Skip to main content
Nucleic Acids Research logoLink to Nucleic Acids Research
. 2013 Nov 20;42(Database issue):D1118–D1123. doi: 10.1093/nar/gkt1129

Therapeutic target database update 2014: a resource for targeted therapeutics

Chu Qin 1,2,3, Cheng Zhang 1,4,5,6, Feng Zhu 7, Feng Xu 4,5, Shang Ying Chen 1, Peng Zhang 1, Ying Hong Li 7, Sheng Yong Yang 2, Yu Quan Wei 2, Lin Tao 1,3,*, Yu Zong Chen 1,2,4,5,*
PMCID: PMC3964951  PMID: 24265219

Abstract

Here we describe an update of the Therapeutic Target Database (http://bidd.nus.edu.sg/group/ttd/ttd.asp) for better serving the bench-to-clinic communities and for enabling more convenient data access, processing and exchange. Extensive efforts from the research, industry, clinical, regulatory and management communities have been collectively directed at the discovery, investigation, application, monitoring and management of targeted therapeutics. Increasing efforts have been directed at the development of stratified and personalized medicines. These efforts may be facilitated by the knowledge of the efficacy targets and biomarkers of targeted therapeutics. Therefore, we added search tools for using the International Classification of Disease ICD-10-CM and ICD-9-CM codes to retrieve the target, biomarker and drug information (currently enabling the search of almost 900 targets, 1800 biomarkers and 6000 drugs related to 900 disease conditions). We added information of almost 1800 biomarkers for 300 disease conditions and 200 drug scaffolds for 700 drugs. We significantly expanded Therapeutic Target Database data contents to cover >2300 targets (388 successful and 461 clinical trial targets), 20 600 drugs (2003 approved and 3147 clinical trial drugs), 20 000 multitarget agents against almost 400 target-pairs and the activity data of 1400 agents against 300 cell lines.

INTRODUCTION

Modern drug development has been primarily focused on targeted therapeutics (1–3) with increasing movement toward stratified and personalized medicines (4–6). Extensive efforts from the research, industry, clinical, regulatory and management communities and the chemistry, biology, pharmaceutics and medicine disciplines have been collectively directed at the discovery, investigation, application, monitoring and management of targeted therapeutics and biomarkers (4,7–10). The knowledge of the efficacy targets and biomarkers is useful not only for the discovery and development of targeted therapeutics (11,12) but also for facilitating the development and practice of stratified and personalized medicines (4,13,14).

In particular, the information of targeted therapeutics and biomarkers may be potentially incorporated into the widely used disease classification systems for more refined classification of disease subclasses and patient subpopulations responsive to a particular treatment so as to better facilitate the diagnosis, prescription, monitoring and management of patient care in stratified and personalized medicines. Although the information about targeted therapeutics and biomarkers can be obtained from the established drug (15), efficacy target (16) and biomarker (17–19) databases, the data retrieval tools of these databases are not specifically designed for optimally supporting such tasks. There is a need to enable data retrieval by using the widely used International Classification of Diseases (ICD) codes (20,21) for facilitating broader, more convenient and automatic data access, processing and exchange by the bench-to-clinic communities, particularly non-domain experts.

To better serve the multiple bench-to-clinic communities and to facilitate the development and practice of stratified and personalized medicines, we made several major improvements to the Therapeutic Target Database (TTD, http://bidd.nus.edu.sg/group/ttd/ttd.asp). First, we added information and search tools based on the ICD codes (22,23) for searching the targets, biomarkers, drugs and other TTD data related to various disease conditions. For more extensive coverage of potential biomarkers and for enabling their convenient access by the ICD codes, we added a significantly higher number (1755) of literature-reported biomarkers for more variety of disease conditions (365) than those in the existing biomarker databases that primarily focus on molecular biomarkers of specific disease classes (17,19) or clinically prioritized sets (18). We also added information and enabled the search of TTD data via drug scaffold names (227 scaffolds for 736 drugs and leads) for facilitating the search of the drugs, targets and diseases related to specific molecular scaffolds. Moreover, we added the Anatomical Therapeutic Chemical (ATC) Classification System codes for 1521 approved drugs for supporting the convenient and automated access of clinical drug data (24).

By using the literature search methods described in our earlier article (16), we also significantly expanded TTD contents to include 388 successful, 461 clinical trial and 1467 research targets; 2003 approved (1008 nature product derived), 3147 clinical trial, 498 discontinued clinical trial and 14 856 experimental drugs, 20 818 multitarget agents against 385 target-pairs and the activity data of 1436 drugs against 274 cell lines. These are compared with the 364 successful, 286 clinical trial and 1331 research targets; 1540 approved (939 natural product derived), 1423 clinical trial, 345 discontinued clinical trial and 14 853 experimental drugs, and 3681 multitarget agents active against 108 target pairs in our last update (16). The statistics of our updated data is summarized in Table 1.

Table 1.

Statistics of the drug targets, drugs and their structure and potency data in 2014 version of TTD database

Category Item 2014 Update 2012 Update
Statistics of drug targets Number of all targets 2360 2025
Number of successful targets 388 364
Number of clinical trial targets 461 286
Number of research targets 1467 1331
Statistics of drugs Number of all drugs 20 667 17 816
Number of approved drugs (no of natural product derived drugs) 2003 (1008) 1540 (939)
Number of clinical trial drugs (no of natural product derived drugs) 3147 (369) 1423 (369)
Number of discontinued drugs 498 345
Number of pre-clinical drugs 163 165
Number of experimental drugs 14 856 14 853
Number of multitarget agents 20 818 3681
Number of drug combinations 115 115
Statistics of drugs with available structure or sequence data Number of small molecular drugs with available structure 17 012 14 170
Number of antisense drugs with available sequence data 652 652
Statistics of drugs with activity data or structure-activity relationship Number of agents with potency data against target 11 810 11 810
Number of agents with potency data against a disease model such as a cell-line, ex vivo, in vivo model 1753 497
Number of quantitative structure-activity relationship qsar models (no of chemical types) 841 (228) 841 (228)

International classification of diseases

ICD has been developed by the World Health Organization (WHO), sponsored by the United Nations, adopted by >110 countries and used by physicians, researchers, nurses, health workers, health information managers, policy makers, insurers and health program managers for defining and studying diseases, monitoring and managing health care and allocating resources (20,21). ICD codes have been regularly revised to the current version ICD-10 (20). But the previous version ICD-9 is still used by some organizations while proceeding with the transition to ICD-10 (the expected completion date for the transition to ICD-10 in the United States is October 1, 2014) (25). ICD-10 is composed of 68 000 alphanumeric codes as compared with the 13 000 numeric codes in ICD-9, thus offering more comprehensive coverage and better representation of medical conditions (20). A number of nations have developed their own adaptations of the ICD codes. For instance, the United States have developed ICD-9 and ICD-10 clinical modification ICD-9-CM (17 000 codes) and ICD-10-CM (155 000 codes) for covering additional morbidity details (26), which were used in TTD because of their more comprehensive coverage.

The ICD-9-CM and ICD-10-CM codes were matched to the TTD target, drug and biomarker entries by the following procedure. First, automated word match was conducted for matching the disease name or names of each TTD target, drug or biomarker entry with the disease descriptions of each ICD codes. Second, each of the fully or partially matched TTD entry was manually checked to either validate the match or to find the right ICD codes. Third, manual search was conducted for every non-matched TTD entries. So far, we were able to find the ICD codes for 785 targets and 3080 drugs related to 732 disease conditions. From the TTD ‘Search drugs and targets by disease or ICD identifier’ field, users can search TTD target and drug entries related to a specific disease or an ICD-9-CM or ICD-10-CM code. The TTD biomarker entries may also be searched by selecting an ICD-9-CM or ICD-10-CM code from the ‘Search for biomarkers’ field. Users may also download from the TTD download page the lists of TTD target, drug and biomarker entries with the corresponding ICD-9-CM and ICD-10-CM codes.

A new ICD version ICD-11 is in development and scheduled for endorsement by WHO in 2015 (WHO. The International Classification of Diseases 11th revision is due by 2015. Retrieved from http://www.who.int/classifications/icd/revision/en/), which offers more refined disease classifications based on more recent scientific understanding of the disease mechanisms. For instance, small cell lung cancer, which represents ∼13% of all lung cancer diagnoses (27), is not explicitly classified in the ICD-10 and earlier ICD versions but is now explicitly represented in the ICD-11 beta draft. Therefore, ICD-11 is expected to be more useful for developing a more refined disease classification system for stratified and personalized medicine. Effort will be made to upgrade TTD to the ICD-11 version on its official release.

Biomarkers

Biomarkers have been developed as non-invasive tests for early detection and indication of disease risks, monitoring of disease progression and recurrence and classification of disease subtypes and patient subpopulations for providing the most appropriate treatments (28–30). As many therapies have been found to elicit markedly different clinical responses in individual patients (31,32), there is a particular need for more biomarkers capable of predicting drug response in individual patients, which has led to intensive efforts in the discovery of such biomarkers (4,33). Table 2 gives examples of the approved and clinically tested biomarkers for facilitating the prescription of a particular drug to specific patient subpopulation. Moreover, there are considerable interests in adopting the multimarker strategy for parallel evaluation of multiple existing and novel biomarkers in the diagnosis and prognostics of diseases and treatment responses in individual patients (34,35). These efforts may be facilitated by significantly expanding biomarker coverage in the biomarker databases. We, therefore, searched literature-reported biomarkers, mapped them to the ICD-9-CM and ICD-10-CM codes and added the relevant information and ICD code search tools in TTD.

Table 2.

Examples of the approved and clinically tested biomarkers for facilitating the prescription of a particular drug to specific patient subpopulation

Disease Therapeutic target Biomarker for the targeted therapeutics Patient subpopulation likely responsive to targeted therapeutics Drug therapy specific for patient subpopulation
Acute promyelocytic leukemia (APL) PML–RAR PML–RAR (gene translocation) APL with PML–RARα t(15:17) translocation Arsenic Trioxide
Alzheimer’s PPAR apolipoprotein E and TOMM40 genotypes and age Mild cognitive impairment due to Alzheimer’s disease Pioglitazone
Breast cancer HER2 HER2 (gene amplification) HER2 amplified and/or over-expressed breast cancer Trastuzumab
Estrogen receptor Estrogen receptor (protein expression) ER overexpressed breast cancer Tamoxifen
PARP BRCA1/2 (mutation) Breast cancer defective in BRCA1 or BRCA2 Olaparib, veliparib
Cystic fibrosis CFTR CFTR G551D mutation Cystic fibrosis patients with CFTR G551D mutation Ivacaftor
Hepatitis C infection HCV non-structural protein 3 IL28B rs12979860 genotype HCV infected patients with IL28B rs12979860 genotype Boceprevir
Melanoma BRAF BRAF V600E (mutation) Melanoma with RAF V600E mutation Vemurafenib, Dabrafenib
MEK BRAF mutations Melanoma with RAF mutations Trametinib
Post-menopausal osteoporosis RANK ligand Post-menopausal women with persistent total hip, femoral neck, or lumbar spine BMD T-scores −1.8 to −4.0, or clinical fracture Post-menopausal osteoporosis at high risk for fractures Denosumab

To broadly cover various types of biomarkers, we conducted comprehensive literature search in the PubMed database (36) by using combination of keywords ‘biomarker’, ‘clinical’, ‘patient’, ‘disease’, ‘drug’ and specific disease names. Additional sources such as the FDA website and the abstracts of the American society of clinical oncology were also systematically searched. Overall we collected 1755 biomarkers for 365 disease conditions, which include both process biomarkers (genetic mutations or alterations, gene amplification and levels of proteins, gene expression, microRNAs, small molecules, or metabolites that capture a molecular/biochemical aspect of disease pathogenesis and the biological responses to the disease process and/or treatment) and global biomarkers (such as tumor sizes, brain structures in neurodegeneration and shape of cells in anemia). These biomarkers may be searched in the ‘Search for biomarkers’ field by using keywords or by selecting an ICD-9-CM or ICD-10-CM code.

Based on the literature descriptions, our collected biomarkers were classified into one or more of the following 12 classes: associative (disease correlation), antecedent (pre-illness risk identification), detective (disease early stage detection), classification (disease categorization and patient assignment for differential treatment), differentiative (differentiation of related diseases), diagnostic (recognition of overt diseases), monitoring (monitoring of disease state or treatment response), pharmacodynamic (examination of the biological basis of drug response variations), pharmacogenomic (genomics-based prediction of drug response, adverse drug reaction and appropriate drug dose), prognostic (prediction of future disease course and response to therapy), surrogate (substitute of a clinical end point for predicting therapeutic benefit) and theragnostic (identification and monitoring of biochemical effects or mode of action of drug and downstream processes) classes.

Apart from the literature-reported biomarkers, the profiles of various known drug resistance mutations (37–39) and drug response regulators (e.g. the genes promoting drug bypass signaling (40,41) or hindering drug actions (42) have been studied for predicting drug resistance, which may be potentially explored as drug response biomarkers (43). Potential biomarkers, particularly multimarkers, have also been predicted from the genetic and gene expression data of patients by using such computational methods as the principal components analysis feature selection method (44), weighted voting classification feature selection method (45), hierarchical clustering feature selection method (46), differentially expressed genes method (47,48) and machine learning feature selection methods (49,50). These potential biomarkers may also be included in TTD and other biomarker databases for facilitating their future exploration.

More refined classification of patient subpopulations for targeted therapeutics

From the examples of the approved and clinically tested drug response biomarkers in Table 2, it seems feasible to incorporate target and biomarker codes into the ICD codes for more refined classification of patient subpopulations responsive to a particular targeted therapy. However, many of the existing biomarkers are based on the profile of a single gene. For highly heterogenetic diseases such as cancers, single-gene biomarkers are highly limited in their coverage of drug escape mechanisms, and multimarkers may be needed for more sufficient coverage of drug escape mechanisms and for more accurate classification of patient subpopulations in stratified and personal medicines. For instance, BRAFV600E inhibitor dabrafenib has shown improved therapeutic effect in BRAFV600E metastatic melanoma patients (51) due in part to its specificity to BRAFV600E tumors with a greater therapeutic window (52). However, drug resistance still emerges (51) partly due to tumor activation of several BRAF inhibitor escape pathways (52–54). Therefore, the use of a single-gene biomarker, BRAFV600E mutation, is insufficient for predicting long-term drug response to dabrafenib therapy, and multimarkers are needed for adequately covering these and other active drug escape mechanisms.

Drug scaffolds

The approved and clinical trial drugs are composed of a limited number of molecular scaffolds (55–57) in contrast to the high number of bioactive molecular scaffolds (58,59). For instance, many drugs have been derived from individual scaffold groups such as macrocycles (60), and 12 FDA-approved anticancer kinase inhibitor drugs (61,62) are grouped into three scaffold groups (63). Investigation and exploration of these highly privileged drug scaffolds are important for discovering new drug-like scaffolds, molecular analogs and drugs. To support the relevant efforts, we searched the literatures for the molecular scaffolds of the approved and clinical trial drugs or their drug leads. By using the combination of keywords drug name or alternative name, ‘scaffold’, ‘discovery’, ‘synthesis’ to search the Pubchem database (36), we found 210 scaffolds for 714 drugs and drug leads. Users can search the TTD drug and target entries related to a drug scaffold by keyword search or by selecting from the list of drug scaffold names in the ‘Search for drug scaffolds’ field.

Remarks

The efforts in the discovery and application of targeted therapeutics increasingly involve collective efforts from multiple bench-to-clinic communities (1–3) and these efforts are increasingly directed at the development of stratified and personalized medicines (4–6). The drug, target, biomarker and other relevant chemical, biological, pharmaceutical and clinical data need to be more integrated and be made easily accessible by the multiple bench-to-clinic communities. These efforts may be partly facilitated by introducing into the relevant databases the ICD code-based data retrieval tools coupled to the other domain knowledge codes such as the codes of drugs (e.g. ATC codes), targets and biomarkers. Continuous efforts will be made to expand the linkage of the ICD and ATC codes to more complete sets of drugs, efficacy targets and biomarkers and to provide the latest and comprehensive information about the drugs, efficacy targets and biomarkers for better serving the multiple bench-to-clinic communities in their collective efforts for the discovery, investigation, application, monitoring and management of targeted therapeutics.

FUNDING

Singapore Academic Research Fund [R-148-000-181-112]; National Natural Science Foundation of China [81202459]; Chongqing Natural Science Foundation [cstc2012jjA10116]; Fundamental Research Funds for the Central Universities [CQDXWL-2012-Z003]; Start up founding of Youth 100 Talents program of Chongqing University [0903005203176]; The Major State Basic Research Development Program of China [2013CB967204]. Funding for open access charge: National Natural Science Foundation of China [81202459].

Conflict of interest statement. None declared.

REFERENCES

  • 1.Zheng CJ, Han LY, Yap CW, Ji ZL, Cao ZW, Chen YZ. Therapeutic targets: progress of their exploration and investigation of their characteristics. Pharmacol. Rev. 2006;58:259–279. doi: 10.1124/pr.58.2.4. [DOI] [PubMed] [Google Scholar]
  • 2.Overington JP, Al-Lazikani B, Hopkins AL. How many drug targets are there? Nat. Rev. Drug Discov. 2006;5:993–996. doi: 10.1038/nrd2199. [DOI] [PubMed] [Google Scholar]
  • 3.Rask-Andersen M, Almen MS, Schioth HB. Trends in the exploitation of novel drug targets. Nat. Rev. Drug Discov. 2011;10:579–590. doi: 10.1038/nrd3478. [DOI] [PubMed] [Google Scholar]
  • 4.La Thangue NB, Kerr DJ. Predictive biomarkers: a paradigm shift towards personalized cancer medicine. Nat. Rev. Clin. Oncol. 2011;8:587–596. doi: 10.1038/nrclinonc.2011.121. [DOI] [PubMed] [Google Scholar]
  • 5.Trusheim MR, Burgess B, Hu SX, Long T, Averbuch SD, Flynn AA, Lieftucht A, Mazumder A, Milloy J, Shaw PM, et al. Quantifying factors for the success of stratified medicine. Nat. Rev. Drug Discov. 2011;10:817–833. doi: 10.1038/nrd3557. [DOI] [PubMed] [Google Scholar]
  • 6.Volzke H, Schmidt CO, Baumeister SE, Ittermann T, Fung G, Krafczyk-Korth J, Hoffmann W, Schwab M, Meyer zu Schwabedissen HE, Dorr M, et al. Personalized cardiovascular medicine: concepts and methodological considerations. Nat. Rev. Cardiol. 2013;10:308–316. doi: 10.1038/nrcardio.2013.35. [DOI] [PubMed] [Google Scholar]
  • 7.Kneller R. The importance of new companies for drug discovery: origins of a decade of new drugs. Nat. Rev. Drug Discov. 2010;9:867–882. doi: 10.1038/nrd3251. [DOI] [PubMed] [Google Scholar]
  • 8.Bunnage ME. Getting pharmaceutical R&D back on target. Nat. Chem. Biol. 2011;7:335–339. doi: 10.1038/nchembio.581. [DOI] [PubMed] [Google Scholar]
  • 9.Chataway J, Fry C, Marjanovic S, Yaqub O. Public-private collaborations and partnerships in stratified medicine: making sense of new interactions. N. Biotechnol. 2012;29:732–740. doi: 10.1016/j.nbt.2012.03.006. [DOI] [PubMed] [Google Scholar]
  • 10.Maliepaard M, Nofziger C, Papaluca M, Zineh I, Uyama Y, Prasad K, Grimstein C, Pacanowski M, Ehmann F, Dossena S, et al. Pharmacogenetics in the evaluation of new drugs: a multiregional regulatory perspective. Nat. Rev. Drug Discov. 2013;12:103–115. doi: 10.1038/nrd3931. [DOI] [PubMed] [Google Scholar]
  • 11.Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–1043. doi: 10.1126/science.1141478. [DOI] [PubMed] [Google Scholar]
  • 12.Chen Z, Cheng K, Walton Z, Wang Y, Ebi H, Shimamura T, Liu Y, Tupper T, Ouyang J, Li J, et al. A murine lung cancer co-clinical trial identifies genetic modifiers of therapeutic response. Nature. 2012;483:613–617. doi: 10.1038/nature10937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hall IP. Stratified medicine: drugs meet genetics. Eur. Respir. Rev. 2013;22:53–57. doi: 10.1183/09059180.00008312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Fugel HJ, Nuijten M, Postma M. Stratified medicine and reimbursement issues. Front Pharmacol. 2012;3:181. doi: 10.3389/fphar.2012.00181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Knox C, Law V, Jewison T, Liu P, Ly S, Frolkis A, Pon A, Banco K, Mak C, Neveu V, et al. DrugBank 3.0: a comprehensive resource for ‘omics' research on drugs. Nucleic Acids Res. 2011;39:D1035–D1041. doi: 10.1093/nar/gkq1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhu F, Shi Z, Qin C, Tao L, Liu X, Xu F, Zhang L, Song Y, Zhang J, Han B, et al. Therapeutic target database update 2012: a resource for facilitating target-oriented drug discovery. Nucleic Acids Res. 2012;40:D1128–D1136. doi: 10.1093/nar/gkr797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yang IS, Ryu C, Cho KJ, Kim JK, Ong SH, Mitchell WP, Kim BS, Oh HB, Kim KH. IDBD: infectious disease biomarker database. Nucleic Acids Res. 2008;36:D455–D460. doi: 10.1093/nar/gkm925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Srivastava S. The early detection research network: 10-year outlook. Clin. Chem. 2013;59:60–67. doi: 10.1373/clinchem.2012.184697. [DOI] [PubMed] [Google Scholar]
  • 19.Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H, Forbes S, Bindal N, Beare D, Smith JA, Thompson IR, et al. Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955–D961. doi: 10.1093/nar/gks1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bramer GR. International statistical classification of diseases and related health problems. Tenth revision. World Health Stat. Q. 1988;41:32–36. [PubMed] [Google Scholar]
  • 21.Wood PH. Applications of the International Classification of Diseases. World Health Stat. Q. 1990;43:263–268. [PubMed] [Google Scholar]
  • 22. (1986) ICD-9-CM. International Classification of Diseases, 9th edn, Clinical Modification: update. Official authorized addendum, effective 1 October 1986. J. Am. Med. Rec. Assoc., 57, S1–S32. [PubMed]
  • 23.Steindel SJ. International classification of diseases, 10th edn, clinical modification and procedure coding system: descriptive overview of the next generation HIPAA code sets. J. Am. Med. Inform. Assoc. 2010;17:274–282. doi: 10.1136/jamia.2009.001230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lim CM, Aryani Md Yusof F, Selvarajah S, Lim TO. Use of ATC to describe duplicate medications in primary care prescriptions. Eur. J. Clin. Pharmacol. 2011;67:1035–1044. doi: 10.1007/s00228-011-1025-4. [DOI] [PubMed] [Google Scholar]
  • 25.Averill R, Bowman S. There are critical reasons for not further delaying the implementation of the new ICD-10 coding system. J. AHIMA. 2012;83:42–48. [PubMed] [Google Scholar]
  • 26.Topaz M, Shafran-Topaz L, Bowles KH. ICD-9 to ICD-10: evolution, revolution, and current debates in the United States. Perspect Health Inf. Manag. 2013;10:1d. [PMC free article] [PubMed] [Google Scholar]
  • 27.Califano R, Abidin AZ, Peck R, Faivre-Finn C, Lorigan P. Management of small cell lung cancer: recent developments for optimal care. Drugs. 2012;72:471–490. doi: 10.2165/11597640-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 28.Robinson WH, Lindstrom TM, Cheung RK, Sokolove J. Mechanistic biomarkers for clinical decision making in rheumatic diseases. Nat. Rev. Rheumatol. 2013;9:267–276. doi: 10.1038/nrrheum.2013.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ludwig JA, Weinstein JN. Biomarkers in cancer staging, prognosis and treatment selection. Nat. Rev. Cancer. 2005;5:845–856. doi: 10.1038/nrc1739. [DOI] [PubMed] [Google Scholar]
  • 30.Walsh P, Elsabbagh M, Bolton P, Singh I. In search of biomarkers for autism: scientific, social and ethical challenges. Nat. Rev. Neurosci. 2011;12:603–612. doi: 10.1038/nrn3113. [DOI] [PubMed] [Google Scholar]
  • 31.Ho C, Laskin J. EGFR-directed therapies to treat non-small-cell lung cancer. Expert. Opin. Investig. Drugs. 2009;18:1133–1145. doi: 10.1517/13543780903066772. [DOI] [PubMed] [Google Scholar]
  • 32.Linardou H, Dahabreh IJ, Bafaloukos D, Kosmidis P, Murray S. Somatic EGFR mutations and efficacy of tyrosine kinase inhibitors in NSCLC. Nat. Rev. Clin. Oncol. 2009;6:352–366. doi: 10.1038/nrclinonc.2009.62. [DOI] [PubMed] [Google Scholar]
  • 33.Blennow K, Hampel H, Zetterberg H. Biomarkers in amyloid-beta immunotherapy trials in alzheimer's disease. Neuropsychopharmacology. 2013;39:189–201. doi: 10.1038/npp.2013.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ortiz A, Massy ZA, Fliser D, Lindholm B, Wiecek A, Martinez-Castelao A, Covic A, Goldsmith D, Suleymanlar G, London GM, et al. Clinical usefulness of novel prognostic biomarkers in patients on hemodialysis. Nat. Rev. Nephrol. 2012;8:141–150. doi: 10.1038/nrneph.2011.170. [DOI] [PubMed] [Google Scholar]
  • 35.Ahmad T, Fiuzat M, Felker GM, O'Connor C. Novel biomarkers in chronic heart failure. Nat. Rev. Cardiol. 2012;9:347–359. doi: 10.1038/nrcardio.2012.37. [DOI] [PubMed] [Google Scholar]
  • 36.NCBI Resource Coordinators. Database resources of the National Center for Biotechnology Information. Nucleic Acids Res. 2013;41:D8–D20. doi: 10.1093/nar/gks1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cools J, Mentens N, Furet P, Fabbro D, Clark JJ, Griffin JD, Marynen P, Gilliland DG. Prediction of resistance to small molecule FLT3 inhibitors: implications for molecularly targeted therapy of acute leukemia. Cancer Res. 2004;64:6385–6389. doi: 10.1158/0008-5472.CAN-04-2148. [DOI] [PubMed] [Google Scholar]
  • 38.Rizvi NA, Rusch V, Pao W, Chaft JE, Ladanyi M, Miller VA, Krug LM, Azzoli CG, Bains M, Downey R, et al. Molecular characteristics predict clinical outcomes: prospective trial correlating response to the EGFR tyrosine kinase inhibitor gefitinib with the presence of sensitizing mutations in the tyrosine binding domain of the EGFR gene. Clin. Cancer Res. 2011;17:3500–3506. doi: 10.1158/1078-0432.CCR-10-2102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Molinari F, Felicioni L, Buscarino M, De Dosso S, Buttitta F, Malatesta S, Movilia A, Luoni M, Boldorini R, Alabiso O, et al. Increased detection sensitivity for KRAS mutations enhances the prediction of anti-EGFR monoclonal antibody resistance in metastatic colorectal cancer. Clin. Cancer Res. 2011;17:4901–4914. doi: 10.1158/1078-0432.CCR-10-3137. [DOI] [PubMed] [Google Scholar]
  • 40.Wang W, Cassidy J, O'Brien V, Ryan KM, Collie-Duguid E. Mechanistic and predictive profiling of 5-Fluorouracil resistance in human cancer cells. Cancer Res. 2004;64:8167–8176. doi: 10.1158/0008-5472.CAN-04-0970. [DOI] [PubMed] [Google Scholar]
  • 41.Dai Z, Barbacioru C, Huang Y, Sadee W. Prediction of anticancer drug potency from expression of genes involved in growth factor signaling. Pharm. Res. 2006;23:336–349. doi: 10.1007/s11095-005-9260-y. [DOI] [PubMed] [Google Scholar]
  • 42.Brase JC, Schmidt M, Fischbach T, Sultmann H, Bojar H, Koelbl H, Hellwig B, Rahnenfuhrer J, Hengstler JG, Gehrmann MC. ERBB2 and TOP2A in breast cancer: a comprehensive analysis of gene amplification, RNA levels, and protein expression and their influence on prognosis and prediction. Clin. Cancer Res. 2010;16:2391–2401. doi: 10.1158/1078-0432.CCR-09-2471. [DOI] [PubMed] [Google Scholar]
  • 43.Zhang J, Jia J, Zhu F, Ma X, Han B, Wei X, Tan C, Jiang Y, Chen Y. Analysis of bypass signaling in EGFR pathway and profiling of bypass genes for predicting response to anticancer EGFR tyrosine kinase inhibitors. Mol. Biosyst. 2012;8:2645–2656. doi: 10.1039/c2mb25165e. [DOI] [PubMed] [Google Scholar]
  • 44.Hilsenbeck SG, Friedrichs WE, Schiff R, O'Connell P, Hansen RK, Osborne CK, Fuqua SA. Statistical analysis of array expression data as applied to the problem of tamoxifen resistance. J. Natl Cancer Inst. 1999;91:453–459. doi: 10.1093/jnci/91.5.453. [DOI] [PubMed] [Google Scholar]
  • 45.Staunton JE, Slonim DK, Coller HA, Tamayo P, Angelo MJ, Park J, Scherf U, Lee JK, Reinhold WO, Weinstein JN, et al. Chemosensitivity prediction by transcriptional profiling. Proc. Natl Acad. Sci. USA. 2001;98:10787–10792. doi: 10.1073/pnas.191368598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, Gascoyne RD, Muller-Hermelink HK, Smeland EB, Giltnane JM, et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N. Engl. J. Med. 2002;346:1937–1947. doi: 10.1056/NEJMoa012914. [DOI] [PubMed] [Google Scholar]
  • 47.Takata R, Katagiri T, Kanehira M, Tsunoda T, Shuin T, Miki T, Namiki M, Kohri K, Matsushita Y, Fujioka T, et al. Predicting response to methotrexate, vinblastine, doxorubicin, and cisplatin neoadjuvant chemotherapy for bladder cancers through genome-wide gene expression profiling. Clin. Cancer Res. 2005;11:2625–2636. doi: 10.1158/1078-0432.CCR-04-1988. [DOI] [PubMed] [Google Scholar]
  • 48.Balko JM, Potti A, Saunders C, Stromberg A, Haura EB, Black EP. Gene expression patterns that predict sensitivity to epidermal growth factor receptor tyrosine kinase inhibitors in lung cancer cell lines and human lung tumors. BMC Genomics. 2006;7:289. doi: 10.1186/1471-2164-7-289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Okano T, Kondo T, Fujii K, Nishimura T, Takano T, Ohe Y, Tsuta K, Matsuno Y, Gemma A, Kato H, et al. Proteomic signature corresponding to the response to gefitinib (Iressa, ZD1839), an epidermal growth factor receptor tyrosine kinase inhibitor in lung adenocarcinoma. Clin. Cancer Res. 2007;13:799–805. doi: 10.1158/1078-0432.CCR-06-1654. [DOI] [PubMed] [Google Scholar]
  • 50.Ma Y, Ding Z, Qian Y, Shi X, Castranova V, Harner EJ, Guo L. Predicting cancer drug response by proteomic profiling. Clin. Cancer Res. 2006;12:4583–4589. doi: 10.1158/1078-0432.CCR-06-0290. [DOI] [PubMed] [Google Scholar]
  • 51.Hauschild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, Millward M, Rutkowski P, Blank CU, Miller WH, Jr, Kaempgen E, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358–365. doi: 10.1016/S0140-6736(12)60868-X. [DOI] [PubMed] [Google Scholar]
  • 52.Cichowski K, Janne PA. Drug discovery: inhibitors that activate. Nature. 2010;464:358–359. doi: 10.1038/464358a. [DOI] [PubMed] [Google Scholar]
  • 53.Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N, Sazegar H, et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010;468:973–977. doi: 10.1038/nature09626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Li QL, Gu FM, Wang Z, Jiang JH, Yao LQ, Tan CJ, Huang XY, Ke AW, Dai Z, Fan J, et al. Activation of PI3K/AKT and MAPK pathway through a PDGFRbeta-dependent feedback loop is involved in rapamycin resistance in hepatocellular carcinoma. PLoS One. 2012;7:e33379. doi: 10.1371/journal.pone.0033379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bemis GW, Murcko MA. The properties of known drugs. 1. Molecular frameworks. J. Med. Chem. 1996;39:2887–2893. doi: 10.1021/jm9602928. [DOI] [PubMed] [Google Scholar]
  • 56.Wang J, Hou T. Drug and drug candidate building block analysis. J. Chem. Inf. Model. 2010;50:55–67. doi: 10.1021/ci900398f. [DOI] [PubMed] [Google Scholar]
  • 57.Duarte CD, Barreiro EJ, Fraga CA. Privileged structures: a useful concept for the rational design of new lead drug candidates. Mini Rev. Med. Chem. 2007;7:1108–1119. doi: 10.2174/138955707782331722. [DOI] [PubMed] [Google Scholar]
  • 58.Koch MA, Schuffenhauer A, Scheck M, Wetzel S, Casaulta M, Odermatt A, Ertl P, Waldmann H. Charting biologically relevant chemical space: a structural classification of natural products (SCONP) Proc. Natl Acad. Sci. USA. 2005;102:17272–17277. doi: 10.1073/pnas.0503647102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kong DX, Jiang YY, Zhang HY. Marine natural products as sources of novel scaffolds: achievement and concern. Drug Discov. Today. 2010;15:884–886. doi: 10.1016/j.drudis.2010.09.002. [DOI] [PubMed] [Google Scholar]
  • 60.Mallinson J, Collins I. Macrocycles in new drug discovery. Future Med. Chem. 2012;4:1409–1438. doi: 10.4155/fmc.12.93. [DOI] [PubMed] [Google Scholar]
  • 61.Newman DJ, Cragg GM. Natural products as sources of new drugs over the last 25 years. J. Nat. Prod. 2007;70:461–477. doi: 10.1021/np068054v. [DOI] [PubMed] [Google Scholar]
  • 62.Butler MS. Natural products to drugs: natural product-derived compounds in clinical trials. Nat. Prod. Rep. 2008;25:475–516. doi: 10.1039/b514294f. [DOI] [PubMed] [Google Scholar]
  • 63.Zhu F, Qin C, Tao L, Liu X, Shi Z, Ma X, Jia J, Tan Y, Cui C, Lin J, et al. Clustered patterns of species origins of nature-derived drugs and clues for future bioprospecting. Proc. Natl Acad. Sci. USA. 2011;108:12943–12948. doi: 10.1073/pnas.1107336108. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Nucleic Acids Research are provided here courtesy of Oxford University Press

RESOURCES