Skip to content

Male Reproductive Organ Cancer

Male reproductive organ cancer encompasses a range of malignancies that originate in the organs of the male reproductive system. This category includes, but is not limited to, prostate cancer, testicular cancer, and penile cancer. Among these, prostate cancer is a prevalent diagnosis in men worldwide, representing a considerable public health concern.

The biological basis of these cancers involves the uncontrolled growth and division of abnormal cells within the affected tissues. This cellular dysregulation is often influenced by a complex interplay of genetic predispositions, environmental factors, and lifestyle choices. Genetic variations, such as single nucleotide polymorphisms (SNPs), have been identified as significant contributors to an individual’s susceptibility to certain male reproductive organ cancers. For example, numerous genome-wide association studies (GWAS) have pinpointed specific genetic variants associated with an increased risk of prostate cancer, including loci on chromosomes 2p15, Xp11.22, 22q13, and other regions[1]. Understanding these genetic underpinnings is crucial for elucidating the molecular pathways involved in cancer development and progression.

From a clinical perspective, early detection and accurate diagnosis are vital for effective management and improved patient outcomes. Advances in screening methods, diagnostic tests, and therapeutic modalities continue to play a crucial role in the fight against these diseases. The clinical relevance of genetic insights extends to personalized medicine, where an individual’s genetic profile can inform risk assessment, guide surveillance strategies, and optimize treatment decisions.

The social importance of addressing male reproductive organ cancer is profound. Beyond the direct health consequences for individuals, these cancers can significantly impact quality of life, emotional well-being, and family dynamics. Efforts to raise public awareness, promote research into prevention and treatment, and advocate for accessible healthcare are essential steps in reducing the societal burden of these diseases.

Genetic studies of male reproductive organ cancer, predominantly focusing on prostate cancer, offer significant insights into disease susceptibility but are subject to several limitations that influence the interpretation and generalizability of their findings. These limitations span methodological approaches, population diversity, and the complex etiology of the disease.

Methodological and Statistical Constraints

Section titled “Methodological and Statistical Constraints”

The discovery of genetic variants associated with male reproductive organ cancer risk often relies on genome-wide association studies (GWAS), which, despite large sample sizes, may still face challenges in statistical power. Initial findings can sometimes exhibit inflated effect sizes, necessitating rigorous validation through extensive replication cohorts and meta-analyses to confirm their authenticity and provide more precise estimates of risk. For instance, the identification of additional risk variants often requires increasing the number of single nucleotide polymorphisms (SNPs) taken forward to large-scale replication studies[2]. Furthermore, the stringent statistical thresholds applied to achieve genome-wide significance, such as p < 5 × 10^-8, while crucial for minimizing false positives, may inadvertently overlook true associations with more modest effects that collectively contribute to disease risk[3]. The ongoing need for international consortia and collaborative studies underscores the inherent statistical challenges in identifying all relevant genetic predispositions [4].

The generalizability of genetic findings for male reproductive organ cancer is often impacted by the specific populations studied. While many studies involve international collaborations across various countries, the ancestral composition of these cohorts can limit the direct applicability of findings to diverse global populations. Different ancestral groups may possess unique genetic backgrounds, leading to variations in allele frequencies or the presence of population-specific risk variants that are not captured in predominantly studied cohorts[5]. Moreover, male reproductive organ cancers, particularly prostate cancer, are heterogeneous diseases encompassing a wide spectrum of clinical presentations and aggressiveness. Most genetic studies identify loci associated with overall disease susceptibility, but they may not consistently differentiate between indolent versus aggressive forms, or specific disease subtypes. This broad phenotyping can obscure genetic variants that are specifically linked to more severe outcomes or influence disease progression, thereby limiting the utility of findings for personalized risk assessment and treatment strategies[1]. The emphasis on specific sub-phenotypes, such as lung cancer in never smokers, highlights the importance of such distinctions in understanding disease etiology[6].

Despite the identification of numerous genetic susceptibility loci, these variants typically account for only a fraction of the total inherited risk for male reproductive organ cancer. This suggests that a significant portion of genetic risk remains undiscovered, pointing to the involvement of other genetic factors, such as rare variants, structural variations, or complex epistatic interactions that are not readily captured by standard GWAS methodologies. Furthermore, the role of environmental factors and their intricate interactions with genetic predispositions is often not fully elucidated in these studies. The absence of comprehensive data on lifestyle, dietary, occupational, or other environmental exposures means that potential gene-environment confounders or synergistic effects on risk are largely unaccounted for. Finally, while studies successfully pinpoint associated genetic loci, the precise biological mechanisms through which many identified variants exert their influence often require further investigation. Many SNPs reside in non-coding regions, making their functional interpretation challenging and necessitating additional research to understand their impact on gene expression, protein function, or broader cellular pathways relevant to cancer development[6].

Genetic variations play a crucial role in an individual’s susceptibility to various conditions, including male reproductive organ cancers. These variants, often single nucleotide polymorphisms (SNPs), can influence gene function, protein activity, or regulatory pathways, thereby impacting cellular processes vital for health. Research efforts continue to uncover specific genetic loci associated with prostate cancer and other related conditions, highlighting the complex genetic architecture underlying these diseases.

KLF12 (Krüppel-like factor 12) is a transcription factor belonging to the KLF family, which plays diverse roles in regulating cell growth, differentiation, and transformation. Variants within or near genes like KLF12 can influence how these processes are controlled, potentially contributing to cancer development. For instance, the KLF family genes, including KLF12, are known to regulate cell growth and transformation, and genomic regions containing such factors are frequently implicated in various cancers[7]. The rs191851063 variant, though its specific impact is not detailed, could modify KLF12’s regulatory functions, thereby affecting cellular pathways critical for maintaining healthy tissue and preventing uncontrolled proliferation in male reproductive organs. Genetic variations across the genome, including those in transcription factors, are widely studied for their associations with prostate cancer susceptibility[3].

The PINX1 gene (PIN2 (TERF1)-Interacting Telomerase Inhibitor 1) is recognized for its role as a tumor suppressor, primarily by inhibiting telomerase activity, an enzyme often overactive in cancer cells that allows for unlimited cell division. Variants likers564906617 in PINX1 could potentially impair this crucial tumor-suppressing function, leading to increased cellular immortality and a higher risk of cancer in male reproductive organs. Similarly, SAMD3 (Sterile Alpha Motif Domain Containing 3) is involved in signal transduction, mediating protein-protein interactions critical for cellular communication and regulation. Dysregulation of these signaling pathways, potentially influenced by variants such asrs75467494 , can contribute to uncontrolled cell growth characteristic of cancers. NWD2 (Nociceptin/Orphanin FQ Receptor 2), a G protein-coupled receptor, also plays a role in various physiological processes, and its altered function due to the rs905839831 variant could affect cell proliferation and survival, thereby influencing cancer risk[1]. Studies have consistently identified multiple genetic loci that contribute to prostate cancer susceptibility, highlighting the complex genetic architecture of the disease[5].

Long intergenic non-coding RNAs (LINC RNAs) such as LINC01182, LINC00504, and LINC01804, along with non-coding elements like RN7SKP181 and RNU5E-7P, represent a significant class of regulatory molecules in the human genome. These non-coding RNAs do not produce proteins but instead influence gene expression, chromatin structure, and various cellular processes, often acting as crucial regulators in cancer development and progression. Variants likers543358139 , rs548212384 , and rs541147603 , located in or near these non-coding regions, can alter their structure or expression, thereby disrupting normal cellular regulation and potentially contributing to the initiation or progression of male reproductive organ cancers. The identification of numerous susceptibility loci for prostate cancer underscores the importance of a wide range of genetic variations, including those in non-coding regions, in disease risk[4]. Genome-wide association studies frequently uncover such regulatory regions that impact cancer risk[8].

Other variants, such as rs148020284 , found near LRATD1 (Lecithin Retinol Acyltransferase Domain Containing 1) and NBAS (Neuroblastoma Amplified Sequence), may influence diverse cellular functions. NBAS is critical for Golgi-ER trafficking and protein assembly, processes vital for cell function and integrity, while LRATD1 is linked to lipid metabolism. Alterations in these pathways can impact cell survival and growth, contributing to cancer. Thers537456754 variant, associated with RAD52P1 (a pseudogene related to DNA repair) and ZNF385B (a zinc finger protein involved in transcription), suggests a role in maintaining genomic stability and gene regulation. Pseudogenes can sometimes modulate the expression of their functional counterparts, and zinc finger proteins are key regulators of gene activity, both critical for preventing cancerous transformation. Lastly, the DTWD2 gene (DTW Domain Containing 2), with variant rs547300358 , is thought to be involved in DNA/RNA processes, potentially affecting gene expression or RNA modification, which are fundamental to cell proliferation and differentiation [1]. These findings collectively emphasize that a broad spectrum of genetic variations, even in less-characterized genes, can have implications for prostate cancer susceptibility and progression[9].

RS IDGeneRelated Traits
rs191851063 KLF12male reproductive organ cancer
rs148020284 LRATD1 - NBASmale reproductive organ cancer
rs543358139 LINC01182 - LINC00504male reproductive organ cancer
rs548212384 RN7SKP181 - LINC02253male reproductive organ cancer
rs75467494 SAMD3male reproductive organ cancer
rs905839831 NWD2male reproductive organ cancer
rs537456754 RAD52P1 - ZNF385Bmale reproductive organ cancer
rs541147603 LINC01804 - RNU5E-7Pmale reproductive organ cancer
rs564906617 PINX1male reproductive organ cancer
rs547300358 DTWD2male reproductive organ cancer

Classification, Definition, and Terminology

Section titled “Classification, Definition, and Terminology”

Defining Male Reproductive Organ Cancers and Core Terminology

Section titled “Defining Male Reproductive Organ Cancers and Core Terminology”

Male reproductive organ cancer refers to the uncontrolled growth of abnormal cells originating in any of the male reproductive organs. While this broad category encompasses various specific cancers, prostate cancer is a prominent example frequently investigated in genetic studies.[1]It is precisely defined by the presence of malignant cells within the prostate gland, a Walnut-sized organ located below the bladder in males, responsible for producing seminal fluid. Key terminology in this field includes “prostate cancer-susceptibility genes” and “susceptibility loci,” which denote specific genetic regions or variants that increase an individual’s predisposition to developing the disease.[1]

These terms are crucial for understanding the conceptual framework of cancer risk, moving beyond purely clinical presentations to include underlying genetic predispositions. The identification of such genes and loci forms the basis of operational definitions for genetic risk factors, distinguishing individuals with higher inherent likelihoods of disease. An “international consortium for prostate cancer genetics” exemplifies a collaborative framework aimed at systematically identifying and characterizing these genetic determinants, highlighting the global effort to standardize nomenclature and research approaches for understanding the etiology of prostate cancer.[1]

Genetic Classification and Susceptibility Loci

Section titled “Genetic Classification and Susceptibility Loci”

The classification of male reproductive organ cancers, particularly prostate cancer, extends beyond traditional pathological staging to include genetic subtypes based on identified “susceptibility loci.” These loci represent specific genomic locations where common sequence variants are associated with an altered risk of developing the disease. For instance, studies have identified significant susceptibility loci on chromosomes 22q13, 2p15, and Xp11.22 that confer risk for prostate cancer.[1]This genetic classification system allows for a more nuanced understanding of disease heterogeneity, moving towards a categorical approach where individuals can be grouped based on their genetic predisposition profiles.

The recognition of these genetic variations introduces a level of severity gradation based on the number and impact of identified risk alleles, although clinical severity is still primarily assessed through tumor characteristics. The nosological system for prostate cancer is thus evolving to integrate both phenotypic and genotypic information, offering a more comprehensive view of disease risk and potential subtypes. For example, the identification of “seven new prostate cancer susceptibility loci”[4]demonstrates the ongoing refinement of these genetic classifications, enriching our understanding of the diverse pathways that can lead to prostate cancer development.

In the context of male reproductive organ cancer research, particularly for identifying genetic susceptibility, precise diagnostic and measurement criteria are applied to establish significant associations. “Genome-wide association studies” (GWAS) represent a primary measurement approach, systematically scanning the entire human genome for common genetic variants that are more frequent in individuals with the disease compared to controls.[3] This methodology relies on stringent statistical thresholds to define “genome-wide significance,” ensuring that observed associations are not due to chance.

A conservative cut-off value, such as a p-value less than 5 × 10⁻⁸, is typically employed as a research criterion to declare a genetic locus significantly associated with cancer risk.[3] This threshold is critical for distinguishing true susceptibility loci from background genetic noise and for validating findings across different study populations. The application of such rigorous statistical biomarkers and thresholds is essential for the robust identification of genetic factors, such as “sequence variants at 22q13” [1]or “four variants associated with prostate cancer susceptibility,”[5] which can then be investigated further for their clinical implications in risk assessment and prevention.

Causes

Genetic Predisposition and Inherited Susceptibility

Section titled “Genetic Predisposition and Inherited Susceptibility”

The development of male reproductive organ cancers, particularly prostate cancer, is significantly influenced by inherited genetic factors. Genome-wide association studies (GWAS) have been instrumental in identifying numerous common genetic variants that collectively contribute to an individual’s susceptibility. This indicates a polygenic risk architecture, where the cumulative effect of multiple genes, each with a small impact, increases the overall likelihood of developing the disease[5]. These studies aim to map the genetic landscape of prostate cancer susceptibility by pinpointing specific genomic regions and variants associated with increased risk.

Research has identified several key susceptibility loci across the human genome. For instance, sequence variants at chromosome 22q13 have been found to be associated with prostate cancer risk[1]. Further international collaborations, such as the PRACTICAL Consortium, have confirmed and expanded upon these findings, identifying multiple novel prostate cancer predisposition loci[10]. Specific large-scale GWAS efforts have, for example, identified four distinct variants [5]and, in another study, seven new susceptibility loci that contribute to prostate cancer risk[4]. These discoveries underscore the complex interplay of inherited genetic factors in determining an individual’s likelihood of developing prostate cancer.

Male reproductive organ cancers, particularly prostate cancer, represent a significant health concern, with their development influenced by a complex interplay of genetic, cellular, and physiological factors. Research into these cancers often focuses on identifying underlying predispositions and understanding how these contribute to disease onset and progression.

The genetic architecture of prostate cancer susceptibility has been extensively investigated through genome-wide association studies (GWAS), leading to the identification of several genomic regions associated with an increased risk. For instance, specific sequence variants located at chromosome 22q13 have been identified as conferring susceptibility to prostate cancer[1]. Further research has revealed additional susceptibility loci, with studies identifying four distinct variants associated with prostate cancer risk[11]. Moreover, common sequence variants on chromosomes 2p15 and Xp11.22 have also been found to confer susceptibility to prostate cancer[5]. These findings, along with the identification of seven new prostate cancer susceptibility loci, underscore the polygenic nature of the disease, where multiple genes and genetic variants contribute to an individual’s overall risk[4].

Cellular and Molecular Underpinnings of Susceptibility

Section titled “Cellular and Molecular Underpinnings of Susceptibility”

The genetic variants identified in prostate cancer susceptibility studies are hypothesized to influence cellular functions and regulatory networks within the prostate gland. While specific molecular pathways and key biomolecules are not detailed in all contexts, the presence of these susceptibility genes implies that their products, such as proteins, enzymes, or transcription factors, may be altered or function atypically. These genetic changes can subtly disrupt the intricate balance of cellular processes, potentially affecting cell growth, differentiation, and programmed cell death. Such molecular alterations at the cellular level can contribute to a pro-cancerous environment, laying the groundwork for tumor initiation and progression.

The cumulative effect of genetic predispositions, alongside other risk factors, contributes to the pathophysiological processes that define prostate cancer. At the organ level, the prostate gland, a vital component of the male reproductive system, undergoes a series of homeostatic disruptions. These disruptions can lead to uncontrolled proliferation of prostate cells, a hallmark of cancer, and can alter the normal tissue interactions within the gland. The disease mechanisms involve the transformation of normal prostate epithelial cells into malignant ones, often progressing from localized tumors to more aggressive forms that can impact the overall function of the reproductive system and potentially lead to systemic consequences if left untreated.

The development of male reproductive organ cancer, particularly prostate cancer, involves a complex interplay of genetic factors and molecular mechanisms that ultimately lead to cellular dysregulation. Research, primarily through genome-wide association studies (GWAS), has identified numerous genetic variants that predispose individuals to these cancers, shedding light on the underlying pathways.

Genetic Predisposition and Gene Regulation

Section titled “Genetic Predisposition and Gene Regulation”

The initiation and progression of male reproductive organ cancers are significantly influenced by an individual’s genetic makeup, with several susceptibility loci identified across the human genome [1]. For prostate cancer, specific chromosomal regions such as 22q13, 2p15, and Xp11.22 harbor variants that confer an increased risk[1]. These genetic variations often occur in non-coding or regulatory regions, where common regulatory variation can impact gene expression in a cell type-dependent manner [6]. This alteration in gene regulation can lead to quantitative or qualitative changes in protein production, disrupting normal cellular pathways and contributing to the processes that drive oncogenesis.

Beyond mere association, understanding the molecular underpinnings of these susceptibility loci reveals how genetic variants contribute to disease. For instance, genetic variations within the prostate stem cell antigen (PSCA) gene have been shown to confer susceptibility to urinary bladder cancer[12]. While specifically linked to bladder cancer in some studies, the gene’s association and its name suggest a role in male reproductive-related tissues, where such variants can lead to altered protein function or expression levels. These molecular changes, stemming from germline genetic architecture, represent a fundamental layer of pathway dysregulation that can predispose cells to malignant transformation.

The identification of specific genetic variants associated with male reproductive organ cancer inherently points to an underlying dysregulation of critical biological pathways. Although the precise signaling cascades, metabolic shifts, or feedback loops are not explicitly detailed in all genetic association studies, the presence of these susceptibility loci implies that the variants perturb normal cellular processes essential for controlled growth, differentiation, and programmed cell death[1]. This inherent dysregulation can manifest as characteristics of cancer, such as uncontrolled cell proliferation, impaired DNA repair, or evasion of immune surveillance, which collectively contribute to the progression of the disease.

Integrated Genetic Risk Factors and Network Interactions

Section titled “Integrated Genetic Risk Factors and Network Interactions”

The etiology of male reproductive organ cancer is often multifactorial, arising from the cumulative effect of multiple genetic risk factors rather than a single causative variant, suggesting complex network interactions. Extensive genome-wide association studies have consistently identified several distinct susceptibility loci, with some research highlighting four new loci and others identifying seven new loci associated with prostate cancer risk[5]. This intricate genetic architecture suggests a systems-level integration where variations across different chromosomal regions, including those on 22q13, 2p15, and Xp11.22, collectively modulate an individual’s overall disease susceptibility[1]. The interplay among these distinct genetic predispositions, potentially affecting various regulatory or functional pathways, ultimately shapes the emergent risk profile for developing male reproductive organ cancer.

Genetic Basis of Drug Response Variability

Section titled “Genetic Basis of Drug Response Variability”

Genetic variants identified in studies, such as those on chromosomes 22q13, 2p15, and Xp11.22, are associated with prostate cancer susceptibility, representing inherited differences that could potentially modulate an individual’s response to therapeutic interventions[1]. While these studies primarily focus on disease risk, the broader field of pharmacogenetics explores how such genetic variations might influence drug absorption, distribution, metabolism, and excretion (pharmacokinetics) or affect drug targets and signaling pathways (pharmacodynamics). The precise mechanisms by which these specific susceptibility variants might impact drug metabolism enzymes, like cytochrome P450s or phase II enzymes, or drug transporters, are areas for further research in the context of male reproductive organ cancers.

Impact on Drug Efficacy and Adverse Reactions

Section titled “Impact on Drug Efficacy and Adverse Reactions”

Polymorphisms in genes, including those like PSCA (Prostate Stem Cell Antigen) which has been associated with cancer susceptibility, highlight the genetic heterogeneity underlying disease[12]. Understanding how these or other genetic variants may affect drug target interactions or the function of drug-metabolizing enzymes and transporters is crucial for predicting therapeutic outcomes and managing adverse reactions. The identification of such variants provides a foundation for investigating their potential to differentiate patient responses to therapies for male reproductive organ cancer, influencing both drug efficacy and the incidence of adverse events.

Towards Personalized Therapeutic Strategies

Section titled “Towards Personalized Therapeutic Strategies”

The discovery of numerous genetic loci linked to prostate cancer risk underscores the potential for personalized medicine approaches in managing male reproductive organ cancers[5]. Integrating information on an individual’s genetic makeup, including variants that might influence drug metabolism or drug targets, could guide drug selection and dosing to optimize therapeutic benefits and minimize adverse effects. The ongoing characterization of these susceptibility loci lays foundational groundwork for future pharmacogenomic applications aimed at tailoring treatment strategies based on a patient’s unique genetic profile.

Frequently Asked Questions About Male Reproductive Organ Cancer

Section titled “Frequently Asked Questions About Male Reproductive Organ Cancer”

These questions address the most important and specific aspects of male reproductive organ cancer based on current genetic research.


Yes, having a close relative like your father with prostate cancer increases your risk. Genetic predispositions, often involving common genetic variations, can run in families, making you more susceptible. However, it doesn’t guarantee you’ll develop it, as many factors are involved. Understanding your family history helps your doctor assess your personal risk.

While genetics play a significant role, lifestyle choices and environmental factors are also important influences on cancer development. Although the exact interplay between your daily habits and specific genetic predispositions is complex and still being researched, maintaining a healthy lifestyle is generally recommended. This can influence your overall risk and potentially mitigate some genetic susceptibilities.

3. Does my ethnic background change my risk for these cancers?

Section titled “3. Does my ethnic background change my risk for these cancers?”

Yes, your ancestral background can affect your risk. Different ethnic groups may possess unique genetic profiles, including variations in allele frequencies or the presence of population-specific risk variants, that influence susceptibility. This means that genetic findings from one population might not directly apply to another, highlighting the importance of diverse research.

Yes, genetic insights can be very useful for personalized risk assessment. A genetic test could identify specific genetic variants that are known to increase susceptibility to certain male reproductive organ cancers, particularly prostate cancer. This information can help guide surveillance strategies and inform discussions with your doctor about early detection.

This often comes down to a complex interplay of genetics and environment. Some men inherit specific genetic variations, like those on chromosomes 2p15, Xp11.22, or 22q13, that make them more susceptible to prostate cancer. Even with similar lifestyles, these underlying genetic differences can significantly influence an individual’s risk compared to others.

While many genetic studies identify loci for overall disease susceptibility, differentiating between indolent (less severe) and aggressive forms is more challenging. Current research is working towards identifying specific genetic variants linked to more severe outcomes or disease progression. This area is crucial for personalized risk assessment and tailoring treatment strategies.

While you can’t change your genes, you can influence your overall risk. Genetic predispositions account for a fraction of the total inherited risk, but environmental factors and lifestyle choices also play a significant role. Engaging in healthy habits and following recommended screening guidelines can help manage and potentially mitigate the impact of inherited susceptibilities.

8. If no one in my family had it, am I in the clear?

Section titled “8. If no one in my family had it, am I in the clear?”

Not necessarily. While a family history certainly increases risk, many cases occur without a clear inherited pattern. Genetic variations, environmental factors, and lifestyle choices all contribute to risk, and some of these can arise spontaneously or without a strong family link. Regular screenings and discussions with your doctor are still important.

Your diet is part of your overall lifestyle, and lifestyle choices can influence cancer risk. Although the exact mechanisms and specific dietary factors are still being fully elucidated, they are considered contributors alongside genetic predispositions. A balanced, healthy diet is generally recommended to support overall health and potentially reduce risk.

Genetic studies, especially large ones, face challenges in confirming findings and ensuring they apply to everyone. Factors like the specific populations studied, stringent statistical thresholds, and the complex, heterogeneous nature of cancer itself can lead to variations in findings. This highlights the ongoing need for extensive replication and diverse international collaborations to refine our understanding.


This FAQ was automatically generated based on current genetic research and may be updated as new information becomes available.

Disclaimer: This information is for educational purposes only and should not be used as a substitute for professional medical advice. Always consult with a healthcare provider for personalized medical guidance.

[1] Sun J, et al. Sequence variants at 22q13 are associated with prostate cancer risk. Cancer Res. 2009;69(2):401-8.

[2] Wang, Y., et al. “Common 5p15.33 and 6p21.33 variants influence lung cancer risk.”Nat Genet, 2008. PMID: 18978787.

[3] Murabito JM. A genome-wide association study of breast and prostate cancer in the NHLBI’s Framingham Heart Study. BMC Med Genet. 2007;8 Suppl 1:S6.

[4] Eeles RA, et al. Identification of seven new prostate cancer susceptibility loci through a genome-wide association study. Nat Genet. 2009;41(10):1116-21.

[5] Gudmundsson, J. “Common sequence variants on 2p15 and Xp11.22 confer susceptibility to prostate cancer.”Nat Genet, 2008.

[6] Li, Y., et al. “Genetic variants and risk of lung cancer in never smokers: a genome-wide association study.”Lancet Oncol, 2010.

[7] Petersen GM, et al. A genome-wide association study identifies pancreatic cancer susceptibility loci on chromosomes 13q22.1, 1q32.1 and 5p15.33. Nat Genet. 2010;42(2):141-6.

[8] Hunter DJ, et al. A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat Genet. 2007;39(7):870-4.

[9] Kiemeney LA, et al. Sequence variant on 8q24 confers susceptibility to urinary bladder cancer. Nat Genet. 2008;40(11):1329-34.

[10] Kote-Jarai, Z., et al. “Multiple novel prostate cancer predisposition loci confirmed by an international study: the PRACTICAL Consortium.”Cancer Epidemiol Biomarkers Prev, vol. 17, no. 8, 2008, pp. 2052-2061.

[11] Gudmundsson J, et al. Genome-wide association and replication studies identify four variants associated with prostate cancer susceptibility. Nat Genet. 2009;41(10):1122-6.

[12] Wu, X. et al. “Genetic variation in the prostate stem cell antigen gene PSCA confers susceptibility to urinary bladder cancer.”Nat Genet, 2009.