1
|
Tom Hu Z, Yu Y, Chen R, Yeh SJ, Chen B, Huang H. Large-scale information retrieval and correction of noisy pharmacogenomic datasets through residual thresholded deep matrix factorization. Brief Bioinform 2025; 26:bbaf226. [PMID: 40420482 DOI: 10.1093/bib/bbaf226] [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: 06/06/2024] [Revised: 01/03/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Pharmacogenomics studies are attracting an increasing amount of interest from researchers in precision medicine. The advances in high-throughput experiments and multiplexed approaches allow the large-scale quantification of drug sensitivities in molecularly characterized cancer cell lines (CCLs), resulting in a number of open drug sensitivity datasets for drug biomarker discovery. However, a significant inconsistency in drug sensitivity values among these datasets has been noted. Such inconsistency indicates the presence of substantial noise, subsequently hindering downstream analyses. To address the noise in drug sensitivity data, we introduce a robust and scalable deep learning framework, Residual Thresholded Deep Matrix Factorization (RT-DMF). This method takes a single drug sensitivity data matrix as its sole input and outputs a corrected and imputed matrix. Deep matrix factorization (DMF) excels at uncovering subtle patterns, due to its minimal reliance on data structure assumptions. This attribute significantly boosts DMF's ability to identify complex hidden patterns among nuisance effects in the data, thereby facilitating the detection of signals that are therapeutically relevant. Furthermore, RT-DMF incorporates an iterative residual thresholding procedure, which plays a crucial role in retaining signals more likely to hold therapeutic importance. Validation using simulated datasets and real pharmacogenomics datasets demonstrates the effectiveness of our approach in correcting noise and imputing missing data in drug sensitivity datasets (open-source package available at https://github.com/tomwhoooo/rtdmf).
Collapse
Affiliation(s)
- Zhiyue Tom Hu
- Division of Biostatistics, University of California Berkeley, Berkeley, CA 94720, United States
| | - Yaodong Yu
- Department of Electrical Engineer and Computer Science, University of California Berkeley, Berkeley, CA 94720, United States
| | - Ruoqiao Chen
- Department of Pharmacology and Toxicology, Michigan State University, MI 48824, United States
| | - Shan-Ju Yeh
- School of Medicine, National Tsing Hua University, Hsinchu 300044, Taiwan R.O.C
| | - Bin Chen
- Department of Pharmacology and Toxicology, Michigan State University, MI 48824, United States
- Department of Pediatrics and Human Development, Michigan State University, MI 48824, United States
| | - Haiyan Huang
- Department of Statistics, University of California Berkeley, Berkeley, CA 94720, United States
| |
Collapse
|
2
|
Popęda M, Żok J, Tomasik B, Duchnowska R, Bieńkowski M. Complete blood counts as potential risk factors of early dissemination to liver and lungs in resected colorectal cancer: a retrospective cohort study. Int J Colorectal Dis 2025; 40:21. [PMID: 39836241 PMCID: PMC11750913 DOI: 10.1007/s00384-024-04802-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE Liver and lung metastases demonstrate distinct biological, particularly immunological, characteristics. We investigated whether preoperative complete blood count (CBC) parameters, which may reflect the immune system condition, predict early dissemination to the liver and lungs in colorectal cancer (CRC). METHODS In this retrospective single-centre study, we included 268 resected CRC cases with complete 2-year follow-up and analysed preoperative CBC for association with early liver or lung metastasis development. Next, selected CBC and clinicopathological parameters were analysed with uni- and multivariable Cox regression. Independent factors affecting liver or lung metastasis-free survival were incorporated into composite scores, which were further evaluated with receiver operating characteristic (ROC) curves and dichotomised using a modified, specificity-focused, Youden approach to identify particularly high-risk patients. RESULTS Compared to metastasis-free patients, early liver metastases were related to decreases in red blood cells, haematocrit, lymphocytes and elevated monocyte-to-lymphocyte ratio, while lung metastases to lower eosinophil counts. A composite score of independent factors (erythrocytopenia, lower lymphocyte count and pN) yielded HR of 8.01 (95% CI 3.45-18.57, p < 0.001) for liver-specific metastasis-free survival (MFS). For lung-specific MFS, the combination of eosinopenia, pN and primary tumour location showed HR of 13.69 (95% CI 4.34-43.20, p < 0.001). CONCLUSION Early CRC metastases to the liver and lungs are associated with partially divergent clinicopathological and peripheral blood features. We propose simple, clinically implementable scores, based on routinely assessed parameters, to identify patients with an increased risk of early dissemination to the liver or lungs. After validation in independent cohorts, these scores may provide easily available prognostic information.
Collapse
Affiliation(s)
- Marta Popęda
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland.
| | - Jolanta Żok
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Bartłomiej Tomasik
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Renata Duchnowska
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Michał Bieńkowski
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
3
|
de Back TR, van Hooff SR, Sommeijer DW, Vermeulen L. Transcriptomic subtyping of gastrointestinal malignancies. Trends Cancer 2024; 10:842-856. [PMID: 39019673 DOI: 10.1016/j.trecan.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024]
Abstract
Gastrointestinal (GI) cancers are highly heterogeneous at multiple levels. Tumor heterogeneity can be captured by molecular profiling, such as genetic, epigenetic, proteomic, and transcriptomic classification. Transcriptomic subtyping has the advantage of combining genetic and epigenetic information, cancer cell-intrinsic properties, and the tumor microenvironment (TME). Unsupervised transcriptomic subtyping systems of different GI malignancies have gained interest because they reveal shared biological features across cancers and bear prognostic and predictive value. Importantly, transcriptomic subtypes accurately reflect complex phenotypic states varying not only per tumor region, but also throughout disease progression, with consequences for clinical management. Here, we discuss methodologies of transcriptomic subtyping, proposed taxonomies for GI malignancies, and the challenges posed to clinical implementation, highlighting opportunities for future transcriptomic profiling efforts to optimize clinical impact.
Collapse
Affiliation(s)
- Tim R de Back
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sander R van Hooff
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Dirkje W Sommeijer
- Flevohospital, Department of Internal Medicine, Hospitaalweg 1, 1315 RA, Almere, The Netherlands
| | - Louis Vermeulen
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Brenner B, Shulman K, Hubert A, Man S, Geva R, Ben-Aharon I, Fennig S, Mishaeli M, Yarom N, Bar-Sela G, Brenner R, Shai A, Baehner FL, Russell C, Soussan-Gutman L, Voet H, Bareket-Samish A, Liebermann N. Treatments and clinical outcomes in stage II colon cancer patients with 12-gene Oncotype DX Colon Recurrence Score® assay-guided therapy: real-world data. ESMO Open 2024; 9:103648. [PMID: 39137478 PMCID: PMC11369429 DOI: 10.1016/j.esmoop.2024.103648] [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] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The 12-gene Oncotype DX Colon Recurrence Score® result quantifies the recurrence risk in stage II/III colon cancer (CC). This real-world study investigated stage II CC patients whose treatment decisions incorporated the Recurrence Score® (RS) result. MATERIALS AND METHODS This retrospective analysis of a prospectively designed cohort included all stage II, mismatch repair-proficient CC patients who underwent 12-gene testing through Clalit between January 2011 and December 2016 and had available data with a minimum 3-year follow-up. RESULTS The analysis included 938 patients {median age 68 [interquartile range (IQR) 60-76] years; 96% T3 tumors}. The median RS was 26 (IQR 19-33) and the three RS categories (0-29, 30-40, 41-100) included 65%, 24%, and 11% of patients, respectively. Chemotherapy (CT) use differed significantly between the three RS categories (14%, 36%, and 60%, respectively; P < 0.001). The CT and observation-only groups were imbalanced with worse clinicopathologic characteristics in the former. Among observation-only patients, Kaplan-Meier (KM) estimates for recurrence-free interval (RFI) and CC-specific survival (CCSS) differed significantly between the three RS categories (P < 0.001). Clinical outcomes by treatment (CT versus observation) within each RS category revealed no differences in RFI and CCSS in the RS 0-29 and 30-40 categories. In contrast, in the RS 41-100 category, the difference in RFI trended toward significance (P = 0.066), and for CCSS, a statistically significant difference was observed, with better outcomes among CT-treated patients (P = 0.035). CONCLUSIONS RS results are prognostic in stage II CC. Among RS 41-100 patients, outcomes were better in CT-treated versus observation-only patients despite worse clinicopathologic characteristics, suggesting that CT confers clinical benefit in high-risk patients.
Collapse
Affiliation(s)
- B Brenner
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva; Faculty of Medicine, Tel Aviv University, Tel Aviv.
| | - K Shulman
- Oncology Department, Lin Medical Center, Haifa; Rappaport Faculty of Medicine, Technion Israeli Institute of Technology, Haifa
| | - A Hubert
- Sharett Institute of Oncology, Hadassah-Hebrew University Hospital, Jerusalem
| | - S Man
- Department of Clinical Oncology and Radiology, Soroka University Medical Center, Beer Sheva
| | - R Geva
- Division of Oncology, Sourasky Medical Center, Tel Aviv
| | - I Ben-Aharon
- Department of Oncology, Rambam Health Care Campus, Haifa
| | - S Fennig
- Institute of Oncology, Kaplan Medical Center, Rehovot
| | - M Mishaeli
- Oncology Department, Meir Medical Center, Kfar Saba
| | - N Yarom
- Oncology Department, Shamir Medical Center, Be'er Ya'akov
| | - G Bar-Sela
- Rappaport Faculty of Medicine, Technion Israeli Institute of Technology, Haifa; Oncology Department, Emek Medical Center, Afula
| | - R Brenner
- Oncology Department, Wolfson Medical Center, Holon
| | - A Shai
- Rappaport Faculty of Medicine, Technion Israeli Institute of Technology, Haifa; Oncology Department, Galilee Medical Center, Nahariya, Israel
| | - F L Baehner
- Medical Department, Exact Sciences, Redwood City, USA
| | - C Russell
- Medical Department, Exact Sciences, Redwood City, USA
| | | | - H Voet
- Environmental Economics and Management, Hebrew University of Jerusalem, Rehovot
| | | | - N Liebermann
- Community Division, Clalit Health Services, Tel Aviv, Israel
| |
Collapse
|
5
|
Shen L, van Gestel A, Prinsen P, Vink G, van Erning FN, Geleijnse G, Kaptein M. Value of Real-World Evidence for Treatment Selection: A Case Study in Colon Cancer. JCO Clin Cancer Inform 2024; 8:e2300186. [PMID: 38753347 DOI: 10.1200/cci.23.00186] [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: 09/17/2023] [Revised: 02/28/2024] [Accepted: 03/25/2024] [Indexed: 01/07/2025] Open
Abstract
PURPOSE Real-world evidence (RWE)-derived from analysis of real-world data (RWD)-has the potential to guide personalized treatment decisions. However, because of potential confounding, generating valid RWE is challenging. This study demonstrates how to responsibly generate RWE for treatment decisions. We validate our approach by demonstrating that we can uncover an existing adjuvant chemotherapy (ACT) guideline for stage II and III colon cancer (CC)-which came about using both data from randomized controlled trials and expert consensus-solely using RWD. METHODS Data from the population-based Netherlands Cancer Registry from a total of 27,056 patients with stage II and III CC who underwent curative surgery were analyzed to estimate the overall survival (OS) benefit of ACT. Focusing on 5-year OS, the benefit of ACT was estimated for each patient using G-computation methods by adjusting for patient and tumor characteristics and estimated propensity score. Subsequently, on the basis of these estimates, an ACT decision tree was constructed. RESULTS The constructed decision tree corresponds to the current Dutch guideline: patients with stage III or stage II with T stage 4 should receive surgery and ACT, whereas patients with stage II with T stage 3 should only receive surgery. Interestingly, we do not find sufficient RWE to conclude against ACT for stage II with T stage 4 and microsatellite instability-high (MSI-H), a recent addition to the current guideline. CONCLUSION RWE, if used carefully, can provide a valuable addition to our construction of evidence on clinical decision making and therefore ultimately affect treatment guidelines. Next to validating the ACT decisions advised in the current Dutch guideline, this paper suggests additional attention should be paid to MSI-H in future iterations of the guideline.
Collapse
Affiliation(s)
- Lingjie Shen
- Department of Methodology and Statistics, Tilburg University, Tilburg, the Netherlands
| | - Anja van Gestel
- Department of Clinical Data Science, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Peter Prinsen
- Department of Clinical Data Science, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Geraldine Vink
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Felice N van Erning
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - Gijs Geleijnse
- Department of Clinical Data Science, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Maurits Kaptein
- Department of Mathematics and Computer Science, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
6
|
Chen L, Wang Y, Cai C, Ding Y, Kim RS, Lipchik C, Gavin PG, Yothers G, Allegra CJ, Petrelli NJ, Suga JM, Hopkins JO, Saito NG, Evans T, Jujjavarapu S, Wolmark N, Lucas PC, Paik S, Sun M, Pogue-Geile KL, Lu X. Machine Learning Predicts Oxaliplatin Benefit in Early Colon Cancer. J Clin Oncol 2024; 42:1520-1530. [PMID: 38315963 PMCID: PMC11095904 DOI: 10.1200/jco.23.01080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 02/07/2024] Open
Abstract
PURPOSE A combination of fluorouracil, leucovorin, and oxaliplatin (FOLFOX) is the standard for adjuvant therapy of resected early-stage colon cancer (CC). Oxaliplatin leads to lasting and disabling neurotoxicity. Reserving the regimen for patients who benefit from oxaliplatin would maximize efficacy and minimize unnecessary adverse side effects. METHODS We trained a new machine learning model, referred to as the colon oxaliplatin signature (COLOXIS) model, for predicting response to oxaliplatin-containing regimens. We examined whether COLOXIS was predictive of oxaliplatin benefits in the CC adjuvant setting among 1,065 patients treated with 5-fluorouracil plus leucovorin (FULV; n = 421) or FULV + oxaliplatin (FOLFOX; n = 644) from NSABP C-07 and C-08 phase III trials. The COLOXIS model dichotomizes patients into COLOXIS+ (oxaliplatin responder) and COLOXIS- (nonresponder) groups. Eight-year recurrence-free survival was used to evaluate oxaliplatin benefits within each of the groups, and the predictive value of the COLOXIS model was assessed using the P value associated with the interaction term (int P) between the model prediction and the treatment effect. RESULTS Among 1,065 patients, 526 were predicted as COLOXIS+ and 539 as COLOXIS-. The COLOXIS+ prediction was associated with prognosis for FULV-treated patients (hazard ratio [HR], 1.52 [95% CI, 1.07 to 2.15]; P = .017). The model was predictive of oxaliplatin benefits: COLOXIS+ patients benefited from oxaliplatin (HR, 0.65 [95% CI, 0.48 to 0.89]; P = .0065; int P = .03), but COLOXIS- patients did not (COLOXIS- HR, 1.08 [95% CI, 0.77 to 1.52]; P = .65). CONCLUSION The COLOXIS model is predictive of oxaliplatin benefits in the CC adjuvant setting. The results provide evidence supporting a change in CC adjuvant therapy: reserve oxaliplatin only for COLOXIS+ patients, but further investigation is warranted.
Collapse
Affiliation(s)
- Lujia Chen
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA
| | | | - Chunhui Cai
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA
| | - Ying Ding
- NRG Oncology Statistics and Data Management Center, Pittsburgh, PA
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA
| | - Rim S. Kim
- NSABP/NRG Oncology, Pittsburgh, PA
- AstraZeneca, Oncology Translational Medicine, Gaithersburg, MD
| | | | - Patrick G. Gavin
- NSABP/NRG Oncology, Pittsburgh, PA
- AstraZeneca Respiratory and Immunology, Gaithersburg, MD
| | - Greg Yothers
- NRG Oncology Statistics and Data Management Center, Pittsburgh, PA
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA
| | - Carmen J. Allegra
- Department of Medicine, University of Florida Health, Gainesville, FL
| | - Nicholas J. Petrelli
- Helen F. Graham Cancer Center and Research Institute at Christiana Care, Newark, DE
| | - Jennifer Marie Suga
- Kaiser Permanente Oncology Clinical Trials, KP NCI Community Oncology Research Program (NCORP), Vallejo, CA
| | - Judith O. Hopkins
- Novant Health Forsyth Medical Cancer Institute/Southeast Clinical Oncology Research NCORP, Kernersville, NC
| | - Naoyuki G. Saito
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | | | | | - Norman Wolmark
- NSABP/NRG Oncology, Pittsburgh, PA
- UPMC Hillman Cancer Center, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Peter C. Lucas
- NSABP/NRG Oncology, Pittsburgh, PA
- UPMC Hillman Cancer Center, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Soonmyung Paik
- NSABP/NRG Oncology, Pittsburgh, PA
- Yonsei University College of Medicine, Yonsei Biomedical Research Institute, Seoul, Republic of South Korea
| | - Min Sun
- UPMC Hillman Cancer Center, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- DeepRx Inc, Pittsburgh, PA
| | | | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA
- DeepRx Inc, Pittsburgh, PA
| |
Collapse
|
7
|
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
|
8
|
McDonald HG, Kerekes DM, Kim J, Khan SA. Precision Oncology in Gastrointestinal and Colorectal Cancer Surgery. Surg Oncol Clin N Am 2024; 33:321-341. [PMID: 38401913 DOI: 10.1016/j.soc.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2024]
Abstract
Precision medicine is used to treat gastrointestinal malignancies including esophageal, gastric, small bowel, colorectal, and pancreatic cancers. Cutting-edge assays to detect and treat these cancers are active areas of research and will soon become standard of care. Colorectal cancer is a prime example of precision oncology as disease site is no longer the final determinate of treatment. Here, the authors describe how leveraging an understanding of tumor biology translates to individualized patient care using evidence-based practices.
Collapse
Affiliation(s)
- Hannah G McDonald
- Department of General Surgery, Division of Surgical Oncology, The University of Kentucky, 800 Rose Street, Lexington, KY 40508, USA
| | - Daniel M Kerekes
- Department of General Surgery, Division of Surgical Oncology, Yale University, 15 York Street, New Haven, CT 06510, USA
| | - Joseph Kim
- Department of General Surgery, Division of Surgical Oncology, The University of Kentucky, 800 Rose Street, Lexington, KY 40508, USA
| | - Sajid A Khan
- Department of Surgery, Yale University, 15 York Street, New Haven, CT 06510, USA.
| |
Collapse
|
9
|
Cao L, Wei S, Yin Z, Chen F, Ba Y, Weng Q, Zhang J, Zhang H. Identifying important microbial biomarkers for the diagnosis of colon cancer using a random forest approach. Heliyon 2024; 10:e24713. [PMID: 38298638 PMCID: PMC10828680 DOI: 10.1016/j.heliyon.2024.e24713] [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: 10/30/2023] [Revised: 12/14/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Colon cancer is one of the most common cancers, with 30-50 % of patients returning or metastasizing within 5 years of treatment. Increasingly, researchers have highlighted the influence of microbes on cancer malignant activity, while no studies have explored the relationship between colon cancer and the microbes in tumors. Here, we used tissue and blood samples from 67 colon cancer patients to identify pathogenic microorganisms associated with the diagnosis and prediction of colon cancer and evaluate the predictive performance of each pathogenic marker and its combination based on the next-generation sequencing data by using random forest algorithms. The results showed that we constructed a database of 13,187 pathogenic microorganisms associated with human disease and identified 2 pathogenic microorganisms (Synthetic.construct_32630 and Dicrocoelium.dendriticum_57078) associated with colon cancer diagnosis, and the constructed diagnostic prediction model performed well for tumor tissue samples and blood samples. In summary, for the first time, we provide new molecular markers for the diagnosis of colon cancer based on the expression of pathogenic microorganisms in order to provide a reference for improving the effective screening rate of colon cancer in clinical practice and ameliorating the personalized treatment of colon cancer patients.
Collapse
Affiliation(s)
- Lichao Cao
- School of Life Sciences, Northwest University, 710127, Xi'an, Shaanxi Province, China
| | - Shangqing Wei
- School of Life Sciences, Northwest University, 710127, Xi'an, Shaanxi Province, China
| | - Zongyi Yin
- Shenzhen University General Hospital, 518071, Shenzhen, Guangdong Province, China
| | - Fang Chen
- Shenzhen Nucleus Gene Technology Co., Ltd., 518071, Shenzhen, Guangdong Province, China
| | - Ying Ba
- Shenzhen Nucleus Gene Technology Co., Ltd., 518071, Shenzhen, Guangdong Province, China
| | - Qi Weng
- Shenzhen Nucleus Gene Technology Co., Ltd., 518071, Shenzhen, Guangdong Province, China
| | - Jiahao Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., 518071, Shenzhen, Guangdong Province, China
| | - Hezi Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., 518071, Shenzhen, Guangdong Province, China
| |
Collapse
|
10
|
Hu ZT, Yu Y, Chen R, Yeh SJ, Chen B, Huang H. Large-Scale Information Retrieval and Correction of Noisy Pharmacogenomic Datasets through Residual Thresholded Deep Matrix Factorization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.07.570723. [PMID: 38106027 PMCID: PMC10723412 DOI: 10.1101/2023.12.07.570723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Pharmacogenomics studies are attracting an increasing amount of interest from researchers in precision medicine. The advances in high-throughput experiments and multiplexed approaches allow the large-scale quantification of drug sensitivities in molecularly characterized cancer cell lines (CCLs), resulting in a number of open drug sensitivity datasets for drug biomarker discovery. However, a significant inconsistency in drug sensitivity values among these datasets has been noted. Such inconsistency indicates the presence of substantial noise, subsequently hindering downstream analyses. To address the noise in drug sensitivity data, we introduce a robust and scalable deep learning framework, Residual Thresholded Deep Matrix Factorization (RT-DMF). This method takes a single drug sensitivity data matrix as its sole input and outputs a corrected and imputed matrix. Deep Matrix Factorization (DMF) excels at uncovering subtle patterns, due to its minimal reliance on data structure assumptions. This attribute significantly boosts DMF's ability to identify complex hidden patterns among nuisance effects in the data, thereby facilitating the detection of signals that are therapeutically relevant. Furthermore, RT-DMF incorporates an iterative residual thresholding (RT) procedure, which plays a crucial role in retaining signals more likely to hold therapeutic importance. Validation using simulated datasets and real pharmacogenomics datasets demonstrates the effectiveness of our approach in correcting noise and imputing missing data in drug sensitivity datasets (open source package available at https://github.com/tomwhoooo/rtdmf).
Collapse
Affiliation(s)
- Zhiyue Tom Hu
- Division of Biostatistics, University of California Berkeley, Berkeley, 94720, U.S.A
| | - Yaodong Yu
- Department of Electrical Engineer and Computer Science, University of California Berkeley, Berkeley, 94720, U.S.A
| | - Ruoqiao Chen
- Department of Pharmacology and Toxicology, Michigan State University, 48824, U.S.A
| | - Shan-Ju Yeh
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan R.O.C
| | - Bin Chen
- Department of Pharmacology and Toxicology, Michigan State University, 48824, U.S.A
- Department of Pediatrics and Human Development, Michigan State University, 48824, U.S.A
| | - Haiyan Huang
- Department of Statistics, University of California Berkeley, Berkeley, 94720, U.S.A
| |
Collapse
|
11
|
Yang G, Yu XR, Weisenberger DJ, Lu T, Liang G. A Multi-Omics Overview of Colorectal Cancer to Address Mechanisms of Disease, Metastasis, Patient Disparities and Outcomes. Cancers (Basel) 2023; 15:cancers15112934. [PMID: 37296894 DOI: 10.3390/cancers15112934] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Human colorectal cancer (CRC) is one of the most common malignancies in men and women across the globe, albeit CRC incidence and mortality shows a substantial racial and ethnic disparity, with the highest burden in African American patients. Even with effective screening tools such as colonoscopy and diagnostic detection assays, CRC remains a substantial health burden. In addition, primary tumors located in the proximal (right) or distal (left) sides of the colorectum have been shown to be unique tumor types that require unique treatment schema. Distal metastases in the liver and other organ systems are the major causes of mortality in CRC patients. Characterizing genomic, epigenomic, transcriptomic and proteomic (multi-omics) alterations has led to a better understanding of primary tumor biology, resulting in targeted therapeutic advancements. In this regard, molecular-based CRC subgroups have been developed that show correlations with patient outcomes. Molecular characterization of CRC metastases has highlighted similarities and differences between metastases and primary tumors; however, our understanding as to how to improve patient outcomes based on metastasis biology is lagging and remains a major obstacle to improving CRC patient outcomes. In this review, we will summarize the multi-omics features of primary CRC tumors and their metastases across racial and ethnic groups, the differences in proximal and distal tumor biology, molecular-based CRC subgroups, treatment strategies and challenges for improving patient outcomes.
Collapse
Affiliation(s)
- Guang Yang
- School of Sciences, China Pharmaceutical University, Nanjing 211121, China
- China Grand Enterprises, Beijing 100101, China
| | - Xi Richard Yu
- China Grand Enterprises, Beijing 100101, China
- Huadong Medicine Co., Ltd., Hangzhou 310011, China
| | - Daniel J Weisenberger
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Tao Lu
- School of Sciences, China Pharmaceutical University, Nanjing 211121, China
- State Key Laboratory of Natural Sciences, China Pharmaceutical University, Nanjing 211121, China
| | - Gangning Liang
- USC Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
12
|
Davey MG, O'Neill M, Regan M, Meshkat B, Nugent E, Joyce M, Hogan AM. Impact of the 12-gene recurrence score in influencing adjuvant chemotherapy prescription in mismatch repair proficient stage II/III colonic carcinoma-a systematic review and meta-analysis. Int J Colorectal Dis 2023; 38:71. [PMID: 36912973 PMCID: PMC10011316 DOI: 10.1007/s00384-023-04364-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2023] [Indexed: 03/14/2023]
Abstract
INTRODUCTION The 12-gene recurrence score (RS) is a clinically validated assay which predicts recurrence risk in patients with stage II/III colon cancer. Decisions regarding adjuvant chemotherapy may be guided using this assay or based on the judgement of tumour board. AIMS To assess the concordance between the RS and MDT decisions regarding adjuvant chemotherapy in colon cancer. METHODS A systematic review was performed in accordance with PRISMA guidelines. Meta-analyses were performed using the Mantel-Haenszel method using the Review Manager version 5.4 software. RESULTS Four studies including 855 patients with a mean age of 68 years (range: 25-90 years) met inclusion criteria. Overall, 79.2% had stage II disease (677/855) and 20.8% had stage III disease (178/855). For the entire cohort, concordant results between the 12-gene assay and MDT were more likely than discordant (odds ratio (OR): 0.38, 95% confidence interval (CI): 0.25-0.56, P < 0.001). Patients were more likely to have chemotherapy omitted than escalated when using the RS (OR: 9.76, 95% CI: 6.72-14.18, P < 0.001). For those with stage II disease, concordant results between the 12-gene assay and MDT were more likely than discordant (OR: 0.30, 95% CI: 0.17-0.53, P < 0.001). In stage II disease, patients were more likely to have chemotherapy omitted than escalated when using the RS (OR: 7.39, 95% CI: 4.85-11.26, P < 0.001). CONCLUSIONS The use of the 12-gene signature refutes the decision of tumour board in 25% of cases, with 75% of discordant decisions resulting in omission of adjuvant chemotherapy. Therefore, it is possible that a proportion of such patients are being overtreated when relying on tumour board decisions alone.
Collapse
Affiliation(s)
- Matthew G Davey
- Department of Surgery, Galway University Hospitals, Galway, H91 YRY71, Ireland.
| | - Maeve O'Neill
- Department of Surgery, Galway University Hospitals, Galway, H91 YRY71, Ireland
| | - Mark Regan
- Department of Surgery, Galway University Hospitals, Galway, H91 YRY71, Ireland
| | - Babak Meshkat
- Department of Surgery, Galway University Hospitals, Galway, H91 YRY71, Ireland
| | - Emmeline Nugent
- Department of Surgery, Galway University Hospitals, Galway, H91 YRY71, Ireland
| | - Myles Joyce
- Department of Surgery, Galway University Hospitals, Galway, H91 YRY71, Ireland
| | - Aisling M Hogan
- Department of Surgery, Galway University Hospitals, Galway, H91 YRY71, Ireland
| |
Collapse
|
13
|
Daza J, Salomé B, Okhawere K, Bane O, Meilika KN, Korn TG, Qi J, Xe H, Patel M, Brody R, Kim-Schulze S, Sfakianos JP, Lewis S, Rich JM, Zuluaga L, Badani KK, Horowitz A. Urine supernatant reveals a signature that predicts survival in clear-cell renal cell carcinoma. BJU Int 2023. [PMID: 36797809 DOI: 10.1111/bju.15989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
OBJECTIVE To profile the cell-free urine supernatant and plasma of a small cohort of clear-cell renal cell carcinoma (ccRCC) patients by measuring the relative concentrations of 92 proteins related to inflammation. Using The Cancer Genome Atlas (TCGA), we then performed a targeted mRNA analysis of genes encoding the above proteins and defined their effects on overall survival (OS). SUBJECTS/PATIENTS AND METHODS Samples were collected prospectively from ccRCC patients. A multiplex proximity extension assay was used to measure the concentrations of 92 inflammation-related proteins in cell-free urine supernatants and plasma. Transcriptomic and clinical information from ccRCC patients was obtained from TCGA. Unsupervised clustering and differential protein expression analyses were performed on protein concentration data. Targeted mRNA analysis on genes encoding significant differentially expressed proteins was performed using TCGA. Backward stepwise regression analyses were used to build a nomogram. The performance of the nomogram and clinical benefit was assessed by discrimination and calibration, and a decision curve analysis, respectively. RESULTS Unsupervised clustering analysis revealed inflammatory signatures in the cell-free urine supernatant of ccRCC patients. Backward stepwise regressions using TCGA data identified transcriptomic risk factors and risk groups associated with OS. A nomogram to predict 2-year and 5-year OS was developed using these risk factors. The decision curve analysis showed that our model was associated with a net benefit improvement compared to the treat-all/none strategies. CONCLUSION We defined four novel biomarkers using proteomic and transcriptomic data that distinguish severity of prognosis in ccRCC. We showed that these biomarkers can be used in a model to predict 2-year and 5-year OS in ccRCC across different tumour stages. This type of analysis, if validated in the future, provides non-invasive prognostic information that could inform either management or surveillance strategies for patients.
Collapse
Affiliation(s)
- Jorge Daza
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bérengère Salomé
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kennedy Okhawere
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Octavia Bane
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirolos N Meilika
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Talia G Korn
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jingjing Qi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hui Xe
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Brody
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P Sfakianos
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara Lewis
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordan M Rich
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Zuluaga
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ketan K Badani
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir Horowitz
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
To YH, Degeling K, McCoy M, Wong R, Jones I, Dunn C, Hong W, Loft M, Gibbs P, Tie J. Real‐world adjuvant chemotherapy treatment patterns and outcomes over time for resected stage II and III colorectal cancer. Asia Pac J Clin Oncol 2022; 19:392-402. [DOI: 10.1111/ajco.13885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/30/2022] [Accepted: 10/09/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Yat Hang To
- The Walter and Eliza Hall Institute Melbourne Victoria Australia
| | - Koen Degeling
- Cancer Health Services Research Centre for Cancer Faculty of Medicine Dentistry and Health Sciences University of Melbourne Melbourne Victoria Australia
- Cancer Health Services Research Centre for Health Policy Melbourne School of Population and Global Health Faculty of Medicine Dentistry and Health Sciences University of Melbourne Melbourne Victoria Australia
| | - Melanie McCoy
- Colorectal Research Unit St John of God Subiaco Hospital Subiaco Western Australia Australia
- Medical School University of Western Australia Crawley Western Australia Australia
| | - Rachel Wong
- The Walter and Eliza Hall Institute Melbourne Victoria Australia
- Department of Medical Oncology Eastern Health Melbourne Victoria Australia
- Eastern Health Clinical School Monash University Melbourne Victoria Australia
- Epworth Healthcare Melbourne Victoria Australia
| | - Ian Jones
- Department of Surgery University of Melbourne Parkville Victoria Australia
- Colorectal Surgery Unit Department of General Surgery Royal Melbourne Hospital Parkville Victoria Australia
| | - Catherine Dunn
- The Walter and Eliza Hall Institute Melbourne Victoria Australia
| | - Wei Hong
- The Walter and Eliza Hall Institute Melbourne Victoria Australia
- Department of Medical Oncology LaTrobe Regional Hospital Traralgon Victoria Australia
- Department of Medical Oncology St Vincent's Hospital Fitzroy Victoria Australia
| | - Matthew Loft
- The Walter and Eliza Hall Institute Melbourne Victoria Australia
- Department of Medical Biology University of Melbourne Melbourne Victoria Australia
| | - Peter Gibbs
- The Walter and Eliza Hall Institute Melbourne Victoria Australia
- Department of Medical Oncology Western Health Melbourne Victoria Australia
- Faculty of Medicine and Health Sciences University of Melbourne Melbourne Victoria Australia
| | - Jeanne Tie
- The Walter and Eliza Hall Institute Melbourne Victoria Australia
- Department of Medical Oncology Peter MacCallum Cancer Centre Parkville Victoria Australia
| |
Collapse
|
15
|
Yu IS, Aubin F, Goodwin R, Loree JM, Mather C, Sheffield BS, Snow S, Gill S. Tumor Biomarker Testing for Metastatic Colorectal Cancer: a Canadian Consensus Practice Guideline. Ther Adv Med Oncol 2022; 14:17588359221111705. [PMID: 35898967 PMCID: PMC9310231 DOI: 10.1177/17588359221111705] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
The systemic therapy management of metastatic colorectal cancer (mCRC) has evolved from primarily cytotoxic chemotherapies to now include targeted agents given alone or in combination with chemotherapy, and immune checkpoint inhibitors. A better understanding of the pathogenesis and molecular drivers of colorectal cancer not only aided the development of novel targeted therapies but led to the discovery of tumor mutations which act as predictive biomarkers for therapeutic response. Mutational status of the KRAS gene became the first genomic biomarker to be established as part of standard of care molecular testing, where KRAS mutations within exons 2, 3, and 4 predict a lack of response to anti- epidermal growth factor receptor therapies. Since then, several other biomarkers have become relevant to inform mCRC treatment; however, there are no published Canadian guidelines which reflect the current standards for biomarker testing. This guideline was developed by a pan-Canadian advisory group to provide contemporary, evidence-based recommendations on the minimum acceptable standards for biomarker testing in mCRC, and to describe additional biomarkers for consideration.
Collapse
Affiliation(s)
- Irene S. Yu
- Department of Medical Oncology, BC Cancer
Surrey, Surrey, BC, Canada
| | - Francine Aubin
- Division of Hematology and Oncology, Department
of Medicine, Centre Hospitalier de l’Université de Montréal, Montreal, QC,
Canada
| | - Rachel Goodwin
- Division of Medical Oncology, Department of
Medicine, Ottawa Hospital Cancer Centre, Ottawa, ON, Canada
| | - Jonathan M. Loree
- Department of Medical Oncology, BC Cancer
Agency - Vancouver Centre, Vancouver, BC, Canada
| | - Cheryl Mather
- Department of Laboratory Medicine and
Pathology, University of Alberta, Edmonton, AB, Canada
| | - Brandon S. Sheffield
- Division of Advanced Diagnostics, William Osler
Health System, Brampton, ON, Canada
| | - Stephanie Snow
- Department of Medicine, Queen Elizabeth II
Health Sciences Centre, Halifax, NS, Canada
| | - Sharlene Gill
- Department of Medical Oncology, BC Cancer
Agency – Vancouver Centre, 600 W 10th Ave, Vancouver, BC, V5Z 4E6,
Canada
| |
Collapse
|
16
|
Shen MH, Huang CJ, Ho TF, Liu CY, Shih YY, Huang CS, Huang CC. Colorectal cancer concurrent gene signature based on coherent patterns between genomic and transcriptional alterations. BMC Cancer 2022; 22:590. [PMID: 35637462 PMCID: PMC9150289 DOI: 10.1186/s12885-022-09627-9] [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: 07/05/2021] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Background The aim of the study was to enhance colorectal cancer prognostication by integrating single nucleotide polymorphism (SNP) and gene expression (GE) microarrays for genomic and transcriptional alteration detection; genes with concurrent gains and losses were used to develop a prognostic signature. Methods The discovery dataset comprised 32 Taiwanese colorectal cancer patients, of which 31 were assayed for GE and copy number variations (CNVs) with Illumina Human HT-12 BeadChip v4.0 and Omni 25 BeadChip v1.1. Concurrent gains and losses were declared if coherent manners were observed between GE and SNP arrays. Concurrent genes were also identified in The Cancer Genome Atlas Project (TCGA) as the secondary discovery dataset (n = 345). Results The “universal” concurrent genes, which were the combination of z-transformed correlation coefficients, contained 4022 genes. Candidate genes were evaluated within each of the 10 public domain microarray datasets, and 1655 (2000 probe sets) were prognostic in at least one study. Consensus across all datasets was used to build a risk predictive model, while distinct relapse-free/overall survival patterns between defined risk groups were observed among four out of five training datasets. The predictive accuracy of recurrence, metastasis, or death was between 61 and 86% (cross-validation area under the receiver operating characteristic (ROC) curve: 0.548-0.833) from five independent validation studies. Conclusion The colorectal cancer concurrent gene signature is prognostic in terms of recurrence, metastasis, or mortality among 1746 patients. Genes with coherent patterns between genomic and transcriptional contexts are more likely to provide prognostication for colorectal cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09627-9.
Collapse
Affiliation(s)
- Ming-Hung Shen
- Department of Surgery, Fu-Jen Catholic University Hospital, No. 69, Guizi Road, Taishan District, New Taipei City, 243, Taiwan.,Ph. D Program in Nutrition and Food Science, College of Human Ecology, Fu-Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City, 242062, Taiwan.,School of Medicine, College of Medicine, Fu-Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City, 242062, Taiwan
| | - Chi-Jung Huang
- Department of Biochemistry, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490, Taiwan.,Department of Medical Research, Cathay General Hospital, No.280, Sec. 4, Renai Rd., Daan Dist., Taipei City, 106, Taiwan
| | - Thien-Fiew Ho
- Division of General Surgery, Cathay General Hospital Sijhih, No. 2, Ln. 59, Jiancheng Rd., Xizhi Dist., New Taipei City, 221, Taiwan
| | - Chih-Yi Liu
- Division of Pathology, Cathay General Hospital Sijhih, No. 2, Ln. 59, Jiancheng Rd., Xizhi Dist., New Taipei City, 221, Taiwan
| | - Ying-Yih Shih
- Division of Hematology and Oncology, Cathay General Hospital Sijhih, No. 2, Ln. 59, Jiancheng Rd., Xizhi Dist., New Taipei City, 221, Taiwan
| | - Ching-Shui Huang
- Department of Surgery, Cathay General Hospital, No.280, Sec. 4, Renai Rd., Daan Dist., Taipei City, 106, Taiwan. .,School of Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei City, 110, Taiwan.
| | - Chi-Cheng Huang
- Department of Surgery, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei City, 11217, Taiwan. .,Comprehensive Breast Health Center, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei City, Taiwan, 11217. .,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, No.17, Xuzhou Rd., Taipei City, 100, Taiwan.
| |
Collapse
|
17
|
Comprehensive analysis of m 6A regulator-based methylation modification patterns characterized by distinct immune profiles in colon adenocarcinomas. Gene 2022; 821:146250. [PMID: 35151825 DOI: 10.1016/j.gene.2022.146250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/05/2022] [Accepted: 01/21/2022] [Indexed: 01/02/2023]
Abstract
Mounting evidences have indicated that RNA N6-methyladenosine (m6A) modification played important roles in tumor formation and growth. However, it is rarely reported that m6A modifications are involved in the immune regulation and tumor microenvironment (TME) formation. In this study, we aimed to investigate the correlation between m6A modifications and TME regulation of colon adenocarcinoma (COAD) by bioinformatic analysis. NMF algorithm was applied to carry out consensus molecular subtype analysis on 36 selected m6A regulators regarding methylation modification, to identify m6A modification patterns and characteristics of m6A related genes in colon adenocarcinoma (COAD). Further, the relative infiltration levels of different immune cell subsets were quantified by ssGSEA and CIBERSORT algorithms, and a m6Sig scoring scheme was constructed to predict the prognosis and evaluate the response to immunotherapy in the patients with COAD. Among 579 COAD samples, we identified three different m6A modification patterns which were related to different biological pathways and clinical outcomes. Then, a scoring scheme termed "m6Sig score" was developed based on m6A-related characteristic genes, and was utilized to score patients with COAD into groups. We found that COAD patients with lower m6Sig scores exhibited prolonged survival and potentiated immune infiltration, which were associated with higher tumor mutation load, lower PD-L1 expression, and higher mutation rates of SMG (such as TTN and KRAS). Moreover, analysis regarding evaluation of immune response revealed that the patients with lower m6Sig scores had higher Immunophenoscore. Collectively, our study provided in depth insight into the interactions between m6A modification and regulation of TME. In addition, the quantitative evaluation of m6A modification patterns in our results may have implications in further immunotherapy for individual COAD patients.
Collapse
|
18
|
Comprehensive Analysis of Colorectal Cancer Immunity and Identification of Immune-Related Prognostic Targets. DISEASE MARKERS 2022; 2022:7932655. [PMID: 35401882 PMCID: PMC8986440 DOI: 10.1155/2022/7932655] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/03/2022] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (COAD) is ranked as the third most common cancer and second in terms of cancer-related deaths worldwide. Due to its poor overall survival and prognosis, the incidents of COAD are significantly increasing. Although treatment methods have greatly been improved in the last decade, it is still not good enough to have satisfactory treatment outcomes. In recent years, immunotherapy has been successful to some extent in the treatment of many cancers but still, many patients do not respond to immunotherapy. Therefore, it is essential to have a deeper understanding of the immune characteristics of the tumor microenvironment and identify meaningful immune targets. In terms of immune targets, COAD has been poorly explored; thus, in the current study, based on the immune cell infiltration score and differentially expressed genes, COAD tumors were classified into hot and cold tumors. The Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis was used to identify hub genes, construct a prognostic model, and screen potential immune targets. In total, 12 genes (CLK3, CYSLTR2, GJA10, CYP4Z1, FAM185A, LINC00324, EEF1A1P34, EEF1B2P8, PTCSC3, MIR6780A, LINC01666, and RNU6.661P) differentially expressed between hot and cold tumors were screened out. Among them, CYSLTR2 was considered as a potential candidate gene, because it showed a significant positive correlation with immune cell infiltration and immune checkpoints (PDCD1, CD274, and CTLA4). Finally, we constructed and validated a new prognostic model for COAD showing 0.854 AUC for the ROC curve, and these results provide sufficient potential to choose CYSLTR2 as an important immune target for the prognosis of COAD.
Collapse
|
19
|
A retrospective analysis using deep-learning models for prediction of survival outcome and benefit of adjuvant chemotherapy in stage II/III colorectal cancer. J Cancer Res Clin Oncol 2022; 148:1955-1963. [PMID: 35332389 DOI: 10.1007/s00432-022-03976-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Most of Stage II/III colorectal cancer (CRC) patients can be cured by surgery alone, and only certain CRC patients benefit from adjuvant chemotherapy. Risk stratification based on deep-learning from haematoxylin and eosin (H&E) images has been postulated as a potential predictive biomarker for benefit from adjuvant chemotherapy. However, very limited success has been achieved in using biomarkers, including deep-learning-based markers, to facilitate the decision for adjuvant chemotherapy despite recent advances of artificial intelligence. METHODS We trained and internally validated CRCNet using 780 Stage II/III CRC patients from Molecular and Cellular Oncology. Independent external validation of the model was performed using 337 Stage II/III CRC patients from The Cancer Genome Atlas (TCGA). RESULTS CRCNet stratified the patients into high, medium, and low-risk subgroups. Multivariate Cox regression analyses confirmed that CRCNet risk groups are statistically significant after adjusting for existing risk factors. The high-risk subgroup significantly benefits from adjuvant chemotherapy. A hazard ratio (chemo-treated vs untreated) of 0.2 (95% Confidence Interval (CI), 0.05-0.65; P = 0.009) and 0.6 (95% CI 0.42-0.98; P = 0.038) are observed in the TCGA and MCO Fluorouracil-treated patients, respectively. Conversely, no significant benefit from chemotherapy is observed in the low- and medium-risk groups (P = 0.2-1). CONCLUSION The retrospective analysis provides further evidence that H&E image-based biomarkers may potentially be of great use in delivering treatments following surgery for Stage II/III CRC, improving patient survival, and avoiding unnecessary treatment and associated toxicity, and warrants further validation on other datasets and prospective confirmation in clinical trials.
Collapse
|
20
|
Wang H, Gao Y, Vafaei S, Yu Q, Zhang J, Wang L. A chemoresistance lncRNA signature for recurrence risk stratification of colon cancer patients with chemotherapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:427-438. [PMID: 35036055 PMCID: PMC8733234 DOI: 10.1016/j.omtn.2021.12.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023]
Abstract
Chemotherapy is considered the nonsurgical treatment of choice for colon cancer patients. However, no precise molecular markers are available to determine which patients can actually benefit from it. In this study, we identified 55 chemotherapy-specific long non-coding RNAs (lncRNAs) of colon cancer patients through a systematic assessment of lncRNA expression profiles from a public database. These were taken from multiple cohorts of colon cancer patients who had received chemotherapy, or not. Based on these data, a chemoresistance lncRNA signature, named CRLSig, was constructed and successfully applied to divide chemotherapy patients into two groups with different recurrence-free survival (RFS) rates. Gene set enrichment analysis revealed that patients with low CRLSig had more infiltrating CD8+ T cells and macrophages, while those with high CRLSig had more infiltrating natural killer T cells. KEGG pathway analysis revealed that the low CRLSig group had more activated metabolic pathways compared with those in the high CRLSig group, indicating better response to chemotherapy. Single-cell sequencing analysis revealed that stromal cells and epithelial cells had higher CRLSig. Thus, we have constructed an auxiliary prognostic tool, CRLSig, able to discriminate patients at high risk of RFS, despite having received standard adjuvant chemotherapy treatment. A CRLSig was constructed for the first time CRLSig revealed chemotherapy patients with different RFS rates Low CRLSig group had more activated metabolic pathways ScRNA-seq analysis revealed stromal cells and epithelial cells had higher CRLSig
Collapse
Affiliation(s)
- Hao Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuzhen Gao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China.,Biomedical Research Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310013, China
| | - Somayeh Vafaei
- Department of Molecular Medicine, Faculty of advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Qiaoyan Yu
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China.,Biomedical Research Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310013, China
| | - Liangjing Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Allar BG, Messaris E, Poylin VY, Schlechter BL, Cataldo TE. Oncotype DX testing does not affect clinical practice in stage IIa colon cancer. Med Oncol 2022; 39:59. [PMID: 35150339 DOI: 10.1007/s12032-022-01660-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 01/18/2022] [Indexed: 11/24/2022]
Abstract
Although studies have demonstrated the 12-gene Oncotype DX Colon Recurrence Score's (RS) validity in predicting recurrence and influence on physician-patient decision-making, its discriminatory power and inability to predict response to treatment make its clinical impact uncertain. We sought to evaluate the influence of RS in the decision to offer adjuvant chemotherapy after resection of stage IIa colon cancer. A review of patients with stage IIa colon cancer who obtained the RS at a tertiary academic medical center was conducted. The main study outcome was decision to start adjuvant chemotherapy. The association between RS and the decision to obtain adjuvant chemotherapy was evaluated utilizing the Wilcoxon rank-sum test and area under the receiver operating characteristic curve. 52 of 105 patients with stage IIa colon cancer underwent RS testing. Overall, seven of 52 patients (13%) received adjuvant chemotherapy. 34 (65%) patients obtained the RS test despite having multiple other recurrence risk factors. There were no significant associations between any patient/tumor characteristic and RS score (all p > 0.08) or starting adjuvant chemotherapy (all p > 0.15). On multivariable analysis, there was no significant effect of RS on the odds of undergoing chemotherapy (OR 1.07, 95% CI 0.98-1.19; p = 0.14). There was no clear association between RS and starting adjuvant chemotherapy (AUC 0.64, 95% CI 0.36-0.91; p = 0.25). RS was not associated with the decision to start adjuvant chemotherapy. Given its lack of association with clinical decision-making and inability to predict clinical outcome, our data suggest the RS should not be obtained in patients with stage IIa colon cancer.
Collapse
Affiliation(s)
- Benjamin G Allar
- Division of Colon and Rectal Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Evangelos Messaris
- Division of Colon and Rectal Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Vitaliy Y Poylin
- Division of Colon and Rectal Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.,Division of Gastrointestinal Surgery, Northwestern Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin L Schlechter
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Gastrointestinal Cancer Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Thomas E Cataldo
- Division of Colon and Rectal Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
22
|
Vogel JD, Felder SI, Bhama AR, Hawkins AT, Langenfeld SJ, Shaffer VO, Thorsen AJ, Weiser MR, Chang GJ, Lightner AL, Feingold DL, Paquette IM. The American Society of Colon and Rectal Surgeons Clinical Practice Guidelines for the Management of Colon Cancer. Dis Colon Rectum 2022; 65:148-177. [PMID: 34775402 DOI: 10.1097/dcr.0000000000002323] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
| | | | | | | | | | | | - Amy J Thorsen
- Colon and Rectal Surgery Associates, Minneapolis, Minnesota
| | | | | | | | | | | |
Collapse
|
23
|
Yothers G, Venook AP, Oki E, Niedzwiecki D, Lin Y, Crager MR, Chao C, Baehner FL, Wolmark N, Yoshino T. Patient-specific meta-analysis of 12-gene colon cancer recurrence score validation studies for recurrence risk assessment after surgery with or without 5FU and oxaliplatin. J Gastrointest Oncol 2022; 13:126-136. [PMID: 35284101 PMCID: PMC8899729 DOI: 10.21037/jgo-21-620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/04/2022] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Individualized estimates of the risk of recurrence in colon cancer patients are needed that reflect current medical practice and available treatment options. METHODS Three validation studies of the 12-gene colon recurrence score assay were used with pre-specified patient-specific meta-analysis (PSMA) methods to integrate the 12-gene Oncotype DX Colon Recurrence Score result (RS) with the clinical and pathology risk factors stage, T-stage, mis-match repair (MMR) status, and number of nodes examined to calculate individualized recurrence risk estimates. Baseline risk estimation used the most recent studies, so the risk estimates reflect current medical practice. The effect of fluorouracil (5FU) was estimated with a meta-analysis of two studies. The effect of oxaliplatin was estimated using one of the RS assay validation studies, in which patients were randomized to 5FU with or without oxaliplatin. RESULTS The RS result and each of the clinical-pathologic factors provided independent prognostic information for recurrence. Among stage II, T3, MMR-proficient patients with ≥12 nodes examined (the most common scenario), patients with RS ≤30 (approximately 48%) have estimated 5-year recurrence risk ≤10% with surgery alone. Among stage IIIA/B, T3, MMR-deficient patients with ≥12 nodes examined, patients with RS ≤19 (approximately 14%) have an estimated 5-year recurrence risk ≤10% with surgery alone. Among stage IIIA/B, T3, MMR-proficient patients with ≥12 nodes examined, those with RS ≤14 (approximately 6%) have estimated 5-year recurrence risk ≤10% with 5FU alone. DISCUSSION The PSMA integrates the 12-gene colon RS result with clinical and pathology factors to provide individualized recurrence risk estimates that reflect current medical practice. The risk estimates are in a range that may help inform treatment decisions for a substantial number of stage II and stage III patients.
Collapse
Affiliation(s)
- Greg Yothers
- NSABP, NRG Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alan P. Venook
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Eiji Oki
- Department of Surgery and Science, Kyushu University, Kyushu, Japan
| | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Yan Lin
- NSABP, NRG Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael R. Crager
- Department of Biostatistics, Precision Oncology, Exact Sciences Corporation, Redwood City, CA, USA
| | - Calvin Chao
- Global Medical Affairs, Precision Oncology, Exact Sciences Corporation, Redwood City, CA, USA
| | - Frederick L. Baehner
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
- Medical Department, Precision Oncology, Exact Sciences Corporation, Redwood City, CA, USA
| | - Norman Wolmark
- NSABP, NRG Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
24
|
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: 11] [Impact Index Per Article: 3.7] [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
|
25
|
Cadham B, Parker B, Yin Y, Gill S, Loree JM, Cheung WY, Davies JM. Evaluation of Clinical and Pathological Factors to Predict High Risk of Recurrence in Patients With Stage II Colon Cancer. Clin Colorectal Cancer 2021; 21:e39-e48. [PMID: 34836806 DOI: 10.1016/j.clcc.2021.09.003] [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: 03/07/2021] [Revised: 08/06/2021] [Accepted: 09/09/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND The ability to predict patients with stage II colon cancer at high risk of recurrence is currently limited to certain clinicopathologic factors. PATIENTS AND METHODS This population-based study reviewed various prognostic factors to identify those associated with worse time to recurrence (TTR) and improved disease-specific survival (DSS), and to subsequently develop a prognostic index (PI) to identify high risk cancers. RESULTS Multivariate analyses revealed factors significant for TTR and DSS. A PI derived from the TTR risk factors identified 3 risk groups using 5-year rate without relapse: 88% for low-risk, 81% for intermediate-risk, and 59% for high risk. CONCLUSION This model was better at identifying high risk patients than using equally weighted standard risk factors.
Collapse
|
26
|
Zhao XF, Yang YS, Gao DZ, Park YK. TMPRSS4 overexpression promotes the metastasis of colorectal cancer and predicts poor prognosis of stage III-IV colorectal cancer. Int J Biol Markers 2021; 36:23-32. [PMID: 34595980 DOI: 10.1177/17246008211046368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To study in detail the expression pattern and prognostic significance of TMPRSS4 in colorectal cancer. METHODS The expression of TMPRSS4 protein was determined using Western blot in the colorectal cancer tissues and normal tissues. Immunohistochemistry was used to detect the TMPRSS4 expression in colorectal cancer tissues, and the clinicopathologic characteristics and prognostic significance were analyzed. RESULTS TMPRSS4 overexpression was associated with tumor budding, lymphovascular invasion, perineural invasion, cancerous emboli, infiltration depth, lymph node metastasis, distant metastasis, and tumor node metastasis stage (P < 0.05 for all). Interestingly, TMPRSS4 expression in the tumor budding, tumor emboli, lymph node, and liver metastatic tumor samples was higher than in the paired primary tumors. In contrast, TMPRSS4 overexpression is inversely correlated with both the overall survival and the disease-free survival of the patients with colorectal cancer (P < 0.05 for both). Also, we found that TMPRSS4 is only of significance in predicting the prognosis of stage III and IV colorectal cancer, not stage I and II. CONCLUSIONS TMPRSS4 was shown to be involved in the whole process of metastasis from tumor budding to lymph node and/or distant metastasis in colorectal cancer and predicted the unfavorable prognosis of stage III-IV, indicating that it is a novel target for the precise treatment of colorectal cancer with lymph node or distant organ metastasis.
Collapse
Affiliation(s)
- Xue-Feng Zhao
- Department of General Surgery, 194043Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Yu-Shen Yang
- Department of General Surgery, 194043Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Da-Zhi Gao
- Department of General Surgery, 194043Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Young Kyu Park
- Department of General Surgery, 65722Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea
| |
Collapse
|
27
|
Loaiza‐Bonilla A, Benson AB, Grothey A, Karimi M, Klempner SJ, Lin D, Mahtani R, Soares HP. Use of Molecular Assays and Circulating Tumor DNA in Early-Stage Colorectal Cancer: A Roundtable Discussion of the Gastrointestinal Cancer Therapy Expert Group. Oncologist 2021; 26:651-659. [PMID: 33650740 PMCID: PMC8342566 DOI: 10.1002/onco.13738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
The use of genomic testing is rapidly emerging as an important clinical tool both for cancer diagnosis and for guiding treatment decisions in a wide range of malignancies, including gastrointestinal (GI) cancers such as colorectal cancer (CRC). Advances in technologies such as polymerase chain reaction and next-generation sequencing methods have made it possible to noninvasively screen for CRC through, for example, the use of blood- or stool-based testing, with high specificity. Tests are also available that can provide prognostic information beyond traditional clinicopathologic factors such as tumor size, grade, and nodal status, which can enable clinicians to more accurately risk stratify patients for recurrence. Lastly, in the setting of resected CRC, tests are now available that can detect circulating tumor DNA as a means for noninvasive minimal/molecular residual disease monitoring, thereby potentially guiding the use of adjuvant chemotherapy and/or escalating or de-escalating therapy. The Gastrointestinal Cancer Therapy Expert Group (GICTEG) recently convened a virtual meeting to discuss current issues related to genomic testing in GI cancer, with the goal of providing guidance on the use of these tests for the practicing community oncologist, for whom GI cancer may be only one of many tumor types encountered. This article provides a summary of the discussion and highlights the key opinions of the GICTEG on this topic. IMPLICATIONS FOR PRACTICE: The Gastrointestinal Cancer Therapy Expert Group seeks to provide practical guidance and opinion on the treatment of gastrointestinal malignancies, including colorectal cancer (CRC), for the practicing community oncologist in situations for which guidelines from established bodies, such as the National Comprehensive Cancer Network and the American Society of Clinical Oncology, may be less clear. In the present report, clinical guidance on the use of molecular assays for a range of clinical indications in CRC is presented, including the use of circulating tumor DNA to detect minimal/molecular residual disease in patients with successfully resected early-stage CRC.
Collapse
Affiliation(s)
- Arturo Loaiza‐Bonilla
- Department of Medical Oncology, Cancer Treatment Centers of AmericaPhiladelphiaPennsylvaniaUSA
| | - Al B. Benson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern UniversityChicagoIllinoisUSA
| | | | - Misagh Karimi
- Department of Medical Oncology & Therapeutics ResearchCity of Hope, Newport Beach, CaliforniaUSA
| | - Samuel J. Klempner
- Department of Medicine, Massachusetts General HospitalBostonMassachusettsUSA
| | - Daniel Lin
- Thomas Jefferson University HospitalPhiladelphiaPennsylvaniaUSA
| | | | | |
Collapse
|
28
|
Carmona-Bayonas A, Valentí V, Jiménez-Fonseca P, Salazar R. SUNRISE-DI study. The daily sunrise is easier to predict than the benefit of adjuvant treatment in colon cancer. ESMO Open 2021; 6:100205. [PMID: 34242967 PMCID: PMC8271109 DOI: 10.1016/j.esmoop.2021.100205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 10/26/2022] Open
Affiliation(s)
- A Carmona-Bayonas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, UMI, IMIB, Murcia, Spain
| | - V Valentí
- Medical Oncology Department, Hospital Del Vendrell, El Vendrell, Tarragona, Spain
| | - P Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - R Salazar
- Medical Oncology Department, Oncobell Program, IDIBELL Institut Català d'Oncologia, Hospital Duran i Reynals, CIBERONC, Barcelona, Spain.
| |
Collapse
|
29
|
Characterization of a Tumor-Microenvironment-Relevant Gene Set Based on Tumor Severity in Colon Cancer and Evaluation of Its Potential for Dihydroartemisinin Targeting. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4812068. [PMID: 34239578 PMCID: PMC8233087 DOI: 10.1155/2021/4812068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/09/2021] [Indexed: 01/12/2023]
Abstract
Colon cancer (COAD) is a leading cause of cancer mortality in the world. Most patients with COAD die as a result of cancer cell metastasis. However, the mechanisms underlying the metastatic phenotype of COAD remain unclear. Instead, particular features of the tumor microenvironment (TME) could predict adverse outcomes including metastasis in patients with COAD, and the role of TME in governing COAD progression is undeniable. Therefore, exploring the role of TME in COAD may help us better understand the molecular mechanisms behind COAD progression which may improve clinical outcomes and quality of patients. Here, we identified a Specific TME Regulatory Network including AEBP1, BGN, POST, and FAP (STMERN) that is highly involved in clinical outcomes of patients with COAD. Comprehensive in silico analysis of our study revealed that the STMERN is highly correlated with the severity of COAD. Meanwhile, our results reveal that the STMERN might be associated with immune infiltration in COAD. Importantly, we show that dihydroartemisinin (DHA) potentially interacts with the STMERN. We suggest that DHA might contribute to immune infiltration through regulating the STMERN in COAD. Taken together, our data provide a set of biomarkers of progression and poor prognosis in COAD. These findings could have potential prognostic and therapeutic implications in the progression of COAD.
Collapse
|
30
|
Oki E, Watanabe J, Sato T, Kagawa Y, Kuboki Y, Ikeda M, Ueno H, Kato T, Kusumoto T, Masuishi T, Yamaguchi K, Kanazawa A, Nishina T, Uetake H, Yamanaka T, Yoshino T. Impact of the 12-gene recurrence score assay on deciding adjuvant chemotherapy for stage II and IIIA/B colon cancer: the SUNRISE-DI study. ESMO Open 2021; 6:100146. [PMID: 33984677 PMCID: PMC8134704 DOI: 10.1016/j.esmoop.2021.100146] [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: 02/07/2021] [Revised: 03/26/2021] [Accepted: 04/12/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Recent advances in adjuvant chemotherapy for early colon cancer have widened physicians' recommendations on the regimen and duration (3 or 6 months) of the treatment. We conducted this prospective study to evaluate whether the 12-gene recurrence score (12-RS) assay affected physicians' recommendations on adjuvant treatment selection. PATIENTS AND METHODS Patients with stage IIIA/IIIB or stage II colon cancer were enrolled. After the patients discussed adjuvant treatment with their treating physicians, the physicians filled in the questionnaire before assay indicating the treatment recommendation. When the 12-RS assay results were available, the physicians again filled in the questionnaire after assay. The primary endpoint was the rate of change in treatment recommendations from before to after the assay, with a threshold rate of change being 20%. Patients with stage IIIA/B to II were enrolled in a ratio of 2 : 1. RESULTS Overall, the treatment recommendations changed in 40% of cases after obtaining 12-RS assay results. Recommendations were changed in 45% (80/178; 95% confidence interval, 37% to 53%; P < 0.001) and 30% (29/97; 95% confidence interval, 21% to 40%; P < 0.001) of patients with stage IIIA/B and II colon cancer, respectively. Patients with stage IIIA/B cancer had significantly more change than those with stage II cancer (P = 0.0148). From before to after the 12-RS assay, the percentage of patients whose physicians reported being confident in their treatment recommendations significantly increased from 54% to 81% in stage IIIA/B (P < 0.001) and from 65% to 83% in stage II (P < 0.001). CONCLUSION Our study confirmed the usefulness of the 12-RS assay in aiding the physician-patient decision-making process for tailoring adjuvant chemotherapy for stage IIIA/B colon cancer.
Collapse
Affiliation(s)
- E Oki
- Department of Surgery and Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - J Watanabe
- Department of Surgery, Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - T Sato
- Department of Colorectal Surgery, Kitasato University Hospital, Kanagawa, Japan
| | - Y Kagawa
- Department of Surgery, Kansai Rosa Hospital, Hyogo, Japan
| | - Y Kuboki
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - M Ikeda
- Department of Surgery, Hyogo College of Medicine, Hyogo, Japan
| | - H Ueno
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - T Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - T Kusumoto
- Department of Gastroenterological Surgery, NHO National Kyushu Medical Center, Fukuoka, Japan
| | - T Masuishi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Aichi, Japan
| | - K Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - A Kanazawa
- Department of Gastroenterological Surgery, Shimane Prefectural Central Hospital, Shimane, Japan
| | - T Nishina
- Department of Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Ehime, Japan
| | - H Uetake
- Department of Specialized Surgeries, Tokyo Medical and Dental University, Tokyo, Japan
| | - T Yamanaka
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan.
| | - T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
31
|
Chang GJ, You YNY, Russell CA, Tierno MB, Turner M, Bennett JP, Lau A, Hochster HS. Young-Onset Colon Cancer and Recurrence Risk by Gene Expression. J Natl Cancer Inst 2021; 112:1170-1173. [PMID: 32040172 PMCID: PMC7669228 DOI: 10.1093/jnci/djaa019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/14/2020] [Accepted: 02/05/2020] [Indexed: 01/29/2023] Open
Abstract
The incidence and mortality from colorectal cancer in younger adults (younger than 55 years) is increasing. We reviewed the complete database of a gene-expression test, Oncotype DX Colon Recurrence Score test, to determine age-related differences in recurrence score (RS) and single-gene results (7 cancer-related of the 12-gene assay). We included 20 478 stage II and III A and B colon cancer patients submitted to Genomic Health. RS results were grouped by low-, intermediate-, and high-risk groups. Single-gene scores were described using median and interquartile range. Of all patients 72.5% and 72.6% of those younger than 40 years had low-risk RS. Comparing older with younger patients, RS or single-gene expression did not differ by age group or stage. Young-onset colon cancer does not differ by expression of the RS component genes. Most patients with stage II and III colon cancer have low-risk disease as measured by the 12-gene assay, regardless of age.
Collapse
Affiliation(s)
- George J Chang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Y Nancy Y You
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | - Anna Lau
- Genomic Health, Inc., Redwood City, CA, USA
| | | |
Collapse
|
32
|
Integrated approaches for precision oncology in colorectal cancer: The more you know, the better. Semin Cancer Biol 2021; 84:199-213. [PMID: 33848627 DOI: 10.1016/j.semcancer.2021.04.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/30/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the most common human malignancies accounting for approximately 10 % of worldwide cancer incidence and mortality. While early-stage CRC is mainly a preventable and curable disease, metastatic colorectal cancer (mCRC) remains an unmet clinical need. Moreover, about 25 % of CRC cases are diagnosed only at the metastatic stage. Despite the extensive molecular and functional knowledge on this disease, systemic therapy for mCRC still relies on traditional 5-fluorouracil (5-FU)-based chemotherapy regimens. On the other hand, targeted therapies and immunotherapy have shown effectiveness only in a limited subset of patients. For these reasons, there is a growing need to define the molecular and biological landscape of individual patients to implement novel, rationally driven, tailored therapies. In this review, we explore current and emerging approaches for CRC management such as genomic, transcriptomic and metabolomic analysis, the use of liquid biopsies and the implementation of patients' preclinical avatars. In particular, we discuss the contribution of each of these tools in elucidating patient specific features, with the aim of improving our ability in advancing the diagnosis and treatment of colorectal tumors.
Collapse
|
33
|
Wang L, Jiang X, Zhang X, Shu P. Prognostic implications of an autophagy-based signature in colorectal cancer. Medicine (Baltimore) 2021; 100:e25148. [PMID: 33787596 PMCID: PMC8021367 DOI: 10.1097/md.0000000000025148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The heterogeneity of colorectal cancer (CRC) poses a significant challenge to the precise treatment of patients. CRC has been divided into 4 consensus molecular subtypes (CMSs) with distinct biological and clinical characteristics, of which CMS4 has the mesenchymal identity and the highest relapse rate. Autophagy plays a vital role in CRC development and therapeutic response. METHODS The gene expression profiles collected from 6 datasets were applied to this study. Network analysis was applied to integrate the subtype-specific molecular modalities and autophagy signature to establish an autophagy-based prognostic signature for CRC (APSCRC). RESULTS Network analysis revealed that 6 prognostic autophagy genes (VAMP7, DLC1, FKBP1B, PEA15, PEX14, and DNAJB1) predominantly regulated the mesenchymal modalities of CRC. The APSCRC was constructed by these 6 core genes and applied for risk calculation. Patients were divided into high- and low-risk groups based on APSCRC score in all cohorts. Patients within the high-risk group showed an unfavorable prognosis. In multivariate analysis, the APSCRC remained an independent predictor of prognosis. Moreover, the APSCRC achieved higher prognostic power than commercialized multigene signatures. CONCLUSIONS We proposed and validated an autophagy-based signature, which is a promising prognostic biomarker of CRC patients. Further prospective studies are warranted to test and validate its efficiency for clinical application.
Collapse
Affiliation(s)
- Liangbin Wang
- Department of Anorectal Surgery, Beilun People's Hospital, Ningbo
| | - Xinlei Jiang
- Tianjin Key Laboratory of Aquatic Science and Technology, School of Environmental and Municipal Engineering, Tianjin Chengjian University, Tianjin
| | - Xingguo Zhang
- Molecular Laboratory, Beilun People's Hospital, Ningbo, P.R. China
| | - Peng Shu
- Molecular Laboratory, Beilun People's Hospital, Ningbo, P.R. China
| |
Collapse
|
34
|
Miyamoto Y, Hiyoshi Y, Sawayama H, Tokunaga R, Baba H. Precision medicine for adjuvant chemotherapy of resected colorectal cancer. Ann Gastroenterol Surg 2020; 4:635-645. [PMID: 33319153 PMCID: PMC7726679 DOI: 10.1002/ags3.12397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/16/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is the most common cancer and the second leading cause of cancer death in Japan. Surgical resection is the only curative option for localized disease. However, undetectable micrometastases remaining after curative surgery may cause disease recurrence. Adjuvant chemotherapy aims to eradicate these micrometastases to improve the cure rate. Unfortunately, few reliable prognostic and predictive markers are available that identify patients at high risk for CRC during early-stage disease. However, promising biomarkers may become available in the near future. Such biomarkers provide information for stratifying a patient's risk and for selecting the optimal treatment. Here, we provide an overview of current relevant prognostic and predictive biomarkers applicable to adjuvant treatment of early-stage CRC and focus on the future of this field.
Collapse
Affiliation(s)
- Yuji Miyamoto
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yukiharu Hiyoshi
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Hiroshi Sawayama
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Ryuma Tokunaga
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Hideo Baba
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
35
|
Navarro S, Cuatrecasas M, Hernández-Losa J, Landolfi S, Musulén E, Ramón Y Cajal S, García-Carbonero R, García-Foncillas J, Pérez-Segura P, Salazar R, Vera R, García-Alfonso P. [Update of the recommendations for the determination of biomarkers in colorectal carcinoma. National Consensus of the Spanish Society of Medical Oncology and the Spanish Society of Pathology]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2020; 54:41-54. [PMID: 33455693 DOI: 10.1016/j.patol.2020.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/02/2020] [Accepted: 07/26/2020] [Indexed: 11/25/2022]
Abstract
This update of the consensus of the Spanish Society of Medical Oncology (Sociedad Española de Oncología Médica - SEOM) and the Spanish Society of Pathology (Sociedad Española de Anatomía Patológica - SEAP), reviews the advances in the analysis of biomarkers in advanced colorectal cancer (CRC) as well as susceptibility markers of hereditary CRC and molecular biomarkers of localized CRC. Recently published information on the essential determination of KRAS, NRAS and BRAF mutations and the possible benefits of determining the amplification of human epidermal growth factor receptor 2 (HER2), the expression of proteins in the DNA repair pathway and the study of NTRK fusions are also evaluated. From a pathological point of view, the importance of analysing the tumour budding and poorly differentiated clusters and its prognostic value in CRC is reviewed, as well as the impact of molecular lymph node analysis on lymph node staging in CRC. The incorporation of pan-genomic technologies, such as next-generation sequencing (NGS) and liquid biopsy in the clinical management of patients with CRC is also outlined. All these aspects are developed in this guide which, like the previous one, will be revised when necessary in the future.
Collapse
Affiliation(s)
- Samuel Navarro
- Departamento de Patología, Universidad de Valencia, Hospital Clínico Universitario de Valencia, CIBERONC, Valencia, España.
| | | | - Javier Hernández-Losa
- Departamento de Patología, Hospital Universitario Vall d'Hebron, CIBERONC, Barcelona, España
| | - Stefania Landolfi
- Departamento de Patología, Hospital Universitario Vall d'Hebron, CIBERONC, Barcelona, España
| | - Eva Musulén
- Departamento de Patología, Hospital Universitari General de Catalunya, Grupo Quirónsalud, Sant Cugat del Vallès, España; Grupo de Epigenética del Cáncer, Institut de Recerca contra la Leucèmia Josep Carreras, Badalona, España
| | - Santiago Ramón Y Cajal
- Departamento de Patología, Hospital Universitario Vall d'Hebron, CIBERONC, Barcelona, España
| | - Rocío García-Carbonero
- Departamento de Oncología Médica, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Madrid, España
| | - Jesús García-Foncillas
- Departamento de Oncología, Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, España
| | - Pedro Pérez-Segura
- Departamento de Oncología Médica, Hospital Clínico Universitario San Carlos, CIBERONC, Madrid, España
| | - Ramón Salazar
- Departamento de Oncología Médica, ICO ĹHospitalet, Oncobell Program (IDIBELL), CIBERONC, Hospitalet de Llobregat, España
| | - Ruth Vera
- Departamento de Oncología Médica, Complejo Hospitalario de Navarra, Navarrabiomed, IDISNA, Pamplona, España
| | - Pilar García-Alfonso
- Departamento de Oncología Médica, Hospital General Universitario Gregorio Marañón, Madrid, España
| |
Collapse
|
36
|
Taieb J, Gallois C. Adjuvant Chemotherapy for Stage III Colon Cancer. Cancers (Basel) 2020; 12:E2679. [PMID: 32961795 PMCID: PMC7564362 DOI: 10.3390/cancers12092679] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
In patients with stage III colon cancer (CC), adjuvant chemotherapy with the combination of oxapliplatin to a fluoropyrimidine (FOLFOX or CAPOX) is a standard of care. The duration of treatment can be reduced from 6 months to 3 months, depending on the regimen, for patients at low risk of recurrence, without loss of effectiveness and allowing a significant reduction in the risk of cumulative sensitive neuropathy. However, our capacity to identify patients that do really need this doublet adjuvant treatment remains limited. In fact, only 30% at the most will actually benefit from this adjuvant treatment, 50% of them being already cured by the surgery and 20% of them experiencing disease recurrence despite the adjuvant treatment. Thus, it is necessary to be able to better predict individually for each patient the risk of recurrence and the need for adjuvant chemotherapy together with the need of new treatment approaches for specific subgroups. Many biomarkers have been described with their own prognostic weight, without leading to any change in clinical practices for now. In this review, we will first discuss the recommendations for adjuvant chemotherapy, and then the different biomarkers described and the future perspectives for the management of stage III CC.
Collapse
Affiliation(s)
- Julien Taieb
- Sorbonne Paris cite, University of Paris, 75006 Paris, France;
- Siric CARPEM, Assistance Publique-Hôpitaux de Paris, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Claire Gallois
- Sorbonne Paris cite, University of Paris, 75006 Paris, France;
- Siric CARPEM, Assistance Publique-Hôpitaux de Paris, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| |
Collapse
|
37
|
Schraa SJ, van Rooijen KL, van der Kruijssen DEW, Rubio Alarcón C, Phallen J, Sausen M, Simmons J, Coupé VMH, van Grevenstein WMU, Elias S, Verkooijen HM, Laclé MM, Bosch LJW, van den Broek D, Meijer GA, Velculescu VE, Fijneman RJA, Vink GR, Koopman M. Circulating tumor DNA guided adjuvant chemotherapy in stage II colon cancer (MEDOCC-CrEATE): study protocol for a trial within a cohort study. BMC Cancer 2020; 20:790. [PMID: 32819390 PMCID: PMC7441668 DOI: 10.1186/s12885-020-07252-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/03/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Accurate detection of patients with minimal residual disease (MRD) after surgery for stage II colon cancer (CC) remains an urgent unmet clinical need to improve selection of patients who might benefit form adjuvant chemotherapy (ACT). Presence of circulating tumor DNA (ctDNA) is indicative for MRD and has high predictive value for recurrent disease. The MEDOCC-CrEATE trial investigates how many stage II CC patients with detectable ctDNA after surgery will accept ACT and whether ACT reduces the risk of recurrence in these patients. METHODS/DESIGN MEDOCC-CrEATE follows the 'trial within cohorts' (TwiCs) design. Patients with colorectal cancer (CRC) are included in the Prospective Dutch ColoRectal Cancer cohort (PLCRC) and give informed consent for collection of clinical data, tissue and blood samples, and consent for future randomization. MEDOCC-CrEATE is a subcohort within PLCRC consisting of 1320 stage II CC patients without indication for ACT according to current guidelines, who are randomized 1:1 into an experimental and a control arm. In the experimental arm, post-surgery blood samples and tissue are analyzed for tissue-informed detection of plasma ctDNA, using the PGDx elio™ platform. Patients with detectable ctDNA will be offered ACT consisting of 8 cycles of capecitabine plus oxaliplatin while patients without detectable ctDNA and patients in the control group will standard follow-up according to guideline. The primary endpoint is the proportion of patients receiving ACT when ctDNA is detectable after resection. The main secondary outcome is 2-year recurrence rate (RR), but also includes 5-year RR, disease free survival, overall survival, time to recurrence, quality of life and cost-effectiveness. Data will be analyzed by intention to treat. DISCUSSION The MEDOCC-CrEATE trial will provide insight into the willingness of stage II CC patients to be treated with ACT guided by ctDNA biomarker testing and whether ACT will prevent recurrences in a high-risk population. Use of the TwiCs design provides the opportunity to randomize patients before ctDNA measurement, avoiding ethical dilemmas of ctDNA status disclosure in the control group. TRIAL REGISTRATION Netherlands Trial Register: NL6281/NTR6455 . Registered 18 May 2017, https://www.trialregister.nl/trial/6281.
Collapse
Affiliation(s)
- S J Schraa
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - K L van Rooijen
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - D E W van der Kruijssen
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - C Rubio Alarcón
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - J Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - M Sausen
- Personal Genome Diagnostics, Baltimore, MD, 21224, USA
| | - J Simmons
- Personal Genome Diagnostics, Baltimore, MD, 21224, USA
| | - V M H Coupé
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - W M U van Grevenstein
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - S Elias
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - H M Verkooijen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - M M Laclé
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - L J W Bosch
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - D van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - G A Meijer
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - V E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - R J A Fijneman
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - G R Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - M Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | | |
Collapse
|
38
|
Chakrabarti S, Peterson CY, Sriram D, Mahipal A. Early stage colon cancer: Current treatment standards, evolving paradigms, and future directions. World J Gastrointest Oncol 2020; 12:808-832. [PMID: 32879661 PMCID: PMC7443846 DOI: 10.4251/wjgo.v12.i8.808] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/16/2020] [Accepted: 08/01/2020] [Indexed: 02/05/2023] Open
Abstract
Colon cancer continues to be one of the leading causes of mortality and morbidity throughout the world despite the availability of reliable screening tools and effective therapies. The majority of patients with colon cancer are diagnosed at an early stage (stages I to III), which provides an opportunity for cure. The current treatment paradigm of early stage colon cancer consists of surgery followed by adjuvant chemotherapy in a select group of patients, which is directed at the eradication of minimal residual disease to achieve a cure. Surgery alone is curative for the vast majority of colon cancer patients. Currently, surgery and adjuvant chemotherapy can achieve long term survival in about two-thirds of colon cancer patients with nodal involvement. Adjuvant chemotherapy is recommended for all patients with stage III colon cancer, while the benefit in stage II patients is not unequivocally established despite several large clinical trials. Contemporary research in early stage colon cancer is focused on minimally invasive surgical techniques, strategies to limit treatment-related toxicities, precise patient selection for adjuvant therapy, utilization of molecular and clinicopathologic information to personalize therapy and exploration of new therapies exploiting the evolving knowledge of tumor biology. In this review, we will discuss the current standard treatment, evolving treatment paradigms, and the emerging biomarkers, that will likely help improve patient selection and personalization of therapy leading to superior outcomes.
Collapse
Affiliation(s)
- Sakti Chakrabarti
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Carrie Y Peterson
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Deepika Sriram
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Amit Mahipal
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
39
|
Parent P, Cohen R, Rassy E, Svrcek M, Taieb J, André T, Turpin A. A comprehensive overview of promising biomarkers in stage II colorectal cancer. Cancer Treat Rev 2020; 88:102059. [DOI: 10.1016/j.ctrv.2020.102059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 02/08/2023]
|
40
|
Auclin E, Taieb J, Lepage C, Aparicio T, Faroux R, Mini E, Folprecht G, Salazar R, Benetkiewicz M, Banzi M, Louvet C, Van Laethem JL, Tabernero J, Hickish T, de Gramont A, André T, Vernerey D. Carcinoembryonic Antigen Levels and Survival in Stage III Colon Cancer: Post hoc Analysis of the MOSAIC and PETACC-8 Trials. Cancer Epidemiol Biomarkers Prev 2020; 28:1153-1161. [PMID: 31263053 DOI: 10.1158/1055-9965.epi-18-0867] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/16/2018] [Accepted: 04/16/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND We explored and validated the association of postoperative carcinoembryonic antigen (CEA) with disease-free survival (DFS) and overall survival (OS) in stage III colon cancer. METHODS Patients with stage III colon cancer from the MOSAIC and PETACC-8 trials were enrolled. The relation between CEA and outcomes was continuously modeled with the restricted cubic splines (RCS) method. Association of CEA with outcomes was assessed by the Kaplan-Meier method, with two risk groups among patients with a CEA level ≤5 ng/mL. Multivariate Cox proportional hazard models were constructed. RESULTS The CEA level was available in 1,292 (96%) and 2,477 (97%) patients in the discovery and validation cohorts. The RCS analysis confirmed that patients with a CEA level >5 ng/mL were at highest risk of recurrence or death and those with a CEA level ≤5 ng/mL presented a heterogeneous risk population. In the discovery cohort, the 3-year DFS rate was 75%, 65%, and 45% in a group of patients with CEA level of 0-1.30 ng/mL (n = 630), 1.30-5 ng/mL (n = 613), and >5 ng/mL (n = 49), respectively (P < 0.001). CEA was independently associated with endpoints. All findings were confirmed in the validation cohort. CONCLUSIONS Postoperative CEA level was highly and independently associated with DFS and OS, especially in patients with a CEA level of ≤5 ng/mL, suggesting that this cutoff is not optimal. IMPACT CEA levels should be applied more accurately in future trials and clinical practice.
Collapse
Affiliation(s)
- Edouard Auclin
- Sorbonne Paris-Cité, Paris Descartes University, Hepato-Gastroenterology and Gastrointestinal Oncology Department, Hôpital Européen Georges Pompidou, Paris, France.,Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France.,University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Julien Taieb
- Sorbonne Paris-Cité, Paris Descartes University, Hepato-Gastroenterology and Gastrointestinal Oncology Department, Hôpital Européen Georges Pompidou, Paris, France
| | - Come Lepage
- Department of Gastroenterology, CHU Le Bocage, INSERM U1231, Dijon, France
| | - Thomas Aparicio
- Department of Gastroenterology and Digestive Oncology, Hôpital Saint Louis, Sorbonne Paris-Cité, Paris Diderot University, Paris, France
| | - Roger Faroux
- Department of Gastroenterology, Centre Hospitalier Départemental Les Oudairies, La Roche-Sur-Yon, France
| | - Enrico Mini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Gunnar Folprecht
- Medical Department I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Ramon Salazar
- Institut Catala d'Oncologia, Oncobell Program, IDIBELL, CIBERONC, University of Barcelona. L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Maria Banzi
- Unit of Medical Oncology, Clinical Cancer Center, AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Christophe Louvet
- Unit of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - Jean-Luc Van Laethem
- Gastroenterology and Digestive Department, Hopital Erasme, Université Libre de Bruxelles, Belgium
| | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), CIBERONC, University of Barcelona, Barcelona, Spain
| | - Tamas Hickish
- Department of Oncology, Royal Bournemouth Hospital and Bournemouth University, Bournemouth, England
| | - Aimery de Gramont
- Department of Oncology, Institut Hospitalier Franco-Britannique, Levallois-Perret, France
| | - Thierry André
- Sorbonne University and Department of Medical Oncology, Hospital Saint Antoine, Paris, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France. .,University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,Oncology Multidisciplinary Research Group (GERCOR), Paris, France
| |
Collapse
|
41
|
Zhang B, Wang L, Liu Z, Shao B, Jiang W, Shu P. Integrated analysis identifies an immune-based prognostic signature for the mesenchymal identity in colorectal cancer. Medicine (Baltimore) 2020; 99:e20617. [PMID: 32569190 PMCID: PMC7310905 DOI: 10.1097/md.0000000000020617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) has been divided into 4 consensus molecular subtypes (CMSs), of which CMS4 has the mesenchymal identity and the highest relapse rate. Our goal is to develop a prognostic signature by integrating the immune system and mesenchymal modalities involved in CMS4. METHODS The gene expression profiles collected from 5 public datasets were applied to this study, including 1280 samples totally. Network analysis was applied to integrate the mesenchymal modalities and immune signature to establish an immune-based prognostic signature for CRC (IPSCRC). RESULTS We identified 6 immune genes as key factors of CMS4 and established the IPSCRC. The IPSCRC could significantly divide patients into high- and low- risk groups in terms of relapse-free survival (RFS) and overall survival (OS) and in discovery (RFS: P < .0001) and 4 independent validation sets (RFS range: P = .01 to <.0001; OS range: P = .02-.0004). After stage stratification, the IPSCRC could still distinguish poor prognosis patients in discovery (RFS: P = .04) and validation cohorts (RFS range: P = .04-.007) within stage II in terms of RFS. Further, in multivariate analysis, the IPSCRC remained an independent predictor of prognosis. Moreover, Macrophage M2 was significantly enriched in the high-risk group, while plasma cells enriched in the low-risk group. CONCLUSION We propose an immune-based signature identified by network analysis, which is a promising prognostic biomarker and help for the selection of CRC patients who might benefit from more rigorous therapies. Further prospective studies are warranted to test and validate its efficiency for clinical application.
Collapse
Affiliation(s)
| | | | | | | | | | - Peng Shu
- Molecular Laboratory, Beilun People's Hospital, Ningbo, China
| |
Collapse
|
42
|
Circulating Tumor DNA as a Novel Biomarker Optimizing Chemotherapy for Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12061566. [PMID: 32545750 PMCID: PMC7352651 DOI: 10.3390/cancers12061566] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022] Open
Abstract
Liquid biopsy is a minimally invasive method for detecting soluble factors, including circulating tumor DNA (ctDNA), in body fluids. ctDNA carrying tumor-specific genetic or epigenetic alterations is released into circulation from tumor cells. ctDNA in the plasma contains somatic mutations that have occurred in the tumor, and reflects tumor progression and therapeutic effects promptly and accurately. Furthermore, ctDNA is useful for early detection of recurrence and estimation of prognosis and may be utilized for diagnosis and personalized medicine for treatment selection. Thus, in the near future, it will be possible to select the most appropriate treatment based on real-time genetic information using ctDNA.
Collapse
|
43
|
García-Alfonso P, García-Carbonero R, García-Foncillas J, Pérez-Segura P, Salazar R, Vera R, Ramón Y Cajal S, Hernández-Losa J, Landolfi S, Musulén E, Cuatrecasas M, Navarro S. Update of the recommendations for the determination of biomarkers in colorectal carcinoma: National Consensus of the Spanish Society of Medical Oncology and the Spanish Society of Pathology. Clin Transl Oncol 2020; 22:1976-1991. [PMID: 32418154 PMCID: PMC7505870 DOI: 10.1007/s12094-020-02357-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/28/2020] [Indexed: 12/16/2022]
Abstract
In this update of the consensus of the Spanish Society of Medical Oncology (Sociedad Española de Oncología Médica—SEOM) and the Spanish Society of Pathology (Sociedad Española de Anatomía Patológica—SEAP), advances in the analysis of biomarkers in advanced colorectal cancer (CRC) as well as susceptibility markers of hereditary CRC and molecular biomarkers of localized CRC are reviewed. Recently published information on the essential determination of KRAS, NRAS and BRAF mutations and the convenience of determining the amplification of human epidermal growth factor receptor 2 (HER2), the expression of proteins in the DNA repair pathway and the study of NTRK fusions are also evaluated. From the pathological point of view, the importance of analysing the tumour budding and poorly differentiated clusters, and its prognostic value in CRC is reviewed, as well as the impact of molecular lymph node analysis on lymph node staging in CRC. The incorporation of pan-genomic technologies, such as next-generation sequencing (NGS) and liquid biopsy in the clinical management of patients with CRC is also outlined. All these aspects are developed in this guide, which, like the previous one, will remain open to any necessary revision in the future.
Collapse
Affiliation(s)
- P García-Alfonso
- Departament of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - R García-Carbonero
- Departament of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Madrid, Spain
| | - J García-Foncillas
- Departament of Medical Oncology, Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - P Pérez-Segura
- Departament of Medical Oncology, Hospital Clínico Universitario San Carlos, CIBERONC, Madrid, Spain
| | - R Salazar
- Departament of Medical Oncology, ICO L'Hospitalet, Oncobell Program (IDIBELL), CIBERONC, Hospitalet de Llobregat, Spain
| | - R Vera
- Departament of Medical Oncology, Complejo Hospitalario de Navarra; Navarrabiomed, IDISNA, Pamplona, Spain
| | - S Ramón Y Cajal
- Department of Pathology, Hospital Universitario Vall D'Hebron, CIBERONC, Barcelona, Spain
| | - J Hernández-Losa
- Department of Pathology, Hospital Universitario Vall D'Hebron, CIBERONC, Barcelona, Spain
| | - S Landolfi
- Department of Pathology, Hospital Universitario Vall D'Hebron, CIBERONC, Barcelona, Spain
| | - E Musulén
- Department of Pathology, Hospital Universitari General de Catalunya, Grupo Quirónsalud, Sant Cugat del Vallès, Spain.,Cancer Epigenetics Group, Institut de Recerca Contra La Leucèmia Josep Carreras, Badalona, Spain
| | - M Cuatrecasas
- Department of Pathology, Hospital Clinic, CIBERehd, Barcelona, Spain
| | - S Navarro
- Department of Pathology, University of Valencia, Hospital Clínico Universitario de Valencia, CIBERONC, Valencia, Spain
| |
Collapse
|
44
|
Shinto E, Yoshida Y, Kajiwara Y, Okamoto K, Mochizuki S, Yamadera M, Shiraishi T, Nagata K, Tsuda H, Hase K, Kishi Y, Ueno H. Clinical Significance of a Gene Signature Generated from Tumor Budding Grade in Colon Cancer. Ann Surg Oncol 2020; 27:4044-4054. [PMID: 32328985 DOI: 10.1245/s10434-020-08498-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Tumor budding, a microscopic finding of dedifferentiation at the invasive margin, has been reported as a definite prognostic marker in colon cancer (CC). Herein, we aimed to generate a molecular budding signature (MBS) based on DNA microarray data and to examine its prognostic significance. METHODS Frozen tissue samples from 85 patients with stage II/III CC were used for DNA microarray analyses. First, we selected candidate genes that were differentially expressed (twofold change) between the invasive frontal regions and corresponding tumor centers of three extremely high-grade budding tumors. Subsequently, using microarray data from whole-tissue sections of the 85 patients, we selected MBS-constituent genes from the candidates based on correlation to the pathological budding grade. The MBS score was calculated using the sum of the logarithm of the expression of each gene. RESULTS We selected seven MBS-constituent genes: MSLN, SLC4A11, WNT11, SCEL, RUNX2, MGAT3, FOXC1. A comparison of relapse-free survival (RFS) rates revealed a significant impact of the MBS score [5-year RFS, 77.4% (score-high) vs. 95.1% (score-low); P = 0.044]. Analyses of public databases revealed that low MBS score patients exhibited better prognosis than those with high-score cancers (GSE14333: 5-year RFS, 83.1% vs. 66.6%, P = 0.028; GSE39582: 5-year disease-free survival, 72.2% vs. 58.1%, P = 0.0005). Multivariate analysis revealed that the MBS score is an independent prognostic indicator in GSE39582 (hazard ratio, 1.611; P = 0.013). CONCLUSIONS We developed a new gene classification method, i.e., MBS, and demonstrated its clinical relevance as an indicator of high recurrence risk of CC.
Collapse
Affiliation(s)
- Eiji Shinto
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | | | - Yoshiki Kajiwara
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Koichi Okamoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Satsuki Mochizuki
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Masato Yamadera
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Takehiro Shiraishi
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Ken Nagata
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hitoshi Tsuda
- Department of Pathology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Kazuo Hase
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
45
|
Postoperative C-reactive protein/albumin ratio is a biomarker of risk of recurrence and need for adjuvant chemotherapy for stage III colorectal cancer. Int J Clin Oncol 2020; 25:1318-1326. [PMID: 32279124 DOI: 10.1007/s10147-020-01672-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/30/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Adjuvant chemotherapy is generally recommended for patients with stage III colorectal cancer. Even with adjuvant chemotherapy, 20-30% of such patients develop recurrences; the risk factors for recurrence are currently unclear. The preoperative systemic inflammation index has been linked to poor prognoses in patients with colorectal cancer; however, the relationship between postoperative systemic inflammation index and recurrence is unclear. We aimed to evaluate the association between preoperative and postoperative systemic inflammation indexes and recurrence in patients with stage III colorectal cancer. METHODS The following laboratory data of 133 patients with stage III colorectal cancer were analyzed: preoperative and postoperative C-reactive protein/albumin ratios (CAR); neutrophil to lymphocyte ratios (NLR); and platelet to lymphocyte ratios (PLR) and their relationships with recurrence analyzed. RESULTS The optimal cutoff values for systemic inflammation indexes were determined by examining receiver operating characteristic curves. Multivariate analyses indicated that N-stage, postoperative complications, preoperative NLR, and postoperative CAR were independent predictors of recurrence-free survival (RFS). Postoperative CAR was also an independent predictor of overall survival (OS). Patients with postoperative CAR ≥ 0.035 who did not receive adjuvant chemotherapy had shorter RFS and OS than those who did. There were no significant differences in RFS and OS between patients with postoperative CAR < 0.035 who did and did not receive adjuvant chemotherapy. CONCLUSIONS Postoperative CAR is strongly associated with poor prognosis in patients with stage III colorectal cancer and is a useful biomarker for determining whether adjuvant chemotherapy should be administered.
Collapse
|
46
|
Zhou R, Sun H, Zheng S, Zhang J, Zeng D, Wu J, Huang Z, Rong X, Bin J, Liao Y, Shi M, Liao W. A stroma-related lncRNA panel for predicting recurrence and adjuvant chemotherapy benefit in patients with early-stage colon cancer. J Cell Mol Med 2020; 24:3229-3241. [PMID: 31989761 PMCID: PMC7077592 DOI: 10.1111/jcmm.14999] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/28/2022] Open
Abstract
The heterogeneity in prognoses and chemotherapeutic responses of colon cancer patients with similar clinical features emphasized the necessity for new biomarkers that help to improve the survival prediction and tailor therapies more rationally and precisely. In the present study, we established a stroma‐related lncRNA signature (SLS) based on 52 lncRNAs to comprehensively predict clinical outcome. The SLS model could not only distinguish patients with different recurrence and mortality risks through univariate analysis, but also served as an independent factor for relapse‐free and overall survival. Compared with the conventionally used TNM stage system, the SLS model clearly possessed higher predictive accuracy. Moreover, the SLS model also effectively screened chemotherapy‐responsive patients, as only patients in the low‐SLS group could benefit from adjuvant chemotherapy. The following cell infiltration and competing endogenous RNA (ceRNA) network functional analyses further confirmed the association between the SLS model and stromal activation‐related biological processes. Additionally, this study also identified three phenotypically distinct colon cancer subtypes that varied in clinical outcome and chemotherapy benefits. In conclusion, our SLS model may be a significant determinant of survival and chemotherapeutic decision‐making in colon cancer and may have a strong clinical transformation value.
Collapse
Affiliation(s)
- Rui Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huiying Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Siting Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingwen Zhang
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenhua Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Akagi T, Inomata M. Essential advances in surgical and adjuvant therapies for colorectal cancer 2018-2019. Ann Gastroenterol Surg 2020; 4:39-46. [PMID: 32021957 PMCID: PMC6992683 DOI: 10.1002/ags3.12307] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/18/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
Surgical resection and adjuvant chemotherapy are the only treatment modalities for localized colorectal cancer that can obtain a "cure." The goal in surgically treating primary colorectal cancer is complete tumor removal along with dissection of systematic D3 lymph nodes. Adjuvant treatment controls recurrence and improves the prognosis of patients after they undergo R0 resection. Various clinical studies have promoted the gradual spread and clinical use of new surgical approaches such as laparoscopic surgery, robotic surgery, and transanal total mesorectal excision (TaTME). Additionally, the significance of adjuvant chemotherapy has been established and it is now recommended in the JSCCR (the Japanese Society for Cancer of the Colon and Rectum) guideline as a standard treatment. Herein, we review and summarize current surgical treatment and adjuvant chemotherapy for localized colorectal cancer and discuss recent advances in personalized medicine related to adjuvant chemotherapy.
Collapse
Affiliation(s)
- Tomonori Akagi
- Department of Gastroenterological and Pediatric SurgeryFaculty of MedicineOita UniversityYufu‐CityJapan
| | - Masafumi Inomata
- Department of Gastroenterological and Pediatric SurgeryFaculty of MedicineOita UniversityYufu‐CityJapan
| |
Collapse
|
48
|
Chan GHJ, Chee CE. Making sense of adjuvant chemotherapy in colorectal cancer. J Gastrointest Oncol 2019; 10:1183-1192. [PMID: 31949938 PMCID: PMC6954995 DOI: 10.21037/jgo.2019.06.03] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
Surgical resection is the only curative treatment for locoregional colon cancer. The goal of adjuvant chemotherapy is to eradicate micro-metastatic disease and improve survival. This has been most clearly demonstrated in stage III (node-positive) disease, whereas benefit of adjuvant chemotherapy in stage II disease remains controversial. In stage III colon cancer, 6 months of adjuvant chemotherapy with oxaliplatin-based chemotherapy have been accepted as the standard for the last 15 years. The recent IDEA collaboration has challenged this in 2018; while overall was a negative non-inferiority study, pre-planned subset analyses do support that for patients with low-risk stage III disease, 3 months of XELOX (capecitabine and oxaliplatin combination) is non-inferior to 6 months. In stage II colon cancer, where the potential benefit of adjuvant chemotherapy is small, the emergence of biomarkers has helped in decision-making. Tumors with deficient mismatch repair protein (dMMR) do not benefit from 5-fluorouracil-based chemotherapy. For patients with high clinicopathological risk stage II disease with proficient mismatch repair proteins and good performance status, six months of adjuvant chemotherapy is still recommended. In the management of rectal cancers, where the risk of local recurrence is higher, chemoradiation (CRT) is often included as neoadjuvant or adjuvant therapy in the management of stage II and III rectal cancer. The benefit of adjuvant chemotherapy in rectal cancer has been extrapolated from adjuvant colon cancer studies with updated results from adjuvant rectal cancer studies demonstrating similar benefits. This review summarizes the current landscape of adjuvant therapy for patients with resected stage II and III colorectal cancer.
Collapse
Affiliation(s)
- Gloria H J Chan
- Department of Hematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Cheng E Chee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| |
Collapse
|
49
|
Differential Survival Benefits of 5-Fluorouracil-Based Adjuvant Chemotherapy for Patients With Microsatellite-Stable Stage III Colorectal Cancer According to the Tumor Budding Status: A Retrospective Analysis. Dis Colon Rectum 2019; 62:1316-1325. [PMID: 31567925 DOI: 10.1097/dcr.0000000000001480] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Recent research has established tumor budding as a prognostic factor and a possible histomorphologic reflection of epithelial-mesenchymal transition in colorectal cancer, highlighting the ability of cancer cells exhibiting epithelial-mesenchymal transition to resist chemotherapy. OBJECTIVE This study aimed to investigate the clinical benefits of adjuvant chemotherapy according to the tumor budding status in microsatellite-stable stage III colorectal cancer. DESIGN This was a retrospective study of 2 cohorts. SETTINGS The study was conducted at the National Defense Medical College in Japan. PATIENTS We reviewed 2 data sets of patients with microsatellite-stable stage III colorectal cancer with curatively intended surgery (R0) from 1999 to 2005 (first cohort; n = 203) and 2006 to 2012 (second cohort; n = 346). In both cohorts, 128 and 203 patients received 5-fluorouracil-based adjuvant chemotherapy and 75 and 143 patients did not. MAIN OUTCOME MEASURES We assessed the benefits of adjuvant chemotherapy according to the grades of tumor budding based on the cancer-specific survival. RESULTS In low-budding tumors, the chemotherapy group exhibited better cancer-specific survival than the surgery-alone group (first cohort, 93.1% vs 65.5%, p = 0.001; second cohort, 94.0% vs 76.0%, p < 0.0001). Conversely, the prognostic difference between the chemotherapy and surgery-alone groups was statistically insignificant in high-budding tumors (first cohort, 59.7% vs 52.4%, p = 0.57; second cohort, 83.1% vs 75.6%, p = 0.19). The multivariate analysis corroborated the benefits of adjuvant chemotherapy in low-budding tumors (first cohort, p = 0.002, HR = 0.28; second cohort, p < 0.0001, HR = 0.23) but not in high-budding tumors. LIMITATIONS Postoperative adjuvant chemotherapy and treatments for recurrence were not homogeneous, and the patient backgrounds differed between the chemotherapy and surgery alone groups. CONCLUSIONS The high-budding group demonstrated resistance to 5-fluorouracil-based chemotherapy, whereas the low-budding group exhibited significant survival benefits from adjuvant chemotherapy in stage III colorectal cancer. See Video Abstract at http://links.lww.com/DCR/B14. BENEFICIOS DE SUPERVIVENCIA DIFERENCIAL DE LA QUIMIOTERAPIA ADYUVANTE BASADA EN 5-FLUOROURACILO PARA PACIENTES CON CÁNCER COLORRECTAL EN ESTADIO III ESTABLE CON MICROSATÉLITE SEGÚN EL ESTADO DE BROTACIÓN DEL TUMOR: UN ANÁLISIS RETROSPECTIVO:: Investigaciones recientes han establecido la aparición de tumores como un factor pronóstico y una posible reflexión histomorfológica de la transición epitelial-mesenquimatosa en el cáncer colorrectal, destacando la capacidad de las células cancerosas que presentan una transición epitelio-mesenquimática para resistir la quimioterapia.El objetivo de este estudio es investigar los beneficios clínicos de la quimioterapia adyuvante según el estado de brotación del tumor en el cáncer colorrectal en estadio III estable con microsatélite.Este fue un estudio retrospectivo de dos cohortes.El estudio se realizó en la Escuela de Medicina de la Defensa Nacional de Japón.Revisamos dos conjuntos de datos de pacientes con cáncer colorrectal en estadio III estable con microsatélite con cirugía de intención curativa (R0) de 1999 a 2005 (primera cohorte; n = 203) y 2006 a 2012 (segunda cohorte; n = 346). En ambas cohortes, 128 y 203 pacientes recibieron quimioterapia adyuvante basada en 5-fluorouracilo y 75 y 143 pacientes no, respectivamente.Evaluamos los beneficios de la quimioterapia adyuvante de acuerdo con los grados de brotación del tumor en función de la supervivencia específica del cáncer.n los tumores con brotes bajos, el grupo de quimioterapia mostró una mejor supervivencia específica al cáncer que el grupo con cirugía sola (primera cohorte, 93.1% vs. 65.5%, p = 0.001; segunda cohorte, 94.0% vs. 76.0%, p < 0.0001). A la inversa, la diferencia pronóstica entre los grupos de quimioterapia y cirugía sola fue estadísticamente insignificante en los tumores de brotes elevados (primera cohorte, 59.7% vs. 52.4%, p = 0.57; segunda cohorte, 83.1% vs. 75.6%, p = 0.19). El análisis multivariado corroboró los beneficios de la quimioterapia adyuvante en los tumores de brotes bajos (primera cohorte, p = 0,002, índice de riesgo: 0,28; segundo cohorte, p <0,0001, índice de riesgo: 0,23) pero no en los tumores de alto brote.a quimioterapia adyuvante postoperatoria y los tratamientos para la recurrencia no fueron homogéneos, y los antecedentes de los pacientes difirieron entre los grupos de quimioterapia y cirugía sola.El grupo de alto brote demostró resistencia a la quimioterapia basada en 5-fluorouracilo, mientras que el grupo de bajo brote mostró beneficios significativos de supervivencia de la quimioterapia adyuvante en el cáncer colorrectal en estadio III. Vea el Resumen del Video en http://links.lww.com/DCR/B14.
Collapse
|
50
|
Bender U, Rho YS, Barrera I, Aghajanyan S, Acoba J, Kavan P. Adjuvant therapy for stages II and III colon cancer: risk stratification, treatment duration, and future directions. Curr Oncol 2019; 26:S43-S52. [PMID: 31819709 PMCID: PMC6878933 DOI: 10.3747/co.26.5605] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background To date, the role of adjuvant systemic therapy in stages ii and iii colon cancer remains a topic of interest and debate. The objective of the present review was to assess the most recent data, specifically addressing methods of risk stratification, duration of therapy, and future directions. Methods PubMed and medline were searched for literature pertinent to adjuvant chemotherapy in either stage ii or stage iii colorectal cancer. Summary Locoregional disease, histopathology, age, laterality, and a number of other biologic and molecular markers appear to have a role in disease risk stratification. The duration of adjuvant therapy for stage iii disease can vary based on risk factors, but use of adjuvant therapy and duration of therapy in stage ii disease remain controversial. Future directions should include genomic assays and improved study design to provide concrete evidence about the duration of adjuvant folfox or capox and about other types of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- U Bender
- Gerald Bronfman Department of Oncology, McGill University Faculty of Medicine, Montreal, QC
| | - Y S Rho
- University of Hawaii Internal Medicine Program, Honolulu, HI, U.S.A
| | - I Barrera
- Gerald Bronfman Department of Oncology, McGill University Faculty of Medicine, Montreal, QC
| | - S Aghajanyan
- Gerald Bronfman Department of Oncology, McGill University Faculty of Medicine, Montreal, QC
| | - J Acoba
- University of Hawaii Internal Medicine Program, Honolulu, HI, U.S.A
- University of Hawaii Cancer Center, Honolulu, HI, U.S.A
| | - P Kavan
- Gerald Bronfman Department of Oncology, McGill University Faculty of Medicine, Montreal, QC
| |
Collapse
|