Skip to main content
PLOS One logoLink to PLOS One
. 2021 Jun 1;16(6):e0252620. doi: 10.1371/journal.pone.0252620

TGF-β inhibition can overcome cancer primary resistance to PD-1 blockade: A mathematical model

Nourridine Siewe 1,*, Avner Friedman 2
Editor: Joseph Najbauer3
PMCID: PMC8168900  PMID: 34061898

Abstract

Immune checkpoint inhibitors have demonstrated, over the recent years, impressive clinical response in cancer patients, but some patients do not respond at all to checkpoint blockade, exhibiting primary resistance. Primary resistance to PD-1 blockade is reported to occur under conditions of immunosuppressive tumor environment, a condition caused by myeloid derived suppressor cells (MDSCs), and by T cells exclusion, due to increased level of T regulatory cells (Tregs). Since TGF-β activates Tregs, TGF-β inhibitor may overcome primary resistance to anti-PD-1. Indeed, recent mice experiments show that combining anti-PD-1 with anti-TGF-β yields significant therapeutic improvements compared to anti-TGF-β alone. The present paper introduces two cancer-specific parameters and, correspondingly, develops a mathematical model which explains how primary resistance to PD-1 blockade occurs, in terms of the two cancer-specific parameters, and how, in combination with anti-TGF-β, anti-PD-1 provides significant benefits. The model is represented by a system of partial differential equations and the simulations are in agreement with the recent mice experiments. In some cancer patients, treatment with anti-PD-1 results in rapid progression of the disease, known as hyperprogression disease (HPD). The mathematical model can also explain how this situation arises, and it predicts that HPD may be reversed by combining anti-TGF-β to anti-PD-1. The model is used to demonstrate how the two cancer-specific parameters may serve as biomarkers in predicting the efficacy of combination therapy with PD-1 and TGF-β inhibitors.

1 Introduction

Immune checkpoint inhibitors, introduced in recent years, have demonstrated impressive clinical response in cancer patients, although resistance may develop over time. But some patients do not respond at all to checkpoint blockade, exhibiting, what is called, primary resistance. Mechanisms of adaptive resistance to PD-1 blockade and potential therapies to overcome it are reviewed in [15], and of primary resistance in [35]. In particular, primary resistance is reported to occur under conditions of immunosuppressive tumor environment, including effective T cells exclusion [4, 5]. Such environment is often caused by increased level of T regulatory cells (Tregs). Indeed, as reported in [6, 7], PD-1 expression balance between effective T cells and Tregs predicts the efficacy of PD-1 blockade therapy. In clinical study of patients with melanoma, PD-1 blockade resulting in decline of PD1+ Tregs predicted more favorable outcome [8]

In some cancer patients, treatment with anti-PD-1 resulted in rapid progression of tumor, known as hyperprogression disease (HPD) [810]. Recent reviews of HPD in cancer patients appeared in [1113], and, of biomarkers for HPD, in [14]. Although the mechanism of HPD is unknown, it has been noted that HPD is associated with increased levels of MDSC and Treg cells [11, 14]. Motivated by the observation that HPD occurs in approximately 10% of anti-PD-1 monoclonal anti-body (mAb)-treated advanced gastric cancer patients, Kamada et al. [15] conducted mice experiments with gastric cancer. They demonstrated that PD-1 blockade activated and expanded tumor infiltration of PD-1+ Tregs to overwhelm tumor PD-1+ effective T cells, as cancer underwent rapid progression.

TGF-β is a pleiotropic cytokine that could suppress immune response by regulating Tregs [16]. Hence TGF-β blockade is likely to enhance immune-checkpoint therapy [17]. Mariathasan et al. [18] and Tauriello et al. [19] identified TGF-β signaling in tumor microenvironment as a determinant of tumor T cell role in affecting poor response to PD-1/PD-L1 blockade. They demonstrated, in mouse models, that combining TGF-β inhibition with immune checkpoint blockade induces complete and durable response to otherwise unresponsive tumor; see also reveiw article [20]. Sow et al. [21] found that combined inhibition of TGF-β signaling and PD-L1 is differentially effective in mouse model.

Streel et al. [22] and Martin et al. [23] have recently demonstrated, in several mouse models, that TGF-β inhibition overcomes primary resistance to PD-1 blockade. More precisely, in some cancers, PD-1 inhibition does not decrease tumor volume, but, in combination with anti-TGF-β, PD-1 blockade significantly improves outcome of treatment compared to treatment with anti-TGF-β alone. In this paper, we develop a mathematical model that explains these experimental results in [15, 22, 23] in terms of two cancer-specific parameters that may serve as cancer biomarkers.

The model is based on two important observations:

  • (i) TGF-β (Tβ) inhibits the killing rate of cancer cells by CD8+ T cells [24]; we represent this inhibition by a factor 1/(1+ζTβTβ), for some constant ζTβ.

  • (ii) The complex Q = PD-1/PD-L1 induces change from pro-inflammatory CD4+ T cells (T1) to regulatory T cells (Tr) [25, 26], at rate modeled by λT1TrT1Q/(KQ+Q), where KQ and λT1Tr are constants.

Anti-PD-1 increases the activation of CD8+ T cells (T8). On the other hand, Tβ contributes to the proliferation of Tr [2728], possibly resulting in only minimal increase (T8).

PD-1 blockade increases the proliferation rate of T1. If T1 were fixed, the loss rate λT1TrT1Q/(KQ+Q) of T1 (to Tr) will also decrease. But since the proliferation of T1 has increased by the PD-1 blockade, the product T1 Q/(KQ+Q) may conceivably increase; in this case the rate of change λT1TrT1Q/(KQ+Q) from T1 to Tr will increase, and, if λT1Tr is sufficiently large, the Tr inhibition of T8 may result in loss of T8, and thus in hyperprogression of cancer.

Myeloid cells play an important immunosuppressive role in the tumor microenvironment. They include MDSCs, M2 macrophages and M2-like TAMs (tumor associated macrophages) [29]. MDSCs secrete IL-10 [30, 31] and TGF-β [28, 32, 33]; M2 macrophages secrete IL-10 [34, 35], and TAMs and M2 macrophages secrete TGF-β [36]. For simplicity we shall represent these three types of myeloid populations by one variable, designated by M2, and will refer to it as MDSC or M2.

The mathematical model is represented by a system of partial differential equations within the tumor compartment. The species in the model include immune cells, CD8+ and CD4+-Th1 T cells, Tregs, immunosuppressive M2 macrophages and pro-inflammatory macrophages M1, and dendritic cells. They also include cytokines that play important role in the interactions among immune cells and cancer cells: CCL2 (MCP-1) and interleukins IL-2, IL-10 and IL-12. CCL2 is produced by cancer cells [37], and it attracts MDSCs into the tumor compartment [3840]. IL-2 is produced by Th1 cells [41] and it enhances the proliferation of T1 and T8, but also Tr, so its effect in clinical trials is not always predictable [42]. IL-12 is produced by dendritic cells and it activates T1 and T8 cells [43]. IL-10 is produced by MDSCs, M2 macrophages and cancer cells [30, 31]. Both IL-10 and Tr inhibit the activation of T1 and T8 by IL-12 [31]. The cancer-specific parameters λT1Tr and ζTβ play a critical role in the model simulations, and are adjusted in order to establish agreement with the experimental results of Streel et al. [22], Martin et al. [23], and Kamada et al. [15]. The model is then used to demonstrate how various other choices of these two parameters determine the efficacy of combination therapy with anti-PD-1 and anti-TGF-β, and how these parameters may serve as prediction biomarkers.

2 Mathematical model

The mathematical model is based on the network shown in Fig 1. Table 1 lists the variables of the model in units of g/cm3. We assume that all species Xi, (i = 1, …, n) are dispersing (or diffusing) with a coefficient δXi, and are dying (or degrading) at rate μXi; cells also undergo advection velocity u that is associated with internal pressure in the tumor compartment, see S1 File. We write the equation for cells Xi in the form

Xit+·(uXi)-δXi2Xi=FXi(X1,,Xn)

where ∇2 is the Laplace operator ∇⋅grad, or j=132xj2. In modeling the structure of FXi we use, for simplicity, the linear mas conservation law, that is, if Xj+XkXm then the rate by which Xm is formed, or Xj is lost, is mXj Xk where m is a positive parameter. In a process where Xi is activated by cytokine Xj, Xj represents molecules that are bound and internalized by Xi, and this internalization may be limited owing to the limited rate of receptor recycling. We then represent the rate of activation by the Michaelis-Menten law mXi(Xj/(K+Xj)) for some positive parameters m, K. A term of the form mXi/(1+Xj/K) means that Xj inhibits the growth of Xi. Finally, an expression of the form ∇⋅(Xi χXj) means that Xi is moving by chemotaxis in the direction of the gradient of chemoattractant Xj with chemotactic force χ, where χ is a positive parameter.

Fig 1. Network describing the interactions between cells and cytokines under treatment with anti-PD-1 and anti-TGF-β.

Fig 1

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

Variables Descriptions Variables Descriptions
M1 density of M1 macrophages M2 density of MDSCs
D density of dedritic cells T1 density of CD4+ T/Th1 cells
T8 density of CD8+ T cells Tr density of Treg cells
C density of cancer cells
I2 concentration of IL-2 I10 concentration of IL-10
I12 concentration of IL-12 P concentration of CCL2 (MCP-1)
Tβ concentration of TGF-β PD concentration of PD-1
PL concentration of PD-L1 Q concentration of PD-1/PD-L1
A1 concentration of anti-PD-1 Aβ concentration of anti-TGF-β

2.1 Equation for tumor cells (C)

We assume a logistic growth for cancer cells with carrying capacity CM, to account for space competition among these cells. Cancer cells are killed by CD8+ T cells, a processed inhibited by the pleiotropic cytokine TGF-β [24], represented by the factor 11+ζTβTβ. We write the equation for C in the following form:

Ct+·(uC)-δC2C=λCC(1-CCM)Growthofcancercells-μT8C1+ζTβTβT8CkillingbyT8-μCCdeath. (1)

2.2 Equation for M1 macrophages (M1)

The equation for M1 macrophages has the following form:

M1t+·(uM1)-δM2M1=λM1M0PKP+PactivationbyCCL2-·(χPM1P)chemoattractionbyCCL2+λM2M1M2I12KI12+I12M2M1byIL-12-λM1M2M1TβKTβ+TβM1M2byTGF-β-μM1M1death (2)

where the first term on the right-hand side represents a source of macrophages differentiated from monocytes that are activated by CCL2 (P) and the second term represents chemoattraction of M1 by CCL2 [44]. The third and fourth terms on the right-hand side represent phenotype changes from M2 to M1 induced by IL-12, and from M1 to M2 induced by TGF-β [44, 45].

2.3 Equation for MDSCs (M2)

Tumor recruits macrophages and “educates” them to become tumor-associated-macrophages (TAMs), which behave like MDSCs [46, 47]; MDSCs are chemotactically attracted by CCL2 [3840]. The equation for M2 is given by:

M2t+·(uM2)-δM2M2=λM2M0PKP+PactivationbyCCL2-·(χPM2P)chemoattratcedbyCCL2-λM2M1M2I12KI12+I12M2M1byIL-12+λM1M2M1TβKTβ+TβM1M2byTGF-β-μM2M2death. (3)

2.4 Equation for CD4+ T/Th1 cells (T1)

The pleiotropic cytokine TGF-β contributes to the development of naive CD4+ T cells, T10 [48]. Naive CD4+ T cells differentiate into Th1 cells under IL-12 inducement [41, 49], and this process is inhibited by IL-10 and Tregs. The proliferation of activated CD4+ T cells is enhanced by IL-2 [42]. Activation and proliferation of T1 cells are inhibited by the complex PD-1/PD-L1 (Q), represented by the factor 11+Q/K^TQ. The complex Q also mediates phenotype change from Th1 cells to Treg cells [25, 26], by a factor λT1TrQKQ+Q; we consider the parameter λT1Tr to be cancer-specific. Hence T1 satisfies the following equation:

T1t+·(uT1)advection-δT2T1diffusion=(λT1I12T10(1+TβKTβ+Tβ)Tβ-augmentedactivationI12KI12+I12activationbyIL-12·11+I10/K^TI10inhibitionbyIL-10·11+Tr/K^TTrinhibitionbyTregs+.λT1I2T1I2KI2+I2IL-2-inducedproliferation)×11+Q/K^TQinhibitionbyQ-λT1TrT1QKQ+QQ-inducedT1Trtransition-μT1T1death. (4)

2.5 Equation for activated CD8+ T cells (T8)

The cytokine TGF-β contributes to the development of inactive CD8+ T cells, T80 [48]. Inactive CD8+ T cells are activated by IL-12 [41, 49], and this process is resisted by IL-10 and Treg cells [27, 31]. IL-2 enhances the proliferation of activated CD8+ T cells [42]. Both processes of activation and proliferation are inhibited by PD-1/PD-L1, by the factor 11+Q/K^TQ. Hence, T8 satisfies the following equation:

T8t+·(uT8)-δT2T8=(λT8I12T80(1+TβKTβ+Tβ)Tβ-augmentedactivationI12KI12+I12activationbyIL-12·11+I10/K^TI10inhibitionbyIL-10·11+Tr/K^TTrinhibitionbyTregs+.λT8I2T8I2KI2+I2IL-2-inducedproliferation)×11+Q/K^TQinhibitionbyQ-μT8T8death. (5)

2.6 Equation for Tregs (Tr)

Naive CD4+ T cells differentiate into Tregs under activation by Fox3+ transcription factor, a process enhanced by TGF-β [27, 28]. The activated Tregs are recruited into tumor by tumor-derived immunosuppressive cytokines IL-6 and CCL2 (P) [3840]; for simplicity, we represent both cytokines by CCL2. IL-2 enhances the proliferation of Tregs within the tumor [42] Representing this chemoattraction by ∇⋅(χP TrP), we get the following equation for Tr:

Trt+·(uTr)-δT2Tr=λTrTβT10TβKTβ+TβTβ-enhancednaiveTcellsactivation+λT1TrT1QKQ+QQ-inducedT1Trtransition+λTrI2TrI2KI2+I2IL-2-inducedproliferation-·(χPTrP)chemoattractionbyCCL2/MCP-1-μTrTrdeath, (6)

where the second term in the right-hand side is the same as in Eq (4).

2.7 Equation for TGF-β (Tβ)

When anti-TGF-β drug is applied, TGF-β is depleted at a rate proportional to Aβ, and the equation for Tβ takes the following form:

Tβt-δTβ2Tβ=λTβCC+λTβM2M2+λTβTrTrsecretionbyC,M2andTr-μTβTβdegradation-μAβTβTβAβdepletionbyanti-TGF-β. (7)

The equations for I2, I10, I12, P, as well as the equations for PD and PL are given in S1 File, and we take

Q=σPDPL, (8)

for some parameter σ.

2.8 Equation for anti-PD-1 (A1)

In mice experiments in [23], anti-PD-1 was injected, intraperitoneally twice a week, begining t0 days after tumor cells implantation, and ending at day t1; in [22] the drug was administered daily. We approximate the effective source of the drug by taking it to be a constant, γA1, so that

cA1(t)={γA1,ift0tt10,otherwise. (8)

The drug is depleted in the process of blocking PD-1, so that

A1t-δA12A1=cA1(t)source-μPDA1PDA1depletionthroughblockingPD-1-μA1A1degradation (9)

2.9 Equation for anti-TGF-β (Tβ)

In [22, 23], anti-TGF-β was administered weekly for the same periods t0tt1 as in (8). We again approximate the effective level of the drug by taking

cAβ(t)={γAβ,ift0tt10,otherwise (10)

where γAβ is some constant. The drug Aβ is depleted in the process of blocking TGF-β, so that

Aβt-δAβ2Aβ=cAβ(t)source-μTβAβTβAβdepletionthroughblockingTGF-β-μAβAβdegradation (11)

2.10 Equation for cells velocity (u)

The velocity u is determined by the condition that the combined density of all cells in the tumor compartment is constant; see S1 File.

To simplify the computations, we assume that the tumor is spherical and that all the densities and concentrations are radially symmetric, that is, functions of (r, t), 0 ≤ rR(t) where r = R(t) is the boundary of the tumor, and that u = u(r, t)er, where er is the unit radial vector.

2.11 Equation for free boundary (R)

We assume that the free boundary r = R(t) moves with the velocity of cells, so that

dR(t)dt=u(R(t),t). (12)

We complement the system by prescribing initial and boundary conditions; see S1 File.

3 Results

All the computations were done using Python 3.5.4. The parameter values of the model equations are estimated in and are listed in S1 File. The technique used in the simulations is also described in S1 File.

3.1 Mouse models and simulations

We define the efficacy of treatment by

efficacy=tumorvolumewithnotreatment-tumorvolumeundertreatmenttumorvolumewithnotreatment×100%, (13)

where both volumes are measured at the last day of treatment. We refer to efficacy as the relative difference (in tumor volume) of treatment to no treatment, in percentage. Negative efficacy means that treatment resulted in increase (rather than decrease) in tumor volume.

Streel et al. [22] and Martin et al. [23] performed mice experiments with different types of cancer, treated with combinations of anti-PD-1 and anti-TGF-β. In Streel et al. [22] (Fig. 2b), mice were implanted with colon cancer cells and treatment began 6 days after infection. The tumor volume in each mouse was measured regularly for 45 days and reported accordingly. They found that there was almost no reduction in the tumor size when treatment was with anti-PD-1 alone, but the tumor volume reduced significantly when anti-PD-1 was combined with anti-TGF-β. Our simulations in Fig 2A show the volume of the tumor in the cases of no treatment and treatment with various combinations of anti-PD-1 and anti-TGF-β. We see that while anti-PD-1 as a single agent does not reduce the cancer volume growth, when given in combination with anti-TGF-β, the growth of the tumor volume is significantly decreased; this is in agreement with Fig. 2b in [22]

Fig 2. Tumor volume under various combinations with anti-PD-1 and anti-TGF-β.

Fig 2

The “%” represents the difference, in volume, of treatment to no treatment, in percentage. All parameters are as in S1 File, with γA1=108g/cm3d and γAβ=2×106g/cm3d. (A) Colon cancer: treatment starts at day 6 which corresponds to the schedule in [22]. (B) Melanoma cancer: treatment starts at day 14 as in [22]. (C) Breast cancer: treatment starts at day 14 as in [23]. (D) Bladder cancer: treatment starts at day 14 which corresponds to the schedule in [23]. (E) Gastric cancer: treatment starts at day 15 which corresponds to the schedule in [15].

In the experiments conducted by Martin et al. [23], mice were implanted with cells from bladder cancer, melanoma or breast cancer, and then treated with anti-PD-1 as a single agent, or with combination of anti-PD-1 and anti-TGF-β. Starting treatment at day 14 post-infection, Martin et al. found, as in [22], that in the case of breast cancer ([23] Fig 4B, 4C) and bladder cancer ([23] Fig 4G, 4H), with anti-PD-1 alone there was hardly any reduction in the tumor volume, but in combination with anti-TGF-β, anti-PD-1 reduced tumor volume significantly; Fig 2C and 2D are in agreement with these results. On the other hand, in the case of melanoma ([23] Fig 4D, 4E), there was primary resistance to anti-PD-1; Fig 2B is in agreement with this result. Note that the cancer-specific parameter ζTβ in Fig 2B is much smaller than the corresponding parameter in Fig 2A, 2D and 2E.

Note that the parameters λT1Tr and ζTβ are the same in Fig 2A, 2C and 2D, but the profiles are taken for different time durations (45, 56 and 32 days, respectively), and this accounts for the somewhat different impressions one may get of the tumor volume growth.

In mice experiments with gastric cancer, Kamada et al. [15] administered anti-PD-1 as a single agent and compared the tumor volume in this case to the tumor volume in the control (no-drug) case. They observed that the tumor volume with anti-PD-1 exceeded the tumor volume in the control case (Fig 6B, 6C in [15]). The simulations in Fig 2E show the same qualitative results. Notice that in these simulations, the parameter ζTβ is the same as in Fig 2A, 2C and 2D, but λT1Tr is much larger than in these figures.

Fig 2E shows also the effect of anti-PD-1 on tumor treated with anti-TGF-β: In the first few weeks, tumor volume slightly increases (hyperprogression of cancer) but later on it decreases, and by day 45 it is significantly decreased under the combined therapy.

3.2 Tumor volume hyperprogression

The simulations in Fig 2 suggest that hyperprogression of cancer under PD-1 inhibition depends on the parameters λT1Tr and ζTβ. Fig 3 shows tumor volume at day 45 for pairs of parameters (ζTβ,λT1Tr) in the range 0<ζTβ<1.5×106 cm3/g, 0<λT1Tr<5×104d1. The color column scales the efficacy, that is, the percentage of increase/decrease of tumor volume relative to the control case; the drug level is taken to be γA1=108g/cm3d. We see that (i) negative efficacy (hyperprogression) increases with both λT1Tr and ζTβ, and (ii) efficacy is positive when either λT1Tr or ζTβ is small. A monotone decreasing curve of the form λT1Tr=f(ζTβ) separates the regions of positive and negative efficacies.

Fig 3. Combined effect of cancer-specific parameters λT1Tr and ζTβ, under treatment with anti-PD-1, at γA1=108g/cm3d.

Fig 3

The color column indicates relative difference of the tumor volume at day 45. Negative values represent parameter ranges of tumor hyperprogression.

Kamada et al. [15] (Fig. 5F) also measured the level of Tregs under treatment with anti-PD-1 as single agent, and compared it with the corresponding level of Tregs in the control case. They found that Tregs level increased by 1/3 more than their corresponding level in the control case. The simulations in Fig 4 show the same level of increase of Tregs under treatment with anti-PD-1, with cancer-specific parameters ζTβ=4×106cm3/g and λT1Tr=103d1. Fig 4 also shows that the Tregs level is very low under treatment with anti-TGF-β, but it increases significantly (although it remains below the control case) in combination with anti-PD-1.

Fig 4. Tregs levels in all treatment combinations with anti-PD-1 and anti-TGF-β.

Fig 4

The bar plots represent the density of Tregs in the control, anti-PD-1 only, anti-TGF-β only, and anti-PD-1+anti-TGF-β cases. Tregs increase with anti-PD-1 as single agent, decrease significantly with anti-TGF-β as single agent, and decrease (but remains below the control case) when anti-PD-1 is combined with anti-TGF-β.

3.3 Efficacy maps

In order to see how the cancer-specific parameters affect the efficacy of treatment, we took 9 pairs (λT1Tr,ζTβ) as in Fig 5 and for each pair, we simulated the model under combination therapy with (γAβ,γA1) that vary in the region 0<γA1<108g/cm3d 0<γAβ<2×106g/cm3d. Then, in Fig 5, we plotted the efficacy of treatment after 45 days. Note that the values of λT1Tr increase along each row, and the values of ζTβ increase along each column. The ranges of λT1Tr and ζTβ, and the ranges of γA1 and γAβ include the values that appear in Fig 2.

Fig 5. Efficacy map, combination of anti-PD-1 with anti-TGF-β.

Fig 5

We vary the cancer-specific parameters λT1Tr{6,6×102,6×103}d1 and ζTβ{4×105,22×105,40×105}cm3/g, and plot efficacy maps for the combination anti-PD-1+anti-TGF-β with doses γA1 between 0<γA1<108g/cm3d and γAβ between 0 < γβ < 2×10−6 g/cm3⋅d, respectively. The color columns indicate the relative difference of the tumor at day 45. Negative values represent anti-PD-1 and anti-TGF-β dose ranges of tumor hyperprogression.

From Fig 5 we see that (i) for any combination (γAβ,γA1), the efficacy increases when λT1Tr and ζTβ are decreased. (ii) For large values of λT1Tr and ζTβ, tumor progression is likely to occur. (iii) For small values of λT1Tr and ζTβ, the efficacy increases as γA1 and γAβ are increased. We also see that efficacy always increases if γAβ is increased. This is not surprising, since if γAβ is increased, the Tβ is decreased and, hence, the killing rate of C by T8, which is proportional to 1/(1+ζTβTβ) is increased.

On the other hand, as seen in the last two columns of Fig 5, for fixed large γAβ, there is an interval (γA1,γA1+) such that the efficacy is decreasing as γA1 increases in this interval. To explain this situation we note that if γA1 is increased then T1 and T8 are increased, but also Tr is increased, at rate λT1TrT1, and hence Tβ is also increased (by Eq (7)). It follows that the killing rate of C, which is proportional to T8/(1+ζTβTβ), may either increase or decrease. Fig 5 shows that, as γA1 increases, this factor decreases as long as γA1 remains in an intermediate interval (γA1,γA1+), and is increased elsewhere.

4 Conclusion

Therapeutic antibodies that block PD-1/PD-L1 induce robust and durable response in some cancer patients, negative response in some patients [12, 14], and no response at all in others [18, 50]. Since substantial proportion of patients have little or no benefits, while treatment with these drugs are costly and might have associated toxicity [51], biomarkers which are likely to predict response rate to PD-1/PD-L1 blockade are highly desirable [51, 52]. You et al. [53] summarizes (in Table 1) clinical outcome of predictive biomarkers for PD-1/PD-L1 blockade, while asserting the need for reliable biomarkers to ensure rational use of this checkpoint blockade. In the present paper we identified two cancer-specific parameters, λT1Tr and ζTβ, and used them in a mathematical model to predict the response rate to treatment with anti-PD-1 as single agent and in combination with anti-TGF-β.

Our simulations, in Figs 2 and 4, show agreement with the experimental results (in mice) reported in [15, 22, 23]. We also show, in Fig 3, that under treatment with anti-PD-1 alone, as the parameters λT1Tr and ζTβ increase the progression of cancer increases, while treatment does not result in progression of cancer if either λT1Tr or ζTβ is small.

The parameters λT1Tr and ζTβ can be viewed as biomarkers, predicting the following:

  • (i) for any combination (γAβ,γA1), the efficacy increases when λT1Tr and ζTβ are decreased.

  • (ii) For large values of λT1Tr and ζTβ, tumor progression is likely to occur.

  • (iii) For small values of λT1Tr and ζTβ, the efficacy increases as γA1 and γAβ are increased.

We also found that while efficacy always inceases when γAβ is increased, there are regions in the (γAβ,γA1)-plane such that efficacy is decreased as γA1 increases: these regions consist of points {(γAβ,γA1):γA1<γA1<γA1+}, where γA1 and γA1+ depend on γAβ.

The mathematical model presented in this paper has several limitations:

  1. We assumed that the densities of immature, or naive, immune cells remain constant throughout the progression of the cancer and that dead cells are quickly removed from the tumor.

  2. In estimating production parameters we made a steady state assumption in some of the differential equations.

  3. Although our mathematical model does not presume any geometric form of the tumor, for simplicity, the simulations have been carried out only in the case of spherical tumor. We note however that spherical cancer models have been used in research as an intermediate between in vitro cancer line cultures and in vivo cancer [54]. Furthermore, spheroids mirror the 3D cellular context and therapeutically relevant pathophysiological gradient of in vivo tumors [55].

Biomarkers are characteristics of the body and they are critical in order to diagnose a disease and/or to measure the effect of a drug on the patient. In the present paper, based on mice experiments, we developed a mathematical model which demonstrates, depending on two parameters, how primary resistance to anti-PD-1 can be overcome by anti-TGF-beta. These parameters may serve as new cancer biomarkers, but our results will first need to be validated by clinical studies.

Supporting information

S1 File. TGF-β inhibition can overcome cancer primary resistance to PD-1 blockade: A mathematical model.

Model equations (Section 1 in S1 File), parameter estimates (Section 2 in S1 File), parameter sensitivity analysis (Section 3 in S1 File), numerical methods used (Section 4 in S1 File) and the parameter values (Tables 1 and 2 in S1 File).

(PDF)

Data Availability

All relevant data are within the manuscript and its Supporting information files.

Funding Statement

This research was supported by the Dean’s Research Initiative Grant #15874 of the College of Science at Rochester Institute of Technology. This work was also supported by the Mathematical Biosciences Institute of The Ohio State University. There was no additional external funding received for this study.

References

  • 1. Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am Soc Clin Oncol Educ Book. 2019;39:147–164. 10.1200/EDBK_240837 [DOI] [PubMed] [Google Scholar]
  • 2. Ren D, Hua Y, Yu B, Ye X, He Z, Li C, et al. Predictive biomarkers and mechanisms underlying resistance to PD1/PD-L1 blockade cancer immunotherapy. Mol Cancer. 2020;19(1):19. 10.1186/s12943-020-1144-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Nowicki TS, Hu-Lieskovan S, Ribas A. Mechanisms of Resistance to PD-1 and PD-L1 Blockade. Cancer J. 2018;24(1):47–53. 10.1097/PPO.0000000000000303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Lei Q, Wang D, Sun K, Wang L, Zhang Y. Resistance Mechanisms of Anti-PD1/PDL1 Therapy in Solid Tumors. Front Cell Dev Biol. 2020;8(672):1–16. 10.3389/fcell.2020.00672 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Haibe Y, El Husseini Z, El Sayed R, Shamseddine A. Resisting Resistance to Immune Checkpoint Therapy: A Systematic Review. Int J Mol Sci. 2020;21(17):6176. 10.3390/ijms21176176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression—implications for anticancer therapy. Nat Rev Clin Oncol. 2019;16(6):356–371. 10.1038/s41571-019-0175-7 [DOI] [PubMed] [Google Scholar]
  • 7. Kumagai S, Togashi Y, Kamada T, Sugiyama E, Nishinakamura H, Takeuchi Y, et al. The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat Immunol. 2020;21(11):1346–1358. 10.1038/s41590-020-0769-3 [DOI] [PubMed] [Google Scholar]
  • 8. Champiat S, Dercle L, Ammari S, Massard C, Hollebecque A, Postel-Vinay S, et al. Hyperprogressive Disease Is a New Pattern of Progression in Cancer Patients Treated by Anti-PD-1/PD-L1. Clin Cancer Res. 2016;23(8):1920–1928. 10.1158/1078-0432.CCR-16-1741 [DOI] [PubMed] [Google Scholar]
  • 9. Champiat S, Ferrara R, Massard C, Besse B, Marabelle A, Soria JC, et al. Hyperprogressive disease: recognizing a novel pattern to improve patient management. Nat Rev Clin Oncol. 2018;15(12):748–762. 10.1038/s41571-018-0111-2 [DOI] [PubMed] [Google Scholar]
  • 10. Kato S, Goodman A, Walavalkar V, Barkauskas DA, Sharabi A, Kurzrock R. Hyperprogressors after Immunotherapy: Analysis of Genomic Alterations Associated with Accelerated Growth Rate. Clin Cancer Res. 2017;23(15):4242–4250. 10.1158/1078-0432.CCR-16-3133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Popat V, Gerber DE. Hyperprogressive disease: a distinct effect of immunotherapy? J Thorac Dis. 2019;11(Suppl 3):S262–S265. 10.21037/jtd.2019.01.97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Sabio E, Chan TA. The good, the bad, and the ugly: hyperprogression in cancer patients following immune checkpoint therapy. Genome Med. 2019;11(1):43. 10.1186/s13073-019-0661-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Denis M, Duruisseaux M, Brevet M, Dumontet C. How Can Immune Checkpoint Inhibitors Cause Hyperprogression in Solid Tumors? Front Immunol. 2020;11(492):1–8. 10.3389/fimmu.2020.00492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Wang X, Wang F, Zhong M, Yarden Y, Fu L. The biomarkers of hyperprogressive disease in PD-1/PD-L1 blockage therapy. Mol Cancer. 2020;19(81):1–15. 10.1186/s12943-020-01200-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, et al. PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. PNAS. 2019;116(20):9999–10008. 10.1073/pnas.1822001116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Bai X, Yi M, Jiao Y, Chu Q, Wu K. Blocking TGF-β Signaling To Enhance The Efficacy Of Immune Checkpoint Inhibitor. Onco Targets Ther. 2019;12:9527–9538. 10.2147/OTT.S224013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Löffek S. Transforming of the Tumor Microenvironment: Implications for TGF- Inhibition in the Context of Immune-Checkpoint Therapy. J Oncol. 2018;2018(9732939):1–9. 10.1155/2018/9732939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGF-β attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. nature. 2018;554(7693):544–548. 10.1038/nature25501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. nature. 2018;554(7693):538–543. 10.1038/nature25492 [DOI] [PubMed] [Google Scholar]
  • 20. Ganesh K, Massaguké J. TGFβ Inhibition and Immunotherapy: Checkmate. Immunity. 2018;48(4):62–628. 10.1016/j.immuni.2018.03.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Sow HS, Ren J, Camps M, Ossendorp F, Ten Dijke PJ. Combined Inhibition of TGF-β Signaling and the PD-L1 Immune Checkpoint Is Differentially Effective in Tumor Models. Cells. 2019;8(4):320. 10.3390/cells8040320 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. de Streel G, Bertrand C, Chalon N, Liénart S, Bricard O, Lecomte S, et al. Selective inhibition of TGF-β1 produced by GARP-expressing Tregs overcomes resistance to PD-1/PD-L1 blockade in cancer. Nature Communications. 2020;11(4545):1–15. 10.1038/s41467-020-17811-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Martin CJ, Datta A, Littlefield C, Kalra A, Chapron C, Wawersik S, et al. Selective inhibition of TGFβ1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape. Sci Transl Med. 2020;12(536):8456. 10.1126/scitranslmed.aay8456 [DOI] [PubMed] [Google Scholar]
  • 24. Thomas DA, Massagué J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8(5):369–380. 10.1016/j.ccr.2005.10.012 [DOI] [PubMed] [Google Scholar]
  • 25. Cai J, Wang D, Zhang G, Guo X. The Role Of PD-1/PD-L1 Axis In Treg Development And Function: Implications For Cancer Immunotherapy. Onco Targets Ther. 2019;12:8437–8445. 10.2147/OTT.S221340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Amarnath S, Mangus CW, Wang JC, Wei F, He A, Kapoor V, et al. The PDL1-PD1 axis converts human TH1 cells into regulatory T cells. Sci Transl Med. 2011;3(111):111–120. 10.1126/scitranslmed.3003130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Whiteside TL. The role of regulatory t cells in cancer immunology. Immunotargets Ther. 2015;4:159–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Umansky V, Blattner C, Gebhardt C, Utikal J. The Role of Myeloid-Derived Suppressor Cells (MDSC) in Cancer Progression. Vaccines (Basel). 2016;4(36):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Fang Z, Wen C, Chen X, Yin R, Zhang C, Wang X, et al. Myeloid-derived suppressor cell and macrophage exert distinct angiogenic and immunosuppressive effects in breast cancer. Oncotarget. 2017;14881(33):54173–54186. 10.18632/oncotarget.17013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Yaseen MM, Abuharfeil NM, Darmani H, Daoud A. Mechanisms of immune suppression by myeloid-derived suppressor cells: the role of interleukin-10 as a key immunoregulatory cytokine. Open Biol. 2017;10(9):200111. 10.1098/rsob.200111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Perrot CY, Javelaud D, Mauviel A. Insights into the transforming growth factor-beta signaling pathway in cutaneous melanoma. Ann Dermatol. 2013;25(2):135–144. 10.5021/ad.2013.25.2.135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32(1):19–25. 10.1016/j.it.2010.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Cantelli G, Crosas-Molist E, Georgouli M, Sanz-Moreno V. TGFb-induced transcription in cancer. Semin Cancer Biol. 2016;42:60–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Qi L, Yu H, Zhang Y, Zhao D, Lv P, Zhong Y, et al. IL-10 secreted by M2 macrophage promoted tumorigenesis through interaction with JAK2 in glioma. Oncotarget. 2016;7(44):71673. 10.18632/oncotarget.12317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Steen EH, Wang X, Balaji S, Butte MJ, Bollyky PL, Keswani SG. The role of the anti-inflammatory cytokine interleukin-10 in tissue fibrosis. Adv Wound Care (New Rochelle). 2020;9(4):184–198. 10.1089/wound.2019.1032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Liu Z, Kuang W, Zhou Q, Zhang Y. TGF-β1 secreted by M2 phenotype macrophages enhances the stemness and migration of glioma cells via the SMAD2/3 signalling pathway. Int J Mol Med. 2018;42(6):3395–3403. 10.3892/ijmm.2018.3923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Labbe K, Danialou G, Gvozdic D, Demoule A, Divangahi M, Boyd JH, et al. Inhibition of monocyte chemoattractant protein-1 prevents diaphragmatic inflammation and maintains contractile function during endotoxemia. Critica Care. 2010;14(R187):1–11. 10.1186/cc9295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Kawakami Y, Yaguchi T, Sumimoto H, Kudo-Saito C, Iwata-Kajihara T, Nakamura S, et al. Improvement of cancer immunotherapy by combining molecular targeted therapy. Front Oncol. 2013;3(136):1–7. 10.3389/fonc.2013.00136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Jobe NP, Rösel D, Dvorankova B, Kodet O, Lacina L, Mateu R, et al. Simultaneous blocking of IL-6 and IL-8 is sufficient to fully inhibit CAF-induced human melanoma cell invasiveness. Histochem Cell Biol. 2016;146(2):205–217. 10.1007/s00418-016-1433-8 [DOI] [PubMed] [Google Scholar]
  • 40. Oelkrug C, Ramage JM. Enhancement of t cell recruitment and infiltration into tumours. Clin Exp Immunol. 2014;178(1):1–8. 10.1111/cei.12382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer. 2013;4(1):36–44. 10.7150/jca.5046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Choudhry H, Helmi N, Abdulaal WH, Zeyadi M, Zamzami MA, Wu W, et al. Prospects of IL-2 in Cancer Immunotherapy. BioMed Res. 2018;2018(9056173). 10.1155/2018/9056173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Vacaflores A, Freedman SN, Chapman NM, Houtman JCD. Pretreatment of activated human CD8 T cells with IL-12 leads to enhanced TCR-induced signaling and cytokine production. Mol Immunol. 2017;81:1–15. 10.1016/j.molimm.2016.11.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Obeid E, Nanda R, Fu YX, Olopade OI. The role of tumor-associated macrophages in breast cancer progression (review). Int J Oncol. 2013;43(1):5–12. 10.3892/ijo.2013.1938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers. 2014;6(3):1670–1790. 10.3390/cancers6031670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Chen D, Roda JM, Marsh CB, Eubank TD, Friedman A. Hypoxia inducible factors-mediated inhibition of cancer by gm-csf: a mathematical model. Bull Math Biol. 2012;74(11):2752–2777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Eubank TD, Roberts RD, Khan M, Curry JM, Nuovo GJ, Kuppusamy P, et al. Granulocyte macrophage colony-stimulating factor inhibits breast cancer growth and metastasis by invoking an anti-angiogenic program in tumor-educated macrophages. Cancer Res. 2009;69(5):2133–2140. 10.1158/0008-5472.CAN-08-1405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Dahmani A, Delisle JB. TGF-β in T Cell Biology: Implications for Cancer Immunotherapy. Cancers (Basel). 2014;10(194):1–21. 10.3390/cancers10060194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Janco JMT, Lamichhane P, Karyampudi L, Knutson KL. Tumor-infiltrating dendritic cells in cancer pathogenesis. J Immunol. 2015;194(7):2985–2991. 10.4049/jimmunol.1403134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Sun JY, Zhang D, Wu S, Xu M, Zhou X, Lu XJ, et al. Resistance to PD-1/PD-L1 blockade cancer immunotherapy: mechanisms, predictive factors, and future perspectives. Biomarker Research. 2020;8(35):1–10. 10.1186/s40364-020-00212-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, et al. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer. 2018;17(1):129. 10.1186/s12943-018-0864-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Cottrell TR, Taube JM. PD-L1 and Emerging Biomarkers in Immune Checkpoint Blockade Therapy. Cancer J. 2018;24(1):41–46. 10.1097/PPO.0000000000000301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. You W, Shang B, Sun J, Liu X, Su L, Jiang S. Mechanistic insight of predictive biomarkers for antitumor PD‑1/PD‑L1 blockade: A paradigm shift towards immunome evaluation (Review). Oncol Rep. 2020;42(2):424–437. 10.3892/or.2020.7643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Weiswald B, Bellet D, Dangles-Marie V. Spherical cancer models in tumor biology. Neoplasia. 2015;17(1):1–15. 10.1016/j.neo.2014.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Klieser W, Kunz-Schughart LA. Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol. 2010;148(1):3–15. 10.1016/j.jbiotec.2010.01.012 [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

S1 File. TGF-β inhibition can overcome cancer primary resistance to PD-1 blockade: A mathematical model.

Model equations (Section 1 in S1 File), parameter estimates (Section 2 in S1 File), parameter sensitivity analysis (Section 3 in S1 File), numerical methods used (Section 4 in S1 File) and the parameter values (Tables 1 and 2 in S1 File).

(PDF)

Data Availability Statement

All relevant data are within the manuscript and its Supporting information files.


Articles from PLoS ONE are provided here courtesy of PLOS

RESOURCES