1
|
Brown M, O'Connor D, Turkington R, Eatock M, Vince R, Hulme C, Bowdery R, Robinson R, Wadsley J, Maraveyas A, Prue G. Feasibility of delivering supervised exercise training following surgical resection and during adjuvant chemotherapy for pancreatic ductal adenocarcinoma (PRECISE): a case series. BMC Sports Sci Med Rehabil 2023; 15:116. [PMID: 37735664 PMCID: PMC10514993 DOI: 10.1186/s13102-023-00722-3] [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: 04/17/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is an aggressive neoplasm, with surgical resection and adjuvant chemotherapy the only curative treatment. Treatment-related toxicities place a considerable burden on patients although exercise training has shown promise is helping to manage such adversities and facilitate rehabilitation. The feasibility and safety of exercise training as a supportive therapy during adjuvant chemotherapy remains unknown. METHODS Patients with PDAC were screened post-surgical resection and enrolled in a 16-week, progressive, concurrent exercise programme alongside their chemotherapy regimen. Feasibility was the primary objective detailing recruitment, retention and adherence rates throughout as well as the safety and fidelity of the intervention. Secondarily, the impact on functional fitness and patient-reported outcomes was captured at baseline, post-intervention and 3-month follow up. RESULTS Eight patients consented to participate in this trial, with five proceeding to enrol in exercise training. Concurrent exercise training is feasible and safe during adjuvant chemotherapy and prevented an expected decline in functional fitness and patient-reported outcomes during this time. DISCUSSION This case series provides preliminary evidence that concurrent exercise training during adjuvant therapy is safe, feasible and well tolerated, preventing an expected decline in functional fitness, muscular strength and health-related quality of life (HRQoL). Given the adverse effects of treatment, these findings are promising and provide further evidence for the inclusion of exercise training as a standard of care for surgical rehabilitation and managing treatment-related toxicities. Future research should explore the impact of exercise training during neoadjuvant chemotherapy, with prehabilitation now standard practice for borderline resectable disease. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04305067, prospectively registered 12/03/2020, https://classic. CLINICALTRIALS gov/ct2/show/NCT04305067 .
Collapse
Affiliation(s)
- Malcolm Brown
- School of Nursing and Midwifery, Queen's University Belfast Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Dominic O'Connor
- School of Health Sciences, The University of Nottingham, Nottingham, England, UK
| | - Richard Turkington
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK
- The Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
| | - Martin Eatock
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK
- The Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, Northern Ireland, UK
| | - Rebecca Vince
- School of Sport, Exercise and Rehabilitation Sciences, University of Hull, Hull, England, UK
| | - Claire Hulme
- Department of Health and Community Sciences, University of Exeter Medical School, Exeter, England, UK
| | - Roy Bowdery
- Pancreatic Cancer UK Research Involvement Network, London, England, UK
| | - Rebecca Robinson
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, England, UK
| | - Jonathan Wadsley
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, England, UK
| | | | - Gillian Prue
- School of Nursing and Midwifery, Queen's University Belfast Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| |
Collapse
|
2
|
Park LK, Lim KH, Volkman J, Abdiannia M, Johnston H, Nigogosyan Z, Siegel MJ, McGill JB, McKee AM, Salam M, Zhang RM, Ma D, Popuri K, Chow VTY, Beg MF, Hawkins WG, Peterson LR, Ippolito JE. Safety, tolerability, and effectiveness of the sodium-glucose cotransporter 2 inhibitor (SGLT2i) dapagliflozin in combination with standard chemotherapy for patients with advanced, inoperable pancreatic adenocarcinoma: a phase 1b observational study. Cancer Metab 2023; 11:6. [PMID: 37202813 DOI: 10.1186/s40170-023-00306-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/02/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy. Thus, there is an urgent need for safe and effective novel therapies. PDAC's excessive reliance on glucose metabolism for its metabolic needs provides a target for metabolic therapy. Preclinical PDAC models have demonstrated that targeting the sodium-glucose co-transporter-2 (SGLT2) with dapagliflozin may be a novel strategy. Whether dapagliflozin is safe and efficacious in humans with PDAC is unclear. METHODS We performed a phase 1b observational study (ClinicalTrials.gov ID NCT04542291; registered 09/09/2020) to test the safety and tolerability of dapagliflozin (5 mg p.o./day × 2 weeks escalated to 10 mg p.o./day × 6 weeks) added to standard Gemcitabine and nab-Paclitaxel (GnP) chemotherapy in patients with locally advanced and/or metastatic PDAC. Markers of efficacy including Response Evaluation Criteria in Solid Tumors (RECIST 1.1) response, CT-based volumetric body composition measurements, and plasma chemistries for measuring metabolism and tumor burden were also analyzed. RESULTS Of 23 patients who were screened, 15 enrolled. One expired (due to complications from underlying disease), 2 dropped out (did not tolerate GnP chemotherapy) during the first 4 weeks, and 12 completed. There were no unexpected or serious adverse events with dapagliflozin. One patient was told to discontinue dapagliflozin after 6 weeks due to elevated ketones, although there were no clinical signs of ketoacidosis. Dapagliflozin compliance was 99.4%. Plasma glucagon increased significantly. Although abdominal muscle and fat volumes decreased; increased muscle-to-fat ratio correlated with better therapeutic response. After 8 weeks of treatment in the study, partial response (PR) to therapy was seen in 2 patients, stable disease (SD) in 9 patients, and progressive disease (PD) in 1 patient. After dapagliflozin discontinuation (and chemotherapy continuation), an additional 7 patients developed the progressive disease in the subsequent scans measured by increased lesion size as well as the development of new lesions. Quantitative imaging assessment was supported by plasma CA19-9 tumor marker measurements. CONCLUSIONS Dapagliflozin is well-tolerated and was associated with high compliance in patients with advanced, inoperable PDAC. Overall favorable changes in tumor response and plasma biomarkers suggest it may have efficacy against PDAC, warranting further investigation.
Collapse
Affiliation(s)
- Lauren K Park
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA
| | - Kian-Huat Lim
- Department of Medicine, Oncology Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jonas Volkman
- Department of Medicine, Oncology Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Mina Abdiannia
- Department of Medicine, Oncology Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Hannah Johnston
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA
| | - Zack Nigogosyan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA
| | - Marilyn J Siegel
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA
| | - Janet B McGill
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Alexis M McKee
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Maamoun Salam
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Rong M Zhang
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Da Ma
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - William G Hawkins
- Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Linda R Peterson
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA.
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA.
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
3
|
Paiella S, Azzolina D, Trestini I, Malleo G, Nappo G, Ricci C, Ingaldi C, Vacca PG, De Pastena M, Secchettin E, Zamboni G, Maggino L, Corciulo MA, Sandini M, Cereda M, Capretti G, Casadei R, Bassi C, Mansueto G, Gregori D, Milella M, Zerbi A, Gianotti L, Salvia R. Body composition parameters, immunonutritional indexes, and surgical outcome of pancreatic cancer patients resected after neoadjuvant therapy: A retrospective, multicenter analysis. Front Nutr 2023; 10:1065294. [PMID: 36860690 PMCID: PMC9968808 DOI: 10.3389/fnut.2023.1065294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Background and aims Body composition parameters and immunonutritional indexes provide useful information on the nutritional and inflammatory status of patients. We sought to investigate whether they predict the postoperative outcome in patients with pancreatic cancer (PC) who received neoadjuvant therapy (NAT) and then pancreaticoduodenectomy. Methods Data from locally advanced PC patients who underwent NAT followed by pancreaticoduodenectomy between January 2012 and December 2019 in four high-volume institutions were collected retrospectively. Only patients with two available CT scans (before and after NAT) and immunonutritional indexes (before surgery) available were included. Body composition was assessed and immunonutritional indexes collected were: VAT, SAT, SMI, SMA, PLR, NLR, LMR, and PNI. The postoperative outcomes evaluated were overall morbidity (any complication occurring), major complications (Clavien-Dindo ≥ 3), and length of stay. Results One hundred twenty-one patients met the inclusion criteria and constituted the study population. The median age at the diagnosis was 64 years (IQR16), and the median BMI was 24 kg/m2 (IQR 4.1). The median time between the two CT-scan examined was 188 days (IQR 48). Skeletal muscle index (SMI) decreased after NAT, with a median delta of -7.8 cm2/m2 (p < 0.05). Major complications occurred more frequently in patients with a lower pre-NAT SMI (p = 0.035) and in those who gained in subcutaneous adipose tissue (SAT) compartment during NAT (p = 0.043). Patients with a gain in SMI experienced fewer major postoperative complications (p = 0.002). The presence of Low muscle mass after NAT was associated with a longer hospital stay [Beta 5.1, 95%CI (1.5, 8.7), p = 0.006]. An increase in SMI from 35 to 40 cm2/m2 was a protective factor with respect to overall postoperative complications [OR 0.43, 95% (CI 0.21, 0.86), p < 0.001]. None of the immunonutritional indexes investigated predicted the postoperative outcome. Conclusion Body composition changes during NAT are associated with surgical outcome in PC patients who receive pancreaticoduodenectomy after NAT. An increase in SMI during NAT should be favored to ameliorate the postoperative outcome. Immunonutritional indexes did not show to be capable of predicting the surgical outcome.
Collapse
Affiliation(s)
- Salvatore Paiella
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy,*Correspondence: Salvatore Paiella ✉
| | - Danila Azzolina
- Department of Environmental and Preventive Science, University of Ferrara, Ferrara, Italy
| | - Ilaria Trestini
- Dietetics Services, Hospital Medical Direction, University Hospital Trust of Verona, Verona, Italy
| | - Giuseppe Malleo
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gennaro Nappo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy,IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Claudio Ricci
- Pancreatic Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, University of Bologna, Bologna, Italy,Department of Internal Medicine and Surgery (DIMEC), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Carlo Ingaldi
- Pancreatic Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, University of Bologna, Bologna, Italy,Department of Internal Medicine and Surgery (DIMEC), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Pier Giuseppe Vacca
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Matteo De Pastena
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Erica Secchettin
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Giulia Zamboni
- Radiology Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Laura Maggino
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Maria Assunta Corciulo
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Marta Sandini
- General Surgery Unit, University of Siena, Siena, Italy
| | - Marco Cereda
- School of Medicine and Surgery, University of Milano-Bicocca and HPB Unit, San Gerardo Hospital Monza, Monza, Italy
| | - Giovanni Capretti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy,IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Riccardo Casadei
- Pancreatic Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, University of Bologna, Bologna, Italy,Department of Internal Medicine and Surgery (DIMEC), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Claudio Bassi
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | | | - Dario Gregori
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona Hospital Trust, Verona, Italy
| | - Alessandro Zerbi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy,IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Luca Gianotti
- School of Medicine and Surgery, University of Milano-Bicocca and HPB Unit, San Gerardo Hospital Monza, Monza, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy,Roberto Salvia ✉
| |
Collapse
|
4
|
Gunesch AN, Sutton TL, Krasnow SM, Deig CR, Sheppard BC, Marks DL, Grossberg AJ. Validation of automated body composition analysis using diagnostic computed tomography imaging in patients with pancreatic cancer. Am J Surg 2022; 224:742-746. [PMID: 35396132 PMCID: PMC9308682 DOI: 10.1016/j.amjsurg.2022.03.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/19/2022] [Accepted: 03/22/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Sarcopenia is associated with complications and inferior oncologic outcomes in solid tumors. Axial computed tomography (CT) scans can be used to evaluate sarcopenia, however manual quantification is laborious. We sought to validate an automated method of quantifying muscle cross-sectional area (CSA) in patients with pancreatic adenocarcinoma (PDAC). METHODS Mid-L3 CT images from patients with PDAC were analyzed: CSAs of skeletal muscle (SM) were measured using manual segmentation and the software AutoMATiCA, and then compared with linear regression. RESULTS Five-hundred-twenty-five unique scans were analyzed. There was robust correlation between manual and automated segmentation for L3 CSA (R2 0.94, P < 0.001). Bland-Altman analysis demonstrated a consistent overestimation of muscle CSA by AutoMATiCA with a mean difference of 5.7%. A correction factor of 1.06 was validated using a unique test dataset of 36 patients with non-PDAC peripancreatic malignancies. CONCLUSIONS Automated muscle CSA measurement with AutoMATiCA is highly efficient and yields results highly correlated with manual measurement. These findings support the potential use of high-throughput sarcopenia analysis with abdominal CT scans for both clinical and research purposes.
Collapse
Affiliation(s)
- Ali N Gunesch
- School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | | | | | | | | | - Daniel L Marks
- Department of Pediatrics, OHSU, Portland, OR, 97239, USA; Brenden Colson Center for Pancreatic Care, OHSU, Portland, OR, 97239, USA
| | - Aaron J Grossberg
- Brenden Colson Center for Pancreatic Care, OHSU, Portland, OR, 97239, USA; Department of Radiation Medicine, OHSU, Portland, OR, 97239, USA.
| |
Collapse
|
5
|
Capurso G, Pecorelli N, Burini A, Orsi G, Palumbo D, Macchini M, Mele R, de Cobelli F, Falconi M, Arcidiacono PG, Reni M. The impact of nutritional status on pancreatic cancer therapy. Expert Rev Anticancer Ther 2022; 22:155-167. [PMID: 34989653 DOI: 10.1080/14737140.2022.2026771] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Pancreatic Ductal Adenocarcinoma (PDAC) is an aggressive disease with poor outcomes. One of the reasons for the dismal prognosis resides in its impressive ability to alter the nutritional status of patients who develop malnutrition, cachexia, anorexia, and sarcopenia in most cases. The ideal way to measure such changes in PDAC patients, in order to readily identify them and avoid complications or discontinuations of treatment is a relatively unexplored area. In addition, most PDAC patients experience pancreatic exocrine insufficiency (PEI) that contributes to the complex puzzle of malnutrition and that can be treated with Pancreatic Enzyme Replacement Therapy (PERT). AREAS COVERED We review current knowledge on the impact of nutritional status on both surgical and medical treatments for PDAC, reporting available data on the causes of malnutrition, characteristics, and advantages of different tools to investigate nutritional status and possible strategies to improve patient outcomes. EXPERT OPINION All PDAC patients should receive a careful nutritional assessment at diagnosis, and this should be repeated alongside their treatment path. Screening tools and biochemical variables or scores are associated with prognosis, but bioimpedance vector analysis (BIVA) and radiological assessment of body composition seem more accurate in predicting clinical outcomes and postoperative complications.
Collapse
Affiliation(s)
- Gabriele Capurso
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy & Endosonography Division, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Nicolò Pecorelli
- Pancreas Translational & Clinical Research Center, Division of Pancreatic Surgery, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Alice Burini
- Nutrition Service, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Giulia Orsi
- Pancreas Translational & Clinical Research Center, Oncology Department, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Diego Palumbo
- Pancreas Translational & Clinical Research Center, Department of Radiology & Center for Experimental Imaging, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Marina Macchini
- Pancreas Translational & Clinical Research Center, Oncology Department, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Roberto Mele
- Nutrition Service, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Francesco de Cobelli
- Pancreas Translational & Clinical Research Center, Department of Radiology & Center for Experimental Imaging, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- Pancreas Translational & Clinical Research Center, Division of Pancreatic Surgery, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy & Endosonography Division, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Michele Reni
- Pancreas Translational & Clinical Research Center, Oncology Department, San Raffaele Scientific Institute IRCCS, Milan, Italy
| |
Collapse
|
6
|
Sinicrope FA, Shi Q, Smyrk TC, Goldberg RM, Cohen SJ, Gill S, Kahlenberg MS, Nair S, Shield AF, Jahagirdar BN, Jacobson SB, Foster NR, Pollak MN, Alberts SR. Association of Adiponectin and Vitamin D With Tumor Infiltrating Lymphocytes and Survival in Stage III Colon Cancer. JNCI Cancer Spectr 2021; 5:pkab070. [PMID: 34485815 PMCID: PMC8410141 DOI: 10.1093/jncics/pkab070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/03/2021] [Accepted: 07/21/2021] [Indexed: 12/14/2022] Open
Abstract
Background Adipocyte-derived adiponectin may play a role in the host inflammatory response to cancer. We examined the association of plasma adiponectin with the density of tumor-infiltrating lymphocytes (TILs) in colon cancers and with vitamin D, clinicopathological features, and patient survival. Methods Plasma adiponectin and 25-hydroxyvitamin D [25(OH)D] were analyzed by radioimmunoassay in 600 patients with stage III colon cancer who received FOLFOX-based adjuvant chemotherapy (NCCTG N0147 [Alliance]). TIL densities were determined in histopathological sections. Associations with disease-free survival (DFS), time to recurrence, and overall survival were evaluated by multivariable Cox regression adjusting for potential confounders (ie, body mass index, race, TILs, and N stage). All statistical tests were 2-sided. Results We found a statistically significant reduction in adiponectin, but not 25(OH)D, levels in tumors with high vs low TIL densities (median = 6845 vs 8984 ng/mL; P = .04). A statistically significant reduction in adiponectin was also observed in obese (body mass index >30 kg/m2) vs nonobese patients (median = 6608 vs 12 351 ng/mL; P < .001), in men vs women (median = 8185 vs 11 567 ng/mL; P < .001), in Blacks vs Whites or Asians (median = 6412 vs 8847 vs 7858 ng/mL; P < .03), and in those with fewer lymph node metastases (N1 vs N2: median = 7768 vs 9253 ng/mL; P = .01). Insufficiency of 25(OH)D (<30 ng/mL) was detected in 291 (48.5%) patients. In multivariable analyses, neither adiponectin nor 25(OH)D were associated with a statistically significant difference in DFS, overall survival , or time to recurrence in models adjusted for potential confounders. We found a statistically significant association of TILs with prognosis, yet no such interaction was observed for the association of adiponectin with TILs for DFS. Conclusions Lower circulating adiponectin levels were associated with a statistically significant increase in TIL densities in colon cancers, indicating an enhanced antitumor immune response. In contrast to TILs, neither adiponectin nor 25(OH)D was independently prognostic.
Collapse
Affiliation(s)
- Frank A Sinicrope
- Division of Oncology and Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| | - Qian Shi
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Thomas C Smyrk
- Division of Oncology and Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| | | | | | - Sharlene Gill
- British Columbia Cancer Agency, Vancouver Cancer Centre, Vancouver, BC, Canada
| | | | | | - Anthony F Shield
- Wayne State University, Karmanos Cancer Institute, Detroit, MI, USA
| | | | - Sawyer B Jacobson
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Nathan R Foster
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | | | - Steven R Alberts
- Division of Oncology and Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| |
Collapse
|