1
|
Gallois C, Sroussi M, André T, Mouillet-Richard S, Agueeff N, Mulot C, Vernerey D, Louvet C, Bachet JB, Dourthe LM, Mazard T, Jary M, Coutzac C, Lecaille C, Tabernero J, Van Laethem JL, Lepage C, Emile JF, de Reyniès A, Taieb J, Laurent-Puig P. Prognostic Models From Transcriptomic Signatures of the Tumor Microenvironment and Cell Cycle in Stage III Colon Cancer From PETACC-8 and IDEA-France Trials. J Clin Oncol 2025:JCO2302262. [PMID: 39889251 DOI: 10.1200/jco.23.02262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 02/02/2025] Open
Abstract
PURPOSE The objective of this work was to establish prognostic models in stage III colon cancer (CC) on the basis of transcriptomic signatures of the tumor microenvironment (TME) and cell cycle from the PETACC-8 (training set) and IDEA-France (validation set) trials. PATIENTS AND METHODS 3'RNA sequencing was performed in 1,733 patients from the PETACC-8 trial and 1,248 patients from the IDEA-France trial. Four transcriptomic signatures were analyzed: T-cell and macrophage M2 signatures, the expression of CXCL13, and a score on the basis of the Oncotype DX CC Recurrence Score using the same formula from the stromal score and the cell cycle score. The Immune Proliferative Stromal (IPS) score was defined as the number of dichotomized signatures that fall under the category of a dismal prognosis (from 0 to 4). Time to recurrence (TTR) was defined as the time from the date of random assignment to local and/or metastatic relapse and/or death because of CC, whichever occurs first. RESULTS High Oncotype-like and M2 scores and low CXCL13 expression and T-cell score were associated with a shorter TTR. A multivariable model including these signatures and all known prognostic factors applied to the IDEA-France cohort by obtaining a value of this model for each patient showed TTR significantly different depending on the quartile of this value and a 3-year rate of patients without recurrence ranging from 56% for the lowest quartile to 89% for the highest quartile (P < .0001). The IPS score was significantly associated with TTR in multivariable analysis. CONCLUSION Using transcriptomic data of patients with stage III CC from two large-scale adjuvant trials, a prognostic model on the basis of signatures of the TME and the cell cycle provides important information in addition to known prognostic factors for patient stratification on risk of recurrence.
Collapse
Affiliation(s)
- Claire Gallois
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Digestive Oncology Department, Institut du Cancer Paris CARPEM, APHP, APHP Centre, Hôpital Européen G. Pompidou, Université Paris Cité, Paris, France
| | - Marine Sroussi
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Institut Chimie Biologie Innovation-Laboratoire de BioChimie, ESPCI, UMR8231 CNRS, Université PSL, Paris, France
| | - Thierry André
- Department of Medical Oncology, Hôpital Saint Antoine, Sorbonne Université, Paris, France
| | - Sophie Mouillet-Richard
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| | - Natacha Agueeff
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University of Besançon, Besançon, France
- Etablissement Français du Sang Bourgogne Franche-Comté, INSERM, Unité Mixte de Recherche 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Franche-Comté University, Besançon University Hospital, Besançon, France
| | - Christophe Louvet
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - Jean-Baptiste Bachet
- Department of Hepato-Gastroenterology and Digestive Oncology, Pitié-Salpêtriére Hospital, APHP, Sorbonne Université, Paris, France
| | | | - Thibault Mazard
- Department of Medical Oncology, IRCM, INSERM, ICM, University of Montpellier, Montpellier, France
| | - Marine Jary
- Department of Digestive and Hepatobiliary Surgery, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Clélia Coutzac
- Department of Medical Oncology, Centre Leon Berard, University Claude Bernard Lyon, Lyon, France
| | - Cédric Lecaille
- Cancerology Department, Bordeaux Nord Polyclinic, Bordeaux, France
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Jean-Luc Van Laethem
- Department of Digestive Oncology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Côme Lepage
- Hepatogastroenterology and Digestive Oncology Department, Dijon Bourgogne Hospital, University of Burgundy and Franche Comté, Dijon, France
| | - Jean-François Emile
- EA4340 BECCOH, Service de Pathologie, Hôpital Ambroise Paré, AP-HP, Université de Versailles SQY, Boulogne, France
| | - Aurélien de Reyniès
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Laboratoire SeqOIA, Paris, France
| | - Julien Taieb
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Digestive Oncology Department, Institut du Cancer Paris CARPEM, APHP, APHP Centre, Hôpital Européen G. Pompidou, Université Paris Cité, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| |
Collapse
|
2
|
Chia JWK, Segelov E, Deng Y, Ho GF, Wang W, Han S, Sharma A, Ding K, Chen G, Jeffery MG, Tham CK, Ahn JB, Nott L, Zielinski R, Chao TY, van Hagen T, Wei PL, Day F, Mehta S, Yau T, Peng J, Hayes TM, Li Y, Gandhi M, Foo EMJ, Rahman N, Rothwell P, Ali R, Simes J, Toh HC. Aspirin after completion of standard adjuvant therapy for colorectal cancer (ASCOLT): an international, multicentre, phase 3, randomised, double-blind, placebo-controlled trial. Lancet Gastroenterol Hepatol 2025:S2468-1253(24)00387-X. [PMID: 39824200 DOI: 10.1016/s2468-1253(24)00387-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND Aspirin is a simple, globally available medication that has been shown to reduce the incidence of colorectal cancer. We aimed to evaluate the safety and efficacy of aspirin in the secondary prevention of colorectal cancer. METHODS This phase 3, randomised, double-blind, placebo-controlled trial was conducted at 66 centres across 11 countries and territories (ten in Asia-Pacific; one in the Middle East). The trial included patients aged 18 years and older with Dukes' C or high-risk Dukes' B colon cancer or Dukes' B or C rectal cancer who had undergone resection and had completed standard adjuvant therapy (at least 3 months of chemotherapy). Patients with contraindications to aspirin, familial syndromes of colorectal cancer, recent other cancers, and clinically significant history of cardiovascular disease or stroke were excluded. Patients were randomly assigned (1:1) to aspirin 200 mg daily or placebo for 3 years, and were followed up for 5 years. Randomisation was stratified by study centre, tumour site and stage, and inclusion of oxaliplatin in adjuvant chemotherapy. The patients, study team, and sponsor were masked to treatment assignment. The primary endpoint was disease-free survival. The primary analysis used a stratified Cox model in those commencing study treatment (modified intention-to-treat population), analysing all events to March 31, 2023. Safety was analysed in the same population. This trial is registered at ClinicalTrials.gov (NCT00565708). The primary analysis has been completed, but translational studies of putative aspirin sensitivity biomarkers are ongoing. FINDINGS Between Feb 25, 2009, and June 30, 2021, 1587 patients underwent randomisation, of whom 1550 were included in the modified intention-to-treat analysis: 791 (51%) in the aspirin group and 759 (49%) in the placebo group. Of these patients, the median age was 57 years (IQR 48-65); 897 (58%) were male and 653 (42%) female; 271 (17%) had Dukes' B colon cancer, 770 (50%) Dukes' C colon cancer, and 509 (33%) rectal cancer. Median follow-up at data cutoff was 59·2 months (IQR 36·7-60·0). 5-year disease-free survival was 77·0% (95% CI 73·6-80·0) in the aspirin group and 74·8% (71·3-77·9) in the placebo group (hazard ratio of 0·91 [95% CI 0·73-1·13]; p=0·38). Any-grade adverse events were reported in 390 (49%) of 791 patients in the aspirin group versus 386 (51%) of 759 in the placebo group. Serious adverse events were reported in 95 (12%) patients in the aspirin group versus 107 (14%) in the placebo group. There were no treatment-related deaths in either group. Among adverse events of special interest, there were no cases of acute myocardial infarction in the aspirin group versus two in the placebo group; no ischaemic cerebrovascular events in the aspirin group versus two in the placebo group; and three major gastrointestinal bleeds in the aspirin group versus one in the placebo group. INTERPRETATION In patients with colorectal cancer, aspirin 200 mg daily for 3 years after completion of standard adjuvant therapy was well tolerated but did not significantly improve disease-free survival. FUNDING SingHealth Foundation, National Medical Research Council Singapore, National Cancer Centre Research Fund, Rising Tide Foundation, Lee Foundation, Lee Kim Tah Foundation, Duke-NUS Khoo Bridge Funding Award, Terry-Fox Run, Silent Foundation, Cancer Australia, Bowel Cancer Australia, and Cancer Council NSW.
Collapse
Affiliation(s)
- John W K Chia
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Eva Segelov
- Department of Clinical Research and Department of Radiation Oncology, Faculty of Medicine, University of Bern, Bern, Switzerland; Australasian Gastro-Intestinal Trials Group (AGITG), GI Cancer Institute @Lifehouse, Camperdown, NSW, Australia
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gwo Fuang Ho
- University Malaya Medical Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Wei Wang
- The First People's Hospital of Foshan, Foshan, China
| | - Shuting Han
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Atul Sharma
- Department of Medical Oncology, AIIMS, New Delhi, India
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gong Chen
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Mark G Jeffery
- Australasian Gastro-Intestinal Trials Group (AGITG), GI Cancer Institute @Lifehouse, Camperdown, NSW, Australia; Oncology Service, Christchurch Hospital, Christchurch, New Zealand
| | - Chee Kian Tham
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Joong Bae Ahn
- Division of Medical Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Louise Nott
- Tasmanian Health Service, Royal Hobart Hospital, Hobart, TAS, Australia
| | - Robert Zielinski
- Central West Cancer Care Centre, Orange Base Hospital, Orange, NSW, Australia; Western Sydney University, Sydney, NSW, Australia
| | - Tsu-Yi Chao
- Hematology & Oncology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Tom van Hagen
- St John of God Subiaco Hospital, Subiaco, WA, Australia
| | - Po-Li Wei
- Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Fiona Day
- Australasian Gastro-Intestinal Trials Group (AGITG), GI Cancer Institute @Lifehouse, Camperdown, NSW, Australia; Medical Oncology, Calvary Mater Newcastle and University of Newcastle, Newcastle, NSW, Australia
| | - Shaesta Mehta
- Department of Digestive Diseases and Clinical Nutrition Member, Disease Management Group - Gastrointestinal Oncology, Tata Memorial Hospital, Mumbai, India
| | - Thomas Yau
- Department of Medicine, Queen Mary Hospital and Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jiewen Peng
- Department of Medical Oncology, Zhongshan City People's Hospital, Zhongshan, China
| | - Theresa M Hayes
- South West Oncology, South West Regional Cancer Centre, Warrnambool, VIC, Australia
| | - Yong Li
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, China
| | - Mihir Gandhi
- Department of Biostatistics, Singapore Clinical Research Institute, Singapore; Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Estelle M J Foo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Nabilah Rahman
- Department of Biostatistics, Singapore Clinical Research Institute, Singapore; Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Peter Rothwell
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Raghib Ali
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK; Public Health Research Centre, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - John Simes
- Australasian Gastro-Intestinal Trials Group (AGITG), GI Cancer Institute @Lifehouse, Camperdown, NSW, Australia; NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
3
|
Seligmann JF, Morton D, Elliott F, Handley K, Gray R, Seymour M, Glimelius B, Magill L, Williams CJM, Quirke P, Bottomley D, Wood HM, Murakami K, Beggs AD, West NP. Neo-adjuvant FOLFOX with and without panitumumab for patients with KRAS-wt locally advanced colon cancer: results following an extended biomarker panel on the FOxTROT Trial embedded phase II population. Ann Oncol 2025:S0923-7534(25)00002-X. [PMID: 39805350 DOI: 10.1016/j.annonc.2024.12.013] [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: 11/07/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The FOxTROT trial has reported advantages of neoadjuvant chemotherapy (NAC) in locally advanced colon cancer (LACC). Here we present results of the embedded randomized phase II trial testing the addition of panitumumab to neoadjuvant FOLFOX compared with FOLFOX alone in RAS and BRAF-wild-type patients and with biomarker hyperselction. PATIENTS AND METHODS Patients had operable, CT-predicted stage T3-4, N0-2, M0 colon adenocarcinoma. KRAS-wt pts allocated to NAC could optionally be sub-randomized 1:1 to FOLFOX ± panitumumab during the pre-operative phase. RAS/BRAF were tested by NGS; EREG/AREG by RNAseq. Primary endpoint is time to recurrence (TTR) in RAS/BRAF-wt patients; secondary endpoints include safety, histological downstaging, disease-free survival (DFS), colon cancer-specific survival (CCSS), overall survival (OS), and impact of primary tumor location and EREG/AREG. RESULTS In total 269 KRAS-wt patients were enrolled into the embedded phase II trial. Extended RAS/BRAF data were available for 232 (83%) patients; 22/232(9.5%) were RAS-mutant; 41/210(20%) were BRAF-mutant. Median follow up was 42 months. In 169 RAS/BRAF-wt patients there was a trend towards reduced recurrences with FOLFOX plus panitumumab compared with FOLFOX (12% vs 21%, HR=0.51, p=0.09); significant improvements were seen for DFS, CCSS and OS. Within the hyperselected EREG/AREG high group, there was significant reduction in recurrences with panitumumab. Panitumumab was not associated with increased pathological regression of the primary tumor (TRG 1-3 16% vs 22%,p=0.27). FOLFOX plus panitumumab was associated with higher rates of grade 3 diarrhoea (8% vs 3%,) and rash (22% vs 2%). CONCLUSION This exploratory analysis from a randomized phase II study shows a non-significant improvement in TTR from the addition of neoadjuvant panitumumab to peri-operative FOLFOX in RAS/BRAF-wt LACC. Hyperselection with EREG/AREG status was associated with increased efficacy. A dedicated prospective trial within a biomarker selected population is under development. CLINICAL TRIALS REGISTRATION ISRCTN87163246.
Collapse
Affiliation(s)
- J F Seligmann
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, UK.
| | - D Morton
- University Hospital Birmingham, Birmingham, UK
| | - F Elliott
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, UK
| | - K Handley
- University of Birmingham Clinical Trials Unit, Birmingham, UK
| | - R Gray
- Clinical Trials Service Unit, University of Oxford, Oxford, UK
| | - M Seymour
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, UK
| | | | - L Magill
- University of Birmingham Clinical Trials Unit, Birmingham, UK
| | - C J M Williams
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, UK
| | - P Quirke
- Division of Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, UK
| | - D Bottomley
- Division of Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, UK
| | - H M Wood
- Division of Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, UK
| | - K Murakami
- Department of Pathology, Tohoku University, Sendai, Japan
| | - A D Beggs
- Institute of Cancer & Genomic Sciences, University of Birmingham, UK
| | - N P West
- Division of Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, UK
| |
Collapse
|
4
|
van den Berg K, van Hellemond IEG, Willems JMWE, Burger JWA, Rutten HJT, Creemers GJ. Neoadjuvant chemotherapy in locally advanced colon cancer: A systematic review with proportional meta-analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 51:109560. [PMID: 39869958 DOI: 10.1016/j.ejso.2024.109560] [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: 08/28/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 01/29/2025]
Abstract
Neoadjuvant chemotherapy is suggested in locally advanced colon cancer. Data on improved long-term oncological outcomes are lacking, which hampers the implementation in clinical practice. This systematic review provides an overview of the benefits and drawbacks of neoadjuvant chemotherapy in patients with locally advanced colon cancer. A systematic literature search was performed using Embase (OVID), MEDLINE (OvidSP), and the Cochrane Library. Studies reporting on the efficacy of neoadjuvant chemotherapy in patients with operable, locally advanced colon cancer without metastases at the time of diagnosis were considered eligible for inclusion. An overview of short- and long-term outcomes of neoadjuvant chemotherapy is provided based on available literature. Additionally, proportional meta-analyses were performed using MedCalc Statistical Software version 19.2.6. A total of 17 unique studies were included in this review, 3 randomised controlled trials and 14 prospective single-arm or retrospective studies. The maximum reported dropout before surgery was 7.8 % in the neoadjuvant chemotherapy group. A histopathological complete response after neoadjuvant chemotherapy was observed in 0-4.8 % of the patients. The occurrence of anastomotic leaks was less than 8 % for both patients treated with neoadjuvant chemotherapy and patients treated with upfront surgery. Neoadjuvant chemotherapy is a safe alternative for adjuvant chemotherapy based on the dropout rate before surgery and the peri-operative morbidity and peri-operative mortality. Robust long-term survival outcomes are lacking and serious concerns regarding the risk of overtreatment have been expressed. Hence, neoadjuvant chemotherapy might be considered in a select group of patients with locally advanced colon cancer.
Collapse
Affiliation(s)
- K van den Berg
- Department of Medical Oncology, Catharina Hospital, Eindhoven, Netherlands; Department of Surgery, Catharina Hospital, Eindhoven, Netherlands.
| | | | - J M W E Willems
- Department of Medical Oncology, Anna Hospital, Geldrop, Netherlands
| | - J W A Burger
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - H J T Rutten
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - G J Creemers
- Department of Medical Oncology, Catharina Hospital, Eindhoven, Netherlands
| |
Collapse
|
5
|
Samaille T, Falcoz A, Cohen R, Laurent-Puig P, André T, Taieb J, Auclin E, Vernerey D. A novel risk classification model integrating CEA, ctDNA, and pTN stage for stage 3 colon cancer: a post hoc analysis of the IDEA-France trial. Oncologist 2024; 29:e1492-e1500. [PMID: 39011625 PMCID: PMC11546770 DOI: 10.1093/oncolo/oyae140] [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: 02/02/2024] [Accepted: 05/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND We assessed the added value of incorporating carcinoembryonic antigen (CEA) to circulating tumor DNA (ctDNA) and pathological TN (pTN) stage for risk classification in stage 3 colon cancer (CC). PATIENTS AND METHODS We retrospectively analyzed postoperative CEA values in patients with CC from the IDEA-France phase 3 trial. The relation between disease-free survival (DFS) and CEA was modeled through restricted cubic splines. Prognostic value of CEA, ctDNA, and pTN was assessed with the Kaplan-Meier method. Multivariate analysis was used to identify prognostic and predictive factors for DFS. RESULTS Among 696 patients (35%), CEA values were retrievable, and for 405 (20%) both CEA and ctDNA were available. An optimized CEA threshold of 2 ng/mL was identified, the 3-year DFS was 66.4% for patients above the threshold and 80.9% for those below (HR, 1.74; 95% CI, 1.33-2.28, P < .001). In multivariate analysis, CEA ≥ 2 ng/mL contributed significantly to model variability, becoming an independent prognostic factor for DFS (HR, 1.82; 95% CI,1.27-2.59), alongside ctDNA (HR, 1.88; 95% CI, 1.16-3.03) and pTN (HR, 1.78; 95% CI, 1.24-2.54). A novel integrated risk classification combining CEA, ctDNA, and pTN stage reclassified 19.8% of pT4/N2 patients as low risk and 2.5% of pT3/N1 patients as high risk. This new classification demonstrated the 3-year DFS of 80.8% for low-risk patients and 55.4% for high-risk patients (HR, 2.66, 95% CI, 1.84-3.86, P < .001). CONCLUSIONS Postoperative CEA value is a prognostic factor for DFS in stage 3 CC, independently of ctDNA and pTN. It advocates for systematic reporting in future adjuvant trials. Integrating both biomarkers with pTN could refine risk classification in stage 3 CC.
Collapse
Affiliation(s)
- Thomas Samaille
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne Université, Paris, France
| | - Antoine Falcoz
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
| | - Romain Cohen
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne Université, Paris, France
| | - Pierre Laurent-Puig
- Institut du cancer Paris CARPEM, Georges Pompidou European Hospital, AP-HP, Université Paris Cité, Paris, France
| | - Thierry André
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne Université, Paris, France
| | - Julien Taieb
- Department of Gastroenterology and GI oncology, Georges Pompidou European Hospital, SIRIC CARPE, Université Paris-Cité, Paris, France
| | - Edouard Auclin
- Department of Medical Oncology, Georges Pompidou European Hospital, AP-HP, Université Paris Cité, Paris, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|
6
|
Valéry M, Tanguy ML, Gelli M, Smolenschi C, Hollebecque A, Boilève A, de Sevilla EF, Tselikas L, Bonnet B, Goéré D, Taïeb J, Boige V, Ducreux M, Malka D. Oxaliplatin-induced peripheral neuropathy with hepatic arterial versus intravenous infusion in metastatic colorectal cancer. Support Care Cancer 2024; 32:660. [PMID: 39283505 DOI: 10.1007/s00520-024-08807-6] [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: 02/04/2024] [Accepted: 08/13/2024] [Indexed: 10/20/2024]
Abstract
BACKGROUND Oxaliplatin, a major drug in metastatic colorectal cancer (mCRC), is responsible for cumulative, dose-limiting peripheral neuropathy (PN). Whether the hepatic arterial infusion (HAI) route can limit oxaliplatin-induced PN in comparison with the intravenous (IV) route has not been specifically explored so far. METHODS We compared the frequency and severity of PN in oxaliplatin-naive patients with mCRC included in trials that evaluated treatment with oxaliplatin administered either by HAI (ACCORD 04, CHOICE, OSCAR, and PACHA-01 trials) or by IV route (FFCD 2000-05 trial). We retrieved anonymized, prospectively collected data from trial databases for the ACCORD 04, CHOICE, and FFCD 2000-05 trials and through a review of Gustave Roussy patients' electronic medical records for PACHA-01 and OSCAR trials. The primary endpoint was the incidence of clinically significant PN (grades 2 to 4) according to the cumulative dose of oxaliplatin received. Secondary endpoints were time to onset of neuropathy as a function of the cumulative dose of oxaliplatin, discontinuation of oxaliplatin for neurotoxicity, and safety. RESULTS A total of 363 patients were included (IV, 300; HAI, 63). In total, 180 patients in the IV group (60%) and 30 patients in the HAI group (48%) developed clinically significant PN, with no significant difference between the two groups (p = 0.23). No difference was shown in the time to onset of PN either (p = 0.23). CONCLUSION The administration of oxaliplatin HAI rather than IV in the treatment of mCRC does not reduce the incidence, precocity, and severity of PN.
Collapse
Affiliation(s)
- Marine Valéry
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Marie-Laure Tanguy
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
- Équipe Labellisée Ligue Contre Le Cancer, Oncostat U1018, INSERM, Université Paris-Saclay, Villejuif, France
| | - Maximiliano Gelli
- Département de Chirurgie Oncologique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Cristina Smolenschi
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
- Département d'Innovation Thérapeutique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Antoine Hollebecque
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
- Département d'Innovation Thérapeutique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Alice Boilève
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
- INSERM Unité Dynamique Des Cellules Tumorales, Université Paris-Saclay, Gustave Roussy, 94805, Villejuif, France
| | | | - Lambros Tselikas
- Département d'Imagerie Médicale et de Radiologie Interventionnelle, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Baptiste Bonnet
- Département d'Imagerie Médicale et de Radiologie Interventionnelle, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Diane Goéré
- Service de Chirurgie Viscérale, Cancérologique et Endocrinienne, Hôpital Universitaire Saint-Louis, Paris, France
| | - Julien Taïeb
- Service d'Oncologie Digestive, Hôpital Européen Georges Pompidou, Paris, France
| | - Valérie Boige
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Michel Ducreux
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
- INSERM Unité Dynamique Des Cellules Tumorales, Université Paris-Saclay, Gustave Roussy, 94805, Villejuif, France
| | - David Malka
- INSERM Unité Dynamique Des Cellules Tumorales, Université Paris-Saclay, Gustave Roussy, 94805, Villejuif, France.
- Département d'Oncologie Médicale, Institut Mutualiste Montsouris, 42 Boulevard Jourdan, 75014 Paris, France.
| |
Collapse
|
7
|
Zhang W, Hou Z, Zhang L, Hong X, Wang W, Wu X, Xu D, Lu Z, Chen J, Peng J. A log odds of positive lymph nodes-based predictive model effectively forecasts prognosis and guides postoperative adjuvant chemotherapy duration in stage III colon cancer: a multi-center retrospective cohort study. BMC Cancer 2024; 24:1088. [PMID: 39223610 PMCID: PMC11370012 DOI: 10.1186/s12885-024-12875-6] [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: 03/31/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The log odds of positive lymph nodes (LODDS) was considered a superior staging system to N stage in colon cancer, yet its value in determining the optimal duration of adjuvant chemotherapy for stage III colon cancer patients has not been evaluated. This study aims to assess the prognostic value of a model that combines LODDS with clinicopathological information for stage III colon cancer patients and aims to stratify these patients using the model, identifying individuals who could benefit from varying durations of adjuvant chemotherapy. METHOD A total of 663 consecutive patients diagnosed with stage III colon cancer, who underwent colon tumor resection between November 2007 and June 2020 at Sun Yat-sen University Cancer Center and Longyan First Affiliated Hospital of Fujian Medical University, were enrolled in this study. Survival outcomes were analyzed using Kaplan-Meier, Cox regression. Nomograms were developed to forecast patient DFS, with the Area Under the Curve (AUC) values of time-dependent Receiver Operating Characteristic (timeROC) and calibration plots utilized to assess the accuracy and reliability of the nomograms. RESULTS Multivariate analysis revealed that perineural invasion (HR = 1.776, 95% CI: 1.052-3.003, P = 0.032), poor tumor differentiation (HR = 1.638, 95% CI: 1.084-2.475, P = 0.019), and LODDS groupings of 2 and 1 (HR = 1.920, 95% CI: 1.297-2.842, P = 0.001) were independent predictors of disease-free survival (DFS) in the training cohort. Nomograms constructed from LODDS, perineural invasion, and poor tumor differentiation demonstrated robust predictive performance for 3-year and 5-year DFS in both training (3-year AUC = 0.706, 5-year AUC = 0.678) and validation cohorts (3-year AUC = 0.744, 5-year AUC = 0.762). Stratification according to this model showed that patients in the high-risk group derived significant benefit from completing 8 cycles of chemotherapy (training cohort, 82.97% vs 67.17%, P = 0.013; validation cohort, 89.49% vs 63.97%, P = 0.030). CONCLUSION The prognostic model, integrating LODDS, pathological differentiation, and neural invasion, demonstrates strong predictive accuracy for stage III colon cancer prognosis. Moreover, stratification via this model offers valuable insights into optimal durations of postoperative adjuvant chemotherapy.
Collapse
Affiliation(s)
- Weili Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicineof Colorectal Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Zhenlin Hou
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicineof Colorectal Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Linjie Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicineof Colorectal Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Xuanlin Hong
- Medical College, Shaoguan University, Shaoguan, Guangdong, 512005, People's Republic of China
| | - Weifeng Wang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicineof Colorectal Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Xiaojun Wu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicineof Colorectal Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Dongbo Xu
- Department of Gastrointestinal Surgery, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, 364000, People's Republic of China
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicineof Colorectal Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Jianxun Chen
- Department of Gastrointestinal Surgery, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, 364000, People's Republic of China.
| | - Jianhong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicineof Colorectal Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China.
| |
Collapse
|
8
|
Dumont F, Kepenekian V, Passot C, Ezanno-Manasterski AC, Pocard M, Raoul JL, Lelièvre B, Hiret S, Senellart H, Pein F, Raimbourg J, Campion L, Thibaudeau E, Paul J, Glehen O. PIPAC in patients with peritoneal metastases from gastrointestinal tract (PIPOX01): An open label, non-comparative phase 1/2 dose escalation and expansion trial. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108468. [PMID: 38878757 DOI: 10.1016/j.ejso.2024.108468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Despite modern systemic chemotherapy, survival remains poor for patients with advanced isolated peritoneal metastases from the gastrointestinal tract. We aimed to assess the safety and efficacy of pressurized intraperitoneal aerosol chemotherapy (PIPAC) with oxaliplatin. PATIENTS AND METHODS We conducted a phase 1/2, open label, non-comparative, dose escalation and expansion trial of PIPAC with oxaliplatin in patients with a peritoneal cancer index (PCI) of more than 5, 13 and 15 for respectively a gastric, small bowel and colorectal primary cancer, and who had received at least three months of systemic chemotherapy. PIPAC cycle lengths were 4-6 weeks with systemic chemotherapy allowed 15 days after each PIPAC. PCI and oxaliplatin tumor concentration were assessed every PIPAC cycle. The main endpoints were tolerability, tumor response, and survival. RESULTS Between 2017 and 2020, 34 patients were enrolled in three centers, in this phase 1/2 study, of whom 25 were evaluable at the recommended dose determined in the phase I trial (90 mg/m2 plus systemic 5-FU). Before inclusion, patients received a median of 2 [1-4] chemotherapy lines and had a median PCI of 22.5 [7-29]. At this dose, the safety profile showed acceptable tolerability. Eight patients (32 %) had grade 3/4 treatment-related adverse events. Minor (grade 1/2) adverse events were mainly abdominal pain (n = 19, 76 %) and nausea (n = 16, 64 %). Median PFS was 6.1 months and median OS was 13 months. CONCLUSION In patients with advanced and refractory peritoneal metastasis, PFS of 6.1 months is encouraging. A prospective randomized phase II study is required.
Collapse
Affiliation(s)
- Frédéric Dumont
- Department of Surgical Oncology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France.
| | - Vahan Kepenekian
- Department of Surgical Oncology, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Christophe Passot
- Oncopharmacology - Pharmacogenetics, Institut de Cancérologie de l'Ouest, Site d'Angers, France
| | | | - Marc Pocard
- Department of Surgery, Hôpital d'Instruction des armées Begin, Saint-Mande, France
| | - Jean-Luc Raoul
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Bénédicte Lelièvre
- Laboratory of Pharmacology and Toxicology, Centre Hospitalier et Universitaire, Angers, France
| | - Sandrine Hiret
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Hélène Senellart
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Francois Pein
- Department of Clinical Research and Innovation, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Judith Raimbourg
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Loic Campion
- Department of Biostatistics and Methodology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Emilie Thibaudeau
- Department of Surgical Oncology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Julie Paul
- Department of Biostatistics and Methodology, Institut de Cancérologie de l'Ouest, Site de Saint Herblain, France
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| |
Collapse
|
9
|
Hu H, Zhang J, Li Y, Wang X, Wang Z, Wang H, Kang L, Liu P, Lan P, Wu X, Zhen Y, Pei H, Huang Z, Zhang H, Chen W, Zeng Y, Lai J, Wei H, Huang X, Chen J, Chen J, Tao K, Xu Q, Peng X, Liang J, Cai G, Ding K, Ding Z, Hu M, Zhang W, Tang B, Hong C, Cao J, Huang Z, Cao W, Li F, Wang X, Wang C, Huang Y, Zhao Y, Cai Y, Ling J, Xie X, Wu Z, Shi L, Ling L, Liu H, Wang J, Huang M, Deng Y. Neoadjuvant Chemotherapy With Oxaliplatin and Fluoropyrimidine Versus Upfront Surgery for Locally Advanced Colon Cancer: The Randomized, Phase III OPTICAL Trial. J Clin Oncol 2024; 42:2978-2988. [PMID: 38564700 DOI: 10.1200/jco.23.01889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/29/2023] [Accepted: 02/02/2024] [Indexed: 04/04/2024] Open
Abstract
PURPOSE The role of neoadjuvant chemotherapy (NAC) in colon cancer remains unclear. This trial investigated whether 3 months of modified infusional fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) or capecitabine and oxaliplatin (CAPOX) as NAC could improve outcomes in patients with locally advanced colon cancer versus upfront surgery. PATIENTS AND METHODS OPTICAL was a randomized, phase III trial in patients with clinically staged locally advanced colon cancer (T3 with extramural spread into the mesocolic fat ≥5 mm or T4). Patients were randomly assigned 1:1 to receive six preoperative cycles of mFOLFOX6 or four cycles of CAPOX, followed by surgery and adjuvant chemotherapy (NAC group), or immediate surgery and the physician's choice of adjuvant chemotherapy (upfront surgery group). The primary end point was 3-year disease-free survival (DFS) assessed in the modified intention-to-treat (mITT) population. RESULTS Between January 2016 and April 2021, of the 752 patients enrolled, 744 patients were included in the mITT analysis (371 in the NAC group; 373 in the upfront surgery group). At a median follow-up of 48.0 months (IQR, 46.0-50.1), 3-year DFS rates were 82.1% in the NAC group and 77.5% in the upfront surgery group (stratified hazard ratio [HR], 0.74 [95% CI, 0.54 to 1.03]). The R0 resection was achieved in 98% of patients who underwent surgery in both groups. Compared with upfront surgery, NAC resulted in a 7% pathologic complete response rate (pCR), significantly lower rates of advanced tumor staging (pT3-4: 77% v 94%), lymph node metastasis (pN1-2: 31% v 46%), and potentially improved overall survival (stratified HR, 0.44 [95% CI, 0.25 to 0.77]). CONCLUSION NAC with mFOLFOX6 or CAPOX did not show a significant DFS benefit. However, this neoadjuvant approach was safe, resulted in substantial pathologic downstaging, and appears to be a viable therapeutic option for locally advanced colon cancer.
Collapse
Affiliation(s)
- Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianwei Zhang
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunfeng Li
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Xiaozhong Wang
- Department of Gastrointestinal Surgery, Shantou Central Hospital, Shantou, People's Republic of China
| | - Ziqiang Wang
- Department of General Surgery, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liang Kang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ping Liu
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaojian Wu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunhuan Zhen
- Department of Colorectal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Haiping Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhongcheng Huang
- Department of General Surgery, Hunan Provincial People's Hospital, Changsha, People's Republic of China
| | - Hao Zhang
- Department of General Surgery, Dongguan Kanghua Hospital, Dongguan, People's Republic of China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yongming Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, People's Republic of China
| | - Jiajun Lai
- Department of Gastrointestinal Surgery, Yuebei People's Hospital, Shaoguan, People's Republic of China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jiansi Chen
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Jigui Chen
- Department of Surgery, The Eighth Hospital of Wuhan, Wuhan, People's Republic of China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qingwen Xu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xiang Peng
- Department of Gastrointestinal Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Junlin Liang
- Department of Coloproctological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guanfu Cai
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangzhou, People's Republic of China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Cancer Center, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zhijie Ding
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University, Xiamen, People's Republic of China
| | - Ming Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wei Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Bo Tang
- Department of General Surgery, The First Hospital Affiliated to Army Medical University, Chongqing, People's Republic of China
| | - Chuyuan Hong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jie Cao
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, People's Republic of China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Wuteng Cao
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Fangqian Li
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinhua Wang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yandong Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiayu Ling
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoyu Xie
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zehua Wu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lishuo Shi
- Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Ling
- Department of Medical Statistics, School of Public Health, and Center for Migrant Health Policy, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Biostatistics Shared Resource, Rutgers Cancer Institute of New Jersey, Rutgers School of Public Health, Brunswick, NJ
| | - Jianping Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meijin Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
10
|
Suzumura H, Terauchi T, Yukisawa S, Kimata M, Shinozaki H. Curative Surgery After Neoadjuvant Chemotherapy for Locally Advanced Sigmoid Colon Cancer With Extensive Abdominal Wall Invasion: A Case Report. Cureus 2024; 16:e67444. [PMID: 39310573 PMCID: PMC11415604 DOI: 10.7759/cureus.67444] [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] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Locally advanced colon cancer (LACC) can be cured under an appropriate treatment strategy, but the decision on the treatment strategy is also important in terms of long-term prognosis. In cases with extensive abdominal wall involvement, it is especially important to secure adequate margins and repair abdominal wall defects. Recently, neoadjuvant chemotherapy (NAC) for LACC has shown promise in improving the chance of cure with tumor shrinkage. Herein, we report a case of curative surgery after NAC for locally advanced sigmoid colon cancer with extensive abdominal wall invasion. A 50-year-old woman visited our hospital with anemia and an abdominal mass. The diagnosis was LACC of the sigmoid colon with abdominal wall invasion (maximum size, 12 cm), and the clinical stage was stage IIIc (T4b[skin]N1bM0). Resection of the involved skin was expected to cause an extensive abdominal wall defect. At first, a colostomy was performed, followed by NAC with leucovorin, 5-fluorouracil, and oxaliplatin (FOLFOX). Ten cycles of chemotherapy were completed without severe adverse events, and the tumor shrank in size by approximately 39%. We performed a curative sigmoidectomy combined with abdominal wall resection with adequate margins. We reconstructed the abdominal wall defect using a left anterolateral thigh skin flap. Pathological examination revealed mucinous carcinoma involving the transverse colon and abdominal wall, with luminal narrowing in the sigmoid colon. The surgical margins were negative, and the tumor was considered to have had a pathological partial response to NAC. Herein, we report a rare case of curative surgery after NAC with FOLFOX for LACC in the sigmoid colon with extensive invasion of the abdominal wall. We reconstructed the extensive abdominal wall defect with a free anterolateral thigh flap. One of the optional treatment strategies for LACC with extensive abdominal wall invasion was reported in our report.
Collapse
Affiliation(s)
| | | | - Seigo Yukisawa
- Department of Medical Oncology, Saiseikai Utsunomiya Hospital, Tochigi, JPN
| | - Masaru Kimata
- Department of Surgery, Saiseikai Utsunomiya Hospital, Tochigi, JPN
| | | |
Collapse
|
11
|
Lecuelle J, Truntzer C, Basile D, Laghi L, Greco L, Ilie A, Rageot D, Emile JF, Bibeau F, Taïeb J, Derangere V, Lepage C, Ghiringhelli F. Machine learning evaluation of immune infiltrate through digital tumour score allows prediction of survival outcome in a pooled analysis of three international stage III colon cancer cohorts. EBioMedicine 2024; 105:105207. [PMID: 38880067 PMCID: PMC11233898 DOI: 10.1016/j.ebiom.2024.105207] [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: 12/18/2023] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND T-cell immune infiltrates are robust prognostic variables in localised colon cancer. Evaluation of prognosis using artificial intelligence is an emerging field. We evaluated whether machine learning analysis improved prediction of patient outcome in comparison with analysis of T cell infiltrate only or in association with clinical variables. METHODS We used data from two phase III clinical trials (Prodige-13 and PETACC08) and one retrospective Italian cohort (HARMONY). Cohorts were split into training (N = 692), internal validation (N = 297) and external validation (N = 672) sets. Tumour slides were stained with CD3mAb. CD3 Machine Learning (CD3ML) score was computed using graphical parameters within the tumour tiles obtained from CD3 slides. CD3 infiltrates in tumour core and invasive margin were automatically detected. Associations of CD3 infiltrates and CD3ML with 5-year Disease-Free Survival (DFS) were examined using univariate and multivariable survival models by Cox regression. FINDINGS CD3 density both in the invasive margin and the tumour core were significantly associated with DFS in the different sets. Similarly, CD3ML score was significantly associated with DFS in all sets. CD3 assessment did not provide added value on top of CD3ML assessment (Likelihood Ratio Test (LRT), p = 0.13). In contrast, CD3ML improved prediction of DFS when combined with a clinical risk stage (LRT, p = 0.001). Stratified by clinical risk score (High or Low), patients with low CD3ML score had better DFS. INTERPRETATION In all tested sets, machine learning analysis of tumour cells improved prediction of prognosis compared to clinical parameters. Adding tumour-infiltrating lymphocytes assessment did not improve prognostic determination. FUNDING This research received no external funding.
Collapse
Affiliation(s)
- Julie Lecuelle
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Caroline Truntzer
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Debora Basile
- Department of Medical Oncology, San Giovanni di Dio Hospital, Crotone, Italy
| | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luana Greco
- Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alis Ilie
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - David Rageot
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Jean-François Emile
- Paris-Saclay University, Versailles SQY University (UVSQ), EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (AP-HP), Ambroise Paré Hospital, Smart Imaging, Service de Pathologie, Boulogne, France
| | - Fréderic Bibeau
- Service d'Anatomie et Cytologie Pathologiques, CHU Côte de Nacre, Normandie Université, Caen, France; Department of Pathology, Besançon University Hospital, Besançon, France
| | - Julien Taïeb
- Institut du Cancer Paris Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique, Sorbonne Université, Université Sorbonne Paris Cité, Université de Paris, Paris, France; Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Valentin Derangere
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France
| | - Come Lepage
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Fédération Francophone de Cancérologie Digestive, Centre de Randomisation Gestion Analyse, EPICAD LNC 1231, Dijon, France; Service d'Hépato-gastroentérologie et Oncologie digestive, CHU de Dijon, France
| | - François Ghiringhelli
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France.
| |
Collapse
|
12
|
Gallois C, Shi Q, Pederson LD, André T, Iveson TJ, Sobrero AF, Alberts S, de Gramont A, Meyerhardt JA, George T, Schmoll HJE, Souglakos I, Harkin A, Labianca R, Sinicrope FA, Oki E, Shields AF, Boukovinas I, Kerr R, Lonardi S, Yothers G, Yoshino T, Goldberg RM, Taieb J, Papamichael D. Oxaliplatin-Based Adjuvant Chemotherapy in Older Patients With Stage III Colon Cancer: An ACCENT/IDEA Pooled Analysis of 12 Trials. J Clin Oncol 2024; 42:2295-2305. [PMID: 38547438 DOI: 10.1200/jco.23.01326] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/30/2023] [Accepted: 02/28/2024] [Indexed: 06/28/2024] Open
Abstract
PURPOSE A number of studies suggest that older patients may have reduced or no benefit from the addition of oxaliplatin to fluoropyrimidines as adjuvant chemotherapy for stage III colon cancer (CC). MATERIALS AND METHODS We studied the prognostic impact of age, as well as treatment adherence/toxicity patterns according to age, in patients with stage III CC who received 3 or 6 months of infusional fluorouracil, leucovorin, and oxaliplatin/capecitabine and oxaliplatin (CAPOX) on the basis of data collected from trials from the ACCENT and IDEA databases. Associations between age and time to recurrence (TTR), disease-free survival (DFS), overall survival (OS), survival after recurrence (SAR), and cancer-specific survival (CSS) were assessed by a Cox model or a competing risk model, stratified by studies and adjusted for sex, performance status, T and N stage, and year of enrollment. RESULTS A total of 17,909 patients were included; 24% of patients were age older than 70 years (n = 4,340). Patients age ≥70 years had higher rates of early treatment discontinuation. Rates of grade ≥3 adverse events were similar between those older and younger than 70 years, except for diarrhea and neutropenia that were more frequent in older patients treated with CAPOX (14.2% v 11.2%; P = .01 and 12.1% v 9.6%; P = .04, respectively). In multivariable analysis, TTR was not significantly different between patients <70 years and those ≥70 years, but DFS, OS, SAR, and CSS were significantly shorter in those patients ≥70 years. CONCLUSION In patients ≥70 years with stage III CC fit enough to be enrolled in clinical trials, oxaliplatin-based adjuvant chemotherapy was well tolerated and led to similar TTR compared with younger patients, suggesting similar efficacy. TTR may be a more appropriate end point for efficacy in this patient population.
Collapse
Affiliation(s)
- Claire Gallois
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, SIRIC CARPEM, Paris-Cité University, Paris, France
| | - Qian Shi
- Department of Quantitative Health Science, Mayo Clinic, Rochester, MN
| | - Levi D Pederson
- Department of Quantitative Health Science, Mayo Clinic, Rochester, MN
| | - Thierry André
- Department of Medical Oncology, Hôpital Saint-Antoine, Sorbonne Université, Paris, France
| | - Timothy J Iveson
- Department of Medical Oncology, University of Southampton, Southampton, United Kingdom
| | | | | | - Aimery de Gramont
- Department of Medical Oncology, Franco-British Institute, Levallois-Perret, France
| | | | - Thomas George
- University of Florida Health Cancer Center, Gainesville, FL
| | - Hans-Joachim E Schmoll
- Department Internal Medicine, Clinic of Internal Medicine IV, University Clinic Halle, Martin-Luther University, Halle, Germany
| | - Ioannis Souglakos
- Department of Medical Oncology, University Hospital of Heraklion, Heraklion, Greece
| | - Andrea Harkin
- Cancer Research UK Glasgow Clinical Trials Unit, Glasgow, United Kingdom
| | - Roberto Labianca
- Cancer Center, Ospedale Papa Giovanni XXIII Bergamo, Bergamo, Italy
| | | | - Eiji Oki
- Department of Surgery and Science, Kyushu University, Fukuoka, Japan
| | | | | | - Rachel Kerr
- Department of Oncology, Oxford University, Oxford, United Kingdom
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IRCCS, Padua, Italy
| | - Greg Yothers
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Richard M Goldberg
- West Virginia University Cancer Institute and the Mary Babb Randolph Cancer Center, Morgantown, WV
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, SIRIC CARPEM, Paris-Cité University, Paris, France
| | | |
Collapse
|
13
|
Wang JL, Chen YS, Huang KC, Yeh CH, Chen MCM, Wu LSH, Chiu YH. Resistant Starch-Encapsulated Probiotics Attenuate Colorectal Cancer Cachexia and 5-Fluorouracil-Induced Microbial Dysbiosis. Biomedicines 2024; 12:1450. [PMID: 39062024 PMCID: PMC11274618 DOI: 10.3390/biomedicines12071450] [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: 05/29/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
5-Fluorouracil (5-FU) is commonly used as the primary chemotherapy for colorectal cancer (CRC). However, it can lead to unwanted chemoresistance. Resistant starch (RS), which functions similarly to fermentable dietary fiber, has the potential to reduce the risk of CRC. The effects of RS on improving CRC-associated cachectic symptoms and 5-FU chemotherapy-induced microbial dysbiosis remain unknown. Female BALB/cByJNarl mice were randomly divided into four groups: one tumor group (with CT26 colonic carcinoma but no treatment) and three CT26 colonic carcinoma-bearing groups that were administered 20 mg/kg 5-FU (T+5-FU group), a probiotic cocktail (4 × 108 CFUs) plus chemotherapy (T+5-FU+Pro), or resistant-starch-encapsulated probiotics plus chemotherapy (T+5-FU+RS-Pro). T+5-FU and T+5-FU+RS-Pro administration significantly suppressed tumor growth and activated apoptotic cell death in CT26-bearing mice. 5-FU-induced increases in inflammatory cytokines and NF-κB signaling were mitigated by the Pro or RS-Pro supplementation. A gut microbial composition comparison indicated that the abundance of intestinal bacteria in the T and T+5-FU groups decreased significantly, while the groups receiving Pro or RS-Pro maintained a greater abundance and healthy gut microbiota composition, suggesting that RS can reduce the microbial dysbiosis that occurs during 5-FU chemotherapy. The use of RS-Pro before chemotherapy should be considered for the regulation of chemotherapy-associated cachectic symptoms, inflammation, and chemotherapy-induced microbial dysbiosis.
Collapse
Affiliation(s)
- Jui-Ling Wang
- Animal Testing Division, National Laboratory Animal Center, National Applied Research Laboratories, Tainan 744, Taiwan;
| | - Yu-Siang Chen
- Department of Microbiology, Soochow University, Taipei 111, Taiwan;
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Kuo-Chin Huang
- Holistic Education Center, Mackay Medical College, New Taipei City 25245, Taiwan;
| | - Chin-Hsing Yeh
- Fecula Biotech Co., Ltd., Tainan 744, Taiwan; (C.-H.Y.); (M.C.-M.C.)
| | | | - Lawrence Shih-Hsin Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| | - Yi-Han Chiu
- Department of Microbiology, Soochow University, Taipei 111, Taiwan;
| |
Collapse
|
14
|
Pathania AS, Chava H, Balusu R, Pasupulati AK, Coulter DW, Challagundla KB. The crosstalk between non-coding RNAs and cell-cycle events: A new frontier in cancer therapy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200785. [PMID: 38595981 PMCID: PMC10973673 DOI: 10.1016/j.omton.2024.200785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
The cell cycle comprises sequential events during which a cell duplicates its genome and divides it into two daughter cells. This process is tightly regulated to ensure that the daughter cell receives identical copied chromosomal DNA and that any errors in the DNA during replication are correctly repaired. Cyclins and their enzyme partners, cyclin-dependent kinases (CDKs), are critical regulators of G- to M-phase transitions during the cell cycle. Mitogenic signals induce the formation of the cyclin/CDK complexes, resulting in phosphorylation and activation of the CDKs. Once activated, cyclin/CDK complexes phosphorylate specific substrates that drive the cell cycle forward. The sequential activation and inactivation of cyclin-CDK complexes are tightly controlled by activating and inactivating phosphorylation events induced by cell-cycle proteins. The non-coding RNAs (ncRNAs), which do not code for proteins, regulate cell-cycle proteins at the transcriptional and translational levels, thereby controlling their expression at different cell-cycle phases. Deregulation of ncRNAs can cause abnormal expression patterns of cell-cycle-regulating proteins, resulting in abnormalities in cell-cycle regulation and cancer development. This review explores how ncRNA dysregulation can disrupt cell division balance and discusses potential therapeutic approaches targeting these ncRNAs to control cell-cycle events in cancer treatment.
Collapse
Affiliation(s)
- Anup S. Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Haritha Chava
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ramesh Balusu
- Department of Hematologic Malignancies and Cellular Therapeutics, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Anil K. Pasupulati
- Department of Biochemistry, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Don W. Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kishore B. Challagundla
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- The Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
15
|
Lecomte T, Tougeron D, Chautard R, Bressand D, Bibeau F, Blanc B, Cohen R, Jacques J, Lagasse JP, Laurent-Puig P, Lepage C, Lucidarme O, Martin-Babau J, Panis Y, Portales F, Taieb J, Aparicio T, Bouché O. Non-metastatic colon cancer: French Intergroup Clinical Practice Guidelines for diagnosis, treatments, and follow-up (TNCD, SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, SFP, AFEF, and SFR). Dig Liver Dis 2024; 56:756-769. [PMID: 38383162 DOI: 10.1016/j.dld.2024.01.208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 01/25/2024] [Accepted: 01/28/2024] [Indexed: 02/23/2024]
Abstract
INTRODUCTION This article is a summary of the French intergroup guidelines regarding the management of non-metastatic colon cancer (CC), revised in November 2022. METHODS These guidelines represent collaborative work of all French medical and surgical societies involved in the management of CC. Recommendations were graded in three categories (A, B, and C) according to the level of evidence found in the literature published up to November 2022. RESULTS Initial evaluation of CC is based on clinical examination, colonoscopy, chest-abdomen-pelvis computed tomography (CT) scan, and carcinoembryonic antigen (CEA) assay. CC is usually managed by surgery and adjuvant treatment depending on the pathological findings. The use of adjuvant therapy remains a challenging question in stage II disease. For high-risk stage II CC, adjuvant chemotherapy must be discussed and fluoropyrimidine monotherapy or oxaliplatin-based chemotherapy proposed according to the type and number of poor prognostic features. Oxaliplatin-based chemotherapy (FOLFOX or CAPOX) is the current standard for adjuvant therapy of patients with stage III CC. However, these regimens are associated with significant oxaliplatin-induced neurotoxicity. The results of the recent IDEA study provide evidence that 3 months of treatment with CAPOX is as effective as 6 months of oxaliplatin-based therapy in patients with low-risk stage III CC (T1-3 and N1). A 6-month oxaliplatin-based therapy remains the standard of care for high-risk stage III CC (T4 and/or N2). For patients unfit for oxaliplatin, fluoropyrimidine monotherapy is recommended. CONCLUSION French guidelines for non-metastatic CC management help to offer the best personalized therapeutic strategy in daily clinical practice. Each individual case must be discussed within a multidisciplinary tumor board and then the treatment option decided with the patient.
Collapse
Affiliation(s)
- Thierry Lecomte
- Department of Hepatogastroenterology and Digestive Oncology, Tours University Hospital, Tours, France; Inserm UMR 1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France.
| | - David Tougeron
- Department of Hepatogastroenterology, Poitiers University Hospital, Poitiers, France
| | - Romain Chautard
- Department of Hepatogastroenterology and Digestive Oncology, Tours University Hospital, Tours, France; Inserm UMR 1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Diane Bressand
- Department of Hepatogastroenterology and Digestive Oncology, Tours University Hospital, Tours, France
| | - Frédéric Bibeau
- Department of Pathology, Besançon University Hospital, Besançon, France
| | - Benjamin Blanc
- Department of Digestive Surgery, Dax Hospital, Dax, France
| | - Romain Cohen
- Sorbonne Université, Department of Medical Oncology, Saint-Antoine hospital, AP-HP, Inserm, Unité Mixte de Recherche Scientifique 938 et SiRIC CURAMUS, Saint-Antoine Research Center, Paris, France
| | - Jérémie Jacques
- Department of Hepatogastroenterology, Limoges University Hospital, Limoges, France
| | - Jean-Paul Lagasse
- Department of Hepatogastroenterology and Digestive Oncology, Orléans University Hospital, Orléans, France
| | - Pierre Laurent-Puig
- Department of Biology, AP-HP, European Georges Pompidou Hospital, Paris, France
| | - Come Lepage
- Department of Hepatogastroenterology and Digestive Oncology, Dijon University Hospital, Dijon, France
| | - Olivier Lucidarme
- Department of Radiology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Jérôme Martin-Babau
- Armoricain Center of Radiotherapy, Radiology and Oncology, Côtes D'Armor Private Hospital, Plérin, France
| | - Yves Panis
- Department of Colorectal Surgery, AP-HP, Beaujon Hospital, Clichy, France
| | - Fabienne Portales
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, Paris, France
| | - Thomas Aparicio
- Department of Gastroenterology and Digestive Oncology, AP-HP, Saint-Louis Hospital, Paris, France
| | - Olivier Bouché
- Department of Digestive Oncology, Reims, CHU Reims, France
| |
Collapse
|
16
|
Lee KH, Yang IJ, Ha GW, Lee J, Park YY, Lee SH, Lee JM, Bae JH, Park EJ, Kim H, Kim KY, An S, Kim IY, Kim JY. mFOLFIRINOX versus mFOLFOX 6 as adjuvant treatment for high-risk stage III colon cancer - the FROST trial: study protocol for a multicenter, randomized controlled, phase II trial. BMC Cancer 2024; 24:397. [PMID: 38553680 PMCID: PMC10979582 DOI: 10.1186/s12885-024-11939-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/31/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND High-risk stage III colon cancer has a considerably poorer prognosis than stage II and low-risk stage III colon cancers. Nevertheless, most guidelines recommend similar adjuvant treatment approaches for all these stages despite the dearth of research focusing on high-risk stage III colon cancer and the potential for improved prognosis with intensive adjuvant treatment. Given the the proven efficacy of triplet chemotherapy in metastatic colorectal cancer treatment, the goal of this study is to evaluate the oncologic efficacy and safety of mFOLFIRINOX in comparison to those of the current standard of care, mFOLFOX 6, as an adjuvant treatment for patients diagnosed with high-risk stage III colon cancer after radical resection. METHODS This multicenter, randomized (1:1), open-label, phase II trial will assess and compare the effectiveness and toxicity of mFOLFIRINOX and mFOLFOX 6 in patients with high-risk stage III colon cancer after radical resection. The goal of the trial is to enroll 312 eligible patients, from 11 institutes, aged between 20 and 70 years, with an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2, or between 70 and 75 with an ECOG performance status of 0. Patients will be randomized into two arms - Arm A, the experimental arm, and Arm B, the reference arm - and will receive 12 cycles of mFOLFIRINOX and mFOLFOX 6 every 2 weeks, respectively. The primary endpoint of this study is the 3-year disease-free survival, and secondary endpoints include the 3-year overall survival and treatment toxicity. DISCUSSION The Frost trial would help determine the oncologic efficacy and safety of adjuvant triplet chemotherapy for high-risk stage III colon cancers and ultimately improve prognoses. TRIAL REGISTRATION ClinicalTrials.gov NCT05179889, registered on 17 December 2021.
Collapse
Affiliation(s)
- Kyung-Ha Lee
- Department of Colorectal Surgery, Chungnam National University Hospital & College of Medicine, Daejeon, Korea
| | - In Jun Yang
- Department of Colorectal Surgery, Chungnam National University Hospital & College of Medicine, Daejeon, Korea
| | - Gi Won Ha
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Korea
| | - Jaeim Lee
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Youn Young Park
- Department of Surgery, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Suk Hwan Lee
- Department of Surgery, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Jong Min Lee
- Department of Surgery, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Jung Hoon Bae
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun Jung Park
- Division of Colorectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyungjin Kim
- Department of Surgery, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Keun Young Kim
- Department of General Surgery, Wonkwang University Hospital & School of Medicine, Iksan, Korea
| | - Sanghyung An
- Department of Surgery, Yonsei University, Wonju College of Medicine, Wonju, Korea
| | - Ik Yong Kim
- Department of Surgery, Yonsei University, Wonju College of Medicine, Wonju, Korea
| | - Ji Yeon Kim
- Department of Colorectal Surgery, Chungnam National University Hospital & College of Medicine, Daejeon, Korea.
| |
Collapse
|
17
|
Protásio BM, de Castria TB, Natalino R, Mangone FR, Saragiotto DF, Sabbaga J, Hoff PM, Chammas R. Prognostic Impact of Primary Tumor Sidedness in Stage III Colorectal Cancer: Real-World Evidence from a Brazilian Cohort. Clin Colorectal Cancer 2024; 23:73-84. [PMID: 38151358 DOI: 10.1016/j.clcc.2023.12.001] [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: 08/18/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Primary tumor sidedness (PTS) is an independent prognostic factor in patients with metastatic colorectal cancer (CRC), with a worse prognosis for right-sided tumors. There are limited data on the prognostic impact of PTS in stage III CRC. The main objective of this study was to analyze the prognostic impact of PTS in stage III CRC. PATIENTS AND METHODS A retrospective and uni-institutional cohort study was performed in an oncology reference center. Patients with stage III CRC treated with a 5-fluorouracil and oxaliplatin-based chemotherapy regimen (mFLOX regimen) from October 2007 to February 2013 were included. The primary outcome was the probability of overall survival (OS) at 5 years stratified by PTS. Secondary outcomes were the probability of disease-free survival (DFS) at 5 years and an analysis of the prognostic impact of clinical and molecular biomarkers. Kaplan‒Meier curves were used, and Cox models were used to evaluate prognostic factors associated with OS and DFS. RESULTS Overall, 265 patients were evaluated. Transverse colon tumors, multicentric tumors, and undetermined primary subsites were excluded, resulting in 234 patients classified according to PTS: 95 with right sidedness (40.6%) and 139 with left sidedness (59.4%). The median follow-up time was 66 months [interquartile range (IQR): 39-81]. The 5-year OS probabilities for right-sided and left-sided tumors were 67% (95% CI: 58%-77%) and 82% (75%-89%), respectively [hazard ratio (HR): 2.02, 95% CI: 1.18-3.46; P = .010]. The 5-year probabilities of DFS for right-sided and left-sided tumors were 58% (49%-69%) and 65% (58%-74%), respectively (HR: 1.29, 0.84-1.97; P = 0.248). CONCLUSION These data suggest that there may be a worse prognosis (inferior OS at 5 years) for resected right-sided stage III CRC patients treated in the real world. However, these data need to be confirmed by prospective studies with a larger number of participants.
Collapse
Affiliation(s)
- Bruno Medonça Protásio
- Programa de Pós-Graduação em Oncologia, Faculdade de Medicina da Universidade de São Paulo- FMUSP, Sao Paulo, Brazil; Núcleo de Oncologia da Bahia- NOB. Grupo Oncoclínicas, Salvador, Brazil.
| | | | - Renato Natalino
- Centro de Investigação Translacional em Oncologia- CTO. Instituto do Câncer do Estado de São Paulo-ICESP. Faculdade de Medicina da Universidade de São Paulo- FMUSP, Sao Paulo, Brazil
| | - Flávia R Mangone
- Centro de Investigação Translacional em Oncologia- CTO. Instituto do Câncer do Estado de São Paulo-ICESP. Faculdade de Medicina da Universidade de São Paulo- FMUSP, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de São Paulo, Sao Paulo, Brazil
| | - Daniel Fernandes Saragiotto
- Departamento de Oncologia do Hospital Santa Catarina Paulista, São Paulo, Brazil; Centro Brasileiro de Radioterapia, Oncologia e Mastologia- CEBROM. Grupo Oncoclínicas, Goiania, Brazil
| | - Jorge Sabbaga
- IDOR- Intituto D'Or de Pesquisa e Ensino, Sao Paulo, Brazil; Disciplina de Oncologia Clínica do Departamento de Radiologia e Oncologia da Faculdade de Medicina da Universidade de São Paulo- FMUSP. Instituto do Câncer do Estado de São Paulo- ICESP, Sao Paulo, Brazil
| | - Paulo M Hoff
- IDOR- Intituto D'Or de Pesquisa e Ensino, Sao Paulo, Brazil; Disciplina de Oncologia Clínica do Departamento de Radiologia e Oncologia da Faculdade de Medicina da Universidade de São Paulo- FMUSP. Instituto do Câncer do Estado de São Paulo- ICESP, Sao Paulo, Brazil
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia- CTO. Instituto do Câncer do Estado de São Paulo-ICESP. Faculdade de Medicina da Universidade de São Paulo- FMUSP, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de São Paulo, Sao Paulo, Brazil
| |
Collapse
|
18
|
Yu Z, Deng P, Chen Y, Lin D, Liu S, Hong J, Guan P, Chen J, Zhong ME, Chen J, Chen X, Sun Y, Wang Y, Wang P, Cai Z, Chan JY, Huang Y, Xiao R, Guo Y, Zeng X, Wang W, Zou Y, Yu Q, Lan P, Teh BT, Wu X, Tan J. Pharmacological modulation of RB1 activity mitigates resistance to neoadjuvant chemotherapy in locally advanced rectal cancer. Proc Natl Acad Sci U S A 2024; 121:e2304619121. [PMID: 38289962 PMCID: PMC10861914 DOI: 10.1073/pnas.2304619121] [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: 04/17/2023] [Accepted: 11/29/2023] [Indexed: 02/01/2024] Open
Abstract
Resistance to neoadjuvant chemotherapy leads to poor prognosis of locally advanced rectal cancer (LARC), representing an unmet clinical need that demands further exploration of therapeutic strategies to improve clinical outcomes. Here, we identified a noncanonical role of RB1 for modulating chromatin activity that contributes to oxaliplatin resistance in colorectal cancer (CRC). We demonstrate that oxaliplatin induces RB1 phosphorylation, which is associated with the resistance to neoadjuvant oxaliplatin-based chemotherapy in LARC. Inhibition of RB1 phosphorylation by CDK4/6 inhibitor results in vulnerability to oxaliplatin in both intrinsic and acquired chemoresistant CRC. Mechanistically, we show that RB1 modulates chromatin activity through the TEAD4/HDAC1 complex to epigenetically suppress the expression of DNA repair genes. Antagonizing RB1 phosphorylation through CDK4/6 inhibition enforces RB1/TEAD4/HDAC1 repressor activity, leading to DNA repair defects, thus sensitizing oxaliplatin treatment in LARC. Our study identifies a RB1 function in regulating chromatin activity through TEAD4/HDAC1. It also provides the combination of CDK4/6 inhibitor with oxaliplatin as a potential synthetic lethality strategy to mitigate oxaliplatin resistance in LARC, whereby phosphorylated RB1/TEAD4 can serve as potential biomarkers to guide the patient stratification.
Collapse
Affiliation(s)
- Zhaoliang Yu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Peng Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yufeng Chen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Dezheng Lin
- Department of Endoscopic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510060, People’s Republic of China
| | - Shini Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Jinghan Hong
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
| | - Peiyong Guan
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore138672, Singapore
| | - Jianfeng Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Min-er Zhong
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jinghong Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Xiaochuan Chen
- Department of Obstetrics and Gynecology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Yichen Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yali Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Peili Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Zerong Cai
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jason Yongsheng Chan
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| | - Yulin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Rong Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yaoyu Guo
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Xian Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Wenyu Wang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Yifeng Zou
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Qiang Yu
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore138672, Singapore
| | - Ping Lan
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Bin Tean Teh
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| | - Xiaojian Wu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jing Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| |
Collapse
|
19
|
Audisio A, Fazio R, Daprà V, Assaf I, Hendlisz A, Sclafani F. Neoadjuvant chemotherapy for early-stage colon cancer. Cancer Treat Rev 2024; 123:102676. [PMID: 38160535 DOI: 10.1016/j.ctrv.2023.102676] [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: 10/31/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Surgery with or without adjuvant chemotherapy is the standard treatment for early-stage colon cancer. However, evidence has recently emerged for neoadjuvant chemotherapy, with the results of randomised clinical trials sparking debates within multidisciplinary teams and splitting the gastrointestinal oncology community. Further to a systematic search of the literature, we provide a thorough and in-depth analysis of the findings from these trials, highlighting the advantages and disadvantages of neoadjuvant chemotherapy. We conclude that, while there is a potential value of moving systemic therapy from the post-operative to the pre-operative setting, the available evidence does not justify a shift in the treatment paradigm of early-stage colon cancer, and surgery with or without adjuvant chemotherapy should remain the standard approach for these patients.
Collapse
Affiliation(s)
- Alessandro Audisio
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium
| | - Roberta Fazio
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium
| | - Valentina Daprà
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium
| | - Irene Assaf
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium; Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alain Hendlisz
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium; Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Francesco Sclafani
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium; Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
20
|
Asada Y, Nakayama F, Takenoya T, Oto I, Nakamura T, Osumi K, Kameyama N. Curative resection after percutaneous drainage followed by preoperative panitumumab monotherapy for locally advanced sigmoid colon cancer with intra-abdominal abscess: a case report. Surg Case Rep 2024; 10:23. [PMID: 38233703 PMCID: PMC10794648 DOI: 10.1186/s40792-023-01800-x] [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: 08/26/2023] [Accepted: 12/25/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND The gold standard treatment for locally advanced colon cancer is curative surgery followed by adjuvant chemotherapy, although this approach is associated with serious concerns, such as high recurrence rates and occasionally unnecessary oversurgery. Neoadjuvant chemotherapy may be a promising strategy for overcoming these issues. This study reports a case of a recurrence-free patient who underwent curative resection without significant organ dysfunction after preoperative chemotherapy for locally advanced sigmoid colon cancer. The tumor coexisted with a large intra-abdominal abscess, and the patient was quite frail at the first visit. We performed percutaneous drainage followed by preoperative panitumumab monotherapy, which yielded favorable outcomes. CASE PRESENTATION A 78-year-old frail woman was emergently transferred to our hospital with fever and abdominal pain. The diagnosis was locally advanced sigmoid colon cancer stage IIIC (T4bN2aM0) with a large intra-abdominal abscess. Immediate curative surgery was inappropriate, considering both tumor progression and the patient's frailty. We performed percutaneous drainage and colostomy construction, which was followed by seven cycles of preoperative panitumumab monotherapy without significant adverse events. After these treatments, inflammation was well controlled, and the tumor shrank remarkably. Furthermore, the patient recovered well from frailty; therefore, curative sigmoidectomy combined with resection of the left ovary and stoma closure was possible without any postoperative complications. The final pathological finding was T3N0M0, stage IIA disease. The patient was recurrence-free and had no significant organ dysfunction 21 months after the curative surgery. CONCLUSIONS The management of intra-abdominal abscesses and tailor-made preoperative chemotherapy based on the patient's frailty may have been the key factors responsible for the favorable course of this patient. Although further research is needed on the appropriateness of percutaneous drainage for malignancies related to intra-abdominal abscesses and preoperative panitumumab use for locally advanced colon cancer, the study findings can serve as reference for managing similar cases in an aging society.
Collapse
Affiliation(s)
- Yusuke Asada
- Department of Surgery, Ogikubo Hospital, 3-1-24 Imagawa, Suginami, Tokyo, 167-0035, Japan.
| | - Fumitaka Nakayama
- Department of Surgery, Ogikubo Hospital, 3-1-24 Imagawa, Suginami, Tokyo, 167-0035, Japan
| | - Takashi Takenoya
- Department of Surgery, Ogikubo Hospital, 3-1-24 Imagawa, Suginami, Tokyo, 167-0035, Japan
| | - Ippei Oto
- Department of Surgery, Ogikubo Hospital, 3-1-24 Imagawa, Suginami, Tokyo, 167-0035, Japan
| | - Tetsuya Nakamura
- Department of Surgery, Ogikubo Hospital, 3-1-24 Imagawa, Suginami, Tokyo, 167-0035, Japan
| | - Koji Osumi
- Department of Surgery, Ogikubo Hospital, 3-1-24 Imagawa, Suginami, Tokyo, 167-0035, Japan
| | - Noriaki Kameyama
- Department of Surgery, Ogikubo Hospital, 3-1-24 Imagawa, Suginami, Tokyo, 167-0035, Japan
| |
Collapse
|
21
|
Rasola C, Laurent-Puig P, André T, Falcoz A, Lepage C, Aparicio T, Bouché O, Lievre A, Mineur L, Bennouna J, Louvet C, Bachet JB, Borg C, Vernerey D, Lonardi S, Taieb J. Time to recurrence and its relation to survival after recurrence in patients resected for stage III colon cancer. Eur J Cancer 2023; 194:113321. [PMID: 37797388 DOI: 10.1016/j.ejca.2023.113321] [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: 06/25/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND It is intuitively thought that early relapse is associated with poor survival after recurrence (SAR) in resected colon cancer (CC) patients, but this has never been formally studied. METHODS We pooled data from stage III patients treated with oxaliplatin-based adjuvant therapy in two phase III trials, to analyse time to recurrence (TTR) and its relationship with SAR. TTR and SAR were also studied according to molecular status (mismatch repair (MMR), RAS, and BRAFV600E). Early relapsing patients were defined as patients having a TTR event within 12 months after starting adjuvant chemotherapy. RESULTS 4548 stage III CC patients were included in the present analysis. Deficient MMR (dMMR) CC patients experienced fewer recurrences than proficient (p)MMR CC patients (18.8% versus 27.6%) but had a significantly shorter median TTR (mTTR; 0.74 versus 1.40 years, p < 0.0001). In pMMR patients, BRAF and RAS mutations were also associated with earlier mTTR as compared to double wild-type (WT) patients (0.99 versus 1.38 versus 1.54 years, respectively, p < 0.0001). Early recurrence occurred in 397 patients and was associated with a median SAR (2.2 versus 3.3 years, p = 0.0007). However, this association was mainly due to pMMR/RAS and BRAFV600E mutated tumours and was not confirmed in dMMR and pMMR/double WT subgroups. CONCLUSION In resected stage III CC treated with standard oxaliplatin-based adjuvant therapy, TTR varies between dMMR, pMMR/RAS, or BRAFV600E mutated and pMMR/double WT tumours. In addition, early relapse is associated with poor survival, mainly due to patients resected for a pMMR/RAS or BRAFV600E mutated tumour.
Collapse
Affiliation(s)
- Cosimo Rasola
- Department of Gastroenterology and GI Oncology, Georges Pompidou European Hospital, SIRIC CARPEM, Université Paris-Cité, Paris, France; Department of Oncology, Veneto Institute of Oncology IRCCS, Padua, Italy; Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris Cité, team Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris CARPEM, AP-HP,Centre Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Thierry André
- Sorbonne Université and Medical Oncology Department, Hôpital Saint-Antoine, Paris, France
| | - Antoine Falcoz
- University Hospital of Besançon, Methodology and Quality of Life Unit in Oncology, Besançon, France; INSERM, Établissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Come Lepage
- Gastroenterology and Digestive Oncology, Hôpital Universitaire Le Bocage, Dijon, France
| | - Thomas Aparicio
- Université Paris-Cité, Gastroenterology Department, Hôpital Saint Louis, APHP, Paris, France
| | | | - Astrid Lievre
- Digestive Unit, Hôpital Universitaire de Pontchaillou, Rennes, France
| | - Laurent Mineur
- Oncology Department, Clinique Sainte-Catherine, Avignon, France
| | - Jaafar Bennouna
- Department of Medical Oncology, Hôpital Foch, Suresnes, France
| | - Christophe Louvet
- Department of Medical Oncology, Institute Mutualiste Montsouris, Paris, France
| | - Jean Baptiste Bachet
- Sorbonne University, Hepatogastroenterology and Digestive Oncology Department, Pitié Salpêtrière hospital, APHP, Paris, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, France
| | - Dewi Vernerey
- University Hospital of Besançon, Methodology and Quality of Life Unit in Oncology, Besançon, France; INSERM, Établissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IRCCS, Padua, Italy; Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Julien Taieb
- Department of Gastroenterology and GI Oncology, Georges Pompidou European Hospital, SIRIC CARPEM, Université Paris-Cité, Paris, France.
| |
Collapse
|
22
|
Soni N, Nandi G, Chaudhary M, Bissa B. The role of ncRNA in the co-regulation of autophagy and exosome pathways during cancer progression. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119523. [PMID: 37348764 DOI: 10.1016/j.bbamcr.2023.119523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023]
Abstract
Since its discovery a few decades ago, autophagy has been recognized as a crucial signaling pathway, linked to the recycling of cellular components in nutrient stress. Autophagy is a two-way sword, playing a dual role in tumorigenesis. In this catabolic process, dysfunctional organelles, biomolecules, and misfolded proteins are sequestered in the autophagosome and sent to the lysosome for degradation. Alongside, there are cellular messengers called exosomes, which are released from cells and are known to communicate and regulate metabolism in recipient cells. Multivesicular bodies (MVB) act as the intricate link between autophagy and exosome pathways. The continuous crosstalk between the two pathways is coordinated and regulated by multiple players among which ncRNA is the emerging candidates. The exosomes carry varied cargo of which non-coding RNA exerts an immediate regulatory effect on recipient cells. ncRNA is known to exhibit dual behavior in both promoting and inhibiting tumor growth. There is increasing evidence for the involvement of ncRNAs' in the regulation of different hallmarks of cancer. Different ncRNAs are involved in the co-regulation of autophagy and exosome pathways and therefore represent a superior therapeutic approach to target cancer chemoresistance. Here, we will discuss the ncRNA involved in regulating autophagy, and exosomes pathways and its relevance in cancer therapeutics.
Collapse
Affiliation(s)
- Naveen Soni
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Gargi Nandi
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Megha Chaudhary
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Bhawana Bissa
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India.
| |
Collapse
|
23
|
Gallois C, Landi M, Taieb J, Sroussi M, Saberzadeh-Ardestani B, Cazelles A, Lonardi S, Bergamo F, Intini R, Maddalena G, Pietrantonio F, Corti F, Ambrosini M, Martinetti A, Germani MM, Boccaccio C, Vetere G, Mouillet-Richard S, de Reynies A, Sinicrope FA, Cremolini C, Laurent-Puig P. Transcriptomic Signatures of MSI-High Metastatic Colorectal Cancer Predict Efficacy of Immune Checkpoint Inhibitors. Clin Cancer Res 2023; 29:3771-3778. [PMID: 37439810 PMCID: PMC10502457 DOI: 10.1158/1078-0432.ccr-22-3964] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/10/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023]
Abstract
PURPOSE Microsatellite instability (MSI) is currently the only predictive biomarker of efficacy of immune checkpoint inhibitors (ICI) in metastatic colorectal cancers (mCRC). However, 10% to 40% of patients with MSI mCRC will experience a primary resistance to ICI. EXPERIMENTAL DESIGN In two cohorts of patients with MSI mCRC treated with ICI (exploratory, N = 103; validation, N = 35), 3' RNA sequencing was performed from primary tumors. Previously described single-cell transcriptomic signatures of tumor microenvironment (TME) were analyzed. RESULTS In the exploratory cohort, the unsupervised clustering allowed the identification of three clusters of tumors with distinct transcriptional profiles: cluster A ("stromalHIGH-proliferationLOW"), cluster B ("stromalHIGH-proliferationMED"), and cluster C ("stromalLOW-proliferationHIGH"), with an enrichment of patients progressing at first disease assessment under ICI in cluster A (30% vs. 12% in cluster B and 8.1% in cluster C; P = 0.074). Progression-free survival (PFS) was also significantly shorter in patients belonging to cluster A, compared with clusters B or C (P < 0.001) with 2-year PFS rates of 33.5%, 80.5%, and 78.3%, respectively. In multivariate analysis, PFS was still significantly longer in patients belonging to cluster B [HR, 0.19; 95% confidence interval (CI), 0.08-0.45; P < 0.001] and cluster C (HR, 0.25; 95% CI, 0.10-0.59; P = 0.02), compared with patients belonging to cluster A. The association of this clustering with PFS under ICI was confirmed in the validation cohort. PFS related to non-ICI-based regimens was not significantly different according to cluster. CONCLUSIONS This unsupervised transcriptomic classification identified three groups of MSI mCRCs with different compositions of TME cells and proliferative capacities of TME/tumor cells. The "stromalHIGH-proliferationLOW" cluster is associated with a poorer prognosis with ICI treatment.
Collapse
Affiliation(s)
- Claire Gallois
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
- Institut du Cancer Paris CARPEM, AP-HP Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Matteo Landi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Julien Taieb
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
- Institut du Cancer Paris CARPEM, AP-HP Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Marine Sroussi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
- Institut Chimie Biologie Innovation – Laboratoire de BioChimie, ESPCI, UMR8231 CNRS, Université PSL, Paris, France
| | - Bahar Saberzadeh-Ardestani
- Gastrointestinal Research Unit, Departments of Medicine and Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Antoine Cazelles
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Sara Lonardi
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IRCCS, Padova, Italy
| | - Francesca Bergamo
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IRCCS, Padova, Italy
| | - Rossana Intini
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IRCCS, Padova, Italy
| | - Giulia Maddalena
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IRCCS, Padova, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Corti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Margherita Ambrosini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Chiara Boccaccio
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Guglielmo Vetere
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Sophie Mouillet-Richard
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Aurélien de Reynies
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Frank A. Sinicrope
- Gastrointestinal Research Unit, Departments of Medicine and Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
- Institut du Cancer Paris CARPEM, APHP.Centre, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
24
|
Piringer G, Gruenberger T, Thaler J, Kührer I, Kaczirek K, Längle F, Viragos-Toth I, Amann A, Eisterer W, Függer R, Andel J, Pichler A, Stift J, Sölkner L, Gnant M, Öfner D. LM02 trial Perioperative treatment with panitumumab and FOLFIRI in patients with wild-type RAS, potentially resectable colorectal cancer liver metastases-a phase II study. Front Oncol 2023; 13:1231600. [PMID: 37621684 PMCID: PMC10446765 DOI: 10.3389/fonc.2023.1231600] [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: 05/30/2023] [Accepted: 07/17/2023] [Indexed: 08/26/2023] Open
Abstract
Background Twenty percent of colorectal cancer liver metastases (CLMs) are initially resectable with a 5-year survival rate of 25%-40%. Perioperative folinic acid, 5-fluorouracil, oxaliplatin (FOLFOX) increases progression-free survival (PFS). In advanced disease, the addition of targeting therapies results in an overall survival (OS) advantage. The aim of this study was to evaluate panitumumab and FOLFIRI as perioperative therapy in resectable CLM. Methods Patients with previously untreated, wild-type Rat sarcoma virus (RAS), and resectable CLM were included. Preoperative four and postoperative eight cycles of panitumumab and folinic acid, 5-fluorouracil, irinotecan (FOLFIRI) were administered. Primary objectives were efficacy and safety. Secondary endpoints included PFS and OS. Results We enrolled 36 patients in seven centers in Austria (intention-to-treat analyses, 35 patients). There were 28 men and seven women, and the median age was 66 years. About 91.4% completed preoperative therapy and 82.9% underwent liver resection. The R0 resection rate was 82.7%. Twenty patients started and 12 patients completed postoperative chemotherapy. The objective radiological response rate after preoperative therapy was 65.7%. About 20% and 5.7% of patients had stable disease and progressive disease, respectively. The most common grade 3 adverse events were diarrhea, rash, and leukopenia during preoperative therapy. One patient died because of sepsis, and one had a pulmonary embolism grade 4. After surgery, two patients died because of hepatic failure. Most common grade 3 adverse events during postoperative therapy were skin toxicities/rash and leukopenia/neutropenia, and the two grade 4 adverse events were stroke and intestinal obstruction. Median PFS was 13.2 months. The OS rate at 12 and 24 months were 85.6% and 73.3%, respectively. Conclusions Panitumumab and FOLFIRI as perioperative therapy for resectable CLM result in a radiological objective response rate in 65.7% of patients with a manageable grade 3 diarrhea rate of 14.3%. Median PFS was 13.2 months, and the 24-month OS rate was 73.3%. These data are insufficient to widen the indication of panitumumab from the unresectable setting to the setting of resectable CLM.
Collapse
Affiliation(s)
- Gudrun Piringer
- Department of Internal Medicine IV, Klinikum Wels-Grieskirchen, Wels, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Thomas Gruenberger
- Department of Surgery, Clinic Favoriten, Hepato-Pancreato-Biliary Center, Health Network Vienna and Sigmund Freud University, Vienna, Austria
| | - Josef Thaler
- Department of Internal Medicine IV, Klinikum Wels-Grieskirchen, Wels, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Irene Kührer
- Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Klaus Kaczirek
- Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Friedrich Längle
- Department of Surgery, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | - Istvan Viragos-Toth
- Department of Surgery, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | - Arno Amann
- Department of Haematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wolfgang Eisterer
- Department of Internal Medicine and Oncology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Reinhold Függer
- Department of General and Visceral Surgery, Congregation Hospital, Linz, Austria
| | - Johannes Andel
- Department of Internal Medicine II, Landeskrankenhaus Steyr, Steyr, Austria
| | - Angelika Pichler
- Department of Hematology and Oncology, Landeskrankenhaus Hochsteiermark, Leoben, Austria
| | - Judith Stift
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Lidija Sölkner
- Department of Statistics, Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria
| | - Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Dietmar Öfner
- Department of Visceral-, Transplant- and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
25
|
Huang S, Ye J, Gao X, Huang X, Huang J, Lu L, Lu C, Li Y, Luo M, Xie M, Lin Y, Liang R. Progress of research on molecular targeted therapies for colorectal cancer. Front Pharmacol 2023; 14:1160949. [PMID: 37614311 PMCID: PMC10443711 DOI: 10.3389/fphar.2023.1160949] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies, accounting for approximately 10% of global cancer incidence and mortality. Approximately 20% of patients with CRC present metastatic disease (mCRC) at the time of diagnosis. Moreover, up to 50% of patients with localized disease eventually metastasize. mCRC encompasses a complex cascade of reactions involving multiple factors and processes, leading to a diverse array of molecular mechanisms. Improved comprehension of the pathways underlying cancer cell development and proliferation, coupled with the accessibility of relevant targeted agents, has propelled advancements in CRC treatment, ultimately leading to enhanced survival rates. Mutations in various pathways and location of the primary tumor in CRC influences the efficacy of targeted agents. This review summarizes available targeted agents for different CRC pathways, with a focus on recent advances in anti-angiogenic and anti-epidermal growth factor receptor agents, BRAF mutations, and human epidermal growth factor receptor 2-associated targeted agents.
Collapse
Affiliation(s)
- Shilin Huang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xing Gao
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xi Huang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Julu Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lu Lu
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Cheng Lu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongqiang Li
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Luo
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mingzhi Xie
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yan Lin
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Rong Liang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
26
|
Zhao X, Cai B, Chen H, Wan P, Chen D, Ye Z, Duan A, Chen X, Sun H, Pan J. Tuna trimmings (Thunnas albacares) hydrolysate alleviates immune stress and intestinal mucosal injury during chemotherapy on mice and identification of potentially active peptides. Curr Res Food Sci 2023; 7:100547. [PMID: 37522134 PMCID: PMC10371818 DOI: 10.1016/j.crfs.2023.100547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
In this study, Tuna trimmings (Thunnas albacares) protein hydrolysate (TPA) was produced by alcalase. The anti-tumor synergistic effect and intestinal mucosa protective effect of TPA on S180 tumor-bearing mice treated with 5-fluorouracil (5-FU) chemotherapy were investigated. The results showed that TPA can enhance the anti-tumor effect of 5-FU chemotherapy, as evident by a significant reduction in tumor volume observed in the medium and high dose TPA+5-FU groups compared to the 5-FU group (p < 0.001). Moreover, TPA significantly elevated the content of total protein and albumin in all TPA dose groups (p < 0.01, p < 0.001), indicating its ability to regulate the nutritional status of the mice. Furthermore, histopathological studies revealed a significant increase in the height of small intestinal villi, crypt depth, mucosal thickness, and villi area in the TPA+5-FU groups compared to the 5-FU group (p < 0.05), suggesting that TPA has a protective effect on the intestinal mucosa. Amino acid analysis revealed that TPA had a total amino acid content of 66.30 g/100 g, with essential amino acids accounting for 30.36 g/100 g. Peptide molecular weight distribution analysis of TPA indicated that peptides ranging from 0.25 to 1 kDa constituted 64.54%. LC-MS/MS analysis identified 109 peptide sequences, which were predicted to possess anti-cancer and anti-inflammatory activities through database prediction. Therefore, TPA has the potential to enhance the antitumor effects of 5-FU, mitigate immune depression and intestinal mucosal damage induced by 5-FU. Thus, TPA could be serve as an adjuvant nutritional support for malnourished patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Xiangtan Zhao
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, China
| | - Bingna Cai
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- Southern Marine Science and Engineering Guangdong Laboratory(Guangzhou), Guangzhou, 511458, China
| | - Hua Chen
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- Southern Marine Science and Engineering Guangdong Laboratory(Guangzhou), Guangzhou, 511458, China
| | - Peng Wan
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- Southern Marine Science and Engineering Guangdong Laboratory(Guangzhou), Guangzhou, 511458, China
| | - Deke Chen
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- Southern Marine Science and Engineering Guangdong Laboratory(Guangzhou), Guangzhou, 511458, China
| | - Ziqing Ye
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, China
| | - Ailing Duan
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, China
| | - Xin Chen
- Foshan University, School of Environment and Chemical Engineering, Foshan, 528000, China
| | - Huili Sun
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
| | - Jianyu Pan
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- Southern Marine Science and Engineering Guangdong Laboratory(Guangzhou), Guangzhou, 511458, China
- Innovation Academy of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, China
| |
Collapse
|
27
|
Dragnev KH, Dragnev CPC, Lubet RA. Major hurdles to the use of tyrosine kinase inhibitors in clinical prevention/interception studies: Do preclinical studies with EGFR inhibitors suggest approaches to overcome some of the limitations. Front Cell Dev Biol 2023; 11:1170444. [PMID: 37169023 PMCID: PMC10165497 DOI: 10.3389/fcell.2023.1170444] [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: 02/20/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
There are major hurdles to the use of tyrosine kinase inhibitors (TKIs) and any other agents with significant toxicities (which means practically the preponderance of potential effective agents) in the context of prevention/anti-progression (interception) studies. We will discuss epidermal growth factor receptor (EGFR) inhibitors as examples, both in a primary prevention setting, where agent(s) are administered to individuals with no cancer but who might be considered at higher risk due to a variety of factors, and in anti-progression/interception studies, where agent(s) are administered to persons with known preinvasive lesions (e.g., colon adenomas, lung nodules, ductal carcinoma in situ (DCIS), or pancreatic intraepithelial neoplasia (PanIN) lesions in the pancreas) in an attempt to reverse or inhibit progression of these lesions. Multiple potential hurdles will be examined, including: a) toxicity of agents, b) the likely range of subtypes of cancers affected by a given treatment (e.g., EGFR inhibitors against EGFR mutant lung adenocarcinomas), c) the availability of practical endpoints besides the blocking of cancer formation or pharmacokinetics related to the agents administered in a primary prevention study, and d) the interpretation of the regression or blockage of new preinvasive lesions in the anti-progression study. Such an anti-progression approach may help address some of the factors commented on regarding primary prevention (toxicity, potential target organ cancer subtypes) but still leaves major questions regarding interpretation of modulation of preinvasive endpoints when it may not be clear how frequently they progress to clinical cancer. Additionally, we address whether certain recent preclinical findings might be able to reduce the toxicities associated with these agents and perhaps even increase their potential efficacy. Antibodies and TKIs other than the EGFR inhibitors are not discussed because few if any had been tested as monotherapies in humans, making their efficacy harder to predict, and because a number have relatively rare but quite striking toxicities. Furthermore, most of the practical hurdles raised regarding the EGFR inhibitors are relevant to the other TKIs. Finally, we briefly discuss whether early detection employing blood or serum samples may allow identification of high-risk groups more amenable to agents with greater toxicity.
Collapse
Affiliation(s)
| | | | - Ronald A. Lubet
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, United States
| |
Collapse
|
28
|
Affiliation(s)
- Isabelle Mahé
- Université Paris Cité, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Louis Mourier, Médecine Interne, Colombes, France
- Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris, France
- INNOVTE, FCRIN, France
| | - Florian Scotté
- Gustave Roussy, Patient Pathway Department, Villejuif, France
| | - Ismail Elalamy
- INNOVTE, FCRIN, France
- Hematology and Thrombosis Center, Hôpital Tenon, Hôpitaux Universitaires de l’Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine, Sorbonne Université, Paris, France
- Research Group “Cancer, Vessel Biology and Therapeutics,” INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Faculty of Medicine, Sorbonne University, Paris, France
| |
Collapse
|
29
|
Morton D, Seymour M, Magill L, Handley K, Glasbey J, Glimelius B, Palmer A, Seligmann J, Laurberg S, Murakami K, West N, Quirke P, Gray R. Preoperative Chemotherapy for Operable Colon Cancer: Mature Results of an International Randomized Controlled Trial. J Clin Oncol 2023; 41:1541-1552. [PMID: 36657089 PMCID: PMC10022855 DOI: 10.1200/jco.22.00046] [Citation(s) in RCA: 143] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 08/15/2022] [Accepted: 10/20/2022] [Indexed: 01/20/2023] Open
Abstract
PURPOSE Neoadjuvant chemotherapy (NAC) has potential advantages over standard postoperative chemotherapy for locally advanced colon cancer but requires formal evaluation. METHODS Patients with radiologically staged T3-4, N0-2, M0 colon cancer were randomly allocated (2:1) to 6 weeks oxaliplatin-fluoropyrimidine preoperatively plus 18 postoperatively (NAC group) or 24 weeks postoperatively (control group). Patients with RAS-wildtype tumors could also be randomly assigned 1:1 to receive panitumumab or not during NAC. The primary end point was residual disease or recurrence within 2 years. Secondary outcomes included surgical morbidity, histopathologic stage, regression grade, completeness of resection, and cause-specific mortality. Log-rank analyses were by intention-to-treat. RESULTS Of 699 patients allocated to NAC, 674 (96%) started and 606 (87%) completed NAC. In total, 686 of 699 (98.1%) NAC patients and 351 of 354 (99.2%) control patients underwent surgery. Thirty patients (4.3%) allocated to NAC developed obstructive symptoms requiring expedited surgery, but there were fewer serious postoperative complications with NAC than with control. NAC produced marked T and N downstaging and histologic tumor regression (all P < .001). Resection was more often histopathologically complete: 94% (648/686) versus 89% (311/351), P < .001. Fewer NAC than control patients had residual or recurrent disease within 2 years (16.9% [118/699] v 21.5% [76/354]; rate ratio, 0.72 [95% CI, 0.54 to 0.98]; P = .037). Tumor regression correlated strongly with freedom from recurrence. Panitumumab did not enhance the benefit from NAC. Little benefit from NAC was seen in mismatch repair-deficient tumors. CONCLUSION Six weeks of preoperative oxaliplatin-fluoropyrimidine chemotherapy for operable colon cancer can be delivered safely, without increasing perioperative morbidity. This chemotherapy regimen, when given preoperatively, produces marked histopathologic down-staging, fewer incomplete resections, and better 2-year disease control. Histologic regression after NAC is a strong predictor of lower postoperative recurrence risk so has potential use as a guide for postoperative therapy. Six weeks of NAC should be considered as a treatment option for locally advanced colon cancer.
Collapse
Affiliation(s)
- Dion Morton
- University Hospital Birmingham, Birmingham, United Kingdom
| | | | - Laura Magill
- University of Birmingham Clinical Trials Unit, Birmingham, United Kingdom
| | - Kelly Handley
- University of Birmingham Clinical Trials Unit, Birmingham, United Kingdom
| | - James Glasbey
- University Hospital Birmingham, Birmingham, United Kingdom
| | | | - Andy Palmer
- University of Birmingham Clinical Trials Unit, Birmingham, United Kingdom
| | | | | | - Keigo Murakami
- Division of Pathology and Data Analytics, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Nick West
- Division of Pathology and Data Analytics, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Philip Quirke
- Division of Pathology and Data Analytics, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Richard Gray
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Affiliation(s)
- Julien Taieb
- Department of Hepato-Gastroenterology and GI Oncology, Georges Pompidou European Hospital, Université Paris Cité, SIRIC CARPEM, Paris, France
| | - Mehdi Karoui
- Department of GI and Oncologic Surgery, Georges Pompidou European Hospital, Université Paris Cité, SIRIC CARPEM, Paris, France
| |
Collapse
|
31
|
Etienne-Grimaldi MC, Pallet N, Boige V, Ciccolini J, Chouchana L, Barin-Le Guellec C, Zaanan A, Narjoz C, Taieb J, Thomas F, Loriot MA. Current diagnostic and clinical issues of screening for dihydropyrimidine dehydrogenase deficiency. Eur J Cancer 2023; 181:3-17. [PMID: 36621118 DOI: 10.1016/j.ejca.2022.11.028] [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: 06/13/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
Fluoropyrimidine drugs (FP) are the backbone of many chemotherapy protocols for treating solid tumours. The rate-limiting step of fluoropyrimidine catabolism is dihydropyrimidine dehydrogenase (DPD), and deficiency in DPD activity can result in severe and even fatal toxicity. In this review, we survey the evidence-based pharmacogenetics and therapeutic recommendations regarding DPYD (the gene encoding DPD) genotyping and DPD phenotyping to prevent toxicity and optimize dosing adaptation before FP administration. The French experience of mandatory DPD-deficiency screening prior to initiating FP is discussed.
Collapse
Affiliation(s)
| | - Nicolas Pallet
- Department of Clinical Chemistry, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France
| | - Valérie Boige
- Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France; Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Joseph Ciccolini
- SMARTc, CRCM INSERM U1068, Université Aix-Marseille, Marseille, France; Laboratory of Pharmacokinetics and Toxicology, Hôpital Universitaire La Timone, F-13385 Marseille, France; COMPO, CRCM INSERM U1068-Inria, Université Aix-Marseille, Marseille, France
| | - Laurent Chouchana
- Regional Center of Pharmacovigilance, Department of Pharmacology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, Université de Paris, Paris, France; French Pharmacovigilance Network, France
| | - Chantal Barin-Le Guellec
- Laboratory of Biochemistry and Molecular Biology, Centre Hospitalo-uinversitaire de Tours, Tours, France; INSERM U1248, IPPRITT, University of Limoges, Limoges, France
| | - Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris University; Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Céline Narjoz
- Department of Clinical Chemistry, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France
| | - Julien Taieb
- SIRIC CARPEM, Université de Paris; Fédération Francophone de Cancérologie Digestive (FFCD), Assistance Publique-Hôpitaux de Paris, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Fabienne Thomas
- Laboratory of Pharmacology, Institut Claudius Regaud, IUCT-Oncopole and CRCT, INSERM UMR1037, Université Paul Sabatier, Toulouse, France
| | - Marie-Anne Loriot
- Department of Clinical Chemistry, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, F-75006 Paris, France.
| | | |
Collapse
|
32
|
Zhang L, Deng Y, Liu S, Zhang W, Hong Z, Lu Z, Pan Z, Wu X, Peng J. Lymphovascular invasion represents a superior prognostic and predictive pathological factor of the duration of adjuvant chemotherapy for stage III colon cancer patients. BMC Cancer 2023; 23:3. [PMID: 36593480 PMCID: PMC9808960 DOI: 10.1186/s12885-022-10416-7] [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: 08/09/2022] [Accepted: 12/06/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Lymphovascular invasion (LVI) and perineural invasion (PNI) can indicate poor survival outcomes in colorectal cancer, but few studies have focused on stage III colon cancer. The current study aimed to confirm the prognostic value of LVI and PNI and identify patients who could benefit from a complete duration of adjuvant chemotherapy based on the two pathological factors. METHODS We enrolled 402 consecutive patients with stage III colon cancer who received colon tumor resection from November 2007 to June 2016 at Sun Yat-sen University Cancer Center. Survival analyses were performed by using Kaplan-Meier method with log-rank tests. Risk factors related to disease-free survival (DFS) and overall survival (OS) were identified through Cox proportional hazards analysis. RESULTS 141 (35.1%) patients presented with LVI, and 108 (26.9%) patients with PNI. The LVI-positive group was associated with poorer 3-year DFS (86.5% vs. 76.3%, P = 0.001) and OS (96.0% vs. 89.1%, P = 0.003) rates compared with the LVI-negative group. The PNI-positive group showed a worse outcome compared with the PNI-negative group in 3-year DFS rate (72.5% vs. 86.7%, P < 0.001). Moreover, LVI-positive group present better 3-year DFS and OS rate in patients completing 6-8 cycles of adjuvant chemotherapy than those less than 6 cycles (3-year DFS: 80.0% vs. 64.9%, P = 0.019; 3-year OS: 93.2% vs. 76.3%, P = 0.002). CONCLUSIONS LVI is a superior prognostic factor to PNI in stage III colon cancer patients undergoing curative treatment. PNI status can noly predict the 3-year DFS wihout affecting the 3-year OS. Furthermore, LVI also represents an effective indicator for adjuvant chemotherapy duration.
Collapse
Affiliation(s)
- Linjie Zhang
- grid.488530.20000 0004 1803 6191Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| | - Yuxiang Deng
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518000 Shenzhen, P. R. China
| | - Songran Liu
- grid.488530.20000 0004 1803 6191Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| | - Weili Zhang
- grid.488530.20000 0004 1803 6191Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| | - Zhigang Hong
- grid.488530.20000 0004 1803 6191Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| | - Zhenhai Lu
- grid.488530.20000 0004 1803 6191Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| | - Zhizhong Pan
- grid.488530.20000 0004 1803 6191Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| | - Xiaojun Wu
- grid.488530.20000 0004 1803 6191Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| | - Jianhong Peng
- grid.488530.20000 0004 1803 6191Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangdong 510060 Guangzhou, P. R. China
| |
Collapse
|
33
|
Crutcher M, Waldman S. Biomarkers in the development of individualized treatment regimens for colorectal cancer. Front Med (Lausanne) 2022; 9:1062423. [DOI: 10.3389/fmed.2022.1062423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
IntroductionColorectal cancer (CRC) is the third most common and second most deadly malignancy in the world with an estimated 1. 9 million cases and 0.9 million deaths in 2020. The 5-year overall survival for stage I disease is 92% compared to a dismal 11% in stage IV disease. At initial presentation, up to 35% of patients have metastatic colorectal cancer (mCRC), and 20–50% of stage II and III patients eventually progress to mCRC. These statistics imply both that there is a proportion of early stage patients who are not receiving adequate treatment and that we are not adequately treating mCRC patients.BodyTargeted therapies directed at CRC biomarkers are now commonly used in select mCRC patients. In addition to acting as direct targets, these biomarkers also could help stratify which patients receive adjuvant therapies and what types. This review discusses the role of RAS, microsatellite instability, HER2, consensus molecular subtypes and ctDNA/CTC in targeted therapy and adjuvant chemotherapy.DiscussionGiven the relatively high recurrence rate in early stage CRC patients as well as the continued poor survival in mCRC patients, additional work needs to be done beyond surgical management to limit recurrence and improve survival. Biomarkers offer both a potential target and a predictive method of stratifying patients to determine those who could benefit from adjuvant treatment.
Collapse
|
34
|
Crutcher MM, Snook AE, Waldman SA. Overview of predictive and prognostic biomarkers and their importance in developing a clinical pharmacology treatment plan in colorectal cancer patients. Expert Rev Clin Pharmacol 2022; 15:1317-1326. [PMID: 36259230 PMCID: PMC9847576 DOI: 10.1080/17512433.2022.2138339] [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: 04/19/2022] [Accepted: 10/14/2022] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the second most common cause of cancer-related death worldwide. Although overall survival for CRC patients has improved with earlier screening, survival continues to vary substantially across stages. Also, while the introduction of targeted therapies, including VEGF and EGFR inhibitors, has contributed to improving survival, better tools are needed to optimize patient selection and maximize therapeutic benefits. Emerging biomarkers can be used to guide pharmacologic decision-making, as well as monitor treatment response, clarify the need for adjuvant therapies, and indicate early signs of recurrence. This is a narrative review examining the current and evolving use of predictive and prognostic biomarkers in colorectal cancer. AREAS COVERED Areas covered include mutations of the MAPK (KRAS, BRAF) and HER2 pathways and their impacts on treatment decisions. In addition, novel methods for assessing tumor mutations and tracking treatment responses are examined. EXPERT OPINION The standard of care pathway for staging, and treatment selection and surveillance, of CRC will expand to include novel biomarkers in the next 5 years. It is anticipated that these new biomarkers will assist in decision-making regarding selection of targeted therapies and, importantly, in risk stratification for treatment decisions in patients at high risk for recurrence.
Collapse
Affiliation(s)
| | - Adam E. Snook
- Departmnet of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
- Department of Microbiology & Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Scott A. Waldman
- Departmnet of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
35
|
Sueda T, Tei M, Mori S, Nishida K, Yoshikawa Y, Nomura M, Matsumura T, Koga C, Miyagaki H, Tsujie M, Akamaru Y. Oncological outcomes following minimally invasive surgery for pathological N2M0 colorectal cancer: A propensity score-matched analysis. Asian J Endosc Surg 2022; 15:781-793. [PMID: 35715936 DOI: 10.1111/ases.13094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Whether minimally invasive surgery (MIS) is safe and effective for patients with N2M0 colorectal cancer (CRC) remains controversial. This study aimed to compare short- and long-term outcomes between MIS and open surgery (Open) groups for patients with pathological (p)N2M0 CRC, and evaluate the oncological outcomes of MIS for pN2M0 CRC. MATERIALS AND METHODS We retrospectively analyzed 125 consecutive patients with pN2M0 CRC who underwent curative surgery between 2010 and 2017, using propensity score-matching (PSM) analysis. RESULTS Median follow-up was 59.4 months. After PSM, we enrolled 68 patients (n = 34 in each group). The conversion rate was 9.6% for the entire patient cohort and 5.9% for the matched cohort. In colon cancer (CC), short-term outcomes were similar between groups. On the other hand, in rectal cancer (RC), estimated blood loss, rate of anastomosis leakage, and length of postsurgical stay were lower in the MIS group than the Open group. R0 resection was achieved in all patients with MIS. No surgical mortality was encountered in any group. No significant differences were found between groups in terms of 3-year local recurrence rate, overall survival, cancer-specific survival, or recurrence-free survival among the entire patient cohort or the matched cohort, regardless of the primary tumor site (CC or RC). Surgical approach (MIS vs Open) had no significant influence on survival outcomes. CONCLUSIONS MIS is a safe and effective option for patients with pN2M0 CRC, with acceptable short- and long-term outcomes comparable to the open approach. MIS can be considered for patients with pN2M0 CRC.
Collapse
Affiliation(s)
- Toshinori Sueda
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Mitsuyoshi Tei
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Soichiro Mori
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Kentaro Nishida
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Yukihiro Yoshikawa
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Masatoshi Nomura
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Tae Matsumura
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Chikato Koga
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Hiromichi Miyagaki
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Masanori Tsujie
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Yusuke Akamaru
- Department of Gastroenterological Surgery, Osaka Rosai Hospital, Osaka, Japan
| |
Collapse
|
36
|
Macchi L, Bao QR, Albertoni L, Fassan M, Chiminazzo V, Scarpa M, Spolverato G, Pucciarelli S. Prognostic significance of additional histologic features for subclassification of pathological T3 colon cancer. Int J Clin Oncol 2022; 27:1428-1438. [PMID: 35716324 PMCID: PMC9393148 DOI: 10.1007/s10147-022-02192-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/18/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Additional histologic features of T3 colon cancer, such as tumour depth invasion beyond muscularis propria and elastic lamina invasion (ELI), have taken interest for a more accurate staging. METHODS Patients with pT3 and pT4a (control group) colon adenocarcinoma were retrospectively collected from our institutional database. The study group was divided according to depth of tumour invasion < 5 mm and ≥ 5 mm, and into ELI - and ELI + . Chi-square test was used to compare the clinicopathological characteristics. OS and DFS were estimated using Kaplan-Meier method and compared with the log-rank test. Univariable and multivariable Cox proportional hazard models were employed to assess the effect on OS and DFS. RESULTS Out of 290 pT3 tumours, 168 (58%) had a depth of tumour invasion < 5 mm and 122 (42%) ≥ 5 mm. The 5-year OS and DFS were 85.2, 68.7 and 60.9%, and 81.4, 73.9 and 60.1% in pT3 < 5 mm, pT3 ≥ 5 mm, and pT4a respectively (p = 0.001, p = 0.072). Considering ELI - (n = 157, 54%) and ELI + (n = 133, 46%), the 5-year OS and DFS were 78.9, 76.7, and 60.9%, and 75.5, 81.5, and 60.1% in ELI - , ELI + and pT4a respectively (p = 0.955, p = 0.462). At multivariable analysis, the depth of invasion was found to be an independent predictive factor for OS (HR 2.04, 95%CI 1.28-3.24, p = 0.003) and DFS (HR 1.98, 95%CI 1.24-3.18, p = 0.004), while ELI did not result a prognostic factor for OS nor DFS. CONCLUSION In pT3 colon cancer, depth of tumour invasion ≥ 5 mm is an independent risk factor for OS and DFS, whereas ELI did not result a prognostic factor affecting OS nor DFS.
Collapse
Affiliation(s)
- Lorenzo Macchi
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Quoc Riccardo Bao
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Laura Albertoni
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padova, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padova, Padua, Italy
- Veneto Institute of Oncology (I.O.V. IRCSS), Padua, Italy
| | - Valentina Chiminazzo
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Marco Scarpa
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Gaya Spolverato
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128, Padua, Italy.
| | - Salvatore Pucciarelli
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| |
Collapse
|
37
|
Modest DP, Karthaus M, Kasper S, Moosmann N, Keitel V, Kiani A, Uhlig J, Jacobasch L, Fischer V Weikersthal L, Fuchs M, Kaiser F, Lerchenmüller C, Sent D, Junghanß C, Held S, Lorenzen S, Kaczirek K, Jung A, Stintzing S, Heinemann V. FOLFOX plus panitumumab or FOLFOX alone as additive therapy following R0/1 resection of RAS wild-type colorectal cancer liver metastases - The PARLIM trial (AIO KRK 0314). Eur J Cancer 2022; 173:297-306. [PMID: 35970102 DOI: 10.1016/j.ejca.2022.07.012] [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: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE This trial investigates the addition of panitumumab to chemotherapy with fluorouracil/folinic acid and oxaliplatin (FOLFOX) in a 2:1 randomised, controlled, open-label, phase II trial in RAS wild-type colorectal cancer patients with R0/1-resected liver metastases. EXPERIMENTAL DESIGN The primary endpoint was progression-free survival (PFS) two years after randomisation. The experimental arm (12 weeks of biweekly mFOLFOX6 plus panitumumab followed by 12 weeks of panitumumab alone) was considered active if the two-year PFS rate was ≥65%. Based on historical data, a two-year PFS rate of 50% was estimated in the control arm (12 weeks of biweekly FOLFOX). The trial was performed with a power of 80% and an alpha of 0.05. Secondary endpoints included overall survival (OS) and toxicity. The trial is registered with ClinicalTrials.gov, NCT01384994. RESULTS The full analysis set consists of 70 patients (pts) in the experimental arm and 36 pts in the control arm. The primary endpoint was missed with a two-year PFS of 35.7% with FOLFOX plus panitumumab and 30.6% in the control arm. In comparative analyses, trends towards improved PFS (HR 0.83; 95%CI, 0.52-1.33; P = 0.44) and OS (HR 0.70; 95% CI, 0.34-1.46; P = 0.34) were observed in favour of the panitumumab-based study arm. No new or unexpected safety signals were observed with FOLFOX plus panitumumab following liver resection. CONCLUSION The PARLIM trial failed to demonstrate a two-year PFS rate of 65% after resection of colorectal liver metastases. The positive trends in survival endpoints may support future trials evaluating treatment with anti-EGFR agents after resection of liver metastases.
Collapse
Affiliation(s)
- Dominik Paul Modest
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Cancer Immunology, Charitéplatz 1, 10117, Berlin, Germany; German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| | - Meinolf Karthaus
- Department of Hematology and Oncology, Munich Hospital Neuperlach, Munich, Germany
| | | | | | - Verena Keitel
- Department of Gastroenterology, University of Düsseldorf, Germany
| | | | | | | | | | - Martin Fuchs
- Department of Gastroenterology, Munich Hospital Bogenhausen, Munich, Germany
| | - Florian Kaiser
- Practice Oncology, Hospital Landshut-Achdorf, Landshut, Germany
| | | | - Dagmar Sent
- Oncological Practice, MVZ Hospital Leverkusen, Leverkusen, Germany
| | | | | | - Sylvie Lorenzen
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany; Department of Internal Medicine III (Haematology/Medical Oncology), Technical University of Munich Hospital Rechts der Isar, Munchen, Bayern, Germany
| | - Klaus Kaczirek
- Department of Surgery, University of Vienna, Vienna, Austria
| | - Andreas Jung
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Ludwig Maximillians Universität (LMU), Munich, Germany
| | - Sebastian Stintzing
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Cancer Immunology, Charitéplatz 1, 10117, Berlin, Germany; German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Volker Heinemann
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany; Department of Medicine III & Comprehensive Cancer Center, Hospital of the University (LMU), München, Germany
| |
Collapse
|
38
|
Pelullo M, Zema S, De Carolis M, Cialfi S, Giuli MV, Palermo R, Capalbo C, Giannini G, Screpanti I, Checquolo S, Bellavia D. 5FU/Oxaliplatin-Induced Jagged1 Cleavage Counteracts Apoptosis Induction in Colorectal Cancer: A Novel Mechanism of Intrinsic Drug Resistance. Front Oncol 2022; 12:918763. [PMID: 35847908 PMCID: PMC9283835 DOI: 10.3389/fonc.2022.918763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022] Open
Abstract
Colorectal cancer (CRC) is characterized by early metastasis, resistance to anti-cancer therapy, and high mortality rate. Despite considerable progress in the development of new treatment options that improved survival benefits in patients with early-stage or advanced CRC, many patients relapse due to the activation of intrinsic or acquired chemoresistance mechanisms. Recently, we reported novel findings about the role of Jagged1 in CRC tumors with Kras signatures. We showed that Jagged1 is a novel proteolytic target of Kras signaling, which induces Jagged1 processing/activation resulting in Jag1-ICD release, which favors tumor development in vivo, through a non-canonical mechanism. Herein, we demonstrate that OXP and 5FU cause a strong accumulation of Jag1-ICD oncogene, through ERK1/2 activation, unveiling a surviving subpopulation with an enforced Jag1-ICD expression, presenting the ability to counteract OXP/5FU-induced apoptosis. Remarkably, we also clarify the clinical ineffectiveness of γ-secretase inhibitors (GSIs) in metastatic CRC (mCRC) patients. Indeed, we show that GSI compounds trigger Jag1-ICD release, which promotes cellular growth and EMT processes, functioning as tumor-promoting agents in CRC cells overexpressing Jagged1. We finally demonstrate that Jagged1 silencing in OXP- or 5FU-resistant subpopulations is enough to restore the sensitivity to chemotherapy, confirming that drug sensitivity/resistance is Jag1-ICD-dependent, suggesting Jagged1 as a molecular predictive marker for the outcome of chemotherapy.
Collapse
Affiliation(s)
- Maria Pelullo
- CLN2S - Center for Life Nano- & Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Sabrina Zema
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Mariangela De Carolis
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Samantha Cialfi
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Maria Valeria Giuli
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Rocco Palermo
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Carlo Capalbo
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Giuseppe Giannini
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Isabella Screpanti
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University, Latina, Italy
- *Correspondence: Saula Checquolo, ; Diana Bellavia,
| | - Diana Bellavia
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy
- *Correspondence: Saula Checquolo, ; Diana Bellavia,
| |
Collapse
|
39
|
André L, Antherieu G, Boinet A, Bret J, Gilbert T, Boulahssass R, Falandry C. Oncological Treatment-Related Fatigue in Oncogeriatrics: A Scoping Review. Cancers (Basel) 2022; 14:2470. [PMID: 35626074 PMCID: PMC9139887 DOI: 10.3390/cancers14102470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 12/17/2022] Open
Abstract
Fatigue is a highly prevalent symptom in both cancer patients and the older population, and it contributes to quality-of-life impairment. Cancer treatment-related fatigue should thus be included in the risk/benefit assessment when introducing any treatment, but tools are lacking to a priori estimate such risk. This scoping review was designed to report the current evidence regarding the frequency of fatigue for the different treatment regimens proposed for the main cancer indications, with a specific focus on age-specific data, for the following tumors: breast, ovary, prostate, urothelium, colon, lung and lymphoma. Fatigue was most frequently reported using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) versions 3 to 5. A total of 324 regimens were analyzed; data on fatigue were available for 217 (67%) of them, and data specific to older patients were available for 35 (11%) of them; recent pivotal trials have generally reported more fatigue grades than older studies, illustrating increasing concern over time. This scoping review presents an easy-to-understand summary that is expected to provide helpful information for shared decisions with patients regarding the anticipation and prevention of fatigue during each cancer treatment.
Collapse
Affiliation(s)
- Louise André
- Hospices Civils de Lyon, Geriatrics Department, Hôpital Lyon Sud, 69230 Saint Genis-Laval, France; (L.A.); (G.A.); (A.B.); (J.B.); (T.G.)
| | - Gabriel Antherieu
- Hospices Civils de Lyon, Geriatrics Department, Hôpital Lyon Sud, 69230 Saint Genis-Laval, France; (L.A.); (G.A.); (A.B.); (J.B.); (T.G.)
| | - Amélie Boinet
- Hospices Civils de Lyon, Geriatrics Department, Hôpital Lyon Sud, 69230 Saint Genis-Laval, France; (L.A.); (G.A.); (A.B.); (J.B.); (T.G.)
| | - Judith Bret
- Hospices Civils de Lyon, Geriatrics Department, Hôpital Lyon Sud, 69230 Saint Genis-Laval, France; (L.A.); (G.A.); (A.B.); (J.B.); (T.G.)
| | - Thomas Gilbert
- Hospices Civils de Lyon, Geriatrics Department, Hôpital Lyon Sud, 69230 Saint Genis-Laval, France; (L.A.); (G.A.); (A.B.); (J.B.); (T.G.)
- Research on Healthcare Professionals and Performance RESHAPE, Inserm U1290, Lyon 1 University, 69008 Lyon, France
| | - Rabia Boulahssass
- Geriatric Coordination Unit for Geriatric Oncology (UCOG) PACA Est CHU de Nice, 06000 Nice, France;
- FHU OncoAge, 06000 Nice, France
- Faculty of Medicine, University of Nice Sofia Antilpolis, 06000 Nice, France
| | - Claire Falandry
- Hospices Civils de Lyon, Geriatrics Department, Hôpital Lyon Sud, 69230 Saint Genis-Laval, France; (L.A.); (G.A.); (A.B.); (J.B.); (T.G.)
- FHU OncoAge, 06000 Nice, France
- CarMeN Laboratory, INSERM U.1060/Université Lyon1/INRA U. 1397/INSA Lyon/Hospices Civils Lyon, Bâtiment CENS-ELI 2D, Hôpital Lyon Sud Secteur 2, 69310 Pierre-Bénite, France
- UCOGIR—Auvergne-Rhône-Alpes Ouest–Guyane, Hôpital Lyon Sud, 69495 Pierre-Bénite, France
- Faculty of Medicine and Maieutics Charles Mérieux, Lyon 1 University, 69310 Pierre-Bénite, France
| |
Collapse
|
40
|
Association between genetic mutations and risk of venous thromboembolism in patients with solid tumor malignancies: A systematic review and meta-analysis. Thromb Res 2022; 213:47-56. [DOI: 10.1016/j.thromres.2022.02.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/02/2022] [Accepted: 02/23/2022] [Indexed: 01/08/2023]
|
41
|
Lipsyc-Sharf M, Ou FS, Yurgelun MB, Rubinson DA, Schrag D, Dakhil SR, Stella PJ, Weckstein DJ, Wender DB, Faggen M, Zemla TJ, Heying EN, Schuetz SR, Noble S, Meyerhardt JA, Bekaii-Saab T, Fuchs CS, Ng K. Cetuximab and Irinotecan With or Without Bevacizumab in Refractory Metastatic Colorectal Cancer: BOND-3, an ACCRU Network Randomized Clinical Trial. Oncologist 2022; 27:292-298. [PMID: 35380713 PMCID: PMC8982431 DOI: 10.1093/oncolo/oyab025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background Combination irinotecan and cetuximab is approved for irinotecan-refractory metastatic colorectal cancer (mCRC). It is unknown if adding bevacizumab improves outcomes. Patients and Methods In this multicenter, randomized, double-blind, placebo-controlled phase II trial, patients with irinotecan-refractory RAS-wildtype mCRC and no prior anti-EGFR therapy were randomized to cetuximab 500 mg/m2, bevacizumab 5 mg/kg, and irinotecan 180 mg/m2 (or previously tolerated dose) (CBI) versus cetuximab, irinotecan, and placebo (CI) every 2 weeks until disease progression or intolerable toxicity. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), objective response rate (ORR), and adverse events (AEs). Results The study closed early after the accrual of 36 out of a planned 120 patients due to changes in funding. Nineteen patients were randomized to CBI and 17 to CI. Baseline characteristics were similar between arms. Median PFS was 9.7 versus 5.5 months for CBI and CI, respectively (1-sided log-rank P = .38; adjusted hazard ratio [HR] = 0.64; 95% confidence interval [CI], 0.25-1.66). Median OS was 19.7 versus 10.2 months for CBI and CI (1-sided log-rank P = .02; adjusted HR = 0.41; 95% CI, 0.15-1.09). ORR was 36.8% for CBI versus 11.8% for CI (P = .13). Grade 3 or higher AEs occurred in 47% of patients receiving CBI versus 35% for CI (P = .46). Conclusion In this prematurely discontinued trial, there was no significant difference in the primary endpoint of PFS between CBI and CI. There was a statistically significant improvement in OS in favor of CBI compared with CI. Further investigation of CBI for the treatment of irinotecan-refractory mCRC is warranted. Clinical Trial Registration: NCT02292758
Collapse
Affiliation(s)
- Marla Lipsyc-Sharf
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fang-Shu Ou
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Douglas A Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Deborah Schrag
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | - Meredith Faggen
- Dana-Farber at South Shore Hospital, South Weymouth, MA, USA
| | - Tyler J Zemla
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Erica N Heying
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Charles S Fuchs
- Yale Cancer Center, New Haven, CT, USA
- Genentech, South San Francisco, CA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
42
|
Wei L, Lin Z, Xie S, Ruan D, Jiang W, Cui Y, Liu S, Wang T, Chen Z, Lin Q. Complete Response With Cetuximab-Based Treatment of Metastatic Colorectal Cancers: Two Case Reports and Literature Review. Front Oncol 2022; 12:798515. [PMID: 35251973 PMCID: PMC8888900 DOI: 10.3389/fonc.2022.798515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/21/2022] [Indexed: 12/24/2022] Open
Abstract
Metastases typically develop before diagnosis and during the treatment of colorectal cancers, while patients with metastatic colorectal cancers (mCRCs) currently have a poor prognosis. In terms of surgical approaches, adjuvant therapies, and targeted therapies, the treatment of mCRCs has had numerous recent advances. As a targeted agent widely used in mCRCs, cetuximab-based treatment is still under dispute due to its side effects and unstable effect. We present two mCRC cases treated with cetuximab-based therapy, of which two patients achieved complete response and without recurrence for over 22 and 84 months, respectively. To better understand the drug usage, we also reviewed the recent achievements and usage precautions of cetuximab in mCRCs. Present and many previous observations support that cetuximab might be a referred drug in the first-line chemotherapy of mCRCs with wild-type RAS and BRAF and proficient mismatch repair.
Collapse
Affiliation(s)
- Li Wei
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zexiao Lin
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sidong Xie
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Danyun Ruan
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wen Jiang
- Department of Research and Development, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Yueli Cui
- Department of Research and Development, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Sisi Liu
- Department of Research and Development, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Tiantian Wang
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Tiantian Wang, ; Zhanhong Chen, ; Qu Lin,
| | - Zhanhong Chen
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Tiantian Wang, ; Zhanhong Chen, ; Qu Lin,
| | - Qu Lin
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Tiantian Wang, ; Zhanhong Chen, ; Qu Lin,
| |
Collapse
|
43
|
Asada Y, Chinen K, Yamataka K, Tokuyama J, Kurihara N, Iida S. Pathological complete response after neoadjuvant chemotherapy with FOLFOX for locally advanced sigmoid colon cancer with diverticulitis: A case report. Int J Surg Case Rep 2022; 90:106685. [PMID: 34952318 PMCID: PMC8714994 DOI: 10.1016/j.ijscr.2021.106685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/28/2022] Open
Abstract
Introduction and importance The standard treatment for locally advanced colon cancer (LACC) without distant metastasis is curative surgery followed by adjuvant chemotherapy, but the long-term outcomes of this strategy are not satisfactory. Neoadjuvant chemotherapy (NAC) is a promising novel option to overcome this issue. Tumor regression is an expected effect of NAC for LACC, but pathological complete response (pCR) is rare. In this report, we present a rare case of pCR after NAC with FOLFOX for LACC in the sigmoid colon. Presentation of case A 66-year-old woman presented to our hospital with fever and abdominal pain. The diagnosis was LACC in the sigmoid colon with possible invasion of the uterus and pelvic wall, stage IIIC (T4bN1bM0). Furthermore, the tumor was complicated by diverticulitis. A colostomy was performed, followed by NAC with FOLFOX. Six cycles were completed without significant adverse events, and the lesion shrunk remarkably. We performed a curative sigmoidectomy without any postoperative complications. Pathological examination revealed no viable cancer cells, indicating pCR. Discussion To the best of our knowledge, this is the first report of pCR after NAC for LACC complicated by diverticulitis. Colostomy before NAC, regimen, and cycle of NAC may be the key to this favorable course. Conclusion We present a rare case of pathological complete after neoadjuvant chemotherapy with FOLFOX for locally advanced colon cancer in the sigmoid colon complicated by diverticulitis. Our experience may be valuable in determining the optimal treatment strategy for LACC complicated by diverticulitis. Neoadjuvant chemotherapy is a promising novel option for locally advanced colon cancer. Pathological complete response is rare. Indications for cases complicated with diverticulitis are unknown. We present a rare case of pCR after NAC for LACC complicated by diverticulitis.
Collapse
Affiliation(s)
- Yusuke Asada
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan.
| | - Katsuya Chinen
- Department of Pathology, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Ken Yamataka
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Jo Tokuyama
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Naoto Kurihara
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Shuhei Iida
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| |
Collapse
|
44
|
Sabbagh C, Manceau G, Mege D, Abdalla S, Voron T, Bridoux V, Lakkis Z, Venara A, Beyer-Berjot L, Diouf M, Karoui M. Is Adjuvant Chemotherapy Necessary for Obstructing Stage II Colon Cancer? Results From a Propensity Score Analysis of the French Surgical Association Database. Ann Surg 2022; 275:149-156. [PMID: 32068553 DOI: 10.1097/sla.0000000000003832] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of this study was to compare the survival of patients with stage II obstructing colon cancer (OCC) who had adjuvant chemotherapy with those who did not. SUMMARY BACKGROUND DATA The need for adjuvant chemotherapy in stage II colon cancer is still debated. METHODS All consecutive patients treated for a stage II OCC in a curative intent (with primary tumor resection) between January 2000 and December 2015 were included in this retrospective, multicenter cohort study which included a propensity score analysis using an odds of treatment weighting (Average Treatment effect on the Treated, ATT). The endpoint was the comparison between the 2 groups for overall survival (OS) and disease-free survival (DFS) according to whether or not patients received adjuvant chemotherapy. RESULTS During the study period, 504 patients underwent a curative colectomy for a stage II OCC. Among these patients, 179 (35.5%) had adjuvant chemotherapy and 325 (64.5%) had no adjuvant treatment. Among the 179 patients who received adjuvant chemotherapy, 108 patients (60%) received oxaliplatin based regimen and 99 patients (55%) completed all scheduled cycles. At multivariate analysis, after weighting by the odds (ATT analysis) and adjustment, adjuvant chemotherapy after resection of a stage II OCC was associated with improvements in OS [hazard ratio (HR) = 0.42 (0.17-0.99), P = 0.0498] and DFS [HR = 0.57 (0.37-0.88), P = 0.0116]. CONCLUSION This study suggests that adjuvant chemotherapy after curative resection of stage II OCC may improve oncological outcomes.
Collapse
Affiliation(s)
- Charles Sabbagh
- Department of Digestive Surgery, Amiens University Hospital, Amiens, France
| | - Gilles Manceau
- Sorbonne Université, Assistance Publique Hôpitaux de Paris, Pitié Salpêtrière University Hospital, Department of Digestive Surgery, Paris, France
| | - Diane Mege
- Department of Digestive Surgery, Timone University Hospital, Marseille, France
| | - Solafah Abdalla
- Université Paris-Sud, Assistance Publique Hôpitaux de Paris, Bicêtre University Hospital, Department of Digestive Surgery, Le Kremlin Bicêtre, France
| | - Thibault Voron
- Sorbonne Université, Assistance Publique Hôpitaux de Paris, Saint Antoine University Hospital, Department of Digestive Surgery, Paris, France
| | - Valérie Bridoux
- Department of Digestive Surgery, Charles Nicolle University Hospital, Rouen, France
| | - Zaher Lakkis
- Department of Digestive Surgery, Besançon University Hospital, Besançon, France
| | - Aurélien Venara
- Department of Digestive Surgery, Angers University Hospital, Angers, France
| | - Laura Beyer-Berjot
- Department of Digestive Surgery, Assistance Publique Hôpitaux de Marseille, North University Hospital, Marseille, France
| | - Momar Diouf
- Department of Clinical Research and Innovation, Amiens University Hospital, France
| | - Mehdi Karoui
- Sorbonne Université, Assistance Publique Hôpitaux de Paris, Pitié Salpêtrière University Hospital, Department of Digestive Surgery, Paris, France
| |
Collapse
|
45
|
Howe JR. The impact of DNA testing on management of patients with colorectal cancer. Ann Gastroenterol Surg 2022; 6:17-28. [PMID: 35106411 PMCID: PMC8786701 DOI: 10.1002/ags3.12526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/18/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Knowledge of the genetic basis of colorectal cancer has evolved over the past decades, allowing for the pre-symptomatic identification of affected patients in those with familial syndromes and to the understanding of the multi-step progression to carcinogenesis in tumors. Knowledge of the genes and pathways involved in colorectal cancer has allowed for targeted therapies in patients in addition to standard chemotherapy for those with metastases. Next-generation sequencing technologies have now also allowed for the sensitive detection of circulating mutations derived from tumors, which can give insight into the presence of residual disease and has implications for changing the standard paradigms for treatment. This article will specifically review advances in targeted therapy in metastatic colon cancer and the progress being made in using circulating tumor DNA in patient management.
Collapse
Affiliation(s)
- James R. Howe
- Department of SurgeryUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
| |
Collapse
|
46
|
Oki E, Ando K, Taniguchi H, Yoshino T, Mori M. Sustainable Clinical Development of Adjuvant Chemotherapy for Colon Cancer. Ann Gastroenterol Surg 2022; 6:37-45. [PMID: 35106413 PMCID: PMC8786685 DOI: 10.1002/ags3.12503] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/28/2021] [Accepted: 08/26/2021] [Indexed: 01/05/2023] Open
Abstract
Numerous clinical studies in an adjuvant setting have been conducted and the combination therapy of 5-fluorouracil and oxaliplatin has been established as the standard treatment for Stage III and as an option for high-risk Stage II patients. Biologics such as bevacizumab and antiepidermal growth factor receptor antibodies have failed to show additional survival benefits. The indication of adjuvant chemotherapy has been determined according to the pathological stage. Nevertheless, a pathological diagnosis does not necessarily result in selection of the optimal treatment. To improve treatment decisions, many trials have aimed to stratify patients into treatment groups using genomic testing. Recently, gene signature, Immunoscore, and circulating tumor DNA (ctDNA) assays have been reported and among them, ctDNA was shown to be a promising accurate predictive marker for recurrence. Treatment of ctDNA-positive patients with aggressive chemotherapy may reduce recurrence rates. The ultimate goal is to accurately predict the risk of recurrence and to prevent recurrence in colon cancer patients. In this review we focus on the clinical development of adjuvant chemotherapy and stratification of patients according to risk of recurrence and the future direction of adjuvant chemotherapy.
Collapse
Affiliation(s)
- Eiji Oki
- Department of Surgery and ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koji Ando
- Department of Surgery and ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | | | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Masaki Mori
- Tokai University School of MedicineIseharaJapan
| |
Collapse
|
47
|
Chen K, Collins G, Wang H, Toh JWT. Pathological Features and Prognostication in Colorectal Cancer. Curr Oncol 2021; 28:5356-5383. [PMID: 34940086 PMCID: PMC8700531 DOI: 10.3390/curroncol28060447] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/01/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023] Open
Abstract
The prognostication of colorectal cancer (CRC) has traditionally relied on staging as defined by the Union for International Cancer Control (UICC) and American Joint Committee on Cancer (AJCC) TNM staging classifications. However, clinically, there appears to be differences in survival patterns independent of stage, suggesting a complex interaction of stage, pathological features, and biomarkers playing a role in guiding prognosis, risk stratification, and guiding neoadjuvant and adjuvant therapies. Histological features such as tumour budding, perineural invasion, apical lymph node involvement, lymph node yield, lymph node ratio, and molecular features such as MSI, KRAS, BRAF, and CDX2 may assist in prognostication and optimising adjuvant treatment. This study provides a comprehensive review of the pathological features and biomarkers that are important in the prognostication and treatment of CRC. We review the importance of pathological features and biomarkers that may be important in colorectal cancer based on the current evidence in the literature.
Collapse
Affiliation(s)
- Kabytto Chen
- Discipline of Surgery, Faculty of Medicine and Health, The University of Sydney, Westmead 2145, Australia; (G.C.); (H.W.)
- Division of Colorectal Surgery, Department of Surgery, Westmead Hospital, Westmead 2145, Australia
| | - Geoffrey Collins
- Discipline of Surgery, Faculty of Medicine and Health, The University of Sydney, Westmead 2145, Australia; (G.C.); (H.W.)
- Division of Colorectal Surgery, Department of Surgery, Westmead Hospital, Westmead 2145, Australia
| | - Henry Wang
- Discipline of Surgery, Faculty of Medicine and Health, The University of Sydney, Westmead 2145, Australia; (G.C.); (H.W.)
- Division of Colorectal Surgery, Department of Surgery, Westmead Hospital, Westmead 2145, Australia
| | - James Wei Tatt Toh
- Discipline of Surgery, Faculty of Medicine and Health, The University of Sydney, Westmead 2145, Australia; (G.C.); (H.W.)
- Division of Colorectal Surgery, Department of Surgery, Westmead Hospital, Westmead 2145, Australia
| |
Collapse
|
48
|
Dashti F, Mirazimi SMA, Rabiei N, Fathazam R, Rabiei N, Piroozmand H, Vosough M, Rahimian N, Hamblin MR, Mirzaei H. The role of non-coding RNAs in chemotherapy for gastrointestinal cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:892-926. [PMID: 34760336 PMCID: PMC8551789 DOI: 10.1016/j.omtn.2021.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers, including colorectal, gastric, hepatic, esophageal, and pancreatic tumors, are responsible for large numbers of deaths around the world. Chemotherapy is the most common approach used to treat advanced GI cancer. However, chemoresistance has emerged as a critical challenge that prevents successful tumor elimination, leading to metastasis and recurrence. Chemoresistance mechanisms are complex, and many factors and pathways are involved. Among these factors, non-coding RNAs (ncRNAs) are critical regulators of GI tumor development and subsequently can induce resistance to chemotherapy. This occurs because ncRNAs can target multiple signaling pathways, affect downstream genes, and modulate proliferation, apoptosis, tumor cell migration, and autophagy. ncRNAs can also induce cancer stem cell features and affect the epithelial-mesenchymal transition. Thus, ncRNAs could possibly act as new targets in chemotherapy combinations to treat GI cancer and to predict treatment response.
Collapse
Affiliation(s)
- Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Nikta Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Fathazam
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haleh Piroozmand
- Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
49
|
Li J, Xie Y, Huang Z, Shen D, Zhuang Z, Zhu M, Huang Y, He R, Wang X, Huang M, Luo Y, Yu H. Current treatment and surveillance modalities are not sufficient for advanced stage III colon cancer: Result from a multicenter cohort analysis. Cancer Med 2021; 10:8924-8933. [PMID: 34786881 PMCID: PMC8683545 DOI: 10.1002/cam4.4417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/02/2021] [Accepted: 10/25/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE We conducted this multicenter cohort study to evaluate the current tumor-node-metastasis staging system and treatment modality by analyzing the survival outcomes of patient groups with stage III and IV colon cancer. PATIENTS AND METHODS Stage III and IV colon cancer patients from the Surveillance, Epidemiology, and End Results (SEER) database (SEER cohort) and prospectively maintained Sun Yat-sen University (SYSU) cohort were included in this study. Kaplan-Meier method was used to estimate the cumulative rate of overall survival (OS) between patient groups, and the inverse probability weighting method was used to calculated age and sex-adjusted survival curves. The Cox regression model was used to identify the risk factors for OS. RESULTS A total of 17,911 and 1135 stage III-IV cases were included in the SEER and SYSU cohorts, respectively. Among them, 1448 and 124 resectable stage IV cases underwent curative-intent treatment in the SEER and SYSU cohorts, respectively. The T4N2b group showed a significantly worse survival outcome compared with the M1a subset receiving curative-intent treatment (HR, 1.46; p < 0.001). This finding was validated in the SYSU cohort, in which the T4N2 group had a worse outcome than that of the M1 group receiving curative-intent treatment (HR, 2.44; p < 0.001). These findings were confirmed in the adjusted survival analysis. In the multivariate analysis, the right-side tumor, poor-undifferentiated tumor, and age over 60 years were identified as independent risk factors for T4N2b patients. Based on this multivariate model, the high-risk T4N2b subgroup had a worse survival outcome compared with resectable M1b patients (HR, 1.24; p = 0.03). CONCLUSION By comparing stage III with stage IV colon cancer patients, we identified a subgroup of stage III patients at a higher risk of death than more advanced stages, implying that current cancer care modalities are not sufficient for these high-risk substages.
Collapse
Affiliation(s)
- Juan Li
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yumo Xie
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Ziying Huang
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Dingcheng Shen
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhuokai Zhuang
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Mingxuan Zhu
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yaoyi Huang
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Rongzhao He
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaolin Wang
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Meijin Huang
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yanxin Luo
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Huichuan Yu
- Department of Colorectal SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseGuangdong Institute of GastroenterologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
50
|
Larribère L, Martens UM. Advantages and Challenges of Using ctDNA NGS to Assess the Presence of Minimal Residual Disease (MRD) in Solid Tumors. Cancers (Basel) 2021; 13:5698. [PMID: 34830853 PMCID: PMC8616165 DOI: 10.3390/cancers13225698] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 12/22/2022] Open
Abstract
The ability to detect minimal residual disease (MRD) after a curative-intent surgery or treatment is of paramount importance, because it offers the possibility to help guide the clinical decisions related adjuvant therapy. Thus, the earlier MRD is detected, the earlier potentially beneficial treatment can be proposed to patients who might need it. Liquid biopsies, and in particular the next-generation sequencing of circulating tumor DNA (ctDNA) in the blood, have been the focus of an increasing amount of research in the past years. The ctDNA detection at advanced cancer stages is practicable for several solid tumors, and complements molecular information on acquired therapy resistance. In the context of MRD, it is by definition more challenging to detect ctDNA, but it is technically achievable and provides information on treatment response and probability of relapse significantly earlier than standard imaging methods. The clinical benefit of implementing this new technique in the routine is being tested in interventional clinical trials at the moment. We propose here an update of the current use of ctDNA detection by NGS as a tool to assess the presence of MRD and improve adjuvant treatment of solid tumors. We also discuss the main limitations and medium-term perspectives of this process in the clinic.
Collapse
Affiliation(s)
- Lionel Larribère
- Department of Hematology and Oncology, Cancer Center Heilbronn-Franken, SLK Clinics Heilbronn GmbH, 74078 Heilbronn, Germany;
- Skin Cancer Unit, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 68167 Mannheim, Germany
| | - Uwe M. Martens
- Department of Hematology and Oncology, Cancer Center Heilbronn-Franken, SLK Clinics Heilbronn GmbH, 74078 Heilbronn, Germany;
- MOLIT Institute for Personalized Medicine GmbH, 74076 Heilbronn, Germany
| |
Collapse
|