Optimizing targeted cancer therapy: Towards clinical application of systems biology approaches

https://doi.org/10.1016/j.critrevonc.2011.05.002Get rights and content

Abstract

In cancer, genetic and epigenetic alterations ultimately culminate in discordant activation of signal transduction pathways driving the malignant process. Pharmacological or biological inhibition of such pathways holds significant promise with respect to devising rational therapy for cancer. Thus, technical concepts pursuing robust characterization of kinase activity in tissue samples from cancer patients have been subject of investigation. In the present review we provide a comprehensive overview of these techniques and discuss their advantages and disadvantages for systems biology approaches to identify kinase targets in oncological disease.

Recent advances in the development and application of array-based peptide-substrate kinase activity screens show great promise in overcoming the discrepancy between the evaluation of aberrant cell signaling in specific malignancies or even individual patients and the currently available ensemble of highly specific targeted treatment strategies. These developments have the potential to result in a more effective selection of kinase inhibitors and thus optimize mechanism-based patient-specific therapeutic strategies. Given the results from current research on the tumor kinome, generating network views on aberrant tumor cell signaling is critical to meet this challenge.

Introduction

Cancer is defined as an uncontrolled proliferation of clonally derived cells. This malignant transformation is characterized by changes in the expression and activity of key mediators of signal transduction coordinating proliferation, migration and cell death [1]. The tremendous significance of changes in cell signaling activity in the malignant transformation of cells has only been fully recognized in the last decade [2], [3], [4]. The identification of disease-related signal transduction effectors culminated into the acknowledgement of protein kinases as one of the most important classes of potential drug targets to date [5]. Significant advances in the development of small molecule inhibitors to counteract aberrant cell signaling promoting tumor cell proliferation have been made since then. Kinase signaling, because of the excellent specific drugability of the ATP-binding pocket, as well as the general importance in virtually all signal transduction pathways, holds great promise here. The first breakthrough was the approval of the Abl kinase inhibitor Imatinib for treatment of BCR-Abl positive chronic myeloid leukemia [6]. Application of this inhibitor is at present part of first-line treatment for chronic myeloid leukemia and has improved patient outcome dramatically.

Despite the successful application of kinase inhibitors for the treatment of specific malignancies, tumor resistance due to acquired mutations in downstream effectors of the targeted molecule and toxicities as a result of limited agent specificity remain important challenges to overcome [7], [8], [9], [10], [11], [12], [13]. Insufficient ways to predict the efficacy of inhibiting specific kinase activity gives rise to a discrepancy between the evaluation of aberrant cell signaling and the currently available ensemble of highly specific targeted treatment strategies. Hence, providing better ways to predict the efficacy of kinase-targeted small molecule inhibitors in the tumor- or patient-specific situation will be one of the most important goals of cancer research in the upcoming decade. Accurate identification of tumor type-specific or even patient-specific aberrations in cell signaling activity that are crucial for tumor progression, is required. In this review we provide an overview of recent developments in kinase activity screening concepts focused on achieving that goal. An introduction concerning aberrant protein kinase activity as the essence of the oncogenic phenotype will be followed by a discussion of the gap in knowledge on anomalous cell signaling in specific malignancies that is becoming ever more evident, thereby severely hampering the implementation of kinome-targeted cancer therapies. After a description of the most apparent challenges that have emerged in recent research, we will address the most encouraging strategies aimed at dissecting the cancer-specific signal transduction network. Promising research focused on allowing us to obtain a better grip on the selection of effective and specific kinome-targeted cancer treatment strategies will be evaluated. Future investigational topics will be discussed.

Section snippets

Molecular biology of cancer cells: targeting signaling kinases

Up until the last decade, cancer research essentially relied on the expression of messenger RNA and, to a lesser extent, protein to elucidate changes in cell signaling [14]. Microarray applications have made it possible to simultaneously study the expression of a tremendous number of genes, thus providing a network view on gene expression. The concept of personal genomics in individualized medicine is aiming at the assessment of risk and the selection of therapeutic approaches based on the

Application of kinome-targeted therapies: opportunities and challenges

Broad spectrum kinase inhibition tends to narrow the therapeutic window of compounds because of serious side effects on non-malignant tissues [47]. Nevertheless, a therapeutic window proves to exist for an increasing number of compounds with a side effect profile that challenges that of the traditional cytotoxic agents [48]. Examples are Imatinib and the relatively broad-spectrum BCR-Abl and Src-family kinase inhibitor Dasatinib, which has been approved for treatment of chronic myeloid leukemia

Quantitative phosphoproteomics

Phosphoproteomics studies using mass spectrometry techniques already provided us with a wealth of insight in reversible phosphorylation events that occur in the mammalian cell. The number of potential phosphorylation sites is estimated to be several hundred thousand in the human proteome, representing a large combinatorial repertoire for proteome regulation [65]. This level of complexity makes it hard to reliably quantify specific phosphorylation states in order to interpret the signaling

Conclusions and future perspectives: bridging the gap

Effective mechanism-based cancer treatment strategies specifically target cell signaling mediators which are critical to cell survival and tumor progression. Increasing numbers of kinase-targeted small molecules with high specificity are currently becoming available. Despite the successful application of relatively broad spectrum kinase inhibitors for the treatment of specific malignancies (Table 1), insufficient ways to predict the efficacy of inhibiting specific kinase activity gives rise to

Conflict of interest statement

The authors declare no conflict of interest.

Role of funding source

The study sponsors did not play a role in the study design, in the collection, analysis and interpretation of data, the writing of the manuscript and in the decision to submit the manuscript for publication.

Reviewers

Helena B. Nader, PhD, Universidade Federal de Sao Paulo, Department of Biochemistry, Rua Tres de Maio 100, Sao Paulo, SP 04044-020, Brazil.

Prof. Anne Hansen Ree, Akershus University Hospital, Department of Oncology, N-1478 Lorenskog, Norway.

Elisa Giovannetti, MD, PhD, Post-Doc Scientific Researcher, VU University, Medical Oncology, De Boelelaan 1117, NL-1081 HV Amsterdam, Netherlands.

Acknowledgements

Arend H. Sikkema is supported by a grant from the Foundation of Pediatric Oncology Groningen, the Netherlands (Grant: SKOG-05-001).

We thank Mrs. J.E. den Dunnen-Briggs for correction of the English grammar.

Arend H. Sikkema, M.Sc., is a Ph.D.-student in the Division of Pediatric Oncology/Hematology, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. He graduated in Medical Biology at the University of Groningen in 2006. His current research focuses on the identification of aberrant kinase activity in pediatric brain tumors.

References (188)

  • S.M. Kornblau et al.

    Simultaneous activation of multiple signal transduction pathways confers poor prognosis in acute myelogenous leukemia

    Blood

    (2006)
  • K. Inoki et al.

    TSC2 mediates cellular energy response to control cell growth and survival

    Cell

    (2003)
  • B.D. Manning et al.

    Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway

    Mol Cell

    (2002)
  • A. Garami et al.

    Insulin activation of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1 and 2

    Mol Cell

    (2003)
  • D.E. McNulty et al.

    Hydrophilic interaction chromatography reduces the complexity of the phosphoproteome and improves global phosphopeptide isolation and detection

    Mol Cell Proteomics

    (2008)
  • K. Schmelzle et al.

    Phosphoproteomic approaches to elucidate cellular signaling networks

    Curr Opin Biotechnol

    (2006)
  • H.C. Harsha et al.

    Phosphoproteomics in cancer

    Mol Oncol

    (2010)
  • S.E. Ong et al.

    Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics

    Mol Cell Proteomics

    (2002)
  • C. Pan et al.

    Global effects of kinase inhibitors on signaling networks revealed by quantitative phosphoproteomics

    Mol Cell Proteomics

    (2009)
  • P.L. Ross et al.

    Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents

    Mol Cell Proteomics

    (2004)
  • F.S. Oppermann et al.

    Large-scale proteomics analysis of the human kinome

    Mol Cell Proteomics

    (2009)
  • P.A. Everley et al.

    Quantitative cancer proteomics: stable isotope labeling with amino acids in cell culture (SILAC) as a tool for prostate cancer research

    Mol Cell Proteomics

    (2004)
  • K. Rikova et al.

    Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer

    Cell

    (2007)
  • T. Sonoda et al.

    Mass-tag technology for monitoring of protein kinase activity using mass spectrometry

    Bioorg Med Chem Lett

    (2004)
  • M.F. Moran et al.

    Emerging applications for phospho-proteomics in cancer molecular therapeutics

    Biochim Biophys Acta

    (2006)
  • R. Linding et al.

    Systematic discovery of in vivo phosphorylation networks

    Cell

    (2007)
  • T. Pawson et al.

    Protein–protein interactions define specificity in signal transduction

    Genes Dev

    (2000)
  • P. Cohen

    Protein kinases—the major drug targets of the twenty-first century?

    Nat Rev Drug Discov

    (2002)
  • B.J. Druker et al.

    Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia

    N Engl J Med

    (2001)
  • P.D. Bonomi et al.

    Selecting patients for treatment with epidermal growth factor tyrosine kinase inhibitors

    Clin Cancer Res

    (2007)
  • A. de Reynies et al.

    KRAS mutation signature in colorectal tumors significantly overlaps with the cetuximab response signature

    J Clin Oncol

    (2008)
  • A. Sartore-Bianchi et al.

    PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies

    Cancer Res

    (2009)
  • C.Q. Zhu et al.

    Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21

    J Clin Oncol

    (2008)
  • J.R. Sierra et al.

    Molecular mechanisms of acquired resistance to tyrosine kinase targeted therapy

    Mol Cancer

    (2010)
  • G. Chambers et al.

    Proteomics: a new approach to the study of disease

    J Pathol

    (2000)
  • D.H. Roukos et al.

    Human genetic and structural genomic variation: would genome-wide association studies be the solution for cancer complexity like Alexander the Great for the “Gordian Knot”?

    Ann Surg Oncol

    (2009)
  • D. Ziogas et al.

    Genetics and personal genomics for personalized breast cancer surgery: progress and challenges in research and clinical practice

    Ann Surg Oncol

    (2009)
  • D. Ziogas et al.

    Challenges in developing robust genetic markers and targets to predict and prevent distant and peritoneal recurrence in gastric cancer

    Ann Surg Oncol

    (2009)
  • M. Kool et al.

    Integrated genomics identifies five medulloblastoma subtypes with distinct genetic profiles, pathway signatures and clinicopathological features

    PLoS One

    (2008)
  • M.C. Thompson et al.

    Genomics identifies medulloblastoma subgroups that are enriched for specific genetic alterations

    J Clin Oncol

    (2006)
  • P.A. Northcott et al.

    Medulloblastoma comprises four distinct molecular variants

    J Clin Oncol

    (2010)
  • L.J. ’t Veer et al.

    Gene expression profiling predicts clinical outcome of breast cancer

    Nature

    (2002)
  • H.Y. Chang et al.

    Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival

    Proc Natl Acad Sci USA

    (2005)
  • L.D. Miller et al.

    An expression signature for p53 status in human breast cancer predicts mutation status, transcriptional effects, and patient survival

    Proc Natl Acad Sci USA

    (2005)
  • H. Bonnefoi et al.

    New predictive factors for chemosensitivity of breast cancers

    Bull Cancer

    (2008)
  • L. Harris et al.

    American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer

    J Clin Oncol

    (2007)
  • S. Paik et al.

    A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer

    N Engl J Med

    (2004)
  • J.J. de Ronde et al.

    Concordance of clinical and molecular breast cancer subtyping in the context of preoperative chemotherapy response

    Breast Cancer Res Treat

    (2010)
  • B. Kreike et al.

    Gene expression profiling and histopathological characterization of triple-negative/basal-like breast carcinomas

    Breast Cancer Res

    (2007)
  • B. Kobe et al.

    Active site-directed protein regulation

    Nature

    (1999)
  • Cited by (16)

    • The active kinome: The modern view of how active protein kinase networks fit in biological research

      2022, Current Opinion in Pharmacology
      Citation Excerpt :

      As such, rather than an inherent mutation in one coding region of a particular kinase, variations in phosphorylation regions may rewire the interaction network of a specific protein kinase. Changes in the interaction network of a protein kinase may explain why inhibition of an individual protein kinase may not be an effective therapeutic strategy [32,33]. Broadly inhibiting protein kinases and phosphorylation targets which are highly perturbed in cancer may correct for aberrations in other cellular signaling networks, reversing pathologic cellular states.

    • Tumor phosphatidylinositol 3-kinase signaling in therapy resistance and metastatic dissemination of rectal cancer: Opportunities for signaling-adapted therapies

      2015, Critical Reviews in Oncology/Hematology
      Citation Excerpt :

      Ideally, to become clinically useful, technologies must provide rapid and high-fidelity analysis of small-quantity samples. Kinase substrate array technologies are high-throughput tools for global profiling of kinase activities in tissue samples without prior knowledge of which signaling pathways are activated [33–36]. Using such arrays, each tumor sample will generate an individual phosphosubstrate signature, theoretically portraying the state of composite information flow through signaling cascades.

    View all citing articles on Scopus

    Arend H. Sikkema, M.Sc., is a Ph.D.-student in the Division of Pediatric Oncology/Hematology, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. He graduated in Medical Biology at the University of Groningen in 2006. His current research focuses on the identification of aberrant kinase activity in pediatric brain tumors.

    Eveline de Bont, M.D., Ph.D., is associate professor and head of the Division of Pediatric Oncology/Hematology, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. She graduated in Medicine at the University of Amsterdam in 1989. She has worked at the Department for Cancer Biology, Harvard School of Public Health, Boston, USA for 1 year in 1997. She obtained her Ph.D. degree in Medical Sciences at the University of Groningen in 1999. Her main research interests are focused on dissecting the role (and mechanism) of VEGF and VEGFR signaling on tumor cells and surrounding tumor stromal cells in pediatric brain tumors and acute myeloid leukemia to develop new therapeutic strategies.

    View full text