Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Clinical Cancer Predictive Biomarkers and Personalized Medicine Research

Mapping of 1p Deletions in Myeloma Identifies FAM46C at 1p12 and CDKN2C at 1p32.3 as Being in Regions Associated with Adverse Survival

Kevin D. Boyd1, Fiona M. Ross2, Brian A. Walker1, Christopher P. Wardell1, William J. Tapper2, Laura Chiecchio2, GianPaolo Dagrada2, Zoe J. Konn2, Walter M. Gregory3, Graham H. Jackson4, J. Anthony Child3, Faith E. Davies1, and Gareth J. Morgan1 on behalf of the NCRI Haematology Oncology Studies Group

Abstract Purpose: Regions on 1p with recurrent deletions in presenting myeloma patients were examined with the purpose of defining the deletions and assessing their survival impact. Experimental Design: mapping, gene expression, FISH, and mutation analyses were conducted on patient samples from the MRC Myeloma IX trial and correlated with clinical outcome data. Results: 1p32.3 was deleted in 11% of cases, and deletion was strongly associated with impaired overall survival (OS) in patients treated with autologous stem cell transplant (ASCT). In patients treated less intensively, del(1)(p32.3) was not associated with adverse progression-free survival (PFS) or OS. The target of homozygous deletions was CDKN2C, however its role in the adverse outcome of cases with hemizygous deletion was less certain. 1p22.1-21.2 was the most frequently deleted region and contained the candidate genes MTF2 and TMED5. No mutations were identified in these genes. 1p12 was deleted in 19% of cases, and deletion was associated with impaired OS in univariate analysis. The target of homozygous deletion was FAM46C, which was mutated in 3.4% of cases. When cases with FAM46C deletion or mutation were considered together, they were strongly associated with impaired OS in the intensive treatment setting. Conclusion: Deletion of 1p32.3 and 1p12 was associated with impaired OS in myeloma patients receiving ASCT. FAM46C was identified as a gene with potential pathogenic and prognostic significance based on the occurrence of recurrent homozygous deletions and mutations. Clin Cancer Res; 17(24); 7776–84. 2011 AACR.

Introduction shown to be important include deletion of 17p, involving the TP53 gene, and gain of 1q (2–7). A prognostic model High-risk genetic lesions can be divided into primary incorporating these lesions with the International Staging genetic events, namely, translocations involving the IGH System (ISS) has been described which identified patients gene at 14q32, and secondary genetic events which usually with poor prognosis at diagnosis (8). constitute chromosomal gains or deletions (1). IGH trans- Deletion of 1p has been identified as a common recurrent locations that have been associated with short survival in genetic event in myeloma that has prognostic significance myeloma include t(4;14), t(14;16), and t(14;20), whereas when detected by conventional cytogenetics (9, 10). Several structural chromosomal abnormalities that have been minimally altered regions on 1p have been identified, including 1p32.3, 1p31.3, 1p22.1-1p21.3, and 1p12 (11). We have previously used an approach of focusing on regions with recurrent homozygous deletions, as these are Authors' Affiliations: 1The Institute of Cancer Research, Haemato-oncol- ogy Research Unit, Division of Molecular Pathology, London; 2University of usually small, pathogenically relevant, and allow for the Southampton, Wessex Regional Genetics Laboratory, Salisbury; 3Clinical definition of critically deregulated genes (12). We described Trials Research Unit, University of Leeds, Leeds; and 4University of New- castle, Newcastle-upon-Tyne, United Kingdom recurrent homozygous deletions of 1p32.3 which incorpo- rate two genes, CDKN2C and FAF1, with homozygous Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). deletion being associated with abrogation of expression of both of these genes (13, 14). We found deletion of 1p32.3 to Corresponding Author: Gareth J. Morgan, The Institute of Cancer Research, Haemato-oncology Research Unit, Division of Molecular Pathol- be associated with impaired survival in 510 newly present- ogy, 15 Cotswold Road, Sutton, Surrey SM2 5NG, United Kingdom. Phone: ing patients treated intensively (14). In another study, 442087224130; Fax: 442087224432; E-mail: [email protected] genome-wide correlation of CGH-defined genomic lesions doi: 10.1158/1078-0432.CCR-11-1791 with survival identified 1p31-32 as the only factor inde- 2011 American Association for Cancer Research. pendently associated with prognosis in 131 patients treated

7776 Clin Cancer Res; 17(24) December 15, 2011

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Survival Impact of 1p12/FAM46C and 1p32.3/CDKN2C in Myeloma

doxorubicin, and dexamethasone) or CTD (cyclophospha- Translational Relevance mide, thalidomide and dexamethasone) followed by ASCT. After the autograft procedure there was a second random- Cytogenetic lesions that are associated with impaired ization to maintenance thalidomide versus no ongoing survival have been identified in myeloma, and there is an therapy. In the nonintensive pathway patients were ran- aim to use such lesions to define high-risk disease in a domized to either MP (melphalan and prednisolone) or risk-adapted therapeutic approach. The lesions generally CTDa (attenuated cyclophosphamide, thalidomide and accepted to constitute high risk lesions are t(4;14), dexamethasone) to maximum response, followed by the t(14;16), t(14;20), del(17p), and þ1q. We identified same maintenance randomization. Irrespective of treat- two regions of 1p that have prognostic impact when ment pathway, all patients underwent a bisphosphonate deleted: 1p32.3 and 1p12. Deletion of 1p32.3 targets randomization to receive zoledronic acid or clodronic acid. CDKN2C, and it only impacts prognosis in patients Median follow-up was 3.7 years. An extended data set for treated with ASCT. Deletion of 1p12 targets FAM46C, 1p12 FISH testing included patients randomized in the which is also frequently mutated. Cases with deletion or Myeloma IX trial along with other diagnostic myeloma mutation of FAM46C also had impaired survival. Dele- samples referred to the Wessex Regional Cytogenetic tion of 1p has prognostic significance in patients treated laboratory. with ASCT, and could be incorporated into the definition of high-risk disease in risk-adapted treatment strategies. Genetic test data sets aspirate and peripheral blood samples were taken from patients after informed consent. Bone marrow was purified for plasma cells using CD138 magnetic with high-dose melphalan and autologous stem cell trans- microbead cell selection (Miltenyi Biotec). 1,140 of 1,960 plantation (ASCT; ref. 15). Two data sets have, therefore, randomized patients provided a bone marrow aspirate found loss of 1p32 to be prognostically significant in sample that was suitable for analysis. A variety of genetic patients treated with ASCT, although its significance outside tests were carried out on the CD138 selected cell fraction. of this clinical context is unknown. We have previously The number of samples tested by each method was decided identified recurrent homozygous deletions of FAM46C at by the quality and quantity of the provided samples, result- 1p12, identifying it as a gene with potential pathogenic ing in several overlapping data sets: 1,140 had FISH carried relevance (11). The next generation sequencing of 38 mye- out, including 859 with results for 1p32.3 and 378 with loma tumours recently found FAM46C to be frequently results for 1p12; 272 had global gene expression arrays mutated, highlighting the potential significance of this gene carried out; 114 had SNP-based gene mapping carried out; (16). It is not known whether abnormalities of 1p12/ 147 had mutation analysis of FAM46C and MTF2. 124 had a FAM46C impact prognosis. We have also previously iden- complete data set of deletional annotation of 1p by map- tified a region from 1p22.1-21.3 as being a region of ping or FISH, plus gene expression and mutation analysis potential significance as, although it did not harbour homo- (Table 1). These various data sets were representative of the zygous mutations, it was the most frequently deleted region overall trial population (Supplementary Data, Table 1). (11). Two candidate genes with downregulated expression within this region (MTF2 and TMED5) were identified. Gene mapping and expression analysis In this study, we conducted an analysis of the survival CD138 selected cells were stored in RLT buffer (Qiagen) associations of genetic lesions of 1p32.3, 1p22.1, and 1p12 at 80C immediately after purification until extraction. in patients treated intensively and nonintensively, with DNA and RNA were extracted using commercial kits as conventional and thalidomide-based chemotherapy regi- previously described (19). 100 ng of tumor RNA was mens, and have integrated gene expression and mutational amplified using the 2-cycle target labeling kit and hybrid- analysis to define deregulated genes on 1p. ized to U133 Plus 2.0 arrays (Affymetrix),

Materials and Methods

Patients Table 1. Genetic test data sets The MRC Myeloma IX trial (ISRCTN68454111) enrolled 1,960 patients with newly diagnosed Data set Number of patients requiring treatment, and received ethical approval from the Myeloma IX clinical trial entrants 1,960 MRC Leukaemia Data Monitoring and Ethics Committee Any FISH carried out 1,140 (MREC 02/8/95). The design and results of Myeloma IX FISH results for 1p32.3 859 have been reported elsewhere, but in summary treatment FISH results for 1p12 378 followed 2 pathways, an intensive or a nonintensive path- Global gene expression arrays 272 way, based on patient age and performance status (17, 18). Gene mapping arrays 114 Patients in the intensive pathway underwent an initial FAM46C/MTF2 mutation analysis 147 randomization to CVAD (cyclophosphamide, vincristine,

www.aacrjournals.org Clin Cancer Res; 17(24) December 15, 2011 7777

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Boyd et al.

washed and scanned following the manufacturer’s proto- BigDye terminators v3.1 (Applied Biosytems) on a 3500 col. Five hundred nanograms of tumor and 500 ng of germ Genetic Analyzer (Applied Biosystems). Variants that were line DNA were used for mapping purposes. DNA was not labeled as SNPs on Ensembl had constitutional DNA digested, amplified, labeled, and hybridized to GeneChip sequenced using DNA extracted from peripheral blood mapping 500K arrays (Affymetrix) as previously described samples. (19, 20). SNP genotypes were derived as previously described using Affymetrix GCOS, and GTYPE software Statistical methods (19). Regions of gain or loss of heterozygosity were iden- Statistical analysis was conducted using R and SPSS tified using dChip 2006 (www.dchip.org), and expression version 19.0 (SPSS Inc.). Survival curves were plotted using data were normalized using dChip. Microarray data are the Kaplan–Meier method. Differences between curves were accessible through Gene Expression Omnibus Series acces- tested for statistical significance using the log-rank test, with sion number GSE15695. P < 0.05 taken as the level of significance. Variables asso- ciated with survival in univariate analysis at the P < 0.05 FISH level were entered into the model for multivariate testing. FISH was carried out on CD138 selected bone marrow Multivariate analysis of variables associated with survival samples. FISH analysis of 1p32.3 used 2 bespoke probes used a backwards elimination Cox proportional hazards (RP11-278J17 and RP11-116M11) grown and labeled in the model, with factors retained if they were significant at laboratory, as previously described (13). FISH for 1p12 used P < 0.05. The test for interaction used a Cox regression the RP11-418J17 probe. A mixture of commercially available model, with x2 values derived from the difference of the log- and bespoke probes were used as previously described to likelihood values. Comparison of categorical variables used detect the presence of an immunoglobulin heavy chain the Fisher exact or x2 tests. Comparison of continuous (IGH) translocation, the common IGH translocation part- variables used the Wilcoxon rank-sum test. The gene expres- ners (MMSET at 4p16, CCND3 at 6p21, CCND1 at 11q13, sion of groups was compared using a t test. MAF at 16q23, and MAFB at 20q12), hyperdiploid status using the iFISH ploidy classification, deletion of 13q14, Results 16q23, 17p13, 22q11, and gain of 1q21 (21, 22). Survival impact and molecular biology of specific Mutation analysis regional loss Genomic DNA underwent whole genome amplification 1p32.3. FISH for 1p32.3 was carried out on 859 using the REPLI-g commercial kit (Qiagen) following the patients, of which 96 (11.2%) had del(1)(p32.3). As pre- manufacturer’s protocol. High resolution melt (HRM) anal- viously reported, intensively treated patients with this dele- ysis was used to screen tumor DNA samples for genetic tion had very short survival [median overall survival (OS) variants of FAM46C, using the Type It HRM commercial kit 34.5 months vs. median not reached, P < 0.001; ref. 11]. In (Qiagen) and the Rotorgene Q Real-Time Cycler (Qiagen). contrast, patients with del(1)(p32.3) treated with a non- Primers were designed to span segments of approximately intensive approach had improved survival compared with 250 bases (Supplementary Data, Table 2). The PCR reaction patients without deletion (median OS 36.4 vs. 31.2 months, comprised 7 pmol of combined forward and reverse primer P ¼ 0.047; Fig. 1); del(1)(p32.3) was not associated with mix, 20 ng of DNA, 5 mLof2 HRM Master Mix and 3.3 mL progression-free survival (PFS) in either treatment pathway. of RNAse-free water. PCR conditions were a 95C hold for A formal test for interaction supported the finding that the 5 minutes, followed by 40 cycles of 95C for 10 seconds, prognostic impact of del(1)(p32.3) varied by treatment 55C for 30 seconds and 72C for 10 seconds. High reso- pathway (x2 13.69, df 1, P < 0.001). We examined whether lution melt analysis was conducted from 65Cto95Cat other variables associated with survival were balanced 0.1C increments. Data were analyzed using the Rotorgene between the intensive and nonintensive arms in patients Q series software version 1.7. Samples identified as variants with del(1)(p32.3), in an attempt to explain this paradox- by HRM underwent purification using the QIAquick PCR ical result. There was no significant bias in frequency of Purification Kit (Qiagen) and were directly sequenced with other high risk genetic lesions including t(4;14), t(14;16), t(14;20), þ1q or del(17p). Similarly, there were no signif- icant differences comparing intensively and nonintensively Table 2. Multivariate analysis of genetic treated patients with del(1)(p32.3) in ISS stratification, variables associated with OS in the intensive length of 1p deletion, rate of homozygous 1p32.3 deletion or rate of cosegregation of 1p12 deletion (Supplementary pathway Data, Table 3). In a multivariate analysis including these genetic variables, del(1)(p32.3) was significantly associated P Genetic variable HR 95% CI with adverse OS in the intensive arm [HR ¼ 1.68; 95% þ(1)(q21) 1.79 1.33–2.42 <0.001 confidence interval (CI) ¼ 1.11–2.56, P ¼ 0.015; Table 2]. del(17)(p13) 1.90 1.21–2.98 0.005 Surprisingly, the association of del(1)(p32.3) with favorable del(1)(p32.3) 1.68 1.11–2.56 0.015 survival in the patients treated nonintensively remained weakly significant in multivariate analysis (HR ¼ 0.63;

7778 Clin Cancer Res; 17(24) December 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Survival Impact of 1p12/FAM46C and 1p32.3/CDKN2C in Myeloma

ing patients that did not proceed to transplant. In this 1.0 landmarked analysis del(1)(p32.3) was associated with Log-rank P = 0.047 impaired PFS and OS (median PFS from time of ASCT 19.0 months vs. 26.0 months, P ¼ 0.033; median OS 0.8 from time of ASCT 38.1 months vs. median not reached, P < 0.001; Fig. 2A and B). A total of 118 patients in the 0.6 nonintensive pathway were less than 70 years of age, including 14 with del(1)(p32.3). Del(1)(p32.3) in these patients was not associated with impaired OS (median 40.7 0.4 months [del(1)(p32.3)] vs. 33.5 months [no del(1) (p32.3)], P ¼ 0.391; Fig. 3). From this we conclude that Proportion of patients del(1)(p32.3) was only associated with impaired survival in 0.2 the context of ASCT. The fact that del(1)(p32.3) was not

del(1)(p32.3) (N = 43) associated with OS in young patients that were not auto- 0.0 No del(1)(p32.3) (N = 306) grafted suggests that this may be a treatment-specific effect. 010203040506070 To investigate the biological basis of this effect, the Survival (mo) deletions were defined in more detail. Of the 114 patients

Figure 1. OS of patients undergoing nonintensive treatment comparing patients with del(1)(p32.3) to those without deletion. A 1.0 95% CI, 0.39–1.00, P ¼ 0.050; Table 3). These data suggest Log-rank P = 0.033 that the paradoxicalsurvival effect in the 2 arms is not due to a 0.8 confounding variable but is associated with the 1p32.3 deletion. The major differences between the intensive and non- 0.6 intensive arms were treatment and patient age. The inten- sive pathway incorporated treatment with ASCT, whereas 0.4 the nonintensive pathway did not. Whether a patient

entered the intensive or nonintensive pathway was decided Proportion of patients by the treating physician based on patient suitability for 0.2 ASCT, with no defined age limit. However, in practice, the majority of patients under the age of 70 were treated del(1)(p32.3) (N = 36) N intensively, whereas the majority of patients more than the 0.0 No del(1)(p32.3) ( = 302) age of 70 were treated nonintensively. This raised the 0 102030405060 question as to whether del(1)(p32.3) was associated with B Survival (mo) adverse prognosis in the context of ASCT or young age, and 1.0 taking this question further we investigated age and ASCT as Log-rank P < 0.001 factors in subgroup analyses. We examined the impact of del (1)(p32.3) in patients that received autograft and in young 0.8 patients treated in the nonintensive pathway. Although there was an intention to treat with ASCT in the intensive pathway, only 346 of 503 (68.6%) analyzable patients in 0.6 the intensive arm received ASCT. The analysis of the asso- ciation of del(1)(p32.3) with survival in the intensive 0.4 pathway was landmarked from time of ASCT, thus exclud- Proportion of patients

0.2 Table 3. Multivariate analysis of genetic del(1)(p32.3) (N = 36) N variables associated with OS in the nonintensive 0.0 No del(1)(p32.3) ( = 302) pathway 0 10203040506070 Survival (mo) Genetic variable HR 95% CI P

Adverse IGH group 2.12 1.52–2.95 <0.001 Figure 2. PFS (A) and OS (B) of patients treated in the intensive pathway, del(1)(p32.3) 0.63 0.39–1.00 0.050 landmarked at time of ASCT and comparing cases with del(1)(p32.3) with patients without deletion.

www.aacrjournals.org Clin Cancer Res; 17(24) December 15, 2011 7779

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Boyd et al.

This is consistent with previously reported data linking high 1.0 CDKN2C expression with a high proliferation rate (13, 23). Log-rank P = 0.391 We have previously not found CDKN2C to be inactivated through mutation or methylation (13). As hemizygous 0.8 deletion was not associated with low expression, low expression was not associated with adverse survival, and 0.6 other mechanisms of allelic inactivation have not been shown, it is difficult to link the adverse survival associated with hemizygous deletion of 1p32.3 to modulation of 0.4 CDKN2C. We examined FAF1, and found that hemizygous deletion of 1p32.3 was associated with decreased expression Proportion of patients of this gene (218080_x_at fold change 1.48 (1.29 to 0.2 1.73), P < 0.001; 224217_s_at fold change 1.66 (1.37 to 2.07), P < 0.001). However, there was no association of del(1)(p32.3) (N = 14) 0.0 No del(1)(p32.3) (N = 104) low expression of these probes with short survival. 1p22.1. 0 102030405060 In a previous report we identified 1p22.1-21.3 Survival (mo) as being the most frequently deleted of 1p, and the meta- analysis of the additional 565 publicly available mapping cases confirmed this, with 22.5% of cases having a deletion Figure 3. OS of patients 70 years or younger in the nonintensive pathway, of 1p22.1 (11). We previously identified MTF2 and TMED5 comparing patients with del(1)(p32.3) with patients without deletion. as candidate genes within this region as both had signifi- cantly downregulated expression in cases with deletion with gene mapping data, 6 had a homozygous deletion of (11). We now report that low expression of MTF2 was not this region, with a further 12 having a hemizygous deletion. associated with impaired OS or PFS. Moreover, no muta- In our data set all the homozygous deletions affected 2 tions of MTF2 were identified by HRM analysis in 147 newly genes, CDKN2C and FAF1, and to try and define the key presenting patients. Low TMED5 expression was weakly deleted gene we examined the mapping of an additional associated with short survival (median OS 29.9 months 565 publicly available myeloma mapping cases [available low expression vs. 51.2 months, high expression, P ¼ through www.broad.mit.edu/mmgp (incorporating the 0.050). However, no mutations were identified in 20 newly Mayo clinic, Carrasco and MMRC reference collections) presenting patients. and the IFM collection at www.ncbi.nlm.nih.gov/geo 1p12. Of 114 cases with mapping data, 1p12 was GSE12896]. These data identified a further 15 cases with homozygously deleted in 2 cases and hemizygously deleted homozygous deletion of 1p32.3. In the majority of cases the in a further 20 cases. In addition, uniparental disomy was a deletions incorporated both CDKN2C and FAF1, but in 3 mechanism of loss of heterozygosity in 4 cases. In an cases, CDKN2C seemed to be the sole target of deletion. extended data set of 378 cases with FISH for 1p12, using The relationship of deletion and gene expression was a probe that was not specific for FAM46C, the rate of 1p12 examined. Although homozygous deletion of 1p32.3 abro- deletion was 19%. When mapping of human myeloma cell gates expression of CDKN2C, hemizygous deletion was not lines (HMCL) was examined, small homozygous deletions associated with decreased expression. Moreover, although of FAM46C were identified in KMS11, LP1, and MM1s, with the homozygously deleted cases with abrogated CDKN2C further hemizygous deletions in H929, JIM1, JIM3, KMS26, expression were associated with short survival, low KMS28, and MM1R. The fact that 2 of 114 presenting cases CDKN2C expression was not associated with impaired had homozygous deletion of this region, compared with 3 survival in other cases. In fact, the converse was true; of 18 HMCLs suggest that this lesion is associated with patients with top quartile CDKN2C expression were asso- disease progression. Mutations of FAM46C were screened ciated with significantly shorter survival than those with for in 147 patients, and 5 (3.4%) were found to harbor bottom quartile expression (median OS 54.8 months low mutations (Table 4). All mutations were single base sub- expression vs. 36.1 months high expression, P ¼ 0.001). stitutions, resulting in either an amino acid change or a stop

Table 4. Annotated FAM46C mutations

Sample no. Base change Amino acid change 1p12 mapping

323 c357 C>G F119L No deletion 1,527 c463 A>T I155F Hemizygous deletion 245 c537 C>A F179C No deletion 326 c872 A>G Y291C No deletion 127 c1068 C>G Y356X Hemizygous deletion

7780 Clin Cancer Res; 17(24) December 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Survival Impact of 1p12/FAM46C and 1p32.3/CDKN2C in Myeloma

impaired survival (median OS 40.9 months vs. 53.0 1.0 months, P ¼ 0.045), whereas there was a stronger associ-

Log-rank P = 0.004 ation with short survival in patients with only del(1)(p32.3) (median OS 21.6 months vs. 51.6 months, P < 0.001). This 0.8 suggests that when deletion alone is being considered, 1p32.3 has the largest prognostic impact. 0.6 Discussion

0.4 In this analysis we identify genes and regions of 1p with prognostic impact and potential pathogenic relevance. Proportion of patients 1p32.3 underwent homozygous deletion in 5.3% of cases, 0.2 with the deletions affecting CDKN2C and also FAF1, although the mapping data of other data sets suggest that FAM46C LOH or mutation (N = 26) 0.0 Normal FAM46C (N = 88) CDKN2C may be the key deleted gene (23). Deletions 0 10203040506070 detected by FISH were associated with impaired survival in Survival (mo) patients treated in the intensive arm of the Myeloma IX trial, but surprisingly the converse was true in patients treated nonintensively, with deletion being associated with a mar- Figure 4. OS of cases with any abnormality of FAM46C (incorporating ginal improvement in OS. Both these associations were true FAM46C LOH and mutation) compared with cases with normal . in multivariate analyses including other genetic lesions, and we found no bias in confounding variables to explain this effect. We examined this paradox in more detail to assess codon. Two of the mutations occurred in samples with whether this was related to patient therapy or patient age, hemizygous deletion of 1p12, whereas 3 did not cosegregate and the association of deletion with impaired survival was with either deletion or uniparental disomy. only observed in patients receiving ASCT, suggesting that The survival of patients with any abnormality of del(1)(p32.3) may specifically impair the response to this FAM46C (incorporating loss of heterozygosity or muta- therapy. However, the low numbers of young patients not tion) was compared with patients with normal FAM46C, autografted in the analysis means that age cannot be and we show that abnormal FAM46C was associated completely discounted as a factor. with impaired survival (median OS 25.7 months vs. Although CDKN2C is likely to be the key deregulated 51.3 months, P ¼ 0.004; Fig. 4). Although this effect was gene in cases with homozygous deletion, it’s involvement in most marked in patients treated intensively (median OS the outcome of cases with hemizygous deletion was less 24.4 months vs. not reached, P ¼ 0.005), a formal test for certain, as in these cases CDKN2C was not underexpressed, interaction of pathway and del(1)(p12) was not signi- and we have previously reported no evidence of allelic ficant (x2 ¼ 3.08; df 1; P ¼ 0.078) which does not support silencing through methylation or mutation (13). Moreover, there being the same differential survival impact in low expression was not associated with short survival but patients treated intensively and nonintensively as was the converse was true, with high expression being linked to evident in the analysis of 1p32.3. The survival impact impaired survival. This is consistent with previously of del(1)(p12) was extended to the FISH data set of reported data showing that myeloma with high prolifera- 378 patients and deletion was found to be weakly asso- tion defined by a gene expression signature was associated ciated with impaired OS (median OS 44.5 months vs. with high CKDN2C expression (23). It is possible, then, that 53.0 months, P ¼ 0.0497). This association was not in patients with hemizygous deletion of 1p32.3, deletion of significant in multivariate analysis when þ1q, del17p other genes may play a role in the clinical outcome. and adverse IGH translocations [t(4;14), t(14;16), and Although homozygous deletions were small and focal, t(14;20)] were included as covariates. mapping analysis showed that hemizygous deletions are often large and involve other regions, including 1p22.1 and 1p12. The relative survival impact of del(1)(p32.3) 1p22.1 was the most frequently deleted region in our and del(1)(p12) mapping data set and in the extended analysis of all publicly Hemizygous deletion are often large, so that 1p deletions available mapping data sets. However, the 2 candidate often encompass both 1p12 and del1p32.3. To assess the genes that we had previously identified within this region relative contributions of these 2 regions to the adverse (MTF2 and TMED5) did not have convincing evidence that prognostic signal, 372 patients with FISH results for both downregulation adversely affected survival. Moreover, we 1p12 and 1p32.3 were examined. 38 (10.2%) had del(1) did not identify any mutations in these genes. Although it (p12) only, 18 (4.8%) had del(1)(p32.3) only and 30 remains likely that this region has pathogenic relevance, a (8.1%) cases had deletion of both regions. The 38 patients key deregulated gene within this region has not been with only del(1)(p12) were weakly associated with identified.

www.aacrjournals.org Clin Cancer Res; 17(24) December 15, 2011 7781

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Boyd et al.

Homozygous deletions of 1p12 affecting only patients, although the association was of borderline signif- FAM46C were observed in 2 presenting myeloma icance, and was not significant in multivariate analysis. patients and 3 HMCLs. In our initial gene mapping and Moreover, when cases with del(1)(p12) alone were com- expression data set we identified the potential impor- pared with patients with del(1)(p32.3) alone, the asso- tance of FAM46C, and this was confirmed in the report ciation of del(1)(p32.3) with short survival was much of the initial genome sequencing of 38 myeloma tumors stronger. A caveat of this extended analysis was that the which found FAM46C mutations in 5 of 38 patient FISH probe for 1p12 did not target FAM46C and may, samplesand4of17HMCLs, and suggested a role for therefore, have missed some FAM46C deleted cases. These this functionally uncharacterized gene in regulation of analyses also did not consider FAM46C mutation events, translation by acting as an mRNA stability factor which were frequent, and when we examined survival (11, 16). We screened presenting myeloma tumors for within patients with complete annotation of deletion variants of FAM46C by high resolution melt analysis, and mutation of FAM46C, patients with any LOH or muta- and found mutations in 5 samples. The mutation rate tion of FAM46C were associated with impaired survival, was 3.4%, which is significant for presenting myeloma although the numbers of patients involved in this analy- samples, but is lower than the 13% reported in the next- sis means that these results should be treated as pre- generation sequencing analysis. All our mutations were liminary. Taken together, these data suggest that when missense mutations, as were 5 of 9 of the mutations deletion alone is being considered, del(1)(p32.3) is the reported previously (16). FAM46C consists of 2 exons, more prognostically important lesion. However, if deletion with only part of exon 2 being translated, and is made up and mutation events are considered together, FAM46C of 391 codons. It is notable that 35.7% (5/14) of muta- may still be prognostically important and future studies tions described in FAM46C in myeloma are in a small incorporating next generation sequencing data will illumi- region between codon 173 and 186, suggesting that this nate this further. may constitute a mutation hotspot (Fig. 5). As FAM46C In summary, mapping of homozygous deletions in shows occasional homozygous deletion in presenting newly presenting myeloma patients identified 2 regions myeloma cases, and has a reported mutation rate of of potential importance, 1p32.3 and 1p12. Deletion of between 3.4% and 13%, it likely represents a gene with 1p32.3 was strongly associated with adverse prognosis, pathogenic significance. but only in patients treated with ASCT, whereas in We examined the prognostic significance of abnormali- patients treated nonintensively its impact was neutral, ties of this gene and region. The association of deletion of or even slightly favorable. Although CDKN2C seemed 1p12 with adverse survival was confirmed by FISH in 379 to be the target of homozygous deletions of 1p32.3, it

Figure 5. Mutations of FAM46C described to date in myeloma.

7782 Clin Cancer Res; 17(24) December 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Survival Impact of 1p12/FAM46C and 1p32.3/CDKN2C in Myeloma

remains unclear if the adverse survival seen in patients Acknowledgments with hemizygous deletion was mediated through CDKN2C dysregulation, or if other genes played a role The authors thank Rebecca Protheroe, David Stockley, Tim Parker, Hazel Robinson, Elisabet Dachs Cabanas, and Christina Rudduck of the LLR UKMF in the observed clinical outcome. In comparison, the Cytogenetics group for probe preparation and FISH scoring, and staff from target of 1p12 deletion was more apparent, as the homo- the Clinical Trials Research Unit, University of Leeds, for trial coordination zygous deletions targeted only one gene, FAM46C,and and data management. The University of Leeds acted as the trial sponsor. this gene was found to be frequently mutated. Del(1) Grant Support (p12) was not associated with adverse survival to the same extent as del(1)(p32.3), but if deletion and muta- The study was supported by the National Institute for Health Research, tion events were both considered, there may be a prog- through the National Cancer Research Network. Main financial support for the MRC Myeloma IX clinical trial was from the UK Medical Research nostic effect. Taken together, these data highlight the Council, with additional funding in the form of unrestricted educational importance of genetic events affecting 1p in myeloma, grants from Novartis, Schering Health Care, Chugai, Pharmion, Celgene, and and show that the clinical relevance of these events needs Ortho Biotech. The LLR UKMF Cytogenetics group was supported by Leu- kemia and Lymphoma Research. The Institute of Cancer Research received to be individualized on the basis of the site of deletion financial support from Myeloma UK and the Biological Research Centre of and treatment context. the National Institute for Health Research at the Royal Marsden Hospital. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in Disclosure of Potential Conflicts of Interest accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Received July 11, 2011; revised September 11, 2011; accepted September No potential conflicts of interest were disclosed. 30, 2011; published OnlineFirst October 12, 2011.

References 1. Bergsagel PL, Kuehl WM. Molecular pathogenesis and a conse- predictor of poor outcome in myeloma patients undergoing an auto- quent classification of multiple myeloma. J Clin Oncol 2005;23: transplant. Biol Blood Marrow Transplant 2007;13:1066–72. 6333–8. 11. Walker BA, Leone PE, Chiecchio L, Dickens NJ, Jenner MW, Boyd KD, 2. Fonseca R, Blood E, Rue M, Harrington D, Oken MM, Kyle RA, et al. et al. A compendium of myeloma-associated chromosomal copy Clinical and biologic implications of recurrent genomic aberrations in number abnormalities and their prognostic value. Blood 2010;116: myeloma. Blood 2003;101:4569–75. e56–65. 3. Avet-Loiseau H, Attal M, Moreau P, Charbonnel C, Garban F, Hulin C, 12. Dickens NJ, Walker BA, Leone PE, Johnson DC, Brito JL, Zeisig A, et al. et al. Genetic abnormalities and survival in multiple myeloma: the Homozygous deletion mapping in myeloma samples identifies genes experience of the Intergroupe Francophone du Myelome. Blood and an expression signature relevant to pathogenesis and outcome. 2007;109:3489–95. Clin Cancer Res 2010;16:1856–64. 4. Ross FM, Chiecchio L, Dagrada G, Protheroe RK, Stockley DM, 13. Leone PE, Walker BA, Jenner MW, Chiecchio L, Dagrada G, Protheroe Harrison CJ, et al. The t(14;20) is a poor prognostic factor in myeloma RK, et al. Deletions of CDKN2C in multiple myeloma: biological and but is associated with long-term stable disease in monoclonal gam- clinical implications. Clin Cancer Res 2008;14:6033–41. mopathies of undetermined significance. Haematologica 2010;95: 14. Walker BA, Wardell CP, Chiecchio L, Smith EM, Boyd KD, Neri A, 1221–5. et al. Aberrant global methylation patterns affect the molecular 5. Hanamura I, Stewart JP, Huang Y, Zhan F, Santra M, Sawyer JR, et al. pathogenesis and prognosis of multiple myeloma. Blood 2010;2: Frequent gain of chromosome band 1q21 in plasma-cell dyscrasias 553–62. detected by fluorescence in situ hybridization: incidence increases 15. Chng WJ, Gertz MA, Chung TH, Van Wier S, Keats JJ, Baker A, et al. from MGUS to relapsed myeloma and is related to prognosis and Correlation between array-comparative genomic hybridization- disease progression following tandem stem-cell transplantation. defined genomic gains and losses and survival: identification of Blood 2006;108:1724–32. 1p31-32 deletion as a prognostic factor in myeloma. Leukemia 6. Fonseca R, Bergsagel PL, Drach J, Shaughnessy J, Gutierrez N, 2010;24:833–42. Stewart AK, et al. International Myeloma Working Group molecular 16. Chapman MA, Lawrence MS, Keats JJ, Cibulskis K, Sougnez C, classification of multiple myeloma: spotlight review. Leukemia Schinzel AC, et al. Initial genome sequencing and analysis of multiple 2009;23:2210–21. myeloma. Nature 2011;471:467–72. 7. Boyd KD, Ross FM, Tapper WJ, Chiecchio L, Dagrada G, Konn ZJ, 17. Morgan GJ, Davies FE, Gregory WM, Cocks K, Bell SE, Szubert AJ, et al. The clinical impact and molecular biology of del(17p) in multiple et al. First-line treatment with zoledronic acid as compared with myeloma treated with conventional or thalidomide-based therapy. clodronic acid in multiple myeloma (MRC Myeloma IX): a randomised Genes Cancer 2011;50:765–74. controlled trial. Lancet 2010;376:1989–99. 8. Boyd KD, Ross FM, Chiecchio L, Dagrada G, Konn ZJ, Tapper WJ, 18. Morgan GJ, Davies FE, Gregory WM, Russell NH, Bell SE, Szubert AJ, et al. A novel prognostic model in myeloma based on co-segregating et al. Cyclophosphamide, thalidomide, and dexamethasone (CTD) as adverse FISH lesions and the ISS: analysis of patients treated in the initial therapy for patients with multiple myeloma unsuitable for autol- MRC Myeloma IX trial. Leukemia 2011 Aug 12. doi: 10.1038/ ogous transplantation. Blood 2011;5:1231–8. leu.2011.204. [Epub ahead of print]. 19. Walker BA, Leone PE, Jenner MW, Li C, Gonzalez D, Johnson DC, et al. 9. Wu KL, Beverloo B, Lokhorst HM, Segeren CM, van der Holt B, Integration of global SNP-based mapping and expression arrays Steijaert MM, et al. Abnormalities of chromosome 1p/q are highly reveals key regions, mechanisms, and genes important in the path- associated with chromosome 13/13q deletions and are an ogenesis of multiple myeloma. Blood 2006;108:1733–43. adverse prognostic factor for the outcome of high-dose chemo- 20. Jenner MW, Leone PE, Walker BA, Ross FM, Johnson DC, Gonzalez therapy in patients with multiple myeloma. Br J Haematol D, et al. Gene mapping and expression analysis of 16q loss of 2007;136:615–23. heterozygosity identifies WWOX and CYLD as being important in 10. Qazilbash MH, Saliba RM, Ahmed B, Parikh G, Mendoza F, Ashraf N, determining clinical outcome in multiple myeloma. Blood 2007;110: et al. Deletion of the short arm of (del 1p) is a strong 3291–300.

www.aacrjournals.org Clin Cancer Res; 17(24) December 15, 2011 7783

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Boyd et al.

21. Chiecchio L, Protheroe RK, Ibrahim AH, Cheung KL, Rudduck C, cell dyscrasias is dependent on genetic context. Haematologica Dagrada GP, et al. Deletion of chromosome 13 detected by conven- 2009;94:1708–13. tional cytogenetics is a critical prognostic factor in myeloma. Leukemia 23. Dib A, Peterson TR, Raducha-Grace L, Zingone A, Zhan F, Hanamura 2006;20:1610–7. I, et al. Paradoxical expression of INK4c in proliferative multiple 22. Chiecchio L, Dagrada GP, Ibrahim AH, Dachs Cabanas E, Protheroe myeloma tumors: bi-allelic deletion vs increased expression. Cell RK, Stockley DM, et al. Timing of acquisition of deletion 13 in plasma Div 2006;1:23.

7784 Clin Cancer Res; 17(24) December 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst October 12, 2011; DOI: 10.1158/1078-0432.CCR-11-1791

Mapping of Chromosome 1p Deletions in Myeloma Identifies FAM46C at 1p12 and CDKN2C at 1p32.3 as Being Genes in Regions Associated with Adverse Survival

Kevin D. Boyd, Fiona M. Ross, Brian A. Walker, et al.

Clin Cancer Res 2011;17:7776-7784. Published OnlineFirst October 12, 2011.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-11-1791

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2011/10/12/1078-0432.CCR-11-1791.DC1

Cited articles This article cites 22 articles, 11 of which you can access for free at: http://clincancerres.aacrjournals.org/content/17/24/7776.full#ref-list-1

Citing articles This article has been cited by 11 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/17/24/7776.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/17/24/7776. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2011 American Association for Cancer Research.