1
|
Luo D, Li Y, Yu X, Ji L, Gong X. Early onset pancreatic cancer: A review. Transl Oncol 2024; 52:102239. [PMID: 39672003 PMCID: PMC11699111 DOI: 10.1016/j.tranon.2024.102239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/17/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024] Open
Abstract
Early-onset pancreatic cancer (EOPC) is usually defined as patients with pancreatic cancer before the age of 50 years, which is relatively rare. However, the research on EOPC is somewhat obscure, and the specific clinical and molecular characteristics of this condition are debated. In this review, we discussed the differences between EOPC and late-onset pancreatic cancer (LOPC) or average-onset pancreatic cancer (AOPC) with a focus on clinical and molecular characteristics, survival outcomes and treatment to promote the diagnosis and treatment of EOPC.
Collapse
Affiliation(s)
- Dong Luo
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Department of General, Visceral and Transplant Surgery, European Pancreas Centre, Heidelberg. University Hospital, Heidelberg, Germany
| | - Yixiong Li
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery Ⅱ, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Liandong Ji
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| | - Xuejun Gong
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
2
|
Renaldi K, William A. The Association between Early-Onset Pancreatic Ductal Adenocarcinoma and Patients Survival: A Systematic Review and Meta-Analysis. F1000Res 2024; 13:976. [PMID: 39355802 PMCID: PMC11443187 DOI: 10.12688/f1000research.153743.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/03/2024] Open
Abstract
Background In recent years, the incidence of early-onset pancreatic cancer (EOPC) has increased. Several studies comparing the survival of patients with EOPC to those with average-onset pancreatic cancer (AOPC) have reported mixed results. We aimed, therefore, to perform a meta-analysis summarizing the current evidence. Methods We searched the MEDLINE and EMBASE databases for relevant articles published through March 2024. Articles comparing the survival of patients with EOPC - defined as pancreatic ductal adenocarcinoma (PDAC) diagnosed at ≤ 50 years of age - and AOPC were included in the present meta-analysis. The primary outcome was the pooled adjusted hazard ratio (aHR), and the risk of bias analysis was performed using the Quality in Prognostic Factor Studies tool. The meta-analysis was performed using a random-effects model. Results A total of 17 studies were eligible for the primary analysis, the results of which indicated that patients with EOPC had a longer overall survival than those with AOPC (aHR = 0.80; 95% confidence interval [CI], 0.74-0.86; P < 0.001). The rate of distant metastasis was higher in EOPC than AOPC; however, patients with EOPC also received more treatments than those with AOPC. Conclusions Patients with EOPC had a better prognosis than those with AOPC. Clinicians must ensure that patients with PDAC receive early and appropriate treatment to improve their survival.
Collapse
Affiliation(s)
- Kaka Renaldi
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine University of Indonesia/Cipto Mangunkusumo National General Hospital, Central Jakarta, Jakarta, 10430, Indonesia
| | - Andy William
- Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| |
Collapse
|
3
|
Bogdanski AM, van Hooft JE, Boekestijn B, Bonsing BA, Wasser MNJM, Klatte DCF, van Leerdam ME. Aspects and outcomes of surveillance for individuals at high-risk of pancreatic cancer. Fam Cancer 2024; 23:323-339. [PMID: 38619782 PMCID: PMC11255004 DOI: 10.1007/s10689-024-00368-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths and is associated with a poor prognosis. The majority of these cancers are detected at a late stage, contributing to the bad prognosis. This underscores the need for novel, enhanced early detection strategies to improve the outcomes. While population-based screening is not recommended due to the relatively low incidence of PDAC, surveillance is recommended for individuals at high risk for PDAC due to their increased incidence of the disease. However, the outcomes of pancreatic cancer surveillance in high-risk individuals are not sorted out yet. In this review, we will address the identification of individuals at high risk for PDAC, discuss the objectives and targets of surveillance, outline how surveillance programs are organized, summarize the outcomes of high-risk individuals undergoing pancreatic cancer surveillance, and conclude with a future perspective on pancreatic cancer surveillance and novel developments.
Collapse
Affiliation(s)
- Aleksander M Bogdanski
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N J M Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Mendis S, Lipton L, To YH, Ananda S, Michael M, McLachlan SA, Thomson B, Loveday B, Knowles B, Fox A, Nikfarjam M, Usatoff V, Shapiro J, Clarke K, Pattison S, Chee CE, Zielinski R, Wong R, Gibbs P, Lee B. Early onset pancreatic cancer-exploring contemporary treatment and outcomes using real-world data. Br J Cancer 2024; 130:1477-1484. [PMID: 38448752 PMCID: PMC11058801 DOI: 10.1038/s41416-024-02619-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Pancreatic cancer incidence is increasing in younger populations. Differences between early onset pancreatic cancer (EOPC) and later onset pancreatic cancer (LOPC), and how these should inform management warrant exploration in the contemporary setting. METHODS A prospectively collected multi-site dataset on consecutive pancreatic adenocarcinoma patients was interrogated. Patient, tumour, treatment, and outcome data were extracted for EOPC (≤50 years old) vs LOPC (>50 years old). RESULTS Of 1683 patients diagnosed between 2016 and 2022, 112 (6.7%) were EOPC. EOPC more frequently had the tail of pancreas tumours, earlier stage disease, surgical resection, and trended towards increased receipt of chemotherapy in the curative setting compared to LOPC. EOPC more frequently received 1st line chemotherapy, 2nd line chemotherapy, and chemoradiotherapy than LOPC in the palliative setting. Recurrence-free survival was improved for the tail of pancreas EOPC vs LOPC in the resected setting; overall survival was superior for EOPC compared to LOPC across the resected, locally advanced unresectable and metastatic settings. CONCLUSIONS EOPC remains a small proportion of pancreatic cancer diagnoses. The more favourable outcomes in EOPC suggest these younger patients are overall deriving benefits from increased treatment in the curative setting and increased therapy in the palliative setting.
Collapse
Affiliation(s)
- Shehara Mendis
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
| | | | - Yat Hang To
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Sumitra Ananda
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Michael Michael
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Sue-Anne McLachlan
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Benjamin Thomson
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Benjamin Loveday
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Brett Knowles
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Epworth Healthcare, Melbourne, VIC, Australia
| | - Adrian Fox
- Department of Hepatobiliary Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Mehrdad Nikfarjam
- University of Melbourne, Parkville, VIC, Australia
- Department of Hepatobiliary Surgery, Austin Health, Heidelberg, VIC, Australia
| | | | - Julia Shapiro
- Department of Medicine, Alfred Hospital, Prahran, VIC, Australia
| | - Kate Clarke
- Department of Medical Oncology, Wellington Hospital, Wellington, New Zealand
| | - Sharon Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Otago, New Zealand
| | - Cheng Ean Chee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Rob Zielinski
- Department of Medical Oncology, Orange Hospital, Orange, NSW, Australia
- Department of Medical Oncology, Dubbo Base Hospital, Dubbo, NSW, Australia
- Department of Medical Oncology, Bathurst Base Hospital, West Bathurst, NSW, Australia
| | - Rachel Wong
- Epworth Healthcare, Melbourne, VIC, Australia
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- Department of Medical Oncology, Eastern Health, Box Hill, VIC, Australia
| | - Peter Gibbs
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Belinda Lee
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Department of Medical Oncology, Northern Hospital, Epping, VIC, Australia
| |
Collapse
|
5
|
Lloyd EG, Henríquez JA, Biffi G. Modelling the micro- and macro- environment of pancreatic cancer: from patients to pre-clinical models and back. Dis Model Mech 2024; 17:dmm050624. [PMID: 38639944 PMCID: PMC11051978 DOI: 10.1242/dmm.050624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with very low survival rates. Over the past 50 years, improvements in PDAC survival have significantly lagged behind the progress made in other cancers. PDAC's dismal prognosis is due to typical late-stage diagnosis combined with lack of effective treatments and complex mechanisms of disease. We propose that improvements in survival are partly hindered by the current focus on largely modelling and targeting PDAC as one disease, despite it being heterogeneous. Implementing new disease-representative pre-clinical mouse models that capture this complexity could enable the development of transformative therapies. Specifically, these models should recapitulate human PDAC late-stage biology, heterogeneous genetics, extensive non-malignant stroma, and associated risk factors and comorbidities. In this Perspective, we focus on how pre-clinical mouse models could be improved to exemplify key features of PDAC micro- and macro- environments, which would drive clinically relevant patient stratification, tailored treatments and improved survival.
Collapse
Affiliation(s)
- Eloise G. Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
6
|
Debernardi S, Liszka L, Ntala C, Steiger K, Esposito I, Carlotti E, Baker A, McDonald S, Graham T, Dmitrovic B, Feakins RM, Crnogorac‐Jurcevic T. Molecular characteristics of early-onset pancreatic ductal adenocarcinoma. Mol Oncol 2024; 18:677-690. [PMID: 38145461 PMCID: PMC10920080 DOI: 10.1002/1878-0261.13576] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 12/26/2023] Open
Abstract
The median age of patients with pancreatic ductal adenocarcinoma (PDAC) at diagnosis is 71 years; however, around 10% present with early-onset pancreatic cancer (EOPC), i.e., before age 50. The molecular mechanisms underlying such an early onset are unknown. We assessed the role of common PDAC drivers (KRAS, TP53, CDKN2A and SMAD4) and determined their mutational status and protein expression in 90 formalin-fixed, paraffin-embedded tissues, including multiple primary and matched metastases, from 37 EOPC patients. KRAS was mutated in 88% of patients; p53 was altered in 94%, and p16 and SMAD4 were lost in 86% and 71% of patients, respectively. Meta-synthesis showed a higher rate of p53 alterations in EOPC than in late-onset PDAC (94% vs. 69%, P = 0.0009) and significantly higher loss of SMAD4 (71% vs. 44%, P = 0.0025). The majority of EOPC patients accumulated aberrations in all four drivers; in addition, high tumour heterogeneity was observed across all tissues. The cumulative effect of an exceptionally high rate of alterations in all common PDAC driver genes combined with high tumour heterogeneity suggests an important mechanism underlying the early onset of PDAC.
Collapse
Affiliation(s)
- Silvana Debernardi
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer InstituteQueen Mary University of LondonUK
| | - Lukasz Liszka
- Department of Pathomorphology and Molecular DiagnosticsMedical University of SilesiaKatowicePoland
| | | | - Katja Steiger
- Institute of Pathology, School of Medicine and HealthTechnical University of MunichGermany
| | - Irene Esposito
- Institute of PathologyHeinrich‐Heine University and University Hospital of DusseldorfGermany
| | - Emanuela Carlotti
- Centre for Tumour Biology, Barts Cancer InstituteQueen Mary University of LondonUK
| | - Ann‐Marie Baker
- Centre for Tumour Biology, Barts Cancer InstituteQueen Mary University of LondonUK
| | - Stuart McDonald
- Centre for Tumour Biology, Barts Cancer InstituteQueen Mary University of LondonUK
| | - Trevor Graham
- Centre for Tumour Biology, Barts Cancer InstituteQueen Mary University of LondonUK
| | - Branko Dmitrovic
- Department of Pathology and Forensic MedicineClinical Hospital Center OsijekCroatia
| | - Roger M. Feakins
- Department of Cellular PathologyRoyal Free London NHS Foundation TrustUK
| | - Tatjana Crnogorac‐Jurcevic
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer InstituteQueen Mary University of LondonUK
| |
Collapse
|
7
|
Chandana SR, Woods LM, Maxwell F, Gandolfo R, Bekaii-Saab T. Risk factors for early-onset pancreatic ductal adenocarcinoma: A systematic literature review. Eur J Cancer 2024; 198:113471. [PMID: 38154392 DOI: 10.1016/j.ejca.2023.113471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Emerging cancer trends suggest an increase in pancreatic cancer incidence in individuals younger than its typical age of onset, potentially reflecting changes in population exposures and lifestyles. PATIENTS AND METHODS We conducted a PRISMA-standard systematic literature review to identify non-heritable risk factors for early-onset pancreatic ductal adenocarcinoma (PDAC) (PROSPERO number: CRD42022299397). Systematic searches of MEDLINE and Embase bibliographic databases were performed (January 2022), and publications were screened against predetermined eligibility criteria; data were extracted using standardised data fields. The STROBE checklist was used to assess the completeness of reporting as a proxy for publication quality. Data were categorised by risk factor and analysed descriptively. RESULTS In total, 24 publications were included. All publications reported observational study data; thresholds for age group comparisons ranged between 40 and 65 years. Lifestyle factors investigated included smoking, alcohol consumption, obesity, physical inactivity, meat intake, socioeconomic status and geographical residence. Clinical factors investigated included pancreatitis, diabetes/insulin resistance, prior cancer and cancer stage at diagnosis, hepatitis B infection, metabolic syndrome and long-term proton pump inhibitor exposure. Publication STROBE scores were 6-21 (maximum, 22). Eight studies reported results adjusted for confounders. Potential non-heritable risk factors for early-onset PDAC that warrant further investigation included smoking, alcohol consumption, pancreatitis and hepatitis B infection. CONCLUSION Evidence for non-heritable risk factors for early-onset PDAC is heterogeneous, but four factors were identified that might aid the identification of at-risk individuals who may benefit from screening and risk reduction strategies.
Collapse
Affiliation(s)
- Sreenivasa R Chandana
- Department of Gastrointestinal Medical Oncology, The Cancer and Hematology Centers, Grand Rapids, MI, USA.
| | - Laura M Woods
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | | | | | | |
Collapse
|
8
|
Zironda A, Zhang C, Day C, McWilliams RR, Starlinger P, Warner SG, Smoot RL, Cleary SP, Kendrick ML, Truty MJ, Thiels CA. Early vs conventional onset pancreatic ductal adenocarcinoma: analysis of surgical and oncologic outcomes in patients undergoing curative intent resection. HPB (Oxford) 2024; 26:145-153. [PMID: 37752029 DOI: 10.1016/j.hpb.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/21/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) impacts patients in their 60s, but its incidence in younger patients is increasing. We hypothesize that younger patients may have worse oncologic outcomes. METHODS Patients who underwent curative pancreatic resection for PDAC between January 2011 and December 2021 at a single institution were analyzed. Early-onset pancreatic cancer (EOPC) was defined as pancreatic cancer diagnosed in patients ≤50 years. Clinical and survival outcomes were compared between EOPC and Conventional Onset Pancreas Cancer (COPC). RESULTS A total of 1133 patients were identified, 65 (5.7%) were EOPC. Preoperative patient characteristics including sex, smoking status, alcohol habitus, diabetes mellitus, CA 19-9, and neoadjuvant therapy were similar between EOPC and COPC (p > 0.05). EOPC patients were more likely non-white (p = 0.03), had lower ASA scores (p = 0.02) and larger median tumor size (33 vs 28 mm, p = 0.04), but had similar pathological stages and rate of R0 resections (p > 0.05). Postoperative outcomes were similar (p > 0.05). There was no statistically significant difference in overall (HR 0.93, CI 0.64, 1.33; p = 0.68) or recurrence free (HR 1.05, CI 0.75, 1.48; p = 0.77) survival between the EOPC and COPC after adjusting for significant factors. CONCLUSION Patients with EOPC who underwent surgical resection had similar oncological outcomes compared to patients with COPC.
Collapse
Affiliation(s)
- Andrea Zironda
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Chi Zhang
- Department of Surgery, Mayo Clinic, Phoenix, AZ, USA; Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Courtney Day
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | | | - Patrick Starlinger
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Susanne G Warner
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Rory L Smoot
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Sean P Cleary
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Micheal L Kendrick
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Mark J Truty
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Cornelius A Thiels
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA.
| |
Collapse
|
9
|
Castet F, Fabregat-Franco C, Castillo G, Navarro V, Sierra A, Acosta DA, López-Valbuena D, Dienstmann R, Tabernero J, Vivancos A, Tian TV, Macarulla T. Clinical and genomic characterisation of early-onset pancreatic cancer. Eur J Cancer 2023; 194:113338. [PMID: 37793216 DOI: 10.1016/j.ejca.2023.113338] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND The incidence of early-onset pancreatic cancer (EOPC) has risen dramatically in recent years. We aimed to characterise the clinical and genomic features of EOPC and evaluate their therapeutic implications. METHODS We performed a comparative, single-centre, retrospective analysis of clinical, germline, and genomic features in EOPC (≤50 years) patients and compared them with a control group of average-onset pancreatic cancer patients (AOPC, ≥70 years). Key molecular findings were compared with an external, publicly available cohort. RESULTS We reviewed 336 patients who met all inclusion criteria (EOPC N = 139, AOPC N = 197). EOPC was associated with smoking status, lower prevalence of diabetes, better performance status, higher CA19.9 levels, and higher albumin levels at diagnosis. After adjustment for baseline covariates, we observed no differences in overall survival (OS). Age was associated with an increase in the incidence of KRASMUT both in our cohort and the validation cohort. EOPC were enriched in potentially actionable alterations according to ESCAT tiers I-IIIA when compared with AOPC in discovery and validation cohorts (19% versus 14% and 14% versus 8%, respectively). In the first-line metastatic setting, EOPC had a longer progression-free survival (hazard ratio [HR] 0.61, 95% confidence interval (CI) 0.43-0.87) and OS (HR 0.65, 95% CI 0.45-0.95), although there were no differences in response rate. After adjusting for the number of treatment lines, EOPC patients who did receive targeted therapies exhibited longer OS compared with EOPC who did not (HR 0.34, 95% CI 0.12-0.93). CONCLUSIONS EOPC patients have improved outcomes in the metastatic setting when compared to AOPC and are enriched for targetable alterations that open opportunities for precision oncology-based approaches.
Collapse
Affiliation(s)
- Florian Castet
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain; Upper Gastrointestinal Cancer Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Carles Fabregat-Franco
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain; Upper Gastrointestinal Cancer Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Gloria Castillo
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Víctor Navarro
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Alexandre Sierra
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Daniel Alejandro Acosta
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Daniel López-Valbuena
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain; Upper Gastrointestinal Cancer Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Josep Tabernero
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain; University of Vic-Central University of Catalonia, Carrer del Dr. Junyent, 1, 08500 Vic, Barcelona, Spain; International Oncology Bureau-Quiron, Plaça d'Alfonso Comín, 5, 08023 Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Tian V Tian
- Upper Gastrointestinal Cancer Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
| | - Teresa Macarulla
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain; Upper Gastrointestinal Cancer Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain; International Oncology Bureau-Quiron, Plaça d'Alfonso Comín, 5, 08023 Barcelona, Spain.
| |
Collapse
|
10
|
Topham JT, Renouf DJ, Schaeffer DF. Circulating tumor DNA: toward evolving the clinical paradigm of pancreatic ductal adenocarcinoma. Ther Adv Med Oncol 2023; 15:17588359231157651. [PMID: 36895849 PMCID: PMC9989430 DOI: 10.1177/17588359231157651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/30/2023] [Indexed: 03/06/2023] Open
Abstract
Over a decade of sequencing-based genomics research has unveiled a diverse somatic mutation landscape across patients with pancreatic ductal adenocarcinoma (PDAC), and the identification of druggable mutations has aligned with the development of novel targeted therapeutics. However, despite these advances, direct translation of years of PDAC genomics research into the clinical care of patients remains a critical and unmet need. Technologies that enabled the initial mapping of the PDAC mutation landscape, namely whole-genome and transcriptome sequencing, remain overly expensive in terms of both time and financial resources. Consequentially, dependence on these technologies to identify the relatively small subset of patients with actionable PDAC alterations has greatly impeded enrollment for clinical trials testing novel targeted therapies. Liquid biopsy tumor profiling using circulating tumor DNA (ctDNA) generates new opportunities by overcoming these challenges while further addressing issues particularly relevant to PDAC, namely, difficulty of obtaining tumor tissue via fine-needle biopsy and the need for faster turnaround time due to rapid disease progression. Meanwhile, ctDNA-based approaches for tracking disease kinetics with respect to surgical and therapeutic interventions offer a means to elevate the current clinical management of PDAC toward higher granularity and accuracy. This review provides a clinically focused summary of ctDNA advances, limitations, and opportunities in PDAC and postulates ctDNA sequencing technology as a catalyst for evolving the clinical decision-making paradigm of this disease.
Collapse
Affiliation(s)
| | - Daniel J Renouf
- Pancreas Centre BC, Vancouver, BC, Canada.,Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Division of Anatomic Pathology, Vancouver General Hospital, 910 West 10th Avenue, Vancouver, BC V5Z 1M9, Canada.,Pancreas Centre BC, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada
| |
Collapse
|
11
|
Topham JT, Tsang ES, Karasinska JM, Metcalfe A, Ali H, Kalloger SE, Csizmok V, Williamson LM, Titmuss E, Nielsen K, Negri GL, Spencer Miko SE, Jang GH, Denroche RE, Wong HL, O'Kane GM, Moore RA, Mungall AJ, Loree JM, Notta F, Wilson JM, Bathe OF, Tang PA, Goodwin R, Morin GB, Knox JJ, Gallinger S, Laskin J, Marra MA, Jones SJM, Schaeffer DF, Renouf DJ. Integrative analysis of KRAS wildtype metastatic pancreatic ductal adenocarcinoma reveals mutation and expression-based similarities to cholangiocarcinoma. Nat Commun 2022; 13:5941. [PMID: 36209277 PMCID: PMC9547977 DOI: 10.1038/s41467-022-33718-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 09/29/2022] [Indexed: 11/15/2022] Open
Abstract
Oncogenic KRAS mutations are absent in approximately 10% of patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) and may represent a subgroup of mPDAC with therapeutic options beyond standard-of-care cytotoxic chemotherapy. While distinct gene fusions have been implicated in KRAS wildtype mPDAC, information regarding other types of mutations remain limited, and gene expression patterns associated with KRAS wildtype mPDAC have not been reported. Here, we leverage sequencing data from the PanGen trial to perform comprehensive characterization of the molecular landscape of KRAS wildtype mPDAC and reveal increased frequency of chr1q amplification encompassing transcription factors PROX1 and NR5A2. By leveraging data from colorectal adenocarcinoma and cholangiocarcinoma samples, we highlight similarities between cholangiocarcinoma and KRAS wildtype mPDAC involving both mutation and expression-based signatures and validate these findings using an independent dataset. These data further establish KRAS wildtype mPDAC as a unique molecular entity, with therapeutic opportunities extending beyond gene fusion events. KRAS wildtype metastatic pancreatic ductal adenocarcinoma (mPDAC) could represent a distinct molecular entity from other PDACs. Here, the authors analyse KRAS wildtype mPDAC tumours using genomics and transcriptomics and find molecular similarities with cholangiocarcinomas.
Collapse
Affiliation(s)
| | - Erica S Tsang
- Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | | | | | - Hassan Ali
- Pancreas Centre BC, Vancouver, BC, Canada
| | - Steve E Kalloger
- Pancreas Centre BC, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada
| | - Veronika Csizmok
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Emma Titmuss
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Karina Nielsen
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Gian Luca Negri
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | | | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - Hui-Li Wong
- Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | | | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | | | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Julie M Wilson
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Oliver F Bathe
- Departments of Surgery and Oncology, Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Patricia A Tang
- Departments of Surgery and Oncology, Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Rachel Goodwin
- The Ottawa Hospital Cancer Centre, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Gregg B Morin
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer J Knox
- University Health Network, University of Toronto, Toronto, ON, Canada
| | - Steven Gallinger
- Ontario Institute for Cancer Research, Toronto, ON, Canada.,University Health Network, University of Toronto, Toronto, ON, Canada
| | - Janessa Laskin
- Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada.,Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Pancreas Centre BC, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada.,Division of Anatomic Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | - Daniel J Renouf
- Pancreas Centre BC, Vancouver, BC, Canada. .,Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada. .,Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
12
|
The CCTG PA.7 phase II trial of gemcitabine and nab-paclitaxel with or without durvalumab and tremelimumab as initial therapy in metastatic pancreatic ductal adenocarcinoma. Nat Commun 2022; 13:5020. [PMID: 36028483 PMCID: PMC9418247 DOI: 10.1038/s41467-022-32591-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/08/2022] [Indexed: 11/08/2022] Open
Abstract
Immunotherapy-based monotherapy treatment in metastatic pancreatic ductal adenocarcinoma (mPDAC) has shown limited benefit outside of the mismatch repair deficiency setting, while safety and efficacy of combining dual-checkpoint inhibitor immunotherapy with chemotherapy remains uncertain. Here, we present results from the CCTG PA.7 study (NCT02879318), a randomized phase II trial comparing gemcitabine and nab-paclitaxel with and without immune checkpoint inhibitors durvalumab and tremelimumab in 180 patients with mPDAC. The primary endpoint was overall survival. Secondary endpoints included progression-free survival and objective response rate. Results of the trial were negative as combination immunotherapy did not improve survival among the unselected patient population (p = 0.72) and toxicity was limited to elevation of lymphocytes in the combination immunotherapy group (p = 0.02). Exploratory baseline circulating tumor DNA (ctDNA) sequencing revealed increased survival for patients with KRAS wildtype tumors in both the combination immunotherapy (p = 0.001) and chemotherapy (p = 0.004) groups. These data support the utility of ctDNA analysis in PDAC and the prognostic value of ctDNA-based KRAS mutation status.
Collapse
|
13
|
Permuth JB, Powers BD, Hodul PJ. A Path Forward for Understanding and Addressing Multifaceted Pancreatic Cancer Disparities. Gastroenterology 2022; 163:51-53. [PMID: 35513007 PMCID: PMC9364138 DOI: 10.1053/j.gastro.2022.04.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 04/28/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Jennifer B Permuth
- Department of Gastrointestinal Oncology and, Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Benjamin D Powers
- Department of Gastrointestinal Oncology and, Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Pamela J Hodul
- Department of Gastrointestinal Oncology and, Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
14
|
Wei Z, Gordon P, Hao C, Huangfu J, Fan E, Zhang X, Yan H, Fan X. Aged Lens Epithelial Cells Suppress Proliferation and Epithelial–Mesenchymal Transition-Relevance for Posterior Capsule Opacification. Cells 2022; 11:cells11132001. [PMID: 35805085 PMCID: PMC9265589 DOI: 10.3390/cells11132001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 02/01/2023] Open
Abstract
Posterior capsule opacification (PCO) is a frequent complication after cataract surgery, and advanced PCO requires YAG laser (Nd: YAG) capsulotomy, which often gives rise to more complications. Lens epithelial cell (LEC) proliferation and transformation (i.e., epithelial–mesenchymal transition (EMT)) are two critical elements in PCO initiation and progression pathogenesis. While PCO marginally impacts aged cataract surgery patients, PCO incidences are exceptionally high in infants and children undergoing cataract surgery. The gene expression of lens epithelial cell aging and its role in the discrepancy of PCO prevalence between young and older people have not been fully studied. Here, we conducted a comprehensive differentially expressed gene (DEG) analysis of a cell aging model by comparing the early and late passage FHL124 lens epithelial cells (LECs). In vitro, TGFβ2, cell treatment, and in vivo mouse cataract surgical models were used to validate our findings. We found that aged LECs decelerated rates of cell proliferation accompanied by dysregulation of cellular immune response and cell stress response. Surprisingly, we found that LECs systematically downregulated epithelial–mesenchymal transition (EMT)-promoting genes. The protein expression of several EMT hallmark genes, e.g., fibronectin, αSMA, and cadherin 11, were gradually decreased during LECs aging. We then confirmed these findings in vitro and found that aged LECs markedly alleviated TGFβ2-mediated EMT. Importantly, we explicitly confirmed the in vitro findings from the in vivo mouse cataract surgery studies. We propose that both the high proliferation rate and EMT-enriched young LECs phenotypic characteristics contribute to unusually high PCO incidence in infants and children.
Collapse
Affiliation(s)
- Zongbo Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
| | - Pasley Gordon
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Caili Hao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
| | - Jingru Huangfu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
| | - Emily Fan
- Lakeside High School at Columbia County, Evans, GA 30809, USA;
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45221, USA;
| | - Hong Yan
- Xi’an Fourth Hospital, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Xingjun Fan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
- Correspondence:
| |
Collapse
|
15
|
Ulanja MB, Moody AE, Beutler BD, Antwi-Amoabeng D, Rahman GA, Alese OB. Early-onset pancreatic cancer: a review of molecular mechanisms, management, and survival. Oncotarget 2022; 13:828-841. [PMID: 35720978 PMCID: PMC9200435 DOI: 10.18632/oncotarget.28242] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Early-onset pancreatic cancer (EOPC) - defined as pancreatic cancer diagnosed before the age of 50 years - is associated with a poor prognosis as compared to later-onset pancreatic cancer (LOPC). Emerging evidence suggests that EOPC may exhibit a genetic signature and tumor biology that is distinct from that of LOPC. We review genetic mutations that are more prevalent in EOPC relative to LOPC and discuss the potential impact of these mutations on treatment and survival. MATERIALS AND METHODS Using PubMed and Medline, the following terms were searched and relevant citations assessed: "early onset pancreatic cancer," "late onset pancreatic cancer," "pancreatic cancer," "pancreatic cancer genes," and "pancreatic cancer targeted therapy." RESULTS Mutations in CDKN2, FOXC2, and SMAD4 are significantly more common in EOPC as compared to LOPC. In addition, limited data suggest that PI3KCA mutations are more frequently observed in EOPC as compared to LOPC. KRAS mutations are relatively rare in EOPC. CONCLUSIONS Genetic mutations associated with EOPC are distinct from those of LOPC. The preponderance of the evidence suggest that poor outcomes in EOPC are related both to advanced stage of presentation and unique tumor biology. The molecular and genetic features of EOPC warrant further investigation in order to optimize management.
Collapse
Affiliation(s)
- Mark B. Ulanja
- Christus Ochsner Saint Patrick Hospital, Lake Charles, LA 70601, USA
| | - Alastair E. Moody
- Department of Anesthesiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Bryce D. Beutler
- Department of Radiology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | | | - Ganiyu A. Rahman
- Department of Surgery, University of Cape Coast, School of Medical Sciences, Cape Coast, Ghana
| | - Olatunji B. Alese
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
16
|
Lin RT, Chen PL, Yang CY, Yeh CC, Lin CC, Huang WH, Chung AK, Lin JT. Risk factors related to age at diagnosis of pancreatic cancer: a retrospective cohort pilot study. BMC Gastroenterol 2022; 22:243. [PMID: 35568803 PMCID: PMC9107247 DOI: 10.1186/s12876-022-02325-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/09/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Increased pancreatic cancer incidence has been observed among younger than in older adults. This pilot study aimed to determine the feasibility of a large study that would compare the age at diagnosis of pancreatic cancer among patients with different risk factors. METHODS We compared the age at diagnosis of pancreatic cancer between groups of pancreatic cancer patients exposed and not exposed to the identified risk factors. We estimated the age at which exposure started, average exposure quantity, and total years of exposure and investigated their relationships with age at diagnosis of pancreatic cancer. RESULTS Sixteen out of 24 (67%) subjects carried known genetic factors and/or had smoking and/or drinking habits; however, an earlier age of pancreatic cancer diagnosis was not observed. Conversely, we found a significant correlation between the age at which alcohol consumption was started and the age at diagnosis of pancreatic cancer (r = 0.8124, P = 0.0043). CONCLUSIONS Our pilot study suggested that a large study following this study design is feasible and that the following should be conducted in a large study: mediation analysis for disease-related factors, advanced genomic analysis for new candidate genes, and the correlation between age of first exposure to risk factors and pancreatic cancer onset.
Collapse
Affiliation(s)
- Ro-Ting Lin
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, 406040 Taiwan
| | - Pei-Lung Chen
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, 100226 Taiwan
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100025 Taiwan
| | - Chi-Ying Yang
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung, 404332 Taiwan
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung, 404332 Taiwan
- Department of Surgery, Asia University Hospital, Taichung, 413505 Taiwan
| | - Chun-Che Lin
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung, 404332 Taiwan
| | - Wen-Hsin Huang
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung, 404332 Taiwan
| | - An-Ko Chung
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100025 Taiwan
| | - Jaw-Town Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, No. 1, Yida Rd., Yanchao Dist., Kaohsiung, 824005 Taiwan
| |
Collapse
|
17
|
Trends in the Incidence and Treatment of Early-Onset Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14020283. [PMID: 35053447 PMCID: PMC8773833 DOI: 10.3390/cancers14020283] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/24/2021] [Accepted: 12/31/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Pancreatic cancer is being diagnosed more frequently in younger individuals. However, limited insight exists into the magnitude of this increase, which subgroups are most affected, and which treatments are utilized in this population. In our study, we aimed to characterize which, and how, subgroups in the United States were affected by pancreatic cancer from 2000 to 2016. Additionally, we aimed to show which therapies were used to treat young patients with pancreatic cancer. Our findings provide valuable information regarding which subgroups face higher rates of this disease and what therapies have historically been used for treatment. Clinicians, scientists, policymakers, and the general population can use this information to develop programs to educate and identify individuals who are at risk for developing pancreatic cancer at an early age, as well as to study whether younger patients should be treated differently than older patients. Abstract Background: Early-onset pancreatic cancer (EOPC) is relatively uncommon. It is unclear if the incidence of EOPC is evolving and how these patients are treated. Methods: We conducted a retrospective, population-based study using SEER 2004–2016. We evaluated annual age-adjusted incidence rate (AAIR), stage at presentation, and race/ethnicity among 7802 patients plus treatment patterns in 7307 patients (excluding neuroendocrine tumors) younger than 50. Results: The AAIR was higher in males while the rate increased faster in females. The AAIR was highest in Non-Hispanic Black patients and increased for all races/ethnicities over time. The percentage of patients diagnosed with distant-stage disease decreased over time but increased for localized-stage disease. Hispanic patients made up a larger proportion of patients over time compared to other groups. For localized-stage disease, primary surgery alone was the most utilized modality of therapy. For regional-stage disease, chemotherapy with radiation was the most utilized modality from 2004–2010, whereas chemotherapy alone was the most utilized from 2011–2016. For distant-stage disease, chemotherapy alone was the most utilized and used increasingly over time. Patients with EOPC received radiation and chemotherapy at similar rates to, and underwent surgery more frequently, than patients 50–69. Conclusions: The AAIR of EOPC increased over time, faster so in females. Groups who experience a higher burden of pancreatic cancer, particularly African Americans, experienced a higher burden of EOPC. Treatment of localized and regional-stage disease did not follow standard treatment guidelines for pancreatic cancer. Our findings indicate that EOPC patients received more treatment than their older counterparts.
Collapse
|
18
|
Lou E. Age is in the Eye of the Beholder: Distinguishing Molecular Signatures in Early-onset Pancreatic Adenocarcinomas. Clin Cancer Res 2021; 27:8-10. [PMID: 33109735 DOI: 10.1158/1078-0432.ccr-20-3683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/15/2020] [Accepted: 10/23/2020] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is more prevalent in older patients, but early-onset cases (<55 years) may be a distinct genetic subpopulation. Differential expression of CDKN2A and transcription factor FOXC2 were found in early-onset cases. This finding opens the door to investigating additional features that distinguish PDAC tumors in this age group.See related article by Tsang et al., p. 246.
Collapse
Affiliation(s)
- Emil Lou
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|