Skip to main content
Endocrinology logoLink to Endocrinology
. 2021 Aug 17;162(11):bqab170. doi: 10.1210/endocr/bqab170

Sex Differences in Cancer Genomes: Much Learned, More Unknown

Chenghao Zhu 1,2,3,4, Paul C Boutros 1,2,3,4,
PMCID: PMC8439393  PMID: 34402895

Abstract

Cancer is a leading cause of death worldwide. Sex influences cancer in a bewildering variety of ways. In some cancer types, it affects prevalence; in others, genomic profiles, response to treatment, or mortality. In some, sex seems to have little or no influence. How and when sex influences cancer initiation and progression remain a critical gap in our understanding of cancer, with direct relevance to precision medicine. Here, we note several factors that complicate our understanding of sex differences: representativeness of large cohorts, confounding with features such as ancestry, age, obesity, and variability in clinical presentation. We summarize the key resources available to study molecular sex differences and suggest some likely directions for improving our understanding of how patient sex influences cancer behavior.

Keywords: cancer, sex difference, genomics, cancer genomics


Cancer is a leading cause of death worldwide, especially in more developed countries and regions (1). It is a group of diseases categorized largely on the basis of the organ and cell-type within which they arise. This leads to substantial heterogeneity between and within cancer types. Some cancer types, such as pancreatic and brain cancers, have a very low survival rate. Other cancer types, like prostate and thyroid cancers, have very good outcomes (2). Cancers ultimately arise from the activation of oncogenes and deactivation of tumor suppressor genes. These can occur through multiple mechanisms: some can be inherited, and others are generated through somatic mutational processes (3). Point mutations and chromosomal translocations can alter the amino acid sequence and protein structure of the protein encoded by proto-oncogenes, activating them (4). Amplifications and translocations in certain situations can also affect the RNA and protein abundance of proto-oncogenes (4). Many cancer genomic studies have been conducted over the past 2 decades, and these have started to quantify the heterogeneity in genomic landscapes between and within cancer types, identifying many previously unrecognized cancer subtypes (5).

In a largely independent line of research, epidemiological studies have uncovered a large number of variables associated with risk of a cancer diagnosis, including sex, age, obesity, and others (6-9). This information can be useful in developing screening protocols or in designing interventions to reduce disease incidence with preventative strategies. It also provides insights for new therapeutic targets because subpopulations with altered cancer risks often have unique genomic alteration patterns. Sex is one of the variables that has a large impact on cancer risk. The prevalence and mortality of cancer in men is higher than women for 9 of the 10 most commonly diagnosed cancers in the United States; thyroid cancer is the sole exception (2). The combined cancer mortality is higher in men than women in most countries and regions of the world, with many African regions and Micronesia/Polynesia as notable exceptions (1).

We start by summarizing the current resources available for research into sex differences in cancer genomics. Next, we review the key limitations to the knowledge we have derived from studies of those resources. Finally, we suggest some likely directions for improving our understanding of how patient sex influences the molecular behavior of cancer.

Is Big Data Big Enough?

A series of multi-institutional cancer genomics projects, some completed and some ongoing, have generated tremendous resources. They are being broadly used by the community to generate novel hypotheses about cancer and its behavior in specific groups of patients. The Cancer Genome Atlas (TCGA) included more than 11 000 primary cancers and paired normal tissue samples derived from cancers of 33 types (10,11). The International Cancer Genome Consortium (ICGC) is an ongoing project with teams in the United States, Canada, European, Asia, and Australia—including TCGA projects. With the underlying idea that no single country can cover all cancer types in depth, ICGC brings together scientists in the world and set a target to study 50 cancer types, with at least 500 patients of each cancer category in its first phase (12). To understand the genomic features and abnormalities across cancer types, the Pan-cancer Analysis of Whole Genomes (PCAWG) project included 2658 tumor samples with whole-genome sequencing data from TCGA and ICGC and preformed comprehensive analysis (13). The Therapeutically Applicable Research to Generate Effective Treatments (TARGET) initiative is aiming at understanding the molecular characteristics of pediatric cancers and currently has studied over 6000 children and adolescents at the time of this writing (14). With the decreased price of sequencing technology, genomic profiling is becoming a routine procedure during diagnosis and decision-making of treatments. The Genomics, Evidence, Neoplasia, Information, Exchange (GENIE) project of American Association for Cancer Research comprises genomic profiling data from over 40 000 clinical patients contributed by 8 institutes from North America and Europe (15). Proteomics is largely absent from the previously mentioned projects. To complement them, the US Clinical Proteome Tumour Analysis Consortium has thus far performed comprehensive proteomic and genomic analysis on over 1300 patients with 12 cancer types, with ongoing efforts to expand this (16). Its international counterpart, the International Cancer Proteome Consortium is rapidly growing.

The proportions of the two main biological sexes (defined by chromosome conformation: XX vs XY) in the previously mentioned major cancer genomic projects are relatively representative of the sex distribution of cancer incidence. The American Cancer Society estimates the cancer incidence and mortality in a yearly bases (2). It was estimated that there will be 1 898 160 total cases of all cancers in the United States in 2021, of which 51.1% (970 250) will arise in males and 48.9% (927 910) will arise in females (2). Excluding cancers arising in the reproductive systems and in breast tissue, there are projected to be 707 390 (57.2%) arising in men and 529 600 (42.8%) arising in women. The sex ratio of cases studied in the previously noted projects are all relatively close to this estimate (Table 1).

Table 1.

Patient number in total and by sex in major cancer genomic projects

Project All cancers Cancers not arising in sex organsa
Both sexes Male, n (%) Female, n (%) Both sexes Male, n (%) Female, n (%)
TCGA 11 167 5346 (47.9) 5817 (52.1) 7906 4699 (59.4) 3203 (40.5)
ICGC 24 289 13 135 (54.1) 10 856 (44.7) 19 301 11 700 (60.6) 7303 (37.8)
TARGET 4866 2650 (54.5) 2177 (44.7) 4718 2568 (54.4) 2111 (44.7)
GENIE 44 756 20 713 (46.3) 24 025 (53.7) 32 512 17 991 (55.3) 14 510 (44.6)
CPTAC 1121 507 (45.2) 602 (53.7) 794 507 (63.9) 287 (36.1)

Abbreviations: CPTAC, Clinical Proteome Tumour Analysis Consortium; GENIE, Genomics, Evidence, Neoplasia, Information, Exchange; ICGC, International Cancer Genome Consortium; TARGET, Therapeutically Applicable Research to Generate Effective Treatments; TCGA, The Cancer Genome Atlas.

aAll cancer types except those originating in reproductive organs (eg, prostate, uterine, ovary) or in the breast.

Surprisingly, despite this relatively balanced sex representation, even the largest public genomic databases provide very modest statistical power. For individual cancer types, excluding cancers of the breast and reproductive organs, the median number of patients in TCGA is 231. ICGC has similar numbers: a median of 205 patients in each study. These numbers are quite modest when one considers the large number of confounding variables at play. Cancers vary in their histopathological and clinical features—stage, grade, subtype, treatment status, etc. Patients vary dramatically in their epidemiologic and lifestyle characteristics—age (as seen in breast and prostate cancer), ancestry, smoking history, obesity (as seen in multiple myeloma, colorectal carcinoma, thyroid carcinoma, carcinoma of the pancreas), mutagen exposure (such as exposure to radioactive substances, X-rays, ultraviolet radiation), exercise and diet patterns, and so forth (17-19).

Controlling for these large numbers of confounders is extremely challenging with current cohort sizes. But the situation is further complicated by the relatively low frequency of many somatic drivers and other mutational features. Many specific mutations only occur in 15% to 30% of patients. Consider a large cohort of 500 tumors arising in men and 500 arising in women; this is much larger than a typical ICGC or TCGA data set. For a driver mutation that occurs in 20% and 30% of male- and female-derived tumors, respectively; that means 100 to 150 patients with the mutation would be available. However statistical tests need to be adjusted for at least a dozen potential confounders, such as tumor stage. Finally, because several hundred driver mutations are now known (20,21), discovery studies incur large multiple hypothesis testing penalties, further reducing statistical power. Thus, differences of sex in cancer genomic abnormalities are highly diverse in different cancer types, strongly influenced by other clinical, epidemiologic, and molecular features, and thus even the largest sample sizes available today remain a critical limitation.

Heterogeneity of Cancer Types in Sex Differences

Men have higher incidence and mortality compared to women in many cancer types, such as colon, lung, liver, and gastric cancer (2). A small number of cancer types show the inverse trend. Thyroid cancer is 2.9 times more frequent in women than in men in general; however, in more aggressive histological thyroid cancer subtypes (eg, anaplastic and medullary thyroid cancer), disease prevalence is comparable between the sexes (22). Gallbladder cancer is 2 to 6 more frequent in females than in males (23). The estimated incidence and mortality of the most popular cancer subtypes in men and women in United States of 2021 are shown in Table 2 [data adopted from American Cancer Society’s estimates (2)].

Table 2.

Estimated incidence and mortality of 10 most common cancer sites/tissues of nonreproductive and nonbreast organs/tissues in men and women in United States, 2021

Site/tissue Incidence, n Mortality, n
Male Female Male Female
Lung and bronchus 119 100 116 660 69 410 62 470
Colon and rectum 79 520 69 980 28 520 24 460
Melanoma of the skin 62 260 43 850 4600 2 580
Urinary bladder 64 280 19 450 12 260 4 940
Non-Hodgkin lymphoma 45 630 35 930 12 170 8 550
Kidney and renal pelvis 48 780 27 300 8790 4 990
Leukemia 35 530 25 560 13 900 9 760
Pancreas 31 950 28 480 25 270 22 950
Oral cavity and pharynx 38 800 15 210 7620 3230
Thyroid 12 150 32 130 1050 1150

Data adapted from Siegel et al (2). With permission from American Cancer Society, New York.

This variability in the clinical manifestations of sex differences between cancer types is mirrored in the cancer genome. Somatic mutation density is elevated in tumors arising in males from stomach and esophageal cancer, hepatocellular carcinoma, bladder urothelial carcinoma, melanoma, and kidney renal papillary cell carcinoma; by contrast, it is elevated in glioblastomas (GBMs) arising in females (24,25). The proportion of the genome with a copy number aberration (PGA) is a measure of total copy number burden in a tumor genome and is a surrogate for genomic instability (26). PGA was higher in tumors arising in males from stomach and esophageal cancers, head and neck squamous cell tumors (HNSCs), and kidney renal clear cell carcinomas. PGA was higher in sarcomas and hepatocellular carcinomas arising in females (24,25).

A trio of comprehensive pan-cancer analyses have been done to evaluate the sex differences in cancer genome using TCGA and PCAWG data (24,25,27). Table 3 summarizes key findings from these 3 studies. Two of the studies used multistage multivariate modeling to explicitly control for clinico-epidemiologic features, while the third used propensity score modeling. In all cases, this multivariate modeling implicitly reduced statistical power to help reduce false positives caused by chance unequal distributions of tumors across various covariates. Nevertheless, despite relatively low statistical power that was variable across cancer types, numerous sex differences were detected. Bladder, skin cutaneous melanoma, kidney papillary, hepatocellular carcinoma, stomach and esophageal cancers (STES), and kidney clear cell tumors all had higher somatic single-nucleotide variant (SNV) density in tumors arising in males, while GBM had higher SNV density when arising in females (24). Stomach and esophageal cancers, HNSC, and kidney clear cell tumors had higher PGA in tumors arising in males, while sarcoma and hepatocellular carcinoma had higher PGA in tumors arising in females. Microsatellite instability is a marker for impaired DNA mismatch repair and is observed in numerous cancer subtypes. It was found that in colon cancer and STES, tumors arising in females had higher microsatellite instability frequency than those arising in males (24).

Table 3.

Summary of genomic features with sex differences identified in TCGA and PCAWG

Cancer subtype Mutation density PGA Genes with sex-different SNV
Bladder urothelial Higher in male
Melanoma Higher in male
Liver hepatocellular Higher in male CTNNB1, BAP1
Glioblastoma Higher in female
Stomach and esophageal Higher in male (MSI adjusted) Higher in male PGM5, GTF3C1, LTBP1, MEGF8, RNF213, EP400, OBSCN, ZFHX3, SLIT2, ZBTB20
Head and neck Higher in male
Kidney renal cell Higher in male BAP1
Sarcoma Higher in female
Thyroid Higher inmale (marginal) TERT
Kidney renal papillary cell TTN
Long adenocarcinoma STK11, SMG1, CNTN5, ZNF521, COL21A1, RBM10, ABCB5, FAM47A, DMD, F8, MED12

Data adapted from Li et al (24,25) and Yuan et al (27) with permission from Elsevier.

Abbreviation: MSI, microsatellite instability; PACWG, Pan-cancer Analysis of Whole Genomes; PGA, proportion of the genome with a copy number aberration; SNV, single-nucleotide variant; TCGA, The Cancer Genome Atlas.

Some cancer types have not displayed significant sex-differences in these large surveys. In acute myeloid leukemia, low-grade glioma (LGG) and pheochromocytoma and paraganglioma, none of the genomic alterations tested were significantly different between tumors arising in males and those arising in females—these included somatic SNV burden, PGA, and specific driver genes (25,27). Interestingly, men also have a higher risk of diagnosis for cancer types without genomic sex-differences such as acute myeloid leukemia and LGG (28,29).

Seeing Through the Variability: Known Sex Differences

Using data collected by the SEER (Surveillance, Epidemiology, and End Results) program has shown that across the 10 most common nonsex-specific cancers, men had more advanced stage disease at diagnosis than women in most. These included lung, melanoma, non-Hodgkin lymphoma, and kidney, thyroid, pancreatic and liver cancer but not bladder, colorectal, and brain cancer (30). In lung cancer, women were more likely to have adenocarcinoma, and men, squamous-cell carcinoma (30). Data collected in England also showed a slightly higher proportion of men diagnosed with stage 1 colorectal cancer and women with stage 2 (31). The same study also observed that the anatomical site tended to be higher (eg, caecum, ascending, transverse colon) in women, and lower (eg, sigmoid colon and rectum) in men (31). It is unclear whether these differences primarily reflect differences in health-seeking behavior, in other lifestyle or social characteristics, in the underlying molecular biology of the tumor, or some other features.

As previously noted, many factors contribute to the tumor development and progression, and could influence our understanding of the sex-difference in cancer genomics. Cancer prevalence and genomic features are associated with ethnic and geographic diversity. In TCGA data, the genome instability of African American patients was higher in breast (BRCA), head and neck and endometrial cancers [uterine corpus endometrial carcinoma (UCEC)], but lower in patients with kidney cancers (32). The elevated genomic instability of African ancestry in BRCA, HNSC, and UCEC was also associated with increased TP53 mutation and CCNE1 amplification (32). Similarly, another study observed correlations between African ancestry and aneuploidy and East Asian ancestry and mutation load (33). Another recent study integrated TCGA and PCAWG data and observed that African ancestry was associated with higher PGA (proportion of genome with a copy number alteration) in HNSC and UCEC, while East Asian ancestry was associated with higher PGA in hepatocellular carcinoma and STES (34). East Asian and African ancestry were also associated with lower SNV density than European ancestry in melanoma (34). It is yet difficult to assess the race disparities in large cancer genomic studies such as TCGA and ICGC. In TCGA studies, somatic mutations with mutational frequency below 10% are not detectable in most cancer types for most non-Caucasian ancestry groups (35). The underrepresentativeness of nonwhite patients makes it difficult to study the genomic disparities in sex while trying to adjust for effects of ancestry. Thus, our understanding of sex differences in cancer will remain at least partially incomplete and uncertain until there is more equal representation in our databases of genomic profiles of patients of all ancestries.

In a similar way, age is associated with the evolution and somatic mutation features of cancer and might interact with sex differences. In TCGA data, genome instability increased with age at both the pan-cancer level as well as in specific cancer types including LGG, ovarian cancer, UCEC and sarcoma (36). Somatic point mutation burden also increased with age at the pan-cancer level and in 18 of 33 TCGA cancer types (36). Driver gene mutations such as mutations in IDH1 and ATRX were negatively associated with age, while PIK3CA were positively associated with age in pan-cancer data (36). These trends were confirmed in ICGC PCAWG whole-genome sequencing data (37). In addition, a positive association between clonal SNVs and indels were also observed pan-cancer and in 2 specific cancer types: medulloblastoma and melanoma (37). Several mutational signatures were associated with age, suggesting different exogenous and endogenous mutational oncogenic processes in older patients relative to younger ones (37). These data highlight that age significantly affects the somatic mutational evolution of cancer and needs to be considered in the context of sex differences.

Besides innate and inheritable factors such as race and age, environmental factors also contribute to cancer risks and are associated with specific patterns of genomic abnormalities. It is well known that smoking and alcohol consumption are associated with mutation rates and types in multiple cancer types such as lung, colon, and oral cancer (38-41). The consumption of tobacco and alcohol itself also shows sex differences, as both smoking and alcohol exposure was higher in men than women in lung and liver cancers (42,43). Diet is another important environmental factor that has a strong association to cancer, but one that is not yet well understood. A study in the Netherlands reported that dietary heme consumption is strongly associated with KRAS and APC mutations in colorectal cancers arising in males but not in those arising in females (44). The same team reported that dietary folate intake was associated with a decreased risk for rectal cancer in men, particularly for patients carrying KRAS point mutations. By contrast, it showed an association with increased risk in female patients independent of KRAS mutation status (45). Another study reported that fish oil consumption was significant associated with a lower risk for colorectal cancer in men but not in women (46). Similarly, dietary fiber intake was associated with a lower risk for recurrence of colorectal adenoma in men but not women (47). Those findings indicated potential sex differences in diet-mutation interactions in cancer that will need to be extensively studied moving forward. Unfortunately, dietary profiling for patients with sequenced cancer genomes has not been a major focus to date. The lack of dietary data in large cancer genomic projects such as TCGA and ICGC makes it almost impossible to achieve a comprehensive understanding of the relationship between diet and cancer genome abnormality. Similarly, physical exercise has protective effect in several caner types, especially in breast and colon cancer (48,49) and has a positive effect when undertaken with or after treatment (50). Growing evidence suggests that exercise alters DNA methylations in patients with different cancer types (51). Obesity, which can be the consequence of unhealthy dietary patterns (high calorie, high fat, high refined sugar, and low fiber), is associated with cancer in a complex way. While obesity is associated with a higher risk for many cancer subtypes (52-54), it was observed that overweight and obese patients experienced statistically significantly higher survival in renal cell cancer, colorectal cancer, and acute myeloid leukemia (55-57). This obesity paradox was also observed in other diseases such as cardiovascular disease (58,59), diabetes (60), and chronic kidney disease (61) but not in prostate and breast cancer (62). Importantly, these lifestyle factors of diet, exercise, and obesity themselves vary with an individual’s age. It is unclear whether their impact is most acute in certain age windows—either in absolute time or relative to a specific landmark in tumor evolution such as the first whole-genome duplication.

Thus, a growing body of evidence shows that mutational processes and specific driver somatic mutations are affected by numerous endogenous and exogenous variables. When evaluating sex differences, it is necessary to consider those variables properly. Sex differences in TMN stage, histology, and anatomical site of various cancer highlight the importance of exogenous factors and also suggest societal and behavioral difference in men and women could underlie at least some proportion of sex differences. To highlight the importance of these differences, imagine that tumors arising in men and women differ in either their stage at diagnosis or their age at diagnosis. If stage or age is itself associated with somatic mutational features, then a univariate uncontrolled analysis will reveal sex differences in cancer genomics, while a statistical control for these features will remove them. It is unclear how we might best understand sex differences that disappear after controlling for other clinico-epidemiologic features, when those clinico-epidemiologic features themselves might differ across the sexes in specific cancer types. Effectively communicating complex multivariate models appears to be an increasingly important skillset in cancer genomics.

Sex Differences and the Selective Pressure of Treatment

Beyond the clinical and molecular differences between tumors arising in males and females, there are also sex differences in drug responses. Females have a slower clearance and have a stronger side effects from many chemotherapy drugs (63). For example, clearance of 5-florouracil is slower in females, often leading to elevated toxicity (64-67). This sex disparity can be explained by the decreased activity of dihydropyrimidine dehydrogenase, which is responsible for degrading 5-florouracil (68,69). Female patients with colorectal cancer suffered from more frequent and severe adverse side effects with 5-florouracil treatment, including stomatitis, leukopenia, alopecia, and diarrhea (66). Paclitaxel, another chemotherapy drug, was shown to have a 20% lower elimination rate in women with solid tumors and causes more toxicity in female patients, including leukopenia (70-72). A pharmacokinetic study of normal liver function showed that females have slower doxorubicin clearance than males (73). In patients receiving cisplatin-based chemotherapy, women had a higher incidence of vomiting and nausea than men, and the side effects were also harder to control (74).

Sex differences also exist in responses to immunotherapy. Cancer cells can harbor immunosuppression ability through pathways such as cytotoxic T-lymphocyte protein-4 and programmed death receptor 1 (75). Immune checkpoint inhibitors are drugs that inhibit these pathways and are emerging as a breakthrough in the management of multiple cancer types (75). Monoclonal antibodies targeting immune checkpoint inhibitors (eg, ipilimumab, tremelimumab, nivolumab) have shown survival benefits for several solid cancer types (76-78). A recent meta-analysis that reviewed Phase 3 clinical studies in melanoma, lung cancer, head and neck carcinoma, kidney, stomach and urothelial cancers concluded that men had a greater improvement in overall survival with immune checkpoint inhibitors relative to women, suggesting a better drug response in men (79). Another meta-analysis that included advanced or metastatic patients reported a better overall survival of male melanoma patients on immune checkpoint inhibitors alone or with other drugs (chemotherapy or other immunological compounds); an inconsistent result was found in non-small cell lung carcinoma under the same setting (80). This topic is now the subject of intense investigation (81,82).

Sex Discordance in Tumor Evolution

One hypothesis for the sex-differences in response to immunotherapy is that sex hormones such as estrogen mediate the activity of immune cells and overall immunity that could impact drug response (83). But, more broadly, immunotherapy resistance has been linked to tumor evolution: under the selective pressure of the treatment, tumor lineages evolve fitness to the their local environment (84). Thus sexual dimorphism in cancer evolution may play a role in the drug response to immunotherapy.

Evidence that sex differences in cancer evolution may come from study of metastasis. A pair of studies have reported different metastatic patterns in mem and women. In colon and rectal cancer, men had more frequent liver metastases while women had more frequent lung metastases (85,86). Cancer metastasis is an evolutionary process in which tumor cells must acquire the ability to disseminate from primary tumor, survive in the circulation, and ultimately adhere to and colonize the target organ or tissue (87). It is possible that the difference in metastasis pattern in men and women indicates a sex difference in cancer evolution and mutation timing.

Although differences in cancer evolution in sex are largely unstudied outside of the broad differences in driver gene frequency and mutation density reported previously, some initial steps have been taken by mining PCAWG and TCGA data (25,88). Using PCAWG data, it was found that, in biliary adenocarcinoma, females had more frequent polyclonal tumors than males, while males had more clonal somatic structural variants (25). In esophageal cancer, structural variants are more frequently clonal in male-derived samples than in female-derived ones (25). Sex differences were also seen in mutation signatures, with different proportions of single and double base substitution signatures observed in male and female tumors (25). In another report, female carried more subclonal but not clonal mutations in both GBM and LGG, and this sex dimorphism is associated with more malignant tumors (88). Females also tend to have more clonal CDH18 mutations in GBM, and males tend to have more clonal ATRX mutations in LGG, suggesting an interplay between sex-biased driver acquisition and evolutionary timing.

It remains largely unknown whether the metastatic pattern is different in men and women in other cancer subtypes and in non-Caucasians. It is also largely unexplored whether sex-specific signatures evolve differently in different subtypes of cancer. Treatment serves as a powerful selective pressure upon the population of cancer cells and understand to what extent this selective pressure molecularly influences tumors arising in men and women differently is a major gap in our knowledge.

Perspectives: Next Steps

The rapid development of high-throughput DNA sequencing has accelerated our knowledge of cancer genomes and our ability to detect sex differences in them. Although large clinical cancer genomic studies integrating multiple institutions and countries have tremendously accelerated our knowledge, we still have only a modest understanding of sex differences in primary human cancer. Our understanding of sex differences in relapsing and metastatic tumors is negligible. Each cancer type of ICGC and TCGA studies has ~200 to 500 patients. After stratifying by sex and adjusting for cofounding variables such as age and race, statistical powering is very limited. Existing cohorts are poorly representative of the global cancer burden, being biased toward large surgically resected tumors arising in individuals of Caucasian ancestry. Our understanding the interplay of clinical, histopathologic, and epidemiologic features with molecular sex differences in cancer is in its infancy. Thus, targeted sequencing of key driver genes with larger patient number within each individual cancer types might be necessary next step. Fortunately, much of these data are starting to be collected through the routine adoption of DNA sequencing in clinical care, particularly for locally advanced and metastatic tumors.

As previously noted, environmental factors such as dietary pattern and lifestyle are associated with disease risk in several cancer subtypes, but their association with cancer genomic alteration has not been well characterized: these data are absent in most large studies. Dietary information is commonly collected through self-reporting in epidemiological and clinical studies, for both the short term (24-h dietary recall or 3-day diet record) and the long term (food frequency questionnaire). Although those measurements suffer from a questionable accuracy because of underreporting of energy intake and potentially certain categories of dietary components, we and others still recommend that these be incorporated into human studies to begin to adjust for their cofounding effects (89). The recent rapid emergence of wearable technology in healthcare is bringing new hope to this area. Several pioneering studies have used wearable cameras to record dietary data and successfully reduced dietary underreporting (90,91). The collection of other lifestyle data such as physical activity and sleeping also rely heavily on self-reporting and suffer similar biases (92). Improved collection of these data in large cohorts may become increasingly feasible in the near future.

The development of cancer is an evolutionary process consists of somatic mutations under Darwinian selection pressure. Cancer evolution and mutation timing play very important roles in aspects such as drug resistance and metastasis. To better understand sex difference in cancer genomics, evolution will increasingly need to be central to data analysis. This requires us identifying not only the specific mutations in a tumor but also when each occurred during a tumor’s life history and what endogenous or exogenous mutational process most likely caused each one. Great caution should be taken to ensure data quality in cancer evolution studies, including sequencing multiple regions of the same sample; phased or single-cell sequencing are immensely advantageous but are generally cost-prohibitive for routine clinical use at this time (93). It is also largely unknown which key genomic variants contribute to driving tumors to metastasize from their primary sites, and how sex plays a role during this process. This requires not only that we collect paired samples of primary and metastatic tumors but will also that we specifically model the selective and adaptive response to treatment, which itself might show sex differences.

Sex and age are perhaps the 2 most fundamental “subtypes” of human physiology. Understanding how sex influences cancer genomes will improve our understanding of disease initiation, progression, and response to therapy.

Acknowledgments

Financial Support: This work was supported by the NIH/NCI under award number P30CA016042. This work was supported by an operating grant from the National Cancer Institute Early Detection Research Network (U01CA214194) to P.C.B. This work was supported by the NIH/NCI to P.C.B. (R01CA244729).

Additional Information

Disclosure Statement: P.C.B. sits on the scientific advisory boards of Sage Bionetworks, Intersect Diagnostics Inc. and BioSymetrics Inc. The relationship may not relate to the subject matter of this manuscript.

Data Availability

Data sharing is not applicable to this article as no data sets were generated or analyzed during the current study.

References

  • 1.Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209-249. [DOI] [PubMed] [Google Scholar]
  • 2.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7-33. [DOI] [PubMed] [Google Scholar]
  • 3.Binkley EM, Wiley LA. Cancer genetics. In: Singh AD, Damato BE, eds. Clinical Ophthalmic Oncology: Basic Principles. Springer International; 2019:79-86. [Google Scholar]
  • 4.Kontomanolis EN, Koutras A, Syllaios A, et al. Role of oncogenes and tumor-suppressor genes in carcinogenesis: a review. Anticancer Res. 2020;40(11):6009-6015. [DOI] [PubMed] [Google Scholar]
  • 5.Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499(7457):214-218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Johnson CM, Wei C, Ensor JE, et al. Meta-analyses of colorectal cancer risk factors. Cancer Causes Control. 2013;24(6):1207-1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chuang SC, La Vecchia C, Boffetta P. Liver cancer: descriptive epidemiology and risk factors other than HBV and HCV infection. Cancer Lett. 2009;286(1):9-14. [DOI] [PubMed] [Google Scholar]
  • 8.Chow WH, Dong LM, Devesa SS. Epidemiology and risk factors for kidney cancer. Nat Rev Urol. 2010;7(5):245-257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev. 2014;23(5):700-713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hutter C, Zenklusen JC. The cancer genome atlas: creating lasting value beyond its data. Cell. 2018;173(2):283-285. [DOI] [PubMed] [Google Scholar]
  • 11.Gao GF, Parker JS, Reynolds SM, et al. ; Genomic Data Analysis Network . Before and after: comparison of legacy and harmonized TCGA genomic data commons’ data. Cell Syst. 2019;9(1):24-34.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hudson (Chairperson) TJ, Anderson W, Aretz A, et al. International Cancer Genome Consortium. International network of cancer genome projects. Nature 2010;464(7291): 993-998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis of whole genomes . Nature 2020;578(7793):82-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ma X, Liu Y, Liu Y, et al. Pan-cancer genome and transcriptome analyses of 1699 paediatric leukaemias and solid tumours. Nature. 2018;555(7696):371-376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.AACR Project GENIE Consortium. AACR project GENIE: powering precision medicine through an International Consortium. Cancer Discov 2017;7(8):818-831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Edwards NJ, Oberti M, Thangudu RR, et al. The CPTAC data portal: a resource for cancer proteomics research. J Proteome Res. 2015;14(6):2707-2713. [DOI] [PubMed] [Google Scholar]
  • 17.Arem H, Loftfield E. Cancer epidemiology: a survey of modifiable risk factors for prevention and survivorship. Am J Lifestyle Med. 2018;12(3):200-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yoon HH, Shi Q, Alberts SR, Goldberg RM, Thibodeau SN, Sargent DJ, Sinicrope FA; for the Alliance for Clinical Trials in Oncology . Racial differences in BRAF/KRAS mutation rates and survival in stage III colon cancer patients. J Natl Cancer Inst 2015;107(10):djv186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Shariat SF, Sfakianos JP, Droller MJ, Karakiewicz PI, Meryn S, Bochner BH. The effect of age and gender on bladder cancer: a critical review of the literature. BJU Int. 2010;105(3):300-308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bailey MH, Tokheim C, Porta-Pardo E, et al. ; MC3 Working Group; Cancer Genome Atlas Research Network . Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173(2):371-385.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rheinbay E, Nielsen MM, Abascal F, et al. ; PCAWG Drivers and Functional Interpretation Working Group; PCAWG Structural Variation Working Group; PCAWG Consortium . Analyses of non-coding somatic drivers in 2658 cancer whole genomes. Nature. 2020;578(7793):102-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Rahbari R, Zhang L, Kebebew E. Thyroid cancer gender disparity. Future Oncol. 2010;6(11):1771-1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Konstantinidis IT, Deshpande V, Genevay M, et al. Trends in presentation and survival for gallbladder cancer during a period of more than 4 decades: a single-institution experience. Arch Surg. 2009;144(5):441-447; discussion 447. [DOI] [PubMed] [Google Scholar]
  • 24.Li CH, Haider S, Shiah YJ, Thai K, Boutros PC. Sex differences in cancer driver genes and biomarkers. Cancer Res. 2018;78(19):5527-5537. [DOI] [PubMed] [Google Scholar]
  • 25.Li CH, Prokopec SD, Sun RX, Yousif F, Schmitz N, Boutros PC; PCAWG Tumour Subtypes and Clinical Translation; PCAWG Consortium . Sex differences in oncogenic mutational processes. Nat Commun. 2020;11(1):4330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lalonde E, Ishkanian AS, Sykes J, et al. Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort study. Lancet Oncol. 2014;15(13):1521-1532. [DOI] [PubMed] [Google Scholar]
  • 27.Yuan Y, Liu L, Chen H, et al. Comprehensive characterization of molecular differences in cancer between male and female patients. Cancer Cell. 2016;29(5):711-722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hossain MJ, Xie L. Sex disparity in childhood and young adult acute myeloid leukemia (AML) survival: Evidence from US population data. Cancer Epidemiol. 2015;39(6):892-900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Claus EB, Black PM. Survival rates and patterns of care for patients diagnosed with supratentorial low-grade gliomas: data from the SEER program, 1973-2001. Cancer. 2006;106(6):1358-1363. [DOI] [PubMed] [Google Scholar]
  • 30.Wainer Z, Wright GM, Gough K, et al. Sex-dependent staging in non-small-cell lung cancer; analysis of the effect of sex differences in the eighth edition of the tumor, node, metastases staging system. Clin Lung Cancer. 2018;19(6):e933-e944. [DOI] [PubMed] [Google Scholar]
  • 31.White A, Ironmonger L, Steele RJC, Ormiston-Smith N, Crawford C, Seims A. A review of sex-related differences in colorectal cancer incidence, screening uptake, routes to diagnosis, cancer stage and survival in the UK. BMC Cancer. 2018;18(1):906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yuan J, Hu Z, Mahal BA, et al. Integrated analysis of genetic ancestry and genomic alterations across cancers. Cancer Cell. 2018;34(4):549-560.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Carrot-Zhang J, Chambwe N, Damrauer JS, et al. ; Cancer Genome Atlas Analysis Network . Comprehensive analysis of genetic ancestry and its molecular correlates in cancer. Cancer Cell. 2020;37(5):639-654.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Li CH, Haider S, Boutros PC. Ancestry influences on the molecular presentation of tumours. bioRxiv. Posted October 4, 2020. doi: 10.1101/2020.08.02.233528 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Spratt DE, Chan T, Waldron L, et al. Racial/ethnic disparities in genomic sequencing. JAMA Oncol. 2016;2(8):1070-1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chatsirisupachai K, Lesluyes T, Paraoan L, Van Loo P, de Magalhães JP. An integrative analysis of the age-associated multi-omic landscape across cancers. Nat Commun. 2021;12(1):2345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Li CH, Haider S, Boutros PC. Age influences on the molecular presentation of tumours. bioRxiv. Posted May 24, 2021. doi: 10.1101/2020.07.07.192237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gibbons DL, Byers LA, Kurie JM. Smoking, p53 mutation, and lung cancer. Mol Cancer Res. 2014;12(1):3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Samowitz WS, Albertsen H, Sweeney C, et al. Association of smoking, CpG island methylator phenotype, and V600E BRAF mutations in colon cancer. J Natl Cancer Inst. 2006;98(23):1731-1738. [DOI] [PubMed] [Google Scholar]
  • 40.Proia NK, Paszkiewicz GM, Nasca MA, Franke GE, Pauly JL. Smoking and smokeless tobacco-associated human buccal cell mutations and their association with oral cancer–a review. Cancer Epidemiol Biomarkers Prev. 2006;15(6):1061-1077. [DOI] [PubMed] [Google Scholar]
  • 41.Ahrendt SA, Chow JT, Yang SC, et al. Alcohol consumption and cigarette smoking increase the frequency of p53 mutations in non-small cell lung cancer. Cancer Res. 2000;60(12):3155-3159. [PubMed] [Google Scholar]
  • 42.O’Keeffe LM, Taylor G, Huxley RR, Mitchell P, Woodward M, Peters SAE. Smoking as a risk factor for lung cancer in women and men: a systematic review and meta-analysis. BMJ Open. 2018;8(10):e021611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rehm J, Shield KD, Weiderpass E. Alcohol consumption: a leading risk factor for cancer. Chem Biol Interact. 2020;331:109280. [DOI] [PubMed] [Google Scholar]
  • 44.Gilsing AM, Fransen F, de Kok TM, et al. Dietary heme iron and the risk of colorectal cancer with specific mutations in KRAS and APC. Carcinogenesis. 2013;34(12):2757-2766. [DOI] [PubMed] [Google Scholar]
  • 45.Brink M, Weijenberg MP, de Goeij AF, et al. Dietary folate intake and k-ras mutations in sporadic colon and rectal cancer in the Netherlands Cohort Study. Int J Cancer. 2005;114(5):824-830. [DOI] [PubMed] [Google Scholar]
  • 46.Kantor ED, Lampe JW, Peters U, Vaughan TL, White E. Long-chain omega-3 polyunsaturated fatty acid intake and risk of colorectal cancer. Nutr Cancer. 2014;66(4):716-727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jacobs ET, Lanza E, Alberts DS, et al. Fiber, sex, and colorectal adenoma: results of a pooled analysis. Am J Clin Nutr. 2006;83(2):343-349. [DOI] [PubMed] [Google Scholar]
  • 48.Howard RA, Freedman DM, Park Y, Hollenbeck A, Schatzkin A, Leitzmann MF. Physical activity, sedentary behavior, and the risk of colon and rectal cancer in the NIH-AARP Diet and Health Study. Cancer Causes Control. 2008;19(9):939-953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bernstein L, Patel AV, Ursin G, et al. Lifetime recreational exercise activity and breast cancer risk among black women and white women. J Natl Cancer Inst. 2005;97(22):1671-1679. [DOI] [PubMed] [Google Scholar]
  • 50.Galvão DA, Newton RU. Review of exercise intervention studies in cancer patients. J Clin Oncol. 2005;23(4):899-909. [DOI] [PubMed] [Google Scholar]
  • 51.Voisin S, Eynon N, Yan X, Bishop DJ. Exercise training and DNA methylation in humans. Acta Physiol (Oxf). 2015;213(1):39-59. [DOI] [PubMed] [Google Scholar]
  • 52.Yang P, Zhou Y, Chen B, et al. Overweight, obesity and gastric cancer risk: results from a meta-analysis of cohort studies. Eur J Cancer. 2009;45(16):2867-2873. [DOI] [PubMed] [Google Scholar]
  • 53.Larsson SC, Wolk A. Overweight, obesity and risk of liver cancer: a meta-analysis of cohort studies. Br J Cancer. 2007;97(7):1005-1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Moghaddam AA, Woodward M, Huxley R. Obesity and risk of colorectal cancer: a meta-analysis of 31 studies with 70 000 events. Cancer Epidemiol Biomarkers Prev. 2007;16(12):2533-2547. [DOI] [PubMed] [Google Scholar]
  • 55.Schlesinger S, Siegert S, Koch M, et al. Postdiagnosis body mass index and risk of mortality in colorectal cancer survivors: a prospective study and meta-analysis. Cancer Causes Control. 2014;25(10):1407-1418. [DOI] [PubMed] [Google Scholar]
  • 56.Hakimi AA, Furberg H, Zabor EC, et al. An epidemiologic and genomic investigation into the obesity paradox in renal cell carcinoma. J Natl Cancer Inst. 2013;105(24):1862-1870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Brunner AM, Sadrzadeh H, Feng Y, et al. Association between baseline body mass index and overall survival among patients over age 60 with acute myeloid leukemia. Am J Hematol. 2013;88(8):642-646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Uretsky S, Messerli FH, Bangalore S, et al. Obesity paradox in patients with hypertension and coronary artery disease. Am J Med. 2007;120(10):863-870. [DOI] [PubMed] [Google Scholar]
  • 59.Oreopoulos A, Padwal R, Kalantar-Zadeh K, Fonarow GC, Norris CM, McAlister FA. Body mass index and mortality in heart failure: a meta-analysis. Am Heart J. 2008;156(1):13-22. [DOI] [PubMed] [Google Scholar]
  • 60.Carnethon MR, De Chavez PJ, Biggs ML, et al. Association of weight status with mortality in adults with incident diabetes. Jama. 2012;308(6):581-590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kalantar-Zadeh K, Streja E, Kovesdy CP, et al. The obesity paradox and mortality associated with surrogates of body size and muscle mass in patients receiving hemodialysis. Mayo Clin Proc. 2010;85(11):991-1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Parekh N, Chandran U, Bandera EV. Obesity in cancer survival. Annu Rev Nutr. 2012;32:311-342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kim HI, Lim H, Moon A. Sex differences in cancer: epidemiology, genetics and therapy. Biomol Ther (Seoul). 2018;26(4): 335-342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Milano G, Etienne MC, Cassuto-Viguier E, et al. Influence of sex and age on fluorouracil clearance. J Clin Oncol. 1992;10(7):1171-1175. [DOI] [PubMed] [Google Scholar]
  • 65.Stein BN, Petrelli NJ, Douglass HO, Driscoll DL, Arcangeli G, Meropol NJ. Age and sex are independent predictors of 5-fluorouracil toxicity: analysis of a large scale Phase III trial. Cancer. 1995;75(1):11-17. [DOI] [PubMed] [Google Scholar]
  • 66.Sloan JA, Goldberg RM, Sargent DJ, et al. Women experience greater toxicity with fluorouracil-based chemotherapy for colorectal cancer. J Clin Oncol. 2002;20(6):1491-1498. [DOI] [PubMed] [Google Scholar]
  • 67.Wang J, Huang Y. Pharmacogenomics of sex difference in chemotherapeutic toxicity. Curr Drug Discov Technol. 2007;4(1):59-68. [DOI] [PubMed] [Google Scholar]
  • 68.Etienne MC, Lagrange JL, Dassonville O, et al. Population study of dihydropyrimidine dehydrogenase in cancer patients. J Clin Oncol. 1994;12(11):2248-2253. [DOI] [PubMed] [Google Scholar]
  • 69.Milano G, Etienne MC, Pierrefite V, Barberi-Heyob M, Deporte-Fety R, Renée N. Dihydropyrimidine dehydrogenase deficiency and fluorouracil-related toxicity. Br J Cancer. 1999;79(3-4):627-630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Joerger M, Huitema AD, van den Bongard DH, Schellens JH, Beijnen JH. Quantitative effect of gender, age, liver function, and body size on the population pharmacokinetics of Paclitaxel in patients with solid tumors. Clin Cancer Res. 2006;12(7 Pt 1):2150-2157. [DOI] [PubMed] [Google Scholar]
  • 71.Yamamoto H, Sekine I, Yamada K, et al. Gender differences in treatment outcomes among patients with non-small cell lung cancer given a combination of carboplatin and paclitaxel. Oncology. 2008;75(3-4):169-174. [DOI] [PubMed] [Google Scholar]
  • 72.Schmetzer O, Flörcken A. Sex differences in the drug therapy for oncologic diseases. Handb Exp Pharmacol 2012;214: 411-442. [DOI] [PubMed] [Google Scholar]
  • 73.Dobbs NA, Twelves CJ, Gillies H, James CA, Harper PG, Rubens RD. Gender affects doxorubicin pharmacokinetics in patients with normal liver biochemistry. Cancer Chemother Pharmacol. 1995;36(6):473-476. [DOI] [PubMed] [Google Scholar]
  • 74.Liaw CC, Wang CH, Chang HK, et al. Gender discrepancy observed between chemotherapy-induced emesis and hiccups. Support Care Cancer. 2001;9(6):435-441. [DOI] [PubMed] [Google Scholar]
  • 75.Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):1-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Antonia SJ, Villegas A, Daniel D, et al. ; PACIFIC Investigators . Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 2017;377(20):1919-1929. [DOI] [PubMed] [Google Scholar]
  • 78.Ribas A, Kefford R, Marshall MA, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol. 2013;31(5):616-622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Conforti F, Pala L, Bagnardi V, et al. Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta-analysis. Lancet Oncol. 2018;19(6):737-746. [DOI] [PubMed] [Google Scholar]
  • 80.Ye Y, Jing Y, Li L, et al. Sex-associated molecular differences for cancer immunotherapy. Nat Commun. 2020;11(1):1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wallis CJD, Butaney M, Satkunasivam R, et al. Association of patient sex with efficacy of immune checkpoint inhibitors and overall survival in advanced cancers: a systematic review and meta-analysis. JAMA Oncol. 2019;5(4):529-536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Botticelli A, Onesti CE, Zizzari I, et al. The sexist behaviour of immune checkpoint inhibitors in cancer therapy? Oncotarget. 2017;8(59):99336-99346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Irelli A, Sirufo MM, D’Ugo C, Ginaldi L, De Martinis M. Sex and gender influences on cancer immunotherapy response. Biomedicines 2020;8(7):232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Venkatesan S, Swanton C, Taylor BS, Costello JF. Treatment-induced mutagenesis and selective pressures sculpt cancer evolution. Cold Spring Harb Perspect Med 2017;7(8):a026617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Riihimäki M, Hemminki A, Sundquist J, Hemminki K. Patterns of metastasis in colon and rectal cancer. Sci Rep. 2016;6:29765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Meltzer S, Bakke KM, Rød KL, et al. Sex-related differences in primary metastatic site in rectal cancer; associated with hemodynamic factors? Clin Transl Radiat Oncol. 2020;21:5-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Birkbak NJ, McGranahan N. Cancer genome evolutionary trajectories in metastasis. Cancer Cell. 2020;37(1):8-19. [DOI] [PubMed] [Google Scholar]
  • 88.Cohen EEW, Soulières D, Le Tourneau C, et al. ; KEYNOTE-040 Investigators . Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study. Lancet. 2019;393(10167):156-167. [DOI] [PubMed] [Google Scholar]
  • 89.Subar AF, Freedman LS, Tooze JA, et al. Addressing current criticism regarding the value of self-report dietary data. J Nutr. 2015;145(12):2639-2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gemming L, Rush E, Maddison R, et al. Wearable cameras can reduce dietary under-reporting: doubly labelled water validation of a camera-assisted 24 h recall. Br J Nutr. 2015;113(2):284-291. [DOI] [PubMed] [Google Scholar]
  • 91.Bulungu ALS, Palla L, Priebe J, et al. Validation of a life-logging wearable camera method and the 24-h diet recall method for assessing maternal and child dietary diversity. Br J Nutr. 2021;125(11):1299-1309. [DOI] [PubMed] [Google Scholar]
  • 92.Prince SA, Adamo KB, Hamel ME, Hardt J, Connor Gorber S, Tremblay M. A comparison of direct versus self-report measures for assessing physical activity in adults: a systematic review. Int J Behav Nutr Phys Act. 2008;5:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tarabichi M, Salcedo A, Deshwar AG, et al. A practical guide to cancer subclonal reconstruction from DNA sequencing. Nat Methods. 2021;18(2):144-155. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no data sets were generated or analyzed during the current study.


Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES