Characterising Somatic Mutations in Cancer Genome by Means of Next-generation Sequencing Mei Ling Chong,

Advanced article Article Contents . Introduction . Targeted Sequencing . WES . WGS . Challenges and Conclusion

Online posting date: 15th February 2012

Cancer Science Institute of Singapore, National University of Singapore,

Singapore

Chee Seng Ku, Cancer Science Institute of Singapore, National University of Singapore, Singapore

Mengchu Wu, Cancer Science Institute of Singapore, National University of Singapore, Singapore

Richie Soong, Cancer Science Institute of Singapore, National University of Singapore, Singapore

Cancer genome sequencing studies have identified tens of thousands of somatic mutations in various human cancers to date. This data has started to generate new insights into mutation patterns and their differences between various cancers. Further, the mutation patterns have also helped to elucidate mechanism involved in generating mutations in cancer genome such as deoxyribonucleic acid (DNA)-repair processes and mutagen exposures. With the introduction of next-generation sequencing technologies, cancer genome sequencing has evolved from a targeted sequencing approach to whole-exome sequencing and whole-genome sequencing (WGS) approaches. However, each sequencing approach has its strengths and limitations. It is widely anticipated that WGS would eventually replace targeted and exome sequencing. WGS offers a unique advantage to study structural variants or rearrangements and fusion genes in a single experiment, in addition to point mutations. However, currently the WGS is still prohibitively expensive for a large number of samples. Despite these technological advances, several challenges still remain, such as discerning driver mutations from benign mutations and

eLS subject area: Genetics & Disease How to cite: Chong, Mei Ling; Ku, Chee Seng; Wu, Mengchu; and Soong, Richie (February 2012) Characterising Somatic Mutations in Cancer Genome by Means of Next-generation Sequencing. In: eLS. John Wiley & Sons, Ltd: Chichester. DOI: 10.1002/9780470015902.a0023379

collection of high-quality primary tumour tissues to minimise tissue heterogeneity. Ultimately, a comprehensive delineation of the somatic mutations in the cancer genome would require WGS of a large number of samples from various cancer types and subtypes. Congruent to this goal, the International Cancer Genome Consortium was initiated and upon completion of the project, its data is expected to further enhance our knowledge and understanding of the biological mechanisms underlying cancer development.

Introduction Identifying somatic mutations in cancer genomes has important implications for personalised cancer medicine. These mutations include single nucleotide substitutions or variations (point mutations), small insertions and deletions (indels), and larger copy number variations and structural rearrangements (Robison, 2010; Stratton, 2011). Mutations in both oncogenes and tumour suppressor genes play an important role in the pathogenesis of cancer; hence identification of these mutations has been a major focus in recent cancer genetics research. Notably, gefitinib was developed to target EGFR as a treatment for lung cancer, with the mutation status of EGFR also used to predict the therapeutic responses (Paez et al., 2004). In a similar vein, the discovery of a high mutation frequency in PIK3CA (Samuels et al., 2004) has also made the PI3K pathway one of the most attractive targets for targeted therapy. Apart from point mutations, structural rearrangement is also a common mechanism that mediates the formation of an

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

1

Characterising Cancer Genome by Means of Next-generation Sequencing

‘oncogenic product’. These rearrangements usually lead to the formation of a fusion gene, which occur more frequent than previously thought in human solid cancers (Mitelman et al., 2004). Some of these fusion genes result in activation of a protein that contributes to oncogenesis, such as the BCR-ABL fusion gene which is targeted by imatinib in the treatment of chronic myeloid leukemia (Stephens et al., 2009). The ETS and EML4-ALK fusion genes were recently discovered in prostate cancer (Tomlins et al., 2005, 2007) and lung adenocarcinoma (Soda et al., 2007), respectively, and are also potential targets for the development of new cancer drugs. These examples have illustrated the importance of identifying various cancer-causing somatic mutations, and thus have prompted the investigation of the cancer genome through various sequencing approaches. As noted earlier, a comprehensive characterisation of the somatic mutation profile of the cancer genome will not only help to understand the biological mechanism underlying cancer development, but also identify novel targets for cancer treatment. The number of human cancer genome sequencing studies published over the past few years has increased, driven mainly by the technological advances in sequence capture or enrichment methods, and next-generation sequencing (NGS) technologies. Several sequencing approaches to decipher the somatic mutation profile of the cancer genome are available, including targeted sequencing, whole-exome sequencing (WES) and whole-genome sequencing (WGS). Table 1 summarises the large-scale targeted sequencing (up to hundreds of genes), WES and WGS studies (as on 31 October 2011) through a PubMed search. In this chapter, we review and discuss the findings of studies identifying somatic mutations of different cancers using these sequencing approaches. We also elaborate on the strengths and limitations of each approach in interrogating the cancer genome.

Targeted Sequencing Targeted sequencing is a hypothesis-driven approach where candidate genes are often selected based on knowledge of previously reported mutated genes or their functional relevance in cancer development, such as the kinome or phosphatome. These targeted sequencing studies were previously performed using polymerase chain reaction (PCR)-based Sanger sequencing. However, the advent of custom-enrichment methods and NGS technologies has made this targeted sequencing approach more technically feasible. Up to several hundreds of genes were sequenced in these studies for different cancer types (Dalgliesh et al., 2010; Ding et al., 2008; Kan et al., 2010). Compared to WES and WGS, the ability to study different cancers and their subtypes is an important advantage of the targeted sequencing approach. This has been demonstrated in a study of 441 human primary tumour samples comprising breast (n=183), lung (n=134), ovarian 2

(n=58), prostate (n=58) and pancreatic (n=8) cancer types and their subtypes (Kan et al., 2010). For example, different subtypes of lung cancer were investigated, such as nonsmall-cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cancer and small cell lung cancer. However, as a ‘compromise’ to the large sample size that was being investigated, this study targeted only the coding exons and flanking splice sites of 1507 genes, which comprised known cancer genes and druggable genes such as protein kinases, E3 ligases, deubiquitinases and G-protein-coupled receptors. It is widely anticipated that the somatic mutation profile in different cancers and their subtypes differs, thus it is important to dissect their genetic heterogeneity. This also has important clinical implications, as genetic heterogeneity in tumour cells is often the cause of patients’ variability in drug responses. Indeed, mutation rates and the sets of mutated genes have been found to vary substantially across cancer types and subtypes. More specifically, Kan et al. found that an average of 1.8 protein-altering mutations per megabase (Mb) of deoxyribonucleic acid (DNA) of tumours analysed, however individual cancers and subtypes showed wide deviations from this background rate. In particular, lung adenocarcinomas and squamous carcinomas were found to have a higher protein-altering mutation rate of 3.5 and 3.9 per Mb, respectively. In contrast, prostate cancers, where  75% carried the TMPRSS2–ERG gene fusion, had a low mutation rate of 0.33 per Mb (Kan et al., 2010). These results are in agreement with an earlier study where comparison between cancer types also revealed that the number of somatic mutations per Mb (or somatic mutation prevalence) ranked highest for lung carcinoma (4.21 per Mb), gastric cancer (2.10 per Mb) and ovarian cancer (1.85 per Mb) (Greenman et al., 2007). Although these studies have documented differences in mutation rates, comparison between studies is challenging as a different set of genes was targeted by these studies. Further, another advantage of studying a large sample size is the ability to identify recurrent mutations (i.e. identical mutations affecting multiple samples) or highly mutated genes (i.e. different mutations affecting the same gene in different samples). Indeed, a total of 37 recurrent mutations distributed in 13 genes were found in the tumours analysed, which included novel mutations in EPHB1, GSK3B and RUNX1T1 (Kan et al., 2010). This also allowed the identification of 77 significantly mutated cancer genes (i.e. having significant prevalence of proteinaltering mutations). This set of significantly mutated genes varied across cancer types and subtypes, where 19 different genes were identified in lung squamous carcinoma, 18 in lung adenocarcinoma and 5 in small cell lung cancer. For example, a G-protein-coupled receptor such as GRM8 was mutated in 8% of NSCLC squamous subtypes and GRM1 in 7% of NSCLC adenocarcinomas (Kan et al., 2010). Similar findings were also observed in an independent study of 188 human lung adenocarcinomas. A total of 26 genes that were mutated at significantly high frequencies

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

Characterising Cancer Genome by Means of Next-generation Sequencing

Table 1 Summary of targeted sequencing, whole-exome sequencing (WES), whole-transcriptome sequencing (WTS) and wholegenome sequencing (WGS) studies of different cancer types and their sample sizes Sequencing approach (number of genes sequenced) Targeted (1507 genes) Targeted (18 191 genes) Targeted (20 661 genes) Targeted (3544 genes) Targeted (518 genes) Targeted (623 genes) Targeted (86 genes) Targeted (20 661 genes) WES WES WES WES WES WES WES WES WES WES WES WES WGS WGS WGS WGS WGS and WTS WGS WGS WGS WGS WGS WGS WGS WGS WGS+WES

Cancer type

Sample size

References

Different cancer types Breast and colorectal cancer Glioblastoma Renal carcinoma Different cancer types Lung cancer Melanoma Pancreatic cancer Acute myeloid leukemia Bladder cancer Chronic lymphocytic leukemia Diffuse large B-cell lymphoma Gastric cancer Hairy cell leukemia Head and neck squamous cell carcinoma Head and neck squamous cell carcinoma Hepatocellular carcinoma Melanoma Renal carcinoma Uterine leiomyomas Acute myeloid leukemia Acute myeloid leukemia Acute myeloid leukemia Breast cancer Breast cancer Breast cancer Chronic lymphocytic leukemia Hepatocellular carcinoma Lung cancer Lung cancer Lung cancer Melanoma Prostate cancer Multiple myeloma

441 22 22 101 (5 cell lines) 210 188 29 24 14 9 5 6 22 1 32

Kan et al. (2010) Wood et al. (2007) Parsons et al. (2008) Dalgliesh et al. (2010) Greenman et al. (2007) Ding et al. (2008) Prickett et al. (2009) Jones et al. (2008) Yan et al. (2011) Gui et al. (2011) Fabbri et al. (2011) Pasqualucci et al. (2011) Wang et al. (2011b) Tiacci et al. (2011) Agrawal et al. (2011)

74

Stransky et al. (2011)

10 14 7 18 1 1 1 24 (9 cell lines) 1 1 4 1 1 2 cell line 1 cell line 1 cell line 7 38

Li et al. (2011) Wei et al. (2011) Varela et al. (2011) Makinen et al. (2011) Ley et al. (2008) Mardis et al. (2009) Link et al. (2011) Stephens et al. (2009) Shah et al. (2009) Ding et al. (2010) Puente et al. (2011) Totoki et al. (2011) Lee et al. (2010) Campbell et al. (2008) Pleasance et al. (2010b) Pleasance et al. (2010a) Berger et al. (2011) Chapman et al. (2011)

WES: whole-exome sequencing; WGS: whole-genome sequencing; WTS: whole-transcriptome sequencing.

were identified in lung adenocarcinomas where the coding exons and splice sites of 623 candidate cancers were sequenced (Ding et al., 2008). Furthermore, 12 genes were found with significantly higher frequencies of nonsense, splice-site and frameshift mutations, suggesting that they are tumour suppressor genes affected by mutations leading to truncated proteins. Although this study only investigated lung adenocarcinomas without comparison to other lung cancer subtypes, the large sample size demonstrated that lung adenocarcinomas are heterogeneous, that is, the samples were found to harbour diverse combinations of mutations, but they are common in the main pathways affected by these mutations. Indeed, genetic alterations in

lung adenocarcinoma frequently occurred in genes of the MAPK signalling, p53 signalling, Wnt signalling, cell cycle and mTOR pathways (Ding et al., 2008). Through another interesting observation, the somatic mutations in the coding exons of 3544 genes have also been investigated in 101 renal cell carcinoma samples (Dalgliesh et al., 2010). The large sample size also allowed the study to prioritise genes with two or more nonsynonymous mutations for follow-up studies in 311 primary renal cell carcinoma samples comprising 246 clear cell renal cell carcinoma and 65 other samples of nonclear-cell histology. The combined initial and follow-up studies identified five genes under ‘positive selection’ (i.e. SETD2, JARID1C,

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

3

Characterising Cancer Genome by Means of Next-generation Sequencing

NF2, UTX and MLL2), which is a clustering of somatic mutations in these genes, suggesting a role in cancer development. All except MLL2 were plausible functional candidates as they had strong evidence for selection by truncating mutations. More interestingly, they are histone modifying genes. Notably, 3% of clear cell renal carcinoma had somatic truncating mutations in SETD2, which encodes a histone H3K36 methyltransferase, and 3% had truncating mutations in JARID1C, which encodes a histone H3K4 demethylase. By contrast, no mutations were found in SETD2 or JARID1C in the subset of nonclear-cell cancers highlighting differences between subtypes. This two-stage study design was also applied in melanoma samples (Prickett et al., 2009). In the first stage, exons comprising the kinase domains of all 86 members of this gene superfamily (protein tyrosine kinases) were sequenced in 29 samples. Subsequently, 19 genes which contained a total of 30 somatic mutations affecting the kinase domains were examined in additional 79 melanoma samples. This led to the identification of ERBB4 as the most highly mutated gene where 19% of the samples were found to have mutations in this gene. See also: Protein Kinases: Signatures in Cancer Collectively, these large-scale targeted sequencing studies have demonstrated their feasibility in interrogating a large sample size. The strength of a large sample size lies in its ability to (1) identify recurrent mutations and highly mutated genes, (2) delineate genetic heterogeneity within a cancer type and (3) compare the similarities and differences of somatic mutation patterns between different cancers and their subtypes. These studies have also lent further support to the notion of genetic heterogeneity within and between cancer types. However, this targeted sequencing approach is deemed ‘incomplete’ in delineating the somatic mutation profile of the cancer genome. It is not only that the noncoding regions were not studied but also that the entire set of genes was incompletely investigated. This has important implications in interpreting the results. For example, Kan et al. have estimated the mutation rates for various cancers and their subtypes and some differences were documented. However, this should be interpreted in the context of the 1507 genes sequenced in the study (Kan et al., 2010). Furthermore, targeted sequencing studies have to be supplemented with microarray experiments to interrogate structural aberrations. Additional experiments using the comparative genome hybridisation array (aCGH) were performed to obtain copy number alteration profiles. This led to several significantly mutated genes and genes harbouring mutations predicted to have functional effects being found to be amplified in the 441 samples of various cancer types, indicating that these genes may function as oncogenes (Kan et al., 2010). Similarly, several known tumour suppressor genes such as PTEN and CDKN2A were frequently deleted in multiple cancers, thus expanding the role of these genes in additional tumour types, which is an advantage of studying different cancers and subtypes. Additionally, several significantly mutated genes were also found in regions of copy number gain (EGFR and KRAS) 4

or loss (CDKN2A, PTPRD and RB1) in lung adenocarcinoma (Ding et al., 2008). See also: Comparative Genomic Hybridization

WES Although targeted sequencing studies have produced interesting results, the limitation that only a subset of genes was investigated must also be recognised. The ascertainment bias in the selection of genes may lead to overlooking key mutations in the cancer genome. Thus, this led to the concept of the WES approach, which is now more technically feasible with the developments of whole-exome enrichment kits and NGS technologies. However, the initial WES studies were conducted using traditional PCR and Sanger sequencing methods (Jones et al., 2008; Parsons et al., 2008; Sjoblom et al., 2006; Wood et al., 2007). These studies have sequenced almost all the Consensus Coding Sequence (CCDS) and RefSeq genes. PCR-based methods were used to amplify the targeted regions and then subjected to Sanger sequencing. However, this is laborious, for example, a total of 125 624 PCR primers was deigned to amplify 6196 RefSeq transcripts (Wood et al., 2007). This is not technically efficient when the entire set of genes is sequenced and it is also not scalable to a large sample size. Nevertheless, these early studies have provided important information on the patterns of somatic mutations in various cancers. See also: Cancer Genome Sequencing The development of multiple commercial exome enrichment kits has circumvented the technical challenges in isolating the entire exome for sequencing. These exome enrichment kits have comprehensive coverage of the CCDS and RefSeq genes, but are varied in their extent of coverage (Clark et al., 2011; Parla et al., 2011; Sulonen et al., 2011). More specifically, the Illumina TruSeq Exome Enrichment Kit (an in-solution sequence capture method for isolating exonic regions using hybrid selection) covers 97.2% of CCDS coding exons (31.3 Mb, hg19) and 96.4% of RefSeq (regGene) coding exons (33.2 Mb, hg19). In addition to comprehensive coverage of the major exon databases, this enrichment kit also provides broad coverage of noncoding DNA in exon-flanking regions (promoters and untranslated regions (UTRs)), and 77.6% of the predicted microribonucleic acid (microRNA) targets (9.0 Mb, hg19) were also captured (http://www.illumina.com/products/truseq_exome_enrichment_kit.ilmn). Thus, these enrichment methods coupled with the existing NGS technologies have made WES more technically feasible and more cost-effective. As a result, a number of cancer sequencing studies have leveraged this technological advancement in interrogating the somatic mutational profile in various cancers. Table 2 summarises the genes harbouring recurrent mutations and/ or highly mutated genes identified by WES studies. Each WES study has identified hundreds of somatic mutations and putative cancer genes. Some of these studies have adopted a two-stage study design with a small

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

Characterising Cancer Genome by Means of Next-generation Sequencing

Table 2 Summary of highly mutated genes or recurrent mutations identified by whole-exome sequencing (WES) Cancer type

Highly mutated genes/recurrent mutations

References

Breast and colorectal cancer Glioblastoma

PIK3CA, FBXW7, KRAS, APC, TP53 TP53, PTEN, CDKN2A, RB1, EGFR, NF1, PIK3CA, PIK3R1, IDH1, CDK4 KRAS, NRAS, TP53, CCND1, DIS3, FAM46C, XBP1, LRRK2, PNRC1, HLA-A, MAGED1, IRF4 ATP2A2, C10orf2, CCND3, DNMT3A, FLT3, GATA2, MLL, NSD1, NPM1, NRAS UTX, TP53, CREBBP, EP300, ARID1A, RB1, HRAS, ERBB3, CHD6, LRP2, FGFR3, MLL, LAMA4, ANK2, NF1, ESPL1, NCOR1, KRAS, MLL3, ANK3, ZFHX3 BRAF ADAMTSL3, AKAP8, ANKLE2, B2M, BCL2, C12orf35, CARD11, CCND3, CD36, CD58, CD79B, CREBBP, DCHS1, DPYD, DSC3, DUSP27, EP300, EZH2, HNF1B, KDM2B, KLF2, MAGEC3, MED12L, MEF2B, MLL2, MTMR8, MYC, MYD88, MYOM2, NOTCH1, OFD1, PIM1, PMS1, PRDM1, TLL2, TMEM30A, TNFAIP3, TP53, TSC22D1 CTNNB1, TP53, ARID2, DMXL1, NLRP1 AKR1B10, BRAF, C12orf63, CCDC63, GRIN2A, KHDRBS2, PCDHB8, PLCB4, PTPRO, SLC17A5, SLC6A11, SYT4, TAS2R60, TMEM132B, UGT2B10, ZNF831 UTX, JARID1C, SETD2, PBRM1 NOTCH1,TGM7, BIRC3, PLEKHG,TP53 ARID1A MED12

Wood et al. (2007) Parsons et al. (2008)

Pancreatic cancer

Acute myeloid leukemia Bladder cancer

Hairy cell leukemia Diffuse large B-cell lymphoma

Hepatocellular carcinoma Melanoma

Renal carcinoma Chronic lymphocytic leukemia Gastric cancer Uterine leiomyomas

Jones et al. (2008)

Yan et al. (2011) Gui et al. (2011)

Tiacci et al. (2011) Pasqualucci et al. (2011)

Li et al. (2011) Wei et al. (2011)

Varela et al. (2011) Fabbri et al. (2011) Wang et al. (2011b) Makinen et al. (2011)

Note: This is not a complete gene list as reported by the studies. Please refer to the original article for further information of the definition of highly mutated genes/recurrent mutations in each study.

discovering sample set in the exome sequencing and subsequent screening of the mutations or promising candidate genes in a larger validation set. Of note, WES of nine transitional cell carcinomas with a follow-up study of all the somatically mutated genes in an additional 88 samples identified mutations in multiple chromatin remodelling genes in 59% of the total 97 samples (Gui et al., 2011). Mutations in the genes involved in the chromatin remodelling process were also reported in other cancers (Dalgliesh et al., 2010; Li et al., 2011; Varela et al., 2011), for example, novel inactivating mutations of ARID2 and ARID1A were discovered in hepatocellular carcinoma and gastric cancer through WES (Li et al., 2011; Wang et al., 2011b). Although WES interrogates only approximately 1–2% of the entire human genome, the several hundred somatic mutations identified have already prohibited all of them to be examined by follow-up studies in a larger sample set. As such, prioritisation criteria would be needed to select a subset of somatic mutations or genes for further studies. This was demonstrated in the WES study of transitional

cell carcinoma of the bladder where 465 predicted somatic mutations were identified in the ‘discovery phase’ (Gui et al., 2011). However, subsequent investigations focused on 328 genes that had at least one nonsilent somatic mutation and their mutation prevalence in 88 additional samples. The combined analysis of the discovery and further studies identified 54 significantly mutated genes. It is also noteworthy that the definition of ‘significantly mutated genes’ differs from one study to another. In this study, a significantly mutated gene was identified if the gene harboured confirmed nonsilent mutations in at least two tumours and if its nonsilent mutation rate was significantly higher than the background. Thus, a direct comparison of the results across the same cancer or different cancers from different studies is inappropriate due to the methodological inconsistencies. The identification of previously wellknown genes for transitional cell carcinoma of the bladder such as TP53, RB1, HRAS, FGFR3 and KRAS, lends further credibility to the approach. Furthermore, the discovery of 49 new frequently mutated genes has

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

5

Characterising Cancer Genome by Means of Next-generation Sequencing

demonstrated that WES is a powerful information generation and discovery tool (Gui et al., 2011). Through another interesting observation, 68 genes that appeared to be somatically mutated at elevated frequency, of which many are not known to be genetically altered in melanoma, were also identified by the WES approach (Wei et al., 2011). Most of the WES studies have employed an almost similar study design, analytical approach and prioritisation pipeline, however, to a different extent of challenges and difficulties. Wei et al. prioritised genes for further scrutiny by searching for new recurrent mutations that occurred in at least two of the 14 samples subjected to WES. Several genes were found to harbour recurrent mutations. Follow-up investigation in an additional 153 melanoma samples identified TRRAP, which harboured a recurrent mutation in approximately 4% of the samples. This recurrent mutation is likely to be functionally important because the likelihood of the occurrence of six identical mutations (i.e. 6 out of 167 samples) is extremely low. Similarly, WES of hepatitis C virus (HCV)-associated hepatocellular carcinomas (HCC) identified five genes, which were somatically mutated in more than one sample out of 10 samples. These genes were selected for further analysis in additional samples and led to the discovery of novel inactivating mutations of ARID2. The relevance of these ARID2 mutations was not limited to HCV-associated HCC, but also to HBV-associated HCC, alcohol-associated HCC and HCC with unknown etiology (Li et al., 2011). In summary, these WES studies have proven powerful to identify recurrent mutations and highly mutated genes. These putative cancer genes warrant subsequent biological functional studies. It is widely believed that the WES approach will be the transient technology in cancer genome sequencing in the next few years, because of its costeffectiveness, being less challenging analytically and having the ability to study a larger sample size than WGS. Although the cost of WGS provided as a commercial service is now below USD5000, the cost incurred for data storage and analysis is still substantial, with the total cost prohibitive for individual laboratories.

WGS The advances of NGS technologies and rapidly declining sequencing costs have enabled the completion of a number of WGS studies of various cancers. A pioneering study was published in 2008, which sequenced the entire genome of acute myeloid leukaemia (AML) with normal cytogenetics (Ley et al., 2008). This and subsequent WGS studies identified an average of up to tens of thousands of somatic mutations in a single cancer genome. These studies also showed that the number of somatic mutations varies widely both within and between types of cancer. Although the numbers varied, the highest number of somatic mutations was identified in lung cancer (Lee et al., 2010; Pleasance et al., 2010b) and melanoma (Pleasance et al., 2010a), in which more than 20 000 somatic mutations were found. 6

The high mutation rate in lung cancer and melanoma supports the notion that recurrent mutagen exposure to tobacco and UV radiation mediates the occurrence of the mutations. On the other hand, approximately 1000 and 7450 somatic mutations per tumour were identified in chronic lymphocytic leukemia (CLL) (Puente et al., 2011) and multiple myeloma (Chapman et al., 2011), respectively. The number of somatic mutations per tumour identified in other solid tumours such as prostate (Berger et al., 2011) and breast cancer (Ding et al., 2010; Shah et al., 2009; Stephens et al., 2009) are comparable to that observed in nonsolid tumours (Chapman et al., 2011; Mardis et al., 2009; Puente et al., 2011). As noted earlier, differences in the number of mutations identified in these studies could be as a consequence of technical and analytical differences between the studies. As a result, a direct comparison of the final results from different studies is not methodologically appropriate. The patterns of somatic mutations provided by WGS are useful in elucidating the mechanisms such as DNA-repair processes and differences in mutagen exposure in generating the mutations in the cancer genome. The most well-established mutation spectrum that have been identified to date are in lung cancer and melanoma. Single nucleotide substitutions such as G/C4T/A transversions are the most frequent changes observed in lung cancer (Lee et al., 2010; Pleasance et al., 2010b). It was shown that G to T transversions were strongly targeted to the nontranscribed DNA strand of the most active genes, which in turn could be eliminated from the transcribed DNA strand by transcription-coupled repair. Moreover, these transversions often occurred within CpG dinucleotides, which is consistent with the known profile of TP53 mutation in lung cancers associated with smoking history (Hainaut et al., 2001). On the other hand, sequencing of melanoma cell lines (COLO-829) and tumour samples revealed mutation patterns characteristic of UV radiation, where most of the somatic base substitutions were C4T/G4A transition (Pleasance et al., 2010a; Wei et al., 2011). Among these mutations, 92% occurred at the 3’ base of pyrimidine dinucleotides, whereas CC-to-TT double substitutions represented 10% of all mutations. Both of these mutation types are characteristics of UVB-induced DNA damage (Pfeifer and Hainaut, 2011). Similarly, Puente et al. (2011) showed that the major substitution in CLL was G4A/C4T transition. Interestingly, they also found a different mutation spectrum between IGHV-mutant and IGHV-wildtype CLL, in which IGHV-mutated cases showed a higher proportion of A4C/T4G mutations than cases with wildtype IGHV. This might imply different molecular mechanisms during tumour development between the two groups. The context and patterns of mutations in IGHV-mutated cases was consistent with error-prone polymerase during the normal process of somatic hypermutation of IGHV genes (Puente et al., 2011). The biological basis of this mutational signature in other cancer types remains unknown and may be due to a defect in DNA repair or a shared mutagenic

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

Characterising Cancer Genome by Means of Next-generation Sequencing

exposure (Table 3). Comparison to the whole-genome mutation spectrum reported for HCC, T4C/A4G transition could be a characteristic mutational signature of HCV-associated cancer, which would be consistent with a previous observation that HCV induces error-prone DNA polymerases that preferentially cause the T4C/A4G mutation (Machida et al., 2004). A further advantage of WGS is its ability to identify structural rearrangements compared to targeted sequencing and WES (Berger et al., 2011; Link et al., 2011; Totoki et al., 2011; Yan et al., 2011). The cancer genome is also characterised by aberrations in somatic rearrangements (Stephens et al., 2009). However, the conventional methods to identify structural rearrangements such as G-banded cytogenetics, spectral karyotyping and fluorescence in situ hybridisation (FISH) are limited by their poor resolution. With the advent of multiple massively parallel sequencing-based strategies, an increasing number of structural rearrangements and fusion genes have been documented in human cancers (Table 4) (Campbell et al., 2008). For instance, fusion of DGKG-BST1 caused by a reciprocal translocation between chromosomes 3 and 4 was identified in one AML patient (Link et al., 2011). Similarly, four novel fusion genes (BCORL1-ELF4, CTNND1STX5, VCL-ADK and CABP2-LOC645332) were also found in hepatocellular carcinoma. Despite these novel findings, none of the fusion

genes were found in an additional validation set of tumours (Totoki et al., 2011). This could be due to the fact that only one sample was used in the discovery set (and the fusion genes might be unique to the samples). More interestingly, Berger et al. (2011) also reported a subset of chromosomal rearrangements showing multiple inter- and intrachromosomal location exchange breakpoint arms without any loss in total genetic material. The authors hypothesised that these events may be due to the simultaneous disruption of many co-localised chromosomes as this pattern is distinct from one in which all breaks occur as reciprocal pairs. The detection of structural rearrangements using NGS methods is more analytically challenging than point mutations. However, this could be improved through the development of more powerful analytical tools. Recently, Wang et al. (2011a) have developed an algorithm, ‘clipping reveals structure’ that uses NGS reads with partial alignments to a reference genome to directly map structural variations at the nucleotide level of resolution.

Challenges and Conclusion Although WES and WGS approaches are now technically feasible, several challenges still remain. These approaches have identified multiple novel mutations and genes for

Table 3 Summary of mutation spectra identified in different cancer types Cancer type

Spectrum

Mutation mechanism

References

Acute myeloid leukemia Bladder cancer Breast cancer Chronic lymphocytic leukemia Diffuse large B-cell lymphoma Multiple myeloma Hepatocellular carcinoma Lung cancer cell line, NCIH209 Lung cancer Melanoma cell line, COLO829 Melanoma Renal carcinoma

G4A/C4T transition C/G4T/A transition C/G4T/A transition G4A/C4T transition CpG transition CpG transition T4C/A4G transition C4T/C4A transversion

Unknown Unknown Unknown Unknown Unknown Unknown DNA repair Tobacco carcinogens

Link et al. (2011) Gui et al. (2011) Ding et al. (2010) Puente et al. (2011) Pasqualucci et al. (2011) Chapman et al. (2011) Totoki et al. (2011) Pleasance et al. (2010b)

G/C4T/A transversion C4T/G4A transition C4T/G4A transition C4T/G4A transition

Tobacco carcinogens UV exposure UV exposure UV exposure

Lee et al. (2010) Pleasance et al. (2010a) Prickett et al. (2009) Dalgliesh et al. (2010)

Table 4 Fusion genes identified in different cancer types Sequencing type WES WGS WGS

Cancer type Acute myeloid leukemia Acute myeloid leukemia Hepatocellular carcinoma

WGS WGS

Lung cancer Prostate cancer

Structural rearrangement MLL-MLLT4 DGKG-BST1 BCORL1-ELF4, CTNND1STX5, VCLADK, CABP2-LOC645332 CREBBP-BTBD12 TMPRSS2-ERG

References Yan et al. (2011) Link et al. (2011) Totoki et al. (2011)

Pleasance et al. (2010b) Berger et al. (2011)

WES: whole-exome sequencing; WGS: whole-genome sequencing. eLS & 2012, John Wiley & Sons, Ltd. www.els.net

7

Characterising Cancer Genome by Means of Next-generation Sequencing

various cancers, but validation using other methods is required as NGS approaches are prone to false-positive results. These methods include Sanger sequencing, Sequenom, real-time PCR or FISH. In addition, selecting the most promising putative cancer genes for subsequent biological functional studies represents another challenge. Most of the top priority candidates are recurrent mutations and highly mutated genes. A further challenge is to distinguish ‘driver’ and ‘passenger’ mutations. Driver mutations confer a growth advantage and may be involved in cancer development, whereas passenger mutations may just evolve during cancer progression without any functional roles (Schweiger et al., 2011; Stratton, 2011). The characteristics of driver mutations are poorly defined; however, mutations recurring in multiple samples suggest functional roles and are thus more likely to be driver mutations. Various bioinformatics and analytical tools have been developed to assess the functional consequences of somatic mutations. Despite this bioinformatics evidence, characterisation of somatic mutations in functional studies is still essential to confirm their role in tumourigenesis. Accurate identification of somatic mutations in primary tumour tissues is another critical challenge because of tissue heterogeneity due to the frequent contamination from noncancerous cells and the genetic heterogeneity between the tumour cells (Meyerson et al., 2010; Robison, 2010). In a hypothetical example to detect a heterozygous mutation, only 25% of sequence reads mapping to the locus are expected to carry the mutant allele in a clinical sample with 50% tumour content. Therefore, a much higher sequencing depth or coverage is needed to detect somatic mutations in primary tumour tissue than germline mutation. In this context, targeted sequencing and WES approaches could achieve a higher coverage than WGS, and may thus alleviate the noise resulting from poor sample purity and quality. Delineating the patterns of somatic mutations in the cancer genome requires a comprehensive interrogation of the cancer genome (the entire genome, exome or a large number of candidate genes) in an adequate sample size of up to hundreds of samples. Currently, WGS is limited by a small sample size whereas targeted sequencing is constrained by interrogating only a ‘small portion’ of the cancer genome. Thus, it is widely anticipated that WES will become a ‘transient technology’ in cancer genome sequencing until WGS becomes more cost-effective and analytically feasible for hundreds of samples. A large sample size comprised of different cancers and cancer subtypes is important to dissect the heterogeneity of their mutational profiles or patterns. Different cancers or cancer subtypes are likely to share some ‘common patterns’ and have ‘specific or unique patterns’ of somatic mutations that are responsible for different clinico-pathological features. Parallel to this aim, the International Cancer Genome Consortium was formed to sequence various cancer types and subtypes. In addition, integrative genomic analysis is desirable to dissect the complexities of cancer genomics. Integrative genomics is the discipline of integrating data 8

from multiple genomic experiments such as from transcriptomics and epigenomics experiments. This is not a new concept, but it is now greatly facilitated by NGS as these experiments can all be conducted by NGS-based methods. See also: Characterising Structural Variation by Means of Next-Generation Sequencing; Next Generation Sequencing Technologies and Their Applications Taken together, this scale of cancer sequencing project is beyond the technical and financial capacity of a single laboratory. Thus, the International Cancer Genome Consortium will eventually obtain a comprehensive description of genomic, transcriptomic and epigenomic changes in 50 different cancer types and subtypes (Hudson et al., 2010). Currently, the Consortium has received commitments from funding organisations in Asia, Australia, Europe and North America for 39 project teams to study over 18 000 tumour genomes. Projects that are currently funded are examining tumours affecting multiple organs such as the bladder, blood, bone, brain, breast, cervix and colon. It is foreseeable that the results generated by the consortium, upon completion of the project, will further enhance our knowledge of somatic mutation profiles of various cancers, the molecular basis of cancers by integrating various omics data and allow identification of potential molecular drug targets.

References Agrawal N, Frederick MJ, Pickering CR et al. (2011) Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 333: 1154– 1157. Berger MF, Lawrence MS, Demichelis F et al. (2011) The genomic complexity of primary human prostate cancer. Nature 470: 214–220. Campbell PJ, Stephens PJ, Pleasance ED et al. (2008) Identification of somatically acquired rearrangements in cancer using genome-wide massively parallel paired-end sequencing. Nature Reviews Genetics 40: 722–729. Chapman MA, Lawrence MS, Keats JJ et al. (2011) Initial genome sequencing and analysis of multiple myeloma. Nature 471: 467–472. Clark MJ, Chen R, Lam HY et al. (2011) Performance comparison of exome DNA sequencing technologies. Nature Biotechnology 29: 908–914. Dalgliesh GL, Furge K, Greenman C et al. (2010) Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463: 360–363. Ding L, Ellis MJ, Li S et al. (2010) Genome remodelling in a basallike breast cancer metastasis and xenograft. Nature 464: 999–1005. Ding L, Getz G, Wheeler DA et al. (2008) Somatic mutations affect key pathways in lung adenocarcinoma. Nature 455: 1069–1075. Fabbri G, Rasi S, Rossi D et al. (2011) Analysis of the chronic lymphocytic leukemia coding genome: role of Notch1 mutational activation. Journal of Experimental Medicine 208: 1389–1401.

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

Characterising Cancer Genome by Means of Next-generation Sequencing

Greenman C, Stephens P, Smith R et al. (2007) Patterns of somatic mutation in human cancer genomes. Nature 446: 153–158. Gui Y, Guo G, Huang Y et al. (2011) Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nature Genetics 43: 875–878. Hainaut P, Olivier M and Pfeifer GP (2001) TP53 mutation spectrum in lung cancers and mutagenic signature of components of tobacco smoke: lessons from the IARC TP53 mutation database. Mutagenesis 16: 551–553. Hudson TJ, Anderson W, Artez A et al. (2010) International network of cancer genome projects. Nature 464: 993–998. Jones S, Zhang X, Parsons DW et al. (2008) Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321: 1801–1806. Kan Z, Jaiswal BS, Stinson J et al. (2010) Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466: 869–873. Lee W, Jiang Z, Liu J et al. (2010) The mutation spectrum revealed by paired genome sequences from a lung cancer patient. Nature 465: 473–477. Ley TJ, Mardis ER, Ding L et al. (2008) DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome. Nature 456: 66–72. Li M, Zhao H, Zhang X et al. (2011) Inactivating mutations of the chromatin remodeling gene arid2 in hepatocellular carcinoma. Nature Genetics 43: 828–829. Link DC, Schuettpelz LG, Shen D et al. (2011) Identification of a novel TP53 cancer susceptibility mutation through whole-genome sequencing of a patient with therapy-related AML. Journal of American Medical Association 305: 1568–1576. Machida K, Cheng KT, Sung VM et al. (2004) Hepatitis C virus induces a mutator phenotype: enhanced mutations of immunoglobulin and protooncogenes. Proceedings of the National Academy of Sciences of the USA 101: 4262–4267. Makinen N, Mehine M, Tolvanen J et al. (2011) MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science 334: 252–255. Mardis ER, Ding L, Dooling DJ et al. (2009) Recurring mutations found by sequencing an acute myeloid leukemia genome. New England Journal of Medicine 361: 1058–1066. Meyerson M, Gabriel S and Getz G (2010) Advances in understanding cancer genomes through second-generation sequencing. Nature Reviews Genetics 11: 685–696. Mitelman F, Johansson B and Mertens F (2004) Fusion genes and rearranged genes as a linear function of chromosome aberrations in cancer. Nature Genetics 36: 331–334. Paez JG, Janne PA, Lee JC et al. (2004) EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304: 1497–1500. Parla JS, Iossifov I, Grabill I et al. (2011) A comparative analysis of exome capture. Genome Biology 12: R97. Parsons DW, Jones S, Zhang X et al. (2008) An integrated genomic analysis of human glioblastoma multiforme. Science 321: 1807–1812. Pasqualucci L, Trifonov V, Fabbri G et al. (2011) Analysis of the coding genome of diffuse large B-cell lymphoma. Nature Genetics 43: 830–837. Pfeifer GP and Hainaut P (2011) Next-generation sequencing: emerging lessons on the origins of human cancer. Current Opinion in Oncology 23: 62–68.

Pleasance ED, Cheetham RK, Stephens PJ et al. (2010a) A comprehensive catalogue of somatic mutations from a human cancer genome. Nature 463: 191–196. Pleasance ED, Stephens PJ, O’Meara S et al. (2010b) A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature 463: 184–190. Prickett TD, Agrawal NS, Wei X et al. (2009) Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4. Nature Genetics 41: 1127–1132. Puente XS, Pinyol M, Quesada V et al. (2011) Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 475: 101–105. Robison K (2010) Application of second-generation sequencing to cancer genomics. Brief Bioinformatics 11: 524–534. Samuels Y, Wang Z, Bardelli A et al. (2004) High frequency of mutations of the PIK3CA gene in human cancers. Science 304: 554. Schweiger MR, Kerick M, Timmermann B et al. (2011) The power of NGS technologies to delineate the genome organization in cancer: from mutations to structural variations and epigenetic alterations. Cancer and Metastasis Review 30: 199–210. Shah SP, Morin RD, Khattra J et al. (2009) Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature 461: 809–813. Sjoblom T, Jones S, Wood LD et al. (2006) The consensus coding sequences of human breast and colorectal cancers. Science 314: 268–274. Soda M, Choi YL, Enomoto M et al. (2007) Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 448: 561–566. Stephens PJ, McBride DJ, Lin ML et al. (2009) Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature 462: 1005–1010. Stransky N, Egloff AM, Tward AM et al. (2011) The mutational landscape of head and neck squamous cell carcinoma. Science 333: 1157–1160. Stratton MR (2011) Exploring the genomes of cancer cells: progress and promise. Science 331: 1553–1558. Sulonen AM, Ellonen P, Almusa H et al. (2011) Comparison of solution-based exome capture methods for next generation sequencing. Genome Biology 12: R94. Tiacci E, Trifonov V, Schiavoni G et al. (2011) BRAF mutations in hairy-cell leukemia. New England Journal of Medicine 364: 2305–2315. Tomlins SA, Laxman B, Dhanasekaran SM et al. (2007) Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature 448: 595–599. Tomlins SA, Rhodes DR, Perner S et al. (2005) Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310: 644–648. Totoki Y, Tatsuno K, Yamamoto S et al. (2011) High-resolution characterization of a hepatocellular carcinoma genome. Nature Genetics 43: 464–469. Varela I, Tarpey P, Raine K et al. (2011) Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469: 539–542. Wang J, Mullighan CG, Easton J et al. (2011a) CREST maps somatic structural variation in cancer genomes with base-pair resolution. Nature Methods 8: 652–654.

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

9

Characterising Cancer Genome by Means of Next-generation Sequencing

Wang K, Kan J, Yuen ST et al. (2011b) Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nature Genetics 43: 1219–1223. Wei X, Walia V, Lin JC et al. (2011) Exome sequencing identifies GRIN2A as frequently mutated in melanoma. Nature Genetics 43: 442–446. Wood LD, Parsons DW, Jones S et al. (2007) The genomic landscapes of human breast and colorectal cancers. Science 318: 1108–1113. Yan XJ, Xu J, Gu ZH et al. (2011) Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nature Genetics 43: 309–315.

10

Further Reading Chin L, Andersen JN and Futreal PA (2011) Cancer genomics: from discovery science to personalized medicine. Nature Medicine 17: 297–303. Macconaill LE and Garraway LA (2010) Clinical implications of the cancer genome. Journal of Clinical Oncology 28: 5219–5228. Taylor BS and Ladanyi M (2011) Clinical cancer genomics: how soon is now? Journal of Pathology 223: 318–326.

eLS & 2012, John Wiley & Sons, Ltd. www.els.net

"Characterising Somatic Mutations in Cancer Genome by Means of ...

Feb 15, 2012 - imise tissue heterogeneity. Ultimately, a comprehensive delineation of the somatic mutations in the cancer gen- ome would require WGS of a ...

143KB Sizes 1 Downloads 247 Views

Recommend Documents

Cancer genome sequencing: a review
The digital nature of next- generation sequencing allows us to ..... MicroRNA Signature of Malignant Mesothelioma with Potential. Diagnostic and Prognostic ...

Induced genome maintenance pathways in pre-cancer ...
cancerous and cancerous lesions deposited in the GEO database. We find that the ...... Figure 1 – Subpathways expression profiles of microarrays studies of. 60.

pdf-1857\from-dna-photolesions-to-mutations-skin-cancer ...
... apps below to open or edit this item. pdf-1857\from-dna-photolesions-to-mutations-skin-cance ... eries-in-photochemical-photobiological-sciences-fr.pdf.

1499499901845-mandalas-by-means-of-antiquated-design ...
Page 3 of 3. Page 3 of 3. 1499499901845-mandalas-by-means-of-antiquated-design-omnigeometry-mandala-designer-online.pdf.

1499499901845-mandalas-by-means-of-antiquated-design ...
There was a problem loading more pages. Retrying... 1499499901845-mandalas-by-means-of-antiquated-design-omnigeometry-mandala-designer-online.pdf.

CALLUS INDUCTION, SOMATIC ... - Semantic Scholar
The best medium for proliferation of callus was standardized as MS + 0.1 mgl-1 2,4-D + ... different combinations tried, MS + 1.9 gl-1 KNO3 was found suitable for ...

MITOCHONDRIAL DNA MUTATIONS IN HUMAN DISEASE.pdf ...
MITOCHONDRIAL DNA MUTATIONS IN HUMAN DISEASE.pdf. MITOCHONDRIAL DNA MUTATIONS IN HUMAN DISEASE.pdf. Open. Extract. Open with.

Concurrency of Mutations, Microsatellites and Predicted Domains in ...
Journal of Proteomics & Bioinformatics - Open Access ... Abstract. Bioinformatics is the field of science in which biology, computer science, and information technology merge to form a single ... fueled a new era in the analysis of biological data.

Characterization of SH2D1A Missense Mutations ...
ase chain reaction-negative for EBV (8–10). Patients infected with EBV mount an ... School, 330 Brookline Ave., Boston, MA 02215. Tel.: 617-667-7147; Fax:.

REPORT Detecting Disease-Causing Mutations in the ...
strategy for detecting mutations that is based on comparing affected haplotypes with closely matched control sequences from healthy ..... Cell lines from these individuals would ... coverage—that is, under the assumption of a Poisson coverage.

pdf-1477\privacy-and-progress-in-whole-genome-sequencing-by ...
... apps below to open or edit this item. pdf-1477\privacy-and-progress-in-whole-genome-sequenc ... ial-commission-for-the-study-of-bioethical-issues.pdf.

Qualitative Modelling of Complex Systems by Means of Fuzzy ...
in these systems, output variables depend on so many different inputs that ...... Static records can then be sorted alphabetically and stored in the so-called input- .... Among the different dialects of fuzzy logic, FIR is quite peculiar, as most of

Relative Contributions of Axonal and Somatic Na ...
Feb 15, 2006 - Tech Drive, Northwestern University, Evanston, IL 60208. E-mail: ... fused with ice-cold (4°C) artificial CSF (ACSF) (in mM: 123.75 NaCl,. 3.5 KCl ...... Raman IM, Bean BP (2001) Inactivation and recovery of sodium currents in.

Optimization of Menu Layouts by Means of Genetic ...
Examples are Apple's Human Interface Guidelines [11] and Sun's Java Look and. Feel Guidelines ..... Apple Computer Inc.: Apple human interface guidelines.

Qualitative Modelling of Complex Systems by Means of ...
les qui només es preocupaven per la realitat diària però quedaven felices per qualsevol explicació que els donés sobre la recerca feta. ...... ___168. Figure 6-5 Trajectory of the considered gas turbine output (7200 data points). ...... best qua

Detection of Web Defacements by Means of Genetic ...
ger the sending of a notification to the administrator of the page. The tool is ... creating computer programs by means of artificial evolu- tion [5]. A population of ...

Enhanced signal detection in neuroimaging by means ...
Available online on ScienceDirect (www.sciencedirect.com). 1053-8119/$ - see front ..... procedure to any other preferred programming language or data processing ..... degree filled with true positives, small to moderately large imaging volumes, weak

Defining new approximations of belief functions by means of ...
representations are sought, i.e. by means of belief functions with a restricted number of focal elements. The second drawback is the lack of intuitive significance for a belief function with several focal elements of different cardinality. As explain

Subclonal mutations in SETBP1 confer a poor ...
Jan 15, 2015 - 3Department of Bioengineering and Therapeutic Sciences, California Institute for Quantitative Biosciences, University of California, San ...

The Costs of Cancer - American Cancer Society Cancer Action Network
solutions designed to eliminate cancer as a major ...... suffering and maintain the best possible quality of life for patients and .... wordpress.com/2016/01/8806-the-burden-of-medical- ... February 22, 2017. https://www.acscan.org/sites/default/.