1
|
Ead AS, Wirkus J, Matsukuma K, Mackenzie GG. A high-fat diet induces changes in mesenteric adipose tissue accelerating early-stage pancreatic carcinogenesis in mice. J Nutr Biochem 2024; 131:109690. [PMID: 38876394 DOI: 10.1016/j.jnutbio.2024.109690] [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: 03/17/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
Increased adiposity is a significant risk factor for pancreatic cancer development. Multiple preclinical studies have documented that high-fat, high calorie diets, rich in omega-6 fatty acids (FA) accelerate pancreatic cancer development. However, the effect of a high-fat, low sucrose diet (HFD), on pancreatic carcinogenesis remains unclear. We evaluated the impact of a HFD on early-stage pancreatic carcinogenesis in the clinically relevant KrasLSL-G12D/+; Ptf1aCre/+ (KC) genetically engineered mouse model, and characterized the role of the mesenteric adipose tissue (MAT). Cohorts of male and female KC mice were randomly assigned to a control diet (CD) or a HFD, matched for FA composition (9:1 of omega-6 FA: omega-3 FA), and fed their diets for 8 weeks. After 8 weeks on a HFD, KC mice had significantly higher body weight, fat mass, and serum leptin compared to CD-fed KC mice. Furthermore, a HFD accelerated pancreatic acinar-to-ductal metaplasia (ADM) and proliferation, associated with increased activation of ERK and STAT3, and macrophage infiltration in the pancreas, compared to CD-fed KC mice. Metabolomics analysis of the MAT revealed sex differences between diet groups. In females, a HFD altered metabolites related to FA (α-linolenic acid and linoleic acid) and amino acid metabolism (alanine, aspartate, glutamate). In males, a HFD significantly affected pathways related to alanine, aspartate, glutamate, linoleic acid, and the citric acid cycle. A HFD accelerates early pancreatic ADM through multifaceted mechanisms, including effects at the tumor and surrounding MAT. The sex-dependent changes in MAT metabolites could explain some of the sex differences in HFD-induced pancreatic ADM.
Collapse
Affiliation(s)
- Aya S Ead
- Department of Nutrition, University of California, Davis, California, USA
| | - Joanna Wirkus
- Department of Nutrition, University of California, Davis, California, USA
| | - Karen Matsukuma
- Department of Pathology and Laboratory Medicine, Davis Medical Center, University of California, Sacramento, California, USA; University of California Davis Comprehensive Cancer Center, University of California, Sacramento, California, USA
| | - Gerardo G Mackenzie
- Department of Nutrition, University of California, Davis, California, USA; University of California Davis Comprehensive Cancer Center, University of California, Sacramento, California, USA.
| |
Collapse
|
2
|
Tan JY, Yeo YH, Ng WL, Chiang CH, Stucky CC, Wasif N, Fong ZV. Pancreatic cancer mortality trends in the United States: how much have we moved the needle? J Gastrointest Oncol 2024; 15:1789-1795. [PMID: 39279936 PMCID: PMC11399823 DOI: 10.21037/jgo-24-213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/27/2024] [Indexed: 09/18/2024] Open
Abstract
Background Despite advances made in pancreatic cancer treatment, the extent of progress made in pancreatic cancer mortality at the population level remains unclear. Our cross-sectional study sought to measure trends in pancreatic cancer mortality in the United States in the last 2 decades. Methods Patients with pancreatic cancer mortality from 1999 to 2020 were analyzed from the Centers for Disease Control and Prevention's Wide-Ranging Online Data for Epidemiologic Research (CDC WONDER). Age-adjusted mortality rates (AAMRs) per 100,000 individuals were measured. We used joinpoint trend analysis to determine average annual percent change (AAPC) in AAMR trends. Results From 1999 to 2020, pancreatic cancer accounted for 809,197 deaths. Overall, the AAMRs of pancreatic cancer increased from 20.74 per 100,000 individuals in 1999 to 21.60 per 100,000 individuals in 2020. The highest AAMR was recorded in non-Hispanic Black males (30.11 per 100,000 individuals), and the lowest, in non-Hispanic White females (18.51 per 100,000 individuals). Patients aged 75-84 years had the highest AAMR (6.87 per 100,000 individuals) compared to the younger patients. The highest AAMR was observed in the Northeast region (22.07 per 100,000 individuals) and rural regions (21.29 per 100,000 individuals). Conclusions There was no improvement in pancreatic cancer mortality in the last two decades. These findings emphasize the importance of efforts to increase access to multidisciplinary cancer care with the realization that without it, improvements in treatment standards will not translate to lower cancer mortality at the population level.
Collapse
Affiliation(s)
- Jia Yi Tan
- Department of Internal Medicine, New York Medical College at Saint Michael's Medical Center, Newark, NJ, USA
| | - Yong Hao Yeo
- Department of Internal Medicine/Pediatrics, Corewell Health, Royal Oak, MI, USA
| | - Wern Lynn Ng
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC) Harrisburg, Harrisburg, PA, USA
| | - Cho Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Chee-Chee Stucky
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Nabil Wasif
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Zhi Ven Fong
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
3
|
Wang H, Shen B, Jia P, Li H, Bai X, Li Y, Xu K, Hu P, Ding L, Xu N, Xia X, Fang Y, Chen H, Zhang Y, Yue S. Guiding post-pancreaticoduodenectomy interventions for pancreatic cancer patients utilizing decision tree models. Front Oncol 2024; 14:1399297. [PMID: 38873261 PMCID: PMC11169653 DOI: 10.3389/fonc.2024.1399297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/29/2024] [Indexed: 06/15/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is frequently diagnosed in advanced stages, necessitating pancreaticoduodenectomy (PD) as a primary therapeutic approach. However, PD surgery can engender intricate complications. Thus, understanding the factors influencing postoperative complications documented in electronic medical records and their impact on survival rates is crucial for improving overall patient outcomes. Methods A total of 749 patients were divided into two groups: 598 (79.84%) chose the RPD (Robotic pancreaticoduodenectomy) procedure and 151 (20.16%) chose the LPD (Laparoscopic pancreaticoduodenectomy) procedure. We used correlation analysis, survival analysis, and decision tree models to find the similarities and differences about postoperative complications and prognostic survival. Results Pancreatic cancer, known for its aggressiveness, often requires pancreaticoduodenectomy as an effective treatment. In predictive models, both BMI and surgery duration weigh heavily. Lower BMI correlates with longer survival, while patients with heart disease and diabetes have lower survival rates. Complications like delayed gastric emptying, pancreatic fistula, and infection are closely linked post-surgery, prompting conjectures about their causal mechanisms. Interestingly, we found no significant correlation between nasogastric tube removal timing and delayed gastric emptying, suggesting its prompt removal post-decompression. Conclusion This study aimed to explore predictive factors for postoperative complications and survival in PD patients. Effective predictive models enable early identification of high-risk individuals, allowing timely interventions. Higher BMI, heart disease, or diabetes significantly reduce survival rates in pancreatic cancer patients post-PD. Additionally, there's no significant correlation between DGE incidence and postoperative extubation time, necessitating further investigation into its interaction with pancreatic fistula and infection.
Collapse
Affiliation(s)
- Haixin Wang
- Department of Cadre Medical, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Bo Shen
- Department of Respiratory and Critical Care Medicine, The Eighth Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Peiheng Jia
- Academy of Military Medical Science, Beijing, China
| | - Hao Li
- Academy of Military Medical Science, Beijing, China
| | - Xuemei Bai
- Academy of Military Medical Science, Beijing, China
| | - Yaru Li
- Academy of Military Medical Science, Beijing, China
| | - Kang Xu
- School of Software, Shandong University, Jinan, China
| | - Pengzhen Hu
- Academy of Military Medical Science, Beijing, China
- Northwestern Polytechnical University School of Life Sciences, Xi'an, China
| | - Li Ding
- Department of Cadre Medical, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Na Xu
- Department of Cadre Medical, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaoxiao Xia
- Department of Cadre Medical, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yong Fang
- College of Mathematics and Systems Science, Shandong University of Science and Technology, Qingdao, China
| | - Hebing Chen
- Academy of Military Medical Science, Beijing, China
| | - Yan Zhang
- Department of Cadre Medical, The First Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shutong Yue
- College of Mathematics and Systems Science, Shandong University of Science and Technology, Qingdao, China
| |
Collapse
|
4
|
Sato K, Hikita H, Shigekawa M, Soma K, Yamauchi R, Sung J, Kato S, Sasaki Y, Kudo S, Fukumoto K, Shirai K, Murai K, Tahata Y, Yoshioka T, Nishio A, Saito Y, Kodama T, Sasaki Y, Tatsumi T, Takehara T. The serum tenascin C level is a marker of metabolic disorder-related inflammation affecting pancreatic cancer prognosis. Sci Rep 2024; 14:12028. [PMID: 38797735 PMCID: PMC11128447 DOI: 10.1038/s41598-024-62498-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
Obesity is a risk factor for pancreatic cancer development, partly due to the tissue environment of metabolic disorder-related inflammation. We aimed to detect a tissue environment marker triggered by obesity-related metabolic disorders related to pancreatic cancer progression. In murine experiments, Bl6/j mice fed a normal diet (ND) or a high-fat diet (HFD) were orthotopically injected with mPKC1, a murine-derived pancreatic cancer cell line. We used stocked sera from 140 pancreatic cancer patients for analysis and 14 colon polyp patients as a disease control. Compared with ND-fed mice, HFD-fed mice exhibited obesity, larger tumors, and worse prognoses. RNA sequencing of tumors identified tenascin C (TNC) as a candidate obesity-related serum tissue environment marker with elevated expression in tumors of HFD-fed mice. Serum TNC levels were greater in HFD-fed mice than in ND-fed mice. In pancreatic cancer patients, serum TNC levels were greater than those in controls. The TNC-high group had more metabolic disorders and greater CA19-9 levels than did the TNC-low group. There was no relationship between serum TNC levels and disease stage. Among 77 metastatic patients treated with chemotherapy, a high serum TNC concentration was an independent poor prognostic factor. Pancreatic cancer patients with high serum TNC levels experienced progression more rapidly.
Collapse
Affiliation(s)
- Katsuhiko Sato
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Minoru Shigekawa
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Kazumasa Soma
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Ryohei Yamauchi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Jihyun Sung
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Seiya Kato
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Yoichi Sasaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Shinnosuke Kudo
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Kenji Fukumoto
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Kumiko Shirai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Kazuhiro Murai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Yuki Tahata
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Teppei Yoshioka
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Akira Nishio
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Yoshinobu Saito
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Yutaka Sasaki
- Osaka Central Hospital, 3-3-30, Umeda, Kitaku, Osaka City, Osaka, Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka, Japan.
| |
Collapse
|
5
|
Fukusada S, Shimura T, Natsume M, Nishigaki R, Okuda Y, Iwasaki H, Sugimura N, Kitagawa M, Katano T, Tanaka M, Ozeki K, Kubota E, Hayashi K, Kataoka H. Osteopontin secreted from obese adipocytes enhances angiogenesis and promotes progression of pancreatic ductal adenocarcinoma in obesity. Cell Oncol (Dordr) 2024; 47:229-244. [PMID: 37640984 DOI: 10.1007/s13402-023-00865-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
PURPOSE Obesity is a risk factor and poor prognostic factor for pancreatic ductal adenocarcinoma (PDAC), but the underlying mechanisms remain unclear. METHODS PDAC cells and obese visceral adipocytes (O-Ad) derived from mice and humans were used to analyze interactions between the two cell types, and human microvascular endothelial cells were used for angiogenesis assay. A xenograft mouse model with subcutaneously injected PDAC cells was used for animal studies. The relationship between visceral fat and prognosis was analyzed using resected tissues from PDAC patients with and without obesity. RESULTS Conditioned media (CM) from O-Ad significantly increased PDAC cell growth and migration and angiogenic capacity in both human and mice cells, and blocking osteopontin (OPN) in O-Ad canceled O-Ad-induced effects in both mouse and human cells. In addition, O-Ad directly increased the migratory and tube-forming capacities of endothelial cells, while blocking OPN canceled these effects. O-Ad increased AKT phosphorylation and VEGFA expression in both PDAC and endothelial cells, and OPN inhibition in O-Ad canceled those O-Ad-induced effects. In the xenograft model, PDAC tumor volume was significantly increased in obese mice compared with lean mice, whereas blocking OPN significantly inhibited obesity-accelerated tumor growth. OPN expression in adipose tissues adjacent to human PDAC tumor was significantly higher in obese patients than in non-obese patients. In PDAC patients with obesity, high OPN expression in adipose tissues was significantly associated with poor prognosis. CONCLUSION Obese adipocytes trigger aggressive transformation in PDAC cells to induce PDAC progression and accelerate angiogenesis via OPN secretion.
Collapse
Affiliation(s)
- Shigeki Fukusada
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Makoto Natsume
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Ruriko Nishigaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yusuke Okuda
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiroyasu Iwasaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Naomi Sugimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Mika Kitagawa
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takahito Katano
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Mamoru Tanaka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Keiji Ozeki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Eiji Kubota
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Kazuki Hayashi
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| |
Collapse
|
6
|
Anstadt EJ, Carmona R, Berlin E, Yegya-Raman N, Venigalla S, Reddy V, Williams GR, Leibensperger MR, Wojcieszynski A, Baumann BC, Lee MK, Plastaras JP, Furth EE, Mell LK, Metz JM, Ben-Josef E. SMAD4 loss predicts worse overall and distant metastasis-free survival in patients with resected pancreatic adenocarcinoma. Cancer 2024; 130:476-484. [PMID: 37823514 DOI: 10.1002/cncr.35058] [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: 04/19/2023] [Revised: 07/20/2023] [Accepted: 08/08/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND In select patients, pancreatic adenocarcinoma remains a local disease, yet there are no validated biomarkers to predict this behavior and who may benefit from aggressive local treatments. This study sought to determine if SMAD4 (mothers against decapentaplegic homolog 4) messenger RNA-sequencing (RNA-seq) expression is a robust method for predicting overall survival (OS) and distant metastasis-free survival (DMFS) in patients with resected pancreatic adenocarcinoma. METHODS Utilizing The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC), 322 patients with resected stage I-III pancreatic adenocarcinoma were identified. In TCGA, multivariable proportional hazards models were used to determine the association of SMAD4 genomic aberrations and RNA-seq expression with OS and DMFS. In the ICGC, analysis sought to confirm the predictive performance of RNA-seq via multivariable models and receiver operator characteristic curves. RESULTS In TCGA, the presence of SMAD4 genomic aberrations was associated with worse OS (hazard ratio [HR], 1.55; 95% CI, 1.00-2.40; p = .048) but not DMFS (HR, 1.33; 95% CI, .87-2.03; p = .19). Low SMAD4 RNA-seq expression was associated with worse OS (HR, 1.83; 95% CI, 1.17-2.86; p = .008) and DMFS (HR, 1.70; 95% CI, 1.14-2.54; p = .009). In the ICGC, increased SMAD4 RNA-seq expression correlated with improved OS (area under the curve [AUC], .92; 95% CI, .86-.94) and DMFS (AUC, .84; 95% CI, .82-.87). CONCLUSIONS In patients with resected pancreatic adenocarcinoma, SMAD4 genomic aberrations are associated with worse OS but do not predict for DMFS. Increased SMAD4 RNA-seq expression is associated with improved OS and DMFS in patients with resected pancreatic adenocarcinoma. This reproducible finding suggests SMAD4 RNA-seq expression may be a useful marker to predict metastatic spread.
Collapse
Affiliation(s)
- Emily J Anstadt
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ruben Carmona
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eva Berlin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nikhil Yegya-Raman
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sriram Venigalla
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vishruth Reddy
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Graeme R Williams
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark R Leibensperger
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrzej Wojcieszynski
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brian C Baumann
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Major K Lee
- Division of Gastrointestinal Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John P Plastaras
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emma E Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Loren K Mell
- Department of Radiation Medicine and Applied Sciences, University of California San Diego School of Medicine, La Jolla, California, USA
| | - James M Metz
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edgar Ben-Josef
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Ruiz CF, Garcia C, Jacox JB, Lawres L, Muzumdar MD. Decoding the obesity-cancer connection: lessons from preclinical models of pancreatic adenocarcinoma. Life Sci Alliance 2023; 6:e202302228. [PMID: 37648285 PMCID: PMC10474221 DOI: 10.26508/lsa.202302228] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Obesity is a metabolic state of energy excess and a risk factor for over a dozen cancer types. Because of the rising worldwide prevalence of obesity, decoding the mechanisms by which obesity promotes tumor initiation and early progression is a societal imperative and could broadly impact human health. Here, we review results from preclinical models that link obesity to cancer, using pancreatic adenocarcinoma as a paradigmatic example. We discuss how obesity drives cancer development by reprogramming the pretumor or tumor cell and its micro- and macro-environments. Specifically, we describe evidence for (1) altered cellular metabolism, (2) hormone dysregulation, (3) inflammation, and (4) microbial dysbiosis in obesity-driven pancreatic tumorigenesis, denoting variables that confound interpretation of these studies, and highlight remaining gaps in knowledge. Recent advances in preclinical modeling and emerging unbiased analytic approaches will aid in further unraveling the complex link between obesity and cancer, informing novel strategies for prevention, interception, and therapy in pancreatic adenocarcinoma and other obesity-associated cancers.
Collapse
Affiliation(s)
- Christian F Ruiz
- https://ror.org/03v76x132 Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- https://ror.org/03v76x132 Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Cathy Garcia
- https://ror.org/03v76x132 Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- https://ror.org/03v76x132 Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Jeremy B Jacox
- https://ror.org/03v76x132 Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- https://ror.org/03v76x132 Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
- https://ror.org/03v76x132 Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
| | - Lauren Lawres
- https://ror.org/03v76x132 Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Mandar D Muzumdar
- https://ror.org/03v76x132 Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- https://ror.org/03v76x132 Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
- https://ror.org/03v76x132 Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
- https://ror.org/03v76x132 Yale Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
8
|
Yang J, Liu S, Li Y, Fan Z, Meng Y, Zhou B, Zhang G, Zhan H. FABP4 in macrophages facilitates obesity-associated pancreatic cancer progression via the NLRP3/IL-1β axis. Cancer Lett 2023; 575:216403. [PMID: 37741433 DOI: 10.1016/j.canlet.2023.216403] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/25/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Obesity is an essential risk factor for pancreatic cancer (PC). Macrophage-induced inflammation plays a pivotal role in obesity-associated carcinogenesis and disease progression; however, the underlying molecular mechanisms remain unclear. In this study, we found that fatty acid-binding protein 4 (FABP4) overexpressed in serum of obese patients and was associated with poor overall survival. In vivo and in vitro experiments have revealed that FABP4 induces macrophage-related inflammation to promote cancer cell migration, invasion and metastasis under obese conditions. Mechanistically, FABP4 participates in transferring saturated fatty acid to induce macrophages pyroptosis in a caspase-1/GSDMD-dependent manner and mediates NOD-like receptor thermal protein domain associated protein 3 (NLRP3)/IL-1β axis in macrophages, which further regulates epithelial-mesenchymal transition signals to promote the migration, invasion, and metastasis of PC cells. Our results suggest that FABP4 in macrophages is a crucial regulator of the NLRP3/IL-1β axis to promote the progression of PC under obese conditions, which could act as a promising molecular target for treating of PC patients with obesity.
Collapse
Affiliation(s)
- Jian Yang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Shujie Liu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yongzheng Li
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yufan Meng
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China.
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
9
|
Babic A, Wang QL, Lee AA, Yuan C, Rifai N, Luo J, Tabung FK, Shadyab AH, Wactawski-Wende J, Saquib N, Kim J, Kraft P, Sesso HD, Buring JE, Giovannucci EL, Manson JE, Stampfer MJ, Ng K, Fuchs CS, Wolpin BM. Sex-Specific Associations between Adiponectin and Leptin Signaling and Pancreatic Cancer Survival. Cancer Epidemiol Biomarkers Prev 2023; 32:1458-1469. [PMID: 37555827 PMCID: PMC10592159 DOI: 10.1158/1055-9965.epi-23-0505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/17/2023] [Accepted: 08/07/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Circulating adiponectin and leptin have been associated with risk of pancreatic cancer. However, the relationship between long-term exposure to these adipokines in the prediagnostic period with patient survival has not been investigated. METHODS Adipokine levels were measured in prospectively collected samples from 472 patients with pancreatic cancer. Because of sex-specific differences in adipokine levels, associations were evaluated separately for men and women. In a subset of 415 patients, we genotyped 23 SNPs in adiponectin receptor genes (ADIPOR1 and ADIPOR2) and 30 SNPs in the leptin receptor gene (LEPR). RESULTS Adiponectin levels were inversely associated with survival in women [HR, 1.71; 95% confidence interval (CI), 1.15-2.54]; comparing top with bottom quartile but not in men (HR, 0.89; 95% CI, 0.46-1.70). The SNPs rs10753929 and rs1418445 in ADIPOR1 were associated with survival in the combined population (per minor allele HR, 0.66; 95% CI, 0.51-0.84, and HR, 1.33; 95% CI, 1.12-1.58, respectively). Among SNPs in LEPR, rs12025906, rs3790431, and rs17127601 were associated with survival in the combined population [HRs, 1.54 (95% CI, 1.25-1.90), 0.72 (95% CI, 0.59-0.88), and 0.70 (95% CI, 0.56-0.89), respectively], whereas rs11585329 was associated with survival in men only (HR, 0.39; 95% CI, 0.23-0.66; Pinteraction = 0.0002). CONCLUSIONS High levels of adiponectin in the prediagnostic period were associated with shorter survival among women, but not among men with pancreatic cancer. Several polymorphisms in ADIPOR1 and LEPR are associated with patient survival. IMPACT Our findings reveal the association between adipokine signaling and pancreatic cancer survival and demonstrate the importance of examining obesity-associated pathways in relation to pancreatic cancer in a sex-specific manner.
Collapse
Affiliation(s)
- Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Qiao-Li Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Alice A. Lee
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Nader Rifai
- Department of Laboratory Medicine, Children’s Hospital Boston, Boston, MA
| | - Juhua Luo
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN
| | - Fred K. Tabung
- Department of Internal Medicine, Ohio State University, Columbus, OH
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, State University of New York, Buffalo, NY
| | - Nazmus Saquib
- College of Medicine, Sulaiman Al Rajhi University, Al Bukairiyah, Kingdom of Saudi Arabia
| | - Jihye Kim
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Howard D. Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Julie E. Buring
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Department of Ambulatory Care and Prevention, Harvard Medical School, Boston, MA
| | - Edward L. Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
| | - JoAnn E. Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
| | - Meir J. Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Charles S. Fuchs
- Hematology and Oncology Product Development, Genentech & Roche, South San Francisco, CA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| |
Collapse
|
10
|
Xu T, Xu X, Liu D, Chang D, Li S, Sun Y, Xie J, Ju S. Visual Investigation of Tumor-Promoting Fibronectin Potentiated by Obesity in Pancreatic Ductal Adenocarcinoma Using an MR/NIRF Dual-Modality Dendrimer Nanoprobe. Adv Healthc Mater 2023; 12:e2300787. [PMID: 37057680 DOI: 10.1002/adhm.202300787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease characterized by dense stroma. Obesity is an important metabolic factor that greatly increases PDAC risk and mortality, worsens progression and leads to poor chemotherapeutic outcomes. With omics analysis, magnetic resonance and near-infrared fluorescence (MR/NIRF) dual-modality imaging and molecular functional verification, obesity as an important risk factor is proved to modulate the extracellular matrix (ECM) components and enhance Fibronectin (FN) infiltration in the PDAC stroma, that promotes tumor progression and worsens response to chemotherapy by reducing drug delivery. In the study, to visually evaluate FN in vivo and guide PDAC therapy, an FN-targeted nanoprobe, NP-CREKA, is synthesized by conjugating gadolinium chelates, NIR797 and fluorescein isothiocyanate to a polyamidoamine dendrimer functionalized with targeting peptides. A dual-modality strategy combining MR and NIRF imaging is applied, allowing effective visualization of FN in orthotopic PDAC with high spatial resolution, ideal sensitivity and excellent penetrability, especially in obese mice. In conclusion, the findings provide new insights into the potential of FN as an ideal target for therapeutic evaluation and improving treatment efficacy in PDAC, hopefully improving the specific management of PDAC in lean and obese hosts.
Collapse
Affiliation(s)
- Tingting Xu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Xiaoxuan Xu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Dongfang Liu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Di Chang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Siqi Li
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Yeyao Sun
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jinbing Xie
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| |
Collapse
|
11
|
Li L, He J. Prognostic Role of Geriatric Nutritional Risk Index in Patients with Pancreatic Cancer: A Meta-Analysis. Nutr Cancer 2023:1-10. [PMID: 37162261 DOI: 10.1080/01635581.2023.2209345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background: The Geriatric Nutritional Risk Index (GNRI) is used to assess the prognosis of patients with pancreatic cancer. Therefore, this meta-analysis was conducted to evaluate the association between the GNRI and prognosis in pancreatic cancer.Methods: We performed a pooled analysis of the hazard ratios (HRs) and 95% confidence intervals (CIs) of the GNRI for survival in pancreatic cancer. Using pooled odds ratios (ORs) and 95% CIs, we investigated the association between the GNRI and clinicopathological characteristics of pancreatic cancer.Results: Six studies were included in this meta-analysis, totaling 1,513 patients. A low GNRI was significantly associated with a poorer overall survival (OS) in the pooled results (HR, 1.95; 95% CI, 1.29-2.94; p = 0.002) in pancreatic cancer. However, GNRI was not significantly associated with progression-free survival (PFS) in pancreatic cancer (HR, 1.57; 95% CI, 0.90-2.73; p = 0.114). The pooled results indicated that a low GNRI was significantly associated with tumor location of pancreas head (OR, 2.18; 95% CI, 1.45-3.29; p < 0.001).Conclusions: This meta-analysis showed that low GNRI was significantly associated with poor OS but not with poor PFS in patients with pancreatic cancer. The GNRI is a novel and effective risk factor and a potential biomarker for the prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Lili Li
- Clinical Laboratory, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Junjun He
- Clinical Laboratory, Traditional Chinese Medical Hospital of Huzhou Affiliated to Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| |
Collapse
|
12
|
Mastracci TL, Apte M, Amundadottir LT, Alvarsson A, Artandi S, Bellin MD, Bernal-Mizrachi E, Caicedo A, Campbell-Thompson M, Cruz-Monserrate Z, El Ouaamari A, Gaulton KJ, Geisz A, Goodarzi MO, Hara M, Hull-Meichle RL, Kleger A, Klein AP, Kopp JL, Kulkarni RN, Muzumdar MD, Naren AP, Oakes SA, Olesen SS, Phelps EA, Powers AC, Stabler CL, Tirkes T, Whitcomb DC, Yadav D, Yong J, Zaghloul NA, Pandol SJ, Sander M. Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases: Workshop Proceedings. Diabetes 2023; 72:433-448. [PMID: 36940317 PMCID: PMC10033248 DOI: 10.2337/db22-0942] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/29/2022] [Indexed: 03/22/2023]
Abstract
The Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases workshop was a 1.5-day scientific conference at the National Institutes of Health (Bethesda, MD) that engaged clinical and basic science investigators interested in diseases of the pancreas. This report provides a summary of the proceedings from the workshop. The goals of the workshop were to forge connections and identify gaps in knowledge that could guide future research directions. Presentations were segregated into six major theme areas, including 1) pancreas anatomy and physiology, 2) diabetes in the setting of exocrine disease, 3) metabolic influences on the exocrine pancreas, 4) genetic drivers of pancreatic diseases, 5) tools for integrated pancreatic analysis, and 6) implications of exocrine-endocrine cross talk. For each theme, multiple presentations were followed by panel discussions on specific topics relevant to each area of research; these are summarized here. Significantly, the discussions resulted in the identification of research gaps and opportunities for the field to address. In general, it was concluded that as a pancreas research community, we must more thoughtfully integrate our current knowledge of normal physiology as well as the disease mechanisms that underlie endocrine and exocrine disorders so that there is a better understanding of the interplay between these compartments.
Collapse
Affiliation(s)
- Teresa L. Mastracci
- Department of Biology, Indiana University–Purdue University Indianapolis, Indianapolis, IN
| | - Minoti Apte
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | | | - Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Mount Sinai Hospital, New York, NY
| | - Steven Artandi
- Department of Internal Medicine, Stanford University, Stanford, CA
| | - Melena D. Bellin
- Departments of Pediatrics and Surgery, University of Minnesota Medical School, Minneapolis, MN
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Andrea Geisz
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Rebecca L. Hull-Meichle
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University, Ulm, Germany
| | - Alison P. Klein
- Department of Pathology and Medicine, Johns Hopkins School of Medicine, Baltimore MD
| | - Janel L. Kopp
- Department of Cellular & Physiological Sciences, The University of British Columbia, Vancouver, Canada
| | | | - Mandar D. Muzumdar
- Departments of Genetics and Internal Medicine (Oncology), Yale University School of Medicine, New Haven, CT
| | | | - Scott A. Oakes
- Department of Pathology, The University of Chicago, Chicago, IL
| | - Søren S. Olesen
- Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Alvin C. Powers
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | - Cherie L. Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Temel Tirkes
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN
| | | | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Jing Yong
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Norann A. Zaghloul
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Stephen J. Pandol
- Department of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Maike Sander
- Department of Pediatrics and Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
13
|
Purinergic receptor: a crucial regulator of adipose tissue functions. Purinergic Signal 2023; 19:273-281. [PMID: 36515790 PMCID: PMC9984650 DOI: 10.1007/s11302-022-09907-w] [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: 11/01/2021] [Accepted: 11/14/2022] [Indexed: 12/15/2022] Open
Abstract
Obesity is a public-health challenge resulting from an imbalance between energy expenditure and calorie intake. This health problem exacerbates a variety of metabolic complications worldwide. Adipose tissue is an essential regulator of energy homeostasis, and the functions within it are regulated by purinergic receptors. A1R, P2X7R, and P2YR mainly mediate energy homeostasis primarily through regulating energy storage and adipokines secretion in white adipose tissue (WAT). P2X5R is a novel-specific cell surface marker in brown/beige adipocytes. A2R is a promising therapeutic target for stimulating energy expenditure in brown adipose tissue (BAT) and also mediating WAT browning. Based on these features, purinergic receptors may be an appropriate target in treating obesity. In this review, the role of purinergic receptors in different types of adipose tissue is summarized. An improved understanding of purinergic receptor functions in adipose tissue may lead to more effective treatment interventions for obesity and its related metabolic disorders.
Collapse
|
14
|
Saeed U, Myklebust TÅ, Robsahm TE, Møller B, Mala T, Skålhegg BS, Yaqub S. Body mass index and pancreatic adenocarcinoma: A nationwide registry-based cohort study. Scand J Surg 2023; 112:11-21. [PMID: 36173093 DOI: 10.1177/14574969221127530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND OBJECTIVE An association between body mass index (BMI) and pancreatic cancer is suggested in observational studies. However, further studies are required to substantiate available evidence. The aim of this study was to explore the association between BMI and pancreatic ductal adenocarcinoma (PDAC) risk, treatment, and mortality. METHODS A registry-based cohort study was performed by combining data from four registries in Norway. Baseline data were collected between 1963 and 1975 with follow-up data collected until 2018. Kaplan-Meier curves and multivariable Cox regressions were estimated. Chi-square tests were used to analyze differences between groups. RESULTS The study cohort consisted of 1,723,692 individuals. A total of 8973 PDAC cases were identified during 55,744,749 person-years of follow-up. A 5 kg/m2 increase in BMI was associated with an increased risk of PDAC if high BMI at young age (16-29 years) (hazard ratio (HR): 1.21, 95% confidence interval (CI): 1.13-1.31), both for men (HR: 1.30, 95% CI: 1.15-1.46) and women (HR: 1.16, 95% CI: 1.05-1.28). In men, there was a 52% increase in risk of early-onset PDAC (<age 50) (HR: 1.52, 95% CI: 1.13-2.03) with 5 kg/m2 increase in BMI. A total of 2645 individuals were diagnosed with stage 1-3 disease, of whom 1131 underwent curative surgery. In all, 49% of normal weight, 38% overweight, and 30% obese individuals with stage 1-3 PDAC underwent surgery with curative intent (p < 0.001). Cancer survival was lower in overweight and obese individuals for both sexes. Analysis adjusted for sex, age, and period of diagnosis confirmed increased risk of cancer death in obese individuals. CONCLUSION This study suggests that increased BMI in young adults may increase the risk of PDAC, and higher BMI in men is associated with an increased risk of early onset PDAC (<age 50). Overweight and obese individuals were less likely to undergo curative surgery and obese individuals had a higher risk of cancer-related death.
Collapse
Affiliation(s)
- Usman Saeed
- Department of Gastrointestinal Surgery ,Oslo University Hospital, PO Box 4950 Nydalen N-0424 Oslo Norway
| | - Tor Å Myklebust
- Department of Registration, Cancer Registry of Norway, Oslo, Norway Department of Research and Innovation, Møre and Romsdal Hospital Trust, Ålesund, Norway
| | - Trude E Robsahm
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Bjørn Møller
- Department of Registration, Cancer Registry of Norway, Oslo, Norway
| | - Tom Mala
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway Institute of Clinical Medicine, University of Oslo, Norway
| | - Bjørn S Skålhegg
- Division for Molecular Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Sheraz Yaqub
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, NorwayInstitute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
15
|
Bulsei J, Chierici A, Alifano M, Castaldi A, Drai C, De Fatico S, Rosso E, Fontas E, Iannelli A. Bariatric surgery reduces the risk of pancreatic cancer in individuals with obesity before the age of 50 years: A nationwide administrative data study in France. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2022; 49:788-793. [PMID: 36376141 DOI: 10.1016/j.ejso.2022.11.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Obesity is a well-established risk factor for pancreatic cancer. Bariatric surgery has demonstrated superior results in terms of weight loss and obesity-related comorbidities compared to medical and behavioral treatments. The aim of this study is to evaluate the effect of bariatric surgery on pancreatic cancer incidence in individuals with obesity. METHOD Individuals with a diagnosis of obesity were retrieved from the French national hospital discharge database. We conducted a cohort study comparing the risk to develop pancreatic cancer in individuals with obesity with and without history of bariatric surgery; the inverse probability of treatment weighting (IPTW) method was performed to assess the uncertainty around the results. Moreover, a subgroup analysis according to age at the time of bariatric surgery was performed to study its impact on the risk of pancreatic cancer. Finally, possible differences depending on the type of bariatric procedure (sleeve gastrectomy vs Roux-en-Y gastric bypass) were also explored. RESULTS 160,129 (Bariatric Surgery group) and 1,263,804 (control group) patients with 5.2 ± 1.9 and 6.0 ± 1.9 years of follow-up respectively were included. A significant reduced risk to develop pancreatic cancer during follow-up was identified for the bariatric surgery group in the overall population (HR: 0.567). However, this reduced risk was only observed in the 18-50 years group. These results were furtherly confirmed after IPTW analysis. No difference was found between different bariatric procedures. CONCLUSION Bariatric surgery has a protective effect against pancreatic cancer in the 18-50 years population. High-quality prospective studies are needed to confirm these results.
Collapse
Affiliation(s)
- Julie Bulsei
- Centre Hospitalier Universitaire de Nice, Department of Clinical Research and Innovation, Université Côte d'Azur, Nice, France
| | - Andrea Chierici
- Service de Chirurgie Digestive, Centre Hospitalier d'Antibes Juan-les-Pins, 107, av. de Nice, 06600, Antibes, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, France; INSERM U1138 Team «Cancer, Immune Control, and Escape», Cordeliers Research Center, University of Paris, France
| | - Antonio Castaldi
- Centre Hospitalier Universitaire de Nice - Digestive Surgery and Liver Transplantation Unit, Archet 2 Hospital, Nice, France
| | - Céline Drai
- Centre Hospitalier Universitaire de Nice - Digestive Surgery and Liver Transplantation Unit, Archet 2 Hospital, Nice, France; Université Côte d'Azur, Nice, France
| | - Serena De Fatico
- Centre Hospitalier Universitaire de Nice - Digestive Surgery and Liver Transplantation Unit, Archet 2 Hospital, Nice, France
| | - Edoardo Rosso
- Unité des Maladies de l'Appareil Digestif et Endocrine, Department of Surgery and Robotics, Centre Hospitalier de Luxembourg, L-1210 Luxembourg, Luxembourg
| | - Eric Fontas
- Centre Hospitalier Universitaire de Nice, Department of Clinical Research and Innovation, Université Côte d'Azur, Nice, France
| | - Antonio Iannelli
- Centre Hospitalier Universitaire de Nice - Digestive Surgery and Liver Transplantation Unit, Archet 2 Hospital, Nice, France; Université Côte d'Azur, Nice, France; Inserm, U1065, Team 8 "Hepatic complications of obesity and alcohol", France.
| |
Collapse
|
16
|
Amanollahi A, Khazdouz M, Malekahmadi M, Klement RJ, Lee D, Khodabakhshi A. Effect of Ketogenic Diets on Cardio-Metabolic Outcomes in Cancer Patients: A Systematic Review and Meta-Analysis of Controlled Clinical Trials. Nutr Cancer 2022; 75:95-111. [PMID: 36110060 DOI: 10.1080/01635581.2022.2117388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this systematic review and meta-analysis of clinical controlled trials (CCTs) we aimed to investigate the efficacy of KDs as an adjuvant therapy on cardiometabolic outcomes in patient with cancer compared to conventional non-ketogenic diets. Only CCTs involving cancer patients that were assigned to either a KD or a standard diet control group were selected. Two reviewers independently extracted the data, and a meta-analysis was performed using a random effects model to estimate weighted mean differences (WMDs) and confidence intervals (CIs) in body composition, metabolite, lipid profile, liver and kidney function parameters and quality of life. This meta-analysis showed a significant reduction in body weight (WMD= -2.99 kg; 95% CI: -4.67, -1.31; and P < 0.001), BMI (WMD= -1.08 kg/m2; 95% CI: -1.81, -0.34; P ≤ 0.002) and fat mass (WMD= -1.48 kg; 95% CI: -2.56, -0.40; and P = 0.007) by a KD. KDs significantly decreased glucose (WMD= -5.22 mg/dl; 95% CI: -9.0, -1.44; and P = 0.007), IGF-1 (WMD= -17.52 ng/ml; 95% CI: -20.24, -14.8; and P ˂0.001) and triglyceride (WMD= -24.46 mg/dl; 95% CI: -43.96, -4.95; and P = 0.014) levels. Furthermore, KDs induced ketosis by increasing β-hydroxybutyrate (WMD= 0.56 mmol/l; 95% CI: 0.37, 0.75; and P < 0.001). There were non-significant pooled effects of KDs on improving insulin, C-reactive protein and cholesterol levels and kidney and liver function. Emotional functioning was even increased significantly in the KD compared to the SD groups. In summary we found that KDs result in a greater reduction in glucose, IGF-1, triglycerides, body weight, BMI, and fat mass in cancer patients compared to traditional non-ketogenic diets and improved emotional functioning. The quality of evidence in the meta-analysis was moderate according to the Nutrigrade assessment.
Collapse
Affiliation(s)
- Alireza Amanollahi
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Khazdouz
- Growth and Development Research Center, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Malekahmadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.,Department of Clinical Nutrition, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Rainer J Klement
- Department of Radiation Oncology, Leopoldina Hospital Schweinfurt, Schweinfurt, Germany
| | - Derek Lee
- Biology Department, Boston College, Chestnut Hill, MA, USA
| | - Adeleh Khodabakhshi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.,Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
17
|
Liu S, Wu D, Fan Z, Yang J, Li Y, Meng Y, Gao C, Zhan H. FABP4 in obesity-associated carcinogenesis: Novel insights into mechanisms and therapeutic implications. Front Mol Biosci 2022; 9:973955. [PMID: 36060264 PMCID: PMC9438896 DOI: 10.3389/fmolb.2022.973955] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022] Open
Abstract
The increasing prevalence of obesity worldwide is associated with an increased risk of various diseases, including multiple metabolic diseases, cardiovascular diseases, and malignant tumors. Fatty acid binding proteins (FABPs) are members of the adipokine family of multifunctional proteins that are related to fatty acid metabolism and are divided into 12 types according to their tissue origin. FABP4 is mainly secreted by adipocytes and macrophages. Under obesity, the synthesis of FABP4 increases, and the FABP4 content is higher not only in tissues but also in the blood, which promotes the occurrence and development of various cancers. Here, we comprehensively investigated obesity epidemiology and the biological mechanisms associated with the functions of FABP4 that may explain this effect. In this review, we explore the molecular mechanisms by which FABP4 promotes carcinoma development and the interaction between fat and cancer cells in obese circumstances here. This review leads us to understand how FABP4 signaling is involved in obesity-associated tumors, which could increase the potential for advancing novel therapeutic strategies and molecular targets for the systematic treatment of malignant tumors.
Collapse
|
18
|
Roalsø MTT, Hald ØH, Alexeeva M, Søreide K. Emerging Role of Epigenetic Alterations as Biomarkers and Novel Targets for Treatments in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14030546. [PMID: 35158814 PMCID: PMC8833770 DOI: 10.3390/cancers14030546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Epigenetic alterations cause changes in gene expression without affecting the DNA sequence and are found to affect several molecular pathways in pancreatic tumors. Such changes are reversible, making them potential drug targets. Furthermore, epigenetic alterations occur early in the disease course and may thus be explored for early detection. Hence, a deeper understanding of epigenetics in pancreatic cancer may lead to improved diagnostics, treatments, and prognostication. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with limited treatment options. Emerging evidence shows that epigenetic alterations are present in PDAC. The changes are potentially reversible and therefore promising therapeutic targets. Epigenetic aberrations also influence the tumor microenvironment with the potential to modulate and possibly enhance immune-based treatments. Epigenetic marks can also serve as diagnostic screening tools, as epigenetic changes occur at early stages of the disease. Further, epigenetics can be used in prognostication. The field is evolving, and this review seeks to provide an updated overview of the emerging role of epigenetics in the diagnosis, treatment, and prognostication of PDAC.
Collapse
Affiliation(s)
- Marcus T. T. Roalsø
- Department of Quality and Health Technology, University of Stavanger, 4036 Stavanger, Norway;
- HPB Unit, Department of Gastrointestinal Surgery, Stavanger University Hospital, 4068 Stavanger, Norway;
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Medicine, Stavanger University Hospital, 4068 Stavanger, Norway
| | - Øyvind H. Hald
- Department of Oncology, University Hospital of North Norway, 9038 Tromsø, Norway;
| | - Marina Alexeeva
- HPB Unit, Department of Gastrointestinal Surgery, Stavanger University Hospital, 4068 Stavanger, Norway;
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Medicine, Stavanger University Hospital, 4068 Stavanger, Norway
| | - Kjetil Søreide
- HPB Unit, Department of Gastrointestinal Surgery, Stavanger University Hospital, 4068 Stavanger, Norway;
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Medicine, Stavanger University Hospital, 4068 Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway
- Correspondence:
| |
Collapse
|
19
|
Gheorghe G, Diaconu CC, Ionescu V, Constantinescu G, Bacalbasa N, Bungau S, Gaman MA, Stan-Ilie M. Risk Factors for Pancreatic Cancer: Emerging Role of Viral Hepatitis. J Pers Med 2022; 12:83. [PMID: 35055398 PMCID: PMC8780367 DOI: 10.3390/jpm12010083] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/09/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most aggressive malignant neoplastic diseases. The incidence and mortality rates of this disease vary depending on geographical area, which might be explained by the different exposure to risk factors. To improve the prognosis of patients with pancreatic cancer, different approaches are needed for an earlier diagnosis. Identification of risk factors and implementation of screening strategies are essential for a better prognosis. Currently, the risk factors for pancreatic cancer fall into two broad categories, namely extrinsic and intrinsic factors. Extrinsic factors include alcohol consumption, smoking, a diet rich in saturated fats, and viral infections such as chronic infection with hepatitis B and C viruses. The pathophysiological mechanisms explaining how these hepatotropic viruses contribute to the development of pancreatic cancer are not fully elucidated. The common origin of hepatocytes and pancreatic cells in the multipotent endodermal cells, the common origin of the blood vessels and biliary ducts of the pancreas and the liver, or chronic inflammatory changes may be involved in this interaction. A careful monitoring of patients with viral liver infections may contribute to the early diagnosis of pancreatic cancer and improve the prognosis of these patients.
Collapse
Affiliation(s)
- Gina Gheorghe
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| | - Camelia Cristina Diaconu
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Vlad Ionescu
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| | - Gabriel Constantinescu
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| | - Nicolae Bacalbasa
- Department of Visceral Surgery, “Carol Davila” University of Medicine and Pharmacy, Center of Excellence in Translational Medicine “Fundeni” Clinical Institute, 022328 Bucharest, Romania;
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - Mihnea-Alexandru Gaman
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Madalina Stan-Ilie
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| |
Collapse
|
20
|
Fu N, Jiang Y, Qin K, Chen H, Deng X, Shen B. Higher body mass index indicated better overall survival in pancreatic ductal adenocarcinoma patients: a real-world study of 2010 patients. BMC Cancer 2021; 21:1318. [PMID: 34886801 PMCID: PMC8656027 DOI: 10.1186/s12885-021-09056-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 11/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The association between body mass index (BMI) and the overall survival (OS) of pancreatic ductal adenocarcinoma (PDAC) patients remains controversial and unclear, METHOD: A total of 2010 patients from a high-volume center were enrolled in the study. The OS of PDAC patients was evaluated based on restricted cubic spline (RCS), propensity score (PS) and multivariable risk adjustment analyses. RESULT BMI was linearly related to the OS (total P = 0.004, nonlinear P = 0.124). BMI was analyzed as categorical data based on X-tile software-defined cutoffs and World Health Organization (WHO)-recommended cutoffs. Adjusted with confounding covariates, higher BMI manifested as a positive prognostic predictor. Furthermore, BMI was proven to be associated with the OS in the PS analysis. (UnderweightXtile vs. NormalXtileP = 0.003, OverweightXtile vs. NormalXtileP = 0.019; UnderweightWHO vs. NormalWHOP < 0.001, OverweightWHO vs. NormalWHOP = 0.024). It was also revealed that patients with higher BMI benefitted more from chemotherapy. (Adjusted hazard ratio (aHR): UnderweightXtile vs. NormalXtile vs. OverweightXtile: 0.565 vs. 0.474 vs. 0.409; UnderweightWHO vs. NormalWHO vs. OverweightWHO: 0.613 vs. 0.464 vs. 0.425). CONCLUSION Among PDAC patients, there was a positive association between BMI and the OS, especially in patients treated with chemotherapy.
Collapse
Affiliation(s)
- Ningzhen Fu
- Department of General Surgery, Pancreatic Disease Center, Shanghai Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, No.197 Ruijin Er Road, Shanghai, 200025, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Jiang
- Department of General Surgery, Pancreatic Disease Center, Shanghai Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, No.197 Ruijin Er Road, Shanghai, 200025, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Qin
- Department of General Surgery, Pancreatic Disease Center, Shanghai Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, No.197 Ruijin Er Road, Shanghai, 200025, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Shanghai Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, No.197 Ruijin Er Road, Shanghai, 200025, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Shanghai Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, No.197 Ruijin Er Road, Shanghai, 200025, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Shanghai Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, No.197 Ruijin Er Road, Shanghai, 200025, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
The Relationship between Nutritional Status and Body Composition with Clinical Parameters, Tumor Stage, CA19-9, CEA Levels in Patients with Pancreatic and Periampullary Tumors. Curr Oncol 2021; 28:4805-4820. [PMID: 34898583 PMCID: PMC8628718 DOI: 10.3390/curroncol28060406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 12/26/2022] Open
Abstract
Recent studies have obtained inadequate data on the association between nutritional status, body composition, clinical parameters and tumor stage in patients withpancreatic and periampullary tumors. The purpose of this study was to assess the relationship between nutritional status (NS), body composition (BC) and selected clinical parameters in patients with pancreatic and periampullary cancer, as well as describe the differences between resection and non-resection groups. This is a prospective study of 76 patients with pancreatic and periampullary tumors. We evaluated NS, BMI, body mass loss (BML) and albumin, total protein, CRP, CEA, CA19-9, lipase, amylase, tumor stage, and BC using bioelectrical impedance (BIA). All subjects were divided into resection (n = 59) and non-resection (n = 17) groups. The non-resection group had a worse NS, as well as increased amylase and WBC, compared to the resection. The selected parameters of BC corresponded to BML albumin, TP, NS, age, BMI, Karnofsky, RBC, HCT and HGB. No associations were found between BC with tumor size, CRP, CA19-9, and CEA. We recorded the relationship between metastasis and NRS, as well as tumor size with SGA. The percentage of BML was positively correlated with age and CRP but negatively correlated with RBC, HGB, HCT and anthropometric measurements. We found many statistical correlations with NS and selected parameters, as well as differences between the resection and non-resection group. The detection of early prognostic factors of nutritional impairments would improve the quality of life and reduce the rate of postoperative complications.
Collapse
|
22
|
Deng GC, Lv Y, Yan H, Sun DC, Qu TT, Pan YT, Han QL, Dai GH. Nomogram to predict survival of patients with advanced and metastatic pancreatic Cancer. BMC Cancer 2021; 21:1227. [PMID: 34781928 PMCID: PMC8594118 DOI: 10.1186/s12885-021-08943-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 10/28/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Nomograms are rarely employed to estimate the survival of patients with advanced and metastatic pancreatic cancer (PC). Herein, we developed a comprehensive approach to using a nomogram to predict survival probability in patients with advanced and metastatic PC. METHODS A total of 323 patients with advanced and metastatic PC were identified from the Chinese People's Liberation Army (PLA) General Hospital. A baseline nomogram was constructed using baseline variables of 323 patients. Additionally, 233 patients, whose tumors showed initial responses to first-line chemotherapy, were enrolled in the chemotherapy response-based model. 128 patients and 108 patients with advanced and metastatic PC from January 2019 to April 2021 were selected for external validating baseline model and chemotherapy response-based model. The 1-year and 2-year survival probability was evaluated using multivariate COX regression models. The discrimination and calibration capacity of the nomograms were assessed using C-statistic and calibration plots. The predictive accuracy and net benefit of the nomograms were evaluated using ROC curve and DCA, respectively. RESULTS In the baseline model, six variables (gender, KPS, baseline TB, baseline N, baseline WBC and baseline CA19-9) were used in the final model. In the chemotherapy response-based model, nine variables (KPS, gender, ascites, baseline N, baseline CA 19-9, baseline CEA, change in CA 19-9 level at week, change in CEA level at week and initial response to chemotherapy) were included in the final model. The C-statistics of the baseline nomogram and the chemotherapy response-based nomogram were 0.67 (95% CI, 0.62-0.71) and 0.74 (95% CI, 0.69-0.77), respectively. CONCLUSION These nomograms were constructed to predict the survival probability of patients of advanced and metastatic PC. The baseline model and chemotherapy response-based model performed well in survival prediction.
Collapse
Affiliation(s)
- G C Deng
- School of Medicine, Nankai University, Tianjin, China
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Y Lv
- School of Medicine, Nankai University, Tianjin, China
| | - H Yan
- School of Medicine, Nankai University, Tianjin, China
| | - D C Sun
- School of Medicine, Nankai University, Tianjin, China
| | - T T Qu
- School of Medicine, Nankai University, Tianjin, China
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Y T Pan
- School of Medicine, Nankai University, Tianjin, China
| | - Q L Han
- School of Medicine, Nankai University, Tianjin, China.
| | - G H Dai
- School of Medicine, Nankai University, Tianjin, China.
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
23
|
Hsieh MC, Zhang L, Velasco-Gonzalez C, Yi Y, Pareti LA, Trapido EJ, Chen VW, Wu XC. Impact of diabetes and modifiable risk factors on pancreatic cancer survival in a population-based study after adjusting for clinical factors. Cancer Causes Control 2021; 33:37-48. [PMID: 34633573 DOI: 10.1007/s10552-021-01497-z] [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: 04/12/2021] [Accepted: 09/22/2021] [Indexed: 01/18/2023]
Abstract
PURPOSES Our study aimed to examine the impact of diabetes, smoking and BMI on pancreatic cancer survival in a population-based setting by adjusting both sociodemographic and clinical factors and measuring their attributable risk. METHODS Data on pancreatic adenocarcinoma patients diagnosed in 2011-2017 were acquired from the Louisiana Tumor Registry. Diabetes, smoking, height, and weight were abstracted from medical records and linked with Hospital Inpatient Discharge Data to enhance the completeness of the diabetes data. The Cox regression model was used to assess effect sizes of diabetes, smoking, and BMI on cancer-specific survival and survival rate. The partial population attributable risk was employed to measure the attributable risk of these risk factors. RESULTS Of the 3,200 eligible patients, 34.6% were diabetics, 23.9% were current smokers, and 52.3% had BMI ≥ 25 kg/m2. After adjusting for sociodemographic and clinical factors, diabetic patients had an increased cancer-specific death risk of 15% (95% CI, 1.06-1.25), 36% (95% CI, 1.19-1.44) for current smokers, and 24% (95% CI, 1.00-1.54) for patients with a BMI ≥ 40 when compared to their counterparts. Diabetic current smokers had significantly lower 2- and 3-year adjusted cancer-specific survival rates, 13.1% and 10.5%, respectively. By eliminating diabetes and modifiable risk factors, an estimated 16.6% (95% CI, 6.9%-25.9%) of the cancer-specific deaths could be avoided during a nine-year observational period between 2011 and 2019. CONCLUSIONS Diabetes and smoking contributed substantially to the reduction of pancreatic cancer survival even after controlling for sociodemographic and clinical factors; however, BMI ≥ 35 was observed to increase risk of mortality among stage III-IV patients only.
Collapse
Affiliation(s)
- Mei-Chin Hsieh
- Louisiana Tumor Registry, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier St., 3rd floor, New Orleans, LA, 70112, USA. .,Epidemiology Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| | - Lu Zhang
- Department of Public Health Sciences, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC, 29634, USA
| | - Cruz Velasco-Gonzalez
- Center for Outcomes and Health Services Research, Ochsner Health System, Jefferson, LA, 70121, USA
| | - Yong Yi
- Louisiana Tumor Registry, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier St., 3rd floor, New Orleans, LA, 70112, USA
| | - Lisa A Pareti
- Louisiana Tumor Registry, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier St., 3rd floor, New Orleans, LA, 70112, USA
| | - Edward J Trapido
- Epidemiology Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Vivien W Chen
- Louisiana Tumor Registry, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier St., 3rd floor, New Orleans, LA, 70112, USA.,Epidemiology Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Xiao-Cheng Wu
- Louisiana Tumor Registry, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier St., 3rd floor, New Orleans, LA, 70112, USA.,Epidemiology Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| |
Collapse
|
24
|
Stolzenberg-Solomon R. Are Prediagnostic Biomarkers of Inflammation and an Empirically Based Proinflammatory Dietary Pattern Associated With Poorer Pancreatic Cancer Survival? J Natl Cancer Inst 2021; 113:1123-1124. [PMID: 33739407 PMCID: PMC8418422 DOI: 10.1093/jnci/djab043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Rachael Stolzenberg-Solomon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Yuan C, Morales-Oyarvide V, Khalaf N, Perez K, Tabung FK, Ho GYF, Kooperberg C, Shadyab AH, Qi L, Kraft P, Sesso HD, Giovannucci EL, Manson JE, Stampfer MJ, Ng K, Fuchs CS, Wolpin BM, Babic A. Prediagnostic Inflammation and Pancreatic Cancer Survival. J Natl Cancer Inst 2021; 113:1186-1193. [PMID: 33739411 DOI: 10.1093/jnci/djab040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 01/21/2021] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chronic inflammation may promote initiation and progression of pancreatic cancer, but no studies have examined the association between inflammation in the period before diagnosis and pancreatic cancer survival. METHODS We prospectively examined the association of prediagnostic plasma levels of C-reactive protein, interleukin-6, and tumor necrosis factor-α receptor 2 with survival among 492 participants from 5 large US prospective cohort studies who developed pancreatic cancer. Using an empirical dietary inflammatory pattern (EDIP) score, we evaluated whether long-term proinflammatory diets were associated with survival among 1153 patients from 2 of the 5 cohorts. Cox proportional hazards regression was used to estimate hazard ratios for death with adjustment for potential confounders. All statistical tests were 2-sided. RESULTS Higher prediagnostic levels of C-reactive protein, interleukin-6, and tumor necrosis factor-α receptor 2 were individually associated with reduced survival (Ptrend = .03, .01, and .04, respectively). Compared with patients with a combined inflammatory biomarker score of 0 (all 3 marker levels below medians), those with a score of 3 (all 3 marker levels above medians) had a hazard ratio for death of 1.57 (95% confidence interval = 1.16 to 2.12; Ptrend = .003), corresponding to median overall survival times of 8 vs 5 months. Patients consuming the most proinflammatory diets (EDIP quartile 4) in the prediagnostic period had a hazard ratio for death of 1.34 (95% confidence interval = 1.13 to 1.59; Ptrend = .01), compared with those consuming the least proinflammatory diets (EDIP quartile 1). CONCLUSION Prediagnostic levels of inflammatory biomarkers and long-term proinflammatory diets were inversely associated with pancreatic cancer survival.
Collapse
Affiliation(s)
- Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Vicente Morales-Oyarvide
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Natalia Khalaf
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Fred K Tabung
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA.,The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gloria Y F Ho
- Department of Occupational Medicine, Epidemiology and Prevention, Feinstein Institute for Medical Research, Northwell Health, Great Neck, NY, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Aladdin H Shadyab
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Lihong Qi
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis School of Medicine, Davis, CA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Preventive Medicine and Division of Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Preventive Medicine and Division of Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Meir J Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Charles S Fuchs
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Diaconescu S, Gîlcă-Blanariu GE, Poamaneagra S, Marginean O, Paduraru G, Stefanescu G. Could the burden of pancreatic cancer originate in childhood? World J Gastroenterol 2021; 27:5322-5340. [PMID: 34539135 PMCID: PMC8409163 DOI: 10.3748/wjg.v27.i32.5322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/08/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
The presence of pancreatic cancer during childhood is extremely rare, and physicians may be tempted to overlook this diagnosis based on age criteria. However, there are primary malignant pancreatic tumors encountered in pediatric patients, such as pancreatoblastoma, and tumors considered benign in general but may present a malignant potential, such as the solid pseudo-papillary tumor, insulinoma, gastrinoma, and vasoactive intestinal peptide secreting tumor. Their early diagnosis and management are of paramount importance since the survival rates tend to differ for various types of these conditions. Many pediatric cancers may present pancreatic metastases, such as renal cell carcinoma, which may evolve with pancreatic metastatic disease even after two or more decades. Several childhood diseases may create a predisposition for the development of pancreatic cancer during adulthood; hence, there is a need for extensive screening strategies and complex programs to facilitate the transition from pediatric to adult healthcare. Nevertheless, genetic studies highlight the fact the specific gene mutations and family aggregations may be correlated with a special predisposition towards pancreatic cancer. This review aims to report the main pancreatic cancers diagnosed during childhood, the most important childhood diseases predisposing to the development of pancreatic malignancies, and the gene mutations associates with pancreatic malignant tumors.
Collapse
Affiliation(s)
- Smaranda Diaconescu
- Department of Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- Department of Pediatric Gastroenterology, St Mary Emergency Children's Hospital, Iasi 700309, Romania
| | - Georgiana Emmanuela Gîlcă-Blanariu
- Department of Gastroenterology and Hepatology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi 700115, Romania
- Department of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| | - Silvia Poamaneagra
- Department of Pediatric Gastroenterology, St Mary Emergency Children's Hospital, Iasi 700309, Romania
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures 540142, Romania
| | - Otilia Marginean
- Department of Pediatrics, Research Center of Disturbance of Growth and Development on Children-Belive, University of Medicine and Pharmacy “Victor Babes” Timisoara, Timisoara 300041, Romania
- First Clinic of Pediatrics, "Louis Turcanu" Emergency Childen's Hospital, Timisoara 300011, Romania
| | - Gabriela Paduraru
- Department of Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- Department of Pediatric Gastroenterology, St Mary Emergency Children's Hospital, Iasi 700309, Romania
| | - Gabriela Stefanescu
- Department of Gastroenterology and Hepatology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi 700115, Romania
- Department of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| |
Collapse
|
27
|
Fan R, Niu J, Ma H, Xie Q, Cheng J, Rao H, Dou X, Xie J, Zhao W, Peng J, Gao Z, Gao H, Chen X, Chen J, Li Q, Tang H, Zhang Z, Ren H, Cheng M, Liang X, Zhu C, Wei L, Jia J, Sun J, Hou J. Association of central obesity with hepatocellular carcinoma in patients with chronic hepatitis B receiving antiviral therapy. Aliment Pharmacol Ther 2021; 54:329-338. [PMID: 34157146 DOI: 10.1111/apt.16469] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/26/2021] [Accepted: 05/23/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Obesity is typically associated with metabolic dysfunction, but its impact on hepatocellular carcinoma (HCC) remains unclear in patients with chronic hepatitis B (CHB). AIM To study the effect of obesity on HCC development in patients with CHB receiving antiviral therapy. METHODS We included patients from a Chinese multicentre, prospective, observational, treated CHB cohort in this study. General obesity was evaluated by body-mass index (BMI). Central obesity was evaluated by waist circumference, waist-to-hip ratio and waist-to-height ratio. RESULTS A total of 5754 nucleos(t)ide analogue treated patients were enrolled in the analysis. The 5-year cumulative incidence of HCC was 2.9%. Waist-to-height ratio performed better in predicting HCC development than BMI, waist circumference or waist-to-hip ratio. Patients with central obesity (defined as waist-to-height ratio >0.5) had significantly higher 5-year incidence of HCC than those without central obesity in the overall population (3.9% vs 2.1%, hazard ratio [HR]: 2.06, P = 0.0001) and 745 propensity score matched pairs (4.7% vs 2.3%, HR: 2.04, P = 0.026), respectively. Besides cirrhosis status and aMAP HCC risk score, central obesity was also independently associated with HCC risk (HR: 1.63, P = 0.013). Waist-to-height ratio gain within 1 year was associated with a significantly higher HCC risk with an adjusted HR value of 1.88 (95% confidence interval: 1.12-3.13, P = 0.017). CONCLUSIONS Central obesity, evaluated by the waist-to-height ratio, was associated with a twofold increase in HCC risk among CHB patients receiving antiviral treatment, highlighting the important role of abnormal metabolic function in the progression of liver disease.
Collapse
Affiliation(s)
- Rong Fan
- Guangzhou, China.,Shenzhen, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wogsland CE, Lien HE, Pedersen L, Hanjra P, Grondal SM, Brekken RA, Lorens JB, Halberg N. High-dimensional immunotyping of tumors grown in obese and non-obese mice. Dis Model Mech 2021; 14:dmm048977. [PMID: 33653826 PMCID: PMC8033414 DOI: 10.1242/dmm.048977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 11/20/2022] Open
Abstract
Obesity is a disease characterized by chronic low-grade systemic inflammation and has been causally linked to the development of 13 cancer types. Several studies have been undertaken to determine whether tumors evolving in obese environments adapt differential interactions with immune cells and whether this can be connected to disease outcome. Most of these studies have been limited to single-cell lines and tumor models and analysis of limited immune cell populations. Given the multicellular complexity of the immune system and its dysregulation in obesity, we applied high-dimensional suspension mass cytometry to investigate how obesity affects tumor immunity. We used a 36-marker immune-focused mass cytometry panel to interrogate the immune landscape of orthotopic syngeneic mouse models of pancreatic and breast cancer. Unanchored batch correction was implemented to enable simultaneous analysis of tumor cohorts to uncover the immunotypes of each cancer model and reveal remarkably model-specific immune regulation. In the E0771 breast cancer model, we demonstrate an important link to obesity with an increase in two T-cell-suppressive cell types and a decrease in CD8 T cells.
Collapse
Affiliation(s)
- Cara E. Wogsland
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Hilde E. Lien
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Line Pedersen
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Pahul Hanjra
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Sturla M. Grondal
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Rolf A. Brekken
- Division of Surgical Oncology, Department of Surgery, and Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James B. Lorens
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| |
Collapse
|
29
|
Petrelli F, Cortellini A, Indini A, Tomasello G, Ghidini M, Nigro O, Salati M, Dottorini L, Iaculli A, Varricchio A, Rampulla V, Barni S, Cabiddu M, Bossi A, Ghidini A, Zaniboni A. Association of Obesity With Survival Outcomes in Patients With Cancer: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e213520. [PMID: 33779745 PMCID: PMC8008284 DOI: 10.1001/jamanetworkopen.2021.3520] [Citation(s) in RCA: 208] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Importance Obesity, defined as a body mass index (BMI) greater than 30, is associated with a significant increase in the risk of many cancers and in overall mortality. However, various studies have suggested that patients with cancer and no obesity (ie, BMI 20-25) have worse outcomes than patients with obesity. Objective To assess the association between obesity and outcomes after a diagnosis of cancer. Data Sources PubMed, the Cochrane Library, and EMBASE were searched from inception to January 2020. Study Selection Studies reporting prognosis of patients with obesity using standard BMI categories and cancer were included. Studies that used nonstandard BMI categories, that were limited to children, or that were limited to patients with hematological malignant neoplasms were excluded. Screening was performed independently by multiple reviewers. Among 1892 retrieved studies, 203 (17%) met inclusion criteria for initial evaluation. Data Extraction and Synthesis The Meta-analysis of Observational Studies in Epidemiology (MOOSE) and Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guidelines were reporting guideline was followed. Data were extracted by multiple independent reviewers. Risk of death, cancer-specific mortality, and recurrence were pooled to provide an adjusted hazard ratio (HR) with a 95% CI . A random-effects model was used for the retrospective nature of studies. Main Outcomes and Measures The primary outcome of the study was overall survival (OS) in patients with cancer, with and without obesity. Secondary end points were cancer-specific survival (CSS) and progression-free survival (PFS) or disease-free survival (DFS). The risk of events was reported as HRs with 95% CIs, with an HR greater than 1 associated with a worse outcome among patients with obesity vs those without. Results A total of 203 studies with 6 320 365 participants evaluated the association of OS, CSS, and/or PFS or DFS with obesity in patients with cancer. Overall, obesity was associated with a reduced OS (HR, 1.14; 95% CI, 1.09-1.19; P < .001) and CSS (HR, 1.17; 95% CI, 1.12-1.23; P < .001). Patients were also at increased risk of recurrence (HR, 1.13; 95% CI, 1.07-1.19; P < .001). Conversely, patients with obesity and lung cancer, renal cell carcinoma, or melanoma had better survival outcomes compared with patients without obesity and the same cancer (lung: HR, 0.86; 95% CI, 0.76-0.98; P = .02; renal cell: HR, 0.74; 95% CI, 0.53-0.89; P = .02; melanoma: HR, 0.74; 95% CI, 0.57-0.96; P < .001). Conclusions and Relevance In this study, obesity was associated with greater mortality overall in patients with cancer. However, patients with obesity and lung cancer, renal cell carcinoma, and melanoma had a lower risk of death than patients with the same cancers without obesity. Weight-reducing strategies may represent effective measures for reducing mortality in these patients.
Collapse
Affiliation(s)
- Fausto Petrelli
- Oncology Unit, Azienda Socio Sanitaria Territoriale Bergamo Ovest, Treviglio, Italy
| | - Alessio Cortellini
- Oncology Unit, Department of Biotechnology and Applied Clinical Sciences, San Salvatore Hospital, University of L’Aquila, L’Aquila, Italy
| | - Alice Indini
- Oncology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Maggiore Policlinico, Milano, Italy
| | - Gianluca Tomasello
- Oncology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Maggiore Policlinico, Milano, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Maggiore Policlinico, Milano, Italy
| | - Olga Nigro
- Oncology Unit, Azienda Socio Sanitaria Territoriale Sette Laghi, Ospedale di Circolo, Varese, Italy
| | - Massimiliano Salati
- Oncology Unit, University Hospital of Modena, Modena Cancer Centre, Modena, Italy
| | - Lorenzo Dottorini
- Oncology Unit, Azienda Socio Sanitaria Territoriale Bergamo Est, Seriate, Italy
| | - Alessandro Iaculli
- Oncology Unit, Azienda Socio Sanitaria Territoriale Bergamo Est, Seriate, Italy
| | - Antonio Varricchio
- Surgical Oncology Unit, Azienda Socio Sanitaria Territoriale Bergamo Ovest, Treviglio, Italy
| | - Valentina Rampulla
- Surgical Oncology Unit, Azienda Socio Sanitaria Territoriale Bergamo Ovest, Treviglio, Italy
| | - Sandro Barni
- Oncology Unit, Azienda Socio Sanitaria Territoriale Bergamo Ovest, Treviglio, Italy
| | - Mary Cabiddu
- Oncology Unit, Azienda Socio Sanitaria Territoriale Bergamo Ovest, Treviglio, Italy
| | - Antonio Bossi
- Endocrine Diseases Unit–Diabetes Regional Center, Azienda Socio Sanitaria Territoriale Bergamo Ovest, Treviglio, Italia
| | | | | |
Collapse
|
30
|
Cha JY, Park JS, Hong YK, Jeun SS, Ahn S. Impact of Body Mass Index on Survival Outcome in Patients with Newly Diagnosed Glioblastoma: A Retrospective Single-Center Study. Integr Cancer Ther 2021; 20:1534735421991233. [PMID: 33543653 PMCID: PMC7869148 DOI: 10.1177/1534735421991233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Introduction: The impact of obesity on survival outcomes in patients with glioblastoma
(GBM) has not been well reported and the results for patients are currently
unclear. We investigated the effect of obesity on survival outcomes in
patients with newly diagnosed GBM. Methods: Using electronic medical records, all GBM patients that visited the Seoul St.
Mary’s Hospital between 2008 and 2018 were reviewed. A total of 177 patients
met our eligibility criteria. The cut-off point for BMI was
23.0 kg/m2 based on previous studies which focused on Asian
populations. Results: A total of 177 patients met our eligibility criteria. The overall median BMI
of patients was 24.5 kg/m2 (range 15.82-39.26). About 62 patients
who had a BMI less than the cut-off value were assigned to the “lower BMI”
group, while 115 patients who had a BMI greater than the cut-off value were
assigned to the “higher BMI” group. In Kaplan-Meier survival analysis, the
median OS of the higher BMI group was longer than that of the lower BMI
group (21.3 months vs 15.3 months, P = .002). In
multivariate Cox regression analysis for OS, lower BMI was associated with
inferior OS (HR 1.48 CI 1.06-2.08, P = .002). Conclusion: Our findings suggest that elevated BMI may be associated with better survival
in patients with newly diagnosed GBM. Additional larger prospective studies
could help validate our findings to confirm the effect of body composition
and survival outcomes in GBM patients.
Collapse
Affiliation(s)
- Jun-Yong Cha
- The Catholic University of Korea, Seoul, South Korea
| | - Jae-Sung Park
- The Catholic University of Korea, Seoul, South Korea
| | - Yong-Kil Hong
- The Catholic University of Korea, Seoul, South Korea
| | - Sin-Soo Jeun
- The Catholic University of Korea, Seoul, South Korea
| | - Stephen Ahn
- The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
31
|
Wirkus J, Ead AS, Mackenzie GG. Impact of dietary fat composition and quantity in pancreatic carcinogenesis: Recent advances and controversies. Nutr Res 2020; 88:1-18. [PMID: 33607535 DOI: 10.1016/j.nutres.2020.12.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/11/2020] [Accepted: 12/20/2020] [Indexed: 12/11/2022]
Abstract
A significant number of pancreatic cancer cases are due to modifiable risk factors, with many being attributed to increased body fatness. This has sparked investigators to examine the role played by high dietary fat intake in pancreatic cancer development and the mechanisms driving this connection. However, there is currently no consensus on how dietary fat quantity and composition specifically affect pancreatic carcinogenesis. The objective of this narrative review is to discuss the link between high total fat consumption and fatty acid composition (saturated, mono-, or poly-unsaturated fats) with pancreatic cancer incidence and progression. Following our detailed analysis of the strengths and weaknesses of recent preclinical and human studies, we discuss existing research gaps and opportunities, and provide recommendations for future studies. Numerous studies suggest that diets high in omega-3 polyunsaturated fatty acids are associated with reduced pancreatic cancer risk. However, the current evidence appears insufficient for a general conclusion regarding the impact of other types of fat in pancreatic carcinogenesis, with many studies providing inconclusive findings due to study limitations. Thus, we recommend future studies to include detailed methodology of the animal experiments, not limited to the diet composition, type of ingredients, formulations, and administration of the diets. Moreover, human studies should include a diverse population and well-characterized biomarkers for accurate determination of dietary fat intake. Ultimately, this will aid the study rigor, and improve our understanding of the impact of fat quantity and composition in pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Joanna Wirkus
- Department of Nutrition, University of California, Davis. One Shields Ave, Davis, CA 95616, USA
| | - Aya S Ead
- Department of Nutrition, University of California, Davis. One Shields Ave, Davis, CA 95616, USA
| | - Gerardo G Mackenzie
- Department of Nutrition, University of California, Davis. One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
32
|
Obesity and Pancreatic Cancer: A Matched-Pair Survival Analysis. J Clin Med 2020; 9:jcm9113526. [PMID: 33142763 PMCID: PMC7693315 DOI: 10.3390/jcm9113526] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Morbid obesity is a risk factor for pancreatic ductal adenocarcinoma (PDAC). However, the impact of obesity on postoperative outcomes and overall survival in patients with PDAC remains a controversial topic. METHODS Patients who underwent pancreatic surgery for PDAC between 1997 and 2018 were included in this study. Matched pairs (1:1) were generated according to age, gender and American Society of Anesthesiologists status. Obesity was defined according to the WHO definition as BMI ≥ 30 kg/m2. The primary endpoint was the difference in overall survival between patients with and without obesity. RESULTS Out of 553 patients, a total of 76 fully matched pairs were generated. Obese patients had a mean BMI-level of 33 compared to 25 kg/m2 in patients without obesity (p = 0.001). The frequency of arterial hypertension (p = 0.002), intraoperative blood loss (p = 0.039), and perineural invasion (p = 0.033) were also higher in obese patients. Clinically relevant postoperative complications (p = 0.163) and overall survival rates (p = 0.885) were comparable in both study groups. Grade II and III obesity resulted in an impaired overall survival, although this was not statistically significant. Subgroup survival analyses revealed no significant differences for completion of adjuvant chemotherapy and curative-intent surgery. CONCLUSIONS Obesity did not affect overall survival and postoperative complications in these patients with PDAC. Therefore, pancreatic surgery should not be withheld from obese patients.
Collapse
|
33
|
Zhao TC, Liang SY, Ju WT, Liu Y, Tan YR, Zhu DW, Zhang CP, Zhang ZY, Zhong LP. Normal BMI predicts the survival benefits of inductive docetaxel, cisplatin, and 5-fluorouracil in patients with locally advanced oral squamous cell carcinoma. Clin Nutr 2020; 39:2751-2758. [PMID: 31839432 DOI: 10.1016/j.clnu.2019.11.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/18/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS This study aimed to evaluate the prognostic value of the body mass index (BMI), as well as the association with docetaxel, cisplatin, and 5-fluorouracil (TPF) induction chemotherapy in patients with locally advanced oral squamous cell carcinoma (OSCC). METHODS This retrospective study enrolled 253 patients with locally advanced OSCC between 2008 and 2010 based on our previous prospective, randomized, phase 3 trial (NCT01542931). Univariate and multivariate Cox regression models, and the Kaplan-Meier method were used for survival analyses. RESULTS Among the 253 patients, the BMI at the time of clinical diagnosis ranged from 13.16 to 34.66 kg/m2. Smoking status among patients showed a marked correlation with a higher BMI status at the time of clinical diagnosis (tobacco status: P < 0.001). The distribution of clinical nodal (cN) stage was significantly different, as patients with higher BMIs generally had earlier cN stages (P < 0.021) among the different BMI groups. The Kaplan-Meier analysis showed that the BMI was significantly correlated with overall survival (OS, P = 0.004), disease-free survival (DFS, P = 0.005), locoregional recurrence-free survival (LRFS, P = 0.003) and distant metastasis-free survival (DMFS, P = 0.007). When the BMI was included in the multivariate Cox regression model adjusted for potentially confounding clinical variables, the BMI was shown to be an independent predictive factor of OS (P = 0.015), DFS (P = 0.015), LRFS (P = 0.009), and DMFS (P = 0.023). The TPF group showed better 5-year clinical survival rates than the control group when analyzing patients with a normal BMI (OS: 64.2% vs. 55.9%; DFS: 54.7% vs. 46.4%; LRFS: 56.6% vs. 49.6%; DMFS: 64.2% vs. 56.0%), but no significant difference was observed. Subgroup survival analysis indicated that patients with a normal BMI and clinical stage IVA disease who accepted TPF induction chemotherapy had a significantly improved OS (HR: 0.425, 95% CI: 0.187-0.966, P = 0.035) and DMFS (HR: 0.425, 95% CI: 0.187-0.966, P = 0.034). CONCLUSION The BMI at the time of clinical diagnosis was showed to be an independent predictive factor for patients with locally advanced OSCC. Compared with normoweight patients, underweight patients may have worse clinical outcomes, while overweight and obese patients have a better prognosis. A normal BMI in clinical stage IVA OSCC patients predicts significant OS and DMFS benefits of TPF induction chemotherapy.
Collapse
Affiliation(s)
- Tong-Chao Zhao
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Si-Yuan Liang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Wu-Tong Ju
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Ying Liu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Yi-Ran Tan
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China.
| | - Dong-Wang Zhu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Chen-Ping Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Zhi-Yuan Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Lai-Ping Zhong
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China.
| |
Collapse
|
34
|
Is Host Metabolism the Missing Link to Improving Cancer Outcomes? Cancers (Basel) 2020; 12:cancers12092338. [PMID: 32825010 PMCID: PMC7564800 DOI: 10.3390/cancers12092338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
For the past 100 years, oncologists have relentlessly pursued the destruction of tumor cells by surgical, chemotherapeutic or radiation oncological means. Consistent with this focus, treatment plans are typically based on key characteristics of the tumor itself such as disease site, histology and staging based on local, regional and systemic dissemination. Precision medicine is similarly built on the premise that detailed knowledge of molecular alterations of tumor cells themselves enables better and more effective tumor cell destruction. Recently, host factors within the tumor microenvironment including the vasculature and immune systems have been recognized as modifiers of disease progression and are being targeted for therapeutic gain. In this review, we argue that—to optimize the impact of old and new treatment options—we need to take account of an epidemic that occurs independently of—but has major impact on—the development and treatment of malignant diseases. This is the rapidly increasing number of patients with excess weight and its’ attendant metabolic consequences, commonly described as metabolic syndrome. It is well established that patients with altered metabolism manifesting as obesity, metabolic syndrome and chronic inflammation have an increased incidence of cancer. Here, we focus on evidence that these patients also respond differently to cancer therapy including radiation and provide a perspective how exercise, diet or pharmacological agents may be harnessed to improve therapeutic responses in this patient population.
Collapse
|
35
|
Abstract
Despite great advances in treatment, cancer remains a leading cause of death worldwide. Diet can greatly impact health, while caloric restriction and fasting have putative benefits for disease prevention and longevity. Strong epidemiological associations exist between obesity and cancer, whereas healthy diets can reduce cancer risk. However, less is known about how diet might impact cancer once it has been diagnosed and particularly how diet can impact cancer treatment. In the present review, we discuss the links between obesity, diet, and cancer. We explore potential mechanisms by which diet can improve cancer outcomes, including through hormonal, metabolic, and immune/inflammatory effects, and present the limited clinical research that has been published in this arena. Though data are sparse, diet intervention may reduce toxicity, improve chemotherapy efficacy, and lower the risk of long-term complications in cancer patients. Thus, it is important that we understand and expand the science of this important but complex adjunctive cancer treatment strategy.
Collapse
Affiliation(s)
- Steven D Mittelman
- Division of Pediatric Endocrinology, University of California, Los Angeles (UCLA), Children's Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA;
| |
Collapse
|
36
|
Shakhbazov R, Pattarabanjird O, Brayman KL, Alekberzade AV, Krylov NN. [Morphometry of adipose tissue for prediction of the outcomes of total pancreatectomy with pancreatic islets autotransplantation in patients with chronic pancreatitis]. Khirurgiia (Mosk) 2020:12-19. [PMID: 32500684 DOI: 10.17116/hirurgia202005112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To compare different clinical and morphometric features of patients undergoing TPAIT for prediction of postoperative outcomes. MATERIAL AND METHODS A retrospective review enrolled patients who underwent TPAIT for the period from January 2007 to October 2017. Morphometric parameters were analyzed using preoperative CT scans and patients were grouped to examine association of these characteristics with postoperative morbidity. Sarcopenia was defined as the presence of a TPA in the lowest sex-specific quartile. The impact of sarcopenia on pancreatic islet features, perioperative blood transfusion, ICU- and hospital-stay, complications, repeated admission within 90 days and islet function was assessed. RESULTS A total of 34 patients were included in this study (12 males and 24 females). At the time of diagnosis, mean age of patients was 43.1 years. Mean body mass index (BMI) in sarcopenic patients was 24.9 kg/m2, mean BMI in those without sarcopenia - 24.8 kg/m2 (p=1.00). Various surgical complications were observed in 11 patients (32.3%). Patients with sarcopenia experienced more complications (83.3%) compared with patients without sarcopenia (50%). However, differences were not significant (p=0.31). Islet characteristics (islet numbers, purity), readmission, ICU- and hospital-stay, incidence of blood transfusion and islet function were also similar in both groups. CONCLUSION Sarcopenia is not a predictor of postoperative complications and islet cell function in chronic pancreatitis patients following TPAIT.
Collapse
Affiliation(s)
| | | | - K L Brayman
- University of Virginia, Charlottesville, VA, USA
| | - A V Alekberzade
- Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - N N Krylov
- Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| |
Collapse
|
37
|
Chung KM, Singh J, Lawres L, Dorans KJ, Garcia C, Burkhardt DB, Robbins R, Bhutkar A, Cardone R, Zhao X, Babic A, Vayrynen SA, Dias Costa A, Nowak JA, Chang DT, Dunne RF, Hezel AF, Koong AC, Wilhelm JJ, Bellin MD, Nylander V, Gloyn AL, McCarthy MI, Kibbey RG, Krishnaswamy S, Wolpin BM, Jacks T, Fuchs CS, Muzumdar MD. Endocrine-Exocrine Signaling Drives Obesity-Associated Pancreatic Ductal Adenocarcinoma. Cell 2020; 181:832-847.e18. [PMID: 32304665 PMCID: PMC7266008 DOI: 10.1016/j.cell.2020.03.062] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/13/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022]
Abstract
Obesity is a major modifiable risk factor for pancreatic ductal adenocarcinoma (PDAC), yet how and when obesity contributes to PDAC progression is not well understood. Leveraging an autochthonous mouse model, we demonstrate a causal and reversible role for obesity in early PDAC progression, showing that obesity markedly enhances tumorigenesis, while genetic or dietary induction of weight loss intercepts cancer development. Molecular analyses of human and murine samples define microenvironmental consequences of obesity that foster tumorigenesis rather than new driver gene mutations, including significant pancreatic islet cell adaptation in obesity-associated tumors. Specifically, we identify aberrant beta cell expression of the peptide hormone cholecystokinin (Cck) in response to obesity and show that islet Cck promotes oncogenic Kras-driven pancreatic ductal tumorigenesis. Our studies argue that PDAC progression is driven by local obesity-associated changes in the tumor microenvironment and implicate endocrine-exocrine signaling beyond insulin in PDAC development.
Collapse
Affiliation(s)
| | - Jaffarguriqbal Singh
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Lauren Lawres
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | | | - Cathy Garcia
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Daniel B Burkhardt
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Rebecca Robbins
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Arjun Bhutkar
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Rebecca Cardone
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiaojian Zhao
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Sara A Vayrynen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Jonathan A Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel T Chang
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Richard F Dunne
- Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Aram F Hezel
- Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Albert C Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joshua J Wilhelm
- Schulze Diabetes Institute and Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN 55454, USA
| | - Melena D Bellin
- Schulze Diabetes Institute and Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN 55454, USA; Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN 55454, USA
| | - Vibe Nylander
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Anna L Gloyn
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | - Richard G Kibbey
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Smita Krishnaswamy
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charles S Fuchs
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT 06511, USA
| | - Mandar Deepak Muzumdar
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT 06511, USA.
| |
Collapse
|
38
|
Lupo F, Piro G, Torroni L, Delfino P, Trovato R, Rusev B, Fiore A, Filippini D, De Sanctis F, Manfredi M, Marengo E, Lawlor RT, Martini M, Tortora G, Ugel S, Corbo V, Melisi D, Carbone C. Organoid-Transplant Model Systems to Study the Effects of Obesity on the Pancreatic Carcinogenesis in vivo. Front Cell Dev Biol 2020; 8:308. [PMID: 32411709 PMCID: PMC7198708 DOI: 10.3389/fcell.2020.00308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related mortality among adults in developed countries. The discovery of the most common genetic alterations as well as the development of organoid models of pancreatic cancer have provided insight into the fundamental pathways driving tumor progression from a normal cell to non-invasive precursor lesion and finally to widely metastatic disease, offering new opportunities for identifying the key driver of cancer evolution. Obesity is one of the most serious public health challenges of the 21st century. Several epidemiological studies have shown the positive association between obesity and cancer-related morbidity/mortality, as well as poorer prognosis and treatment outcome. Despite strong evidence indicates a link between obesity and cancer incidence, the molecular basis of the initiating events remains largely elusive. This is mainly due to the lack of an accurate and reliable model of pancreatic carcinogenesis that mimics human obesity-associated PDAC, making data interpretation difficult and often confusing. Here we propose a feasible and manageable organoid-based preclinical tool to study the effects of obesity on pancreatic carcinogenesis. Therefore, we tracked the effects of obesity on the natural evolution of PDAC in a genetically defined transplantable model of the syngeneic murine pancreatic preneoplastic lesion (mP) and tumor (mT) derived-organoids that recapitulates the progression of human disease from early preinvasive lesions to metastatic disease. Our results suggest that organoid-derived transplant in obese mice represents a suitable system to study early steps of pancreatic carcinogenesis and supports the hypothesis that inflammation induced by obesity stimulates tumor progression and metastatization during pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Francesca Lupo
- Section of Anatomical Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Geny Piro
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lorena Torroni
- Unit of Epidemiology and Medical Statistics, University of Verona, Verona, Italy
| | - Pietro Delfino
- Section of Anatomical Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Rosalinda Trovato
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Borislav Rusev
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Alessandra Fiore
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Dea Filippini
- Section of Anatomical Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Francesco De Sanctis
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | | | - Maurizio Martini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Vincenzo Corbo
- Section of Anatomical Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy.,ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Davide Melisi
- Section of Medical Oncology, Department of Oncology, University of Verona, Verona, Italy
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
39
|
Bagni K, Chen IM, Johansen AZ, Dehlendorff C, Jensen BV, Hansen CP, Preus Hasselby J, Holländer NH, Nissen M, Bjerregaard JK, Pfeiffer P, Yilmaz MK, Rasmussen LS, Nielsen SE, Johansen JS. Prognostic impact of Charlson's Age-Comorbidity Index and other risk factors in patients with pancreatic cancer. Eur J Cancer Care (Engl) 2020; 29:e13219. [PMID: 31908093 DOI: 10.1111/ecc.13219] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 12/01/2019] [Accepted: 12/11/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Few studies have evaluated the impact of risk factors and comorbidity on overall survival (OS) in patients with pancreatic ductal adenocarcinoma (PDAC). The aim was to investigate the prognostic importance of Charlson's age-comorbidity index (CACI) and other risk factors on prognosis in a clinical real-world cohort of PDAC patients. METHODS A total of 1,159 patients with PDAC who had received at least one cycle of adjuvant or palliative chemotherapy were included from the Danish BIOPAC study. We analysed OS according to CACI, tobacco smoking, alcohol intake, performance status (PS), BMI and diabetes. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated for OS using Cox proportional hazards regression. RESULTS At the end of follow-up, 994 (86%) patients had died. The median OS was 298 days for all patients (range 3-3010) and shortest in patients with stage IV. No association with short OS was seen for CACI > 2, diabetes, alcohol abuse, tobacco smoking, hypertension, and high BMI. Multivariate analysis showed that stage (IV vs. I: HR = 9.05, 95% CI 5.17-15.84), PS (2 vs. 0: HR = 3.67, 2.92-4.61) and treatment with angiotensin-converting enzyme inhibitors (yes vs. no: HR = 1.31, 1.06-1.61) were independent negative prognostic factors. CONCLUSIONS We found that CACI, diabetes, tobacco smoking, alcohol abuse, hypertension, and high BMI were not associated with OS in a real-world cohort of patients with PDAC treated with chemotherapy. Only stage and PS were prognostic parameters.
Collapse
Affiliation(s)
- Karin Bagni
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Inna M Chen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Astrid Z Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Dehlendorff
- Statistics and Pharmacoepidemiology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Benny V Jensen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Carsten P Hansen
- Department of Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jane Preus Hasselby
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels H Holländer
- Department of Oncology at, Zealand University Hospital, Naestved, Denmark
| | - Mette Nissen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Oncology at, Zealand University Hospital, Naestved, Denmark
| | | | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Mette K Yilmaz
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Svend E Nielsen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Oncology and Palliative Care, North Zealand University Hospital, Hillerød, Denmark
| | - Julia S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Afzal A, Suhong L, Gage BF, Schoen MW, Carson K, Thomas T, Sanfilippo K. Splanchnic vein thrombosis predicts worse survival in patients with advanced pancreatic cancer. Thromb Res 2019; 185:125-131. [PMID: 31812026 DOI: 10.1016/j.thromres.2019.11.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Pancreatic cancer is a thrombogenic malignancy with nearly half of venous thrombotic events occurring in the splanchnic circulation. The effect of splanchnic vein thrombosis on mortality in pancreatic cancer is unknown. We studied the effect of splanchnic vein thrombosis on mortality in veterans with advanced pancreatic adenocarcinoma, and explored the association of anticoagulant therapy on mortality and hemorrhage. METHODS Using International Classification of Diseases (ICD) codes, we identified eligible patients and outcomes in the Veterans Health Administration database. Using Cox proportional hazards regression, we analyzed the association between splanchnic vein thrombosis and mortality among patients with advanced pancreatic cancer. We used propensity score inverse probability-of-treatment weighting to balance the groups who did and did not receive anticoagulation. To understand the role of anticoagulant therapy, we used Cox proportional hazards regression to analyze mortality and competing risk analysis to assess the risk of hemorrhage. RESULTS Of the patients with advanced pancreatic cancer (N = 6164), 122 developed splanchnic vein thrombosis. Splanchnic vein thrombosis was associated with a two-fold increase in mortality, aHR 2.02, 95% CI 1.65-2.47. The finding held true after restricting the analysis to patients undergoing treatment for pancreatic cancer, and after adjusting for immortal time bias by a 30-day landmark analysis. Anticoagulant therapy did not affect mortality (aHR 0.99, 95% CI 0.65-1.51), and increased the risk of hemorrhage (aHR 2.7, 95% CI 1.02-7.07). CONCLUSION Splanchnic vein thrombosis predicts worse survival in patients with advanced pancreatic adenocarcinoma. Anticoagulant therapy may not mitigate this increased mortality, and increases the risk of hemorrhage.
Collapse
Affiliation(s)
- Amber Afzal
- St Louis Veterans Health Administration Medical Center Research Service, St. Louis, MO, United States of America; Division of Hematology, Department of Medicine, Washington University School of Medicine in St Louis, MO, United States of America.
| | - Luo Suhong
- St Louis Veterans Health Administration Medical Center Research Service, St. Louis, MO, United States of America
| | - Brian F Gage
- Division of General Medical Sciences, Department of Medicine, Washington University School of Medicine in St Louis, MO, United States of America
| | - Martin W Schoen
- St Louis Veterans Health Administration Medical Center Research Service, St. Louis, MO, United States of America
| | - Kenneth Carson
- St Louis Veterans Health Administration Medical Center Research Service, St. Louis, MO, United States of America
| | - Theodore Thomas
- St Louis Veterans Health Administration Medical Center Research Service, St. Louis, MO, United States of America
| | - Kristen Sanfilippo
- St Louis Veterans Health Administration Medical Center Research Service, St. Louis, MO, United States of America; Division of Hematology, Department of Medicine, Washington University School of Medicine in St Louis, MO, United States of America
| |
Collapse
|
41
|
Hamada T, Yuan C, Bao Y, Zhang M, Khalaf N, Babic A, Morales-Oyarvide V, Cochrane BB, Gaziano JM, Giovannucci EL, Kraft P, Manson JE, Ng K, Nowak JA, Rohan TE, Sesso HD, Stampfer MJ, Amundadottir LT, Fuchs CS, De Vivo I, Ogino S, Wolpin BM. Prediagnostic Leukocyte Telomere Length and Pancreatic Cancer Survival. Cancer Epidemiol Biomarkers Prev 2019; 28:1868-1875. [PMID: 31427306 PMCID: PMC6825575 DOI: 10.1158/1055-9965.epi-19-0577] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/12/2019] [Accepted: 08/13/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Leukocyte telomere length has been associated with risk of subsequent pancreatic cancer. Few prospective studies have evaluated the association of prediagnostic leukocyte telomere length with pancreatic cancer survival. METHODS We prospectively examined the association of prediagnostic leukocyte telomere length with overall survival (OS) time among 423 participants diagnosed with pancreatic adenocarcinoma between 1984 and 2008 within the Health Professionals Follow-up Study, Nurses' Health Study, Physicians' Health Study, and Women's Health Initiative. We measured prediagnostic leukocyte telomere length in banked blood samples using quantitative PCR. Cox proportional hazards models were used to estimate HRs for OS with adjustment for potential confounders. We also evaluated 10 SNPs at the telomerase reverse transcriptase locus. RESULTS Shorter prediagnostic leukocyte telomere length was associated with reduced OS among patients with pancreatic cancer (P trend = 0.04). The multivariable-adjusted HR for OS comparing the lowest with highest quintiles of leukocyte telomere length was 1.39 (95% confidence interval, 1.01-1.93), corresponding to a 3-month difference in median OS time. In an analysis excluding cases with blood collected within 2 years of cancer diagnosis, the association was moderately stronger (HR, 1.55; 95% confidence interval, 1.09-2.21; comparing the lowest with highest quintiles; P trend = 0.01). No prognostic association or effect modification for the prognostic association of prediagnostic leukocyte telomere length was noted in relation to the studied SNPs. CONCLUSIONS Prediagnostic leukocyte telomere length was associated with pancreatic cancer survival. IMPACT Prediagnostic leukocyte telomere length can be a prognostic biomarker in pancreatic cancer.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ying Bao
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mingfeng Zhang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Natalia Khalaf
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Vicente Morales-Oyarvide
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | | | - J Michael Gaziano
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Jamaica Plain, Massachusetts
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meir J Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, Connecticut
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
- Smilow Cancer Hospital, New Haven, Connecticut
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Zhou B, Wu D, Liu H, Du LT, Wang YS, Xu JW, Qiu FB, Hu SY, Zhan HX. Obesity and pancreatic cancer: An update of epidemiological evidence and molecular mechanisms. Pancreatology 2019; 19:941-950. [PMID: 31447281 DOI: 10.1016/j.pan.2019.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/04/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
Despite advances in therapy and achievements in translational research, pancreatic cancer (PC) remains an invariably fatal malignancy. Risk factors that affect the incidence of PC include diabetes, smoking, obesity, chronic pancreatitis, and diet. The growing worldwide obesity epidemic is associated with an increased risk of the most common cancers, including PC. Chronic inflammation, hormonal effects, circulating adipokines, and adipocyte-mediated inflammatory and immunosuppressive microenvironment are involved in the association of obesity with PC. Herein, we systematically review the epidemiology of PC and the biological mechanisms that may account for this association. Included in this review is a discussion of adipokine-mediated inflammation, lipid metabolism, and the interactions of adipocytes with cancer cells. We consider the influence of bariatric surgery on the risk of PC risk as well as potential molecular targets of therapy. Our review leads us to conclude that targeting adipose tissue to achieve weight loss may represent a new therapeutic strategy for preventing and treating PC.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China; Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Dong Wu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Han Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Lu-Tao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China; Tumor Marker Detection Engineering Laboratory of Shandong Province, Jinan, Shandong Province, 250033, China
| | - Yun-Shan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China; Tumor Marker Detection Engineering Laboratory of Shandong Province, Jinan, Shandong Province, 250033, China
| | - Jian-Wei Xu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Fa-Bo Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China; Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - San-Yuan Hu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Han-Xiang Zhan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
43
|
Zamorano AS, Hagemann AR, Morrison L, Lee JA, Liao LM, Brinton LA, Park Y, Toriola AT. Pre-diagnosis body mass index, physical activity and ovarian cancer mortality. Gynecol Oncol 2019; 155:105-111. [DOI: 10.1016/j.ygyno.2019.07.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/18/2019] [Accepted: 07/28/2019] [Indexed: 12/24/2022]
|
44
|
Obesity-Induced Adipose Tissue Inflammation as a Strong Promotional Factor for Pancreatic Ductal Adenocarcinoma. Cells 2019; 8:cells8070673. [PMID: 31277269 PMCID: PMC6678863 DOI: 10.3390/cells8070673] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to soon become the second leading cause of cancer related deaths in the United States. This may be due to the rising obesity prevalence, which is a recognized risk factor for PDAC. There is great interest in deciphering the underlying driving mechanisms of the obesity–PDAC link. Visceral adiposity has a strong correlation to certain metabolic diseases and gastrointestinal cancers, including PDAC. In fact, our own data strongly suggest that visceral adipose tissue inflammation is a strong promoter for PDAC growth and progression in a genetically engineered mouse model of PDAC and diet-induced obesity. In this review, we will discuss the relationship between obesity-associated adipose tissue inflammation and PDAC development, with a focus on the key molecular and cellular components in the dysfunctional visceral adipose tissue, which provides a tumor permissive environment.
Collapse
|
45
|
Park Y, Peterson LL, Colditz GA. The Plausibility of Obesity Paradox in Cancer-Point. Cancer Res 2019; 78:1898-1903. [PMID: 29654151 DOI: 10.1158/0008-5472.can-17-3043] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
In contrast to the convincing evidence that obesity (measured by body mass index, BMI) increases the risk of many different types of cancer, there is an ambiguity in the role of obesity in survival among cancer patients. Some studies suggested that higher BMI decreased mortality risk in cancer patients, a phenomenon called the obesity paradox. The spurious positive association between BMI and cancer survival is likely to be explained by several methodologic limitations including confounding, reverse causation, and collider stratification bias. Also, the inadequacy of BMI as a measure of body fatness in cancer patients commonly experiencing changes in body weight and body composition may have resulted in the paradox. Other factors contributing to the divergent results in literature are significant heterogeneity in study design and method (e.g., study population, follow-up length); time of BMI assessment (pre-, peri-, or post-diagnosis); and lack of consideration for variability in the strength and directions of associations by age, sex, race/ethnicity, and cancer subtype. Robust but practical methods to accurately assess body fatness and body compositions and weight trajectories in cancer survivors are needed to advance this emerging field and to develop weight guidelines to improve both the length and the quality of cancer survival. Cancer Res; 78(8); 1898-903. ©2018 AACR.
Collapse
Affiliation(s)
- Yikyung Park
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri.
| | - Lindsay L Peterson
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Graham A Colditz
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
46
|
Duconseil P, Garnier J, Weets V, Ewald J, Marchese U, Gilabert M, Moureau-Zabotto L, Poizat F, Giovannini M, Delpero JR, Turrini O. Effect of clinical status on survival in patients with borderline or locally advanced pancreatic adenocarcinoma. World J Surg Oncol 2019; 17:95. [PMID: 31164144 PMCID: PMC6549256 DOI: 10.1186/s12957-019-1637-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
Objective To determine the effect of clinical status (weight variation and performance status [PS]) at diagnosis and during induction treatment on resectability and overall survival (OS) rates in patients with borderline resectable (BRPC) or locally advanced pancreatic cancer (LAPC). Methods From 2005 to 2017, 454 consecutive patients were diagnosed with LAPC or BRPC. We evaluated the PS (0–1 or 2–3), body mass index at diagnosis, and weight loss (WL) > 5% at initial staging and after induction treatment and separated continuous weight loss (CWL) from weight stabilization. Results A total of 294 patients (64.8%) presented with WL, and 57 patients (12.6%) presented with a PS of 2–3. At restaging, 60 patients (13.2%) presented with CWL. Independent factors that poorly influenced the OS were a PS of 2–3 at diagnosis (P < .01), CWL at restaging (P < .01), and absence of resection (P < .01). Factors independently impeding resection were LAPC (P < .01), PS > 1 at diagnosis (P < .01), and CWL (P = .01). In total, 142 patients (31.3%) underwent pancreatectomy. Independent factors that poorly influenced the OS in the resected group were PS > 0 at diagnosis (P = .01) and obesity (P < .01). For the 312 unresected cancer patients (68.7%), CWL (P < .01) was identified as an independent factor that poorly influenced the OS. Conclusion Clinical parameters that are easy to measure and monitor are independent factors of poor prognosis. The variation of weight during the induction treatment, more than WL at diagnosis, significantly precluded resection and was an independent factor of shorter OS in unresected patients.
Collapse
Affiliation(s)
- Pauline Duconseil
- Department of Surgical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France.
| | - Jonathan Garnier
- Department of Surgical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France
| | - Victoria Weets
- Department of Surgical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France
| | - Jacques Ewald
- Department of Surgical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France
| | - Ugo Marchese
- Department of Surgical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France
| | - Marine Gilabert
- Department of Oncology, Institut Paoli-Calmettes, Marseille, France
| | | | - Flora Poizat
- Department of Pathology, Institut Paoli-Calmettes, Marseille, France
| | - Marc Giovannini
- Department of Endoscopy, Institut Paoli-Calmettes, Marseille, France
| | - Jean-Robert Delpero
- Department of Surgical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France
| | - Olivier Turrini
- Department of Surgical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France
| |
Collapse
|
47
|
Seika P, Klein F, Pelzer U, Pratschke J, Bahra M, Malinka T. Influence of the body mass index on postoperative outcome and long-term survival after pancreatic resections in patients with underlying malignancy. Hepatobiliary Surg Nutr 2019; 8:201-210. [PMID: 31245400 DOI: 10.21037/hbsn.2019.02.05] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background While the long-term survival rate among patients with pancreatic and periampullary carcinomas remains low, it can be influenced by various factors. The purpose of this retrospective study was to investigate the effects of body mass index (BMI) on postoperative complications and patient survival after pancreatic resections for underlying malignancy over a 20-year observation period. Methods We analyzed 1,384 patients, 918 patients with pancreatic ductal adenocarcinoma (PDAC) (66.3%), 229 patients with distal cholangiocarcinoma (16.5%), 206 ampullary carcinoma patients (14.8%), and 31 duodenal carcinoma patients (2.2%). Patients were classified into four groups (group 1 <18.5; group 2, 18.5-25.0; group 3, 25.1-30.0; group 4 >30.0) according to their BMI (kg/m2). We analyzed differences in postoperative complications, postoperative length of hospital stays, reoperations, postoperative mortality and survival rate among the groups. Results Within a mean observation period of 687.7 [2-8,500] days, 735 (53.1%) patients died. There were important differences in postoperative complications (group 1, 16.2%; group 2, 20.3%; group 3, 27.2%, group 4, 41.6%) with the type of postoperative complications also varying between the groups. Overall 1-, 5-, 10- and 15-year survival rates were 66.4%, 25.5%, 17.9%, and 12.1%, respectively, with survival rates varying amongst the four groups. Conclusions Patients with a BMI between 18.5 and 30 show better postoperative outcomes, regarding complications, hospitalization duration, and reoperation rates than underweight or obese patients. Short-term survival depends strongly on postoperative complications while patients with a higher BMI show better long-term survival rates.
Collapse
Affiliation(s)
- Philippa Seika
- Department of Surgery, Charité Campus Mitte and Charité Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fritz Klein
- Department of Surgery, Charité Campus Mitte and Charité Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Pelzer
- Department of Hematology/Oncology/Tumorimmunology, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Charité Campus Mitte and Charité Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Bahra
- Department of Surgery, Charité Campus Mitte and Charité Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Malinka
- Department of Surgery, Charité Campus Mitte and Charité Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
48
|
Hendifar AE, Petzel MQB, Zimmers TA, Denlinger CS, Matrisian LM, Picozzi VJ, Rahib L. Pancreas Cancer-Associated Weight Loss. Oncologist 2019; 24:691-701. [PMID: 30591550 PMCID: PMC6516128 DOI: 10.1634/theoncologist.2018-0266] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/18/2018] [Indexed: 12/17/2022] Open
Abstract
Unintentional weight loss in patients with pancreatic cancer is highly prevalent and contributes to low therapeutic tolerance, reduced quality of life, and overall mortality. Weight loss in pancreatic cancer can be due to anorexia, malabsorption, and/or cachexia. Proper supportive care can stabilize or reverse weight loss in patients and improve outcomes. We review the literature on supportive care relevant to pancreatic cancer patients, and offer evidence-based recommendations that include expert nutritional assessment, counseling, supportive measures to ensure adequate caloric intake, pancreatic enzyme supplementation, nutritional supplement replacement, orexigenic agents, and exercise. Pancreatic Cancer Action Network-supported initiatives will spearhead the dissemination and adoption of these best supportive care practices. IMPLICATIONS FOR PRACTICE: Weight loss in pancreatic cancer patients is endemic, as 85% of pancreatic cancer patients meet the classic definition of cancer cachexia. Despite its significant prevalence and associated morbidity, there is no established approach to this disease entity. It is believed that this is due to an important knowledge gap in understanding the underlying biology and lack of optimal treatment approaches. This article reviews the literature regarding pancreas cancer-associated weight loss and establishes a new framework from which to view this complex clinical problem. An improved approach and understanding will help educate clinicians, improve clinical care, and provide more clarity for future clinical investigation.
Collapse
Affiliation(s)
| | | | - Teresa A Zimmers
- Indiana University, Simon Cancer Center, Indianapolis, Indiana, USA
| | | | - Lynn M Matrisian
- Pancreatic Cancer Action Network, Manhattan Beach, California, USA
| | | | - Lola Rahib
- Pancreatic Cancer Action Network, Manhattan Beach, California, USA
| |
Collapse
|
49
|
Luo G, Liu N. An integrative theory for cancer (Review). Int J Mol Med 2018; 43:647-656. [PMID: 30483756 PMCID: PMC6317675 DOI: 10.3892/ijmm.2018.4004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022] Open
Abstract
In the integrative theory, chronic irritations induce tumors with genetic alterations and rapid proliferative ability. Tumor cells reprogram the metabolism and employ aerobic glycolysis to sustain rapid growth. The host provides both the nutrients and exhaust system to support tumor growth via the tumor microenvironment. Under certain conditions, such as aging, diabetes, obesity and a high‑fat diet, the exhaust system is impaired, triggering a metabolic imbalance between the tumor and host. This is similar to a problematic car with an advanced motor with an out‑of‑date exhaust system. The metabolic imbalance causes a metabolic catastrophe, making tumor cells reside in a dismal environment and forcing them to invade, metastasize and undergo necrosis. Tumor necrosis, particularly in metastases, leads to non‑specific systemic inflammation, which is the major cause of cancer‑related mortality. On the whole, the integrative theory views cancer in an integrative manner and proposes that both genetic alterations and tumor‑host interaction as regards metabolism and immunology determine the destiny of the tumor and host. Although cancer is a genetic disease, tumor biology is basically the nature of the host.
Collapse
Affiliation(s)
- Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Na Liu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P.R. China
| |
Collapse
|
50
|
Murphy N, Jenab M, Gunter MJ. Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions. Nat Rev Gastroenterol Hepatol 2018; 15:659-670. [PMID: 29970888 DOI: 10.1038/s41575-018-0038-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Excess adiposity is a risk factor for several cancers of the gastrointestinal system, specifically oesophageal adenocarcinoma and colorectal, small intestine, pancreatic, liver, gallbladder and stomach cancers. With the increasing prevalence of obesity in nearly all regions of the world, this relationship could represent a growing source of cancers of the digestive system. Experimental and molecular epidemiological studies indicate important roles for alterations in insulin signalling, adipose tissue-derived inflammation and sex hormone pathways in mediating the association between adiposity and gastrointestinal cancer. The intestinal microbiome, gut hormones and non alcoholic fatty liver disease (NAFLD) also have possible roles. However, important gaps remain in our knowledge. For instance, our understanding of how adiposity throughout the life course is related to the risk of gastrointestinal cancer development and of how obesity influences gastrointestinal cancer prognosis and survival is limited. Nonetheless, the increasing use of state-of-the-art analytical methods (such as omics technologies, Mendelian randomization and MRI) in large-scale epidemiological studies offers exciting opportunities to advance our understanding of the complex relationship between adiposity and gastrointestinal cancers. Here, we examine the epidemiology of associations between obesity and gastrointestinal cancer, explore potential mechanisms underlying these relationships and highlight important unanswered research questions.
Collapse
Affiliation(s)
- Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Mazda Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France.
| |
Collapse
|