ArticlePDF Available

Female Immunity Protects from Cutaneous Squamous Cell Carcinoma

Authors:

Abstract and Figures

Purpose: Cancer susceptibility and mortality are higher in males, and the mutational and transcriptomic landscape of cancer differs by sex. The current assumption is that men are at higher risk of epithelial cancers as they expose more to carcinogens and accumulate more damage than women. We present data showing women present with less aggressive primary cutaneous squamous cell carcinoma (cSCC) and early strong immune activation. Experimental design: We explored clinical and molecular sexual disparity in immunocompetent and immunosuppressed patients with primary cSCC (N = 738, N = 160), advanced-stage cSCC (N = 63, N = 20) and FVB/N mice exposed to equal doses of DMBA, as well as in human keratinocytes by whole-exome, bulk, and single-cell RNA sequencing. Results: We show cSCC is more aggressive in men, and immunocompetent women develop mild cSCC, later in life. To test whether sex drives disparity, we exposed male and female mice to equal doses of carcinogen, and found males present with more aggressive, metastatic cSCC than females. Critically, females activate cancer immune-related expression pathways and CD4 and CD8 T-cell infiltration independently of mutations, a response that is absent in prednisolone-treated animals. In contrast, males increase the rate of mitosis and proliferation in response to carcinogen. Women's skin and keratinocytes also activate immune-cancer fighting pathways and immune cells at UV radiation-damaged sites. Critically, a compromised immune system leads to high-risk, aggressive cSCC specifically in women. Conclusions: This work shows the immune response is sex biased in cSCC and highlights female immunity offers greater protection than male immunity.
Content may be subject to copyright.
Female immunity protects from cutaneous squamous cell
carcinoma
Timothy Budden1,2, Caroline Gaudy-Marqueste3, Sarah Craig1,2, Yuan Hu4,5, Charles H.
Earnshaw1,2, Shilpa Gurung1,2, Amelle Ra6, Victoria Akhras6, Patrick Shenjere7, Ruth
Green8, Lynne Jamieson8, John Lear9, Luisa Motta8, Carlos Caulín4,5, Deemesh Oudit10,
Simon J Furney11,12, Amaya Virós1,2,9,*
1Skin Cancer and Ageing Lab, Cancer Research UK Manchester Institute, The University of
Manchester, Manchester, UK
2National Institute for Health Research Manchester Biomedical Research Centre, Manchester
3Aix Marseille Univ, APHM, CRCM Inserm U1068, CNRS U7258, CHU Timone, Dermatology and
Skin Cancer Department, Marseille, France
4Department of Otolaryngology, Head and Neck Surgery, University of Arizona, Tucson, AZ, USA
5 University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
6Department of Dermatology, St. George’s NHS Foundation Trust, London, UK
7Department of Histopathology, The Christie Hospital NHS Foundation Trust, Manchester, UK
8Department of Histopathology, Salford Royal NHS Foundation Trust, The University of
Manchester, Manchester UK
9Department of Dermatology, Salford Royal NHS Foundation Trust, The University of Manchester,
Manchester UK
10Department of Plastic and Reconstructive Surgery, The Christie Hospital NHS Foundation Trust,
Manchester, UK
11Genomic Oncology Research Group, Department of Physiology and Medical Physics, Royal
College of Surgeons in Ireland, Dublin, Ireland
12Centre for Systems Medicine, Royal College of Surgeons in Ireland Dublin, Ireland
Abstract
Purpose—Cancer susceptibility and mortality are higher in males, and the mutational and
transcriptomic landscape of cancer differs by sex. The current assumption is that men are at higher
* Corresponding author: Amaya Virós, MD, PhD, Wellcome Trust Clinician Scientist, Skin Cancer and Ageing Lab, Cancer
Research UK Manchester Institute, Alderley Park. Alderley SK10 4TG, United Kingdom. Telephone: +44 (0) 7878794211, +44 (0)
161 306 6038 Amaya.viros@cruk.manchester.ac.uk.
Conflicts of interest: No competing interests
Author contributions
Conceptualization: A.V.; Writing of the original manuscript and generation of figures T.B. and A.V.; Collection of data: T.B., A.V.,
S.C., C.E., A.R., V.A., P.B., D.O., R.G., L.J., L.M., J.L., S.G., Y.H., C.C., C.G.-M.; Analysis of data: T.B., A.V., L.M., C.C, S.J.F.;
Tools and methods: T.B., A.V., S.J.F.; Software: T.B., S.J.F.; Funding acquisition, project administration, resources, supervision: A.V.
Europe PMC Funders Group
Author Manuscript
Clin Cancer Res. Author manuscript; available in PMC 2022 September 22.
Published in final edited form as:
Clin Cancer Res
. 2021 June 01; 27(11): 3215–3223. doi:10.1158/1078-0432.CCR-20-4261.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
risk of epithelial cancers as they expose more to carcinogens and accumulate more damage than
women. We present data showing women present less aggressive primary cutaneous squamous cell
carcinoma (cSCC) and early strong immune activation.
Methods—We explored clinical and molecular sexual disparity in immunocompetent and
immunosuppressed primary cSCC patients (N=738, N=160), advanced stage cSCC (N=63, N=20)
and FVB/N mice exposed to equal doses of DMBA, as well as in human keratinocytes by whole
exome, bulk and single cell RNA sequencing.
Results—We show cSCC is more aggressive in men, and immunocompetent women develop
mild cSCC, later in life. To test if sex drives disparity, we exposed male and female mice to equal
doses of carcinogen, and found males present more aggressive, metastatic cSCC than females.
Critically, females activate cancer immune-related expression pathways and CD4 and CD8 T
cell infiltration independently of mutations, a response that is absent in prednisolone treated
animals. In contrast, males increase the rate of mitosis and proliferation in response to carcinogen.
Women’s skin and keratinocytes also activate immune-cancer fighting pathways and immune
cells at ultraviolet radiation-damaged sites. Critically, a compromised immune system leads to
high-risk, aggressive cSCC specifically in women.
Conclusions—This work shows the immune response is sex biased in cSCC, and highlights
female immunity offers greater protection than male immunity.
Key words
cancer sex bias; cutaneous SCC; cancer immunity activation; non-melanoma cancer; cancer and
immunosuppression
Introduction
Many diseases show sex disparity in their epidemiology, clinical course and outcome(1),
and cancer incidence is higher in men even after adjusting for risk factors. Cancer
mortality also affects men disproportionately and women respond better to some cancer
therapies(2–4). Squamous cell carcinomas (SCCs) arise in epithelia from the head and neck,
lung, bladder, esophagus and skin, have a strong male bias and are primarily caused by
carcinogens such as tobacco, alcohol and ultraviolet radiation (UVR). The higher SCC
male incidence and mortality is thought to reflect the increased lifetime exposure of men
to carcinogens and delayed clinical care; and molecular studies show a higher burden of
carcinogen-driven mutations in male tumors(5–9). SCCs arising on the skin, cutaneous SCC
(cSCC)(10–12), are more frequent in men, the 2nd most frequent malignancy in humans and
the most common malignancy in patients with a compromised immune system(10,13,14).
Previous studies indicate male mice may be inherently more susceptible to cSCC(15,16),
so here we examined if the higher incidence and mortality of cSCC in men is due to
increased vulnerability to epithelial neoplasia reflected in the molecular landscape, rather
than due to increased exposure to a carcinogen. To test this hypothesis, we used the
most frequent cSCC mouse model driven by the topical cutaneous application of DMBA,
a polycyclic aromatic hydrocarbon that induces epidermal
Hras
mutations, followed by
topical tetradecanoyl-phorbol acetate (TPA), which induces inflammation and epidermal
proliferation, leading to epidermal papillomas and cSCC(17). The advantages of this model
Budden et al. Page 2
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
are that the genomic landscape of DMBA/TPA-induced cSCC displays significant overlap
with human cutaneous SCC, and that it allows the
in vivo
study of cSCC controlling for age,
strain, susceptibility and dose of carcinogen(18). We explored the clinical and molecular
sex bias of cSCC carcinogenesis in animals exposed to the same dose of carcinogen,
and confirmed the molecular findings in human skin and keratinocytes. Additionally, we
examined the relationship between age at diagnosis, histological grade of cSCC and immune
status in human cSCC patient cohorts to explore the role of immunity by sex.
Methods
Animal experiments
Experiments were performed in 4-week-old FVB/N male and female mice, and DMBA
(25mg/ml) and TPA (0.02mg/ml) in acetone applied once a week, two days apart for 6
weeks, followed by TPA weekly for 10 weeks or until tumor development. All procedures
involving animals were performed under the Home Office approved project license
P8ADED6C8, and UK Home Office regulations under the Animals (Scientific Procedures)
Act 1986. The study received ethical approval by the Cancer Research UK Manchester
Institute’s Animal Welfare and Ethics Review Body (AWERB).
Molecular analyses
DNA from the largest sized mouse tumor, adjacent treated skin and a kidney was sequenced,
patterns of single nucleotide variation, insertion and deletions in tumors and skin were
analysed and oncoplots generated of the top 20 frequently mutated genes by sex and
histology to identify patterns of mutations. Paired-end RNA sequencing was performed from
fresh whole DMBA/TPA treated skin from the back and normal skin from the abdomen.
Human single cell RNA-seq data from male and female keratinocytes was analysed from a
study of squamous cell carcinoma(19), downloaded from GEO database (GSE144236).
Histology
Mouse tumors were classified as papillomas, keratoacanthoma, well, moderately and poorly
differentiated cSCC, and visceral organs stained with H&E and pan-keratin to confirm
metastasis. Immunohistochemistry of tumors and skin was performed with anti-phospho-
Histone H3 (Ser10) (06-570) CD4 Antibody (14-9766-82) ThermoFisher / eBioscience and
CD8a Antibody (14-0808-82) ThermoFisher / eBioscience.
Human clinical data
A retrospective, non-interventional review of clinical data: age, sex, immune status and
histological grade of human primary cSCC, metastatic cSCC to the lymph node and
cSCC treated with systemic therapy from 3 UK NHS centers and one French hospital,
from immune-competent and immune-suppressed patients diagnosed by dermatologists
and pathologists under routine diagnostic practice were collected. The review at Salford
Royal is part of a larger skin cancer study IRAS approval: 216310, REC reference:
16/LO/2098. The Christie and St.George’s clinicians collected age, sex and immune status
within the SCC care audit. No patient consent of retrospectively obtained, anonymized
variables was required by local hospital and National Health Research Authority UK.
Budden et al. Page 3
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
The clinical data collection in the French cohort was approved by tumour board local,
national regulations, given Comite de Protection des Personnes Sud Méditerranée and Aix-
Marseile University Hospital approval. Publicly available genomic data from single cell
RNA seq analysis of human skin was accessed by GEO database (GSE144236). cSCC
were classified as keratoacanthoma (KA)/ well differentiated invasive SCC (1), moderately
differentiated SCC (2) and poorly differentiated SCC (3). The immune status of patients
was retrieved from clinical histories, and immunosuppressed patients were organ transplant
recipients on immunosuppressive medication, with white blood cell dyscrasia, systemic
cancer treatment with immunotherapy, radiotherapy or chemotherapy in the past 10 years,
chronic inflammatory disorders and autoimmune disorders on systemic immunosuppressive
medication.
Results
Immune competent men develop more aggressive cSCC than women
To explore if men are more susceptible to aggressive cSCC than women, we studied the
relationship between sex, age and histological grade in consecutive cSCC samples excised
from immunocompetent patients (n = 738; men: 459, women: 279, Supplementary Table
1A). We confirmed a male preponderance of cSCC in this cohort of immunocompetent
men(10) (62.2% men, Z-test p <0.0001), and found the median age of diagnosis to be similar
in men and women (median age men = 79, women = 81, Mann Whitney test p=0.10, Fig.
1A). Strikingly, men more frequently presented cSCC that was less differentiated, of higher
histological grade compared to cSCC of women (Fig. 1B, Mann Whitney test p=0.0002).
Furthermore, when we restricted our study by sex to the more aggressive variants of cSCC,
which have the highest risk of metastasis(14), we found that immunocompetent females
diagnosed with higher grade cSCC were older than men (median age men =80, median
women =83, p=0.04, Fig. 1C). Additionally, a review of the prevalence of metastatic cSCC
to the lymph nodes in a tertiary cancer hospital revealed male preponderance (men: 25,
women: 4, Z-test p = 0.0001, Supplementary Table 1B), and men developed aggressive
primary cSCC at a younger age than women (women: 86 (71-91), men 73 (41-91), p=0.01,
Figure 1D). Furthermore, we investigated sex bias in a cohort of patients with metastatic or
advanced stage cSCC that required systemic treatment, which showed more men received
systemic treatment than women (men: 62.2%, Z-test p = 0.0011; Supplementary Table 1C,
Figure 1E), and intriguingly, female cSCC was histologically more aggressive in this cohort
of poor outcome (Mann Whitney test p = 0.01, Figure 1F). These data show that men are at
higher risk of aggressive cSCC.
Male mice are more susceptible to chemically-induced aggressive cSCC
To determine if the increased susceptibility to aggressive cSCC in men is due to a greater
exposure to carcinogens or due to biological sex differences, we exposed immunocompetent
male and female mice, matched for age and strain, to equal doses of the carcinogen DMBA/
TPA, which promotes cSCC in mice. We recorded earliest lesion incidence and burden, and
found males developed more papillomas and cSCC earlier than female animals, although
this difference was not statistically significant (p=0.10, Fig. 2A). Animals presented a
range of squamoproliferative lesions, including epidermal hyperplasia, papillomas, well-
Budden et al. Page 4
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
differentiated, invasive SCC and more aggressive, undifferentiated SCC. However, compared
to females, males presented more advanced, aggressive subtypes of disease (Fisher exact
test, p=0.04) (Fig. 2B). Specifically, male cSCC presented more mesenchymal, spindle
features, compared to female cSCC. Importantly, only males presented metastatic lung
cSCC deposits (Fig. 2B, 2C). Taken together, these data show male mice developed more
aggressive primaries and metastatic cSCC than females exposed to the same dose of
carcinogen.
DNA damage accumulates equally in male and female animal skin and cSCC
Increased mutation burden underpins more aggressive forms of epithelial cancer(20), and
human male epithelial tumors have a higher mutation burden(5–9). Therefore, we examined
if the more aggressive male cSCCs in our animals are due to greater mutation accumulation,
or less DNA repair, leading to higher mutation burden. For this, we compared the tumor
mutation burden (TMB) in DMBA/TPA-induced mouse cSCCs (DT-cSCC), and found deep
targeted exome sequencing of DT-cSCC revealed the number of total mutations increased
with increasing histological grade, but did not differ by sex (Fig. 2D, 2E). Furthermore, we
found no differences in the pattern and types of mutations by sex (Supplementary Table 2,
Supplementary Fig. 1A).
Mouse tumors display a range of morphological features within the same histological grade,
and cancer-driving mutations can associate specific histological features(21). Therefore,
to ensure specimens were exactly comparable between males and females, we focused
on mutations accrued in clinically and histologically normal DMBA/TPA-treated skin (DT-
skin). We found males and females accumulate the same amount of genetic damage in
DT-skin (Fig. 2D), and similar to DT-cSCCs, there was no sex disparity in the number,
pattern and types of mutations (Supplementary Fig. 1B, Supplementary Table 2). These
data indicate carcinogens damage male and female DNA similarly, and the more aggressive
phenotype of male cSCC is independent of mutation burden.
Unique transcriptomic and immune changes by sex in mice
Male animals develop more aggressive cSCC independently of the mutation spectrum.
Therefore, we explored if the transcriptomic response to carcinogen exposure differed by
sex. To reduce the gene expression variability due to histological differences between
tumors, we compared the histologically normal, carcinogen exposed DT-skin of males
and females. We first investigated the autosomal gene expression changes in DT-skin
and normal skin, and then studied whether DT-skin gene expression varied by sex. We
found the most significant changes in DT-skin involved critical cancer immune regulatory
pathways including the interferon gamma and interferon alpha response pathways; as well
as the inflammation-related and allograft rejection genes (Fig. 3A, Supplementary Table
3A). In contrast, normal, untreated skin expressed genes involved in RAS signaling (Fig.
3A, 3B). We then investigated sex-specific changes in DT-skin, and found female DT-skin
presented more transcriptomic changes than male skin overall, including higher expression
of critical genes involved in cancer immunity, compared to males. We noted specifically the
cytokine interferon gamma (
Ifng
), which is known to drive potent antitumor activity(22–24),
to be increased in female carcinogen-treated skin. Intriguingly, we additionally observed
Budden et al. Page 5
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
female upregulation of the G1 to S-phase tumor suppressor, senescence-inducing cell
cycle regulator
Cdkn2a
(Fig. 3C, Supplementary Table 3B). Cdkn2a can exert ample anti-
tumor effects(25,26), and when expressed in keratinocytes restricts proliferation, increases
senescence and differentiation, and reduces tumor growth(27), consistent with its tumor-
suppressive roles. Furthermore, recent work shows natural and immune checkpoint cancer
immune control is achieved via Cdkn2a-dependent signaling, and interferons directly
activate
Cdkn2a
(28). We therefore examined cSCC incidence, histological grade and
metastasis in an available model of spontaneous carcinogenesis in
Cdkn2afl/fl
animals(29),
and found that although the study was not powered to detect sex differences,
Cdkn2afl/fl
female mice presented more spindle, aggressive primary tumour and metastatic cSCC than
Cdkn2awt
females (two-way Anova p =0.0796, Supplementary Fig. 1C, Supplementary
Table 4).
We next investigated if, in addition to immune-gene expression changes, female animals
present a unique immune cell landscape compared to males. For this, we studied the
proportion and subtypes of adaptive and innate immune cells by gene expression, which
revealed a trend in female skin for increased antigen-presenting CD1 cells and CD4 / CD8
tumor lymphocytes. In contrast, male animal skin tended to be enriched for macrophages
(Supplementary Fig. 1D) although these were not significant (p>0.5, Mann Whitney test).
We explored gene expression by immunohistochemistry staining of CD4+ and CD8+ T
cells in DT-cSCC and DT-skin by sex, and confirmed female tumors and female DT-skin
were more infiltrated with immune cells than male DT-cSCC and DT-skin (Fig. 3D, 3E. To
confirm the immune response to the DT challenge is sex-specific, we compared the early
inflammatory and CD4+ cell infiltration in immunocompetent and immunosuppressed mice.
We confirmed that DT-skin from immunocompetent females presented a denser immune
cell (Supplementary Fig. 1E, 1F) and CD4+ infiltration (Supplementary Fig. 1G, 1H) across
all the layers of the skin, compared to males. However, immunosuppressed females and
males, treated with oral prednisolone in addition to DMBA/TPA, had an equally reduced
inflammatory cell response and CD4+ infiltration (Supplementary Fig. 1E-I). These data
indicate the early inflammatory response to carcinogen in immunocompetent animals differs
by sex, and this difference is lost in immunosuppressed animals.
Tumors in animals arise weeks after being exposed to the DMBA carcinogen, but the total
number of mutations per cSCC rises progressively in tumors of increasing histological grade
(Fig. 2E). We hypothesized the selection and growth of the more mutated clones leading to
more aggressive cSCC occurs due to increased cell division and a proliferation advantage
of these clones. As males present more aggressive disease, and female epidermis expresses
higher levels of the cell cycle regulator
Cdkn2a
, we investigated if a higher epidermal
proliferation rate in the male epidermis could underpin speedier clonal selection and disease
progression of male disease. For this, we compared the rate of the mitosis-specific marker
phospho-histone 3 (phospho-H3) in male and female epidermis and observed that DT-skin,
compared to animal-matched non-DT skin was thickened, presenting increased number of
layers, independently of sex (Supplementary Fig. 1J, p<0.0001). Next, we compared male
to female DT-epidermis and found that phosphoH3 in male epidermis was increased in
males (Fig. 3F, 3G, p<0.0001). We compared the expression of phospho-H3 in a subset of
cSCCs, and again observed a trend for higher expression in males (Supplementary Fig. 1K,
Budden et al. Page 6
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
1L). Thus, male and female mouse epidermis modulate the rate of epidermal proliferation
differently following exposure to a chemical carcinogen.
Taken together, these findings indicate the transcriptome and immune cell response of
male and female mice differs at the earliest stage of carcinogenesis, independently of
DNA damage. Males present higher rates of epidermal proliferation following chemical
carcinogen exposure compared to females. In contrast, females strongly upregulate
immune responses and cancer-linked lymphocytes, implicating immunity in delayed female
tumorigenesis.
Human female skin upregulates immune-related cancer defense pathways
To validate the role of immune-related expression changes and immune cells modulating
carcinogenesis in females, we compared the sex-specific changes in single cell RNA
of normal male and female human keratinocytes obtained from cSCC-adjacent skin(19).
Human cSCC is driven by ultraviolet light and arise in fields of severely sun-damaged skin,
so we reasoned cSCC-adjacent keratinocytes, from sun exposed anatomic sites of adults,
will accumulate significant carcinogen exposure. Indeed, we found that similar to animal
DT-skin, the most prominently upregulated pathways in human keratinocytes overlapped
with mouse skin. Strikingly, the most prominent pathway expression changes in all female
keratinocyte subtypes were up-regulation of interferon gamma (IFNG), interferon alpha and
allograft rejection pathway genes (Fig. 4A, Supplementary Table 5). In further agreement
with the mouse experiment, we found female human sun-exposed epidermis and cSCCs had
a trend to higher counts of CD4, CD8 T cells and CD1C dendritic cells, whilst male skin and
cSCC showed a trend for more macrophages (Mann-Whitney p=0.38, Supplementary Fig.
2A, 2B).
The use of the human cSCC single-cell RNAseq data allows expression patterns to be
isolated to specific cell types, and this reveals that in human skin and cSCC,
IFNG
is
almost exclusively expressed by T Cells. Cytokines
CXCL10
and
CXCL9
, which can
chemoattract T-cells, are expressed from multiple cell types including keratinocytes and
other immune cells such as Langerhans cells. The immune pathway enrichment of human
female keratinocytes (Fig. 4A) is particularly enriched for genes that play a role in antigen
presentation such as HLA genes and
TAP1
, suggesting a complex multicellular immune
response in human skin including alterations in cytokine production, T cell recruitment
and IFNG production, and increased antigen presentation of keratinocytes for immune
recognition in female skin after UV damage (Supplementary Fig. 2C, 2D). Overall, these
data indicate that the biological response to chemical and ultraviolet light carcinogen
exposure in keratinocytes varies by sex in mice and humans.
Female immunity inhibits aggressive cSCC
Because we found the immune response is critical to the carcinogen response in female
animals, and immune competent women have less aggressive forms of cSCC (Fig. 1B),
we reasoned women with compromised immune systems would present more aggressive
cSCC, comparable to immunocompetent men. To test this hypothesis, we investigated sex,
age and histological grade in cSCC specimens excised from a cohort of immunosuppressed
Budden et al. Page 7
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
patients (IS, n= 160, men =106 and women n=54, Supplementary Table 1, 6). Strikingly,
we discovered cSCC appeared at a younger age in both immunosuppressed men and
women, supporting the critical role of immunity in delaying cancer in both sexes (median
age men =72, women =71, p=0.25, Fig. 4B). We then compared the histological grade
in immunosuppressed and immunocompetent women, which revealed immunosuppressed
women present more histologically aggressive disease (p=0.029, Fig. 4C), of equivalent
grade to men. By contrast, men present aggressive disease regardless of their underlying
immune status (Fig. 4C).
Next, we restricted our study by sex to the more aggressive, risky variants of cSCC in
the immunosuppressed population, and found, as expected, that aggressive disease occurred
earlier in life in both sexes. However, the lower age at diagnosis particularly occurred in
women, suggesting the loss of immunity is more damaging to females (median age men
=73, median women =70, p=0.04, Fig. 4D). Taken together, these data show the immune
system is critical to contain carcinogenesis, and validate the immune response underpins less
aggressive disease in females.
Discussion
The molecular landscape of cancer differs by sex, and men are more frequently diagnosed
with cancer and die more of cancer than women. Men traditionally are exposed to more
carcinogens due to professional hazards and behavior, but in risk-adjusted epidemiological
studies, there is still an unexplained greater proportion of men with cancer compared to
women(2–4). Furthermore, there is significant variation in the DNA alterations and RNA
landscape by sex in most cancers, suggesting there are sex-specific susceptibilities and
biological processes driving female and male cancer(5–9). However, comparing human
tumors from different sexes is a limited approach as human tumors are not matched for age,
histological grade, underlying susceptibility to cancer, and dose of carcinogen.
In this study we show immunocompetent women have less aggressive cSCC than men.
We compared the development and the molecular hallmarks of cSCC in male and female
animals using a well-established mouse model of DMBA/TPA carcinogenesis, adjusted
for carcinogen exposure(18), showing that similar to other studies using UVB as the
carcinogenic challenge(15,16), males present histologically more aggressive cSCC and
metastasis.”
We show that the rate of DNA damage accumulation is equal in male and female mice
exposed to equal doses of DMBA. This indicates both sexes repair damage similarly
following exposure to the chemical carcinogen. Intriguingly, we observe the overall
mutation burden (TMB) increases as tumors become more aggressive, or with increasing
histological grade in both sexes; despite animals not receiving additional exposures to
carcinogen. This suggests that in DMBA-driven cSCC there is a selection for clones with
more mutations as cell division progresses and tumors advance. Previous work comparing
mutation burden in adjacent areas of early dysplasia, intermediate dysplasia and primary
cutaneous melanoma reveal increasing mutation burden with oncogenic stage(30); raising
the hypothesis that areas with early damage must receive additional carcinogenic exposure
Budden et al. Page 8
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
to drive cancer progression. However, our study indicates carcinogenic damage conferring
full oncogenic potential may occur at the early stage and will depend on successful
expansion of clones with higher TMB, as we discontinued carcinogen exposure before cSCC
onset.
Intriguingly, we find it is the transcriptional response to carcinogen exposure that differs
between the sexes. At the earliest stage of disease, female animals increase cancer immune-
related responses and recruit an immune cell landscape involved in cancer defense, which is
reversed in animals on prednisolone with a compromised immune system. Male animals,
by contrast, have more macrophages, which are linked to poor prognosis in cancer,
and comparatively show few immune-related gene expression changes. The macrophage
male sex bias, although from a small cohort, is mirrored in the human immune cell
cSCC landscape as well as in a mouse hepatocellular carcinogenesis model driven by
diethylnitrosamine(31), indicating they may be specifically involved in the male response to
extrinsic damage.
One critical gene expression difference we found between male and female animals is
the up-regulation of
Cdkn2a
in females, a fundamental cell cycle regulator that exerts
profound influence in both cell proliferation(25,26) and cancer-immune responses(27,28).
Intriguingly, our preliminary data show the histological grade and rate of metastasis is more
advanced in
Cdkn2afl/fl
than in
Cdkn2af
wild type females, suggesting
Cdnkn2a
expression
may play a critical role restricting female cSCC carcinogenesis. In further support for a
central role of
Cdnkn2a
, we found the mitotic rate of carcinogen-exposed epithelium was
higher in male epidermis, indicating greater proliferation, than female skin. Additionally,
epidemiological studies indicate that
CDKN2A
germline loss-of-function carriers, who
are at high risk for cSCC and melanoma, are particularly susceptible to tobacco-induced
lung cancers(32). This further supports a central role for CDKN2A in the response to
environmental damage beyond skin carcinogenesis.
We validated the relevance of the
in vivo
findings in human female and male epidermal
keratinocytes, which originated from a sun-exposed site(19), and in human cSCCs.
Keratinocytes from women mirror the response observed in female mouse epidermis,
up-regulating critical cancer-immune pathways, genes and immune cells following UVR,
compared to men. The use of human cSCC single-cell RNAseq data allows expression
patterns to be isolated to specific cell types, indicating the cytokine differences are expressed
by multiple cell types, including keratinocytes and immune cells, in keeping with a complex
multicellular immune response in human skin which requires further study.
Similar to the mouse model, although cSCC arises at a similar age in immunocompetent
men and women, men have significantly more aggressive disease, and the aggressive tumors
arise at a younger age in men. Restricting our analysis to immunosuppressed patients, we
show cSCC arises in younger men and women. Importantly, cSCC in immunosuppressed
women are significantly more aggressive, similar to male disease, whilst male disease
is aggressive regardless of immune status. Although our study includes a broad and
heterogeneous range of diagnoses and medication driving immune suppression in our
patients, these data show the immune response plays a critical role constraining cSCC
Budden et al. Page 9
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
progression at the earliest stage of disease in both sexes, as the age of incidence drops
in both men and women. However, the data indicate the mechanisms restricting tumor
progression are more robust in females. Thus, this study, adjusted for risk factors, implicates
distinct biology driving male and female cancer; and shows differences in immune responses
between the sexes. Future studies should be conducted to identify the potential regulators
of sex-linked SCC dimorphism, to understand if the sex bias in cSCC onset and outcome is
due to chromosomal content or due to hormonal causes, and if our findings extend to other
epithelial and non-epithelial cancers with a strong male bias.
These results are strongly aligned with clinical observations revealing higher incidence
of excess immunity-linked disease in females and unique immune responses to infectious
disease by sex(33). Personalized medicine approaches stratify cancer patients by genotype;
however, to date, the potential for cancer stratification and therapy by sex has not been
explored. Further work will be necessary to identify the potential regulators of sex-linked
cSCC dimorphism.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgements
We acknowledge the contribution of CRUK Manchester Institute and Facilities, in particular
the Histology department led by Garry Ashton. We are grateful to the Khavari Lab for
advice and shared data. We acknowledge the support of the National Institute for Health
Research Manchester Biomedical Research Centre, Manchester.
Funding
AV: Wellcome Beit Fellow, personally funded by a Wellcome Intermediate Fellowship
(110078/Z/15/Z), additional work funded by Cancer Research UK (A27412) Leo
Pharma Foundation and Royal Society RGS\R1\201222. SJF: European Commission (FP7-
PEOPLE-2013-IEF – 6270270) and the Royal College of Surgeons in Ireland StAR
programme. The Irish Centre for High End Computing (https://www.ichec.ie/) provided
HPC infrastructure.
References
1. Sampathkumar NK, Bravo JI, Chen Y, Danthi PS, Donahue EK, Lai RW, et al. Widespread
sex dimorphism in aging and age-related diseases. Hum Genet. 2020; 139: 333–56. [PubMed:
31677133]
2. Clocchiatti, A, Cora, E, Zhang, Y, Dotto, GP. Nat Rev Cancer. Vol. 16. Nature Publishing Group;
2016. 330–9. [Internet]
3. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018: Cancer Statistics, 2018. CA Cancer J Clin.
2018; 68: 7–30. DOI: 10.3322/caac.21442 [PubMed: 29313949]
4. Pearce MS, Parker L. Childhood cancer registrations in the developing world: Still more boys than
girls. Int J Cancer. 2001; 91: 402–6. [PubMed: 11169966]
Budden et al. Page 10
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
5. Dunford A, Weinstock DM, Savova V, Schumacher SE, Cleary JP, Yoda A, et al. Tumor-suppressor
genes that escape from X-inactivation contribute to cancer sex bias. Nat Genet. 2017; 49: 10–6.
[PubMed: 27869828]
6. Yuan, Y, Liu, L, Chen, H, Wang, Y, Xu, Y, Mao, H. , et al. Cancer Cell. Vol. 29. Elsevier Inc; 2016.
Comprehensive Characterization of Molecular Differences in Cancer between Male and Female
Patients; 711–22. [Internet]
7. Li CH, Haider S, Shiah YJ, Thai K, Boutros PC. Sex differences in cancer driver genes and
biomarkers. Cancer Res. 2018; 78: 5527–37. [PubMed: 30275052]
8. Lopes-Ramos CM, Kuijjer ML, Ogino S, Fuchs CS, DeMeo DL, Glass K, et al. Gene regulatory
network analysis identifies sex-linked differences in colon cancer drug metabolism. Cancer Res.
2018; 78: 5538–47. [PubMed: 30275053]
9. Lotz, M, Budden, T, Furney, SJ, Virós, A. Br J Dermatol. England: 2020.
10. Venables ZC, Autier P, Nijsten T, Wong KF, Langan SM, Rous B, et al. Nationwide Incidence
of Metastatic Cutaneous Squamous Cell Carcinoma in England. JAMA Dermatology. 2019; 155:
298–306. [PubMed: 30484823]
11. Karia, PS, Han, J, Schmults, CD. J Am Acad Dermatol. Vol. 68. Elsevier Inc; 2013. 957–66.
[Internet]
12. Schmults CD, Karia PS, Carter JB, Han J, Qureshi AA. Factors predictive of recurrence and
death from cutaneous squamous cell carcinoma: A 10-year, single-institution cohort study. JAMA
Dermatology. 2013; 149: 541–7. [PubMed: 23677079]
13. Stang A, Khil L, Kajüter H, Pandeya N, Schmults CD, Ruiz ES, et al. Incidence and mortality
for cutaneous squamous cell carcinoma: comparison across three continents. J Eur Acad Dermatol
Venereol. 2019; 33 (Suppl 8) 6–10.
14. Manyam BV, Garsa AA, Chin R-I, Reddy CA, Gastman B, Thorstad W, et al. A multi-institutional
comparison of outcomes of immunosuppressed and immunocompetent patients treated with
surgery and radiation therapy for cutaneous squamous cell carcinoma of the head and neck. Cancer
United States. 2017; 123: 2054–60.
15. Dehelean CA, Soica C, Pinzaru I, Coricovac D, Danciu C, Pavel I, et al. Sex differences and
pathology status correlated to the toxicity of some common carcinogens in experimental skin
carcinoma. Food Chem Toxicol an Int J Publ Br Ind Biol Res Assoc England. 2016; 95: 149–58.
16. Thomas-Ahner JM, Wulff BC, Tober KL, Kusewitt DF, Riggenbach JA, Oberyszyn TM. Gender
differences in UVB-induced skin carcinogenesis, inflammation, and DNA damage. Cancer Res
United States. 2007; 67: 3468–74.
17. Abel EL, Angel JM, Kiguchi K, DiGiovanni J. Multi-stage chemical carcinogenesis in mouse skin:
fundamentals and applications. Nat Protoc. 2009; 4: 1350–62. [PubMed: 19713956]
18. Nassar D, Latil M, Boeckx B, Lambrechts D, Blanpain C. Genomic landscape of carcinogen-
induced and genetically induced mouse skin squamous cell carcinoma. Nat Med. 2015; 21: 946–
54. [PubMed: 26168291]
19. Ji AL, Rubin AJ, Thrane K, Jiang S, Reynolds DL, Meyers RM, et al. Multimodal Analysis
of Composition and Spatial Architecture in Human Squamous Cell Carcinoma. Cell. 2020; 182:
497–514. e22 [PubMed: 32579974]
20. Frese KK, Tuveson DA. Maximizing mouse cancer models. Nat Rev Cancer. 2007; 7: 654–8. DOI:
10.1038/nrc2192
21. Fu Y, Jung AW, Torne RV, Gonzalez S, Vöhringer H, Shmatko A, et al. Pan-cancer computational
histopathology reveals mutations, tumor composition and prognosis. Nat Cancer. 2020; 1: 800–10.
DOI: 10.1038/s43018-020-0085-8 [PubMed: 35122049]
22. Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al. IFNgamma and
lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature
England. 2001; 410: 1107–11.
23. Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ, et al. Demonstration of
an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl
Acad Sci U S A. 1998; 95: 7556–61. [PubMed: 9636188]
24. Zaidi MR, Merlino G. The two faces of interferon-γ in cancer. Clin cancer Res an Off J Am Assoc
Cancer Res. 2011; 17: 6118–24.
Budden et al. Page 11
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
25. Lee S, Schmitt CA. The dynamic nature of senescence in cancer. Nat Cell Biol England. 2019; 21:
94–101.
26. Pérez-Mancera PA, Young ARJ, Narita M. Inside and out: the activities of senescence in cancer.
Nat Rev Cancer England. 2014; 14: 547–58.
27. Azazmeh N, Assouline B, Winter E, Ruppo S, Nevo Y, Maly A, et al. Chronic expression of
p16(INK4a) in the epidermis induces Wnt-mediated hyperplasia and promotes tumor initiation.
Nat Commun. 2020; 11 2711 [PubMed: 32483135]
28. Brenner E, Schörg BF, Ahmetlić F, Wieder T, Hilke FJ, Simon N, et al. Cancer immune control
needs senescence induction by interferon-dependent cell cycle regulator pathways in tumours. Nat
Commun. 2020; 11 1335 [PubMed: 32165639]
29. Li Z, Gonzalez CL, Wang B, Zhang Y, Mejia O, Katsonis P, et al. Cdkn2a suppresses metastasis in
squamous cell carcinomas induced by the gain-of-function mutant p53(R172H). J Pathol England.
2016; 240: 224–34.
30. Shain AH, Yeh I, Kovalyshyn I, Sriharan A, Talevich E, Gagnon A, et al. The Genetic Evolution of
Melanoma from Precursor Lesions. N Engl J Med United States. 2015; 373: 1926–36.
31. Romualdo GR, Prata GB, da Silva TC, Fernandes AAH, Moreno FS, Cogliati B, et al. Fibrosis-
associated hepatocarcinogenesis revisited: Establishing standard medium-term chemically-induced
male and female models. PLoS One. 2018; 13 e0203879 [PubMed: 30212575]
32. Helgadottir H, Höiom V, Jönsson G, Tuominen R, Ingvar C, Borg A, et al. High risk of tobacco-
related cancers in CDKN2A mutation-positive melanoma families. J Med Genet. 2014; 51: 545–
52. [PubMed: 24935963]
33. Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol England. 2016;
16: 626–38.
Budden et al. Page 12
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Translational relevance
Sex bias affects cancer incidence, mortality and therapy response; and the molecular
landscape of cancer differs by sex. However, it is not known if the sex discrepancy
is due to a difference in behaviour and exposure to carcinogens, or due to sex-linked
susceptibility. This work reveals men are more susceptible to cutaneous aggressive
squamous cell carcinoma, in contrast to women who activate stronger immune responses
when challenged with the same carcinogens. The loss of immunity particularly affects
women.
Personalised medicine approaches stratify cancer patients by genotype; however, to
date, the potential for cancer stratification, prevention strategies and therapy by sex
and immune competency has not been explored. We show men and immunosuppressed
women are at higher risk of aggressive disease and may benefit from targeted prevention
programmes, closer surveillance and stratification for adjuvant therapy.
Budden et al. Page 13
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 1. Men have more aggressive cSCC.
(A) Age of immunocompetent patients diagnosed with primary cSCC by sex, n.s
not significant, Mann-Whitney p = 0.1. (B) Histological grade of primary cSCC
by sex in immunocompetent patients (median, interquartile range (IQR). (C) Age of
immunocompetent women and men diagnosed with moderately and poorly differentiated
primary cSCC. (D) Lymph node metastatic cSCC by sex and age at time of primary cSCC
diagnosis in immunocompetent patients. (E) Median age at primary diagnosis of a cohort of
Budden et al. Page 14
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
advanced stage cSCC treated with systemic agents. (F) Histological grade of cSCC by sex at
the time of primary tumour diagnosis (all tests Mann-Whitney).
Budden et al. Page 15
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 2. Male animals are more susceptible to chemically-induced aggressive cSCC.
(A) Days to first lesion in male and female skin exposed to DMBA/TPA, n.s not significant
Mann Whitney test p=0.1 (B) Histological grade of cSCC tumors by sex (Fisher exact test,
p = 0.04). (C) Representative H&E of papilloma (scale bar: 400μm), keratoacanthoma (scale
bar: 500μm), well differentiated cSCC (scale bar: 500μm), poorly differentiated cSCC (scale
bar: 500μm) and insert (box, scale bar: 100μm) and lung metastasis stain for pan-cytokeratin
(brown, scale bar: 1mm). (D) Tumor mutation burden in DMBA/TPA exposed skin (Treated
Skin) and DMBA/TPA-induced cSCC (Tumor) by sex, Mann Whitney test p = 0.7, p = 0.8
Budden et al. Page 16
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
(E) Tumor mutation burden in DMBA/TPA lesions by histological grade and sex. Error bars
represent standard error of the mean (bar).
Budden et al. Page 17
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 3. Female mouse skin upregulates immune cancer pathways and cells in response to
chemical carcinogenesis.
(A) Gene pathways differentially expressed by sex in normal skin (left) and in DMBA/
TPA-exposed skin (treated skin, right). Size of spheres represent numbers of enriched
genes in pathway, color represents significance of enrichment (red most significant). (B)
Differentially expressed genes enriched in pathways in normal skin and (C) treated skin.
Negative fold change (blue) represents genes expressed higher in female skin, positive fold
change (red) represents genes expressed higher in male skin. (D) Quantification of CD4
and CD8 in tumors and DMBA/TPA tumors and treated skin by sex, Mann Whitney test.
Budden et al. Page 18
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
(E) Representative images of H&E and CD4 and CD8 T cell IHC in male and female
cSCC (scale bar: 200μm). (F) Quantification of mitotic cell marker phospho-histone 3 (Ph3)
immunohistochemistry (IHC) intensity score low (1), moderate (2), high (3) and very high
(4), Mann Whitney test. (G) Photomicrographs of H&E (scale bar: 50μm) and mitotic cells
(Ph3, scale bar 15μm) IHC by sex in normal and DMBA/TPA-skin.
Budden et al. Page 19
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 4. cSCC histological grade of human cSCC is tied to female immunity.
(A) Pathways enriched for genes differentially expressed by sex in human adult
keratinocytes from UVR-exposed, tumor-adjacent skin by sex. Female pathways (left)
represent gene pathways expressed higher in female keratinocytes. Male pathways (right)
represent gene pathways expressed higher in male keratinocytes. Keratinocytes grouped into
basal, cycling and differentiated cells. Size of spheres represent numbers of enriched genes
in pathway, color represents significance (- log10(False discovery rate) of enrichment (red
most significant). (B) Age comparison of immunocompetent (IC) and immunosuppressed
(IS) patients diagnosed with primary cSCC by sex, Mann-Whitney (C) Histological grade
comparison of primary cSCC by sex in immunocompetent (IC) and immunosuppressed
Budden et al. Page 20
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
(IS) patients (median, interquartile range (IQR), Mann-Whitney. (D) Age comparison
of immunocompetent (IC) and immunosuppressed (IS) women and men diagnosed with
moderately and poorly differentiated primary cSCC, Mann-Whitney.
Budden et al. Page 21
Clin Cancer Res
. Author manuscript; available in PMC 2022 September 22.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
... Several mouse models recapitulate gender disparities in human disease [74][75][76][77], thereby allowing for the study of sex-based differences and their impact on the immune system. In an epithelial squamous cell carcinoma mouse model, female mice, upon exposure to a carcinogen, activate immune response pathways and upregulate CD4 + and CD8 + T regardless of any mutation in DNA caused by the carcinogen [78]. However, the male-sexed mice, upon exposure to a carcinogen, show a higher rate of mitosis and proliferation of cancer cells, which leads to less differentiated, higher-stage epithelial squamous cell carcinomas [78]. ...
... In an epithelial squamous cell carcinoma mouse model, female mice, upon exposure to a carcinogen, activate immune response pathways and upregulate CD4 + and CD8 + T regardless of any mutation in DNA caused by the carcinogen [78]. However, the male-sexed mice, upon exposure to a carcinogen, show a higher rate of mitosis and proliferation of cancer cells, which leads to less differentiated, higher-stage epithelial squamous cell carcinomas [78]. These findings indicate that immune responses may endow female-sexed mice with some degree of protection against carcinogen exposures which is highly relevant to bladder cancer. ...
... These findings indicate that immune responses may endow female-sexed mice with some degree of protection against carcinogen exposures which is highly relevant to bladder cancer. Furthermore, the studies in the in vivo models suggest that there are differences in the tumor's immune response between males and females [74][75][76][77][78][79]. Thus, the immune response may represent a major biological factor that contributes to sex and gender disparities in cancer patients. ...
Article
Full-text available
Sex and gender disparities in bladder cancer have long been a subject of interest to the cancer research community, wherein men have a 4 times higher incidence rate than women, and female patients often present with higher-grade disease and experience worse outcomes. Despite the known differences in disease incidence and clinical outcomes between male and female bladder cancer patients, clinical management remains the same. In this review, we critically analyze studies that report on the biological differences between men and women and evaluate how these differences contribute to sex and gender disparities in bladder cancer. Distinct characteristics of the male and female immune systems, differences in circulating hormone levels and hormone receptor expression, and different genetic and epigenetic alterations are major biological factors that all likely contribute to disparate incidence rates and outcomes for male and female bladder cancer patients. Future preclinical and clinical studies in this area should employ experimental approaches that account for and consider sex and gender disparities in bladder cancer, thereby facilitating the development of precision medicine for the effective treatment of bladder cancer in all patients.
... Ultraviolet (UV) radiation-induced somatic mutations are considered an early event for cSCC carcinogenesis, and immunosuppression is a key risk factor for their development [5]. This risk factor lends to being the most common cancer among immunocompromised individuals [6]. Metastasises are found in up to 5% of cases, and their five-year survival is 70% [7]. ...
... The anatomical distribution of cSCC between men and women, where men are more prone to head and neck cSCC (HNcSCC) and women lower limb cSCC, lends itself to several hypotheses for this phenomenon [9]. For example, men are less compliant with sun protection measures [11] and more likely to perform outdoor work [12], although a laboratory mice study shows male rats had more aggressive and clinically poorer outcomes histologically when cSCC tumour grade and overall risk profiles were matched between sexes [6]. Emerging evidence also demonstrates that males are disproportionately affected for cancer incidence, have poorer treatment responses, and have worse outcomes even when adjusting for known epidemiological risk factors [13][14][15]. ...
... It has long been thought that men are at a higher risk of HNcSCC due to occupational and lifestyle reasons, including the likelihood of performing outdoor work [92], having male-pattern baldness (scalp cSCC) [93], shorter haircuts (ear cSCC) [94] and not wearing lipsticks (external lip cSCC) [95]. Despite this, epidemiological studies adjusting for these risk factors still demonstrate that males suffer disproportionately for the prevalence of cSCCs and mortality across cancer subtypes [6,93]. For example, Duran et al. found poorer outcomes of non-scalp cSCC in men which potentially confounds the hypothesis that male-pattern baldness is a significant risk factor for poor outcomes in cSCC [94]. ...
Article
Full-text available
The incidence of head and neck cutaneous squamous cell carcinoma (HNcSCC) is unevenly distributed between men and women. At present, the mechanism behind this disparity remains elusive. This study conducted a systematic review and meta-analysis of proportions to investigate the disparity between sexes for patients with HNcSCC. PubMed, Scopus, EMBASE, MEDLINE, Emcare and CINAHL were searched in November 2021 and June 2022 (N > 50, English, human), and studies which examined the association between sex and HNcSCC were included. Analysis was conducted using RStudio with data and forest plots displaying males as a proportion of total patients with HNcSCC. Two independent researchers performed study selection, data extraction, data analysis and risk of bias. Eighty-two studies (1948 to 2018) comprising approximately 186,000 participants (67% male, 33% female) from 29 countries were included. Significantly more males had HNcSCC overall (71%; CI: 67–74). Males were also significantly more affected by cSCC of the ear (92%; CI: 89–94), lip (74%; CI: 66–81), and eyelid (56%; CI: 51–62). This study found HNcSCC disproportionately affected males overall and across all subtypes. Improving our understanding of sex-specific mechanisms in HNcSCC will better inform our preventive, therapeutic and prognostic practices.
... Further investigation into the biological mechanism revealed a potentially altered CD8 T-cell response. A study by Budden et al. [38] reported increased aggressiveness of CSCC and increased metastatic potential in CSCC in males compared to females. Validation of these clinical findings in murine models revealed increased anti-tumour immunity as well as increased CD4 and CD8 T-cell infiltration. ...
... Validation of these clinical findings in murine models revealed increased anti-tumour immunity as well as increased CD4 and CD8 T-cell infiltration. While this generally leads to milder CSCC in women [38], the cases included in our study present with advanced disease and exclusion of T-cell infiltrates could have occurred during progression of the lesions. However, due to the low numbers of samples and lack of histopathological confirmation of these findings, further investigation in a validation cohort is required to support this hypothesis. ...
Article
Full-text available
Simple Summary Cutaneous squamous cell carcinomas (CSCC) are the second most common skin cancer amongst Caucasians, accounting for up to 20–25% of skin cancers. Whilst a majority of CSCCs can be cured with surgery alone, approximately 3–5% of patients develop advanced CSCC, which encompass locally advanced tumours or tumours with distant metastatic spread. As CSCCs are highly immunogenic, there is a strong rationale for treatment with immunotherapy. Several phase II clinical trials have demonstrated the benefit of immunotherapy in patients with advanced CSCC. However, only half of patients with advanced CSCC respond to immunotherapy, and thus there is a need to identify predictors of response. In this study, we demonstrated inferior clinical outcomes in female patients with advanced CSCC treated with immunotherapy compared to their male counterparts. This clinical finding is supported with translational assays on pre-treatment biopsies, demonstrating the presence of fewer anti-tumour immune cells in the tumours of female patients. Abstract Approximately 3–5% of patients with cutaneous squamous cell carcinoma (CSCC) develop advanced disease, accounting for roughly 1% of all cancer deaths in Australia. Immunotherapy has demonstrated significant clinical benefit in advanced CSCC in several key phase II studies; however, there are limited data for patients treated outside of clinical trials. This is particularly relevant in advanced CSCC, which is most often seen in elderly patients with significant comorbidities. Thus, we aim to describe our experience with immunotherapy in a cohort of patients with advanced CSCC in Australia. We retrospectively reviewed all advanced CSCC patients treated with immunotherapy within the Illawarra and Shoalhaven Local Health District. Among the 51 patients treated with immunotherapy, there was an objective response rate (ORR) of 53% and disease control rate (DCR) of 67%. Our most significant predictor of response was sex, with male patients more likely to have better responses compared to female patients (DCR 85% vs. 41%, p < 0.0001), as well as improved progression-free survival (HR 4.6, 95%CI 1.9–10.8, p = 0.0007) and overall survival (HR 3.0, 95%CI 1.3–7.1, p = 0.006). Differential expression analysis of 770 immune-related genes demonstrated an impaired CD8 T-cell response in female patients. Our observed ORR of 53% is similar to that described in current literature with durable responses seen in the majority of patients.
... It may be that there are molecular influences determining some of this observed difference. 12 The relative risk of developing a primary cSCC was greatest in the southern and coastal regions of the study area for those over 60 years of age. This was not the case for those 40-60 years, where further inland saw the greatest relative risk. ...
Article
Full-text available
Background/objectives: To describe the incidence of primary cutaneous squamous cell carcinoma in coastal NSW Australia. Methods: The design is a case-controlled study of reported cSCC from 2016 to 2019 within a defined region of coastal southern NSW. Participants include all reported pathological diagnoses of cSCC in patients greater than 20 years of age. The main outcome measures the incidence and relative risk of cSCC. Results: The overall age-adjusted incidence rate of primary cSCC was 856//year. Men over 60 years of age had an age-adjusted incidence rate of 2875/106 /year. Histologically diagnosed invasive SCC samples were included using SNOMED clinical term codes. Keratoacanthomas and SCC in situ SNOWMED codes were not included. SCC in situ results was found within the sample analysis and was offset by including one SCC per annum per person. Conclusions: The rates of cSCC are far higher than previously reported and demand a reappraisal of our national management of this disease.
... The relevance of invasive cSCC is denoted by a range of twice the incidence of melanoma in European Caucasians of up to 10 times in fair-skinned populations in Australia 2 . Recent findings also suggest a sex-biased susceptibility of cSCC highlighting a certain protective role of female immunity 3 . ...
Article
Full-text available
Advanced cutaneous squamous cell carcinoma (cSCC) encompasses unresectable and metastatic disease. Although immune checkpoint inhibition has been approved for this entity recently, a considerable proportion of cases is associated with significant morbidity and mortality. Clinical, histopathological, and radiological criteria are used for current diagnostics, classification, and therapeutic decision-making. The identification of complex molecular biomarkers to accurately stratify patients is a not yet accomplished requirement to further shift current diagnostics and care to a personalized precision medicine. This article highlights new insights into the mutational profile of cSCC, summarizes current diagnostic and therapeutic standards, and discusses emerging diagnostic approaches with emphasis on liquid biopsy and tumor tissue-based analyses.
... measures, in this study only 1 female sample was sequenced. Others have highlighted that age, 203 male sex and immunosuppression are among risk factors for metastasis [32] while another study 204 reports sex determined immunological mechanisms as a determinant of the development of 205 metastasis in cSCC [33]. Two patients were immunocompromised; one patient was on long-term 206 . ...
Preprint
Full-text available
Metastatic cutaneous squamous cell carcinoma (cSCC) is associated with a high risk of recurrence and poor prognosis. There is limited published data exploring whole genome sequencing (WGS). The aim of this project was to provide the first comprehensive genomic understanding of the state of metastatic cSCC. In this study, we used WGS on matched tumor and blood DNA to detect somatic genetic alterations from 25 patients with regional metastases of head and neck cSCC. Our computational analyses interrogate clinical impacts of these genetic alterations on metastatic cSCC across the cohort for both the coding and non-coding genome. In the non-coding genome, 3’UTR regions of EVC (48%), PPP1R1A (48%) and LUM (16%) were significantly functionally altered (Q-value < 0.05). Further, significant functional alterations are observed in the tumor suppressing lncRNA LINC01003 (68% of specimens, Q-value: 0.0158). In addition, significant recurrent copy number loss in tumor suppressor genes KANSL1 and PTPRD and gain in CALR, CCND1 and FGF3 was observed for coding regions. SNV’s driver analyses predicted TP53, CDKN2A , as potential drivers of the metastasis cSCC (using 3 different tools). Indel signature analysis highlight dominance of ID signature 13 followed by ID8 & ID9. Interestingly, ID 9 has previously been shown to have no association with skin melanoma, unlike ID 13 and 8, suggesting some point of difference between these two skin-based diseases. The overall landscape of variation in metastatic cSCC is dominated by cell cycle and DNA repair disruption.
Article
Background: Cutaneous squamous cell carcinoma (cSCC) is a common skin cancer, affecting more than 2 million people worldwide yearly and metastasising in 2-5% of patients. However, current clinical staging systems do not provide estimates of absolute metastatic risk, hence missing the opportunity for more personalised treatment advice. We aimed to develop a clinico-pathological model that predicts the probability of metastasis in patients with cSCC. Methods: Nationwide cohorts from (1) all patients with a first primary cSCC in The Netherlands in 2007-2008 and (2) all patients with a cSCC in 2013-2015 in England were used to derive nested case-control cohorts. Pathology records of primary cSCCs that originated a loco-regional or distant metastasis were identified, and these cSCCs were matched to primary cSCCs of controls without metastasis (1:1 ratio). The model was developed on the Dutch cohort (n = 390) using a weighted Cox regression model with backward selection and validated on the English cohort (n = 696). Model performance was assessed using weighted versions of the C-index, calibration metrics, and decision curve analysis; and compared to the Brigham and Women's Hospital (BWH) and the American Joint Committee on Cancer (AJCC) staging systems. Members of the multidisciplinary Skin Cancer Outcomes (SCOUT) consortium were surveyed to interpret metastatic risk cutoffs in a clinical context. Findings: Eight out of eleven clinico-pathological variables were selected. The model showed good discriminative ability, with an optimism-corrected C-index of 0.80 (95% Confidence interval (CI) 0.75-0.85) in the development cohort and a C-index of 0.84 (95% CI 0.81-0.87) in the validation cohort. Model predictions were well-calibrated: the calibration slope was 0.96 (95% CI 0.76-1.16) in the validation cohort. Decision curve analysis showed improved net benefit compared to current staging systems, particularly for thresholds relevant for decisions on follow-up and adjuvant treatment. The model is available as an online web-based calculator (https://emc-dermatology.shinyapps.io/cscc-abs-met-risk/). Interpretation: This validated model assigns personalised metastatic risk predictions to patients with cSCC, using routinely reported histological and patient-specific risk factors. The model can empower clinicians and healthcare systems in identifying patients with high-risk cSCC and offering personalised care/treatment and follow-up. Use of the model for clinical decision-making in different patient populations must be further investigated. Funding: PPP Allowance made available by Health-Holland, Top Sector Life Sciences & Health, to stimulate public-private partnerships.
Article
Full-text available
Cutaneous squamous cell carcinoma (cSCC) is a disease with globally rising incidence and poor prognosis for patients with advanced or metastatic disease. Epithelial-mesenchymal transition (EMT) is a driver of metastasis in many carcinomas, and cSCC is no exception. We aimed to provide a systematic overview of the clinical and experimental evidence for EMT in cSCC, with critical appraisal of type and quality of the methodology used. We then used this information as rationale for potential drug targets against advanced and metastatic cSCC. All primary literature encompassing clinical and cell-based or xenograft experimental studies reporting on the role of EMT markers or related signalling pathways in the progression of cSCC were considered. A screen of 3443 search results yielded 86 eligible studies comprising 44 experimental studies, 22 clinical studies, and 20 studies integrating both. From the clinical studies a timeline illustrating the alteration of EMT markers and related signalling was evident based on clinical progression of the disease. The experimental studies reveal connections of EMT with a multitude of factors such as genetic disorders, cancer-associated fibroblasts, and matrix remodelling via matrix metalloproteinases and urokinase plasminogen activator. Additionally, EMT was found to be closely tied to environmental factors as well as to stemness in cSCC via NFκB and β-catenin. We conclude that the canonical EGFR, canonical TGF-βR, PI3K/AKT and NFκB signalling are the four signalling pillars that induce EMT in cSCC and could be valuable therapeutic targets. Despite the complexity, EMT markers and pathways are desirable biomarkers and drug targets for the treatment of advanced or metastatic cSCC. Graphical Abstract
Article
Full-text available
We use deep transfer learning to quantify histopathological patterns across 17,355 hematoxylin and eosin-stained histopathology slide images from 28 cancer types and correlate these with matched genomic, transcriptomic and survival data. This approach accurately classifies cancer types and provides spatially resolved tumor and normal tissue distinction. Automatically learned computational histopathological features correlate with a large range of recurrent genetic aberrations across cancer types. This includes whole-genome duplications, which display universal features across cancer types, individual chromosomal aneuploidies, focal amplifications and deletions, as well as driver gene mutations. There are widespread associations between bulk gene expression levels and histopathology, which reflect tumor composition and enable the localization of transcriptomically defined tumor-infiltrating lymphocytes. Computational histopathology augments prognosis based on histopathological subtyping and grading, and highlights prognostically relevant areas such as necrosis or lymphocytic aggregates. These findings show the remarkable potential of computer vision in characterizing the molecular basis of tumor histopathology. Two papers by Kather and colleagues and Gerstung and colleagues develop workflows to predict a wide range of molecular alterations from pan-cancer digital pathology slides.
Article
Full-text available
To define the cellular composition and architecture of cutaneous squamous cell carcinoma (cSCC), we combined single-cell RNA sequencing with spatial transcriptomics and multiplexed ion beam imaging from a series of human cSCCs and matched normal skin. cSCC exhibited four tumor subpopulations, three recapitulating normal epidermal states, and a tumor-specific keratinocyte (TSK) population unique to cancer, which localized to a fibrovascular niche. Integration of single-cell and spatial data mapped ligand-receptor networks to specific cell types, revealing TSK cells as a hub for intercellular communication. Multiple features of potential immunosuppression were observed, including T regulatory cell (Treg) co-localization with CD8 T cells in compartmentalized tumor stroma. Finally, single-cell characterization of human tumor xenografts and in vivo CRISPR screens identified essential roles for specific tumor subpopulation-enriched gene networks in tumorigenesis. These data define cSCC tumor and stromal cell subpopulations, the spatial niches where they interact, and the communicating gene networks that they engage in cancer.
Article
Full-text available
p16INK4a (CDKN2A) is a central tumor suppressor, which induces cell-cycle arrest and senescence. Cells expressing p16INK4a accumulate in aging tissues and appear in premalignant lesions, yet their physiologic effects are poorly understood. We found that prolonged expression of transgenic p16INK4a in the mouse epidermis induces hyperplasia and dysplasia, involving high proliferation rates of keratinocytes not expressing the transgene. Continuous p16INK4a expression increases the number of epidermal papillomas formed after carcinogen treatment. Wnt-pathway ligands and targets are activated upon prolonged p16INK4a expression, and Wnt inhibition suppresses p16INK4a-induced hyperplasia. Senolytic treatment reduces p16INK4a-expressing cell numbers, and inhibits Wnt activation and hyperplasia. In human actinic keratosis, a precursor of squamous cell carcinoma, p16INK4a-expressing cells are found adjacent to dividing cells, consistent with paracrine interaction. These findings reveal that chronic p16INK4a expression is sufficient to induce hyperplasia through Wnt-mediated paracrine stimulation, and suggest that this tumor suppressor can promote early premalignant epidermal lesion formation.
Article
Full-text available
Background Many cancer types display sex and age disparity in incidence and outcome. The mutational load of tumours, including melanoma, varies according to sex and age. However, there are no tools to systematically explore if clinical variables such as age and sex determine the genomic landscape of cancer. Objectives To establish a mathematical approach using melanoma mutational data to analyze how sex and age shape the tumour genome. Methods We model how age‐related (clock‐like) somatic mutations that arise during cell division, and extrinsic (environmental ultraviolet light) mutations accumulate in cancer genomes. Results Melanoma is primarily driven by cell‐intrinsic age‐related mutations and extrinsic ultraviolet light‐induced mutations, and we show these mutation types differ in magnitude, chronology and by sex in the distinct molecular melanoma subtypes. Our model confirms age and sex are determinants of cellular mutation rate, shaping the final mutation composition. We show mathematically for the first time how, similar to non‐cancer tissues, melanoma genomes reflect a decline in cell division during ageing. We find clock‐like mutations strongly correlate with the acquisition of ultraviolet light‐induced mutations, but critically, males present a higher number and rate of cell division‐linked mutations. Conclusions These data indicate the contribution of environmental damage to melanoma likely extends beyond genetic damage to affect cell division. Sex and age determine the final mutational composition of melanoma.
Article
Full-text available
Immune checkpoint blockade (ICB)-based or natural cancer immune responses largely eliminate tumours. Yet, they require additional mechanisms to arrest those cancer cells that are not rejected. Cytokine-induced senescence (CIS) can stably arrest cancer cells, suggesting that interferon-dependent induction of senescence-inducing cell cycle regulators is needed to control those cancer cells that escape from killing. Here we report in two different cancers sensitive to T cell-mediated rejection, that deletion of the senescence-inducing cell cycle regulators p16Ink4a/p19Arf (Cdkn2a) or p21Cip1 (Cdkn1a) in the tumour cells abrogates both the natural and the ICB-induced cancer immune control. Also in humans, melanoma metastases that progressed rapidly during ICB have losses of senescence-inducing genes and amplifications of senescence inhibitors. Metastatic cells also resist CIS. Such genetic and functional alterations are infrequent in metastatic melanomas regressing during ICB. Thus, activation of tumour-intrinsic, senescence-inducing cell cycle regulators is required to stably arrest cancer cells that escape from eradication.
Article
Full-text available
Background: Population-based incidence and mortality studies of cutaneous squamous cell carcinoma (SCC) have been few owing to the commonness of the disease, and rare deaths making accurate mortality statistics difficult. Objectives: Our aim was to summarize SCC incidence and mortality in populations across three continents, exemplified by Australia, the United States (US) and Germany. Methods: We estimated age-specific and age-standardized (Australian Standard 2001 Population) incidence and mortality rates per 100 000 person-years. Results: Squamous cell carcinoma incidence is plateauing or falling in Australia, stable in the United States (2013-2015) and rising in Germany (2007-2015). Current incidence estimates in men and women are 341 and 209, 497 and 296, and 54 and 26, respectively, for the three countries. Incidence increases strongly with age in all countries. Mortality of non-melanoma skin cancer appears to be increasing in Germany and stable in Australia (unavailable for the US population). Conclusions: Squamous cell carcinoma is an important health issue, particularly among older men, with incidence exceeding most other cancers. More precise and uniform population-based studies of incidence and mortality are needed to better quantify the impact of SCC on healthcare systems worldwide and to gauge the effect of new treatments such as anti-PD1 therapy on mortality.
Article
Full-text available
Although aging is a conserved phenomenon across evolutionary distant species, aspects of the aging process have been found to differ between males and females of the same species. Indeed, observations across mammalian studies have revealed the existence of longevity and health disparities between sexes, including in humans (i.e. with a female or male advantage). However, the underlying mechanisms for these sex differences in health and lifespan remain poorly understood, and it is unclear which aspects of this dimorphism stem from hormonal differences (i.e. predominance of estrogens vs. androgens) or from karyotypic differences (i.e. XX vs. XY sex chromosome complement). In this review, we discuss the state of the knowledge in terms of sex dimorphism in various aspects of aging and in human age-related diseases. Where the interplay between sex differences and age-related differences has not been explored fully, we present the state of the field to highlight important future research directions. We also discuss various dietary, drug or genetic interventions that were shown to improve longevity in a sex-dimorphic fashion. Finally, emerging tools and models that can be leveraged to decipher the mechanisms underlying sex differences in aging are also briefly discussed.
Article
Cellular senescence is implicated in physiological and pathological processes spanning development, wound healing, age-related decline in organ functions and cancer. Here, we discuss cell-autonomous and non-cell-autonomous properties of senescence in the context of tumour formation and anticancer therapy, and characterize these properties, such as reprogramming into stemness, tissue remodelling and immune crosstalk, as far more dynamic than suggested by the common view of senescence as an irreversible, static condition.
Article
Importance Cutaneous squamous cell carcinoma (cSCC) is the most common skin cancer with metastatic potential, but epidemiologic data are poor. Changes to the National Cancer Registration and Analysis Service (NCRAS) in England have allowed more accurate data analysis of primary and metastatic cSCC since 2013. Objective To assess the national incidence of cSCC and metastatic cSCC (mcSCC) in England from 2013 through 2015. Design, Setting, and Participants This national population-based study identified a cohort of patients with cSCC and mcSCC in England from January 1, 2013, through December 31, 2015. Patients were identified using diagnostic codes derived from pathology reports in the NCRAS. Data were analyzed from March 1, 2017, through March 1, 2018. Main Outcomes and Measures Incidence rates across sex and risk factors for cSCC were derived from the NCRAS data. Risk of occurrence of mcSCC among the population with cSCC was assessed with Cox proportional hazards regression analysis to determine indicators of mcSCC. Results Among the 76 977 patients with first primary cSCC in 2013 through 2015 (62.7% male; median age, 80 years [interquartile range, 72-86 years]), the age-standardized rates for the first registered cSCC in England from 2013 through 2015 were 77.3 per 100 000 person-years (PY) (95% CI, 76.6-78.0) in male patients and 34.1 per 100 000 PY (95% CI, 33.7-34.5) in female patients. Increased primary cSCC tumor count was observed in older, white male patients in lower deprivation quintiles. After a maximum follow-up of 36 months, cumulative incidence of mcSCC developed in 1.1% of women and 2.4% of men with a primary cSCC. Significant increases in the risk of metastasis with adjusted hazard rates of approximately 2.00 were observed in patients who were aged 80 to 89 years (hazard ratio [HR], 1.23; 95% CI, 1.07-1.43), 90 years or older (HR, 1.35; 95% CI, 1.09-1.66), male (HR, 1.79; 95% CI, 1.52-2.10), immunosuppressed (HR, 1.99; 95% CI, 1.64-2.42), and in higher deprivation quintiles (HR for highest quintile, 1.64; 95% CI, 1.35-2.00). Primary cSCC located on the ear (HR, 1.70; 95% CI, 1.42-2.03) and lip (HR, 1.85; 95% CI, 1.29-2.63) were at highest risk of metastasis. Conclusions and Relevance This study presents the first national study of the incidence of mcSCC. With limited health care resources and an aging population, accurate epidemiologic data are essential for informing future health care planning, identifying high-risk patients, and evaluating skin cancer prevention policies.
Article
Understanding sex differences in colon cancer is essential to advance disease prevention, diagnosis, and treatment. Males have a higher risk of developing colon cancer and a lower survival rate than women. However, the molecular features that drive these sex differences are poorly understood. In this study, we use both transcript-based and gene regulatory network methods to analyze RNA-seq data from The Cancer Genome Atlas for 445 patients with colon cancer. We compared gene expression between tumors in men and women and observed significant sex differences in sex chromosome genes only. We then inferred patient-specific gene regulatory networks and found significant regulatory differences between males and females, with drug and xenobiotics metabolism via cytochrome P450 pathways more strongly targeted in females. This finding was validated in a dataset of 1,193 patients from five independent studies. While targeting, the drug metabolism pathway did not change overall survival for males treated with adjuvant chemotherapy, females with greater targeting showed an increase in 10-year overall survival probability, 89% [95% confidence interval (CI), 78-100] survival compared with 61% (95% CI, 45-82) for women with lower targeting, respectively (P = 0.034). Our network analysis uncovers patterns of transcriptional regulation that differentiate male and female colon cancer and identifies differences in regulatory processes involving the drug metabolism pathway associated with survival in women who receive adjuvant chemotherapy. This approach can be used to investigate the molecular features that drive sex differences in other cancers and complex diseases.Significance: A network-based approach reveals that sex-specific patterns of gene targeting by transcriptional regulators are associated with survival outcome in colon cancer. This approach can be used to understand how sex influences progression and response to therapies in other cancers. Cancer Res; 78(19); 5538-47.