1
|
Spinelli A, Carrano FM, Laino ME, Andreozzi M, Koleth G, Hassan C, Repici A, Chand M, Savevski V, Pellino G. Artificial intelligence in colorectal surgery: an AI-powered systematic review. Tech Coloproctol 2023; 27:615-629. [PMID: 36805890 DOI: 10.1007/s10151-023-02772-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023]
Abstract
Artificial intelligence (AI) has the potential to revolutionize surgery in the coming years. Still, it is essential to clarify what the meaningful current applications are and what can be reasonably expected. This AI-powered review assessed the role of AI in colorectal surgery. A Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)-compliant systematic search of PubMed, Embase, Scopus, Cochrane Library databases, and gray literature was conducted on all available articles on AI in colorectal surgery (from January 1 1997 to March 1 2021), aiming to define the perioperative applications of AI. Potentially eligible studies were identified using novel software powered by natural language processing (NLP) and machine learning (ML) technologies dedicated to systematic reviews. Out of 1238 articles identified, 115 were included in the final analysis. Available articles addressed the role of AI in several areas of interest. In the preoperative phase, AI can be used to define tailored treatment algorithms, support clinical decision-making, assess the risk of complications, and predict surgical outcomes and survival. Intraoperatively, AI-enhanced surgery and integration of AI in robotic platforms have been suggested. After surgery, AI can be implemented in the Enhanced Recovery after Surgery (ERAS) pathway. Additional areas of applications included the assessment of patient-reported outcomes, automated pathology assessment, and research. Available data on these aspects are limited, and AI in colorectal surgery is still in its infancy. However, the rapid evolution of technologies makes it likely that it will increasingly be incorporated into everyday practice.
Collapse
Affiliation(s)
- A Spinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, MI, Italy.
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, MI, Italy.
| | - F M Carrano
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - M E Laino
- Artificial Intelligence Center, Humanitas Clinical and Research Center-IRCCS, Via A. Manzoni 56, 20089, Rozzano, MI, Italy
| | - M Andreozzi
- Department of Clinical Medicine and Surgery, University "Federico II" of Naples, Naples, Italy
| | - G Koleth
- Department of Gastroenterology and Hepatology, Hospital Selayang, Selangor, Malaysia
| | - C Hassan
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - A Repici
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - M Chand
- Wellcome EPSRC Centre for Interventional and Surgical Sciences (WEISS), University College London, London, UK
| | - V Savevski
- Artificial Intelligence Center, Humanitas Clinical and Research Center-IRCCS, Via A. Manzoni 56, 20089, Rozzano, MI, Italy
| | - G Pellino
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
- Colorectal Surgery, Vall d'Hebron University Hospital, Universitat Autonoma de Barcelona UAB, Barcelona, Spain
| |
Collapse
|
2
|
del Carmen S, Corchete LA, González Velasco C, Sanz J, Alcazar JA, García J, Rodríguez AI, Vidal Tocino R, Rodriguez A, Pérez-Romasanta LA, Sayagués JM, Abad M. High-Risk Clinicopathological and Genetic Features and Outcomes in Patients Receiving Neoadjuvant Radiochemotherapy for Locally Advanced Rectal Cancer. Cancers (Basel) 2021; 13:3166. [PMID: 34202891 PMCID: PMC8269103 DOI: 10.3390/cancers13133166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 01/16/2023] Open
Abstract
Administering preoperative radiochemotherapy (RCT) in stage II-III tumors to locally advanced rectal carcinoma patients has proved to be effective in a high percentage of cases. Despite this, 20-30% of patients show no response or even disease progression. At present, preoperative response is assessed by a combination of imaging and tumor regression on histopathology, but recent studies suggest that various genetic abnormalities may be associated with the sensitivity or resistance of rectal cancer tumor cells to neoadjuvant therapy. In the present study we investigated the relationship between genetic lesions detected by high-density single-nucleotide polymorphisms (SNP) arrays 6.0 and response to neoadjuvant RCT, evaluated according to Dworak criteria in 39 rectal cancer tumors before treatment. The highest frequency of copy-number (CN) losses detected corresponded to chromosomes 18q (n = 27; 69%), 1p (n = 22; 56%), 15q (n = 19; 49%), 8p (n = 18; 48%), 4q (n = 17; 46%), and 22q (n = 17; 46%); in turn, CN gains more frequently involved chromosomes 20p (n = 22; 56%), 8p (n = 20; 51%), and 15q (n = 16; 41%). There was a significant association between alterations in the 1p, 3q, 7q, 12p, 17q, 20p, and 22q chromosomal regions and the degree of response to therapy prior to surgery. However, 4q, 15q11.1, and 15q14 chromosomal region alterations were identified as important by five prediction algorithms, i.e., those with the greatest influence on predicting the tumor response to treatment with preoperative RCT. Multivariate analysis of prognostic factors showed that gains on 15q11.1 and carcinoembryonic antigen (CEA) levels serum at diagnosis were the only independent variables predicting disease-free survival (DFS). Lymph node involvement also showed a prognostic impact on overall survival (OS) in the multivariate analysis. A deep-learning-based algorithm showed a 100% success rate in predicting both DFS and OS at 60 months after diagnosis of the disease. In summary, our results indicate the existence of an association between tumor genetic abnormalities at diagnosis, response to neoadjuvant therapy, and survival of patients with locally advanced rectal cancer. In addition to the clinical and biological characteristics of locally advanced rectal cancer patients, these could be used in the future as therapeutic and prognostic biomarkers, to identify patients sensitive or resistant to preoperative treatment, helping guide therapeutic decision-making. Additional prospective studies in larger series of patients are required to confirm the clinical utility of the newly identified biomarkers.
Collapse
Affiliation(s)
- Sofía del Carmen
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (S.d.C.); (C.G.V.); (J.S.); (A.R.)
| | - Luís Antonio Corchete
- Cancer Research Center and Hematology Service and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain;
| | - Cristina González Velasco
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (S.d.C.); (C.G.V.); (J.S.); (A.R.)
| | - Julia Sanz
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (S.d.C.); (C.G.V.); (J.S.); (A.R.)
| | - José Antonio Alcazar
- General and Gastrointestinal Surgery Service and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (J.A.A.); (J.G.)
| | - Jacinto García
- General and Gastrointestinal Surgery Service and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (J.A.A.); (J.G.)
| | - Ana Isabel Rodríguez
- Radiation Oncology Service and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (A.I.R.); (L.A.P.-R.)
| | - Rosario Vidal Tocino
- Medical Oncology Service and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain;
| | - Alba Rodriguez
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (S.d.C.); (C.G.V.); (J.S.); (A.R.)
| | - Luis Alberto Pérez-Romasanta
- Radiation Oncology Service and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (A.I.R.); (L.A.P.-R.)
| | - José María Sayagués
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (S.d.C.); (C.G.V.); (J.S.); (A.R.)
| | - Mar Abad
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007 Salamanca, Spain; (S.d.C.); (C.G.V.); (J.S.); (A.R.)
| |
Collapse
|
3
|
Eslamizadeh S, Zare AA, Talebi A, Tabaeian SP, Eshkiki ZS, Heydari-Zarnagh H, Akbari A. Differential Expression of miR-20a and miR-145 in Colorectal Tumors as Potential Location-specific miRNAs. Microrna 2020; 10:66-73. [PMID: 33349227 DOI: 10.2174/2211536609666201221123604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/16/2020] [Accepted: 11/27/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs), as tissue specific regulators of gene transcription, may be served as biomarkers for Colorectal Cancer (CRC). OBJECTIVE This study aimed to investigate the potential role of the cancer-related hsa-miRNAs as biomarkers in Colon Cancer (CC) and Rectal Cancer (RC). METHODS A total of 148 CRC samples (74 rectum and 74 colon) and 74 adjacent normal tissues were collected to examine the differential expression of selected ten hsa-miRNAs using quantitative Reverse Transcriptase PCR (qRT-PCR). RESULTS The significantly elevated levels of miR-21, miR-133b, miR-18a, miR-20a, and miR-135b, and decreased levels of miR-34a, miR-200c, miR-145, and let-7g were detected in colorectal tumors compared to the healthy tissues (P<0.05). Hsa-miR-20a was significantly overexpressed in rectum compared to colon (p =0.028) from a cut-off value of 3.15 with a sensitivity of 66% and a specificity of 60% and an AUC value of 0.962. Also, hsa-miR-145 was significantly overexpressed in colon compared to the rectum (p =0.02) from a cut-off value of 3.9 with a sensitivity of 55% and a specificity of 61% and an AUC value of 0.91. CONCLUSION In conclusion, hsa-miR-20a and hsa-miR-145, as potential tissue-specific biomarkers for distinguishing RC and CC, improve realizing the molecular differences between these local tumors.
Collapse
Affiliation(s)
- Sara Eslamizadeh
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Zare
- Young Researchers and Elites club, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Atefeh Talebi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Zahra Shokati Eshkiki
- Alimentary Tract Research Center, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hafez Heydari-Zarnagh
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Kamran SC, Lennerz JK, Margolis CA, Liu D, Reardon B, Wankowicz SA, Van Seventer EE, Tracy A, Wo JY, Carter SL, Willers H, Corcoran RB, Hong TS, Van Allen EM. Integrative Molecular Characterization of Resistance to Neoadjuvant Chemoradiation in Rectal Cancer. Clin Cancer Res 2019; 25:5561-5571. [PMID: 31253631 PMCID: PMC6744983 DOI: 10.1158/1078-0432.ccr-19-0908] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/08/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Molecular properties associated with complete response or acquired resistance to concurrent chemotherapy and radiotherapy (CRT) are incompletely characterized.Experimental Design: We performed integrated whole-exome/transcriptome sequencing and immune infiltrate analysis on rectal adenocarcinoma tumors prior to neoadjuvant CRT (pre-CRT) and at time of resection (post-CRT) in 17 patients [8 complete/partial responders, 9 nonresponders (NR)]. RESULTS CRT was not associated with increased tumor mutational burden or neoantigen load and did not alter the distribution of established somatic tumor mutations in rectal cancer. Concurrent KRAS/TP53 mutations (KP) associated with NR tumors and were enriched for an epithelial-mesenchymal transition transcriptional program. Furthermore, NR was associated with reduced CD4/CD8 T-cell infiltrates and a post-CRT M2 macrophage phenotype. Absence of any local tumor recurrences, KP/NR status predicted worse progression-free survival, suggesting that local immune escape during or after CRT with specific genomic features contributes to distant progression. CONCLUSIONS Overall, while CRT did not impact genomic profiles, CRT impacted the tumor immune microenvironment, particularly in resistant cases.
Collapse
Affiliation(s)
- Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital, Boston, Massachusetts
| | - Claire A Margolis
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - David Liu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Brendan Reardon
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Stephanie A Wankowicz
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Emily E Van Seventer
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Adam Tracy
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Scott L Carter
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ryan B Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Eliezer M Van Allen
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
5
|
do Canto LM, Larsen SJ, Catin Kupper BE, Begnami MDFDS, Scapulatempo-Neto C, Petersen AH, Aagaard MM, Baumbach J, Aguiar S, Rogatto SR. Increased Levels of Genomic Instability and Mutations in Homologous Recombination Genes in Locally Advanced Rectal Carcinomas. Front Oncol 2019; 9:395. [PMID: 31192117 PMCID: PMC6527873 DOI: 10.3389/fonc.2019.00395] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/26/2019] [Indexed: 12/11/2022] Open
Abstract
Pre-operative 5-fluoracil-based chemoradiotherapy (nCRT) is the standard treatment for patients with locally advanced rectal cancer (LARC). Patients with pathological complete response (pCR–0% of tumor cells in the surgical specimen after nCRT) have better overall survival and lower risk of recurrence in comparison with incomplete responders (pIR). Predictive biomarkers to be used for new therapeutic strategies and capable of stratifying patients to avoid overtreatment are needed. We evaluated the genomic profiles of 33 pre-treatment LARC biopsies using SNP array and targeted-next generation sequencing (tNGS). Based on the large number of identified genomic alterations, we calculated the genomic instability index (GII) and three homologous recombination deficiency (HRD) scores, which have been reported as impaired DNA repair markers. We observed high GII in our LARC cases, which was confirmed in 165 rectal cancer cases from TCGA. Patients with pCR presented higher GII compared with pIR. Moreover, a negative correlation between GII and the fraction of tumor cells remaining after surgery was observed (ρ = –0.382, P = 0.02). High HRD scores were detected in 61% of LARC, of which 70% were incomplete responders. Using tNGS (105 cancer-related genes, 13 involved in HR and 5 in mismatch repair pathways), we identified 23% of cases with mutations in HR genes, mostly in pIR cases (86% of mutated cases). In agreement, the analysis of the TCGA dataset (N = 145) revealed 21% of tumors with mutations in HR genes. The HRD scores were shown to be predictive of better response to PARP-inhibitors and platinum-based chemotherapy in breast and ovarian cancer. Our results suggest that the same strategy could be applied in a set of LARC patients with HRD. In conclusion, we identified high genomic instability in LARC, which was related to alterations in the HR pathway, especially in pIR. These findings suggest that patients with impaired HRD would clinically benefit from PARP-inhibitors and platinum-based therapy.
Collapse
Affiliation(s)
- Luisa Matos do Canto
- International Research Center-Center for International Private Enterprise (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil.,Department of Clinical Genetics, Vejle Hospital, Vejle, Denmark
| | - Simon J Larsen
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | | | | | | | | | - Mads M Aagaard
- Department of Clinical Genetics, Vejle Hospital, Vejle, Denmark
| | - Jan Baumbach
- TUM School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
| | - Samuel Aguiar
- Department of Pelvic Surgery, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Silvia R Rogatto
- Department of Clinical Genetics, Vejle Hospital, Vejle, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.,Danish Colorectal Cancer Center South, Vejle, Denmark
| |
Collapse
|
6
|
Wan JF, Li XQ, Zhang J, Yang LF, Zhu J, Li GC, Liang LP, Shen LJ, Zhang H, Li J, Zhang YT, Chen CY, Zhang Z. Aneuploidy of chromosome 8 and mutation of circulating tumor cells predict pathologic complete response in the treatment of locally advanced rectal cancer. Oncol Lett 2018; 16:1863-1868. [PMID: 30008877 DOI: 10.3892/ol.2018.8831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Identifying patients who may or may not achieve pathologic complete response (pathCR) allows for treatment with alternative approaches in the preoperative setting. The aim of the current study was to investigate whether aneuploidy of chromosome 8 and mutations of circulating tumor cells (CTCs) could predict the response of patients with rectal cancer to preoperative chemoradiotherapy. A total of 33 patients with locally advanced rectal cancer (cT3-T4 and/or cN+) treated with neoadjuvant chemoradiotherapy between September 2014 and March 2015 were recruited. Blood samples were collected from 33 patients with pre-chemoradiotherapy rectal cancer. It was demonstrated that ≥5 copies of chromosome 8 was associated with pathCR (univariate logistic regression, P=0.042). Of the 6 patients whose CTCs had <5 copies of chromosome 8, 3 achieved pathCR (3/6, 50%), and of the 27 patients whose CTCs had ≥5 copies of chromosome 8 obtained 3 pathCR (3/27, 11.1%; Chi-square test, P=0.0255). Of the 33 patients with mutations assessed, 8 significant nonsynonymous mutations in CTCs were identified as associated with pathCR (Chi-square test, P-values range, 0.0004-0.0298; mutations in ARID1A, HDAC1, APC, ERBB3, TP53, AMER1 and AR). These results suggest that ≥5 copies of chromosome 8 and 8 nonsynonymous mutations in ARID1A, HDAC1, APC, ERBB3, TP53, AMER1 AR in CTCs were associated with pathCR. This conclusion should be validated further in larger prospective studies and the long-term follow-up survival data of this study will also be reported in the future.
Collapse
Affiliation(s)
- Jue-Feng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Xue-Qin Li
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Jing Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Li-Feng Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Ji Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Gui-Chao Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Li-Ping Liang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Li-Jun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Jing Li
- Department of Medical Research, Shanghai MajorMed Diagnostics Company, Shanghai 201321, P.R. China
| | - Yi-Tong Zhang
- Department of Medical Research, Shanghai MajorMed Diagnostics Company, Shanghai 201321, P.R. China
| | - Chang-Yue Chen
- Department of Medical Research, Shanghai MajorMed Diagnostics Company, Shanghai 201321, P.R. China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
7
|
Cosgrove CM, Tritchler DL, Cohn DE, Mutch DG, Rush CM, Lankes HA, Creasman WT, Miller DS, Ramirez NC, Geller MA, Powell MA, Backes FJ, Landrum LM, Timmers C, Suarez AA, Zaino RJ, Pearl ML, DiSilvestro PA, Lele SB, Goodfellow PJ. An NRG Oncology/GOG study of molecular classification for risk prediction in endometrioid endometrial cancer. Gynecol Oncol 2018; 148:174-180. [PMID: 29132872 PMCID: PMC5756518 DOI: 10.1016/j.ygyno.2017.10.037] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/24/2017] [Accepted: 10/27/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The purpose of this study was to assess the prognostic significance of a simplified, clinically accessible classification system for endometrioid endometrial cancers combining Lynch syndrome screening and molecular risk stratification. METHODS Tumors from NRG/GOG GOG210 were evaluated for mismatch repair defects (MSI, MMR IHC, and MLH1 methylation), POLE mutations, and loss of heterozygosity. TP53 was evaluated in a subset of cases. Tumors were assigned to four molecular classes. Relationships between molecular classes and clinicopathologic variables were assessed using contingency tests and Cox proportional methods. RESULTS Molecular classification was successful for 982 tumors. Based on the NCI consensus MSI panel assessing MSI and loss of heterozygosity combined with POLE testing, 49% of tumors were classified copy number stable (CNS), 39% MMR deficient, 8% copy number altered (CNA) and 4% POLE mutant. Cancer-specific mortality occurred in 5% of patients with CNS tumors; 2.6% with POLE tumors; 7.6% with MMR deficient tumors and 19% with CNA tumors. The CNA group had worse progression-free (HR 2.31, 95%CI 1.53-3.49) and cancer-specific survival (HR 3.95; 95%CI 2.10-7.44). The POLE group had improved outcomes, but the differences were not statistically significant. CNA class remained significant for cancer-specific survival (HR 2.11; 95%CI 1.04-4.26) in multivariable analysis. The CNA molecular class was associated with TP53 mutation and expression status. CONCLUSIONS A simple molecular classification for endometrioid endometrial cancers that can be easily combined with Lynch syndrome screening provides important prognostic information. These findings support prospective clinical validation and further studies on the predictive value of a simplified molecular classification system.
Collapse
Affiliation(s)
| | - David L Tritchler
- NRG Oncology Statistics and Data Management Center, Buffalo, NY, United States
| | - David E Cohn
- The Ohio State University, Columbus, OH, United States
| | - David G Mutch
- Washington University School of Medicine, St. Louis, MO, United States
| | - Craig M Rush
- The Ohio State University, Columbus, OH, United States
| | - Heather A Lankes
- Gynecologic Oncology Group Tissue Bank, Biopathology Center, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - William T Creasman
- Department of Obstetrics & Gynecology, Medical University of South Carolina, Charleston, SC, United States
| | - David S Miller
- Department of Obstetrics & Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Nilsa C Ramirez
- Gynecologic Oncology Group Tissue Bank, Biopathology Center, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | | | - Matthew A Powell
- Washington University School of Medicine, St. Louis, MO, United States
| | | | - Lisa M Landrum
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | | | | | - Richard J Zaino
- Anatomic Pathology, Penn State Milton S. Hersey Medical Center, Hershey, PA, United States
| | - Michael L Pearl
- Stony Brook University Hospital, Stony Brook, NY, United States
| | - Paul A DiSilvestro
- Women and Infants Hospital of Rhode Island, Providence, RI, United States
| | | | | |
Collapse
|
8
|
Karagkounis G, Kalady MF. Molecular Biology: Are We Getting Any Closer to Providing Clinically Useful Information? Clin Colon Rectal Surg 2017; 30:415-422. [PMID: 29184477 DOI: 10.1055/s-0037-1606373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Advances in molecular biology and biomarker research have significantly impacted our understanding and treatment of multiple solid malignancies. In rectal cancer, where neoadjuvant chemoradiation is widely used for locally advanced disease, most efforts have focused on the identification of predictors of response in an attempt to appropriately select patients for multimodality therapy. A variety of biomarkers have been studied, including genetic mutations, chromosomal copy number alterations, and single as well as multigene expression patterns. Also, as transanal resection of rectal tumors requires accurate preoperative detection of lymph node metastasis, the identification of biomarkers of regional nodal involvement has been another important field of active research. While preliminary results have been promising, lack of external validation means has a limited translation to clinical use. This review summarizes recent developments in rectal cancer biomarker research, highlighting the challenges associated with their adoption, and evaluating their potential for clinical use.
Collapse
Affiliation(s)
- Georgios Karagkounis
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Matthew F Kalady
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
9
|
Arriba M, García JL, Inglada-Pérez L, Rueda D, Osorio I, Rodríguez Y, Álvaro E, Sánchez R, Fernández T, Pérez J, Hernández JM, Benítez J, González-Sarmiento R, Urioste M, Perea J. DNA copy number profiling reveals different patterns of chromosomal instability within colorectal cancer according to the age of onset. Mol Carcinog 2016; 55:705-716. [PMID: 25808986 DOI: 10.1002/mc.22315] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/19/2015] [Accepted: 02/21/2015] [Indexed: 12/13/2022]
Abstract
Chromosomal instability resulting in copy number alterations is a hallmark of colorectal cancer (CRC). However, few studies have attempted to characterize the chromosomal changes occurring in early-onset CRC in order to compare them with those taking place within the more extensively studied late-onset CRC subset. Our aim was to characterize the genomic profiles of these two groups of colorectal tumors and to compare them to each other. Array comparative genomic hybridization profiling of 146 colorectal tumors (60 early-onset and 86 late-onset) in combination with an unsupervised analysis was used to define common and specific copy number alterations. We found a number of important differences between the chromosomal instability profiles of each age subset. Thus, losses at 1p36, 1p12, 1q21, 9p13, 14q11, 16p13, and 16p12 were significantly more frequent in younger patients, whereas gains at 7q11 and 7q22 were more frequent in older patients. Moreover, the unsupervised analysis stratified the tumors into two clusters, each one of which was enriched in patients from one of the age subsets. Our findings confirm the existence of substantial differences between the chromosomal instability profiles of the two groups which are more important from a qualitative point of view. Further studies are needed to understand the clinicopathological implications of these dissimilarities.
Collapse
Affiliation(s)
- María Arriba
- Centre for Biomedical Research of 12 de Octubre University Hospital, Madrid, Spain
| | - Juan L García
- Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca-USAL-CSIC, Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Salamanca, Spain
| | - Lucía Inglada-Pérez
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
| | - Daniel Rueda
- Molecular Biology Laboratory, 12 de Octubre University Hospital, Madrid, Spain
| | - Irene Osorio
- Department of Surgery, 12 de Octubre University Hospital, Madrid, Spain
| | - Yolanda Rodríguez
- Department of Pathology, 12 de Octubre University Hospital, Madrid, Spain
| | - Edurne Álvaro
- Department of Surgery, Infanta Leonor University Hospital, Madrid, Spain
| | - Ricard Sánchez
- Centre for Biomedical Research of 12 de Octubre University Hospital, Madrid, Spain
| | - Tamara Fernández
- Department of Surgery, 12 de Octubre University Hospital, Madrid, Spain
| | - Jessica Pérez
- Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca-USAL-CSIC, Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Salamanca, Spain
| | - Jesús M Hernández
- Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca-USAL-CSIC, Salamanca, Spain
| | - Javier Benítez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
- Human Genetics Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rogelio González-Sarmiento
- Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca-USAL-CSIC, Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Salamanca, Spain
| | - Miguel Urioste
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
- Familial Cancer Clinical Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - José Perea
- Centre for Biomedical Research of 12 de Octubre University Hospital, Madrid, Spain
- Department of Surgery, 12 de Octubre University Hospital, Madrid, Spain
| |
Collapse
|
10
|
Ryan JE, Warrier SK, Lynch AC, Ramsay RG, Phillips WA, Heriot AG. Predicting pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer: a systematic review. Colorectal Dis 2016; 18:234-46. [PMID: 26531759 DOI: 10.1111/codi.13207] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 09/17/2015] [Indexed: 02/08/2023]
Abstract
AIM Approximately 20% of patients treated with neoadjuvant chemoradiotherapy (nCRT) for locally advanced rectal cancer achieve a pathological complete response (pCR) while the remainder derive the benefit of improved local control and downstaging and a small proportion show a minimal response. The ability to predict which patients will benefit would allow for improved patient stratification directing therapy to those who are likely to achieve a good response, thereby avoiding ineffective treatment in those unlikely to benefit. METHOD A systematic review of the English language literature was conducted to identify pathological factors, imaging modalities and molecular factors that predict pCR following chemoradiotherapy. PubMed, MEDLINE and Cochrane Database searches were conducted with the following keywords and MeSH search terms: 'rectal neoplasm', 'response', 'neoadjuvant', 'preoperative chemoradiation', 'tumor response'. After review of title and abstracts, 85 articles addressing the prediction of pCR were selected. RESULTS Clear methods to predict pCR before chemoradiotherapy have not been defined. Clinical and radiological features of the primary cancer have limited ability to predict response. Molecular profiling holds the greatest potential to predict pCR but adoption of this technology will require greater concordance between cohorts for the biomarkers currently under investigation. CONCLUSION At present no robust markers of the prediction of pCR have been identified and the topic remains an area for future research. This review critically evaluates existing literature providing an overview of the methods currently available to predict pCR to nCRT for locally advanced rectal cancer. The review also provides a comprehensive comparison of the accuracy of each modality.
Collapse
Affiliation(s)
- J E Ryan
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Epworth Healthcare, Melbourne, Victoria, Australia.,Austin Academic Centre, University of Melbourne, Parkville, Victoria, Australia
| | - S K Warrier
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - A C Lynch
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - R G Ramsay
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - W A Phillips
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Cancer Biology and Surgical Oncology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - A G Heriot
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Molinari C, Matteucci F, Caroli P, Passardi A. Biomarkers and Molecular Imaging as Predictors of Response to Neoadjuvant Chemoradiotherapy in Patients With Locally Advanced Rectal Cancer. Clin Colorectal Cancer 2015; 14:227-38. [PMID: 26170142 DOI: 10.1016/j.clcc.2015.05.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022]
Abstract
Standard treatment of patients with locally advanced rectal cancer (LARC) includes neoadjuvant chemoradiotherapy (NCRT) followed by surgery. Tumor regression after NCRT varies substantially among individuals and pathological complete response is a known prognostic factor for LARC. The identification of a predictive model for response to chemoradiotherapy would help clinicians to identify patients who would probably benefit from multimodal treatment and to perform an early assessment of individual prognosis. Carcinoembryonic antigen has proven to be a good predictor of response in several clinical trials. Other widely studied predictive models in LARC include molecular biomarkers, analyzed at various levels and by different techniques, and molecular imaging, in particular magnetic resonance imaging and positron emission tomography/computed tomography. Although none of the studied markers have been approved in clinical practice, their evaluation in larger, prospective trials and in combined predictive models could be of use to define tailored therapeutic strategies.
Collapse
Affiliation(s)
- Chiara Molinari
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Federica Matteucci
- Diagnostic Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Caroli
- Diagnostic Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| |
Collapse
|
12
|
Copy number alterations and allelic ratio in relation to recurrence of rectal cancer. BMC Genomics 2015; 16:438. [PMID: 26048403 PMCID: PMC4458034 DOI: 10.1186/s12864-015-1550-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/17/2015] [Indexed: 12/16/2022] Open
Abstract
Background In rectal cancer, total mesorectal excision surgery combined with preoperative (chemo)radiotherapy reduces local recurrence rates but does not improve overall patient survival, a result that may be due to the harmful side effects and/or co-morbidity of preoperative treatment. New biomarkers are needed to facilitate identification of rectal cancer patients at high risk for local recurrent disease. This would allow for preoperative (chemo)radiotherapy to be restricted to high-risk patients, thereby reducing overtreatment and allowing personalized treatment protocols. We analyzed genome-wide DNA copy number (CN) and allelic alterations in 112 tumors from preoperatively untreated rectal cancer patients. Sixty-six patients with local and/or distant recurrent disease were compared to matched controls without recurrence. Results were validated in a second cohort of tumors from 95 matched rectal cancer patients. Additionally, we performed a meta-analysis that included 42 studies reporting on CN alterations in colorectal cancer and compared results to our own data. Results The genomic profiles in our study were comparable to other rectal cancer studies. Results of the meta-analysis supported the hypothesis that colon cancer and rectal cancer may be distinct disease entities. In our discovery patient study cohort, allelic retention of chromosome 7 was significantly associated with local recurrent disease. Data from the validation cohort were supportive, albeit not statistically significant, of this finding. Conclusions We showed that retention of heterozygosity on chromosome 7 may be associated with local recurrence in rectal cancer. Further research is warranted to elucidate the mechanisms and effect of retention of chromosome 7 on the development of local recurrent disease in rectal cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1550-0) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Predictive and prognostic biomarkers for neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Crit Rev Oncol Hematol 2015; 96:67-80. [PMID: 26032919 DOI: 10.1016/j.critrevonc.2015.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/03/2015] [Accepted: 05/05/2015] [Indexed: 02/08/2023] Open
Abstract
Locally advanced rectal cancer is regularly treated with trimodality therapy consisting of neoadjuvant chemoradiation, surgery and adjuvant chemotherapy. There is a need for biomarkers to assess treatment response, and aid in stratification of patient risk to adapt and personalise components of the therapy. Currently, pathological stage and tumour regression grade are used to assess response. Experimental markers include proteins involved in cell proliferation, apoptosis, angiogenesis, the epithelial to mesenchymal transition and microsatellite instability. As yet, no single marker is sufficiently robust to have clinical utility. Microarrays that screen a tumour for multiple promising candidate markers, gene expression and microRNA profiling will likely have higher yield and it is expected that a combination or panel of markers would prove most useful. Moving forward, utilising serial samples of circulating tumour cells or circulating nucleic acids can potentially allow us to demonstrate tumour heterogeneity, document mutational changes and subsequently measure treatment response.
Collapse
|
14
|
Poulsen LØ, Qvortrup C, Pfeiffer P, Yilmaz M, Falkmer U, Sorbye H. Review on adjuvant chemotherapy for rectal cancer - why do treatment guidelines differ so much? Acta Oncol 2015; 54:437-46. [PMID: 25597332 DOI: 10.3109/0284186x.2014.993768] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The use of postoperative adjuvant chemotherapy is controversial for rectal adenocarcinoma. Both international and national guidelines display a great span varying from recommending no adjuvant chemotherapy at all, over single drug 5-fluororuacil (5-FU), to combinations of 5-FU/oxaliplatin. METHODS A review of the literature was made identifying 24 randomized controlled trials on adjuvant treatment of rectal cancer based on about 10 000 patients. The trials were subdivided into a number of clinically relevant subgroups. RESULTS As regards patients treated with preoperative (chemo) radiotherapy, four randomized studies were found where use of adjuvant chemotherapy showed no benefit in survival. Three trials were found in which a subset of patients received preoperative (chemo) radiotherapy. Two of these trials showed a statistically significant benefit of adjuvant chemotherapy. Twenty trials were identified in which the patients did not receive preoperative (chemo) radiotherapy, including five Asian studies in which a statistically significant benefit from adjuvant chemotherapy was reported. CONCLUSIONS Most of the data found did not support the use of postoperative adjuvant chemotherapy for patients already treated with preoperative (chemo) radiotherapy. For patients not treated preoperatively, several studies support the use of single agent 5-FU chemotherapy. Treatment guidelines seem to differ according to if preoperative chemoradiation is considered of importance for use of adjuvant chemotherapy and if adjuvant colon cancer studies are considered transferrable to rectal cancer patients regardless of the molecular differences.
Collapse
Affiliation(s)
- Laurids Ø Poulsen
- Department of Oncology, Aalborg University Hospital , Aalborg , Denmark
| | | | | | | | | | | |
Collapse
|
15
|
González-González M, Garcia J, Alcazar JA, Gutiérrez ML, Gónzalez LM, Bengoechea O, Abad MM, Santos-Briz A, Blanco O, Martín M, Rodríguez A, Fuentes M, Muñoz-Bellvis L, Orfao A, Sayagues JM. Association between the cytogenetic profile of tumor cells and response to preoperative radiochemotherapy in locally advanced rectal cancer. Medicine (Baltimore) 2014; 93:e153. [PMID: 25474426 PMCID: PMC4616389 DOI: 10.1097/md.0000000000000153] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neoadjuvant radiochemotherapy to locally advanced rectal carcinoma patients has proven efficient in a high percentage of cases. Despite this, some patients show nonresponse or even disease progression. Recent studies suggest that different genetic alterations may be associated with sensitivity versus resistance of rectal cancer tumor cells to neoadjuvant therapy. We investigated the relationship between intratumoral pathways of clonal evolution as assessed by interphase fluorescence in situ hybridization (51 different probes) and response to neoadjuvant radiochemotherapy, evaluated by Dworak criteria in 45 rectal cancer tumors before (n = 45) and after (n = 31) treatment. Losses of chromosomes 1p (44%), 8p (53%), 17p (47%), and 18q (38%) and gains of 1q (49%) and 13q (75%) as well as amplification of 8q (38%) and 20q (47%) chromosomal regions were those specific alterations found at higher frequencies. Significant association (P < 0.05) was found between alteration of 1p, 1q, 11p, 12p, and 17p chromosomal regions and degree of response to neoadjuvant therapy. A clear association was observed between cytogenetic profile of the ancestral tumor cell clone and response to radiochemotherapy; cases presenting with del(17p) showed a poor response to neoadjuvant treatment (P = 0.03), whereas presence of del(1p) was more frequently observed in responder patients (P = 0.0002). Moreover, a significantly higher number of copies of chromosomes 8q (P = 0.004), 13q (P = 0.003), and 20q (P = 0.002) were found after therapy versus paired pretreatment rectal cancer samples. Our results point out the existence of an association between tumor cytogenetics and response to neoadjuvant therapy in locally advanced rectal cancer. Further studies in larger series of patients are necessary to confirm our results.
Collapse
Affiliation(s)
- María González-González
- From the Servicio General de Citometría, Departamento de Medicina and Centro de Investigación del Cáncer (IBMCC-CSIC/USAL), Hospital Universitario de Salamanca-IBSAL, Universidad de Salamanca (MG-G, MLG, MF, AO, MS); Servicio de Cirugía General y Aparato digestivo (JC, JAA, LMG, LM-B); Servicio de Patología, (OB, MMA, AS-B, OB); Servicio de Radio-diagnóstico, Hospital Universitario de Salamanca-IBSAL (MM); and Servicio de Oncología Radioterápica, Hospital Universitario de Salamanca (AR), Salamanca, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Can We Predict Response and/or Resistance to Neoadjuvant Chemoradiotherapy in Patients with Rectal Cancer? CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0210-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
17
|
Milgrom SA, Garcia-Aguilar J. Molecular biomarkers as predictors of response to neoadjuvant chemoradiation therapy in rectal cancer. SEMINARS IN COLON AND RECTAL SURGERY 2013. [DOI: 10.1053/j.scrs.2013.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
18
|
Torino F, Sarmiento R, Gasparini G. The contribution of targeted therapy to the neoadjuvant chemoradiation of rectal cancer. Crit Rev Oncol Hematol 2013; 87:283-305. [DOI: 10.1016/j.critrevonc.2013.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 12/24/2012] [Accepted: 02/13/2013] [Indexed: 12/26/2022] Open
|
19
|
Gantt GA, Kalady MF. Molecular markers for targeted neoadjuvant rectal cancer therapy. COLORECTAL CANCER 2013. [DOI: 10.2217/crc.13.35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
SUMMARY Neoadjuvant chemoradiotherapy is the standard of care for locally advanced rectal cancer. While neoadjuvant chemoradiation has been demonstrated to improve oncological outcomes, there is a wide spectrum of responses to therapy. The ability to predict who will respond favorably or unfavorably to neoadjuvant therapy could prevent unnecessary morbidity and potentially lead to novel therapeutic targets. A number of individual biomarkers and multigene signatures have been investigated as potential means of predicting response to neoadjuvant chemoradiation. While promising, none of these predictive biomarkers have yet been introduced clinically. This review summarizes both individual and multigene biomarkers for rectal cancer response to neoadjuvant chemoradiation.
Collapse
Affiliation(s)
- Gerald A Gantt
- Department of Colorectal Surgery, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Matthew F Kalady
- Department of Stem Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
20
|
Salawu A, Ul-Hassan A, Hammond D, Fernando M, Reed M, Sisley K. High quality genomic copy number data from archival formalin-fixed paraffin-embedded leiomyosarcoma: optimisation of universal linkage system labelling. PLoS One 2012; 7:e50415. [PMID: 23209738 PMCID: PMC3510175 DOI: 10.1371/journal.pone.0050415] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/19/2012] [Indexed: 01/01/2023] Open
Abstract
Most soft tissue sarcomas are characterized by genetic instability and frequent genomic copy number aberrations that are not subtype-specific. Oligonucleotide microarray-based Comparative Genomic Hybridisation (array CGH) is an important technique used to map genome-wide copy number aberrations, but the traditional requirement for high-quality DNA typically obtained from fresh tissue has limited its use in sarcomas. Although large archives of Formalin-fixed Paraffin-embedded (FFPE) tumour samples are available for research, the degradative effects of formalin on DNA from these tissues has made labelling and analysis by array CGH technically challenging. The Universal Linkage System (ULS) may be used for a one-step chemical labelling of such degraded DNA. We have optimised the ULS labelling protocol to perform aCGH on archived FFPE leiomyosarcoma tissues using the 180k Agilent platform. Preservation age of samples ranged from a few months to seventeen years and the DNA showed a wide range of degradation (when visualised on agarose gels). Consistently high DNA labelling efficiency and low microarray probe-to-probe variation (as measured by the derivative log ratio spread) was seen. Comparison of paired fresh and FFPE samples from identical tumours showed good correlation of CNAs detected. Furthermore, the ability to macro-dissect FFPE samples permitted the detection of CNAs that were masked in fresh tissue. Aberrations were visually confirmed using Fluorescence in situ Hybridisation. These results suggest that archival FFPE tissue, with its relative abundance and attendant clinical data may be used for effective mapping for genomic copy number aberrations in such rare tumours as leiomyosarcoma and potentially unravel clues to tumour origins, progression and ultimately, targeted treatment.
Collapse
Affiliation(s)
- Abdulazeez Salawu
- Department of Oncology, The University of Sheffield, Medical School, Sheffield, United Kingdom.
| | | | | | | | | | | |
Collapse
|
21
|
Izumi K, Conlin LK, Berrodin D, Fincher C, Wilkens A, Haldeman-Englert C, Saitta SC, Zackai EH, Spinner NB, Krantz ID. Duplication 12p and Pallister-Killian syndrome: A case report and review of the literature toward defining a Pallister-Killian syndrome minimal critical region. Am J Med Genet A 2012; 158A:3033-45. [DOI: 10.1002/ajmg.a.35500] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 04/21/2012] [Indexed: 11/10/2022]
|
22
|
Chen Z, Liu Z, Deng X, Warden C, Li W, Garcia-Aguilar J. Chromosomal copy number alterations are associated with persistent lymph node metastasis after chemoradiation in locally advanced rectal cancer. Dis Colon Rectum 2012; 55:677-85. [PMID: 22595848 PMCID: PMC3356567 DOI: 10.1097/dcr.0b013e31824f873f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lymph node metastasis is an important indicator of oncologic outcome for patients with rectal cancer. Identifying predictive biomarkers of lymph node metastasis could therefore be clinically useful. OBJECTIVE The aim of this study was to assess whether chromosomal copy number alterations can assist in predicting persistent lymph node metastasis in patients with locally advanced rectal cancer treated with preoperative chemoradiation therapy. DESIGN This is a nonrandomized, prospective phase II study. SETTING This study took place in a multi-institutional setting. PATIENTS Ninety-five patients with stage II (cT3-4, cN0) or stage III (any cT, cN1-2) rectal cancer were included. INTERVENTION Patients were treated with preoperative chemoradiation therapy followed by total mesorectal excision. Pretreatment biopsy tumor DNA and surgical margin control DNA were extracted and analyzed by oligonucleotide array-based comparative genomic hybridization. Chromosomal copy number alterations were correlated with persistent lymph node metastasis. Finally, a model for predicting persistent lymph node metastasis was built. MAIN OUTCOME MEASURES The primary outcomes assessed were whether chromosomal copy number alterations are associated with persistent lymph node metastasis in patients with rectal cancer and the accuracy of oligonucleotide array-based comparative genomic hybridization for predicting lymph node metastasis. RESULTS Twenty-five of 95 (26%) patients had lymph node metastasis after chemoradiation. Losses of 28 chromosomal regions, most notably in chromosome 4, were significantly associated with lymph node metastasis. Our predictive model contained 65 probes and predicted persistent lymph node metastasis with 68% sensitivity, 93% specificity, and positive and negative predictive values of 77% and 89%. The use of this model accurately predicted lymph node status (positive or negative) after chemoradiation therapy in 82 of 95 patients (86%). LIMITATIONS The patient cohort was not completely homogeneous, which may have influenced their clinical outcome. In addition, although we performed rigorous, statistically sound internal validation, external validation will be important to further corroborate our findings. CONCLUSIONS Copy number alterations can help identify patients with rectal cancer who are at risk of lymph node metastasis after chemoradiation.
Collapse
Affiliation(s)
- Zhenbin Chen
- Department of Surgery, City of Hope, Duarte, CA 91010, USA
| | - Zheng Liu
- Bioinformatics Core, Department of Molecular Medicine, City of Hope, Duarte, CA 91010, USA
| | - Xutao Deng
- Bioinformatics Core, Department of Molecular Medicine, City of Hope, Duarte, CA 91010, USA
| | - Charles Warden
- Bioinformatics Core, Department of Molecular Medicine, City of Hope, Duarte, CA 91010, USA
| | - Wenyan Li
- Department of Surgery, City of Hope, Duarte, CA 91010, USA
| | - Julio Garcia-Aguilar
- Department of Surgery, City of Hope, Duarte, CA 91010, USA,Corresponding Author: Julio Garcia-Aguilar, MD, PhD, Chair, Department of Surgery, City of Hope, 1500 E. Duarte Road, CA 91010. Tel: (626) 471-9309. Fax: (626) 301-8113.,
| |
Collapse
|
23
|
Perotti D, Spreafico F, Torri F, Gamba B, D'Adamo P, Pizzamiglio S, Terenziani M, Catania S, Collini P, Nantron M, Pession A, Bianchi M, Indolfi P, D'Angelo P, Fossati-Bellani F, Verderio P, Macciardi F, Radice P. Genomic profiling by whole-genome single nucleotide polymorphism arrays in Wilms tumor and association with relapse. Genes Chromosomes Cancer 2012; 51:644-53. [PMID: 22407497 DOI: 10.1002/gcc.21951] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 02/01/2012] [Accepted: 02/01/2012] [Indexed: 01/21/2023] Open
Abstract
Despite the excellent survival rate of Wilms tumor (WT) patients, only approximately one-half of children who suffer tumor recurrence reach second durable remission. This underlines the need for novel markers to optimize initial treatment. We investigated 77 tumors using Illumina 370CNV-QUAD genotyping BeadChip arrays and compared their genomic profiles to detect copy number (CN) abnormalities and allelic ratio anomalies associated with the following clinicopathological variables: relapse (yes vs. no), age at diagnosis (≤ 24 months vs. >24 months), and disease stage (low stage, I and II, vs. high stage, III and IV). We found that CN gains at chromosome region 1q21.1-q31.3 were significantly associated with relapse. Additional genetic events, including allelic imbalances at chromosome arms 1p, 1q, 3p, 3q, and 14q were also found to occur at higher frequency in relapsing tumors. Interestingly, allelic imbalances at 1p and 14q also showed a borderline association with higher tumor stages. No genetic events were found to be associated with age at diagnosis. This is the first genome wide analysis with single nucleotide polymorphism (SNP) arrays specifically investigating the role of genetic anomalies in predicting WT relapse on cases prospectively enrolled in the same clinical trial. Our study, besides confirming the role of 1q gains, identified a number of additional candidate genetic markers, warranting further molecular investigations.
Collapse
Affiliation(s)
- Daniela Perotti
- Department of Preventive and Predictive Medicine, Unit of Molecular Bases of Genetic Risk and Genetic Testing, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Grade M, Wolff HA, Gaedcke J, Ghadimi BM. The molecular basis of chemoradiosensitivity in rectal cancer: implications for personalized therapies. Langenbecks Arch Surg 2012; 397:543-55. [PMID: 22382702 PMCID: PMC3314820 DOI: 10.1007/s00423-012-0929-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 02/14/2012] [Indexed: 12/11/2022]
Abstract
Introduction Preoperative chemoradiotherapy represents the standard treatment for patients with locally advanced rectal cancer. Unfortunately, the response of individual tumors to multimodal treatment is not uniform and ranges from complete response to complete resistance. This poses a particular problem for patients with a priori resistant tumors because they may be exposed to irradiation and chemotherapy, treatment regimens that are both expensive and at times toxic, without benefit. Accordingly, there is a strong need to establish molecular biomarkers that predict the response of an individual patient’s tumor to multimodal treatment and that indicate treatment-associated toxicities prior to therapy. Such biomarkers may guide clinicians in choosing the best possible treatment for each individual patient. In addition, these biomarkers could be used to identify novel molecular targets and thereby assist in implementing novel strategies to sensitize a priori resistant tumors to multimodal treatment regimens. Objective The aim of this review is to summarize recent findings about the molecular basis of treatment resistance and treatment toxicity in patients with rectal cancer. Whole-genome, as well as single-biomarker or multibiomarker, analyses and their potential implications will be highlighted. At the end, we will outline a future vision of rectal cancer treatment in the era of personalized medicine.
Collapse
Affiliation(s)
- Marian Grade
- Department of General and Visceral Surgery, University Medical Center Göttingen, Robert-Koch Str. 40, 37075 Göttingen, Germany.
| | | | | | | |
Collapse
|