WO2012071665A1 - Prognostic markers of inherited variations in the ugt2b genes for prostate cancer recurrence - Google Patents

Prognostic markers of inherited variations in the ugt2b genes for prostate cancer recurrence Download PDF

Info

Publication number
WO2012071665A1
WO2012071665A1 PCT/CA2011/001338 CA2011001338W WO2012071665A1 WO 2012071665 A1 WO2012071665 A1 WO 2012071665A1 CA 2011001338 W CA2011001338 W CA 2011001338W WO 2012071665 A1 WO2012071665 A1 WO 2012071665A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
prostate cancer
risk
genes
ugt2b
Prior art date
Application number
PCT/CA2011/001338
Other languages
French (fr)
Inventor
Eric Levesque
Chantal Guillemette
Yves Fradet
Louis Lacombe
Original Assignee
Universite Laval
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universite Laval filed Critical Universite Laval
Publication of WO2012071665A1 publication Critical patent/WO2012071665A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the present invention relates to the use of markers to identify relevant patterns of inherited variation in genes involved in sex-steroid metabolism, and more particularly the UGT2B gene, optionally with the SRD5A gene, for use in prognosticating or predicting the recurrence of prostate cancer.
  • PCa Prostate cancer
  • Prostate cancer an androgen- and estrogen-related cancer
  • PCa is the sixth most common cancer in the world and the second leading cause of cancer death in North American men.
  • the specific mortality of PCa is directly related to both the stage at time of diagnosis and age at onset.
  • age, ethnicity, genetic and dietary factors are known.
  • androgens are centrally implicated in the development and progression of this hormone- dependent cancer.
  • an association between plasma androgen levels and PCa susceptibility has not been consistently observed across studies.
  • PCa recurrence risk is assessed based on clinical stage, biochemical prostate-specific antigen (PSA) level, and Gleason scores.
  • PSA prostate-specific antigen
  • Gleason scores are assessed based on clinical stage, biochemical prostate-specific antigen (PSA) level, and Gleason scores.
  • Patients with localized (clinical stage T1 -T2) and locally advanced (T3) PCa are frequently treated with radical prostatectomy (RP), a potentially curative procedure.
  • RP radical prostatectomy
  • BCR biochemical recurrence
  • TMM Tumor/Nodes/Metastasis
  • PSA serum prostate-specific antigen
  • Androgen hormones such as testosterone (T) and 5a-dihydrotestosterone (DHT) have been clearly implicated in development of PCa. Circulating T, secreted by the testis, and adrenal steroid precursors are among factors influencing androgen levels in the prostate. Indeed, the conversion of T by 5a-reductases (SRD5A1 and SRD5A2) leads to DHT, the more potent androgen receptor (AR) endogenous agonist in target cells.
  • SRD5A1 and SRD5A2 5a-reductases
  • SRD5A2 is the major 5a-reductase enzyme expressed in the prostate compared to SRD5A1 1 . However, while the expression of SRD5A2 decreases in prostate cancer cells, SRD5A1 is increased in tumoral tissues 1"3 . This imbalance in the expression of SRD5A genes in PCa tumors illustrates the complex relation between 5a-reductases, DHT synthesis and PCa progression. In concert with SRD5A genes, the active androgen level in target cells is controlled by steroid metabolizing UDP-glucuronosyltransferases 2B genes (UGT2B). These phase II metabolism enzymes conjugate androgens, and thus contribute to the irreversible transformation of steroids into biologically inactive
  • ADT Androgen deprivation therapy
  • RP primary therapy
  • UDP-glucuronosyltransferases are conjugating enzymes that, in concert with other steroidogenesis enzymes, contribute to the maintenance of intracellular levels of sex-steroid hormones in target cells. This enzymatic process generates glucuronide derivatives that are biologically inactive and are subsequently excreted in bile or urine.
  • UGTs are expressed in almost all tissues, including hormone target organs such as the prostate. Five UGT isoforms are mainly responsible for inactivating androgens and estrogens. The UGT1A1 isoform is primarily involved in the glucuronidation of estradiol, whereas the UGT2B class of enzymes (namely UGT2B7, UGT2B15, UGT2B17, and UGT2B28) is mainly involved in androgen catabolism in hormone-dependent cells.
  • the present invention identifies germline polymorphic CNV events in UGT2B17 and UGT2B28 that are significantly associated with biochemical recurrence in clinically localized PCa after surgical treatment. These deletions are linked to altered levels of circulating sex steroids, supporting the hypothesis that reduced androgen glucuronidation by UGT-mediated inactivation leads to an increased PSA recurrence.
  • Our data indicates that co-occurrence of deleted copies of the UGT2B17 and UGT2B28 genes are important predictors for PSA recurrence independently of the well-established clinico-pathological markers; Gleason score, PSA values and stages at the time of diagnosis.
  • SRD5A genes are also well recognized molecular targets in PCa prevention trials 7,35 .
  • SRD5A1 and SRD5A2 genes in relation to PCa progression are presented. The association between the known SRD5A2 V89L polymorphism and BCR as proposed by other groups 12,13 is also described.
  • the association between inherited genetic alterations in androgen metabolizing genes and the risk of relapse was determined after RP as the sole initial curative intent.
  • the clinical consequence of simultaneous occurrence of variations in the androgen biosynthetic SRD5A genes and in UGT2B sex- steroid inactivating genes is used to improve the statistical significance of the method for predicting the likelihood of a human subject to develop PCa recurrence or the risk of relapse after RP.
  • a biomarker for predicting the likelihood of prostate cancer development, its progression or recurrence in a human subject comprising the identification of at least one copy number variation(s) in at least one of a UGT2B gene.
  • the invention provides the identification of CNV(s) in at least one of a UGT2B gene of a human subject for predicting the likelihood of prostate cancer, its progression or recurrence in said human subject.
  • an in-vitro method for predicting risk for PCa in a human subject comprising the steps of: a) obtaining a nucleic acid from a non-tumor or tumor sample from said human subject; and b) determining an individual's genetic variation (or haplotypes, CNV or deletion) in a UGT2B gene; whereby the identification of at least one genetic variation in said UGT2B gene in said subject's nucleic acid is an indication that said subject has an increased risk of developing prostate cancer or of experiencing progression thereof.
  • UGT2B risk alleles may be defined as one deletion of UGT2B28 or UGT2B17 and/or two or more deletions in UGT genes.
  • the invention further provides the method as defined herein wherein said identification of genetic variation in said UGT2B17 or UGT2B28 comprises identifying at least one CNV in said nucleic acids, said CNV being selected from the group consisting of: rs3100645; and rs1 1249532; and associated reference sequences thereof.
  • the method of the invention further comprises the identification of the cooccurrence of a genetic variation (or haplotype) in SRD5A1 or SRD5A2 gene; whereby the co-occurrence of at least one genetic variation in UGT and SRD5A1 or SRD5A2 in said subject's nucleic acid is an indication that said subject has a further increased likelihood that the PCa will recur.
  • a genetic variation or haplotype
  • the invention further provides the method as defined herein wherein said identification of genetic variation in said SRD5A1 or SRD5A2 comprises identifying at least one single nucleotide polymorphism (SNP) in said nucleic acids, said SNP being selected from the group consisting of: rs166050; rs2208532; rs523349; rs4952197;
  • SNP single nucleotide polymorphism
  • the present invention also provides a method for adapting a course of treatment of prostate cancer in a human subject, comprising the steps of: a) providing a prognosis or predicting the likelihood of a human subject to develop prostate cancer recurrence in accordance with the method as defined herein; and b) adapting a course of treatment according to whether said subject has an increased likelihood that the cancer will recur. Particularly, adaptation of course of treatment is realized after the subject has undergone radical prostatectomy.
  • the invention further provides a kit for predicting the likelihood of a human subject to develop prostate cancer recurrence by detecting a CNV in a nucleic acid sample, the kit comprising reagents for determining the individual's genetic variations (or haplotypes) in UGT2B17 and UGT2B28 gene.
  • the kit comprises a reagent for detecting a reference sequence selected from the group consisting of: rs3100645; and rs1 1249532, and associated reference sequences thereof.
  • the kit further comprises reagents for determining the individual's genetic variations (or haplotypes) in SRD5A1 or SRD5A2 gene.
  • the kit further comprises reagents for detecting a SNP in a reference sequence selected from the group consisting of: rs518673; rs166050; rs12470143; rs2208532; rs523349; rs4952197 and rs676033 or their associated SNPs.
  • Figure 1 PCR amplifications of the UGT2B17 and UGT2B28 genes.
  • FIG. Kaplan-Meier curves of Biochemical Recurrence-Free Survival (bRFS) for copy-number variations of UGT2B genes.
  • FIG. 3 Kaplan-Meier curves of bRFS for co-occurrence of UGT2B risk allele.
  • UGT2B risk alleles were defined as 1 deletion of UGT2B28 and/or two or more deletions in UGT genes.
  • UGT2B risk alleles were defined as two or more deletions in UGT genes.
  • Figure 4. Association analysis of common UGT2B17 and UGT2B28 gene deletions and biochemical failure in additional cohorts of PCa cases. Odds ratios and confidence intervals were calculated to combine data from the 1 ,161 PCa cases.
  • UGT2B risk alleles were defined as 1 deletion of UGT2B28 and/or two or more deletions in UGT genes.
  • Figure 5 Prognostic impact of inclusion of genetic markers, UGT2B17 and UGT2B28 gene deletions, in the evaluation of the D'Amico risk classification of PCa recurrence after RP.
  • Figure 6. Kaplan-Meier Biochemical Recurrence-Free Survival Analysis for other UGT genes.
  • Figure 7 Risk of recurrence associated with known clinical and pathological prognostic variables (A) and SRD5A genes (B). Boxes represent hazard ratios (HR) and their 95% CI. PSA categories are in ng/ml. Reference categories (HR: 1.00) are: PSA at diagnosis ⁇ 10ng/ml, pG ⁇ 6, and pT ⁇ T2b. Genetic linkage between htSNPs tested for each SRD5A gene is represented in the triangles on the left in panel B.
  • Figure 8 Kaplan-Meier estimates of recurrence-free survival for A) SRD5A2, B) SRD5A 1 and C) protective htSNPs in both SRD5A genes. Log rank (LR) P values are shown in each frame.
  • SRD5A2 genes stratified by status for UGT2B risk alleles. Boxes represent hazard ratios (HR) and their 95% CI in multivariate analysis. Black boxes were analyzed only for SRD5A genotypes, white boxes are for SRD5A minor alleles + no UGT2B risk alleles, and grey boxes are for SRD5A minor alleles + UGT2B risk alleles. Boxes indicate HR and error bars indicate the 95% confidence intervals. The reference category is set at one. Models used for multivariate analysis are indicated in Table 13.
  • ADT androgen-deprivation therapy
  • AR androgen receptor
  • BCR biochemical recurrence
  • bRFS biochemical recurrence-free survival
  • CNV copy number variation
  • DHT 5a-dihydrotestoterone
  • HR hazard ratio
  • htSNP haplotype-tagging SNP
  • MAF minor allele frequencies
  • PCa prostate cancer
  • PSA prostate-specific antigen
  • SRD5A1 5a-reductase type 1
  • SRD5A2 5a-reductase type 2
  • RP radical prostatectomy
  • SNP Single nucleotide polymorphism
  • T Testosterone
  • TNM Tumor/Nodes/Metastasis staging system
  • UGT UDP-glucuronosyltransferase.
  • identify means the act of searching for and finding a specific marker.
  • Other terms that may be used for the same purpose are: “detect”, “locate”, “expose”, “observe”, “recognize”, “uncover” or “reveal”.
  • development or “developing” when used in the context of cancer means the development, progression, recurrence, biochemical recurrence, death from or metastasis of such a cancer.
  • sample means a patient or subject's nucleic acid- containing sample originating indistinctly from a tumor or a non-tumor sample.
  • the subject's non-tumor sample can be selected from: tissue, or biological fluid.
  • tissue can be selected from the group consisting of: skin, lymph node, hair and buccal smear.
  • the biological fluid can be selected from the group consisting of: sputum, saliva, blood, serum, urine, semen and plasma.
  • the non-tumor or tumor sample may originate from a biopsy.
  • CNVs Copy-number variations
  • a form of structural variation consisting in alterations of the DNA of a genome that results in the cell having an abnormal number of copies of one or more sections of the DNA.
  • CNVs correspond to relatively large regions of the genome that have been deleted (fewer than the normal number) or duplicated (more than the normal number) on certain chromosomes.
  • the chromosome that normally has sections in order as A-B-C-D might instead have sections A-B-C-C-D (a duplication of "C") or A-B-D (a deletion of "C”).
  • SNPs single-nucleotide polymorphisms
  • an in-vitro method for predicting the risk for PCa in a human subject comprising the steps of: a) obtaining a nucleic acid from a non-tumor or tumor sample from said human subject; and b) determining the individual's genetic variation status such as for example a copy number variation in UGT2B17 gene and/or UGT2B28 gene; whereby the determination of at least one genetic variation in one of the UGT2B17 or UGT2B28 gene is an indication that said subject has an increased likelihood that the prostate cancer will recur.
  • the invention further provides the method as defined herein wherein said identification of genetic variation status in said UGT2B17 or UGT2B28 comprises identifying at least one copy number variation (CNV) in said nucleic acids, said CNV being selected from the group consisting of: rs3100645; and rs1 1249532.
  • CNV copy number variation
  • the genetic variation in UGT2B17 is a copy number variation consisting of one or two deletions.
  • the genetic variation in UGT2B28 is a copy number variation consisting of one or two deletions.
  • the method of the invention comprises the identification of the co-occurrence of the genetic variations in UGT2B17 and UGT2B28 genes, e.g. when both UGT2B17 and UGT2B28 genes harbor at least one CNV selected from the group consisting of: rs3100645; and rs 1249532.
  • each of UGT2B17 and UGT2B28 can harbor one or two deletions.
  • the method of the invention further comprises the identification of the co- occurrence of a genetic variation in SRD5A1 or SRD5A2 gene; whereby the cooccurrence of at least one genetic variation in UGT2B and either SRD5A1 or SRD5A2 or both in said subject's nucleic acid is an indication that said subject has a further increased likelihood that the prostate cancer will recur.
  • the invention further provides the method as defined herein wherein said identification of genetic variation in said SRD5A1 or SRD5A2 comprises identifying at least one single nucleotide polymorphism (SNP) in said nucleic acids, said SNP being selected from the group consisting of: rs166050; rs2208532; rs523349; rs4952197;
  • SNP single nucleotide polymorphism
  • the invention also provides the method as defined herein, wherein the subject's nucleic acid-containing sample is a tumor or a non-tumor sample.
  • the subject's non-tumor sample is selected from: tissue, or biological fluid.
  • tissue is selected from the group consisting of: skin, lymph node, hair and buccal smear.
  • the biological fluid is selected from the group consisting of: sputum, saliva, blood, serum, urine, semen and plasma.
  • the non-tumor or tumor sample originates from a biopsy.
  • the invention also provides the method as defined herein wherein in step a), the nucleic acid is DNA or RNA.
  • the DNA and the marker is detected with the use of a nucleic acid primer. More particularly, the DNA is amplified by PCR.
  • the genetic variation of DNA or RNA is detected with the use of a methodology selected from the group consisting of: direct sequencing, pyrosequencing, massively parallel sequencing, high-throughput sequencing, high performance liquid chromatography (HPLC) fragment analysis, mass spectrometry, branch DNA and capillary electrophoresis.
  • the DNA or RNA of the marker is detected with the use of a nucleic acid primer and probe combination.
  • the DNA or RNA is amplified prior to incubation with the probe, such as by PCR. More particularly, amplified PCR products and the genetic variation are analyzed by mass spectrometry.
  • the present invention also provides for a method for adapting a course of treatment of prostate cancer in a human subject, comprising the steps of: a) predicting the likelihood of prostate cancer progression or recurrence in a human subject as defined herein; and b) adapting a course of treatment according to whether said subject has an increased or decreased likelihood that the cancer will progress, or recur.
  • the subject when the likelihood of recurrence is increased, the subject is prescribed at least one of: hormone therapy, chemotherapy, surgery, radiotherapy, brachytherapy or active surveillance (watchful waiting). More particularly, steps a) and b) are carried out after the subject has undergone radical prostatectomy.
  • the invention also provides the method as defined herein, wherein, when the likelihood of recurrence is increased, the subject is prescribed inducer (or inductive) UGT2B therapy and/or 5a-reductase inhibitors therapy.
  • the present invention provides for a kit for predicting the likelihood of a human subject to develop prostate cancer or a recurrence thereof, said kit comprising reagents for determining the individual's genetic variations (or haplotypes) in the UGT2B gene.
  • the kit comprises reagents for detecting a CNV in a reference sequence selected from the group consisting of: rs3100645 and/or rs1 1249532; and associated reference sequences thereof.
  • the invention also provides the kit as defined herein, comprising a PCR primer, wherein the primer is selected from the group consisting of: SEQ ID Nos. 1 , 2, 7 and 8. Additionally, the kit may comprise a PCR probe, wherein the probe hybridizes with a sequence selected from rs3100645 and/or rs1 1249532; and associated reference sequences thereof.
  • the kit further comprises reagents for determining the individual's genetic variations (or haplotypes, SNP) in SRD5A1 and/or SRD5A2 genes, such as reagents for detecting a SNP in a reference sequence selected from the group consisting of: rs518673; rs166050; rs12470143; rs2208532; rs523349; rs4952197 and rs676033 or their associated SNPs.
  • reagents for determining the individual's genetic variations (or haplotypes, SNP) in SRD5A1 and/or SRD5A2 genes such as reagents for detecting a SNP in a reference sequence selected from the group consisting of: rs518673; rs166050; rs12470143; rs2208532; rs523349; rs4952197 and rs676033 or their associated SNPs.
  • the invention further provides the kit as defined herein, comprising PCR primer- probe set for determining the individual's SNPs in SRD5A1 and/or SRD5A2 genes, wherein the primer is selected from the group consisting of: SEQ ID Nos. 12 to 50; and the probe is selected from the group consisting of: SEQ ID Nos. 51 to 69.
  • the kit as defined herein, comprising PCR primer- probe set for determining the individual's SNPs in SRD5A1 and/or SRD5A2 genes, wherein the primer is selected from the group consisting of: SEQ ID Nos. 12 to 50; and the probe is selected from the group consisting of: SEQ ID Nos. 51 to 69.
  • the primary outcome variable was biochemical recurrence-free survival (bRFS), also called 'PSA failure' or 'biochemical relapse'.
  • bRFS biochemical recurrence-free survival
  • bRFS was defined as (1 ) two consecutive PSA values > 0.3 pg/L, (2) one PSA value ⁇ 0.3 pg/L followed by hormonotherapy (androgen deprivation therapy or ADT) or radiation therapy, and (3) a single last-recorded PSA value > 0.3 pg/L after prostatectomy.
  • bRFS was defined as the period of time elapsed between the surgical procedure and the date of PSA failure.
  • cohorts C and D the bRFS was defined as previously described. 8 36
  • Common amplification steps used are: one heating step at 95° C for 30 sec, 40 cycles of denaturing at 95°C, annealing and elongation at 72°C and a single final extension step of 7 min at 72°C (see Figure 1 and Table 1 ).
  • Amplification of the exon 6 can be performed to confirm the absence of the studied gene (UGT2B17 and UGT2B28) if deemed necessary, in case of uncertainties only. With these genotyping strategies, we were able to detect precisely the number of deletions (from 0 to 4).
  • Kaplan-Meier analysis for bRFS analysis was performed to compare genotypes using the log-rank test followed by a pairwise multiple comparison using the Sidak multiple-comparison method. Results that were significantly associated with bRFS in univariate analysis were then evaluated in a multivariate Cox regression analysis.
  • UGT2B28 Haplotype analysis was performed for cohort A only. After this initial screen, the other genetic variables were not further studied in other cohorts due to lack of association. Clinical variables were compared across genotypes using the chi-square test, Fisher's exact test evaluated exactly, or the Monte Carlo approximation, as appropriate. A p value of less than 0.05 was considered statistically significant, and all tests were two- sided. All statistical analyses were performed using SAS® Statistical Software version 9.2 (SAS Institute, Cary, NC, USA).
  • Figure 2 displays the Kaplan-Meier curves of bRFS for UGT2B17 and UGT2B28 genes for all studied patients for whom this analysis was achievable (cohorts A, B and C).
  • Individuals of Asian Ancestry (cohort C) were included in our study because they have the highest reported frequencies of UGT2B deletions.
  • Polymorphisms in SRD5A genes were chosen according to one or more of the following criteria: i) to be likely functional (with supportive data in the literature), ii) to have previously been associated with PCa risk, aggressiveness, age at onset, BCR or ADT efficiency, and iii) to explain most of the haplotype diversity in the CEU (Utah residents with Northern and Western European Ancestry) Hapmap population.
  • Genomic DNA was purified using the QIAamp DNA Blood Mini Kit (Qiagen Inc., Mississauga, On, Canada) and stored at -20°C.
  • Qiagen Inc. Mississauga, On, Canada
  • PCR amplifications were performed using Sequenom iPLEX matrix- assisted laser desorption/ionization-time-of-flight mass spectrometry.
  • Negative controls were present for every run of analyses and quality controls (random replicates of known genotypes) were successfully performed in 5% of the study cohort.
  • each htSNP was considered using 3 models since the function of most htSNPs remains unknown.
  • the genomic model considered the htSNP as a categorical variable with a common allele homozygote, heterozygote and a minor allele homozygote.
  • the dominant model considered the htSNP with only 2 categories: one with a common allele homozygote and one with at least one minor allele.
  • the recessive model also considered the htSNP with only 2 categories: one with at least one common allele and one minor allele homozygote.
  • Cox regression was performed on each htSNP considering the 3 above mentioned models with adjustment for clinicopathological variables: PSA level at diagnosis, age at diagnosis, smoking status, pathological Gleason grade, pathological stage and neoadjuvant ADT. All co-variables were treated as categorical, and for PSA level, Gleason scores and stage, they were used as described by the D'Amico risk classification.
  • the censoring variable was BCR, which was defined as 1 ) two consecutive PSA values > 0.3 pg/L, 2) one PSA value > 0.3 pg/L followed by ADT, and 3) a last-recorded PSA value > 0.3 pg/L after prostatectomy.
  • htSNPs were selected with a strategy to maximize gene coverage and to reflect adequately the Caucasian haplotype genetic diversity.
  • the htSNPs strategy allowed us to study 109 genetic variations in both genes (Table 13).
  • V89L polymorphism One of the best characterized SNP in the SRD5A2 gene, the functional V89L polymorphism, is associated with a decreased enzymatic activity. 44,45 This polymorphism has been associated with prostate cancer risk in some studies 32,34 agressiveness 32 33 , and with conflicting results with biochemical recurrence 12 13 .
  • One important finding of our study is a significant association between the V89L (rs523349) polymorphism and the risk of BCR after prostatectomy (2.1 -fold risk of recurrence for homozygotes rs523349C). This result is in agreement with Shibata and colleagues 27 and with a recent large study that associated this SNP with an aggressive form of PCa 33 .
  • this data set included a large sample size combined with relevant information on clinical and pathological covariates as well as a significant median follow- up time of 7.4 years.
  • relevant information on clinical and pathological covariates included a large sample size combined with relevant information on clinical and pathological covariates as well as a significant median follow- up time of 7.4 years.
  • some clinically relevant events such as metastasis, androgen-deprivation resistance, or death related to the localized features of the tumors, prevented us from addressing the association between inherited variations in candidate genes and risk for these events.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present application discloses that reduced copy number variations (CNVs) in UGT2B17 and UGT2B28 were significantly associated with bRFS while no significant association was observed for additional UGT variants tested. The co-occurrence of two or more deletions of UGT2B17 and UGT2B28 genes resulted in an HR of 1.377 (95% CI [1.105; 1.716]; p=0.004) for Caucasians and 2.151 (95% CI [1.239; 3.372]; p=0.006) for Asians. In all studied men, risk of BCR was 1.510; 95% CI [1.178 - 1.950]; p=0.001 ). The addition of the UGT2B CNV status in the D'Amico risk classification further enhanced the risk prediction mainly for the low and intermediate risk categories (Log-rank test, p=0.022 and 0.011, respectively). In addition, after adjusting for all clinicopathologic risk factors, we found a strong association between the risk of BCR and 7 SNPs in SRD5A genes. The combination of 2 SNPs respectively in SRD5A1 and SRD5A2 were highly favourable, reducing drastically the risk of BCR for carriers of 3-4 alleles (HR=0.34; 95% CN0.18- 0.64; P=9x10"4). Other variations in the SRD5A2 gene were associated with an increased rate of BCR, namely the coding SNPs rs523349 with a HR of 2.12 (95% CI, 1.21 -3.75; P=0.009) and reaching 4.97 when combined with deleted copies of UGT2B genes (95% CI, 2.38-10.36; P=0.00002). Further, BCR-free survival rate was significantly reduced to 27% in patients with unfavourable genotypes compared to 75% for patients with favourable genotypes (P=7x10-6). This study is the first to recognize CNV in genes at the end of the androgenic signal as significant and independent predictors of PCa recurrence after prostatectomy. In addition, inherited polymorphisms in the SRD5A and UGT2B genes are independent predictors of biochemical recurrence after radical prostatectomy.

Description

Prognostic Markers of Inherited variations in the UGT2B genes for Prostate
Cancer Recurrence
Cross-reference to related applications [0001] This application claims priority from US provisional patent application 61/419,057 filed on December 2, 2010, the content of which is incorporated by reference in its entirety.
Field of the invention
[0002] The present invention relates to the use of markers to identify relevant patterns of inherited variation in genes involved in sex-steroid metabolism, and more particularly the UGT2B gene, optionally with the SRD5A gene, for use in prognosticating or predicting the recurrence of prostate cancer.
Background of the invention
[0003] Prostate cancer (PCa), an androgen- and estrogen-related cancer, is the sixth most common cancer in the world and the second leading cause of cancer death in North American men. The specific mortality of PCa is directly related to both the stage at time of diagnosis and age at onset. Of the several known risk factors, the most significant are age, ethnicity, genetic and dietary factors. In the early stages of carcinogenesis, androgens are centrally implicated in the development and progression of this hormone- dependent cancer. However, an association between plasma androgen levels and PCa susceptibility has not been consistently observed across studies.
[0004] Currently, PCa recurrence risk is assessed based on clinical stage, biochemical prostate-specific antigen (PSA) level, and Gleason scores. Patients with localized (clinical stage T1 -T2) and locally advanced (T3) PCa are frequently treated with radical prostatectomy (RP), a potentially curative procedure. However, it is estimated that over 30% of men undergoing RP will have disease relapse, also referred to as biochemical recurrence (BCR) as the first clinical indication of rising serum level of PSA .
[0005] Currently, the Tumor/Nodes/Metastasis (TNM) staging system, the Gleason score, and pre-treatment serum prostate-specific antigen (PSA) are the most important factors influencing both the likelihood of more extensive disease and the probability of
subsequent relapse following RP. Indeed, these tools include both nomograms and risk tables incorporating clinical variables that can predict, although still imperfectly, the likelihood of tumor recurrence and provide crucial prognosis information to guide clinicians in their therapeutic decisions. The risk of disease progression greatly differs between individuals and the heterogeneity in clinical behaviour further emphasizes the need to find novel markers of progression. Even if cases of PCa are considered localized at the time of diagnosis, the rate of BCR after RP is still significant and occur most often in the 5 years after surgery. The persistence of tumor cells in a state of either complete or near dormancy prior to metastatic progression is likely accountable for disease recurrence while these residual cells are most probably responsive to hormones.
[0006] However, in two recently published large-scale population trials, PSA screening was shown to have little impact on decreasing cancer-related mortality, and was accompanied by a risk of overdiagnosis, overtreatment, and increased complications. Hence, the heterogeneity observed in clinical behavior emphasizes the need to identify additional tools suitable for use in clinical practice, such as molecular biomarkers.
[0007] Androgen hormones, such as testosterone (T) and 5a-dihydrotestosterone (DHT), have been clearly implicated in development of PCa. Circulating T, secreted by the testis, and adrenal steroid precursors are among factors influencing androgen levels in the prostate. Indeed, the conversion of T by 5a-reductases (SRD5A1 and SRD5A2) leads to DHT, the more potent androgen receptor (AR) endogenous agonist in target cells.
SRD5A2 is the major 5a-reductase enzyme expressed in the prostate compared to SRD5A11. However, while the expression of SRD5A2 decreases in prostate cancer cells, SRD5A1 is increased in tumoral tissues1"3. This imbalance in the expression of SRD5A genes in PCa tumors illustrates the complex relation between 5a-reductases, DHT synthesis and PCa progression. In concert with SRD5A genes, the active androgen level in target cells is controlled by steroid metabolizing UDP-glucuronosyltransferases 2B genes (UGT2B). These phase II metabolism enzymes conjugate androgens, and thus contribute to the irreversible transformation of steroids into biologically inactive
glucuronide metabolites4,5,6. Consequently, the intracellular hormonal level is determined by the balance between SRD5A and UGT2B enzymatic activities. [0008] Androgen deprivation therapy (ADT) is the standard of care for metastatic PCa and is also used to treat asymptomatic patients with PSA recurrence after failed primary therapy (RP), further reinforcing the initial androgen dependency of these cells.
Finasteride, a 5a-reductase type 2 inhibitor, and Dutasteride, a dual 5a-reductase inhibitor currently used in the clinic, have been recently shown to be effective chemopreventive medications reducing by almost 25% the risk of PCa incidence7 8. Despite this well recognized hormonal dependence of prostate cancer cells in the early cancer stage, very few studies have investigated the associations between polymorphisms in the androgen biosynthesis pathway and clinical outcome after surgical procedure8"14. To date, common polymorphisms such as those in sex-steroids biosynthesis pathways have been extensively studied in relation to risk of PCa15"19. However, almost all of these studies did not address the association between polymorphisms in genes regulating hormonal exposure with PCa recurrence and survival, and were not designed to do so. Long-term longitudinal studies are thus still required to systematically evaluate the impact of a patient's genetic profile on risk of recurrence.
[0009] One of the most significant challenges in oncology remains our inability to predict PCa disease recurrence and clinical outcome due to a lack of key prognostic markers. The prediction of recurrence following RP is clearly important for personalized treatments and follow-up strategies due to striking heterogeneity in prostate cancer clinical behaviour. Our work support that inherited variations in sex-steroid biosynthesis enzymes are relevant candidates as novel predictive markers.
[0010] Over the last several decades, a number of studies have addressed the role of inherited variations in hormone-related genes and the associated risk of developing prostate cancer.15,16,20,21 However, to our knowledge, only two studies have looked at the prognostic value of hormone-related gene polymorphisms in PCa progression and mortality.11 ,22 In the first study, Lindstrom and colleagues studied the role of germline variations in AR genes and the biosynthetic genes CYP17 and SRD5A2, and suggested that polymorphisms in the AR genes affect hormonal treatment and ultimately PCa death.11 More recently, overexpression of the androgen/estrogen inactivating enzyme HSD17B4 was associated with a poor clinical outcome.22 Despite the fundamental role of androgens in PCa development, very scarce data are available regarding the
consequence of genetic variations in the steroidogenic pathway and the associated risk of PCa progression following treatment, particularly for early stage disease. No other predictive or prognostic markers have been identified in the steroidogenic pathway, although several prognostic biomarkers have been identified in other cellular pathways, including cell cycle genes, tumor suppressor genes, cytokines, and transcription factors. [0011] UDP-glucuronosyltransferases (UGTs) are conjugating enzymes that, in concert with other steroidogenesis enzymes, contribute to the maintenance of intracellular levels of sex-steroid hormones in target cells. This enzymatic process generates glucuronide derivatives that are biologically inactive and are subsequently excreted in bile or urine. UGTs are expressed in almost all tissues, including hormone target organs such as the prostate. Five UGT isoforms are mainly responsible for inactivating androgens and estrogens. The UGT1A1 isoform is primarily involved in the glucuronidation of estradiol, whereas the UGT2B class of enzymes (namely UGT2B7, UGT2B15, UGT2B17, and UGT2B28) is mainly involved in androgen catabolism in hormone-dependent cells.
Common polymorphisms in two of these genes (UGT2B15 and UGT2B17) have been associated with prostate cancer risk in some but not all studies.18,23"30 Interestingly, copy number variations (CNV) of the major UGT2B17 and UGT2B28 androgen metabolizing genes have been reported, and more than 50% of Caucasians and 75% of Asians have a reduced copy number in at least one of these genes.31 However, the impact of common germline polymorphic variations or deletions in UGTs has not yet been addressed in PCa progression and mortality.
Summary of the invention
[0012] In the present invention, functional genetic variations in each of the following sex- steroid metabolizing genes UGT1A1 , UGT2B7, UGT2B15, UGT2B17, and UGT2B28, were studied to identify potential molecular biomarkers that are associated with an increased risk of biochemical PSA recurrence after prostatectomy. These UGT candidates were selected due to their prostate-specific role in steroid catabolism. Four populations of patients (three Caucasians and 1 Asian) comprising a total of 1161 men with clinically localized and locally advanced prostate cancer were studied.
[0013] The present invention identifies germline polymorphic CNV events in UGT2B17 and UGT2B28 that are significantly associated with biochemical recurrence in clinically localized PCa after surgical treatment. These deletions are linked to altered levels of circulating sex steroids, supporting the hypothesis that reduced androgen glucuronidation by UGT-mediated inactivation leads to an increased PSA recurrence. Our data indicates that co-occurrence of deleted copies of the UGT2B17 and UGT2B28 genes are important predictors for PSA recurrence independently of the well-established clinico-pathological markers; Gleason score, PSA values and stages at the time of diagnosis.
[0014] To further improve the prognosis significance of inherited variation in the sex- steroid metabolizing genes, genetic variation in the androgen biosynthesis SRD5A genes were combined with CNV in the androgen inactivating UGT2B genes. Together, these genes may alter systemic hormone bioavailability and influence the tumoral
microenvironment exposure to hormones, and thus could modify the risk of PCa recurrence after RP. This is based on the fact that both classes of genes have a well characterized physiological role in DHT homeostasis, are associated with PCa risk and, more recently with cancer progression 12. 3.18·23.24·26.27·29·32·33.46, SRD5A genes are also well recognized molecular targets in PCa prevention trials7,35. In one aspect of the invention, the detailed genetic analyses of the SRD5A1 and SRD5A2 genes in relation to PCa progression are presented. The association between the known SRD5A2 V89L polymorphism and BCR as proposed by other groups12,13 is also described. In one particular aspect, the association between inherited genetic alterations in androgen metabolizing genes and the risk of relapse was determined after RP as the sole initial curative intent. In a particular embodiment, the clinical consequence of simultaneous occurrence of variations in the androgen biosynthetic SRD5A genes and in UGT2B sex- steroid inactivating genes is used to improve the statistical significance of the method for predicting the likelihood of a human subject to develop PCa recurrence or the risk of relapse after RP. [0015] According to a first aspect of the present invention, there is provided a biomarker for predicting the likelihood of prostate cancer development, its progression or recurrence in a human subject, comprising the identification of at least one copy number variation(s) in at least one of a UGT2B gene.
[0016] According to a further aspect, the invention provides the identification of CNV(s) in at least one of a UGT2B gene of a human subject for predicting the likelihood of prostate cancer, its progression or recurrence in said human subject. [0017] There is therefore provided an in-vitro method for predicting risk for PCa in a human subject, said method comprising the steps of: a) obtaining a nucleic acid from a non-tumor or tumor sample from said human subject; and b) determining an individual's genetic variation (or haplotypes, CNV or deletion) in a UGT2B gene; whereby the identification of at least one genetic variation in said UGT2B gene in said subject's nucleic acid is an indication that said subject has an increased risk of developing prostate cancer or of experiencing progression thereof. Particularly, UGT2B risk alleles may be defined as one deletion of UGT2B28 or UGT2B17 and/or two or more deletions in UGT genes.
[0018] The invention further provides the method as defined herein wherein said identification of genetic variation in said UGT2B17 or UGT2B28 comprises identifying at least one CNV in said nucleic acids, said CNV being selected from the group consisting of: rs3100645; and rs1 1249532; and associated reference sequences thereof.
[0019] The method of the invention further comprises the identification of the cooccurrence of a genetic variation (or haplotype) in SRD5A1 or SRD5A2 gene; whereby the co-occurrence of at least one genetic variation in UGT and SRD5A1 or SRD5A2 in said subject's nucleic acid is an indication that said subject has a further increased likelihood that the PCa will recur.
[0020] The invention further provides the method as defined herein wherein said identification of genetic variation in said SRD5A1 or SRD5A2 comprises identifying at least one single nucleotide polymorphism (SNP) in said nucleic acids, said SNP being selected from the group consisting of: rs166050; rs2208532; rs523349; rs4952197;
rs518673, rs12470143 and rs676033; and associated reference sequences thereof.
[0021] The present invention also provides a method for adapting a course of treatment of prostate cancer in a human subject, comprising the steps of: a) providing a prognosis or predicting the likelihood of a human subject to develop prostate cancer recurrence in accordance with the method as defined herein; and b) adapting a course of treatment according to whether said subject has an increased likelihood that the cancer will recur. Particularly, adaptation of course of treatment is realized after the subject has undergone radical prostatectomy. [0022] The invention further provides a kit for predicting the likelihood of a human subject to develop prostate cancer recurrence by detecting a CNV in a nucleic acid sample, the kit comprising reagents for determining the individual's genetic variations (or haplotypes) in UGT2B17 and UGT2B28 gene.
[0023] Particularly, the kit comprises a reagent for detecting a reference sequence selected from the group consisting of: rs3100645; and rs1 1249532, and associated reference sequences thereof.
[0024] Particularly, the kit further comprises reagents for determining the individual's genetic variations (or haplotypes) in SRD5A1 or SRD5A2 gene. Particularly, the kit further comprises reagents for detecting a SNP in a reference sequence selected from the group consisting of: rs518673; rs166050; rs12470143; rs2208532; rs523349; rs4952197 and rs676033 or their associated SNPs.
Detailed description of the invention
Description of the figures
[0025] Figure 1. PCR amplifications of the UGT2B17 and UGT2B28 genes.
[0026] Figure 2. Kaplan-Meier curves of Biochemical Recurrence-Free Survival (bRFS) for copy-number variations of UGT2B genes.
[0027] Figure 3. Kaplan-Meier curves of bRFS for co-occurrence of UGT2B risk allele. In Caucasians, UGT2B risk alleles were defined as 1 deletion of UGT2B28 and/or two or more deletions in UGT genes. In Asians, UGT2B risk alleles were defined as two or more deletions in UGT genes. [0028] Figure 4. Association analysis of common UGT2B17 and UGT2B28 gene deletions and biochemical failure in additional cohorts of PCa cases. Odds ratios and confidence intervals were calculated to combine data from the 1 ,161 PCa cases. UGT2B risk alleles were defined as 1 deletion of UGT2B28 and/or two or more deletions in UGT genes. [0029] Figure 5. Prognostic impact of inclusion of genetic markers, UGT2B17 and UGT2B28 gene deletions, in the evaluation of the D'Amico risk classification of PCa recurrence after RP. [0030] Figure 6. Kaplan-Meier Biochemical Recurrence-Free Survival Analysis for other UGT genes.
[0031] Figure 7. Risk of recurrence associated with known clinical and pathological prognostic variables (A) and SRD5A genes (B). Boxes represent hazard ratios (HR) and their 95% CI. PSA categories are in ng/ml. Reference categories (HR: 1.00) are: PSA at diagnosis < 10ng/ml, pG < 6, and pT < T2b. Genetic linkage between htSNPs tested for each SRD5A gene is represented in the triangles on the left in panel B.
[0032] Figure 8. Kaplan-Meier estimates of recurrence-free survival for A) SRD5A2, B) SRD5A 1 and C) protective htSNPs in both SRD5A genes. Log rank (LR) P values are shown in each frame.
[0033] Figure 9. Risk of recurrence associated with htSNPs in A) SRD5A1 and B)
SRD5A2 genes stratified by status for UGT2B risk alleles. Boxes represent hazard ratios (HR) and their 95% CI in multivariate analysis. Black boxes were analyzed only for SRD5A genotypes, white boxes are for SRD5A minor alleles + no UGT2B risk alleles, and grey boxes are for SRD5A minor alleles + UGT2B risk alleles. Boxes indicate HR and error bars indicate the 95% confidence intervals. The reference category is set at one. Models used for multivariate analysis are indicated in Table 13.
[0034] Figure 10. Kaplan-Meier curves of biochemical recurrence-free survival for carriers of protective alleles of both SRD5A genes and of UGT2B risk alleles. Log-rank P values (LR) are shown in each frame. Risk alleles for UGT2B are defined as one deleted copy of UGT2B28 or >2 deleted copies of UGT2B17 and UGT2B28. 0= no protective SRD5A alleles; 1 -2= 1 or 2 protective SRD5A alleles and 3-4= 3 or 4 protective SRD5A alleles.
Definitions and abbreviations
[0035] ADT: androgen-deprivation therapy; AR: androgen receptor; BCR: biochemical recurrence; bRFS: biochemical recurrence-free survival, CNV: copy number variation; DHT: 5a-dihydrotestoterone; HR: hazard ratio; htSNP: haplotype-tagging SNP; MAF: minor allele frequencies; PCa: prostate cancer; PSA: prostate-specific antigen; SRD5A1 : 5a-reductase type 1 ; SRD5A2: 5a-reductase type 2; RP: radical prostatectomy; SNP: Single nucleotide polymorphism; T: Testosterone; TNM: Tumor/Nodes/Metastasis staging system; UGT: UDP-glucuronosyltransferase. [0036] The term "identify" as used herein, means the act of searching for and finding a specific marker. Other terms that may be used for the same purpose are: "detect", "locate", "expose", "observe", "recognize", "uncover" or "reveal".
[0037] The term "development" or "developing" when used in the context of cancer means the development, progression, recurrence, biochemical recurrence, death from or metastasis of such a cancer.
[0038] The term 'sample" as used herein, means a patient or subject's nucleic acid- containing sample originating indistinctly from a tumor or a non-tumor sample. The subject's non-tumor sample can be selected from: tissue, or biological fluid. Particularly, the tissue can be selected from the group consisting of: skin, lymph node, hair and buccal smear. Alternatively, the biological fluid can be selected from the group consisting of: sputum, saliva, blood, serum, urine, semen and plasma. Alternatively, the non-tumor or tumor sample may originate from a biopsy.
[0039] The terms "genetic variation" or "haplotype" used herein in conjunction with UGTs mean "Copy-number variations" (CNVs), e.g. a form of structural variation consisting in alterations of the DNA of a genome that results in the cell having an abnormal number of copies of one or more sections of the DNA. CNVs correspond to relatively large regions of the genome that have been deleted (fewer than the normal number) or duplicated (more than the normal number) on certain chromosomes. For example, the chromosome that normally has sections in order as A-B-C-D might instead have sections A-B-C-C-D (a duplication of "C") or A-B-D (a deletion of "C"). When used in conjunction with SDR5A gene, the terms "genetic variation" or "haplotype" refer to single-nucleotide polymorphisms (SNPs), which are DNA sequence variations occurring when a single nucleotide base— A, T, C or G— in the genome, or other shared sequence, differs between members of a biological species or paired chromosomes in an individual.
Detailed description of particular embodiments
Genetic variation status, copy number variation and reference sequence.
[0040] In accordance with particular aspects of the present invention, there is provided an in-vitro method for predicting the risk for PCa in a human subject, said method comprising the steps of: a) obtaining a nucleic acid from a non-tumor or tumor sample from said human subject; and b) determining the individual's genetic variation status such as for example a copy number variation in UGT2B17 gene and/or UGT2B28 gene; whereby the determination of at least one genetic variation in one of the UGT2B17 or UGT2B28 gene is an indication that said subject has an increased likelihood that the prostate cancer will recur.
[0041] The invention further provides the method as defined herein wherein said identification of genetic variation status in said UGT2B17 or UGT2B28 comprises identifying at least one copy number variation (CNV) in said nucleic acids, said CNV being selected from the group consisting of: rs3100645; and rs1 1249532. [0042] Particularly, the genetic variation in UGT2B17 is a copy number variation consisting of one or two deletions. Still, particularly, the genetic variation in UGT2B28 is a copy number variation consisting of one or two deletions.
[0043] Still, more particularly, the method of the invention comprises the identification of the co-occurrence of the genetic variations in UGT2B17 and UGT2B28 genes, e.g. when both UGT2B17 and UGT2B28 genes harbor at least one CNV selected from the group consisting of: rs3100645; and rs 1249532. Particularly, each of UGT2B17 and UGT2B28 can harbor one or two deletions.
Detection of co-occurrence with other markers
[0044] The method of the invention further comprises the identification of the co- occurrence of a genetic variation in SRD5A1 or SRD5A2 gene; whereby the cooccurrence of at least one genetic variation in UGT2B and either SRD5A1 or SRD5A2 or both in said subject's nucleic acid is an indication that said subject has a further increased likelihood that the prostate cancer will recur.
[0045] The invention further provides the method as defined herein wherein said identification of genetic variation in said SRD5A1 or SRD5A2 comprises identifying at least one single nucleotide polymorphism (SNP) in said nucleic acids, said SNP being selected from the group consisting of: rs166050; rs2208532; rs523349; rs4952197;
rs518673, rs 12470143 and rs676033. Sample
[0046] The invention also provides the method as defined herein, wherein the subject's nucleic acid-containing sample is a tumor or a non-tumor sample. Particularly, the subject's non-tumor sample is selected from: tissue, or biological fluid. Particularly, the tissue is selected from the group consisting of: skin, lymph node, hair and buccal smear. Alternatively, the biological fluid is selected from the group consisting of: sputum, saliva, blood, serum, urine, semen and plasma. Alternatively, the non-tumor or tumor sample originates from a biopsy.
Methodology
[0047] The invention also provides the method as defined herein wherein in step a), the nucleic acid is DNA or RNA. Particularly, the DNA and the marker is detected with the use of a nucleic acid primer. More particularly, the DNA is amplified by PCR.
[0048] Particularly, the genetic variation of DNA or RNA is detected with the use of a methodology selected from the group consisting of: direct sequencing, pyrosequencing, massively parallel sequencing, high-throughput sequencing, high performance liquid chromatography (HPLC) fragment analysis, mass spectrometry, branch DNA and capillary electrophoresis. Alternatively, the DNA or RNA of the marker is detected with the use of a nucleic acid primer and probe combination. Particularly, the DNA or RNA is amplified prior to incubation with the probe, such as by PCR. More particularly, amplified PCR products and the genetic variation are analyzed by mass spectrometry.
Treatment regimen.
[0049] The present invention also provides for a method for adapting a course of treatment of prostate cancer in a human subject, comprising the steps of: a) predicting the likelihood of prostate cancer progression or recurrence in a human subject as defined herein; and b) adapting a course of treatment according to whether said subject has an increased or decreased likelihood that the cancer will progress, or recur.
[0050] Particularly, when the likelihood of recurrence is increased, the subject is prescribed at least one of: hormone therapy, chemotherapy, surgery, radiotherapy, brachytherapy or active surveillance (watchful waiting). More particularly, steps a) and b) are carried out after the subject has undergone radical prostatectomy. [0051] The invention also provides the method as defined herein, wherein, when the likelihood of recurrence is increased, the subject is prescribed inducer (or inductive) UGT2B therapy and/or 5a-reductase inhibitors therapy.
Kit and reagents
[0052] Furthermore, the present invention provides for a kit for predicting the likelihood of a human subject to develop prostate cancer or a recurrence thereof, said kit comprising reagents for determining the individual's genetic variations (or haplotypes) in the UGT2B gene. Particularly, the kit comprises reagents for detecting a CNV in a reference sequence selected from the group consisting of: rs3100645 and/or rs1 1249532; and associated reference sequences thereof.
[0053] The invention also provides the kit as defined herein, comprising a PCR primer, wherein the primer is selected from the group consisting of: SEQ ID Nos. 1 , 2, 7 and 8. Additionally, the kit may comprise a PCR probe, wherein the probe hybridizes with a sequence selected from rs3100645 and/or rs1 1249532; and associated reference sequences thereof.
[0054] Particularly, the kit further comprises reagents for determining the individual's genetic variations (or haplotypes, SNP) in SRD5A1 and/or SRD5A2 genes, such as reagents for detecting a SNP in a reference sequence selected from the group consisting of: rs518673; rs166050; rs12470143; rs2208532; rs523349; rs4952197 and rs676033 or their associated SNPs.
[0055] The invention further provides the kit as defined herein, comprising PCR primer- probe set for determining the individual's SNPs in SRD5A1 and/or SRD5A2 genes, wherein the primer is selected from the group consisting of: SEQ ID Nos. 12 to 50; and the probe is selected from the group consisting of: SEQ ID Nos. 51 to 69. Example 1 - Copy number variations in the UGT2B genes
Patients and methods
Description of Study Cohorts.
[0056] The copy number variations in the UGT2B genes were studied in 1 161 unrelated men obtained from four independent cohorts; cohort A was composed of 526 Caucasian patients with localized prostate cancer; cohort B comprised 213 Caucasian patients with locally advanced disease, cohort C comprised 320 Asian patients with localized prostate cancer and cohort D was composed of 102 Caucasian Slovenian men (Table 2). Cohorts A and B patients underwent open radical prostatectomy for clinically localized and locally advanced adenocarcinoma of the prostate at the CHUQ - Hotel-Dieu de Quebec hospital (Quebec, Qc, Canada) between 1982 and 2002. Cohorts C and D have been described elsewhere.8,36 Each participant gave written informed consent before surgery for subsequent genetic analyses. The local research ethical committee approved the research protocol. Detailed clinical information was obtained retrospectively from medical records. All patients were followed postoperatively with serial PSA measurements.
Definitions.
[0057] The primary outcome variable was biochemical recurrence-free survival (bRFS), also called 'PSA failure' or 'biochemical relapse'. In Caucasians, bRFS was defined as (1 ) two consecutive PSA values > 0.3 pg/L, (2) one PSA value≥ 0.3 pg/L followed by hormonotherapy (androgen deprivation therapy or ADT) or radiation therapy, and (3) a single last-recorded PSA value > 0.3 pg/L after prostatectomy. bRFS was defined as the period of time elapsed between the surgical procedure and the date of PSA failure. In cohorts C and D, the bRFS was defined as previously described. 8 36
Genotyping.
[0058] For cohorts A and B, peripheral blood was collected on the morning of a preoperative ambulatory clinic visit. All samples were kept frozen at -80°C until the time of study. Genomic DNA was extracted using a QIAamp DNA Blood mini kit (Qiagen Inc, Mississauga, Ontario, Canada) and stored at -80°C. For cohorts C and D, DNA was prepared as previously described. 8 36 [0059] The presence of at least one copy of UGT2B17 and UGT2B28 is first assessed by PCR amplifications of the exon 1 of each gene and the corresponding deletion site, using PCR conditions and specific primers as previously described 31.
[0060] Common amplification steps used are: one heating step at 95° C for 30 sec, 40 cycles of denaturing at 95°C, annealing and elongation at 72°C and a single final extension step of 7 min at 72°C (see Figure 1 and Table 1 ). [0061] Amplification of the exon 6 can be performed to confirm the absence of the studied gene (UGT2B17 and UGT2B28) if deemed necessary, in case of uncertainties only. With these genotyping strategies, we were able to detect precisely the number of deletions (from 0 to 4).31 [0062] For UGT1A1 (rs34815109), UGT2B15 (rs1902023), and UGT2B7 (rs7439366), after PCR amplification (for primers and amplification strategies, see 31 ,37), products were purified and sequenced with an ABI 3730x1 genetic analyzer (Applied Biosystems, Foster City, CA, USA) and assessed only in Cohort A. The sequences were analyzed using Staden preGap and Gap4 programs (Open Source Technology Group,
http://staden.sourceforge.net/) to identify the variant sequence. PCR amplicons were sequenced, and amplicons resulting in ambiguous sequencing chromatograms were systematically re-amplified and re-sequenced.
Statistical Analysis
[0063] Based on our sample size, we were able to detect a hazard ratio of 1.50, for a minor allele with frequency of 5%, with over 80% statistical power. All genes were tested for consistency with Hardy-Weinberg equilibrium using chi-square analysis. Each gene was treated as a categorical variable, with a common (reference) homozygote, a heterozygote, and a rare homozygote. A rare homozygote was combined with a heterozygote if the frequency of the rare homozygote was below 0.10. For UGT2B17, it was previously established that deletion of one allele is not associated with significantly lower intraprostatic mRNA levels compared to individuals homozygous for the insertion allele (ins/ins)35. Therefore, individuals with one deletion allele of UGT2B17 were combined with subjects with no deletion. For UGT2B28, the rare del/del genotype was combined with ins/del heterozygote because the frequency of the null genotype was below 0.10. Initial PSA level, Gleason score at biopsy, and tumor clinical stages were treated as categorical variables using the well-recognized D'Amico risk classification. In cohorts C and D, the pathological stage was used for multivariate analysis because tumor clinical stage was not available.
[0064] Kaplan-Meier analysis for bRFS analysis was performed to compare genotypes using the log-rank test followed by a pairwise multiple comparison using the Sidak multiple-comparison method. Results that were significantly associated with bRFS in univariate analysis were then evaluated in a multivariate Cox regression analysis. We included the covariates included in Table 2 in the models and UGT2B17 and UGT2B28 genetic status. The proportional hazard assumption of Cox models was verified graphically by log-minus-log vs. log (time) plots for each variable. Kaplan-Meier curves and univariate analysis for bRFS were also performed for CNVs of UGT2B17 and
UGT2B28. Haplotype analysis was performed for cohort A only. After this initial screen, the other genetic variables were not further studied in other cohorts due to lack of association. Clinical variables were compared across genotypes using the chi-square test, Fisher's exact test evaluated exactly, or the Monte Carlo approximation, as appropriate. A p value of less than 0.05 was considered statistically significant, and all tests were two- sided. All statistical analyses were performed using SAS® Statistical Software version 9.2 (SAS Institute, Cary, NC, USA).
RESULTS
[0065] Patient's characteristics are presented in Table 2. Overall, 130 patients (24.7%) experienced PSA failure in cohort A, 132 (62%) in cohort B, 1 16 (36.3%) in cohort C and 34 (33%) in cohort D.
[0066] Preoperative PSA values and pathological biopsy Gleason scores were both associated with bRFS. Men with PSA values >20 were associated with an HR of 2.120 (95% [1.542; 2.913], p = 0.000004) for bRFS in multivariate analysis (Table 4), whereas a Gleason score >8 is associated with an HR of 2.773 (95% CI [2.005; 3.835], p < 0.0001 ) and clinical T stage with an HR of 2.046 (95% CI [1.264; 3.313], p < 0.004); similar to previous reports. 38,39 Reduced CNVs in UGT2B17 and UGT2B28 were significantly associated with bRFS (Table 3 and Figure 2) while no significant association was observed for additional UGT variants tested in cohort A (Table 5). Haplotype analyses of the UGT2B locus in this cohort also revealed that haplotypes 3 and 5, containing UGT2B deletions, were associated with bRFS (Table 6). Based on these initial results, additional cohorts were tested. The distributions of genotype frequencies are shown in Table 7 and are consistent with previous reports.26,31 ,40,41
[0067] Figure 2 displays the Kaplan-Meier curves of bRFS for UGT2B17 and UGT2B28 genes for all studied patients for whom this analysis was achievable (cohorts A, B and C). In cohorts A and B, multivariate analyses indicated that individuals with one UGT2B28 or two UGT2B17 deleted alleles had an increased likelihood of PSA failure with HR values of 1 .390 (95% CI [1.064; 1.815], p = 0.016) and 1.385 (95% CI [0.961 ; 1.997], p = 0.08), respectively (Table 3). Individuals of Asian Ancestry (cohort C) were included in our study because they have the highest reported frequencies of UGT2B deletions. 41 Comparison of UGT2B17 CNVs minor allele frequencies (MAF) revealed that the UGT2B17 deletion genotype prevailed in this population with a MAF of 0.73, compared to 0.27 in Caucasians (Table 7). In this population, 57% of individuals (n=185) have a UGT2B17 null genotype and, therefore, constitute the most common genotype in this ethnic group. For UGT2B28, the MAF is 0.15 in Asians which is similar to the frequency of 0.13 observed in
Caucasians. Six subjects with the combined UGT2B17 and UGT2B28 null genotypes were identified, indicating for the first time that concurrent deletions in these UGT2B genes are rare but not lethal. In the population of Asians, data revealed that two deleted copies of the UGT2B17 gene is associated with biochemical failure with an HR of 1 .791 (95% CI [1.107; 2.898]; p=0.018). Patients genotyped with two or more deletions of UGT2B17 and UGT2B28 genes have an increased risk of BCR with HR of 2.151 (95% CI [1.239; 3.372]; p=0.006) for bRFS (Figure 3 and Table 3). Multivariate analysis of all 1161 patients lead to an OR of 1.510 (95% CI [1 .178 - 1.950]; p=0.001 ) associated with the combined effect of UGT2B deletions (Figure 4). These results support that UGT2B17 and UGT2B28 CNVs are determinants of PSA failure after prostatectomy. [0068] To further demonstrate the clinical relevance of UGT2B deletions on bRFS, we used the D'Amico risk classification and further stratified each category for the status for UGT2B risk alleles in Caucasians (cohorts A and B) (Figure 5). The addition of the information regarding CNV status led to an improved risk prediction, particularly for the low and intermediate risk categories (Log-rank (LR) test, p=0.022 and 0.01 1 , respectively) (Figure 5). This further supports that genetic deletions in the UGT2B pathway are independent predictors of bRFS whereas their incorporation along with current clinico- pathological prognostic markers did not changed the risk prediction for the high risk subgroup (LR=0.49). Interestingly, the presence of the UGT2B17 gene deletion is enriched in high risk patients, OR 3.83 (95% CI [2.31 1 ; 6.373]; p=2X10"7), which is not the case for UGT2B28, OR 0.968 (95% CI [0.67; 1 .398]; p=0.863). Thus, the presence of UGT2B17 deletions would be associated with adverse clinical features. No other significant interaction was noted between gene markers, PSA, Gleason score and TNM staging (Table 8). DISCUSSION
[0069] We assessed the prognostic value of common genetic variants of five major sex- steroid metabolizing UGT isoforms in clinically localized and locally advanced prostate cancer patients following prostatectomy. We analyzed the samples of 1 161 patients with localized and locally advanced prostate cancer from four independent cohorts, for which prostatectomy was the sole initial therapeutic intervention. To our knowledge, there are no previous reports studying this inherited variation in UGT2B genes for prostate cancer recurrence. Using this data set, we observed a positive correlation between individuals with one or two deletion alleles for UGT2B17 and UGT2B28 and bRFS. However, no significant association between selected SNPs in other UGT genes and bRFS was observed in our study. Studying genes involved in the end of the androgenic signal is biologically relevant in the initial phase of disease development because prostate cancer cells are hormone responsive at this stage, and hormonal manipulation is a cornerstone of disease management. [0070] Our findings demonstrate that the presence of at least one deleted allele of the sex-steroid metabolizing gene UGT2B28, or the co-occurrence of two deletions in the UGT2B17 and UGT2B28 genes is predictive of early PSA failure after prostatectomy in both Asians and Caucasians. Recently, CNV in UGT2B17 was shown to be associated with osteoporosis in Asians,42 linking CNV for the first time with a complex metabolic disease. Of all genetic process involved in prostate cancer progression, CNV is certainly one of the most drastic molecular events. However, to date, very limited data have looked at the association between CNV and cancer risk and more importantly, none have been linked to cancer progression. CNV of UGT2B17 was shown to be associated with prostate cancer risk in some studies but not in others. 8'26,27,29 However, none of these studies looked simultaneously at the chromosomal neighbors UGT2B15 and/or UGT2B28, which may contribute to the discrepancies observed.
[0071] Incorporation of CNV in UGT genes and other relevant genetic variants in a nomogram will ultimately provide a relevant clinical translation that could help guide treatment decisions. Indeed, we also revealed that the incorporation of UGT CNV status to the well recognized d'Amico risk classification system, improved stratification of low and intermediate risk patients. [0072] This data thus implicates CNV in sex-steroid metabolizing genes as biomarkers for prostate cancer recurrence, and provides support for the significance of sex-steroids in the pathogenesis and recurrence of this prostate cancer.
Example 2 - Genotyping of SRD5A genes polymorphisms Methods
Clinical Data and Outcome Collections
[0073] A second study was mostly composed of Caucasians, and included 526 men who underwent RP at I'Hotel-Dieu de Quebec Hospital (QC, Canada) between February 1999 and December 2002. Each participant provided written consent before surgery for the analysis of their genome and the research protocol was approved by the research ethical committee at the Centre Hospitalier Universitaire de Quebec (CHUQ, QC, Canada). All patients were followed postoperatively with serial PSA measurements and detailed clinical information was available.
DNA Isolation and Genetic Analysis
[0074] Polymorphisms in SRD5A genes were chosen according to one or more of the following criteria: i) to be likely functional (with supportive data in the literature), ii) to have previously been associated with PCa risk, aggressiveness, age at onset, BCR or ADT efficiency, and iii) to explain most of the haplotype diversity in the CEU (Utah residents with Northern and Western European Ancestry) Hapmap population. For both SRD5A genes, a region covering all the exons, introns and 5 kb of the 5' and 3' sections of the genes was screened using a haplotype tagging SNPs (htSNPs) strategy to maximize coverage, using HapMap Phase 2 0, and data from the CEU unrelated subjects based on a r2 >0.80 and a minimum minor allele frequency >0.05. Determination of UGT2B genetic deletion status has been described elsewhere 31. [0075] Peripheral blood was collected on the morning of a preoperative clinic visit and kept frozen at -80°C until analysis. Genomic DNA was purified using the QIAamp DNA Blood Mini Kit (Qiagen Inc., Mississauga, On, Canada) and stored at -20°C. For htSNPs in SRD5A genes, PCR amplifications were performed using Sequenom iPLEX matrix- assisted laser desorption/ionization-time-of-flight mass spectrometry. For oligos sequence, see Table 12. Negative controls were present for every run of analyses and quality controls (random replicates of known genotypes) were successfully performed in 5% of the study cohort.
Statistical Analysis
[0076] Based on our sample size, we were able to detect a hazard ratio of 1.50, for a minor allele with frequency of 5%, with over 80% statistical power. Allelic frequencies and Hardy-Weinberg equilibrium were computed with PLINK (version 1.07) 43. To analyze their association with BCR, each htSNP was considered using 3 models since the function of most htSNPs remains unknown. The genomic model considered the htSNP as a categorical variable with a common allele homozygote, heterozygote and a minor allele homozygote. The dominant model considered the htSNP with only 2 categories: one with a common allele homozygote and one with at least one minor allele. Finally, the recessive model also considered the htSNP with only 2 categories: one with at least one common allele and one minor allele homozygote. Cox regression was performed on each htSNP considering the 3 above mentioned models with adjustment for clinicopathological variables: PSA level at diagnosis, age at diagnosis, smoking status, pathological Gleason grade, pathological stage and neoadjuvant ADT. All co-variables were treated as categorical, and for PSA level, Gleason scores and stage, they were used as described by the D'Amico risk classification. The censoring variable was BCR, which was defined as 1 ) two consecutive PSA values > 0.3 pg/L, 2) one PSA value > 0.3 pg/L followed by ADT, and 3) a last-recorded PSA value > 0.3 pg/L after prostatectomy.
[0077] Kaplan-Meier analyses were also processed for every htSNP (log-rank), but only results for htSNPs significantly associated with BCR in Cox regression multivariate analysis are shown. These statistical analyses were performed using PASW statistics 17 (SPSS Inc., Chicago, IL) and R version 2.10.0. Results
[0078] All 526 patients had initially RP as curative intent enabling a precise pathological evaluation. Clinical and pathological characteristics of the study cohort are shown in Table 9. The median follow-up time of the cohort is 7.4 years (range: 0.5 to 10.2 years). The cohort had mainly organ-confined and locally advanced tumors, as PCa cases were composed of pT2 (60%) and pT3 (37%) pathological tumor stages (Table 9). Overall, 130 cancer cases experienced BCR (25%), with a median time to relapse of 2.1 years. [0079] Risk of recurrence associated with known clinical and pathological prognostic variables are shown in Figure 7. The risk of recurrence increased with higher PSA values at diagnosis with a risk of relapse of HR= 1.5 (P=0.081 ) and HR=2.1 (P=0.003) for PSA values of >10-20 ng/mL and >20 ng/mL, respectively. Gleason scores of 7 and≥ 8 were also positively associated with relapse with HR of 2.6 (P=0.002) and 5.4 (P=0.0000003), respectively.
[0080] A total of 19 htSNPs distributed across the two SRD5A genes were studied herein, of which 2 were functional coding SNPs. htSNPs were selected with a strategy to maximize gene coverage and to reflect adequately the Caucasian haplotype genetic diversity. The htSNPs strategy allowed us to study 109 genetic variations in both genes (Table 13).
[0081] After analyses with a Cox regression multivariate model, adjusted for all clinical and pathological factors known to affect BCR, 7 htSNPs, 2 in SRD5A1 and 5 in SRD5A2, were positively associated (P<0.05) with the risk of relapse. Their relative frequencies in cancer patients with and without relapse, and the corresponding hazard ratios (95% CI) are displayed in Figure 7B and Table 10. Genetic linkage between the 19 htSNPs tested in both SRD5A genes is also represented.
[0082] Among 7 htSNPs tested in SRD5A1 , the rs518673 showed a P=0.008 by log-rank test for recurrence-free survival while the rs166050 was almost significant with a (log-rank test P=0.051 ; Figure 8). The SRD5A1 rs166050 gene polymorphism was associated with an increased recurrence risk of HR=1 .89 (95% CI, 1 .03-3.45; P=0.039), while the rs518673 in SRD5A1 was associated with a decreased recurrence risk (HR=0.63, 95% CI, 0.44-0.91 ; P=0.014; Table 10).
[0083] Of the 5 SRD5A2 htSNPs associated with the risk of BCR, a significant association was observed with the nonsynonymous SNP V89L (rs523349) while no association was seen with the other known coding variation A49T (rs9282858) (HR=0.81 , 95% CI, 0.36- 1.85; P=0.62) (Table 10). Additional variations in SRD5A2 were also associated with increased risk of recurrence; rs2208532, rs676033 and rs4952197 with HR of 1 .65, 1.77 and 2.05, respectively (Figure 7B and Table 10). In contrast, the SRD5A2 htSNP rs 12470143 was associated with a reduced risk of recurrence; HR of 0.64 (95% CI, 0.44- 0.93; P=0.019). Kaplan-Meier and log-rank P values are illustrated in Figure 8B. [0084] By combining protective alleles in SRD5A1 (rs518673 T) and SRD5A2
(rs12470143 A), the effect was shown to be greater in Kaplan-Meier analysis with the maximum protection conferred by 3 or 4 alleles (Figure 8C) and remained an independent predictor of recurrence in Cox proportional hazards analysis (HR=0.34, 0.18-0.64;
P=0.0009; Table 11 ). We also discovered a significant allele-dose-dependent relationship with recurrence in which each additional allele diminishing by 26% the risk of recurrence of BCR (P=0.004).
[0085] Here, we outlined a significant association of BCR with multiple genetic
polymorphisms in both SRD5A genes in a cohort of 526 PCa cases. Remarkably, of the 19 htSNPs tested, 7 inherited variations (37%) were shown to affect the risk of recurrence after RP either by conferring protection or an elevated risk, independently of known clinical and pathological predictors of prostate cancer recurrence. Our study is the first to show strong positive associations of multiple genetically unlinked SRD5A2 variations with BCR and to argue for a significant role of SRD5A1 in the risk of recurrence after RP.
These findings thus define that genetic variations in both 5a-reductase type 1 and type 2 pathways play a critical role in prostate cancer recurrence after RP while the effect was magnified with an increasing number of SRD5A adverse genotypes. Furthermore, a protective effect by two unlinked SRD5A genetic variants, rs518673T and rs12470143A, on recurrence was revealed. Reduced occurrence of BCR was shown to be maximal in patients with 3 to 4 of these protective alleles in both SRD5A genes.
[0086] One of the best characterized SNP in the SRD5A2 gene, the functional V89L polymorphism, is associated with a decreased enzymatic activity.44,45 This polymorphism has been associated with prostate cancer risk in some studies32,34 agressiveness32 33, and with conflicting results with biochemical recurrence12 13. One important finding of our study is a significant association between the V89L (rs523349) polymorphism and the risk of BCR after prostatectomy (2.1 -fold risk of recurrence for homozygotes rs523349C). This result is in agreement with Shibata and colleagues 27 and with a recent large study that associated this SNP with an aggressive form of PCa33. However, for the non-synonymous SRD5A2 A49T variant (rs9282858), a functional SNP associated in some studies with the risk of PCa46, no association was evidenced with recurrence-free survival after RP indicating that this SNP would have no obvious role in PCa recurrence. This result is consistent with a meta-analysis investigating this SNP46. Example 3 - Co-occurrence of polymorphism in the SRD5A and copy number variations UGT2B genes
[0087] As presented herein before, patients with two or more deleted copies of the UGT2B17 and/or UGT2B28 genes have an increased risk of recurrence (HR of 2.12 (95% CI [1 .34; 3.37]). Again, the presence of a SRD5A risk alleles combined with deletions in UGT2B genes further increased this risk (Figure 9 and Table 14). For example, when combining the group of patients carrying risk alleles in UGT2B and SRD5A2 rs523349C, those individuals had more than 70% BCR rates compared to 25% for the other groups (Log-rank P=0.000007; Figure 9). The HR values rose from 2.12 (95% CI; 1.21 -3.75; P=0.009) for individuals homozygous SRD5A2 rs523349C to 4.97 (95% CI, 2.38 - 10.36; P=0.00002; Figure 9) for patients with deletions in the UGT2B genes. Similarly, the presence of a rs2880532 SRD5A2 risk allele combined with deleted copies of UGT2B are associated with more than 50% BCR, compared to less than 25% for the other groups (Log-rank P=0002; Figure 9). Patients homozygous for rs2208532G and rs676033A had HR values of 1 .65 and 1.77 respectively; compared to risks of 2.85 and 3.85 when combined with UGT2B risk alleles (Figure 9).
[0088] In patients with unfavourable UGT2B genotypes (e.g. deleted copies) but carrying 3-4 protective SRD5A alleles, BCR-free recurrence reached 80.8% compared to 37.5% for those patients with no protective allele (LR: 0.001 ; Table 11 and Figure 10).
Comparable BCR-free survival rates (LR=0.17) were observed in patients with no deleted copies of UGT2B presented, regardless of the presence of SRD5A protective alleles.
[0089] As a result, we observed an additive effect of UGT2B CNVs with htSNPs in SRD5A genes. Indeed, as presented hereinbefore, we demonstrated that deletions of both UGT2B17 and UGT2B28 are associated with risk of recurrence of PCa after RP. As both SRD5A and UGT enzymes are the rate limiting steps in T and DHT
biotransformation, we confirmed that genetic alterations in both pathways are additive and strikingly modify the risk of biochemical relapse after RP.
[0090] Of note, this data set included a large sample size combined with relevant information on clinical and pathological covariates as well as a significant median follow- up time of 7.4 years. However the limited number of some clinically relevant events such as metastasis, androgen-deprivation resistance, or death related to the localized features of the tumors, prevented us from addressing the association between inherited variations in candidate genes and risk for these events.
[0091] This data clearly demonstrated for the first time that multiple genetic markers in SRD5A and UGT2B genes contribute to biochemical recurrence risk after RP. In localized disease clinical settings, these markers are independent of current predictors of recurrence such as Gleason score and PSA level and predict risk better than the pathological stage. These findings ultimately help refine our ability to identify individuals at low or high risk of cancer relapse after RP, beyond known prognostic variables, and for whom a more personalized approach might optimize outcome, especially in the context of PCa chemoprevention using of 5a-reductase inhibitors therapy. Indeed, the inclusion of such markers is critical in predicting clinical outcome and may lead to informed decisions such as for endocrine treatment with major implications on the patient's recurrence-free survival and quality of life.
References
1. Thomas LN, Lazier CB, Gupta R, et al: Differential alterations in 5alpha-reductase type 1 and type 2 levels during development and progression of prostate cancer. Prostate 63:231-9, 2005
2. Montgomery RB, Mostaghel EA, Vessella R, et al: Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res 68:4447-54, 2008
3. Titus MA, Gregory CW, Ford OH, 3rd, et al: Steroid 5alpha-reductase isozymes I and II in recurrent prostate cancer. Clin Cancer Res 11 :4365-71 , 2005
4. Belanger A, Pelletier G, Labrie F, et al: Inactivation of androgens by UDP- glucuronosyltransferase enzymes in humans. Trends Endocrinol Metab. 14:473-9., 2003 5. Beaulieu M, Levesque E, Hum DW, et al: Isolation and characterization of a novel cDNA encoding a human UDP-glucuronosyltransferase active on C19 steroids. J Biol Chem 271 :22855-62, 1996
6. Levesque E, Turgeon D, Carrier JS, et al: Isolation and characterization of the UGT2B28 cDNA encoding a novel human steroid conjugating UDP-glucuronosyltransferase. Biochemistry 40:3869-81 , 2001
7. Andriole GL, Bostwick DG, Brawley OW, et al: Effect of dutasteride on the risk of prostate cancer. N Engl J Med 362:1192-202, 2010 8. Celhar T, Gersak K, Ovcak Z, et al: The presence of the CYP1 1A1 (TTTTA)6 allele increases the risk of biochemical relapse in organ confined and low-grade prostate cancer. Cancer Genet Cytogenet 187:28-33, 2008
9. Edwards SM, Badzioch MD, Minter R, et al: Androgen receptor polymorphisms: association with prostate cancer risk, relapse and overall survival. Int J Cancer 84:458-65, 1999
10. Hamada A, Danesi R, Price DK, et al: Association of a CYP17 polymorphism with overall survival in Caucasian patients with androgen-independent prostate cancer. Urology 70:217- 20, 2007
11. Lindstrom S, Adami HO, Baiter KA, et al: Inherited variation in hormone-regulating genes and prostate cancer survival. Clin Cancer Res 13:5156-61 , 2007
12. Nam RK, Toi A, Vesprini D, et al: V89L polymorphism of type-2, 5-alpha reductase enzyme gene predicts prostate cancer presence and progression. Urology 57:199-204, 2001
13. Shibata A, Garcia Ml, Cheng I, et al: Polymorphisms in the androgen receptor and type II 5 alpha-reductase genes and prostate cancer prognosis. Prostate 52:269-78, 2002
14. Strom SS, Gu Y, Zhang H, et al: Androgen receptor polymorphisms and risk of biochemical failure among prostatectomy patients. Prostate 60:343-51 , 2004
15. Cunningham JM, Hebbring SJ, McDonnell SK, et al: Evaluation of genetic variations in the androgen and estrogen metabolic pathways as risk factors for sporadic and familial prostate cancer. Cancer Epidemiol Biomarkers Prev. 16:969-78., 2007
16. Cussenot O, Azzouzi AR, Nicolaiew N, et al: Combination of polymorphisms from genes related to estrogen metabolism and risk of prostate cancers: the hidden face of estrogens. J
Clin Oncol 25:3596-602, 2007
17. Mononen N, Schleutker J: Polymorphisms in genes involved in androgen pathways as risk factors for prostate cancer. J Urol 181 :1541 -9, 2009
18. Park JY, Tanner JP, Sellers TA, et al: Association between polymorphisms in HSD3B1 and
UGT2B17 and prostate cancer risk. Urology. 70:374-9., 2007
19. Setlur SR, Chen CX, Hossain RR, et al: Genetic variation of genes involved in dihydrotestosterone metabolism and the risk of prostate cancer. Cancer Epidemiol
Biomarkers Prev 19:229-39, 2010
20. Stanbrough M, Bubley GJ, Ross K, et al: Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res. 66:2815-
25., 2006
21. Giovannucci E, Stampfer MJ, Krithivas K, et al: The CAG repeat within the androgen receptor gene and its relationship to prostate cancer. Proc Natl Acad Sci U S A 94:3320-3, 1997
22. Rasiah KK, Gardiner-Garden M, Padilla EJ, et al: HSD17B4 overexpression, an independent biomarker of poor patient outcome in prostate cancer. Mol Cell Endocrinol 301 :89-96, 2009 23. Gallagher CJ, Kadlubar FF, Muscat JE, et al: The UGT2B17 gene deletion polymorphism and risk of prostate cancer: A case-control study in Caucasians. Cancer Detect Prev 32:186- 187, 2008
24. Gsur A, Preyer M, Haidinger G, et al: A polymorphism in the UDP-Glucuronosyltransferase 2B15 gene (D85Y) is not associated with prostate cancer risk. Cancer Epidemiol Biomarkers
Prev 1 1 :497-8, 2002
25. Hajdinjak T, Zagradisnik B: Prostate cancer and polymorphism D85Y in gene for dihydrotestosterone degrading enzyme UGT2B15: Frequency of DD homozygotes increases with Gleason Score. Prostate. 59:436-9., 2004
26. Karypidis AH, Olsson M, Andersson SO, et al: Deletion polymorphism of the UGT2B17 gene is associated with increased risk for prostate cancer and correlated to gene expression in the prostate. Pharmacogenomics J 8: 147-51 , 2008
27. Olsson M, Lindstrom S, Haggkvist B, et al: The UGT2B 7 gene deletion is not associated with prostate cancer risk. Prostate 4:4, 2008
28. Okugi H, Nakazato H, Matsui H, et al: Association of the polymorphisms of genes involved in androgen metabolism and signaling pathways with familial prostate cancer risk in a
Japanese population. Cancer Detect Prev 30:262-8, 2006
29. Park J, Chen L, Ratnashinge L, et al: Deletion polymorphism of UDP- glucuronosyltransferase 2B17 and risk of prostate cancer in African American and Caucasian men. Cancer Epidemiol Biomarkers Prev. 15:1473-8., 2006
30. Park J, Chen L, Shade K, et al: Asp85tyr polymorphism in the udp-glucuronosyltransferase (UGT) 2B15 gene and the risk of prostate cancer. J Urol. 171 :2484-8., 2004
31. Menard V, Eap O, Harvey M, et al: Copy-number variations (CNVs) of the human sex steroid metabolizing genes UGT2B17 and UGT2B28 and their associations with a UGT2B15 functional polymorphism. Hum Mutat 30: 1310-1319, 2009
32. Cicek MS, Conti DV, Curran A, et al: Association of prostate cancer risk and aggressiveness to androgen pathway genes: SRD5A2, CYP17, and the AR. Prostate 59:69-76, 2004
33. Cussenot O, Azzouzi AR, Nicolaiew N, et al: Low-activity V89L variant in SRD5A2 is associated with aggressive prostate cancer risk: an explanation for the adverse effects observed in chemoprevention trials using 5-alpha-reductase inhibitors. Eur Urol 52:1082-7,
2007
34. Salam MT, Ursin G, Skinner EC, et al: Associations between polymorphisms in the steroid 5- alpha reductase type II (SRD5A2) gene and benign prostatic hyperplasia and prostate cancer. Urol Oncol 23:246-53, 2005
35. Thompson IM, Goodman PJ, Tangen CM, et al: The influence of finasteride on the development of prostate cancer. N Engl J Med 349:215-24, 2003 36. Huang SP, Huang LC, Ting WC, et al: Prognostic significance of prostate cancer susceptibility variants on prostate-specific antigen recurrence after radical prostatectomy. Cancer Epidemiol Biomarkers Prev 18:3068-74, 2009
37. Levesque E, Delage R, Benoit-Biancamano MO, et al: The impact of UGT1A8, UGT1A9, and UGT2B7 genetic polymorphisms on the pharmacokinetic profile of mycophenolic acid after a single oral dose in healthy volunteers. Clin Pharmacol Ther 81 :392-400, 2007
38. Haukaas SA, Halvorsen OJ, Daehlin L, et al: Is preoperative serum prostate-specific antigen level significantly related to clinical recurrence after radical retropubic prostatectomy for localized prostate cancer? BJU Int 97:51 -5, 2006
39. D'Amico AV, Whittington R, Malkowicz SB, et al: Predicting prostate specific antigen outcome preoperatively in the prostate specific antigen era. J Urol 166:2185-8, 2001 40. Levesque E, Beaulieu M, Green MD, et al: Isolation and characterization of UGT2B15(Y85): a UDP-glucuronosyltransferase encoded by a polymorphic gene. Pharmacogenetics 7:317- 25., 1997
41. McCarroll SA, Hadnott TN, Perry GH, et al: Common deletion polymorphisms in the human genome. Nat Genet 38:86-92, 2006
42. Yang TL, Chen XD, Guo Y, et al: Genome-wide Copy-Number-Variation Study Identified a Susceptibility Gene, UGT2B17, for Osteoporosis. Am J Hum Genet, 2008
43. Makridakis N, Ross RK, Pike MC, et al: A prevalent missense substitution that modulates activity of prostatic steroid 5alpha-reductase. Cancer Res 57: 1020-2, 1997
44. Makridakis NM, di Salle E, Reichardt JK: Biochemical and pharmacogenetic dissection of human steroid 5 alpha-reductase type II. Pharmacogenetics 10:407-13, 2000
45. Pearce CL, Van Den Berg DJ, Makridakis N, et al: No association between the SRD5A2 gene A49T missense variant and prostate cancer risk: lessons learned. Hum Mol Genet 17:2456-61 , 2008
46. Nadeau et al., J Clin Endocrinol Metab. 201 1 Sep;96(9):E 1550-7. Epub 201 1 Jul 6

Claims

1. An in-vitro method for predicting risk for prostate cancer in a human subject, said method comprising the steps of:
a) obtaining a nucleic acid from sample from said human subject; and b) determining the individual's UGT2B genetic variation status;
whereby the identification of at least one genetic variation in said UGT2B gene in said subject's nucleic acid is an indication that said subject has an increased risk of developing prostate cancer or progression thereof.
2. The method of claim 1 , wherein said UGT2B gene is selected from the group consisting of: UGT2B17 and UGT2B28.
3. The method of claim 2, wherein said genetic variation in UGT2B17 is one or two deletions,
4. The method of claim 2 or 3, wherein said genetic variation in UGT2B28 is one or two deletions.
5. The method of claim 2, 3 or 4, wherein said identification of genetic variation in said UGT2B17 or UGT2B28 comprises identifying at least one copy number variation (CNV) in said gene, said CNV being selected from the group consisting of: rs3100645; and rs1 1249532, and associated reference sequences thereof.
6. The method of claim 2, 3, 4 or 5, comprising at least one genetic variation in both UGT2B17 and UGT2B28 genes.
7. The method of claim 1 to 6, further comprising at least one genetic variation in SRD5A1 or SRD5A2 gene or both; whereby the co-occurence of at least one genetic variation in UGT and SRD5A1 or SRD5A2 in said subject's nucleic acid is an indication that said subject has a further increased likelihood that the prostate cancer will recur.
8. The method of claim 7, wherein said identification of genetic variation in said SRD5A1 or SRD5A2 comprises identifying at least one single nucleotide polymorphism (SNP) in said nucleic acids, said SNP being selected from the group consisting of: rs166050; rs2208532; rs523349; rs4952197; rs12470143; rs518673 and rs676033, and associated reference sequences thereof.
9. The method of claim 1 , wherein in step a) said nucleic acid is DNA or RNA.
10. The method of claim 9, wherein said genetic variation of DNA or RNA is detected with the use of a nucleic acid probe.
11. The method of claim 10, wherein said DNA is amplified by PCR prior to incubation with the probe.
12. The method of any one of claims 1 to 10, wherein said genetic variation of DNA or RNA is detected with the use of a methodology selected from the group consisting of: direct sequencing, pyrosequencing, massively parallel sequencing, high-throughput sequencing, high performance liquid chromatography (HPLC) fragment analysis, mass spectrometry, branch DNA and capillary electrophoresis.
13. The method of claim 12, wherein said methodology is carried out on DNA or RNA with prior amplification.
14. The method of claim 13, wherein said genetic variation in said DNA is detected with the use of a specific primer and a specific probe.
15. The method of claim 14, wherein the primer is selected from the group consisting of: SEQ ID Nos. 1 , 2, 7 or 8, and the probe hybridizes with a sequence selected from rs3100645 and/or rs1 1249532m, and associated reference sequences thereof.
16. The method of claim 1 , wherein the subject's nucleic acid-containing sample is a tumor or a non-tumor sample.
17. The method of claim 16, wherein the subject's non-tumor sample is selected from the group consisting of: tissue or biological fluid.
18. The method of claim 17, wherein said tissue sample is selected from the group consisting of: skin, lymph node, hair and buccal smear.
19. The method of claim 17, wherein said biological fluid sample is selected from the group consisting of: sputum, saliva, blood, serum, urine, semen and plasma.
20. The method of claim 17, wherein said tumor sample is obtained from a biopsy.
21. A method for adapting a course of treatment of prostate cancer in a human subject, comprising the steps of:
a) providing a prognosis or predicting the likelihood of a human subject to develop prostate cancer recurrence in accordance with any one of claims 1 to 20; and
b) adapting a course of treatment according to whether said subject has an increased likelihood that the cancer will recur.
22. The method according to claim 21 , wherein, when the likelihood of recurrence is increased, the subject is prescribed at least one of: hormone therapy, chemotherapy, surgery, radiotherapy, brachytherapy or active surveillance (watchful waiting).
23. The method according to claim 20 or 21 , wherein steps a) and b) are performed after the subject has undergone radical prostatectomy.
24. The method according to any one of claims 21 to 23, wherein, when the likelihood of recurrence is increased, the subject is prescribed UGT2B inducer therapy.
25. The method according to any one of claims 21 to 23, wherein, when the likelihood of recurrence is increased, the subject is prescribed 5a-reductase inhibitors therapy.
26. A kit for predicting the likelihood of a human subject to develop prostate cancer recurrence by detecting a genetic variation according to any one of claims 1 to 8; said kit comprising reagents for determining the individual's genetic variations (or haplotypes) in UGT2B gene.
27. The kit of claim 26, comprising a reagent for detecting a reference sequence selected from the group consisting of: rs3100645; and rs1 1249532; and associated sequences thereof.
28. The kit of claim 25 or 26, comprising a PCR primer-probe set, wherein the primer is selected from the group consisting of: SEQ ID Nos. 1 , 2, 7 or 8 and the probe hybridizes with a sequence selected from rs3100645 and/or rs1 1249532, and associated reference sequences thereof.
29. The kit of claim 28, further comprising reagents for determining the individual's genetic variations (or haplotypes) in SRD5A1 or SRD5A2 gene.
Particularly, the kit further comprises reagents for detecting a SNP in a reference sequence selected from the group consisting of: rs518673; rs166050; rs12470143;
rs2208532; rs523349; rs4952197 and rs676033 or their associated SNPs.
30. The kit of claim 29, comprising PCR primer-probe set, wherein the primer is selected from the group consisting of: SEQ ID Nos. 12 to 50; and the probe is selected from the group consisting of: SEQ ID Nos. 51 to 69.
31. A method of predicting the response to hormonotherapy for treating prostate cancer in a subject, the method comprising carrying the steps according to any one of claims 1 to 25; whereby the determination of at least one genetic variation in said UGT2B gene in said subject's nucleic acid is an indication that said treatment regimen is not efficacious for treating prostate cancer in the subject.
32. A method of predicting the response to 5-alpha inhibitor therapy for treating prostate cancer in a subject, the method comprising carrying the steps according to any one of claims 7 or 8; whereby the determination of at least one genetic variation in said SRD5A1 or SRD5A2 gene in said subject's nucleic acid is an indication that said treatment regimen is not efficacious for treating prostate cancer in the subject.
33. A biomarker for predicting the likelihood of prostate cancer progression or recurrence in a human subject, comprising a genetic variation in UGT2B17 or UGT2B28 gene of said human subject.
34. A genetic variation in UGT2B17 or UGT2B28 gene of a human subject for use as a biomarker for predicting the likelihood of prostate cancer progression or recurrence in said human subject.
PCT/CA2011/001338 2010-12-02 2011-12-02 Prognostic markers of inherited variations in the ugt2b genes for prostate cancer recurrence WO2012071665A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US41905710P 2010-12-02 2010-12-02
US61/419,057 2010-12-02

Publications (1)

Publication Number Publication Date
WO2012071665A1 true WO2012071665A1 (en) 2012-06-07

Family

ID=46171129

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2011/001338 WO2012071665A1 (en) 2010-12-02 2011-12-02 Prognostic markers of inherited variations in the ugt2b genes for prostate cancer recurrence

Country Status (1)

Country Link
WO (1) WO2012071665A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020036655A3 (en) * 2018-04-25 2020-04-02 Juneau Biosciences, L.L.C. Methods of using genetic markers associated with endometriosis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999037986A2 (en) * 1998-01-23 1999-07-29 University Of Southern California Androgen-metabolic gene mutations and prostate cancer risk
US20060051763A1 (en) * 2002-09-25 2006-03-09 Anu-Maria Loukola Detection methods

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999037986A2 (en) * 1998-01-23 1999-07-29 University Of Southern California Androgen-metabolic gene mutations and prostate cancer risk
US20060051763A1 (en) * 2002-09-25 2006-03-09 Anu-Maria Loukola Detection methods

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MENARD V. ET AL.: "Copy number variations (CNVs) of the human sex steroid metabolizing genes UGT2B 17 and UGT2B28 and their associations with UGT2B 15 functional polymorphism.", HUMAN MUTATION, vol. 30, no. ISS.10, October 2009 (2009-10-01), pages 1310 - 1319 *
NADEAU G ET AL.: "Deletions of the androgen-metabolizing UGT2B genes have an effect on circulating steroid levels and biochemical recurrence after radical prostatectomy in localized prostate cancer.", THE J. OF CLINICAL ENDOCRINOLOGY & METABOLISM., vol. 96, no. 9, September 2011 (2011-09-01), pages E1550 - E1557 *
PARK J ET AL.: "ASP85TYR polymorphism in the UDP-glucuronosvltlansferase (UGT) 2B15 gene and the risk of prostate cancer.", THE JOURNAL OF UROLOGY, vol. 171, no. ISS.6, June 2004 (2004-06-01), pages 2484 - 2488 *
PARK JONG. ET AL.: "Deletion polymorphism of UDP-Glucuronosvltlansferase 2B17 and risk of prostate cancer inJAfrican and Caucasian men.", CANCER EPIDEMIOL BIOMARKERS PREV., vol. 15, 2006, pages 1473 - 1478 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020036655A3 (en) * 2018-04-25 2020-04-02 Juneau Biosciences, L.L.C. Methods of using genetic markers associated with endometriosis

Similar Documents

Publication Publication Date Title
Cunningham et al. Evaluation of genetic variations in the androgen and estrogen metabolic pathways as risk factors for sporadic and familial prostate cancer
Huiping et al. High frequency of LOH, MSI and abnormal expression of FHIT in gastric cancer
Cicek et al. Association of prostate cancer risk and aggressiveness to androgen pathway genes: SRD5A2, CYP17, and the AR
AU2008212117B2 (en) Genetic variants contributing to risk of prostate cancer
Beuten et al. Single and multigenic analysis of the association between variants in 12 steroid hormone metabolism genes and risk of prostate cancer
Kwon et al. Androgen metabolism and JAK/STAT pathway genes and prostate cancer risk
Lindström et al. Systematic replication study of reported genetic associations in prostate cancer: Strong support for genetic variation in the androgen pathway
Audet-Walsh et al. SRD5A polymorphisms and biochemical failure after radical prostatectomy
Karakas et al. Molecular mechanisms involving prostate cancer racial disparity
Setlur et al. Genetic variation of genes involved in dihydrotestosterone metabolism and the risk of prostate cancer
Schatzl et al. Association of polymorphisms within androgen receptor, 5α‐reductase, and PSA genes with prostate volume, clinical parameters, and endocrine status in elderly men
Audet-Walsh et al. The impact of germline genetic variations in hydroxysteroid (17-beta) dehydrogenases on prostate cancer outcomes after prostatectomy
US20220002810A1 (en) Methods and compositions for correlating genetic markers with prostate cancer risk
Wright et al. CYP17 polymorphisms and prostate cancer outcomes
WO2013049926A1 (en) PROGNOSTIC MARKERS OF INHERITED VARIATIONS IN THE 17β-HYDROXYSTEROID DEHYDROGENASE (HSD17B) GENES FOR PROSTATE CANCER
Ziaran et al. Biomarkers for determination prostate cancer: implication for diagnosis and prognosis
Mononen et al. Polymorphisms in genes involved in androgen pathways as risk factors for prostate cancer
Karatzas et al. Genetic polymorphisms in the UDP-glucuronosyltransferase 1A1 (UGT1A1) gene and prostate cancer risk in Caucasian men
US20130149703A1 (en) &#34;markers for prostate cancer progression&#34;
Habibi et al. Genetic variations in UGT2B28, UGT2B17, UGT2B15 genes and the risk of prostate cancer: A case-control study
Sun et al. Genetic variants in MMP9 and TCF2 contribute to susceptibility to lung cancer
Choubey et al. SRD5A2 gene polymorphisms and the risk of benign prostatic hyperplasia but not prostate cancer
Novelli et al. Pharmacogenetics of human androgens and prostatic diseases
Rajender et al. Longer (TA) n repeat but not A49T and V89L polymorphisms in SRD5A2 gene may confer prostate cancer risk in South Indian men
WO2012071665A1 (en) Prognostic markers of inherited variations in the ugt2b genes for prostate cancer recurrence

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11845458

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11845458

Country of ref document: EP

Kind code of ref document: A1