Skip to main content
Log in

Modeling age-dependent radiation-induced second cancer risks and estimation of mutation rate: an evolutionary approach

  • Original Paper
  • Published:
Radiation and Environmental Biophysics Aims and scope Submit manuscript

Abstract

Although the survival rate of cancer patients has significantly increased due to advances in anti-cancer therapeutics, one of the major side effects of these therapies, particularly radiotherapy, is the potential manifestation of radiation-induced secondary malignancies. In this work, a novel evolutionary stochastic model is introduced that couples short-term formalism (during radiotherapy) and long-term formalism (post-treatment). This framework is used to estimate the risks of second cancer as a function of spontaneous background and radiation-induced mutation rates of normal and pre-malignant cells. By fitting the model to available clinical data for spontaneous background risk together with data of Hodgkin’s lymphoma survivors (for various organs), the second cancer mutation rate is estimated. The model predicts a significant increase in mutation rate for some cancer types, which may be a sign of genomic instability. Finally, it is shown that the model results are in agreement with the measured results for excess relative risk (ERR) as a function of exposure age and that the model predicts a negative correlation of ERR with increase in attained age. This novel approach can be used to analyze several radiotherapy protocols in current clinical practice and to forecast the second cancer risks over time for individual patients.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  • Aleman BM, van den Belt-Dusebout AW, Klokman WJ, van’t Veer MB, Bartelink H, van Leeuwen FE (2003) Long-term cause-specific mortality of patients treated for Hodgkins disease. J Clin Oncol 21(18):3431–3439

    Article  Google Scholar 

  • Armitage P (1985) Multistage models of carcinogenesis. Environ Health Perspect 63:195

    Article  Google Scholar 

  • Armitage P, Doll R (1954) The age distribution of cancer and a multi-stage theory of carcinogenesis. Br J Cancer 8(1):1

    Article  Google Scholar 

  • Athar BS, Paganetti H (2011) Comparison of second cancer risk due to out-of-field doses from 6-mv imrt and proton therapy based on 6 pediatric patient treatment plans. Radiother Oncol 98(1):87–92

    Article  Google Scholar 

  • Bhatia S, Sklar C (2002) Second cancers in survivors of childhood cancer. Nat Rev Cancer 2(2):124–132

    Article  Google Scholar 

  • Bhatia S, Yasui Y, Robison LL, Birch JM, Bogue MK, Diller L, DeLaat C, Fossati-Bellani F, Morgan E, Oberlin O, Gregory Reaman JT, Frederick B Ruymann, Meadows AT (2003) High risk of subsequent neoplasms continues with extended follow-up of childhood Hodgkin’s disease: report from the late effects study group. J Clin Oncol 21(23):4386–4394

    Article  Google Scholar 

  • Dores GM, Metayer C, Curtis RE, Lynch CF, Clarke EA, Glimelius B, Storm H, Pukkala E, van Leeuwen FE, Holowaty EJ, Andersson M, Wiklund T, Joensuu T, van’t Veer MB, Stovall M, Gospodarowicz M, Travis LB (2002) Second malignant neoplasms among long-term survivors of Hodgkin’s disease: a population-based evaluation over 25 years. J Clin Oncol 20(16):3484–3494

    Article  Google Scholar 

  • Durrett R (2008) Probability models for DNA sequence evolution, probability and its applications, vol 12, 2nd edn. Springer, Berlin

    Book  Google Scholar 

  • Hall EJ (2006) Intensity-modulated radiation therapy, protons, and the risk of second cancers. Int J Radiat Oncol Biol Phys 65(1):1–7

    Article  ADS  Google Scholar 

  • Hodgson DC, Gilbert ES, Dores GM, Schonfeld SJ, Lynch CF, Storm H, Hall P, Langmark F, Pukkala E, Andersson M et al (2007a) Long-term solid cancer risk among 5-year survivors of Hodgkin’s lymphoma. J Clin Oncol 25(12):1489–1497

    Article  Google Scholar 

  • Hodgson DC, Koh ES, Tran TH, Heydarian M, Tsang R, Pintilie M, Xu T, Huang L, Sachs RK, Brenner DJ (2007b) Individualized estimates of second cancer risks after contemporary radiation therapy for Hodgkin lymphoma. Cancer 110(11):2576–2586

    Article  Google Scholar 

  • Huang L, Snyder AR, Morgan WF (2003) Radiation-induced genomic instability and its implications for radiation carcinogenesis. Oncogene 22(37):5848–5854

    Article  Google Scholar 

  • Iwasa Y, Michor F, Komarova NL, Nowak MA (2005) Population genetics of tumor suppressor genes. J Theor Biol 233(1):15–23

    Article  MathSciNet  Google Scholar 

  • Komarova NL, Sengupta A, Nowak MA (2003) Mutation-selection networks of cancer initiation: tumor suppressor genes and chromosomal instability. J Theor Biol 223(4):433–450

    Article  MathSciNet  Google Scholar 

  • Kunkel TA (2004) DNA replication fidelity. J Biol Chem 279(17):16895–16898

    Article  Google Scholar 

  • Lindsay K, Wheldon E, Deehan C, Wheldon T (2001) Radiation carcinogenesis modelling for risk of treatment-related second tumours following radiotherapy. Br J Radiol 74(882):529–536

    Article  Google Scholar 

  • Little MP (2009) Cancer and non-cancer effects in Japanese atomic bomb survivors. J Radiol Prot 29(2A):A43

    Article  Google Scholar 

  • Manem V, Kohandel M, Hodgson D, Sharpe M, Sivaloganathan S (2014a) The effect of radiation quality on the risks of second malignancies.Int J Radiat Biol 1–39 [Epub ahead of print]

  • Manem VS, Dhawan A, Kohandel M, Sivaloganathan S (2014b) Efficacy of dose escalation on TCP, recurrence and second cancer risks: a mathematical study. Br J Radiol 87(1043):20140377

    Article  Google Scholar 

  • Michor F, Iwasa Y, Nowak MA (2004) Dynamics of cancer progression. Nat Rev Cancer 4(3):197–205

    Article  Google Scholar 

  • Moran PAP (1962) The statistical processes of evolutionary theory, 1st edn. Clarendon Press and Oxford University Press, Oxford

    MATH  Google Scholar 

  • Moteabbed M, Yock TI, Paganetti H (2014) The risk of radiation-induced second cancers in the high to medium dose region: a comparison between passive and scanned proton therapy, IMRT and VMAT for pediatric patients with brain tumors. Phys Med Biol 59(12):2883

    Article  Google Scholar 

  • Nowak MA, Komarova NL, Sengupta A, Jallepalli PV, Shih IM, Vogelstein B, Lengauer C (2002) The role of chromosomal instability in tumor initiation. Proc Natl Acad Sci 99(25):16226–16231

    Article  ADS  Google Scholar 

  • Paganetti H, Athar BS, Moteabbed M, Adams JA, Schneider U, Yock TI (2012) Assessment of radiation-induced second cancer risks in proton therapy and IMRT for organs inside the primary radiation field. Phys Med Biol 57(19):6047

    Article  Google Scholar 

  • Preston D, Ron E, Tokuoka S, Funamoto S, Nishi N, Soda M, Mabuchi K, Kodama K (2007) Solid cancer incidence in atomic bomb survivors: 1958–1998. Radiat Res 168(1):1–64

    Article  Google Scholar 

  • Preston DL, Kusumi S, Tomonaga M, Izumi S, Ron E, Kuramoto A, Kamada N, Dohy H, Matsui T, Nonaka H et al (1994) Cancer incidence in atomic bomb survivors. Part iii: leukemia, lymphoma and multiple myeloma, 1950–1987. Radiat Res 137(2s):S68–S97

    Article  Google Scholar 

  • Sachs RK, Brenner DJ (2005) Solid tumor risks after high doses of ionizing radiation. Proc Natl Acad Sci USA 102(37):13040–13045

    Article  ADS  Google Scholar 

  • Sachs RK, Shuryak I, Brenner D, Fakir H, Hlatky L, Hahnfeldt P (2007) Second cancers after fractionated radiotherapy: stochastic population dynamics effects. J Theor Biol 249(3):518–531

    Article  MathSciNet  Google Scholar 

  • Schneider U (2009) Mechanistic model of radiation-induced cancer after fractionated radiotherapy using the linear–quadratic formula. Med Phys 36(4):1138–1143

    Article  Google Scholar 

  • Schneider U, Lomax A, Besserer J, Pemler P, Lombriser N, Kaser-Hotz B (2007) The impact of dose escalation on secondary cancer risk after radiotherapy of prostate cancer. Int J Radiat Oncol Biol Phys 68(3):892–897

    Article  Google Scholar 

  • Schneider U, Sumila M, Robotka J, Weber D, Gruber G (2014) Radiation-induced second malignancies after involved-node radiotherapy with deep-inspiration breath-hold technique for early stage Hodgkin lymphoma: a dosimetric study. Radiat Oncol 9(1):58

    Article  Google Scholar 

  • Shuryak I, Hahnfeldt P, Hlatky L, Sachs RK, Brenner DJ (2009a) A new view of radiation-induced cancer: integrating short-and long-term processes. Part i: approach. Radiat Environ Biophys 48(3):263–274

    Article  Google Scholar 

  • Shuryak I, Hahnfeldt P, Hlatky L, Sachs RK, Brenner DJ (2009b) A new view of radiation-induced cancer: integrating short-and long-term processes. Part ii: second cancer risk estimation. Radiat Environ Biophys 48(3):275–286

    Article  Google Scholar 

  • Sigurdson AJ, Jones IM (2003) Second cancers after radiotherapy: Any evidence for radiation-induced genomic instability? Oncogene 22(45):7018–7027

    Article  Google Scholar 

  • Travis LB, Hill D, Dores GM, Gospodarowicz M, van Leeuwen FE, Holowaty E, Glimelius B, Andersson M, Pukkala E, Lynch CF, Pee D, Smith SA, Veer MBV, Joensuu T, Storm H, Stovall M, JDB, Gilbert E, Gail MH (2005) Cumulative absolute breast cancer risk for young women treated for Hodgkin lymphoma. J Natl Cancer Inst 97(19):1428–1437

    Article  Google Scholar 

  • van Leeuwen FE, Klokman WJ, Stovall M, Dahler EC, van’t Veer MB, No-ordijk EM, Crommelin MA, Aleman BMP, Broeks A, Gospodarowicz M, Travis LB, Russell NS (2003) Roles of radiation dose, chemotherapy, and hormonal factors in breast cancer following Hodgkin’s disease. J Natl Cancer Inst 95(13):971–980

    Article  Google Scholar 

  • Wang J, Pulido JS, O’Neill BP, Johnston PB (2014) Second malignancies in patients with primary central nervous system lymphoma. Neuro Oncol pii:nou105 [Epub ahead of print]

  • Xu XG, Bednarz B, Paganetti H (2008) A review of dosimetry studies on external-beam radiation treatment with respect to second cancer induction. Phys Med Biol 53(13):R193

    Article  ADS  Google Scholar 

  • Yahalom J (2009) Role of radiation therapy in Hodgkin’s lymphoma. Cancer J 15(2):155–160

    Article  Google Scholar 

  • Yeoh KW, Mikhaeel NG (2010) Role of radiotherapy in modern treatment of Hodgkin’s lymphoma. Adv Hematology 2011:258797

    Google Scholar 

  • Yorke E, Kutcher G, Jackson A, Ling C (1993) Probability of radiation-induced complications in normal tissues with parallel architecture under conditions of uniform whole or partial organ irradiation. Radiother Oncol 26(3):226–237

    Article  Google Scholar 

  • Zhang Y, Goddard K, Spinelli JJ, Gotay C, McBride ML (2012) Risk of late mortality and second malignant neoplasms among 5-year survivors of young adult cancer: a report of the childhood, adolescent, and young adult cancer survivors research program. J Cancer Epidemiol 2012:103032

    Google Scholar 

Download references

Acknowledgments

M. Kohandel and S. Sivaloganathan are supported by an NSERC/CIHR Collaborative Health Research grant. The authors thank DC Hodgson from the Princess Margaret Hospital, Toronto for fruitful discussions.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kamran Kaveh.

Appendices

Appendix 1: Evolutionary dynamics of two-hit process

Consider a population of N cells (inside a niche) governed by the Moran process (types—0, 1, 2 indicating normal, pre-malignant and malignant phenotypes). At each time step, a cell is randomly chosen, based on its fitness, to reproduce and another cell is randomly chosen to die. In the presence of mutations, such as in the model presented in this paper, at each time step, there is an alternative possibility (instead of death–birth) that a normal cell may transform into a pre-malignant cell or alternatively a pre-malignant cell may transform into a malignant cell. The mutation rate for the first hit is \(u_{1}\) and the rate for the second hit (pre-malignant into malignant) is \(u_{2}\) (or \(u_{r}\) after radiation). As discussed in the text, let \(f_{0}(t, u_{1},u_{2},r,N)\) be the probability that a first malignant cell has emerged before time t given that the initial population consists of all normal cells in the niche. Similarly, let \(f_{1}(t, u_{1}, u_{2}, r, N)\) be the probability that a malignant cell emerges in a niche of size N before time t, beginning with one pre-malignant cell and N − 1 normal cells. It is straightforward to show that the two probabilities satisfy the continuous time limit of the Kolmogorov equations,

$$\begin{aligned} \frac{{\rm d}f_{0}(t)}{{\rm d}t}&= Nu_{1} f_{1}(t)\left(1 - f_{0}(t) \right), \\ \frac{{\rm d}f_{1}(t)}{{\rm d}t}&= ru_{2} - (1-r +2ru_{2})f_{1}(t) - r(1-u_{2})f_{1}^{2}(t), \end{aligned}$$
(9)

with the initial condition \(f_{0}(0) = 0\). The solutions of the above set of equations can be directly obtained under a branching process approximation, i.e., independence of two lineages starting from two different cells, one can then obtain an analytical solution for these probabilities,

$$\begin{aligned} f_{1}(t, u_{1},u_{2},r,N)&= \frac{\exp \{( r(1-u_{2})(a + b)t \}- 1}{(1/a)\exp \{ r(1-u_{2})(a + b)t \} +(1/b)}, \\ f_{0}(t, u_{1},u_{2},r,N)&= 1 - \exp \left( -Nu_{1}\left\{ \frac{a + b}{c} \ln \left[ \frac{{\rm e}^{ct} + a/b}{1 + a/b}\right] - bt\right\} \right), \end{aligned}$$
(10)

with a, b, c given as,

$$\begin{aligned} a&= \frac{1}{2(1-u_{2})}\left[ -\left( \frac{1-r}{r} + 2u_{2}\right) + \sqrt{\left( \frac{1-r}{r} + 2u_{2}\right) ^{2} + 4u_{2}}\,\right], \\ b&= \frac{1}{2(1-u_{2})}\left[ \left( \frac{1-r}{r} + 2u_{2}\right) + \sqrt{\left( \frac{1-r}{r} + 2u_{2}\right) ^{2} + 4u_{2}}\,\right], \\ c&= r(1-u_{2})(a + b). \end{aligned}$$
(11)

As discussed in the main body of the paper, both the cumulative age incidence of different cancer types and excess relative risk of cancer (second cancer in this case) is expressed in terms of functions \(f_{0}(t,u_{1},u_{2},r,N)\) and \(f_{1}(t,u_{1},u_{2},r,N)\), which lead to a simple form for the ERR. As is obvious, there is no dependence on the total number of niches \(\tilde{N}\), and the dependence on the niche size, N, is very weak. However, for more realistic fits, the value of the proliferation strength r is not extremely close to unity and thus the above approximation might not be appropriate. However, it is easy to see from the above approximate results that any quantity of interest (i.e., number of background age incidences or ERR) is represented by very weak functions of the niche size, N. Thus, the present rough estimates for the value of N do not change any of the important conclusions resulting from this work.

Appendix 2: Initiation–inactivation–repopulation model

In this section, the initiation–inactivation–repopulation (Sachs and Brenner 2005) formalism is briefly reviewed to estimate the number of pre-malignant cells at the end of radiation treatment. The effect of radiation is simplified into two mechanism: (1) cell killing which is modeled by the linear–quadratic approximation and gives the number of either normal or pre-malignant cells killed due to radiation dose. (2) The cell initiation which is assumed to be a linear function of the dose with a small constant mutation rate per dose rate. Two populations of normal stem cells and pre-malignant stem cells are assumed. Normal stem cells grow logistically while they can die during the radiation exposure times, where cell death is given by linear–quadratic formula. They can also transform into pre-malignant cells during the exposure time. The population of pre-malignant cells has a similar growth form while its proliferation is regulated by the population of normal cells and has the same cell death rate due to radiation and also a positive rate due to normal stem cell initiation. The above can be written in the form of the following coupled system of ordinary differential equations.

$$\begin{aligned} \frac{{\rm d}n(t)}{{\rm d}t}&= r_{0}n(t)\left( 1 - \frac{n(t)}{K}\right) - (\alpha + \beta D)\cdot \frac{{\rm d}D}{{\rm d}t}n(t) - \gamma \frac{{\rm d}D}{{\rm d}t}n(t), \\ \frac{{\rm d}m(t)}{{\rm d}t}&= r_{0}\lambda m(t)\left( 1 - \frac{n(t)}{K}\right) - (\alpha + \beta D) \cdot \frac{{\rm d}D}{{\rm d}t}m(t) + \gamma \frac{{\rm d}D}{{\rm d}t}n(t), \end{aligned}$$
(12)

where \(r_{0}\) is the normal cell repopulation rate while \(r_{0}\lambda\) is the pre-malignant repopulation rate. \({\rm d}D/{\rm d}t\) is the dose delivery rate, \(\lambda\) represents the relative proliferation rate of pre-malignant cells to the normal cells and K is the carrying capacity of normal cells. \(\alpha\) and \(\beta\) are the cell killing rates from linear–quadratic dose dependence (LQ). The number of pre-malignant cells after the total radiation dose has been applied and is the quantity of importance here. This can be analytically calculated for the simplified case of an acute dose, \({\rm d}D/{\rm d}t = {{\rm const}}.\) applied in a finite interval of time

$$\begin{aligned} M&= m(\infty ) = N(\exp (\gamma D) - 1), \\&\sim N\gamma D, \end{aligned}$$
(13)

where D is the total dose. The total number of the cells \(N_{{\rm tot}}\), is given by the number of niche times the niches size, \(N\tilde{N}\). For the values of the radiation-induced initiation rate \(\gamma\), this will result in \(M/N_{{\rm tot}} \sim 10^{-6}\) (per Gy). Since we are interested on an estimate for number of radiation-induced pre-malignant cells, the details of cell killing mechanism in LQ formula does not appear in Eq. (13).

The above formalism can be expanded to incorporate fractionation therapy, i.e., non-constant dose–delivery rate which has been discussed in the literature. In the present work, there is no need to deal with these details. However, as a future direction of research, all the details of radiotherapy (fractionation protocol and the dose–volume histogram for each patient) can be used as input to estimate the ERR as a function of exposure age and attained age.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kaveh, K., Manem, V.S.K., Kohandel, M. et al. Modeling age-dependent radiation-induced second cancer risks and estimation of mutation rate: an evolutionary approach. Radiat Environ Biophys 54, 25–36 (2015). https://doi.org/10.1007/s00411-014-0576-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00411-014-0576-z

Keywords

Navigation