115
Views
3
CrossRef citations to date
0
Altmetric
Review

Dysregulation of the homeobox transcription factor gene HOXB13: role in prostate cancer

&
Pages 193-201 | Published online: 05 Aug 2014

Abstract

Prostate cancer (PC) is the most common noncutaneous cancer in men, and epidemiological studies suggest that about 40% of PC risk is heritable. Linkage analyses in hereditary PC families have identified multiple putative loci. However, until recently, identification of specific risk alleles has proven elusive. Cooney et al used linkage mapping and segregation analysis to identify a putative risk locus on chromosome 17q21-22. In search of causative variant(s) in genes from the candidate region, a novel, potentially deleterious G84E substitution in homeobox transcription factor gene HOXB13 was observed in multiple hereditary PC families. In follow-up testing, the G84E allele was enriched in cases, especially those with an early diagnosis or positive family history of disease. This finding was replicated by others, confirming HOXB13 as a PC risk gene. The HOXB13 protein plays diverse biological roles in embryonic development and terminally differentiated tissue. In tumor cell lines, HOXB13 participates in a number of biological functions, including coactivation and localization of the androgen receptor and FOXA1. However, no consensus role has emerged and many questions remain. All HOXB13 variants with a proposed role in PC risk are predicted to damage the protein and lie in domains that are highly conserved across species. The G84E variant has the strongest epidemiological support and lies in a highly conserved MEIS protein-binding domain, which binds cofactors required for activation. On the basis of epidemiological and biological data, the G84E variant likely modulates the interaction between the HOXB13 protein and the androgen receptor, as well as affecting FOXA1-mediated transcriptional programming. However, further studies of the mutated protein are required to clarify the mechanisms by which this translates into PC risk.

Introduction

Prostate cancer (PC) is the most common noncutaneous cancer in men in the Western world, with about 240,000 new cases diagnosed in 2011.Citation1 Twin studies,Citation2Citation4 segregation analysis,Citation5Citation11 and linkage of high-risk or hereditary families all suggest that there are hereditary components to the disease.Citation12Citation18 However, replication of linkage results has proven difficult, and few high-risk alleles have been conclusively identified. Genome-wide association studies have identified more than 75 common alleles that are also associated with increased PC risk, and although many of these have been validated, each is predicted to make a small contribution to disease risk.Citation19Citation25 Finally, family-based association studies have contributed additional validated loci.Citation26,Citation27

Despite this enormous amount of work, the greatest challenge remains: how can we accurately and efficiently identify the subset of men who will go on to die of metastatic disease versus those with slow-growing indolent PC? While a variety of approaches have been tested, including linkage analysis of high-risk families using various measures of aggressiveness as the phenotypeCitation28Citation32 and scans based on Gleason grade among family members,Citation33Citation35 few replicated loci have been identified.

Thus, much current research in the field is focused on 1) identifying genetic factors that predispose to any level of disease risk; 2) understanding the tumor biology and genetic factors that contribute to tumor aggressiveness and metastatic progression; and 3) identifying a panel of genetic markers that predict aggressive forms of PC. In 2012, significant progress was made on all of these questions with the discovery of mutations in homeobox transcription factor gene HOXB13 that were associated with both familial and population-based risk. We summarize the state of the field with regard to HOXB13 in the following review.

Initial findings

HOXB13 was identified as a risk factor for PC following a series of studies led by Ewing et al.Citation36 These investigators followed up on their initial findings of linkage in high-risk families at chromosome 17q21-23.Citation37,Citation38 The initial linkage peak was identified in a scan of 175 hereditary pedigrees and was replicated by the International Consortium for Prostate Cancer Genetics, a large collaborative group with more than 2,000 hereditary PC families.Citation39,Citation40 Fine mapping of the region narrowed the focus to chromosome 17q21-22, a region with more than 200 genes in the minimal recombination region.Citation41 With this smaller region identified, the group evaluated the coding sequence of 202 candidate genes in search of potential PC susceptibility alleles or genes.Citation36 Targeted sequencing of the youngest cases in families with evidence of linkage focused attention on the HOXB13 gene, as four of the sequenced families harbored a novel (G84E) substitution that was predicted to be deleterious. In follow-up genotyping, all 18 cases in these four families were observed to carry the mutation. Analysis of a population of 5,083 unrelated individuals with PC suggested a frequency of about 1.4%, compared to 0.1% in 1,401 healthy male controls from the general population. The carrier rate was highest in younger cases who reported a family history of PC (3.1%). Thus, cases from the general population, as well as cases from families with history of PC, were significantly enriched for the G84E variant, with all P<0.001. Interestingly, while the G84E mutation appeared to be the most common risk variant in this initial study, two additional variants, R229G and G216C, were found in African American families. Three individuals in one family segregated a c.685 C>G transversion, which encodes the R229G variant at a highly conserved position. Additional rare variants were found in other families, but G84E was most frequent and most strongly implicated in PC.

HOXB13 findings replicated

The strength of the epidemiological findings and intriguing biological role of HOXB13 generated great interest among PC researchers. Many groups who were underwhelmed by earlier reports of PC candidate risk genes mobilized to replicate these findings. The story was compelling for several reasons. First, the chromosome 17 region that harbors the gene was initially selected for intense study because multiple studies of high-risk PC families had already uncovered evidence for genetic linkage at the locus.Citation41,Citation42 Second, in the data presented by Ewing et al,Citation36 multiple large families faithfully segregated the same mutation, which strengthened the result. The finding was upheld in multiple subsequent studies, and several are described below, as well as in .

Table 1 HOXB13 risk allele studies cited in this review

Stott-Miller et alCitation43 reported results from a population-based, case–control study of men in western Washington State, where they observed the G84E mutation in 1.3% of cases versus 0.4% of controls, which represents a 3.3-fold higher relative risk in carriers versus noncarriers. The initial finding of a stronger association in the setting of positive family history was also replicated. Furthermore, risk estimates for the variant were also higher among men with higher versus lower Gleason score at diagnosis (odds ratio [OR] =4.13, 95% confidence interval [CI] =1.38–12.38 versus OR =2.71, 95% CI =0.88–8.30) and advanced versus prostate-confined disease.Citation43 While these differences were not statistically significant, they may suggest that HOXB13 G84E is a marker for disease aggressiveness. The same year, Witte et alCitation44 evaluated two cohorts, a family-based study of brothers and a case–control study of 2,665 men with more aggressive disease. In their study, the mutation was detected in 1.47% of cases, in stark contrast with 0.34% among unaffected brothers in the family-based analysis, and there was a complete absence of the variant in controls from the case–control arm.Citation44 Again, the mutation was more common among men with early-onset disease or a positive family history.Citation44

Not surprisingly, given the association with family history, the result has also replicated in family studies. One of the first such studies was an examination of familial PC from Vanderbilt University.Citation45 These investigators genotyped 930 controls without a personal or family history of PC and 928 familial cases, and observed the mutation in 16 of the familial cases versus only two controls (OR =7.9, 95% CI =1.8–34.5). The carrier rate was 1.9% in all probands, but it went up 30% in cases who had ≥3 affected family members.Citation45 The primary result was also confirmed in the much larger International Consortium for Prostate Cancer Genetics study in which investigators genotyped the G84E variant and 14 other single-nucleotide polymorphisms (SNPs) in 2,443 families.Citation46 They showed that at least one mutation carrier was present in 112 families (4.6%) and that within the carrier families, the G84E variant was more common among men with disease than among those without. The mutation was in the same haplotype in the majority (95%) of carriers, strongly suggesting a founder effect. Overall, these findings suggest that nearly 5% of cases from high-risk families carry the G84E mutation.

Several reports have addressed penetrance of the allele in addition to risk. An Australian study found that 19 of 1,384 probands (1.4%) carried the G84E mutation. Six of those men had a family history of PC. The age-specific incidence for carriers in Australia was estimated to be 16.4-fold higher (95% CI =2.5–107.2) than expected levels based on birth year. This study also estimated birth year-specific cumulative risk for carriers and found that penetrance was 60% by age 80, which is higher than other estimates.Citation47 By comparison, Karlsson et alCitation48 examined 5,003 cases and 4,693 controls from Sweden and showed that the G84E mutation was present in 1.3% of population controls and 4.6% of cases, providing a strong association with PC risk (OR =3.4, 95% CI =2.2–5.4). Again, the strongest associations were for those with early-onset disease or a positive family history. Among cases aged 35–55 years, the carrier frequency was 10.3% (OR =8.6, 95% CI =5.1–14.0). In this situation, the investigators estimated the collective risk of G84E carriers to be 33% by 80 years of age, compared to the 12% risk that was observed in this population among noncarriers.Citation48

The carrier frequency for men undergoing diagnostic biopsy is much lower than that reported in case–control or PC family-based studies. For instance, Schroeck et alCitation49 observed that among 938 biopsied men, the prevalence of the G84E mutation was 0.42%. Three of the four carriers had PC. None of 301 men with a positive family history and one of 226 men diagnosed before age 55 were carriers, yielding an overall prevalence of 0.44% and raising questions about the utility of clinical testing for this variant in this setting.

HOXB13 within and across populations

It is clear that the frequency of the G84E allele varies significantly between populations. In a multinational study of 2,508 subjects from the REDUCE (Reduction by Dutasteride of Prostate Cancer Events) trial, the authors reported that the G84E mutation frequency was 1.06% in biopsy-negative men from Northern Europe, followed by 0.60% and 0.31% in Europe and North America, respectively.Citation50 No carriers were observed among populations from a variety of other regions, including southern and eastern Europe, South Africa, Latin America, and Australia.Citation50 In Caucasians alone, the mutation frequency was 0.99% in biopsy-positive men versus 0.24% in controls. All mutation carriers had a common haplotype that was once again indicative of a founder effect.Citation50

Because the G84E risk allele is entirely absent in some populations, some have suggested that other variants may confer risk. Several candidate alleles have been identified in addition to those found in African American families in the initial report by Ewing et al.Citation36 For instance, Lin and colleaguesCitation51 studied a cohort of Chinese men and reported that the G84E mutation was not found in their study population. However, a G135E mutation was found twice in 575 cases but not in any of 1,536 controls, a significant difference (P=0.027). However, further epidemiological evidence is required to establish a role for these variants.

A consensus is emerging that HOXB13 is a critical player in PC risk. But the mutation is generally rare in most populations. The gene may be more relevant for northern European populations, as well as in individuals with strong family history or early disease onset. Though the gene’s epidemiological role is becoming clearer, the mechanism by which the G84E allele confers risk remains a crucial question.

The homeobox gene family

HOXB13 is a member of the homeobox (HOX) gene family, which is a large group of clustered, paralogous transcription factors that are crucial for embryonic development along the anterior–posterior axis.Citation52 HOX genes are defined by a characteristic 180-base sequence called the homeobox, which encodes a helix–turn–helix protein motif that mediates binding to enhancer DNA sequences.Citation52 HOX genes are further distinguished by collinearity, an incompletely understood phenomenon in which genes in an HOX cluster are sequentially expressed along the anteroposterior axis, ie, upstream genes are expressed in anterior segments, and downstream genes are expressed in posterior segments (see ).Citation52

Figure 1 The genomic architecture of HOX genes and schematic of gene expression in embryonic development.

Notes: (A) Humans have four clusters of HOX genes on different chromosomes, labeled Cluster A through Cluster D. Each cluster comprises a variable subset of the 13 homology-based HOX gene subtypes. The relative order of the subtypes is preserved across clusters. This conserved ordering is attributed to the genomic duplication-based mechanism by which the clusters arose. (B) During embryonic development, HOX genes are expressed sequentially in partially overlapping zones along the anterior-posterior axis, a phenomenon called collinearity. Accordingly, HOX13 paralogs, including HOXB13, are expressed in posterior regions of the embryo and direct development of the spinal cord, digestive tract, and urogenital system.
Abbreviation: HOX, homeobox gene.
Figure 1 The genomic architecture of HOX genes and schematic of gene expression in embryonic development.

Function of HOX genes

HOX genes play many roles in both embryonic development and differentiated tissues. Much of this functional diversity is explained by the large size of the HOX gene family, tightly regulated spatial and temporal expression patterns, and slight variations in homeodomain sequences that result in specificity for different enhancer DNA sequences.Citation52 The number of HOX genes, as well as the number of HOX gene clusters, varies widely across phyla.Citation53 Genomic amplification and evolutionary divergence have yielded paralogous HOX gene homology subtypes that are defined by sequence similarity of the homeobox sequence.Citation53 In addition, larger genomic events have led to amplification of entire HOX gene clusters, whereas subsequent losses have resulted in clusters that contain only a subset of the homology subtypes.Citation53 Together, the increase in number of genes and clusters allows HOX genes to play many diverse roles. In spite of this variability, the fundamental mechanism of HOX-mediated anterior–posterior patterning is highly conserved across species, which manifests in vertebrates as typical clustered genomic architecture and collinear pattern of expression, but most importantly as extensive coding sequence homology, especially of the characteristic homeobox domain.Citation52

Organization of human HOX genes

Like all tetrapods, humans have four HOX clusters, labeled A–D. Each cluster consists of 9–11 genes that are a subset of the 13 HOX subtypes (). All four clusters have a HOX13 paralog, and these genes are the furthest downstream constituents of each cluster. As predicted by the collinearity principle of HOX genes, HOX13 paralogs are expressed in the most posterior segments of the developing embryo. In 1996, Zeltser et alCitation54 identified a novel gene about 70 kb downstream of the other known HOXB cluster constituents on chromosome 17. Due to high homeodomain sequence identity with HOX13 paralogs A, C, and D, it was dubbed HOXB13.Citation54 In accordance with the principle of collinearity, this downstream position yields HOXB13 expression in posterior structures, where it plays a role in the development of the spinal cord, digestive tract, and urogenital system, including the prostate.Citation54,Citation55

Role of HOXB13

Early studies in mice clarified the role of HOXB13 in normal development and tissue function. Sreenath, et alCitation55 used Northern blotting and in situ hybridization to confirm that murine HOXB13 is expressed during posterior segment development. By sampling these tissues at multiple developmental time points, they found that the HOXB13 transcript appears early in posterior organogenesis, and that expression persists into adulthood in distal colon tissues, as well as in an androgen-independent manner in all four lobes of the murine prostate.Citation55 Murine knockout studies conducted by Economides and CapecchiCitation56 demonstrated that the HOX13 paralogs HOXB13 and HOXD13 apparently cooperate in normal prostate development, as double-knockout mice had malformed ducts with severe truncations and branching defects. However, mice homozygous for homeobox domain-damaging mutations in HOXB13 exhibited multiple structural and functional defects of the secretory cells in the murine ventral prostate, suggesting that HOXB13 is the crucial mediator of terminal differentiation in these cells.Citation56 Examination of seminal fluid from knockout mice showed no evidence of characteristic ventral prostate secretory proteins such as the kazal-type protease inhibitor p12 or the spermine binding protein p25.Citation56 Furthermore, histopathological examination of the ducts revealed cuboid rather than the normal columnar epithelial cell morphology, accompanied by loss of polarity evidenced by CD44 expression; both of these features are consistent with precancerous and neoplastic lesions.Citation56

Additional explorations have built on these basic clues about the gene’s biological role, and mounting evidence suggests that the gene might play a complex-but-plausible role in the carcinogenesis of prostate tumors. The earliest studies focused on the role of HOXB13 in growth regulation via androgen receptor (AR) signaling or cell cycle regulation, but more-recent studies have highlighted many additional roles in PC oncogenesis. However, the picture remains clouded by incongruous results.

HOXB13 and prostate cancer

Overexpression and knockdown studies in PC cell lines have suggested that HOXB13 functions in both normal adult tissue and prostate cancer through the AR, but conflicting findings make it difficult to dissect its physiological role. Jung et alCitation57 showed that HOXB13 was expressed only in cells that also expressed the AR. In their experiments, overexpression of the gene inhibited growth of PC cell lines, which was associated with a dose-dependent disruption of stimulatory AR signaling, as assayed by expression of downstream reporter genes.Citation57 In contrast, Norris et alCitation58 found that HOXB13 knockdown via RNA interference inhibited prostate cellline growth. Under HOXB13 knockdown conditions, they observed repressed transcription of some androgen-regulated genes, but found that HOXB13 acted as an obligate coactivator for expression of others by modulating recruitment of AR and other coregulators to target sequences.Citation58 These authors used a yeast two-hybrid assay and fluorescence resonance energy transfer to confirm that the homeodomain of HOXB13 physically interacts with the AR.Citation58 Intriguingly, Kim et alCitation59 observed that in the setting of AR expression, the HOXB13 transcript repressed the cell cycle, whereas HOXB13 exerted a stimulatory effect on the cell cycle when AR was absent. Indeed, in AR-negative cells, increased HOXB13 expression promoted growth via inhibition of the tumor suppressor p21 and resultant activation of RB-E2F signaling.Citation59 The authors speculated that this role for HOXB13 may help explain persistent growth of androgen-independent tumors.Citation59 These findings may also shed light on the conflicting results of expression studies. One possibility is that heterogeneity between PC cell lines results in context-specific response to HOXB13 perturbations, as noted in the presence versus absence of the AR.

In addition, HOXB13, like other HOX genes in other tissues, has been linked to cell-cycle regulation pathways that are important in PC. Jung et alCitation60 found that overexpressed HOXB13 in PC cell lines inhibited the transcription factor T-cell factor 4. Removal of this pro-cell division signal induced arrested growth at the G1 phase due to suppressed expression of the key cell-cycle regulators cyclin D1 and c-myc.Citation60 Building on these results, Hamid et alCitation61 explored the effects of HOXB13 overexpression and knockdown on the cell cycle. They also noted overexpression-induced arrest at the G1 phase and associated ubiquitination and degradation of cyclin D1, as well as decreased activation of the stimulatory RB protein.Citation61 In contrast, knockdown resulted in upregulation of cyclins, increased activation of RB, and more-rapid cell growth.Citation61

HOXB13 has been tied to PC biology in several other ways. McMullin et al,Citation62 for example, showed that the expression of HOXB13 itself is regulated by FOXA1, a transcription factor with prostate-specific effects on expression that has been tied to many aspects of prostate biology and tumor development. The gene has also been linked to PC risk loci identified in genome-wide association studies. For instance, Huang and CaiCitation63 recently found that the risk allele for rs339331 at chromosome 6q22 enhances the binding of HOXB13, AR, and FOXA1 to the promoter for the RFX6 gene, which stimulates tumor progression and aggressiveness. Since HOXB13 plays many roles in adult prostate tumors, it is likely that it interacts with additional risk loci. It is furthermore probable that the HOXB13 G84E risk allele modulates these interactions, although these effects have not yet been rigorously investigated.

Epigenetic modifications that alter HOXB13 expression may also be important in PC. In AR-negative tumor cell lines, Ren et alCitation64 found that histone modifications prevented expression of HOXB13. In these cells, transcription factor YY1 recruited the suppressive histone deacetylase HDAC4 to the HOXB13 promoter.Citation64 However, administration of the histone deacetylase inhibitor sodium butyrate suppressed repressive histone modifications and resulted in a dramatic increase in HOXB13 and concomitant inhibition of cell proliferation.Citation64 Liu et alCitation65 noted a similar effect in which all-trans retinoic acid increased HOXB13 expression and decreased cell growth via histone methylation modifications mediated by EZH2 and DNMT3b.

HOXB13 may be involved in castration resistance and metastasis of PC, as well. In another study of androgen-independent prostate cell lines, the gene potently stimulated tumor invasion and metastasis by decreasing intracellular zinc concentrations, and resulted in increased nuclear factor-κB signaling.Citation66

Due to these many roles in prostate tumor biology, HOXB13 expression has been suggested as a biomarker for PC diagnosis or detection of recurrence. In microarray studies, Edwards et alCitation67 found that the gene is ubiquitously expressed in the human prostate but significantly overexpressed in prostate tumor tissue versus normal prostate and noncancerous samples from other lineages. This differential expression was confirmed via reverse-transcription polymerase chain reaction in 37 primary prostate tumor specimens versus 16 adjacent normal samples plus four subjects without evidence of malignancy.Citation67 Kim and colleagues also detected overexpression, but only in castration-resistant tumors.Citation59 In an explicit evaluation of HOXB13 as a PC biomarker in 57 human PC tumors, Jeong et alCitation68 saw a positive correlation between expression and Gleason grade, as well as with preoperative prostate-specific antigen measurements. Furthermore, four subjects with tumors refractory to androgen deprivation had very high expression of the gene.Citation68 All of these results have led to speculation that HOXB13 or other related markers might have utility in screening or diagnostic testing.

HOXB13 in other tumors

Reports of HOXB13 function in malignancies of other tissues are similarly contradictory. Because expression of the gene is maintained in the adult distal colon, it is perhaps unsurprising that the gene has been implicated in colon cancer. In colorectal cell lines, HOXB13 expression seems to inhibit growth by some of the same mechanisms observed in prostate tumors, namely suppression of TCF4 and c-myc expression.Citation69 The gene may similarly act as a tumor suppressor in renal cell carcinoma.Citation70 However, although the gene has been investigated in tumors of the endometrium, ovaries, cervix, and breast, its role in these malignancies is less clear.Citation70Citation73 In breast cancer cell lines, for example, Ma et alCitation74 noted that ectopic expression of HOXB13 promoted tumor cell motility and invasion. Furthermore, the ratio of HOXB13:IL17BR expression predicted clinical response to tamoxifen, and a subsequent study found that HOXB13 causes tamoxifen resistance via modulation of ERα and IL-6 expression.Citation74Citation76

The role of HOXB13 G84E in prostate cancer risk

The HOXB13 G84E variant, as well as other, rarer potential risk alleles in the gene, are computationally predicted to impair the structure or function of the encoded protein. The variants fall in domains that are highly conserved across species.Citation36 The G84E variant lies in a domain that binds MEIS proteins, which are critical coactivators of HOX gene binding that modulate target specificity and downstream expression.Citation77 Other, less-proven potential risk alleles, including two found only in patients of African descent, lie in the highly conserved homeobox domain.Citation36

It is difficult to fully assess the mechanistic underpinnings of PC risk variants in HOXB13, because it is not even certain whether the gene acts as a tumor suppressor or oncogene. These contradictory results may be attributable to context-specific roles for HOXB13, including the polar opposite effects of HOXB13 on the cell cycle in the presence versus the absence of the AR, as noted by Kim et al.Citation59 The original report on G84E noted the conflicts in the literature and suggested that the variant may induce a gain of function, since both inherited and acquired truncating mutations are absent.Citation36 However, inherited truncating mutations may not be evolutionarily tolerated due to the critical role of HOXB13 in embryonic development. Furthermore, some other cancer risk genes, including BRCA1, are not commonly somatically mutated.

It is clear that the HOXB13 gene plays a complex and crucial role in many pathways implicated in PC oncogenesis, progression, and metastasis. HOXB13 is preferentially recruited to the risk allele of an established PC-associated SNP, enhancing expression of RFX6, which is a driver of prostate cancer cell migration and a predictor of disease progression.Citation63 To better understand the role of the gene in PC risk, future research must clarify its role in normal prostate tissue, as well as AR-positive and AR-negative cancer cell lines. Only then will we be able to piece together the gene’s wide-ranging roles in androgen receptor signaling, cell-cycle regulation, and castration-resistant disease. Finally, studies of HOXB13 function in cell lines with and without the G84E variant will enhance our understanding of PC oncogenesis and progression.

Acknowledgments

We gratefully acknowledge the many men who have participated in the cited studies by providing DNA samples, tumor tissue, and medical information. We also express our gratitude to the Intramural Program of the National Human Genome Research Institute of the National Institutes of Health (EAO and BD) and to the National Institutes of Health Oxford Cambridge Graduate Scholars Program (BD).

Disclosure

The authors report no conflicts of interest in this work.

References

  • SiegelRNaishadhamDJemalACancer statistics, 2013CA Cancer J Clin2013631113023335087
  • GrönbergHDamberLDamberJEStudies of genetic factors in prostate cancer in a twin populationJ Urol19941525 Pt 114841487 discussion 1487–14897933190
  • LichtensteinPHolmNVVerkasaloPKEnvironmental and heritable factors in the causation of cancer – analyses of cohorts of twins from Sweden, Denmark, and FinlandN Engl J Med20003432788510891514
  • VerkasaloPKKaprioJKoskenvuoMPukkalaEGenetic predisposition, environment and cancer incidence: a nationwide twin study in Finland, 1976–1995Int J Cancer199983674374910597189
  • CarterBSBeatyTHSteinbergGDChildsBWalshPCMendelian inheritance of familial prostate cancerProc Natl Acad Sci U S A1992898336733711565627
  • ConlonEMGoodeELGibbsMOligogenic segregation analysis of hereditary prostate cancer pedigrees: evidence for multiple loci affecting age at onsetInt J Cancer2003105563063512740911
  • GongGOakley-GirvanIWuAHSegregation analysis of prostate cancer in 1,719 white, African-American and Asian-American families in the United States and CanadaCancer Causes Control200213547148212146852
  • GrönbergHDamberLDamberJEIseliusLSegregation analysis of prostate cancer in Sweden: support for dominant inheritanceAm J Epidemiol199714675525579326432
  • MacInnisRJAntoniouACEelesRAProstate cancer segregation analyses using 4390 families from UK and Australian population-based studiesGenet Epidemiol2010341425019492347
  • PakkanenSBaffoe-BonnieABMatikainenMPSegregation analysis of 1,546 prostate cancer families in Finland shows recessive inheritanceHum Genet2007121225726717203302
  • RischNThe genetic epidemiology of cancer: interpreting family and twin studies and their implications for molecular genetic approachesCancer Epidemiol Biomarkers Prev200110773374111440958
  • EastonDFSchaidDJWhittemoreASIsaacsWJInternational Consortium for Prostate Cancer GeneticsWhere are the prostate cancer genes? – A summary of eight genome wide searchesProstate200357426126914601022
  • FoulkesWDInherited susceptibility to common cancersN Engl J Med2008359202143215319005198
  • LangebergWJIsaacsWBStanfordJLGenetic etiology of hereditary prostate cancerFront Biosci2007124101411017485361
  • NupponenNNCarptenJDProstate cancer susceptibility genes: many studies, many results, no answersCancer Metastasis Rev2001203–415516412085959
  • OstranderEAMarkianosKStanfordJLFinding prostate cancer susceptibility genesAnnu Rev Genomics Hum Genet2004515117515485346
  • PomerantzMMFreedmanMLGenetics of prostate cancer riskMt Sinai J Med201077664365421105126
  • VerhageBAKiemeneyLAInherited predisposition to prostate cancerEur J Epidemiol200318111027103614620936
  • EelesRAKote-JaraiZAl OlamaAAUK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons’ Section of OncologyUK ProtecT Study CollaboratorsPRACTICAL ConsortiumIdentification of seven new prostate cancer susceptibility loci through a genome-wide association studyNat Genet200941101116112119767753
  • EelesRAKote-JaraiZGilesGGUK Genetic Prostate Cancer Study CollaboratorsBritish Association of Urological Surgeons’ Section of OncologyUK ProtecT Study CollaboratorsMultiple newly identified loci associated with prostate cancer susceptibilityNat Genet200840331632118264097
  • EelesRAOlamaAABenllochSCOGS–Cancer Research UK GWAS–ELLIPSE (part of GAME-ON) InitiativeAustralian Prostate Cancer BioresourceUK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons’ Section of OncologyUK ProtecT (Prostate testing for cancer and Treatment) Study CollaboratorsPRACTICAL (Prostate Cancer Association Group to Investigate Cancer-Associated Alterations in the Genome) ConsortiumIdentification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping arrayNat Genet2013454385391391e123535732
  • Kote-JaraiZEastonDFStanfordJLPRACTICAL ConsortiumMultiple novel prostate cancer predisposition loci confirmed by an international study: the PRACTICAL ConsortiumCancer Epidemiol Biomarkers Prev20081782052206118708398
  • Kote-JaraiZOlamaAAGilesGGSeven prostate cancer susceptibility loci identified by a multi-stage genome-wide association studyNat Genet201143878579121743467
  • Kote-JaraiZSaundersEJLeongamornlertDACOGS-CRUK GWAS-ELLIPSE (Part of GAME-ON) InitiativeUK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons’ Section of OncologyUK ProtecT Study CollaboratorsPRACTICAL ConsortiumFine-mapping identifies multiple prostate cancer risk loci at 5p15, one of which associates with TERT expressionHum Mol Genet201322122520252823535824
  • LuLCancel-TassinGValeriAInternational Consortium for Prostate Cancer GeneticsChromosomes 4 and 8 implicated in a genome wide SNP linkage scan of 762 prostate cancer families collected by the ICPCGProstate201272441042621748754
  • JinGLuLCooneyKAInternational Consortium for Prostate Cancer GeneticsValidation of prostate cancer risk-related loci identified from genome-wide association studies using family-based association analysis: evidence from the International Consortium for Prostate Cancer Genetics (ICPCG)Hum Genet201213171095110322198737
  • TeerlinkCCThibodeauSNMcDonnellSKInternational Consortium for Prostate Cancer GeneticsAssociation analysis of 9,560 prostate cancer cases from the International Consortium of Prostate Cancer Genetics confirms the role of reported prostate cancer associated SNPs for familial diseaseHum Genet2014133334735624162621
  • LangeEMHoLABeebe-DimmerJLGenome-wide linkage scan for prostate cancer susceptibility genes in men with aggressive disease: significant evidence for linkage at chromosome 15q12Hum Genet2006119440040716508751
  • SchaidDJMcDonnellSKZarfasKEInvestigators of the International Consortium for Prostate Cancer GeneticsPooled genome linkage scan of aggressive prostate cancer: results from the International Consortium for Prostate Cancer GeneticsHum Genet2006120447148516932970
  • SlagerSLSchaidDJCunninghamJMConfirmation of linkage of prostate cancer aggressiveness with chromosome 19qAm J Hum Genet200372375976212563560
  • SlagerSLZarfasKEBrownWMGenome-wide linkage scan for prostate cancer aggressiveness loci using families from the University of Michigan Prostate Cancer Genetics ProjectProstate200666217317916173044
  • StanfordJLMcDonnellSKFriedrichsenDMProstate cancer and genetic susceptibility: a genome scan incorporating disease aggressivenessProstate200666331732516245279
  • GoddardKAWitteJSSuarezBKCatalonaWJOlsonJMModel-free linkage analysis with covariates confirms linkage of prostate cancer to chromosomes 1 and 4Am J Hum Genet20016851197120611309685
  • SchaidDJStanfordJLMcDonnellSKGenome-wide linkage scan of prostate cancer Gleason score and confirmation of chromosome 19qHum Genet2007121672973517486369
  • WitteJSGoddardKAContiDVGenomewide scan for prostate cancer-aggressiveness lociAm J Hum Genet2000671929910825281
  • EwingCMRayAMLangeEMGermline mutations in HOXB13 and prostate-cancer riskN Eng J Med20123662141149
  • LangeEMGillandersEMDavisCCGenome-wide scan for prostate cancer susceptibility genes using families from the University of Michigan prostate cancer genetics project finds evidence for linkage on chromosome 17 near BRCA1Prostate200357432633414601029
  • PenneyKLSalinasCAPomerantzMEvaluation of 8q24 and 17q risk loci and prostate cancer mortalityClin Cancer Res20091593223323019366828
  • GillandersEMXuJChangBLCombined genome-wide scan for prostate cancer susceptibility genesJ Natl Cancer Inst200496161240124715316059
  • XuJDimitrovLChangBLACTANE ConsortiumA combined genomewide linkage scan of 1,233 families for prostate cancer-susceptibility genes conducted by the international consortium for prostate cancer geneticsAm J Hum Genet200577221922915988677
  • LangeEMRobbinsCMGillandersEMFine-mapping the putative chromosome 17q21-22 prostate cancer susceptibility gene to a 10 cM region based on linkage analysisHum Genet20071211495517120048
  • LangeEMChenHBrierleyKLinkage analysis of 153 prostate cancer families over a 30-cM region containing the putative susceptibility locus HPCXClin Cancer Res19995124013402010632333
  • Stott-MillerMKaryadiDMSmithTHOXB13 mutations in a population-based, case-control study of prostate cancerProstate201373663464123129385
  • WitteJSMeffordJPlummerSJHOXB13 mutation and prostate cancer: studies of siblings and aggressive diseaseCancer Epidemiol Biomarkers Prev201322467568023396964
  • BreyerJPAvrittTGMcReynoldsKMDupontWDSmithJRConfirmation of the HOXB13 G84E germline mutation in familial prostate cancerCancer Epidemiol Biomarkers Prev20122181348135322714738
  • XuJLangeEMLuLInternational Consortium for Prostate Cancer GeneticsHOXB13 is a susceptibility gene for prostate cancer: results from the International Consortium for Prostate Cancer Genetics (ICPCG)Hum Genet2013132151423064873
  • MacInnisRJSeveriGBagliettoLPopulation-based estimate of prostate cancer risk for carriers of the HOXB13 missense mutation G84EPLoS One201382e5472723457453
  • KarlssonRAlyMClementsMA population-based assessment of germline HOXB13 G84E mutation and prostate cancer riskEur Urol201465116917622841674
  • SchroeckFRZuhlkeKASiddiquiJSiddiquiRCooneyKAWeiJTTesting for the recurrent HOXB13 G84E germline mutation in men with clinical indications for prostate biopsyJ Urol2013189384985323036981
  • ChenZGreenwoodCIsaacsWBThe G84E mutation of HOXB13 is associated with increased risk for prostate cancer: results from the REDUCE trialCarcinogenesis20133461260126423393222
  • LinXQuLChenZA novel germline mutation in HOXB13 is associated with prostate cancer risk in Chinese menProstate201373216917522718278
  • McGinnisWKrumlaufRHomeobox genes and axial patterningCell19926822833021346368
  • Garcia-FernàndezJThe genesis and evolution of homeobox gene clustersNat Rev Genet200561288189216341069
  • ZeltserLDesplanCHeintzNHoxb-13: A new Hox gene in a distant region of the HOXB cluster maintains colinearityDevelopment19661228247524848756292
  • SreenathTOroszAFujitaKBieberichCJAndrogen-independent expression of hoxb-13 in the mouse prostateProstate199941320320710517879
  • EconomidesKDCapecchiMRHoxb13 is required for normal differentiation and secretory function of the ventral prostateDevelopment2003130102061206912668621
  • JungCKimRSZhangHJLeeSJJengMHHOXB13 induces growth suppression of prostate cancer cells as a repressor of hormone-activated androgen receptor signalingCancer Res200464249185919215604291
  • NorrisJDChangCYWittmannBMThe homeodomain protein HOXB13 regulates the cellular response to androgensMol Cell200936340541619917249
  • KimSDParkRYKimYRHOXB13 is co-localized with androgen receptor to suppress androgen-stimulated prostate-specific antigen expressionAnat Cell Biol201043428429321267402
  • JungCKimRSLeeSJWangCJengMHHOXB13 homeodomain protein suppresses the growth of prostate cancer cells by the negative regulation of T-cell factor 4Cancer Res20046493046305115126340
  • HamidSMCicekSKaramilSHOXB13 contributes to G1/S and G2/M checkpoint controls in prostateMol Cell Endocrinol20143831–2384724325868
  • McMullinRPDobiAMuttonLNA FOXA1-binding enhancer regulates Hoxb13 expression in the prostate glandProc Natl Acad Sci U S A201010719810320018680
  • HuangHCaiBG84E mutation in HOXB13 is firmly associated with prostate cancer risk: a meta-analysisTumour Biol20143521177118224026887
  • RenBYuGTsengGCMCM7 amplification and overexpression are associated with prostate cancer progressionOncogene20062571090109816247466
  • LiuZRenGShangguanCATRA inhibits the proliferation of DU145 prostate cancer cells through reducing the methylation level of HOXB13 genePLoS One201277e4094322808286
  • KimYRKimIJKangTWHOXB13 downregulates intracellular zinc and increases NF-κB signaling to promote prostate cancer metastasisOncogene Epub1072013
  • EdwardsSCampbellCFlohrPExpression analysis onto microarrays of randomly selected cDNA clones highlights HOXB13 as a marker of human prostate cancerBr J Cancer200592237638115583692
  • JeongTOOhKJXuan NguyenNTEvaluation of HOXB13 as a molecular marker of recurrent prostate cancerMol Med Rep20125490190422293681
  • JungCKimRSZhangHHOXB13 is downregulated in colorectal cancer to confer TCF4-mediated transactivationBr J Cancer200592122233223915928669
  • OkudaHToyotaMIshidaWEpigenetic inactivation of the candidate tumor suppressor gene HOXB13 in human renal cell carcinomaOncogene200625121733174216278676
  • YamashitaTTazawaSYaweiZSuppression of invasive characteristics by antisense introduction of overexpressed HOX genes in ovarian cancer cellsInt J Oncol200628493193816525643
  • MiaoJWangZProvencherHHOXB13 promotes ovarian cancer progressionProc Natl Acad Sci U S A200710443170931709817942676
  • LópezRGarridoEPiñaPHOXB homeobox gene expression in cervical carcinomaInt J Gynecol Cancer200616132933516445654
  • MaXJWangZRyanPDA two-gene expression ratio predicts clinical outcome in breast cancer patients treated with tamoxifenCancer Cell20045660761615193263
  • JerevallPLBrommessonSStrandCExploring the two-gene ratio in breast cancer – independent roles for HOXB13 and IL17BR in prediction of clinical outcomeBreast Cancer Res Treat2008107222523417453342
  • ShahNJinKCruzLAHOXB13 mediates tamoxifen resistance and invasiveness in human breast cancer by suppressing ERα and inducing IL-6 expressionCancer Res201373175449545823832664
  • WilliamsTMWilliamsMEInnisJWRange of HOX/TALE superclass associations and protein domain requirements for HOXA13:MEIS interactionDev Biol2005277245747115617687