Elsevier

Biosystems

Volume 83, Issues 2–3, February–March 2006, Pages 81-90
Biosystems

Cancer: A Systems Biology disease

https://doi.org/10.1016/j.biosystems.2005.05.014Get rights and content

Abstract

Cancer research has focused on the identification of molecular differences between cancerous and healthy cells. The emerging picture is overwhelmingly complex. Molecules out of many parallel signal transduction pathways are involved. Their activities appear to be controlled by multiple factors. The action of regulatory circuits, cross-talk between pathways and the non-linear reaction kinetics of biochemical processes complicate the understanding and prediction of the outcome of intracellular signaling. In addition, interactions between tumor and other cell types give rise to a complex supra-cellular communication network. If cancer is such a complex system, how can one ever predict the effect of a mutation in a particular gene on a functionality of the entire system? And, how should one go about identifying drug targets?

Here, we argue that one aspect is to recognize, where the essence resides, i.e. recognize cancer as a Systems Biology disease. Then, more cancer biologists could become systems biologists aiming to provide answers to some of the above systemic questions. To this aim, they should integrate the available knowledge stemming from quantitative experimental results through mathematical models. Models that have contributed to the understanding of complex biological systems are discussed. We show that the architecture of a signaling network is important for determining the site at which an oncologist should intervene. Finally, we discuss the possibility of applying network-based drug design to cancer treatment and how rationalized therapies, such as the application of kinase inhibitors, may benefit from Systems Biology.

Introduction

During the micro-evolutionary process of malignant transformation, cancer cells accumulate multiple genetic alterations that provide them with several capabilities (Cahill et al., 1999). The latter include the escape from normal growth control, evasion of the suicidal apoptotic program, induction of sustained angiogenesis, the ability to metastasise, and to invade healthy tissues (Hanahan and Weinberg, 2000). In the past decades, cancer researchers have collected an enormous amount of information about the differences between cancer cells and their healthy counterparts, with the ultimate goal of identifying drug targets. This has, for instance, led to the identification of genes that are causally implicated in human cancer and to the discovery of the mutations in those genes. A cancer gene census was recently compiled (Futreal et al., 2004), which currently contains 300 genes. The vast majority of these genes function in signal transduction processes within or between cells, govern cell cycle progression, apoptosis, angiogenesis, and infiltration (Vogelstein and Kinzler, 2004).

Although knowledge of the molecular cell biology of cancer is enormous, at the same time, the emerging complexity of the entire ‘cancer system’ overwhelms us, leaving an enormous gap in our understanding and predictive power. In this paper, we will discuss aspects of this complexity, and how one can deal with it to answer questions that are relevant for the treatment of cancer.

Section snippets

Zooming in: complex signaling networks

Many signaling molecules (proteins, lipids, and ions) have been identified, and for many, the way they communicate with each other through signal transduction pathways has been elucidated. Signaling pathways consist of multiple sequential events, including covalent modifications (e.g. phosphorylation), recruitment, allosteric activation or inhibition, and binding of proteins (Alberts et al., 2002). The kinetics of these reactions are often non-linear, as a result of the properties of the

Zooming out: complexity increases

The architecture of signal transduction pathways is not where the complexity of cancer ends. Being parts of the cell, signaling networks are affected by additional levels of organization, for instance, as many proteins are not uniformly distributed over the cell. Areas with high protein concentrations might lead to macromolecular crowding (Ellis and Minton, 2003) and cause steep spatial gradients of activated signaling proteins (Brown and Kholodenko, 1999).

Numerous interactions at the

Cancer: a Systems Biology disease

Above, we have discussed that cancer involves a number of molecular processes at the same time. Looking back, it is clear that the interactions of these processes lead to new functionalities that would not otherwise be possible. An example is the formation of a life-threatening tumor requiring the formation of new blood vessels, which requires the cancer cells to become insensitive to growth inhibition and the endothelial cells to become activated to new blood vessel formation. Indeed, cancer

Towards integration

It appears that the many supra-cellular interactions, which add to the intra-cellular signaling, are all at least to some extent relevant for the development of cancer. As more relevant genes, proteins and interactions are being identified, the total picture will continue to grow even more complex. If we also take into account the non-linearity of the biochemical processes and the fact that different processes act on different time scales (signaling, cell cycle, and angiogenic switch), then it

Network-based drug design

The inhibition of enzymes that contribute to aberrant signaling and cancer progression holds promise for the treatment of cancer. Here, one should perhaps also think about a substantial reduction in the rate of tumor progression and metastasis instead of only about a successful total elimination of tumors. Such new therapies could benefit directly from analyzing networks and whole systems, rather than all their individual parts of systems. Which clinically relevant questions may be answered by

Kinase inhibitors: smart drugs?

Conventional cancer treatment relies on surgery, radiotherapy, and chemotherapy (DeVita et al., 2001). The development of rationalized cancer therapy, based on the knowledge of the biology of cancer, may enhance the arsenal of oncologists. Inhibition of constitutive pathways, e.g. by monoclonal antibodies or tyrosine kinase inhibitors, is a strategy that is already employed in clinical trials (Mendelsohn and Baselga, 2000, Sebolt-Leopold, 2000, Shawver et al., 2002). The success of drugs in

References (74)

  • R. Heinrich et al.

    Mathematical models of protein kinase signal transduction

    Mol. Cell

    (2002)
  • B.P. Ingalls et al.

    Sensitivity analysis of stoichiometric networks: an extension of metabolic control analysis to non-steady state trajectories

    J. Theor. Biol.

    (2003)
  • D. Kahn et al.

    Control theory of regulatory cascades

    J. Theor. Biol.

    (1991)
  • B.N. Kholodenko et al.

    Quantification of short term signaling by the epidermal growth factor receptor

    J. Biol. Chem.

    (1999)
  • J.G. Koster et al.

    Kinetics of histone gene expression during early development of Xenopus laevis

    J. Theor. Biol.

    (1988)
  • J. Lankelma et al.

    Simulation model of doxorubicin activity in islets of human breast cancer cells

    Biochim. Biophys. Acta

    (2003)
  • A. Mantovani et al.

    Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes

    Trends Immunol.

    (2002)
  • P. Mendes

    Biochemistry by numbers: simulation of biochemical pathways with Gepasi 3

    Trends Biochem. Sci.

    (1997)
  • M.A. Peletier et al.

    Control of spatially heterogeneous and time-varying cellular reaction networks: a new summation law

    J. Theor. Biol.

    (2003)
  • K.A. Reijenga et al.

    Control analysis for autonomously oscillating biochemical networks

    Biophys. J.

    (2002)
  • L.K. Shawver et al.

    Smart drugs: tyrosine kinase inhibitors in cancer therapy

    Cancer Cell

    (2002)
  • J.W. Stucki et al.

    Mathematical modeling of the regulation of caspase-3 activation and degradation

    J. Theor. Biol.

    (2005)
  • B. Alberts et al.

    Cell communication

    Molecular Biology of the Cell

    (2002)
  • A.R. Asthagiri et al.

    A computational study of feedback effects on signal dynamics in a mitogen-activated protein kinase (mapk) pathway model

    Biotechnol. Prog.

    (2001)
  • M. Bentele et al.

    Mathematical modeling reveals threshold mechanism in CD95-induced apoptosis

    J. Cell Biol.

    (2004)
  • U.S. Bhalla et al.

    Emergent properties of networks of biological signaling pathways

    Science

    (1999)
  • E.C. Butcher et al.

    Systems biology in drug discovery

    Nat. Biotechnol.

    (2004)
  • S.L. Campbell et al.

    Increasing complexity of Ras signaling

    Oncogene

    (1998)
  • M. Cascante et al.

    Metabolic control analysis in drug discovery and disease

    Nat. Biotechnol.

    (2002)
  • M. Chaplain et al.

    Mathematical modelling of tumour-induced angiogenesis: network growth and structure

    Cancer Treat. Res.

    (2004)
  • R. Christopher et al.

    Data-driven computer simulation of human cancer cell

    Ann. N. Y. Acad. Sci.

    (2004)
  • DeVita, V.T., Hellman, S., Rosenberg, S.A. Cancer: Principles and Practice of Oncology, sixth ed. Lippincott Williams &...
  • R.J. Ellis et al.

    Cell biology: join the crowd

    Nature

    (2003)
  • Fell, D.A. Understanding the Control of Metabolism. Portland Press, London,...
  • N. Ferrara

    Vascular endothelial growth factor as a target for anticancer therapy

    Oncologist

    (2004)
  • J.E. Ferrell

    Building a cellular switch: more lessons from a good egg

    Bioessays

    (1999)
  • P.A. Futreal et al.

    A census of human cancer genes

    Nat. Rev. Cancer

    (2004)
  • Cited by (339)

    View all citing articles on Scopus
    View full text