Elsevier

Seminars in Cancer Biology

Volume 53, December 2018, Pages 110-124
Seminars in Cancer Biology

Molecular pathway activation – New type of biomarkers for tumor morphology and personalized selection of target drugs

https://doi.org/10.1016/j.semcancer.2018.06.003Get rights and content

Abstract

Anticancer target drugs (ATDs) specifically bind and inhibit molecular targets that play important roles in cancer development and progression, being deeply implicated in intracellular signaling pathways. To date, hundreds of different ATDs were approved for clinical use in the different countries. Compared to previous chemotherapy treatments, ATDs often demonstrate reduced side effects and increased efficiency, but also have higher costs. However, the efficiency of ATDs for the advanced stage tumors is still insufficient. Different ATDs have different mechanisms of action and are effective in different cohorts of patients. Personalized approaches are therefore needed to select the best ATD candidates for the individual patients. In this review, we focus on a new generation of biomarkers – molecular pathway activation – and on their applications for predicting individual tumor response to ATDs. The success in high throughput gene expression profiling and emergence of novel bioinformatic tools reinforced quick development of pathway related field of molecular biomedicine. The ability to quantitatively measure degree of a pathway activation using gene expression data has revolutionized this field and made the corresponding analysis quick, robust and inexpensive. This success was further enhanced by using machine learning algorithms for selection of the best biomarkers. We review here the current progress in translating these studies to clinical oncology and patient-oriented adjustment of cancer therapy.

Introduction

For about seven decades, chemotherapy remains a key treatment for many cancers, often with impressive success rates. For example, the use of cisplatin regimens of testicular cancer treatment turned almost complete mortality to ∼90–95% disease-specific survival [1,2]. However, many cancer types and advancer individual tumors remain incurable and/or unresponsive using standard chemotherapy approaches. Moreover, chemotherapy frequently causes severe side effects, which may dramatically decrease the quality of life of a patient [3,4]. The chemical compounds included in standard chemotherapy cocktails have numerous, sometimes poorly characterized molecular targets in both cancerous and normal proliferating cells, which makes it difficult to simulate and predict the activity of drugs to an individual patient, and in standard practice clinicians routinely use clinical or morphological predictive factors like stage, grade, proliferative activity, etc [5,6]. These predictive factors are mostly inaccurate and poorly applicable for tracing the individual patient response to chemotherapy drugs and regimens.

To address specific activities of tumor marker proteins frequently overexpressed in cancer, a new generation of drugs termed Target drugs was created that target one or a few specific molecules in a cell [7]. Drugs can also target abnormal fusion proteins, such as chimeric fusions AML1-ETO or BCR-ABL frequently formed in leukemias, and the corresponding drugs imatinib and oridonin [[8], [9], [10]].

Target drugs consist of either specific monoclonal antibodies Mabs or low molecular weight inhibitor molecules, e.g. specific kinase inhibitors Nibs [11]. Different target drugs have different mechanisms of action and are effective to different cohorts of patients. For now, more than a hundred of different target drugs have been approved for use in cancer treatment and are present on the global pharmaceutical market (e.g., see www.drugbank.ca). For several cancer types, the emergence of target drugs was highly beneficial. For example, trastuzumab (anti-HER2 monoclonal antibody) and several other new anti-HER2 medications at least doubled median survival time in patients with metastatic HER2-positive breast cancer and improved 5-year survival in early stage disease to ∼90-95% [12,13]. Interestingly, before the introduction of trastuzumab, HER2-positive cancers had the worst prognosis across all breast cancer subtypes, whereas now the situation has reverted [14]. Melanoma patients for decades had no treatment opportunities except dacarbazine chemotherapy, which resulted in only a tiny, less than 10%, chance of short-lasting (∼5-6 month long) response and very low median survival of less than a year. Now, in the case of BRAF-mutated tumor, they can receive vemurafenib (anti-BRAF target drug) and have ∼50% chance of response [15], or, irrespectively of BRAF mutation, immune checkpoint inhibitor target therapies with ∼20% chance of long-term (>5 years) disease control [16]. Target drugs were also of a great benefit for advanced kidney cancers, which used to be mostly uncurable before [17]. Compared to the previous chemotherapy treatments, target drugs often cause reduced side effects and increased efficiency, but also have higher costs.

However, many individual advancer tumor cases and even entire cancer types (e.g. glioblastoma and pancreatic cancer) remain poorly responding to the target therapies [18]. Importantly, the results of clinical trials clearly evidence that in many cases the drugs considered inefficient for an overall cancer type, show significant benefit for a small fraction of the patients. For example, anti-EGFR drugs gefitinib and erlotinib showed no overall benefit in large randomized trials on patients with non-small cell lung cancer. However, it was observed that ∼10-15% of the patients did respond to the treatment and had longer survival characteristics. Further investigation revealed that the latter group of patients had activating mutation of EGFR gene and that this mutation, therefore, can predict response to the EGFR-targeting therapies [19]. Of note, this approach doesn’t work for colorectal cancer, where EGFR-mutated status has little predictive power for anti-EGFR drugs cetuximab and panitumumab. However, discovery of the role of KRAS mutation in the resistance to the EGFR-targeting antibodies helped to identify a group of patients that can benefit from this type of treatment - patients with wild-type status of KRAS. Moreover, further investigations showed that for the KRAS-mutant tumors (∼40% of colorectal cancer), anti-EGFR antibodies cause harm and even decrease survival [20].

It is, therefore, of great importance to identify strong predictive markers of target drug efficacy, for as many cancer-drug combinations as possible. Several molecular tests have been designed to identify individualized cancer treatments [21,22]. Like the examples mentioned above, these tests mostly utilize data on single somatic mutations or expressions of single or few genes. Unfortunately, most of those predictor features are applicable only to a relatively small number of patients – because they cover only a minor fraction of target drugs, in a minor fraction of cancer types. Thus, the problem of the efficient ATD selection remains largely unsolved. The price for incorrect or inefficient treatment is losing valuable patients’ time, decreased quality and duration of life and spending excessive financial resources. It became clear nowadays that somewhat more universal methods are needed to rank the maximum number of the existing drugs for an individual patient. Solving this task would certainly require high-throughput screening of the patient-specific genetic information, including deep assessment of the patient’s tumor specific gene expression profile. In this review, we focus on the recent advancements in the application to a tumor biology of a next level of the molecular data analysis - activation of intracellular molecular pathways. We outline here the methods utilized for molecular pathway activation analysis and their applications for personalizing the use of target drugs.

Section snippets

Molecular pathways and cancer biology

Intracellular molecular pathways (IMPs) combine gene products implicated in certain molecular functions. IMPs are involved in all major events in the normal and cancer cells. The best-known IMPs are signaling, metabolic, DNA repair and cytoskeleton organization pathways [23]. The metabolic pathway combines biochemical reactions in a continuously connected network having a biologically significant output. The signaling pathway, in turn, consolidates gene products involved in signal transduction.

Pathway analysis and gene expression data

In various experimental models, molecular pathway activation can be measured through the extensive screening of protein phosphorylation, by interrogating switches of transcription factor binding sites, using specific gene expression or protein aggregation biosensors, by measuring physiological outputs and by many other methods [48,49]. However, those approaches have an obvious strong limitation that they can’t be reproduced for the biomaterial of tumor patients, especially formalin-fixed,

Pathways as tumor biomarkers

Finding reliable and accurate molecular markers of cancer remains one the major priorities of the contemporary molecular medicine. Every year, thousands of reports communicate new RNA, DNA mutation or methylation, protein and non-protein biochemical biomarkers sensitive to cancer development [83]. Most of the expression biomarkers represent RNA or protein products for the individual genes. However, there is a clear trend towards finding next-level biomarkers of cancer consisting of gene

Pathway-based scoring of cancer drug efficiencies

Overall, the pathway-based methods of predicting drug efficiency can be classified in two major groups: (i) those using PAS signatures for the IMPs as more or less simple statistical coincidence biomarkers of drug response and (ii) those using knowledge of specific drug molecular mechanisms and PAS values to simulate activities of target drugs in an individual tumor.

Translation to personalized medicine

The drug scoring methods reviewed here rely on the high throughput gene expression data. However, the first published study utilizing molecular profiling to find potential targets and select treatments was a recent pilot prospective investigation utilizing molecular profiling of tumors by immunohistochemistry, fluorescence in situ hybridization (FISH) and DNA microarrays to interrogate mutation profiles, completed by the Translational Genomics Research Institute. This multicenter (9 sites in

Challenges and opportunities

Tumor represents a mixture of different cell populations with distinct phenotypes, genotypes and epigenetic modifications. The apparent heterogeneity of high-grade tumors may be due to various factors like influence of the different cancer stem cells niches, alternative mutation profiles and differential vascularization features. This creates obvious obstacles to the molecular diagnostics of tumor, drug scoring and cancer treatment. Current RNAseq technology is an effective tool to study the

Conclusion

A rapidly growing field of molecular pathway analytics highly benefits from transition to the new level of data analysis. Studies of single genes (including expression, mutation, epigenetic regulation, molecular interactions, etc) are being replaced by the attempts of building functional models involving hundreds and thousands of gene products in their interaction networks. The emergence of the related techniques balancing on the interface of advanced computer technologies and high throughput

Acknowledgment

This study was supported by the Russian Science Foundation grant 18-15-00061.

References (155)

  • J. Oldenburg et al.

    On behalf of: SWENOTECA (Swedish Norwegian Testicular Cancer Group), the Italian Germ Cell Cancer Group (IGG), Spanish Germ Cell Cancer Group (SGCCG), Personalizing, not patronizing: the case for patient autonomy by unbiased presentation of management options in stage I testicular cancer

    Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO.

    (2014)
  • T.A. Ahles et al.

    Quality of life of long-term survivors of breast cancer and lymphoma treated with standard-dose chemotherapy or local therapy

    J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol.

    (2005)
  • A.E. Kayl et al.

    Side-effects of chemotherapy and quality of life in ovarian and breast cancer patients

    Curr. Opin. Obstet. Gynecol.

    (2006)
  • N.V. Zhukov et al.

    Targeted therapy in the treatment of solid tumors: practice contradicts theory

    Biochem. Biokhimii͡a

    (2008)
  • J. Sjöström

    Predictive factors for response to chemotherapy in advanced breast cancer

    Acta Oncol. Stockh. Swed.

    (2002)
  • C. Sawyers

    Targeted cancer therapy

    Nature

    (2004)
  • B.J. Druker et al.

    Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome

    N. Engl. J. Med.

    (2001)
  • 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. Spirin et al.

    Synergistic suppression of t(8;21)-positive leukemia cell growth by combining oridonin and MAPK1/ERK2 inhibitors

    Oncotarget

    (2017)
  • S. Aggarwal

    Targeted cancer therapies

    Nat. Rev. Drug Discov.

    (2010)
  • C.A. Hudis

    Trastuzumab – mechanism of action and use in clinical practice

    N. Engl. J. Med.

    (2007)
  • R. Nahta et al.

    Trastuzumab: triumphs and tribulations

    Oncogene

    (2007)
  • A.A. Onitilo et al.

    Breast cancer subtypes based on ER/PR and Her2 expression: comparison of clinicopathologic features and survival

    Clin. Med. Res.

    (2009)
  • P.B. Chapman et al.

    BRIM-3 study group, improved survival with vemurafenib in melanoma with BRAF V600E mutation

    N. Engl. J. Med.

    (2011)
  • P.A. Prieto et al.

    CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma

    Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res.

    (2012)
  • M. Ghidini et al.

    Clinical development of mTor inhibitors for renal cancer

    Expert Opin. Investig. Drugs

    (2017)
  • Institute for Quality and Efficiency in Health Care

    Curation vs. Palliation: An Attempt to Clarify Terms

    (2014)
  • A. Grothey et al.

    Explaining the unexplainable: EGFR antibodies in colorectal cancer

    J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol.

    (2012)
  • J. Hornberger et al.

    Economic analysis of targeting chemotherapy using a 21-gene RT-PCR assay in lymph-node-negative, estrogen-receptor-positive, early-stage breast cancer

    Am. J. Manag. Care

    (2005)
  • C. Le Tourneau et al.

    Randomised proof-of-concept phase II trial comparing targeted therapy based on tumour molecular profiling vs conventional therapy in patients with refractory cancer: results of the feasibility part of the SHIVA trial

    Br. J. Cancer.

    (2014)
  • M.V. Blagosklonny

    MTOR-driven quasi-programmed aging as a disposable soma theory: blind watchmaker vs. intelligent designer

    Cell Cycle Georget. Tex.

    (2013)
  • C. Sonnenschein et al.

    The aging of the 2000 and 2011 hallmarks of cancer reviews: a critique

    J. Biosci.

    (2013)
  • A.M. Aliper et al.

    Mathematical justification of expression-based pathway activation scoring (PAS)

    Methods Mol. Biol. Clifton NJ

    (2017)
  • N. Borisov et al.

    Systems-level interactions between insulin-EGF networks amplify mitogenic signaling

    Mol. Syst. Biol.

    (2009)
  • N.B. Kuzmina et al.

    Handling complex rule-based models of mitogenic cell signaling (on the example of ERK activation upon EGF stimulation)

    Int. Proc. Chem. Biol. Environ. Eng.

    (2011)
  • M. Filteau et al.

    Systematic identification of signal integration by protein kinase A

    Proc. Natl. Acad. Sci.

    (2015)
  • D. Branzei et al.

    Regulation of DNA repair throughout the cell cycle

    Nat. Rev. Mol. Cell Biol.

    (2008)
  • M. Malumbres et al.

    Cell cycle, CDKs and cancer: a changing paradigm

    Nat. Rev. Cancer

    (2009)
  • K. Vermeulen et al.

    The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer

    Cell Prolif.

    (2003)
  • D. Croft et al.

    The reactome pathway knowledgebase

    Nucleic Acids Res.

    (2014)
  • R. Elkon et al.

    SPIKE--a database, visualization and analysis tool of cellular signaling pathways

    BMC Bioinform.

    (2008)
  • A. Nakaya et al.

    KEGG OC: a large-scale automatic construction of taxonomy-based ortholog clusters

    Nucleic Acids Res.

    (2013)
  • A. Nikitin et al.

    Pathway studio--the analysis and navigation of molecular networks

    Bioinform. Oxf. Engl.

    (2003)
  • Uni Prot Consortium

    Ongoing and future developments at the universal protein Resource

    Nucleic Acids Res.

    (2011)
  • S. Mathivanan et al.

    An evaluation of human protein-protein interaction data in the public domain

    BMC Bioinform.

    (2006)
  • R. Elkon et al.

    SPIKE--a database, visualization and analysis tool of cellular signaling pathways

    BMC Bioinform.

    (2008)
  • A. Bauer-Mehren et al.

    Pathway databases and tools for their exploitation: benefits, current limitations and challenges

    Mol. Syst. Biol.

    (2009)
  • H. Nakagawa et al.

    Genome sequencing analysis of liver cancer for precision medicine

    Semin. Cancer Biol.

    (2018)
  • J.R. Ramroop et al.

    Impact of phosphoproteomics in the era of precision medicine for prostate cancer

    Front. Oncol.

    (2018)
  • G.M. Otto et al.

    Seq-ing answers: uncovering the unexpected in global gene regulation

    Curr. Genet.

    (2018)
  • Cited by (68)

    • Immunotherapy-related biomarkers: Confirmations and uncertainties

      2023, Critical Reviews in Oncology/Hematology
    • Gene Expression Signature of Endometrial Samples from Women with and without Endometriosis

      2021, Journal of Minimally Invasive Gynecology
      Citation Excerpt :

      Therefore, although the final signature genes were not determined to be differential in the previous studies of endometriosis, these genes could be tightly connected with the maintenance of a healthy endometrium state and normal menstrual cycle. The proposed 2-step framework of the next-generation sequencing data-driven approach could be used in further research of complex gene expression biomarkers at the levels of individual genes or molecular pathways that determine the onset and progression of endometriosis [29]. Moreover, the final 5-gene signature supports the implementation of nonsurgical diagnostics of endometriosis through endometrial sampling.

    View all citing articles on Scopus
    View full text