1
|
Papaccio F, Cabeza-Segura M, García-Micó B, Gimeno-Valiente F, Zúñiga-Trejos S, Gambardella V, Gutiérrez-Bravo MF, Martinez-Ciarpaglini C, Rentero-Garrido P, Fleitas T, Roselló S, Carbonell-Asins JA, Huerta M, Moro-Valdezate D, Roda D, Tarazona N, Sánchez Del Pino MM, Cervantes A, Castillo J. Decoding chromosomal instability insights in CRC by integrating omics and patient-derived organoids. J Exp Clin Cancer Res 2025; 44:77. [PMID: 40022181 PMCID: PMC11869439 DOI: 10.1186/s13046-025-03308-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/28/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Chromosomal instability (CIN) is involved in about 70% of colorectal cancers (CRCs) and is associated with poor prognosis and drug resistance. From a clinical perspective, a better knowledge of these tumour's biology will help to guide therapeutic strategies more effectively. METHODS We used high-density chromosomal microarray analysis to evaluate CIN level of patient-derived organoids (PDOs) and their original mCRC tissues. We integrated the RNA-seq and mass spectrometry-based proteomics data from PDOs in a functional interaction network to identify the significantly dysregulated processes in CIN. This was followed by a proteome-wGII Pearson correlation analysis and an in silico validation of main findings using functional genomic databases and patient-tissues datasets to prioritize the high-confidence CIN features. RESULTS By applying the weighted Genome Instability Index (wGII) to identify CIN, we classified PDOs and demonstrated a good correlation with tissues. Multi-omics analysis showed that our organoids recapitulated genomic, transcriptomic and proteomic CIN features of independent tissues cohorts. Thanks to proteotranscriptomics, we uncovered significant associations between mitochondrial metabolism and epithelial-mesenchymal transition in CIN CRC PDOs. Correlating PDOs wGII with protein abundance, we identified a subset of proteins significantly correlated with CIN. Co-localisation analysis in PDOs strengthened the putative role of IPO7 and YAP, and, through in silico analysis, we found that some of the targets give significant dependencies in cell lines with CIN compatible status. CONCLUSIONS We first demonstrated that PDO models are a faithful reflection of CIN tissues at the genetic and phenotypic level. Our new findings prioritize a subset of genes and molecular processes putatively required to cope with the burden on cellular fitness imposed by CIN and associated with disease aggressiveness.
Collapse
Affiliation(s)
- Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081, Baronissi, Italy.
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain.
| | - Manuel Cabeza-Segura
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
| | - Blanca García-Micó
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Francisco Gimeno-Valiente
- Cancer Evolution and Genome Instability Laboratory, University College London Cancer Institute, London, UK
| | - Sheila Zúñiga-Trejos
- Bioinformatic Unit, INCLIVA Biomedical Research Institute, Avda. Menéndez y Pelayo 3, 46010, Valencia, Spain
| | - Valentina Gambardella
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - María Fernanda Gutiérrez-Bravo
- Experimental and Applied Biomedicine Research Group, Health Sciences Faculty, Universidad Particular Internacional SEK (UISEK), Quito, 170302, Ecuador
| | - Carolina Martinez-Ciarpaglini
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Department of Pathology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
| | - Pilar Rentero-Garrido
- Precision Medicine Unit, INCLIVA Biomedical Research Institute, Avda. Menéndez y Pelayo 4, 46010, Valencia, Spain
| | - Tania Fleitas
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Susana Roselló
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | | | - Marisol Huerta
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
| | - David Moro-Valdezate
- Department of General Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Desamparados Roda
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Noelia Tarazona
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Manuel M Sánchez Del Pino
- Institute of Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100, Burjassot, Spain.
- Department of Biochemistry and Molecular Biology, University of Valencia, 46100, Burjassot, Spain.
| | - Andrés Cervantes
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain.
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Josefa Castillo
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Avda. Blasco Ibañez 17, 46010, Valencia, Spain.
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
- Department of Biochemistry and Molecular Biology, University of Valencia, 46100, Burjassot, Spain.
| |
Collapse
|
2
|
Zhao Y, Xia S, Zhao X, Song Z, Wang F, Mao L, He Z, Du X. DNA ploidy combined with tumor stroma as a biomarker for predicting the prognosis of stage II colorectal cancer patients and identifying candidates for chemotherapy. World J Surg Oncol 2025; 23:49. [PMID: 39940036 PMCID: PMC11823223 DOI: 10.1186/s12957-025-03693-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
PURPOSE The efficacy of postoperative adjuvant chemotherapy in patients with stage II colorectal cancer has been a subject of debate. This study aimed to evaluate the prognostic and predictive significance of DNA ploidy and stroma ratio in patients diagnosed with stage II colorectal cancer (CRC). METHODS Clinical data and tumor tissues from 179 patients with stage II CRC were collected retrospectively. DNA ploidy (P) and stroma (S) were assessed using automatic image analysis tools powered by machine learning. RESULTS Patients were categorized into three risk groups: PS-low (diploid and low stroma, PS-L), PS-intermediate (non-diploid or high stroma, PS-M), and PS-high (non-diploid and high-stroma, PS-H). According to the univariable model, the PS-H group exhibited significantly poorer 5-year overall survival rates at 73.0% compared to 87.8%, with a hazard ratio (HR) of 2.281 (95% CI: 0.946-5.502, P = 0.066), as well as lower 5-year disease-free survival rates at 69.4% versus 86.6%, HR = 2.323 (95% CI: 1.016-5.308, P = 0.046) among stage II colorectal cancer patients. Notably, chemotherapy was associated with improved overall survival [HR = 83.460 (95% CI: 0.179-38925.833), P = 0.003] and disease-free survival [HR = 8.628 (95% CI: 1.059-70.265), P = 0 .044] in individuals within the PS-high group. CONCLUSION While ploidy and stroma alone do not possess predictive power regarding survival outcomes for stage II colorectal cancer patients, those receiving chemotherapy within the PS-H group demonstrated enhanced survival rates. Therefore, combining assessments of ploidy and stroma may serve as an adjunctive tool in clinical decision-making processes to guide chemotherapy treatment strategies for patients diagnosed with stage II colorectal cancer.
Collapse
Affiliation(s)
- Yunshan Zhao
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Haidian District, 28 Fuxing Road, Beijing, 100853, China
| | - Shaoyou Xia
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Haidian District, 28 Fuxing Road, Beijing, 100853, China
| | - Xudong Zhao
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Haidian District, 28 Fuxing Road, Beijing, 100853, China
| | - Zhigang Song
- Department of Pathology, First Medical Center of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Fei Wang
- My-BioMed Technology (Guangzhou) Co., Ltd., Guangzhou, 510535, China
| | - Lijun Mao
- My-BioMed Technology (Guangzhou) Co., Ltd., Guangzhou, 510535, China
| | - Zufeng He
- My-BioMed Technology (Guangzhou) Co., Ltd., Guangzhou, 510535, China
| | - Xiaohui Du
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Haidian District, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
3
|
Silva MPME, Sabbaga J, Najman HL, Nascimento CDC, Cotta-Pereira RL, Nicoluzzi JEL, Braghiroli MI. FROM ONCOLOGIST TO SURGEON - GENETICS IN COLORECTAL METASTASIS FOR SURGEONS. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2025; 37:e1869. [PMID: 39936820 PMCID: PMC11810113 DOI: 10.1590/0102-6720202400075e1869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/30/2024] [Indexed: 02/13/2025]
Abstract
Colorectal cancer (CRC) is a common disease, with incidence in Brazil of 45,630 new cases per 100,000 inhabitants between 2023-2025. Risk factors for CRC can be evaluated between environmental and hereditary and their mode of presentation are classified as sporadic, inherited and familial. Sporadic disease is characterized by the absence of a family history and accounts for approximately 70% of all colorectal cancers, being more common over 50 years of age, with dietary and environmental factors implicated in its pathogenesis. Sporadic disease is characterized by the absence of a family history and accounts for approximately 70% of all colorectal cancers, being more common over 50 years of age, with dietary and environmental factors implicated in its pathogenesis. The percentage of patients with a true hereditary genetic predisposition is less than 10%, and these are related to the presence or absence of colonic polyps as an important manifestation of the disease. Non-polyposis diseases are known as hereditary non-polypomatous colorectal cancer (HNPCC) or Lynch syndrome, and polyposis diseases are familial adenomatous polyposis (FAP), MUTYH-associated polyposis (MAP), and hamartomatous polyposis syndromes (e.g., Peutz-Jeghers, juvenile polyposis, phosphatase and tensin homologue - PTEN, Cowden syndrome). These diseases are linked to a high risk of developing cancer. With the development of treatments in metastatic disease and the use of targeted therapies and their biomarkers, it was possible to evaluate them within clinical studies both in the primary tumor and in the correspondence of metastases.
Collapse
Affiliation(s)
- Marília Polo Mingueti E Silva
- D'Or Institute for Research and Education - São Paulo (SP), Brazil
- D'Or Institute for Research and Education - Rio de Janeiro (RJ), Brazil
- Universidade de São Paulo, Faculty of Medicine, Cancer Institute - São Paulo (SP), Brazil
| | - Jorge Sabbaga
- D'Or Institute for Research and Education - São Paulo (SP), Brazil
- D'Or Institute for Research and Education - Rio de Janeiro (RJ), Brazil
- Universidade de São Paulo, Faculty of Medicine, Cancer Institute - São Paulo (SP), Brazil
| | | | | | | | | | - Maria Ignez Braghiroli
- D'Or Institute for Research and Education - São Paulo (SP), Brazil
- D'Or Institute for Research and Education - Rio de Janeiro (RJ), Brazil
- Universidade de São Paulo, Faculty of Medicine, Cancer Institute - São Paulo (SP), Brazil
| |
Collapse
|
4
|
Schettini F, Sirico M, Loddo M, Williams GH, Hardisty KM, Scorer P, Thatcher R, Rivera P, Milani M, Strina C, Ferrero G, Ungari M, Bottin C, Zanconati F, de Manzini N, Aguggini S, Tancredi R, Fiorio E, Fioravanti A, Scaltriti M, Generali D. Next-generation sequencing-based evaluation of the actionable landscape of genomic alterations in solid tumors: the "MOZART" prospective observational study. Oncologist 2025; 30:oyae206. [PMID: 39177668 PMCID: PMC11783315 DOI: 10.1093/oncolo/oyae206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND The identification of the most appropriate targeted therapies for advanced cancers is challenging. We performed a molecular profiling of metastatic solid tumors utilizing a comprehensive next-generation sequencing (NGS) assay to determine genomic alterations' type, frequency, actionability, and potential correlations with PD-L1 expression. METHODS A total of 304 adult patients with heavily pretreated metastatic cancers treated between January 2019 and March 2021 were recruited. The CLIA-/UKAS-accredit Oncofocus assay targeting 505 genes was used on newly obtained or archived biopsies. Chi-square, Kruskal-Wallis, and Wilcoxon rank-sum tests were used where appropriate. Results were significant for P < .05. RESULTS A total of 237 tumors (78%) harbored potentially actionable genomic alterations. Tumors were positive for PD-L1 in 68.9% of cases. The median number of mutant genes/tumor was 2.0 (IQR: 1.0-3.0). Only 34.5% were actionable ESCAT Tier I-II with different prevalence according to cancer type. The DNA damage repair (14%), the PI3K/AKT/mTOR (14%), and the RAS/RAF/MAPK (12%) pathways were the most frequently altered. No association was found among PD-L1, ESCAT, age, sex, and tumor mutational status. Overall, 62 patients underwent targeted treatment, with 37.1% obtaining objective responses. The same molecular-driven treatment for different cancer types could be associated with opposite clinical outcomes. CONCLUSIONS We highlight the clinical value of molecular profiling in metastatic solid tumors using comprehensive NGS-based panels to improve treatment algorithms in situations of uncertainty and facilitate clinical trial recruitment. However, interpreting genomic alterations in a tumor type-specific manner is critical.
Collapse
Affiliation(s)
- Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”47014, Meldola, Italy
| | - Marco Loddo
- Oncologica UK Ltd, Cambridge CB10 1XL, United Kingdom
| | | | | | - Paul Scorer
- Oncologica UK Ltd, Cambridge CB10 1XL, United Kingdom
| | | | - Pablo Rivera
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Manuela Milani
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Carla Strina
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Giuseppina Ferrero
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Marco Ungari
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Cristina Bottin
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Nicolò de Manzini
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Sergio Aguggini
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Richard Tancredi
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Elena Fiorio
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, 37134, Verona, Italy
| | | | - Maurizio Scaltriti
- Neurosurgery Unit, ASST Cremona, 26100, Cremona, Italy
- AstraZeneca, Gaithersburg, MD 20876, United States
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| |
Collapse
|
5
|
Phillips AF, Zhang R, Jaffe M, Schulz R, Carty MC, Verma A, Feinberg TY, Arensman MD, Chiu A, Letso R, Bosco N, Queen KA, Racela AR, Stumpff J, Andreu-Agullo C, Bettigole SE, Depetris RS, Drutman S, Su SM, Cogan DA, Eng CH. Targeting chromosomally unstable tumors with a selective KIF18A inhibitor. Nat Commun 2025; 16:307. [PMID: 39747049 PMCID: PMC11697083 DOI: 10.1038/s41467-024-55300-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Chromosome instability is a prevalent vulnerability of cancer cells that has yet to be fully exploited therapeutically. To identify genes uniquely essential to chromosomally unstable cells, we mined the Cancer Dependency Map for genes essential in tumor cells with high levels of copy number aberrations. We identify and validate KIF18A, a mitotic kinesin, as a vulnerability of chromosomally unstable cancer cells. Knockdown of KIF18A leads to mitotic defects and reduction of tumor growth. Screening of a chemical library for inhibitors of KIF18A enzymatic activity identified a hit that was optimized to yield VLS-1272, which is orally bioavailable, potent, ATP non-competitive, microtubule-dependent, and highly selective for KIF18A versus other kinesins. Inhibition of KIF18A's ATPase activity prevents KIF18A translocation across the mitotic spindle, resulting in chromosome congression defects, mitotic cell accumulation, and cell death. Profiling VLS-1272 across >100 cancer cell lines demonstrates that the specificity towards cancer cells with chromosome instability differentiates KIF18A inhibition from other clinically tested anti-mitotic drugs. Treatment of tumor xenografts with VLS-1272 results in mitotic defects leading to substantial, dose-dependent inhibition of tumor growth. The strong biological rationale, robust preclinical data, and optimized compound properties enable the clinical development of a KIF18A inhibitor in cancers with high chromosomal instability.
Collapse
Affiliation(s)
| | | | - Mia Jaffe
- Volastra Therapeutics, New York, NY, USA
| | | | | | | | | | | | - Alan Chiu
- Volastra Therapeutics, New York, NY, USA
| | - Reka Letso
- Volastra Therapeutics, New York, NY, USA
| | | | - Katelyn A Queen
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Allison R Racela
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Li W, Lan J, Zhou C, Yang R, Wang J, He J, Xiao B, Ou Q, Fang Y, Fan W, Lin J, Pan Z, Peng J, Wu X. Chromosomal instability is associated with prognosis and efficacy of bevacizumab after resection of colorectal cancer liver metastasis. Ann Med 2024; 56:2396559. [PMID: 39247989 PMCID: PMC11385633 DOI: 10.1080/07853890.2024.2396559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 09/10/2024] Open
Abstract
INTRODUCTION Individualized treatment of colorectal cancer liver metastases (CRLM) remains challenging due to differences in the severity of metastatic disease and tumour biology. Exploring specific prognostic risk subgroups is urgently needed. The current study aimed to investigate the prognostic value of chromosomal instability (CIN) in patients with initially resectable CRLM and the predictive value of CIN for the efficacy of bevacizumab. METHODS Ninety-one consecutive patients with initially resectable CRLM who underwent curative liver resection from 2006 to 2018 at Sun Yat-sen University Cancer Center were selected for analysis. CIN was evaluated by automated digital imaging systems. Immunohistochemistry (IHC) was performed to detect interleukin-6 (IL-6), vascular endothelial growth factor A (VEGFA) and CD31 expression in paraffin-embedded specimens. Recurrence-free survival (RFS) and overall survival (OS) were analysed using the Kaplan-Meier method and Cox regression models. RESULTS Patients with high chromosomal instability (CIN-H) had a worse 3-year RFS rate (HR, 1.953; 95% CI, 1.001-3.810; p = 0.049) and a worse 3-year OS rate (HR, 2.449; 95% CI, 1.150-5.213; p = 0.016) than those with low chromosomal instability (CIN-L). CIN-H was identified as an independent prognostic factor for RFS (HR, 2.569; 95% CI, 1.078-6.121; p = 0.033) and OS (HR, 3.852; 95% CI, 1.173-12.645; p = 0.026) in the multivariate analysis. The protein levels of IL-6, VEGFA and CD31 were upregulated in patients in the CIN-H group compared to those in the CIN-L group in both primary tumour and liver metastases tissues. Among them, 22 patients with recurrent tumours were treated with first-line bevacizumab treatment and based on the clinical response assessment, disease control rates were adversely associated with chromosomal instability (p = 0.043). CONCLUSIONS Our study showed that high chromosomal instability is a negative prognostic factor for patients with initially resectable CRLM after liver resection. CIN may have positive correlations with angiogenesis through expression of IL-6-VEGFA axis and be used as a potential predictor of efficacy of bevacizumab.
Collapse
Affiliation(s)
- Weihao Li
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jin Lan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chi Zhou
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Rong Yang
- Department of Intensive Care Unit, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiayu Wang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, P. R. China
| | - Jiahua He
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Binyi Xiao
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qingjian Ou
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yujing Fang
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wenhua Fan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Junzhong Lin
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhizhong Pan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jianhong Peng
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiaojun Wu
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
7
|
Fang X, Yu WY, Zhu CM, Zhao N, Zhao W, Xie TT, Wei LJ, Sun XR, Xie J, Zhao Y. Chromosome instability functions as a potential therapeutic reference by enhancing chemosensitivity to BCL-XL inhibitors in colorectal carcinoma. Acta Pharmacol Sin 2024; 45:2420-2431. [PMID: 39187678 PMCID: PMC11489767 DOI: 10.1038/s41401-024-01372-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Chromosome instability (CIN) and subsequent aneuploidy are prevalent in various human malignancies, influencing tumor progression such as metastases and relapses. Extensive studies demonstrate the development of chemoresistance in high-CIN tumors, which poses significant therapeutic challenges. Given the association of CIN with poorer prognosis and suppressed immune microenvironment observed in colorectal carcinoma (CRC), here we aimed to discover chemotherapeutic drugs exhibiting increased inhibition against high-CIN CRC cells. By using machine learning methods, we screened out two BCL-XL inhibitors Navitoclax and WEHI-539 as CIN-sensitive reagents in CRC. Subsequent analyses using a CIN-aneuploidy cell model confirmed the vulnerability of high-CIN CRC cells to these drugs. We further revealed the critical role of BCL-XL in the viability of high-CIN CRC cells. In addition, to ease the evaluation of CIN levels in clinic, we developed a three-gene signature as a CIN surrogate to predict prognosis, chemotherapeutic and immune responses in CRC samples. Our results demonstrate the potential value of CIN as a therapeutic target in CRC treatment and the importance of BCL-XL in regulating survival of high-CIN CRC cells, therefore representing a valuable attempt to translate a common trait of heterogeneous tumor cells into an effective therapeutic target.
Collapse
Affiliation(s)
- Xiao Fang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Clinical Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Wen-Ying Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Chun-Miao Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Nan Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Wei Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Ting-Ting Xie
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Li-Jie Wei
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Xi-Ran Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Juan Xie
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Ya Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China.
| |
Collapse
|
8
|
Devillers R, Dos Santos A, Destombes Q, Laplante M, Elowe S. Recent insights into the causes and consequences of chromosome mis-segregation. Oncogene 2024; 43:3139-3150. [PMID: 39278989 DOI: 10.1038/s41388-024-03163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
Mitotic cells face the challenging task of ensuring accurate and equal segregation of their duplicated, condensed chromosomes between the nascent daughter cells. Errors in the process result in chromosome missegregation, a significant consequence of which is the emergence of aneuploidy-characterized by an imbalance in chromosome number-and the associated phenomenon of chromosome instability (CIN). Aneuploidy and CIN are common features of cancer, which leverages them to promote genome heterogeneity and plasticity, thereby facilitating rapid tumor evolution. Recent research has provided insights into how mitotic errors shape cancer genomes by inducing both numerical and structural chromosomal changes that drive tumor initiation and progression. In this review, we survey recent findings regarding the mitotic causes and consequences of aneuploidy. We discuss new findings into the types of chromosome segregation errors that lead to aneuploidy and novel pathways that protect genome integrity during mitosis. Finally, we describe new developments in our understanding of the immediate consequences of chromosome mis-segregation on the genome stability of daughter cells.
Collapse
Affiliation(s)
- Romain Devillers
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Alexsandro Dos Santos
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Quentin Destombes
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Mathieu Laplante
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Sabine Elowe
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada.
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada.
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada.
- Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec City, QC, Canada.
| |
Collapse
|
9
|
Karami Fath M, Nazari A, Parsania N, Behboodi P, Ketabi SS, Razmjouei P, Farzam F, Shafagh SG, Nabi Afjadi M. Centromeres in cancer: Unraveling the link between chromosomal instability and tumorigenesis. Med Oncol 2024; 41:254. [PMID: 39352464 DOI: 10.1007/s12032-024-02524-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/23/2024] [Indexed: 11/14/2024]
Abstract
Centromeres are critical structures involved in chromosome segregation, maintaining genomic stability, and facilitating the accurate transmission of genetic information. They are key in coordinating the assembly and help keep the correct structure, location, and function of the kinetochore, a proteinaceous structure vital for ensuring proper chromosome segregation during cell division. Abnormalities in centromere structure can lead to aneuploidy or chromosomal instability, which have been implicated in various diseases, including cancer. Accordingly, abnormalities in centromeres, such as structural rearrangements and dysregulation of centromere-associated proteins, disrupt gene function, leading to uncontrolled cell growth and tumor progression. For instance, altered expression of CENP-A, CENP-E, and others such as BUB1, BUBR1, MAD1, and INCENP, have been shown to ascribe to centromere over-amplification, chromosome missegregation, aneuploidy, and chromosomal instability; this, in turn, can culminate in tumor progression. These centromere abnormalities also promoted tumor heterogeneity by generating genetically diverse cell populations within tumors. Advanced techniques like fluorescence in situ hybridization (FISH) and chromosomal microarray analysis are crucial for detecting centromere abnormalities, enabling accurate cancer classification and tailored treatment strategies. Researchers are exploring strategies to disrupt centromere-associated proteins for targeted cancer therapies. Thus, this review explores centromere abnormalities in cancer, their molecular mechanisms, diagnostic implications, and therapeutic targeting. It aims to advance our understanding of centromeres' role in cancer and develop advanced diagnostic tools and targeted therapies for improved cancer management and treatment.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ahmad Nazari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Parsania
- Department of Brain and Cognitive Sciences, Cell Science Research Center, ROYAN Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Paria Behboodi
- Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Pegah Razmjouei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
10
|
González A, Badiola I, Fullaondo A, Rodríguez J, Odriozola A. Personalised medicine based on host genetics and microbiota applied to colorectal cancer. ADVANCES IN GENETICS 2024; 112:411-485. [PMID: 39396842 DOI: 10.1016/bs.adgen.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) ranks second in incidence and third in cancer mortality worldwide. This situation, together with the understanding of the heterogeneity of the disease, has highlighted the need to develop a more individualised approach to its prevention, diagnosis and treatment through personalised medicine. This approach aims to stratify patients according to risk, predict disease progression and determine the most appropriate treatment. It is essential to identify patients who may respond adequately to treatment and those who may be resistant to treatment to avoid unnecessary therapies and minimise adverse side effects. Current research is focused on identifying biomarkers such as specific mutated genes, the type of mutations and molecular profiles critical for the individualisation of CRC diagnosis, prognosis and treatment guidance. In addition, the study of the intestinal microbiota as biomarkers is being incorporated due to the growing scientific evidence supporting its influence on this disease. This article comprehensively addresses the use of current and emerging diagnostic, prognostic and predictive biomarkers in precision medicine against CRC. The effects of host genetics and gut microbiota composition on new approaches to treating this disease are discussed. How the gut microbiota could mitigate the side effects of treatment is reviewed. In addition, strategies to modulate the gut microbiota, such as dietary interventions, antibiotics, and transplantation of faecal microbiota and phages, are discussed to improve CRC prevention and treatment. These findings provide a solid foundation for future research and improving the care of CRC patients.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Iker Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | | | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
11
|
Peng J, Zhang W, He J, Wang W, Li W, Mao L, Dong Y, Lu Z, Pan Z, Zhou C, Wu X. Combination of DNA ploidy, stroma, and nucleotyping predicting prognosis and tailoring adjuvant chemotherapy duration in stage III colon cancer. Ther Adv Med Oncol 2024; 16:17588359241260575. [PMID: 38894737 PMCID: PMC11185039 DOI: 10.1177/17588359241260575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction DNA ploidy (P), stroma fraction (S), and nucleotyping (N) collectively known as PSN, have proven prognostic accuracy in stage II colorectal cancer (CRC). However, few studies have reported on the prognostic value of the PSN panel in stage III colon cancer patients receiving capecitabine and oxaliplatin adjuvant chemotherapy. Objectives This study aimed to validate PSN's prognostic impact on stage III colon cancer, identifying candidates for optimized adjuvant chemotherapy duration. Design A retrospective analysis was conducted on a cohort of stage III colon cancer patients from April 2008 to June 2020. Methods Postoperative pathological samples from stage III colon cancer patients who underwent radical surgery and postoperative adjuvant chemotherapy at Sun Yat-sen University Cancer Center were retrospectively collected. Automated digital imaging assessed PSN, categorizing risk groups. Kaplan-Meier, Cox regression, and time-dependent receiver operating characteristic analysis compared model validity. Results Significant differences in 5-year disease-free survival (DFS) and overall survival (OS) were noted among PSN-based low-, moderate-, and high-risk groups (DFS: 92.10% versus 83.62% versus 79.80%, p = 0.029; OS: 96.69% versus 93.99% versus 90.12%, p = 0.016). PSN emerged as an independent prognostic factor for DFS [hazard ratio (HR) = 1.409, 95% confidence interval (CI): 1.002-1.981, p = 0.049] and OS (HR = 1.720, 95% CI: 1.127-2.624, p = 0.012). The PSN model, incorporating perineural invasion and tumor location, displayed superior area under the curve for 5-year (0.692 versus 0.553, p = 0.020) and 10-year (0.694 versus 0.532, p = 0.006) DFS than TNM stage. In the PSN high-risk group, completing eight cycles of adjuvant chemotherapy significantly improved 5-year DFS and OS compared to four to seven cycles (DFS: 89.43% versus 71.52%, p = 0.026; OS: 96.77% versus 85.46%, p = 0.007). Conclusion The PSN panel effectively stratifies stage III colon cancer, aiding in optimized adjuvant chemotherapy duration determination.
Collapse
Affiliation(s)
- Jianhong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, P.R. China
| | - Weili Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, P.R. China
| | - Jiahua He
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, P.R. China
| | - Weifeng Wang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, P.R. China
| | - Weihao Li
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, P.R. China
| | - Lijun Mao
- My-BioMed Technology (Guangzhou) Co., Ltd, Guangzhou, Guangdong, P.R. China
| | - Yuejin Dong
- My-BioMed Technology (Guangzhou) Co., Ltd, Guangzhou, Guangdong, P.R. China
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, P.R. China
| | - Zhizhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, P.R. China
| | - Chi Zhou
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaojun Wu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
12
|
Benmokhtar S, Laraqui A, Hilali F, Bajjou T, El Zaitouni S, Jafari M, Baba W, Elannaz H, Lahlou IA, Hafsa C, Oukabli M, Mahfoud T, Tanz R, Ichou M, Ennibi K, Dakka N, Sekhsokh Y. RAS/RAF/MAPK Pathway Mutations as Predictive Biomarkers in Middle Eastern Colorectal Cancer: A Systematic Review. Clin Med Insights Oncol 2024; 18:11795549241255651. [PMID: 38798959 PMCID: PMC11128178 DOI: 10.1177/11795549241255651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Background This review article aims to investigate the prevalence and spectrum of rat sarcoma (RAS) and V-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF) mutations, and their connection with geographical location, clinicopathological features, and other relevant factors in colorectal cancer (CRC) patients in the Middle East. Methods A systematic literature review, employing the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) framework, was conducted to investigate the association between the frequency of relevant mutations and the descriptive clinicopathological characteristics of CRC patients. Multiple electronic databases, including PubMed, Science Direct, Web of Science, Scopus, and Google Scholar, were searched to analyze the relevant literature. Results A total of 19 eligible studies comprising 2960 patients with CRC were included in this review. A comprehensive analysis of the collected literature data as well as descriptive and methodological insights is provided. Men were predominant in reviewed studies for the region, accounting for 58.6%. Overall, RAS mutation prevalence was 38.1%. Kirsten RAS Viral Oncogene Homolog (KRAS) mutations were the most common, accounting for 37.1% of cases and distributed among different exons, with the G12D mutation being the most frequent in exon 2 (23.2%) followed by G12V (13.7%), G13D (10.1%), G12C (5.1%), G12A (5.04%), and G12S (3.6%). Neuroblastoma RAS Viral Oncogene Homolog (NRAS) mutations were identified in 3.3% of tumor samples, with the most common mutation site located in exons 2, 3, and 4, and codon 61 being the most common location for the region. The total mutation frequency in the BRAF gene was 2.6%, with the V600E mutation being the most common. Conclusion The distribution patterns of RAS and BRAF mutations among CRC patients exhibit notable variations across diverse ethnic groups. Our study sheds light on this phenomenon by demonstrating a higher prevalence of KRAS mutations in CRC patients from the Middle East, as compared with those from other regions. The identification of these mutations and geographical differences is important for personalized treatment planning and could potentially aid in the development of novel targeted therapies. The distinct distribution patterns of RAS and BRAF mutations among CRC patients across different ethnic groups, as well as the regional variability in mutation prevalence, highlight the need for further research in this area.
Collapse
Affiliation(s)
- Soukaina Benmokhtar
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
- Laboratory of Biology of Human Pathologies and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Abdelilah Laraqui
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
- Laboratory of Biology of Human Pathologies and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Farida Hilali
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Tahar Bajjou
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Sara El Zaitouni
- Laboratory of Biology of Human Pathologies and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Meryem Jafari
- Laboratory of Biology of Human Pathologies and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Walid Baba
- Laboratory of Biology of Human Pathologies and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Hicham Elannaz
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Idriss Amine Lahlou
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Chahdi Hafsa
- Department of Medical Oncology, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Mohamed Oukabli
- Department of Pathology, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Tarik Mahfoud
- Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Rachid Tanz
- Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Mohamed Ichou
- Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Khaled Ennibi
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
- Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Nadia Dakka
- Laboratory of Biology of Human Pathologies and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Yassine Sekhsokh
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| |
Collapse
|
13
|
Liu L, Zhang P, Liu Z, Sun T, Qiao H. Joint global and local interpretation method for CIN status classification in breast cancer. Heliyon 2024; 10:e27054. [PMID: 38562500 PMCID: PMC10982965 DOI: 10.1016/j.heliyon.2024.e27054] [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: 06/14/2023] [Revised: 12/10/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Breast cancer is among the cancer types with the highest numbers of new cases. The study of this disease from a microscopic perspective has been a prominent research topic. Previous studies have shown that microRNAs (miRNAs) are closely linked to chromosomal instability (CIN). Correctly predicting CIN status from miRNAs can help to improve the survival of breast cancer patients. In this study, a joint global and local interpretation method called GL_XGBoost is proposed for predicting CIN status in breast cancer. GL_XGBoost integrates the eXtreme Gradient Boosting (XGBoost) and SHapley Additive exPlanation (SHAP) methods. XGBoost is used to predict CIN status from miRNA data, whereas SHAP is used to select miRNA features that have strong relationships with CIN. Furthermore, SHAP's rich visualization strategies enhance the interpretability of the entire model at the global and local levels. The performance of GL_XGBoost is validated on the TCGA-BRCA dataset, and it is shown to have an accuracy of 78.57% and an area under the curve value of 0.87. Rich visual analysis is used to explain the relationships between miRNAs and CIN status from different perspectives. Our study demonstrates an intuitive way of exploring the relationship between CIN and cancer from a microscopic perspective.
Collapse
Affiliation(s)
- Liangliang Liu
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, PR China
| | - Pei Zhang
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, PR China
| | - Zhihong Liu
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, PR China
| | - Tong Sun
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, PR China
| | - Hongbo Qiao
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, PR China
| |
Collapse
|
14
|
Cheng F, Li P, Xu S, Zhang C, Liang H, Ding Z. A pair of primary colorectal cancer-derived and corresponding synchronous liver metastasis-derived organoid cell lines. Aging (Albany NY) 2024; 16:4396-4422. [PMID: 38407980 PMCID: PMC10968669 DOI: 10.18632/aging.205595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024]
Abstract
Proper preclinical models for the research of colorectal cancer (CRC) and CRC liver metastases (CLM) are a clear and unmet need. Patient-derived organoids have recently emerged as a robust preclinical model, but are not available to all scientific researchers. Here, we present paired 3D organoid cell lines of CWH22 (CRC-derived) and CLM22 (CLM-derived) with sound background information and the short tandem repeats are identical to those of the normal tissue. Morphological and immunohistochemical staining, along with whole-exome sequencing (WES), confirmed that the organoids exhibited the same differentiation, molecular expression, and mutation status as the corresponding tumor tissue. Both organoids possessed mutated APC/KRAS/SMAD4/CDKN1B/KMT2C genes and wild-type TP53 and PIK3CA; stably secreted the tumor markers CEA and CA19-9, and possessed sound proliferation rates in vitro, as well as subcutaneous tumorigenicity and liver metastatic abilities in vivo. IC50 assays confirmed that both cell lines were sensitive to 5-fluorouracil, oxaliplatin, SN-38, and sotorasib. WES and karyotype analyses revealed the genomic instability status as chromosome instability. The corresponding adherent cultured CWH22-2D/CLM22-2D cells were established and compared with commonly used CRC cell lines from the ATCC. Both organoids are publicly available to all researchers and will be useful tools for specific human CRC/CLM studies both in vitro and in vivo.
Collapse
Affiliation(s)
- Fangling Cheng
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, China
| | - Pengcheng Li
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, China
| | - Sanpeng Xu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chao Zhang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huifang Liang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, China
| | - Zeyang Ding
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, China
| |
Collapse
|
15
|
Ye J, Zhang J, Ding W. DNA methylation modulates epigenetic regulation in colorectal cancer diagnosis, prognosis and precision medicine. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:34-53. [PMID: 38464391 PMCID: PMC10918240 DOI: 10.37349/etat.2024.00203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/11/2023] [Indexed: 03/12/2024] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease influenced by the interplay of genetic and environmental factors. The clinical heterogeneity of CRC cannot be attributed exclusively to genetic diversity and environmental exposures, and epigenetic markers, especially DNA methylation, play a critical role as key molecular markers of cancer. This review compiles a comprehensive body of evidence underscoring the significant involvement of DNA methylation modifications in the pathogenesis of CRC. Moreover, this review explores the potential utility of DNA methylation in cancer diagnosis, prognostics, assessment of disease activity, and prediction of drug responses. Recognizing the impact of DNA methylation will enhance the ability to identify distinct CRC subtypes, paving the way for personalized treatment strategies and advancing precision medicine in the management of CRC.
Collapse
Affiliation(s)
- Jingxin Ye
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
- Department of Gastroenterology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian 223800, Jiangsu Province, China
| | - Jianfeng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Weifeng Ding
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
16
|
Trembath HE, Yeh JJ, Lopez NE. Gastrointestinal Malignancy: Genetic Implications to Clinical Applications. Cancer Treat Res 2024; 192:305-418. [PMID: 39212927 DOI: 10.1007/978-3-031-61238-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Advances in molecular genetics have revolutionized our understanding of the pathogenesis, progression, and therapeutic options for treating gastrointestinal (GI) cancers. This chapter provides a comprehensive overview of the molecular landscape of GI cancers, focusing on key genetic alterations implicated in tumorigenesis across various anatomical sites including GIST, colon and rectum, and pancreas. Emphasis is placed on critical oncogenic pathways, such as mutations in tumor suppressor genes, oncogenes, chromosomal instability, microsatellite instability, and epigenetic modifications. The role of molecular biomarkers in predicting prognosis, guiding treatment decisions, and monitoring therapeutic response is discussed, highlighting the integration of genomic profiling into clinical practice. Finally, we address the evolving landscape of precision oncology in GI cancers, considering targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Hannah E Trembath
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Jen Jen Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Nicole E Lopez
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA.
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA.
| |
Collapse
|
17
|
Tijhuis AE, Foijer F. Characterizing chromosomal instability-driven cancer evolution and cell fitness at a glance. J Cell Sci 2024; 137:jcs260199. [PMID: 38224461 DOI: 10.1242/jcs.260199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Chromosomal instability (CIN), an increased rate of chromosome segregation errors during mitosis, is a hallmark of cancer cells. CIN leads to karyotype differences between cells and thus large-scale heterogeneity among individual cancer cells; therefore, it plays an important role in cancer evolution. Studying CIN and its consequences is technically challenging, but various technologies have been developed to track karyotype dynamics during tumorigenesis, trace clonal lineages and link genomic changes to cancer phenotypes at single-cell resolution. These methods provide valuable insight not only into the role of CIN in cancer progression, but also into cancer cell fitness. In this Cell Science at a Glance article and the accompanying poster, we discuss the relationship between CIN, cancer cell fitness and evolution, and highlight techniques that can be used to study the relationship between these factors. To that end, we explore methods of assessing cancer cell fitness, particularly for chromosomally unstable cancer.
Collapse
Affiliation(s)
- Andréa E Tijhuis
- European Research Institute for the Biology of Ageing , University Medical Center Groningen, University of Groningen,9713 AV Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing , University Medical Center Groningen, University of Groningen,9713 AV Groningen, The Netherlands
| |
Collapse
|
18
|
Huo A, Xiong X. PAICS as a potential target for cancer therapy linking purine biosynthesis to cancer progression. Life Sci 2023; 331:122070. [PMID: 37673296 DOI: 10.1016/j.lfs.2023.122070] [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: 07/04/2023] [Revised: 09/02/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
Tumor cells are required to undergo metabolic reprogramming for rapid development and progression, and one of the metabolic characteristics of cancer cells is the excessive synthesis and utilization of nucleotides. Abnormally increased nucleotides and their metabolites not only directly accelerate tumor cell progression but also indirectly act on stromal cells in the tumor microenvironment (TME) via a paracrine manner to regulate tumor progression. Purine nucleotides are mainly produced via de novo nucleotide synthesis in tumor cells; therefore, intervening in their synthesis has emerged as a promising strategy in anti-tumor therapy. De novo purine synthesis is a 10-step reaction catalyzed by six enzymes to synthesize inosine 5-monophosphate (IMP) and subsequently synthesize AMP and GMP. Phosphoribosylaminoimidazole carboxylase/phosphori-bosylaminoimidazole succinocarboxamide synthetase (PAICS) is a bifunctional enzyme that catalyzes de novo purine synthesis. Aberrantly elevated PAICS expression in various tumors is associated with poor prognosis. Evidence suggests that PAICS and its catalytic product, N-succinylcarboxamide-5-aminoimidazole ribonucleotide (SAICAR), could inhibit tumor cell apoptosis and promote the growth, epithelial-mesenchymal transition (EMT), invasion, and metastasis by regulating signaling pathways such as pyruvate kinase M2 (PKM2), extracellular signal-related kinases 1 and 2 (ERK1/2), focal adhesion kinase (FAK) and so on. This review summarizes the structure, biological functions and the molecular mechanisms of PAICS in cancer development and discusses its potential to be a target for tumor therapy.
Collapse
Affiliation(s)
- Anqi Huo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, China; The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, China; Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
19
|
Ying Y, Zhang Z, Xing N, Qian Z, Wang B, Zeng S, Xu C. Preoperative urine sediment chromosomal instability level predicts urothelial cancer prognosis. Urol Oncol 2023; 41:433.e1-433.e7. [PMID: 37652824 DOI: 10.1016/j.urolonc.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/01/2023] [Accepted: 06/24/2023] [Indexed: 09/02/2023]
Abstract
PURPOSE Urothelial carcinomas (UCs) are often characterized by frequent recurrences after surgery, making UC one of the costliest cancers. Chromosomal instability (CIN) has been proven to be a hallmark of UCs and is related to the prognosis of many cancer types. In this study, we evaluated CIN of urine sediments as a prognostic indicator for UCs. METHODS Patients with UC were prospectively recruited. Preoperative urine samples were collected for whole genome sequencing and urine cytology tests. Patients underwent standard-of-care treatment and were followed up until disease relapse or study ended. Concordance and accuracy of CIN alone or in combination with cytology in predicting disease relapse were assessed. The value of CIN combined with European Organization for Research and Treatment of Cancer (EORTC) model were also analyzed. RESULTS A total of 137 patients with UCs were included in this study. Median follow-up was 44.2 months and 41.61% patients suffered from cancer relapse. Patients with CIN-high indicated higher relapse rate, and this distinction was significant for patients underwent transurethral resection of bladder tumor (57.89% vs. 34.29%, P = 0.016). Combination of cytology and CIN result could further classified patients into subgroups with distinct relapse risks. Meanwhile, the combination of CIN and EORTC model significantly improved the prediction accuracy compared with EORTC alone (Harrel's C-index: 0.71 vs. 0.65). CONCLUSION CIN level of preoperative urine exfoliated cells had robust prognostic value for bladder cancer patients underwent TURBT. The prognostic model by combining CIN and EORTC may help in stratifying patients to optimize follow-up regimen.
Collapse
Affiliation(s)
- Yidie Ying
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, P. R. China
| | - Zhensheng Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, P. R. China
| | - Naidong Xing
- Department of Urology, Qilu Hospital of Shandong University, Ji'nan, Shandong, P. R. China
| | - Ziliang Qian
- Suzhou Hongyuan Biotech Inc., Biobay, Suzhou, P. R. China
| | - Baiyun Wang
- Suzhou Hongyuan Biotech Inc., Biobay, Suzhou, P. R. China
| | - Shuxiong Zeng
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, P. R. China.
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, P. R. China.
| |
Collapse
|
20
|
Andrade JR, Gallagher AD, Maharaj J, McClelland SE. Disentangling the roles of aneuploidy, chromosomal instability and tumour heterogeneity in developing resistance to cancer therapies. Chromosome Res 2023; 31:28. [PMID: 37721639 PMCID: PMC10506951 DOI: 10.1007/s10577-023-09737-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/26/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
Aneuploidy is defined as the cellular state of having a number of chromosomes that deviates from a multiple of the normal haploid chromosome number of a given organism. Aneuploidy can be present in a static state: Down syndrome individuals stably maintain an extra copy of chromosome 21 in their cells. In cancer cells, however, aneuploidy is usually present in combination with chromosomal instability (CIN) which leads to a continual generation of new chromosomal alterations and the development of intratumour heterogeneity (ITH). The prevalence of cells with specific chromosomal alterations is further shaped by evolutionary selection, for example, during the administration of cancer therapies. Aneuploidy, CIN and ITH have each been individually associated with poor prognosis in cancer, and a wealth of evidence suggests they contribute, either alone or in combination, to cancer therapy resistance by providing a reservoir of potential resistant states, or the ability to rapidly evolve resistance. A full understanding of the contribution and interplay between aneuploidy, CIN and ITH is required to tackle therapy resistance in cancer patients. However, these characteristics often co-occur and are intrinsically linked, presenting a major challenge to defining their individual contributions. Moreover, their accurate measurement in both experimental and clinical settings is a technical hurdle. Here, we attempt to deconstruct the contribution of the individual and combined roles of aneuploidy, CIN and ITH to therapy resistance in cancer, and outline emerging approaches to measure and disentangle their roles as a step towards integrating these principles into cancer therapeutic strategy.
Collapse
Affiliation(s)
- Joana Reis Andrade
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M6BQ, England
| | - Annie Dinky Gallagher
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M6BQ, England
| | - Jovanna Maharaj
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M6BQ, England
| | | |
Collapse
|
21
|
van den Brink A, Suárez Peredo Rodríguez MF, Foijer F. Chromosomal instability and inflammation: a catch-22 for cancer cells. Chromosome Res 2023; 31:19. [PMID: 37561163 PMCID: PMC10415485 DOI: 10.1007/s10577-023-09730-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
Chromosomal instability (CIN), an increased rate of chromosomal segregation abnormalities, drives intratumor heterogeneity and affects most human cancers. In addition to chromosome copy number alterations, CIN results in chromosome(s) (fragments) being mislocalized into the cytoplasm in the form of micronuclei. Micronuclei can be detected by cGAS, a double-strand nucleic acid sensor, which will lead to the production of the second messenger 2'3'-cGAMP, activation of an inflammatory response, and downstream immune cell activation. However, the molecular network underlying the CIN-induced inflammatory response is still poorly understood. Furthermore, there is emerging evidence that cancers that display CIN circumvent this CIN-induced inflammatory response, and thus immune surveillance. The STAT1, STAT3, and NF-κB signaling cascades appear to play an important role in the CIN-induced inflammatory response. In this review, we discuss how these pathways are involved in signaling CIN in cells and how they are intertwined. A better understanding of how CIN is being signaled in cells and how cancer cells circumvent this is of the utmost importance for better and more selective cancer treatment.
Collapse
Affiliation(s)
- Anouk van den Brink
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Maria F Suárez Peredo Rodríguez
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands.
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands.
| |
Collapse
|
22
|
Kleo K, Jovanovic VM, Arndold A, Lehmann A, Lammert H, Berg E, Harloff H, Treese C, Hummel M, Daum S. Response prediction in patients with gastric and esophagogastric adenocarcinoma under neoadjuvant chemotherapy using targeted gene expression analysis and next-generation sequencing in pre-therapeutic biopsies. J Cancer Res Clin Oncol 2023; 149:1049-1061. [PMID: 35246724 PMCID: PMC9984352 DOI: 10.1007/s00432-022-03944-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/02/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Perioperative chemo-(radio-) therapy is the accepted standard in European patients with locally advanced adenocarcinoma of the esophagogastric junction or stomach (AEG/AS). However, 30-85% of patients do not respond to this treatment. The aim of our study was the identification of predictive biomarkers in pre-therapeutic endoscopic tumor biopsies from patients with histopathologic response (Becker-1) versus non-response (Becker-2/3) to preoperative chemotherapy. METHODS Formalin-fixed paraffin-embedded biopsies from 36 Caucasian patients (Becker-1 n = 11, Becker-2 n = 7, Becker-3 n = 18) with AEG/AS, taken prior to neoadjuvant chemotherapy were selected. For RNA expression analysis, we employed the NanoString nCounter System. To identify genomic alterations like single nucleotide variants (SNV), copy number variation (CNV) and fusion events, we used Illumina TST170 gene panel. For HER2 and FGFR2 protein expression, immunostaining was performed. Furthermore, we analyzed the microsatellite instability (MSI) and Epstein-Barr virus (EBV) infection status by EBER in situ hybridization. RESULTS Heat map and principal component analyses showed no clustering by means of gene expression according to regression grade. Concerning two recently proposed predictive markers, our data showed equal distribution for MSI (Becker-1: 2; Becker-2: 1; Becker-3: 3; out of 29 tested) and EBV infection was rare (1/32). We could not reveal discriminating target genes concerning SNV, but found a higher mutational burden in non-responders versus responders and fusion (in 6/14) and CNV events (in 5/14) exclusively in Becker-3. CONCLUSIONS Although we could not identify discriminating target genes, our data suggest that molecular alterations are in general more prevalent in patients with AEG/AS belonging to the non-responding Becker group 3.
Collapse
Affiliation(s)
- Karsten Kleo
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Vladimir M Jovanovic
- Institute of Informatics, Bioinformatics Solution Center, Freie Universität (FU), Takustr. 9, 14195, Berlin, Germany
| | - Alexander Arndold
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Annika Lehmann
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Hedwig Lammert
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Erika Berg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Hannah Harloff
- Medical Department, Division of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Christoph Treese
- Medical Department, Division of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
- Experimental and Clinical Research Center, Charité University Medicine, Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Chariteplatz 1, 10117, Berlin, Germany
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Michael Hummel
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Severin Daum
- Medical Department, Division of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany.
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
23
|
Wen Z, Wang S, Yang DM, Xie Y, Chen M, Bishop J, Xiao G. Deep learning in digital pathology for personalized treatment plans of cancer patients. Semin Diagn Pathol 2023; 40:109-119. [PMID: 36890029 DOI: 10.1053/j.semdp.2023.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Over the past decade, many new cancer treatments have been developed and made available to patients. However, in most cases, these treatments only benefit a specific subgroup of patients, making the selection of treatment for a specific patient an essential but challenging task for oncologists. Although some biomarkers were found to associate with treatment response, manual assessment is time-consuming and subjective. With the rapid developments and expanded implementation of artificial intelligence (AI) in digital pathology, many biomarkers can be quantified automatically from histopathology images. This approach allows for a more efficient and objective assessment of biomarkers, aiding oncologists in formulating personalized treatment plans for cancer patients. This review presents an overview and summary of the recent studies on biomarker quantification and treatment response prediction using hematoxylin-eosin (H&E) stained pathology images. These studies have shown that an AI-based digital pathology approach can be practical and will become increasingly important in improving the selection of cancer treatments for patients.
Collapse
Affiliation(s)
- Zhuoyu Wen
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shidan Wang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Donghan M Yang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yang Xie
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mingyi Chen
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Justin Bishop
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
24
|
Intra-tumor heterogeneity, turnover rate and karyotype space shape susceptibility to missegregation-induced extinction. PLoS Comput Biol 2023; 19:e1010815. [PMID: 36689467 PMCID: PMC9917311 DOI: 10.1371/journal.pcbi.1010815] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/10/2023] [Accepted: 12/12/2022] [Indexed: 01/24/2023] Open
Abstract
The phenotypic efficacy of somatic copy number alterations (SCNAs) stems from their incidence per base pair of the genome, which is orders of magnitudes greater than that of point mutations. One mitotic event stands out in its potential to significantly change a cell's SCNA burden-a chromosome missegregation. A stochastic model of chromosome mis-segregations has been previously developed to describe the evolution of SCNAs of a single chromosome type. Building upon this work, we derive a general deterministic framework for modeling missegregations of multiple chromosome types. The framework offers flexibility to model intra-tumor heterogeneity in the SCNAs of all chromosomes, as well as in missegregation- and turnover rates. The model can be used to test how selection acts upon coexisting karyotypes over hundreds of generations. We use the model to calculate missegregation-induced population extinction (MIE) curves, that separate viable from non-viable populations as a function of their turnover- and missegregation rates. Turnover- and missegregation rates estimated from scRNA-seq data are then compared to theoretical predictions. We find convergence of theoretical and empirical results in both the location of MIE curves and the necessary conditions for MIE. When a dependency of missegregation rate on karyotype is introduced, karyotypes associated with low missegregation rates act as a stabilizing refuge, rendering MIE impossible unless turnover rates are exceedingly high. Intra-tumor heterogeneity, including heterogeneity in missegregation rates, increases as tumors progress, rendering MIE unlikely.
Collapse
|
25
|
Castellanos G, Valbuena DS, Pérez E, Villegas VE, Rondón-Lagos M. Chromosomal Instability as Enabling Feature and Central Hallmark of Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:189-211. [PMID: 36923397 PMCID: PMC10010144 DOI: 10.2147/bctt.s383759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/11/2022] [Indexed: 03/11/2023]
Abstract
Chromosomal instability (CIN) has become a topic of great interest in recent years, not only for its implications in cancer diagnosis and prognosis but also for its role as an enabling feature and central hallmark of cancer. CIN describes cell-to-cell variation in the number or structure of chromosomes in a tumor population. Although extensive research in recent decades has identified some associations between CIN with response to therapy, specific associations with other hallmarks of cancer have not been fully evidenced. Such associations place CIN as an enabling feature of the other hallmarks of cancer and highlight the importance of deepening its knowledge to improve the outcome in cancer. In addition, studies conducted to date have shown paradoxical findings about the implications of CIN for therapeutic response, with some studies showing associations between high CIN and better therapeutic response, and others showing the opposite: associations between high CIN and therapeutic resistance. This evidences the complex relationships between CIN with the prognosis and response to treatment in cancer. Considering the above, this review focuses on recent studies on the role of CIN in cancer, the cellular mechanisms leading to CIN, its relationship with other hallmarks of cancer, and the emerging therapeutic approaches that are being developed to target such instability, with a primary focus on breast cancer. Further understanding of the complexity of CIN and its association with other hallmarks of cancer could provide a better understanding of the cellular and molecular mechanisms involved in prognosis and response to treatment in cancer and potentially lead to new drug targets.
Collapse
Affiliation(s)
- Giovanny Castellanos
- Maestría en Ciencias Biológicas, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia.,School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Duván Sebastián Valbuena
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Erika Pérez
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Victoria E Villegas
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| |
Collapse
|
26
|
Talaat IM, Elemam NM, Zaher S, Saber-Ayad M. Checkpoint molecules on infiltrating immune cells in colorectal tumor microenvironment. Front Med (Lausanne) 2022; 9:955599. [PMID: 36072957 PMCID: PMC9441912 DOI: 10.3389/fmed.2022.955599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancer types worldwide, with a high mortality rate due to metastasis. The tumor microenvironment (TME) contains multiple interactions between the tumor and the host, thus determining CRC initiation and progression. Various immune cells exist within the TME, such as tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and tumor-associated neutrophils (TANs). The immunotherapy approach provides novel opportunities to treat solid tumors, especially toward immune checkpoints. Despite the advances in the immunotherapy of CRC, there are still obstacles to successful treatment. In this review, we highlighted the role of these immune cells in CRC, with a particular emphasis on immune checkpoint molecules involved in CRC pathogenesis.
Collapse
Affiliation(s)
- Iman M. Talaat
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Noha M. Elemam
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Shroque Zaher
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Maha Saber-Ayad
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
27
|
Richardson TE, Walker JM, Abdullah KG, McBrayer SK, Viapiano MS, Mussa ZM, Tsankova NM, Snuderl M, Hatanpaa KJ. Chromosomal instability in adult-type diffuse gliomas. Acta Neuropathol Commun 2022; 10:115. [PMID: 35978439 PMCID: PMC9386991 DOI: 10.1186/s40478-022-01420-w] [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: 07/12/2022] [Accepted: 08/04/2022] [Indexed: 11/14/2022] Open
Abstract
Chromosomal instability (CIN) is a fundamental property of cancer and a key underlying mechanism of tumorigenesis and malignant progression, and has been documented in a wide variety of cancers, including colorectal carcinoma with mutations in genes such as APC. Recent reports have demonstrated that CIN, driven in part by mutations in genes maintaining overall genomic stability, is found in subsets of adult-type diffusely infiltrating gliomas of all histologic and molecular grades, with resulting elevated overall copy number burden, chromothripsis, and poor clinical outcome. Still, relatively few studies have examined the effect of this process, due in part to the difficulty of routinely measuring CIN clinically. Herein, we review the underlying mechanisms of CIN, the relationship between chromosomal instability and malignancy, the prognostic significance and treatment potential in various cancers, systemic disease, and more specifically, in diffusely infiltrating glioma subtypes. While still in the early stages of discovery compared to other solid tumor types in which CIN is a known driver of malignancy, the presence of CIN as an early factor in gliomas may in part explain the ability of these tumors to develop resistance to standard therapy, while also providing a potential molecular target for future therapies.
Collapse
Affiliation(s)
- Timothy E. Richardson
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, Annenberg Building, 15th Floor, 1468 Madison Avenue, New York, NY 10029 USA
| | - Jamie M. Walker
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, Annenberg Building, 15th Floor, 1468 Madison Avenue, New York, NY 10029 USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Kalil G. Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA 15213 USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA 15232 USA
| | - Samuel K. McBrayer
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Mariano S. Viapiano
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210 USA
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, NY 13210 USA
| | - Zarmeen M. Mussa
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, Annenberg Building, 15th Floor, 1468 Madison Avenue, New York, NY 10029 USA
| | - Nadejda M. Tsankova
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, Annenberg Building, 15th Floor, 1468 Madison Avenue, New York, NY 10029 USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health, New York City, NY 10016 USA
| | - Kimmo J. Hatanpaa
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| |
Collapse
|
28
|
Fatemi N, Tierling S, Es HA, Varkiani M, Nazemalhosseini Mojarad E, Asadzadeh Aghdaei H, Walter J, Totonchi M. DNA Methylation Biomarkers in Colorectal Cancer: Clinical Applications for Precision Medicine. Int J Cancer 2022; 151:2068-2081. [PMID: 35730647 DOI: 10.1002/ijc.34186] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/29/2022] [Accepted: 06/08/2022] [Indexed: 11/06/2022]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer death worldwide that is attributed to gradual long-term accumulation of both genetic and epigenetic changes. To reduce the mortality rate of CRC and to improve treatment efficacy, it will be important to develop accurate noninvasive diagnostic tests for screening, acute, and personalized diagnosis. Epigenetic changes such as DNA methylation play an important role in the development and progression of CRC. Over the last decade, a panel of DNA methylation markers has been reported showing a high accuracy and reproducibility in various semi-invasive or noninvasive biosamples. Research to obtain comprehensive panels of markers allowing a highly sensitive and differentiating diagnosis of CRC is ongoing. Moreover, the epigenetic alterations for cancer therapy, as a precision medicine strategy will increase their therapeutic potential over time. Here, we discuss the current state of DNA methylation-based biomarkers and their impact on CRC diagnosis. We emphasize the need to further identify and stratify methylation-biomarkers and to develop robust and effective detection methods that are applicable for a routine clinical setting of CRC diagnostics particularly at the early stage of the disease.
Collapse
Affiliation(s)
- Nayeralsadat Fatemi
- Basic & Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology & Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sascha Tierling
- Department of Genetics/Epigenetics, Faculty NT, Life Sciences, Saarland University, Saarbrücken, Germany
| | | | - Maryam Varkiani
- Department of Molecular Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Ehsan Nazemalhosseini Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic & Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology & Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jörn Walter
- Department of Genetics/Epigenetics, Faculty NT, Life Sciences, Saarland University, Saarbrücken, Germany
| | - Mehdi Totonchi
- Basic & Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology & Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
29
|
Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers (Basel) 2022; 14:cancers14041028. [PMID: 35205776 PMCID: PMC8869923 DOI: 10.3390/cancers14041028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Late-stage colorectal cancer treatment often involves chemotherapy and radiation that can cause dose-limiting toxicity, and therefore there is great interest in developing targeted therapies for this disease. Immunotherapy is a targeted therapy that uses peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer. Here, we discuss the preclinical and clinical development of immunotherapy for treatment of colorectal cancer and provide an overview of predictive biomarkers for such treatments. We also consider open questions including optimal combination treatments and sensitization of colorectal cancer patients with proficient mismatch repair enzymes. Abstract Though early-stage colorectal cancer has a high 5 year survival rate of 65–92% depending on the specific stage, this probability drops to 13% after the cancer metastasizes. Frontline treatments for colorectal cancer such as chemotherapy and radiation often produce dose-limiting toxicities in patients and acquired resistance in cancer cells. Additional targeted treatments are needed to improve patient outcomes and quality of life. Immunotherapy involves treatment with peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer cells. Preclinical and clinical investigations of immunotherapy for treatment of colorectal cancer including immune checkpoint blockade, adoptive cell therapy, monoclonal antibodies, oncolytic viruses, anti-cancer vaccines, and immune system modulators have been promising, but demonstrate limitations for patients with proficient mismatch repair enzymes. In this review, we discuss preclinical and clinical studies investigating immunotherapy for treatment of colorectal cancer and predictive biomarkers for response to these treatments. We also consider open questions including optimal combination treatments to maximize efficacy, minimize toxicity, and prevent acquired resistance and approaches to sensitize mismatch repair-proficient patients to immunotherapy.
Collapse
|
30
|
Kohlruss M, Krenauer M, Grosser B, Pfarr N, Jesinghaus M, Slotta-Huspenina J, Novotny A, Hapfelmeier A, Schmidt T, Steiger K, Gaida MM, Reiche M, Bauer L, Ott K, Weichert W, Keller G. Diverse 'just-right' levels of chromosomal instability and their clinical implications in neoadjuvant treated gastric cancer. Br J Cancer 2021; 125:1621-1631. [PMID: 34671125 PMCID: PMC8651679 DOI: 10.1038/s41416-021-01587-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The Cancer Genome Atlas (TCGA) consortium described EBV positivity(+), high microsatellite instability (MSI-H), genomic stability (GS) and chromosomal instability (CIN) as molecular subtypes in gastric carcinomas (GC). We investigated the predictive and prognostic value of these subtypes with emphasis on CIN in the context of neoadjuvant chemotherapy (CTx) in GC. METHODS TCGA subgroups were determined for 612 resected adenocarcinomas of the stomach and gastro-oesophageal junction (291 without, 321 with CTx) and 143 biopsies before CTx. EBV and MSI-H were analysed by standard assays. CIN was detected by multiplex PCRs analysing 22 microsatellite markers. Besides the TCGA classification, CIN was divided into four CIN-subgroups: low, moderate, substantial, high. Mutation profiling was performed for 52 tumours by next-generation sequencing. RESULTS EBV(+) (HR, 0.48; 95% CI, 0.23-1.02), MSI-H (HR, 0.56; 95% CI, 0.35-0.89) and GS (HR, 0.72; 95% CI, 0.45-1.13) were associated with increased survival compared to CIN in the resected tumours. Considering the extended CIN-classification, CIN-substantial was a negative prognostic factor in uni- and multivariable analysis in resected tumours with CTx (each p < 0.05). In biopsies before CTx, CIN-high predicted tumour regression (p = 0.026), but was not prognostically relevant. CONCLUSION A refined CIN classification reveals tumours with different biological characteristics and potential clinical implications.
Collapse
Affiliation(s)
- Meike Kohlruss
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Marie Krenauer
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Bianca Grosser
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,grid.419801.50000 0000 9312 0220Institute of Pathology and Molecular Diagnostics, University Hospital Augsburg, Augsburg, Germany
| | - Nicole Pfarr
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Moritz Jesinghaus
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,grid.411067.50000 0000 8584 9230Institute of Pathology, University Hospital Marburg, Marburg, Germany
| | - Julia Slotta-Huspenina
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Novotny
- grid.6936.a0000000123222966Department of Surgery, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Hapfelmeier
- grid.6936.a0000000123222966Institute of Medical Informatics, Statistics and Epidemiology, Technical University of Munich, Munich, Germany ,grid.6936.a0000000123222966Institute of General Practice and Health Services Research, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Schmidt
- grid.7700.00000 0001 2190 4373Department of Surgery, University of Heidelberg, Heidelberg, Germany ,grid.411097.a0000 0000 8852 305XDepartment of Surgery, Universitätsklinikum Köln, Köln, Germany
| | - Katja Steiger
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,German Cancer Consortium (DKTK), Partner Site Munich, Institute of Pathology, Munich, Germany
| | - Matthias M. Gaida
- grid.7700.00000 0001 2190 4373Institute of Pathology, University of Heidelberg, Heidelberg, Germany ,grid.410607.4Institute of Pathology, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | - Magdalena Reiche
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Lukas Bauer
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Ott
- grid.477776.20000 0004 0394 5800Department of Surgery, RoMed Klinikum Rosenheim, Rosenheim, Germany
| | - Wilko Weichert
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,German Cancer Consortium (DKTK), Partner Site Munich, Institute of Pathology, Munich, Germany
| | - Gisela Keller
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
31
|
Wills C, He Y, Summers MG, Lin Y, Phipps AI, Watts K, Law PJ, Al-Tassan NA, Maughan TS, Kaplan R, Houlston RS, Peters U, Newcomb PA, Chan AT, Buchanan DD, Gallinger S, Marchand LL, Pai RK, Shi Q, Alberts SR, Gray V, West HD, Escott-Price V, Dunlop MG, Cheadle JP. A genome-wide search for determinants of survival in 1926 patients with advanced colorectal cancer with follow-up in over 22,000 patients. Eur J Cancer 2021; 159:247-258. [PMID: 34794066 PMCID: PMC9132154 DOI: 10.1016/j.ejca.2021.09.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND While genome-wide association studies (GWAS) have identified germline variants influencing the risk of developing colorectal cancer (CRC), there has been limited examination of the possible role of inherited variation as a determinant of patient outcome. PATIENTS AND METHODS We performed a GWAS for overall survival (OS) in 1926 patients with advanced CRC from the COIN and COIN-B clinical trials. For single nucleotide polymorphisms (SNPs) showing an association with OS (P < 1.0 × 10-5), we conducted sensitivity analyses based on the time from diagnosis to death and sought independent replications in 5675 patients from the Study of Colorectal Cancer in Scotland (SOCCS) and 16,964 patients from the International Survival Analysis in Colorectal cancer Consortium (ISACC). We analysed the Human Protein Atlas to determine if ERBB4 expression was associated with survival in 438 patients with colon adenocarcinomas. RESULTS The most significant SNP associated with OS was rs79612564 in ERBB4 (hazard ratio [HR] = 1.24, 95% confidence interval [CI] = 1.16-1.32, P = 1.9 × 10-7). SNPs at 17 loci had suggestive associations for OS and all had similar effects on the time from diagnosis to death. No lead SNPs were independently replicated in the meta-analysis of all patients from SOCCS and ISACC. However, rs79612564 was significant in stage-IV patients from SOCCS (P = 2.1 × 10-2) but not ISACC (P = 0.89) and SOCCS combined with COIN and COIN-B attained genome-wide significance (P = 1.7 × 10-8). Patients with high ERBB4 expression in their colon adenocarcinomas had worse survival (HR = 1.50, 95% CI = 1.1-1.9, P = 4.6 × 10-2). CONCLUSIONS Genetic and expression data support a potential role for rs79612564 in the receptor tyrosine kinase ERBB4 as a predictive biomarker of survival.
Collapse
Affiliation(s)
- Christopher Wills
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Yazhou He
- Colon Cancer Genetics Group, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK; Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, China
| | - Matthew G Summers
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Yi Lin
- Epidemiology Department, University of Washington, Seattle, WA, USA; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Amanda I Phipps
- Epidemiology Department, University of Washington, Seattle, WA, USA; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Katie Watts
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Nada A Al-Tassan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Timothy S Maughan
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Richard Kaplan
- MRC Clinical Trials Unit, University College of London, 125 Kingsway, London, WC2B 6NH, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Ulrike Peters
- Epidemiology Department, University of Washington, Seattle, WA, USA; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Polly A Newcomb
- Epidemiology Department, University of Washington, Seattle, WA, USA; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia; Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Steve Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Loic L Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Qian Shi
- Department of Quantitative Science, Mayo Clinic, Rochester, MN, USA
| | | | - Victoria Gray
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Hannah D West
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Valentina Escott-Price
- Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Malcolm G Dunlop
- Colon Cancer Genetics Group, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Jeremy P Cheadle
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
32
|
Peng J, Li W, Fan W, Zhang R, Li X, Xiao B, Dong Y, Wan D, Pan Z, Lin J, Wu X. Prognostic value of a novel biomarker combining DNA ploidy and tumor burden score for initially resectable liver metastases from patients with colorectal cancer. Cancer Cell Int 2021; 21:554. [PMID: 34688293 PMCID: PMC8542290 DOI: 10.1186/s12935-021-02250-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/07/2021] [Indexed: 11/23/2022] Open
Abstract
Background Colorectal cancer liver metastases (CRLM) has not been identified as a unified disease entity due to the differences in the severity of metastatic disease and tumor aggressiveness. A screen for specific prognostic risk subgroups is urgently needed. The current study aimed to investigate the prognostic value of DNA ploidy, stroma fraction and nucleotyping of initially resectable liver metastases from patients with CRLM. Methods One hundred thirty-nine consecutive patients with initially resectable CRLM who underwent curative liver resection from 2006 to 2018 at Sun Yat-sen University Cancer Center were selected for analysis. DNA ploidy, stroma fraction and nucleotyping of liver metastases were evaluated using automated digital imaging systems. Recurrence-free survival (RFS) and overall survival (OS) were analyzed using the Kaplan-Meier method and Cox regression models. Results DNA ploidy was identified as an independent prognostic factor for RFS (HR, 2.082; 95% CI 1.053–4.115; P = 0.035) in the multivariate analysis, while stroma-tumor fraction and nucleotyping were not significant prognostic factors. A significant difference in 3-year RFS was observed among the low-, moderate- and high-risk groups stratified by a novel parameter combined with the tumor burden score (TBS) and DNA ploidy (72.5% vs. 63.2% vs. 37.3%, P = 0.007). The high-risk group who received adjuvant chemotherapy had a significantly better 3-year RFS rate than those without adjuvant chemotherapy (46.7% vs. 24.8%; P = 0.034). Conclusions Our study showed that DNA ploidy of liver metastases is an independent prognostic factor for patients with initially resectable CRLM after liver resection. The combination of DNA ploidy and TBS may help to stratify patients into different recurrence risk groups and may guide postoperative treatment among the patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02250-x.
Collapse
Affiliation(s)
- Jianhong Peng
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Weihao Li
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Wenhua Fan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Rongxin Zhang
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Xinyue Li
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Binyi Xiao
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Yuejin Dong
- NingBo Meishan FTZ MBM Clinical Lab Co., Ltd, Ningbo, 315832, Zhejiang, P. R. China
| | - Desen Wan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Zhizhong Pan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
| | - Junzhong Lin
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
| | - Xiaojun Wu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
| |
Collapse
|
33
|
Sharma S, George P, Waddell N. Precision diagnostics: Integration of tissue pathology and genomics in cancer. Pathology 2021; 53:809-817. [PMID: 34635323 DOI: 10.1016/j.pathol.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/09/2022]
Abstract
Traditionally, cancer diagnosis and management has been reactionary in that symptoms lead to investigations, then a diagnosis is followed by clinical management. This process is heavily dependent on tissue diagnosis mainly by histopathology and to a lesser extent, cytopathology. However, in recent times there has been a shift towards precision medicine to enable prevention, prediction and personalisation in healthcare. The core of precision medicine is optimising therapeutic benefit for patients, by using genomic and molecular profiling, analogously termed precision pathology. This review explores (1) the evolution of pathology from a para-clinical discipline to a mainstream medical field integral to oncology tumour boards; (2) its critical role in preventative, diagnostic, therapeutic and follow-up cancer care; (3) the future of tissue pathology in the era of precision oncology; and (4) how pathologists may evolve to future-proof their profession.
Collapse
Affiliation(s)
- Sowmya Sharma
- Medlab Pathology, Auburn, NSW, Australia; QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, Qld, Australia; Faculty of Medicine, University of Queensland, Brisbane, Qld, Australia.
| | - Peter George
- Medlab Pathology, Auburn, NSW, Australia; genomiQa, Brisbane, Qld, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, Qld, Australia; Faculty of Medicine, University of Queensland, Brisbane, Qld, Australia; genomiQa, Brisbane, Qld, Australia
| |
Collapse
|
34
|
Genomic instability in lower-grade glioma: Prediction of prognosis based on lncRNA and immune infiltration. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:431-443. [PMID: 34553030 PMCID: PMC8430277 DOI: 10.1016/j.omto.2021.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/17/2021] [Indexed: 12/11/2022]
Abstract
Glioma is the most common type of primary malignant tumor in the central nervous system. Tumor recurrence and progression are common in lower-grade glioma (LGG). Immune checkpoint blockade (ICB), as an emerging immunotherapy, is expected to improve the prognosis of patients undergoing conventional treatment, but it currently performs poorly in glioma. We divided patients into genome-stable and -unstable groups according to the somatic mutation count and then found that the expression of CDC20 was positively correlated with genomic instability. We compared the differences in long non-coding RNA (lncRNA) expression and immune infiltration between the two groups. Five lncRNAs and three immune cell types were identified to construct risk models and a nomogram combing clinical features. Through internal and external validation, the models exhibited sufficient ability to predict the prognosis and the possible response to ICB therapy of patients. This study provided a potential predictive approach for the precise application of ICB and support for improving the prognosis of LGG patients.
Collapse
|
35
|
Patil S, Jahagirdar S, Khot M, Sengupta K. Studying the Role of Chromosomal Instability (CIN) in GI Cancers Using Patient-derived Organoids. J Mol Biol 2021; 434:167256. [PMID: 34547328 DOI: 10.1016/j.jmb.2021.167256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/28/2021] [Accepted: 09/13/2021] [Indexed: 01/10/2023]
Abstract
Chromosomal instability (CIN) is associated with the initiation and progression of gastrointestinal (GI) tract cancers. Cancers of the GI tract are typically characterized by altered chromosome numbers. While the dynamics of CIN have been extensively characterized in 2D monolayer cell cultures derived from GI tumors, the tumor microenvironment and 3D tumor architecture also contribute to the progression of CIN, which is not captured in 2D cell culture systems. To overcome these limitations, self-organizing cellular structures that retain organ-specific 3D architecture, namely organoids, have been derived from various tissues of the GI tract. Organoids derived from normal tissue and patient tumors serve as a useful paradigm to study the crosstalk between tumor cells in the context of a tissue microenvironment and its impact on chromosomal stability. Such a paradigm, therefore, has a considerable advantage over 2D cell culture systems in drug screening and personalized medicine. Here, we review the importance of patient-derived tumor organoids (PDTOs) as a model to study CIN in cancers of the GI tract.
Collapse
Affiliation(s)
- Shalaka Patil
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Pune 411008, India. https://twitter.com/@ShalakaPatil11
| | - Sanika Jahagirdar
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Pune 411008, India. https://twitter.com/@SanikaJag
| | - Maithilee Khot
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Pune 411008, India. https://twitter.com/@MaithileeKhot
| | - Kundan Sengupta
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Pune 411008, India.
| |
Collapse
|
36
|
Wang XF, Yang SA, Gong S, Chang CH, Portilla JM, Chatterjee D, Irianto J, Bao H, Huang YC, Deng WM. Polyploid mitosis and depolyploidization promote chromosomal instability and tumor progression in a Notch-induced tumor model. Dev Cell 2021; 56:1976-1988.e4. [PMID: 34146466 DOI: 10.1016/j.devcel.2021.05.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/18/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Ploidy variation is a cancer hallmark and is frequently associated with poor prognosis in high-grade cancers. Using a Drosophila solid-tumor model where oncogenic Notch drives tumorigenesis in a transition-zone microenvironment in the salivary gland imaginal ring, we find that the tumor-initiating cells normally undergo endoreplication to become polyploid. Upregulation of Notch signaling, however, induces these polyploid transition-zone cells to re-enter mitosis and undergo tumorigenesis. Growth and progression of the transition-zone tumor are fueled by a combination of polyploid mitosis, endoreplication, and depolyploidization. Both polyploid mitosis and depolyploidization are error prone, resulting in chromosomal copy-number variation and polyaneuploidy. Comparative RNA-seq and epistasis analysis reveal that the DNA-damage response genes, also active during meiosis, are upregulated in these tumors and are required for the ploidy-reduction division. Together, these findings suggest that polyploidy and associated cell-cycle variants are critical for increased tumor-cell heterogeneity and genome instability during cancer progression.
Collapse
Affiliation(s)
- Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Sheng-An Yang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Shangyu Gong
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Chih-Hsuan Chang
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Juan Martin Portilla
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Deeptiman Chatterjee
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Hongcun Bao
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA; Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
37
|
Comprehensive Analysis of Differentially Expressed Long Noncoding RNA-mRNA in the Adenoma-Carcinoma Sequence of DNA Mismatch Repair Proficient Colon Cancer. JOURNAL OF ONCOLOGY 2021; 2021:9977695. [PMID: 34211553 PMCID: PMC8208869 DOI: 10.1155/2021/9977695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/08/2021] [Indexed: 12/24/2022]
Abstract
DNA proficient mismatch repair colon cancer (pMMR CC) is the most common subtype of sporadic CC. We aimed to investigate the role of long noncoding RNAs (lncRNAs) in pMMR CC carcinogenesis. In the present study, we conducted transcriptomic analysis of lncRNAs-mRNAs in five low-grade intraepithelial neoplasia (LGIN), five high-grade intraepithelial neoplasia (HGIN), four pMMR CC, and five normal control (NC) tissues. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathway, and coexpression network analyses were performed to elucidate the functions of lncRNAs and mRNAs as well as their interactions. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to validate five dysregulated lncRNAs in a large set of colon tissues. Receiver-operating characteristic (ROC) curves were employed to evaluate the performance of the candidate lncRNAs. A set of 5783 differentially expressed lncRNAs and 4483 differentially expressed mRNAs were detected among the LGIN, HGIN, pMMR CC, and NC samples. These differentially expressed lncRNAs and mRNAs were assigned to 275 significant GO terms and 179 significant KEGG enriched pathways. qRT-PCR confirmed that the expression of five selected lncRNAs (ENST00000521815, ENST00000603052, ENST00000609220, NR_026543, and ENST00000545920) were consistent with the microarray data. ROC analysis showed that four lncRNAs (ENST00000521815, ENST00000603052, ENST00000609220, and NR_026543) had larger area under the ROC curve (AUC) values compared to serum carcinoembryonic antigens, thereby distinguishing NC from pMMR CC. In conclusion, several lncRNAs play various roles in the adenoma-carcinoma sequence and may serve as potential biomarkers for the early diagnosis of pMMR CC.
Collapse
|
38
|
Rao CV, Xu C, Farooqui M, Zhang Y, Asch AS, Yamada HY. Survival-Critical Genes Associated with Copy Number Alterations in Lung Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13112586. [PMID: 34070461 PMCID: PMC8197496 DOI: 10.3390/cancers13112586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/19/2022] Open
Abstract
Chromosome Instability (CIN) in tumors affects carcinogenesis, drug resistance, and recurrence/prognosis. Thus, it has a high impact on outcomes in clinic. However, how CIN occurs in human tumors remains elusive. Although cells with CIN (i.e., pre/early cancer cells) are proposed to be removed by apoptosis and/or a surveillance mechanism, this surveillance mechanism is poorly understood. Here we employed a novel data-mining strategy (Gene Expression to Copy Number Alterations [CNA]; "GE-CNA") to comprehensively identify 1578 genes that associate with CIN, indicated by genomic CNA as its surrogate marker, in human lung adenocarcinoma. We found that (a) amplification/insertion CNA is facilitated by over-expressions of DNA replication stressor and suppressed by a broad range of immune cells (T-, B-, NK-cells, leukocytes), and (b) deletion CNA is facilitated by over-expressions of mitotic regulator genes and suppressed predominantly by leukocytes guided by leukocyte extravasation signaling. Among the 39 CNA- and survival-associated genes, the purine metabolism (PPAT, PAICS), immune-regulating CD4-LCK-MEC2C and CCL14-CCR1 axes, and ALOX5 emerged as survival-critical pathways. These findings revealed a broad role of the immune system in suppressing CIN/CNA and cancer development in lung, and identified components representing potential targets for future chemotherapy, chemoprevention, and immunomodulation approaches for lung adenocarcinoma.
Collapse
Affiliation(s)
- Chinthalapally V. Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA;
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA;
- VA Medical Center, Oklahoma City, OK 73104, USA
- Correspondence: (C.V.R.); (H.Y.Y.)
| | - Chao Xu
- Hudson College of Public Health, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA;
| | - Mudassir Farooqui
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Yuting Zhang
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA;
| | - Adam S. Asch
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA;
| | - Hiroshi Y. Yamada
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA;
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA;
- Correspondence: (C.V.R.); (H.Y.Y.)
| |
Collapse
|
39
|
Xu Z, Verma A, Naveed U, Bakhoum SF, Khosravi P, Elemento O. Deep learning predicts chromosomal instability from histopathology images. iScience 2021; 24:102394. [PMID: 33997679 PMCID: PMC8099498 DOI: 10.1016/j.isci.2021.102394] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/03/2021] [Accepted: 04/01/2021] [Indexed: 12/13/2022] Open
Abstract
Chromosomal instability (CIN) is a hallmark of human cancer yet not readily testable for patients with cancer in routine clinical setting. In this study, we sought to explore whether CIN status can be predicted using ubiquitously available hematoxylin and eosin histology through a deep learning-based model. When applied to a cohort of 1,010 patients with breast cancer (Training set: n = 858, Test set: n = 152) from The Cancer Genome Atlas where 485 patients have high CIN status, our model accurately classified CIN status, achieving an area under the curve of 0.822 with 81.2% sensitivity and 68.7% specificity in the test set. Patch-level predictions of CIN status suggested intra-tumor heterogeneity within slides. Moreover, presence of patches with high predicted CIN score within an entire slide was more predictive of clinical outcome than the average CIN score of the slide, thus underscoring the clinical importance of intra-tumor heterogeneity.
Collapse
Affiliation(s)
- Zhuoran Xu
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York 10065, USA
- Pathology and Laboratory Medicine, Weill Cornell Medicine, New York 10065, USA
| | - Akanksha Verma
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York 10065, USA
| | - Uska Naveed
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York 10065, USA
| | - Samuel F. Bakhoum
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York 10021, USA
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York 10021, USA
| | - Pegah Khosravi
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York 10065, USA
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York 10021, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York 10065, USA
| |
Collapse
|
40
|
Baudoin NC, Bloomfield M. Karyotype Aberrations in Action: The Evolution of Cancer Genomes and the Tumor Microenvironment. Genes (Basel) 2021; 12:558. [PMID: 33921421 PMCID: PMC8068843 DOI: 10.3390/genes12040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/27/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is a disease of cellular evolution. For this cellular evolution to take place, a population of cells must contain functional heterogeneity and an assessment of this heterogeneity in the form of natural selection. Cancer cells from advanced malignancies are genomically and functionally very different compared to the healthy cells from which they evolved. Genomic alterations include aneuploidy (numerical and structural changes in chromosome content) and polyploidy (e.g., whole genome doubling), which can have considerable effects on cell physiology and phenotype. Likewise, conditions in the tumor microenvironment are spatially heterogeneous and vastly different than in healthy tissues, resulting in a number of environmental niches that play important roles in driving the evolution of tumor cells. While a number of studies have documented abnormal conditions of the tumor microenvironment and the cellular consequences of aneuploidy and polyploidy, a thorough overview of the interplay between karyotypically abnormal cells and the tissue and tumor microenvironments is not available. Here, we examine the evidence for how this interaction may unfold during tumor evolution. We describe a bidirectional interplay in which aneuploid and polyploid cells alter and shape the microenvironment in which they and their progeny reside; in turn, this microenvironment modulates the rate of genesis for new karyotype aberrations and selects for cells that are most fit under a given condition. We conclude by discussing the importance of this interaction for tumor evolution and the possibility of leveraging our understanding of this interplay for cancer therapy.
Collapse
Affiliation(s)
- Nicolaas C. Baudoin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
41
|
Monteverde T, Sahoo S, La Montagna M, Magee P, Shi L, Lee D, Sellers R, Baker AR, Leong HS, Fassan M, Garofalo M. CKAP2L Promotes Non-Small Cell Lung Cancer Progression through Regulation of Transcription Elongation. Cancer Res 2021; 81:1719-1731. [PMID: 33472893 DOI: 10.1158/0008-5472.can-20-1968] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 12/04/2020] [Accepted: 01/12/2021] [Indexed: 12/24/2022]
Abstract
Chromosomal instability (CIN) is a driver of clonal diversification and intratumor heterogeneity, providing genetic diversity that contributes to tumor progression. It is estimated that approximately 80% of solid cancers, including non-small cell lung cancer (NSCLC), exhibit features of CIN, which affects tumor growth and response to therapy. However, the molecular mechanisms connecting CIN to tumor progression are still poorly understood. Through an RNAi screen performed on genes involved in CIN and overexpressed in human lung adenocarcinoma samples, we identified the cytoskeleton-associated protein 2-like (CKAP2L) as a potential oncogene that promotes lung cancer proliferation and growth in vitro and in vivo. Mechanistically, CKAP2L directly interacted with RNA Pol II and regulated transcription elongation of key genes involved in spindle assembly checkpoint, chromosome segregation, cell cycle, and E2F signaling. Furthermore, depletion of CKAP2L increased the sensitivity of NSCLC cells to alvocidib, a pan-CDK inhibitor, leading to a significant reduction of cell proliferation and an increase in cell death. Altogether, these findings shed light on the molecular mechanisms through which CKAP2L, a protein involved in CIN, promotes cancer progression and suggest that its inhibition represents a novel therapeutic strategy in NSCLC. SIGNIFICANCE: These findings demonstrate the oncogenic function of CKAP2L through regulation of transcription elongation and suggest that targeting CKAP2L could enhance therapeutic response in patients with NSCLC.
Collapse
Affiliation(s)
- Tiziana Monteverde
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence at Manchester and University College London, United Kingdom
| | - Sudhakar Sahoo
- Computational Biology Support, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Manuela La Montagna
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence at Manchester and University College London, United Kingdom
| | - Peter Magee
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence at Manchester and University College London, United Kingdom
| | - Lei Shi
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence at Manchester and University College London, United Kingdom
| | - Dave Lee
- Computational Biology Support, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Robert Sellers
- Computational Biology Support, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Alexander R Baker
- Visualisation, Irradiation & Analysis, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Hui Sun Leong
- Computational Biology Support, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy
| | - Michela Garofalo
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom.
- Cancer Research UK Lung Cancer Centre of Excellence at Manchester and University College London, United Kingdom
| |
Collapse
|
42
|
Zhao JM, Qi TG. The role of TXNL1 in disease: treatment strategies for cancer and diseases with oxidative stress. Mol Biol Rep 2021; 48:2929-2934. [PMID: 33660093 DOI: 10.1007/s11033-021-06241-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 02/18/2021] [Indexed: 12/17/2022]
Abstract
Thioredoxin-like protein-1 (TXNL1; also known as thioredoxin-related 32 kDa protein, TRP32) is a thioredoxin involved in the regulation of oxidative stress, which protects cells from damage through redox balance. Studies have shown that TXNL1 has a variety of functions, including cell signal transduction, cell cycle regulation, protein synthesis, modification and degradation, vesicle transport, transcriptional regulation, cell apoptosis, virus replication and oxidative stress regulation, etc., and plays an important role in the occurrence and development of human diseases. Therefore, TXNL1 has a strong correlation with the treatment of cancer and oxidative stress diseases. In this paper, the basic structure, function and potential application value of TXNL1 in diseases are reviewed, so as to open up new targets for the treatment of cancer and oxidative stress-related diseases.
Collapse
Affiliation(s)
- Jin-Ming Zhao
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Tong-Gang Qi
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| |
Collapse
|
43
|
Jeusset LM, Guppy BJ, Lichtensztejn Z, McDonald D, McManus KJ. Reduced USP22 Expression Impairs Mitotic Removal of H2B Monoubiquitination, Alters Chromatin Compaction and Induces Chromosome Instability That May Promote Oncogenesis. Cancers (Basel) 2021; 13:cancers13051043. [PMID: 33801331 PMCID: PMC7958346 DOI: 10.3390/cancers13051043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Chromosome instability (CIN) is an enabling feature of oncogenesis associated with poor patient outcomes, whose genetic determinants remain largely unknown. As mitotic chromatin compaction defects can compromise the accuracy of chromosome segregation into daughter cells and drive CIN, characterizing the molecular mechanisms ensuring accurate chromatin compaction may identify novel CIN genes. In vitro, histone H2B monoubiquitination at lysine 120 (H2Bub1) impairs chromatin compaction, while in vivo H2Bub1 is rapidly depleted from chromatin upon entry into mitosis, suggesting that H2Bub1 removal may be a pre-requisite for mitotic fidelity. The deubiquitinating enzyme USP22 catalyzes H2Bub1 removal in interphase and may also be required for H2Bub1 removal in early mitosis to maintain chromosome stability. In this study, we demonstrate that siRNA-mediated USP22 depletion increases H2Bub1 levels in early mitosis and induces CIN phenotypes associated with mitotic chromatin compaction defects revealed by super-resolution microscopy. Moreover, USP22-knockout models exhibit continuously changing chromosome complements over time. These data identify mitotic removal of H2Bub1 as a critical determinant of chromatin compaction and faithful chromosome segregation. We further demonstrate that USP22 is a CIN gene, indicating that USP22 deletions, which are frequent in many tumor types, may drive genetic heterogeneity and contribute to cancer pathogenesis.
Collapse
Affiliation(s)
- Lucile M. Jeusset
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E0J9, Canada
| | - Brent J. Guppy
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E0J9, Canada
| | - Zelda Lichtensztejn
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
| | - Darin McDonald
- Department of Oncology, University of Alberta, Edmonton, AB T6G2H7, Canada;
| | - Kirk J. McManus
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E0J9, Canada
- Correspondence: ; Tel.: +1-(204)-787-2833
| |
Collapse
|
44
|
Badic B, Tixier F, Cheze Le Rest C, Hatt M, Visvikis D. Radiogenomics in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13050973. [PMID: 33652647 PMCID: PMC7956421 DOI: 10.3390/cancers13050973] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/07/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal carcinoma is characterized by intratumoral heterogeneity that can be assessed by radiogenomics. Radiomics, high-throughput quantitative data extracted from medical imaging, combined with molecular analysis, through genomic and transcriptomic data, is expected to lead to significant advances in personalized medicine. However, a radiogenomics approach in colorectal cancer is still in its early stages and many problems remain to be solved. Here we review the progress and challenges in this field at its current stage, as well as future developments. Abstract The steady improvement of high-throughput technologies greatly facilitates the implementation of personalized precision medicine. Characterization of tumor heterogeneity through image-derived features—radiomics and genetic profile modifications—genomics, is a rapidly evolving field known as radiogenomics. Various radiogenomics studies have been dedicated to colorectal cancer so far, highlighting the potential of these approaches to enhance clinical decision-making. In this review, a general outline of colorectal radiogenomics literature is provided, discussing the current limitations and suggested further developments.
Collapse
Affiliation(s)
- Bogdan Badic
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Correspondence: ; Tel.: +33-298-347-215
| | - Florent Tixier
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Catherine Cheze Le Rest
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Department of Nuclear Medicine, University Hospital of Poitiers, 86021 Poitiers, France
| | - Mathieu Hatt
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Dimitris Visvikis
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| |
Collapse
|
45
|
Novel Genetic and Epigenetic Biomarkers of Prognostic and Predictive Significance in Stage II/III Colorectal Cancer. Mol Ther 2020; 29:587-596. [PMID: 33333293 DOI: 10.1016/j.ymthe.2020.12.017] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/15/2020] [Accepted: 12/09/2020] [Indexed: 12/18/2022] Open
Abstract
The therapeutic strategies of stage II/III colorectal cancer (CRC) patients after curative surgery remain controversial. In the clinical decision-making process, oncologists need to answer questions such as whether adjuvant chemotherapy is necessary or which therapeutic regimen should be given to each patient. At present, whether adjuvant chemotherapy should be applied is primarily based on histopathological features and clinical risk factors. However, only a fraction of patients can benefit from it. More rigorous stratifying biomarkers are urgently needed to help further distinguishing these populations of patients. Recent progress in next-generation sequencing and high-throughput technologies has greatly promoted biomarker discovery as well as our understanding of the underlying mechanisms in CRC. Novel genetic and epigenetic biomarkers that are associated with prognosis or therapeutic responses have emerged. In this review, we discuss the strategies of biomarker discovery and summarize the status and assess the utility of previously published biomarkers in CRC.
Collapse
|
46
|
Zhou J, Zhou XA, Zhang N, Wang J. Evolving insights: how DNA repair pathways impact cancer evolution. Cancer Biol Med 2020; 17:805-827. [PMID: 33299637 PMCID: PMC7721097 DOI: 10.20892/j.issn.2095-3941.2020.0177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Abstract
Viewing cancer as a large, evolving population of heterogeneous cells is a common perspective. Because genomic instability is one of the fundamental features of cancer, this intrinsic tendency of genomic variation leads to striking intratumor heterogeneity and functions during the process of cancer formation, development, metastasis, and relapse. With the increased mutation rate and abundant diversity of the gene pool, this heterogeneity leads to cancer evolution, which is the major obstacle in the clinical treatment of cancer. Cells rely on the integrity of DNA repair machineries to maintain genomic stability, but these machineries often do not function properly in cancer cells. The deficiency of DNA repair could contribute to the generation of cancer genomic instability, and ultimately promote cancer evolution. With the rapid advance of new technologies, such as single-cell sequencing in recent years, we have the opportunity to better understand the specific processes and mechanisms of cancer evolution, and its relationship with DNA repair. Here, we review recent findings on how DNA repair affects cancer evolution, and discuss how these mechanisms provide the basis for critical clinical challenges and therapeutic applications.
Collapse
Affiliation(s)
- Jiadong Zhou
- Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xiao Albert Zhou
- Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ning Zhang
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.,Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China
| | - Jiadong Wang
- Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
47
|
Vargas-Rondón N, Pérez-Mora E, Villegas VE, Rondón-Lagos M. Role of chromosomal instability and clonal heterogeneity in the therapy response of breast cancer cell lines. Cancer Biol Med 2020; 17:970-985. [PMID: 33299647 PMCID: PMC7721098 DOI: 10.20892/j.issn.2095-3941.2020.0028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/20/2020] [Indexed: 01/08/2023] Open
Abstract
Objective Chromosomal instability (CIN) is a hallmark of cancer characterized by cell-to-cell variability in the number or structure of chromosomes, frequently observed in cancer cell populations and is associated with poor prognosis, metastasis, and therapeutic resistance. Breast cancer (BC) is characterized by unstable karyotypes and recent reports have indicated that CIN may influence the response of BC to chemotherapy regimens. However, paradoxical associations between extreme CIN and improved outcome have been observed. Methods This study aimed to 1) evaluate CIN levels and clonal heterogeneity (CH) in MCF7, ZR-751, MDA-MB468, BT474, and KPL4 BC cells treated with low doses of tamoxifen (TAM), docetaxel (DOC), doxorubicin (DOX), Herceptin (HT), and combined treatments (TAM/DOC, TAM/DOX, TAM/HT, HT/DOC, and HT/DOX) by using fluorescence in situ hybridization (FISH), and 2) examine the association with response to treatments by comparing FISH results with cell proliferation. Results Intermediate CIN was linked to drug sensitivity according to three characteristics: estrogen receptor α (ERα) and HER2 status, pre-existing CIN level in cancer cells, and the CIN induced by the treatments. ERα+/HER2- cells with intermediate CIN were sensitive to treatment with taxanes (DOC) and anthracyclines (DOX), while ERα-/HER2-, ERα+/HER2+, and ERα-/HER2+ cells with intermediate CIN were resistant to these treatments. Conclusions A greater understanding of CIN and CH in BC could assist in the optimization of existing therapeutic regimens and/or in supporting new strategies to improve cancer outcomes.
Collapse
Affiliation(s)
- Natalia Vargas-Rondón
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| | - Erika Pérez-Mora
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| | - Victoria E. Villegas
- Biology Program, Faculty of Natural Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| |
Collapse
|
48
|
Kour A, Sambyal V, Guleria K, Singh NR, Uppal MS, Manjari M, Sudan M. In silico pathway analysis based on chromosomal instability in breast cancer patients. BMC Med Genomics 2020; 13:168. [PMID: 33167967 PMCID: PMC7653868 DOI: 10.1186/s12920-020-00811-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/11/2020] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Complex genomic changes that arise in tumors are a consequence of chromosomal instability. In tumor cells genomic aberrations disrupt core signaling pathways involving various genes, thus delineating of signaling pathways can help understand the pathogenesis of cancer. The bioinformatics tools can further help in identifying networks of interactions between the genes to get a greater biological context of all genes affected by chromosomal instability. METHODS Karyotypic analyses was done in 150 clinically confirmed breast cancer patients and 150 age and gender matched healthy controls after 72 h Peripheral lymphocyte culturing and GTG-banding. Reactome database from Cytoscape software version 3.7.1 was used to perform in-silico analysis (functional interaction and gene enrichment). RESULTS Frequency of chromosomal aberrations (structural and numerical) was found to be significantly higher in patients as compared to controls. The genes harbored by chromosomal regions showing increased aberration frequency in patients were further analyzed in-silico. Pathway analysis on a set of genes that were not linked together revealed that genes HDAC3, NCOA1, NLRC4, COL1A1, RARA, WWTR1, and BRCA1 were enriched in the RNA Polymerase II Transcription pathway which is involved in recruitment, initiation, elongation and dissociation during transcription. CONCLUSION The current study employs the information inferred from chromosomal instability analysis in a non-target tissue for determining the genes and the pathways associated with breast cancer. These results can be further extrapolated by performing either mutation analysis in the genes/pathways deduced or expression analysis which can pinpoint the relevant functional impact of chromosomal instability.
Collapse
Affiliation(s)
- Akeen Kour
- Human Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vasudha Sambyal
- Human Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India.
| | - Kamlesh Guleria
- Human Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Neeti Rajan Singh
- Department of Surgery, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, Punjab, India
| | - Manjit Singh Uppal
- Department of Surgery, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, Punjab, India
| | - Mridu Manjari
- Department of Pathology, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, Punjab, India
| | - Meena Sudan
- Department of Radiotherapy, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, Punjab, India
| |
Collapse
|
49
|
Genetic Alterations of Metastatic Colorectal Cancer. Biomedicines 2020; 8:biomedicines8100414. [PMID: 33066148 PMCID: PMC7601984 DOI: 10.3390/biomedicines8100414] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/05/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Genome sequencing studies have characterized the genetic alterations of different tumor types, highlighting the diversity of the molecular processes driving tumor development. Comprehensive sequencing studies have defined molecular subtypes of colorectal cancers (CRCs) through the identification of genetic events associated with microsatellite stability (MSS), microsatellite-instability-high (MSI-H), and hypermutation. Most of these studies characterized primary tumors. Only recent studies have addressed the characterization of the genetic and clinical heterogeneity of metastatic CRC. Metastatic CRC genomes were found to be not fundamentally different from primary CRCs in terms of the mutational landscape or of genes that drive tumorigenesis, and a genomic heterogeneity associated with tumor location of primary tumors helps to define different clinical behaviors of metastatic CRCs. Although CRC metastatic spreading was traditionally seen as a late-occurring event, growing evidence suggests that this process can begin early during tumor development and the clonal architecture of these tumors is consistently influenced by cancer treatment. Although the survival rate of patients with metastatic CRC patients improved in the last years, the response to current treatments and prognosis of many of these patients remain still poor, indicating the need to discover new improvements for therapeutic vulnerabilities and to formulate a rational prospective of personalized therapies.
Collapse
|
50
|
Gemble S, Basto R. CHRONOCRISIS: When Cell Cycle Asynchrony Generates DNA Damage in Polyploid Cells. Bioessays 2020; 42:e2000105. [PMID: 32885500 DOI: 10.1002/bies.202000105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/19/2020] [Indexed: 12/16/2022]
Abstract
Polyploid cells contain multiple copies of all chromosomes. Polyploidization can be developmentally programmed to sustain tissue barrier function or to increase metabolic potential and cell size. Programmed polyploidy is normally associated with terminal differentiation and poor proliferation capacity. Conversely, non-programmed polyploidy can give rise to cells that retain the ability to proliferate. This can fuel rapid genome rearrangements and lead to diseases like cancer. Here, the mechanisms that generate polyploidy are reviewed and the possible challenges upon polyploid cell division are discussed. The discussion is framed around a recent study showing that asynchronous cell cycle progression (an event that is named "chronocrisis") of different nuclei from a polyploid cell can generate DNA damage at mitotic entry. The potential mechanisms explaining how mitosis in non-programmed polyploid cells can generate abnormal karyotypes and genetic instability are highlighted.
Collapse
Affiliation(s)
- Simon Gemble
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, Paris, 75005, France
| | - Renata Basto
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, Paris, 75005, France
| |
Collapse
|