1
|
Berliner JL, Cummings SA, Boldt Burnett B, Ricker CN. Risk assessment and genetic counseling for hereditary breast and ovarian cancer syndromes-Practice resource of the National Society of Genetic Counselors. J Genet Couns 2021; 30:342-360. [PMID: 33410258 DOI: 10.1002/jgc4.1374] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022]
Abstract
Cancer risk assessment and genetic counseling for hereditary breast and ovarian cancer (HBOC) are a communication process to inform and prepare patients for genetic test results and the related medical management. An increasing number of healthcare providers are active in the delivery of cancer risk assessment and testing, which can have enormous benefits for enhanced patient care. However, genetics professionals remain key in the multidisciplinary care of at-risk patients and their families, given their training in facilitating patients' understanding of the role of genetics in cancer development, the potential psychological, social, and medical implications associated with cancer risk assessment and genetic testing. A collaborative partnership of non-genetics and genetics experts is the ideal approach to address the growing number of patients at risk for hereditary breast and ovarian cancer. The goal of this practice resource is to provide allied health professionals an understanding of the key components of risk assessment for HBOC as well as the use of risk models and published guidelines for medical management. We also highlight what patient types are appropriate for genetic testing, what are the most appropriate test(s) to consider, and when to refer individuals to a genetics professional. This practice resource is intended to serve as a resource for allied health professionals in determining their approach to delivering comprehensive care for families and individuals facing HBOC. The cancer risk and prevalence figures in this document are based on cisgender women and men; the risks for transgender or non-binary individuals have not been studied and therefore remain poorly understood.
Collapse
Affiliation(s)
- Janice L Berliner
- Genetic Counseling Department, Bay Path University, East Longmeadow, MA, USA
| | | | | | - Charité N Ricker
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Nevler A, Muller AJ, Cozzitorto JA, Goetz A, Winter JM, Yeo TP, Lavu H, Yeo CJ, Prendergast GC, Brody JR. A Sub-Type of Familial Pancreatic Cancer: Evidence and Implications of Loss-of-Function Polymorphisms in Indoleamine-2,3-Dioxygenase-2. J Am Coll Surg 2018; 226:596-603. [PMID: 29426021 PMCID: PMC6047862 DOI: 10.1016/j.jamcollsurg.2017.12.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Variation in an individual’s genetic status can impact the development of pancreatic ductal adenocarcinoma; however, the m ajority of familial pancreatic cancers (FPC) cannot yet be attributed to a specific inherited mutation. We present data suggesting a correlation between loss-of-function single nucleotide polymorphisms (SNPs) in an immune regulator gene, indoleamine-2,3-dioxygenase-2 (IDO2), and an increased risk of FPC. STUDY DESIGN Germline DNA from patients who underwent resection for pancreatic ductal adenocarcinoma (n = 79) was sequenced for the IDO2 SNPs R248W and Y359Stop. Genotypes resulting in inactivation of IDO2 (Y325X homozygous, R248W homozygous) were labeled as homozygous, and the other genotypes were grouped as wild-type or heterozygous. Genotype distributions of each SNP were analyzed for Hardy-Weinberg deviation. A genotype frequency set from the 1000 Genomes Project (n = 99) was used as a genetic control for genotype distribution comparisons. RESULTS A significant 2-fold increase in the overall prevalence of the Y359Stop homozygous genotype compared with the expected Hardy-Weinberg equilibrium was noted (p < 0.05). Familial pancreatic cancer was noted in 15 cases (19%) and comparison of the FPC cohort set to the genetic control set showed a 3-fold increase in Y359Stop homozygous rates (p = 0.054). Overall in our cohort, the homozygous genotype group was associated with increased risk of FPC (odds ratio 5.4; 95% CI 1.6 to 17.6; p < 0.01). Sex, age at diagnosis, and history of tobacco use were not found to be significantly associated with FPC. CONCLUSIONS Our preliminary data suggest a strong association between the IDO2 inactivating Y359Stop SNP and an increased risk of FPC when compared with the control group. Future studies will evaluate the value of IDO2 genotyping as a prognostic, early detection marker for pancreatic ductal adenocarcinoma and a predictive marker for novel immune checkpoint therapies.
Collapse
Affiliation(s)
- Avinoam Nevler
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA
| | - Alexander J Muller
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA; Sidney Kimmel Medical College and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA; Lankenau Institute for Medical Research, Wynnewood, PA
| | - Joseph A Cozzitorto
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA
| | - Austin Goetz
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA
| | - Jordan M Winter
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA; Sidney Kimmel Medical College and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Theresa P Yeo
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA; Sidney Kimmel Medical College and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Harish Lavu
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA; Sidney Kimmel Medical College and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Charles J Yeo
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA; Sidney Kimmel Medical College and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - George C Prendergast
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA; Sidney Kimmel Medical College and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA; Lankenau Institute for Medical Research, Wynnewood, PA
| | - Jonathan R Brody
- Jefferson Pancreas, Biliary and Related Cancer Center and Department of Surgery, Thomas Jefferson University, Philadelphia, PA; Sidney Kimmel Medical College and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA.
| |
Collapse
|
3
|
Lee MV, Katabathina VS, Bowerson ML, Mityul MI, Shetty AS, Elsayes KM, Balachandran A, Bhosale PR, McCullough AE, Menias CO. BRCA-associated Cancers: Role of Imaging in Screening, Diagnosis, and Management. Radiographics 2017; 37:1005-1023. [PMID: 28548905 DOI: 10.1148/rg.2017160144] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Harmful mutations of the BRCA tumor suppressor genes result in a greater lifetime risk for malignancy-breast and ovarian cancers in particular. An increased risk for male breast, fallopian tube, primary peritoneal, pancreatic, prostate, and colon cancers also has been reported. The BRCA gene is inherited in an autosomal dominant pattern and tends to be highly penetrant; thus, there is an increased incidence of these cancers in affected families. Compared with sporadic tumors, BRCA-associated malignancies have unique manifestations, clinical features, and pathologic profiles. Manifestation at an early patient age, high-grade tumors, and an aggressive clinical course are common features of BRCA-associated malignancies. Understanding the behavior of these cancers aids in identification of affected individuals and families, who can then make informed decisions regarding their future health. Enhanced screening, prophylactic surgery, and chemoprevention are options for managing cancer risk factors in these individuals. Imaging has an important role in the screening, evaluation, staging, and follow-up of BRCA-associated malignancies. Supplemental screening of BRCA mutation carriers often begins at an early age and is critical for early and accurate cancer diagnoses. The authors review the etiopathogenesis and imaging features of BRCA-associated malignancies, the importance of a multidisciplinary approach to determining the diagnosis, and the treatment of patients who have these mutations to improve their outcomes. © RSNA, 2017.
Collapse
Affiliation(s)
- Michelle V Lee
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Venkata S Katabathina
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Michyla L Bowerson
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Marina I Mityul
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Anup S Shetty
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Khaled M Elsayes
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Aparna Balachandran
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Priya R Bhosale
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Ann E McCullough
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| | - Christine O Menias
- From the Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St Louis, MO 63110 (M.V.L., M.I.M., A.S.S.); Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, Tex (V.S.K.); Department of Radiology, Brigham and Women's Hospital, Boston, Mass (M.L.B.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (K.M.E., A.B., P.R.B.); and Departments of Pathology (A.E.M.) and Radiology (C.O.M.), Mayo Clinic, Scottsdale, Ariz
| |
Collapse
|
4
|
Graffeo R, Livraghi L, Pagani O, Goldhirsch A, Partridge AH, Garber JE. Time to incorporate germline multigene panel testing into breast and ovarian cancer patient care. Breast Cancer Res Treat 2016; 160:393-410. [DOI: 10.1007/s10549-016-4003-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023]
|
5
|
Norris AL, Roberts NJ, Jones S, Wheelan SJ, Papadopoulos N, Vogelstein B, Kinzler KW, Hruban RH, Klein AP, Eshleman JR. Familial and sporadic pancreatic cancer share the same molecular pathogenesis. Fam Cancer 2015; 14:95-103. [PMID: 25240578 DOI: 10.1007/s10689-014-9755-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is nearly uniformly lethal, with a median overall survival in 2014 of only 6 months. The genetic progression of sporadic PDAC (SPC) is well established, with common somatic alterations in KRAS, p16/CDKN2A, TP53, and SMAD4/DPC4. Up to 10 % of all PDAC cases occur in families with two or more affected first-degree relatives (familial pancreatic cancer, FPC), but these cases do not appear to present at an obviously earlier age of onset. This is unusual because most familial cancer syndrome patients present at a substantially younger age than that of corresponding sporadic cases. Here we collated the reported age of onset for FPC and SPC from the literature. We then used an integrated approach including whole exomic sequencing, whole genome sequencing, RNA sequencing, and high density SNP microarrays to study a cohort of FPC cell lines and corresponding germline samples. We show that the four major SPC driver genes are also consistently altered in FPC and that each of the four detection strategies was able to detect the mutations in these genes, with one exception. We conclude that FPC undergoes a similar somatic molecular pathogenesis as SPC, and that the same gene targets can be used for early detection and minimal residual disease testing in FPC patients.
Collapse
Affiliation(s)
- Alexis L Norris
- Department of Pathology, The Sol Goldman Center for Pancreatic Cancer Research, Johns Hopkins University School of Medicine, Room 344, Cancer Research Building-II, 1550 Orleans Street, Baltimore, MD, 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Luo G, Lu Y, Jin K, Cheng H, Guo M, Liu Z, Long J, Liu C, Ni Q, Yu X. Pancreatic cancer: BRCA mutation and personalized treatment. Expert Rev Anticancer Ther 2015; 15:1223-31. [PMID: 26402249 DOI: 10.1586/14737140.2015.1086271] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The highly heterozygous nature of pancreatic cancer is partially responsible for its therapeutic ineffectiveness and resistance. Therefore, the ability to identify subgroups of pancreatic cancer with unique biological characteristics and treatment response is urgently needed. In addition to breast and ovarian cancer, pancreatic cancer is the third most common cancer type that is related to the early onset (BRCA) gene mutation in breast cancer. Mounting evidence has demonstrated that BRCA1/2-mutant breast and ovarian cancers are highly sensitive to DNA damage-related treatment, including poly(ADP-ribose) polymerase inhibitors (PARPi) and platinum-based agents. Preliminary evidence also showed promising results for DNA damage-related treatment in BRCA1/2-mutant pancreatic cancer. Importantly, several prospective clinical trials of PARPi-based regimens are underway for BRCA1/2-mutated pancreatic cancer. Pancreatic cancer with a BRCA1/2 mutation is a small subgroup with a promising therapeutic strategy.
Collapse
|
7
|
Abstract
Cancer is caused by the accumulation of inherited and/or acquired alterations in specific genes. The recent decline in the cost of DNA sequencing has allowed tumor sequencing to be conducted on a large scale, which, in turn, has led to an unprecedented understanding of the genetic events that drive neoplasia. This understanding, when integrated with meticulous histologic analyses and with clinical findings, has direct clinical implications. The recent sequencing of all of the major types of cystic and noncystic neoplasms of the pancreas has revealed opportunities for molecular diagnoses and for personalized treatment. This review summarizes the results from these recent studies focusing on the clinical relevance of genomic data.
Collapse
|
8
|
Familial Pancreatic Cancer: Challenging Diagnostic Approach and Therapeutic Management. J Gastrointest Cancer 2014; 45:256-61. [DOI: 10.1007/s12029-014-9609-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
9
|
Abstract
Pancreatic cancer is a leading cause of cancer death, and it has the poorest prognosis of any major tumour type. Familial pancreatic cancer registries are important for investigating the genetic aetiology of this devastating disease. Using data from our familial pancreatic cancer registry and other registries, this Review discusses the usefulness of family registries in the study of pancreatic and other cancers, and also how such registries provide a unique opportunity for laboratory, population and clinical research.
Collapse
Affiliation(s)
- Alison P Klein
- Department of Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA.
| |
Collapse
|
10
|
Raymond VM, Stoffel EM. Familial gastric and pancreatic cancers: Diagnosis and screening. Am Soc Clin Oncol Educ Book 2013:0011300044. [PMID: 23714452 DOI: 10.14694/edbook_am.2013.33.e44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Screening for gastric and pancreatic cancers in asymptomatic individuals is not routinely practiced in the United States. While there is insufficient evidence that general population screening would reduce morbidity and/or mortality associated with these cancers, the utility of screening for individuals at increased risk warrants further study. Clinical challenges include identifying high risk individuals who would be most likely to benefit from screening and determining which screening modalities and intervals would be most effective.
Collapse
Affiliation(s)
- Victoria M Raymond
- From the Divisions of Gastroenterology and Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
11
|
Amino-Functionalized Silica Nanoparticles: In Vitro Evaluation for Targeted Delivery and Therapy of Pancreatic Cancer. JOURNAL OF NANOTECHNOLOGY 2013. [DOI: 10.1155/2013/768724] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We report a method of synthesis and optimization of amino-functionalized silica nanoparticles (SiNPs) and their in vitro evaluation as targeted delivery vehicles for the potential treatment of pancreatic cancer. SiNPs can efficiently encapsulate doxorubicin and can be attached to a targeting moiety such as anti-Claudin-4 (CLN4). The preferential uptake in pancreatic cancer cells, where CLN4 is overexpressed, of SiNPs when conjugated to CLN4 antibody (compared to nonconjugated SiNPs) was confirmed by confocal microscopy. SiNPs encapsulating doxorubicin had greater efficacy in MTT assays than free doxorubicin, and when conjugated to CLN4, the efficacy was dramatically increased (at 1 μM). No apparent carrier toxicity was observed when void SiNPs were used. SiNPs carrying a chemotherapeutic drug have the potential to be used as a targeted therapy for lethal cancers, such as pancreatic cancer. Also, incorporation of fluorescent probes in these SiNPs creates the possibility of their use as an imaging probe for diagnostic purposes.
Collapse
|
12
|
Abstract
Hereditary breast and ovarian cancer due to mutations in the BRCA1 and BRCA2 genes is the most common cause of hereditary forms of both breast and ovarian cancer. The overall prevalence of BRCA1/2 mutations is estimated to be from 1 in 400 to 1 in 800 with a higher prevalence in the Ashkenazi Jewish population (1 in 40). Estimates of penetrance (cancer risk) vary considerably depending on the context in which they were derived and have been shown to vary within families with the same BRCA1/2 mutation. This suggests there is no exact risk estimate that can be applied to all individuals with a BRCA1/2 mutation. The likelihood of harboring a BRCA1 or BRCA2 mutation is dependent on one's personal and/or family history of cancer and can be estimated using various mutation probability models. For those individuals who have a BRCA1 or BRCA2 mutation, several screening and primary prevention options have been suggested, including prophylactic surgery and chemoprevention. Once a BRCA1 or BRCA2 mutation has been identified in a family, testing of at-risk relatives can identify those family members who also have the familial mutation and thus need increased surveillance and early intervention when a cancer is diagnosed.
Collapse
|
13
|
Couch FJ, Wang X, Bamlet WR, de Andrade M, Petersen GM, McWilliams RR. Association of mitotic regulation pathway polymorphisms with pancreatic cancer risk and outcome. Cancer Epidemiol Biomarkers Prev 2010; 19:251-7. [PMID: 20056645 DOI: 10.1158/1055-9965.epi-09-0629] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Mitosis is a highly regulated process that serves to ensure the fidelity of cell division. The disruption of mitotic regulators leading to aneuploidy and polyploidy is commonly observed in cancer cells. Single nucleotide polymorphisms (SNP) in regulators of mitosis may promote chromosome missegregation and influence pancreatic cancer and/or survival. METHODS Thirty-four SNPs, previously associated with breast cancer risk, from 33 genes involved in the regulation of mitosis, were investigated for associations with pancreatic cancer risk in 1,143 Caucasian patients with pancreatic adenocarcinoma and 1,097 unaffected controls from the Mayo Clinic. Associations with survival from pancreatic cancer were also assessed using 1,030 pancreatic cancer cases with known outcome. RESULTS Two SNPs in the APC (rs2431238) and NIN (rs10145182) loci, of 34 examined, were significantly associated with pancreatic cancer risk (P = 0.035 and P = 0.038, respectively). Further analyses of individuals categorized by smoking and body mass index identified several SNPs displaying significant associations (P < 0.05) with pancreatic cancer risk, including APC rs2431238 in individuals with high body mass index (>/=30; P = 0.031) and NIN rs10145182 in ever smokers (P = 0.01). In addition, survival analyses detected significant associations between SNPs in EIF3S10 and overall survival (P = 0.009), SNPs from five genes and survival in resected cancer cases (P < 0.05), and SNPs from two other genes (P < 0.05) and survival of locally advanced cancer cases. CONCLUSION Common variation in genes encoding regulators of mitosis may independently influence pancreatic cancer susceptibility and survival.
Collapse
Affiliation(s)
- Fergus J Couch
- Department of Laboratory Medicine and Pathology, Stabile 2-42, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| | | | | | | | | | | |
Collapse
|
14
|
McWilliams RR, Petersen GM, Rabe KG, Holtegaard LM, Lynch PJ, Bishop MD, Highsmith WE. Cystic fibrosis transmembrane conductance regulator (CFTR) gene mutations and risk for pancreatic adenocarcinoma. Cancer 2010; 116:203-9. [PMID: 19885835 DOI: 10.1002/cncr.24697] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene are common in white persons and are associated with pancreatic disease. The purpose of this case-control study was to determine whether CFTR mutations confer a higher risk of pancreatic cancer. METHODS In a case-control study, the authors compared the rates of 39 common cystic fibrosis-associated CFTR mutations between 949 white patients with pancreatic adenocarcinoma and 13,340 white controls from a clinical laboratory database for prenatal testing for CFTR mutations. The main outcome measure was the CFTR mutation frequency in patients and controls. RESULTS Overall, 50 (5.3%) of 949 patients with pancreatic cancer carried a common CFTR mutation versus 510 (3.8%) of 13,340 controls (odds ratio [OR], 1.40; 95% confidence interval [CI], 1.04-1.89; P = .027). Among patients who were younger when their disease was diagnosed (<60 years), the carrier frequency was higher than in controls (OR, 1.82; 95% CI, 1.14-2.94; P = .011). In patient-only analyses, the presence of a mutation was associated with younger age (median 62 vs 67 years; P = .034). In subgroups, the difference was seen only among ever-smokers (60 vs 65 years, P = .028). Subsequent sequencing analysis of the CFTR gene detected 8 (16%) compound heterozygotes among the 50 patients initially detected to have 1 mutation. CONCLUSIONS Carrying a disease-associated mutation in CFTR is associated with a modest increase in risk for pancreatic cancer. Those affected appear to be diagnosed at a younger age, especially among smokers. Clinical evidence of antecedent pancreatitis was uncommon among both carriers and noncarriers of CFTR mutations.
Collapse
|
15
|
Couch FJ, Wang X, McWilliams RR, Bamlet WR, de Andrade M, Petersen GM. Association of breast cancer susceptibility variants with risk of pancreatic cancer. Cancer Epidemiol Biomarkers Prev 2009; 18:3044-8. [PMID: 19843670 DOI: 10.1158/1055-9965.epi-09-0306] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND A number of susceptibility genes are common to breast and pancreatic cancer. Recently, several breast cancer susceptibility loci have been identified through genome-wide association studies. Here we evaluated possible associations between these single nucleotide polymorphisms (SNP) and pancreatic cancer risk. METHODS Ten SNPs from FGFR2, TOX3, MAP3K1, H19, LSP1, chromosome 8q24, CASP8, and LUM were investigated for associations with pancreatic cancer risk following genotyping in 1,143 Caucasian individuals with pancreatic adenocarcinoma and 1,097 unaffected controls from a clinic-based pancreatic cancer case-control study. RESULTS CASP8 rs1045485 [odds ratio (OR), 0.78; 95% confidence interval (95% CI), 0.65-0.9; P = 0.005] and MAP3K1 rs889312 (OR, 0.85; 95% CI, 0.74-0.97; P = 0.017) showed evidence of association with risk of pancreatic cancer. The CASP8 rs1045485 association was evident in ever smokers (P = 0.002), but not in nonsmokers (P = 0.55), and the effect was strongest in heavy smokers (OR, 0.52; 95% CI, 0.29-0.93; P = 0.03). In contrast the MAP3K1 rs889312 association was only evident in nonsmokers (OR, 0.78; 95% CI, 0.64-0.95; P = 0.01). In addition, evaluation of the influence of the 10 SNPs on survival detected significant associations between outcome for locally advanced pancreatic cancer cases and both 8q rs6983561 (P = 0.045) and LUM rs2268578 (P = 0.02). CONCLUSION Association studies in a large pancreatic case-control study indicate that SNPs associated with breast cancer may also be associated with pancreatic cancer susceptibility and survival.
Collapse
Affiliation(s)
- Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Pancreatic cancer is an extraordinarily deadly disease and is responsible for over 220,000 deaths worldwide each year. One of the greatest risk factors for developing pancreatic cancer is a positive family history. Hereditary pancreatitis patients have a greatly elevated pancreatic cancer risk and individuals with cystic fibrosis may rarely develop this cancer, but often at very young ages. Various genetically linked cancer syndromes have been associated with pancreatic cancer in mutation-positive family members. Finally, familial pancreatic cancer-defined as families with two or more first-degree relatives who have pancreatic cancer but do not have a known cancer syndrome-is a known entity whose disease-causing mutation remains unidentified. This article describes research to date on hereditary pancreatic cancer, addresses how best clinicians should recognise hereditary forms of pancreatic cancer and explains the emotional burden of discovering a potentially lethal mutation. Many controversies and unanswered questions in hereditary pancreatic cancer remain.
Collapse
|
17
|
McWilliams RR, Bamlet WR, de Andrade M, Rider DN, Cunningham JM, Petersen GM. Nucleotide excision repair pathway polymorphisms and pancreatic cancer risk: evidence for role of MMS19L. Cancer Epidemiol Biomarkers Prev 2009; 18:1295-302. [PMID: 19318433 DOI: 10.1158/1055-9965.epi-08-1109] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Nucleotide excision repair is a vital response to DNA damage, including damage from tobacco exposure. Single nucleotide polymorphisms (SNP) in the nucleotide excision repair pathway may encode alterations that affect DNA repair function and therefore influence the risk of pancreatic cancer development. METHODS A clinic-based case-control study in non-Hispanic white persons compared 1,143 patients with pancreatic adenocarcinoma with 1,097 healthy controls. Twenty-seven genes directly and indirectly involved in the nucleotide excision repair pathway were identified and 236 tag-SNPs were selected from 26 of these (one had no SNPs identified). Association studies were done at the gene level by principal components analysis, whereas recursive partitioning analysis was utilized to identify potential gene-gene and gene-environment interactions within the pathway. At the individual SNP level, adjusted additive, dominant, and recessive models were investigated, and gene-environment interactions were also assessed. RESULTS Gene level analyses showed an association of the MMS19L genotype (chromosome 10q24.1) with altered pancreatic cancer risk (P = 0.023). Haplotype analysis of MMS19L also showed a significant association (P = 0.0132). Analyses of seven individual SNPs in this gene showed both protective and risk associations for minor alleles, broadly distributed across patient subgroups defined by smoking status, sex, and age. CONCLUSION In a candidate pathway SNP association study analysis, common variation in a nucleotide excision repair gene, MMS19L, was associated with the risk of pancreatic cancer.
Collapse
|
18
|
Gemmel C, Eickhoff A, Helmstädter L, Riemann JF. Pancreatic cancer screening: state of the art. Expert Rev Gastroenterol Hepatol 2009; 3:89-96. [PMID: 19210116 DOI: 10.1586/17474124.3.1.89] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer is a devastating disease with a median survival of approximately 6 months after diagnosis. Many factors are associated with a worse outcome; examples include late diagnosis, low resection rate, aggressive tumor behavior and a lack of an effective chemotherapy regimen. Owing to the low prevalence of pancreatic cancer relative to the diagnostic accuracy of present detection methods and the absence of promising treatment modalities, even in early stages, it is currently neither advisable nor cost effective to screen the general population. Efforts are focused on early screening of selected high-risk-cohorts, who account for approximately 10% of patients with pancreatic cancer. These include patients with chronic pancreatitis, individuals with a family history of pancreatic cancer, patients with hereditary pancreatitis, Peutz-Jeghers syndrome, cystic fibrosis or familial atypical multiple mole melanoma. At present, a multimodal-screening approach of endoscopic ultrasound, computed tomography and endoscopic retrograde cholangiopancreatography appears to be the most effective method to screen for pancreatic cancer in high-risk patients. Continued efforts are needed to elucidate effective testing to identify patients with nonhereditary risk factors who will benefit from screening protocols. A combined approach of serum markers, genetic markers and specific imaging studies may prove to be the future of pancreatic screening.
Collapse
Affiliation(s)
- Christian Gemmel
- Medizinische Klinik C, Klinikum der Stadt Ludwigshafen gGmbH, Bremserstrasse 79, D-67063 Ludwigshafen, Germany.
| | | | | | | |
Collapse
|
19
|
Tan AC, Fan JB, Karikari C, Bibikova M, Garcia EW, Zhou L, Barker D, Serre D, Feldmann G, Hruban RH, Klein AP, Goggins M, Couch FJ, Hudson TJ, Winslow RL, Maitra A, Chakravarti A. Allele-specific expression in the germline of patients with familial pancreatic cancer: an unbiased approach to cancer gene discovery. Cancer Biol Ther 2008; 7:135-44. [PMID: 18059179 PMCID: PMC4104667 DOI: 10.4161/cbt.7.1.5199] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Physiologic allele-specific expression (ASE) in germline tissues occurs during random X-chromosome inactivation and in genomic imprinting, wherein the two alleles of a gene in a heterozygous individual are not expressed equally. Recent studies have confirmed the existence of ASE in apparently non-imprinted autosomal genes; however, the extent of ASE in the human genome is unknown. We explored ASE in lymphoblastoid cell lines of 145 individuals using an oligonucleotide array based assay. ASE of autosomal genes was found to be a very common phenomenon in approximately 20% of heterozygotes at 78% of SNPs at 84% of the genes examined. Comparison of 100 affected individuals from familial pancreatic cancer kindreds and 45 controls revealed three types of changes in the germline: (a) loss of ASE, (b) gain of ASE, and, (c) rare instances of "extreme" (near monoallelic) ASE. The latter changes identified heterozygous deleterious mutations in a subset of these genes. Consequently, an ASE assay efficiently identifies candidate disease genes with altered germline expression properties as compared to controls, and provides insights into mechanisms that confer an inherited disease risk for pancreatic cancer.
Collapse
Affiliation(s)
- Aik Choon Tan
- The Institute for Computational Medicine; Johns Hopkins University; Baltimore, Maryland USA
| | | | | | | | | | - Lixin Zhou
- Illumina Inc.; San Diego, California USA
| | | | - David Serre
- McGill University and Genome Quebec Innovation Center; Montreal, Quebec, Canada
| | | | | | | | | | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology; Mayo Clinic College of Medicine; Rochester, Minnesota USA
| | | | - Raimond L. Winslow
- The Institute for Computational Medicine; Johns Hopkins University; Baltimore, Maryland USA
| | - Anirban Maitra
- The Institute for Computational Medicine; Johns Hopkins University; Baltimore, Maryland USA
- The Sol Goldman Pancreatic Cancer Research Center
- McKusick-Nathans Institute of Genetic Medicine; Johns Hopkins University School of Medicine; Baltimore, Maryland USA
| | - Aravinda Chakravarti
- The Institute for Computational Medicine; Johns Hopkins University; Baltimore, Maryland USA
- McKusick-Nathans Institute of Genetic Medicine; Johns Hopkins University School of Medicine; Baltimore, Maryland USA
| |
Collapse
|
20
|
Feldmann G, Beaty R, Hruban RH, Maitra A. Molecular genetics of pancreatic intraepithelial neoplasia. ACTA ACUST UNITED AC 2007; 14:224-32. [PMID: 17520196 PMCID: PMC2666331 DOI: 10.1007/s00534-006-1166-5] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Accepted: 04/11/2006] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recent evidence suggests that noninvasive precursor lesions, classified as pancreatic intraepithelial neoplasia (PanIN), can progress to invasive pancreatic cancer. This review will discuss the major genetic alterations in PanIN lesions. METHODS A comprehensive review of the literature was performed in order to find studies on the molecular profile of human PanIN lesions. In addition, recent publications on genetically engineered mouse models of preinvasive neoplasia and pancreatic cancers were reviewed. RESULTS PanINs demonstrate abnormalities at the genomic (DNA), transcriptomic (RNA), and proteomic levels, and there is a progressive accumulation of molecular alterations that accompany the histological progression from low-grade PanIN-1A to high-grade PanIN-3 lesions. Molecular changes in PanINs can be classified as "early" (KRAS2 mutations, telomere shortening, p21(WAF1/CIP1) up-regulation, etc.), "intermediate" (cyclin D1 up-regulation, expression of proliferation antigens, etc.), or "late" (BRCA2 and TP53 mutations, DPC4/SMAD4/MADH4 inactivation, etc.). All the genetic changes observed in PanINs are also found in invasive ductal adenocarcinomas, where they usually occur at a higher frequency. Genetically engineered mice expressing mutant Kras in the pancreas, with or without additional genetic alterations, provide a unique in vivo platform to study the pancreatic cancer progression model. CONCLUSIONS Molecular studies have been instrumental in establishing that PanIN lesions are the noninvasive precursors for invasive ductal adenocarcinomas. The availability of molecular date provides the basis for designing rational early detection strategies and therapeutic intervention trials before pancreatic neoplasms invade, with the intention of alleviating the dismal prognosis associated with this disease.
Collapse
Affiliation(s)
- Georg Feldmann
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Ross Bldg 632, Johns Hopkins University School of Medicine, 720 Rutland Ave., Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
21
|
Abstract
BACKGROUND Pancreatic cancer is a lethal disease, with near uniform 5-year mortality rates. The key to improving survival of pancreatic cancer rests upon early detection of this neoplasm at a resectable, and hence potentially curable, stage. METHODS We review the current state of the literature vis-à-vis the three common precursor lesions of pancreatic adenocarcinoma: pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasm, and mucinous cystic neoplasm. We also discuss two clinical scenarios of emerging importance, namely asymptomatic pancreatic cysts ('pancreatic incidentalomas') and the significance of precursor lesions in familial pancreatic cancer kindreds. RESULTS Pancreatic intraepithelial neoplasias are the microscopic precursor lesions of pancreatic adenocarcinomas, while intraductal papillary mucinous neoplasms and mucinous cystic neoplasms are macroscopic, cystic precursor lesions. All three noninvasive entities demonstrate a multistep morphologic and genetic progression that culminates in frank invasive adenocarcinoma. Despite these commonalities, each precursor lesion harbors a unique repertoire of clinicopathologic and genetic characteristics that has an impact on natural history and prognosis of these lesions. Due to improvements in radiological techniques, asymptomatic pancreatic cysts are being increasingly discovered in the general population; intraductal papillary mucinous neoplasms and mucinous cystic neoplasms are the most common underlying histology in resected incidentalomas of the pancreas. Pancreatic asymptomatic cysts present an enormous challenge in terms of accurate diagnosis and management stratification. Incorporating molecular signatures of cystic precursor lesions into the diagnostic algorithm will likely become a standard of care for asymptomatic pancreatic cysts. High-risk individuals from familial pancreatic cancer kindreds are another group of individuals where knowledge of precursor lesions has had a therapeutic impact; sensitive imaging technologies have enabled the identification and subsequent resection of pancreatic cancer precursors in these high-risk individuals, preventing the progression to invasive cancer. CONCLUSIONS Precursor lesions of pancreatic adenocarcinomas represent a unique opportunity for diagnosis and intervention for a malignancy with near uniform lethality. Further studies on these precursors will enable the development of rational early detection and therapeutic strategies in order to ameliorate pancreatic cancer survival.
Collapse
Affiliation(s)
- Mansher Singh
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | |
Collapse
|
22
|
Couch FJ, Johnson MR, Rabe KG, Brune K, de Andrade M, Goggins M, Rothenmund H, Gallinger S, Klein A, Petersen GM, Hruban RH. The prevalence of BRCA2 mutations in familial pancreatic cancer. Cancer Epidemiol Biomarkers Prev 2007; 16:342-6. [PMID: 17301269 DOI: 10.1158/1055-9965.epi-06-0783] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mutations in the BRCA2 gene have been implicated in pancreatic cancer susceptibility through studies of high-risk breast and ovarian cancer families. To determine the contribution of mutations in BRCA2 to familial pancreatic cancer, we screened affected probands from 151 high-risk families identified through pancreatic cancer clinics for germ-line BRCA2 mutations. Of these families, 118 had two or more first- and second-degree relatives with pancreatic cancer, and an additional 33 had two or more affected second-degree relatives. The average age of onset for pancreatic cancer was 62.8 years. Five BRCA2 truncating mutations were identified, three in families with two or more first- and second-degree relatives with pancreatic cancer. Three of the families with mutations had a history of breast cancer but not ovarian cancer. Four of five families with mutations were identified through probands with early-onset (<55 years) pancreatic cancer. The results of this study were combined with those from a BRCA2 mutation study of 29 other families from the same Johns Hopkins University National Familial Pancreatic Tumor Registry to estimate the frequency of BRCA2 mutations. A total of 10 carriers from 180 families were identified, suggesting that BRCA2 mutations account for 6% of moderate and high-risk pancreatic cancer families.
Collapse
Affiliation(s)
- Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
McWilliams RR, Bamlet WR, Rabe KG, Olson JE, de Andrade M, Petersen GM. Association of family history of specific cancers with a younger age of onset of pancreatic adenocarcinoma. Clin Gastroenterol Hepatol 2006; 4:1143-7. [PMID: 16861052 PMCID: PMC1899874 DOI: 10.1016/j.cgh.2006.05.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Pancreatic adenocarcinoma has been associated with several familial cancer syndromes that also predispose to other malignancies. Younger ages of onset of pancreatic cancer (PC) have been reported in families with these syndromes. METHODS Six hundred twenty-four consecutive patients (probands) from the Mayo Clinic Pancreatic Cancer Patient Registry who completed questionnaires were analyzed for family history of cancer and cigarette smoking. The ages at diagnosis of those probands who reported a family history (first- or second-degree relative) of PC, breast, ovarian, colorectal cancer, or melanoma were compared with those probands who did not. Multivariable regression analyses were performed with age at diagnosis as the primary outcome variable. RESULTS As expected, smokers had a younger median age of onset of PC than nonsmokers in dose-dependent fashion (P = .0003). After controlling for tobacco exposure and gender, those probands with a family history of breast (-3.23 years, P = .001), ovarian (-5.63 years, P = .005), colorectal (-3.19 years, P = .002) cancers, and melanoma (-5.75 years, P = .017) had a younger age of onset of PC than those who did not. Those with a family history of PC (-.61 years, P = .65) exhibited no difference. Probands reporting other cancers in relatives showed no difference (+.78 years, P = .49) in age of onset of PC. CONCLUSION A family history of cancers (breast, ovarian, colorectal, melanoma) associated with specific cancer syndromes that are known to contribute also to PC risk is associated with a younger onset of PC. A family history of PC does not appear to affect age of onset of PC.
Collapse
Affiliation(s)
- Robert R McWilliams
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 2006; 20:1218-49. [PMID: 16702400 DOI: 10.1101/gad.1415606] [Citation(s) in RCA: 846] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death in the United States with a median survival of <6 mo and a dismal 5-yr survival rate of 3%-5%. The cancer's lethal nature stems from its propensity to rapidly disseminate to the lymphatic system and distant organs. This aggressive biology and resistance to conventional and targeted therapeutic agents leads to a typical clinical presentation of incurable disease at the time of diagnosis. The well-defined serial histopathologic picture and accompanying molecular profiles of PDAC and its precursor lesions have provided the framework for emerging basic and translational research. Recent advances include insights into the cancer's cellular origins, high-resolution genomic profiles pointing to potential new therapeutic targets, and refined mouse models reflecting both the genetics and histopathologic evolution of human PDAC. This confluence of developments offers the opportunity for accelerated discovery and the future promise of improved treatment.
Collapse
Affiliation(s)
- Aram F Hezel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
25
|
Abstract
Evaluation of: Canto MI, Goggins M, Yeo CJ et al.: Screening for pancreatic neoplasia in high-risk individuals: an EUS-based approach. Clin. Gastroenterol. Hepatol. 2(7), 606-621 (2004). Endoscopic ultrasound was utilized in the screening of individuals at high risk for pancreatic cancer. Patients with abnormal endoscopic ultrasound findings were further evaluated with fine-needle biopsy, endoscopic retrograde cholangipancreatography, and computerized tomography scanning. A total of 38 patients were enrolled in this prospective screening program. A total of seven patients were identified with suspicious neoplastic lesions and underwent pancreatic resection. No significant morbidity or mortality resulted from the screening tests or subsequent intervention including surgery. Surgical pathology revealed two neoplastic masses (invasive carcinoma and intraductal pancreatic mucinous neoplasm), three pancreatic intraepithelial neoplastic lesions, and two benign lesions for a diagnostic yield of 13.1% (five of 38). The patient with invasive carcinoma remains disease free over 5 years following surgery. The results of this study suggest a reasonable approach to detecting early neoplastic lesions in asymptomatic individuals. Such early intervention efforts hold much potential in reducing the mortality of this aggressive disease. Further studies are needed to confirm the suggested clinical benefit of screening and to answer questions related to specific screening protocols.
Collapse
Affiliation(s)
- Ted A James
- Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo NY 14263, USA
| | | |
Collapse
|
26
|
Grønborg M, Kristiansen TZ, Iwahori A, Chang R, Reddy R, Sato N, Molina H, Jensen ON, Hruban RH, Goggins MG, Maitra A, Pandey A. Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach. Mol Cell Proteomics 2005; 5:157-71. [PMID: 16215274 DOI: 10.1074/mcp.m500178-mcp200] [Citation(s) in RCA: 368] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Quantitative proteomics can be used as a screening tool for identification of differentially expressed proteins as potential biomarkers for cancers. Candidate biomarkers from such studies can subsequently be tested using other techniques for use in early detection of cancers. Here we demonstrate the use of stable isotope labeling with amino acids in cell culture (SILAC) method to compare the secreted proteins (secretome) from pancreatic cancer-derived cells with that from non-neoplastic pancreatic ductal cells. We identified 145 differentially secreted proteins (>1.5-fold change), several of which were previously reported as either up-regulated (e.g. cathepsin D, macrophage colony stimulation factor, and fibronectin receptor) or down-regulated (e.g. profilin 1 and IGFBP-7) proteins in pancreatic cancer, confirming the validity of our approach. In addition, we identified several proteins that have not been correlated previously with pancreatic cancer including perlecan (HSPG2), CD9 antigen, fibronectin receptor (integrin beta1), and a novel cytokine designated as predicted osteoblast protein (FAM3C). The differential expression of a subset of these novel proteins was validated by Western blot analysis. In addition, overexpression of several proteins not described previously to be elevated in human pancreatic cancer (CD9, perlecan, SDF4, apoE, and fibronectin receptor) was confirmed by immunohistochemical labeling using pancreatic cancer tissue microarrays suggesting that these could be further pursued as potential biomarkers. Lastly the protein expression data from SILAC were compared with mRNA expression data obtained using gene expression microarrays for the two cell lines (Panc1 and human pancreatic duct epithelial), and a correlation coefficient (r) of 0.28 was obtained, confirming previously reported poor associations between RNA and protein expression studies.
Collapse
Affiliation(s)
- Mads Grønborg
- Department of Biological Chemistry, McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
McWilliams RR, Rabe KG, Olswold C, De Andrade M, Petersen GM. Risk of malignancy in first-degree relatives of patients with pancreatic carcinoma. Cancer 2005; 104:388-94. [PMID: 15912495 DOI: 10.1002/cncr.21166] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Approximately 5-10% of pancreatic carcinoma (PC) patients report a family history of the disease. In some families, mutations of tumor suppressor genes have been elucidated, but for most the causative gene remains unidentified. Counseling the families of PC patients regarding their risk of cancer remains problematic because little information is available. METHODS The authors analyzed family history questionnaires completed by 426 unselected, sequential Mayo Clinic patients with PC. The prevalence of malignancy reported among 3355 of their first-degree relatives was compared with the Surveillance, Epidemiology, and End Results Project (SEER) 9 (2000) registry. Age-adjusted and gender-adjusted standardized incidence ratios (SIRs) were generated. RESULTS Greater than 130,000 person-years at risk for cancer among the first-degree relatives were analyzed. The risk of PC was found to be increased among the first-degree relatives of patients with PC (SIR of 1.88; 95% confidence interval [95% CI], 1.27-2.68), as was the risk of liver carcinoma (SIR of 2.70; 95% CI, 1.51-4.46). Lymphoma (SIR of 0.28; 95% CI, 0.12-0.55), bladder carcinoma (SIR of 0.55; 95% CI, 0.31-0.89), breast carcinoma (SIR of 0.73; 95% CI, 0.57-0.92), lung carcinoma (SIR of 0.62; 95% CI, 0.47-0.80), and prostate carcinoma (SIR of 0.71; 95% CI, 0.54-0.92) were found to be underrepresented. When the proband was age < 60 years, the risk of PC to first-degree relatives was found to be increased further (SIR of 2.86; 95% CI, 1.15-5.89). In this subgroup, no other malignancies were found to be significantly increased, although the risks of melanoma (SIR of 1.73; 95% CI, 0.70-3.57), ovarian carcinoma (SIR of 2.20; 95% CI, 0.72-5.12), and colon carcinoma (SIR of 1.37; 95% CI, 0.80-2.19) were suggestive. CONCLUSIONS There was a nearly twofold increased risk of PC in the first-degree relatives of PC probands. This risk was found to increase nearly threefold when patients were diagnosed before age 60 years. At the current time, in the absence of a pedigree suggestive of known familial cancer syndromes, the current study results do not support targeted screening for other malignancies in the first-degree relatives of patients with sporadic PC.
Collapse
Affiliation(s)
- Robert R McWilliams
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA.
| | | | | | | | | |
Collapse
|
28
|
Martin ST, Matsubayashi H, Rogers CD, Philips J, Couch FJ, Brune K, Yeo CJ, Kern SE, Hruban RH, Goggins M. Increased prevalence of the BRCA2 polymorphic stop codon K3326X among individuals with familial pancreatic cancer. Oncogene 2005; 24:3652-6. [PMID: 15806175 DOI: 10.1038/sj.onc.1208411] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Germline BRCA2 mutations predispose to the development of pancreatic cancer. A polymorphic stop codon in the coding region of BRCA2 (K3326X) has been described, and although an initial epidemiological study suggested it was not disease causing, subsequent studies have been inconclusive. To investigate the biological significance of the K3326X polymorphism, we determined its prevalence in patients with sporadic and familial pancreatic cancer. Using a case-control design, we studied 250 patients with resected sporadic pancreatic adenocarcinomas, 144 patients with familial pancreatic adenocarcinoma, 115 spouses of patients with pancreatic cancer, and a disease control group of 135 patients without a personal history of cancer who had undergone cholecystectomy for non-neoplastic disease. The K3326X polymorphism was detected using heteroduplex analysis and DNA sequencing. The BRCA2 K3326X polymorphism was significantly more prevalent in individuals with familial pancreatic cancer: 8/144 (5.6%) vs 3/250 controls (1.2%) (odds ratio, 4.84; 95% CI, 1.27-18.55, P<0.01). One K3326X carrier with familial pancreatic cancer carried an alteration (IVS 16-2A>G) suspected to be deleterious. Excluding this case did not alter the significance of the association (OR: 4.24, P<0.01). In contrast, there was no difference in prevalence among individuals with sporadic pancreatic cancer - 7/250 (OR: 2.37, 95% CI: 0.61-9.27). The increased prevalence of the BRCA2 K3326X polymorphism in patients with familial pancreatic cancer suggests that this polymorphism is deleterious and contributes to pancreatic cancer risk.
Collapse
Affiliation(s)
- Sean T Martin
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21205-2196, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Cancer genetics is increasingly becoming integrated into the practice of modern medical oncology. The ability to distinguish a growing proportion of the 5% to 10% of all cancers that develop in individuals who have inherited a genetic mutation conferring heightened susceptibility to specific cancers may permit targeted efforts in cancer surveillance and prevention. While these individuals comprise a small proportion of the overall burden of cancer, strategies successful in reducing their remarkable cancer risks may be generalizable to the broader population. In this review, we highlight the most common hereditary cancer syndromes, most attributable to genes inherited in an autosomal dominant manner with incomplete penetrance, and a number of rare syndromes in which particular progress has been made. The prevalence, penetrance, tumor spectrum, and underlying genetic defects are discussed and summarized in a large table in which a more comprehensive enumeration of syndromes is provided.
Collapse
Affiliation(s)
- Judy E Garber
- Dana-Farber Cancer Institute, 44 Binney Street, SM 209, Boston, MA 02115, USA.
| | | |
Collapse
|
30
|
James TA, Sheldon DG, Rajput A, Kuvshinoff BW, Javle MM, Nava HR, Smith JL, Gibbs JF. Risk factors associated with earlier age of onset in familial pancreatic carcinoma. Cancer 2004; 101:2722-6. [PMID: 15534880 DOI: 10.1002/cncr.20700] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND An estimated 5-10% of all pancreatic adenocarcinomas have a hereditary association. The objective of the current study was to characterize the clinical and pathologic features of familial pancreatic carcinoma and to determine potential differences in demographics, risk factors, and outcomes between familial and sporadic pancreatic carcinoma populations. METHODS A retrospective review was performed to identify patients diagnosed with pancreatic carcinoma who had an associated familial disposition. Demographic analyses and assessment of clinical features and treatment outcomes were performed for the familial subgroup, and the results were compared with observations made in the nonfamilial, or 'sporadic', population. RESULTS Thirty of 826 patients (3.6%) had familial pancreatic carcinoma. Baseline demographics, resectability, and metastases were similar in both the familial cohort and the sporadic cohort. The mean age of onset was slightly lower in the familial cohort (57.6 years, compared with 61 years in the sporadic cohort). However, the familial population had a significantly greater proportion of patients who were diagnosed at age <50 years compared with the sporadic population (36.7% vs. 18.3%; P=0.017). A positive smoking history was more commonly associated with familial pancreatic carcinoma (87% vs. 66%; P=0.06). The overall median survival durations were 7 months and 6 months for the familial group and the sporadic group, respectively. CONCLUSIONS Patients with familial pancreatic carcinoma present at an earlier age compared with their counterparts who have nonfamilial disease. Smoking may play a significant role in the risk or promotion of pancreatic carcinoma in patients with an inherited predisposition.
Collapse
Affiliation(s)
- Ted A James
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Pancreatic cancer (PC) is the most fatal of all gastrointestinal cancers, wherein its mortality compares strikingly with its incidence. Unfortunately, 80-90% of PCs are diagnosed in the nonresectable stage. While the lifetime risk of PC in developed countries is approximately 1-3%, it is the fifth most common cause of cancer deaths among both males and females in Western countries. It occurs in excess in Jews. Approximately 5-10% of PC shows familial clustering. Examination of such familial clusters must take into consideration cancers of diverse anatomic sites, such as malignant melanoma in the familial atypical multiple melanoma (FAMMM) syndrome due to the CDKN2A (p16) germline mutation, and combinations of colorectal and endometrial carcinoma, ovarian carcinoma, and several other cancers in hereditary nonpolyposis colorectal cancer (HNPCC), which are due to mismatch repair germline mutations, the most common of which are MSH2 and MLH1 . Other hereditary disorders predisposing to PC include Peutz-Jeghers syndrome, due to the STK11 mutation, familial pancreatitis due to the cationic trypsinogen gene, site-specific familial pancreatic cancer which may be due to the 4q32-34 mutation, hereditary breast-ovarian cancer (HBOC) syndrome that is due to BRCA2 and possibly some families with HBOC that is due to BRCA1 , familial adenomatous polyposis due to the ATP gene, and ataxia telangiectasia due to the ATM germline mutation. This extant heterogeneity mandates that the physician be knowledgeable about these PC-prone syndromes which play such an important role when considering the differential diagnosis of hereditary PC. Unfortunately, there are no PC screening programs with acceptable sensitivity and specificity. However, the gold standard for screening at this time is endoscopic ultrasound. Clearly, there is a great need for the development of novel screening approaches with acceptable sensitivity and specificity. Further research is needed to elucidate those etiologic factors that contribute to the apparent excess of PC in Ashkenazi Jews. Attention should also be given to the search for mutations predisposing to PC in Jews so that opportunities to learn more about the disease's pathogenesis, as well as screening and control, may take place.
Collapse
Affiliation(s)
- Henry T Lynch
- Department of Preventive Medicine and Public Health, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68131, USA.
| | | | | | | |
Collapse
|