Skip to main content

Advertisement

Log in

Modeling tumour heterogeneity of PD-L1 expression in tumour progression and adaptive therapy

  • Published:
Journal of Mathematical Biology Aims and scope Submit manuscript

Abstract

Although PD-1/PD-L1 inhibitors show potent and durable anti-tumour effects in some refractory tumours, the response rate in overall patients is unsatisfactory, which in part due to the inherent heterogeneity of PD-L1. In order to establish an approach for predicting and estimating the dynamic alternation of PD-L1 heterogeneity during cancer progression and treatment, this study establishes a comprehensive modelling and computational framework based on a mathematical model of cancer cell evolution in the tumour-immune microenvironment, and in combination with epigenetic data and overall survival data of clinical patients from The Cancer Genome Atlas. Through PD-L1 heterogeneous virtual patients obtained by the computational framework, we explore the adaptive therapy of administering anti-PD-L1 according to the dynamic of PD-L1 state among cancer cells. Our results show that in contrast to the continuous maximum tolerated dose treatment, adaptive therapy is more effective for PD-L1 positive patients, in that it prolongs the survival of patients by administration of drugs at lower dosage.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11

Similar content being viewed by others

References

  • Ai L, Xu A, Xu J (2020) Roles of PD-1/PD-L1 pathway: signaling, cancer, and beyond. In: Xu J (eds) Regulation of cancer immune checkpoints. Advances in Experimental Medicine and Biology, p 1248

  • Avanzini S, Kurtz DM, Chabon JJ, Moding EJ, Hori SS, Gambhir SS, Alizadeh AA, Diehn M, Reiter JG (2020) A mathematical model of ctDNA shedding predicts tumor detection size. Sci Adv 6(50):4308

    Google Scholar 

  • Bassanelli M, Sioletic S, Martini M, Giacinti S, Viterbo A, Staddon A, Liberati F, Ceribelli A (2018) Heterogeneity of PD-L1 expression and relationship with biology of NSCLC. Anticancer Res 38(7):3789–3796

    Google Scholar 

  • Bertucci F, Finetti P, Perrot D, Leroux A, Collin F, Le Cesne A, Coindre J-M, Blay J-Y, Birnbaum D, Mamessier E (2017) PDL1 expression is a poor-prognosis factor in soft-tissue sarcomas. Oncoimmunology 6(3):1278100

    Google Scholar 

  • Biernacki C, Celeux G, Govaert G (2003) Choosing starting values for the EM algorithm for getting the highest likelihood in multivariate Gaussian mixture models. Comput Stat Data Anal 41(3–4):561–575

    MathSciNet  MATH  Google Scholar 

  • Billon E, Finetti P, Bertucci A, Niccoli P, Birnbaum D, Mamessier E, Bertucci F (2019) PDL1 expression is associated with longer postoperative, survival in adrenocortical carcinoma. Oncoimmunology 8(11):1655362

    Google Scholar 

  • Brahmer JR, Tykodi SS, Chow LQ, Hwu W-J, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K et al (2012) Safety and activity of anti-pd-l1 antibody in patients with advanced cancer. N Engl J Med 366(26):2455–2465

    Google Scholar 

  • Bylicki O, Paleiron N, Rousseau-Bussac G, Chouaïd C (2018) New PDL1 inhibitors for non-small cell lung cancer: focus on pembrolizumab. OncoTargets Ther 11:4051

    Google Scholar 

  • Chen Y, Lai X (2022) Modeling the effect of gut microbiome on therapeutic efficacy of immune checkpoint inhibitors against cancer. Math Biosci 350:108868

    MathSciNet  MATH  Google Scholar 

  • Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, Sabedot TS, Malta TM, Pagnotta SM, Castiglioni I et al (2016) TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res 44(8):71

    Google Scholar 

  • Das P, Gopalan V, Islam M, Pillai S (2022) The role of cancer stem cells in disease progression and therapy resistance, In: Frontiers in Stem Cell and Regenerative Medicine Research, Vol 10, pp 42–60. https://doi.org/10.2174/9789811464706122100004

  • de Pillis LG, Radunskaya AE, Wiseman CL (2005) A validated mathematical model of cell-mediated immune response to tumor growth. Cancer Res 65(17):7950–7958

    Google Scholar 

  • Dempster AP, Laird NM, Rubin DB (1977) Maximum likelihood from incomplete data via the EM algorithm. J R Stat Soc Ser B Methodol 39(1):1–22

    MathSciNet  MATH  Google Scholar 

  • Dritschel H, Waters SL, Roller A, Byrne HM (2018) A mathematical model of cytotoxic and helper T cell interactions in a tumor microenvironment. Lett Biomath 5(S1):66

    Google Scholar 

  • Du H, Xie S, Guo W, Che J, Zhu L, Hang J, Li H (2021) Development and validation of an autophagy-related prognostic signature in esophageal cancer. Ann Transl Med 9(4):66

    Google Scholar 

  • El-Khoueiry AB, Melero I, Yau TC, Crocenzi TS, Kudo M, Hsu C, Choo S, Trojan J, Welling T, Meyer T et al (2018) Impact of antitumor activity on survival outcomes, and nonconventional benefit, with nivolumab (NIVO) in patients with advanced hepatocellular carcinoma (aHCC): subanalyses of CheckMate-040. Am Soc Clin Oncol 6:66

    Google Scholar 

  • Filippova N, Yang X, An Z, Nabors LB, Pereboeva L (2018) Blocking PD1/PDL1 interactions together with MLN4924 therapy is a potential strategy for glioma treatment. J Cancer Sci Ther 10(8):190

    Google Scholar 

  • Galante A, Tamada K, Levy D (2012) B7–H1 and a mathematical model for cytotoxic T cell and tumor cell interaction. Bull Math Biol 74(1):91–102

    MathSciNet  MATH  Google Scholar 

  • Gao L, Guo Q, Li X, Yang X, Ni H, Wang T, Zhao Q, Liu H, Xing Y, Xi T et al (2019) MiR-873/PD-L1 axis regulates the stemness of breast cancer cells. EBioMedicine 41:395–407

    Google Scholar 

  • Ghosh C, Luong G, Sun Y (2021) A snapshot of the PD-1/PD-L1 pathway. J Cancer 12(9):2735

    Google Scholar 

  • Gordon RE, Lane BP (1980) Duration of cell cycle and its phases measured in synchronized cells of squamous cell carcinoma of rat trachea. Cancer Res 40(12):4467–4472

    Google Scholar 

  • Han Y, Liu D, Li L (2020) PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res 10(3):727

    Google Scholar 

  • Inman BA, Sebo TJ, Frigola X, Dong H, Bergstralh EJ, Frank I, Fradet Y, Lacombe L, Kwon ED (2007) PD-L1 (B7–H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata. Am Cancer Soc 109(8):1499–1505

    Google Scholar 

  • Kaveh K, Fu F (2021) Immune checkpoint therapy modeling of PD-1/PD-L1 blockades reveals subtle difference in their response dynamics and potential synergy in combination. arXiv preprint arXiv:2103.12186

  • Kim Y, Friedman A (2010) Interaction of tumor with its micro-environment: a mathematical model. Bull Math Biol 72(5):1029–1068

    MathSciNet  MATH  Google Scholar 

  • Kim E, Brown JS, Eroglu Z, Anderson AR (2021) Adaptive therapy for metastatic melanoma: predictions from patient calibrated mathematical models. Cancers 13(4):823

    Google Scholar 

  • Kinjyo I, Qin J, Tan S-Y, Wellard CJ, Mrass P, Ritchie W, Cavanagh LL, Tomura M, Sakaue-Sawano A, Kanagawa O et al (2015) Real-time tracking of cell cycle progression during CD8+ effector and memory T-cell differentiation. Nat Commun 6(1):1–13

    Google Scholar 

  • Kozłowska E, Suwiński R, Giglok M, Świerniak A, Kimmel M (2020) Mathematical model predicts response to chemotherapy in advanced non-resectable non-small cell lung cancer patients treated with platinum-based doublet. PLoS Comput Biol 16(10):1008234

    Google Scholar 

  • Kumar B, Ghosh A, Datta C, Pal DK (2019) Role of PDL1 as a prognostic marker in renal cell carcinoma: a prospective observational study in eastern India. Ther Adv Urol 11:1756287219868859

    Google Scholar 

  • Lai X, Friedman A (2017) Combination therapy of cancer with cancer vaccine and immune checkpoint inhibitors: a mathematical model. PLoS ONE 12(5):0178479

    Google Scholar 

  • Lai X, Stiff A, Duggan M, Wesolowski R, Carson WE III, Friedman A (2018) Modeling combination therapy for breast cancer with bet and immune checkpoint inhibitors. Proc Natl Acad Sci USA 115(21):5534–5539

    Google Scholar 

  • Lei J (2020a) A general mathematical framework for understanding the behavior of heterogeneous stem cell regeneration. J Theor Biol 492:1–35

  • Lei J (2020b) Evolutionary dynamics of cancer: from epigenetic regulation to cell population dynamics—mathematical model framework, applications, and open problems. Sci China Math 63:411–424

  • Leschiera E, Lorenzi T, Shen S, Almeida L, Audebert C (2022) A mathematical model to study the impact of intra-tumour heterogeneity on anti-tumour CD8+ T cell immune response. J Theor Biol 66:111028

    MATH  Google Scholar 

  • Li M, Li A, Zhou S, Xu Y, Xiao Y, Bi R, Yang W (2018) Heterogeneity of PD-L1 expression in primary tumors and paired lymph node metastases of triple negative breast cancer. BMC Cancer 18(1):1–9

    Google Scholar 

  • Li H, Wang Z, Zhang Y, Sun G, Ding B, Yan L, Liu H, Guan W, Hu Z, Wang S et al (2019) The immune checkpoint regulator PDL1 is an independent prognostic biomarker for biochemical recurrence in prostate cancer patients following adjuvant hormonal therapy. J Cancer 10(14):3102

    Google Scholar 

  • Liao K-L, Bai X-F, Friedman A (2014) Mathematical modeling of interleukin-27 induction of anti-tumor t cells response. PLoS ONE 9(3):91844

    Google Scholar 

  • Lorenzi T, Chisholm RH, Clairambault J (2016) Tracking the evolution of cancer cell populations through the mathematical lens of phenotype-structured equations. Biol Direct 11(1):1–17

    Google Scholar 

  • Lote H, Cafferkey C, Chau I (2015) Pd-1 and pd-l1 blockade in gastrointestinal malignancies. Cancer Treat Rev 41(10):893–903

    Google Scholar 

  • Macallan DC, Busch R, Asquith B (2019) Current estimates of T cell kinetics in humans. Curr Opin Syst Biol 18:77–86

    Google Scholar 

  • Mahasa KJ, Ouifki R, Eladdadi A, de Pillis L (2016) Mathematical model of tumor-immune surveillance. J Theor Biol 404:312–330

    MathSciNet  MATH  Google Scholar 

  • Marino S, Hogue IB, Ray CJ, Kirschner DE (2008) A methodology for performing global uncertainty and sensitivity analysis in systems biology. J Theor Biol 254(1):178–196

    MathSciNet  MATH  Google Scholar 

  • McLaughlin J, Han G, Schalper KA, Carvajal-Hausdorf D, Pelekanou V, Rehman J, Velcheti V, Herbst R, LoRusso P, Rimm DL (2016) Quantitative assessment of the heterogeneity of PD-L1 expression in non-small cell lung cancer (NSCLC). JAMA Oncol 2(1):46–54

    Google Scholar 

  • Mellman I, Coukos G, Dranoff G (2011) Cancer immunotherapy comes of age. Nature 480(7378):480–489

    Google Scholar 

  • Mu C-Y, Huang J-A, Chen Y, Chen C, Zhang X-G (2011) High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol 28(3):682–688

    Google Scholar 

  • Muenst S, Schaerli A, Gao F, Däster S, Trella E, Droeser R, Muraro M, Zajac P, Zanetti R, Gillanders W et al (2014) Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 146(1):15–24

    Google Scholar 

  • Nakamura S, Hayashi K, Imaoka Y, Kitamura Y, Akazawa Y, Tabata K, Groen R, Tsuchiya T, Yamasaki N, Nagayasu T et al (2017) Intratumoral heterogeneity of programmed cell death ligand-1 expression is common in lung cancer. PLoS ONE 12(10):0186192

    Google Scholar 

  • Nakanishi J, Wada Y, Matsumoto K, Azuma M, Kikuchi K, Ueda S (2007) Overexpression of b7–h1 (pd-l1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother 56(8):1173–1182

    Google Scholar 

  • Nikolopoulou E, Johnson L, Harris D, Nagy J, Stites E, Kuang Y (2018) Tumour-immune dynamics with an immune checkpoint inhibitor. Lett Biomath 6:66

    MathSciNet  Google Scholar 

  • Rehman JA, Han G, Carvajal-Hausdorf DE, Wasserman BE, Pelekanou V, Mani NL, McLaughlin J, Schalper KA, Rimm DL (2017) Quantitative and pathologist-read comparison of the heterogeneity of programmed death-ligand 1 (PD-L1) expression in non-small cell lung cancer. Mod Pathol 30(3):340–349

    Google Scholar 

  • Riley JL (2009) PD-1 signaling in primary T cells. Immunol Rev 229(1):114–125

    Google Scholar 

  • Sabatier R, Finetti P, Mamessier E, Adelaide J, Chaffanet M, Ali HR, Viens P, Caldas C, Birnbaum D, Bertucci F (2015) Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget 6(7):5449

    Google Scholar 

  • Sauce D, Almeida JR, Larsen M, Haro L, Autran B, Freeman GJ, Appay V (2007) PD-1 expression on human CD8 T cells depends on both state of differentiation and activation status. AIDS 21(15):2005–2013

    Google Scholar 

  • Soliman H, Khalil F, Antonia S (2014) PD-L1 expression is increased in a subset of basal type breast cancer cells. PLoS ONE 9(2):1–10

    Google Scholar 

  • Staňková K, Brown JS, Dalton WS, Gatenby RA (2019) Optimizing cancer treatment using game theory: a review. JAMA Oncol 5(1):96–103

    Google Scholar 

  • Stiehl T, Wang W, Lutz C, Marciniak-Czochra A (2020) Mathematical modeling provides evidence for niche competition in human AML and serves as a tool to improve risk stratification. Cancer Res 80(18):3983–3992

    Google Scholar 

  • Sun X, Bao J, Shao Y (2016) Mathematical modeling of therapy-induced cancer drug resistance: connecting cancer mechanisms to population survival rates. Sci Rep 6(1):1–12

    Google Scholar 

  • Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB et al (2012) Safety, activity, and immune correlates of anti-pd-1 antibody in cancer. N Engl J Med 366(26):2443–2454

    Google Scholar 

  • Tsur N, Kogan Y, Avizov-Khodak E, Vaeth D, Vogler N, Utikal J, Lotem M, Agur Z (2019) Predicting response to pembrolizumab in metastatic melanoma by a new personalization algorithm. J Transl Med 17(1):1–15

    Google Scholar 

  • Tubiana M (1989) Tumor cell proliferation kinetics and tumor growth rate. Acta Oncol 28(1):113–121

    Google Scholar 

  • Uhercik M, Sanders AJ, Owen S, Davies EL, Sharma AK, Jiang WG, Mokbel K (2017) Clinical significance of PD1 and PDL1 in human breast cancer. Anticancer Res 37(8):4249–4254

    Google Scholar 

  • Yang K, Xu J, Liu Q, Li J, Xi Y (2019) Expression and significance of CD47, PD1 and PDL1 in T-cell acute lymphoblastic lymphoma/leukemia. Pathol Res Pract 215(2):265–271

    Google Scholar 

  • Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, Wu K (2018) Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer 17:129–143

    Google Scholar 

  • Zuazo M, Gato-Cañas M, Llorente N, Ibañez-Vea M, Arasanz H, Kochan G, Escors D (2017) Molecular mechanisms of programmed cell death-1 dependent T cell suppression: relevance for immunotherapy. Ann Transl Med 5(19):66

    Google Scholar 

Download references

Acknowledgements

We acknowledge TCGA database for providing their platforms and contributors for uploading their meaningful datasets. This work is supported by the National Natural Science Foundation of China (No. 12171478), the Fundamental Research Funds for the Central Universities (19XNLG14) and the Research Funds of Renmin University of China.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xiulan Lai.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Appendices

Appendix A: Parameter estimation

Parameters in the equation of C

In modelling tumour-immune surveillance (Mahasa et al. 2016), the per capita growth rate of tumour cells was estimated to be \(0.5822\mathrm{day}^{-1}\). We accordingly take the cancer cell basic production rate to be \(\bar{\beta }_{C}=0.5822/24\mathrm{h}^{-1}=0.0243\mathrm{h}^{-1}\). In modelling anti-tumour T cells response (Liao et al. 2014), the death rate of tumour cells was estimated to be \(0.173\mathrm{day}^{-1}\). We accordingly take the apoptosis rate of proliferating cancer cells to be \(\mu _{C}=0.173/24\mathrm{h}^{-1}=0.0072\mathrm{h}^{-1}\). We further assume that the apoptosis rate of non-proliferating cancer cells is much lower than that of the proliferating ones, and take \(\bar{\kappa }_{C}=\mu _{C}/10 = 7.2\times 10^{-4}\mathrm{h}^{-1}\).

In studying the impact of intra-tumour heterogeneity on anti-tumour CD\(8^+\) T cell immune response (Leschiera et al. 2022), the mean cell cycle time of tumour cells was estimated to be \(24\mathrm{h}\), where the duration interval was \(17-48\mathrm{h}\) (Tubiana 1989; Gordon and Lane 1980). Hence we take \(\tau _{C}=24\mathrm{h}\). In modelling tumour-immune surveillance (Mahasa et al. 2016), the reciprocal carrying capacity of the tumour cells was estimated to be \(2.33\times 10^{-8}{\mathrm{cell}}^{-1}\). We take tumor carrying capacity as \({\hat{C}}_{*}=1/(2.33\times 10^{-8}\mathrm{cell}^{-1})=4.3\times 10^7{\mathrm{cell}}\).

Parameters in the equations of \(T_0\) and T

In metastatic melanoma microenvironment (Tsur et al. 2019), activation rate of naive antigen-specific CD\(8^+\) T cells was estimated to be \(0.8318\mathrm{day}^{-1}\). In the tumour microenvironment (Dritschel et al. 2018), the death rate of helper T cells was estimated to be \(0.1\mathrm{day}^{-1}\). Hence we take the resting T cell basic proliferation rate as \(\beta _{T}=0.8318/24\mathrm{h}^{-1}=0.0347\mathrm{h}^{-1}\), and the apoptosis rate of proliferating T cell as \(\mu _{0}=0.1/24\mathrm{h}^{-1}=0.0042\mathrm{h}^{-1}\). By Kinjyo et al. (2015), the T cell cycle time is \(14.3\pm 4.4\mathrm{h}\). We accordingly take \(\tau _{0}=14.3\mathrm{h}\).

In the estimation of T cell kinetics in humans (Macallan et al. 2019), the proliferation rate of memory T cell ranges \(0.006-0.16{\mathrm{day}}^{-1}\). Here we take the coefficient of resting T cell differentiation rate as \(\bar{\kappa }_T = 0.104/24\mathrm{h}^{-1}=0.0043 \mathrm{h}^{-1}\). In modelling tumour-immune surveillance (Mahasa et al. 2016), the per capita death rate of CTLs was estimated to be \(0.02\mathrm{day}^{-1}\); the binding rate of CTLs to tumour cells was \(1.3\times 10^{-7}{\mathrm{day}}^{-1}\). We accordingly take the apoptosis rate of effector T cell as \(\mu _{T}=0.02/24{\mathrm{h}}^{-1}=8.3\times 10^{-4}{\mathrm{h}}^{-1}\); the killing rate of effector T cells as \(\eta _{0}=1.3\times 10^{-7}/24\mathrm{h}^{-1}\) \(=5.4\times 10^{-9}\) \({\mathrm{h}}^{-1}\).

Appendix B: Cell-based stochastic simulation

We have three epigenetic states, \(x_0\), \(x_1\) and \(x_2\), in the differential-integral equations model (2). It is very expensive to solve the system numerically, such as using the Euler method, due to the high dimensional integration. Therefore, we apply the method of cell-based stochastic simulation proposed in Lei (2020a). By this approach, we model the growth process of a multiple-cell system with a collection of epigenetic states. The cell-based stochastic simulation tracks the behaviours of each cell according to its own epigenetic states. The sketch of the numerical scheme is given as follows.

\(\mathbf {Initialize}\) the time \(t = 0\), the cancer cell number \(Q_C\) (cancer cells pool: \(\Sigma _C=\left\{ C_i(\mathbf {x}_i,A_i) \right\} _{i=1}^{Q_C}\)), the \(T_0\) cell number \(Q_{T_0}\) (resting T cells pool: \(\Sigma _{T_0}=\left\{ T_{0i}(\mathbf {x}_i,A_i) \right\} _{i=1}^{Q_{T_0}})\), the T cell number \(Q_T\) (T cell pool: \(\Sigma _T=\left\{ T_i(\mathbf {x}_i,A_i) \right\} _{i=1}^{Q_{T}}\)). At the initial state, all cells are at the resting phase, and the corresponding age at the proliferating phase is \(A_i=0\).

\(\mathbf {for}\) t from 0 to T with step \(\bigtriangleup t\) \(\mathbf {do}\)

\(\mathbf {for}\) cancer cells in \(\Sigma _C\) \(\mathbf {do}\)

  • Calculate the proliferation rate \(\beta _C\), the apoptosis rate of proliferating cells \(\mu _C\), and the death rate \(\kappa _C\), the killing rate of cancer cells by effector T cells \(\eta \).

  • Determine the cell fate during the time interval \((t,t+\Delta t)\):

  • When the cell is at the resting phase, undergo death with a probability \(\kappa _C\Delta t\), be killed by effector T cell with a probability \(\eta \Delta t\) or enter the proliferation phase with a probability \(\beta _C\Delta t\). If the cell enters the proliferation phase, set the age \(A_i=0\).

  • When the cell is at the proliferating phase, if the age \(A_i<\tau \), the cell is either removed (through apoptosis) with a probability \(\mu _C\Delta t\) or remains unchanged and \(A_i=A_i+\Delta t\); if the age \(A_i \ge \tau \), the cell undergoes mitosis and divides into two cells. When mitosis occurs, the epigenetic state of each daughter cell is determined according to the inheritance probability functions \(p_0\left( {x_{0}}, {y}\right) \) and \(p_1\left( {x_{1}}, {y}\right) \).

\(\mathbf {end ~ for }\)

\(\mathbf {for}\) resting T cells in \(\Sigma _{T_0}\) \(\mathbf {do}\)

  • Calculate the proliferation rate \(\beta _{T}\), the apoptosis rate \(\mu _{0}\), and the differentiation rate \(\kappa _{T}\).

  • Determine the cell fate during the time interval \((t, t+\Delta t)\):

  • When the cell is at the resting phase, undergo differentiation with a probability \(\kappa _T\Delta t\) or enter the proliferation phase with a probability \(\beta _T\Delta t\). If the cell enters the proliferation phase, set the age \(A_i=0\).

  • When the cell is at the proliferating phase, if the age \(A_i<\tau \), the cell is either removed (through apoptosis) with a probability \(\mu _0\Delta t\) or remains unchanged and \(A_i=A_i+\Delta t\); if the age \(A_i \ge \tau \), the cell undergoes mitosis and divides into two cells. When mitosis occurs, the epigenetic state of each daughter cell is determined according to the inheritance probability function \(p_2\left( \mathbf {x}, \mathbf {y}\right) \).

\(\mathbf {end ~ for }\)

\(\mathbf {for}\) effector T cells in \(\Sigma _{T}\) \(\mathbf {do}\)

  • Calculate the apoptosis rate \(\mu _T\).

  • Determine the cell fate during the time interval \((t, t+\Delta t)\):

  • The cell is removed (through apoptosis) with a probability \(\mu _T\Delta t\).

\(\mathbf {end ~ for }\)

\(\mathbf {Update}\) the system with the caner cell number, the resting T cell number, the effector T cell number, the epigenetic states of all surviving cells, and the ages of the proliferating phase cells, and set \(t=t+\Delta t\).

\(\mathbf {end ~ for }\)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, S., Lei, J. & Lai, X. Modeling tumour heterogeneity of PD-L1 expression in tumour progression and adaptive therapy. J. Math. Biol. 86, 38 (2023). https://doi.org/10.1007/s00285-023-01872-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00285-023-01872-1

Keywords

Mathematics Subject Classification

Navigation