microarray.bib

@comment{{This file has been generated by bib2bib 1.97}}
@comment{{Command line: bib2bib ../bibli.bib -c 'subject:"microarray" or keywords:"microarray"' -ob tmp.bib}}
@inproceedings{Aliferis2002Machine,
  author = {Aliferis, C.F. and Hardin, D.P. and Massion, P.},
  title = {Machine {L}earning {M}odels {F}or {L}ung {C}ancer {C}lassification
	{U}sing {A}rray {C}omparative {G}enomic {H}ybridization},
  booktitle = {Proceedings of the 2002 {A}merican {M}edical {I}nformatics {A}ssociation
	({AMIA}) {A}nnual {S}ymposium},
  year = {2002},
  pages = {7-11},
  abstract = {Array {CGH} is a recently introduced technology that measures changes
	in the gene copy number of hundreds of genes in a single experiment.
	{T}he primary goal of this study was to develop machine learning
	models that classify non-small {L}ung {C}ancers according to histopathology
	types and to compare several machine learning methods in this learning
	task. {DNA} from tumors of 37 patients (21 squamous carcinomas, and
	16 adenocarcinomas) were extracted and hybridized onto a 452 {BAC}
	clone array. {T}he following algorithms were used: {KNN}, {D}ecision
	{T}ree {I}nduction, {S}upport {V}ector {M}achines and {F}eed-{F}orward
	{N}eural {N}etworks. {P}erformance was measured via leave-one-out
	classification accuracy. {T}he best multi-gene model found had a
	leave-one-out accuracy of 89.2\%. {D}ecision {T}rees performed poorer
	than the other methods in this learning task and dataset. {W}e conclude
	that gene copy numbers as measured by array {CGH} are, collectively,
	an excellent indicator of histological subtype. {S}everal interesting
	research directions are discussed.},
  pdf = {../local/Aliferis2002Machine.pdf},
  file = {Aliferis2002Machine.pdf:local/Aliferis2002Machine.pdf:PDF},
  keywords = {biosvm microarray, cgh},
  owner = {jeanphilippevert}
}
@article{Bao2002Identifying,
  author = {Bao, L. and Sun, Z.},
  title = {Identifying genes related to drug anticancer mechanisms using support
	vector machine},
  journal = {F{EBS} {L}ett.},
  year = {2002},
  volume = {521},
  pages = {109--114},
  abstract = {In an effort to identify genes related to the cell line chemosensitivity
	and to evaluate the functional relationships between genes and anticancer
	drugs acting by the same mechanism, a supervised machine learning
	approach called support vector machine was used to label genes into
	any of the five predefined anticancer drug mechanistic categories.
	{A}mong dozens of unequivocally categorized genes, many were known
	to be causally related to the drug mechanisms. {F}or example, a few
	genes were found to be involved in the biological process triggered
	by the drugs (e.g. {DNA} polymerase epsilon was the direct target
	for the drugs from {DNA} antimetabolites category). {DNA} repair-related
	genes were found to be enriched for about eight-fold in the resulting
	gene set relative to the entire gene set. {S}ome uncharacterized
	transcripts might be of interest in future studies. {T}his method
	of correlating the drugs and genes provides a strategy for finding
	novel biologically significant relationships for molecular pharmacology.},
  pdf = {../local/bao02.pdf},
  file = {bao02.pdf:local/bao02.pdf:PDF},
  keywords = {biosvm microarray},
  subject = {biokernel},
  url = {http://www.elsevier.com/febs/402/19/42/article.html}
}
@article{Ben-Dor2000Tissue,
  author = {Ben-Dor, A. and Bruhn, L. and Friedman, N. and Nachman, I. and Schummer,
	M. and Yakhini, Z.},
  title = {Tissue Classification with Gene Expression Profiles},
  journal = {J. Comput. Biol.},
  year = {2000},
  volume = {7},
  pages = {559-583},
  number = {3-4},
  abstract = {Constantly improving gene expression profiling technologies are expected
	to provide understanding and insight into cancer-related cellular
	processes. {G}ene expression data is also expected to significantly
	aid in the development of efficient cancer diagnosis and classification
	platforms. {I}n this work we examine three sets of gene expression
	data measured across sets of tumor(s) and normal clinical samples:
	{T}he first set consists of 2,000 genes, measured in 62 epithelial
	colon samples ({A}lon et al., 1999). {T}he second consists of approximately
	equal to 100,000 clones, measured in 32 ovarian samples (unpublished
	extension of data set described in {S}chummer et al. (1999)). {T}he
	third set consists of approximately equal to 7,100 genes, measured
	in 72 bone marrow and peripheral blood samples ({G}olub et al, 1999).
	{W}e examine the use of scoring methods, measuring separation of
	tissue type (e.g., tumors from normals) using individual gene expression
	levels. {T}hese are then coupled with high-dimensional classification
	methods to assess the classification power of complete expression
	profiles. {W}e present results of performing leave-one-out cross
	validation ({LOOCV}) experiments on the three data sets, employing
	nearest neighbor classifier, {SVM} ({C}ortes and {V}apnik, 1995),
	{A}da{B}oost ({F}reund and {S}chapire, 1997) and a novel clustering-based
	classification technique. {A}s tumor samples can differ from normal
	samples in their cell-type composition, we also perform {LOOCV} experiments
	using appropriately modified sets of genes, attempting to eliminate
	the resulting bias. {W}e demonstrate success rate of at least 90%
	in tumor versus normal classification, using sets of selected genes,
	with, as well as without, cellular-contamination-related members.
	{T}hese results are insensitive to the exact selection mechanism,
	over a certain range.},
  pdf = {../local/Ben-Dor2000Tissue.pdf},
  file = {Ben-Dor2000Tissue.pdf:local/Ben-Dor2000Tissue.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {jeanphilippevert},
  url = {http://www.liebertonline.com/doi/abs/10.1089/106652700750050943}
}
@article{Bozdech2004Antioxidant,
  author = {Bozdech, Z. and Ginsburg, H.},
  title = {Antioxidant defense in {P}lasmodium falciparum - data mining of the
	transcriptome},
  journal = {Malaria {J}ournal},
  year = {2004},
  volume = {3},
  pages = {23},
  number = {1},
  abstract = {The intraerythrocytic malaria parasite is under constant oxidative
	stress originating both from endogenous and exogenous processes.
	{T}he parasite is endowed with a complete network of enzymes and
	proteins that protect it from those threats, but also uses redox
	activities to regulate enzyme activities. {I}n the present analysis,
	the transcription of the genes coding for the antioxidant defense
	elements are viewed in the time-frame of the intraerythrocytic cycle.
	{T}ime-dependent transcription data were taken from the transcriptome
	of the human malaria parasite {P}lasmodium falciparum. {W}hereas
	for several processes the transcription of the many participating
	genes is coordinated, in the present case there are some outstanding
	deviations where gene products that utilize glutathione or thioredoxin
	are transcribed before the genes coding for elements that control
	the levels of those substrates are transcribed. {S}uch insights may
	hint to novel, non-classical pathways that necessitate further investigations.},
  doi = {10.1186/1475-2875-3-23},
  pdf = {../local/Bozdech2004Antioxidant.pdf},
  file = {Bozdech2004Antioxidant.pdf:local/Bozdech2004Antioxidant.pdf:PDF},
  keywords = {microarray plasmodium},
  owner = {vert},
  url = {http://www.malariajournal.com/content/3/1/23}
}
@article{Bozdech2003Transcriptome,
  author = {Bozdech, Z. and Llinas, M. and Pulliam, B. L. and Wong, E. D. and
	Zhu, J. and DeRisi, J. L.},
  title = {The {T}ranscriptome of the {I}ntraerythrocytic {D}evelopmental {C}ycle
	of {P}lasmodium falciparum },
  journal = {P{L}o{S} {B}iology},
  year = {2003},
  volume = {1},
  pages = {e5},
  number = {1},
  abstract = {Plasmodium falciparum is the causative agent of the most burdensome
	form of human malaria, affecting 200-300 million individuals per
	year worldwide. {T}he recently sequenced genome of {P}. falciparum
	revealed over 5,400 genes, of which 60{percnt} encode proteins of
	unknown function. {I}nsights into the biochemical function and regulation
	of these genes will provide the foundation for future drug and vaccine
	development efforts toward eradication of this disease. {B}y analyzing
	the complete asexual intraerythrocytic developmental cycle ({IDC})
	transcriptome of the {HB}3 strain of {P}. falciparum, we demonstrate
	that at least 60{percnt} of the genome is transcriptionally active
	during this stage. {O}ur data demonstrate that this parasite has
	evolved an extremely specialized mode of transcriptional regulation
	that produces a continuous cascade of gene expression, beginning
	with genes corresponding to general cellular processes, such as protein
	synthesis, and ending with {P}lasmodium-specific functionalities,
	such as genes involved in erythrocyte invasion. {T}he data reveal
	that genes contiguous along the chromosomes are rarely coregulated,
	while transcription from the plastid genome is highly coregulated
	and likely polycistronic. {C}omparative genomic hybridization between
	{HB}3 and the reference genome strain (3{D}7) was used to distinguish
	between genes not expressed during the {IDC} and genes not detected
	because of possible sequence variations. {G}enomic differences between
	these strains were found almost exclusively in the highly antigenic
	subtelomeric regions of chromosomes. {T}he simple cascade of gene
	regulation that directs the asexual development of {P}. falciparum
	is unprecedented in eukaryotic biology. {T}he transcriptome of the
	{IDC} resembles a "just-in-time" manufacturing process whereby induction
	of any given gene occurs once per cycle and only at a time when it
	is required. {T}hese data provide to our knowledge the first comprehensive
	view of the timing of transcription throughout the intraerythrocytic
	development of {P}. falciparum and provide a resource for the identification
	of new chemotherapeutic and vaccine candidates.},
  comment = {(JP Vert) The paper that monitors the 48h cell cycle of P. falciparum},
  doi = {10.1371/journal.pbio.0000005},
  pdf = {../local/Bozdech2003Transcriptome.pdf},
  file = {Bozdech2003Transcriptome.pdf:local/Bozdech2003Transcriptome.pdf:PDF},
  keywords = {microarray plasmodium},
  owner = {vert},
  url = {http://dx.doi.org/10.1371/journal.pbio.0000005 }
}
@article{Bozdech2003Expression,
  author = {Bozdech, Z. and Zhu, J. and Joachimiak, M. and Cohen, F. and Pulliam,
	B. and DeRisi, J.},
  title = {Expression profiling of the schizont and trophozoite stages of {P}lasmodium
	falciparum with a long-oligonucleotide microarray},
  journal = {Genome {B}iology},
  year = {2003},
  volume = {4},
  pages = {R9},
  number = {2},
  abstract = {B{ACKGROUND}:{T}he worldwide persistence of drug-resistant {P}lasmodium
	falciparum, the most lethal variety of human malaria, is a global
	health concern. {T}he {P}. falciparum sequencing project has brought
	new opportunities for identifying molecular targets for antimalarial
	drug and vaccine development.{RESULTS}:{W}e developed a software
	package, {A}rray{O}ligo{S}elector, to design an open reading frame
	({ORF})-specific {DNA} microarray using the publicly available {P}.
	falciparum genome sequence. {E}ach gene was represented by one or
	more long 70 mer oligonucleotides selected on the basis of uniqueness
	within the genome, exclusion of low-complexity sequence, balanced
	base composition and proximity to the 3' end. {A} first-generation
	microarray representing approximately 6,000 {ORF}s of the {P}. falciparum
	genome was constructed. {A}rray performance was evaluated through
	the use of control oligonucleotide sets with increasing levels of
	introduced mutations, as well as traditional northern blotting. {U}sing
	this array, we extensively characterized the gene-expression profile
	of the intraerythrocytic trophozoite and schizont stages of {P}.
	falciparum. {T}he results revealed extensive transcriptional regulation
	of genes specialized for processes specific to these two stages.{CONCLUSIONS}:{DNA}
	microarrays based on long oligonucleotides are powerful tools for
	the functional annotation and exploration of the {P}. falciparum
	genome. {E}xpression profiling of trophozoites and schizonts revealed
	genes associated with stage-specific processes and may serve as the
	basis for future drug targets and vaccine development.},
  doi = {10.1186/gb-2003-4-2-r9},
  pdf = {../local/Bozdech2003Expression.pdf},
  file = {Bozdech2003Expression.pdf:local/Bozdech2003Expression.pdf:PDF},
  keywords = {microarray plasmodium},
  owner = {vert},
  url = {http://genomebiology.com/2003/4/2/R9}
}
@article{Brown2000Knowledge-based,
  author = {Brown, M. P. and Grundy, W. N. and Lin, D. and Cristianini, N. and
	Sugnet, C. W. and Furey, T. S. and Ares, M. and Haussler, D.},
  title = {Knowledge-based analysis of microarray gene expression data by using
	support vector machines.},
  journal = {Proc. {N}atl. {A}cad. {S}ci. {USA}},
  year = {2000},
  volume = {97},
  pages = {262-7},
  number = {1},
  month = {Jan},
  abstract = {We introduce a method of functionally classifying genes by using gene
	expression data from {DNA} microarray hybridization experiments.
	{T}he method is based on the theory of support vector machines ({SVM}s).
	{SVM}s are considered a supervised computer learning method because
	they exploit prior knowledge of gene function to identify unknown
	genes of similar function from expression data. {SVM}s avoid several
	problems associated with unsupervised clustering methods, such as
	hierarchical clustering and self-organizing maps. {SVM}s have many
	mathematical features that make them attractive for gene expression
	analysis, including their flexibility in choosing a similarity function,
	sparseness of solution when dealing with large data sets, the ability
	to handle large feature spaces, and the ability to identify outliers.
	{W}e test several {SVM}s that use different similarity metrics, as
	well as some other supervised learning methods, and find that the
	{SVM}s best identify sets of genes with a common function using expression
	data. {F}inally, we use {SVM}s to predict functional roles for uncharacterized
	yeast {ORF}s based on their expression data.},
  pdf = {../local/Brown2000Knowledge-based.pdf},
  file = {Brown2000Knowledge-based.pdf:local/Brown2000Knowledge-based.pdf:PDF},
  keywords = {biosvm microarray},
  url = {http://www.pnas.org/cgi/content/abstract/97/1/262}
}
@article{Brown2000Exploring,
  author = {P.O. Brown and D. Botstein},
  title = {Exploring the new world of the genome with {DNA} microarrays},
  journal = {Nat. {G}enet.},
  year = {2000},
  volume = {21},
  pages = {33--37},
  pdf = {../local/brow00b.pdf},
  file = {brow00b.pdf:local/brow00b.pdf:PDF},
  subject = {microarray},
  url = {http://www.nature.com/ng/journal/v21/n1s/abs/ng0199supp_33.html}
}
@article{Burckin2005Exploring,
  author = {Burckin, T. and Nagel, R. and Mandel-Gutfreund, Y. and Shiue, L.
	and Clark, T. A. and Chong, J.-L. and Chang, T.-H. and Squazzo, S.
	and Hartzog, G. and Ares, M.},
  title = {Exploring functional relationships between components of the gene
	expression machinery.},
  journal = {Nat. {S}truct. {M}ol. {B}iol.},
  year = {2005},
  volume = {12},
  pages = {175-82},
  number = {2},
  month = {Feb},
  abstract = {Eukaryotic gene expression requires the coordinated activity of many
	macromolecular machines including transcription factors and {RNA}
	polymerase, the spliceosome, m{RNA} export factors, the nuclear pore,
	the ribosome and decay machineries. {Y}east carrying mutations in
	genes encoding components of these machineries were examined using
	microarrays to measure changes in both pre-m{RNA} and m{RNA} levels.
	{W}e used these measurements as a quantitative phenotype to ask how
	steps in the gene expression pathway are functionally connected.
	{A} multiclass support vector machine was trained to recognize the
	gene expression phenotypes caused by these mutations. {I}n several
	cases, unexpected phenotype assignments by the computer revealed
	functional roles for specific factors at multiple steps in the gene
	expression pathway. {T}he ability to resolve gene expression pathway
	phenotypes provides insight into how the major machineries of gene
	expression communicate with each other.},
  doi = {10.1038/nsmb891},
  pdf = {../local/Burckin2005Exploring.pdf},
  file = {Burckin2005Exploring.pdf:local/Burckin2005Exploring.pdf:PDF},
  keywords = {biosvm microarray},
  pii = {nsmb891},
  url = {http://dx.doi.org/10.1038/nsmb891}
}
@article{Bussemaker2001Regulatory,
  author = {Bussemaker, H. J. and Li, H. and Siggia, E. D.},
  title = {Regulatory element detection using correlation with expression},
  journal = {Nat. {G}enet.},
  year = {2001},
  volume = {27},
  pages = {167--174},
  pdf = {../local/buss01.pdf},
  file = {buss01.pdf:local/buss01.pdf:PDF},
  subject = {microarray},
  url = {http://www.nature.com/cgi-taf/DynaPage.taf?file=/ng/journal/v27/n2/full/ng0201_167.html&filetype=pdf}
}
@article{Chen2011Removing,
  author = {Chao Chen and Kay Grennan and Judith Badner and Dandan Zhang and
	Elliot Gershon and Li Jin and Chunyu Liu},
  title = {Removing batch effects in analysis of expression microarray data:
	an evaluation of six batch adjustment methods.},
  journal = {PLoS One},
  year = {2011},
  volume = {6},
  pages = {e17238},
  number = {2},
  abstract = {The expression microarray is a frequently used approach to study gene
	expression on a genome-wide scale. However, the data produced by
	the thousands of microarray studies published annually are confounded
	by "batch effects," the systematic error introduced when samples
	are processed in multiple batches. Although batch effects can be
	reduced by careful experimental design, they cannot be eliminated
	unless the whole study is done in a single batch. A number of programs
	are now available to adjust microarray data for batch effects prior
	to analysis. We systematically evaluated six of these programs using
	multiple measures of precision, accuracy and overall performance.
	ComBat, an Empirical Bayes method, outperformed the other five programs
	by most metrics. We also showed that it is essential to standardize
	expression data at the probe level when testing for correlation of
	expression profiles, due to a sizeable probe effect in microarray
	data that can inflate the correlation among replicates and unrelated
	samples.},
  doi = {10.1371/journal.pone.0017238},
  institution = {National Ministry of Education Key Laboratory of Contemporary Anthropology,
	Fudan University, Shanghai, People's Republic of China.},
  keywords = {Bayes Theorem; Case-Control Studies; Data Interpretation, Statistical;
	Gene Expression Profiling, standards/statistics /&/ numerical data;
	Humans; Microarray Analysis, standards/statistics /&/ numerical data;
	ROC Curve; Reference Standards; Research Design; Sample Size; Selection
	Bias; Validation Studies as Topic},
  language = {eng},
  medline-pst = {epublish},
  owner = {jp},
  pmid = {21386892},
  timestamp = {2012.02.29},
  url = {http://dx.doi.org/10.1371/journal.pone.0017238}
}
@article{Chiang2001Visualizing,
  author = {Chiang, D. Y. and Brown, P. O. and Eisen, M. B.},
  title = {Visualizing associations between genome sequences and gene expression
	data using genome-mean expression profiles},
  journal = {Bioinformatics},
  year = {2001},
  volume = {17},
  pages = {49S--55S},
  pdf = {../local/chia01.pdf},
  file = {chia01.pdf:local/chia01.pdf:PDF},
  subject = {microarray},
  url = {http://bioinformatics.oupjournals.org/cgi/reprint/17/suppl_1/S49.pdf}
}
@article{Chu1998Transcriptional,
  author = {S. Chu and J. DeRisi and M. Eisen and J. Mulholland and D. Botstein
	and P.O. Brown and I. Herskowitz},
  title = {The {T}ranscriptional {P}rogram of {S}porulation in {B}udding {Y}east},
  journal = {Science},
  year = {1998},
  volume = {282},
  pages = {699--705},
  pdf = {../local/chu98.pdf},
  file = {chu98.pdf:local/chu98.pdf:PDF},
  owner = {phupe},
  subject = {microarray},
  timestamp = {2009.10.15},
  url = {http://www.sciencemag.org/cgi/reprint/282/5389/699.pdf}
}
@article{DeRisi1997Exploring,
  author = {DeRisi, J. L. and Iyer, V. R. and Brown, P. O.},
  title = {Exploring the metabolic and genetic control of gene expression on
	a genomic scale},
  journal = {Science},
  year = {1997},
  volume = {278},
  pages = {680--686},
  number = {5338},
  pdf = {../local/deri97.pdf},
  file = {deri97.pdf:local/deri97.pdf:PDF},
  subject = {microarray},
  url = {http://www.sciencemag.org/cgi/reprint/278/5338/680.pdf}
}
@article{Dong2005Fast,
  author = {Jian-xiong Dong and Adam Krzyzak and Ching Y Suen},
  title = {Fast {SVM} training algorithm with decomposition on very large data
	sets.},
  journal = {I{EEE} {T}rans {P}attern {A}nal {M}ach {I}ntell},
  year = {2005},
  volume = {27},
  pages = {603-18},
  number = {4},
  month = {Apr},
  abstract = {Training a support vector machine on a data set of huge size with
	thousands of classes is a challenging problem. {T}his paper proposes
	an efficient algorithm to solve this problem. {T}he key idea is to
	introduce a parallel optimization step to quickly remove most of
	the nonsupport vectors, where block diagonal matrices are used to
	approximate the original kernel matrix so that the original problem
	can be split into hundreds of subproblems which can be solved more
	efficiently. {I}n addition, some effective strategies such as kernel
	caching and efficient computation of kernel matrix are integrated
	to speed up the training process. {O}ur analysis of the proposed
	algorithm shows that its time complexity grows linearly with the
	number of classes and size of the data set. {I}n the experiments,
	many appealing properties of the proposed algorithm have been investigated
	and the results show that the proposed algorithm has a much better
	scaling capability than {L}ibsvm, {SVM}light, and {SVMT}orch. {M}oreover,
	the good generalization performances on several large databases have
	also been achieved.},
  keywords = {Algorithms, Animals, Antibiotics, Antineoplastic, Artificial Intelligence,
	Automated, Automatic Data Processing, Butadienes, Chloroplasts, Comparative
	Study, Computer Simulation, Computer-Assisted, Database Management
	Systems, Databases, Diagnosis, Disinfectants, Dose-Response Relationship,
	Drug, Drug Toxicity, Electrodes, Electroencephalography, Ethylamines,
	Expert Systems, Factual, Feedback, Fungicides, Gene Expression Profiling,
	Genes, Genetic Markers, Humans, Image Enhancement, Image Interpretation,
	Implanted, Industrial, Information Storage and Retrieval, Kidney,
	Kidney Tubules, MEDLINE, Male, Mercuric Chloride, Microarray Analysis,
	Molecular Biology, Motor Cortex, Movement, Natural Language Processing,
	Neural Networks (Computer), Non-P.H.S., Non-U.S. Gov't, Numerical
	Analysis, Pattern Recognition, Plant Proteins, Predictive Value of
	Tests, Proteins, Proteome, Proximal, Puromycin Aminonucleoside, Rats,
	Reproducibility of Results, Research Support, Sensitivity and Specificity,
	Signal Processing, Sprague-Dawley, Subcellular Fractions, Terminology,
	Therapy, Time Factors, Toxicogenetics, U.S. Gov't, User-Computer
	Interface, 15794164}
}
@article{Ein-Dor2005Outcome,
  author = {Ein-Dor, L. and Kela, I. and Getz, G. and Givol, D. and Domany, E.},
  title = {Outcome signature genes in breast cancer: is there a unique set?},
  journal = {Bioinformatics},
  year = {2005},
  volume = {21},
  pages = {171--178},
  number = {2},
  month = {Jan},
  abstract = {MOTIVATION: Predicting the metastatic potential of primary malignant
	tissues has direct bearing on the choice of therapy. Several microarray
	studies yielded gene sets whose expression profiles successfully
	predicted survival. Nevertheless, the overlap between these gene
	sets is almost zero. Such small overlaps were observed also in other
	complex diseases, and the variables that could account for the differences
	had evoked a wide interest. One of the main open questions in this
	context is whether the disparity can be attributed only to trivial
	reasons such as different technologies, different patients and different
	types of analyses. RESULTS: To answer this question, we concentrated
	on a single breast cancer dataset, and analyzed it by a single method,
	the one which was used by van't Veer et al. to produce a set of outcome-predictive
	genes. We showed that, in fact, the resulting set of genes is not
	unique; it is strongly influenced by the subset of patients used
	for gene selection. Many equally predictive lists could have been
	produced from the same analysis. Three main properties of the data
	explain this sensitivity: (1) many genes are correlated with survival;
	(2) the differences between these correlations are small; (3) the
	correlations fluctuate strongly when measured over different subsets
	of patients. A possible biological explanation for these properties
	is discussed. CONTACT: eytan.domany@weizmann.ac.il SUPPLEMENTARY
	INFORMATION: http://www.weizmann.ac.il/physics/complex/compphys/downloads/liate/},
  doi = {10.1093/bioinformatics/bth469},
  pdf = {../local/Ein-Dor2005Outcome.pdf},
  file = {Ein-Dor2005Outcome.pdf:Ein-Dor2005Outcome.pdf:PDF},
  institution = {Department of Physics of Complex Systems, Weizmann Institute of Science
	Rehovot 76100, Israel.},
  keywords = {breastcancer, microarray, featureselection},
  language = {eng},
  medline-pst = {ppublish},
  owner = {jp},
  pii = {bth469},
  pmid = {15308542},
  timestamp = {2010.10.12},
  url = {http://dx.doi.org/10.1093/bioinformatics/bth469}
}
@article{Eisen1998Cluster,
  author = {Eisen, M. B. and Spellman, P. T. and Brown, P. O. and Botstein, D.},
  title = {Cluster analysis and display of genome-wide expression patterns},
  journal = {Proc. Natl. Acad. Sci. USA},
  year = {1998},
  volume = {95},
  pages = {14863--14868},
  month = {Dec},
  pdf = {../local/Eisen1998Cluster.pdf},
  file = {Eisen1998Cluster.pdf:Eisen1998Cluster.pdf:PDF},
  subject = {microarray},
  url = {http://www.pnas.org/cgi/reprint/95/25/14863.pdf}
}
@article{Fan2006Concordance,
  author = {Fan, C. and Oh, D.S. and Wessels, L. and Weigelt, B. and Nuyten,
	D.S.A. and Nobel, A.B. and van't Veer, L.J. and Perou, C.M.},
  title = {Concordance among gene-expression-based predictors for breast cancer},
  journal = {N. Engl. J. Med.},
  year = {2006},
  volume = {355},
  pages = {560},
  number = {6},
  doi = {10.1056/NEJMoa052933},
  pdf = {../local/Fan2006Concordance.pdf},
  file = {Fan2006Concordance.pdf:Fan2006Concordance.pdf:PDF},
  keywords = {breastcancer, microarray},
  owner = {jp},
  publisher = {Mass Med Soc},
  timestamp = {2011.01.13},
  url = {http://dx.doi.org/10.1056/NEJMoa052933}
}
@article{Ferea1999Systematic,
  author = {Ferea, T. L. and Botstein, D. and Brown, P. O. and Rosenzweig, R.
	F.},
  title = {Systematic changes in gene expression patterns following adaptive
	evolution in yeast},
  journal = {Proc. {N}atl. {A}cad. {S}ci. {USA}},
  year = {1999},
  volume = {96},
  pages = {9721--9726},
  number = {17},
  pdf = {../local/fere99.pdf},
  file = {fere99.pdf:local/fere99.pdf:PDF},
  subject = {microarray},
  url = {http://www.pnas.org/cgi/reprint/96/17/9721.pdf}
}
@article{Friedman2000Using,
  author = {Friedman, N. and Linial, M. and Nachman, I. and Pe'er, D.},
  title = {Using {B}ayesian Networks to Analyze Expression Data},
  journal = {J. Comput. Biol.},
  year = {2000},
  volume = {7},
  pages = {601--620},
  number = {3-4},
  abstract = {D{NA} hybridization arrays simultaneously measure the expression level
	for thousands of genes. {T}hese measurements provide a "snapshot"
	of transcription levels within the cell. {A} major challenge in computational
	biology is to uncover, from such measurements, gene/protein interactions
	and key biological features of cellular systems. {I}n this paper,
	we propose a new framework for discovering interactions between genes
	based on multiple expression measurements. {T}his framework builds
	on the use of {B}ayesian networks for representing statistical dependencies.
	{A} {B}ayesian network is a graph-based model of joint multivariate
	probability distributions that captures properties of conditional
	independence between variables. {S}uch models are attractive for
	their ability to describe complex stochastic processes and because
	they provide a clear methodology for learning from (noisy) observations.
	{W}e start by showing how {B}ayesian networks can describe interactions
	between genes. {W}e then describe a method for recovering gene interactions
	from microarray data using tools for learning {B}ayesian networks.
	{F}inally, we demonstrate this method on the {S}. cerevisiae cell-cycle
	measurements of {S}pellman et al. (1998).},
  doi = {10.1089/106652700750050961},
  pdf = {../local/Friedman2000Using.pdf},
  file = {Friedman2000Using.pdf:local/Friedman2000Using.pdf:PDF},
  keywords = {biogm},
  subject = {microarray},
  url = {http://dx.doi.org/10.1089/106652700750050961}
}
@article{Garnis2006High,
  author = {C. Garnis and W. W. Lockwood and E. Vucic and Y. Ge and L. Girard
	and J. D. Minna and A. F. Gazdar and S. Lam and C. MacAulay and W.
	L. Lam},
  title = {High resolution analysis of non-small cell lung cancer cell lines
	by whole genome tiling path array {CGH}.},
  journal = {Int. J. Cancer},
  year = {2006},
  volume = {118},
  pages = {1556--1564},
  number = {6},
  abstract = {Chromosomal regions harboring tumor suppressors and oncogenes are
	often deleted or amplified. Array comparative genomic hybridization
	detects segmental DNA copy number alterations in tumor DNA relative
	to a normal control. The recent development of a bacterial artificial
	chromosome array, which spans the human genome in a tiling path manner
	with >32,000 clones, has facilitated whole genome profiling at an
	unprecedented resolution. Using this technology, we comprehensively
	describe and compare the genomes of 28 commonly used non-small cell
	lung carcinoma (NSCLC) cell models, derived from 18 adenocarcinomas
	(AC), 9 squamous cell carcinomas and 1 large cell carcinoma. Analysis
	at such resolution not only provided a detailed genomic alteration
	template for each of these model cell lines, but revealed novel regions
	of frequent duplication and deletion. Significantly, a detailed analysis
	of chromosome 7 identified 6 distinct regions of alterations across
	this chromosome, implicating the presence of multiple novel oncogene
	loci on this chromosome. As well, a comparison between the squamous
	and AC cells revealed alterations common to both subtypes, such as
	the loss of 3p and gain of 5p, in addition to multiple hotspots more
	frequently associated with only 1 subtype. Interestingly, chromosome
	3q, which is known to be amplified in both subtypes, showed 2 distinct
	regions of alteration, 1 frequently altered in squamous and 1 more
	frequently altered in AC. In summary, our data demonstrate the unique
	information generated by high resolution analysis of NSCLC genomes
	and uncover the presence of genetic alterations prevalent in the
	different NSCLC subtypes.},
  doi = {10.1002/ijc.21491},
  institution = {British Columbia Cancer Research Centre, Vancouver, BC, Canada. cgarnis@bccrc.ca},
  keywords = {Carcinoma, Non-Small-Cell Lung, genetics/pathology; Cell Line, Tumor;
	Chromosomes, Artificial, Bacterial, genetics; Gene Amplification;
	Gene Dosage; Gene Expression Profiling; Genome, Human, genetics;
	Humans; Loss of Heterozygosity; Lung Neoplasms, genetics/pathology;
	Microarray Analysis, methods; Nucleic Acid Hybridization, methods},
  language = {eng},
  medline-pst = {ppublish},
  owner = {jp},
  pmid = {16187286},
  timestamp = {2010.01.08},
  url = {http://dx.doi.org/10.1002/ijc.21491}
}
@article{Gasch2001Genomic,
  author = {A.P. Gasch and M. Huang and S. Metzner and D. Botstein and S.J. Elledge
	and P.O. Brown},
  title = {Genomic expression responses to {DNA}-damaging agents and the regulatory
	role of the yeast {ATR} homolog {M}ec1p},
  journal = {Mol. {B}iol. {C}ell},
  year = {2001},
  volume = {12},
  pages = {2987--3003},
  number = {10},
  pdf = {../local/gasc01.pdf},
  file = {gasc01.pdf:local/gasc01.pdf:PDF},
  subject = {microarray},
  url = {http://www.molbiolcell.org/cgi/content/full/12/10/2987}
}
@article{Gasch2000Genomic,
  author = {Gasch, A. P. and Spellman, P. T. and Kao, C. M. and Carmel-Harel,
	O. and Eisen, M. B. and Storz, G. and Botstein, D. and Brown, P.
	O.},
  title = {Genomic {E}xpression {P}rograms in the {R}esponse of {Y}east {C}ells
	to {E}nvironmental {C}hanges},
  journal = {Mol. {B}iol. {C}ell},
  year = {2000},
  volume = {11},
  pages = {4241--4257},
  month = {Dec},
  pdf = {../local/gasc00.pdf},
  file = {gasc00.pdf:local/gasc00.pdf:PDF},
  subject = {microarray},
  url = {http://www.molbiolcell.org/cgi/reprint/11/12/4241.pdf}
}
@article{Golub1999Molecular,
  author = {Golub, T. R. and Slonim, D. K. and Tamayo, P. and Huard, C. and Gaasenbeek,
	M. and Mesirov, J. P. and Coller, H. and Loh, M. L. and Downing,
	J. R. and Caligiuri, M. A. and Bloomfield, C. D. and Lander, E. S.},
  title = {Molecular classification of cancer: class discovery and class prediction
	by gene expression monitoring},
  journal = {Science},
  year = {1999},
  volume = {286},
  pages = {531--537},
  abstract = {Although cancer classification has improved over the past 30 years,
	there has been no general approach for identifying new cancer classes
	(class discovery) or for assigning tumors to known classes (class
	prediction). Here, a generic approach to cancer classification based
	on gene expression monitoring by DNA microarrays is described and
	applied to human acute leukemias as a test case. A class discovery
	procedure automatically discovered the distinction between acute
	myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) without
	previous knowledge of these classes. An automatically derived class
	predictor was able to determine the class of new leukemia cases.
	The results demonstrate the feasibility of cancer classification
	based solely on gene expression moni- toring and suggest a general
	strategy for discovering and predicting cancer classes for other
	types of cancer, independent of previous biological knowledge.},
  doi = {10.1126/science.286.5439.531},
  pdf = {../local/Golub1999Molecular.pdf},
  file = {Golub1999Molecular.pdf:Golub1999Molecular.pdf:PDF},
  keywords = {csbcbook, csbcbook-ch3, csbcbook-ch4},
  subject = {microarray},
  url = {http://dx.doi.org/10.1126/science.286.5439.531}
}
@article{Gross2000Identification,
  author = {C. Gross and M. Kelleher and V.R. Iyer and P.O. Brown and D.R. Winge},
  title = {Identification of the copper regulon in {S}accharomyces cerevisiae
	by {DNA} microarrays},
  journal = {J. {B}iol. {C}hem.},
  year = {2000},
  volume = {275},
  pages = {32310--32316},
  number = {41},
  pdf = {../local/gros00.pdf},
  file = {gros00.pdf:local/gros00.pdf:PDF},
  subject = {microarray},
  url = {http://www.jbc.org/cgi/content/full/275/41/32310}
}
@article{Haasdonk2005Feature,
  author = {Bernard Haasdonk},
  title = {Feature space interpretation of {SVM}s with indefinite kernels.},
  journal = {I{EEE} {T}rans {P}attern {A}nal {M}ach {I}ntell},
  year = {2005},
  volume = {27},
  pages = {482-92},
  number = {4},
  month = {Apr},
  abstract = {Kernel methods are becoming increasingly popular for various kinds
	of machine learning tasks, the most famous being the support vector
	machine ({SVM}) for classification. {T}he {SVM} is well understood
	when using conditionally positive definite (cpd) kernel functions.
	{H}owever, in practice, non-cpd kernels arise and demand application
	in {SVM}s. {T}he procedure of "plugging" these indefinite kernels
	in {SVM}s often yields good empirical classification results. {H}owever,
	they are hard to interpret due to missing geometrical and theoretical
	understanding. {I}n this paper, we provide a step toward the comprehension
	of {SVM} classifiers in these situations. {W}e give a geometric interpretation
	of {SVM}s with indefinite kernel functions. {W}e show that such {SVM}s
	are optimal hyperplane classifiers not by margin maximization, but
	by minimization of distances between convex hulls in pseudo-{E}uclidean
	spaces. {B}y this, we obtain a sound framework and motivation for
	indefinite {SVM}s. {T}his interpretation is the basis for further
	theoretical analysis, e.g., investigating uniqueness, and for the
	derivation of practical guidelines like characterizing the suitability
	of indefinite {SVM}s.},
  doi = {10.1109/TPAMI.2005.78},
  pdf = {../local/Haasdonk2005Feature.pdf},
  file = {Haasdonk2005Feature.pdf:local/Haasdonk2005Feature.pdf:PDF},
  keywords = {Algorithms, Animals, Antibiotics, Antineoplastic, Artificial Intelligence,
	Automated, Automatic Data Processing, Butadienes, Chloroplasts, Cluster
	Analysis, Comparative Study, Computer Simulation, Computer-Assisted,
	Computing Methodologies, Database Management Systems, Databases,
	Diagnosis, Disinfectants, Dose-Response Relationship, Drug, Drug
	Toxicity, Electrodes, Electroencephalography, Ethylamines, Expert
	Systems, Factual, Feedback, Fungicides, Gene Expression Profiling,
	Genes, Genetic Markers, Humans, Image Enhancement, Image Interpretation,
	Implanted, Industrial, Information Storage and Retrieval, Kidney,
	Kidney Tubules, MEDLINE, Male, Mercuric Chloride, Microarray Analysis,
	Molecular Biology, Motor Cortex, Movement, Natural Language Processing,
	Neural Networks (Computer), Non-P.H.S., Non-U.S. Gov't, Numerical
	Analysis, Pattern Recognition, Plant Proteins, Predictive Value of
	Tests, Proteins, Proteome, Proximal, Puromycin Aminonucleoside, Rats,
	Reproducibility of Results, Research Support, Sensitivity and Specificity,
	Signal Processing, Sprague-Dawley, Subcellular Fractions, Terminology,
	Therapy, Time Factors, Toxicogenetics, U.S. Gov't, User-Computer
	Interface, 15794155},
  url = {http://dx.doi.org/10.1109/TPAMI.2005.78}
}
@article{Haferlach2005AML,
  author = {Torsten Haferlach and Alexander Kohlmann and Susanne Schnittger and
	Martin Dugas and Wolfgang Hiddemann and Wolfgang Kern and Claudia
	Schoch},
  title = {A{ML} {M}3 and {AML} {M}3 variant each have a distinct gene expression
	signature but also share patterns different from other genetically
	defined {AML} subtypes.},
  journal = {Genes {C}hromosomes {C}ancer},
  year = {2005},
  volume = {43},
  pages = {113-27},
  number = {2},
  month = {Jun},
  abstract = {Acute promyelocytic leukemia ({APL}) with t(15;17) appears in two
	phenotypes: {AML} {M}3, with abnormal promyelocytes showing heavy
	granulation and bundles of {A}uer rods, and {AML} {M}3 variant ({M}3v),
	with non- or hypogranular cytoplasm and a bilobed nucleus. {W}e investigated
	the global gene expression profiles of 35 {APL} patients (19 {AML}
	{M}3, 16 {AML} {M}3v) by using high-density {DNA}-oligonucleotide
	microarrays. {F}irst, an unsupervised approach clearly separated
	{APL} samples from other {AML}s characterized genetically as t(8;21)
	(n = 35), inv(16) (n = 35), or t(11q23)/{MLL} (n = 35) or as having
	a normal karyotype (n = 50). {S}econd, we found genes with functional
	relevance for blood coagulation that were differentially expressed
	between {APL} and other {AML}s. {F}urthermore, a supervised pairwise
	comparison between {M}3 and {M}3v revealed differential expression
	of genes that encode for biological functions and pathways such as
	granulation and maturation of hematologic cells, explaining morphologic
	and clinical differences. {D}iscrimination between {M}3 and {M}3v
	based on gene signatures showed a median classification accuracy
	of 90\% by use of 10-fold {CV} and support vector machines. {A}dditional
	molecular mutations such as {FLT}3-{LM}, which were significantly
	more frequent in {M}3v than in {M}3 ({P} < 0.0001), may partly contribute
	to the different phenotypes. {H}owever, linear regression analysis
	demonstrated that genes differentially expressed between {M}3 and
	{M}3v did not correlate with {FLT}3-{LM}.},
  doi = {10.1002/gcc.20175},
  pdf = {../local/Haferlach2005AML.pdf},
  file = {Haferlach2005AML.pdf:local/Haferlach2005AML.pdf:PDF},
  keywords = {biosvm microarray},
  url = {http://dx.doi.org/10.1002/gcc.20175}
}
@article{Haferlach2005global,
  author = {Torsten Haferlach and Alexander Kohlmann and Susanne Schnittger and
	Martin Dugas and Wolfgang Hiddemann and Wolfgang Kern and Claudia
	Schoch},
  title = {A global approach to the diagnosis of leukemia using gene expression
	profiling.},
  journal = {Blood},
  year = {2005},
  volume = {106},
  pages = {1189-1198},
  number = {4},
  month = {Aug},
  abstract = {Accurate diagnosis and classification of leukemias are the bases for
	the appropriate management of patients. {T}he diagnostic accuracy
	and efficiency of present methods may be improved by the use of microarrays
	for gene expression profiling. {W}e analyzed gene expression profiles
	in bone marrow and peripheral blood samples from 937 patients with
	all clinically relevant leukemia subtypes (n=892) and non-leukemic
	controls (n=45) by {U}133{A} and {B} {G}ene{C}hips ({A}ffymetrix).
	{F}or each subgroup differentially expressed genes were calculated.
	{C}lass prediction was performed using support vector machines. {P}rediction
	accuracies were estimated by 10-fold cross validation and assessed
	for robustness in a 100-fold resampling approach using randomly chosen
	test-sets consisting of 1/3 of the samples. {A}pplying the top 100
	genes of each subgroup an overall prediction accuracy of 95.1\% was
	achieved which was confirmed by resampling (median, 93.8\%; 95\%
	confidence interval, 91.4\%-95.8\%). {I}n particular, {AML} with
	t(15;17), t(8;21), or inv(16), {CLL}, and {P}ro-{B}-{ALL} with t(11q23)
	were classified with 100\% sensitivity and 100\% specificity. {A}ccordingly,
	cluster analysis completely separated all of the 13 subgroups analyzed.
	{G}ene expression profiling can predict all clinically relevant subentities
	of leukemia with high accuracy.},
  doi = {10.1182/blood-2004-12-4938},
  pdf = {../local/Haferlach2005global.pdf},
  file = {Haferlach2005global.pdf:local/Haferlach2005global.pdf:PDF},
  keywords = {biosvm microarray},
  pii = {2004-12-4938},
  url = {http://dx.doi.org/10.1182/blood-2004-12-4938}
}
@article{Hanisch2002Co-clustering,
  author = {D. Hanisch and A. Zien and R. Zimmer and T. Lengauer},
  title = {Co-clustering of biological networks and gene expression data},
  journal = {Bioinformatics},
  year = {2002},
  annote = {To appear},
  subject = {microarraybionet},
  url = {http://cartan.gmd.de/~hanisch/paper/CoClustering.pdf}
}
@article{Ioannidis2005Microarrays,
  author = {Ioannidis, J. P. A.},
  title = {Microarrays and molecular research: noise discovery?},
  journal = {Lancet},
  year = {2005},
  volume = {365},
  pages = {454},
  number = {9458},
  pdf = {../local/Ioannidis2005Microarrays.pdf},
  file = {Ioannidis2005Microarrays.pdf:Ioannidis2005Microarrays.pdf:PDF},
  keywords = {microarray},
  owner = {jp},
  timestamp = {2011.01.12}
}
@article{Ishkanian2004tiling,
  author = {Ishkanian, A. S. and Malloff, C. A. and Watson, S. K. and DeLeeuw,
	R. J. and Chi, B. and Coe, B. P. and Snijders, A. and Albertson,
	D. G. and Pinkel, D. and Marra, M. A. and Ling, V. and MacAulay,
	C. and Lam, W. L.},
  title = {A tiling resolution {DNA} microarray with complete coverage of the
	human genome},
  journal = {Nat. Genet.},
  year = {2004},
  volume = {36},
  pages = {299--303},
  number = {3},
  month = {Mar},
  abstract = {We constructed a tiling resolution array consisting of 32,433 overlapping
	BAC clones covering the entire human genome. This increases our ability
	to identify genetic alterations and their boundaries throughout the
	genome in a single comparative genomic hybridization (CGH) experiment.
	At this tiling resolution, we identified minute DNA alterations not
	previously reported. These alterations include microamplifications
	and deletions containing oncogenes, tumor-suppressor genes and new
	genes that may be associated with multiple tumor types. Our findings
	show the need to move beyond conventional marker-based genome comparison
	approaches, that rely on inference of continuity between interval
	markers. Our submegabase resolution tiling set for array CGH (SMRT
	array) allows comprehensive assessment of genomic integrity and thereby
	the identification of new genes associated with disease.},
  doi = {10.1038/ng1307},
  pdf = {../local/Ishkanian2004tiling.pdf},
  file = {Ishkanian2004tiling.pdf:Ishkanian2004tiling.pdf:PDF},
  institution = {British Columbia Cancer Research Centre, 601 West 10th Avenue, Vancouver,
	British Columbia V5Z 1L3, Canada.},
  keywords = {csbcbook, microarray},
  owner = {jp},
  pii = {ng1307},
  pmid = {14981516},
  timestamp = {2009.10.08},
  url = {http://dx.doi.org/10.1038/ng1307}
}
@article{Kuhn2001Global,
  author = {K. M. Kuhn and J. L. DeRisi and P. O. Brown and P. Sarnow},
  title = {Global and specific translational regulation in the genomic response
	of {S}accharomyces cerevisiae to a rapid transfer from a fermentable
	to a nonfermentable carbon source},
  journal = {Mol. {C}ell. {B}iol.},
  year = {2001},
  volume = {21},
  pages = {916--927},
  number = {3},
  pdf = {../local/kuhn01.pdf},
  file = {kuhn01.pdf:local/kuhn01.pdf:PDF},
  subject = {microarray},
  url = {http://mcb.asm.org/cgi/content/full/21/3/916?view=full&pmid=11154278}
}
@article{LeRoch2003Discovery,
  author = {Le Roch, K. G. and Zhou, Y. and Blair, P. L. and Grainger, M. and
	Moch, J. K. and Haynes, J. D. and De la Vega, P. and Holder, A. A.
	and Batalov, S. and Carucci, D. J. and Winzeler, E. A.},
  title = {Discovery of Gene Function by Expression Profiling of the Malaria
	Parasite Life Cycle},
  journal = {Science},
  year = {2003},
  volume = {301},
  pages = {1503-1508},
  number = {5639},
  abstract = {The completion of the genome sequence for {P}lasmodium falciparum,
	the species responsible for most malaria human deaths, has the potential
	to reveal hundreds of new drug targets and proteins involved in pathogenesis.
	{H}owever, only approximately 35% of the genes code for proteins
	with an identifiable function. {T}he absence of routine genetic tools
	for studying {P}lasmodium parasites suggests that this number is
	unlikely to change quickly if conventional serial methods are used
	to characterize encoded proteins. {H}ere, we use a high-density oligonucleotide
	array to generate expression profiles of human and mosquito stages
	of the malaria parasite's life cycle. {G}enes with highly correlated
	levels and temporal patterns of expression were often involved in
	similar functions or cellular processes.},
  doi = {10.1126/science.1087025},
  pdf = {../local/LeRoch2003Discovery.pdf},
  file = {LeRoch2003Discovery.pdf:LeRoch2003Discovery.pdf:PDF},
  keywords = {microarray plasmodium},
  owner = {vert},
  url = {http://www.sciencemag.org/cgi/content/full/301/5639/1503}
}
@article{Levy2007Diploid,
  author = {Samuel Levy and Granger Sutton and Pauline C Ng and Lars Feuk and
	Aaron L Halpern and Brian P Walenz and Nelson Axelrod and Jiaqi Huang
	and Ewen F Kirkness and Gennady Denisov and Yuan Lin and Jeffrey
	R MacDonald and Andy Wing Chun Pang and Mary Shago and Timothy B
	Stockwell and Alexia Tsiamouri and Vineet Bafna and Vikas Bansal
	and Saul A Kravitz and Dana A Busam and Karen Y Beeson and Tina C
	McIntosh and Karin A Remington and Josep F Abril and John Gill and
	Jon Borman and Yu-Hui Rogers and Marvin E Frazier and Stephen W Scherer
	and Robert L Strausberg and J. Craig Venter},
  title = {The diploid genome sequence of an individual human.},
  journal = {PLoS Biol},
  year = {2007},
  volume = {5},
  pages = {e254},
  number = {10},
  month = {Sep},
  abstract = {Presented here is a genome sequence of an individual human. It was
	produced from approximately 32 million random DNA fragments, sequenced
	by Sanger dideoxy technology and assembled into 4,528 scaffolds,
	comprising 2,810 million bases (Mb) of contiguous sequence with approximately
	7.5-fold coverage for any given region. We developed a modified version
	of the Celera assembler to facilitate the identification and comparison
	of alternate alleles within this individual diploid genome. Comparison
	of this genome and the National Center for Biotechnology Information
	human reference assembly revealed more than 4.1 million DNA variants,
	encompassing 12.3 Mb. These variants (of which 1,288,319 were novel)
	included 3,213,401 single nucleotide polymorphisms (SNPs), 53,823
	block substitutions (2-206 bp), 292,102 heterozygous insertion/deletion
	events (indels)(1-571 bp), 559,473 homozygous indels (1-82,711 bp),
	90 inversions, as well as numerous segmental duplications and copy
	number variation regions. Non-SNP DNA variation accounts for 22\%
	of all events identified in the donor, however they involve 74\%
	of all variant bases. This suggests an important role for non-SNP
	genetic alterations in defining the diploid genome structure. Moreover,
	44\% of genes were heterozygous for one or more variants. Using a
	novel haplotype assembly strategy, we were able to span 1.5 Gb of
	genome sequence in segments >200 kb, providing further precision
	to the diploid nature of the genome. These data depict a definitive
	molecular portrait of a diploid human genome that provides a starting
	point for future genome comparisons and enables an era of individualized
	genomic information.},
  doi = {10.1371/journal.pbio.0050254},
  institution = {J. Craig Venter Institute, Rockville, Maryland, USA. slevy@jcvi.org},
  keywords = {Base Sequence; Chromosome Mapping, instrumentation/methods; Chromosomes,
	Human; Chromosomes, Human, Y, genetics; Diploidy; Gene Dosage; Genome,
	Human; Genotype; Haplotypes; Human Genome Project; Humans; INDEL
	Mutation; In Situ Hybridization, Fluorescence; Male; Microarray Analysis;
	Middle Aged; Molecular Sequence Data; Pedigree; Phenotype; Polymorphism,
	Single Nucleotide; Reproducibility of Results; Sequence Analysis,
	DNA, instrumentation/methods},
  language = {eng},
  medline-pst = {ppublish},
  owner = {philippe},
  pii = {07-PLBI-RA-1258},
  pmid = {17803354},
  timestamp = {2010.07.28},
  url = {http://dx.doi.org/10.1371/journal.pbio.0050254}
}
@article{Mavroforakis2005Significance,
  author = {Michael Mavroforakis and Harris Georgiou and Nikos Dimitropoulos
	and Dionisis Cavouras and Sergios Theodoridis},
  title = {Significance analysis of qualitative mammographic features, using
	linear classifiers, neural networks and support vector machines.},
  journal = {Eur {J} {R}adiol},
  year = {2005},
  volume = {54},
  pages = {80-9},
  number = {1},
  month = {Apr},
  abstract = {Advances in modern technologies and computers have enabled digital
	image processing to become a vital tool in conventional clinical
	practice, including mammography. {H}owever, the core problem of the
	clinical evaluation of mammographic tumors remains a highly demanding
	cognitive task. {I}n order for these automated diagnostic systems
	to perform in levels of sensitivity and specificity similar to that
	of human experts, it is essential that a robust framework on problem-specific
	design parameters is formulated. {T}his study is focused on identifying
	a robust set of clinical features that can be used as the base for
	designing the input of any computer-aided diagnosis system for automatic
	mammographic tumor evaluation. {A} thorough list of clinical features
	was constructed and the diagnostic value of each feature was verified
	against current clinical practices by an expert physician. {T}hese
	features were directly or indirectly related to the overall morphological
	properties of the mammographic tumor or the texture of the fine-scale
	tissue structures as they appear in the digitized image, while others
	contained external clinical data of outmost importance, like the
	patient's age. {T}he entire feature set was used as an annotation
	list for describing the clinical properties of mammographic tumor
	cases in a quantitative way, such that subsequent objective analyses
	were possible. {F}or the purposes of this study, a mammographic image
	database was created, with complete clinical evaluation descriptions
	and positive histological verification for each case. {A}ll tumors
	contained in the database were characterized according to the identified
	clinical features' set and the resulting dataset was used as input
	for discrimination and diagnostic value analysis for each one of
	these features. {S}pecifically, several standard methodologies of
	statistical significance analysis were employed to create feature
	rankings according to their discriminating power. {M}oreover, three
	different classification models, namely linear classifiers, neural
	networks and support vector machines, were employed to investigate
	the true efficiency of each one of them, as well as the overall complexity
	of the diagnostic task of mammographic tumor characterization. {B}oth
	the statistical and the classification results have proven the explicit
	correlation of all the selected features with the final diagnosis,
	qualifying them as an adequate input base for any type of similar
	automated diagnosis system. {T}he underlying complexity of the diagnostic
	task has justified the high value of sophisticated pattern recognition
	architectures.},
  doi = {10.1016/j.ejrad.2004.12.015},
  pdf = {../local/Mavroforakis2005Significance.pdf},
  file = {Mavroforakis2005Significance.pdf:local/Mavroforakis2005Significance.pdf:PDF},
  keywords = {Algorithms, Animals, Antibiotics, Antineoplastic, Artificial Intelligence,
	Butadienes, Chloroplasts, Comparative Study, Computer Simulation,
	Computer-Assisted, Diagnosis, Disinfectants, Dose-Response Relationship,
	Drug, Drug Toxicity, Electrodes, Electroencephalography, Ethylamines,
	Expert Systems, Feedback, Fungicides, Gene Expression Profiling,
	Genes, Genetic Markers, Humans, Implanted, Industrial, Information
	Storage and Retrieval, Kidney, Kidney Tubules, MEDLINE, Male, Mercuric
	Chloride, Microarray Analysis, Molecular Biology, Motor Cortex, Movement,
	Natural Language Processing, Neural Networks (Computer), Non-P.H.S.,
	Non-U.S. Gov't, Plant Proteins, Predictive Value of Tests, Proteins,
	Proteome, Proximal, Puromycin Aminonucleoside, Rats, Reproducibility
	of Results, Research Support, Sprague-Dawley, Subcellular Fractions,
	Terminology, Therapy, Time Factors, Toxicogenetics, U.S. Gov't, User-Computer
	Interface, 15797296},
  pii = {S0720-048X(05)00023-9},
  url = {http://dx.doi.org/10.1016/j.ejrad.2004.12.015}
}
@article{Meireles2003Differentially,
  author = {Meireles, S.I. and Carvalho, A.F. and Hirata, R. and Montagnini,
	A.L. and Martins, W.K. and Runza, F.B. and Stolf, B.S. and Termini,
	L. and Neto, C.E. and Silva, R.L. and Soares, F.A. and Neves, E.J.
	and Reis, L.F.},
  title = {Differentially expressed genes in gastric tumors identified by c{DNA}
	array.},
  journal = {Cancer {L}ett.},
  year = {2003},
  volume = {190},
  pages = {199-211},
  number = {2},
  month = {Feb},
  abstract = {Using c{DNA} fragments from the {FAPESP}/l{ICR} {C}ancer {G}enome
	{P}roject, we constructed a c{DNA} array having 4512 elements and
	determined gene expression in six normal and six tumor gastric tissues.
	{U}sing t-statistics, we identified 80 c{DNA}s whose expression in
	normal and tumor samples differed more than 3.5 sample standard deviations.
	{U}sing {S}elf-{O}rganizing {M}ap, the expression profile of these
	c{DNA}s allowed perfect separation of malignant and non-malignant
	samples. {U}sing the supervised learning procedure {S}upport {V}ector
	{M}achine, we identified trios of c{DNA}s that could be used to classify
	samples as normal or tumor, based on single-array analysis. {F}inally,
	we identified genes with altered linear correlation when their expression
	in normal and tumor samples were compared. {F}urther investigation
	concerning the function of these genes could contribute to the understanding
	of gastric carcinogenesis and may prove useful in molecular diagnostics.},
  doi = {10.1016/S0304-3835(02)00587},
  pdf = {../local/Meireles2003Differentially.pdf},
  file = {Meireles2003Differentially.pdf:local/Meireles2003Differentially.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {jeanphilippevert},
  url = {http://dx.doi.org/10.1016/S0304-3835(02)00587-6}
}
@article{Michiels2005Prediction,
  author = {Michiels, S. and Koscielny, S. and Hill, C.},
  title = {Prediction of cancer outcome with microarrays: a multiple random
	validation strategy},
  journal = {Lancet},
  year = {2005},
  volume = {365},
  pages = {488--492},
  number = {9458},
  abstract = {BACKGROUND: General studies of microarray gene-expression profiling
	have been undertaken to predict cancer outcome. Knowledge of this
	gene-expression profile or molecular signature should improve treatment
	of patients by allowing treatment to be tailored to the severity
	of the disease. We reanalysed data from the seven largest published
	studies that have attempted to predict prognosis of cancer patients
	on the basis of DNA microarray analysis. METHODS: The standard strategy
	is to identify a molecular signature (ie, the subset of genes most
	differentially expressed in patients with different outcomes) in
	a training set of patients and to estimate the proportion of misclassifications
	with this signature on an independent validation set of patients.
	We expanded this strategy (based on unique training and validation
	sets) by using multiple random sets, to study the stability of the
	molecular signature and the proportion of misclassifications. FINDINGS:
	The list of genes identified as predictors of prognosis was highly
	unstable; molecular signatures strongly depended on the selection
	of patients in the training sets. For all but one study, the proportion
	misclassified decreased as the number of patients in the training
	set increased. Because of inadequate validation, our chosen studies
	published overoptimistic results compared with those from our own
	analyses. Five of the seven studies did not classify patients better
	than chance. INTERPRETATION: The prognostic value of published microarray
	results in cancer studies should be considered with caution. We advocate
	the use of validation by repeated random sampling.},
  doi = {10.1016/S0140-6736(05)17866-0},
  institution = {Biostatistics and Epidemiology Unit, Institut Gustave Roussy, Villejuif,
	France.},
  keywords = {featureselection, breastcancer, microarray},
  language = {eng},
  medline-pst = {ppublish},
  owner = {jp},
  pii = {S0140-6736(05)17866-0},
  pmid = {15705458},
  timestamp = {2010.10.12},
  url = {http://dx.doi.org/10.1016/S0140-6736(05)17866-0}
}
@techreport{Mukherjee1998Support,
  author = {S. Mukherjee and P. Tamayo and J. P. Mesirov and D. Slonim and A.
	Verri and T. Poggio},
  title = {Support vector machine classification of microarray data},
  institution = {C.B.L.C.},
  year = {1998},
  number = {182},
  note = {A.I. Memo 1677},
  pdf = {../local/Mukherjee1998Support.pdf},
  file = {Mukherjee1998Support.pdf:local/Mukherjee1998Support.pdf:PDF},
  keywords = {biosvm microarray},
  subject = {biokernel},
  url = {http://citeseer.nj.nec.com/437379.html}
}
@article{ODonnell2005Gene,
  author = {Rebekah K O'Donnell and Michael Kupferman and S. Jack Wei and Sunil
	Singhal and Randal Weber and Bert O'Malley and Yi Cheng and Mary
	Putt and Michael Feldman and Barry Ziober and Ruth J Muschel},
  title = {Gene expression signature predicts lymphatic metastasis in squamous
	cell carcinoma of the oral cavity.},
  journal = {Oncogene},
  year = {2005},
  volume = {24},
  pages = {1244-51},
  number = {7},
  month = {Feb},
  abstract = {Metastasis via the lymphatics is a major risk factor in squamous cell
	carcinoma of the oral cavity ({OSCC}). {W}e sought to determine whether
	the presence of metastasis in the regional lymph node could be predicted
	by a gene expression signature of the primary tumor. {A} total of
	18 {OSCC}s were characterized for gene expression by hybridizing
	{RNA} to {A}ffymetrix {U}133{A} gene chips. {G}enes with differential
	expression were identified using a permutation technique and verified
	by quantitative {RT}-{PCR} and immunohistochemistry. {A} predictive
	rule was built using a support vector machine, and the accuracy of
	the rule was evaluated using crossvalidation on the original data
	set and prediction of an independent set of four patients. {M}etastatic
	primary tumors could be differentiated from nonmetastatic primary
	tumors by a signature gene set of 116 genes. {T}his signature gene
	set correctly predicted the four independent patients as well as
	associating five lymph node metastases from the original patient
	set with the metastatic primary tumor group. {W}e concluded that
	lymph node metastasis could be predicted by gene expression profiles
	of primary oral cavity squamous cell carcinomas. {T}he presence of
	a gene expression signature for lymph node metastasis indicates that
	clinical testing to assess risk for lymph node metastasis should
	be possible.},
  doi = {10.1038/sj.onc.1208285},
  pdf = {../local/O'Donnell2005Gene.pdf},
  file = {O'Donnell2005Gene.pdf:local/O'Donnell2005Gene.pdf:PDF},
  keywords = {biosvm microarray},
  pii = {1208285},
  url = {http://dx.doi.org/10.1038/sj.onc.1208285}
}
@article{Ogawa2000New,
  author = {Nobuo Ogawa and Joseph DeRisi and Patrick O. Brown},
  title = {New {C}omponents of a {S}ystem for {P}hosphate {A}ccumulation and
	{P}olyphosphate {M}etabolism in {S}accharomyces cerevisiae {R}evealed
	by {G}enomic {E}xpression {A}nalysis},
  journal = {Mol. {B}iol. {C}ell},
  year = {2000},
  volume = {11},
  pages = {4309--4321},
  month = {Dec},
  pdf = {../local/ogaw00.pdf},
  file = {ogaw00.pdf:local/ogaw00.pdf:PDF},
  subject = {microarray},
  url = {http://www.molbiolcell.org/cgi/reprint/11/12/4309.pdf}
}
@article{Pavey2004Microarray,
  author = {Pavey, S. and Johansson, P. and Packer, L. and Taylor, J. and Stark,
	M. and Pollock, P.M. and Walker, G.J. and Boyle, G.M. and Harper,
	U. and Cozzi, S.J. and Hansen, K. and Yudt, L. and Schmidt, C. and
	Hersey, P. and Ellem, K.A. and O'Rourke, M.G. and Parsons, P.G. and
	Meltzer, P. and Ringner, M. and Hayward, N.K.},
  title = {Microarray expression profiling in melanoma reveals a {BRAF} mutation
	signature},
  journal = {Oncogene},
  year = {2004},
  volume = {23},
  pages = {4060-4067},
  number = {23},
  month = {May},
  abstract = {We have used microarray gene expression profiling and machine learning
	to predict the presence of {BRAF} mutations in a panel of 61 melanoma
	cell lines. {T}he {BRAF} gene was found to be mutated in 42 samples
	(69%) and intragenic mutations of the {NRAS} gene were detected in
	seven samples (11%). {N}o cell line carried mutations of both genes.
	{U}sing support vector machines, we have built a classifier that
	differentiates between melanoma cell lines based on {BRAF} mutation
	status. {A}s few as 83 genes are able to discriminate between {BRAF}
	mutant and {BRAF} wild-type samples with clear separation observed
	using hierarchical clustering. {M}ultidimensional scaling was used
	to visualize the relationship between a {BRAF} mutation signature
	and that of a generalized mitogen-activated protein kinase ({MAPK})
	activation (either {BRAF} or {NRAS} mutation) in the context of the
	discriminating gene list. {W}e observed that samples carrying {NRAS}
	mutations lie somewhere between those with or without {BRAF} mutations.
	{T}hese observations suggest that there are gene-specific mutation
	signals in addition to a common {MAPK} activation that result from
	the pleiotropic effects of either {BRAF} or {NRAS} on other signaling
	pathways, leading to measurably different transcriptional changes.},
  doi = {10.1038/sj.onc.1207563},
  pdf = {../local/Pavey2004Microarray.pdf},
  file = {Pavey2004Microarray.pdf:local/Pavey2004Microarray.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {jeanphilippevert},
  url = {http://dx.doi.org/10.1038/sj.onc.1207563}
}
@article{Peng2003Molecular,
  author = {Peng, S. and Xu, Q. and Ling, X.B. and Peng, X. and Du, W. and Chen,
	L.},
  title = {Molecular classification of cancer types from microarray data using
	the combination of genetic algorithms and support vector machines.},
  journal = {F{EBS} {L}ett.},
  year = {2003},
  volume = {555},
  pages = {358-362},
  number = {2},
  abstract = {Simultaneous multiclass classification of tumor types is essential
	for future clinical implementations of microarray-based cancer diagnosis.
	{I}n this study, we have combined genetic algorithms ({GA}s) and
	all paired support vector machines ({SVM}s) for multiclass cancer
	identification. {T}he predictive features have been selected through
	iterative {SVM}s/{GA}s, and recursive feature elimination post-processing
	steps, leading to a very compact cancer-related predictive gene set.
	{L}eave-one-out cross-validations yielded accuracies of 87.93% for
	the eight-class and 85.19% for the fourteen-class cancer classifications,
	outperforming the results derived from previously published methods.},
  doi = {10.1016/S0014-5793(03)01275-4},
  pdf = {../local/Peng2003Molecular.pdf},
  file = {Peng2003Molecular.pdf:local/Peng2003Molecular.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {jeanphilippevert},
  url = {http://dx.doi.org/10.1016/S0014-5793(03)01275-4}
}
@article{Pilpel2001Identifying,
  author = {Pilpel, Y. and Sudarsanam, P. and Church, G. M.},
  title = {Identifying regulatory networks by combinatorial analysis of promoter
	elements},
  journal = {Nature},
  year = {2001},
  volume = {29},
  pages = {153--159},
  pdf = {../local/pilp01.pdf},
  file = {pilp01.pdf:local/pilp01.pdf:PDF},
  subject = {microarray},
  url = {http://www.nature.com/cgi-taf/DynaPage.taf?file=/ng/journal/v29/n2/full/ng724.html&filetype=PDF}
}
@article{Pochet2004Systematic,
  author = {Pochet, N. and De Smet, F. and Suykens, J. A. K. and De Moor, B.
	L. R.},
  title = {Systematic benchmarking of microarray data classification: assessing
	the role of non-linearity and dimensionality reduction},
  journal = {Bioinformatics},
  year = {2004},
  volume = {20},
  pages = {3185-3195},
  number = {17},
  month = {Nov},
  abstract = {Motivation: {M}icroarrays are capable of determining the expression
	levels of thousands of genes simultaneously. {I}n combination with
	classification methods, this technology can be useful to support
	clinical management decisions for individual patients, e.g. in oncology.
	{T}he aim of this paper is to systematically benchmark the role of
	non-linear versus linear techniques and dimensionality reduction
	methods. {R}esults: {A} systematic benchmarking study is performed
	by comparing linear versions of standard classification and dimensionality
	reduction techniques with their non-linear versions based on non-linear
	kernel functions with a radial basis function ({RBF}) kernel. {A}
	total of 9 binary cancer classification problems, derived from 7
	publicly available microarray datasets, and 20 randomizations of
	each problem are examined. {C}onclusions: {T}hree main conclusions
	can be formulated based on the performances on independent test sets.
	(1) {W}hen performing classification with least squares support vector
	machines ({LS}-{SVM}s) (without dimensionality reduction), {RBF}
	kernels can be used without risking too much overfitting. {T}he results
	obtained with well-tuned {RBF} kernels are never worse and sometimes
	even statistically significantly better compared to results obtained
	with a linear kernel in terms of test set receiver operating characteristic
	and test set accuracy performances. (2) {E}ven for classification
	with linear classifiers like {LS}-{SVM} with linear kernel, using
	regularization is very important. (3) {W}hen performing kernel principal
	component analysis (kernel {PCA}) before classification, using an
	{RBF} kernel for kernel {PCA} tends to result in overfitting, especially
	when using supervised feature selection. {I}t has been observed that
	an optimal selection of a large number of features is often an indication
	for overfitting. {K}ernel {PCA} with linear kernel gives better results.
	{A}vailability: {M}atlab scripts are available on request. {S}upplementary
	information: http://www.esat.kuleuven.ac.be/~npochet/{B}ioinformatics/},
  doi = {10.1093/bioinformatics/bth383},
  pdf = {../local/Pochet2004Systematic.pdf},
  file = {Pochet2004Systematic.pdf:local/Pochet2004Systematic.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {jeanphilippevert},
  url = {http://dx.doi.org/10.1093/bioinformatics/bth383}
}
@article{Ramaswamy2001Multiclass,
  author = {Ramaswamy, S. and Tamayo, P. and Rifkin, R. and Mukherjee, S. and
	Yeang, C.H. and Angelo, M. and Ladd, C. and Reich, M. and Latulippe,
	E. and Mesirov, J.P. and Poggio, T. and Gerald, W. and Loda, M. and
	Lander, E.S. and Golub, T.R.},
  title = {Multiclass cancer diagnosis using tumor gene expression signatures},
  journal = {Proc. {N}atl. {A}cad. {S}ci. {USA}},
  year = {2001},
  volume = {98},
  pages = {15149-15154},
  number = {26},
  month = {Dec},
  abstract = {The optimal treatment of patients with cancer depends on establishing
	accurate diagnoses by using a complex combination of clinical and
	histopathological data. {I}n some instances, this task is difficult
	or impossible because of atypical clinical presentation or histopathology.
	{T}o determine whether the diagnosis of multiple common adult malignancies
	could be achieved purely by molecular classification, we subjected
	218 tumor samples, spanning 14 common tumor types, and 90 normal
	tissue samples to oligonucleotide microarray gene expression analysis.
	{T}he expression levels of 16,063 genes and expressed sequence tags
	were used to evaluate the accuracy of a multiclass classifier based
	on a support vector machine algorithm. {O}verall classification accuracy
	was 78%, far exceeding the accuracy of random classification (9%).
	{P}oorly differentiated cancers resulted in low-confidence predictions
	and could not be accurately classified according to their tissue
	of origin, indicating that they are molecularly distinct entities
	with dramatically different gene expression patterns compared with
	their well differentiated counterparts. {T}aken together, these results
	demonstrate the feasibility of accurate, multiclass molecular cancer
	classification and suggest a strategy for future clinical implementation
	of molecular cancer diagnostics.},
  doi = {10.1073/pnas.211566398},
  pdf = {../local/Ramaswamy2001Multiclass.pdf},
  file = {Ramaswamy2001Multiclass.pdf:local/Ramaswamy2001Multiclass.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {vert},
  url = {http://dx.doi.org/10.1073/pnas.211566398}
}
@phdthesis{Reyal2009Analyse,
  author = {Reyal, F.},
  title = {Analyse du profil d'expression par la technique des puces {\`a} ADN.
	Application \`a la caract\'erisation mol\'eculaire et \`a la d\'etermination
	du pronostic des cancers canalaires infiltrants du sein.},
  school = {Universit\'e Paris 11},
  year = {2009},
  keywords = {breastcancer, microarray},
  owner = {jp},
  timestamp = {2009.10.31}
}
@article{Schena1995Quantitative,
  author = {M. Schena and D. Shalon and R. W. Davis and P. O. Brown},
  title = {Quantitative monitoring of gene expression patterns with a complementary
	DNA microarray.},
  journal = {Science},
  year = {1995},
  volume = {270},
  pages = {467--470},
  number = {5235},
  month = {Oct},
  abstract = {A high-capacity system was developed to monitor the expression of
	many genes in parallel. Microarrays prepared by high-speed robotic
	printing of complementary DNAs on glass were used for quantitative
	expression measurements of the corresponding genes. Because of the
	small format and high density of the arrays, hybridization volumes
	of 2 microliters could be used that enabled detection of rare transcripts
	in probe mixtures derived from 2 micrograms of total cellular messenger
	RNA. Differential expression measurements of 45 Arabidopsis genes
	were made by means of simultaneous, two-color fluorescence hybridization.},
  doi = {10.1126/science.270.5235.467},
  pdf = {../local/Schena1995Quantitative.pdf},
  file = {Schena1995Quantitative.pdf:Schena1995Quantitative.pdf:PDF},
  institution = {Department of Biochemistry, Beckman Center, Stanford University Medical
	Center, CA 94305, USA.},
  keywords = {microarray},
  owner = {jp},
  pmid = {7569999},
  timestamp = {2009.02.08},
  url = {http://dx.doi.org/10.1126/science.270.5235.467}
}
@article{Segal2005From,
  author = {Segal, E. and Friedman, N. and Kaminski, N. and Regev, A. and Koller,
	D.},
  title = {From signatures to models: understanding cancer using microarrays},
  journal = {Nat {G}enet},
  year = {2005},
  volume = {37},
  pages = {S38-45},
  number = {6 Suppl},
  abstract = {Genomics has the potential to revolutionize the diagnosis and management
	of cancer by offering an unprecedented comprehensive view of the
	molecular underpinnings of pathology. {C}omputational analysis is
	essential to transform the masses of generated data into a mechanistic
	understanding of disease. {H}ere we review current research aimed
	at uncovering the modular organization and function of transcriptional
	networks and responses in cancer. {W}e first describe how methods
	that analyze biological processes in terms of higher-level modules
	can identify robust signatures of disease mechanisms. {W}e then discuss
	methods that aim to identify the regulatory mechanisms underlying
	these modules and processes. {F}inally, we show how comparative analysis,
	combining human data with model organisms, can lead to more robust
	findings. {W}e conclude by discussing the challenges of generalizing
	these methods from cells to tissues and the opportunities they offer
	to improve cancer diagnosis and management.},
  doi = {10.1038/ng1561},
  pdf = {../local/Segal2005From.pdf},
  file = {Segal2005From.pdf:Segal2005From.pdf:PDF},
  keywords = {microarray},
  url = {http://dx.doi.org/10.1038/ng1561}
}
@article{Selinger2000RNA,
  author = {Douglas W. Selinger and Kevin J. Cheung and Rui Mei and Erik M. Johansson
	and Craig S. Richmond and Frederick R. Blattner and David J. Lockhart
	and George M. Church},
  title = {R{NA} expression analysis using a 30 base pair resolution {E}scherichia
	coli genome array},
  journal = {Nat. {B}iotechnol.},
  year = {2000},
  volume = {18},
  pages = {1262--1268},
  pdf = {../local/seli00.pdf},
  file = {seli00.pdf:local/seli00.pdf:PDF},
  subject = {microarray},
  url = {http://www.nature.com/cgi-taf/DynaPage.taf?file=/nbt/journal/v18/n12/full/nbt1200_1262.html&filetype=PDF}
}
@article{Sherlock2001Stanford,
  author = {G. Sherlock and T. Hernandez-Boussard and A. Kasarskis and G. Binkley
	and J.C. Matese and S.S. Dwight and M. Kaloper and S. Weng and H.
	Jin and C.A. Ball and M.B. Eisen and P.T. Spellman},
  title = {The {S}tanford {M}icroarray {D}atabase},
  journal = {Nucleic {A}cids {R}es.},
  year = {2001},
  volume = {29},
  pages = {152--155},
  number = {1},
  month = {Jan},
  pdf = {../local/sher01.pdf},
  file = {sher01.pdf:local/sher01.pdf:PDF},
  subject = {microarray},
  url = {http://genome-www5.Stanford.EDU/MicroArray/SMD/SMD.pdf}
}
@article{Spellman1998Comprehensive,
  author = {Spellman, P.T. and Sherlock, G. and Zhang, M.Q. and Iyer, V.R. and
	Anders, K. and Eisen, M.B. and Brown, P.O. and Botstein, D. and Futcher,
	B.},
  title = {Comprehensive {I}dentification of {C}ell {C}ycle-regulated {G}enes
	of the {Y}east {S}accharomyces cerevisiae by {M}icroarray {H}ybridization},
  journal = {Mol. {B}iol. {C}ell},
  year = {1998},
  volume = {9},
  pages = {3273--3297},
  pdf = {../local/spel98.pdf},
  file = {spel98.pdf:local/spel98.pdf:PDF},
  subject = {microarray},
  url = {http://www.molbiolcell.org/cgi/reprint/9/12/3273.pdf}
}
@article{Statnikov2005comprehensive,
  author = {Statnikov, A. and Aliferis, C. F. and Tsamardinos, I. and Hardin,
	D. and Levy, S.},
  title = {A comprehensive evaluation of multicategory classification methods
	for microarray gene expression cancer diagnosis},
  journal = {Bioinformatics},
  year = {2005},
  note = {To appear},
  abstract = {Motivation: {C}ancer diagnosis is one of the most important emerging
	clinical applications of gene expression microarray technology. {W}e
	are seeking to develop a computer system for powerful and reliable
	cancer diagnostic model creation based on microarray data. {T}o keep
	a realistic perspective on clinical applications we focus on multicategory
	diagnosis. {I}n order to equip the system with the optimum combination
	of classifier, gene selection and cross-validation methods, we performed
	a systematic and comprehensive evaluation of several major algorithms
	for multicategory classification, several gene selection methods,
	multiple ensemble classifier methods, and two cross validation designs
	using 11 datasets spanning 74 diagnostic categories and 41 cancer
	types and 12 normal tissue types.{R}esults: {M}ulticategory {S}upport
	{V}ector {M}achines ({MC}-{SVM}s) are the most effective classifiers
	in performing accurate cancer diagnosis from gene expression data.
	{T}he {MC}-{SVM} techniques by {C}rammer and {S}inger, {W}eston and
	{W}atkins, and one-versus-rest were found to be the best methods
	in this domain. {MC}-{SVM}s outperform other popular machine learning
	algorithms such as {K}-{N}earest {N}eighbors, {B}ackpropagation and
	{P}robabilistic {N}eural {N}etworks, often to a remarkable degree.
	{G}ene selection techniques can significantly improve classification
	performance of both {MC}-{SVM}s and other non-{SVM} learning algorithms.
	{E}nsemble classifiers do not generally improve performance of the
	best non-ensemble models. {T}hese results guided the construction
	of a software system {GEMS} ({G}ene {E}xpression {M}odel {S}elector)
	that automates high-quality model construction and enforces sound
	optimization and performance estimation procedures. {T}his is the
	first such system to be informed by a rigorous comparative analysis
	of the available algorithms and datasets.{A}vailability: {T}he software
	system {GEMS} is available for download from http://www.gems-system.org
	for non-commercial use.},
  pdf = {../local/Statnikov2005comprehensive.pdf},
  file = {Statnikov2005comprehensive.pdf:local/Statnikov2005comprehensive.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {jeanphilippevert},
  url = {http://bioinformatics.oupjournals.org/cgi/content/abstract/bti033v1}
}
@article{Tavazoie1999Systematic,
  author = {Tavazoie, S. and Hughes, J. D. and Campbell, M. J. and Cho, R. J.
	and Church, G. M.},
  title = {Systematic determination of genetic network architecture},
  journal = {Nat. Genet.},
  year = {1999},
  volume = {22},
  pages = {281--285},
  doi = {doi:10.1038/10343},
  pdf = {../local/Tavazoie1999Systematic.pdf},
  file = {Tavazoie1999Systematic.pdf:local/Tavazoie1999Systematic.pdf:PDF},
  subject = {microarray},
  url = {http://dx.doi.org/10.1038/10343}
}
@article{Thukral2005Prediction,
  author = {Sushil K Thukral and Paul J Nordone and Rong Hu and Leah Sullivan
	and Eric Galambos and Vincent D Fitzpatrick and Laura Healy and Michael
	B Bass and Mary E Cosenza and Cynthia A Afshari},
  title = {Prediction of nephrotoxicant action and identification of candidate
	toxicity-related biomarkers.},
  journal = {Toxicol {P}athol},
  year = {2005},
  volume = {33},
  pages = {343-55},
  number = {3},
  abstract = {A vast majority of pharmacological compounds and their metabolites
	are excreted via the urine, and within the complex structure of the
	kidney,the proximal tubules are a main target site of nephrotoxic
	compounds. {W}e used the model nephrotoxicants mercuric chloride,
	2-bromoethylamine hydrobromide, hexachlorobutadiene, mitomycin, amphotericin,
	and puromycin to elucidate time- and dose-dependent global gene expression
	changes associated with proximal tubular toxicity. {M}ale {S}prague-{D}awley
	rats were dosed via intraperitoneal injection once daily for mercuric
	chloride and amphotericin (up to 7 doses), while a single dose was
	given for all other compounds. {A}nimals were exposed to 2 different
	doses of these compounds and kidney tissues were collected on day
	1, 3, and 7 postdosing. {G}ene expression profiles were generated
	from kidney {RNA} using 17{K} rat c{DNA} dual dye microarray and
	analyzed in conjunction with histopathology. {A}nalysis of gene expression
	profiles showed that the profiles clustered based on similarities
	in the severity and type of pathology of individual animals. {F}urther,
	the expression changes were indicative of tubular toxicity showing
	hallmarks of tubular degeneration/regeneration and necrosis. {U}se
	of gene expression data in predicting the type of nephrotoxicity
	was then tested with a support vector machine ({SVM})-based approach.
	{A} {SVM} prediction module was trained using 120 profiles of total
	profiles divided into four classes based on the severity of pathology
	and clustering. {A}lthough mitomycin {C} and amphotericin {B} treatments
	did not cause toxicity, their expression profiles were included in
	the {SVM} prediction module to increase the sample size. {U}sing
	this classifier, the {SVM} predicted the type of pathology of 28
	test profiles with 100\% selectivity and 82\% sensitivity. {T}hese
	data indicate that valid predictions could be made based on gene
	expression changes from a small set of expression profiles. {A} set
	of potential biomarkers showing a time- and dose-response with respect
	to the progression of proximal tubular toxicity were identified.
	{T}hese include several transporters ({S}lc21a2, {S}lc15, {S}lc34a2),
	{K}im 1, {IGF}bp-1, osteopontin, alpha-fibrinogen, and {G}stalpha.},
  doi = {10.1080/01926230590927230},
  keywords = {Algorithms, Animals, Antibiotics, Antineoplastic, Artificial Intelligence,
	Butadienes, Chloroplasts, Comparative Study, Computer Simulation,
	Computer-Assisted, Diagnosis, Disinfectants, Dose-Response Relationship,
	Drug, Drug Toxicity, Electrodes, Electroencephalography, Ethylamines,
	Expert Systems, Feedback, Fungicides, Gene Expression Profiling,
	Genes, Genetic Markers, Humans, Implanted, Industrial, Information
	Storage and Retrieval, Kidney, Kidney Tubules, MEDLINE, Male, Mercuric
	Chloride, Microarray Analysis, Molecular Biology, Motor Cortex, Movement,
	Natural Language Processing, Neural Networks (Computer), Non-P.H.S.,
	Non-U.S. Gov't, Plant Proteins, Predictive Value of Tests, Proteins,
	Proteome, Proximal, Puromycin Aminonucleoside, Rats, Reproducibility
	of Results, Research Support, Sprague-Dawley, Subcellular Fractions,
	Terminology, Therapy, Time Factors, Toxicogenetics, U.S. Gov't, User-Computer
	Interface, 15805072},
  pii = {X3U2206L2747H31G},
  url = {http://dx.doi.org/10.1080/01926230590927230}
}
@article{Tothill2005expression-based,
  author = {Richard W Tothill and Adam Kowalczyk and Danny Rischin and Alex Bousioutas
	and Izhak Haviv and Ryan K van Laar and Paul M Waring and John Zalcberg
	and Robyn Ward and Andrew V Biankin and Robert L Sutherland and Susan
	M Henshall and Kwun Fong and Jonathan R Pollack and David D L Bowtell
	and Andrew J Holloway},
  title = {An expression-based site of origin diagnostic method designed for
	clinical application to cancer of unknown origin.},
  journal = {Cancer {R}es.},
  year = {2005},
  volume = {65},
  pages = {4031-40},
  number = {10},
  month = {May},
  abstract = {Gene expression profiling offers a promising new technique for the
	diagnosis and prognosis of cancer. {W}e have applied this technology
	to build a clinically robust site of origin classifier with the ultimate
	aim of applying it to determine the origin of cancer of unknown primary
	({CUP}). {A} single c{DNA} microarray platform was used to profile
	229 primary and metastatic tumors representing 14 tumor types and
	multiple histologic subtypes. {T}his data set was subsequently used
	for training and validation of a support vector machine ({SVM}) classifier,
	demonstrating 89\% accuracy using a 13-class model. {F}urther, we
	show the translation of a five-class classifier to a quantitative
	{PCR}-based platform. {S}electing 79 optimal gene markers, we generated
	a quantitative-{PCR} low-density array, allowing the assay of both
	fresh-frozen and formalin-fixed paraffin-embedded ({FFPE}) tissue.
	{D}ata generated using both quantitative {PCR} and microarray were
	subsequently used to train and validate a cross-platform {SVM} model
	with high prediction accuracy. {F}inally, we applied our {SVM} classifiers
	to 13 cases of {CUP}. {W}e show that the microarray {SVM} classifier
	was capable of making high confidence predictions in 11 of 13 cases.
	{T}hese predictions were supported by comprehensive review of the
	patients' clinical histories.},
  doi = {10.1158/0008-5472.CAN-04-3617},
  pdf = {../local/Tothill2005expression-based.pdf},
  file = {Tothill2005expression-based.pdf:Tothill2005expression-based.pdf:PDF},
  keywords = {biosvm microarray},
  pii = {65/10/4031},
  url = {http://dx.doi.org/10.1158/0008-5472.CAN-04-3617}
}
@article{Tsai2004Gene,
  author = {Tsai, C.A. and Chen, C.H. and Lee, T.C. and Ho, I.C. and Yang, U.C.
	and Chen, J.J.},
  title = {Gene selection for sample classifications in microarray experiments.},
  journal = {D{NA} {C}ell {B}iol.},
  year = {2004},
  volume = {23},
  pages = {607-614},
  number = {10},
  abstract = {D{NA} microarray technology provides useful tools for profiling global
	gene expression patterns in different cell/tissue samples. {O}ne
	major challenge is the large number of genes relative to the number
	of samples. {T}he use of all genes can suppress or reduce the performance
	of a classification rule due to the noise of nondiscriminatory genes.
	{S}election of an optimal subset from the original gene set becomes
	an important prestep in sample classification. {I}n this study, we
	propose a family-wise error ({FWE}) rate approach to selection of
	discriminatory genes for two-sample or multiple-sample classification.
	{T}he {FWE} approach controls the probability of the number of one
	or more false positives at a prespecified level. {A} public colon
	cancer data set is used to evaluate the performance of the proposed
	approach for the two classification methods: k nearest neighbors
	(k-{NN}) and support vector machine ({SVM}). {T}he selected gene
	sets from the proposed procedure appears to perform better than or
	comparable to several results reported in the literature using the
	univariate analysis without performing multivariate search. {I}n
	addition, we apply the {FWE} approach to a toxicogenomic data set
	with nine treatments (a control and eight metals, {A}s, {C}d, {N}i,
	{C}r, {S}b, {P}b, {C}u, and {A}s{V}) for a total of 55 samples for
	a multisample classification. {T}wo gene sets are considered: the
	gene set omega{F} formed by the {ANOVA} {F}-test, and a gene set
	omega{T} formed by the union of one-versus-all t-tests. {T}he predicted
	accuracies are evaluated using the internal and external crossvalidation.
	{U}sing the {SVM} classification, the overall accuracies to predict
	55 samples into one of the nine treatments are above 80% for internal
	crossvalidation. {O}mega{F} has slightly higher accuracy rates than
	omega{T}. {T}he overall predicted accuracies are above 70% for the
	external crossvalidation; the two gene sets omega{T} and omega{F}
	performed equally well.},
  doi = {10.1089/1044549042476947},
  pdf = {../local/Tsai2004Gene.pdf},
  file = {Tsai2004Gene.pdf:local/Tsai2004Gene.pdf:PDF},
  keywords = {biosvm microarray},
  owner = {jeanphilippevert},
  url = {http://dx.doi.org/10.1089/1044549042476947}
}
@article{Wang2005Gene-expression,
  author = {Wang, Y. and Klijn, J.G.M. and Zhang, Y. and Sieuwerts, A.M. and
	Look, M.P. and Yang, F. and Talantov, D. and Timmermans, M. and Meijer-van
	Gelder, M.E. and Yu, J. and Jatkoe, T. and Berns, E.M.J.J. and Atkins,
	D. and Foekens, J.A.},
  title = {Gene-expression profiles to predict distant metastasis of lymph-node-negative
	primary breast cancers},
  journal = {Lancet},
  year = {2005},
  volume = {365},
  pages = {671--679},
  number = {9460},
  abstract = {BACKGROUND: Genome-wide measures of gene expression can identify patterns
	of gene activity that subclassify tumours and might provide a better
	means than is currently available for individual risk assessment
	in patients with lymph-node-negative breast cancer. METHODS: We analysed,
	with Affymetrix Human U133a GeneChips, the expression of 22000 transcripts
	from total RNA of frozen tumour samples from 286 lymph-node-negative
	patients who had not received adjuvant systemic treatment. FINDINGS:
	In a training set of 115 tumours, we identified a 76-gene signature
	consisting of 60 genes for patients positive for oestrogen receptors
	(ER) and 16 genes for ER-negative patients. This signature showed
	93\% sensitivity and 48\% specificity in a subsequent independent
	testing set of 171 lymph-node-negative patients. The gene profile
	was highly informative in identifying patients who developed distant
	metastases within 5 years (hazard ratio 5.67 [95\% CI 2.59-12.4]),
	even when corrected for traditional prognostic factors in multivariate
	analysis (5.55 [2.46-12.5]). The 76-gene profile also represented
	a strong prognostic factor for the development of metastasis in the
	subgroups of 84 premenopausal patients (9.60 [2.28-40.5]), 87 postmenopausal
	patients (4.04 [1.57-10.4]), and 79 patients with tumours of 10-20
	mm (14.1 [3.34-59.2]), a group of patients for whom prediction of
	prognosis is especially difficult. INTERPRETATION: The identified
	signature provides a powerful tool for identification of patients
	at high risk of distant recurrence. The ability to identify patients
	who have a favourable prognosis could, after independent confirmation,
	allow clinicians to avoid adjuvant systemic therapy or to choose
	less aggressive therapeutic options.},
  doi = {10.1016/S0140-6736(05)17947-1},
  pdf = {../local/Wang2005Gene-expression.pdf},
  file = {Wang2005Gene-expression.pdf:local/Wang2005Gene-expression.pdf:PDF},
  keywords = {microarray, breastcancer},
  owner = {jp},
  pii = {S0140673605179471},
  pmid = {15894094},
  timestamp = {2006.07.06},
  url = {http://dx.doi.org/10.1016/S0140-6736(05)17947-1}
}
@article{Wang2005Gene,
  author = {Yu Wang and Igor V Tetko and Mark A Hall and Eibe Frank and Axel
	Facius and Klaus F X Mayer and Hans W Mewes},
  title = {Gene selection from microarray data for cancer classification--a
	machine learning approach.},
  journal = {Comput. {B}iol. {C}hem.},
  year = {2005},
  volume = {29},
  pages = {37-46},
  number = {1},
  month = {Feb},
  abstract = {A {DNA} microarray can track the expression levels of thousands of
	genes simultaneously. {P}revious research has demonstrated that this
	technology can be useful in the classification of cancers. {C}ancer
	microarray data normally contains a small number of samples which
	have a large number of gene expression levels as features. {T}o select
	relevant genes involved in different types of cancer remains a challenge.
	{I}n order to extract useful gene information from cancer microarray
	data and reduce dimensionality, feature selection algorithms were
	systematically investigated in this study. {U}sing a correlation-based
	feature selector combined with machine learning algorithms such as
	decision trees, naïve {B}ayes and support vector machines, we show
	that classification performance at least as good as published results
	can be obtained on acute leukemia and diffuse large {B}-cell lymphoma
	microarray data sets. {W}e also demonstrate that a combined use of
	different classification and feature selection approaches makes it
	possible to select relevant genes with high confidence. {T}his is
	also the first paper which discusses both computational and biological
	evidence for the involvement of zyxin in leukaemogenesis.},
  doi = {10.1016/j.compbiolchem.2004.11.001},
  pdf = {../local/Wang2005Gene.pdf},
  file = {Wang2005Gene.pdf:local/Wang2005Gene.pdf:PDF},
  keywords = {biosvm microarray},
  pii = {S1476-9271(04)00108-2},
  url = {http://dx.doi.org/10.1016/j.compbiolchem.2004.11.001}
}
@article{Wirapati2008Meta-analysis,
  author = {Wirapati, P. and Sotiriou, C. and Kunkel, S. and Farmer, P. and Pradervand,
	S. and Haibe-Kains, B. and Desmedt, C. and Ignatiadis, M. and Sengstag,
	T. and Sch\"utz, F. and Goldstein, D. R. and Piccart, M. and Delorenzi,
	M.},
  title = {Meta-analysis of gene expression profiles in breast cancer: toward
	a unified understanding of breast cancer subtyping and prognosis
	signatures.},
  journal = {Breast Cancer Res.},
  year = {2008},
  volume = {10},
  pages = {R65},
  number = {4},
  abstract = {INTRODUCTION: Breast cancer subtyping and prognosis have been studied
	extensively by gene expression profiling, resulting in disparate
	signatures with little overlap in their constituent genes. Although
	a previous study demonstrated a prognostic concordance among gene
	expression signatures, it was limited to only one dataset and did
	not fully elucidate how the different genes were related to one another
	nor did it examine the contribution of well-known biological processes
	of breast cancer tumorigenesis to their prognostic performance. METHOD:
	To address the above issues and to further validate these initial
	findings, we performed the largest meta-analysis of publicly available
	breast cancer gene expression and clinical data, which are comprised
	of 2,833 breast tumors. Gene coexpression modules of three key biological
	processes in breast cancer (namely, proliferation, estrogen receptor
	[ER], and HER2 signaling) were used to dissect the role of constituent
	genes of nine prognostic signatures. RESULTS: Using a meta-analytical
	approach, we consolidated the signatures associated with ER signaling,
	ERBB2 amplification, and proliferation. Previously published expression-based
	nomenclature of breast cancer 'intrinsic' subtypes can be mapped
	to the three modules, namely, the ER-/HER2- (basal-like), the HER2+
	(HER2-like), and the low- and high-proliferation ER+/HER2- subtypes
	(luminal A and B). We showed that all nine prognostic signatures
	exhibited a similar prognostic performance in the entire dataset.
	Their prognostic abilities are due mostly to the detection of proliferation
	activity. Although ER- status (basal-like) and ERBB2+ expression
	status correspond to bad outcome, they seem to act through elevated
	expression of proliferation genes and thus contain only indirect
	information about prognosis. Clinical variables measuring the extent
	of tumor progression, such as tumor size and nodal status, still
	add independent prognostic information to proliferation genes. CONCLUSION:
	This meta-analysis unifies various results of previous gene expression
	studies in breast cancer. It reveals connections between traditional
	prognostic factors, expression-based subtyping, and prognostic signatures,
	highlighting the important role of proliferation in breast cancer
	prognosis.},
  doi = {10.1186/bcr2124},
  pdf = {../local/Wirapati2008Meta-analysis.pdf},
  file = {Wirapati2008Meta-analysis.pdf:Wirapati2008Meta-analysis.pdf:PDF},
  institution = {Swiss Institute of Bioinformatics, 'Batiment Genopode', University
	of Lausanne, 1015 Lausanne, Switzerland. Pratyaksha.Wirapati@isb-sib.ch},
  keywords = {microarray, breastcancer},
  language = {eng},
  medline-pst = {ppublish},
  owner = {jp},
  pii = {bcr2124},
  pmid = {18662380},
  timestamp = {2010.10.13},
  url = {http://dx.doi.org/10.1186/bcr2124}
}
@article{Zhou2005LS,
  author = {Xin Zhou and K. Z. Mao},
  title = {L{S} {B}ound based gene selection for {DNA} microarray data.},
  journal = {Bioinformatics},
  year = {2005},
  volume = {21},
  pages = {1559-64},
  number = {8},
  month = {Apr},
  abstract = {M{OTIVATION}: {O}ne problem with discriminant analysis of {DNA} microarray
	data is that each sample is represented by quite a large number of
	genes, and many of them are irrelevant, insignificant or redundant
	to the discriminant problem at hand. {M}ethods for selecting important
	genes are, therefore, of much significance in microarray data analysis.
	{I}n the present study, a new criterion, called {LS} {B}ound measure,
	is proposed to address the gene selection problem. {T}he {LS} {B}ound
	measure is derived from leave-one-out procedure of {LS}-{SVM}s (least
	squares support vector machines), and as the upper bound for leave-one-out
	classification results it reflects to some extent the generalization
	performance of gene subsets. {RESULTS}: {W}e applied this {LS} {B}ound
	measure for gene selection on two benchmark microarray datasets:
	colon cancer and leukemia. {W}e also compared the {LS} {B}ound measure
	with other evaluation criteria, including the well-known {F}isher's
	ratio and {M}ahalanobis class separability measure, and other published
	gene selection algorithms, including {W}eighting factor and {SVM}
	{R}ecursive {F}eature {E}limination. {T}he strength of the {LS} {B}ound
	measure is that it provides gene subsets leading to more accurate
	classification results than the filter method while its computational
	complexity is at the level of the filter method. {AVAILABILITY}:
	{A} companion website can be accessed at http://www.ntu.edu.sg/home5/pg02776030/lsbound/.
	{T}he website contains: (1) the source code of the gene selection
	algorithm; (2) the complete set of tables and figures regarding the
	experimental study; (3) proof of the inequality (9). {CONTACT}: ekzmao@ntu.edu.sg.},
  doi = {10.1093/bioinformatics/bti216},
  pdf = {../local/Zhou2005LS.pdf},
  file = {Zhou2005LS.pdf:local/Zhou2005LS.pdf:PDF},
  keywords = {biosvm featureselection microarray},
  pii = {bti216},
  url = {http://dx.doi.org/10.1093/bioinformatics/bti216}
}
@article{Zhu2000Two,
  author = {Zhu, G. and Spellman, P. T. and Volpe, T. and Brown, P. O. and Botstein,
	D. and Davis, T. N. and Futcher, B.},
  title = {Two yeast forkhead genes regulate the cell cycle and pseudohyphal
	growth},
  journal = {Nature},
  year = {2000},
  volume = {406},
  pages = {90--94},
  pdf = {../local/zhu00.pdf},
  file = {zhu00.pdf:local/zhu00.pdf:PDF},
  subject = {microarray},
  url = {http://www.nature.com/cgi-taf/DynaPage.taf?file=/nature/journal/v406/n6791/full/406090a0_fs.html&content_filetype=pdf}
}
@comment{{jabref-meta: selector_author:}}
@comment{{jabref-meta: selector_journal:Adv. Drug Deliv. Rev.;Am. J. Hu
m. Genet.;Am. J. Pathol.;Ann. Appl. Stat.;Ann. Math. Statist.;Ann. N. 
Y. Acad. Sci.;Ann. Probab.;Ann. Stat.;Artif. Intell. Med.;Bernoulli;Bi
ochim. Biophys. Acta;Bioinformatics;Biometrika;BMC Bioinformatics;Br. 
J. Pharmacol.;Breast Cancer Res.;Cell;Cell. Signal.;Chem. Res. Toxicol
.;Clin. Cancer Res.;Combinator. Probab. Comput.;Comm. Pure Appl. Math.
;Comput. Chem.;Comput. Comm. Rev.;Comput. Stat. Data An.;Curr. Genom.;
Curr. Opin. Chem. Biol.;Curr. Opin. Drug Discov. Devel.;Data Min. Know
l. Discov.;Electron. J. Statist.;Eur. J. Hum. Genet.;FEBS Lett.;Found.
 Comput. Math.;Genome Biol.;IEEE T. Neural Networ.;IEEE T. Pattern. An
al.;IEEE T. Signal. Proces.;IEEE Trans. Inform. Theory;IEEE Trans. Kno
wl. Data Eng.;IEEE/ACM Trans. Comput. Biol. Bioinf.;Int. J. Comput. Vi
sion;Int. J. Data Min. Bioinform.;Int. J. Qantum Chem.;J Biol Syst;J. 
ACM;J. Am. Soc. Inf. Sci. Technol.;J. Am. Stat. Assoc.;J. Bioinform. C
omput. Biol.;J. Biol. Chem.;J. Biomed. Inform.;J. Cell. Biochem.;J. Ch
em. Inf. Comput. Sci.;J. Chem. Inf. Model.;J. Clin. Oncol.;J. Comput. 
Biol.;J. Comput. Graph. Stat.;J. Eur. Math. Soc.;J. Intell. Inform. Sy
st.;J. Mach. Learn. Res.;J. Med. Chem.;J. Mol. BIol.;J. R. Stat. Soc. 
Ser. B;Journal of Statistical Planning and Inference;Mach. Learn.;Math
. Program.;Meth. Enzymol.;Mol. Biol. Cell;Mol. Biol. Evol.;Mol. Cell. 
Biol.;Mol. Syst. Biol.;N. Engl. J. Med.;Nat. Biotechnol.;Nat. Genet.;N
at. Med.;Nat. Methods;Nat. Rev. Cancer;Nat. Rev. Drug Discov.;Nat. Rev
. Genet.;Nature;Neural Comput.;Neural Network.;Neurocomputing;Nucleic 
Acids Res.;Pattern Anal. Appl.;Pattern Recognit.;Phys. Rev. E;Phys. Re
v. Lett.;PLoS Biology;PLoS Comput. Biol.;Probab. Theory Relat. Fields;
Proc. IEEE;Proc. Natl. Acad. Sci. USA;Protein Eng.;Protein Eng. Des. S
el.;Protein Sci.;Protein. Struct. Funct. Genet.;Random Struct. Algorit
hm.;Rev. Mod. Phys.;Science;Stat. Probab. Lett.;Statistica Sinica;Theo
r. Comput. Sci.;Trans. Am. Math. Soc.;Trends Genet.;}}
@comment{{jabref-meta: selector_keywords:biogm;biosvm;breastcancer;cgh;
chemogenomics;chemoinformatics;csbcbook;csbcbook-ch1;csbcbook-ch2;csbc
book-ch3;csbcbook-ch4;csbcbook-ch5;csbcbook-ch6;csbcbook-ch7;csbcbook-
ch8;csbcbook-ch9;csbcbook-mustread;dimred;featureselection;glycans;her
g;hic;highcontentscreening;image;immunoinformatics;kernel-theory;kerne
lbook;lasso;microarray;ngs;nlp;plasmodium;proteomics;PUlearning;rnaseq
;segmentation;sirna;}}
@comment{{jabref-meta: selector_booktitle:Adv. Neural. Inform. Process 
Syst.;}}

This file was generated by bibtex2html 1.97.