1
|
Guo Y, Hu C, Cai K, Long G, Cai D, Yu Z, Huang X, Cai Z, Hu P, Chen Y, Gao F, Wu X. KRAS inhibitors may prevent colorectal cancer metachronous metastasis by suppressing TGF‑β mediated epithelial‑mesenchymal transition. Mol Med Rep 2025; 31:24. [PMID: 39540351 DOI: 10.3892/mmr.2024.13389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
In colorectal cancer (CRC), KRAS mutations enhance metachronous metastasis, a condition without prognostic biomarkers or preventive measures. The present study demonstrated that KRAS mutation may be a risk factor for CRC metachronous metastasis through meta‑analysis of public databases. A risk scoring model was constructed using machine learning for predicting metachronous metastasis in KRAS‑mutant CRC. Wound healing and Transwell assay indicated that KRAS inhibitors strongly suppress migration and invasion capabilities of high‑risk CRC cells and these findings were validated through ex vivo organoid and a mouse model of splenic‑liver metastasis. Mechanistically, RNA sequencing, reverse transcription‑quantitative PCR and western blot analyses revealed that KRAS inhibitors suppressed epithelial‑mesenchymal transition (EMT) and transforming growth factor β (TGF‑β) signaling. Notably, addition of TGF‑β1 protein partially reversed the inhibitory effects of KRAS inhibitors on CRC. These results suggested that KRAS inhibitors may prevent CRC metachronous metastasis by downregulating TGF‑β‑mediated EMT, suggesting they can be used prophylactically in high‑risk KRAS‑mutant CRC.
Collapse
Affiliation(s)
- Yaoyu Guo
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Chuling Hu
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Kuntai Cai
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Guojie Long
- Department of General Surgery (Department of Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Du Cai
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Zhaoliang Yu
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Xinxin Huang
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Zerong Cai
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Peishan Hu
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Yufeng Chen
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Feng Gao
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Xiaojian Wu
- Guangdong Institute of Gastroenterology, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| |
Collapse
|
2
|
Schmauch B, Elsoukkary SS, Moro A, Raj R, Wehrle CJ, Sasaki K, Calderaro J, Sin-Chan P, Aucejo F, Roberts DE. Combining a deep learning model with clinical data better predicts hepatocellular carcinoma behavior following surgery. J Pathol Inform 2024; 15:100360. [PMID: 38292073 PMCID: PMC10825615 DOI: 10.1016/j.jpi.2023.100360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/10/2023] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is among the most common cancers worldwide, and tumor recurrence following liver resection or transplantation is one of the highest contributors to mortality in HCC patients after surgery. Using artificial intelligence (AI), we developed an interdisciplinary model to predict HCC recurrence and patient survival following surgery. We collected whole-slide H&E images, clinical variables, and follow-up data from 300 patients with HCC who underwent transplant and 169 patients who underwent resection at the Cleveland Clinic. A deep learning model was trained to predict recurrence-free survival (RFS) and disease-specific survival (DSS) from the H&E-stained slides. Repeated cross-validation splits were used to compute robust C-index estimates, and the results were compared to those obtained by fitting a Cox proportional hazard model using only clinical variables. While the deep learning model alone was predictive of recurrence and survival among patients in both cohorts, integrating the clinical and histologic models significantly increased the C-index in each cohort. In every subgroup analyzed, we found that a combined clinical and deep learning model better predicted post-surgical outcome in HCC patients compared to either approach independently.
Collapse
Affiliation(s)
| | - Sarah S. Elsoukkary
- Owkin Lab, Owkin, Inc., New York, NY, USA
- Department of Pathology, Cleveland Clinic, Cleveland, OH, USA
| | - Amika Moro
- Department of Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Roma Raj
- Department of Surgery, Cleveland Clinic, Cleveland, OH, USA
| | | | - Kazunari Sasaki
- Department of Surgery, Stanford University, Palo Alto, CA, USA
| | - Julien Calderaro
- Department of Pathology, Henri Mondor University Hospital, Créteil, France
| | | | | | | |
Collapse
|
3
|
Salmaso B, Scarpa M, Pellegrini V, Stepanyan A, Salmaso R, Kotsafti A, Scognamiglio F, Gregori D, Rivella G, De Simoni O, Becherucci G, Negro S, Vignotto C, Spolverato G, Ruffolo C, Angriman I, Bergamo F, Chiminazzo V, Maretto I, Zizzo M, Marchegiani F, Facci L, Brignola S, Businello G, Licia L, Guzzardo V, Dal Santo L, Carlotta C, Massani M, Pozza A, Cataldo I, Stecca T, Dei Tos AP, Zagonel V, Pilati P, Franzato B, Scapinello A, Pozza G, Godina M, Pirozzolo G, Recordare A, Mondi I, Lio CD, Merenda R, Bordignon G, Verdi D, Saadeh L, Guerriero S, Piccioli A, Noaro G, Cola R, Portale G, Cipollari C, Zuin M, Candioli S, Gavagna L, Ricagna F, Ortenzi M, Guerrieri M, Tagliente G, Tomassi M, Tedeschi U, Porzionato A, Agostini M, Bao RQ, Cavallin F, Tussardi G, Di Camillo B, Bardini R, Castagliuolo I, Pucciarelli S, Fassan M, Scarpa M. IMMUNOREACT 9 metachronous rectal cancers have high HLA-ABC expression on healthy epithelium but a lower infiltration of CD3+ T cells than primary lesions. Sci Rep 2024; 14:29821. [PMID: 39616209 PMCID: PMC11608249 DOI: 10.1038/s41598-024-80299-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
Lynch syndrome is rarely associated with rectal cancer (RC) and thus, metachronous RC has been scarcely investigated. This study aimed to analyze the mucosal immune microenvironment in sporadic and metachronous RC. We analyzed the mucosal immune microenvironment in the 25 metachronous RCs present in the IMMUNOREACT 1 and 2 multicentre observational studies (624 patients). A panel of immune markers was retrospectively investigated at immunohistochemistry: CD3, CD4, CD8, CD8b, Tbet, FoxP3, PD-L1, MSH6, and PMS2 and CD80. Single-cell suspensions were subjected to flow-cytometry to determine the proportion of epithelial cells (pan-cytokeratin) acting as antigen-presenting cells (expressing CD80, CD86, HLA-ABC) and the proportion of activated CD8 + T cells (CD8 + positive for CD28, CD38), inhibitory T cells (CD3 + CTLA-4+) of activated CD4 + T helper cells (CD4 + CD25+) and activated T regulatory cells (CD4 + CD25 + FoxP3+). No mismatch repair gene deficiencies were observed in the patients. The previous history of colorectal adenoma was significantly more frequent in metachronous RC. In healthy epithelial cells, HLA-ABC expression was significantly higher in patients with metachronous RC. In therapy-naïve metachronous RC patients, a significantly lower level of circulating lymphocytes and CD3 + T-cell infiltration in the healthy mucosa surrounding the RC was observed compared to patients with non-metachronous cancer. Our study supports the hypothesis that metachronous RC can occur in a cancerization field in patients with weak systemic and local immune systems. The peculiar site of RC makes the mismatch-repair genes deficiency in metachronous cancer onset less relevant.
Collapse
Affiliation(s)
| | | | | | - Astghik Stepanyan
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Roberta Salmaso
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | | | - Dario Gregori
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Giorgio Rivella
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | - Giulia Becherucci
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Silvia Negro
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Chiara Vignotto
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Gaya Spolverato
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Cesare Ruffolo
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Imerio Angriman
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | | | - Isacco Maretto
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Maurizio Zizzo
- Arcispedale Santa Maria Nuova IRCCS, Surgery, Reggio Emilia, Italy
| | | | - Luca Facci
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | | | | | - Vincenza Guzzardo
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Luca Dal Santo
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | | | - Anna Pozza
- Azienda ULSS 2 Marca Trevigiana, Treviso, Italy
| | | | | | | | | | | | | | | | - Giulia Pozza
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | | | | | | | | | | | | | | | - Luca Saadeh
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | | | | | | | | | | | | | | | | | | | - Monica Ortenzi
- Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Torrette, Italy
| | - Mario Guerrieri
- Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Torrette, Italy
| | | | | | | | - Andrea Porzionato
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Marco Agostini
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Riccardo Quoc Bao
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | - Gaia Tussardi
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | - Barbara Di Camillo
- Department of Information Engineering DEI, University of Padua, Padua, Italy
| | - Romeo Bardini
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy
| | | | | | - Matteo Fassan
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Azienda ULSS 2 Marca Trevigiana, Treviso, Italy
| | - Marco Scarpa
- Azienda Ospedale Università di Padova, via Giustiniani 2, Padua, 35128, Italy.
| |
Collapse
|
4
|
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
|
5
|
Adegoke NA, Gide TN, Mao Y, Quek C, Patrick E, Carlino MS, Lo SN, Menzies AM, Pires da Silva I, Vergara IA, Long G, Scolyer RA, Wilmott JS. Classification of the tumor immune microenvironment and associations with outcomes in patients with metastatic melanoma treated with immunotherapies. J Immunother Cancer 2023; 11:e007144. [PMID: 37865395 PMCID: PMC10603328 DOI: 10.1136/jitc-2023-007144] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 10/23/2023] Open
Abstract
BACKGROUND Tumor microenvironment (TME) characteristics are potential biomarkers of response to immune checkpoint inhibitors in metastatic melanoma. This study developed a method to perform unsupervised classification of TME of metastatic melanoma. METHODS We used multiplex immunohistochemical and quantitative pathology-derived assessment of immune cell compositions of intratumoral and peritumoral regions of metastatic melanoma baseline biopsies to classify TME in relation to response to anti-programmed cell death protein 1 (PD-1) monotherapy or in combination with anti-cytotoxic T-cell lymphocyte-4 (ipilimumab (IPI)+PD-1). RESULTS Spatial profiling of CD8+T cells, macrophages, and melanoma cells, as well as phenotypic PD-1 receptor ligand (PD-L1) and CD16 proportions, were used to identify and classify patients into one of three mutually exclusive TME classes: immune-scarce, immune-intermediate, and immune-rich tumors. Patients with immune-rich tumors were characterized by a lower proportion of melanoma cells and higher proportions of immune cells, including higher PD-L1 expression. These patients had higher response rates and longer progression-free survival (PFS) than those with immune-intermediate and immune-scarce tumors. At a median follow-up of 18 months (95% CI: 6.7 to 49 months), the 1-year PFS was 76% (95% CI: 64% to 90%) for patients with an immune-rich tumor, 56% (95% CI: 44% to 72%) for those with an immune-intermediate tumor, and 33% (95% CI: 23% to 47%) for patients with an immune-scarce tumor. A higher response rate was observed in patients with an immune-scarce or immune-intermediate tumor when treated with IPI+PD-1 compared with those treated with PD-1 alone. CONCLUSIONS Our study provides an automatic TME classification method that may predict the clinical efficacy of immunotherapy for patients with metastatic melanoma.
Collapse
Affiliation(s)
- Nurudeen A Adegoke
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Tuba N Gide
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yizhe Mao
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ellis Patrick
- Centre for Cancer Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Mathematics and Statistics, The University of Sydney, Sydney, New South Wales, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Westmead and Blacktown Hospitals, Sydney, New South Wales, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alexander Maxwell Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Ines Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Westmead and Blacktown Hospitals, Sydney, New South Wales, Australia
| | - Ismael A Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Georgina Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Predictive and Prognostic Value of Oncogene Mutations and Microsatellite Instability in Locally-Advanced Rectal Cancer Treated with Neoadjuvant Radiation-Based Therapy: A Systematic Review and Meta-Analysis. Cancers (Basel) 2023; 15:cancers15051469. [PMID: 36900260 PMCID: PMC10001009 DOI: 10.3390/cancers15051469] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Markers of pathological complete response (pCR) to preoperative radiation-based therapy in locally advanced rectal cancer (LARC) are strongly needed. This meta-analysis aimed at elucidating the predictive/prognostic role of tumor markers in LARC. We systematically reviewed the impact of RAS, TP53, BRAF, PIK3CA, and SMAD4 mutations and MSI status on response (pCR, downstaging) and prognosis (risk of recurrence, survival) in LARC according to PRISMA guidelines and the PICO model. PubMed, Cochrane Library, and Web of Science Core Collection were systematically searched to identify relevant studies published before October 2022. KRAS mutations were significantly associated with the risk of not achieving pCR after preoperative treatment (summary OR = 1.80, 95% CI: 1.23-2.64). This association was even more significant in patients not receiving cetuximab (summary OR = 2.17, 95% CI: 1.41-3.33) than in patients receiving cetuximab (summary OR = 0.89, 95% CI: 0.39-20.05). MSI status was not associated with pCR (summary OR = 0.80, 95% CI: 0.41-1.57). No effect of KRAS mutation or MSI status on downstaging was detected. Meta-analysis of survival outcomes was not possible due to the large heterogeneity among studies in endpoint assessment. The minimum number of eligible studies to assess the predictive/prognostic role of TP53, BRAF, PIK3CA, and SMAD4 mutations was not reached. KRAS mutation, but not MSI status, proved to be a detrimental marker for response to preoperative radiation-based therapy in LARC. Translating this finding into the clinic could improve the management of LARC patients. More data are needed to clarify the clinical impact of TP53, BRAF, PIK3CA, and SMAD4 mutations.
Collapse
|
7
|
Graham Martínez C, Barella Y, Kus Öztürk S, Ansems M, Gorris MA, van Vliet S, Marijnen CA, Nagtegaal ID. The immune microenvironment landscape shows treatment-specific differences in rectal cancer patients. Front Immunol 2022; 13:1011498. [PMID: 36238289 PMCID: PMC9552175 DOI: 10.3389/fimmu.2022.1011498] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
Neoadjuvant therapy is the cornerstone of modern rectal cancer treatment. Insights into the biology of tumor responses are essential for the successful implementation of organ-preserving strategies, as different treatments may lead to specific tumor responses. In this study, we aim to explore treatment-specific responses of the tumor microenvironment. Patients with locally advanced adenocarcinoma of the rectum who had received neo-adjuvant chemotherapy (CT), neo-adjuvant radiochemotherapy (RCT), neo-adjuvant radiotherapy with a long-interval (LRT) or short-interval (SRT) or no neoadjuvant therapy (NT) as control were included. Multiplex-immunofluorescence was performed to determine the presence of cytotoxic T-cells (T-cyt; CD3+CD8+), regulatory T-cells (T-reg; CD3+FOXP3+), T-helper cells (T-helper; CD3+CD8-FOXP3-), B cells (CD20+), dendritic cells (CD11c+) and tumor cells (panCK+). A total of 80 rectal cancer patients were included. Treatment groups were matched for gender, tumor location, response to therapy, and TNM stage. The pattern of response (shrinkage vs. fragmentation) was, however, different between treatment groups. Our analyses reveal that RCT-treated patients exhibited lower stromal T-helper, T-reg, and T-cyt cells compared to other treatment regimens. In conclusion, we demonstrated treatment-specific differences in the immune microenvironment landscape of rectal cancer patients. Understanding the underlying mechanisms of this landscape after a specific therapy will benefit future treatment decisions.
Collapse
Affiliation(s)
- Cristina Graham Martínez
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
- *Correspondence: Cristina Graham Martínez,
| | - Yari Barella
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Sonay Kus Öztürk
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Marleen Ansems
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Mark A.J Gorris
- Department of Tumor Immunology, Radboud University Medical Centre, Nijmegen, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Shannon van Vliet
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Corrie A.M Marijnen
- Department of Radiotherapy, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
8
|
Lee HH, Chen CH, Huang YH, Chiang CH, Huang MY. Biomarkers of Favorable vs. Unfavorable Responses in Locally Advanced Rectal Cancer Patients Receiving Neoadjuvant Concurrent Chemoradiotherapy. Cells 2022; 11:cells11101611. [PMID: 35626648 PMCID: PMC9139800 DOI: 10.3390/cells11101611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer is the second leading cause of cancer death globally. The gold standard for locally advanced rectal cancer (LARC) nowadays is preoperative concurrent chemoradiation (CCRT). Approximately three quarters of LARC patients do not achieve pathological complete response and hence suffer from relapse, metastases and inevitable death. The exploration of trustworthy and timely biomarkers for CCRT response is urgently called for. This review focused upon a broad spectrum of biomarkers, including circulating tumor cells, DNA, RNA, oncogenes, tumor suppressor genes, epigenetics, impaired DNA mismatch repair, patient-derived xenografts, in vitro tumor organoids, immunity and microbiomes. Utilizing proper biomarkers can assist in categorizing appropriate patients by the most efficient treatment modality with the best outcome and accompanied by minimal side effects. The purpose of this review is to inspect and analyze accessible data in order to fully realize the promise of precision oncology for rectal cancer patients.
Collapse
Affiliation(s)
- Hsin-Hua Lee
- Ph.D. Program in Environmental and Occupational Medicine, National Health Research Institutes, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Hung Chen
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Radiation Oncology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 801, Taiwan
| | - Yu-Hsiang Huang
- Post-Graduate Year Training, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Cheng-Han Chiang
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
| | - Ming-Yii Huang
- Ph.D. Program in Environmental and Occupational Medicine, National Health Research Institutes, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-3121101 (ext. 7158)
| |
Collapse
|