Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Precision Molecular Pathology of Prostate Cancer
Precision Molecular Pathology of Prostate Cancer
Precision Molecular Pathology of Prostate Cancer
Ebook1,350 pages13 hours

Precision Molecular Pathology of Prostate Cancer

Rating: 0 out of 5 stars

()

Read preview

About this ebook

This volume focuses on our current understanding of the molecular underpinnings of prostate cancer and their potential application for precision medicine approaches.  The emergence and applications of new technologies has allowed for a rapid expansion of our understanding of the molecular basis of prostate cancer and has revealed a remarkable genetic heterogeneity that may underlie the clinically variable behavior of the disease.  The book consists of five sections which provide insight about the following: (1) General principles; (2) Molecular signatures of primary prostate cancer; (3) Molecular signatures of advanced prostate cancer; (4) Key molecular pathways in prostate cancer development and progression; (5) and Precision medicine approach: Diagnosis, treatment, prognosis.  Precision Molecular Pathology of Prostate Cancer is an important resource for the practicing oncologist, urologist, and pathologist, and will also be useful for researchers in the prostate cancer community.
LanguageEnglish
PublisherSpringer
Release dateFeb 13, 2018
ISBN9783319640969
Precision Molecular Pathology of Prostate Cancer

Related to Precision Molecular Pathology of Prostate Cancer

Titles in the series (2)

View More

Related ebooks

Medical For You

View More

Related articles

Reviews for Precision Molecular Pathology of Prostate Cancer

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Precision Molecular Pathology of Prostate Cancer - Brian D. Robinson

    Part IGeneral Principles

    © Springer International Publishing AG 2018

    Brian D. Robinson, Juan Miguel Mosquera, Jae Y. Ro and Mukul Divatia (eds.)Precision Molecular Pathology of Prostate CancerMolecular Pathology Libraryhttps://doi.org/10.1007/978-3-319-64096-9_1

    1. Precision Medicine in Prostate Cancer: Approach to the Patient

    Beerinder S. Karir¹, Bishoy M. Faltas¹ and Scott T. Tagawa¹  

    (1)

    Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York Presbyterian Hospital, New York, NY, USA

    Scott T. Tagawa

    Email: stt2007@med.cornell.edu

    Introduction

    Precision Medicine in Prostate Cancer

    Initial Clinical Encounter

    Tissue for Genomic Profiling and Need for Serial Biopsies

    Understanding Precision Medicine Reports

    Communicating Genomic Information to Clinicians and Patients

    Unique Issues Related to Incidental Discovery of Germline Mutations

    Future Direction

    Resources for Patients and Clinicians

    References

    Keywords

    Next-generation sequencingPrecision medicineGenomic profilingPatient preferencesData privacy

    Introduction

    The advent of genomic discoveries and decreasing cost of next-generation sequencing (NGS) technologies has ushered in a new era of precision medicine [1]. The early effects of this paradigm shift in oncology are beginning to impact patient care and thus increase the relevance of a discussion on the approach to patients with prostate cancer (PC). Bringing the application of precision medicine to the clinic raises new challenges related to informed consent prior to testing, effectively communicating the results of cancer genomic testing to the patient, understanding and managing the patient’s expectations, and working with the patient to select the best treatment options based on genomic tests (or not) [2, 3]. This introductory chapter will cover the approach to men with PC undergoing genomic testing of their tumors (see Table 1.1 for summary).

    Table 1.1

    Summary: approach to patient

    Precision Medicine in Prostate Cancer

    Prostate cancer (PC) is the most common non-skin cancer and the second leading cause of cancer death in men in the United States [4]. This disease is on the forefront of precision medicine with multiple opportunities for benefiting from translational genomics [5]. While any given man might benefit from individualized tumor testing, two very important clinical subsets within PC that might benefit most from additional molecular analysis are patients with clinically localized indolent disease that are probably best served without treatment and those with more aggressive, particularly advanced disease with no curative therapy.

    Newly Diagnosed Prostate Cancer

    Every year more than 1,000,000 men in the United States undergo prostatic biopsies based on elevated levels of prostate-specific antigen (PSA) . Among the newly diagnosed PC patients, many patients have clinically diagnosed (presumed) low-volume and low- to intermediate-risk Gleason scores (3 + 3 or 3 + 4 in a small percent of cores). This subset of PC patients presents a clinical dilemma for patients and their treating physicians. Various non-genomic biomarkers like Prostate Health Index (PHI) have been shown to better identify patients with aggressive disease, but there is still an unmet need for better biomarkers [6]. Genomic biomarkers including gene panels like the Decipher genomic classifier and Oncotype DX have demonstrated the ability to better stratify PC patients [7]. After various positive validating studies, Oncotype DX has recently been included under Medicare coverage thereby making such PC genomic-based diagnostics reimbursable [8]. In addition to gene panels, various other genomic biomarkers like urine TMPRSS2-ERG fusion transcripts and long noncoding RNAs(lncRNAs) may show promise in identifying PC patients who may need aggressive treatments [9, 10].

    Advanced Prostate Cancer

    On the opposite end of the spectrum, advanced PC represents another disease state that could benefit from precision medicine approaches. Prostate tumors may remain responsive to androgen deprivation therapy for years (variable among patients) until it evolves into the castration-resistant state (CRPC) , which generally is still driven by the AR pathway. The median overall survival after diagnosis of CRPC is 18–32 months. Although newer-generation hormonal, cytotoxic, immunotherapeutic, and bone-targeted drugs have increased survival in CRPC patients leading to their FDA approval, the development of resistant PC disease remains inevitable. Using a combination of improved biopsy techniques and NGS technologies, molecular characterization of such advanced prostate tumors is increasingly being done. Recently, one such multi-institutional study found that 90% of advanced PC tumor harbor molecular alteration with potential targeting agent/drugs [11]. Another study that included mostly prostate cancer patients also demonstrated targetable alterations with an in-depth analysis of an exceptional responder based upon this mutation [12]. This highlights the importance of precision medicine for subclassification of prostate disease into molecularly defined subgroups with each subtype amenable to different targeted therapies [13].

    Initial Clinical Encounter

    The initial patient visit to a physician’s office generally includes comprehensive elicitation of disease history including diagnosis and initial treatments. Successful integration of precision medicine into oncology clinic will further emphasize clinical data recording and sharing. Without accurate linkage of genomic data to clinical data, even the latest genomic technologies will have a limited impact on patient outcomes [14].

    Family History in Prostate Disease

    Within PC patients’ histories, ethnicity is relevant as PC may sometimes harbor genetic determinants. Various single nucleotide polymorphism (SNPs) and copy number variants (CNVs) have been shown to be determinants of familial risk of prostate cancer [15]. Additionally, it is being increasingly realized that germline alterations like BRCA2, ATM, and BRCA1 mutations also play an important role in PC pathogenesis [11, 16]. So, any family history of such gene abnormalities can warrant increased level of diagnostic and therapeutic interventions.

    Informed Consent

    Precision medicine informed consents are important part of this new paradigm. Informed consents must delineate all the details including risks and also elaborate upon likelihood of finding somatic molecular alterations of unknown significance as well as incidental germline mutations. Pretest counseling should focus on addressing the key components of informed consent.

    Patient Preferences and Data Privacy

    One of the most important parts of the consent is the preferences of patients and families regarding level of detail and the scope of genetic information resulting from molecular testing, especially regarding incidental findings. This issue is discussed in detail in a separate section. Risks due to testing procedures (i.e., biopsy procedures) as well as data privacy should also be clearly detailed. Patients should also be made aware of existing legal protection against discrimination and the provisions of the Genetic Information Nondiscrimination Act (GINA). GINA protects US citizens from discrimination and restricts insurers from limiting coverage/altering premiums based on such genetic information. It prevents insurers from requiring policyholders to undergo genetic testing but could make testing a requirement for treatment [17].

    Actionability of Precision Medicine Results

    Due to enormous media attention generated by the precision medicine initiative started by President Obama [18], patients have high expectations from genomic profiling and its implications especially in terms of cancer cure [19]. These expectations, especially as they relate to the actionability of results, should be addressed up front within the framework of the consent process. The possibilities of nonactionable genomic results, biopsy failure (poor tissue quality/tumor content), analytical validity issues, and turnaround time should be discussed with the patient during this process [20]. Realistic expectations set through early patient education lead to better patient compliance and satisfaction. When managed appropriately, the potential for personal benefit from targeted therapy raises hopes and drives enhanced participation of patients in clinical trials [3].

    It is important to understand that the definition of actionable molecular alterations is dependent on several molecular, patient-specific, and practical factors. A recent survey of practicing oncologists who had just received their patients’ cancer genome sequencing reports showed that 78% did not expect to implement any changes to the current treatment plan [21]. In this study, barriers to actionability included lack of local clinical trials (41%), absence of actionable mutation (33%), and good response to ongoing treatment (16%). In light of these findings, physicians need to explain to patients all the factors that could limit actionability of precision medicine test results.

    Turnaround Time

    Patient’s expectations about the turnaround time for genomic profile results also need to be recalibrated. Presently, waiting time ranging from weeks to months is needed starting with acquisition of tumor sample plus germline sample to generation of a precision medicine report. This is acceptable to stable patients and their treating physicians, but for patients with progressive advanced cancer, such long waiting time may not be clinically useful. Though the latest NGS methodologies have significantly shortened turnaround times, bottlenecks in the process still remain. These include sample acquisition and logistics and data analysis and interpretation [20].

    Tissue for Genomic Profiling and Need for Serial Biopsies

    Successful application of precision medicine requires availability of tissue of origin and/or metastatic site [22]. In many cases with a distant history of prostatectomy, tissue acquisition is not feasible thus leaving only the option of metastatic site biopsy. As most common site for PC metastasis is the bone which is a difficult organ to biopsy, metastatic site biopsies in PC have been very daunting process until lately. However, advancements in biopsy technology have increased the chances of successful tissue procurement from a PC patient [11, 23].

    Over time, the true success of precision medicine may hinge on our ability to get serial biopsies to see real-time genomic evolution of the prostate disease. Liquid biopsy technologies such as circulating tumor cells (CTCs) or cell-free DNA (cfDNA) are a good surrogate for tissue biopsies [24]. Though true utility of liquid biopsies needs further validation [25], application of liquid biopsies seems immense extending to CTC-derived xenografts [26]. Another useful application of CTCs for prostate cancer patients may be in generation of patient-derived organoid cultures. Such cultures have shown to recapitulate the entire molecular diversity of prostate disease and hold promise for use in genetic and pharmacologic studies [27].

    Understanding Precision Medicine Reports

    All procedures for precision medicine outside of a research setting should be performed in CLIA-certified labs. After running the tissue sample through sequencing pipelines, genomic data is streamed through analytical/bioinformatic pipeline. The entire process needs to be standardized throughout for validity. Eventually a precision medicine report on patient’s tumor-specific somatic alterations is generated and usually contains the following elements:

    Somatic alterations in clinically relevant genes—these alterations occur in genes that are potentially actionable as drug targets or confer resistance or susceptibility to treatment.

    Somatic alterations of unknown significance in known cancer genes—these alterations occur in genes that are cancer associated, but their impact on the disease is not fully understood.

    Somatic alterations of unknown significance—these alterations are not known to have any effect on the disease but are profiled in the event that, in future, progress in scientific knowledge could determine their role.

    In addition to these, details on quality control metrics like depth and coverage of sequencing are often provided [12].

    Communicating Genomic Information to Clinicians and Patients

    A recent study done at Duke Medical Centre has found a number of challenges faced by institution when implementing genomic testing into patient care [28]. This necessitates a policy and education program to improve clinician support, enabling them to effectively deliver precision medicine care. One such problem is that precision medicine reports may or may not yield actionable somatic alterations in cancer-related genes. If such molecular alterations are found, these can either be targeted by FDA-approved drug for PC or other cancer types (i.e., off-label use), or there may be approved or investigational drug available as a clinical trial. Such drugs/trials are often enlisted in precision medicine reports made available to treating physician. But procuring the drugs targeting actionable genomic alteration can be a big hurdle. This can be especially problematic if PC is an off-label use of the drug [29].

    For effectively communicating genomic results to patients, the treating physician needs to ensure proper patient education (starting with pretest counseling) and decision support systems are in place. This may require strong collaboration among genetic counselors, physicians, and nurses. Realistic expectations set during informed consent education can be especially helpful if sequencing results yield no obvious actionable alterations. In the case of incidental germline finding , the role of genetic counselors is very important to facilitate family communication [30] (see next section). To conclude, the proper utilization of cancer genomic medicine needs to be accompanied by careful thought about how the genetic test results will be communicated to patients in order to maximize their benefit.

    Unique Issues Related to Incidental Discovery of Germline Mutations

    Genome sequencing provides unprecedented opportunities to study the genomic landscapes and identify the actionable driver mutations for targeted therapy in precision medicine clinics. Because some NGS approaches rely on comparison between germline and somatic variants, germline alterations may be incidentally discovered. These alterations may be associated with inherited health risk or familial susceptibility to cancer. In the setting of cancer, some patients may find it burdensome to bear the knowledge of such inherited health risk in family [31]. This may have psychological consequences associated with the guilt of passing the inherited risk or increased cost of health care. This knowledge is perceived as an obligation to family and is difficult to refuse. Providing patients with simple summaries to share with their families and making local genetic counseling resources available at point of contact for the family can be helpful during the process [32]. Implications of reporting such incidental discoveries of germline mutations are very complex. Discussing these issues during the informed consent for sequencing highly penetrant disease genes and genetic counseling is essential to address the challenges faced in this situation [33]. Overall, the likelihood of finding incidental genetic variants does not appear to significantly discourage patients from adopting genomic profiling though the extent of incidental findings patients wish to be disclosed varies significantly [34].

    Future Direction

    Successful application of precision medicine approaches in the routine clinical care of patients requires not only a wider availability of next-generation sequencing technologies but also resourceful databases possessing consolidated clinical information [14]. As we proceed ahead, the missing metrics of clinical data will need to be filled in and clinical information annotated with genomic data. Another issue will be to improve our ability to complete the entire process of genomic sequencing, generating reports, and matching/administering drugs targeting driver alterations within a rapid turnaround time . This may further require sophisticated rapid machine learning methods [35]. In addition to therapeutic benefits, the promise of precision medicine in prostate cancer will also lie in discovering and validating molecular biomarkers that distinguish aggressive from indolent disease and those that predict treatment resistance [36, 37]. Finally regarding our heightened expectations, we will need to be cautious in terms of seeking quick results through this new paradigm of precision medicine. As Amara’s law correctly states We tend to overestimate the effect of a technology in the short run and underestimate the effect in the long run. We may have outclassed Moore’s law for NGS method cost efficacy, but regarding patient outcomes, it may take few years before we realize the full potential of precision medicine for prostate cancer patients.

    Resources for Patients and Clinicians

    My Cancer Genome—http://​www.​mycancergenome.​org/​: This is a personalized cancer medicine knowledge resource for physicians, patients, caregivers, and researchers. It provides latest information on what mutations make cancers grow and related therapeutic implications, including available clinical trials.

    cBioPortal—http://​www.​cbioportal.​org/​: This portal maintained by Memorial Sloan Kettering Cancer Center stores genomic data from large-scale, integrated cancer genomic data sets. It allows explorative genomic data analysis

    COSMIC database—http://​cancer.​sanger.​ac.​uk/​cosmic: COSMIC is a freely available online database of somatically acquired mutations found in human cancer. It is maintained by Sanger Institute, UK.

    National Cancer Institute Cancer Genetics Services Directory—http://​www.​cancer.​gov/​about-cancer/​causes-prevention/​genetics/​directory: This NCI directory lists professionals who provide services related to cancer genetics (cancer risk assessment, genetic counseling, genetic susceptibility testing, and others).

    National Society of Genetic Counselors (NSGC)—http://​nsgc.​org/​p/​cm/​ld/​fid=​164: For finding genetic counselors in a local area, United States.

    References

    1.

    Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372(9):793–5.CrossrefPubMedPubMedCentral

    2.

    Hollebecque A, Massard C, Soria J-C. Implementing precision medicine initiatives in the clinic: a new paradigm in drug development. Curr Opin Oncol. 2014;26(3):340–6.CrossrefPubMed

    3.

    Miller FA, Hayeems RZ, Bytautas JP, Bedard PL, Ernst S, Hirte H, et al. Testing personalized medicine: patient and physician expectations of next-generation genomic sequencing in late-stage cancer care. Eur J Hum Genet. 2013;22(3):391–5.CrossrefPubMedPubMedCentral

    4.

    Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29.CrossrefPubMed

    5.

    Beltran H, Rubin MA. New strategies in prostate cancer: translating genomics into the clinic. Clin Cancer Res. 2013;19(3):517–23.CrossrefPubMed

    6.

    De la Calle C, Patil D, Wei JT, Scherr DS, Sokoll L, Chan DW, et al. Multicenter evaluation of the prostate health index to detect aggressive prostate cancer in biopsy naïve men. J Urol. 2015;5347(15):183–4.

    7.

    Cooperberg MR, Davicioni E, Crisan A, Jenkins RB, Ghadessi M, Karnes RJ. Combined value of validated clinical and genomic risk stratification tools for predicting prostate cancer mortality in a high-risk prostatectomy cohort. Eur Urol. 2015;67(2):326–33.CrossrefPubMed

    8.

    Genomic Health Announces Favorable Draft Local Coverage Determination from Palmetto GBA® on Medicare Coverage for the Oncotype DX® Prostate Cancer Test [Internet]. [cited 2015 May 27]. Available from: http://​www.​prnewswire.​com/​news-releases/​genomic-health-announces-favorable-draft-local-coverage-determination-from-palmetto-gba-on-medicare-coverage-for-the-oncotype-dx-prostate-cancer-test-300087008.​html.

    9.

    Tomlins SA, Aubin SMJ, Siddiqui J, Lonigro RJ, Sefton-Miller L, Miick S, et al. Urine TMPRSS2:ERG fusion transcript stratifies prostate cancer risk in men with elevated serum PSA. Sci Transl Med. 2011;3(94):94ra72.CrossrefPubMedPubMedCentral

    10.

    Prensner JR, Zhao S, Erho N, Schipper M, Iyer MK, Dhanasekaran SM, et al. RNA biomarkers associated with metastatic progression in prostate cancer: a multi-institutional high-throughput analysis of SChLAP1. Lancet Oncol. 2014;15(13):1469–80.CrossrefPubMedPubMedCentral

    11.

    Robinson D, Van Allen EM, Wu Y-M, Schultz N, Lonigro RJ, Mosquera J-M, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161(5):1215–28.CrossrefPubMedPubMedCentral

    12.

    Beltran H, Eng K, Mosquera JM, Sigaras A, Romanel A, Rennert H, et al. Whole-exome sequencing of metastatic cancer and biomarkers of treatment response. JAMA Oncol. 2015 1(4):466-474

    13.

    Roychowdhury S, Chinnaiyan AM. Advancing precision medicine for prostate cancer through genomics. J Clin Oncol. 2013;31(15):1866–73.CrossrefPubMedPubMedCentral

    14.

    Rubin MA. Health: make precision medicine work for cancer care. Nature. 2015;520(7547):290–1.CrossrefPubMed

    15.

    Demichelis F, Stanford JL. Genetic predisposition to prostate cancer: update and future perspectives. Urol Oncol. 2015;33(2):75–84.CrossrefPubMed

    16.

    Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, Garofalo A, Gulati R, Carreira S, Eeles R, Elemento O, Rubin MA, Robinson D, Lonigro R, Hussain M, Chinnaiyan A, Vinson J, Filipenko J, Garraway L, Taplin ME, AlDubayan S, Han GC, Beightol M, Morrissey C, Nghiem B, Cheng HH, Montgomery B, Walsh T, Casadei S, Berger M, Zhang L, Zehir A, Vijai J, Scher HI, Sawyers C, Schultz N, Kantoff PW, Solit D, Robson M, Van Allen EM, Offit K, de Bono J, Nelson PS. Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer. N Engl J Med. 2016 Aug 4;375(5):443-53.

    17.

    Cornetta K, Brown CG. Balancing personalized medicine and personalized care. Acad Med. 2013;88(3):309–13.CrossrefPubMedPubMedCentral

    18.

    Kaiser J. Obama gives East Room rollout to Precision Medicine Initiative [Internet]. Science. 2015 [cited 2015 Jun 29]. Available from: http://​news.​sciencemag.​org/​biology/​2015/​01/​obama-gives-east-room-rollout-precision-medicine-initiative.

    19.

    Reardon S. Precision-medicine plan raises hopes. Nature. 2015;517:540.CrossrefPubMed

    20.

    Simon R, Roychowdhury S. Implementing personalized cancer genomics in clinical trials. Nat Rev Drug Discov. 2013;12(5):358–69.CrossrefPubMed

    21.

    Bartnik N, Roberts J, Gornick M, Le L, Chinnaiyan A. Are oncologists using genome sequencing results to inform patient care? 2015 ASCO Annual Meeting Abstracts [Internet]. J Clin Oncol. 2015 [cited 2015 May 13]. p. suppl; abstr e12525. Available from: http://​abstracts.​asco.​org/​156/​AbstView_​156_​151842.​html.

    22.

    Cohen RL, Settleman J. From cancer genomics to precision oncology—tissue’s still an issue. Cell. 2014;157(7):1509–14.CrossrefPubMed

    23.

    Van Allen EM, Foye A, Wagle N, Kim W, Carter SL, McKenna A, et al. Successful whole-exome sequencing from a prostate cancer bone metastasis biopsy. Prostate Cancer Prostatic Dis. 2014;17(1):23–7.CrossrefPubMed

    24.

    Toss A, Mu Z, Fernandez S, Cristofanilli M. CTC enumeration and characterization: moving toward personalized medicine. Ann Transl Med. 2014;2(11):108.PubMedPubMedCentral

    25.

    Gold B, Cankovic M, Furtado LV, Meier F, Gocke CD. Do circulating tumor cells, exosomes, and circulating tumor nucleic acids have clinical utility?: a report of the association for molecular pathology. J Mol Diagn. 2015;17(3):209–24.CrossrefPubMedPubMedCentral

    26.

    Yap TA, Lorente D, Omlin A, Olmos D, de Bono JS. Circulating tumor cells: a multifunctional biomarker. Clin Cancer Res. 2014;20(10):2553–68.CrossrefPubMed

    27.

    Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159(1):176–87.CrossrefPubMedPubMedCentral

    28.

    Katsanis SH, Minear MA, Vorderstrasse A, Yang N, Reeves JW, Rakhra-Burris T, et al. Perspectives on genetic and genomic technologies in an academic medical center: the duke experience. J Pers Med. 2015;5(2):67–82.CrossrefPubMedPubMedCentral

    29.

    Rodon J, Soria JC, Berger R, Batist G, Tsimberidou A, Bresson C, et al. Challenges in initiating and conducting personalized cancer therapy trials: perspectives from WINTHER, a Worldwide Innovative Network (WIN) Consortium Trial. Ann Oncol. 2015;26(8):1791–8.CrossrefPubMedPubMedCentral

    30.

    Forrest K, Simpson SA, Wilson BJ, van Teijlingen ER, McKee L, Haites N, et al. To tell or not to tell: barriers and facilitators in family communication about genetic risk. Clin Genet. 2003;64(4):317–26.CrossrefPubMed

    31.

    Stadler ZK, Schrader KA, Vijai J, Robson ME, Offit K. Cancer genomics and inherited risk. J Clin Oncol. 2014;32(7):687–98.CrossrefPubMed

    32.

    Communicating Genetic Test Results to Patients and Their Families [Internet]. [cited 2015 Jun 2]. Available from: https://​am.​asco.​org/​communicating-genetic-test-results-patients-and-their-families.

    33.

    Everett JN, Gustafson SL, Raymond VM. Traditional roles in a non-traditional setting: genetic counseling in precision oncology. J Genet Couns. 2014;23(4):655–60.CrossrefPubMedPubMedCentral

    34.

    Yushak M, Bouberhan S, Han G, Epstein L, Mougalian S, Abu-Khalaf M, et al. Patient preferences for disclosure of incidental genetic information discovered through high-throughput sequencing [Internet]. 2015 [cited 2015 Apr 28]. Available from: http://​www.​abstractsonline.​com/​Plan/​ViewAbstract.​aspx?​mID=​3682&​sKey=​ec0677d3-a055–44be-bfb3–7420b2d03869&​cKey=​9ca3a8ba-f3e7–428b-a162–67ffd36a260a&​mKey=​19573a54-ae8f-4e00–9c23-bd6d62268424.

    35.

    Shrager J, Tenenbaum JM. Rapid learning for precision oncology. Nat Rev Clin Oncol. 2014;11(2):109–18.CrossrefPubMed

    36.

    Rubin MA. Toward a prostate cancer precision medicine. Urol Oncol. 2015;33(2):73–4.CrossrefPubMed

    37.

    Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371(11):1028–38.CrossrefPubMedPubMedCentral

    © Springer International Publishing AG 2018

    Brian D. Robinson, Juan Miguel Mosquera, Jae Y. Ro and Mukul Divatia (eds.)Precision Molecular Pathology of Prostate CancerMolecular Pathology Libraryhttps://doi.org/10.1007/978-3-319-64096-9_2

    2. Epidemiology and Etiology

    Padraic O’Malley¹  

    (1)

    Department of Urology, Queen Elizabeth II Health Sciences Centre, Dalhousie University, Halifax, Nova Scotia, Canada

    Padraic O’Malley

    Email: pomalley@dal.ca

    Introduction: Incidence and Mortality

    Etiology and Risk Factors

    Conclusions

    References

    Keywords

    AgeFamily historyRace and ethnicityMetabolicSmoking

    Introduction: Incidence and Mortality

    In 2016, in the USA, approximately 180,890 men are expected to be diagnosed with prostate cancer, and almost 26,120 men are expected to lose their life to prostate cancer [1]. It is the number one non-cutaneous, solid tumor in men and the second most common cause of cancer death in the USA among men [2]. The estimated number of new cases worldwide in 2012 was 1,112,000, making it the second most common cancer diagnosed in men [3]. 759,000 of these cases are estimated to be seen in developed countries and only 353,000 in developing countries [3]. The estimated cases of prostate cancer deaths were 307,500 worldwide, with 142,000 in developed countries and 165,500 in developing countries [3]. Cumulative lifetime risk for prostate cancer incidence varied markedly between developed countries at 8.8% versus only 1.7% in developing countries. However, lifetime mortality risks were less disparate at 0.8% and 0.6%, respectively [3]. The incidence of prostate cancer underwent a dramatic increase in the USA in the early 1990s (Fig. 2.1) [2] with the widespread introduction of transurethral prostatectomy and then the prostate-specific antigen (PSA) test , leading to an almost threefold increase in incidence in 1975 to its peak in 1993 [2, 4, 5].

    ../images/324579_1_En_2_Chapter/324579_1_En_2_Fig1_HTML.gif

    Fig. 2.1

    Trends in incidence rates for selected cancers in men, USA, 1975–2011. Rates are age adjusted to the 2000 US standard population and adjusted for delays in reporting. Asterisk includes intrahepatic bile duct

    This dramatic rise was not seen in other high-income countries with less widespread adoption of PSA testing, such as those in Western Europe, and these countries demonstrated a gradual increase in incidence instead [6]. As such, due to screening and PSA utilization, as well as no doubt due to the variable risk of disease in certain population, there are marked variations, as much as 25-fold, in incidence and mortality globally (Fig. 2.2) [3, 6].

    ../images/324579_1_En_2_Chapter/324579_1_En_2_Fig2_HTML.gif

    Fig. 2.2

    Prostate cancer incidence and mortality rates by world area. (Reproduced from Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015 Feb 4;65(2):87–108)

    Fortunately, we have also seen a decline in mortality rates, particularly in developed countries. Beginning in 1996, the mortality rates were seen to decline after the introduction of PSA testing and continued to fall, perhaps due to this testing and/or due to improved treatment strategies, most likely in the metastatic setting [7, 8]. The effect of radical treatment on localized disease is less clear as to its overall benefit given the conflicting results of the Prostate Cancer Intervention Versus Observation Trial (PIVOT) and the Scandinavian Prostate Cancer Group 4 randomized trial [9, 10]. In the metastatic setting, however, we have seen the advent of many new therapeutics in the last 25 years including docetaxel and most recently abiraterone and enzalutamide, as well as combination therapy as seen in the recent CHAARTED and STAMPEDE studies and the benefits in overall survival these treatments have offered [11–13].

    Etiology and Risk Factors

    Over the last few decades, we have gained an increasing understanding of prostate cancer. Although we do not know the exact etiology of prostate cancer, we have identified a number of risk factors through epidemiological studies that may provide insight into possible mechanisms that would account for it. These factors can be divided into two distinct groups as seen in Table 2.1.

    Table 2.1

    Risk factors for prostate cancer by category

    To begin let us examine each category’s individual risk factors and the epidemiological evidence behind their possible role in prostate cancer:

    Modifiable

    Age

    Although several guidelines suggest screening men as young as 40 if they have high-risk features, prostate cancer is a disease of older men. Prostate cancer does have the steepest age-incidence curve of all malignancies, predominantly in the seventh decade [14–17]. The incidence of prostate cancer in men 45–54 years of age has remained fairly stable at approximately 35.7 per 100,000 in 2009–2011 [18]. There is a moderate increase in the following decade to 236 per 100,000. However, the subsequent two decades, 65–74 and 75–84, are both significantly higher at 609 and 769 per 100,000, respectively, in the modern era (Fig. 2.3). In North America, SEER data suggests approximately 0.6% of prostate cancer diagnosis are made before age 45, a further 9.7% before age 55, and 86% between the age of 55–84 years [19]. So while there has been an increase in younger men being diagnosed with prostate cancer, the lion’s share of new cases is still in the seventh and eight decades of life. The role of aging in causing prostate cancer is most likely through similar mechanisms common to many malignancies. We know that the process of aging itself leads to a myriad of changes in the genome including telomere shortening, epigenetic changes including methylation and demethylation, senescence, and alterations in gene expression. Why prostate cancer is so sensitive to these influences has not yet been elucidated.

    ../images/324579_1_En_2_Chapter/324579_1_En_2_Fig3_HTML.jpg

    Fig. 2.3

    Prostate cancer incidence rates per 100,000 men by age in the UK, 2009–2011. Data from the Cancer Research UK—UK National Statistics Office [18]. http://​www.​cancerresearchuk​.​org/​health-professional/​cancer-statistics/​statistics-by-cancer-type/​prostate-cancer/​incidence#heading-One

    Twin studies in monozygotic twins have demonstrated epigenetic changes occur with aging and that these are a result of both environmental changes and stochastic processes as well [20]. In addition, the multifocal nature of prostate cancer demonstrates the field effect that arises from accumulation of these changes [21]. This field effect has been termed by Damaschke et al. as Age-Related Epigenetic Alterations inducing Susceptibility (AREAS) as it is related more to the field effect generated by alterations associated with age rather than alterations associated with the presence of a neoplastic nidus [22]. Specific epigenetic changes will be discussed in subsequent chapters.

    Family History and Genetic Alterations (Inherited and Acquired)

    Suffice it to say that there is an abundance of epidemiological evidence to support both familial and genetic components to the development of prostate cancer. As early as the mid-twentieth century, familial clustering was noted and demonstrated an increased risk for the development of prostate cancer for relatives of men with prostate cancer [23]. In more contemporary studies, this risk increases with a greater number of relatives affected, the closer the degree of relation, and younger age of the relatives at diagnosis [24]. Early age of development of aggressive prostate cancer belies a genetically driven phenotype [25]. A number of prostate cancer susceptibility genes have been identified by looking at familial prostate cancers specifically. These and other genetic alterations both inherited and acquired will be discussed in much greater detail in this book.

    The technological advancements in the last decade have also allowed us to begin performing genome-wide association studies (GWAS) to identify correlation between disease and common variants in the genome [26]. Most genetic variants discovered have been modest in their effect size, most likely because GWAS have been performed in sporadic cases. The utility of GWAS may have greater implications and utilizations in family cases or specific racial groups [27]. Replications of previously identified genetic variants were highest in African American men followed by among men with a family history. The large majority of these variants are found in chromosome 8q24 [28]. Family linkage studies have further identified variants such as HOXB13 that are associated with hereditary prostate cancer [29]. Furthermore, BRCA mutations have been clearly seen to increase the risk for developing prostate cancer and more aggressive disease [30, 31]. BRCA1 mutation carriers have roughly a 3.5-fold increased risk, while men with BRCA2 have an 8.6-fold increased risk in men younger than 65 years of age [32, 33]. Moving forward we will hope to be able to identify further genetic variants that are common to the pathway that leads to progression to cancer from a preneoplastic, benign state. Furthermore, we would like to be able to do so on a more individualized level utilizing the knowledge previously gained from the study of specific high-risk groups.

    Race and Ethnicity

    Along with family history, we also inherit our forbearer ethnic and racial identity . Clearly, certain populations have a much higher risk not only of developing but also a risk of dying from prostate cancer [6]. At particular risk are men of African American and Caribbean descent. These men have been shown to present with more aggressive disease [34] and suffer the highest prostate cancer-related death rates among all ethnic groups [35]. Clearly there are other key factors besides racial and ethnic biological variability which contribute to these outcomes including factors such as disparities in access to appropriate health care [36] and greater prevalence in this population of anterior tumors that are prone to under-sampling [37]. Important to note is that there is limited evidence to suggest black men in the USA of either African or Caribbean have any significant variability from one another in terms of prevalence of significant disease (see Table 2.2) [38–40]. In comparison to previously published studies from West Africa, the rates were similar in US-born men of West African background [38, 41–43]. This raises two important points. First, black men inherently have more biologically aggressive disease regardless of their specific ancestry. Second, the demonstration that rates of advanced disease at presentation are being essentially equivalent between a screened and an unscreened population suggests a shorter lead time in black men [38, 44].

    Table 2.2

    Rates of advanceda prostate cancer among Black racial groups of varying geography and ancestry

    aDefined as Gleason >7

    Conversely men of Asian background have a relatively lower risk of developing prostate cancer as well as improved prostate cancer-specific and overall survival in several clinical settings [45, 46]. However, this has yet to be consolidated with several studies that have shown worse clinical outcomes in US Asian men [47] and the higher prevalence of higher-grade disease on autopsy in Asian men [48]. Nonetheless, consideration of a patient’s race and its implications on their biology are important and may become of greater importance as studies are beginning to now look at novel biomarkers within racial subgroups in the current age of personalized medicine.

    Modifiable

    We move on now to the modifiable drivers of prostate cancer biology, those which we can potentially manipulate and perhaps improve prognosis with. Let us begin first by examining those that center around our body’s metabolism, primarily: obesity, exercise, diet, and metabolic syndrome.

    Obesity, Exercise, Diet, and Diabetes (Metabolic)

    Body mass index (BMI), the old standard for gauging obesity, has been correlated with colon and breast cancer risk in middle- and older-age men [49]. It was suggested as a putative risk factor for prostate cancer. Wynder proposed a role for overnutrition in the development of prostate cancer in 1976 [50]. In 1984, the prospectively conducted Seventh-day Adventist study by Snowdon et al. identified a higher rate of fatal prostate cancer in men with body weights greater than 130% of ideal body weight [51]. Several important studies in the last few decades have demonstrated the clear association between obesity and increased risk and death from prostate cancer [52–54]. Furthermore, there is clear evidence demonstrating that obesity is also associated with progression of low-risk cancer in men on active surveillance [55]. Although the risk of obesity may be modest, it has been shown to be consistent [54, 56, 57]. Further confounding the issue is the lower PSA as a result of obesity may lead to a detection bias when PSA is the main driving force behind biopsy utilization [58–60]. Unlike inheritable or acquired genetic alterations, obesity more than likely drives prostate cancer risk and development via a hormonal mechanism. However, the end result on a molecular level results in a perturbation of genomic expression which leads to events such as increase tumor proliferation, reduced apoptosis, and a transition to a castrate resistant state [61]. So, if obesity promotes disease development and impacts prognosis, what can we do to alter this?

    Several studies have looked at the role of exercise and diet in reducing prostate cancer incidence and prognosis [62]. Three large prospective population studies have demonstrated a decreased risk of aggressive prostate cancer [63–65]. The underlying mechanistic effects of exercise have not been examined in great detail. Of course, it is difficult to tease out the effects of exercise from those independent of its role in reducing obesity. There is some suggestion that there may be a role of altered vascular permeability and at least temporal resolution of hypoxia in the tumor microenvironment as seen in orthotopic animal models [66]. Other effects may be more endocrine related such as affecting adipokines and the insulin-like growth factor axis [67].

    Many studies have examined the role of dietary components on prostate cancer susceptibility. Intake of red meat [68–71], green tea [72, 73], dairy products [74, 75], eggs [70, 76], selenium [77, 78], etc. has all been examined. However, results have invariably been inconsistent among the majority of studies. Debate has existed, for instance, whether it is the high levels of dietary branched fatty acids and the upregulation of α[alpha]-methyl-CoA racemase (AMACR) or whether the heterocyclic amines produced from cooking at high temperatures is the driving factor behind the association of red meat with prostate cancer [79, 80]. Meanwhile, soy products have been associated with a decreased risk owing to their phytoestrogens, either due to altering the level of circulating androgens, effects on the estrogen receptor directly, or apoptotic effects [81–83]. Micronutrients have also been examined extensively, in particular selenium and vitamin E. However, the largest randomized trial demonstrated initially no significant difference in prostate cancer development with the use of selenium, vitamin E, both, or placebo [84]. Furthermore, it was subsequently found that vitamin E was associated with an increased risk [85]. The idea of nutritional and metabolic effects and prevention strategies are not dead; however, as researchers have turned to metabolomics and the role of diet affecting these, in particular, much attention has been given to diabetic medications , especially metformin. There are currently a number of trials assessing the role of metformin in both prostate cancer prevention and prostate cancer progression in the active surveillance setting. Perhaps a better understanding of the role of the human body’s metabolism in prostate cancer will allow us to return to dietary and lifestyle modifications which may impact significantly on the disease.

    Smoking

    Smoking is a well-described risk factor for a number of malignancies including primary lung and urothelial carcinoma. Its association with prostate cancer has been under increasing scrutiny in the last decade [86]. A meta-analysis by Huncharek et al., published in 2010, examined the risk of prostate cancer from studies performed up to and including 2007 [87]. This meta-analysis demonstrated a 14% increased risk of prostate cancer death associated with current smoking and as high as 24–30% in those who were heavier smokers [87]. A more recent meta-analysis by Islami, Moreira, Boffetta, and Freedland examined prostate cancer mortality, incidence, and population attributable risk (PAR) [86]. Meta-regression analysis showed no association between smoking and prostate cancer risk (p = 0.09) and if anything perhaps a trend toward an inverse relationship. However, current (RR 1.24, 95% CI 1.18–1.31) and ever having smoked (RR 1.18, 95% CI 1.11–1.24) both showed a significant correlation with prostate cancer mortality. Furthermore, the total number of prostate cancer deaths attributable to cigarette smoking in the USA and Europe was approximately 10,400 deaths per year. In addition to higher prostate cancer mortality, smoking has been seen to be associated with more advanced disease at the time of surgery and subsequently higher risk of recurrence, metastasis, and death [88–92].

    The possible pathways by which smoking may lead to worse outcomes in prostate cancer may be due to CpG hypermethylation of several genes which in turn leads to tumor angiogenesis [93, 94]. Other putative mechanisms include increased heme oxygenase expression (HO-1), altered adhesion molecules and extracellular matrix, and smoking-induced inflammation [95–98]. Indeed smokers have a greater degree of inflammatory changes in the prostate than non-smokers [98], and inflammation may well have a role in prostate cancer progression and/or initiation [99].

    Conclusions

    Clearly there are a number of risk factors, both non-modifiable and modifiable, which have significant impact on a man’s risk of developing prostate cancer and risk of dying from prostate cancer. However, we have yet to derive a single genomic pathway which drives prostate cancer similar to the VHL gene in renal cell carcinoma. Furthermore, the exact mechanisms through which these risk factors impose an increased risk are relatively unknown, and most proposals of mechanisms of action are somewhat speculative. Perhaps this explains then to some degree the amazing degree of clinical heterogeneity we see in patients. Examination of genetic and genomic events in the modern era, however, may allow us to reverse engineer/discover what those mechanisms may be. The high-throughput processing and immense degree of information gained from molecular studies are rapidly opening up new areas of discovery and thought into prostate cancer biology. By understanding this biology better, we may hopefully treat these patients more efficiently and one day prevent these cancers from having the significant impact they have now on men’s health and lives. Once we can understand the biology, we can truly influence the modifiable risk factor component of the equation.

    References

    1.

    Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30.CrossrefPubMed

    2.

    Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29.CrossrefPubMed

    3.

    Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.CrossrefPubMed

    4.

    Potosky AL, Kessler L, Gridley G, Brown CC, Horm JW. Rise in prostatic cancer incidence associated with increased use of transurethral resection. J Natl Cancer Inst. 1990;82(20):1624–8.CrossrefPubMed

    5.

    Potosky AL, Miller BA, Albertsen PC, Kramer BS. The role of increasing detection in the rising incidence of prostate cancer. JAMA. 1995;273(7):548–52.CrossrefPubMed

    6.

    Center MM, Jemal A, Lortet-Tieulent J, Ward E, Ferlay J, Brawley O, et al. International variation in prostate cancer incidence and mortality rates. Eur Urol. 2012;61(6):1079–92.CrossrefPubMed

    7.

    Hoeksema MJ, Law C. Cancer mortality rates fall: a turning point for the nation. J Natl Cancer Inst. 1996;88(23):1706–7.CrossrefPubMed

    8.

    Coleman MP, Forman D, Bryant H, Butler J, Rachet B, Maringe C, et al. Cancer survival in Australia, Canada, Denmark, Norway, Sweden, and the UK, 1995-2007 (the International Cancer Benchmarking Partnership): an analysis of population-based cancer registry data. Lancet. 2011;377(9760):127–38.CrossrefPubMedPubMedCentral

    9.

    Wilt TJ, Brawer MK, Jones KM, Barry MJ, Aronson WJ, Fox S, et al. Radical prostatectomy versus observation for localized prostate cancer. N Engl J Med. 2012;367(3):203–13.CrossrefPubMedPubMedCentral

    10.

    Bill-Axelson A, Holmberg L, Ruutu M, Garmo H, Stark JR, Busch C, et al. Radical prostatectomy versus watchful waiting in early prostate cancer. N Engl J Med. 2011;364(18):1708–17.CrossrefPubMed

    11.

    Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351(15):1502–12.CrossrefPubMed

    12.

    de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364(21):1995–2005.CrossrefPubMedPubMedCentral

    13.

    Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367(13):1187–97.CrossrefPubMed

    14.

    Armitage P, Doll R. The age distribution of cancer and a multi-stage theory of carcinogenesis. Br J Cancer. 1954;8(1):1–12.CrossrefPubMedPubMedCentral

    15.

    Armitage P, Doll R. A two-stage theory of carcinogenesis in relation to the age distribution of human cancer. Br J Cancer. 1957;11(2):161–9.CrossrefPubMedPubMedCentral

    16.

    Sweeney C. Prostate cancer therapy: going forwards by going backwards. Lancet Oncol. 2013;14(2):104–5.CrossrefPubMed

    17.

    Sweeney CJ, Chamberlain D. Insights into E3805: the CHAARTED trial. Future Oncol. 2015;11(6):897–9.CrossrefPubMed

    18.

    Statistics OfN. Cancer incidence and mortality in the United Kingdom, 2008-10. Office for National Statistics Statistical Bulletin 2012.

    19.

    Kohler BA, Sherman RL, Howlader N, Jemal A, Ryerson AB, Henry KA, et al. Annual report to the nation on the status of cancer, 1975-2011, featuring incidence of breast cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer Inst. 2015;107(6):djv048.CrossrefPubMedPubMedCentral

    20.

    Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A. 2005;102(30):10604–9.CrossrefPubMedPubMedCentral

    21.

    Epstein JI, Carmichael MJ, Partin AW, Walsh PC. Small high grade adenocarcinoma of the prostate in radical prostatectomy specimens performed for nonpalpable disease: pathogenetic and clinical implications. J Urol. 1994;151(6):1587–92.CrossrefPubMed

    22.

    Damaschke NA, Yang B, Bhusari S, Svaren JP, Jarrard DF. Epigenetic susceptibility factors for prostate cancer with aging. Prostate. 2013;73(16):1721–30.CrossrefPubMedPubMedCentral

    23.

    Woolf CM. An investigation of the familial aspects of carcinoma of the prostate. Cancer. 1960;13:739–44.CrossrefPubMed

    24.

    Bratt O, Damber JE, Emanuelsson M, Gronberg H. Hereditary prostate cancer: clinical characteristics and survival. J Urol. 2002;167(6):2423–6.CrossrefPubMed

    25.

    Carter BS, Beaty TH, Steinberg GD, Childs B, Walsh PC. Mendelian inheritance of familial prostate cancer. Proc Natl Acad Sci U S A. 1992;89(8):3367–71.CrossrefPubMedPubMedCentral

    26.

    Ishak MB, Giri VN. A systematic review of replication studies of prostate cancer susceptibility genetic variants in high-risk men originally identified from genome-wide association studies. Cancer Epidemiol Biomarkers Prev. 2011;20(8):1599–610.CrossrefPubMed

    27.

    Stadler ZK, Vijai J, Thom P, Kirchhoff T, Hansen NAL, Kauff ND, et al. Genome-wide association studies of cancer predisposition. Hematol Oncol Clin North Am. 2010;24(5):973–96.CrossrefPubMed

    28.

    Freedman ML, Haiman CA, Patterson N, McDonald GJ, Tandon A, Waliszewska A, et al. Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proc Natl Acad Sci U S A. 2006;103(38):14068–73.CrossrefPubMedPubMedCentral

    29.

    Ewing CM, Ray AM, Lange EM, Zuhlke KA, Robbins CM, Tembe WD, et al. Germline mutations in HOXB13 and prostate-cancer risk. N Engl J Med. 2012;366(2):141–9.CrossrefPubMedPubMedCentral

    30.

    Easton DF, Schaid DJ, Whittemore AS, Isaacs WJ, International Consortium for Prostate Cancer G. Where are the prostate cancer genes?—a summary of eight genome wide searches. Prostate. 2003;57(4):261–9.CrossrefPubMed

    31.

    Ostrander EA, Kwon EM, Stanford JL. Genetic susceptibility to aggressive prostate cancer. Cancer Epidemiol Biomarkers Prev. 2006;15(10):1761–4.CrossrefPubMed

    32.

    Kote-Jarai Z, Leongamornlert D, Saunders E, Tymrakiewicz M, Castro E, Mahmud N, et al. BRCA2 is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients. Br J Cancer. 2011;105(8):1230–4.CrossrefPubMedPubMedCentral

    33.

    Leongamornlert D, Mahmud N, Tymrakiewicz M, Saunders E, Dadaev T, Castro E, et al. Germline BRCA1 mutations increase prostate cancer risk. Br J Cancer. 2012;106(10):1697–701.CrossrefPubMedPubMedCentral

    34.

    Fedewa SA, Etzioni R, Flanders WD, Jemal A, Ward EM. Association of insurance and race/ethnicity with disease severity among men diagnosed with prostate cancer, National Cancer Database 2004-2006. Cancer Epidemiol Biomarkers Prev. 2010;19(10):2437–44.CrossrefPubMed

    35.

    Edwards BK, Noone AM, Mariotto AB, Simard EP, Boscoe FP, Henley SJ, et al. Annual Report to the Nation on the status of cancer, 1975-2010, featuring prevalence of comorbidity and impact on survival among persons with lung, colorectal, breast, or prostate cancer. Cancer. 2014;120(9):1290–314.CrossrefPubMed

    36.

    Talcott JA, Spain P, Clark JA, Carpenter WR, Do YK, Hamilton RJ, et al. Hidden barriers between knowledge and behavior: the North Carolina prostate cancer screening and treatment experience. Cancer. 2007;109(8):1599–606.CrossrefPubMed

    37.

    Sundi D, Kryvenko ON, Carter HB, Ross AE, Epstein JI, Schaeffer EM. Pathological examination of radical prostatectomy specimens in men with very low risk disease at biopsy reveals distinct zonal distribution of cancer in black American men. J Urol. 2014;191(1):60–7.CrossrefPubMed

    38.

    Fedewa SA, Jemal A. Prostate cancer disease severity and country of origin among black men in the United States. Prostate Cancer Prostatic Dis. 2013;16(2):176–80.CrossrefPubMed

    39.

    Coard KC, Skeete DH. A 6-year analysis of the clinicopathological profile of patients with prostate cancer at the University Hospital of the West Indies, Jamaica. BJU Int. 2009;103(11):1482–6.CrossrefPubMed

    40.

    Kampel LJ, Tse K, Joseph M. Clinicopathologic findings in Jamaican men with prostate cancer. ASCO Meeting Abstracts. 2011;29(7_suppl):214.

    41.

    Yarney J, Vanderpuye V, Mensah J. Clinicopathologic features and determinants of Gleason score of prostate cancer in Ghanaian men. Urol Oncol. 2013;31(3):325–30.CrossrefPubMed

    42.

    Kabore FA, Zango B, Sanou A, Yameogo C, Kirakoya B. Prostate cancer outcome in Burkina Faso. Infect Agent Cancer. 2011;6 Suppl 2:S6.

    43.

    Obiorah CC, Nwosu SO. A histopathological study of carcinoma of the prostate in Port Harcourt, Nigeria. Niger J Clin Pract. 2011;14(3):363–7.CrossrefPubMed

    44.

    Thompson IM, Pauler DK, Goodman PJ, Tangen CM, Lucia MS, Parnes HL, et al. Prevalence of prostate cancer among men with a prostate-specific antigen level < or =4.0 ng per milliliter. N Engl J Med. 2004;350(22):2239–46.CrossrefPubMed

    45.

    Fukagai T, Namiki TS, Carlile RG, Yoshida H, Namiki M. Comparison of the clinical outcome after hormonal therapy for prostate cancer between Japanese and Caucasian men. BJU Int. 2006;97(6):1190–3.CrossrefPubMed

    46.

    Trinh QD, Nguyen PL, Leow JJ, Dalela D, Chao GF, Mahal BA, et al. Cancer-specific mortality of Asian Americans diagnosed with cancer: a nationwide population-based assessment. J Natl Cancer Inst. 2015;107(6):djv054.CrossrefPubMed

    47.

    Lichtensztajn DY, Gomez SL, Sieh W, Chung BI, Cheng I, Brooks JD. Prostate cancer risk profiles of Asian-American men: disentangling the effects of immigration status and race/ethnicity. J Urol. 2014;191(4):952–6.CrossrefPubMed

    48.

    Zlotta AR, Egawa S, Pushkar D, Govorov A, Kimura T, Kido M, et al. Prevalence of prostate cancer on autopsy: cross-sectional study on unscreened Caucasian and Asian men. J Natl Cancer Inst. 2013;105(14):1050–8.CrossrefPubMed

    49.

    Giovannucci E, Ascherio A, Rimm EB, Colditz GA, Stampfer MJ, Willett WC. Physical activity, obesity, and risk for colon cancer and adenoma in men. Ann Intern Med. 1995;122(5):327–34.CrossrefPubMed

    50.

    Wynder EL. Nutrition and cancer. Fed Proc. 1976;35(6):1309–15.PubMed

    51.

    Snowdon DA, Phillips RL, Choi W. Diet, obesity, and risk of fatal prostate cancer. Am J Epidemiol. 1984;120(2):244–50.CrossrefPubMed

    52.

    Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348(17):1625–38.CrossrefPubMed

    53.

    Reeves GK, Pirie K, Beral V, Green J, Spencer E, Bull D, et al. Cancer incidence and mortality in relation to body mass index in the Million Women Study: cohort study. BMJ. 2007;335(7630):1134.CrossrefPubMedPubMedCentral

    54.

    Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371(9612):569–78.CrossrefPubMed

    55.

    Bhindi B, Kulkarni GS, Finelli A, Alibhai SM, Hamilton RJ, Toi A, et al. Obesity is associated with risk of progression for low-risk prostate cancers managed expectantly. Eur Urol. 2014;66(5):841–8.CrossrefPubMed

    56.

    Bergstrom A, Pisani P, Tenet V, Wolk A, Adami HO. Overweight as an avoidable cause of cancer in Europe. Int J Cancer. 2001;91(3):421–30.CrossrefPubMed

    57.

    MacInnis RJ, English DR. Body size and composition and prostate cancer risk: systematic review and meta-regression analysis. Cancer Causes Control. 2006;17(8):989–1003.CrossrefPubMed

    58.

    Rodriguez C, Freedland SJ, Deka A, Jacobs EJ, McCullough ML, Patel AV, et al. Body mass index, weight change, and risk of prostate cancer in the Cancer Prevention Study II Nutrition Cohort. Cancer Epidemiol Biomarkers Prev. 2007;16(1):63–9.CrossrefPubMed

    59.

    Gong Z, Neuhouser ML, Goodman PJ, Albanes D, Chi C, Hsing AW, et al. Obesity, diabetes, and risk of prostate cancer: results from the prostate cancer prevention trial. Cancer Epidemiol Biomarkers Prev. 2006;15(10):1977–83.CrossrefPubMed

    60.

    Wright ME, Chang SC, Schatzkin A, Albanes D, Kipnis V, Mouw T, et al. Prospective study of adiposity and weight change in relation to prostate cancer incidence and mortality. Cancer. 2007;109(4):675–84.CrossrefPubMed

    61.

    Allott EH, Masko EM, Freedland SJ. Obesity and prostate cancer: weighing the evidence. Eur Urol. 2013;63(5):800–9.CrossrefPubMed

    62.

    Masko EM, Allott EH, Freedland SJ. The relationship between nutrition and prostate cancer: is more always better? Eur Urol. 2013;63(5):810–20.CrossrefPubMed

    63.

    Nilsen TI, Romundstad PR, Vatten LJ. Recreational physical activity and risk of prostate cancer: a prospective population-based study in Norway (the HUNT study). Int J Cancer. 2006;119(12):2943–7.CrossrefPubMed

    64.

    Johnsen NF, Tjonneland A, Thomsen BL, Christensen J, Loft S, Friedenreich C, et al. Physical activity and risk of prostate cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Int J Cancer. 2009;125(4):902–8.CrossrefPubMed

    65.

    Patel AV, Rodriguez C, Jacobs EJ, Solomon L, Thun MJ, Calle EE. Recreational physical activity and risk of prostate cancer in a large cohort of U.S. men. Cancer Epidemiol Biomarkers Prev. 2005;14(1):275–9.PubMed

    66.

    McCullough DJ, Stabley JN, Siemann DW, Behnke BJ. Modulation of blood flow, hypoxia, and vascular function in orthotopic prostate tumors during exercise. J Natl Cancer Inst. 2014;106(4):dju036.CrossrefPubMedPubMedCentral

    67.

    Mina DS, Connor MK, Alibhai SM, Toren P, Guglietti C, Matthew AG, et al. Exercise effects on adipokines and the IGF axis in men with prostate cancer treated with androgen deprivation: a randomized study. Can Urol Assoc J. 2013;7(11-12):E692–8.CrossrefPubMedCentral

    68.

    Punnen S, Hardin J, Cheng I, Klein EA, Witte JS. Impact of meat consumption, preparation, and mutagens on aggressive prostate cancer. PLoS One. 2011;6(11):e27711.CrossrefPubMedPubMedCentral

    69.

    Major JM, Cross AJ, Watters JL, Hollenbeck AR, Graubard BI, Sinha R. Patterns of meat intake and risk of prostate cancer among African-Americans in a large prospective study. Cancer Causes Control. 2011;22(12):1691–8.CrossrefPubMedPubMedCentral

    70.

    Richman EL, Kenfield SA, Stampfer MJ, Giovannucci EL, Chan JM. Egg, red meat, and poultry intake and risk of lethal prostate cancer in the prostate-specific antigen-era: incidence and survival. Cancer Prev Res. 2011;4(12):2110–21.Crossref

    71.

    Alexander DD, Morimoto LM, Mink PJ, Cushing CA. A review and meta-analysis of red and processed meat consumption and breast cancer. Nutr Res Rev. 2010;23(2):349–65.CrossrefPubMed

    72.

    Montague JA, Butler LM, Wu AH, Genkinger JM, Koh WP, Wong AS, et al. Green and black tea intake in relation to prostate cancer risk among Singapore Chinese. Cancer Causes Control. 2012;23(10):1635–41.CrossrefPubMedPubMedCentral

    73.

    Ozten-Kandas N, Bosland MC. Chemoprevention of prostate cancer: natural compounds, antiandrogens, and antioxidants—in vivo evidence. J Carcinog. 2011;10:27.CrossrefPubMedPubMedCentral

    74.

    Newmark HL, Heaney RP. Dairy products and prostate cancer risk. Nutr Cancer. 2010;62(3):297–9.CrossrefPubMed

    75.

    Lampe JW. Dairy products and cancer. J Am Coll Nutr. 2011;30(5 Suppl 1):464S–70S.CrossrefPubMed

    76.

    Richman EL, Kenfield SA, Stampfer MJ, Giovannucci EL, Zeisel SH, Willett WC, et al. Choline intake and risk of lethal prostate cancer: incidence and survival. Am J Clin Nutr. 2012;96(4):855–63.CrossrefPubMedPubMedCentral

    77.

    Sonn GA, Aronson W, Litwin MS. Impact of diet on prostate cancer: a review. Prostate Cancer Prostatic Dis. 2005;8(4):304–10.CrossrefPubMed

    78.

    Hurst R, Hooper L, Norat T, Lau R, Aune D, Greenwood DC, et al. Selenium and prostate cancer: systematic review and meta-analysis. Am J Clin Nutr. 2012;96(1):111–22.CrossrefPubMed

    79.

    Luo J, Zha S, Gage WR, Dunn TA, Hicks JL, Bennett CJ, et al. Alpha-methylacyl-CoA racemase: a new molecular marker for prostate cancer. Cancer Res. 2002;62(8):2220–6.PubMed

    80.

    Augustsson K, Skog K, Jagerstad M, Dickman PW, Steineck G. Dietary heterocyclic amines and cancer of the colon, rectum, bladder, and kidney: a population-based study. Lancet. 1999;353(9154):703–7.CrossrefPubMed

    81.

    Strom SS, Yamamura Y, Duphorne CM, Spitz MR, Babaian RJ, Pillow PC, et al. Phytoestrogen intake and prostate cancer: a case-control study using a new database. Nutr Cancer. 1999;33(1):20–5.CrossrefPubMed

    82.

    Severson RK, Nomura AM, Grove JS, Stemmermann GN. A prospective study of demographics, diet, and prostate cancer among men of Japanese ancestry in Hawaii. Cancer Res. 1989;49(7):1857–60.PubMed

    83.

    Gronberg H. Prostate cancer epidemiology. Lancet. 2003;361(9360):859–64.CrossrefPubMed

    84.

    Lippman SM, Klein EA, Goodman PJ, Lucia MS, Thompson IM, Ford LG, et al. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2009;301(1):39–51.CrossrefPubMed

    85.

    Klein EA, Thompson IM Jr, Tangen CM, Crowley JJ, Lucia MS, Goodman PJ, et al. Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2011;306(14):1549–56.CrossrefPubMedPubMedCentral

    86.

    Islami F, Moreira DM, Boffetta P, Freedland SJ. A systematic review and meta-analysis of tobacco use and prostate cancer mortality and incidence in prospective cohort studies. Eur Urol. 2014;66(6):1054–64.CrossrefPubMedPubMedCentral

    87.

    Huncharek M, Haddock KS, Reid R, Kupelnick B. Smoking as a risk factor for prostate cancer: a meta-analysis of 24 prospective cohort studies. Am J Public Health. 2010;100(4):693–701.CrossrefPubMedPubMedCentral

    88.

    Zapata DF, Howard LE, Aronson WJ, Kane CJ, Terris MK, Amling CL, et al. Smoking is a predictor of adverse pathological features at radical prostatectomy: results from the Shared Equal Access Regional Cancer Hospital database. Int J Urol. 2015;22(7):658–62.CrossrefPubMed

    89.

    Roberts WW, Platz EA, Walsh PC. Association of cigarette smoking with extraprostatic prostate cancer in young men. J Urol. 2003;169(2):512–6. discussion 6CrossrefPubMed

    90.

    Rieken M, Shariat SF, Kluth LA, Fajkovic H, Rink M, Karakiewicz PI, et al. Association of cigarette smoking and smoking cessation with biochemical recurrence of prostate cancer in patients treated with radical prostatectomy. Eur Urol. 2015;68(6):949–56.CrossrefPubMed

    91.

    Joshu CE, Mondul AM, Meinhold CL, Humphreys EB, Han M, Walsh PC, et al. Cigarette smoking and prostate cancer recurrence after prostatectomy. J Natl Cancer Inst. 2011;103(10):835–8.CrossrefPubMedPubMedCentral

    92.

    Moreira DM, Aronson WJ, Terris MK, Kane CJ, Amling CL, Cooperberg MR, et al. Cigarette smoking is associated with an increased risk of biochemical disease recurrence, metastasis, castration-resistant prostate cancer, and mortality after radical prostatectomy: results from the SEARCH database. Cancer. 2014;120(2):197–204.CrossrefPubMed

    93.

    Enokida H, Shiina H, Urakami S, Terashima M, Ogishima T, Li LC, et al. Smoking influences aberrant CpG hypermethylation of multiple genes in human prostate carcinoma. Cancer. 2006;106(1):79–86.CrossrefPubMed

    94.

    Lee J, Cooke JP. Nicotine and pathological angiogenesis. Life Sci. 2012;91(21-22):1058–64.CrossrefPubMedPubMedCentral

    95.

    Birrane G, Li H, Yang S, Tachado SD, Seng S. Cigarette smoke induces nuclear translocation of heme oxygenase 1 (HO-1) in prostate cancer cells: nuclear HO-1 promotes vascular endothelial growth factor secretion. Int J Oncol. 2013;42(6):1919–28.CrossrefPubMedPubMedCentral

    96.

    Yang S, Long M, Tachado SD, Seng S. Cigarette smoke modulates PC3 prostate cancer cell migration by altering adhesion molecules and the extracellular matrix. Mol Med Rep. 2015;12(5):6990–6.CrossrefPubMedPubMedCentral

    97.

    Arnson Y, Shoenfeld Y, Amital H. Effects of tobacco smoke on immunity, inflammation and autoimmunity. J Autoimmun. 2010;34(3):J258–65.CrossrefPubMed

    98.

    Moreira D, Nickel JC, Gerber L, Muller R, Andriole G, Castro-Santamaria R, et al. Smoking is associated with acute prostatic inflammation in men with a negative prostate biopsy: results from the REDUCE study. J Urol. 2013;189(4):E480.Crossref

    99.

    Mahmud SM, Franco EL, Aprikian AG. Use of nonsteroidal anti-inflammatory drugs and prostate cancer risk: a meta-analysis. Int J Cancer. 2010;127(7):1680–91.CrossrefPubMed

    © Springer International Publishing AG 2018

    Brian D. Robinson, Juan Miguel Mosquera, Jae Y. Ro and Mukul Divatia (eds.)Precision Molecular Pathology of Prostate CancerMolecular Pathology Libraryhttps://doi.org/10.1007/978-3-319-64096-9_3

    3. High-Grade Prostatic Intraepithelial Neoplasia

    Fadi Brimo¹  

    (1)

    Department of Pathology, McGill University Health Center, Montreal, QC, Canada

    Fadi Brimo

    Email: fadi.brimo@mcgill.ca

    Definition, Incidence and Evidence of Preneoplastic Process

    Molecular Features of HGPIN

    Clinical Significance of HGPIN

    References

    Keywords

    Prostatic intraepithelial neoplasiaTelomere shorteningCytogenetic alterationsDysregulation of annexinMammalian target of rapamycin (mTOR) pathwayMicroRNA expression profilesTMPRSS2-ERG fusion

    Definition, Incidence and Evidence of Preneoplastic Process

    Prostatic intraepithelial neoplasia [1] is defined as a noninvasive neoplastic transformation of the lining epithelium of prostatic ducts and acini [2–6]. It is the only well-established preinvasive lesion of prostatic adenocarcinoma Prostate cancer (Pca). Although PIN was originally divided into three grades (I, II and III), the poor reproducibility and lack of clinical significance of a diagnosis of PIN I have resulted in the virtual disappearance of this diagnosis from contemporary pathology reporting [7, 8]. Currently, the term PIN is used as a synonym for high-grade PIN (HGPIN) which includes PIN II and III.

    HGPIN is characterized by glands of medium to large size with an intact or fragmented basal cell layer in which a neoplastic cellular proliferation replaces the secretory epithelium. The neoplastic cells have basophilic cytoplasm, enlarged nuclei and prominent nucleoli at 200× magnification. Four main architectural patterns have been described: flat, tufting, micropapillary and cribriform. Other unusual patterns include the signet ring, small cell, mucinous, foamy and inverted patterns [4].

    The incidence of a diagnosis of HGPIN varies significantly in the literature and ranges from 0.7 to 20% in needle biopsies and 3 to 33% in transurethral resection specimens [5, 9]. Isolated HGPIN is reported in 5–10% of needle biopsies with a mean incidence of 5.2%. Such variations are due to difference in the studied population and the inconsistent application of diagnostic criteria [4].

    There is spatial, epidemiological , morphological and molecular evidence suggesting HGPIN to be the precursor of Pca. As an example, autopsy series report the incidence and extent of HGPIN to increase with age and to predate the onset of carcinoma by more than 5 years [10, 11]. The severity, multicentricity and frequency of HGPIN in prostates with cancer are also greatly increased compared with that of prostates without cancer [4, 11]. Also, similar to Pca, HGPIN preferentially involves the peripheral rather than the transition zone and occurs at a higher prevalence in African Americans compared to other races, with the lowest incidence in Asian population [10–13]. The finding of foci of HGPIN from which budding-off of rare invasive carcinoma glands occurs is further histological evidence that HGPIN is a true precursor of cancer. Such foci, referred to in the literature as HGPIN with microinvasive carcinoma, are present in 2% of high-power microscopic field of PIN and are seen in equal frequency in all architectural patterns [4, 5]. Further evidence suggesting a strong relation between HGPIN and adenocarcinoma is the fact that both lesions share similar molecular anomalies. Those molecular features are highlighted below.

    Molecular Features of HGPIN

    Telomere Shortening

    It has been shown that telomere shortening is an early event in prostatic neoplasia that occurs frequently in HGPIN and Pca [14]. Using FISH technique applied to 6 prostatectomies with 11 HGPIN lesions and 20 needle biopsies with HGPIN without cancer, Meeker et al. [15] confirmed the presence of a significant telomere shortening

    Enjoying the preview?
    Page 1 of 1