1
|
Milan N, Navarria F, Cecchin E, De Mattia E. Somatic pharmacogenomics in the treatment prognosis of locally advanced rectal cancer patients: a narrative review of the literature. Expert Rev Clin Pharmacol 2024; 17:683-719. [PMID: 39046146 DOI: 10.1080/17512433.2024.2375449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Standard treatment for patients with locally advanced rectal cancer (LARC) includes neoadjuvant chemoradiotherapy (nCRT) with fluoropyrimidines, followed by surgical excision. The newly introduced therapeutic strategies propose intensified regimens or more conservative approaches based on risk stratification algorithms that currently include clinicoradiological criteria but not molecular variables. How to better stratify patients is a burning clinical question, and pharmacogenomics may prove useful in identifying new genetic markers that could be incorporated into clinical algorithms to personalize nCRT. An emerging area could be the evaluation of somatic mutations as potential genetic markers that correlate with patient prognosis. Tumor mutations in the RAS/BRAF genes, as well as microsatellite instability (MSI) status, are currently used in treatment selection for colorectal cancer (CRC); however, their clinical value in LARC is still unclear. AREA COVERED This literature review discusses the relevant findings on the prognostic role of mutations in the key oncogenes RAS, KRAS, BRAF, PIK3CA, SMAD4 and TP53, including MSI status in LARC patients treated with nCRT. EXPERT OPINION KRAS proved to be the most promising marker, consistently associated with poorer disease-free survival and overall survival. Therefore, KRAS could be a good candidate for integration into the risk stratification algorithm to develop a personalized treatment.
Collapse
Affiliation(s)
- Noemi Milan
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Federico Navarria
- Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Elena De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
2
|
Shu P, Liu N, Luo X, Tang Y, Chen Z, Li D, Miao D, Duan J, Yan O, Sheng L, Ouyang G, Wang S, Jiang D, Deng X, Wang Z, Li Q, Wang X. An immune-related gene prognostic prediction risk model for neoadjuvant chemoradiotherapy in rectal cancer using artificial intelligence. Front Oncol 2024; 14:1294440. [PMID: 38406803 PMCID: PMC10889124 DOI: 10.3389/fonc.2024.1294440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/24/2024] [Indexed: 02/27/2024] Open
Abstract
Background This study aimed to establish and validate a prognostic model based on immune-related genes (IRGPM) for predicting disease-free survival (DFS) in patients with locally advanced rectal cancer (LARC) undergoing neoadjuvant chemoradiotherapy, and to elucidate the immune profiles associated with different prognostic outcomes. Methods Transcriptomic and clinical data were sourced from the Gene Expression Omnibus (GEO) database and the West China Hospital database. We focused on genes from the RNA immune-oncology panel. The elastic net approach was employed to pinpoint immune-related genes significantly impacting DFS. We developed the IRGPM for rectal cancer using the random forest technique. Based on the IRGPM, we calculated prognostic risk scores to categorize patients into high-risk and low-risk groups. Comparative analysis of immune characteristics between these groups was conducted. Results In this study, 407 LARC samples were analyzed. The elastic net identified a signature of 20 immune-related genes, forming the basis of the IRGPM. Kaplan-Meier survival analysis revealed a lower 5-year DFS in the high-risk group compared to the low-risk group. The receiver operating characteristic (ROC) curve affirmed the model's robust predictive capability. Validation of the model was performed in the GSE190826 cohort and our institution's cohort. Gene expression differences between high-risk and low-risk groups predominantly related to cytokine-cytokine receptor interactions. Notably, the low-risk group exhibited higher immune scores. Further analysis indicated a greater presence of activated B cells, activated CD8 T cells, central memory CD8 T cells, macrophages, T follicular helper cells, and type 2 helper cells in the low-risk group. Additionally, immune checkpoint analysis revealed elevated PDCD1 expression in the low-risk group. Conclusions The IRGPM, developed through random forest and elastic net methodologies, demonstrates potential in distinguishing DFS among LARC patients receiving standard treatment. Notably, the low-risk group, as defined by the IRGPM, showed enhanced activation of adaptive immune responses within the tumor microenvironment.
Collapse
Affiliation(s)
- Pei Shu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ning Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Luo
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, China
- School of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Yuanling Tang
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhebin Chen
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, China
- School of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Dandan Li
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dong Miao
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, China
- School of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Jiayu Duan
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ouying Yan
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Leiming Sheng
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ganlu Ouyang
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Sen Wang
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, China
- School of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Dan Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Sichuan University-University of Oxford Huaxi Joint Center for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiangbing Deng
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ziqiang Wang
- Sichuan University-University of Oxford Huaxi Joint Center for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qingyun Li
- Genecast Biotechnology Co., Ltd., Xishan District, Wuxi, Jiangsu, China
| | - Xin Wang
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan University-University of Oxford Huaxi Joint Center for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Liu Y, Yang Y, Ni F, Tai G, Yu C, Jiang X, Wang D. Research on radiotherapy related genes and prognostic target identification of rectal cancer based on multi-omics. J Transl Med 2023; 21:856. [PMID: 38012642 PMCID: PMC10680259 DOI: 10.1186/s12967-023-04753-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Radiosensitivity of rectal cancer is related to the radiotherapy efficacy and prognosis of patients with rectal cancer, and the genes and molecular mechanisms related to radiosensitivity of rectal cancer have not been clarified. We explored the radiosensitivity related genes of rectal cancer at a multi omics level. METHODS mRNA expression data and rectum adenocarcinoma (READ) data were obtained from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus Database (GEO) (GSE150082, GSE60331, GSE46862, GSE46862). Differentially expressed genes between radiotherapy sensitive group and radiotherapy insensitive group were screened. GO analysis and KEGG pathway analysis were performed for differentially expressed genes. Among the differentially expressed genes, five core genes associated with rectal cancer prognosis were selected using random survival forest analysis. For these five core genes, drug sensitivity analysis, immune cell infiltration analysis, TISIDB database immune gene correlation analysis, GSEA enrichment analysis, construction of Nomogram prediction model, transcriptional regulatory network analysis, and qRT-PCR validation was performed on human rectal adenocarcinoma tissue. RESULTS We found that 600 up-regulated genes and 553 down-regulated genes were significantly different between radiotherapy sensitive group and radiotherapy insensitive group in rectal cancer. Five key genes, TOP2A, MATR3, APOL6, JOSD1, and HOXC6, were finally screened by random survival forest analysis. These five key genes were associated with different immune cell infiltration, immune-related genes, and chemosensitivity. A comprehensive transcriptional regulatory network was constructed based on these five core genes. qRT-PCR revealed that MATR3 expression was different in rectal cancer tissues and adjacent non-cancerous tissues, while APOL6, HOXC6, JOSD1, and TOP2A expression was not different. CONCLUSION Five radiosensitivity-related genes related to the prognosis of rectal cancer: TOP2A, MATR3, APOL6, JOSD1, HOXC6, are involved in multiple processes such as immune cell infiltration, immune-related genes, chemosensitivity, signaling pathways and transcriptional regulatory networks and may be potential biomarkers for radiotherapy of rectal cancer.
Collapse
Affiliation(s)
- Yi Liu
- Department of Radiotherapy, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Yanguang Yang
- Department of Radiotherapy, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Feng Ni
- Department of Radiotherapy, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Guomei Tai
- Department of Radiotherapy, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Cenming Yu
- Department of Radiotherapy, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Xiaohui Jiang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China.
| | - Ding Wang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China.
| |
Collapse
|
4
|
Sartorius D, Blume ML, Fleischer JR, Ghadimi M, Conradi LC, De Oliveira T. Implications of Rectal Cancer Radiotherapy on the Immune Microenvironment: Allies and Foes to Therapy Resistance and Patients' Outcome. Cancers (Basel) 2023; 15:5124. [PMID: 37958298 PMCID: PMC10650490 DOI: 10.3390/cancers15215124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Aside from surgical resection, locally advanced rectal cancer is regularly treated with neoadjuvant chemoradiotherapy. Since the concept of cancer treatment has shifted from only focusing on tumor cells as drivers of disease progression towards a broader understanding including the dynamic tumor microenvironment (TME), the impact of radiotherapy on the TME and specifically the tumor immune microenvironment (TIME) is increasingly recognized. Both promoting as well as suppressing effects on anti-tumor immunity have been reported in response to rectal cancer (chemo-)radiotherapy and various targets for combination therapies are under investigation. A literature review was conducted searching the PubMed database for evidence regarding the pleiotropic effects of (chemo-)radiotherapy on the rectal cancer TIME, including alterations in cytokine levels, immune cell populations and activity as well as changes in immune checkpoint proteins. Radiotherapy can induce immune-stimulating and -suppressive alterations, potentially mediating radioresistance. The response is influenced by treatment modalities, including the dosage administered and the highly individual intrinsic pre-treatment immune status. Directly addressing the main immune cells of the TME, this review aims to highlight therapeutical implications since efficient rectal cancer treatment relies on personalized strategies combining conventional therapies with immune-modulating approaches, such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany; (D.S.); (M.L.B.); (J.R.F.); (M.G.)
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany; (D.S.); (M.L.B.); (J.R.F.); (M.G.)
| |
Collapse
|
5
|
Tse BCY, Bergamin S, Steffen P, Hruby G, Pavlakis N, Clarke SJ, Evans J, Engel A, Kneebone A, Molloy MP. CD11c + and IRF8 + cell densities in rectal cancer biopsies predict outcomes of neoadjuvant chemoradiotherapy. Oncoimmunology 2023; 12:2238506. [PMID: 37485033 PMCID: PMC10361136 DOI: 10.1080/2162402x.2023.2238506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 07/09/2023] [Accepted: 07/16/2023] [Indexed: 07/25/2023] Open
Abstract
Approximately 20% of locally advanced rectal cancer (LARC) patients treated preoperatively with chemoradiotherapy (CRT) achieve pathologically confirmed complete regression. However, there are no clinically implemented biomarkers measurable in biopsies that are predictive of tumor regression. Here, we conducted multiplexed immunophenotyping of rectal cancer diagnostic biopsies from 16 LARC patients treated preoperatively with CRT. We identified that patients with greater tumor regression had higher tumor infiltration of pan-T cells and IRF8+HLA-DR+ cells prior to CRT. High IRF8+HLA-DR+ cell density was further associated with prolonged disease-specific survival with 83% survival at 5 y compared to 28% in patients with low infiltration. Contrastingly, low CD11c+ myeloid cell infiltration prior to CRT was a putative biomarker associated with longer 3- and 5-y disease-free survival. The results demonstrate the potential use of rectal cancer diagnostic biopsies to measure IRF8+ HLA-DR+ cells as predictors of CRT-induced tumor regression and CD11c+ myeloid cells as predictors of LARC patient survival.
Collapse
Affiliation(s)
- Benita C. Y. Tse
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Sarah Bergamin
- Department of Radiation Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Pascal Steffen
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - George Hruby
- Department of Radiation Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| | - Stephen J. Clarke
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| | - Justin Evans
- Colorectal Surgical Unit, Royal North Shore Hospital, Sydney, Australia
| | - Alexander Engel
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Colorectal Surgical Unit, Royal North Shore Hospital, Sydney, Australia
| | - Andrew Kneebone
- Department of Radiation Oncology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| | - Mark P. Molloy
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| |
Collapse
|
6
|
Takenaka IKTM, Bartelli TF, Defelicibus A, Sendoya JM, Golubicki M, Robbio J, Serpa MS, Branco GP, Santos LBC, Claro LCL, Dos Santos GO, Kupper BEC, da Silva IT, Llera AS, de Mello CAL, Riechelmann RP, Dias-Neto E, Iseas S, Aguiar S, Nunes DN. Exome and Tissue-Associated Microbiota as Predictive Markers of Response to Neoadjuvant Treatment in Locally Advanced Rectal Cancer. Front Oncol 2022; 12:809441. [PMID: 35392220 PMCID: PMC8982181 DOI: 10.3389/fonc.2022.809441] [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: 11/05/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical and pathological responses to multimodal neoadjuvant therapy in locally advanced rectal cancers (LARCs) remain unpredictable, and robust biomarkers are still lacking. Recent studies have shown that tumors present somatic molecular alterations related to better treatment response, and it is also clear that tumor-associated bacteria are modulators of chemotherapy and immunotherapy efficacy, therefore having implications for long-term survivorship and a good potential as the biomarkers of outcome. Here, we performed whole exome sequencing and 16S ribosomal RNA (rRNA) amplicon sequencing from 44 pre-treatment LARC biopsies from Argentinian and Brazilian patients, treated with neoadjuvant chemoradiotherapy or total neoadjuvant treatment, searching for predictive biomarkers of response (responders, n = 17; non-responders, n = 27). In general, the somatic landscape of LARC was not capable to predict a response; however, a significant enrichment in mutational signature SBS5 was observed in non-responders (p = 0.0021), as well as the co-occurrence of APC and FAT4 mutations (p < 0.05). Microbiota studies revealed a similar alpha and beta diversity of bacteria between response groups. Yet, the linear discriminant analysis (LDA) of effect size indicated an enrichment of Hungatella, Flavonifractor, and Methanosphaera (LDA score ≥3) in the pre-treatment biopsies of responders, while non-responders had a higher abundance of Enhydrobacter, Paraprevotella (LDA score ≥3) and Finegoldia (LDA score ≥4). Altogether, the evaluation of these biomarkers in pre-treatment biopsies could eventually predict a neoadjuvant treatment response, while in post-treatment samples, it could help in guiding non-operative treatment strategies.
Collapse
Affiliation(s)
| | - Thais F Bartelli
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Alexandre Defelicibus
- Laboratory of Bioinformatics and Computational Biology, International Center for Research, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Juan M Sendoya
- Laboratorio de Terapia Molecular y Celular - Genomics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariano Golubicki
- Oncology Unit, Hospital de Gastroenterología Carlos Bonorino Udaondo, Buenos Aires, Argentina.,Clinical Oncology, Intergrupo Argentino para el Tratamiento de los Tumores Gastrointestinales (IATTGI), Buenos Aires, Argentina
| | - Juan Robbio
- Clinical Oncology, Intergrupo Argentino para el Tratamiento de los Tumores Gastrointestinales (IATTGI), Buenos Aires, Argentina
| | - Marianna S Serpa
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Gabriela P Branco
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Luana B C Santos
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Laura C L Claro
- Department of Pathology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Bruna E C Kupper
- Colorectal Cancer Department, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Israel T da Silva
- Laboratory of Bioinformatics and Computational Biology, International Center for Research, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Andrea S Llera
- Laboratorio de Terapia Molecular y Celular - Genomics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Celso A L de Mello
- Department of Clinical Oncology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Emmanuel Dias-Neto
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil.,Laboratory of Neurosciences (LIM-27) Alzira Denise Hertzog Silva, Institute of Psychiatry, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Soledad Iseas
- Oncology Unit, Hospital de Gastroenterología Carlos Bonorino Udaondo, Buenos Aires, Argentina
| | - Samuel Aguiar
- Colorectal Cancer Department, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Diana Noronha Nunes
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil.,National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation (INCITO), São Paulo, Brazil
| |
Collapse
|
7
|
Nikolic A, Krivokapic Z. Nucleic acid-based markers of response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Surg Oncol 2022; 41:101743. [PMID: 35358913 DOI: 10.1016/j.suronc.2022.101743] [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: 10/07/2021] [Revised: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
Abstract
The progress that has been made in the treatment of rectal cancer has mostly resulted from multimodality strategy approach that combines surgery, chemotherapy and radiotherapy. In locally advanced rectal cancer (LARC), surgery remains the primary treatment, while neoadjuvant chemoradiotherapy (nCRT) is used to downsize or downstage the tumor before surgical resection. Highly variable response to nCRT observed in LARC patients raises the need for biomarkers to enable prediction and evaluation of treatment response in a more efficient and timely manner than currently available tools. The search for predictive biomarkers continues beyond blood proteins, which have failed in subsequent validation studies. This review presents nucleic acids-based markers and their predictive potential in LARC patients. Most of the candidate biomarkers come from relatively small single-institution studies. The only candidate biomarker that emerged as relevant in more than a single study was elevated levels of Fusobacterium nucleatum nucleic acids in tumor tissue. Considering that this marker is easily accessible through non-invasive analysis of faecal samples, its predictive potential is worth further validation. The other candidate nucleic acid-based biomarkers require more consistent studies on larger cohorts before they can be considered for use in clinical setting.
Collapse
Affiliation(s)
- Aleksandra Nikolic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.
| | - Zoran Krivokapic
- Clinic for Digestive Surgery, Clinical Center of Serbia, Belgrade, Serbia; Faculty of Medicine, University of Belgrade, Belgrade, Serbia; Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
8
|
Iseas S, Sendoya JM, Robbio J, Coraglio M, Kujaruk M, Mikolaitis V, Rizzolo M, Cabanne A, Ruiz G, Salanova R, Gualdrini U, Méndez G, Antelo M, Carballido M, Rotondaro C, Viglino J, Eleta M, Di Sibio A, Podhajcer OL, Roca E, Llera AS, Golubicki M, Abba MC. Prognostic Impact of An Integrative Landscape of Clinical, Immune, and Molecular Features in Non-Metastatic Rectal Cancer. Front Oncol 2022; 11:801880. [PMID: 35071006 PMCID: PMC8777220 DOI: 10.3389/fonc.2021.801880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Rectal Cancer (RC) is a complex disease that involves highly variable treatment responses. Currently, there is a lack of reliable markers beyond TNM to deliver a personalized treatment in a cancer setting where the goal is a curative treatment. Here, we performed an integrated characterization of the predictive and prognostic role of clinical features, mismatch-repair deficiency markers, HER2, CDX2, PD-L1 expression, and CD3-CD8+ tumor-infiltrating lymphocytes (TILs) coupled with targeted DNA sequencing of 76 non-metastatic RC patients assigned to total mesorectal excision upfront (TME; n = 15) or neoadjuvant chemo-radiotherapy treatment (nCRT; n = 61) followed by TME. Eighty-two percent of RC cases displayed mutations affecting cancer driver genes such as TP53, APC, KRAS, ATM, and PIK3CA. Good response to nCRT treatment was observed in approximately 40% of the RC cases, and poor pathological tumor regression was significantly associated with worse disease-free survival (DFS, HR = 3.45; 95%CI = 1.14-10.4; p = 0.028). High neutrophils-platelets score (NPS) (OR = 10.52; 95%CI=1.34-82.6; p = 0.025) and KRAS mutated cases (OR = 5.49; 95%CI = 1.06-28.4; p = 0.042) were identified as independent predictive factors of poor response to nCRT treatment in a multivariate analysis. Furthermore, a Cox proportional-hazard model showed that the KRAS mutational status was an independent prognostic factor associated with higher risk of local recurrence (HR = 9.68; 95%CI = 1.01-93.2; p <0.05) and shorter DFS (HR = 2.55; 95%CI = 1.05-6.21; p <0.05), while high CEA serum levels were associated with poor DFS (HR = 2.63; 95%CI = 1.01-6.85; p <0.05). Integrated clinical and molecular-based unsupervised analysis allowed us to identify two RC prognostic groups (cluster 1 and cluster 2) associated with disease-specific OS (HR = 20.64; 95%CI = 2.63-162.2; p <0.0001), metastasis-free survival (HR = 3.67; 95%CI = 1.22-11; p = 0.012), local recurrence-free survival (HR = 3.34; 95%CI = 0.96-11.6; p = 0.043) and worse DFS (HR = 2.68; 95%CI = 1.18-6.06; p = 0.012). The worst prognosis cluster 2 was enriched by stage III high-risk clinical tumors, poor responders to nCRT, with low TILs density and high frequency of KRAS and TP53 mutated cases compared with the best prognosis cluster 1 (p <0.05). Overall, this study provides a comprehensive and integrated characterization of non-metastatic RC cases as a new insight to deliver a personalized therapeutic approach.
Collapse
Affiliation(s)
- Soledad Iseas
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Juan M. Sendoya
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Juan Robbio
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
- Unidad de Investigación Traslacional, Laboratorio de Biología Molecular GENUIT, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Mariana Coraglio
- Proctology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Mirta Kujaruk
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Vanesa Mikolaitis
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Mariana Rizzolo
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Ana Cabanne
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Gonzalo Ruiz
- Biomakers Molecular Pathology and Research, Buenos Aires, Argentina
| | - Rubén Salanova
- Biomakers Molecular Pathology and Research, Buenos Aires, Argentina
| | - Ubaldo Gualdrini
- Proctology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Guillermo Méndez
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Marina Antelo
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Marcela Carballido
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Cecilia Rotondaro
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Julieta Viglino
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Martín Eleta
- Imaxe Image Diagnosis Center, Buenos Aires, Argentina
| | | | - Osvaldo L. Podhajcer
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Enrique Roca
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Andrea S. Llera
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Mariano Golubicki
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
- Unidad de Investigación Traslacional, Laboratorio de Biología Molecular GENUIT, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Martín Carlos Abba
- Basic and Applied Immunological Research Center, School of Medical Sciences, National University of La Plata, La Plata, Argentina
| |
Collapse
|
9
|
Ono Y, Cates JMM, Gonzalez RS. Can histologic features predict neoadjuvant therapy response in rectal adenocarcinoma? Pathol Res Pract 2021; 226:153608. [PMID: 34530256 DOI: 10.1016/j.prp.2021.153608] [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: 07/24/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/07/2023]
Abstract
Current standard therapy for locally advanced rectal cancer (LARC) is neoadjuvant therapy followed by surgical resection; however, treatment response is variable among patients. This study aimed to identify histologic features that predict tumor response. This retrospective study included 105 patients with LARC, all of whom underwent biopsy followed by neoadjuvant therapy and subsequent surgical resection. Each patient's initial biopsy was evaluated for tumor grade, tumor budding, intraepithelial lymphocytes, intraepithelial neutrophils, desmoplasia, apoptosis, adjacent stromal lymphocytes, signet ring cells, mucinous features, tumoral Paneth cells, dirty necrosis, microscopic ulceration, and prominent lymphoid aggregates. These histologic features, along with patient age at diagnosis and tumor microsatellite status, were compared to tumor regression grades from the respective resection specimens. No histologic factors in tumor biopsies predictive of treatment response in post-therapy resection specimens were identified. Histologic features in pre-therapy biopsy samples of LARC do not predict subsequent response to neoadjuvant therapy. Effective and reliable methods to predict response to neoadjuvant therapy in rectal cancer remain elusive.
Collapse
Affiliation(s)
- Yuho Ono
- Beth Israel Deaconess Medical Center, Department of Pathology, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Justin M M Cates
- Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Raul S Gonzalez
- Beth Israel Deaconess Medical Center, Department of Pathology, 330 Brookline Avenue, Boston, MA 02215, United States.
| |
Collapse
|
10
|
Momma T, Okayama H, Kanke Y, Fukai S, Onozawa H, Fujita S, Sakamoto W, Saito M, Ohki S, Kono K. Validation of Gene Expression-Based Predictive Biomarkers for Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Cancers (Basel) 2021; 13:cancers13184642. [PMID: 34572869 PMCID: PMC8467397 DOI: 10.3390/cancers13184642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (nCRT) followed by surgery is widely used for patients with locally advanced rectal cancer. However, response to nCRT varies substantially among patients, highlighting the need for predictive biomarkers that can distinguish non-responsive from responsive patients before nCRT. This study aimed to build novel multi-gene assays for predicting nCRT response, and to validate our signature and previously-reported signatures in multiple independent cohorts. METHODS Three microarray datasets of pre-therapeutic biopsies containing a total of 61 non-responders and 53 responders were used as the discovery cohorts to screen for genes that were consistently associated with nCRT response. The predictive values of signatures were tested in a meta-analysis using six independent datasets as the validation cohorts, consisted of a total of 176 non-responders and 99 responders. RESULTS We identified four genes, including BRCA1, GPR110, TNIK, and WDR4 in the discovery cohorts. Although our 4-gene signature and nine published signatures were evaluated, they were unable to predict nCRT response in the validation cohorts. CONCLUSIONS Although this is one of the largest studies addressing the validity of gene expression-based classifiers using pre-treatment biopsies from patients with rectal cancer, our findings do not support their clinically meaningful values to be predictive of nCRT response.
Collapse
Affiliation(s)
- Tomoyuki Momma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Hirokazu Okayama
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
- Correspondence: ; Tel.: +81-24-547-1259
| | - Yasuyuki Kanke
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Satoshi Fukai
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Hisashi Onozawa
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Shotaro Fujita
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Wataru Sakamoto
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Motonobu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Shinji Ohki
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
- Hospital Director, Shirakawa Kosei General Hospital, 2-1 Kamiyajiro, Shirakawa, Fukushima 961-0005, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| |
Collapse
|
11
|
Chen W, Farchoukh L, Seigh L, Hartman DJ, Pai RK. Combined histopathological risk score using TP53 protein expression, CD8 + T cell density and intratumoral budding is an independent predictor of neoadjuvant therapy response in rectal adenocarcinoma. Histopathology 2021; 79:826-835. [PMID: 34121230 DOI: 10.1111/his.14430] [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: 03/05/2021] [Revised: 04/29/2021] [Accepted: 06/11/2021] [Indexed: 12/17/2022]
Abstract
AIMS Neoadjuvant therapy is the recommended treatment for locally advanced rectal adenocarcinoma; however, there remains significant variability in response to therapy. Tumour protein 53 (TP53) has been associated with therapy response and prognosis with conflicting data. Recently, we demonstrated that immune cell density and intratumoral budding (ITB) are predictive factors in rectal cancer. We investigated the predictive value of TP53 immunohistochemistry with CD8+ T cell density and ITB on pretreatment biopsies of rectal adenocarcinoma for response to neoadjuvant therapy. METHODS AND RESULTS Pretreatment biopsies of rectal adenocarcinoma from 117 patients with neoadjuvant therapy were analysed for TP53 expression by immunohistochemistry, ITB, CD8+ T cell density and mismatch repair protein (MMR) status. Most rectal adenocarcinomas displayed aberrant TP53 expression (86 of 117, 74%). Compared to wild-type TP53, aberrant TP53 expression was associated with proficient MMR status (P = 0.003) and low CD8+ T cell density (P = 0.001). Aberrant TP53 was significantly associated with a partial to poor response to neoadjuvant therapy [odds ratio (OR) = 2.42, 95% confidence interval (CI) = 1.04-5.62, P = 0.04]. A combined histopathological risk score (HRS) was created using CD8+ T cell density, ITB and TP53 expression. Patients were separated into low (none to one factor) and high (two to three factors) HRS categories. In the multivariable model, patients with a high HRS were 3.25-fold more likely to have a partial or poor response to neoadjuvant therapy (95% CI = 1.48-7.11, P = 0.003). CONCLUSIONS Our study demonstrates that aberrant TP53 expression, high ITB and low CD8+ T cell density in pretreatment biopsies can help predict response to neoadjuvant therapy. These biomarkers may be helpful in identifying patients at risk for therapy resistance.
Collapse
Affiliation(s)
- Wei Chen
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lama Farchoukh
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lindsey Seigh
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Douglas J Hartman
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
12
|
The concept and use of the neoadjuvant rectal score as a composite endpoint in rectal cancer. Lancet Oncol 2021; 22:e314-e326. [PMID: 34048686 DOI: 10.1016/s1470-2045(21)00053-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 12/20/2022]
Abstract
There is no universally accepted instrument to use as a validated surrogate endpoint for overall survival in phase 2 and phase 3 multimodal rectal cancer trials using chemoradiotherapy. Efforts are hampered by the inaccuracy of clinical TNM staging, the variability of indications for neoadjuvant treatment, and diverse definitions of tumour regression grade. Pathological complete response is commonly used, but fails to capture information from the majority of patients. The neoadjuvant rectal score categorises response and downstaging from the entire trial population to identify whether or not a novel treatment group in a chemoradiation trial is superior by predicting overall survival outcomes. Additionally, the neoadjuvant rectal score assesses the difference between initial clinical and pathological T stage and the presence or absence of nodal involvement after treatment. The neoadjuvant rectal score has been conceptually, but incompletely, statistically validated by two independent trial datasets. However, a fundamental weakness of the score is that no preoperative phase 3 trials in locally advanced rectal cancer in the past 20 years have provided a significant benefit in overall survival to statistically validate the neoadjuvant rectal score as a surrogate endpoint for overall survival. We review the robustness, practical value, applicability, generalisability, advantages, and disadvantages of the neoadjuvant rectal score as a surrogate endpoint for overall survival and recommend how this score could be improved and be acceptable as a standard endpoint in studies investigating neoadjuvant chemotherapy and chemoradiation in patients with rectal cancer.
Collapse
|
13
|
Cheong C, Shin JS, Suh KW. Prognostic value of changes in serum carcinoembryonic antigen levels for preoperative chemoradiotherapy response in locally advanced rectal cancer. World J Gastroenterol 2020; 26:7022-7035. [PMID: 33311947 PMCID: PMC7701949 DOI: 10.3748/wjg.v26.i44.7022] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/31/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Preoperative chemoradiotherapy (CRT) is a standard treatment modality for locally advanced rectal cancer. However, CRT alone cannot improve overall survival. Approximately 20% of patients with CRT-resistant tumors show disease progression. Therefore, predictive factors for treatment response are needed to identify patients who will benefit from CRT. We theorized that the prognosis may vary if patients are classified according to pre- to post-CRT changes in carcinoembryonic antigen (CEA) levels.
AIM To identify patients with locally advanced rectal cancer for preoperative chemoradiotherapy based on carcinoembryonic antigen levels.
METHODS We retrospectively included locally advanced rectal cancer patients who underwent preoperative CRT and curative resection between 2011 and 2017. Patients were assigned to groups A, B, and C based on pre- and post-CRT serum CEA levels: Both > 5; pre > 5 and post ≤ 5; and both ≤ 5 ng/mL, respectively. We compared the response to CRT based on changes in serum CEA levels. Receiver operating characteristic curve analysis was performed to determine optimal cutoff for neutrophil–lymphocyte ratio and platelet–lymphocyte ratio. Multivariate logistic regression analysis was used to evaluate the prognostic factors for pathologic complete response (pCR)/good response.
RESULTS The cohort comprised 145 patients; of them, 27, 43, and 65 belonged to groups A, B, and C, respectively, according to changes in serum CEA levels before and after CRT. Pre- (P < 0.001) and post-CRT (P < 0.001) CEA levels and the ratio of down-staging (P = 0.013) were higher in Groups B and C than in Group A. The ratio of pathologic tumor regression grade 0/1 significantly differed among the groups (P = 0.003). Group C had the highest number of patients showing pCR (P < 0.001). Most patients with pCR showed pre- and post-CRT CEA levels < 5 ng/mL (P < 0.001, P = 0.008). Pre- and post-CRT CEA levels were important risk factors for pCR (OR = 18.71; 95%CI: 4.62–129.51, P < 0.001) and good response (OR = 5.07; 95%CI: 1.92–14.83, P = 0.002), respectively. Pre-CRT neutrophil–lymphocyte ratio and post-CRT T ≥ 3 stage were also prognostic factors for pCR or good response.
CONCLUSION Pre- and post-CRT CEA levels, as well as change in CEA levels, were prognostic markers for treatment response to CRT and may facilitate treatment individualization for rectal cancer.
Collapse
Affiliation(s)
- Chinock Cheong
- Department of Surgery, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, South Korea
| | - Jun Sang Shin
- Department of Surgery, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, South Korea
| | - Kwang Wook Suh
- Department of Surgery, Ajou University School of Medicine, Suwon 16499, Gyeonggi-do, South Korea
| |
Collapse
|