have mutations in both . This process of REVIEW mutation followed by clonal expansion contin- ues, with mutations in genes such as PIK3CA, SMAD4,andTP53,eventuallygeneratingama- Cancer Genome Landscapes lignant tumor that can invade through the under- lying basement membrane and metastasize to Bert Vogelstein, Nickolas Papadopoulos, Victor E. Velculescu, Shibin Zhou, lymph nodes and distant organs such as the Luis A. Diaz Jr., Kenneth W. Kinzler* liver (11). The mutations that confer a selec- tive growth advantage to the tumor cell are called Over the past decade, comprehensive sequencing efforts have revealed the genomic landscapes “driver” mutations. It has been estimated (12) of common forms of human cancer. For most cancer types, this landscape consists of a small that each driver mutation provides only a small number of “mountains” (genes altered in a high percentage of tumors) and a much larger number selective growth advantage to the cell, on the of “hills” (genes altered infrequently). To date, these studies have revealed ~140 genes that, order of a 0.4% increase in the difference be- when altered by intragenic mutations, can promote or “drive” tumorigenesis. A typical tumor tween cell birth and cell death. Over many years, contains two to eight of these “driver ” mutations; the remaining mutations are passengers however, this slight increase, compounded once that confer no selective growth advantage. Driver genes can be classified into 12 signaling or twice per week, can result in a large mass, pathways that regulate three core cellular processes: cell fate, cell survival, and genome containing billions of cells. maintenance. A better understanding of these pathways is one of the most pressing needs in basic The number of mutations in certain tumors of cancer research. Even now, however, our knowledge of cancer genomes is sufficient to guide self-renewing tissues is directly correlated with the development of more effective approaches forreducingcancermorbidity and mortality. age (13). When evaluated through linear regres- sion, this correlation implies that more than half en years ago, the idea that all of the genes Certain tumor types display many more or of the somatic mutations identified in these tu- altered in cancer could be identified at many fewer mutations than average (Fig. 1B). mors occur during the preneoplastic phase; that base-pair resolution would have seemed Notable among these outliers are melanomas is, during the growth of normal cells that con- T on April 30, 2013 like science fiction. Today, such genome-wide and lung tumors, which contain ~200 nonsyn- tinuously replenish gastrointestinal and genito- analysis, through sequencing of the exome (see onymous mutations per tumor (table S1C). These urinary epithelium and other tissues. All of these Box 1, Glossary, for definitions of terms used in larger numbers reflect the involvement of potent pre-neoplastic mutations are “passenger” muta- this Review) or of the whole genome, is routine. mutagens (ultraviolet light and cigarette smoke, tions that have no effect on the neoplastic pro- The prototypical exomic studies of cancer respectively) in the pathogenesis of these tumor cess. This result explains why a colorectal tumor evaluated ~20 tumors at a cost of >$100,000 per types. Accordingly, lung cancers from smokers in a 90-year-old patient has nearly twice as many case (1–3). Today, the cost of this sequencing have 10 times as many somatic mutations as mutations as a morphologically identical colorec- has been reduced 100-fold, and studies reporting those from nonsmokers (4). Tumors with defects tal tumor in a 45-year-old patient. This finding the sequencing of more than 100 tumors of a in DNA repair form another group of outliers also partly explains why advanced brain tumors given type are the norm (table S1A). Although (5). For example, tumors with mismatch repair (glioblastomas) and pancreatic cancers (pancre- www.sciencemag.org vast amounts of data can now be readily ob- defects can harbor thousands of mutations (Fig. atic ductal adenocarcinomas) have fewer mu- tained, deciphering this information in meaning- 1B), even more than lung tumors or melanomas. tations than colorectal tumors; glial cells of ful terms is still challenging. Here, we review Recent studies have shown that high numbers the brain and epithelial cells of the pancreatic what has been learned about cancer genomes of mutations are also found in tumors with ducts do not replicate, unlike the epithelial cells from these sequencing studies—and, more im- genetic alterations of the proofreading domain lining the crypts of the colon. Therefore, the gate- portantly, what this information has taught us of DNA polymerases POLE or POLD1 (6, 7). keeping mutation in a pancreatic or brain can-

about cancer biology and future cancer manage- At the other end of the spectrum, pediatric tu- cer is predicted to occur in a precursor cell that Downloaded from ment strategies. mors and leukemias harbor far fewer point mu- contains many fewer mutations than are present tations: on average, 9.6 per tumor (table S1C). The in a colorectal precursor cell. This line of rea- How Many Genes Are Subtly Mutated basis for this observation is considered below. soning also helps to explain why pediatric can- in a Typical Human Cancer? cers have fewer mutations than adult tumors. In common solid tumors such as those derived Mutation Timing Pediatric cancers often occur in non–self-renewing from the colon, breast, brain, or pancreas, an When do these mutations occur? Tumors evolve tissues, and those that arise in renewing tissues average of 33 to 66 genes display subtle somatic from benign to malignant lesions by acquiring (such as leukemias) originate from precursor mutations that would be expected to alter their aseriesofmutationsovertime,aprocessthat cells that have not renewed themselves as often products (Fig. 1A). About 95% of these has been particularly well studied in colorectal as in adults. In addition, pediatric tumors, as well mutations are single-base substitutions (such as tumors (8, 9). The first, or “gatekeeping,” mu- as adult leukemias and lymphomas, may require C>G), whereas the remainder are deletions or tation provides a selective growth advantage fewer rounds of clonal expansion than adult solid insertions of one or a few bases (such as CTT>CT) to a normal epithelial cell, allowing it to out- tumors (8, 14). Genome sequencing studies of (table S1B). Of the base substitutions, 90.7% re- grow the cells that surround it and become a leukemia patients support the idea that muta- sult in missense changes, 7.6% result in nonsense microscopic clone (Fig. 2). Gatekeeping muta- tions occur as random events in normal precur- changes, and 1.7% result in alterations of splice tions in the colon most often occur in the APC sor cells before these cells acquire an initiating sites or untranslated regions immediately adjacent gene (10). The small adenoma that results from mutation (15). to the start and stop codons (table S1B). this mutation grows slowly, but a second mu- When during tumorigenesis do the remaining tation in another gene, such as KRAS,unleashes somatic mutations occur? Because mutations in The Ludwig Center and The Howard Hughes Medical Institute a second round of clonal growth that allows tumors occur at predictable and calculable rates at Johns Hopkins Kimmel Cancer Center, Baltimore, MD an expansion of cell number (9). The cells with (see below), the number of somatic mutations in 21287, USA. only the APC mutation may persist, but their cell tumors provides a clock, much like the clock *Corresponding author. E-mail: [email protected] numbers are small compared with the cells that used in evolutionary biology to determine species

1546 29 MARCH 2013 VOL 339 SCIENCE www.sciencemag.org SPECIALSECTION

divergence time. The number of mutations has been measured in tumors representing progressive A stages of colorectal and pancreatic cancers (11, 16). Applying the evolutionary clock model to these data leads to two unambiguous conclusions: First, it takes decades to develop a full-blown, meta- static cancer. Second, virtually all of the mutations in metastatic lesions were already present in a large number of cells in the primary tumors. The timing of mutations is relevant to our understanding of metastasis, which is responsible for the death of most patients with cancer. The primary tumor can be surgically removed, but the residual metastatic lesions—often undetectable and widespread—remain and eventually enlarge, com- promising the function of the lungs, liver, or other organs. From a genetics perspective, it would seem that there must be mutations that convert a primary cancer to a metastatic one, just as there are mutations that convert a normal cell to a be- B 1500 nign tumor, or a benign tumor to a malignant one 1000 (Fig. 2). Despite intensive effort, however, con- sistent genetic alterations that distinguish cancers 500 that metastasize from cancers that have not yet 250 on April 30, 2013 metastasized remain to be identified. One potential explanation invokes mutations 225 or epigenetic changes that are difficult to iden- tify with current technologies (see section on “dark 200 matter” below). Another explanation is that meta- static lesions have not yet been studied in suf- 175 ficient detail to identify these genetic alterations, particularly if the mutations are heterogeneous 150 in nature. But another possible explanation is that there are no metastasis genes. A malignant www.sciencemag.org 125 primary tumor can take many years to metasta- size, but this process is, in principle, explicable 100 by stochastic processes alone (17, 18). Advanced

(median +/- one quartile) 75 tumors release millions of cells into the circula- tion each day, but these cells have short half-lives,

50 and only a miniscule fraction establish metastatic Non-synonymous mutations per tumor

lesions (19). Conceivably, these circulating cells Downloaded from 25 may, in a nondeterministic manner, infrequently and randomly lodge in a capillary bed in an organ 0 that provides a favorable microenvironment for growth. The bigger the primary tumor mass, the more likely that this process will occur. In this Breast Gastric

Prostate scenario, the continual evolution of the primary Rhabdoid Melanoma tumor would reflect local selective advantages Glioblastoma Glioblastoma Lung (SCLC) Lung Hepatocellular Lung (NSCLC) Lung Head and neck and Head Neuroblastoma rather than future selective advantages. The idea Colorectal (MSI) Medulloblastoma Colorectal (MSS)

Esophageal (EAC) that growth at metastatic sites is not dependent on Esophageal (ESCC) Endometrial (serous) additional genetic alterations is also supported by Acute myeloid leukemia Acute myeloid Non-Hodgkin lymphoma recent results showing that even normal cells, Endometrial (endometrioid) Pancreatic adenocarcinoma Pancreatic Ovarian (high-grade serous) (high-grade Ovarian

Lung (never smoked NSCLC) smoked (never Lung when placed in suitable environments such as Acute lymphoblastic leukemia lymphoblastic Acute Chronic lymphocytic leukemia lymph nodes, can grow into organoids, complete Mutagens Adult solid tumors Liquid Pediatric with a functioning vasculature (20). Fig. 1. Number of somatic mutations in representative human cancers, detected by genome- Other Types of Genetic Alterations in Tumors wide sequencing studies. (A)Thegenomesofadiversegroupofadult (right) and pediatric (left) cancers have been analyzed. Numbers in parentheses indicate the median number of nonsynonymous Though the rate of point mutations in tumors is mutations per tumor. (B)Themediannumberofnonsynonymousmutationspertumorinavarietyof similar to that of normal cells, the rate of chro- tumor types. Horizontal bars indicate the 25 and 75% quartiles. MSI, microsatellite instability; SCLC, mosomal changes in cancer is elevated (21). small cell lung cancers; NSCLC, non–small cell lung cancers; ESCC, esophageal squamous cell carcinomas; Therefore, most solid tumors display widespread MSS, microsatellite stable; EAC, esophageal adenocarcinomas. The published data on which this figure is changes in number (aneuploidy),

CREDIT: FIG. 1A, E. COOK based are provided in table S1C. as well as deletions, inversions, translocations,

www.sciencemag.org SCIENCE VOL 339 29 MARCH 2013 1547 Fig. 2. Genetic alterations and the progression of colorectal cancer. nents of these pathways can be altered in any individual tumor. Patient age indicates The major signaling pathways that drive tumorigenesis are shown at the transi- the time intervals during which the driver genes are usually mutated. Note that tions between each tumor stage. One of several driver genes that encode compo- this model may not apply to all tumor types. TGF-b,transforminggrowthfactor–b.

and other genetic abnormalities. When a large mutations, as well as the numerous epigenetic those mutations that truncate the encoded protein part of a chromosome is duplicated or deleted, it changes found in cancers, will be discussed later. within its N-terminal 1600 amino acids are driver is difficult to identify the specific “target” gene(s) gene mutations. Missense mutations throughout on the chromosome whose gain or loss confers a Drivers Versus Passenger Mutations the gene, as well as protein-truncating mutations in growth advantage to the tumor cell. Target genes Though it is easy to define a “driver gene muta- the C-terminal 1200 amino acids, are passenger on April 30, 2013 are more easily identified in the case of chro- tion” in physiologic terms (as one conferring a gene mutations. mosome translocations, homozygous deletions, selective growth advantage), it is more difficult Numerous statistical methods to identify driver and gene amplifications. Translocations generally to identify which somatic mutations are drivers genes have been described. Some are based on fuse two genes to create an oncogene (such as and which are passengers. Moreover, it is im- the frequency of mutations in an individual gene BCR-ABL in chronic myelogenous leukemia) but, portant to point out that there is a fundamental compared with the mutation frequency of other in a small number of cases, can inactivate a tumor difference between a driver gene and a driver genes in the same or related tumors after correc- suppressor gene by truncating it or separating it gene mutation. A driver gene is one that con- tion for sequence context and gene size (22, 23). from its promoter. Homozygous deletions often tains driver gene mutations. But driver genes Other methods are based on the predicted effects involve just one or a few genes, and the target is may also contain passenger gene mutations. For of mutation on the encoded protein, as inferred always a tumor suppressor gene. Amplifications example, APC is a large driver gene, but only from biophysical studies (24–26). All of these www.sciencemag.org contain an oncogene whose protein product is methods are useful for prioritiz- abnormally active simply because the tumor ing genes that are most likely cell contains 10 to 100 copies of the gene per to promote a selective growth ad- cell, compared with the two copies present in Translocations vantage when mutated. When normal cells. 80.0 Deletions the number of mutations in a gene Most solid tumors have dozens of translo- 70.0 Amplifications is very high, as with TP53 or

cations; however, as with point mutations, the Indels KRAS,anyreasonablestatistic Downloaded from majority of translocations appear to be passen- 60.0 SBS will indicate that the gene is ex- gers rather than drivers. The breakpoints of the 50.0 tremely likely to be a driver gene. translocations are often in “gene deserts” devoid These highly mutated genes have 40.0 of known genes, and many of the translocations been termed “mountains” (1). Un- and homozygous deletions are adjacent to frag- 30.0 fortunately, however, genes with ile sites that are prone to breakage. Cancer cells 20.0 more than one, but still relatively can, perhaps, survive such chromosome breaks few mutations (so called “hills”) more easily than normal cells because they con- 10.0 numerically dominate cancer ge- Number of alterations per tumor tain mutations that incapacitate genes like TP53, nome landscapes (1). In these which would normally respond to DNA damage cases, methods based on muta- by triggering cell death. Studies to date indicate tion frequency and context alone that there are roughly 10 times fewer genes af- cannot reliably indicate which Breast cancer Glioblastoma fected by chromosomal changes than by point genes are drivers, because the Colorectal cancer Pancreatic cancer Medulloblastoma mutations. Figure 3 shows the types and distri- background rates of mutation bution of genetic alterations that affect protein- Fig. 3. Total alterations affecting protein-coding genes in vary so much among different pa- coding genes in five representative tumor types. selected tumors. Average number and types of genomic altera- tients and regions of the genome. Protein-coding genes account for only ~1.5% of tions per tumor, including single-base substitutions (SBS), small Recent studies of normal cells the total genome, and the number of alterations insertions and deletions (indels), amplifications, and homozygous have indicated that the rate of in noncoding regions is proportionately higher deletions, as determined by genome-wide sequencing studies. For mutation varies by more than than the number affecting coding regions. The colorectal, breast, and pancreatic ductal cancer, and medulloblastomas, 100-fold within the genome (27). vast majority of the alterations in noncoding re- translocations are also included. The published data on which this In tumor cells, this variation can

gions are presumably passengers. These noncoding figure is based are provided in table S1D. be higher and may affect whole CREDIT: FIG. 2, E. COOK

1548 29 MARCH 2013 VOL 339 SCIENCE www.sciencemag.org SPECIALSECTION

Box 1. Glossary

Adenoma: Abenigntumorcomposedofepithelialcells. Human leukocyte antigen (HLA): Aproteinencodedby Passenger mutation (passenger): A mutation that genes that determine an individual’scapacitytorespondto has no direct or indirect effect on the selective growth Alternative lengthening of telomeres (ALT): Aprocess specific antigens or reject transplants from other individuals. advantage of the cell in which it occurred. of maintaining telomeres independent of telomerase, the enzyme normally responsible for telomere replication. Homozygous deletion: Deletion of both copies of a Primary tumor: The original tumor at the site where gene segment (the one inherited from the mother, as tumor growth was initiated. This can be defined for solid Amplification: Ageneticalterationproducingalarge well as that inherited from the father). tumors, but not for liquid tumors. number of copies of a small segment (less than a few Indel: Amutationduetosmallinsertionordeletionof megabases) of the genome. Promoter: A region within or near the gene that one or a few nucleotides. helps regulate its expression. Angiogenesis: the process of forming vascular con- Karyotype: Display of the of a cell on a duits, including veins, arteries, and lymphatics. Rearrangement: A mutation that juxtaposes nucleo- microscopic slide, used to evaluate changes in chromosome tides that are normally separated, such as those on two number as well as structural alterations of chromosomes. Benign tumor: An abnormal proliferation of cells different chromosomes. driven by at least one mutation in an oncogene or tumor Kinase: Aproteinthatcatalyzestheadditionofphos- suppressor gene. These cells are not invasive (i.e., they Selective growth advantage (s): The difference between phate groups to other molecules, such as or cannot penetrate the basementmembraneliningthem), birth and death in a cell population. In normal adult lipids. These proteins are essential to nearly all signal which distinguishes them from malignant cells. cells in the absence of injury, s = 0.000000. transduction pathways.

Carcinoma: A type of malignant tumor composed of Liquid tumors: Tumors composed of hematopoietic (blood) Self-renewing tissues: Tissues whose cells normally on April 30, 2013 epithelial cells. cells, such as leukemias. Though lymphomas generally form repopulate themselves, such as those lining the solid masses in lymph nodes, they are often classified as gastrointestinal or urogenital tracts, as well as blood Clonal mutation: Amutationthatexistsinthevast liquid tumors because of their derivation from hemato- cells. majority of the neoplastic cells within a tumor. poietic cells and ability to travel through lymphatics. Single-base substitution (SBS): Asingle-nucleotide Driver gene mutation (driver): A mutation that Malignant tumor: An abnormal proliferation of cells substitution (e.g., C to T) relative to a reference sequence directly or indirectly confers a selective growth advantage driven by mutations in oncogenes or tumor suppressor or, in the case of somatic mutations, relative to the to the cell in which it occurs. genes that has already invaded their surrounding stroma. germline genome of the person with a tumor. It is impossible to distinguish an isolated benign tumor cell www.sciencemag.org Driver gene: Agenethatcontainsdrivergenemutations from an isolated malignant tumor cell. This distinction can Solid tumors: Tumors that form discrete masses, such (Mut-Driver gene) or is expressed aberrantly in a fashion be made only through examination of tissue architecture. as carcinomas or sarcomas. that confers a selective growth advantage (Epi-Driver gene). Metastatic tumor: A malignant tumor that has migrated Somatic mutations: Mutations that occur in any non– Epi-driver gene: Agenethatisexpressedaberrantlyin away from its primary site, such as to draining lymph germ cell of the body after conception, such as those that cancers in a fashion that confers a selective growth advantage. nodes or another organ. initiate tumorigenesis.

Epigenetic: Changes in gene expression or cellular Methylation: Covalent addition of a methyl group to a Splice sites: Small regions of genes that are juxtaposed Downloaded from phenotype caused by mechanisms other than changes protein, DNA, or other molecule. to the exons and direct exon splicing. in the DNA sequence. Missense mutation: Asingle-nucleotidesubstitution(e.g., Stem cell: An immortal cell that can repopulate a par- Exome: The collection of exons in the . C to T) that results in an amino acid substitution (e.g., ticular cell type. Exome sequencing generally refers to the collection of histidine to arginine). exons that encode proteins. Subclonal mutation: Amutationthatexistsinonlya Mut-driver gene: A gene that contains driver gene subset of the neoplastic cells within a tumor. mutations. Gatekeeper: A gene that, when mutated, initiates tumori- genesis. Examples include RB,mutationsofwhichini- Translocation: Aspecifictypeofrearrangementwhere Nonsense mutation: A single-nucleotide substitution tiate retinoblastomas, and VHL,whosemutationsinitiate regions from two nonhomologous chromosomes are (e.g., C to T) that results in the production of a stop codon. renal cell carcinomas. joined. Nonsynonymous mutation: Amutationthataltersthe Germline genome: An individual’sgenome,asinherited encoded amino acid sequence of a protein. These include Tumor suppressor gene: A gene that, when inacti- from their parents. missense, nonsense, splice site, translation start, transla- vated by mutation, increases the selective growth ad- tion stop, and indel mutations. vantage of the cell in which it resides. Germline variants: Variations in sequences observed in different individuals. Two randomly chosen individuals Oncogene: Agenethat,whenactivatedbymutation,in- Untranslated regions: Regions within the exons differ by ~20,000 genetic variations distributed through- creases the selective growth advantage of the cell in which at the 5′ and 3′ ends of the gene that do not encode out the exome. it resides. amino acids.

www.sciencemag.org SCIENCE VOL 339 29 MARCH 2013 1549 = Missense mutation = Truncating mutation

N ABD RBD C2 Helical Kinase C N C Substrate binding sites PIK3CA 1068 aa IDH1 414 aa

928 aa 213 aa N T-Ag and E1A-binding E4F1 binding C N 5 aa repeats CCT BC C RB1 VHL on April 30, 2013 Fig. 4. Distribution of mutations in two oncogenes (PIK3CA and IDH1) collected from genome-wide studies annotated in the COSMIC database (release and two tumor suppressor genes (RB1 and VHL). The distribution of missense version 61). For PIK3CA and IDH1,mutationsobtainedfromtheCOSMICdatabase mutations (red arrowheads) and truncating mutations (blue arrowheads) in rep- were randomized by the Excel RAND function, and the first 50 are shown. For RB1 resentative oncogenes and tumor suppressor genes are shown. The data were and VHL,allmutationsrecordedinCOSMICareplotted.aa,aminoacids.

regions of the genome in an apparently random rantly in tumors but not frequently mutated; they sor gene, we analogously require that >20% of fashion (28). Thus, at best, methods based on mu- are altered through changes in DNA methyla- the recorded mutations in the gene are inac- tation frequency can only prioritize genes for fur- tion or chromatin modification that persist as the tivating. This “20/20 rule” is lenient in that all ther analysis but cannot unambiguously identify tumor cell divides. well-documented cancer genes far surpass these www.sciencemag.org driver genes that are mutated at relatively low criteria (table S2A). frequencies. ARatiometricMethodtoIdentifyand The following examples illustrate the value Further complicating matters, there are two Classify Mut-Driver Genes of the 20/20 rule. When IDH1 mutations were distinct meanings of the term “driver gene” If mutation frequency, corrected for mutation first identified in brain tumors, their role in tu- that are used in the cancer literature. The driver- context, gene length, and other parameters, can- morigenesis was unknown (2, 31). Initial func- versus-passenger concept was originally used to not reliably identify modestly mutated driver tional studies suggested that IDH1 was a tumor

distinguish mutations that caused a selective genes, what can? In our experience, the best suppressor gene and that mutations inactivated Downloaded from growth advantage from those that did not (29). way to identify Mut-driver genes is through this gene (32). However, nearly all of the muta- According to this definition, a gene that does not their pattern of mutation rather than through tions in IDH1 were at the identical amino acid, harbor driver gene mutations cannot be a driver their mutation frequency. The patterns of mu- codon 132 (Fig. 4). As assessed by the 20/20 gene. But many genes that contain few or no tations in well-studied oncogenes and tumor rule, this distribution unambiguously indicated driver gene mutations have been labeled driver suppressor genes are highly characteristic and that IDH1 was an oncogene rather than a tumor genes in the literature. These include genes that nonrandom. Oncogenes are recurrently mu- suppressor gene, and this conclusion was even- are overexpressed, underexpressed, or epigenet- tated at the same amino acid positions, where- tually supported by biochemical experiments ically altered in tumors, or those that enhance as tumor suppressor genes are mutated through (33, 34). Another example is provided by muta- or inhibit some aspect of tumorigenicity when protein-truncating alterations throughout their tions in NOTCH1.Inthiscase,somefunctional their expression is experimentally manipulated. length (Fig. 4 and table S2A). studies suggested that NOTCH1 was an onco- Though a subset of these genes may indeed On the basis of these mutation patterns rather gene, whereas others suggested it was a tumor play an important role in the neoplastic pro- than frequencies, we can determine which of the suppressor gene (35, 36). The situation could be cess, it is confusing to lump them all together 18,306 mutated genes containing a total of clarified through the application of the 20/20 as driver genes. 404,863 subtle mutations that have been recorded rule to NOTCH1 mutations in cancers. In “liq- To reconcile the two connotations of driver in the Catalogue of Somatic Mutations in Cancer uid tumors” such as lymphomas and leuke- genes, we suggest that genes suspected of increas- (COSMIC) database (30)areMut-drivergenes mias, the mutations were often recurrent and did ing the selective growth advantage of tumor cells and whether they are likely to function as onco- not truncate the predicted protein (37). In squa- be categorized as either “Mut-driver genes” or genes or tumor suppressor genes. To be classified mous cell carcinomas, the mutations were not “Epi-driver genes.” Mut-driver genes contain a as an oncogene, we simply require that >20% of recurrent and were usually inactivating (38–40). sufficient number or type of driver gene muta- the recorded mutations in the gene are at re- Thus, the genetic data clearly indicated that tions to unambiguously distinguish them from current positions and are missense (see legend to NOTCH1 functions differently in different tumor other genes. Epi-driver genes are expressed aber- table S2A). To be classified as a tumor suppres- types. The idea that the same gene can function

1550 29 MARCH 2013 VOL 339 SCIENCE www.sciencemag.org SPECIALSECTION in completely opposite ways in different cell frequencies had already been identified in tumors was mysterious before the discovery of mutations types is important for understanding cell signal- of other organs. In other words, the number of of these genes and their perfect correlation with the ing pathways. frequently altered Mut-driver genes (mountains) ALT phenotype (64). A final example is provided is nearing saturation. More mountains will un- by IDH1 and IDH2,whosemutationshavestim- How Many Mut-Driver Genes Exist? doubtedly be discovered, but these will likely be ulated the burgeoning field of tumor metabolism Though all 20,000 protein-coding genes have been in uncommon tumor types that have not yet (65)andhavehadfascinatingimplicationsfor evaluated in the genome-wide sequencing studies been studied in depth. epigenetics (66, 67). of 3284 tumors, with a total of 294,881 muta- The newly discovered Mut-driver genes that The Mut-driver genes listed in table S2A tions reported, only 125 Mut-driver genes, as de- have been detected through genome-wide se- are affected by subtle mutations: base substi- fined by the 20/20 rule, have been discovered to quencing have often proved illuminating. For ex- tutions, intragenic insertions, or deletions. As date (table S2A). Of these, 71 are tumor sup- ample, nearly half of these genes encode proteins noted above, Mut-driver genes can also be al- pressor genes and 54 are oncogenes. An impor- that directly regulate chromatin through modifi- tered by less subtle changes, such as transloca- tant but relatively small fraction (29%) of these cation of or DNA. Examples include the tions, amplifications, and large-scale deletions. genes was discovered to be mutated through un- histones HIST1H3B and , as well as the As with point mutations, it can be difficult to biased genome-wide sequencing; most of these proteins DNMT1 and TET1, which covalently distinguish Mut-driver genes that are altered by genes had already been identified by previous, modify DNA, EZH2, SETD2, and KDM6A, these types of changes from genes that contain more directed investigations. which, in turn, methylate or demethylate histones only passenger mutations. Genes that are not How many more Mut-driver genes are yet to (53–57). These discoveries have profound impli- point-mutated, but are recurrently amplified (e.g., be discovered? We believe that a plateau is being cations for understanding the mechanistic basis of MYC family genes) or homozygously deleted reached, because the same Mut-driver genes keep the epigenetic changes that are rampant in tumors (e.g., MAP2K4)andthatmeetothercriteria(e.g., being “rediscovered” in different tumor types. (58). The discovery of genetic alterations in genes being the only gene in the amplicon or homo- For example, MLL2 and MLL3 mutations were encoding mRNA splicing factors, such as SF3B1 zygously deleted region) are listed in table originally discovered in medulloblastomas (41) and U2AF1 (59–61), was similarly stunning, as S2B. This adds 13 Mut-driver genes—10 onco- and were subsequently discovered to be mutated mutations in these genes would be expected to genes that are amplified and 3 tumor suppressor on April 30, 2013 in non-Hodgkin lymphomas, prostate cancers, lead to a plethora of nonspecific cellular stresses genes that are homozygously deleted—to the breast cancers, and other tumor types (42–45). rather than to promote specific tumor types. An- 125 driver genes that are affected by subtle mu- Similarly, ARID1A mutations were first discov- other example is provided by mutations in the tations, for a total of 138 driver genes discov- ered to be mutated in clear-cell ovarian cancers cooperating proteins ATRX and DAXX (62). ered to date (table S2). (46, 47)andweresubsequentlyshowntobemu- Tumors with mutations in these genes all have a Translocations provide similar challenges for tated in tumors of several other organs, including specific type of telomere elongation process termed driver classification. An important discovery re- those of the stomach and liver (48–50). In recent “ALT” (for “alternative lengthening of telomeres”) lated to this point is chromothripsis (68), a rare studies of several types of lung cancer (4, 51, 52), (63). Though the ALT phenotype had been rec- cataclysmic event involving one or a small num- nearly all genes found to be mutated at significant ognized for more than a decade, its genetic basis ber of chromosomes that results in a large number of chromosomal rearrangements. www.sciencemag.org This complicates any inferences about 100 causality, in the same way that mis- match repair deficiency compromises Oncogene mutations the interpretation of point mutations. Oncogene + tumor suppressor gene mutations However, for completeness, all fu- 80 sion genes that have been identified

in at least three independent tu- Downloaded from mors are listed in table S3. Virtually 60 all of these genes were discovered through conventional approaches be- fore the advent of genome-wide DNA sequencing studies, with some 40 notable exceptions such as those de- scribed in (6)and(69). The great Fraction of tumors (%) majority of these translocations are 20 found in liquid tumors (leukemias and lymphomas) (table S3C) or mesenchymal tumors (table S3B)

0 and were initially identified through 012345678 012345678 012345678 012345678 012345678 karyotypic analyses. A relatively Medulloblastoma Pancreatic Cancer Glioblastoma Colorectal Cancer Breast Cancer small number of recurrent fusions, Number of driver gene mutations per tumor the most important of which in- clude ERG in prostate cancers (70) Fig. 5. Number and distribution of driver gene mutations in five tumor types. The total number of driver and ALK in lung cancers (71), have gene mutations [in oncogenes and tumor suppressor genes (TSGs)] is shown, as well as the number of oncogene been described in more common mutations alone. The driver genes are listed in tables S2A and S2B. Translocations are not included in this figure, tumors (table S3A). because few studies report translocations along with the other types of genetic alterations on a per-case basis. In the Genes exist that predispose to tumor types shown here, translocations affecting driver genes occur in less than 10% of samples. The published data cancer when inherited in mutant on which this figure is based are provided in table S1E. form in the germ line, but are not

www.sciencemag.org SCIENCE VOL 339 29 MARCH 2013 1551 somatically mutated in cancer to a substantial mors is not as carefully evaluated as in (79)will terpret than the somatic mutations in cancers. degree. These genes generally do not confer an have higher false-negative rates. Moreover, these The first examples of light coming to such dark increase in selective growth advantage when they technical problems are exacerbated in whole- matter have recently been published: Recurrent are abnormal, but they stimulate tumorigenesis genome studies compared with exomic analyses, mutations in the promoter of the TERT gene, en- in indirect ways (such as by increasing genetic in- because the sequence coverage of the former coding the catalytic subunit of telomerase, have stability, as discussed later in this Review). For is often lower than that of the latter (generally been identified and shown to activate its tran- completeness, these genes and the hereditary syn- 30-fold in whole-genome studies versus more scription (81, 82). dromes for which they are responsible are listed than 100-fold in exomic studies). Mut-driver genes other than those listed in in table S4. Conceptual issues also limit the number of table S2 will undoubtedly be discovered as detectable drivers. Virtually all studies, either at genome-wide sequencing continues. However, Dark Matter the whole-genome or whole-exome level, have based on the trends noted above, most of the Classic epidemiologic studies have suggested focused on the coding regions. The reason for Mut-driver genes will likely be mountains in that solid tumors ordinarily require five to eight “hits,” now interpreted as alterations in driver genes, to develop (72). Is this number compat- Intratumoral heterogeneity Intermetastatic heterogeneity ible with the molecular genetic data? In pediatric within a primary tumor between two metastases tumors such as medulloblastomas, the number A B of driver gene mutations is low (zero to two), as Clone 1 Clone 2 expected from the discussion above (Fig. 5). Metastasis 1 In common adult tumors—such as pancreatic, Liver Founder colorectal, breast, and brain cancers—the num- cells ber of mutated driver genes is often three to six, Pancreas Metastasis 2 but several tumors have only one or two driver gene mutations (Fig. 5). How can this be ex- Clone 4 Clone 3 on April 30, 2013 plained, given the widely accepted notion that Primary tumor tumor development and progression require mul- tiple, sequential genetic alterations acquired over decades? Intrametastatic heterogeneity within metastatic lesions Interpatient heterogeneity First, technical issues explain some of the “missing mutations.” Genome-wide sequenc- C D Patient 1 Patient 2 ing is far from perfect, at least with the tech- nologies available today. Some regions of the genome are not well represented because their sequences are difficult to amplify, capture, or www.sciencemag.org unambiguously map to the genome (73–76). Second, there is usually a wide distribution in the number of times that a specific nucleotide in a given gene is observed in the sequence data, so some regions will not be well represented by Fig. 6. Four types of genetic heterogeneity in tumors, illustrated by a primary tumor in chance factors alone (77). Finally, primary tu- the pancreas and its metastatic lesions in the liver. Mutations introduced during primary

mors contain not only neoplastic cells, but also tumor cell growth result in clonal heterogeneity. At the top left, a typical tumor is represented by Downloaded from stromal cells that dilute the signal from the mu- cells with a large fraction of the total mutations (founder cells) from which subclones are derived. tated base, further reducing the probability of The differently colored regions in the subclones represent stages of evolution within a subclone. (A) finding a mutation (78). Intratumoral: heterogeneity among the cells of the primary tumor. (B)Intermetastatic:heterogeneity What fraction of mutations are missed by among different metastatic lesionsinthesamepatient.Inthecaseillustratedhere,eachmetastasiswas these three technical issues? A recent study derived from a different subclone. (C)Intrametastatic:heterogeneityamongthecellsofeachmetastasis of pancreatic cancers is informative in this develops as the metastases grow. (D)Interpatient:heterogeneityamongthetumorsofdifferent regard. Biankin et al.usedimmunohistochem- patients. The mutations in the founder cells of the tumors of these two patientsarealmostcompletely ical and genetic analyses to select a set of pri- distinct (see text). mary tumor samples enriched in neoplastic cells (79). They used massively parallel sequenc- ing to analyze the exomes of these samples, this is practical; it is difficult enough to iden- rare tumor types or small hills in common tu- then compared their mutational data with a set tify driver gene mutations when they qualita- mor types; thus, these genes are unlikely to ac- of pancreatic cancer cell lines and xenografts tively alter the sequence of the encoded protein. count for the bulk of the presumptive dark matter. in which mutations had previously been iden- Trying to make sense of intergenic or intronic Other types of dark matter can be envisioned, tified, using conventional Sanger sequenc- mutations is much more difficult. Based on however. Copy-number alterations are ubiqui- ing, and confirmed to be present in the primary analogous studies of the identifiable mutations tous in cancers, at either the whole-chromosome tumors (3, 16). Only 159 (63%) of the expected in patients with monogenic diseases, more than or subchromosomal levels. These alterations could 251 driver gene mutations were identified in 80% of mutations should be detectable through subtly change the expression of their driver the primary tumors studied by next-generation analysis of the coding regions (80). However, genes. Recent studies have suggested that the sequencing alone, indicating a false-negative this still leaves some mutations as unidentifiable loss of one copy of chromosomes containing rate of 37%. Genome-wide studies in which “dark matter,” even in the germline genomes of several tumor suppressor genes, each plausi-

the proportion of neoplastic cells within tu- heritable cases, which are usually easier to in- bly connected to neoplasia but not altered by CREDIT: FIG. 6, E. COOK

1552 29 MARCH 2013 VOL 339 SCIENCE www.sciencemag.org SPECIALSECTION mutation, may confer a selective growth advan- tially distinct and, in general, will display more consistent with the observation that in patients tage (83, 84). differences than neighboring cells (16). This responsive to targeted agents, the response is The most obvious source of dark matter is in phenomenon is analogous to speciation, wherein often seen in all metastatic lesions rather than Epi-driver genes. Human tumors contain large organisms on different islands are more likely to just a small subset (98). numbers of epigenetic changes affecting DNA diverge from one another than are organisms on 3) Intrametastatic: heterogeneity among the or chromatin proteins. For example, a recent the same island. cells of an individual metastasis. Each metasta- study of colorectal cancers showed that more In studies that have evaluated intratumoral sis is established by a single cell (or small group than 10% of the protein-coding genes were differ- heterogeneity by genome-wide sequencing, the of cells) with a set of founder mutations. As it entially methylated when compared with normal majority of somatic mutations are present in all grows, the metastasis acquires new mutations with colorectal epithelial cells (85). Some of these tumor cells. These mutations form the trunk of each cell division. Though the founder muta- changes (i.e., those in Epi-driver genes) are likely the somatic evolutionary tree. What is the im- tions may make the lesion susceptible to antitu- to provide a selective growth advantage (86, 87). portance of the mutations in the branches (i.e., mor agents, the new mutations provide the seeds For example, epigenetic silencing of CDK2NA those that are not shared by all tumor cells)? for drug resistance. Unlike primary tumors, the and MLH1 is much more common than muta- From a medical perspective, these mutations metastatic lesions generally cannot be removed tional inactivation of either of these two well- are often meaningless because the primary tu- by surgery and must be treated with systemic recognized driver genes (85)However,thereisa mors are surgically removed. How much het- therapies. Patients with complete responses to critical difference between a genetic and an epi- erogeneity existed in the various branches before targeted therapies invariably relapse. Most of the genetic change in a gene. Unlike the sequence surgery is not important. However, this het- initial lesions generally recur, and the time frame of a gene in a given individual, methylation is erogeneity provides the seeds for intermeta- at which they recur is notably similar. This time plastic, varying with cell type, developmental stastic heterogeneity, which is of great clinical course can be explained by the presence of resist- stage, and patient age (21). The methylation importance. ance mutations that existed within each metastasis state of the normal precursor cells that initiate 2) Intermetastatic: heterogeneity among dif- before the onset of the targeted therapy (99–102). tumorigenesis is unknown; these cells, such as ferent metastatic lesions of the same patient. Calculations show that any metastatic lesion of a normal stem cells, may represent only a tiny The vast majority of cancer patients die because size visible on medical imaging has thousands on April 30, 2013 fraction of the cells in a normal organ. This their tumors were not removed before metas- of cells (among the billions present) that are al- plasticity also means that methylation can change tasis to surgically inaccessible sites, such as ready resistant to virtually any drug that can be under microenvironmental cues, such as those the liver, brain, lung, or bone. Patients who re- imagined (99, 101, 102). Thus, recurrence is sim- associated with low nutrient concentrations or lapse with a single metastatic lesion can often ply a matter of time, entirely predictable on the abnormal cell contacts. It is therefore difficult still be cured by surgery or radiotherapy, but basis of known mutation frequencies and tumor to know whether specific epigenetic changes single metastases are the exception rather than cell growth rates. This “fait accompli” can be cir- observed in cancer cells reflect, rather than the rule. A typical patient on a clinical trial has a cumvented, in principle, by treatment with multi- contribute to, the neoplastic state. Criteria for dozen or more metastatic lesions large enough ple agents, as it is unlikely that a single tumor cell distinguishing epigenetic changes that exert a to be visualized by imaging, and many more will be resistant to multiple drugs that act on selective growth advantage from those that do that are smaller. If each of the metastatic le- different targets. www.sciencemag.org not (passenger epigenetic changes) have not yet sions in a single patient was founded by a cell 4) Interpatient: heterogeneity among the tu- been formulated. Given that Epi-driver genes with a very different genetic constitution, then mors of different patients. This type of hetero- are likely to compose a major component of the chemotherapeutic cures would be nearly im- geneity has been observed by every oncologist; dark matter, further research on this topic is possible to achieve: Eradicating a subset of the no two cancer patients have identical clinical essential (58). metastatic lesions in a patient will not be ade- courses, with or without therapy. Some of these quate for long-term survival. differences could be related to host factors, such Genetic Heterogeneity

How much heterogeneity is there among dif- as germline variants that determine drug half- Downloaded from The mutations depicted in Fig. 1 are clonal; that is, ferent metastatic lesions? In short, a lot. It is not life or vascular permeability to drugs or cells, they are present in the majority of the neoplastic uncommon for one metastatic lesion to have 20 and some could be related to nongenetic factors cells in the tumors. But additional, subclonal (i.e., clonal genetic alterations not shared by other (103). However, much of this interpatient heter- heterogeneous within the tumor) mutations are metastases in the same patient (16, 97). Because ogeneity is probably related to somatic mutations important for understanding tumor evolution. they are clonal, these mutations occurred in the within tumors. Though several dozen somatic Four types of genetic heterogeneity are relevant founder cell of the metastasis; that is, the cell mutations may be present in the breast cancers to tumorigenesis (Fig. 6): that escaped from the primary tumor and multi- from two patients, only a small number are in the 1) Intratumoral: heterogeneity among the plied to form the metastasis. The founder cell for same genes, and in the vast majority of cases, cells of one tumor. This type of heterogeneity each metastasis is present in different, geograph- these are the Mut-driver genes (1, 104, 105). Even has been recognized for decades. For example, ically distinct areas of the primary tumors, as in these driver genes, the actual mutations are it is rare to see a cytogenetic study of a solid expected (16). often different. Mutations altering different do- tumor in which all of the tumor cells display the This potentially disastrous situation is tem- mains of a protein would certainly not be expected same karyotype (88). The same phenomenon pered by the fact that the heterogeneity appears to have identical effects on cellular properties, as has been noted for individual genes [e.g., (89)] largely confined to passenger gene mutations. experimentally confirmed (106). Though it may and more recently has been observed throughout In most of the studies documenting heteroge- seem that different mutations in adjacent codons the genome (16, 90–96). This kind of heteroge- neity in malignancies, the Mut-driver genes are would have identical effects, detailed studies of neity must exist: Every time a normal (or tumor) present in the trunks of the trees, though ex- large numbers of patients have shown that this cell divides, it acquires a few mutations, and ceptions have been noted (95). These findings need not be the case. For example, a Gly12→Asp12 the number of mutations that distinguish any are consistent with the idea, discussed above, (G12D) mutation of KRAS does not have the two cells simply marks the time from their last that the genetic alterations required for meta- same clinical implications as a G13D mutation common ancestor (their founder cell). Cells at stasis were present (i.e., selected for) before of the same gene (107). Interpatient heterogene- the opposite ends of large tumors will be spa- metastasis actually occurred. The data are also ity has always been one of the major obstacles

www.sciencemag.org SCIENCE VOL 339 29 MARCH 2013 1553 to designing uniformly effective treatments for cell death in response to such alterations, per- by epigenetic alterations affecting DNA and chro- cancer. Efforts to individualize treatments based haps as a protective mechanism against cancer. matin proteins. What better way to subvert this on knowledge of the genomes of cancer pa- In contrast, cancer cells have evolved to tolerate normal mechanism for controlling tissue archi- tients are largely based on an appreciation of genome complexity by acquiring mutations in tecture than to debilitate the epigenetic modifying this heterogeneity. genes such as TP53 (110). Thus, genomic com- apparatus itself? plexity is, in part, the result of cancer, rather than 2) Cell survival: Though cancer cells di- Signaling Pathways in Tumors the cause. vide abnormally because of cell-autonomous al- The immense complexity of cancer genomes To appreciate the second concept, one must terations, such as those controlling cell fate, their that could be inferred from the data described take the 30,000-foot view. A jungle might look surrounding stromal cells are perfectly normal above is somewhat misleading. After all, even chaotic at ground level, but the aerial view shows and do not keep pace. The most obvious ram- advanced tumors are not completely out of aclearorder,withalltheanimalsgatheringat ification of this asymmetry is the abnormal vas- control, as evidenced by the dramatic responses the streams at certain points in the day, and all culature of tumors. As opposed to the well-ordered to agents that target mutant BRAF in mela- the streams converging at a river. There is order network of arteries, veins, and lymphatics that nomas (108)ormutantALK in lung cancers in cancer, too. Mutations in all of the 138 driver control nutrient concentrations in normal tissues, (109). Albeit transient, these responses mean genes listed in table S2 do one thing: cause a the vascular system in cancers is tortuous and that interference with even a single mutant gene selective growth advantage, either directly or lacks uniformity of structure (112, 113). Normal product is sufficient to stop cancer in its tracks, indirectly. Moreover, there appears to be only a cells are always within 100 mmofacapillary, at least transiently. How can the genomic com- limited number of cellular signaling pathways but this is not true for cancer cells (114). As a plexity of cancer be reconciled with these clin- through which a growth advantage can be in- result, a cancer cell acquiring a mutation that ical observations? curred (Fig. 7 and table S5). allows it to proliferate under limiting nutrient Two concepts bear on this point. The first, All of the known driver genes can be classi- concentrations will have a selective growth ad- mentioned above, is that >99.9% of the altera- fied into one or more of 12 pathways (Fig. 7). vantage, thriving in environments in which its tions in tumors (including point mutations, copy- The discovery of the molecular components of sister cells cannot. Mutations of this sort occur, number alterations, translocations, and epigenetic these pathways is one of the greatest achievements for example, in the EGFR, HER2, FGFR2, PDGFR, on April 30, 2013 changes distributed throughout the genome, of biomedical research, a tribute to investigators TGFbR2, MET, KIT, RAS, RAF, PIK3CA,and not just in the coding regions) are immaterial to working in fields that encompass biochemistry, PTEN genes (table S2A). Some of these genes neoplasia. They are simply passenger changes cell biology, and development, as well as cancer. encode receptors for the growth factors them- that mark the time that has elapsed between These pathways can themselves be further or- selves, whereas others relay the signal from the successive clonal expansions. Normal cells also ganized into three core cellular processes: growth factor to the interior of the cell, stim- undergo genetic alterations as they divide, both 1) Cell fate: Numerous studies have demon- ulating growth when activated (115, 116). For at the nucleotide and chromosomal levels. How- strated the opposing relationship between cell instance, mutations in KRAS or BRAF genes ever, normal cells are programmed to undergo division and differentiation, the arbiters of cell confer on cancer cells the ability to grow in glu- fate. Dividing cells that are re- cose concentrations that are lower than those sponsible for populating normal required for the growth of normal cells or of www.sciencemag.org tissues (stem cells) do not differ- cancer cells that do not have mutations in these entiate, and vice versa. Regen- genes (117, 118). Progression through the cell

erative medicine is based on this cycle (and its antithesis, apoptosis) can be di- apoptosis

Cell cycle/ Cell distinction, predicated on ways rectly controlled by intracellular metabolites, RAS to get differentiated cells to de- and driver genes that directly regulate the cell differentiate into stem cells, then cycle or apoptosis, such as CDKN2A, MYC,and PI3K

forcing the stem cells to differ- BCL2,areoftenmutatedincancers.Another Downloaded from

NOTCH entiate into useful cell types for gene whose mutations enhance cell survival is transplantation back into the pa- VHL,theproductofwhichstimulatesangiogen- STAT Selective HH tient. Many of the genetic alter- esis through the secretion of vascular endothelial growth ations in cancer abrogate the growth factor. What better way to provision advantage precise balance between differ- growth factors to a rogue tumor than to lure the PK APC MA entiation and division, favoring unsuspecting vasculature to its hideout? modificationChromatin the latter. This causes a selective 3) Genome maintenance: As a result of the ␤ scriptional regulation F- growth advantage, because dif- exotic microenvironments in which they re- G T Tran- ferentiating cells eventually die side, cancer cells are exposed to a variety of or become quiescent. Pathways toxic substances, such as reactive oxygen spe- DNA damage control G that function through this process cies. Even without microenvironmental poi- M en ain ome include APC, HH, and NOTCH, sons, cells make mistakes while replicating their tena nce all of which are well known to DNA or during division (119, 120), and check- control cell fate in organisms points exist to either slow down such cells or ranging from worms to mammals make them commit suicide (apoptosis) under Fig. 7. Cancer cell signaling pathways and the cellular pro- (111). Genes encoding chromatin- such circumstances (110, 121, 122). Although it cesses they regulate. All of the driver genes listed in table S2 modifying enzymes can also be is good for the organism to remove these dam- can be classified into one or more of 12 pathways (middle ring) included in this category. In nor- aged cells, tumor cells that can survive the dam- that confer a selective growth advantage (inner circle; see main text). mal development, the heritable age will, by definition, have a selective growth These pathways can themselves be further organized into three core switch from division to differen- advantage. Therefore, it is not surprising that cellular processes (outer ring). Thepublicationsonwhichthisfigure tiation is not determined by muta- genes whose mutations abrogate these checkpoints, is based are provided in table S5. tion, as it is in cancer, but rather such as TP53 and ATM,aremutatedincancers

1554 29 MARCH 2013 VOL 339 SCIENCE www.sciencemag.org SPECIALSECTION

(123). Defects in these genes can also indirectly confer a selective growth advantage by allow- ing cells that have a gross chromosomal change favoring growth, such as a translocation or an extra chromosome, to survive and divide. Anal- ogously, genes that control point mutation rates, such as MLH1 or MSH2,aremutatedincan- cers (table S2A) or in the germ line of patients predisposed to cancers (table S4) because they accelerate the acquisition of mutations that func- tion through processes that regulate cell fate or survival. What better way to promote cancer than by increasing the rate of occurrence of the muta- tions that drive the process? Because the protein products of genes reg- ulating cell fate, cell survival, and genome main- tenance often interact with one another, the pathways within them overlap; they are not as discrete as might be inferred from the description above. However, grouping genes into pathways makes perfect sense from a genetics standpoint. Given that cancer is a genetic disease, the prin- ciples of genetics should apply to its pathogenesis. When performing a conventional mutagenesis on April 30, 2013 screen in bacteria, yeast, fruit flies, or worms, one expects to discover mutations in several different genes that confer similar phenotypes. The products of these genes often interact with one another and define a biochemical or de- velopmental pathway. Therefore, it should not be surprising that several different genes can result in the same selective growth advantage for cancer cells and that the products of these genes interact. The analogy between cancer www.sciencemag.org pathways and biochemical or developmental pathways in other organisms goes even deeper: The vast majority of our knowledge of the func- tion of driver genes has been derived from the study of the pathways through which their homo- logs work in nonhuman organisms. Though the

functions are not identical to those in human Downloaded from cells, they are highly related and have provided the starting point for analogous studies in hu- man cells. Fig. 8. Signal transduction pathways affected by mutations in human cancer. Two represent- Recognition of these pathways also has im- ative pathways from Fig. 7 (RAS and PI3K) are illustrated. The signal transducers are color coded: portant ramifications for our ability to understand red indicates protein components encoded by the driver genes listed in table S2; yellow balls interpatient heterogeneity. One lung cancer might denote sites of phosphorylation. Examples of therapeutic agents that target some of the signal have an activating mutation in a receptor for a transducers are shown. RTK, receptor tyrosine kinase; GDP, guanosine diphosphate; MEK, MAPK stimulatory growth factor, making it able to grow kinase; ERK, extracellular signal–regulated kinase; NFkB, nuclear factor kB; mTOR, mammalian in low concentrations of epidermal growth factor target of rapamycin. (EGF). A second lung cancer might have an ac- tivating mutation in KRAS,whoseproteinproduct same tumor—and this has been experimentally clinical care of cancer patients. The recognition normally transmits the signal from the epidermal confirmed (124, 125). Apart from being intel- that certain tumors contain activating mutations in growth factor receptor (EGFR) to other cell sig- lectually satisfying, knowledge of these path- driver genes encoding protein kinases has led to naling molecules. A third lung cancer might have ways has implications for cancer therapy, as the development of small-molecule inhibitor an inactivating mutation in NF1,aregulatory discussed in the next section. drugs targeting those kinases. protein that normally inactivates the KRAS pro- Representative examples of this type of tein. Finally, a fourth lung cancer might have a APerspectiveonGenome-BasedMedicine genome-based medicine include the use of EGFR mutation in BRAF,whichtransmitsthesignal in Oncology kinase inhibitors to treat cancers with EGFR from KRAS to downstream kinases (Fig. 8). One gene mutations (126), the aforementioned ana- would predict that mutations in the various Opportunities plastic lymphoma kinase (ALK) inhibitors to components of a single pathway would be mu- Though cancer genome sequencing is a relatively treat cancers with ALK gene translocations

CREDIT: FIG. 8, A. DIXON tually exclusive—that is, not occurring in the new endeavor, it has already had an impact on the (109), and specific inhibitors of mutant BRAF

www.sciencemag.org SCIENCE VOL 339 29 MARCH 2013 1555 to treat cancers with BRAF mutations (108). with small drugs is notoriously difficult because targeted therapeutic approaches ever be ex- Before instituting treatment with such agents, small compounds can only inhibit one of these pected to induce long-term remissions, even cures, it is imperative to determine whether the can- interactions (130, 131). rather than the short-term remissions now being cer harbors the mutations that the drug targets. Though one can at least imagine the devel- achieved? The saviors are pathways; every tu- Only a small fraction of lung cancer patients have opment of drugs that inhibit nonenzymatic pro- mor suppressor gene inactivation is expected to EGFR gene mutations or ALK gene transloca- tein functions, the second challenge evident from result in the activation of some growth-promoting tions, and only these patients will respond to the table S2 poses even greater difficulties: A large signal downstream of the pathway. An exam- drugs. Treating lung cancer patients without these fraction of the Mut-driver genes encode tumor ple is provided by PTEN mutations: Inactivation particular genetic alterations would be detri- suppressors. Drugs generally interfere with pro- of the tumor suppressor gene PTEN results in mental, as such patients would develop the tein function; they cannot, in general, replace the activation of the AKT kinase (Fig. 8). Similarly, toxic side effects of the drugs while their tumors function of defective genes such as those result- inactivation of the tumor suppressor gene CDKN2A progressed. ing from mutations in tumor suppressor genes. results in activation of kinases, such as cyclin- Asecondtypeofgenome-basedmedicine Unfortunately, tumor suppressor gene–inactivating dependent kinase 4, that promote cell cycle focuses on the side effects and metabolism of mutations predominate over oncogene-activating traverse (132). Furthermore, inactivation of tu- the therapeutic agents, rather than the genetic mutations in the most common solid tumors: mor suppressor gene APC results in constitutive alterations they target. At present, the dose of Few individual tumors contain more than one activity of oncogenes such as CTNNB1 and cancer drugs given to patients is based on the oncogene mutation (Fig. 5). CMYC (133–135). patients’ size (body weight or surface area). The relatively small number of oncogene We believe that greater knowledge of these But the therapeutic ratio of cancer drugs (ratio mutations in tumors is important in light of the pathways and the ways in which they function of the concentration that causes side effects to intrametastatic heterogeneity described earlier. is the most pressing need in basic cancer re- the concentration required to kill tumor cells) To circumvent the inevitable development of re- search. Successful research on this topic should is generally low, particularly for conventional sistance to targeted therapies, it will likely be allow the development of agents that target, al- (nontargeted) therapeutic agents. Small changes necessary to treat patients with two or more beit indirectly, defective tumor suppressor genes. in circulating concentrations of these drugs can drugs. The probability that a single cancer cell Indeed, there are already examples of such in- on April 30, 2013 make the difference between substantial tumor within a large metastatic lesion will be resistant direct targeting. Inactivating mutations of the regression and intolerable side effects. Interroga- to two agents that target two independent path- tumor suppressor genes BRCA1 or BRCA2 lead tion of the germline status of the genes encoding ways is exponentially less than the probability to activation of downstream pathways required drug-metabolizing enzymes could substantially that the cell will be resistant to a single agent. to repair DNA damage in the absence of BRCA improve the outcomes of treatment by informing However, if the cancer cell does not contain more function. Thus, cancer cells with defects in BRCA1 drug dosing (127). Optimally, this genome inter- than one targetable genetic alteration (i.e., an on- or BRCA2 are more susceptible to DNA dam- rogation would be accompanied by pharmaco- cogene mutation), then this combination strategy aging agents or to drugs that inhibit enzymes kinetic measurements of drug concentrations is not feasible. that facilitate the repair of DNA damage such in each patient. The additional cost of such Given the paucity of oncogene alterations in as PARP [poly(adenosine diphosphate–ribose) analyses would be small compared with the ex- common solid tumors and these principles, can polymerase] (136). PARP inhibitors have shown www.sciencemag.org orbitant costs of new cancer therapies—for re- cently approved drugs, the cost is estimated to be $200,000 to $300,000 per quality life year produced (128). Box 2. Highlights

Challenges 1. Most human cancers are caused by two to eight sequential alterations that develop over the

One challenge of genome-based medicine in course of 20 to 30 years. Downloaded from oncology is already apparent from the oppor- tunities described above: All of the clinically 2. Each of these alterations directly or indirectly increases the ratio of cell birth to cell death; that approved drugs that target the products of ge- is, each alteration causes a selective growth advantage to the cell in which it resides. netically altered genes are directed against ki- 3. The evidence to date suggests that there are ~140 genes whose intragenic mutations contribute nases. One reason for this is that kinases are to cancer (so-called Mut-driver genes). There are probably other genes (Epi-driver genes) that are relatively easy to target with small molecules altered by epigenetic mechanisms and cause a selective growth advantage, but the definitive and have been extensively studied at the bio- identification of these genes has been challenging. chemical, structural, and physiologic levels (129). 4. The known driver genes function through a dozen signaling pathways that regulate three core But another reason has far deeper ramifications. cellular processes: cell fate determination, cell survival, and genome maintenance. The vast majority of drugs on the market today, for cancer or other diseases, inhibit the actions 5. Every individual tumor, even of the same histopathologic subtype as another tumor, is distinct of their protein targets. This inhibition occurs with respect to its genetic alterations, but the pathways affected in different tumors are similar. because the drugs interfere with the protein’s 6. Genetic heterogeneity among the cells of an individual tumor always exists and can impact the enzymatic activity (such as the phosphorylation response to therapeutics. catalyzed by kinases) or with the binding of the protein to a small ligand (such as with G protein– 7. In the future, the most appropriate management plan for a patient with cancer will be informed by an coupled receptors). Only 31 of the oncogenes assessment of the components of the patient’sgermlinegenomeandthegenomeofhisorhertumor. listed in tables S2 and S3 have enzymatic activ- 8. The information from cancer genome studies can also be exploited to improve methods for ities that are targetable in this manner. Many prevention and early detection of cancer, which will be essential to reduce cancer morbidity and others participate in protein complexes, involv- mortality. ing large interfaces and numerous weak inter- actions. Inhibiting the function of such proteins

1556 29 MARCH 2013 VOL 339 SCIENCE www.sciencemag.org SPECIALSECTION encouraging results in clinical trials when used by the human genome (141), this condition is not alter the function of a limited number of path- in patients whose tumors have inactivating mu- limiting. Second, as most proteins affected by ways. Moreover, we know that this process tations of BRCA genes (137). mutations are intracellular, these mutations will takes decades to develop and that the incurable Further progress in this area will require not be visible to the immune system unless the stage, metastasis, occurs only a few years before more detailed information about the signaling mutant residue is presented in the context of a death. In other words, of the one million people pathways through which cancer genes function human leukocyte antigen (HLA) protein. Based that will die from cancer this year, the vast ma- in human cancer cells, as well as in model or- on in silico analyses of binding affinities, it has jority will die only because their cancers were ganisms. One of the lessons of molecular biol- been estimated that a typical breast or colorectal not detected in the first 90% of the cancers’ ogy over the past two decades is that pathway cancer contains 7 to 10 mutant proteins that can lifetimes, when they were amenable to the sur- functions are different, depending on the orga- bind to an individual patient’sHLAtype(142). geons’ scalpel. nism, cell type, and precise genetic alterations in These theoretical predictions have recently gained This new knowledge of cancer (Box 2) has that cell (138). A pertinent example of this prin- experimental support. Studies of mouse tumors reinvigorated the search for cures for advanced ciple is provided by results of treatment with have identified mutant genes and shown that the cancers, but has not yet permeated other fields of drugs inhibiting mutant BRAF kinase activity. corresponding peptides can induce antitumor im- applied cancer research. A common and limited In the majority of patients with melanomas har- munity when administered as vaccines (143). set of driver genes and pathways is responsible for boring (V600E; V, Val; E, Glu) mutations in the Moreover, clinical trials of brain cancer patients most common forms of cancer (table S2); these BRAF gene, these drugs induce dramatic (though immunized against a mutant peptide have yielded genes and pathways offer distinct potential for transient) remissions (108). But the same drugs encouraging results (144). early diagnosis. The genes themselves, the pro- have no therapeutic effect in colorectal cancer As with all cancer therapies that are attract- teins encoded by these genes, and the end products patients harboring the identical BRAF mutations ive in concept, obstacles abound in practice. If a of their pathways are, in principle, detectable in (139). This observation has been attributed to the tumor expresses a mutant protein that is recog- many ways, including analyses of relevant body expression of EGFR, which occurs in some co- nizable as foreign, why has the host immune fluids, such as urine for genitourinary cancers, lorectal cancers but not in melanoma and is system not eradicated that tumor already? In- sputum for lung cancers, and stool for gastro- thought to circumvent the growth-inhibitory ef- deed, immunoediting in cancers has been shown intestinal cancers (150). Equally exciting are the on April 30, 2013 fects of the BRAF inhibitors. With this example to exist, resulting in the down-regulation or ab- possibilities afforded by molecular imaging, in mind, no one should be surprised that a new sence of mutant epitopes that should have, and which not only indicate the presence of a cancer drug that works well in an engineered tumor in perhaps did, elicit an immune response during but also reveal its precise location and extent. mice fails in human trials; the organism is dif- tumor development (145, 146). Additionally, tu- Additionally, research into the relationship be- ferent, the cell type is usually different, and the mors can lose immunogenicity through a variety tween particular environmental influences (diet precise genetic constitutions are always differ- of genetic alterations, thereby precluding the and lifestyle) and the genetic alterations in can- ent. The converse of this statement—that a drug presentation of epitopes that would otherwise be cer is sparse, despite its potential for prevent- that fails in animal trials will not necessarily fail recognized as foreign (147). Though these theo- ative measures. in human trials—has important practical conse- retical limitations are disheartening, recent studies The reasons that society invests so much quences. In our view, if the biochemical and on immune regulation in humans portend cau- more in research on cures for advanced can- www.sciencemag.org conceptual bases for a drug’sactionsaresolid tious optimism (148, 149). cers than on prevention or early detection are and the drug is shown to be safe in animals, complex. Economic issues play a part: New then a human trial may be warranted, even if it Other Ways to Reduce Morbidity and drugs are far more lucrative for industry than does not shrink tumors in mice. Mortality Through Knowledge of new tests, and large individual costs for treat- Cancer Genomics ing patients with advanced disease have be- Genome-Based Medicines of the Future When we think about eradicating cancer, we come acceptable, even in developing countries

Cancer genomes can also be exploited for the generally think about curing advanced cases— (151). From a technical standpoint, the develop- Downloaded from development of more effective immunother- those that cannot be cured by surgery alone be- ment of new and improved methods for early apies. As noted above, typical solid tumors con- cause they have already metastasized. This is a detection and prevention will not be easy, but tain 30 to 70 mutations that alter the amino acid curious way of thinking about this disease. When there is no reason to assume that it will be more sequences of the proteins encoded by the af- we think of cardiovascular or infectious dis- difficult than the development of new therapies fected genes. Each of these alterations is foreign eases, we first consider ways to prevent them aimed at treating widely metastatic disease. to the immune system, as none have been en- rather than drugs to cure their most advanced Our point is not that strenuous efforts to de- countered during embryonic or postnatal life. forms. Today, we are in no better position to cure velop new therapies for advanced cancer pa- Therefore, these alterations, in principle, pro- polio or massive myocardial infarctions than we tients should be abandoned. These will always vide a “holy grail” for tumor immunology: truly were a thousand years ago. But we can pre- be required, no matter our arsenal of early de- tumor-specific antigens. These antigens could vent these diseases entirely (vaccines), reduce tection or preventative measures. Instead, we are be incorporated into any of the numerous plat- incidence (dietary changes, statins), or miti- suggesting that “plan A” should be prevention forms that already exist for the immunother- gate severity (stents, thrombolytic agents) and and early detection, and “plan B” (therapy for apy of cancer. These include administration of thereby make a major impact on morbidity advanced cancers) should be necessary only vaccines containing the mutant peptide, viruses and mortality. when plan A fails. To make plan A viable, gov- encoding the mutant peptides on their surfaces, This focus on curing advanced cancers might ernment and philanthropic organizations must dendritic cells presenting the mutated peptide, have been reasonable 50 years ago, when the dedicate a much greater fraction of their resources and antibodies or T cells with reactivity directed molecular pathogenesis of cancers was mysteri- to this cause, with long-term considerations in against the mutant peptides (140). ous and when chemotherapeutic agents against mind. We believe that cancer deaths can be re- To realize these sorts of therapeutics, several advanced cancers were showing promise. But duced by more than 75% in the coming decades conditions must be met. First, the mutant protein this mindset is no longer acceptable. We now (152), but that this reduction will only come must be expressed. As cancer cells generally ex- know precisely what causes cancer: a sequential about if greater efforts are made toward early press about half of the proteins that are encoded series of alterations in well-defined genes that detection and prevention.

www.sciencemag.org SCIENCE VOL 339 29 MARCH 2013 1557 References and Notes 60. T. A. Graubert et al., Nat. Genet. 44,53(2012). 117. J. Yun et al., Science 325,1555(2009). 1. L. D. Wood et al., Science 318,1108(2007). 61. K. Yoshida et al., Nature 478, 64 (2011). 118. H. Ying et al., Cell 149,656(2012). 2. D. W. Parsons et al., Science 321,1807(2008). 62. Y. Jiao et al., Science 331,1199(2011). 119. D. J. Araten et al., Cancer Res. 65,8111(2005). 3. S. Jones et al., Science 321,1801(2008). 63. A. J. Cesare, R. R. Reddel, Nat. Rev. Genet. 11, 120. T. A. Kunkel, Cold Spring Harb. Symp. Quant. Biol. 74, 4. R. Govindan et al., Cell 150,1121(2012). 319 (2010). 91 (2009). 5. R. Gryfe, S. Gallinger, Surgery 130, 17 (2001). 64. C. M. Heaphy et al., Science 333,425(2011). 121. B.-B. S. Zhou, S. J. Elledge, Nature 408,433(2000). 6. The Cancer Genome Atlas Network, Nature 487, 65. M. G. Vander Heiden, L. C. Cantley, C. B. Thompson, 122. R. H. Medema, L. Macůrek, Oncogene 31,2601 330 (2012). Science 324,1029(2009). (2012). 7. C. Palles et al., Nat. Genet. 45,136(2013). 66. C. Lu et al., Nature 483,474(2012). 123. F. A. Derheimer, M. B. Kastan, FEBS Lett. 584,3675 8. P. C. Nowell, Science 194, 23 (1976). 67. S. Turcan et al., Nature 483,479(2012). (2010). 9. E. R. Fearon, B. Vogelstein, Cell 61,759(1990). 68. P. J. Stephens et al., Cell 144, 27 (2011). 124. G. Ciriello, E. Cerami, C. Sander, N. Schultz, Genome Res. 10. K. W. Kinzler, B. Vogelstein, Nature 386,761 69. A. J. Bass et al., Nat. Genet. 43,964(2011). 22,398(2012). (1997). 70. S. A. Tomlins et al., Science 310,644(2005). 125. C. H. Yeang, F. McCormick, A. Levine, FASEB J. 22,2605 11. S. Jones et al., Proc. Natl. Acad. Sci. U.S.A. 105, 71. M. Soda et al., Nature 448,561(2007). (2008). 4283 (2008). 72. P. Armitage, R. Doll, Br. J. Cancer 8,1(1954). 126. S. V. Sharma, D. W. Bell, J. Settleman, D. A. Haber, 12. I. Bozic et al., Proc. Natl. Acad. Sci. U.S.A. 107,18545 73. R. Böttcher et al., Nucleic Acids Res. 40, e125 (2012). Nat. Rev. Cancer 7,169(2007). (2010). 74. M. Forster et al., Nucleic Acids Res. 41,e16(2013). 127. H. L. McLeod, Science 339,1563(2013). 13. C. Tomasetti, B. Vogelstein, G. Parmigiani, Proc. Natl. 75. J. Ding et al., Bioinformatics 28,167(2012). 128. D. W. Brock, Oncologist 15 (suppl. 1), 36 (2010). Acad. Sci. U.S.A. 110,1999(2013). 76. M. A. Beal, T. C. Glenn, C. M. Somers, Mutat. Res. 129. M. A. Lemmon, J. Schlessinger, Cell 141,1117 14. E. Laurenti, J. E. Dick, Ann. N.Y. Acad. Sci. 1266, 750, 96 (2012). (2010). 68 (2012). 77. F. Mertes et al., Brief. Funct. Genomics 10,374 130. M. R. Arkin, J. A. Wells, Nat. Rev. Drug Discov. 3,301 15. J. S. Welch et al., Cell 150,264(2012). (2011). (2004). 16. S. Yachida et al., Nature 467,1114(2010). 78. M. Gundry, J. Vijg, Mutat. Res. 729,1(2012). 131. R. J. Bienstock, Curr. Pharm. Des. 18,1240(2012). 17. R. S. Kerbel, Adv. Cancer Res. 55, 87 (1990). 79. A. V. Biankin et al., Nature 491,399(2012). 132. A. Besson, S. F. Dowdy, J. M. Roberts, Dev. Cell 14,159 18. R. Bernards, R. A. Weinberg, Nature 418,823 80. H. Yan, K. W. Kinzler, B. Vogelstein, Science 289, (2008). (2002). 1890 (2000). 133. P. J. Morin et al., Science 275,1787(1997). 19. M. Yu, S. Stott, M. Toner, S. Maheswaran, D. A. Haber, 81. F. W. Huang et al., Science 339,957(2013). 134. T. C. He et al., Science 281,1509(1998). J. Cell Biol. 192,373(2011). 82. S. Horn et al., Science 339,959(2013). 135. M. van de Wetering et al., Cell 111,241(2002). 20. J. Komori, L. Boone, A. DeWard, T. Hoppo, E. Lagasse, 83. W. Xue et al., Proc. Natl. Acad. Sci. U.S.A. 109,8212 136. H. Farmer et al., Nature 434,917(2005).

Nat. Biotechnol. 30,976(2012). (2012). 137. S. Irshad, A. Ashworth, A. Tutt, Expert Rev. Anticancer Ther. on April 30, 2013 21. M. Pelizzola, J. R. Ecker, FEBS Lett. 585,1994 84. N. L. Solimini et al., Science 337,104(2012). 11,1243(2011). (2011). 85. A. D. Beggs et al., J. Pathol. 10.1002/path.4132 138. D. A. Grueneberg et al., Proc. Natl. Acad. Sci. U.S.A. 105, 22. G. Parmigiani et al., Genomics 93, 17 (2009). (2012). 16472 (2008). 23. M. Meyerson, S. Gabriel, G. Getz, Nat. Rev. Genet. 11, 86. A. P. Feinberg, B. Tycko, Nat. Rev. Cancer 4,143 139. M. Mao et al., Clin. Cancer Res. 19,657(2013). 685 (2010). (2004). 140. J. M. Kirkwood et al., CA Cancer J. Clin. 62,309 24. H. Carter et al., Cancer Res. 69,6660(2009). 87. P. A. Jones, S. B. Baylin, Cell 128,683(2007). (2012). 25. A. Youn, R. Simon, Bioinformatics 27,175(2011). 88. M. Höglund, D. Gisselsson, T. Säll, F. Mitelman, Cancer 141. T. Barrett et al., Nucleic Acids Res. 39,D1005(2011). 26. J. S. Kaminker, Y. Zhang, C. Watanabe, Z. Zhang, Genet. Cytogenet. 135,103(2002). 142. N. H. Segal et al., Cancer Res. 68,889(2008). Nucleic Acids Res. 35,W595(2007). 89. D. Shibata, J. Schaeffer, Z. H. Li, G. Capella, M. Perucho, 143. J. C. Castle et al., Cancer Res. 72,1081(2012). 27. J. J. Michaelson et al., Cell 151,1431(2012). J. Natl. Cancer Inst. 85,1058(1993). 144. J. H. Sampson et al., J. Clin. Oncol. 28,4722

28. S. Nik-Zainal et al., Cell 149,979(2012). 90. A. Sottoriva, I. Spiteri, D. Shibata, C. Curtis, S. Tavaré, (2010). www.sciencemag.org 29. S. Thiagalingam et al., Nat. Genet. 13,343(1996). Cancer Res. 73, 41 (2013). 145. H. Matsushita et al., Nature 482,400(2012). 30. S. A. Forbes et al., Nucleic Acids Res. 39,D945 91. S. P. Shah et al., Nature 461,809(2009). 146. M. DuPage, C. Mazumdar, L. M. Schmidt, A. F. Cheung, (2011). 92. K. Anderson et al., Nature 469,356(2011). T. Jacks, Nature 482,405(2012). 31. H. Yan et al., N. Engl. J. Med. 360,765(2009). 93. N. Navin et al., Nature 472, 90 (2011). 147. M. Challa-Malladi et al., Cancer Cell 20,728(2011). 32. S. Zhao et al., Science 324,261(2009). 94. S. Nik-Zainal et al., Cell 149,994(2012). 148. F. S. Hodi et al., N. Engl. J. Med. 363,711(2010). 33. P. S. Ward et al., Cancer Cell 17,225(2010). 95. M. Gerlinger et al., N. Engl. J. Med. 366,883 149. S. L. Topalian et al., N. Engl. J. Med. 366,2443 34. L. Dang et al., Nature 465,966(2010). (2012). (2012). 35. W. S. Pear, J. C. Aster, Curr. Opin. Hematol. 11, 96. X. Xu et al., Cell 148,886(2012). 150. B. K. Dunn, K. Jegalian, P. Greenwald, Recent Results 426 (2004). 97. P. J. Campbell et al., Nature 467,1109(2010). Cancer Res. 188, 21 (2011). 36. M. Nicolas et al., Nat. Genet. 33,416(2003). 98. N. Wagle et al., J. Clin. Oncol. 29,3085(2011). 151. L. C. Lopes, S. Barberato-Filho, A. C. Costa, C. G. S. Osorio- Downloaded from 37. A. P. Weng et al., Science 306,269(2004). 99. N. L. Komarova, D. Wodarz, Proc. Natl. Acad. Sci. U.S.A. de-Castro, Rev. Saude Publica 44, 620 (2010). 38. N. Agrawal et al., Science 333,1154(2011). 102,9714(2005). 152. G. A. Colditz, K. Y. Wolin, S. Gehlert, Sci. Transl. Med. 39. N. Stransky et al., Science 333,1157(2011). 100. A. B. Turke et al., Cancer Cell 17, 77 (2010). 4,127rv4(2012). 40. N. Agrawal et al., Cancer Discovery 2,899(2012). 101. R. Durrett, S. Moseley, Theor. Popul. Biol. 77, 42 (2010). 41. D. W. Parsons et al., Science 331,435(2011). 102. L. A. Diaz Jr. et al., Nature 486,537(2012). Acknowledgments: We thank M. Nowak and I. Bozic for 42. L. Pasqualucci et al., Nat. Genet. 43,830(2011). 103. A. Kreso et al., Science 339,543(2013). critical reading of the manuscript, S. Gabelli for assisting 43. R. D. Morin et al., Nature 476,298(2011). 104. P. J. Stephens et al., Nature 486,400(2012). with the production of Fig. 8, and A. Dixon, V. Ferranta, 44. C. S. Grasso et al., Nature 487,239(2012). 105. The Cancer Genome Atlas Network, Nature 490,61 and E. Cook for artwork. This work was supported by The 45. M. J. Ellis et al., Nature 486,353(2012). (2012). Virginia and D.K. Ludwig Fund for Cancer Research; The 46. K. C. Wiegand et al., N. Engl. J. Med. 363,1532 106. H. Pang et al., Cancer Res. 69,8868(2009). Lustgarten Foundation for Pancreatic Cancer Research; and (2010). 107. J. Chen, Y. Ye, H. Sun, G. Shi, Cancer Chemother. NIH grants CA 43460, CA 47345, CA 62924, and CA 121113. 47. S. Jones et al., Science 330,228(2010). Pharmacol. 71,265(2013). All authors are Founding Scientific Advisors of Personal 48. S. Jones et al., Hum. Mutat. 33,100(2012). 108. P. B. Chapman et al., N. Engl. J. Med. 364,2507 Genome Diagnostics (PGDx), a company focused on the 49. K. Wang et al., Nat. Genet. 43,1219(2011). (2011). identification of genetic alterations in human cancer for 50. J. Huang et al., Nat. Genet. 44,1117(2012). 109. E. L. Kwak et al., N. Engl. J. Med. 363,1693(2010). diagnostic and therapeutic purposes. All authors are also 51. M. Imielinski et al., Cell 150,1107(2012). 110. M. Ljungman, D. P. Lane, Nat. Rev. Cancer 4,727 members of the Scientific Advisory Board of Inostics, a 52. C. M. Rudin et al., Nat. Genet. 44,1111(2012). (2004). company that is developing technologies for the molecular 53. F. Delhommeau et al., N. Engl. J. Med. 360,2289 111. N. Perrimon, C. Pitsouli, B. Z. Shilo, Cold Spring Harb. diagnosis of cancer. All authors own stock in PGDx and (2009). Perspect. Biol. 4,a005975(2012). Inostics. The terms of these arrangements are being 54. J. Schwartzentruber et al., Nature 482,226(2012). 112. R. S. Kerbel, N. Engl. J. Med. 358,2039(2008). managed by Johns Hopkins University, in accordance 55. G. Wu et al., Nat. Genet. 44,251(2012). 113. A. S. Chung, N. Ferrara, Annu. Rev. Cell Dev. Biol. 27, with their conflict-of-interest policies. 56. T. J. Ley et al., N. Engl. J. Med. 363,2424(2010). 563 (2011). 57. G. L. Dalgliesh et al., Nature 463,360(2010). 114. J. W. Baish et al., Proc. Natl. Acad. Sci. U.S.A. 108, Supplementary Materials 58. M. L. Suvà, N. Riggi, B. E. Bernstein, Science 339, 1799 (2011). www.sciencemag.org/cgi/content/full/339/6127/1546/DC1 1567 (2013). 115. N. E. Hynes, H. A. Lane, Nat. Rev. Cancer 5,341(2005). 59. E. Papaemmanuil et al., N. Engl. J. Med. 365,1384 116. N. Turner, R. Grose, Nat. Rev. Cancer 10,116 Tables S1 to S5 (2011). (2010). 10.1126/science.1235122

1558 29 MARCH 2013 VOL 339 SCIENCE www.sciencemag.org