (CCR7) deficiency leads to delayed
Sci 2022;23:10125. development of joint damage and functional deficits in a murine
61. Shekhar AC, Pritee C, Manish S, Amit R, Singh GI. Role of matrix model of osteoarthritis. J Orthop Res 2018;36:864–75.
metalloproteinase-17 in knee osteoarthritis and its association 73. Endres M, Andreas K, Kalwitz G, Freymann U, Neumann K, Ringe
with disease severity. Res J Biotechnol 2022;17:57–63. J, et al. Chemokine profile of synovial fluid from normal, os­
62. Sarkar A, Chakraborty D, Kumar V, Malhotra R, Biswas S. teoarthritis and rheumatoid arthritis patients: CCL25, CXCL10
Upregulation of leucine-rich alpha-2 glycoprotein: a key reg­ and XCL1 recruit human subchondral mesenchymal progenitor
ulator of inflammation and joint fibrosis in patients with severe cells. Osteoarthr Cartil 2010;18:1458–66.
knee osteoarthritis. Front Immunol 2022;13, 1028994. 74. Angelini F, Widera P, Mobasheri A, Blair J, Struglics A, Uebelhoer
63. Brennan-Speranza TC, Mor D, Mason RS, Bartlett JR, Duque G, M, et al. Osteoarthritis endotype discovery via clustering
Levinger I, et al. Skeletal muscle vitamin D in patients with end of biochemical marker data. Ann Rheum Dis 2022;81:
stage osteoarthritis of the knee. J Steroid Biochem Mol Biol 666–75.
2017;173:180–4. 75. Goode AP, Hu D, George SZ, Schwartz TA, Kraus VB, Huebner
64. Chaudhry SS, Cain SA, Morgan A, Dallas SL, Shuttleworth CA, JL, et al. Biomarker clusters differentiate phenotypes of
Kielty CM. Fibrillin-1 regulates the bioavailability of TGFbeta1. J lumbar spine degeneration and low back pain: The Johnston
Cell Biol 2007;176:355–67. County Osteoarthritis Project. Osteoarthr Cartil Open 2022;4,
65. Liang Y, Xu K, Liu W, Liu X, Yuan P, Xu P, et al. Monomeric C- 100270.
reactive protein level is associated with osteoarthritis. Exp Ther 76. Rockel JS, Layeghifard M, Rampersaud YR, Perruccio AV,
Med 2022;23:277. Mahomed NN, Davey JR, et al. Identification of a differential
66. Rong G, Zhang T, Xu Y, Zhang Z, Gui B, Hu K, et al. High levels of metabolite-based signature in patients with late-stage knee
TDO2 in relation to pro-inflammatory cytokines in synovium osteoarthritis. Osteoarthr Cartil Open 2022;4, 100258.

You might also like