Skip to main content
PLOS One logoLink to PLOS One
. 2025 Sep 22;20(9):e0332763. doi: 10.1371/journal.pone.0332763

Modeling treatment of osteoarthritis with standard therapy and senolytic drugs

Nourridine Siewe 1,*,#, Avner Friedman 2,#
Editor: Xindie Zhou3
PMCID: PMC12453251  PMID: 40982544

Abstract

Osteoarthritis (OA), the most common form of joint disease, involves the progressive degradation of articular cartilage and is a major cause of chronic disability in aging populations. Since OA is associated with severe deficiency of collagen type II, clinical trials considered treatment of OA by injection with undenatured collagen type II (UC-II). Recent studies consider also injection of senolytic drugs, like fisetin, that eliminates senescent chondrocytes in aging patients, to reduce the negative effect of these senescent cells on cartilage structure. In this paper we develop a mathematical model of OA for men and, separately, for women, and use the model to assess the efficacy of treatment by UC-II and by fisetin, alone or in combination. Our computations show the benefits of starting treatment early. They also show that although the effect of treatment by fisetin on slowing the progression of OA is much smaller compared to UC-II treatment, its effect in combination with UC-II is significantly increased.

Introduction

Arthritis is a term commonly used to mean any disorder that affects the joint; symptoms include join pain and stiffness. Osteoarthritis (OA) is the most common joint disease affecting 500 million people worldwide [1]. The disease wears down the protective cartilage that cushions the end of bone of a joint, and the damage cannot be reversed [2]. Cartilage is a spongy tissue containing up to 80% water, chondrocyte cells, and collagen produced by the chondrocytes; 90–95% of the collagen are collagen type II [3]. Chondrocytes receive nutrients and oxygen by diffusion from the synovial fluid and from the adjacent subchondral bone.

Articular cartilages are a class of cartilages with specific arrangement of chondrocytes and collagen fibers; they is found in the knee and hip joints, and in finger joints. Meniscus is the articular cartilage in the knee joint, below the synovial fluid, that functions as shock absorber.

Cellular senescence is a permanent arrest of normal cell cycle, while maintaining cell viability. Senescence cells secrete senescence-associated secretory phenotype (SASP) that include inflammatory proteins and MMP1, MMP13 [4]; the level of MMP13 is highly expressed in OA cartilage [5].

Cellular senescence is the primary hallmark of aging. OA in age-related cartilage degenerative disease is most commonly caused by chondrocyte senescence [6]. OA of knee joints affect most adults 65 and older, 33% of men and 42% of women in the United States; and knee arthritis is the most common cause of disability affecting 1 in 5 adults in the United States [7].

Arthritis can be inflammatory or non-inflammatory. In most of the population that develop OA after age 50, the disease is associated with age and it is non-inflammatory [8]. In inflammatory OA, the body immune system is causing the inflammation, and there remain considerable gaps in understanding its role in disease progression and how best to target inflammatory response for therapeutic intervention [9].

In this paper we developed a model of non-inflammatory arthritis for population over 50.

Chondrocyte senescence results in decline of chondrocyte cells; for example, there is a 30% fall of chondrocytes in articular cartilage of the hip joint between ages 30 to 70 [10]. Recent studies reveal the presence of mesenchymal stem cells (MSC) in the synovial fluid, synovial membrane, and articular cartilage [11]. Although MSC may sustain the source of chondrocytes or contribute to senescence in OA, because of lack of data we do not include MSC in our model, and treat the source of chondrocytes as a fixed parameter.

Chondrocyte hypertrophy is a process by which chondrocyte cells undergo 10 to 20-fold enlargement [12]. Hypertrophic chondrocytes are master regulators of endochondral ossification, i.e., chondrocyte-to-osteoblast transdifferentiation [12]. Hyertrophic chondrocytes lie near the boundary between cartilage and bone, and secrete factors that promote cartilage to bone transition [4]; they play an essential role in bone formation in development [4,5]. Chondrocytes transition to hypertrophic state in fracture healing [4,5].

Calcified cartilage is a layer of hard connective tissue between articular cartilage and sub-chondral bone; the calcification results from deposition of insoluble calcium salt in the bone matrix [13]. Calcium crystals in calcified cartilage play a role in pathogenesis of OA [14], Hypertrophic chondrocytes serve a critical intermediate in cartilage calcification and in OA lesions [15,16]; they mediate calcified cartilage through membrane bound enzymes and secreted matrix vesicles [17]. Pathological calcification is the hallmark of OA; calcification can be observed both at cartilage surface and in deeper levels [18]. Meniscal calcification is a predisposing factor for cartilage lesions, and is a target for disease modifying drugs of OA [19].

Collagen type II suppresses chondrocyte hypertrophy and OA progression [20], and degradation of collagen type II is associated with articular cartilage lesions [21]. In OA, the highly expressed MMP13 degrades the ECM structure, which results in decreased of collagen type II [22], hence it is an attractive target for treatment of OA [23].

Histological assessment of OA in mouse is given in [24]; in particular, grade 4 of OA is when lesion reaches the calcified cartilage for 25–50% of the quadrant width, grade 5 for 50–75%, and grade 6 when the width is above 75%.

Several mathematical and theoretical models have been proposed over the years to better understand the biomechanics and pathology of articular cartilage. A foundational study by Hayes et al. (1972) developed a mathematical model for indentation tests of articular cartilage, providing early insight into its mechanical behavior [25]. Later, Parra et al. (2011) introduced a model that quantifies the risk of cartilage failure by treating the tissue as an elastic material subjected to cyclic loading, highlighting the role of mechanical fatigue [26]. Kapitanov et al. (2016) advanced the field by proposing a PDE-based model that explicitly couples mechanical stress with biological processes in cartilage lesion formation [27]. In 2019, Campbell et al. focused on the regenerative aspects, modeling the effects of growth factors on cartilage repair following cell implantation [28], while Cope et al. critically reviewed existing osteoarthritis (OA) models and emphasized the need for a unified, physiologically accurate model [29]. Eschweiler et al. (2021) provided a comprehensive overview of cartilage biomechanics, summarizing key concepts relevant to both modeling and clinical application [30]. Most recently, Owida et al. (2022) reviewed the advancements in biomimetic strategies for cartilage tissue engineering, discussing their implications for future therapeutic approaches [31].

In this paper we develop a mathematical model of OA that includes chondrocytes (C), senescent chondrocytes (Cs), hypertrophic chondrocytes (Ch), calcified cartilage (Cc), MMP13 (MP), collagen II (L2) and undenatured collagen type II (U2). (Undenatured collagen type II is a specific form of collagen II derived from chicken stertum cartilage. This form of collagen reduces joint pains associated with immune response, when injected to OA patients.) The model includes treatment of OA by U2 injection and, separately, by senolytic drug fisetin. We demonstrate how the model can qualitatively replicate reported clinical studies with U2 injection. We then conduct clinical studies in-silico with these drugs, separately and in combination, and demonstrate the synergy of the combination.

1 Mathematical model

Table 1 lists the model variables in units of g/cm3. Fig 1 shows the interactions among the model variables. Chondrocytes (C) can become senescent (Cs) and senescent cells secrete MMP13 (MP). Chondrocytes can become hyperthophic (Ch), a process blocked by collagen type II (L2). MP blocks L2, which results in abnormal growth in Ch and formation of calcified cartilage (Cc).

Table 1. Variables of the model. Densities and concentrations are in units of g/cm3.

Descriptions Variables Descriptions Variables
chondrocyte cells C senescence chondrocytes C s
hypertrophic chondrocytes C h calcified cartilage C c
MMP13 M P collagen type II L 2
senolytic drug fisetin D undenatured collagen type II U 2

Fig 1. Interactions among cells and proteins.

Fig 1

C = chondrocytes, Cs = senescent chondrocytes, Ch = hypertrophic chondrocytes, Cc = calcified cartilage, MP = MMP13, L2 = collagen type II.

Fig 2 shows normal knee joint anatomy, and a simplified geometry used in the model.

Fig 2. (a) Knee joint anatomy (adapted from [32]).

Fig 2

(b) simplified geometry of articular cartilage.

The mathematical model is based on the network in Fig 1, and it takes place in the strip Ω={0xh} of the simplified geometry shown in Fig 2. We consider aging population between 50 and 90 years.

Model equations in males and females

In [33], a meta-analysis of animal models is conducted, highlighting that during the postmenopausal period, as estrogen levels decrease, there is a corresponding reduction in collagen type II in cartilage. The study in [34] further demonstrates that MMP-13 levels also increase in cartilage in murine subjects following menopause. The impact of estrogen depletion during postmenopause may help explain why the global prevalence of OA in women over the age of 60 is nearly twice as high as in men [34].

Based on Fig 1 and [33,34], we write the model equations as follows.

Equations for cells (Cs, C and Ch) and calcified cartilage (Cc).

We denote the source of chondrocytes in health by A, and write the following equations:

CtδC2Cx2=AλCCsCλCChC11+L2/KL2μCC (1)
CstδC2Csx2=λCCsCμCsCs (2)
ChtδC2Chx2=λCChC11+L2/KL2λChCcChtμChCh (3)
CctδCc2Ccx2=λChCcCht. (4)

Here, λCCs is the rate at which chondrocytes become senescent, and λCCh/(1+L2/KL2) is the rate at which chondrocytes become hypertrophic under blockage by collagen type II; KL2 is the half-saturation of L2. The term λChCcCh/t models calcified cartilage formation induced by chondrocyte hypertrophy dynamics, where Cc arises not from a direct ChCc transition but as a consequence of Ch activity [16]. The parameters μC, μCs and μCh are death rates, and λChCc is the rate of increase of calcified cartilage. The immune cells clear senescent cells [35], and since the strength of the immune system declines with age [36], μCs=μCs(t) is an increasing function of t; we take μCs(t) a putative increasing function

μCs(t)=μCs(0)1+t/(90 years) d1. (5)

Equations for proteins.

MPtδMP2MPx2={λCsMPCsμL2MPMPL2μMPMP,for males,λCsMPCsμL2MPMPL2μMPMP+dpMP(t)MP,for females, (6)
L2tδL22L2x2={λCL2CμMPL2L2MPμL2L2,for males,λCL2CμMPL2L2MPμL2L2dpL2(t)L2,for females. (7)

The parameter λCsMP denotes the production rate of MMP13 by senescent chondrocytes, and λCL2 represents the secretion rate of collagen type II by chondrocytes. The parameters μL2MP and μMPL2 describe the mutual elimination rates between collagen type II and MMP13. Degradation rates are μMP for MMP13 and μL2 for collagen type II, while dpMP(t) and dpL2(t) are postmenopausal monotone decreasing functions, with dpMP(0)>0, dpL2(0)>0 (age 50) and dpMP(t)~0, dpL2(t)~0 if t10 years (age 60). We take

dpX(t)=αwX1+t2/TwX2,X=MP,L2, (8)

where αwX and TwX are postmenopausal effect and time-scale postmenopausal effect of species X.

Equations with drugs.

CstδC2Csx2=λCCsCμCsCsμCsDCsD (9)
MPtδMP2MPx2={λCsMPCs11+αDD(t)μL2MPMPL2μMPMP,for males,λCsMPCs11+αDD(t)μL2MPMPL2μMPMP+dpMP(t)MP,for females, (10)
L2tδL22L2x2={αU2U2(t)+λCL2CμMPL2L2MPμL2L2,for males,αU2U2(t)+λCL2CμMPL2L2MPμL2L2dpL2(t)L2,for females. (11)

The parameter μCsD denotes the rate at which senescent chondrocytes (Cs) are eliminated due to D. The constants αD and αU2 scale the effects of D and U2, respectively.

The equations for the senolytic drug D, and UC-II (U2) (L2 injection) take the form

DtδD2Dx2=γDhD(t)μDCsDCsωDD (12)
U2(t)=γU2hU2(t), (13)

where μDCs is the rate of lost of D while eliminating Cs, ωD is the washout rate of D, and

hU2(t)={1,if the drug U2 is given (daily) at time t0,otherwise;

if D is administered only at days t0 and t1, then during the period t0tt2 (where t2>t1),

hD(t)={eμDt,for t0tt1eμDt+eμD(tt1),for t1<t<t2,

where μD is the degradation rate of the drug D.The parameters αD and αU2 are constants representing the effects of the respective drugs, ωD is the washout rate of D; t0, t1 and t2 are time points for delivering of D, and the parameters γD and γU2 are the doses of the drugs.

Boundary conditions.

We assume no-flux boundary conditions for all the viable cells:

Xx=0 on x=0 and x=h for cells X=C,Cs,Ch, (14)

and

Xn+αX=0 on x=0 and x=h for proteins X=L2,MP and drug D, (15)

for some α>0, where /n is the outward normal derivative.

Eq (14) means that cells X do not flow in or out of the cartilage, while Eq (15) means that proteins X leak out of the cartilage at rate α.

We take initial conditions in units of g/cm3 as follows:

C(0)= 3×102, Cs(0)=4.5×103, Ch(0)=2.7×104, Cc(0)=0,MP(0)= 2.6×107, L2(0)=1.7×104. (16)

Decrease in the growth of Cc means slower growth in calcified cartilage, which results in slower growth in the progression of OA [24]. We accordingly define the efficacy of the drug X for a treatment beginning at t0 and ending at t by the relative reduction in calcified cartilage:

Efficacy=Cc(t;t0,no drug)Cc(t;t0,X)Cc(t;t0,no drug)(×100)%. (17)

This definition is the same for men and women. But in considering clinical studies with mixed population of men and women, we define E-efficacy as follows:

E-efficacy= # of males# of males and females×(Efficacy for males) +# of females# of males and females×(Efficacy for females) (18)

In Fig 3, we display the profiles of all the model variables for both males and females. From the non-linear system (1)(7) it is difficult to deduce the relative differences between men and women. But, since L2 is decreasing faster in women, both C and Cs profiles are larger for men, as they should be by Eqs (1)(2). However, since C and L2 are both larger for men, it is not clear if C/(1+L2/KL2) is generally larger or smaller for men than for women, which would affect the profiles of Ch and Cc of men versus women. Fig 3 shows, that the profiles of Ch and Cc are larger for women, indicating that women are more susceptible to OA. The profile of MP reflects a sharp postmenopausal effect, where MP first sharply increases and decreases, and then, after age 70, slowly increases.

Fig 3. Profiles of the average density over the strip Ω={0xh} of the model variables for males and females in the control case (no drugs).

Fig 3

2 Drugs

Undenatured type II collagen (UC-II)

Treatment 1 (T1), from [37]

30 males and 22 females, aged between 40 and 75 years, received either UC-II (40 mg/day) or placebo for 90 days. At day 90, the WOMAC score of pain level for the group receiving UC-II was 33% reduced from the group that received placebo. In our simulations, we take the average age of all participants at the clinical study to be 63 years.

Treatment 2 (T2), from [38]

89 males and 97 females, aged between 40 and 75 years, received either UC-II (40 mg/day) or placebo for 90 days. At day 90, the WOMAC score of pain level for the group receiving UC-II was 32% reduced from the group that received placebo. In our simulations, we take the average age of all participants at the clinical study to be 63 years.

Treatment 3 (T3), from [39]

36 males and 69 females, aged between 60 and 80 years, received either UC-II (40 mg/day) or placebo for 90 days. At day 90, the WOMAC score of pain level for the group receiving UC-II was 22.16% reduced from the group that received placebo. In our simulations, we take the average age of all participants at the clinical study to be 70 years.

Treatment 4 (T4), from [40]

39 participants, all females, aged between 40 and 80 years, received either UC-II (40 mg/day) or placebo for 90 days. At day 90, the WOMAC score of pain level for the group receiving UC-II was 25.5% reduced from the group that received placebo. In our simulations, we take the average age of all participants at the clinical study to be 65 years.

Model’s simulations of Treatments T1–T4

In Figs 47 we used the model equations to simulate the treatments T1–T4, and in the last panel of each figure we computed the E-efficacy of the treatment. Table 2 summarizes the data and experiment results of each treatment, and also includes (in the last column) the E-efficacy of the treatment computed by model’s simulations. The protocol of treatment (amount and duration of the drug) was the same in all treatments. Treatment efficacy and treatment-associated pain are two different consequences of a treatment. Note that the E-efficacy (from the model) is very small (0.15–0.22%), because the treatment was taken over a relatively very short time, 90 days. On the other hand, the pain level was significantly reduced (22.16–33%). What is interesting to note is that the E-efficacy is positively correlated to WOMAC reduction: E-efficacy increases when WOMAC reduction increases. On the other hand WOMAC is positively correlated to the biological marker CTX-II, which has been shown to be associated with the severity of OA [41]. Hence the E-efficacy of treatment with UC-II, as derived by the model, is positively correlated to reduction in the severity of OA.

Fig 4. Replicating Treatment 1 [37].

Fig 4

Profiles of the variables with and without the drugs. The extreme right panel provides a zoomed-in view of the profile of the weighted average of Cc around the treatment period (90 days of treatment between years 62.75–63) relative to the male-to-female ratio. The E-efficacy of the treatment is 0.22%.

Fig 7. Replicating Treatment 4 [40].

Fig 7

Profiles of the variables with and without the drugs. The extreme right panel provides a zoomed-in view of the profile of the weighted average of Cc around the treatment period (90 days of treatment between years 64.75–65) relative to the male-to-female ratio. The E-efficacy of the treatment is 0.20%.

Table 2. Summary of the comparison between model simulations and actual WOMAC score reports.

Source M/F AVG age WOMAC reduction E-efficacy (from model) Figure
T3 [39] 36/69 70 22.16% 0.15% Fig 6
T4 [40] 0/39 65 25.5% 0.20% Fig 7
T2 [38] 89/97 63 32% 0.20% Fig 5
T1 [37] 30/22 63 33% 0.22% Fig 4

Fig 5. Replicating Treatment 2 [38].

Fig 5

Profiles of the variables with and without the drugs. The extreme right panel provides a zoomed-in view of the profile of the weighted average of Cc around the treatment period (90 days of treatment between years 62.75–63) relative to the male-to-female ratio. The E-efficacy of the treatment is 0.20%.

Fig 6. Replicating Treatment 3 [39].

Fig 6

Profiles of the variables with and without the drugs. The extreme right panel provides a zoomed-in view of the profile of the weighted average of Cc around the treatment period (90 days of treatment between years 69.75–70) relative to the male-to-female ratio. The E-efficacy of the treatment is 0.15%.

Senolytics

Clinical trials with UBX0101. Clinical trials with senolytic molecule UBX0101 passed phase I, but failed in phase II [42].

Clinical Trials with senolytic drug fisetin. A total of 30 male and 44 female participants, aged between 40 and 80 years, were enrolled in the study [43]. Participants received either fisetin (100 mg/day) or a placebo for two consecutive days, followed by 28-day break, and then an additional two consecutive days of treatment. The primary outcome of the trial was assessment of treatment-emergent adverse events. Secondary outcome, assessed at multiple time points, WOMAC score, patient-reported outcomes (PRO) for knee pain, and levels of proinflammatory markers associated with cellular senescence. The results demonstrated that the study successfully met the primary objective of the clinical trial, with no serious side effects reported over a period of more than 12 months. According to the treatment protocol, the sparingly administered drug was not expected to have a significant impact on slowing the progression of OA. Nevertheless, notable improvements were observed after six months in four key physical function tests related to the study knee; see Table 3. This suggests that cartilage degeneration was slowed during approximately six months. Based on these findings, we conclude that fisetin shows some promise, and may merit advancement to the next phase of clinical trials.

Table 3. Outcome measures from clinical trials with fisetin in [43].

Outcome Measures (since treatment initiation) Placebo Fisetin
Proinflammatory markers associated with senescent factors (0.5, 1.5, 6, 12 months) 10.5, 6.4, 12.0, 8.8 14.0, 8.9, 32.5, 9.2
Change in physical functions of the study knee (Time-up-and-go Test, 6, 12 months) 6.09, 6.0 6.8, 5.93
Change in physical functions of the study knee (Fast 4-meter Walk, 6, 12 months) 21.7, 21.8 22.1, 21.7
Change in physical functions of the study knee (LEK, 6, 12 months) 0.182, 0.180 0.204, 0.204
Change in physical functions of the study knee (Stair-Climbing Test, 6, 12 months) 8.66, 8.78 8.88, 8.66
Change in muscle strength (Isokinetic Dynamometry, 6, 12 months) 83.1, 85.0 81.2, 81.2
WOMAC (6, 12, 18 months) 17.1, 15.4, 17.4 15.2, 19.6, 17.6
Change in the quality of articular cartilage in the study knee with quantitative magnetic resonance imaging (MRI, 6, 12 months) 150.3, 144.8 149.6, 156.1

Clinical studies in-silico

Based on clinical studies with UC-II in T1–T4 and on clinical trial phase 1 with fisetin, we use our model to compute the E-efficacy of each of these treatments alone, and in combination.

In these studies we do not use the standard clinical trials schedule of 3 month on/off but 6 month for UC-II as in clinical studies T1–T4 [3740] and schedules for fisetin based on clinical trials in [44].

In Fig 8, we present a simulation of treatment outcomes using various combinations of UC-II and fisetin, evaluating E-efficacy at age 63 for patients who began treatment at age 62. UC-II is administered daily in cycles of 6 months on and 6 months off, following protocols T1–T4. Fisetin is administered daily at a dose of 100 mg for the first two days of each month, repeated monthly, similarly to [43]. The results indicate that fisetin alone does not significantly improve E-efficacy. However, when combined with UC-II, a synergistic effect is observed, resulting in enhanced E-efficacy compared to UC-II treatment alone.

Fig 8. Treatment with UC-II and fisetin.

Fig 8

UC-II is taken daily at a dose of 40 mg for a period of 6 months, followed by a discontinuation phase of 6 months. Fisetin (D) is taken during the first 2 days of the month, repeated each month.

In Table 4, we present the computed E-efficacy at age 90 for individuals who began treatment at the ages indicated in the top horizontal row and discontinued treatment at ages 50, 55, 60, ..., 85, and 90 (shown in the vertical column). UC-II is administered daily in 6-month cycles (6 months on, 6 months off), while fisetin is administered daily during the first two days of each month, repeated monthly.

Table 4. Efficacy of Cc reduction (in %) at year 90 in males and females with OA treated with UC-II and fisetin (D).

UC-II is administered daily for 6 months, followed by a 6-month break, in repeated cycle. Fisetin is taken during the first two days of each month and repeated monthly. Treatment begins at various times (Treatment Initiation) and ends at different times (Endtime).

(a) Males
Years 50 55 60 65 70 75 80 85 90 Initiation
50 UC-II 0.00
D 0.00
UC-II+D 0.00
55 UC-II 16.43 0.00
D 17.87 0.00
UC-II+D 53.77 0.00
60 UC-II 29.30 19.15 0.00
D 17.82 11.54 0.00
UC-II+D 72.97 51.35 0.00
65 UC-II 41.14 32.97 21.40 0.00
D 17.77 11.50 7.01 0.00
UC-II+D 84.90 70.07 48.50 0.00
70 UC-II 52.46 45.20 35.74 22.93 0.00
D 17.73 11.47 6.98 3.98 0.00
UC-II+D 87.73 80.75 65.66 44.59 0.00
75 UC-II 63.08 56.28 47.73 37.10 23.38 0.00
D 17.69 11.44 6.96 3.96 2.07 0.00
UC-II+D 91.88 84.51 74.55 59.27 39.15 0.00
80 UC-II 72.29 65.72 57.59 47.82 36.22 22.10 0.00
D 17.67 11.42 6.95 3.95 2.06 0.95 0.00
UC-II+D 94.03 86.71 77.68 65.96 50.55 31.79 0.00
85 UC-II 78.84 72.38 64.43 54.99 44.10 31.77 17.90 0.00
D 17.66 11.41 6.94 3.95 2.06 0.95 0.35 0.00
UC-II+D 95.23 88.19 78.87 68.15 54.78 39.24 22.15 0.00
90 UC-II 81.39 74.95 67.04 57.69 46.95 34.97 22.04 9.04 0.00
D 17.65 11.41 6.94 3.94 2.06 0.95 0.35 0.07 0.00
UC-II+D 95.95 88.69 79.58 68.59 55.62 40.83 25.10 9.90 0.00
↑ Endtime
(b) Females
Years 50 55 60 65 70 75 80 85 90 Initiation
50 UC-II 0.00
D 0.00
UC-II+D 0.00
55 UC-II 8.74 0.00
D 4.97 0.00
UC-II+D 23.71 0.00
60 UC-II 18.77 12.92 0.00
D 4.95 2.43 0.00
UC-II+D 42.70 29.93 0.00
65 UC-II 29.95 24.83 16.47 0.00
D 4.94 2.42 1.27 0.00
UC-II+D 59.80 50.31 34.52 0.00
70 UC-II 41.57 36.71 29.43 19.03 0.00
D 4.93 2.41 1.27 0.70 0.00
UC-II+D 73.08 65.11 53.41 35.84 0.00
75 UC-II 52.90 48.15 41.25 32.21 20.34 0.00
D 4.93 2.41 1.26 0.70 0.39 0.00
UC-II+D 82.17 74.90 64.88 51.72 33.83 0.00
80 UC-II 62.96 58.24 51.50 42.94 32.58 19.88 0.00
D 4.92 2.41 1.26 0.70 0.38 0.20 0.00
UC-II+D 87.65 80.69 71.37 59.78 45.86 28.68 0.00
85 UC-II 70.24 65.54 58.85 50.46 40.58 29.30 16.53 0.00
D 4.92 2.40 1.26 0.69 0.38 0.20 0.09 0.00
UC-II+D 90.40 83.56 74.50 63.44 50.66 36.35 20.52 0.00
90 UC-II 73.08 68.39 61.71 53.36 43.57 32.56 20.60 8.49 0.00
D 4.92 2.40 1.26 0.69 0.38 0.20 0.09 0.02 0.00
UC-II+D 90.85 84.19 75.31 64.47 51.91 38.07 23.48 9.30 0.00
↑ Endtime

Table 4 shows that combination of UC-II and fisetin is synergetic, in the sense that

[Efficacy of (UC-II+fisetin)] > [Efficacy of UC-II] + [Efficacy of fisetin]. (19)

Table 4 also shows the importance of starting treatment early, and continuing with it as long as possible. For example, if treatment began at age 50, the Efficacy achieved by age 70 is 87.73% for men and 73.06% for women, but if treatment began at age 60, then Efficacy by age 70 is only 65.66% for men and 53.41% for women.

In OA clinical trials [45], fisetin was administered in one experiment 2 days per month and in another experiment daily, for 90 days.

We can use the model to perform various other trials in-silico with more frequent injections of fisetin than in Table 4. We maintained the treatment protocol with UC-II as specified in Table 4, and evaluated the effect of administering fisetin at a dosage of 100 g/day with increased frequency. In Table 5, fisetin is administered during the first two days of each week, repeated weekly from the initiation of treatment until age 90.

Table 5. Efficacy of Cc reduction (in %) at year 90 in males and females with OA treated with UC-II and fisetin (D).

UC-II is administered daily for 6 months, followed by a 6-month break, in repeated cycle. Fisetin is taken during the first two days of each week and repeated weekly. Treatment begins at various times (Treatment Initiation) and ends at various times (Endtime).

(a) Males
Years 50 55 60 65 70 75 80 85 90 Initiation
50 UC-II 0.00
D 0.00
UC-II+D 0.00
55 UC-II 16.43 0.00
D 18.93 0.00
UC-II+D 55.62 0.00
60 UC-II 29.30 19.15 0.00
D 18.87 12.18 0.00
UC-II+D 74.72 52.82 0.00
65 UC-II 41.14 32.97 21.40 0.00
D 18.81 12.13 7.37 0.00
UC-II+D 86.10 71.41 49.62 0.00
70 UC-II 52.46 45.20 35.74 22.93 0.00
D 18.76 12.10 7.34 4.16 0.00
UC-II+D 89.43 81.79 66.63 45.39 0.00
75 UC-II 63.08 56.28 47.73 37.10 23.38 0.00
D 18.73 12.07 7.32 4.15 2.16 0.00
UC-II+D 91.49 84.93 75.27 59.92 39.67 0.00
80 UC-II 72.29 65.72 57.59 47.82 36.22 22.10 0.00
D 18.70 12.05 7.30 4.14 2.15 0.99 0.00
UC-II+D 94.28 86.96 77.97 66.42 50.93 32.08 0.00
85 UC-II 78.84 72.38 64.43 54.99 44.10 31.77 17.90 0.00
D 18.68 12.03 7.29 4.13 2.14 0.98 0.36 0.00
UC-II+D 95.32 88.37 79.00 68.34 55.05 39.43 22.27 0.00
90 UC-II 81.39 74.95 67.04 57.69 46.95 34.97 22.04 9.04 0.00
D 18.68 12.03 7.29 4.13 2.14 0.98 0.36 0.08 0.00
UC-II+D 96.01 88.82 79.69 68.75 55.84 40.98 25.18 9.92 0.00
↑ Endtime
(b) Females
Years 50 55 60 65 70 75 80 85 90 Initiation
50 UC-II 0.00
D 0.00
UC-II+D 0.00
55 UC-II 8.74 0.00
D 5.28 0.00
UC-II+D 24.61 0.00
60 UC-II 18.77 12.92 0.00
D 5.26 2.57 0.00
UC-II+D 43.99 30.85 0.00
65 UC-II 29.95 24.83 16.47 0.00
D 5.24 2.56 1.34 0.00
UC-II+D 61.17 51.49 35.38 0.00
70 UC-II 41.57 36.71 29.43 19.03 0.00
D 5.23 2.55 1.33 0.73 0.00
UC-II+D 74.28 66.18 54.34 36.53 0.00
75 UC-II 52.90 48.15 41.25 32.21 20.34 0.00
D 5.22 2.54 1.33 0.73 0.40 0.00
UC-II+D 83.11 75.74 65.63 52.36 34.30 0.00
80 UC-II 62.96 58.24 51.50 42.94 32.58 19.88 0.00
D 5.22 2.54 1.32 0.73 0.40 0.21 0.00
UC-II+D 88.36 81.32 71.93 60.25 46.24 28.95 0.00
85 UC-II 70.24 65.54 58.85 50.46 40.58 29.30 16.53 0.00
D 5.21 2.54 1.32 0.73 0.40 0.21 0.09 0.00
UC-II+D 90.78 84.05 74.93 63.79 50.92 36.54 20.63 0.00
90 UC-II 73.08 68.39 61.71 53.36 43.57 32.56 20.60 8.49 0.00
D 5.21 2.54 1.32 0.72 0.40 0.21 0.09 0.02 0.00
UC-II+D 91.27 84.51 75.59 64.75 52.13 38.22 23.56 9.32 0.00
↑ Endtime

In Table 5, the E-efficacy of fisetin alone (D), and consequently that of the combination UC-II+D, exceeds the efficacy observed in Table 4, where fisetin was administered only during the first two days of each month. For example, if the combined therapy began at age 50, the Efficacy achieved by age 70 was 89.43% for men and 74.28% for women, instead of 87.73% and 73.06% when fisetin was administered only the first two days of each month.

We can use the model to compute the Efficacy of other schedules of fisetin administration. For example, in Table 6 fisetin is administered daily for 7 days followed by 7 days off. This treatment is shown to result in very small improvement over Table 5 of Efficacy by age 90.

Table 6. Efficacy of Cc reduction (in %) at year 90 in males and females with OA treated with UC-II and fisetin (D).

UC-II is administered daily for 6 months, followed by a 6-month break, in repeated cycle. Fisetin is taken daily for 7 days, followed by a 7-day break, in a repeating cycle. Treatment begins at various times (Treatment Initiation) and ends at year 90 (Endtime).

(a) Males
Years 50 55 60 65 70 75 80 85 90 Initiation
90 UC-II 81.39 74.95 67.04 57.69 46.95 34.97 22.04 9.04 0.00
D 18.78 12.09 7.32 4.14 2.15 0.98 0.36 0.08 0.00
UC-II+D 95.98 88.84 79.79 68.69 55.88 41.02 25.21 9.93 0.00
↑ Endtime
(b) Females
Years 50 55 60 65 70 75 80 85 90 Initiation
90 UC-II 73.08 68.39 61.71 53.36 43.57 32.56 20.60 8.49 0.00
D 5.20 2.54 1.33 0.73 0.40 0.21 0.09 0.02 0.00
UC-II+D 91.37 84.60 75.64 64.83 52.19 38.26 23.58 9.33 0.00
↑ Endtime

3 Conclusion

Osteoarthritis (OA) is the most prevalent form of joint disease, characterized by the progressive breakdown of articular cartilage and the associated joint pain, stiffness, and inflammation. The cartilage, a spongy tissue composed primarily of water, collagen, and chondrocytes, provides the mechanical buffer that facilitates pain-free joint motion. In OA, this protective tissue gradually deteriorates, and since cartilage has limited regenerative capacity, the damage is effectively irreversible. Understanding the biological progression and treatment dynamics of OA is essential for developing long-term management strategies, especially given its rising prevalence in aging populations.

In this study, we developed a mathematical model of osteoarthritis that captures key components of the cartilage structure in aging population. The model includes chondrocytes in their healthy, senescent, and hypertrophic states, along with calcified cartilage, matrix metalloproteinase-13 (MMP13), and collagen type II. The model was extended to simulate the effects of two therapeutic approaches: intra-articular injection of undenatured collagen type II (UC-II) and treatment with the senolytic drug fisetin. These therapies were evaluated individually and in combination to understand their potential synergy in mitigating disease progression.

We defined the concept of Efficacy of treatment for men and for women, and E-efficacy for a mixed group of men and women. We then demonstrated that the reduced pain associated with treatment by UC-II as documented in several clinical studies is proportional to the E-efficacy as computed by the mathematical model (Table 2).

In Tables 4, 5 and 6, we conducted a series of in-silico clinical studies to explore how the timing, duration, and frequency of treatment affected therapeutic outcomes. These simulations examined treatment initiation at various ages and tracked efficacy over multi-decade time horizons. In Table 4 we followed treatment schedules as in clinical studies, where UC-II was injected continuously for the first 6 months of each year and fisetin was injected in two consecutive days every 28 days. Further simulations explored alternative administration schedules, such as bi-weekly dosing during the first two days of each week (Table 5). These regimens demonstrated higher efficacy compared to monthly schedules, indicating that more frequent drug delivery can enhance therapeutic outcomes.

The main results of this paper are:

  • (i) Efficacy of treatment with UC-II, as derived by the model, is positively correlated to the severity of OA.

  • (ii) Early and continuous treatment significantly improved efficacy, particularly when treatment initiated before age 60; delayed treatment initiation led to notably diminished outcomes.

  • (iii) Efficacy of the combination therapy of UC-II with fisetin consistently exceeded the sum of the treatment Efficacy by UC-II alone and by fisetin alone, highlighting the potential for drug synergy in OA management.

These results reinforce the importance of early treatment and choice of treatment protocol to achieve maximum clinical benefits.

The model has several limitations.

  1. OA is a complex disease involving cartilage degradation, subchondral bone changes, synovial inflammation, and pain mechanisms independent of the structure damage. Accordingly, the severity of OA has been measured by different criteria. In this paper we consider aging patients, above 50 years, for whom the disease is mostly non-inflammatory [8], and we focus on the destruction of the cartilage, measuring the severity of OA in terms of cartilage calcification as in [24]. Cartilage calcification is promoted by hypertrophic chondrocytes, which in health are restricted to the subchondral region. We tacitly assumed that in OA subchondral bone changes result in constant transition from chondrocytes to hypertrophic chondrocytes. In health, the level of hypertrophic chondrocytes is controlled by collagen type II, but in OA the level of collagen type II is highly diminished, due to senescence in the aging patients, hence the concentration of hypertrophic chondrocytes keeps increasing and, correspondingly, also cartilage calcification. We also assumed that pain mechanisms independent of structural damage does not affect the cartilage calcification process.

  2. Most of the model parameters are either taken from published biological data, or estimated using such data. However, some parameters (listed “this work” in Table 7) are unknown, including arguably the most important parameter λChCc, which determines the rate of transition from chondrocyte hyperthophy to cartilage calcification. We performed sensitivity analysis in Fig 9, demonstrating that changes of ±50% of parameters result in expected changes in cartilage calcification. Hence, model simulations can be useful as qualitative results, but only in restricted range (±50%) about the chosen parameters.

  3. Key parameters like drug effectiveness coefficients are not known, as those indicated in Table 7 by “this work”. Hence the results of clinical studies in Tables 4–6 lack experimental validation. When these parameters are revised based on new experiments, the corresponding revised Tables 4–6 would become predictive.

Table 7. Parameters for the model.

Parameters Descriptions Values references
λCCh rate of CCh transition 3.26×102 d−1 [62]est.
λCCs rate of CCs transition 6×103 d−1 [10]est.
λChCc rate of Cc formation 104 d−1 this work
λCsMP rate of production of MP by Cs 6.013×105 d−1 est.
λCL2 rate of production of L2 by C 1.87×108 d−1 est.
μC death rate of C 0.03 d−1 [62]
μCs death rate of Cs at t = 0 0.04 d−1 [35,62]est.
γD dose of fisetin 1.52×106 g/cm3 d−1 [54][55]
γU2 dose of U2 6.1×107 g/cm3 d−1 [37,38]
μL2MP elimination rate of MP by L2 4.897×104 cm3/g d−1 est.
μMPL2 elimination rate of L2 by MP 100.2 cm3/g d−1 est.
μDCs absorption rate of D by Cs 3×102 cm3/g d−1 this work
μCsD elimination rate of Cs by D 103 cm3/g d−1 this work
δC diffusion rate of C, Cs and Ch 8.64×107 cm2 d−1 [52]est.
δCc diffusion rate of Cc 0 cm2 d−1
δMP diffusion rate of MP 6.6×102 cm2 d−1 [4951]est.
δL2 diffusion rate of L2 4.78×102 cm2 d−1 [4951]est.
A source of chondrocytes 1.41×103 g/cm3 d−1 [62]est.
KL2 half-saturation of L2 1.7×104 g/cm3 [59,60]est.
TwMP,TwL2 time-scale in postmenopausal effect cartilage 12 years this work
αD factor of effect of D 2×106 this work
αU2 factor of effect of U2 0.05 this work
αwMP rate of postmenopausal effect on MP 2.5 d−1 this work
αwL2 rate of postmenopausal effect on L2 2.5×104 d−1 this work
h width of articular cartilage 0.2 cm [48]

Fig 9. Parameter sensitivity analysis for the calcified cartilage, Cc, after 1 year.

Fig 9

The p-values are less than 0.01.

The model developed in this paper, while comprehensive, simplifies complex biological processes and does not account for all systemic factors involved in OA or drug metabolism. Patient heterogeneity beyond sex and age – such as genetic variability, comorbid conditions, and lifestyle – was not included but could significantly impact treatment outcomes. Furthermore, our predictions are constrained by the accuracy of parameter estimates and assumptions within the model; in particular, the assumption that progression state of OA is measured simply by the density of calcified cartilage. The present paper can serve as a first step in future work that will involve refining these aspects, incorporating patient-specific data, and validating predictions through experimental and clinical collaborations.

4 Parameter sensitivity analysis

We performed sensitivity analysis with respect to the calcified cartilage density after 1 year, for a male OA patient, for the parameters λCCs, λCCh, λChCc, λCsMP, λCL2, μC, μCs, μCh, μL2MP, μMP, μMPL2, μL2, and A.

The computations were done using Latin Hypercube Sampling/Partial Rank Correlation Coefficient (LHS/PRCC) with a Matlab package by [46,47]. The range for the parameters in the sensitivity analysis was between ±50% of their baseline values in Table 7.

The system dynamics reveal several key relationships governing calcified cartilage density (Cc). The baseline transition rate from chondrocytes to hypertrophic chondrocytes (λCCh) demonstrates the strongest positive correlation with Cc, as it directly controls the production flux through Ch/t. While the calcified cartilage production rate (λChCc) shows expected positive correlation, its effect is secondary to λCCh due to this hierarchical dependence. The chondrocyte production rate (A) also significantly influences Cc by determining the progenitor population available for differentiation.

Conversely, the elimination rate of hypertrophic chondrocytes (μCh) produces the most pronounced negative effect on Cc accumulation. Similar depletion effects occur through increased chondrocyte death rate (μC), which reduces the source population. Additional negative correlations emerge with elevated elimination of senescent chondrocytes (μCs) and increased collagen type II secretion (λCL2). These relationships collectively demonstrate that Cc accumulation depends fundamentally on the balance between chondrocyte production (A) and differentiation (λCCh) versus elimination processes (μC, μCh, μCs).

5 Parameter estimations

The average thickness (width) of the femoral articular cartilage is h = 1.98 mm [48].

Estimates for the diffusion coefficients δX

Diffusion coefficients of MMP and collagen type II. Young [49] established the following formula for estimating the diffusion coefficient δp of protein p:

δp=MV1/3Mp1/3δV, (20)

where MV and δV are respectively the molecular weight and diffusion coefficient of VEGF, Mp is the molecular weight of p, MV=24kDa [50] and δV=8.64×102 cm2 d−1 [51].

The molecular weight of MMP-13 is 53,820 Da, and of collagen type II (COL2A1) is 141,785 Da [50]. Hence,

δMP=6.6×102 cm2 d1,δL2=4.78×102 cm2 d1.

Diffusion coefficients of chondrocytes and calcified cartilage. We take

δC=δCh=δCs=8.64×107 cm2 d1,

the same as for macrophages [52].

The calcified cartilage is hard bone-like connective tissue [53], so we take

δCc=0.

Drugs

Fisetin was used in clinical trials at dose of 100 mg/day [43].

The adult human body average volume is approximately 66×103 g/cm3 [54,55]. Hence the average dose density of fisetin in the body is

γD=100×103 g d16.6×104 cm3=1.52×106 g/cm3 d1.

The terminal half-life of fisetin is approximately 3 hours [56]. Hence

μD=ln20.125=5.55 d1.

We take

ωD=0.01 d1.

The standard daily dose of UC-II is 40 mg [37,38]. Hence

γU2=40×103 g d16.6×104 cm3=6.1×107 g/cm3 d1.

Estimates by equations

In what follows, we shall use the “steady state” equations of the model to estimate some of the unknown parameters; this means the following:

We set the right-hand side of an equation to zero, and take all species X (cells and proteins) in this equation, at their “average” values X = X0 in OA of the control case (no drugs); for example, Cs0=15100C0.

Eq (3): The lifespan of hypertrophic chondrocytes is approximately 1 day [57,58]. We take the half-life to be tCh1/2=0.25 day. Hence,

μCh=ln20.25=2.77 d1.

The level of collagen type II in normal healthy femoral cartilage ranges between 139.9–179.5 μg/ml [59] (Table VII), and it degrades, on the average, by about 6 times in OA at age >70 [60] (Table 1). We take L2=1.7×104 g/cm3 in health,

L2=1.7×1046=2.83×105 g/cm3 in OA,

and

KL2=1.7×104 g/cm3.

In health there is only a small amount Ch [61].

We take Ch0=1%C0, where C0 = 0.027 g/cm3 by [62]; hence Ch0=2.7×104 g/cm3. Assuming a nearly steady state of Eq (3), we have

λCChC011+L2/KL2μChCh00, or equivalently, λCChμChCh0(1+L2/KL2)C0,

where μCh=2.77 d−1. Hence,

λCCh=3.26×102 d1.

Eq (2): Senescent cells have a relatively short half-life, presumably because they are efficiently cleared by immune cells [35]. With μC=0.03 d−1 [62], we take

μCs(0)=0.04 d1,

and, by Eq (5),

μCs(t)=0.041+t/(90 years) d1.

From the average steady state of Eq (2), we get

λCCs=μCsCsC.

Between 30 and 70 years of age there is a fall of 30% from C to Cs [10]. We assume

CsC15100 on average.

Hence,

λCCs=0.15×μCs=6×103 d1.

From steady state of Eq (1), AλCCsCλCChC(12)μCC=0, where μC=0.03 d−1, C0 = 0.027 g/cm3. Hence

A=1.41×103 g/cm3 d1.

Eqs (6) and (7): The half-life of MMP-13 is approximately 70 minutes (0.05 days) [63]. Hence,

μMP=ln20.05=13.86 d1.

The average level of MMP in the synovial fluid is 16.17 ng/ml in rheumatoid arthritis (RA) patient and 0.75 ng/ml in health [64]. We assume that the level of MMP in OA is the same as in RA and take

MP=16.17×109 g/cm3 in OA.

The half-life of cartilage collagen type II is approximately 117 years (42,705 days) [65]. Hence,

μL2=ln242705=1.62×105 d1.

In steady state of Eq (7), we have λCL2C0μMPL2L20MP0μL2L20=0, with C0 = 0.027 g/cm3, L20=2.83×105 g/cm3, μL2=1.62×105 d−1, and MP0=16.17×109 g/cm3 in OA. We assume that the loss rate of active L2 by MP is smaller than the degradation rate of L2, and take

10μMPL2MP0=μL2, so that μMPL2=100.2cm3/g d1.

Then

λCL2=1.87×108 d1.

In steady state of Eq (6), we have λCsMPCs0μL2MPMP0L20μMPMP0=0, with Cs0=0.15C0=0.15×0.027=4.1×103 g/cm3, μMP=13.86 d−1. We assume that the rate by which MP is decreased by blocking L2 is smaller than the degradation rate of MP and take

10μL2MPL20=μMP, so that μL2MP=4.897×104cm3/g d1.

Then

λCsMP=6.013×105 d1.

Data Availability

All relevant data are within the manuscript.

Funding Statement

Research reported in this publication was supported by the National Institute Of General Medical Sciences of the National Institutes of Health under Award Number R16GM154782. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Mobasheri A, Thudium CS, Bay-Jensen A-C, Maleitzke T, Geissler S, Duda GN, et al. Biomarkers for osteoarthritis: current status and future prospects. Best Pract Res Clin Rheumatol. 2023;37(2):101852. doi: 10.1016/j.berh.2023.101852 [DOI] [PubMed] [Google Scholar]
  • 2.Clinic M. Osteoarthritis, symptoms and causes: overview. Mayo Foundation for Medical Education and Research (MFMER). [cited Jan 2025]
  • 3.Ouyang Z, Dong L, Yao F, Wang K, Chen Y, Li S, et al. Cartilage-related collagens in osteoarthritis and rheumatoid arthritis: from pathogenesis to therapeutics. Int J Mol Sci. 2023;24(12):9841. doi: 10.3390/ijms24129841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kodama J, Wilkinson KJ, Iwamoto M, Otsuru S, Enomoto-Iwamoto M. The role of hypertrophic chondrocytes in regulation of the cartilage-to-bone transition in fracture healing. Bone Rep. 2022;17:101616. doi: 10.1016/j.bonr.2022.101616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rim YA, Nam Y, Ju JH. The role of chondrocyte hypertrophy and senescence in osteoarthritis initiation and progression. Int J Mol Sci. 2020;21(7):2358. doi: 10.3390/ijms21072358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Liu Y, Zhang Z, Li T, Xu H, Zhang H. Senescence in osteoarthritis: from mechanism to potential treatment. Arthritis Res Ther. 2022;24(1):174. doi: 10.1186/s13075-022-02859-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lespasio MJ, Piuzzi NS, Husni ME, Muschler GF, Guarino A, Mont MA. Knee Osteoarthritis: A Primer. Perm J. 2017;21:16–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Pella Regional Health Center. Arthritis: Non-inflammatory vs. inflammatory. [cited 2025 Jan]. https://www.pellahealth.org/services/rheumatology/arthritis–non–inflammatory–vs–inflammatory/
  • 9.Knights AJ, Redding SJ, Maerz T. Inflammation in osteoarthritis: the latest progress and ongoing challenges. Curr Opin Rheumatol. 2023;35(2):128–34. doi: 10.1097/BOR.0000000000000923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Loeser RF. Aging and osteoarthritis: the role of chondrocyte senescence and aging changes in the cartilage matrix. Osteoarthritis Cartilage. 2009;17(8):971–9. doi: 10.1016/j.joca.2009.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Xu X, Xu L, Xia J, Wen C, Liang Y, Zhang Y. Harnessing knee joint resident mesenchymal stem cells in cartilage tissue engineering. Acta Biomater. 2023;168:372–87. [DOI] [PubMed] [Google Scholar]
  • 12.Hallett SA, Ono W, Ono N. The hypertrophic chondrocyte: to be or not to be. Histol Histopathol. 2021;36(10):1021–36. doi: 10.14670/HH-18-355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Evans LAE, Pitsillides AA. Structural clues to articular calcified cartilage function: A descriptive review of this crucial interface tissue. J Anat. 2022 October;241(4):875–95. [DOI] [PMC free article] [PubMed]
  • 14.Stücker S, Bollmann M, Garbers C, Bertrand J. The role of calcium crystals and their effect on osteoarthritis pathogenesis. Best Pract Res Clin Rheumatol. 2021;35(4):101722. doi: 10.1016/j.berh.2021.101722 [DOI] [PubMed] [Google Scholar]
  • 15.Yao J, Xu J, Huo Z, Yi Y, Xu D, Wang X, et al. Unveiling the role of hypertrophic chondrocytes in abnormal cartilage calcification: insights into osteoarthritis mechanisms. Eur Cell Mater. 2024;47:265–81. doi: 10.22203/ecm.v047a17 [DOI] [Google Scholar]
  • 16.van der Kraan PM, van den Berg WB. Chondrocyte hypertrophy and osteoarthritis: role in initiation and progression of cartilage degeneration?. Osteoarthritis Cartilage. 2012;20(3):223–32. doi: 10.1016/j.joca.2011.12.003 [DOI] [PubMed] [Google Scholar]
  • 17.Rashid H, Chen H, Javed A. Runx2 is required for hypertrophic chondrocyte mediated degradation of cartilage matrix during endochondral ossification. Matrix Biol Plus. 2021;12:100088. doi: 10.1016/j.mbplus.2021.100088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bernabei I, So A, Busso N, Nasi S. Cartilage calcification in osteoarthritis: mechanisms and clinical relevance. Nat Rev Rheumatol. 2023;19(1):10–27. doi: 10.1038/s41584-022-00875-4 [DOI] [PubMed] [Google Scholar]
  • 19.Sun Y, Mauerhan DR. Meniscal calcification, pathogenesis and implications. Curr Opin Rheumatol. 2012;24(2):152–7. [DOI] [PubMed] [Google Scholar]
  • 20.Lian C, Wang X, Qiu X, Wu Z, Gao B, Liu L, et al. Collagen type II suppresses articular chondrocyte hypertrophy and osteoarthritis progression by promoting integrin β1-SMAD1 interaction. Bone Res. 2019;7:8. doi: 10.1038/s41413-019-0046-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tchetina EV, Squires G, Poole AR. Increased type II collagen degradation and very early focal cartilage degeneration is associated with upregulation of chondrocyte differentiation related genes in early human articular cartilage lesions. J Rheumatol. 2005;32(5):876–86. [PubMed] [Google Scholar]
  • 22.Chen K, Xu M, Lu F, He Y. Development of matrix metalloproteinases-mediated extracellular matrix remodeling in regenerative medicine: a mini review. Tissue Eng Regen Med. 2023;20(5):661–70. doi: 10.1007/s13770-023-00536-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hu Q, Ecker M. Overview of MMP-13 as a promising target for the treatment of osteoarthritis. Int J Mol Sci. 2021;22(4):1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Glasson SS, Chambers MG, Van Den Berg WB, Little CB. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the mouse. Osteoarthritis Cartilage. 2010;18 Suppl 3:S17-23. doi: 10.1016/j.joca.2010.05.025 [DOI] [PubMed] [Google Scholar]
  • 25.Hayes WC, Keer LM, Herrmann G, Mockros LF. A mathematical analysis for indentation tests of articular cartilage. J Biomech. 1972;5(5):541–51. doi: 10.1016/0021-9290(72)90010-3 [DOI] [PubMed] [Google Scholar]
  • 26.Landinez-Parra NS, Garzón-Alvarado DA, Vanegas-Acosta JC. A phenomenological mathematical model of the articular cartilage damage. Comput Methods Programs Biomed. 2011;104(3):e58-74. doi: 10.1016/j.cmpb.2011.02.003 [DOI] [PubMed] [Google Scholar]
  • 27.Kapitanov GI, Wang X, Ayati BP, Brouillette MJ, Martin JA. Linking cellular and mechanical processes in articular cartilage lesion formation: a mathematical model. Front Bioeng Biotechnol. 2016;4:80. doi: 10.3389/fbioe.2016.00080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Campbell K, Naire S, Kuiper JH. A mathematical model of cartilage regeneration after chondrocyte and stem cell implantation - I: the effects of growth factors. J Tissue Eng. 2019;10:2041731419827791. doi: 10.1177/2041731419827791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cope PJ, Ourradi K, Li Y, Sharif M. Models of osteoarthritis: the good, the bad and the promising. Osteoarthritis Cartilage. 2019;27(2):230–9. doi: 10.1016/j.joca.2018.09.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Eschweiler J, Horn N, Rath B, Betsch M, Baroncini A, Tingart M, et al. The biomechanics of cartilage-an overview. Life (Basel). 2021;11(4):302. doi: 10.3390/life11040302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Abu Owida H. Recent biomimetic approaches for articular cartilage tissue engineering and their clinical applications: narrative review of the literature. Adv Orthop. 2022;2022:8670174. doi: 10.1155/2022/8670174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Keil D. Anatomy of the Knee. 2021. https://yoganatomy.com
  • 33.Gilmer G, Bean AC, Iijima H, Jackson N, Thurston RC, Ambrosio F. Uncovering the “riddle of femininity” in osteoarthritis: a systematic review and meta-analysis of menopausal animal models and mathematical modeling of estrogen treatment. Osteoarthritis Cartilage. 2023;31(4):447–57. doi: 10.1016/j.joca.2022.12.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sasono B, Rantam FA, Suroto H, Notobroto HB, Am A. The effect of estrogen on type 2 collagen levels in the joint cartilage of post-menopausal murine subjects. J hard tissue biol. 2019;28(3):245–50. doi: 10.2485/jhtb.28.245 [DOI] [Google Scholar]
  • 35.Childs BG, Durik M, Baker DJ, van Deursen JM. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat Med. 2015;21(12):1424–35. doi: 10.1038/nm.4000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Li Y, Tian X, Luo J, Bao T, Wang S, Wu X. Molecular mechanisms of aging and anti-aging strategies. Cell Commun Signal. 2024;22(1):285. doi: 10.1186/s12964-024-01663-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Crowley DC, Lau FC, Sharma P, Evans M, Guthrie N, Bagchi M. Safety and efficacy of undenatured type II collagen in the treatment of osteoarthritis of the knee: a clinical trial. Int J Med Sci. 2009;6(6):312–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lugo JP, Saiyed ZM, Lane NE. Efficacy and tolerability of an undenatured type II collagen supplement in modulating knee osteoarthritis symptoms: a multicenter randomized, double-blind, placebo-controlled study. Nutr J. 2016;15(14). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sadigursky D, Magnavita VFS, Sá CKCD, Monteiro HDS, Braghiroli OFM, Matos MAA. Undenatured collagen type ii for the treatment of osteoarthritis of the knee. Acta Ortop Bras. 2022;30(2):e240572. doi: 10.1590/1413-785220223002240572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Santana ETN, da Cunha Machado S, Brandao Lima VN, DeSantana Filho VJ, Dos Santos Maciel LY, de Farias Neto JP, et al. Comparison between exercise therapy and non-hydrolyzed collagen (UC-II) in functionality and quality of life in women with knee osteoarthritis: a randomized controlled clinical trial. Wien Klin Wochenschr. 2023;135(11–12):291–300. [DOI] [PubMed] [Google Scholar]
  • 41.Bihlet AR, Byrjalsen I, Bay-Jensen A-C, Andersen JR, Christiansen C, Riis BJ, et al. Associations between biomarkers of bone and cartilage turnover, gender, pain categories and radiographic severity in knee osteoarthritis. Arthritis Res Ther. 2019;21(1):203. doi: 10.1186/s13075-019-1987-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lane N, Hsu B, Visich J, Xie B, Khan A, Dananberg J. A phase 2, randomized, double-blind, placebo-controlled study of senolytic molecule UBX0101 in the treatment of painful knee osteoarthritis. Osteoarthritis and Cartilage. 2021;29:S52–3. doi: 10.1016/j.joca.2021.02.077 [DOI] [Google Scholar]
  • 43.Evans TA. Senolytic drugs attenuate osteoarthritis-related articular cartilage degeneration: a clinical trial (NCT04210986, protocol no 2019-16). Steadman Philippon Research Institute. 2024. https://www.ngram.com/trials/NCT04210986#record–history
  • 44.University of Kentucky. Trial: NCT04770064. https://cdekpharmacypurdueedu/trial/NCT04770064/
  • 45.Center for Research Innovation in Biotechnology (CRIB) College of Pharmacy. Targeting senescence to reduce osteoarthritis pain and cartilage breakdown (ROPE). https://cdek.pharmacy.purdue.edu/trial/NCT04770064/
  • 46.Kirschner DE. Uncertainty and sensitivity functions and implementation. 2007. http://malthus.micro.med.umich.edu/lab/usadata/
  • 47.Marino S, Hogue IB, Ray CJ, Kirschner DE. A methodology for performing global uncertainty and sensitivity analysis in systems biology. J Theor Biol. 2008;254(1):178–96. doi: 10.1016/j.jtbi.2008.04.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Guo J, Yan P, Qin Y, Liu M, Ma Y, Li J. Automated measurement and grading of knee cartilage thickness: a deep learning-based approach. Front Med. 2024;11:1337993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Young ME. Estimation of diffusion coefficients of proteins. Biotech Bioeng. 1980;XXII:947–55. [Google Scholar]
  • 50.Hornbeck PV, Zhang B, Murray B, Kornhauser JM, Latham V, Skrzypek E. PhosphoSitePlus 2014 : mutations, PTMs and recalibrations. Nucleic Acids Res. 2015;43(Database issue):D512-20. doi: 10.1093/nar/gku1267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Liao K-L, Bai X-F, Friedman A. Mathematical modeling of interleukin-27 induction of anti-tumor T cells response. PLoS One. 2014;9(3):e91844. doi: 10.1371/journal.pone.0091844 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Friedman A, Siewe N. Chronic hepatitis b virus and liver fibrosis: a mathematical model. PLoS ONE. 2018;13(4):1–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Arkill KP, Winlove CP. Solute transport in the deep and calcified zones of articular cartilage. Osteoarthritis Cartilage. 2008;16(6):708–14. doi: 10.1016/j.joca.2007.10.001 [DOI] [PubMed] [Google Scholar]
  • 54.Sendroy J Jr, Collison HA. Determination of human body volume from height and weight. J Applied Physiol. 1966;21(1). [DOI] [PubMed] [Google Scholar]
  • 55.Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R, Frabetti F, et al. An estimation of the number of cells in the human body. Ann Hum Biol. 2013;40(6):463–71. doi: 10.3109/03014460.2013.807878 [DOI] [PubMed] [Google Scholar]
  • 56.Kumar RM, Kumar H, Bhatt T, Jain R, Panchal K, Chaurasiya A, et al. Fisetin in cancer: attributes, developmental aspects, and nanotherapeutics. Pharmaceuticals (Basel). 2023;16(2):196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yokoyama Y, Kameo Y, Sunaga J, Maki K, Adachi T. Chondrocyte hypertrophy in the growth plate promotes stress anisotropy affecting long bone development through chondrocyte column formation. Bone. 2024;182:117055. doi: 10.1016/j.bone.2024.117055 [DOI] [PubMed] [Google Scholar]
  • 58.Farnum CE, Tinsley M, Hermanson JW. Forelimb versus hindlimb skeletal development in the big brown bat, Eptesicus fuscus: functional divergence is reflected in chondrocytic performance in Autopodial growth plates. Cells Tissues Organs. 2008;187(1):35–47. doi: 10.1159/000109962 [DOI] [PubMed] [Google Scholar]
  • 59.Hollander AP, Heathfield TF, Webber C, Iwata Y, Bourne R, Rorabeck C, et al. Increased damage to type II collagen in osteoarthritic articular cartilage detected by a new immunoassay. J Clin Invest. 1994;93(4):1722–32. doi: 10.1172/JCI117156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hollander AP, Pidoux I, Reiner A, Rorabeck C, Bourne R, Poole AR. Damage to type II collagen in aging and osteoarthritis starts at the articular surface, originates around chondrocytes, and extends into the cartilage with progressive degeneration. J Clin Invest. 1995;96(6):2859–69. doi: 10.1172/JCI118357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chen N, Wu RWH, Chan WCW, Chan D. Hypertrophic chondrocytes at the junction of musculoskeletal structures. Bone Reports. 2023;19:101698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Moise N, Friedman A. Rheumatoid arthritis - a mathematical model. J Theor Biol. 2019;461:17–33. doi: 10.1016/j.jtbi.2018.10.039 [DOI] [PubMed] [Google Scholar]
  • 63.Yamamoto K, Okano H, Miyagawa W, Visse R, Shitomi Y, Santamaria S, et al. MMP-13 is constitutively produced in human chondrocytes and co-endocytosed with ADAMTS-5 and TIMP-3 by the endocytic receptor LRP1. Matrix Biol. 2016;56:57–73. doi: 10.1016/j.matbio.2016.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Stojanovic SK, Stamenkovic BN, Cvetkovic JM, Zivkovic VG, Apostolovic MRA. Matrix metalloproteinase-9 level in synovial fluid-association with joint destruction in early rheumatoid arthritis. Medicina (Kaunas). 2023;59(1):167. doi: 10.3390/medicina59010167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Verzijl N, DeGroot J, Thorpe SR, Bank RA, Shaw JN, Lyons TJ, et al. Effect of collagen turnover on the accumulation of advanced glycation end products. J Biol Chem. 2000;275(50):39027–31. doi: 10.1074/jbc.M006700200 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Xindie Zhou

6 Aug 2025

PONE-D-25-32495Modeling treatment of osteoarthritis with standard therapy and senolytic drugsPLOS ONE

Dear Dr. Siewe,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by Sep 20 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Xindie Zhou

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. Please note that PLOS One has specific guidelines on code sharing for submissions in which author-generated code underpins the findings in the manuscript. In these cases, we expect all author-generated code to be made available without restrictions upon publication of the work. Please review our guidelines at https://journals.plos.org/plosone/s/materials-and-software-sharing#loc-sharing-code and ensure that your code is shared in a way that follows best practice and facilitates reproducibility and reuse.

3. Please update your submission to use the PLOS LaTeX template. The template and more information on our requirements for LaTeX submissions can be found at http://journals.plos.org/plosone/s/latex.

4. Thank you for stating the following financial disclosure: [Research reported in this publication was supported by the National Institute Of General Medical Sciences of the National Institutes of Health under Award Number R16GM154782. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.]. 

Please state what role the funders took in the study.  If the funders had no role, please state: ""The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.""

If this statement is not correct you must amend it as needed.

Please include this amended Role of Funder statement in your cover letter; we will change the online submission form on your behalf.

5. We note that your Data Availability Statement is currently as follows: [All relevant data are within the manuscript and its Supporting Information files.]

Please confirm at this time whether or not your submission contains all raw data required to replicate the results of your study. Authors must share the “minimal data set” for their submission. PLOS defines the minimal data set to consist of the data required to replicate all study findings reported in the article, as well as related metadata and methods (https://journals.plos.org/plosone/s/data-availability#loc-minimal-data-set-definition).

For example, authors should submit the following data:

- The values behind the means, standard deviations and other measures reported;

- The values used to build graphs;

- The points extracted from images for analysis.

Authors do not need to submit their entire data set if only a portion of the data was used in the reported study.

If your submission does not contain these data, please either upload them as Supporting Information files or deposit them to a stable, public repository and provide us with the relevant URLs, DOIs, or accession numbers. For a list of recommended repositories, please see https://journals.plos.org/plosone/s/recommended-repositories.

If there are ethical or legal restrictions on sharing a de-identified data set, please explain them in detail (e.g., data contain potentially sensitive information, data are owned by a third-party organization, etc.) and who has imposed them (e.g., an ethics committee). Please also provide contact information for a data access committee, ethics committee, or other institutional body to which data requests may be sent. If data are owned by a third party, please indicate how others may request data access.

6. Thank you for stating the following in the Acknowledgments Section of your manuscript: [Research reported in this publication was supported by the National Institute Of General

Medical Sciences of the National Institutes of Health under Award Number R16GM154782.

The content is solely the responsibility of the authors and does not necessarily represent the

official views of the National Institutes of Health.]

We note that you have provided funding information that is not currently declared in your Funding Statement. However, funding information should not appear in the Acknowledgments section or other areas of your manuscript. We will only publish funding information present in the Funding Statement section of the online submission form.

Please remove any funding-related text from the manuscript and let us know how you would like to update your Funding Statement. Currently, your Funding Statement reads as follows: [Research reported in this publication was supported by the National Institute Of General Medical Sciences of the National Institutes of Health under Award Number R16GM154782. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.]. 

Please include your amended statements within your cover letter; we will change the online submission form on your behalf.

7. If the reviewer comments include a recommendation to cite specific previously published works, please review and evaluate these publications to determine whether they are relevant and should be cited. There is no requirement to cite these works unless the editor has indicated otherwise. 

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: This review highlights the manuscript's ambitious attempt to model a complex disease but identifies several critical issues that would need to be addressed before publication. The primary concerns center on model validation, parameter uncertainty, and the disparity between the mathematical and clinical applicability of the results.

The authors have undertaken a challenging modeling problem, but the work would benefit significantly from more rigorous validation approaches and better integration with clinical outcomes. The very low efficacy values (0.15-0.22%) compared to substantial clinical improvements (22-33% reduction in WOMAC scores) suggest that the model may not be capturing the most clinically relevant aspects of OA progression and treatment response.

Summary

This manuscript presents a mathematical model of osteoarthritis (OA) progression that incorporates cellular senescence and treatment effects of undenatured collagen type II (UC-II) and the senolytic drug fisetin. The authors develop separate models for males and females, validate them against clinical trial data, and conduct in silico trials to assess treatment efficacy and synergy.

Novel Integration: The incorporation of cellular senescence mechanisms into OA modeling is timely and biologically relevant, addressing a significant gap in current therapeutic approaches.

Sex-Specific Modeling: The differentiation between male and female models, particularly accounting for postmenopausal effects on collagen degradation and MMP-13 levels, is a valuable contribution.

Clinical Validation: The authors attempt to validate their model against four published clinical trials (T1-T4), demonstrating a correlation between computed E-efficacy and reported improvements in WOMAC scores. Extensive parameter estimation based on literature values and steady-state assumptions demonstrates thorough model development.

Major Concerns

1. Model Assumptions and Simplifications

Critical Issue: The fundamental assumption that OA progression is solely measured by calcified cartilage density (Cc) is oversimplified and potentially problematic. OA is a complex, multifactorial disease involving:

Cartilage degradation beyond calcification, Synovial inflammation, Subchondral bone changes, and Pain mechanisms independent of structural damage. However, authors acknowledge this limitation but don't adequately address how this affects model validity.

The model contains numerous parameters (>25) estimated from disparate sources, steady-state assumptions, and author assumptions; however, no sensitivity analysis is provided. For example, the choice of λ_{Ch, Cc} = 10^4 d^{-1} appears arbitrary without justification.

The correlation between E-efficacy and WOMAC scores shows very low efficacy values (0.15-0.22%) for treatments that demonstrated improvements of 22-33% in WOMAC scores. This raises questions about the model's predictive validity and whether it captures meaningful therapeutic effects.

Furthermore, the relationship between senescent cell elimination and improvement in OA progression is assumed rather than established. Clinical trial data for fisetin show minimal effects, yet the model predicts synergy. The correlation between model predictions and clinical outcomes is weak.

Minor Issues

The authors claim model validation based on four trials, but the correlation is weak.

Treatment protocols in Tables 4-6 don't match standard clinical practice, and the 6-month on/off UC-II protocol lacks clinical justification.

No confidence intervals or uncertainty quantification provided.

No statistical comparison between treatment groups

Key parameters like drug effectiveness coefficients lack experimental validation

Reviewer #2: I have extensively read through the manuscript titled, "Modeling treatment of osteoarthritis with standard therapy and senolytic drugs", and I am highly impressed with the presentation of the work. The manuscript presents a comprehensive mathematical model of osteoarthritis (OA) incorporating biological mechanisms such as chondrocyte senescence, hypertrophy, calcified cartilage formation, and the roles of collagen type II and MMP13. It evaluates treatment effects using UC-II and senolytic drugs (fisetin), both independently and in combination, providing a novel in-silico framework validated against clinical trial data. The work is well-structured, technically rigorous, and contributes valuable computational insight into OA treatment planning.

However, here are some few suggested issues related to the work to improve it:

1. The study uses calcified cartilage (Cc) as a proxy for OA severity and compares it to WOMAC pain scores. The authors should explicitly discuss the biological or clinical rationale for using Cc density as a surrogate for clinical pain outcomes, even if only qualitatively.

2. While the mixed-type boundary condition is mathematically acceptable, more biological interpretation or justification would strengthen the modeling assumptions, especially for protein/drug diffusion.

3. Consider including a sensitivity analysis to identify which parameters have the greatest influence on model outcomes, particularly on efficacy metrics. Even a local sensitivity analysis would improve robustness.

4. In some places, there are unclear or potentially redundant terms in equations (e.g., repeated ∂Ch/∂t in Eq. 2.3). Carefully review all equations for typographical consistency.

5. Ensure that all variables shown in the schematic diagram (Figure 1) are labeled and defined in the caption or immediately adjacent text.

6. Define all symbols the first time they appear (e.g., KL2 in Eq. 2.1), even if they are listed in Table 1. This will improve readability for interdisciplinary readers.

Reviewer #3: The article its good and the results it's correct so we can accept this article with minor correction

Minor correction

The Authors must add recent references

10.3934/MATH.2023821

10.3390/sym15020286

10.1155/2022/2138165

10.1155/2022/5468696

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

Decision Letter 1

Xindie Zhou

4 Sep 2025

Modeling treatment of osteoarthritis with standard therapy and senolytic drugs

PONE-D-25-32495R1

Dear Dr. Siewe,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager® and clicking the ‘Update My Information' link at the top of the page. For questions related to billing, please contact billing support.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Xindie Zhou

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewer #1:

Reviewer #2:

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: I Don't Know

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: No

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The authors don't address how clinicians or researchers should interpret or apply model predictions given the weak correlation with established clinical measures.

The work shows promise as a foundation for OA modeling, but requires additional development to demonstrate its value for clinical decision-making or therapeutic development.

Reviewer #2: (No Response)

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

**********

Acceptance letter

Xindie Zhou

PONE-D-25-32495R1

PLOS ONE

Dear Dr. Siewe,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

You will receive further instructions from the production team, including instructions on how to review your proof when it is ready. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few days to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

You will receive an invoice from PLOS for your publication fee after your manuscript has reached the completed accept phase. If you receive an email requesting payment before acceptance or for any other service, this may be a phishing scheme. Learn how to identify phishing emails and protect your accounts at https://explore.plos.org/phishing.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Xindie Zhou

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    Attachment

    Submitted filename: Response to Reviewers_OA.pdf

    pone.0332763.s001.pdf (95.1KB, pdf)

    Data Availability Statement

    All relevant data are within the manuscript.


    Articles from PLOS One are provided here courtesy of PLOS

    RESOURCES