Context.—

Human papillomavirus (HPV) is a well-known cause of squamous cell carcinomas of anogenital and oropharyngeal regions, where treatment strategies and prognosis depend on HPV status. The significance of HPV status in tumors arising along the urinary tract is not well established.

Objective.—

To provide detailed clinical, morphologic, immunohistochemical, and molecular analysis of HPV+ urinary tract carcinomas (UTCs).

Design.—

We identified and retrospectively examined 12 HPV+ UTCs, confirmed by high-risk HPV in situ hybridization.

Results.—

The HPV+ UTCs originated from the urethra (9) and urinary bladder (3); 5 of 12 (42%) presented with nodal metastasis. On morphology, HPV+ UTCs were predominantly basaloid; well-differentiated squamous areas were focally seen. Available immunohistochemistry (IHC) showed strong staining for p16 (11 of 11), p63 (12 of 12), cytokeratin (CK) 903 (11 of 11), and CK5/6 (11 of 11); variable staining for GATA3 (8 of 12) and CK7 (4 of 11); and rare uroplakin II staining (1 of 12). Molecular analysis revealed the most frequently altered genes: KMT2C (42%), PIK3CA (42%), and KMT2D (25%). In contrast to published conventional urothelial and squamous cell carcinoma molecular data, TERTp mutation was rare (8%), and no TP53 or CDKN2A aberrations were identified. During available follow-up (11 of 12; median, 39 months), 6 patients required treatment for recurrence; ultimately, 1 died of disease, 2 were alive with disease, and 8 had no evidence of disease. Finally, we provide 11 HPV squamous predominant UTCs for IHC and molecular comparisons; notably, a subset of HPV UTCs was positive for p16 IHC (27%), making p16 IHC a less-specific surrogate marker for HPV status at this site.

Conclusions.—

HPV+ UTCs show distinct clinical, morphologic, and molecular characteristics, suggesting important roles for HPV in UTC.

Human papillomavirus (HPV) is a nonenveloped, double-stranded DNA virus belonging to the Papillomaviridae family of viruses and is the most common sexually transmitted virus worldwide, estimated to affect more than 80% of women and men by age 45 years.1,2  There have been more than 150 HPV genotypes described, which are classified into low-risk and high-risk by ability to cause malignant transformation. High-risk HPV (hrHPV), most frequently the HPV16 and HPV18 types, are well-known causes for cervical, anogenital, and oropharyngeal cancers.3,4  Overall, 4.8% of the global cancer burden is attributed to HPV infection, although incidence varies substantially by geographic region and is estimated to be up to 15.5% in sub-Saharan Africa and India.5 

Since discovery of the existence of HPV in cervical squamous cell carcinoma (SCC) by Harald Zur Hausen in 1983, HPV-driven oncogenesis has been an active area of research, with numerous mechanisms described in recent reviews.6–8  The most well-known mechanism is via expression of HPV oncoproteins E6 and E7, which causes inactivation and degradation of p53 and retinoblastoma (pRB) tumor suppressor proteins, respectively, thereby causing resistance to cell death and uncontrolled cell proliferation.9–11  Inactivation of pRB causes increased expression of p16, which is widely used as a surrogate immunohistochemistry (IHC) marker for HPV detection.12,13 

HPV-associated SCCs of anogenital and oropharyngeal regions are biologically unique entities. They often show basaloid morphology and less keratinization than their HPV-negative counterparts.12  Clinical presentation, treatment strategies, and prognosis of SCC in these regions are dependent on HPV status.7,14  In oropharyngeal sites, while HPV-associated SCCs more frequently involve lymph nodes, they usually have a better treatment response, with higher radiosensitivity and improved outcome.7,15–18  The molecular alterations in HPV-associated SCC are also distinct, with absent or rare TP53 and CDKN2A alterations, which are commonly altered in HPV-negative SCC.19,20 

HPV presence has been reported among urinary tract carcinomas (UTCs) in prior studies, with varying detection rates ranging from 0% to 100%, largely attributed to differences in cohorts and detection methods.21–25  The significance of HPV status on the pathogenesis, treatment, and outcome of UTCs is not well understood. The Cancer Genome Atlas study on invasive urothelial bladder carcinomas revealed rare HPV transcripts (in 2.6% of cases) predominantly in tumors with squamous differentiation.26  Clinical and pathologic features of HPV-positive carcinomas in the urinary bladder have been sparsely described in the literature, most notably with suggested association to neurogenic bladder and repetitive catheterization.27  Similar to HPV-associated SCC of oropharyngeal and anogenital areas, the previously reported HPV-associated bladder carcinomas have shown a basaloid morphology and have aberrant p16 expression by IHC.27  A separate study examining a series of urethral carcinomas described similar basaloid features and aggressive behavior with propensity for lymph node and distant organ metastasis, and HPV association was identified in approximately 30%, though HPV-positive cases were not examined separately from HPV-negative cases.28  Detailed molecular analysis of HPV-associated UTCs has not been performed to date. Given the potential parallels to HPV-associated oropharyngeal carcinomas and possible clinical management and outcome implications, further understanding of HPV-associated UTCs is necessary.

In this study, we explore the role of hrHPV infection on UTC pathogenesis by performing detailed clinical, histopathologic, and molecular characterization of a series of HPV-positive (HPV+) UTCs.

Case Selection

Institutional review board approval was obtained for this retrospective study. We identified 12 urethral and urinary bladder cases in which HPV association was confirmed by in situ hybridization for hrHPV in our surgical case files. In brief, we reviewed the pathology reports of all penile, urinary bladder, ureter, and renal pelvis cases in which “HPV” was written in the final diagnosis or diagnosis comment from 2000–2020. Cases with a suspicion of direct extension from the anogenital tract, including penile skin, foreskin or glans mucosa, vulva, cervix, and anus, were excluded. Of the 15 candidate cases, hrHPV in situ hybridization (hrHPV-ISH) was already performed on the case or subsequent case from the same location and was positive in 9 (Supplemental Table 1, see the supplemental digital content containing 3 tables and 2 figures at https://meridian.allenpress.com/aplm in the January 2025 table of contents). For the remaining cases, we performed hrHPV-ISH as part of this study (see below), which resulted in 3 additional positive cases (total 12). The original diagnoses for these cases included urothelial carcinoma, carcinoma with urothelial and squamous features, and SCC (Supplemental Table 1).

For comparison, tumors with predominantly squamous features were preferentially selected owing to HPV+ carcinomas typically showing squamous features. We conducted another database search among all of the bladder and urethral cases accessioned between years 2000 and 2020, using the search term squamous written in the final diagnosis. This search identified 200 cases, of which 11 had molecular sequencing data already available. hrHPV-ISH was performed as part of this study and was negative in all 11 cases. These cases were originally diagnosed as SCC, carcinoma with extensive squamous differentiation, and urothelial carcinoma with extensive squamous differentiation (Supplemental Table 1). Relevant clinical history was obtained from the electronic medical record.

Morphology Review

All available slides were reviewed. Basaloid features and squamous differentiation were documented as percentage involvement of total tumor area for each case. Basaloid features were defined as nests and ribbons of hyperchromatic tumor cells with high nuclear-to-cytoplasmic ratio, frequent mitotic figures, and necrosis.29  Only moderate to well-differentiated areas of squamous differentiation were quantified and were defined by the presence of intercellular bridges, intracellular keratinization, or the formation of squamous pearls. Representative formalin-fixed paraffin-embedded (FFPE) tumor sections of either the primary resection, biopsy, or a recurrence, based on tissue availability, were selected to perform clinically validated IHC and comprehensive molecular testing.

Immunohistochemistry

Immunohistochemical staining for uroplakin II (ready to use [RTU]; BC21, Biocare Medical, Pacheco, California), GATA3 (RTU; L50-823, Ventana Medical Systems, Tucson, Arizona), cytokeratin (CK) 7 (1:100; OV-TL12/30, Dako, Carpinteria, California), CK5/6 (1:200; D5-16B4, Millipore, Billerica, Massachusetts), CK903 (RTU; 34BE12, Enzo Diagnostics, Farmingdale, New York), p63 (RTU; 4A4, Ventana Medical), p16 (RTU; E6H4, Ventana Medical), p53 (1:100; DO-7, Biocare Medical) was performed by using tissue microarrays containing triplicate 1.5-mL punches of tumor from FFPE tissue or on whole slide sections, if not already performed during clinical evaluation. Immunohistochemical staining for uroplakin II, GATA3, CK7, CK5/6, CK903, and p63 was scored as strong and diffuse (++), patchy and weak (+/−), and negative (−). Aberrant p16 staining was defined as blocklike strong nuclear staining.30  p53 wild-type pattern was defined as heterogeneous distribution of variable-intensity nuclear staining. Aberrant p53 expression included overexpression (diffuse strong) and null pattern, as previously described.31 

HPV In Situ Hybridization

For all cases, in situ hybridization for hrHPV E6/E7 mRNA was performed to confirm presence/absence of hrHPV, using the RNAscope 2.5 VS Probe-HPV-HR18 (Advanced Cell Diagnostics Inc, Hayward, California) according to the manufacturer’s instructions and 4-μm FFPE tissue sections. The HPV genotypes included in this assay were HPV 16, 18, 26, 31, 33, 35, 39, 45, 51, 52, 53, 56, 58, 59, 66, 68, 73, and 82. Positive staining was identified as punctate nuclear and/or cytoplasmic dotlike staining. The slides were classified as positive or negative.

Molecular Analysis

Genomic DNA was extracted from punch biopsies or macrodissected unstained sections, targeting tumor-only from FFPE tissue. Sequencing libraries were prepared from genomic DNA, and target enrichment was performed by hybrid capture using a custom oligonucleotide library (Roche Nimblegen, Madison, Wisconsin). Capture-based next-generation sequencing was performed by using a clinically validated DNA sequencing assay that targets the coding regions of 529 cancer-related genes and select introns from 47 genes with a total sequencing footprint of 2.9 Mb (gene list https://genomics.ucsf.edu/content/ucsf-500-cancer-gene-panel-test-ucsf500-uc500, version 3; UCSF Clinical Cancer Genomic Laboratory, San Francisco, California). Sequencing of captured libraries was performed on a NovaSeq 6000 (Illumina, San Diego, California) as previously described.32  Molecular analysis was performed blinded to the component being analyzed and the HPV status. Somatic single-nucleotide variants and indels were visualized and verified with the Integrative Genomics Viewer. Genome-wide copy number analysis based on on-target and off-target reads was performed by using CNVkit and Nexus Copy Number (Biodiscovery, Hawthorne, California). Molecular alterations were manually classified as pathogenic or likely pathogenic on the basis of information from the following databases: cBioPortal for Cancer Genomics, ClinVar, and PubMed. Only pathogenic or likely pathogenic molecular alterations are reported and included in the analysis. Tumor mutation burden was calculated from the number of mutations in the coding regions of the genes in the UCSF500 panel, counting single-nucleotide variants and small indels and excluding known germline variants. Microsatellite instability (MSI) was analyzed by MSIsensor v0.2. Samples with 30% or more MSI detection were deemed MSI-high; 20% to less than 30%, indeterminate; and less than 20%, microsatellite stable.

Statistics

Categorical and quantitative variables were evaluated by using 2-tailed Fisher exact test and 2-tailed Student t test, respectively. Results were considered statistically significant if P < .05.

Clinicopathologic Features of HPV+ UTCs

We identified 12 cases of HPV+ UTC: 9 primary resections (75%) and 3 transurethral resections/biopsies (25%), which were treated with primary chemotherapy and/or radiation therapy (Table 1). The cases came from 5 female and 7 male patients with an average age of 66 years at initial diagnosis/primary resection. The primary sites at presentation were penile urethra (7 [58%]), female urethra (2 [17%]), and urinary bladder (3 [25%]); one of the urinary bladder cases also involved the female urethra but was classified as urinary bladder, as the mass was centered in the bladder (case 12). Notably, nearly half the patients (5 of 12, 42%) had lymph node metastases at presentation, including 2 who presented with a bulky groin mass and small primary tumor (cases 5 and 6). Two cases had a history of long-standing instrumentation (1 for urethral stricture disease, case 7; and 1 for neurogenic bladder, case 9). Additionally, 1 patient had concomitant B-cell lymphoma in the same groin lymph node with metastatic HPV+ UTC at initial diagnosis, and the same chemotherapy regimen was used to treat both tumors (case 5).

Table 1.

Clinical Features of Human Papillomavirus–Positive Urinary Tract Carcinomas

Clinical Features of Human Papillomavirus–Positive Urinary Tract Carcinomas
Clinical Features of Human Papillomavirus–Positive Urinary Tract Carcinomas

Morphology

On hematoxylin-eosin–stained sections, the HPV+ tumors frequently and extensively showed characteristic basaloid morphology, which was present in all cases and comprised on average 70% (range, 5%–100%) of total tumor area (Figure 1, A and B; Supplemental Table 1). Moderate to well-differentiated squamous areas were seen focally in 8 cases (67%), with total area of squamous differentiation in each tumor averaging 8% (range, 0%–30%) (Figure 1, C). The remaining tumor areas were considered “poorly differentiated,” characterized by sheets and nests of cells with high-grade nuclei and moderate amounts of eosinophilic cytoplasm but without definite keratinization, making it challenging to classify as either urothelial or squamous (Figure 1, D). Two tumors showed noninvasive flat lesions, mimicking conventional urothelial carcinoma in situ (Figure 2, A), and 8 cases showed a noninvasive papillary component, mimicking conventional papillary urothelial carcinoma (Figure 2, B). Noninvasive lesions were also hrHPV positive by ISH (Figure 2, C and D).

Figure 1.

Morphology of HPV+ urinary tract carcinoma. A, Basaloid morphology with nests and ribbons of cells with high nuclear-to-cytoplasmic ratio and frequent mitotic figures. B, Extensive necrosis was common. C, Moderate to well-differentiated squamous areas were seen focally. D, Poorly differentiated morphology with sheets and nests of cells with high-grade nuclei and a moderate amount of eosinophilic cytoplasm without keratinization (hematoxylin-eosin, original magnifications ×80 [A], ×40 [B], and ×100 [C and D]). Abbreviation: HPV, human papillomavirus.

Figure 1.

Morphology of HPV+ urinary tract carcinoma. A, Basaloid morphology with nests and ribbons of cells with high nuclear-to-cytoplasmic ratio and frequent mitotic figures. B, Extensive necrosis was common. C, Moderate to well-differentiated squamous areas were seen focally. D, Poorly differentiated morphology with sheets and nests of cells with high-grade nuclei and a moderate amount of eosinophilic cytoplasm without keratinization (hematoxylin-eosin, original magnifications ×80 [A], ×40 [B], and ×100 [C and D]). Abbreviation: HPV, human papillomavirus.

Close modal
Figure 2.

Noninvasive HPV+ lesions. A and C, Flat lesions mimicking urothelial carcinoma in situ. B and D, Papillary lesions mimicking papillary urothelial carcinoma. High-risk HPV-ISH is positive in noninvasive lesions (hematoxylin-eosin, original magnifications ×50 [A] and ×20 [B]; original magnifications ×50 [C] and ×20 [D]). Abbreviations: HPV, human papillomavirus; ISH, in situ hybridization.

Figure 2.

Noninvasive HPV+ lesions. A and C, Flat lesions mimicking urothelial carcinoma in situ. B and D, Papillary lesions mimicking papillary urothelial carcinoma. High-risk HPV-ISH is positive in noninvasive lesions (hematoxylin-eosin, original magnifications ×50 [A] and ×20 [B]; original magnifications ×50 [C] and ×20 [D]). Abbreviations: HPV, human papillomavirus; ISH, in situ hybridization.

Close modal

Immunohistochemical Features

We evaluated a panel of IHC markers commonly used for squamous and urothelial differentiation (Table 2): uroplakin II, CK7, GATA3, p63, CK903, and CK5/6. p63, CK903, and CK5/6 were strongly expressed in all cases with stains available. GATA3 was weakly expressed in 8 of 12 (67%) and CK7 was strongly expressed in 4 of 11 (36%) cases. Uroplakin II was weak and rarely seen, appearing in only 1 case (8%).

Table 2.

Immunohistochemical Profile of HPV+ UTCs

Immunohistochemical Profile of HPV+ UTCs
Immunohistochemical Profile of HPV+ UTCs

Molecular Findings

The most frequent pathogenic variants in HPV+ UTC were seen in KMT2C (42%), PIK3CA (42%), and KMT2D (25%), followed by TGFBR2 (17%) genes (Figure 3). One tumor (8%) showed a TERT promoter mutation (TERTp). None of the tumors in the HPV+ group had any alteration in TP53 or CDKN2A genes. Copy number analysis showed 1 tumor (8%) with deep deletion in RB1 gene, 1 (8%) with MYCL, and 1 (8%) with EGFR focal amplifications. Tumor mutational burden was variable, averaging 11.7 mutations/megabase (range, 3.4–23.4). All HPV+ tumors were microsatellite stable.

Figure 3.

Alterations identified on UCSF500 Next-Generation Sequencing (San Francisco, California) testing of HPV+ urinary tract carcinoma. Abbreviation: HPV, human papillomavirus.

Figure 3.

Alterations identified on UCSF500 Next-Generation Sequencing (San Francisco, California) testing of HPV+ urinary tract carcinoma. Abbreviation: HPV, human papillomavirus.

Close modal

Follow-up

Follow-up data were available for 11 patients (Table 1). During follow-up (median, 39 months; range, 9–115 months), 6 of 11 (55%) required treatment for recurrence. Ultimately, 8 patients (73%) had no evidence of disease, 2 (18%) were alive with disease, and 1 patient (9%) died of disease at the end of the follow-up period (Table 1).

Comparison to Squamous Predominant HPV UTCs

We evaluated 11 squamous predominant HPV UTCs to identify contrasting features that might aid in the detection of HPV+ UTCs. Detailed clinical, morphologic, immunohistochemical, and molecular features of these are presented in Supplemental Tables 1 through 3 and Supplemental Figures 1 and 2.

Notably, HPV+ UTCs showed more characteristic basaloid features than HPV squamous predominant UTCs (70% versus 15% of total tumor area, P < .001) and significantly fewer moderate to well-differentiated squamous areas (8% versus 67% of total tumor area, P < .001). On the other hand, HPV UTCs showed significant immunohistochemical overlap with the HPV+ UTCs: diffuse positive CK5/6, CK903, and p63 staining was seen in all the cases. GATA3 and CK7 was variably expressed in 7 (64%) and 8 (73%), respectively. Uroplakin II expression was the only marker to show significant difference, with 54% positivity in HPV UTCs and 8% positivity in HPV+ UTCs (P = .02).

Finally, in contrast to the HPV+ UTCs, our HPV UTCs showed mutational profiles similar to that more commonly reported in urothelial and squamous carcinomas of the lower urinary tract, with most frequently altered genes including TERTp (54%), TP53 (45%), PIK3CA (45%), CDKN2A (36%), RB1 (27%), KMT2D (27%), and KDM6A (27%) (Supplemental Figure 2).26,33,34  The degree of tumor mutational burden was similar to that of the HPV+ group (average, 13.4 mt/bp; range, 6.9–27.5 mt/bp; P = .56). Like the HPV+ group, all HPV cases were microsatellite stable.

Utilization of p16 and p53 Immunohistochemistry for Detection of HPV+ UTCs

Since p53, and indirectly, p16 proteins are the main targets of HPV oncoproteins E6 and E7, IHC was performed to determine whether their expression is altered in HPV+ UTCs and whether they can be useful tools for diagnosis. All the HPV+ tumors showed aberrant, blocklike p16 expression by IHC (Figure 4, A). Three HPV tumors (27%) also showed aberrant p16 staining (Figure 4, B), all of which on molecular analysis had a loss-of-function mutation in the RB1 gene (Figure 4, C). Overall, the sensitivity and the specificity of p16 IHC were 100% and 73%, respectively, for detection of HPV+ UTCs.

Figure 4.

p16 and p53 IHC staining of HPV+ and HPV urinary tract carcinomas (UTCs). A and B, Blocklike p16 positivity was seen in all HPV+ tumors (A) and in 27% of HPV tumors (B). C, Correlation of p16 and p53 IHC with molecular alterations in RB1 and TP53 in UTCs. D and E, Aberrant p53 staining was seen in 64% of HPV cases in the form of overexpression (D) and null pattern (E) (original magnification ×200 [A through D]). Abbreviations: HPV, human papillomavirus; IHC, immunohistochemical.

Figure 4.

p16 and p53 IHC staining of HPV+ and HPV urinary tract carcinomas (UTCs). A and B, Blocklike p16 positivity was seen in all HPV+ tumors (A) and in 27% of HPV tumors (B). C, Correlation of p16 and p53 IHC with molecular alterations in RB1 and TP53 in UTCs. D and E, Aberrant p53 staining was seen in 64% of HPV cases in the form of overexpression (D) and null pattern (E) (original magnification ×200 [A through D]). Abbreviations: HPV, human papillomavirus; IHC, immunohistochemical.

Close modal

All HPV+ tumors had a wild-type p53 expression pattern, consistent with their molecular profile showing lack of TP53 mutations. Seven HPV tumors (64%) showed aberrant p53 staining: 4 of 7 as overexpression (Figure 4, D) and 3 of 7 as the null pattern (Figure 4, E). All 4 cases with overexpression had a corresponding missense mutation in the TP53 gene on molecular analysis, whereas 1 of 3 cases with the null pattern contained a loss-of-function mutation in TP53 gene (Figure 4, C). The sensitivity and the specificity of p53 IHC for detection of TP53 mutation were 100% and 88%, respectively.

HPV-associated UTCs are uncommon, with limited studies to date. In this study, we found that HPV+ UTCs show clinical, morphologic, and molecular characteristics that are similar to what has been reported in HPV+ SCCs of other sites and are also distinct from their HPV UTC counterparts. At presentation, it is well known that HPV+ oropharyngeal SCCs frequently present with lymph node metastases in which the primary tumor can be small and hardly detectable by clinical examination alone.35  Indeed, 2 of our cases presented with bulky lymphadenopathy with a small primary site in the urethra.

On morphology, it is also well known that HPV-associated SCCs of varying anatomic locations commonly share characteristic basaloid morphology.12,36  In our series of 12 HPV+ UTCs, all showed similar overtly basaloid components (average 70% of total tumor area), albeit in 3 cases, the basaloid component was only focal (5%–10%). Notably, the remaining tumor areas were typically “poorly differentiated,” with areas of more obvious squamous differentiation only focal to absent (Supplemental Table 1). Review of the morphology for this study highlighted that these poorly differentiated areas could be interpreted as poorly differentiated SCC or poorly differentiated urothelial carcinoma. Likewise, the basaloid components of an HPV+ UTC can also mimic conventional high-grade urothelial carcinoma, particularly if there is papillary configuration of the surface tumor, which can resemble an in situ papillary urothelial carcinoma component. Indeed, a subset of our cases was initially called urothelial carcinoma by unsuspecting pathologists. In our own clinical practice, we would recommend testing (1) in any urethral tumor (given that about 30% of urethral carcinomas are HPV+ in the literature);28  (2) in any bulky urinary bladder tumor in a female patient to also exclude possibility of gynecologic tract origin; and (3) in the presence of basaloid appearance with focal or absent squamous features. Typically, we would sign these cases out as “HPV+ carcinomas with squamous features” with a descriptive comment to ensure origin from other sites has been excluded. While they are morphologically, immunohistochemically, and molecularly similar to HPV+ SCC in other sites, given the unique location in the urothelial tract in which the background epithelium is urothelial rather than squamous, and the morphologic overlap between urothelial carcinomas with squamous differentiation and SCCs, we defer official naming to a consensus of urologic pathologists. Finally, while GATA3, p63, CK903, CK5/6, CK7, and uroplakin II may be used to support a urothelial/squamous origin of a poorly differentiated carcinoma, they would not be helpful in distinguishing HPV+ from HPV cases.

HPV+ UTCs and HPV UTCs showed distinct molecular profiles. While there were some commonly overlapping, recurrently altered genes including PIK3CA and KMT2D, the HPV+ tumors rarely showed TERTp, TP53, or CDKN2A alterations, which were more common in HPV UTCs. These findings in our HPV UTCs are similar to what has been reported for urothelial and non–HPV-associated SCCs of the urinary bladder and in other sites.20,26  The absence or rarity of alterations in CDKN2A, TP53, and TERTp in HPV+ UTCs relative to HPV UTCs is likely due to HPV pathogenesis and the impact of the E6 and E7 oncoproteins.

First, the HPV E7 oncoprotein binds to the phosphorylated form of pRb and accelerates its degradation, which causes the release of transcription factor E2F and cell cycle progression.37  p16 is a product of CDKN2A, which functions as a tumor suppressor protein, and CDKN2A is commonly mutated in cancers including urothelial carcinomas.26  p16 inhibits the cyclin-dependent kinases CDK4 and CDK6, thereby preventing phosphorylation of pRb and progression to S phase of the cell cycle. When HPV E7 oncoprotein causes degradation of pRb and progression of cell cycle, this in turn increases p16 expression as a feedback mechanism.37  Increased p16 expression is easily detected on IHC and is often used as a surrogate marker for HPV infection. However, expression of p16 is not only dependent on the function of HPV E7 protein, as mutations in RB1 gene can also have a similar effect as E7 protein, with decreased pRB expression and increased p16 expression. Indeed, in 27% of our HPV cases, p16 expression was found to be aberrant and all had an associated loss-of-function mutation in the RB1 gene. Inactivating mutations and deep deletions in the RB1 gene are reported in 10% and 15% of urinary bladder urothelial carcinomas, respectively.26  Therefore, use of p16 IHC should be limited to screening for HPV and needs to be followed up with hrHPV in situ hybridization. RB1 loss was also seen in one of the HPV+ UTCs in our cases, which is surprising because RB1 loss would seem redundant in HPV+ tumors owing to the function of HPV E7 protein on pRB protein; however, similar findings have also been reported in HPV+ head and neck SCC.20  In our HPV+ case with RB1 loss, the loss was due to deep deletion, whereas in the HPV cases, the RB1 loss was due to point mutations. HPV DNA integration into the host genome is frequently associated with chromosomal structural changes including deletions.38,39  It would be interesting to learn if the deep deletion in RB1 gene is related to HPV insertion into the genome, though this was not possible in our assay.

Second, the HPV E6 oncoprotein induces degradation of the tumor suppressor protein p53, leading to resistance to cell death.40  IHC for p53 was variable in all our HPV+ cases, suggesting that not all p53 is degraded. Aberrant p53 expression was seen in 4 HPV cases as overexpression pattern, all of which had associated missense mutations in TP53 gene. The HPV-associated carcinomas of anogenital and oropharyngeal regions rarely have TP53 mutations, likely because the p53 function is already altered by HPV oncoprotein E6. Similarly, no TP53 mutations were seen in our cohort of HPV+ UTCs.

Third, TERTp mutations are one of the most common molecular alterations in urothelial and squamous cell carcinomas of the urinary tract.26,34  A similar finding to ours was previously reported for penile and head and neck SCC: TERTp mutations were frequent in non–HPV-associated SCC and significantly less frequent in the HPV-associated tumors.41,42  Interestingly, one of the many functions of the HPV E6 oncoproteins is to promote telomerase activity by multiple mechanisms involving the regulation of transcriptional, epigenetic, and posttranscriptional processes of the TERT gene.43,44  Furthermore, HPV DNA has been shown to integrate near the TERT locus, with viral enhancers activating TERTp in cervical cancer.45,46  Therefore, we hypothesize that HPV in our HPV+ UTC cases may increase telomerase activation via multiple mechanisms, resulting in cellular immortalization without the need for TERTp mutations. TERT expression studies would be necessary to confirm this, which are beyond the scope of this study.

Limitations to our study include insufficient number of cases and an insufficiently robust control group required to determine the effect of HPV on clinical outcomes in HPV+ UTCs. In the oropharynx, HPV+ oropharyngeal SCC usually presents at a lower pathologic T stage though with more common lymph node involvement.47,48  Treatment response and outcomes are better in HPV+ oropharyngeal SCC, and studies are underway to determine if less aggressive treatment strategies can be applied for these patients.15,17  Regardless of their mutational burden, HPV-associated cancers are also more immunogenic owing to expression of viral antigens and therefore good candidates for immunotherapy.16  Unlike in oropharyngeal and anogenital sites where HPV+ SCCs are the most common malignancy, HPV+ UTC is rare, making comparative studies difficult. The clinical features and the outcome of the HPV+ UTCs are not clear in the current literature.49–52  In our study, the HPV+ cases had more frequent lymph node involvement at presentation and better apparent disease outcomes than our HPV cases; however, it would be presumptuous to draw this comparison owing to differences in primary location of our HPV+ and HPV UTCs, different treatments received, and also the potentially heterogeneous nature of the HPV group due to the inability to distinguish with certainty a pure SCC from a urothelial carcinoma with extensive squamous differentiation in which the conventional urothelial component was not sampled. Additionally, there is significant selection bias in the HPV UTC cases to worse outcomes, as sequencing was performed at the request of treating oncologists in most cases. Nevertheless, molecularly, conventional urothelial carcinoma and SCCs of the urothelial tract share common alterations in TP53, CDKN2A, and TERT genes in the literature26,34,53  and in our HPV UTC cohort, which were rarely seen in our HPV+ UTCs. The parallels seen in our study to the genetics of HPV-associated tumors in oropharyngeal and anogenital sites further support the molecular findings presented here.20,41  Finally, the molecular profile of urethral tumors has been largely unknown before this study and therefore our study provides new molecular data on these cases.

In conclusion, HPV+ UTCs have a morphologic, immunohistochemical, and molecular presentation similar to HPV+ SCCs of other sites, though they may be difficult to distinguish from conventional urothelial carcinoma owing to overlapping morphologic and immunohistochemical features. Given their rarity in the urothelial tract, more studies are needed to better define these tumors’ behavior and treatment strategies.

The authors would like to thank Jolene Guerin for technical support in construction of the tissue microarrays and Tom Rocereto for support in retrieval of slides and tissue blocks.

1.
Kombe Kombe
 
AJ,
Li
 
B,
Zahid
 
A,
et al.
Epidemiology and burden of human papillomavirus and related diseases, molecular pathogenesis, and vaccine evaluation
.
Front Public Health
.
2020
;
8
:
552028
.
2.
Chesson
 
HW,
Dunne
 
EF,
Hariri
 
S,
Markowitz
 
LE.
The estimated lifetime probability of acquiring human papillomavirus in the United States
.
Sex Transm Dis
.
2014
;
41
(
11
):
660
664
.
3.
Serrano
 
B,
Brotons
 
M,
Bosch
 
FX,
Bruni
 
L.
Epidemiology and burden of HPV-related disease
.
Best Pract Res Clin Obstet Gynaecol
.
2018
;
47
:
14
26
.
4.
Mahal
 
BA,
Catalano
 
PJ,
Haddad
 
RI,
et al.
Incidence and demographic burden of HPV-associated oropharyngeal head and neck cancers in the United States
.
Cancer Epidemiol Biomarkers Prev
.
2019
;
28
(
10
):
1660
1667
.
5.
Forman
 
D,
de Martel
 
C,
Lacey
 
CJ,
et al.
Global burden of human papillomavirus and related diseases
.
Vaccine
.
2012
;
30
(
suppl 5
):
F12
F23
.
6.
Roden
 
RBS,
Stern
 
PL.
Opportunities and challenges for human papillomavirus vaccination in cancer
.
Nat Rev Cancer
.
2018
;
18
(
4
):
240
254
.
7.
Lechner
 
M,
Liu
 
J,
Masterson
 
L,
Fenton
 
TR.
HPV-associated oropharyngeal cancer: epidemiology, molecular biology and clinical management
.
Nat Rev Clin Oncol
.
2022
;
19
(
5
):
306
327
.
8.
Szymonowicz
 
KA,
Chen
 
J.
Biological and clinical aspects of HPV-related cancers
.
Cancer Biol Med
.
2020
;
17
(
4
):
864
878
.
9.
Yugawa
 
T,
Kiyono
 
T.
Molecular mechanisms of cervical carcinogenesis by high-risk human papillomaviruses: novel functions of E6 and E7 oncoproteins
.
Rev Med Virol
.
2009
;
19
(
2
):
97
113
.
10.
Moody
 
CA,
Laimins
 
LA.
Human papillomavirus oncoproteins: pathways to transformation
.
Nat Rev Cancer
.
2010
;
10
(
8
):
550
560
.
11.
Munger
 
K,
Werness
 
BA,
Dyson
 
N,
Phelps
 
WC,
Harlow
 
E,
Howley
 
PM.
Complex formation of human papillomavirus E7 proteins with the retinoblastoma tumor suppressor gene product
.
EMBO J
.
1989
;
8
(
13
):
4099
4105
.
12.
Gondim
 
DD,
Haynes
 
W,
Wang
 
X,
Chernock
 
RD,
El-Mofty
 
SK,
Lewis
 
JS
.
Histologic typing in oropharyngeal squamous cell carcinoma: a 4-year prospective practice study with p16 and high-risk HPV mRNA testing correlation
.
Am J Surg Pathol
.
2016
;
40
(
8
):
1117
11124
.
13.
Rabban
 
JT,
Soslow
 
R,
Zaloudek
 
C.
Immunohistology of the female genital tract. In:
Dabbs
 
DJ
, ed.
Diagnostic Immunohistochemistry
. 3rd ed.
Philadelphia, PA: W.B. Saunders
;
2010
:
690
-
762
.
14.
Symer
 
MM,
Yeo
 
HL.
Recent advances in the management of anal cancer
.
F1000Res
.
2018
;
7
:
F1000 Faculty Rev-1572
.
15.
Golusinski
 
P,
Corry
 
J,
Poorten
 
VV,
et al.
De-escalation studies in HPV-positive oropharyngeal cancer: how should we proceed
?
Oral Oncol
.
2021
;
123
:
105620
.
16.
Shamseddine
 
AA,
Burman
 
B,
Lee
 
NY,
Zamarin
 
D,
Riaz
 
N.
Tumor immunity and immunotherapy for HPV-related cancers
.
Cancer Discov
.
2021
;
11
(
8
):
1896
1912
.
17.
Chera
 
BS,
Amdur
 
RJ,
Green
 
R,
et al.
Phase II trial of de-intensified chemoradiotherapy for human papillomavirus-associated oropharyngeal squamous cell carcinoma
.
J Clin Oncol
.
2019
;
37
(
29
):
2661
2669
.
18.
Liu
 
C,
Mann
 
D,
Sinha
 
UK,
Kokot
 
NC.
The molecular mechanisms of increased radiosensitivity of HPV-positive oropharyngeal squamous cell carcinoma (OPSCC): an extensive review
.
J Otolaryngol Head Neck Surg
.
2018
;
47
(
1
):
59
.
19.
Rusan
 
M,
Li
 
YY,
Hammerman
 
PS.
Genomic landscape of human papillomavirus-associated cancers
.
Clin Cancer Res
.
2015
;
21
(
9
):
2009
2019
.
20.
Cancer Genome Atlas Network
.
Comprehensive genomic characterization of head and neck squamous cell carcinomas
.
Nature
.
2015
;
517
(
7536
):
576
582
.
21.
Shigehara
 
K,
Sasagawa
 
T,
Kawaguchi
 
S,
et al.
Etiologic role of human papillomavirus infection in bladder carcinoma
.
Cancer
.
2011
;
117
(
10
):
2067
2076
.
22.
Jorgensen
 
KR,
Jensen
 
JB.
Human papillomavirus and urinary bladder cancer revisited
.
APMIS
.
2020
;
128
(
2
):
72
79
.
23.
Kim
 
SH,
Joung
 
JY,
Chung
 
J,
Park
 
WS,
Lee
 
KH,
Seo
 
HK.
Detection of human papillomavirus infection and p16 immunohistochemistry expression in bladder cancer with squamous differentiation
.
PLoS One
.
2014
;
9
(
3
):
e93525
.
24.
Lu
 
QL,
Lalani el
 
N,
Abel
 
P.
Human papillomavirus 16 and 18 infection is absent in urinary bladder carcinomas
.
Eur Urol
.
1997
;
31
(
4
):
428
432
.
25.
Khatami
 
A,
Salavatiha
 
Z,
Razizadeh
 
MH.
Bladder cancer and human papillomavirus association: a systematic review and meta-analysis
.
Infect Agent Cancer
.
2022
;
17
(
1
):
3
.
26.
Robertson
 
AG,
Kim
 
J,
Al-Ahmadie
 
H,
et al.
Comprehensive molecular characterization of muscle-invasive bladder cancer
.
Cell
.
2017
;
171
(
3
):
540
556 e25
.
27.
Blochin
 
EB,
Park
 
KJ,
Tickoo
 
SK,
Reuter
 
VE,
Al-Ahmadie
 
H.
Urothelial carcinoma with prominent squamous differentiation in the setting of neurogenic bladder: role of human papillomavirus infection
.
Mod Pathol
.
2012
;
25
(
11
):
1534
1542
.
28.
Zhang
 
M,
Adeniran
 
AJ,
Vikram
 
R,
et al.
Carcinoma of the urethra
.
Hum Pathol
.
2018
;
72
:
35
44
.
29.
Wain
 
SL,
Kier
 
R,
Vollmer
 
RT,
Bossen
 
EH.
Basaloid-squamous carcinoma of the tongue, hypopharynx, and larynx: report of 10 cases
.
Hum Pathol
.
1986
;
17
(
11
):
1158
1166
.
30.
Sano
 
T,
Oyama
 
T,
Kashiwabara
 
K,
Fukuda
 
T,
Nakajima
 
T.
Expression status of p16 protein is associated with human papillomavirus oncogenic potential in cervical and genital lesions
.
Am J Pathol
.
1998
;
153
(
6
):
1741
1748
.
31.
Hodgson
 
A,
Xu
 
B,
Downes
 
MR.
p53 immunohistochemistry in high-grade urothelial carcinoma of the bladder is prognostically significant
.
Histopathology
.
2017
;
71
(
2
):
296
304
.
32.
Chan
 
E,
Garg
 
K,
Stohr
 
BA.
Integrated immunohistochemical and molecular analysis improves diagnosis of high-grade carcinoma in the urinary bladder of patients with prior radiation therapy for prostate cancer
.
Mod Pathol
.
2020
;
33
(
9
):
1802
1810
.
33.
Warrick
 
JI,
Hu
 
W,
Yamashita
 
H,
et al.
FOXA1 repression drives lineage plasticity and immune heterogeneity in bladder cancers with squamous differentiation
.
Nature Commun
.
2022
;
13
(
1
):
6575
.
34.
Hurst
 
CD,
Cheng
 
G,
Platt
 
FM,
et al.
Molecular profile of pure squamous cell carcinoma of the bladder identifies major roles for OSMR and YAP signalling
.
J Pathol Clin Res
.
2022
;
8
(
3
):
279
293
.
35.
Carpen
 
T,
Sjoblom
 
A,
Lundberg
 
M,
et al.
Presenting symptoms and clinical findings in HPV-positive and HPV-negative oropharyngeal cancer patients
.
Acta Otolaryngol
.
2018
;
138
(
5
):
513
518
.
36.
Eich
 
ML,
Del Carmen Rodriguez Pena
 
M,
Schwartz
 
L,
et al.
Morphology, p16, HPV, and outcomes in squamous cell carcinoma of the penis: a multi-institutional study
.
Hum Pathol
.
2020
;
96
:
79
86
.
37.
Khleif
 
SN,
DeGregori
 
J,
Yee
 
CL,
et al.
Inhibition of cyclin D-CDK4/CDK6 activity is associated with an E2F-mediated induction of cyclin kinase inhibitor activity
.
Proc Natl Acad Sci U S A
.
1996
;
93
(
9
):
4350
4354
.
38.
Gao
 
G,
Wang
 
J,
Kasperbauer
 
JL,
et al.
Whole genome sequencing reveals complexity in both HPV sequences present and HPV integrations in HPV-positive oropharyngeal squamous cell carcinomas
.
BMC Cancer
.
2019
;
19
(
1
):
352
.
39.
Nkili-Meyong
 
AA,
Moussavou-Boundzanga
 
P,
Labouba
 
I,
et al.
Genome-wide profiling of human papillomavirus DNA integration in liquid-based cytology specimens from a Gabonese female population using HPV capture technology
.
Sci Rep
.
2019
;
9
(
1
):
1504
.
40.
Werness
 
BA,
Levine
 
AJ,
Howley
 
PM.
Association of human papillomavirus types 16 and 18 E6 proteins with p53
.
Science
.
1990
;
248
(
4951
):
76
79
.
41.
Kim
 
SK,
Kim
 
JH,
Han
 
JH,
et al.
TERT promoter mutations in penile squamous cell carcinoma: high frequency in non-HPV-related type and association with favorable clinicopathologic features
.
J Cancer Res Clin Oncol
.
2021
;
147
(
4
):
1125
1135
.
42.
Morris
 
LGT,
Chandramohan
 
R,
West
 
L,
et al.
The molecular landscape of recurrent and metastatic head and neck cancers: insights from a precision oncology sequencing platform
.
JAMA Oncol
.
2017
;
3
(
2
):
244
255
.
43.
Tornesello
 
ML,
Cerasuolo
 
A,
Starita
 
N,
et al.
The molecular interplay between human oncoviruses and telomerase in cancer development
.
Cancers (Basel)
.
2022
;
14
(
21
):
5257
.
44.
Porter
 
VL,
Marra
 
MA.
The drivers, mechanisms, and consequences of genome instability in HPV-driven cancers
.
Cancers (Basel)
.
2022
;
14
(
19
):
4623
.
45.
McMurray
 
HR,
McCance
 
DJ.
Human papillomavirus type 16 E6 activates TERT gene transcription through induction of c-Myc and release of USF-mediated repression
.
J Virol
.
2003
;
77
(
18
):
9852
9861
.
46.
Ferber
 
MJ,
Montoya
 
DP,
Yu
 
C,
et al.
Integrations of the hepatitis B virus (HBV) and human papillomavirus (HPV) into the human telomerase reverse transcriptase (hTERT) gene in liver and cervical cancers
.
Oncogene
.
2003
;
22
(
24
):
3813
3820
.
47.
Bauwens
 
L,
Baltres
 
A,
Fiani
 
DJ,
et al.
Prevalence and distribution of cervical lymph node metastases in HPV-positive and HPV-negative oropharyngeal squamous cell carcinoma
.
Radiother Oncol
.
2021
;
157
:
122
129
.
48.
Lowy
 
DR,
Munger
 
K.
Prognostic implications of HPV in oropharyngeal cancer
.
N Engl J Med
.
2010
;
363
(
1
):
82
84
.
49.
Ohadian Moghadam
 
S,
Mansori
 
K,
Nowroozi
 
MR,
Afshar
 
D,
Abbasi
 
B,
Nowroozi
 
A.
Association of human papilloma virus (HPV) infection with oncological outcomes in urothelial bladder cancer
.
Infect Agent Cancer
.
2020
;
15
:
52
.
50.
Sarier
 
M,
Sepin
 
N,
Keles
 
Y,
et al.
Is there any association between urothelial carcinoma of the bladder and human papillomavirus: a case-control study
.
Urol Int
.
2020
;
104
(
1-2
):
81
86
.
51.
Cai
 
T,
Mazzoli
 
S,
Meacci
 
F,
et al.
Human papillomavirus and non-muscle invasive urothelial bladder cancer: potential relationship from a pilot study
.
Oncol Rep
.
2011
;
25
(
2
):
485
489
.
52.
Musangile
 
FY,
Matsuzaki
 
I,
Okodo
 
M,
et al.
Detection of HPV infection in urothelial carcinoma using RNAscope: clinicopathological characterization
.
Cancer Med
.
2021
;
10
(
16
):
5534
5544
.
53.
Cowan
 
M,
Springer
 
S,
Nguyen
 
D,
et al.
High prevalence of TERT promoter mutations in primary squamous cell carcinoma of the urinary bladder
.
Mod Pathol
.
2016
;
29
(
5
):
511
515
.

Author notes

Supplemental digital content is available for this article at https://meridian.allenpress.com/aplm in the January 2025 table of contents.

This study was funded by the University of California San Francisco Department of Pathology Research Endowment award to Kayraklioglu.

Competing Interests

The authors have no relevant financial interest in the products or companies described in this article.

This work was presented as a platform presentation at the 2022 United States and Canadian Academy of Pathology Annual meeting; March 21, 2022; Los Angeles, California.

Supplementary data