Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

CLINICAL CANCER RESEARCH | TRANSLATIONAL CANCER MECHANISMS AND THERAPY

Whole-exome Sequencing in Penile Squamous Cell Carcinoma Uncovers Novel Prognostic Categorization and Drug Targets Similar to Head and Neck Squamous Cell Carcinoma A C Jad Chahoud1, Frederico O. Gleber-Netto2, Barrett Z. McCormick3, Priya Rao2, Xin Lu4, Ming Guo2, Maggaret B. Morgan2, Randy A. Chu2, Magaly Martinez-Ferrer5, Agda Karina Eterovic6, Curtis R. Pickering2, and Curtis A. Pettaway2

ABSTRACT ◥ Purpose: Penile squamous cell carcinoma (PSCC) is rare with mutation signatures are most comparable with HNSC signatures. limited treatment options. We report the first whole-exome PSCC samples showed an enrichment of two distinct mutational sequencing (WES) analysis and compare the molecular landscape signatures, the first, associated with oncogenic activity of of PSCC with other squamous cell carcinomas (SCC), with the goal AID/APOBEC, and the second, associated with defective DNA to identify common novel targets. mismatch repair and microsatellite instability. MP1 enrichment Experimental Design: PSCC and matched normal penile tissues was positively correlated with increased tumor mutation burden from 34 prospectively followed patients, underwent genomic WES (TMB; CC, 0.71; P < 0.0001) and correlated with significantly and human papilloma virus testing. We performed tumor mutation worse survival in comparison with those with the MP2 subset signature estimation by two methods, first to identify APOBEC- [HR, 10.2 (1.13–92.9); P ¼ 0.039]. We show that a subset of PSCC related mutation enrichments and second to classify PSCC- (38%), with enrichment of APOBEC-related mutation signature, enriched mutational patterns based on their association with the had significantly higher TMB and worse overall survival in Catalogue of Somatic Mutations in Cancer mutation signatures. We comparison with non-APOBEC–enriched subset [HR, 2.41 performed an extensive genomic comparison between our PSCC (1.11–6.77); P ¼ 0.042]. cohort and other SCCs in The Cancer Genome Atlas studies. Conclusions: This study identified novel druggable targets and Results: We identified that most PSCC samples showed enrich- similarities in mutational signatures between PSCC and HNSC with ment for Notch pathway (n ¼ 24, 70.6%) alterations, comparable potential clinical implications. with head and neck squamous cell carcinoma (HNSC). PSCC See related commentary by McGregor and Sonpavde, p. 2375

Introduction estimated median overall survival (OS) of 5 months (7) and very limited treatment options (8). Thus, it is important to perform Penile squamous cell carcinoma (PSCC) is a rare cancer in the molecular investigations to identify recurrent alterations associated United States with approximately 2,000 new cases and 400 deaths with PSCC and potential molecular targets for drug development in yearly (1). The incidence of PSCC is significantly higher in developing this rare disease with fatal outcomes (9, 10). The current evidence countries worldwide (2). Human papillomavirus (HPV) is the known investigating the genomic landscape of PSCC has been limited by small etiology for approximately 30%–50% of PSCC cases globally (3–5). þ sample size, limited HPV-positive (HPV ) cases, and heterogeneous The current treatment approach for locally advanced PSCC, using molecular testing methods (11–16). Prior efforts have identified two neoadjuvant platinum-based chemotherapy followed by surgical con- different pathways for penile oncogenesis, one that is HPV related and solidation offers long-term disease control for less than 30% of patients a second that is not. In addition, only a few recurrent point mutations with locally advanced PSCC (6); unfortunately, the majority of the in known tumor suppressor have been reported in PSCCC, patients will have disease progression or relapse. Relapsed PSCC after including TP53, CDKN2A, and PIK3CA (11–16). Therefore, the extent first-line chemotherapy is associated with poor survival, with an to which other genes are contributing to the disease, and thus, constitute additional targets for potential therapeutic exploration is 1 2 H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. The not well characterized. Therefore, a pressing need exists for the 3 University of Texas MD Anderson Cancer Center, Houston, Texas. Baptist identification of new therapies, based on an improved insight of the Health MD Anderson, Jacksonville, Florida. 4The University of Notre Dame, Notre Dame, Indiana. 5The University of Puerto Rico Medical Sciences Campus, disease biology. Other studies have found strong evidence for molec- San Juan, Puerto Rico. 6Viracor Eurofins Clinical Diagnostics, Lee’s Summit, ular convergence between squamous cell carcinomas (SCC) from Missouri. different anatomic sites (17), but this has not been explored yet in F.O. Gleber-Netto contributed equally to this article. PSCC. In this study, we performed the largest whole-exome analysis to date of 34 PSCCs to explore the genetics of this rare cancer. Corresponding Author: Curtis A. Pettaway, The University of Texas MD Ander- son Cancer Center, Houston, TX 77030. Phone: 1 (713) 482-9138; Fax: (713) 794- 4824; E-mail: [email protected] Materials and Methods Clin Cancer Res 2021;27:1–11 PSCC cohort doi: 10.1158/1078-0432.CCR-20-4004 PSCC and matched normal penile tissues (n ¼ 34 pairs) were 2021 American Association for Cancer Research. obtained from the University of Texas MD Anderson Cancer Center

AACRJournals.org | OF1

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Chahoud et al.

unstained tissue sections were used for specimen preparation. Briefly, Translational Relevance the FFPE tissue sections were deparaffinized by xylene (3) for 10 The translation of molecular research findings in cancer therapy minutes each, followed by an ethanol wash (3) to remove paraffin. has proven to be successful, yet remains challenging. This is Deparaffinized tissue pellets placed in 1.5 mL microcentrifuge tube particularly ambitious for rare deadly cancers, like penile squa- were air dried and resuspended in 200 mL of Lysis Reagent (10% mous cell carcinoma (PSCC). Novel molecular evidence can be proteinase K in an ATL Buffer, Qiagen). The mixture was incubated in valuable to improve prognostication, identify therapy targets, and a56C water bath for 1 hour and diluted into 2 mL with 50% ethanol. assess similarities between PSCC and other squamous cell tumors. After deparaffinization and specimen pretreatment, 1 mL solution was We identified that PSCC shares considerable molecular and his- applied to Cobas 4800 system, following the manufacturer’s instruc- tologic similarity with head and neck squamous cell carcinoma tion. Cobas HPV assay is an FDA-approved HPV testing assay for (HNSC), specifically involving the Notch pathway, suggesting that cervical cancer screening in Pap cytology specimens. The Cobas 4800 clinical developments from the much larger HNSC patient cohort system is a highly automated HPV testing platform, including DNA could help expedite the translation to the less common PSCC extraction, a real-time PCR thermocycler, and an analyzer for the population. In this context, we provide novel evidence that distinct testing result reporting. In each testing run, positive and negative molecular subtypes with prognostic implication in PSCC were controls, as well as an internal control with b-globin amplification to most similarly enriched in HNSC in comparison with other tumors determine the presence of DNA, were included. The testing results with squamous cell histology. Our data provide a basis to inves- were reported as HPV 16 and 18 genotypes, as well as other collectively tigate molecularly targeted strategies in PSCC and suggest potential 12 high-risk HPV types (HPV 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 66, for drug development research using molecular-specific trials and 68). The FFPE specimen in Cobas 4800 system for HPV testing across PSCC and HNSC may provide advantages to both cancers. was validated in-house in our institution.

Exome capture and sequencing Genomic DNA was extracted from tumor and peripheral nonma- (UT MDACC, Houston, TX) tissue bank of prospectively collected lignant frozen tissue using the QIAmp DNA Tissue Kit (Qiagen) samples. The diagnosis of PSCC was made based on the review of an following the manufacturer’s instructions. Genomic DNA was quan- expert pathologist specializing in genitourinary cancers (P. Rao), who tified by PicoGreen (Invitrogen) and quality was accessed using Geno- assessed and reported the tumor histologic features per the American mic DNA Tape for the 2200 TapeStation (Agilent Technologies). Only Joint Committee on Cancer 8th edition (18). All specimens were samples with more than 200 ng of genomic DNA (optimal quantity obtained from patients who were evaluated at UT MDACC (Houston, threshold) and with DNA integrity number higher than 7 (optimal TX), and following informed consent under protocols approved by the quality threshold) were considered for library preparation. Exome institutional review board. Paired PSCC and normal tissues were capture wasperformed with the SeqCapEZ Exome Probes v3.0 (Roche), obtained snap frozen from the same patient at the time of surgery. which covers 64 megabases (Mb) of (GRCh37). The Most tumors were collected prior to receiving chemotherapy (9/10), captured libraries were sequenced on a HiSeq 4000 (Illumina Inc.) for 2 when indicated clinically. The percentage of malignant tissue was 100 paired end reads, using Cycle Sequencing v3 Reagents (Illumina). calculated by histologic examination, following hematoxylin and eosin Only sequenced samples with >80% of data with quality score ≥ Q30 (H&E) staining. All PSCCs analyzed comprised at least 50% malignant (99.9% of base call accuracy) were used in the analysis. penile cells. The studies were conducted in accordance with the Declaration of Helsinki and informed written consent was obtained Data processing from each subject or each subject’s guardian. TheresultingrawoutputBCLfiles containing the sequence data were converted into FASTQ files using CASAVA 1.8.2. FASTQ files Assessment of HPV status were aligned to the reference genome (human Hg19) using For IHC for p16INK4a, all samples were tested, and staining was BWA (22). The aligned BAM files were subjected to mark performed on a BenchMark Autostainer (Ventana Medical Systems) duplication, realignment, and recalibration using Picard and as described by the manufacturer’s protocol using a prediluted mouse GATK (23) before any downstream analyses. The mean number mAb (CINtec p16 Histology, clone E6H4, Ventana Medical Systems). of total reads generated for the 34 tumor samples was 185.1 26.6 Microscopic slide evaluation for p16INK4a staining patterns was clas- million, with mean of 99.3% 0.1% of DNA mapping rate and sified as follows: pattern 0, complete absence of p16INK4a staining in mean coverage of 141.120.5. For the 34 paired normal tissue all the neoplastic cells; pattern 1, spotty and discontinuous individual samples, the mean number of reads generated was 99.8 14.8 staining in some of the neoplastic cells; pattern 2, a more extensive, million, with 99% 0.1% of mean DNA mapping rate and mean albeit discontinuous staining pattern with small clusters of positive coverage of 70.29.2. Somatic mutation calls were performed neoplastic cells; and pattern 3, entire and continuous cytoplasmic or using MuTect (24), and insertions and deletion (indel) calls using nuclear staining in all neoplastic cells, except in hyperkeratotic or Pindel (24). Resulting data were combined into mutation annota- parakeratotic areas when present. On the basis of prior analyses, only tion format (MAF) files, and then results were manually filtered on pattern 3 was considered as positive for p16INK4a overexpres- the basis of the following parameters: nucleotide changes were sion (19, 20). The p16INK4a staining patterns were confirmed by an considered only when we observed a minimum of 20 coverage experienced pathologist (P. Rao). for tumor samples and 10 for normal samples, and with variant HPV genotyping for all samples was performed using Cobas HPV allelefraction(VAF)higherorequalto0.02fortumorsandlower Assay (Roche Diagnostics) as described elsewhere (21). First, the than or equal to 0.02 for normal samples for single-nucleotide formalin-fixed, paraffin-embedded (FFPE) tissue sections were pre- variants (SNV). For INDELs, the minimum VAF considered was pared from each block (5 mm) with the last section stained with H&E 0.05. All variants described as SNPs at 1000 Genome Project and evaluated to determine the quality of the specimen. Two to three database (25), ESP6500 (26), and EXAC (27), with frequency

OF2 Clin Cancer Res; 27(9) May 1, 2021 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Clinical Implications of WES in Penile Squamous Carcinoma

reported as higher than 0.01, were excluded. All events occurring in MAF files of each cohort were analyzed by using the Maftools R regions with local repeats were also excluded. package, in which we searched for differentially mutant genes between PSCC and squamous TCGA cohorts. Finally, we estimated APOBEC Data analysis and COSMIC mutation signatures, as described previously, for all PSCC whole-exome analysis TCGA squamous tumors and PSCC samples together, by compiling PSCC filtered variants organized in an MAF file were analyzed using mutation information in a single MAF file. the R package, Maftools (28), which congregates multiple genomic analytic tools, as briefly described below. Estimation of hypermutated Statistical analysis genomic regions was performed by assessing interevent distances in Associations between categorical variables were assessed by Fisher each . Genomic areas exhibiting six or more consecutive exact test, or x2 test, whenever appropriate. Associations between events with an average intermutation distance of 1,000 bp were categorical and continuous variables were assessed by Mann–Whitney considered hypermutated. PSCC driver genes were estimated by U test for pairwise comparison and by Kruskal–Wallis test followed by mutation positional analysis, which searches for mutation enrichment Tukey test for three or more categorical features. Correlations between in specific loci, or hotspots, as described by Tamborero and collea- continuous variables were assessed by Pearson correlation test. For gues (29). We also assessed the enrichment level of 10 oncogenic survival analysis, we performed the log-rank test and univariate Cox signaling pathways (RTK/RAS pathway, Wnt pathway, Notch path- regression. Analyses were performed using JMP 15.0 Software. way, Hippo pathway, PI3K pathway, cell-cycle pathway, Myc pathway, TGFb pathway, p53 pathway, and Nrf2 pathway) in our PSCC samples Study approval on the basis of the curated set of genes described by Sanchez-Vega and Collection and use of patient samples for this study were approved colleagues (30). In our work, we considered all missense and truncated by the UT MDACC (Houston, TX) Institutional Review Board (PA15- mutations occurring in genes described in their set. 0138). Estimation of gene druggability was performed by accessing data from the DGIdb database that include gene–drug interaction infor- mation (31). Tumor variants were classified into one of the 96 Results nucleotide substitution classes that consider the mutant base and the PSCC cohort characteristics immediate 30 and 50 nucleotides around it. The classified variants were Thirty-four untreated primary PSCC specimens and corresponding then used for tumor mutation signature estimation by two methods. matching normal penile tissue samples were subjected to whole-exome The first method aimed to identify APOBEC-related mutation enrich- sequencing (WES). Clinicopathologic characteristics of these patients ments. APOBEC-induced mutations tend to be characterized by an are described in Table 1. The median age at diagnosis for this cohort enrichment of TpCpW motif, in which W corresponds to an A or T. was 61 years (range, 39–83 years). Twenty-one patients were Cauca- Using the approach proposed by Roberts and colleagues (32), the sians (62%), 11 were Hispanic (32%), and 2 (6%) were African- þ enrichment of C>T mutations associated with the TpCpW motif was Americans. The cohort included 10 (29%) tumors that were HPV compared with all C>T events and other background events to as assessed by Cobas HPV test, but only eight of those were p16 positive þ calculate an enrichment score. This enrichment score was then (p16 ) by IHC staining. Most patients (n ¼ 24, 70%) had usual SCC evaluated by Fisher exact test, and then, samples were categorized histology, while six (17%) tumors were basaloid, two (6%) were into APOBEC-enriched and non-APOBEC–enriched groups. The papillary not otherwise specified (NOS), and two (6%) were classified second method of signature estimation focused on the identification as verrucous carcinoma. Pathologic primary cancer staging associated of PSCC mutational patterns and association of these patterns with the with high metastatic potential risk was observed in (T2–T4) 24 patients version 3 of the mutation signatures [part of the v89 Catalogue of (71%) with T2–T4 tumors, in 25 patients (74%) with grade 2–3 tumors, Somatic Mutations in Cancer (COSMIC), released in May 2019] from in 16 (47%) patients with lymphovascular invasion, and in 12 (46%) COSMIC database (cancer.sanger.ac.uk; ref. 33). Initial step involves patients with perineural invasion. On the other hand, most patients the estimation of the number of different mutation patterns among the (n ¼ 18, 53%) had clinically N0 disease at the time of diagnosis, defined PSCC samples by nonnegative matrix factorization analysis and as no evidence of palpable lymph nodes based on physical exam and cophenetic correlation. Then, the nucleotide substitution classes are nodal imaging at diagnosis. Pathologically, 21 patients (68%) with decomposed into the estimated number of PSCC signatures and finally enough follow-up were pN0. In this cohort, 10 (29.4%) patients compared with COSMIC mutation signatures and the best match is received neoadjuvant chemotherapy, the majority (n ¼ 9, 90%) identified by cosine similarity calculation. received the combination chemotherapy regimen with cisplatin, ifos- famide, and paclitaxel, followed by consolidation inguinal and pelvic Pan-squamous whole-exome data comparison lymph node dissection in all 10 patients. The median follow-up time Aiming to compare PSCC genomic data with other known for the entire cohort was 32.2 months, during this period, 5 (14.7%) squamous carcinomas, we retrieved MAF files from The Cancer patients died [only 3 (8.8%) died from PSCC], 3 (8.8%) patients were Genome Atlas (TCGA) studies with carcinoma cohorts (34) using alive with disease, and 26 (76.4%) remained alive without evidence of TCGA mutations R package, which included the following sites: disease. Only 8 (24%) patients had disease progression or recurrence. A bladder (dx.doi.org/10.7908/C1MW2GGF), cervix (dx.doi.org/ detailed description of the clinical and pathologic characteristics is 10.7908/C1MG7NV6), esophagus (dx.doi.org/10.7908/C1BV7FZC), presented in Supplementary Table S1. head and neck (dx.doi.org/10.7908/C18C9VM5), and lung (dx.doi. org/10.7908/C134WXV). Because these studies included several Landscape of somatic mutations and enriched mutational histologic variants of carcinomas, we further filtered these MAF files patterns in PSCC by including only those with squamous histology, as described by WES was performed on 34 tumor–normal pairs. All samples were Campbell and colleagues (35). The list of included samples is described sequenced using a whole-exome capture system covering 64 Mb of in the Supplementary Table S1. the human genome (CRCh37). Sequencing depth of targeted

AACRJournals.org Clin Cancer Res; 27(9) May 1, 2021 OF3

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Chahoud et al.

Table 1. Patient characteristics. 99.1% 0.1%, respectively. Collectively, the samples presented 4,275 somatic exonic mutations, including 3,717 missense, 327 Characteristic n (%) nonsense, 1,607 silent, 75 splice site, 95 frameshift indels, and 61 Age at diagnosis, median (range) 61 (39–83) in-frame indels. The full set of nucleotide variants found in PSCC Race samples is described in the Supplementary Table S2. The 30 most Caucasian 21 (62%) frequent exonic mutations identified in PSCC are described in the African-American 2 (6%) oncoplot in Fig. 1A. Hispanic 11 (32%) Patient’s tumor mutation burden (TMB) was determined by divid- Neonatal circumcision (no) 21 (61%) ing the total number of SNVs by the amount of the genome covered by Tobacco use (yes) 24 (70%) whole exome sequencing (64 Mb). TMB was then represented by the Penile trauma (yes) 3 (9%) number of SNVs per Mb. The mean TMB among patients with PSCC Balanitis (yes) 8 (24%) was 1.96 (ranging from 0.01 to 21.2). On the basis of Tukey outlier test, Phimosis (yes) 9 (26%) fi Clinical T stage 2 patients (14T and 27T) were classi ed as hypermutators (Fig. 1B). fi fi 1 7 (21%) Analysis of intermutation distances further con rmed this classi ca- 2 17 (50%) tion by identifying areas of hypermutated loci (six consecutive muta- 3 6 (18%) tions with an average distance of less than or equal to 1,000 bp) only in 4 4 (12%) these two samples (Fig. 1C). Clinical N stage On the basis of the manually curated gene set presented by Sanchez- 0 18 (53%) Vega and colleagues (30), we observed that multiple genes associated 1 7 (20%) with oncogenic pathways were highly mutated in PSCC samples 2 3 (9%) (Supplementary Fig. S1). Notch pathway was the most frequently 3 6 (18%) ¼ Pathologic T stage affected in PSCC samples, in which 70.6% of samples (n 24) had 1 13 (38%) mutations in at least one of the 17 Notch genes, including NOTCH1 ¼ ¼ ¼ 2 8 (24%) (n 15, 44.1%), EP300 (n 5, 14.7%), and FBXW7 (n 5, 3 11(32%) 14.7%; Fig. 1D). Other highly affected pathways were: Hippo pathway 4 2 (6%) with 17 mutant genes among 20 (58.8%) patients, in which 12 (35.3%) Pathologic N stage exhibited FAT1 mutations; RTK-RAS with 22 mutant genes among 16 0 21 (68%) patients (47.1%), which included three (8.8%) cases with HRAS 1 3 (10%) mutations and other three with NF1 mutations; p53 pathway with 2 3 (10%) only three mutant genes among 15 patients (44.1%), but with TP53 3 4 (12%) mutations involving 12 (35.3%) of them; and PI3K, cell cycle, and Wnt Lymphovascular invasion (yes) 17 (50%) Perineural invasion (yes) 17 (50%) pathways were mutated in 11 (32.3%) patients, and PIK3CA, CDKN2A ¼ ¼ HPV status (PCR Cobas) (n 10, 29.4%), and CHD8 (n 3, 8.8%) mutations were the HPV negative 24 (70%) most frequent for each pathway, respectively. Nrf2 (n ¼ 4, HPVþ 10 (30%) 11.7%), TGFb (n ¼ 3, 8.8%), and MYC (n ¼ 3, 8.8%) were less p16 IHC negative 22 (65%) enriched among PSCC samples, and no gene was mutated in more p16 IHC positive 12 (35%) than two samples. TMB was significantly higher among patients Anatomic site of tumor with mutations in Wnt [mutation (Mut) ¼ 4.15 5.8 vs. wild-type Glans only (including foreskin) 19 (56%) (WT) ¼ 0.9 0.7; P ¼ 0.0003], RTK-RAS (Mut ¼ 3.16 5.1 vs. Shaft only 3 (9%) WT ¼ 0.90 0.7; P ¼ 0.010), Notch (Mut ¼ 2.44 0.8 vs. Glans and shaft 12 (35%) WT ¼ 0.80 0.3; P ¼ 0.014), and p53 (Mut ¼ 2.01 1.6 vs. WT ¼ Surgery type ¼ Partial penectomy 18 (53%) 1.92 4.7; P 0.013) pathways. Radical penectomy 13 (38%) Positional enrichment analysis indicated that mutations in PIK3CA Wide local excision 3 (9%) and CDKN2A were significantly enriched in specific loci Grade (FDR ¼ 0.007 and FDR ¼ 0.039, respectively), suggesting these may 1 9 (26%) be potential driver mutations in PSCC (Supplementary Fig. S2). The 2 18 (53%) PIK3CA mutations, p.E545K/G, E542K, and E453K, are all considered 3 7 (21%) gain-of-function oncogenic mutations by OncoKB, while the Neoadjuvant chemotherapy (yes) 10 (29.4%) CDKN2A mutations, p.R80X, p.E88X, p.G65fs, p.D108G, and ORR 80% p.R115fs, are classified as likely loss-of-function and likely oncogen- Number of patients with disease recurrence/progression 8 (24%) Follow-up (months), median (range) 32.2 (2–77) ic (36). The most frequent SNVs among patients with PSCC were > Disease status characterized by C T (mean of 48% of all SNVs/patient) changes, Dead of disease 3 (8.8%) followed by C>G (16.5%), C>A (15.4%), T>C (12.1%), T>G (4.2%), Dead of other causes 2 (5.8%) and T>A (3.8%; Fig. 1A). Alive with disease 3 (8.8%) We estimated the mutational patterns for each patient based on the Alive, no evidence of disease 26 (76.4%) frequency of single-base substitutions in the context of immediate 50 and 30 bases, classifying 96 different mutation types. By performing Abbreviation: ORR, overall response rate. cophenetic analysis, we identified two distinct mutation patterns among PSCC samples, which we called mutation pattern 1 (MP1) regions was 141.120.2 fortumorsamplesand70.29.1 and 2 (MP2; Fig. 2A and B). Enrichment of each of these two for normal samples, with a mapping rate of 99.3% 0.1% and mutation patterns for each individual sample is described in

OF4 Clin Cancer Res; 27(9) May 1, 2021 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Clinical Implications of WES in Penile Squamous Carcinoma

Figure 1. Mutation characteristics of PSCC. A, Oncoplot displays the mutation status and mutation type for the top 30 mutat- ed genes among patients with PSCC and their corresponding HPV, p16, smoking status, and histologic subtype. Horizon- tal bar plot shows the total number of mutations per tumor (top). Bar plot dis- plays the frequency/number of mutant patients per gene (vertical, right). Bar plot shows the overall nucleotide change type frequency in each tumor (bottom). TMB and mutation density. B, Violin plot shows the distribution of number of mutations per sequenced Mb among PSCC samples (mean ¼ 1.96, black line; 1st and 3rd quartiles, dotted lines; minimum, 0.01; and maximum, 21.2). Hypermutator phenotype was tested by Tukey outlier test revealing two hypermutator samples (samples 14T and 27T, red dots). C, Hypermutated regions or kataegis loci (defined as six consecutive mutations with an average distance of 1,000 bp) are repre- sented in the rainfall plots (x-axis, chro- mosome location and y-axis, distance between single-nucleotide variations) as highlighted in dashed areas and arrows at 2, 3, and 22 for sample 14T and at chromosome 9 for sample 27T. Dots represent all nucleo- tide changes for each sample and are colored according to the type of nucle- otide change. D, Ring plots represent the frequency of samples (n ¼ 34) with mutation in any gene in the described pathways. The curated gene lists for each pathway were determined by Sanchez-Vega and colleagues (30), and are depicted in detail in the Supplemen- tary Fig. S1. Pap. NOS, papillary carcino- ma NOS; SCC usual, SCC usual type.

the Fig. 2C. MP1 was the most prevalent in nine PSCC samples, (0.618) and SBS7a (0.644) COSMIC signatures. SBS13 is also whileMP2wasmoreprevalentintheother25cases.These associated with AID/APOBEC cytidine deaminases family, and mutation patterns were then compared with known cancer muta- both are related to local hypermutation phenomenon called kataegis tion signatures from COSMIC database (Mutation Signatures V3, (C>G – TC context). Interestingly, MP1 enrichment was positively May 2019). Cosine similarity scores indicated the closest matches correlated with increased TMB (CC ¼ 0.71; P < 0.0001). By between PSCC mutation patterns and COSMIC signatures the other side, SBS7a signature is commonly found in tumors (Fig. 2D). The MP1 showed the highest cosine similarity to the associated with sun exposure and are likely caused by UV radiation SBS2 (0.767) COSMIC signature, which has as a proposed etiology (C>T TC/CC). the activity of AID/APOBEC family of cytidine deaminases (C>T – The MP2 had the highest cosine similarity with the signature SBS6 TC context). MP1 also presented high cosine similarity with SBS13 (0.836), which is associated with defective DNA mismatch repair

AACRJournals.org Clin Cancer Res; 27(9) May 1, 2021 OF5

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Chahoud et al.

Figure 2. Mutational patterns in PSCC. Mutational patterns, inferred by cophenetic correlation, indicated two main mutation signatures defined as MP1 (A), characterized by an enrichment of C>TandC>G changes at TCN context (N, any of the four bases), and at lower frequency C>A changes at the same TCN context and MP2 (B), characterized by an enrichment of C>T changes, mainly at NCG context. C, The MP1 and MP2 were compared with each single-base substitution signatures (v3) from COSMIC database by estimation of their cosine similarity, represented in the heatmap. MP1 was highly similar to signatures SBS2 (CS ¼ 0.767), SBS7a (CS ¼ 0.644), and SBS13 (CS ¼ 0.618). SBS2 and SBS13 signatures are attributed to the activity of APOBEC family of cytidine deaminases, while SBS7a is attributed to UV light exposure. MP2 was highly similar to signatures SBS6 (CS ¼ 0.836), SBS1 (CS ¼ 0.796), and SBS15 (CS ¼ 0.708). SBS6 and SBS15 are associated with defective mismatch repair system, while SBS1 is linked to deamination of 5-methylcytosine to thymine. D, Nine penile cancer samples showed enrichment of MP1 (mustard bars) and 25 showed enrichment of MP2 (green bars). APOBEC enrichment score (estimated by an independent approach) is shown in the y-axis and is represented as red diamonds for significantly enriched samples and as gray diamonds for not enriched samples. High APOBEC enrichment scores were observed among tumors exhibiting MP1. E, Kaplan–Meier plot shows OS curves for penile cancer samples with enrichment of mutation profiles 1 (mustard line) and 2 (green line). OS of patients with penile cancer with tumors enriched for mutation profile 1 was significantly lower (four deaths, 44.4%) than patients with enrichment of mutation profile 2 (one death, 4%). F, Kaplan–Meier plot shows OS curves for APOBEC-enriched (red line) and non-APOBEC–enriched (gray line) penile cancer samples. Patients with APOBEC-enriched tumors (four deaths, 30.8%) showed worse OS than patients with non-APOBEC–enriched tumors (one death, 4.8%), but the difference was not significant. CI, confidence interval; nt, nucleotide.

OF6 Clin Cancer Res; 27(9) May 1, 2021 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Clinical Implications of WES in Penile Squamous Carcinoma

system, microsatellite instability, and small indels, as well as the other Because MP1 score was highly enriched by mutational signatures enriched signature SBS15 (0.708). Another highly enriched signature associated with APOBEC cytidine deaminases activity, we also in MP2 was the SBS1 (cosine similarity ¼ 0.796), which is associated explored the impact of APOBEC scores in these samples. High with deamination of 5-methylcytosine to thymine, especially in the APOBEC score was significantly associated with mortality (P ¼ NpCpG context. 0.009) and presence of lymph node metastasis (P ¼ 0.024). Increased Considering that the MP1 was highly associated with mutation APOBEC score was significantly associated with poor OS by univariate signatures associated with APOBEC cytidine deaminases activity, we Cox regression [HR, 2.41 (1.11–6.77); Wald test P ¼ 0.042]. When also assessed the enrichment of APOBEC-related mutagenesis pattern samples were dichotomized in APOBEC-enriched and non-APOBEC, by using an algorithm described by Roberts and colleagues (2013), a trend toward worse survival among APOBEC-enriched samples was which considers the rate of TpCpW (in which W may be an adenine or observed, although not statistically significant (Fig. 2F; P ¼ 0.060; HR, a thymine) changes as a marker of APOBEC-related mutagenesis 6.2; 95% confidence interval, 0.7–56). process (32). We estimated that 38% (n ¼ 13) of PSCC samples had an enriched APOBEC-related mutation signature (Fig. 2C) and supports Druggable genes the existence of this subset of PSCC. These APOBEC-enriched samples Among the mutant genes found in PSCC samples, 11 have been had a significantly higher TMB (3.87 5.43) compared with non- described in the DGIdb database as targetable by known drugs APOBEC–enriched samples (0.78 0.55; P < 0.0001). (PIK3CA, 338 drugs; TP53, 193 drugs; CACNA1C, 44 drugs, CDKN2A, 31 drugs; NOTCH1, 26 drugs; FBXW7, 11 drugs; EP300, 10 drugs; Clinicopathologic associations MUC2, nine drugs; CASP8, six drugs; LAMA1, one drug; and TTN, one High p16 immunoexpression is a clinically approved surrogate drug). Drug–gene interaction information is detailed in the Supple- marker of HPV infection for cervical and oropharyngeal tumors. mentary Table S3. From these drug–gene interactions, two of these Although HPV detection and p16 staining were associated in the have been the basis for the FDA drug authorization of a PI3Ka-specific majority of PSCC cases, in two samples (17T and 2T) HPV was inhibitor and CDK4/6 inhibitor to treat patients with advanced breast detected by HPV PCR testing, but the tumors were p16 negative by cancer (37, 38). In addition, deleterious mutations in DNA damage IHC. For both cases, nonsense CDKN2A mutations were detected, responsive (DDR) genes are frequently associated with response to which could explain the absence of p16 immunoexpression. However, PARP inhibitors and platinum chemotherapy (39–41). The DDR the case 17T showed TP53 and CASP8 mutations, which are rare pathway is important in tumor biology, allowing cancer cells a þ among HPV tumors from TCGA database. Whether these two cases mechanism to resist damage by chemotherapy and radiotherapy (42). were HPV driven is unknown. BRCA1/2 are the most well-described genes in the pathway, but several HPV detection was significantly associated with mutations in the others (ATM, ATR, PALB2, etc.) are involved with DDR and are ARPP21, CMYA5, and RPGRIP1 genes (all mutant patients were mutated in many cancers. However, much remains unknown about þ HPV ; P ¼ 0.004, P ¼ 0.020, and P ¼ 0.020, respectively). P53 their prevalence in PSCC, therefore, we report the prevalence of likely pathway genes (TP53 and CHECK2) were mutated in only two pathogenic variants in DDR genes. We identified nine genes (BRCA1/ þ (20%) HPV cases, while more than half (n ¼ 13, 54.2%) of the 2, ARID1A, ATR, CHEK2, PARP1, FANCA, PALB2, and RAD51)as HPV-negative cases had mutations in one of these genes (P ¼ 0.128). DDR-related on the basis of PubMed searches and the NCBI Gene and þ No Nrf2 pathway genes were mutated in HPV PSCC. Biosystems Databases. A total of 26 pathogenic or likely pathogenic p16 immunoexpression was significantly associated with CSPG4 (3/ variants in DDR genes were identified in 8 of 34 (23.5%) patients. þ þ 4 mutant patients were p16 ; P ¼ 0.032) and TP53 (all p16 tumors These variants were found in the following genes: ARID1AM 2 patients þ were WT; P ¼ 0.030). Although not significant (P ¼ 0.072), no p16 (5.9%), BRCA2 2 patients (5.9%), ATR 2 patients (5.9%), CHEK2 2 case presented CDKN2A mutations. P53 pathway genes affected 14 patients (5.9%), PARP1 2 patients (5.9%), FANCA 1 patient (2.9%), p16-negative tumors (53.8%), but only one (12.5%) p16-positive case PALB2 1 patient (2.9%), and RAD51 1 patient (2.9%). þ (P ¼ 0.053). No Nrf2 pathway mutations were detected among p16 tumors. Comparison between PSCC and other squamous carcinomas Local recurrence was observed in 20% (n ¼ 3) of patients with from TCGA mutations in p53 pathway, while no local recurrence was detected in We compared the genomic alterations of PSCC with head and patients with no p53 pathway mutations (P ¼ 0.076). In fact, all local neck squamous cell carcinoma (HNSC; n ¼ 501), cervical carcinoma recurrence cases occurred among TP53-mutant patients (P ¼ 0.037). (n ¼ 241), esophageal carcinoma (n ¼ 95), bladder carcinoma Although development of regional recurrence was rare (n ¼ 3), 2 (n ¼ 47), and lung squamous carcinoma (LUSC; n ¼ 471) using (66.7%) of these patients were TGFb pathway mutants (P ¼ 0.015) and scalable available whole-exome data (34). TMB of PSCC tumors was all had Notch pathway mutations (P ¼ 0.538). the lowest of all squamous tumors analyzed, being significantly lower Patients with mutations in the Notch pathway showed a trend than any other squamous carcinoma investigated (Fig. 3A). When toward worse OS (log-rank P ¼ 0.150), and all deceased patients (n ¼ comparing the TMB of PSCC tumors with other solid malignancies 5) were mutant for this pathway (20.9% of all Notch pathway–mutant (including nonsquamous tumors from the abovementioned sites) and patients). On the other hand, patients with mutations in the PI3K lymphomas, we found that PSCC has an intermediate TMB among pathway tended to have improved progression-free survival when TCGA-sequenced tumors (Supplementary Fig. S3). compared with patients with no mutations in this pathway (log-rank Considering the 30 genes with highest average mutation frequency P ¼ 0.196). Progression occurred in 1 mutant patient (9.1%), while 7 among squamous carcinomas from TCGA, PSCC cohort had a similar (30.4%) patients without PI3K pathway mutations progressed. mutation frequency in 22 of them (73.3%; Supplementary Table S4). Considering the mutational pattern scores, enrichment of MP1 was The other eight genes that differentially mutated between PSCC and significantly associated with worse OS status (Fig. 2E; P ¼ 0.011). In TCGA squamous cohorts were TP53, TTN, CSMD3, RYR2, KMT2D, fact, univariate Cox regression showed that increased MP1 score was FAT1, CDKN2A, and NOTCH1 (Fig. 3B). In addition, PSCC showed associated with lower OS [HR, 10.2 (1.13–92.9); Wald test P ¼ 0.039]. mutations in KDM6A, CASP8, LAMA1, DNAH6, and MUC2 genes

AACRJournals.org Clin Cancer Res; 27(9) May 1, 2021 OF7

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Chahoud et al.

Figure 3. Genomic comparison between PSCCs and TCGA squamous carcinomas. PSCC samples (n ¼ 34) were compared with HNSC (n ¼ 501), bladder (BLCA) (n ¼ 47), esophageal (ESCA) (n ¼ 95), cervical (CESC) (n ¼ 241), and LUSC (n ¼ 471) squamous cell carcinomas. A, Median TMB of PSCC [1.28 mutations (mut)/Mb, based on a 50-Mb sequencing coverage] was significantly lower than median TMB in LUSC (4.42 mut/Mb; P < 0.0001) and bladder carcinoma (2.8 mut/Mb; P ¼ 0.001), but not significantly different from median TMB in esophageal carcinoma (1.84 mut/Mb; P > 0.999), HNSC (2.04 mut/Mb; P ¼ 0.121), and cervical carcinoma (1.74 mut/Mb; P ¼ 0.491) samples. B, Mutation frequency of 13 genes significantly different between PSCC and other squamous tumors. The heatmap depicts their mutation frequency

according to each cohort. Significant differences (based on Padj cutoff of 0.05) between PSCC and other squamous cohorts are represented with asterisks. C, Whole- exome data from PSCC, bladder carcinoma, cervical carcinoma, esophageal carcinoma, HNSC, and LUSC were combined and analyzed as a unique cohort. Mutational patterns were inferred by cophenetic correlation, revealing three different mutational patterns among these tumors, named Sq-MP1, Sq-MP2, and Sq-MP3. Mutation pattern distribution in PSCC (Sq-MP1 ¼ 23.5%, Sq-MP2 ¼ 67.6%, and Sq-MP3 ¼ 8.8%) was significantly different from bladder carcinoma (Sq-MP1 ¼ 53.2%, Sq-MP2 ¼ 31.9%, and Sq-MP3 ¼ 14.9%; P ¼ 0.006), cervical carcinoma (Sq-MP1 ¼ 61%, Sq-MP2 ¼ 38.2%, and Sq-MP3 ¼ 0.8%; P < 0.0001), and LUSC (Sq-MP1 ¼ 8.7%, Sq-MP2 ¼ 5.1%, and Sq-MP3 ¼ 86.2%; P < 0.0001), but similar to esophageal carcinoma (Sq-MP1 ¼ 14.7%, Sq-MP2 ¼ 45%, and Sq-MP3 ¼ 13.9%; P ¼ 0.387) and HNSC (Sq-MP1 ¼ 21.3%, Sq-MP2 ¼ 52.5%, and Sq-MP3 ¼ 19.8%; P ¼ 0.208) samples. D, APOBEC enrichment classification of PSCC samples (APOBEC enriched ¼ 38.2% and non- APOBEC ¼ 61.8%) was significantly different of cervical carcinoma (APOBEC enriched ¼ 75.1% and non-APOBEC ¼ 24.9%; P < 0.0001) and bladder carcinoma (APOBEC enriched ¼ 78.7% and non-APOBEC ¼ 21.3%; P ¼ 0.0004), but similar to LUSC (APOBEC enriched ¼ 30.3% and non-APOBEC ¼ 69.7%; P ¼ 0.340), esophageal carcinoma (APOBEC enriched ¼ 37.9% and non-APOBEC ¼ 62.1%; P ¼ 1.00), and HNSC (APOBEC enriched ¼ 44.9% and non-APOBEC ¼ 55.1%; P ¼ 0.480).

that are not frequently mutated in the majority of TCGA squamous bladder carcinoma (n ¼ 13, 27.7%). HNSC (n ¼ 85, 17%), esophageal tumors (Fig. 3B). Patients with PSCC had increased NOTCH1 muta- carcinoma (n ¼ 13, 13.7%), and cervical carcinoma (n ¼ 44, 18.3%) tion frequency compared with patients with bladder carcinoma, cohorts showed lower number of DDR-mutant patients, but no cohort cervical carcinoma, esophageal carcinoma, and LUSC. Similarly, FAT1 was significantly different from PSCC cohort. mutants were more frequent in PSCC than in cervical carcinoma and To assess the genomic similarity between PSCC and other squa- esophageal carcinoma cohorts, CDKN2A was more frequent in PSCC mous tumors, we evaluated the frequency of the 96 mutation types in than cervical carcinoma; DNAH6 in PSCC than HNSC, and CASP8 PSCC compared with the squamous TCGA cohorts. By performing was more frequent in PSCC than esophageal carcinoma and LUSC. cophenetic analysis, we estimated three different mutation patterns TP53, TTN, CSND3, RYR2, and KMT2D were significantly less fre- among the six squamous cohorts, which we named Sq-MP1, Sq-MP2, quent among PSCC samples. In addition, we compared the mutation and Sq-MP3. As described previously, these mutation patterns were frequency of DDR (BRCA1, BRCA2, ARID1A, ATR, CHEK2, PARP1, compared with COSMIC signatures, which indicated that Sq-MP1 best FANCA, PALB2, and RAD51) genes between PSCC and the squamous match [cosine similarity (CS) ¼ 0.755] was SBS2 (APOBEC cytidine cohorts. PSCC cohort exhibited a high number of patients with mutant deaminase – C>T), Sq-MP2 best match (CS ¼ 0.879) was SBS6 DDR genes (n ¼ 8, 23.5%), similarly to LUSC (n ¼ 116, 24.6%) and (defective DNA mismatch repair), and Sq-MP3 best match (CS ¼

OF8 Clin Cancer Res; 27(9) May 1, 2021 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Clinical Implications of WES in Penile Squamous Carcinoma

0.947) was SBS4 (exposure to tobacco smoking mutagens; Supple- for functional analysis to confirm this hypothesis. Also, a significant mentary Fig. S4). PSCC samples were primarily enriched by Sq-MP2 (n proportion of patients (23.5%) was found to have pathogenic or likely ¼ 23, 67.6%) followed by Sq-MP1 (n ¼ 8, 23.5%) and Sq-MP3 (n ¼ 3, pathogenic variants in DDR genes, which could indicate another 8%; Fig. 3C). Distribution of each signature among cohorts showed a potential clinical benefit from PARP inhibition alone or in combina- significant difference between PSCC and LUSC (P < 0.0001), bladder tion with immune checkpoint inhibition strategies (46, 47). These carcinoma (P ¼ 0.006), and cervical carcinoma (P < 0.0001) cohorts. findings could lay the foundation for precision trials using targeted However, no significant difference between PSCC and HNSC (P ¼ drugs in patients with chemotherapy refractory PSCC. 0.208) and esophageal carcinoma (P ¼ 0.387) was observed, suggesting This study builds on the prior molecular efforts in PSCC, specifically a similar mutation signature between these three cohorts. We further the work published by Ali and colleagues (16), Jacob and collea- compared the background of mutations among these cohorts by gues (13), and Feber and colleagues (14), and addresses some of these assessing the APOBEC mutation signature estimation, as described studies limitations. Our results differed as the sequencing approaches previously (Fig. 3D). PSCC samples differed significantly from bladder presented by Ali and colleagues and Jacob and colleagues were not carcinoma (P ¼ 0.0002) and cervical carcinoma (P < 0.0001) cohorts comparable with our methodologic approach. In this study, we that exhibited a high enrichment of APOBEC-related mutations. No employed WES, which covers most of the exonic regions in the human differences were observed when PSCC was compare with LUSC, genome providing coverage of more than 20,000 human genes. Ali and esophageal carcinoma, and HNSC cohorts. colleagues and Jacob and colleagues used a target sequencing tech- nology called “comprehensive genomic profiling.” Despite its name, this technology covers alterations in approximately 300 human genes, Discussion which represents only 1.5% of the genome coverage we had in our Our study provides several novel insights into PSCC biology and study. In this way, target sequencing technologies are not appropriate highlights the similarities with HNSC, taking advantage of WES to explore overall tumor mutation profile because only a selected analysis, in a rare cancer with limited therapeutic choices (7, 8, 10, 43). fraction of the existent genes is being represented. Beyond covering less The goal was to exploit this technology to gain potential insights into of the genome, some of the genes included in such target sequencing new therapeutic strategies in this rare and lethal cancer. To this extent, approaches are not even fully covered by the assay, underrepresenting while our sample size was small, it represents the largest PSCC cohort the mutation frequency of certain genes. reported using the WES approach. We have also analyzed the data in On the other hand, Feber and colleagues performed whole-exome the context of the HPV status and genotype (11, 15, 16). Another analysis, however, there are important technical differences between limitation to our study is that even though our sample is reflective of the studies to highlight (14). While the mean sequencing depth the stage at presentation, but only a limited number had advanced generated in our study was of 141, in Feber and colleagues’ study, stage and no other penile cancer histology beside squamous cell, which the mean sequencing depth was only 60. This difference in the limits the generalization of our conclusions. This highlights the amount of sequencing data generated can have an important impact on importance of ongoing international collaborative efforts aiming to the final mutation profile results. In their study, the mean mutation develop the PSCC tumor tissue and blood banking infrastructure that rate per patient was 30, which is much lower than we what found, with would allow the field to overcome this limitation. a mean of 125.7 mutations per patient. Even adjusting the mutation In summary, our systematic analyses of PSCCC genomics identified rate per Mb sequenced, their mutation rate, 1.78 (0.72–7.5) mutations that the NOTCH signaling deregulation pathway is implicated in more per Mb, was lower than what we found in our study, 1.96 (0.01–21.2). than half of PSCC samples showing an enrichment of Notch pathway The differences can also be highlighted at the gene level. The most (n ¼ 24, 70.6%) alterations. Others have shown enrichment of the frequently mutated genes in their analysis, FAT1 and TP53, affected Notch pathway at lower rates, this is related to the sequencing only four of the 27 sequenced samples, which represents only 14.8% of technique and analysis used that limited their investigation. This is their cases. On the other hand, FAT1 and TP53, which were among of interest for future clinical investigation, as recent work showed most commonly mutated genes in our cohort, affected 35% of our similar findings with oncogenic mutations found in 67% of human samples. These differences indicate that the mutation frequency in HNSC cases that converge onto the NOTCH signaling pathway. Thus, their study is probably underestimated. Another possible effect of the NOTCH inactivation is a hallmark of both PSCC and HNSC (44). In low sequencing depth in their study is the presence of false positive the era of targeted therapy, a rare cancer like PSCC, could be included findings. Two of their cases (7.4%) showed GPS1 (what they called with other SCCs umbrella clinical trials targeting a specific common CSN1) mutations. None of our PSCC cases showed such mutation, and targetable alteration. The above findings provide the potential ratio- the mutation frequency among the other TCGA squamous tumors was nale for the ongoing and future clinical trials in patients with pretty low or absent: bladder carcinoma ¼ 6.4% (3/47), cervical NOTCH1-mutated HNSC using PI3K/mTOR inhibitors, to potentially carcinoma ¼ 1.2% (3/241), esophageal carcinoma ¼ no mutations include patients with PSCC along with HNSC cohorts (44). It is known found, HNSC ¼ 0.2% (1/501), and LUSC ¼ 1.3% (6/471). Considering that the aberrant expression of the essential NOTCH coactivator of the its low frequency of mutations in this gene in squamous tumors, we Mastermind-like (MAML) family provides an alternative mechanism suspect that the true frequency of GPS1 mutations in PSCC is in the to activate NOTCH signaling in human cancers. Interestingly, the low single-digit range, and it will require a larger cohort of PSCC cases work by Necchi and colleagues identified that the MAML2 (HR, to be confident in the true mutation frequency. 10.411; P ¼ 0.003) was associated with poor OS in patients with penile Furthermore, by performing cophenetic analysis, we identified two cancer that received cisplatin chemotherapy (45). distinct mutation patterns among PSCC samples. The first (MP1), Our data also provide some evidence that PIK3CA and CDKN2A exhibited the highest cosine similarity to the SBS2 and SBS13 COSMIC could be driver mutations in PSCC given positional enrichment signatures, both associated with oncogenic activity of AID/APOBEC analysis indicating that mutations in PIK3CA and CDKN2A were family of cytidine deaminases and related to local hypermutation significantly enriched in specific protein loci. Future studies should phenomenon called kataegis (C>G – TC context). On the other hand, include serial sampling from patients with PSCC or precursors lesions the second (MP2), was more common and had the highest cosine

AACRJournals.org Clin Cancer Res; 27(9) May 1, 2021 OF9

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Chahoud et al.

similarity to SBS6 COSMIC signature, associated with defective DNA better characterization of these findings.Thisworkisafirst step mismatch repair system and microsatellite instability. Interestingly, toward providing the field with the rationale for new therapeutic MP1 enrichment was positively correlated with increased TMB strategies. (CC ¼ 0.71; P < 0.0001) and correlated with significantly worse survival of patients with PSCC in comparison with those in the MP2 Authors’ Disclosures subset. This study is the first to show the existence of a subset of PSCC J. Chahoud reported grants from ASCO CCF during the conduct of the study and (38%) with enrichment of APOBEC-related mutation signatures, other from Pfizer outside the submitted work. C.R. Pickering reported grants from – higher TMB, and worse OS in comparison with the non-APOBEC NIH outside the submitted work. C.A. Pettaway reported personal fees from Wolters enriched subset. These findings can provide the rationale for drug Kluwer outside the submitted work. No disclosures were reported by the other investigation, to assess whether patients with MP1 subset could authors. clinically benefit from immunotherapy with check point inhibitors, while those with the MP2 subset could benefit from PARP inhibitors. Authors’ Contributions Finally, considering the limited genomic understanding of PSCC, J. Chahoud: Conceptualization, data curation, formal analysis, investigation, we aimed to compare these tumors with other well-characterized writing–original draft, writing–review and editing. F.O. Gleber-Netto: Conceptual- squamous carcinomas using TCGA data. On our initial analysis, we ization, data curation, software, formal analysis, methodology, writing–original draft, identified that PSCC appears to be genomically distinct in ways writing–review and editing. B.Z. McCormick: Data curation, writing–review and from other squamous solid tumors, with a significantly lower TMB editing. P. Rao: Data curation, formal analysis, investigation, writing–review and – and having the highest enrichment of NOTCH1 mutations, but editing. X. Lu: Formal analysis, methodology, writing review and editing. M. Guo: Investigation, writing–review and editing. M.B. Morgan: Data curation, software, similar to HNSC. When we evaluated the 96 mutation types in formal analysis, writing–review and editing. R. Chu: Data curation, software, formal PSCC compared with the squamous TCGA cohorts, by performing analysis, writing–review and editing. M. Martinez-Ferrer: Software, visualization, cophenetic analysis and comparison with COSMIC signatures, writing–review and editing. A.K. Eterovic: Resources, software, writing–review PSCC appeared to have a mutation signature that was significantly and editing. C.R. Pickering: Data curation, software, formal analysis, supervision, different from LUSC, bladder carcinoma, and cervical carcinoma methodology, writing–original draft, writing–review and editing. C.A. Pettaway: cohorts, but similar to the mutation signature of HNSC and Conceptualization, data curation, software, formal analysis, supervision, funding acquisition, investigation, methodology, writing–original draft, writing–review esophageal carcinoma. On this note, it is of interest that the and editing. first-line chemotherapy (utilized in the InPACT trial) regimen for metastatic penile cancer using cisplatin, paclitaxel, and ifosfamide Acknowledgments has been shown to have objective responses in 50% of patients and fi This work was supported by funding from the University of Texas MD Anderson was initially chosen because of its ef cacy in HNSC (6, 48, 49). We Cancer Center Human Papillomavirus related cancers moonshot program and the further compared the APOBEC mutation signature estimation of Conquer Cancer Foundation (CCF). J. Chahoud was supported by the CCF American PSCC samples, which differed significantly from bladder carcinoma Cancer Society of Clinical Oncology young investigator award. and cervical carcinoma cohorts, but appeared similar to LUSC, esophageal carcinoma, and HNSC cohorts. These findings highlight The costs of publication of this article were defrayed in part by the payment of page that the genetic signature of PSCC is most similar to HNSC and, to a charges. This article must therefore be hereby marked advertisement in accordance lesser degree, to esophageal carcinoma. with 18 U.S.C. Section 1734 solely to indicate this fact. Future collaborative efforts examining larger cohorts, coupled with functional molecular analysis and taking benefit of recently Received October 18, 2020; revised December 8, 2020; accepted January 6, 2021; available preclinical models (50) are needed for both validation and published first January 13, 2021.

References 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;69: 9. Chahoud J, Pickering CR, Pettaway CA. Genetics and penile cancer: recent 7–34. developments and implications. Curr Opin Urol 2019;29:364–70. 2. Montes Cardona CE, Andres García-Perdomo H. Incidence of penile cancer 10. Aydin AM, Chahoud J, Adashek JJ, Azizi M, Magliocco A, Ross JS, et al. worldwide: systematic review and meta-analysis. Rev Panam Salud Publica 2017; Understanding genomics and the immune environment of penile cancer to 41:117. improve therapy. Nat Rev Urol 2020;17:555–70. 3. Alemany L, Cubilla A, Halec G, Kasamatsu E, Quiros B, Masferrer E, et al. Role of 11. McDaniel AS, Hovelson DH, Cani AK, Liu C-J, Zhai Y, Zhang Y, et al. Genomic human papillomavirus in penile carcinomas worldwide. Eur Urol 2016;69:953–61. profiling of penile squamous cell carcinoma reveals new opportunities for 4. Olesen TB, Sand FL, Rasmussen CL, Albieri V, Toft BG, Norrild B, et al. targeted therapy. Cancer Res 2015;75:5219–27. Prevalence of human papillomavirus DNA and p16INK4a in penile cancer and 12. Marchi FA, Martins DC, Barros-Filho MC, Kuasne H, Busso Lopes penile intraepithelial neoplasia: a systematic review and meta-analysis. AF,BrentaniH,etal.Multidimensional integrative analysis uncovers Lancet Oncol 2019;20:145–58. driver candidates and biomarkers in penile carcinoma. Sci Rep 2017;7: 5. Feber A, Arya M, de Winter P, Saqib M, Nigam R, Malone PR, et al. Epigenetics 1–11. markers of metastasis and HPV-induced tumorigenesis in penile cancer. 13. Jacob JM, Ferry EK, Gay LM, Elvin JA, Vergilio Jo-A, Ramkissoon S, et al. Clin Cancer Res 2015;21:1196–206. Comparative genomic profiling of refractory and metastatic penile and non- 6. Pagliaro LC, Williams DL, Daliani D, Williams MB, Osai W, Kincaid M, et al. penile cutaneous squamous cell carcinoma: implications for selection of systemic Neoadjuvant paclitaxel, ifosfamide, and cisplatin chemotherapy for metastatic therapy. J Urol 2018;201:541–8. penile cancer: a phase II study. J Clin Oncol 2010;28:3851–7. 14. Feber A, Worth DC, Chakravarthy A, de Winter P, Shah K, Arya M, et al. CSN1 7. Wang J, Pettaway CA, Pagliaro LC. Treatment for metastatic penile cancer after somatic mutations in penile squamous cell carcinoma. Cancer Res 2016;76: first-line chemotherapy failure: analysis of response and survival outcomes. 4720–7. Urology 2015;85:1104–10. 15. Busso-Lopes AF, Marchi FA, Kuasne H, Scapulatempo-Neto C, Trindade-Filho 8. Chahoud J, Tamil M, Necchi A. Second line salvage systemic therapy for JCS, de Jesus CMN, et al. Genomic profiling of human penile carcinoma predicts advanced penile cancer. Urol Oncol 2020;20:30366–5. worse prognosis and survival. Cancer Prev Res 2015;8:149–56.

OF10 Clin Cancer Res; 27(9) May 1, 2021 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Clinical Implications of WES in Penile Squamous Carcinoma

16. Ali SM, Pal SK, Wang K, Palma NA, Sanford E, Bailey M, et al. Comprehensive 34. Ellrott K, Bailey MH, Saksena G, Covington KR, Kandoth C, Stewart C, et al. genomic profiling of advanced penile carcinoma suggests a high frequency of Scalable open science approach for mutation calling of tumor exomes using clinically relevant genomic alterations. Oncologist 2016;21:33–9. multiple genomic pipelines. Cell Syst 2018;6:271–81. 17. Cancer Genome Atlas Network. Comprehensive genomic characterization of 35. Campbell JD, Yau C, Bowlby R, Pickering CR, Chen Z, Van Waes C. Genomic, head and neck squamous cell carcinomas. Nature 2015;517:576–82. pathway network, and immunologic features distinguishing squamous carcino- 18. Pettaway CA, Srigley JR, Brookland RK, Choyke PL, Ryan CJ, Humphrey PA, mas. Cell Rep. 2018;23:194–212. et al. Penis AJCC Cancer Staging Manual 2016:3–5. 36. Chakravarty D, Gao J, Phillips S, Kundra R, Zhang H, Wang J, et al. 19. Cubilla AL, Lloveras B, Alejo M, Clavero O, Chaux A, Kasamatsu E, et al. Value of OncoKB: a precision oncology knowledge base. JCO Precis Oncol 2017;1: p16INK4a in the pathology of invasive penile squamous cell carcinomas. Am J PO.17.00011. Surg Pathol 2011;35:253–61. 37. Sobhani Navid, D’Angelo Alberto, Pittacolo Matteo, Roviello Giandomenico, 20. Chaux A, Pfannl R, Lloveras B, Alejo M, Clavero O, Lezcano C, et al. Distinctive Miccoli Anna, Corona Silvia Paola, et al. Updates on the CDK4/6 inhibitory association of p16INK4a overexpression with penile intraepithelial neoplasia strategy and combinations in breast cancer. Cells 2019;8:321. depicting warty and/or basaloid features: a study of 141 cases evaluating a new 38. Andre F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. Alpelisib nomenclature. Am J Surg Pathol 2010;34:385–92. for PIK3CA -mutated, hormone receptor–positive advanced breast cancer. 21. Kerr DA, Sweeney B, Arpin RN, Ring M, Pitman MB, Wilbur DC, et al. N Engl J Med 2019;380:1929–40. Automated extraction of formalin-fixed, paraffin-embedded tissue for high- 39. Brown JS, O’Carrigan B, Jackson SP, Yap TA. Targeting DNA repair in cancer: risk human papillomavirus testing of head and neck squamous cell carci- beyond PARP inhibitors. Cancer Discov 2017;7:20–37. nomas using the Roche Cobas 4800 system. Arch Pathol Lab Med 2016;140: 40. Romero D. DDR signature to predict response to ICI. Nat Rev Clin Oncol 2018; 844–8. 15:346. 22. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler 41. Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kania BE, Moran MM, et al. transform. Bioinformatics 2009;25:1754–60. Alterations in DNA damage response and repair genes as potential marker of 23. DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. A clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers. J Clin framework for variation discovery and genotyping using next-generation DNA Oncol 2018;36:1685–94. sequencing data. Nat Genet 2011;43:491–8. 42. O’Connor MJ. Targeting the DNA damage response in cancer. Mol Cell 2015;60: 24. Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, et al. 547–60. Sensitive detection of somatic point mutations in impure and heterogeneous 43. Ahmed ME, Falasiri S, Hajiran A, Chahoud J, Spiess PE. The immune micro- cancer samples. Nat Biotechnol 2013;31:213–9. environment in penile cancer and rationale for immunotherapy. J Clin Med 25. The 1000 Genomes Project Consortium, Auton A, Brooks LD, Durbin RM, 2020;9:3334. Garrison EP, Kang HM, et al. A global reference for human genetic variation. 44.SambandamV,FrederickMJ,ShenLi,TongP,RaoX,PengS,etal.PDK1 Nature 2015;526:68–74. mediates NOTCH1-mutated head and neck squamous carcinoma vulner- 26. NHLBI GO Exome Sequencing Project (ESP). Exome variant server. Available ability to therapeutic PI3K/mTOR inhibition. Clin Cancer Res 2019;25: from: http://evs.gs.washington.edu/EVS/. 3329–40. 27. Karczewski KJ, Weisburd B, Thomas B, Solomonson M, Ruderfer DM, Kavanagh 45. Necchi A, Eigl BJ, Yang ES-H, Bae S, Chandrashekar D, Chen D, et al. Gene D, et al. The ExAC browser: displaying reference data information from over 60 expression profiling of advanced penile squamous cell carcinoma receiving 000 exomes. Nucleic Acids Res 2017;45:D840–5. cisplatin-based chemotherapy improves prognostication and identifies potential 28. Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and therapeutic targets. Eur Urol Focus 2018;4:733–6. comprehensive analysis of somatic variants in cancer. Genome Res 2018;28: 46. Cesaire M, Thariat J, Candeias SM, Stefan D, Saintigny Y, Chevalier F. Com- 1747–56. bining PARP inhibition, radiation, and immunotherapy: a possible strategy to 29. Tamborero D, NL-B AG-P. OncodriveCLUST: exploiting the positional clus- improve the treatment of cancer? Int J Mol Sci 2018;19;3793. tering of somatic mutations to identify cancer genes. Bioinformatics 2013;29: 47. F€arkkil€a A, Gulhan DC, Casado J, Jacobson CA, Nguyen H, Kochupurakkal B, 2238–44. et al. Immunogenomic profiling determines responses to combined PARP and 30. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, et al. PD-1 inhibition in ovarian cancer. Nat Commun 2020;11:1–13. Oncogenic signaling pathways in The Cancer Genome Atlas. Cell 2018;173: 48. Canter DJ, Nicholson S, Watkin N, Hall E, Pettaway C. The International Penile 321–37. Advanced Cancer Trial (InPACT): rationale and current status. Eur Urol Focus 31. Griffith M, Griffith OL, Coffman AC, Weible JV, McMichael JF, Spies NC, et al. 2019;5:706–9. DGIdb: mining the druggable genome. Nat Methods 2013;10:1209–10. 49. Shin DM, Glisson BS, Khuri FR, Ginsberg L, Papadimitrakopoulou V, Lee JJ, et al. 32. Roberts SA, Lawrence MS, Klimczak LJ, Grimm SA, Fargo D, Stojanov P, et al. An Phase II trial of paclitaxel, ifosfamide, and cisplatin in patients with recurrent APOBEC cytidine deaminase mutagenesis pattern is widespread in human head and neck squamous cell carcinoma. J Clin Oncol 1998;16:1325–30. cancers. Nat Genet 2013;45:970–6. 50. Huang T, Cheng Xi, Chahoud J, Sarhan A, Tamboli P, Rao P, et al. Effective 33. Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: the combinatorial immunotherapy for penile squamous cell carcinoma. catalogue of somatic mutations in cancer. Nucleic Acids Res 2019;47:D941–7. Nat Commun 2020;11.

AACRJournals.org Clin Cancer Res; 27(9) May 1, 2021 OF11

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst January 13, 2021; DOI: 10.1158/1078-0432.CCR-20-4004

Whole-exome Sequencing in Penile Squamous Cell Carcinoma Uncovers Novel Prognostic Categorization and Drug Targets Similar to Head and Neck Squamous Cell Carcinoma

Jad Chahoud, Frederico O. Gleber-Netto, Barrett Z. McCormick, et al.

Clin Cancer Res Published OnlineFirst January 13, 2021.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-20-4004

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2021/01/12/1078-0432.CCR-20-4004.DC1

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 Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2021/04/09/1078-0432.CCR-20-4004. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2021 American Association for Cancer Research.