1
|
Dai Z, Peng X, Cui X, Guo Y, Zhang J, Shen X, Liu CY, Liu Y. Innovative molecular subtypes of multiple signaling pathways in colon cancer and validation of FMOD as a prognostic-related marker. J Cancer Res Clin Oncol 2023; 149:13087-13106. [PMID: 37474678 DOI: 10.1007/s00432-023-05163-6] [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: 06/06/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
PURPOSE Colon cancer is highly heterogeneous in terms of the immune and stromal microenvironment, genomic integrity, and oncogenic properties; therefore, molecular subtypes of the four characteristic dimensions are expected to provide novel clues for immunotherapy of colon cancer. METHODS According to the enrichment of four dimensions, we performed consensus cluster analysis and identified three robust molecular subtypes for colon cancer, namely immune enriched, immune deficiency, and stroma enriched. We characterized and validated the immune infiltration, gene mutations, copy number variants, methylation, protein expression, and clinical features in different datasets. Finally, we developed an 8-gene risk prognostic model and proposed the innovative RiskScore. In addition, a nomogram model was constructed combining clinical characteristics and RiskScore to validate its excellent clinical predictive power. RESULTS Combining clinical patient tissue samples and histochemical microarray data, we found that high FMOD expression in tumor epithelial cells was associated with poorer patient prognosis, but FMOD expression in the mesenchyme was not associated with prognosis. In pan-cancer, RiskScore, a prognostic model constructed based on characteristic pathway scores, was a poor prognostic factor for malignancy and was negatively associated with immunotherapy response. CONCLUSION The identification of molecular subtypes could provide innovative ideas for immunotherapy of colon cancer.
Collapse
Affiliation(s)
- Zhujiang Dai
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Xiang Peng
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Xuewei Cui
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yuegui Guo
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Jie Zhang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Xia Shen
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China
| | - Chen-Ying Liu
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China.
| | - Yun Liu
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
- Shanghai Colorectal Cancer Research Center, Shanghai, 200092, China.
| |
Collapse
|
2
|
Greco L, Rubbino F, Dal Buono A, Laghi L. Microsatellite Instability and Immune Response: From Microenvironment Features to Therapeutic Actionability-Lessons from Colorectal Cancer. Genes (Basel) 2023; 14:1169. [PMID: 37372349 DOI: 10.3390/genes14061169] [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: 04/27/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Microsatellite instability (MSI) can be found in 15-20% of all colorectal cancers (CRC) and is the key feature of a defective DNA mismatch repair (MMR) system. Currently, MSI has been established as a unique and pivotal biomarker in the diagnosis, prognosis, and treatment of CRC. MSI tumors display a strong lymphocytic activation and a shift toward a tumoral microenvironment restraining metastatic potential and ensuing in a high responsiveness to immunotherapy of MSI CRC. Indeed, neoplastic cells with an MMR defect overexpress several immune checkpoint proteins, such as programmed death-1 (PD-1) and programmed death-ligand 1(PD-L1), that can be pharmacologically targeted, allowing for the revival the cytotoxic immune response toward the tumor. This review aims to illustrate the role of MSI in the tumor biology of colorectal cancer, focusing on the immune interactions with the microenvironment and their therapeutic implications.
Collapse
Affiliation(s)
- Luana Greco
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Federica Rubbino
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Arianna Dal Buono
- Division of Gastroenterology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
3
|
Wang HL, Ye ZM, He ZY, Huang L, Liu ZH. m6A-related lncRNA-based immune infiltration characteristic analysis and prognostic model for colonic adenocarcinoma. Hereditas 2023; 160:6. [PMID: 36755298 PMCID: PMC9909974 DOI: 10.1186/s41065-023-00267-y] [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: 05/02/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Colonic adenocarcinoma (COAD) is a common gastrointestinal tract tumor, and its occurrence and progression are typically associated with genomic instability, tumor-suppressor gene and oncogene mutations, and tumor mutational load. N6-methyladenosine (m6A) modification of RNAs and long non-coding RNA (lncRNA) expression are important in tumorigenesis and progression. However, the regulatory roles of m6A-associated lncRNAs in the tumor microenvironment, stratification of prognosis, and immunotherapy are unclear. METHODS We screened 43 prognostic lncRNAs linked to m6A and performed consistent molecular typing of COAD using consensus clustering. The single-sample Gene Set Enrichment Analysis and ESTIMATE algorithms were used to assess the immune characteristics of different subgroups. Covariation between methylation-related prognostic lncRNAs was eliminated by least absolute shrinkage and selection operator Cox regression. A nomogram was created and evaluated by combining the methylation-related prognostic lncRNA model with other clinical factors. The relationship between the prognostic model grouping and microsatellite instability, immunophenotype score, and tumor mutation burden was validated using R scripts. Finally, we used a linkage map to filter sensitive medicines to suppress the expression of high-risk genes. Three m6A-associated lncRNA modes were identified in 446 COAD specimens with different clinical endpoints and biological statuses. Risk scores were constructed based on the m6A-associated lncRNA signature genes. Patients with lower risk scores showed superior immunotherapy responses and clinical benefits compared to those with higher risk scores. Lower risk scores were also correlated with higher immunophenotype scores, tumor mutation burden, and mutation rates in significantly mutated genes (e.g., FAT4 and MUC16). Piperidolate, quinostatin, and mecamylamin were screened for their abilities to suppress the expression of high-risk genes in the model. CONCLUSIONS Quantitative assessment of m6A-associated lncRNAs in single tumors can enhance the understanding of tumor microenvironment profiles. The prognostic model constructed using m6A-associated lncRNAs may facilitate prognosis and immunotherapy stratification of patients with COAD; finally, three drugs with potential therapeutic value were screened based on the model.
Collapse
Affiliation(s)
- Hao-lun Wang
- grid.256607.00000 0004 1798 2653Graduate School of Guangxi Medical University, Nanning, 530021 China
| | - Zhuo-miao Ye
- grid.452223.00000 0004 1757 7615Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008 China
| | - Zi-yun He
- grid.256607.00000 0004 1798 2653Graduate School of Guangxi Medical University, Nanning, 530021 China
| | - Lu Huang
- grid.413431.0Day-Care Unit, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Zhi-hui Liu
- grid.413431.0Day-Care Unit, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, 530021 China
| |
Collapse
|
4
|
Jiang A, Song J, Fang X, Fang Y, Wang Z, Liu B, Wu Z, Qu L, Luo P, Wang L. A novel thinking: DDR axis refines the classification of ccRCC with distinctive prognosis, multi omics landscape and management strategy. Front Public Health 2022; 10:1029509. [PMID: 36478716 PMCID: PMC9720257 DOI: 10.3389/fpubh.2022.1029509] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/02/2022] [Indexed: 11/22/2022] Open
Abstract
Background DNA damage response and repair (DDR) related signatures play an important role in maintaining genome stability and other biological processes. It also affects the occurrence, development, and treatment of cancer. However, in renal cell carcinoma (RCC), especially clear cell renal carcinoma (ccRCC), the potential association between DDR-related signatures and tumor heterogeneity and tumor microenvironment (TME) remains unclear. Methods Utilizing unsupervised clustering algorithm, we divided RCC into two subgroups, DCS1 and DCS2, according to the differences in DDR gene expression, and compared the characteristics of the two subgroups through multiple dimensions. Results Compared with DCS1, DCS2 patients have higher clinical stage/grade and worse prognosis, which may be related to active metabolic status and immunosuppression status. At the same time, the high mutation rate in DCS2 may also be an important reason for the prognosis. We also analyzed the sensitivity of the two subgroups to different therapeutic agents and established a subtypes' biomarkers-based prognostic system with good validation results to provide ideas for clinical diagnosis and treatment. Finally, we identified a pivotal role for DDX1 in the DDR gene set, which may serve as a future therapeutic target. Conclusion This study showed that DDR has an important impact on the development and treatment of RCC. DCS2 subtypes have a poor prognosis, and more personalized treatment and follow-up programs may be needed. The assessment of DDR gene mutations in patients may be helpful for clinical decision-making. DDX1 may be one of the effective targets for RCC treatment in the future.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai, China
| | - Jiaao Song
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai, China
| | - Xiao Fang
- Department of Urology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai, China
| | - Zheng Wang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai, China
| | - Bing Liu
- Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhenjie Wu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai, China
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China,*Correspondence: Le Qu
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China,Peng Luo
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai, China,Linhui Wang
| |
Collapse
|
5
|
Laycock A, Kang A, Ang S, Texler M, Bentel J. Lack of correlation between MSH3 immunohistochemistry and microsatellite analysis for the detection of elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) in colorectal cancers. Hum Pathol 2021; 118:9-17. [PMID: 34537247 DOI: 10.1016/j.humpath.2021.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022]
Abstract
Immunohistochemical evaluation of mismatch repair protein (MMR) expression is an important screening tool in diagnostic pathology, where it is routinely used to identify subsets of colorectal cancers (CRCs) with either inherited or sporadic forms of microsatellite instability (MSI). MSH3 is not included in current MMR panels, although aberrant MSH3 expression is reported to occur in 40-60% of CRCs and is associated with elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) and a worse prognosis. In this study, we applied MSH3 immunohistochemistry and tetranucleotide MSI analysis to a cohort of 250 unselected CRCs to evaluate the potential use of the methods in routine practice. Partial, complete, and focal loss of nuclear MSH3 and its cytoplasmic mislocalization were evident in 67% of tumors, whereas MSI was evident in two to six of a panel of six tetranucleotide repeats in 46% of cases. However, concordance between MSH3 immunohistochemistry and tetranucleotide MSI results was only 61%, indicating the unsuitability of this combination of tests in routine pathology practice. MSH3 immunostaining was compromised in areas of tissue crush and autolysis, which are common in biopsy and surgical samples, potentially mitigating against its routine use. Although tetranucleotide MSI is clearly evident in a subset of CRCs, further development of validated sets of tetranucleotide repeats and either MSH3 or other immunohistochemical markers will be required to include EMAST testing in the routine evaluation of CRCs in clinical practice.
Collapse
Affiliation(s)
- Andrew Laycock
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia; University of Notre Dame, Fremantle, 6160 Western Australia, Australia; Curtin University, Perth, 6102 Western Australia, Australia.
| | - Alexandra Kang
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| | - Sophia Ang
- Clinical Services, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| | - Michael Texler
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| | - Jacqueline Bentel
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| |
Collapse
|
6
|
Effects of a Novel Thiadiazole Derivative with High Anticancer Activity on Cancer Cell Immunogenic Markers: Mismatch Repair System, PD-L1 Expression, and Tumor Mutation Burden. Pharmaceutics 2021; 13:pharmaceutics13060885. [PMID: 34203761 PMCID: PMC8232699 DOI: 10.3390/pharmaceutics13060885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/31/2022] Open
Abstract
Microsatellite instability (MSI), tumor mutation burden (TMB), and programmed cell death ligand-1 (PD-L1) are particularly known as immunotherapy predictive biomarkers. MSI and TMB are closely related to DNA mismatch repair (MMR) pathway functionality, while the PD-L1 checkpoint mediates cancer cell evasion from immune surveillance via the PD-L1/PD-1 axis. Among all the novel triazolo[3,4-b]thiadiazole derivatives, the compound KA39 emerged as the most potent anticancer agent. In the present study, potential alterations in MSI, TMB, and/or PD-L1 expression upon cell treatment with KA39 are explored. We tested three MMR-deficient (DLD-1, LS174T, and DU-145) and two MMR-proficient (HT-29 and PC-3) human cancer cell lines. Our findings support KA39-induced PD-L1 overexpression in all cancer cell lines, although the most outstanding increase was observed in MMR-proficient HT-29 cells. MSI analysis showed that KA39 affects the MMR system, impairing its recognition or repair activity, particularly in MMR-deficient DLD-1 and DU-145 cells, enhancing oligonucleotide production. There were no remarkable alterations in the TMB between untreated and treated cells, indicating that KA39 does not belong to mutagenic agents. Taking together the significant in vitro anticancer activity with PD-L1 upregulation and MSI increase, KA39 should be investigated further for its implication in chemo-immunotherapy of cancer.
Collapse
|
7
|
Cini G, Carnevali I, Sahnane N, Chiaravalli AM, Dell'Elice A, Maestro R, Pin E, Bestetti I, Radovic S, Armelao F, Viel A, Tibiletti MG. Lynch syndrome and Muir-Torre phenotype associated with a recurrent variant in the 3'UTR of the MSH6 gene. Cancer Genet 2021; 254-255:1-10. [PMID: 33516942 DOI: 10.1016/j.cancergen.2021.01.005] [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: 08/07/2020] [Revised: 12/18/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
Abstract
A MSH6 3'UTR variant (c.*23_26dup) was found in 13 unrelated families consulted for Lynch/Muir-Torre Syndrome. This variant, which is very rare in the genomic databases, was absent in healthy controls and strongly segregated with the disease in the studied pedigrees. All tumors were defective for MSH2/MSH6/MSH3 proteins expression, but only MSH2 somatic pathogenic mutations were found in 5 of the 12 sequenced tumors. Moreover, we had no evidence of MSH6 transcript decrease in carriers, whereas MSH2 transcript was downregulated. Additional evaluations performed in representative carriers, including karyotype, arrayCGH and Linked-Reads whole genome sequencing, failed to evidence any MSH2 germline pathogenic variant. Posterior probability of pathogenicity for MSH6 c.*23_26dup was obtained from a multifactorial analysis incorporating segregation and phenotypic data and resulted >0.999, allowing to classify the variant as pathogenic (InSiGHT Class 5). Carriers shared a common haplotype involving MSH2/MSH6 loci, then a cryptic disease-associated variant, linked with MSH6 c.*23_26dup, cannot be completely excluded. Even if it is not clear whether the MSH6 variant is pathogenic per se or simply a marker of a disease-associated MSH2/MSH6 haplotype, all data collected on patients and pedigrees prompted us to manage the variant as pathogenic and to offer predictive testing within these families.
Collapse
Affiliation(s)
- Giulia Cini
- Unit of Functional Oncogenomics and Genetics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy
| | - Ileana Carnevali
- Department of Pathology, Circolo Hospital ASST Settelaghi, via O. Rossi 9, 21100, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, via O. Rossi 9, 21100, Varese, Italy
| | - Nora Sahnane
- Department of Pathology, Circolo Hospital ASST Settelaghi, via O. Rossi 9, 21100, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, via O. Rossi 9, 21100, Varese, Italy
| | - Anna Maria Chiaravalli
- Department of Pathology, Circolo Hospital ASST Settelaghi, via O. Rossi 9, 21100, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, via O. Rossi 9, 21100, Varese, Italy
| | - Anastasia Dell'Elice
- Unit of Functional Oncogenomics and Genetics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy
| | - Roberta Maestro
- Unit of Functional Oncogenomics and Genetics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy
| | - Elisa Pin
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, The Royal Institute of Technology KTH, Tomtebodavägen 23B, 171 65 Solna, Stockholm, Sweden
| | - Ilaria Bestetti
- Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, Via Zucchi, 18 - 20095 Cusano Milanino (MI); Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20133 Milan, Italy
| | | | - Franco Armelao
- U.O. Gastroenterologia ed Endoscopia Digestiva, Ospedale S. Chiara, APSS, Via A. de Gasperi 79 - 38123, Trento, Italy
| | - Alessandra Viel
- Unit of Functional Oncogenomics and Genetics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081 Aviano, Italy.
| | - Maria Grazia Tibiletti
- Department of Pathology, Circolo Hospital ASST Settelaghi, via O. Rossi 9, 21100, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, via O. Rossi 9, 21100, Varese, Italy
| |
Collapse
|
8
|
Prognostic and Predictive Cross-Roads of Microsatellite Instability and Immune Response to Colon Cancer. Int J Mol Sci 2020; 21:ijms21249680. [PMID: 33353162 PMCID: PMC7766746 DOI: 10.3390/ijms21249680] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
Understanding molecular features of colon cancer has shed light on its pathogenesis and progression. Over time, some of these features acquired clinical dignity and were incorporated in decision making. Namely, microsatellite instability (MSI) due to mismatch repair of defects, which primarily was adopted for the diagnosis of Lynch syndrome, became recognized as the biomarker of a different disease type, showing a less aggressive behavior. MSI tumors harbor high amounts of tumor infiltrating lymphocytes (TILs) due to their peculiar load in neoantigens. However, microsatellite stable colon cancer may also show high amounts of TILs, and this feature is as well associated with better outcomes. High TIL loads are in general associated with a favorable prognosis, especially in stage II colon cancer, and therein identifies a patient subset with the lowest probability of relapse. With respect to post-surgical adjuvant treatment, particularly in stage III, TILs predictive ability seems to weaken along with the progression of the disease, being less evident in high risk patients. Moving from cohort studies to the analysis of a series from clinical trials contributed to increase the robustness of TILs as a biomarker. The employment of high TIL densities as an indicator of good prognosis in early-stage colon cancers is strongly advisable, while in late-stage colon cancers the employment as an indicator of good responsiveness to post-surgical therapy requires refinement. It remains to be clarified whether TILs could help in identifying those patients with node-positive cancers to whom adjuvant treatment could be spared, at least in low-risk groups as defined by the TNM staging system.
Collapse
|
9
|
Tabibzadeh A, Tameshkel FS, Moradi Y, Soltani S, Moradi-Lakeh M, Ashrafi GH, Motamed N, Zamani F, Motevalian SA, Panahi M, Esghaei M, Ajdarkosh H, Mousavi-Jarrahi A, Niya MHK. Signal transduction pathway mutations in gastrointestinal (GI) cancers: a systematic review and meta-analysis. Sci Rep 2020; 10:18713. [PMID: 33127962 PMCID: PMC7599243 DOI: 10.1038/s41598-020-73770-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
The present study was conducted to evaluate the prevalence of the signaling pathways mutation rate in the Gastrointestinal (GI) tract cancers in a systematic review and meta-analysis study. The study was performed based on the PRISMA criteria. Random models by confidence interval (CI: 95%) were used to calculate the pooled estimate of prevalence via Metaprop command. The pooled prevalence indices of signal transduction pathway mutations in gastric cancer, liver cancer, colorectal cancer, and pancreatic cancer were 5% (95% CI: 3-8%), 12% (95% CI: 8-18%), 17% (95% CI: 14-20%), and 20% (95% CI: 5-41%), respectively. Also, the mutation rates for Wnt pathway and MAPK pathway were calculated to be 23% (95% CI, 14-33%) and 20% (95% CI, 17-24%), respectively. Moreover, the most popular genes were APC (in Wnt pathway), KRAS (in MAPK pathway) and PIK3CA (in PI3K pathway) in the colorectal cancer, pancreatic cancer, and gastric cancer while they were beta-catenin and CTNNB1 in liver cancer. The most altered pathway was Wnt pathway followed by the MAPK pathway. In addition, pancreatic cancer was found to be higher under the pressure of mutation compared with others based on pooled prevalence analysis. Finally, APC mutations in colorectal cancer, KRAS in gastric cancer, and pancreatic cancer were mostly associated gene alterations.
Collapse
Affiliation(s)
- Alireza Tabibzadeh
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Safarnezhad Tameshkel
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Yousef Moradi
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Saber Soltani
- Department of Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Maziar Moradi-Lakeh
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
- Preventive Medicine and Public Health Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - G Hossein Ashrafi
- Cancer Theme SEC Faculty, Kingston University, Penrhyn Road, London, KT1 2EE, UK
| | - Nima Motamed
- Department of Social Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Abbas Motevalian
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Panahi
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Esghaei
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Ajdarkosh
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
10
|
Cavalleri T, Bianchi P, Basso G, Celesti G, Grizzi F, Bossi P, Greco L, Pitrone C, Valtorta E, Mauri G, Truini M, Dall'Olio FG, Brandi G, Sartore-Bianchi A, Ricciardiello L, Torri V, Rimassa L, Siena S, Mantovani A, Malesci A, Laghi L. Combined Low Densities of FoxP3 + and CD3 + Tumor-Infiltrating Lymphocytes Identify Stage II Colorectal Cancer at High Risk of Progression. Cancer Immunol Res 2019; 7:751-758. [PMID: 30804005 DOI: 10.1158/2326-6066.cir-18-0661] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/07/2018] [Accepted: 02/19/2019] [Indexed: 11/16/2022]
Abstract
The densities of CD3+ and CD8+ tumor-infiltrating lymphocytes (TILs), combined with tumor-node-metastasis (TNM) staging, have prognostic value for patients with nonmetastatic colorectal cancer. We compared the prognostic value of CD3+ and FoxP3+ TILs at the invasive front, TNM classifiers, and microsatellite (MS) status in a trial set of patients with stage II and III colorectal cancer (n = 413), by recursive partitioning with a classification and regression tree (CART). Significant prognostic factors and interactions were reassessed by logistic regression and Cox proportional-hazards modeling in the trial and a validation set (n = 215) of patients with stage II colorectal cancer. In the trial set, CART indicated that TIL numbers were of value only in predicting recurrence risk for stage II cancers, where low densities of FoxP3+ TILs ranked first and low densities of CD3+ TILs further stratifying risk. Multivariate analysis showed that TILs interacted with tumor stage (FoxP3+, P = 0.06; CD3+, P = 0.02) and MS instability (MSI; FoxP3+; P = 0.02). In stage II MS-stable cancers, concomitant low densities of both FoxP3+ and CD3+ TILs identified patients with the highest progression risk in the trial [HR 7.24; 95% confidence interval (CI), 3.41-15.4; P < 0.001] and the validation (HR 15.16; 95% CI, 3.43-66.9; P < 0.001) sets. FoxP3+ and CD3+ TIL load in colorectal cancer was more informative than other prognostic factors before the cancer progressed to lymph nodes. This prognostic information about TILs, including FoxP3+ cells, suggests that randomized controlled trials might be refined to include interactions between TNM status, molecular classifiers, and postsurgical treatments.
Collapse
Affiliation(s)
- Tommaso Cavalleri
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Paolo Bianchi
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Gianluca Basso
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Giuseppe Celesti
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Fabio Grizzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Paola Bossi
- Department of Pathology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Luana Greco
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Calogero Pitrone
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Emanuele Valtorta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Gianluca Mauri
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Dipartimento di Oncologia ed Emato-Oncologia, Milano, Italy
| | - Mauro Truini
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Dipartimento di Oncologia ed Emato-Oncologia, Milano, Italy
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Valter Torri
- Laboratory of Methodology for Biomedical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Lorenza Rimassa
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Dipartimento di Oncologia ed Emato-Oncologia, Milano, Italy
| | - Alberto Mantovani
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
- Department of Biotechnologies and Translational Medicine, Humanitas University, Pieve Emanuele, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alberto Malesci
- Department of Internal Medicine, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | | |
Collapse
|
11
|
Basso G, Bianchi P, Malesci A, Laghi L. Hereditary or sporadic polyposis syndromes. Best Pract Res Clin Gastroenterol 2017; 31:409-417. [PMID: 28842050 DOI: 10.1016/j.bpg.2017.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 01/31/2023]
Abstract
Polyposis syndromes are encountered in endoscopy practice, and are considered rare entities, accounting for ≤1% of colorectal cancer. Polyposis can occur within inherited syndromes or as "sporadic" cases of unknown etiology. Their proper characterization is relevant for patient management, and should nowadays drive appropriate genetic tests which have a key role in clinical practice for driving surveillance and colorectal cancer prevention, enlarged to relatives. Polyposis classification is based upon polyp number and histology, familial and personal history. This review will explore the polyposis nosology and their genetic determinants in the emerging scenario of Next Generation Sequencing which allow testing multiples genes in parallel. This capability will likely continue to increase the range of polyposis predisposing genes, contributing to define new clinical entities.
Collapse
Affiliation(s)
- Gianluca Basso
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Paolo Bianchi
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Alberto Malesci
- Department of Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20133 Milan, Italy; Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy; Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy; Hereditary Cancer Genetics Clinic, Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy.
| |
Collapse
|
12
|
Koi M, Garcia M, Choi C, Kim HR, Koike J, Hemmi H, Nagasaka T, Okugawa Y, Toiyama Y, Kitajima T, Imaoka H, Kusunoki M, Chen YH, Mukherjee B, Boland CR, Carethers JM. Microsatellite Alterations With Allelic Loss at 9p24.2 Signify Less-Aggressive Colorectal Cancer Metastasis. Gastroenterology 2016; 150:944-55. [PMID: 26752111 PMCID: PMC4808397 DOI: 10.1053/j.gastro.2015.12.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 12/20/2015] [Accepted: 12/22/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Molecular events that lead to recurrence and/or metastasis after curative treatment of patients with colorectal cancers (CRCs) are poorly understood. Patients with stage II or III primary CRC with elevated microsatellite alterations at selected tetranucleotide repeats and low levels of microsatellite instability (E/L) are more likely to have disease recurrence after treatment. Hypoxia and/or inflammation not only promote metastasis, but also induce elevated microsatellite alterations at selected tetranucleotide repeats by causing deficiency of MSH3 in the cancer cell nucleus. We aimed to identify genetic alterations associated with metastasis of primary colorectal tumors to liver and to determine their effects on survival. METHODS We obtained 4 sets of primary colorectal tumors and matched liver metastases from hospitals in Korea and Japan. Intragenic microsatellites with large repeats at 141 loci were examined for frame-shift mutations and/or loss of heterozygosity (LOH) as possible consequences of MSH3 deficiency. Highly altered loci were examined for association with E/L in liver metastases. We analyzed data from 156 of the patients with stage II or III primary colorectal tumors to determine outcomes and whether altered loci were associated with E/L. RESULTS LOH at several loci at chromosome 9p24.2 (9p24.2-LOH) was associated with E/L in liver metastases (odds ratio = 10.5; 95% confidence interval: 2.69-40.80; P = .0007). We found no significant difference in the frequency of E/L, 9p24.2-LOH, mutations in KRAS or BRAF, or the combination of E/L and 9p24.2-LOH, between primary colorectal tumors and their matched metastases. Patients with stage II or III colorectal tumors with E/L and 9p24.2-LOH had increased survival after CRC recurrence (hazard ratio = 0.25; 95% CI: 0.12-0.50; P = .0001), compared with patients without with E/L and 9p24.2-LOH. E/L with 9p24.2-LOH appeared to be an independent prognostic factor for overall survival of patients with stage III CRC (hazard ratio = 0.06; 95% CI: 0.01-0.57; P = .01). CONCLUSIONS E/L with 9p24-LOH appears to be a biomarker for less aggressive metastasis from stage III primary colorectal tumors.
Collapse
Affiliation(s)
- Minoru Koi
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Gastrointestinal Cancer Research Laboratory, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - C Richard Boland
- Gastrointestinal Cancer Research Laboratory, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas.
| | - John M Carethers
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
13
|
Galdiero MR, Bianchi P, Grizzi F, Di Caro G, Basso G, Ponzetta A, Bonavita E, Barbagallo M, Tartari S, Polentarutti N, Malesci A, Marone G, Roncalli M, Laghi L, Garlanda C, Mantovani A, Jaillon S. Occurrence and significance of tumor-associated neutrophils in patients with colorectal cancer. Int J Cancer 2016; 139:446-56. [PMID: 26939802 DOI: 10.1002/ijc.30076] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/01/2016] [Accepted: 02/24/2016] [Indexed: 11/06/2022]
Abstract
Inflammatory cells are an essential component of the tumor microenvironment. Neutrophils have emerged as important players in the orchestration and effector phase of innate and adaptive immunity. The significance of tumor-associated neutrophils (TAN) in colorectal cancer (CRC) has been the subject of conflicting reports and the present study was designed to set up a reliable methodology to assess TAN infiltration in CRC and to evaluate their clinical significance. CD66b and myeloperoxidase (MPO) were assessed as candidate neutrophil markers in CRC using immunohistochemistry. CD66b was found to be a reliable marker to identify TAN in CRC tissues, whereas MPO also identified a subset of CD68(+) macrophages. CRC patients (n = 271) (Stages I-IV) were investigated retrospectively by computer-assisted imaging on whole tumor sections. TAN density dramatically decreases in Stage IV patients as compared to Stage I-III. At Cox analysis, higher TAN density was associated with better prognosis. Importantly, multivariate analysis showed that prognostic significance of TAN can be influenced by clinical stage and 5-fluorouracil(5-FU)-based chemotherapy. On separate analysis of Stage III patients (n = 178), TAN density had a dual clinical significance depending on the use of 5-FU-based chemotherapy. Unexpectedly, higher TAN density was associated with better response to 5-FU-based chemotherapy. Thus, TAN are an important component of the immune cell infiltrate in CRC and assessment of TAN infiltration may help identify patients likely to benefit from 5-FU-based chemotherapy. These results call for a reassessment of the role of neutrophils in cancer using rigorous quantitative methodology.
Collapse
Affiliation(s)
- Maria Rosaria Galdiero
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy.,Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Paolo Bianchi
- Molecular Gastroenterology, Department of Gastroenterology & Clinical Investigation Laboratory, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Fabio Grizzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Giuseppe Di Caro
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Gianluca Basso
- Molecular Gastroenterology, Department of Gastroenterology & Clinical Investigation Laboratory, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Andrea Ponzetta
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Eduardo Bonavita
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Marialuisa Barbagallo
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Silvia Tartari
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Nadia Polentarutti
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy
| | - Alberto Malesci
- Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy.,Department of Biotechnologies and Translational Medicine, University of Milan, Rozzano, Milan, 20089, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Massimo Roncalli
- Department of Pathology, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, 20089, Italy
| | - Luigi Laghi
- Hereditary Cancer Genetic Clinic & Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan 20089, Italy
| | - Cecilia Garlanda
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, 20089, Italy
| | - Alberto Mantovani
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, 20089, Italy
| | - Sébastien Jaillon
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Rozzano, Milan, 20089, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, 20089, Italy
| |
Collapse
|
14
|
Germini DE, Mader AMAA, Gomes LGL, Teodoro TR, Franco MIF, Waisberg J. Detection of DNA repair protein in colorectal cancer of patients up to 50 years old can increase the identification of Lynch syndrome? Tumour Biol 2015; 37:2757-64. [PMID: 26408182 DOI: 10.1007/s13277-015-4108-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/20/2015] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to compare the results of protein level of the DNA mismatch repair genes with the clinical diagnosis of Lynch syndrome according to the Amsterdam II criteria in patients 50 years and younger who underwent surgery for colorectal cancer. The subjects of analysis were 48 patients 50 years old and younger. Immunohistochemistry assays were performed to detect proteins from the DNA mismatch repair genes. Clinicopathological data and Amsterdam II criteria for the diagnosis of hereditary nonpolyposis colorectal cancer were obtained by analyzing medical records. Two (4 %) patients satisfied the Amsterdam II criteria for Lynch syndrome, and both presented levels of all of the studied mismatch repair proteins. A total of 13 (27 %) patients exhibited the absence of protein levels of the studied mismatch repair genes. None of these patients were considered suspicious for Lynch syndrome according to the Amsterdam II criteria. Screening for the level of proteins of the mismatch repair system in all colorectal cancer patients 50 years and younger can increase the identification of patients with suspicion of Lynch syndrome.
Collapse
Affiliation(s)
- Demétrius Eduardo Germini
- Department of Surgery, São Paulo State Civil Servant Hospital, Rua Augusto de Miranda, 1303 ap. 22 Pompéia, São Paulo, SP, 05026-001, Brazil.
| | | | - Luiz Guilherme Lisboa Gomes
- Department of Surgery, ABC Medical School, Avenida Príncipe de Gales, 821, Santo André, SP, 09080-650, Brazil
| | - Thérèse Rachel Teodoro
- Department of Morphology, ABC Medical School, Avenida Príncipe de Gales, 821, 09080650, Santo André, SP, Brazil
| | - Maria Isete Fares Franco
- Department of Pathology, São Paulo State Civil Servant Hospital, Avenida Ibirapuera, 981, São Paulo, SP, 04029-000, Brazil
| | - Jaques Waisberg
- Department of Surgery, ABC Medical School, Avenida Príncipe de Gales, 821, Santo André, SP, 09080-650, Brazil
| |
Collapse
|
15
|
Pinheiro M, Pinto C, Peixoto A, Veiga I, Lopes P, Henrique R, Baldaia H, Carneiro F, Seruca R, Tomlinson I, Kovac M, Heinimann K, Teixeira MR. Target gene mutational pattern in Lynch syndrome colorectal carcinomas according to tumour location and germline mutation. Br J Cancer 2015; 113:686-92. [PMID: 26247575 PMCID: PMC4647680 DOI: 10.1038/bjc.2015.281] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/17/2015] [Accepted: 06/25/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND We previously reported that the target genes in sporadic mismatch repair (MMR)-deficient colorectal carcinomas (CRCs) in the distal colon differ from those occurring elsewhere in the colon. This study aimed to compare the target gene mutational pattern in microsatellite instability (MSI) CRC from Lynch syndrome patients stratified by tumour location and germline mutation, as well as with that of sporadic disease. METHODS A series of CRC from Lynch syndrome patients was analysed for MSI in genes predicted to be selective MSI targets and known to be involved in several pathways of colorectal carcinogenesis. RESULTS The most frequently mutated genes belong to the TGF-β superfamily pathway, namely ACVR2A and TGFBR2. A significantly higher frequency of target gene mutations was observed in CRC from patients with germline mutations in MLH1 or MSH2 when compared with MSH6. Mutations in microsatellite sequences (A)7 of BMPR2 and (A)8 of MSH3 were significantly more frequent in the distal CRC. Additionally, we observed differences in MSH3 and TGFBR2 mutational frequency between Lynch syndrome and sporadic MSI CRC regarding tumour location. CONCLUSIONS Our results indicate that the pattern of genetic changes differs in CRC depending on tumour location and between Lynch syndrome and sporadic MSI CRC, suggesting that carcinogenesis can occur by different pathways even if driven by generalised MSI.
Collapse
Affiliation(s)
- Manuela Pinheiro
- Department of Genetics, Portuguese Oncology Institute, Rua Doutor António Bernardino Almeida, 4200-072 Porto, Portugal
| | - Carla Pinto
- Department of Genetics, Portuguese Oncology Institute, Rua Doutor António Bernardino Almeida, 4200-072 Porto, Portugal
| | - Ana Peixoto
- Department of Genetics, Portuguese Oncology Institute, Rua Doutor António Bernardino Almeida, 4200-072 Porto, Portugal
| | - Isabel Veiga
- Department of Genetics, Portuguese Oncology Institute, Rua Doutor António Bernardino Almeida, 4200-072 Porto, Portugal
| | - Paula Lopes
- Department of Pathology, Portuguese Oncology Institute, Rua Doutor António Bernardino Almeida, 4200-072 Porto, Portugal
| | - Rui Henrique
- Department of Pathology, Portuguese Oncology Institute, Rua Doutor António Bernardino Almeida, 4200-072 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, University of Porto, Largo Professor Abel Salazar, 4099-003 Porto, Portugal
| | - Helena Baldaia
- Department of Pathology, Centro Hospitalar de São João, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Fátima Carneiro
- Department of Pathology, Centro Hospitalar de São João, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Medical Faculty of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Raquel Seruca
- Medical Faculty of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Ian Tomlinson
- Molecular and Population Genetics Laboratory, Nuffield Department of Clinical Medicine, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
- Oxford NIHR Comprehensive Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Michal Kovac
- Research Group Human Genomics, Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
- Medical Genetics, University Hospital Basel, Burgfelderstrasse 101, 4055 Basel, Switzerland
| | - Karl Heinimann
- Research Group Human Genomics, Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
- Medical Genetics, University Hospital Basel, Burgfelderstrasse 101, 4055 Basel, Switzerland
| | - Manuel R Teixeira
- Department of Genetics, Portuguese Oncology Institute, Rua Doutor António Bernardino Almeida, 4200-072 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, University of Porto, Largo Professor Abel Salazar, 4099-003 Porto, Portugal
| |
Collapse
|
16
|
Di Caro G, Bergomas F, Grizzi F, Doni A, Bianchi P, Malesci A, Laghi L, Allavena P, Mantovani A, Marchesi F. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin Cancer Res 2014; 20:2147-58. [PMID: 24523438 DOI: 10.1158/1078-0432.ccr-13-2590] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Tumor-infiltrating T lymphocytes (TIL) play a key role in the clinical outcome of human colorectal cancer; however, the dynamics of their recruitment along colorectal cancer clinical progression have not been fully elucidated. Tertiary lymphoid tissue (TLT) is an ectopic organized lymph node-like structure that typically forms at sites of chronic inflammation and is involved in adaptive immune responses. Its occurrence in cancer is sporadically documented and its role and clinical relevance is largely unknown. EXPERIMENTAL DESIGN The occurrence of TLT, the correlation with TILs, and the clinical relevance were evaluated retrospectively, in a cohort study involving a consecutive series of 351 patients with stage II and III colorectal cancer. The role of TLT in lymphocyte recruitment was assessed in a preclinical model of colorectal cancer. RESULTS In both human colorectal cancer and in a murine model of colorectal cancer, we identified organized TLT, highly vascularized (including high endothelial venules), and correlated with the density of CD3(+) TILs. Intravenous injection in mice of GFP splenocytes resulted in homing of lymphocytes to TLT, suggesting an active role of TLT in the recruitment of lymphocytes to tumor areas. Accordingly, TLT density and TIL infiltration correlated and were coordinated in predicting better patient's outcome among patients with stage II colorectal cancer. CONCLUSIONS We provide evidence that TLT is associated with lymphocyte infiltration in colorectal cancer, providing a pathway of recruitment for TILs. TLT cooperates with TILs in a coordinated antitumor immune response, when identifying patients with low-risk early-stage colorectal cancer, thus, representing a novel prognostic biomarker for colorectal cancer.
Collapse
Affiliation(s)
- Giuseppe Di Caro
- Authors' Affiliations: Departments of Immunology and Inflammation; and Gastroenterology; Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano; and Department of Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Park JM, Huang S, Tougeron D, Sinicrope FA. MSH3 mismatch repair protein regulates sensitivity to cytotoxic drugs and a histone deacetylase inhibitor in human colon carcinoma cells. PLoS One 2013; 8:e65369. [PMID: 23724141 PMCID: PMC3665625 DOI: 10.1371/journal.pone.0065369] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 04/28/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND MSH3 is a DNA mismatch repair (MMR) gene that undergoes frequent somatic mutation in colorectal cancers (CRCs) with MMR deficiency. MSH3, together with MSH2, forms the MutSβ heteroduplex that interacts with interstrand cross-links induced by drugs such as cisplatin. To date, the impact of MSH3 on chemosensitivity is unknown. METHODS We utilized isogenic HCT116 (MLH1-/MSH3-) cells where MLH1 is restored by transfer of chromosome 3 (HCT116+ch3) and also MSH3 by chromosome 5 (HCT116+3+5). We generated HCT116+3+5, SW480 (MLH1+/MSH3+) and SW48 (MLH1-/MSH3+) cells with shRNA knockdown of MSH3. Cells were treated with 5-fluorouracil (5-FU), SN-38, oxaliplatin, or the histone deacetylase (HDAC) inhibitor PCI-24781 and cell viability, clonogenic survival, DNA damage and apoptosis were analyzed. RESULTS MSH3-deficient vs proficient CRC cells showed increased sensitivity to the irinotecan metabolite SN-38 and to oxaliplatin, but not 5-FU, as shown in assays for apoptosis and clonogenic survival. In contrast, suppression of MLH1 attenuated the cytotoxic effect of 5-FU, but did not alter sensitivity to SN-38 or oxaliplatin. The impact of MSH3 knockdown on chemosensitivity to SN-38 and oxaliplatin was maintained independent of MLH1 status. In MSH3-deficient vs proficient cells, SN-38 and oxaliplatin induced higher levels of phosphorylated histone H2AX and Chk2, and similar results were found in MLH1-proficient SW480 cells. MSH3-deficient vs proficient cells showed increased 53BP1 nuclear foci after irradiation, suggesting that MSH3 can regulate DNA double strand break (DSB) repair. We then utilized PCI-24781 that interferes with homologous recombination (HR) indicated by a reduction in Rad51 expression. The addition of PCI-24781 to oxaliplatin enhanced cytotoxicity to a greater extent compared to either drug alone. CONCLUSION MSH3 status can regulate the DNA damage response and extent of apoptosis induced by chemotherapy. The ability of MSH3 to regulate chemosensitivity was independent of MLH1 status. PCI-24781-mediated impairment of HR enhanced oxaliplatin sensitivity, suggesting that reduced DSB repair capacity may be contributory.
Collapse
Affiliation(s)
- Jae Myung Park
- Mayo Clinic and Mayo Cancer Center, Rochester, Minnesota, United States of America
| | - Shengbing Huang
- Mayo Clinic and Mayo Cancer Center, Rochester, Minnesota, United States of America
| | - David Tougeron
- Mayo Clinic and Mayo Cancer Center, Rochester, Minnesota, United States of America
- Department of Gastroenterology, Poitiers, France
| | - Frank A. Sinicrope
- Mayo Clinic and Mayo Cancer Center, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
18
|
Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood 2013; 122:1256-65. [PMID: 23699601 DOI: 10.1182/blood-2013-02-483727] [Citation(s) in RCA: 309] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a genetically heterogeneous cancer composed of at least 2 molecular subtypes that differ in gene expression and distribution of mutations. Recently, application of genome/exome sequencing and RNA-seq to DLBCL has revealed numerous genes that are recurrent targets of somatic point mutation in this disease. Here we provide a whole-genome-sequencing-based perspective of DLBCL mutational complexity by characterizing 40 de novo DLBCL cases and 13 DLBCL cell lines and combining these data with DNA copy number analysis and RNA-seq from an extended cohort of 96 cases. Our analysis identified widespread genomic rearrangements including evidence for chromothripsis as well as the presence of known and novel fusion transcripts. We uncovered new gene targets of recurrent somatic point mutations and genes that are targeted by focal somatic deletions in this disease. We highlight the recurrence of germinal center B-cell-restricted mutations affecting genes that encode the S1P receptor and 2 small GTPases (GNA13 and GNAI2) that together converge on regulation of B-cell homing. We further analyzed our data to approximate the relative temporal order in which some recurrent mutations were acquired and demonstrate that ongoing acquisition of mutations and intratumoral clonal heterogeneity are common features of DLBCL. This study further improves our understanding of the processes and pathways involved in lymphomagenesis, and some of the pathways mutated here may indicate new avenues for therapeutic intervention.
Collapse
|
19
|
Lu Y, Soong TD, Elemento O. A novel approach for characterizing microsatellite instability in cancer cells. PLoS One 2013; 8:e63056. [PMID: 23671654 PMCID: PMC3646030 DOI: 10.1371/journal.pone.0063056] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 04/01/2013] [Indexed: 11/18/2022] Open
Abstract
Microsatellite instability (MSI) is characterized by the expansion or contraction of DNA repeat tracts as a consequence of DNA mismatch repair deficiency (MMRD). Accurate detection of MSI in cancer cells is important since MSI is associated with several cancer subtypes and can help inform therapeutic decisions. Although experimental assays have been developed to detect MSI, they typically depend on a small number of known microsatellite loci or mismatch repair genes and have limited reliability. Here, we report a novel genome-wide approach for MSI detection based on the global detection of insertions and deletions (indels) in microsatellites found in expressed genes. Our large-scale analyses of 20 cancer cell lines and 123 normal individuals revealed striking indel features associated with MSI: there is a significant increase of short microsatellite deletions in MSI samples compared to microsatellite stable (MSS) ones, suggesting a mechanistic bias of repair efficiency between insertions and deletions in normal human cells. By incorporating this observation into our MSI scoring metric, we show that our approach can correctly distinguish between MSI and MSS cancer cell lines. Moreover, when we applied this approach to primal tumor samples, our metric is also well consistent with diagnosed MSI status. Thus, our study offers new insight into DNA mismatch repair system, and also provides a novel MSI diagnosis method for clinical oncology with better reliability.
Collapse
Affiliation(s)
- Yuheng Lu
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, New York, United States of America
| | - T. David Soong
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, New York, United States of America
- Institute of Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, United States of America
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, New York, United States of America
- Institute of Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
20
|
Campregher C, Schmid G, Ferk F, Knasmüller S, Khare V, Kortüm B, Dammann K, Lang M, Scharl T, Spittler A, Roig AI, Shay JW, Gerner C, Gasche C. MSH3-deficiency initiates EMAST without oncogenic transformation of human colon epithelial cells. PLoS One 2012; 7:e50541. [PMID: 23209772 PMCID: PMC3507781 DOI: 10.1371/journal.pone.0050541] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 10/25/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIM Elevated microsatellite instability at selected tetranucleotide repeats (EMAST) is a genetic signature in certain cases of sporadic colorectal cancer and has been linked to MSH3-deficiency. It is currently controversial whether EMAST is associated with oncogenic properties in humans, specifically as cancer development in Msh3-deficient mice is not enhanced. However, a mutator phenotype is different between species as the genetic positions of repetitive sequences are not conserved. Here we studied the molecular effects of human MSH3-deficiency. METHODS HCT116 and HCT116+chr3 (both MSH3-deficient) and primary human colon epithelial cells (HCEC, MSH3-wildtype) were stably transfected with an EGFP-based reporter plasmid for the detection of frameshift mutations within an [AAAG]17 repeat. MSH3 was silenced by shRNA and changes in protein expression were analyzed by shotgun proteomics. Colony forming assay was used to determine oncogenic transformation and double strand breaks (DSBs) were assessed by Comet assay. RESULTS Despite differential MLH1 expression, both HCT116 and HCT116+chr3 cells displayed comparable high mutation rates (about 4×10(-4)) at [AAAG]17 repeats. Silencing of MSH3 in HCECs leads to a remarkable increased frameshift mutations in [AAAG]17 repeats whereas [CA]13 repeats were less affected. Upon MSH3-silencing, significant changes in the expression of 202 proteins were detected. Pathway analysis revealed overexpression of proteins involved in double strand break repair (MRE11 and RAD50), apoptosis, L1 recycling, and repression of proteins involved in metabolism, tRNA aminoacylation, and gene expression. MSH3-silencing did not induce oncogenic transformation and DSBs increased 2-fold. CONCLUSIONS MSH3-deficiency in human colon epithelial cells results in EMAST, formation of DSBs and significant changes of the proteome but lacks oncogenic transformation. Thus, MSH3-deficiency alone is unlikely to drive human colon carcinogenesis.
Collapse
Affiliation(s)
- Christoph Campregher
- Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Gerald Schmid
- Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Franziska Ferk
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Siegfried Knasmüller
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Vineeta Khare
- Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Benedikt Kortüm
- Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Kyle Dammann
- Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Michaela Lang
- Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Theresa Scharl
- ACIB GmbH, c/o Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
- Department of Statistics and Probability Theory, University of Technology, Vienna, Austria
| | - Andreas Spittler
- Department of Surgery, Research Laboratories & Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Andres I. Roig
- Department of Cell Biology, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jerry W. Shay
- Department of Cell Biology, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christopher Gerner
- Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christoph Gasche
- Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| |
Collapse
|