201
|
Herreros-Villanueva M, Chen CC, Tsai EM, Er TK. Endometriosis-associated ovarian cancer: What have we learned so far? Clin Chim Acta 2019; 493:63-72. [PMID: 30776361 DOI: 10.1016/j.cca.2019.02.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023]
Abstract
Endometriosis is defined as the presence of ectopic endometrial tissue outside of the uterine cavity, most commonly in the ovaries and peritoneum. It is a complex disease that is influenced by multiple factors. It is also a common gynecological disorder and affects approximately 10-15% of all women of reproductive age. Recent molecular and pathological studies indicate that endometriosis may serve as a precursor of ovarian cancer (endometriosis-associated ovarian cancer, EAOC), particularly endometrioid and clear cell ovarian cancers. Although histological and epidemiological studies have demonstrated that endometriosis has a malignant potential, the molecular mechanism that underlies the malignant transformation of endometriosis is still controversial, and the precise mechanism of carcinogenesis must be fully elucidated. Currently, the development and improvement of a new sequencing technology, next-generation sequencing (NGS), has been increasingly relevant in cancer genomics research. Recently, NGS has also been utilized in clinical oncology to advance the personalized treatment of cancer. In addition, the sensitivity, speed, and cost make NGS a highly attractive platform compared to other sequencing modalities. For this reason, NGS may lead to the identification of driver mutations and underlying pathways associated with EAOC. Here, we present an overview of the molecular pathways that have led to the current opinions on the relationship between endometriosis and ovarian cancer.
Collapse
Affiliation(s)
- M Herreros-Villanueva
- Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastián, Spain
| | - Chih-Chieh Chen
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Rapid Screening Research Center for Toxicology and Biomedicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tze-Kiong Er
- Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung, Taiwan; Deparment of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan; Deparment of Biotechnology, Asia University, Taichung, Taiwan; Deparment of Nursing, Asia University, Taichung, Taiwan.
| |
Collapse
|
202
|
Bhalla A, Zulfiqar M, Bluth MH. Molecular Diagnostics in Colorectal Carcinoma: Advances and Applications for 2018. Clin Lab Med 2019; 38:311-342. [PMID: 29776633 DOI: 10.1016/j.cll.2018.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The molecular pathogenesis and classification of colorectal carcinoma are based on the traditional adenomaecarcinoma sequence, serrated polyp pathway, and microsatellite instability (MSI). The genetic basis for hereditary nonpolyposis colorectal cancer is the detection of mutations in the MLH1, MSH2, MSH6, PMS2, and EPCAM genes. Genetic testing for Lynch syndrome includes MSI testing, methylator phenotype testing, BRAF mutation testing, and molecular testing for germline mutations in MMR genes. Molecular makers with predictive and prognostic implications include quantitative multigene reverse transcriptase polymerase chain reaction assay and KRAS and BRAF mutation analysis. Mismatch repair-deficient tumors have higher rates of programmed death-ligand 1 expression. Cell-free DNA analysis in fluids are proving beneficial for diagnosis and prognosis in these disease states towards effective patient management.
Collapse
Affiliation(s)
- Amarpreet Bhalla
- Department of Pathology and Anatomical Sciences, Jacobs School of Buffalo, Buffalo, NY 14203, USA.
| | | | - Martin H Bluth
- Department of Pathology, Wayne State University School of Medicine, 540 East Canfield Street, Detroit, MI 48201, USA; Pathology Laboratories, Michigan Surgical Hospital, 21230 Dequindre Road, Warren, MI 48091, USA
| |
Collapse
|
203
|
Situ Y, Chung L, Lee CS, Ho V. MRN (MRE11-RAD50-NBS1) Complex in Human Cancer and Prognostic Implications in Colorectal Cancer. Int J Mol Sci 2019; 20:E816. [PMID: 30769804 PMCID: PMC6413120 DOI: 10.3390/ijms20040816] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023] Open
Abstract
The MRE11-RAD50-NBS1 (MRN) complex has been studied in multiple cancers. The identification of MRN complex mutations in mismatch repair (MMR)-defective cancers has sparked interest in its role in colorectal cancer (CRC). To date, there is evidence indicating a relationship of MRN expression with reduced progression-free survival, although the significance of the MRN complex in the clinical setting remains controversial. In this review, we present an overview of the function of the MRN complex, its role in cancer progression, and current evidence in colorectal cancer. The evidence indicates that the MRN complex has potential utilisation as a biomarker and as a putative treatment target to improve outcomes of colorectal cancer.
Collapse
Affiliation(s)
- Yiling Situ
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia.
| | - Liping Chung
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia.
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia.
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia.
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia.
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia.
- Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia.
- Faculty of Medicine, South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia.
- Faculty of Medicine and Health, Central Clinical School, University of Sydney, Camperdown, NSW 2050, Australia.
| | - Vincent Ho
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia.
| |
Collapse
|
204
|
Yurgelun MB, Kastrinos F. Tumor Testing for Microsatellite Instability to Identify Lynch Syndrome: New Insights Into an Old Diagnostic Strategy. J Clin Oncol 2019; 37:263-265. [PMID: 30550362 DOI: 10.1200/jco.18.01664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Matthew B Yurgelun
- 1 Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.,2 Brigham & Women's Hospital, Boston, MA
| | - Fay Kastrinos
- 3 Columbia University Irving Medical Center and the Herbert Irving Comprehensive Cancer Center, New York, NY.,4 Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
205
|
Raynes Y, Weinreich DM. Genomic clustering of fitness-affecting mutations favors the evolution of chromosomal instability. Evol Appl 2019; 12:301-313. [PMID: 30697341 PMCID: PMC6346662 DOI: 10.1111/eva.12717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/16/2018] [Accepted: 09/16/2018] [Indexed: 12/19/2022] Open
Abstract
Most solid cancers are characterized by chromosomal instability (CIN)-an elevated rate of large-scale chromosomal aberrations and ploidy changes. Chromosomal instability may arise through mutations in a range of genomic integrity loci and is commonly associated with fast disease progression, poor prognosis, and multidrug resistance. However, the evolutionary forces promoting CIN-inducing alleles (hereafter, CIN mutators) during carcinogenesis remain poorly understood. Here, we develop a stochastic, individual-based model of indirect selection experienced by CIN mutators via genomic associations with fitness-affecting mutations. Because mutations associated with CIN affect large swaths of the genome and have the potential to simultaneously comprise many individual loci, we show that indirect selection on CIN mutators is critically influenced by genome organization. In particular, we find strong support for a key role played by the spatial clustering of loci with either beneficial or deleterious mutational effects. Genomic clustering of selected loci allows CIN mutators to generate favorable chromosomal changes that facilitate their rapid expansion within a neoplasm and, in turn, accelerate carcinogenesis. We then examine the distribution of oncogenic and tumor-suppressing loci in the human genome and find both to be potentially more clustered along the chromosome than expected, leading us to speculate that human genome may be susceptible to CIN hitchhiking. More quantitative data on fitness effects of individual mutations will be necessary, though, to assess the true levels of clustering in the human genome and the effectiveness of indirect selection for CIN. Finally, we use our model to examine how therapeutic strategies that increase the deleterious burden of genetically unstable cells by raising either the rate of CIN or the cost of deleterious mutations affect CIN evolution. We find that both can inhibit CIN hitchhiking and delay carcinogenesis in some circumstances, yet, in line with earlier work, we find the latter to be considerably more effective.
Collapse
Affiliation(s)
- Yevgeniy Raynes
- Department of Ecology and Evolutionary Biology, Center for Computational Molecular BiologyBrown UniversityProvidenceRhode Island
| | - Daniel M. Weinreich
- Department of Ecology and Evolutionary Biology, Center for Computational Molecular BiologyBrown UniversityProvidenceRhode Island
| |
Collapse
|
206
|
Liu F, Tong T, Huang D, Yuan W, Li D, Lin J, Cai S, Xu Y, Chen W, Sun Y, Zhuang J. CapeOX perioperative chemotherapy versus postoperative chemotherapy for locally advanced resectable colon cancer: protocol for a two-period randomised controlled phase III trial. BMJ Open 2019; 9:e017637. [PMID: 30700474 PMCID: PMC6352769 DOI: 10.1136/bmjopen-2017-017637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Adjuvant chemotherapy with the CapeOX regimen is now widely used for treating colorectal cancer. However, prior studies have demonstrated better efficacy of pre-operative/neoadjuvant chemotherapy without increase of safety risks. METHODS AND ANALYSIS This multicentre, open-label, parallel-group, randomised, controlled, phase III study aims to compare the efficacy and safety of perioperative CapeOX chemotherapy with the postoperative one for treating patients with locally advanced R0 resectable colon cancers in China. In total 1370 eligible patients will be randomised to: the test group, up to four cycles (every 3 weeks is a cycle, Q3W) of chemotherapy plus radical surgery plus up to four cycles of post-operative chemotherapy; or the control group, radical surgery first, then up to eight cycles of chemotherapy. In each cycle, oxaliplatin will be given at a dose of 130 mg/m2 through continuous IV infusion for 2 hours on the first day. From day 1 to day 14, capecitabine will be taken orally every morning and evening at a dose of 1000mg/m2/d. The primary outcome measure is the 3-year disease free survival. The objective response rate, R0 resection rate, overall survival, as well as the adverse events will also be measured as second endpoints. The study may include two periods. If results of period 1 are not favourable, period 2 will be initiated, recruiting genetically sensitive patients and repeating the same process with period 1. ETHICS AND DISSEMINATION Informed consent will be required from, and provided, by all subjects. The study protocol has been approved by the independent ethics committee of Shanghai Fudan University Cancer Centre. This study will clearly demonstrate the potential benefit of perioperative chemotherapy with the CapeOX regimen. Results will be shared among all the participating centres, and with policymakers and the academic community to promote the clinical management of colon cancer. TRIAL REGISTRATION NUMBER NCT03125980.
Collapse
Affiliation(s)
- Fangqi Liu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department Of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tong Tong
- Department Of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Dan Huang
- Department Of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Weitang Yuan
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dechuan Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jianjiang Lin
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department Of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department Of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenbin Chen
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yueming Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Zhuang
- Department of General Surgery, Zhengzhou University Cancer Hospital, Zhengzhou, China
| |
Collapse
|
207
|
Gandhi JS, Goswami M, Sharma A, Tanwar P, Gupta G, Gupta N, Pasricha S, Mehta A, Singh S, Agarwal M, Gupta N. Clinical Impact of Mismatch Repair Protein Testing on Outcome of Early Staged Colorectal Carcinomas. J Gastrointest Cancer 2019; 49:406-414. [PMID: 28585041 DOI: 10.1007/s12029-017-9954-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Colorectal cancer is the third most common cancer in men and second most common in women globally. In the present study, we aimed to analyse the proportion of patients with loss of immunostaining for mismatch repair (MMR) proteins in all newly diagnosed stage II cases of colorectal cancer for the purpose of prognostication, for determination of further chemotherapeutic strategy and for familial screening. METHOD From January 2014 to December 2015, 62 consecutive newly diagnosed cases of stage II colorectal cancer were included in the study. Details of each patient related to their demographic profile and tumour profile were recorded. All the cases were grossed and staged according to College of American Pathologist (CAP) guidelines. The expression of MMR proteins (which was earlier validated on normal as well as tumour tissue) in FFPE tumour tissue using IHC for mut L homologue 1 (MLH1), mut S homologue 2 (MSH2), mut S homologue 6 (MSH6) and post-meiotic segregation increased 2 (PMS2) was studied. Information regarding stage, treatment, clinical outcome and overall survival was retrieved when available. RESULTS Out of a total of 371 cases, 62 (16.7%) cases were of stage II CRC, out of which 43 (12%) were treatment naive. Among the selected 62 cases, 26 (41.9%) demonstrated loss of MMR proteins and 36 (58.0%) cases had intact nuclear expression. Out of the cases with MMR loss, 38.4% showed loss of MLH1 and PMS2, 30.7% showed loss of MSH2 and MSH6, 26.9% showed isolated loss of PMS2 and 3.8% showed isolated loss of MSH6. Right-sided location (57.6%) was more common than left-sided (19.2%) and transverse colon (23.0%). Majority of the cases were moderately differentiated (65.3%) in morphology. There was no intratumoural infiltrate in most of the cases (53.8%), and only 3.8% cases showed marked intratumoural infiltrate. Also, peritumoural lymphocytic infiltrate was mild to moderate in most of the cases (26.9%) and marked Crohn's-like infiltrate was seen in only 7.6% cases. CONCLUSION Our study shows that the routine evaluation of MMR proteins is achievable and essential for the purpose of prognostication, planning of treatment strategies and ascertaining a hereditary basis of CRC. The incidence of MMR protein loss was quite high in our study compared to other studies probably due to a difference in ethnicity. Though a right-sided predominance was supported, none of the typical morphological features of microsatellite instability (MSI) tumours were substantiated by our study, highlighting the lack of importance of histology for predicting MSI, and emphasising the point that MSI testing should be done as a routine procedure in all stage II CRC. A short follow-up was done for all our cases and comparison between the survival of the chemotherapy treated MSI cases versus those which were treatment naïve was performed and revealed that chemotherapy (CT) did not provide additional benefit to survival; MSI tumours in general are a better prognostic category and do not require additional chemotherapy.
Collapse
Affiliation(s)
- Jatin Sundersham Gandhi
- Department of Histopathology and Cytopathology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India.
| | - Malini Goswami
- Department of Histopathology and Cytopathology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Anila Sharma
- Department of Histopathology and Cytopathology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Parul Tanwar
- Department of Histopathology and Cytopathology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Gurudutt Gupta
- Department of Histopathology and Cytopathology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Nikhil Gupta
- Department of Gastrointestinal Surgery, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Sunil Pasricha
- Department of Histopathology and Cytopathology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Anurag Mehta
- Department of Histopathology and Cytopathology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Shivender Singh
- Department of Gastrointestinal Surgery, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Mohit Agarwal
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| | - Nitin Gupta
- Department of Gastroenterology, Rajiv Gandhi Cancer Institute and Research Centre, Sector 5, Rohini, New Delhi, India
| |
Collapse
|
208
|
Lehrer S, Rheinstein PH. Inverse Relationship Between Polio Incidence in the US and Colorectal Cancer. In Vivo 2019; 32:1485-1489. [PMID: 30348706 DOI: 10.21873/invivo.11404] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIM Polio is predominantly an enteric viral infection that was progressively eradicated in the United States after the introduction of polio vaccine in the early 1950s. U.S. colorectal cancer rates have dropped steadily for individuals born between 1890 and 1950, but have been increasing for every generation born since 1950. Moreover, the lowest worldwide age adjusted rates of colorectal cancer in 2012 were in sub-Saharan Africa, Gambia and Mozambique, where polio has not been eradicated. In the current study, poliomyelitis incidence in US states before the introduction of polio vaccine was analyzed. MATERIALS AND METHODS Reported cases of poliomyelitis per 100,000 population by state 1932-1951 were from Centers for Disease Control. Colorectal cancer deaths per 100,000 in men (2005-2009) by US State are from the American Cancer Society. US state overweight and obesity data are from the Centers for Disease Control and Prevention (CDC). Smoking data are from the CDC. RESULTS By US state, colorectal cancer incidence per 100,000 in men for 2005-2009 was inversely correlated with reported cases of poliomyelitis per 100,000 for 1932-1951 (r=-0.311, p=0.032). Colorectal cancer deaths per 100,000 in men in 2005-2009 were also inversely correlated with reported cases of poliomyelitis per 100,000 by state for 1932-1951 (r=-0.493, p<0.001). The relationship between colorectal cancer deaths and polio incidence was significant (β=-0.196, p=0.028) and independent of the effects of smoking (β=0.289, p=0.012) and overweight (β=0.547, p<0.001). The relationship in females with colorectal cancer was identical. CONCLUSION Polio virus infection of cells of the colon may induce some degree of resistance to the development of colon cancer decades later. The effect of polio virus infection seems to be especially potent in reducing the rate of death from colon cancer.
Collapse
Affiliation(s)
- Steven Lehrer
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, U.S.A.
| | | |
Collapse
|
209
|
Next Generation Sequencing (NGS): A Revolutionary Technology in Pharmacogenomics and Personalized Medicine in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1168:9-30. [DOI: 10.1007/978-3-030-24100-1_2] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
210
|
Kim K, Castro EJT, Shim H, Advincula JVG, Kim YW. Differences Regarding the Molecular Features and Gut Microbiota Between Right and Left Colon Cancer. Ann Coloproctol 2018; 34:280-285. [PMID: 30630301 PMCID: PMC6347335 DOI: 10.3393/ac.2018.12.17] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022] Open
Abstract
For many years, developmental and physiological differences have been known to exist between anatomic segments of the colorectum. Because of different outcomes, prognoses, and clinical responses to chemotherapy, the distinction between right colon cancer (RCC) and left colon cancer (LCC) has gained attention. Furthermore, variations in the molecular features and gut microbiota between right and LCCs have recently been a hot research topic. CpG island methylator phenotype-high, microsatellite instability-high colorectal cancers are more likely to occur on the right side whereas tumors with chromosomal instability have been detected in approximately 75% of LCC patients and 30% of RCC patients. The mutation rates of oncogenes and tumor suppressor genes also differ between RCC and LCC patients. Biofilm is more abundant in RCC patients than LLC patients, as are Prevotella, Selenomonas, and Peptostreptococcus. Conversely, Fusobacterium, Escherichia/Shigella, and Leptotrichia are more abundant in LCC patients compared to RCC patients. Distinctive characteristics are apparent in terms of molecular features and gut microbiota between right and LCC. However, how or to what extent these differences influence diverging oncologic outcomes remains unclear. Further clinical and translational studies are needed to elucidate the causative relationship between primary tumor location and prognosis.
Collapse
Affiliation(s)
- Kwangmin Kim
- Big Data Research Group, Yonsei University Wonju College of Medicine, Wonju, Korea.,Division of Acute Care Surgery, Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | | | - Hongjin Shim
- Division of Trauma Surgery and Surgical Critical Care, Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | | | - Young-Wan Kim
- Big Data Research Group, Yonsei University Wonju College of Medicine, Wonju, Korea.,Division of Colorectal Surgery, Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
211
|
Digiacomo N, Bolzacchini E, Veronesi G, Cerutti R, Sahnane N, Pinotti G, Bregni M, Artale S, Verusio C, Crivelli F, Capella C, Sessa F, Furlan D. Neuroendocrine Differentiation, Microsatellite Instability, and Tumor-infiltrating Lymphocytes in Advanced Colorectal Cancer With BRAF Mutation. Clin Colorectal Cancer 2018; 18:e251-e260. [PMID: 30638691 DOI: 10.1016/j.clcc.2018.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Approximately 10% of metastatic colorectal cancer (mCRC) cases will harbor the BRAF p.V600E mutation (BRAF-mCRC) and have been associated with a poor prognosis. Although they are usually considered a unique clinical entity, biologic heterogeneity has been described. We performed an extensive clinicopathologic study of a multicenter series of BRAF-mCRC to highlight differences between tumors with microsatellite instability (MSI) and microsatellite stable tumors, focusing on both inflammatory profiles and neuroendocrine differentiation. METHODS We included 59 BRAF-mCRC cases and collected the clinical data (ie, surgery, treatment, and follow-up). We evaluated MSI status, budding, lympho-angioinvasion, neuroinvasion, extent of active stroma, CD3+ and CD8+ intratumoral and peritumoral lymphocytes, programmed cell death ligand 1, p53, Ki-67, synaptophysin, and CDX2 expression. RESULTS The 22 MSI BRAF-mCRC cases were associated with the right side (P < .0001), an expansive grown pattern (P < .01), programmed cell death ligand 1 expression (P < .0001), high CD8 T-cell content (P = .0001), and lymph node metastases (P < .029). The 37 MSS BRAF-mCRC cases were characterized by a greater stromal component (P = .0002), pulmonary metastases (P = .095), and p53 and synaptophysin immunoreactivity (P = .004 and P = .001, respectively). Univariate analysis demonstrated that MSI and a high CD8 T-cell content were associated with a 34% (hazard ratio [HR], 0.66; 95% confidence interval [CI], 0.34-1.28; P = .2) and 33% (HR, 0.67; 95% CI, 0.45-0.99; P = .04) reduction in the risk of death, respectively. The combined presence of MSI and CD8 T-cell content decreased the hazard of mortality ≤ 63% (HR, 0.37; 95% CI, 0.14-0.97; P = .2), which was slightly reduced after multivariate analysis. CONCLUSION A simultaneous evaluation of MSI, CD8 T-cell content, and neuroendocrine markers could allow for the identification of subsets of BRAF-mCRC with a different prognosis and potential eligibility for specific treatments.
Collapse
Affiliation(s)
- Nunzio Digiacomo
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | - Giovanni Veronesi
- Research Center in Epidemiology and Preventive Medicine (EPIMED), Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Roberta Cerutti
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Nora Sahnane
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | | | - Marco Bregni
- Unit of Oncology, ASST Valle Olona, Varese, Italy
| | | | | | | | - Carlo Capella
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Daniela Furlan
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy; Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
212
|
Baudrin LG, Deleuze JF, How-Kit A. Molecular and Computational Methods for the Detection of Microsatellite Instability in Cancer. Front Oncol 2018; 8:621. [PMID: 30631754 PMCID: PMC6315116 DOI: 10.3389/fonc.2018.00621] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/30/2018] [Indexed: 12/31/2022] Open
Abstract
Microsatellite instability (MSI) is a genomic alteration in which microsatellites, usually of one to four nucleotide repeats, accumulate mutations corresponding to deletions/insertions of a few nucleotides. The MSI phenotype has been extensively characterized in colorectal cancer and is due to a deficiency of the DNA mismatch repair system. MSI has recently been shown to be present in most types of cancer with variable frequencies (from <1 to 30%). It correlates positively to survival outcome and predicts the response to immune checkpoint blockade therapy. The different methods developed for MSI detection in cancer require taking into consideration two critical parameters which influence method performance. First, the microsatellite markers used should be chosen carefully to ensure they are highly sensitive and specific for MSI detection. Second, the analytical method used should be highly resolute to allow clear identification of MSI and of the mutant allele genotype, and should present the lowest limit of detection possible for application in samples with low mutant allele frequency. In this review, we describe all the different molecular and computational methods developed to date for the detection of MSI in cancer, how they have evolved and improved over the years, and their advantages and drawbacks.
Collapse
Affiliation(s)
- Laura G Baudrin
- Laboratoire de Génomique, Fondation Jean Dausset-CEPH, Paris, France.,Laboratoire d'Excellence GenMed Paris, Paris, France
| | - Jean-François Deleuze
- Laboratoire de Génomique, Fondation Jean Dausset-CEPH, Paris, France.,Centre National de Recherche en Génomique Humaine, CEA-Institut François Jacob, Evry, France
| | - Alexandre How-Kit
- Laboratoire de Génomique, Fondation Jean Dausset-CEPH, Paris, France
| |
Collapse
|
213
|
[Predictive and prognostic value of MSI phenotype in adjuvant colon cancer: Who and how to treat?]. Bull Cancer 2018; 106:129-136. [PMID: 30527814 DOI: 10.1016/j.bulcan.2018.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/06/2018] [Accepted: 10/25/2018] [Indexed: 12/31/2022]
Abstract
The MSI phenotype in colon cancer is a good prognostic factor, with an impact probably more pronounced for stage II than stage III tumor. This survival advantage may be related to the tumor-infiltrating lymphocytes observed in MSI tumors, thus explaining the existence of a probably more effective anti-tumor immune response. In addition, the MSI status would also be a biomarker able to predict the lack of efficacy of adjuvant 5-fluorouracil (5FU) chemotherapy. In contrast, as observed in MSS colon cancer, the MSI tumors would have a survival benefit with the addition of oxaliplatin to adjuvant 5FU chemotherapy. Based on these data, the "French National Thesaurus of Digestive Oncology" suggests for patients with MSI colon cancer, an adjuvant chemotherapy combining fluoropyrimidine and oxaliplatin for stage III, and surgery alone without adjuvant chemotherapy for stage II (excepted for pT4b tumors in which the combination of fluoropyrimidine and oxaliplatin may be a therapeutic option). Beyond these recommendations, the discussion of adjuvant treatment in MSI tumors should also include other factors such as the patient's age and comorbidities. The duration of the adjuvant treatment (3 or 6 months) and the regimen used (FOLFOX or XELOX) should be based on the recommendations of the international IDEA consortium pending the results of the translational studies of this trial. Finally, the promising results of immunotherapy in metastatic MSI colorectal led to the development of clinical trials evaluating "immune checkpoint blockers" in combination with FOLFOX in the treatment of stage III MSI colon cancer.
Collapse
|
214
|
Papke DJ, Nowak JA, Yurgelun MB, Frieden A, Srivastava A, Lindeman NI, Sholl LM, MacConaill LE, Dong F. Validation of a targeted next-generation sequencing approach to detect mismatch repair deficiency in colorectal adenocarcinoma. Mod Pathol 2018; 31:1882-1890. [PMID: 29955144 DOI: 10.1038/s41379-018-0091-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/11/2018] [Accepted: 05/13/2018] [Indexed: 12/30/2022]
Abstract
Mismatch repair protein deficiency is a hallmark of cancers associated with Lynch syndrome and is a biomarker for response to immunotherapy. With the increasing adoption of cancer next-generation sequencing, there has been a movement to develop screening approaches that take advantage of the unique mutational signatures of mismatch repair-deficient tumors. Here, we develop a sequencing-based metric that distinguishes mismatch repair-deficient from mismatch repair-proficient colorectal adenocarcinomas with comparison to immunohistochemical staining. We find that a single criterion of three or more single base pair insertion or deletion mutations per megabase sequenced, occurring in mononucleotide repeat regions of four or more nucleotides, is sufficient to detect mismatch repair deficiency with 96% sensitivity and 100% specificity in a training set of 241 cancers and 96% sensitivity and 99% specificity in a validation set of 436 additional cancers. Using data from the same cohort, we also find that sequencing information from only three genes-ARID1A, KMT2D, and SOX9-is sufficient to detect mismatch repair-deficient colorectal adenocarcinomas with 76% sensitivity and 98% specificity in the validation set. These findings support the notion that targeted next-generation sequencing already being performed for clinical or research purposes can also be used to accurately detect mismatch repair deficiency in colorectal adenocarcinomas.
Collapse
Affiliation(s)
- David J Papke
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan A Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alexander Frieden
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amitabh Srivastava
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura E MacConaill
- Center for Cancer Genome Discovery, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
215
|
Yurgelun MB, Hampel H. Recent Advances in Lynch Syndrome: Diagnosis, Treatment, and Cancer Prevention. Am Soc Clin Oncol Educ Book 2018; 38:101-109. [PMID: 30231390 DOI: 10.1200/edbk_208341] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Identification of individuals with inherited predispositions to cancer, including Lynch syndrome, can help prevent cancer and cancer-related death by allowing for the uptake of specific cancer prevention and screening as well as the use of therapies directed toward the underlying neoplastic process for individuals with advanced cancer. In the 25 years since the discovery of microsatellite instability (MSI) and the first recognition of germline mismatch repair (MMR) gene variants as the etiologic basis of Lynch syndrome, there has been tremendous progress in the understanding of the spectrum of cancer risk associated with Lynch syndrome as well as in cancer prevention and risk-reduction strategies. The past few years, in particular, have brought transformative changes in the treatment of Lynch syndrome-associated cancers with immune checkpoint inhibitors. In parallel, advances in next-generation sequencing (NGS) technologies now allow rapid and scalable somatic and germline sequencing that promises to help identify Lynch syndrome in individuals who otherwise lack classic phenotypes. Last, real progress is being made to understand more sophisticated methods of precision cancer prevention, including chemotherapeutic prevention agents (e.g., aspirin) and strategies that leverage the immune system to facilitate primary cancer prevention in otherwise-healthy Lynch syndrome carriers.
Collapse
Affiliation(s)
- Matthew B Yurgelun
- From the Dana-Farber Cancer Institute, Brigham & Women's Hospital, and Harvard Medical School, Boston, MA; The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Heather Hampel
- From the Dana-Farber Cancer Institute, Brigham & Women's Hospital, and Harvard Medical School, Boston, MA; The Ohio State University Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
216
|
How the BRAF V600E Mutation Defines a Distinct Subgroup of Colorectal Cancer: Molecular and Clinical Implications. Gastroenterol Res Pract 2018; 2018:9250757. [PMID: 30598662 PMCID: PMC6287148 DOI: 10.1155/2018/9250757] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
The BRAF oncogene is an integral component of the MAP kinase pathway, and an activating V600E mutation occurs in 15% of sporadic colorectal cancer. This is an early event in serrated pathway tumourigenesis, and the BRAF V600E has been commonly associated with the CpG island methylator phenotype, microsatellite instability (MSI), and a consistent clinical presentation including a proximal location and predilection for elderly females. A proportion of the BRAF mutant lesions remain as microsatellite stable (MSS), and in contrast to the MSI cancers, they have an aggressive phenotype and correlate with poor patient outcomes. Recent studies have found that they have clinical and molecular features of both the BRAF mutant/MSI and the conventional BRAF wild-type cancers and comprise a distinct colorectal cancer subgroup. This review highlights the importance of the BRAF mutation occurring in colorectal cancer stratified for molecular background and discusses its prognostic and clinical significance.
Collapse
|
217
|
Complexity of genome sequencing and reporting: Next generation sequencing (NGS) technologies and implementation of precision medicine in real life. Crit Rev Oncol Hematol 2018; 133:171-182. [PMID: 30661654 DOI: 10.1016/j.critrevonc.2018.11.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/23/2018] [Indexed: 12/17/2022] Open
Abstract
The finalization of the Human Genome Project in 2003 paved the way for a deeper understanding of cancer, favouring a faster progression towards "personalized" medicine. Research in oncology has progressively focused on the sequencing of cancer genomes, to better understand the genetic basis of tumorigenesis and identify actionable alterations to guide cancer therapy. Thanks to the development of next-generation-sequencing (NGS) techniques, sequencing of tumoral DNA is today technically easier, faster and cheaper. Commercially available NGS panels enable the detection of single or global genomic alterations, namely gene mutation and mutagenic burden, both on germline and somatic DNA, potentially predicting the response or resistance to cancer treatments. Profiling of tumor DNA is nowadays a standard in cancer research and treatment. In this review we discuss the history, techniques and applications of NGS in cancer care, under a "personalized tailored therapy" perspective.
Collapse
|
218
|
Hissong E, Crowe EP, Yantiss RK, Chen YT. Assessing colorectal cancer mismatch repair status in the modern era: a survey of current practices and re-evaluation of the role of microsatellite instability testing. Mod Pathol 2018; 31:1756-1766. [PMID: 29955148 DOI: 10.1038/s41379-018-0094-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 01/05/2023]
Abstract
Results of DNA mismatch repair testing are used to detect Lynch syndrome and have prognostic and therapeutic implications among patients with sporadic colorectal carcinomas. Immunohistochemistry for mismatch repair proteins (MLH1, PMS2, MSH2, MSH6) and PCR for microsatellite instability are two established methods for assessing mismatch repair function. Older literature suggested a discordance rate of approximately 5% between these assays, leading some institutions to perform dual testing on all cases. Although universal mismatch repair testing is now recommended by multiple professional organizations, none provide guidelines regarding preferred assays. We surveyed 96 academic and nonacademic institutions to assess Lynch syndrome screening practices and evaluated discordance rates between immunohistochemistry and PCR among 809 colorectal cancers tested in our own institution. Our survey demonstrated no significant differences between academic and nonacademic practices with respect to testing strategies. Eighty six percent performed universal screening, and usually (76%) employed immunohistochemistry on initial biopsy samples. Only 20% employed PCR; these were mostly academic practices that used both immunohistochemistry and PCR (p < 0.01 compared with the nonacademic groups). Loss of MLH1/PMS2 staining was often (90%) followed by either BRAF mutational analysis or MLH1 methylation assays. Only 24% adhered to WHO recommendations to assign histologic grade based on mismatch repair status. We found only 3 cases (0.4%) with discordant immunohistochemistry and PCR results in our own practice: 1 reflected decreased MSH-6 staining in a neoadjuvantly treated microsatellite stable tumor, 1 MLH1-deficient tumor showed diminished MLH1/PMS2 in the tumor compared with internal control, and 1 case reflected an error in the molecular laboratory. Overall, our results showed extremely low discordance between methods assessing mismatch repair status and would suggest immunohistochemistry as the preferred single screening test. PCR can be reserved for cases that show equivocal immunostaining patterns.
Collapse
Affiliation(s)
- Erika Hissong
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Elizabeth P Crowe
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Rhonda K Yantiss
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Yao-Tseng Chen
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
219
|
Wong HL, Christie M, Gately L, Tie J, Lee B, Semira C, Lok SW, Wong R, Gibbs P. Mismatch repair deficiency assessment by immunohistochemistry: for Lynch syndrome screening and beyond. Future Oncol 2018; 14:2725-2739. [DOI: 10.2217/fon-2018-0319] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
While mismatch repair (MMR) deficiency has been studied extensively, the assessment of MMR status in colorectal and other cancers remains highly relevant, particularly in light of recent data demonstrating that MMR deficiency is a strong predictor for treatment benefit with immune checkpoint inhibitors across multiple tumor types. In colorectal cancer, there is a growing consensus in support of routine MMR testing for Lynch syndrome screening, to inform prognosis and adjuvant chemotherapy use in early stage disease, and to predict response to immunotherapy in advanced disease. Here, we provide a review of the Ventana MMR Immunohistochemistry Panel, which was recently approved by the US FDA for use in Lynch syndrome screening.
Collapse
Affiliation(s)
- Hui-li Wong
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Christie
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Pathology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Lucy Gately
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, St. Vincent's Health, Fitzroy, Victoria, Australia
| | - Jeanne Tie
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Department of Medical Oncology, Western Health Medical School, University of Melbourne, Footscray, Victoria, Australia
| | - Belinda Lee
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Department of Medical Oncology, Northern Health, Epping, Victoria, Australia
| | - Christine Semira
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Sheau Wen Lok
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Rachel Wong
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, Eastern Health, Box Hill, Victoria, Australia
| | - Peter Gibbs
- Systems Biology & Personalised Medicine Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, Western Health Medical School, University of Melbourne, Footscray, Victoria, Australia
| |
Collapse
|
220
|
Salman P, Panay S, Fernández R, Mahave M, Soza-Ried C. Evidence of response to pembrolizumab in a patient with Lynch syndrome-related metastatic colon cancer. Onco Targets Ther 2018; 11:7295-7300. [PMID: 30425520 PMCID: PMC6205820 DOI: 10.2147/ott.s167645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patients with Lynch Syndrome (LS) are at high risk of developing colorectal cancer at an early age. Germline mutations in DNA mismatch repair genes and microsatellite instability are clear signatures of this autosomal dominant disorder. Here, we report the clinical history of a 38-year-old patient with LS-related metastatic colon cancer treated in Chile with immunotherapy (pembrolizumab). The patient exhibited a pathogenic deletion in Epithelial cell Adhesion Molecule (EPCAM) and mutS homolog 2 (MSH2) genes, and after diagnosis received 12 cycles of FOLFOX. The tumor mass, however, continued to grow, and a new metastatic mucinous adenocarcinoma of 13 mm appeared at the level of the 11th right dorsal vertebra. To treat these lesions, the patient received immunotherapy scheme with pembrolizumab (200 mg every 21 days). After only four cycles, the patient’s symptoms improved and the lesions showed less metabolic activity. After 12 cycles with pembrolizumab, the patient started palliative radiation and systemic second-line treatment with FOLFIRI and Avastin. The immunotherapy scheme with pembrolizumab was capable of delaying the second-line treatment for at least 8 months, becoming a useful therapeutic option for this patient. Thus, our study highlights the importance of implementing immunotherapy treatment programs for LS-colorectal cancer patients in South American countries.
Collapse
Affiliation(s)
- Pamela Salman
- Department of Medical Oncology, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile,
| | - Sergio Panay
- Department of Medical Oncology, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile,
| | - René Fernández
- Department of Nuclear Medicine, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Mauricio Mahave
- Department of Medical Oncology, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile,
| | - Cristian Soza-Ried
- Department of Medical Oncology, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile,
| |
Collapse
|
221
|
Bando H, Okamoto W, Fukui T, Yamanaka T, Akagi K, Yoshino T. Utility of the quasi-monomorphic variation range in unresectable metastatic colorectal cancer patients. Cancer Sci 2018; 109:3411-3415. [PMID: 30142704 PMCID: PMC6215873 DOI: 10.1111/cas.13774] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/22/2018] [Accepted: 08/09/2018] [Indexed: 11/26/2022] Open
Abstract
Microsatellite Instability (MSI) status is an established predictive biomarker for the treatment of the anti-programmed death 1 (PD-1) antibody. The current approach to determine the MSI status in tumours requires matched normal DNA. Some mononucleotide microsatellite markers are known to have few variant alleles in both Caucasians and Asians. Therefore, the length of these microsatellite makers is almost confined within the quasi-monomorphic variation range (QMVR). Considering the application of MSI testing for various types of cancers, a simple, sensitive and inexpensive method is desired. This study assessed the clinical utility of the QMVR for determining the MSI status in patients with unresectable metastatic colorectal cancer (mCRC). The study enrolled 435 patients with mCRC. The concordance of the MSI status in mCRC between the standard method using tumour DNA plus matched normal DNA and the testing method using only tumour DNA was evaluated. Eleven (2.5%) MSI-high cases were detected by both the standard and testing methods. The sensitivity and specificity of the testing method were both 100%, indicating complete concordance between the methods. Among the mononucleotide markers, three and two patients showed discordance for NR-21 and BAT-25, respectively. Results from MSI testing with normal tissue indicated that four of five patients had rare germline variants outside the QMVR. For BAT-26, NR-24 and MONO-27, all patients showed complete concordance. Using the QMVR, the MSI status of mCRC can be determined without matched normal DNA.
Collapse
Affiliation(s)
- Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Wataru Okamoto
- Biobank Translational Research Support Section, Translational Research Management Division, Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takafumi Fukui
- Biomedical Business Department, FALCO Biosystems, Kyoto, Japan
| | - Takeharu Yamanaka
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kiwamu Akagi
- Division of Molecular Diagnosis, Saitama Cancer Center, Saitama, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
222
|
Sato K, Kawazu M, Yamamoto Y, Ueno T, Kojima S, Nagae G, Abe H, Soda M, Oga T, Kohsaka S, Sai E, Yamashita Y, Iinuma H, Fukayama M, Aburatani H, Watanabe T, Mano H. Fusion Kinases Identified by Genomic Analyses of Sporadic Microsatellite Instability-High Colorectal Cancers. Clin Cancer Res 2018; 25:378-389. [PMID: 30279230 DOI: 10.1158/1078-0432.ccr-18-1574] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/31/2018] [Accepted: 09/27/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Colorectal cancers with microsatellite instability-high (MSI-H) status, due to mismatch repair deficiency, are associated with poor patient outcomes after relapse. We aimed to identify novel therapeutic targets for them. EXPERIMENTAL DESIGN We performed MSI analyses of over 2,800 surgically resected colorectal tumors obtained from consecutive patients treated in Japan from 1998 through June 2016. Whole-exome sequencing, transcriptome sequencing, and methylation analyses were performed on 149 of 162 tumors showing MSI in BAT25 and BAT26 loci. We analyzed patient survival times using Bonferroni-adjusted log-rank tests. RESULTS Sporadic MSI-H colorectal cancers with promoter methylation of MLH1 (called MM) had a clinicopathological profile that was distinct from that of colorectal cancers of patients with germline mutations (Lynch syndrome, LS-associated) or somatic, Lynch-like mutations in mismatch repair genes. MM tumors had more insertions and deletions and more recurrent mutations in BRAF and RNF43 than LS-associated or Lynch-like MSI-H tumors. Eleven fusion kinases were exclusively detected in MM MSI-H colorectal cancers lacking oncogenic KRAS/BRAF missense mutations and were associated with worse post-relapse prognosis. We developed a simple method to identify MM tumors and applied it to a validation cohort of 28 MSI-H colorectal cancers, identifying 16 MM tumors and 2 fusion kinases. CONCLUSIONS We discovered that fusion kinases are frequently observed among sporadic MM MSI-H colorectal cancers. The new method to identify MM tumors enables us to straightforwardly group MSI-H patients into candidates of LS or fusion kinase carriers.
Collapse
Affiliation(s)
- Kazuhito Sato
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahito Kawazu
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Yoko Yamamoto
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihide Ueno
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinya Kojima
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Genta Nagae
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Abe
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Manabu Soda
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takafumi Oga
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinji Kohsaka
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eirin Sai
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Yamashita
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hisae Iinuma
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Watanabe
- Department of Surgical Oncology and Vascular Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Mano
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
223
|
Cheah PL, Li J, Looi LM, Teoh KH, Ong DBL, Arends MJ. DNA mismatch repair and CD133-marked cancer stem cells in colorectal carcinoma. PeerJ 2018; 6:e5530. [PMID: 30221090 PMCID: PMC6138039 DOI: 10.7717/peerj.5530] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022] Open
Abstract
Background Except for a few studies with contradictory observations, information is lacking on the possibility of association between DNA mismatch repair (MMR) status and the presence of cancer stem cells in colorectal carcinoma (CRC), two important aspects in colorectal carcinogenesis. Methods Eighty (40 right-sided and 40 left-sided) formalin-fixed, paraffin-embedded primary CRC were immunohistochemically studied for CD133, a putative CRC stem cell marker, and MMR proteins MLH1, MSH2, MSH6 and PMS2. CD133 expression was semi-quantitated for proportion of tumor immunopositivity on a scale of 0-5 and staining intensity on a scale of 0-3 with a final score (units) being the product of proportion and intensity of tumor staining. The tumor was considered immunopositive only when the tumor demonstrated moderate to strong intensity of CD133 staining (a decision made after analysis of CD133 expression in normal colon). Deficient MMR (dMMR) was interpreted as unequivocal loss of tumor nuclear staining for any MMR protein despite immunoreactivity in the internal positive controls. Results CD133 was expressed in 36 (90.0%) left-sided and 28 (70.0%) right-sided tumors (p < 0.05) and CD133 score was significantly higher in left- (mean ± SD = 9.6 ± 5.3 units) compared with right-sided tumors (mean ± SD = 6.8 ± 5.6 units) p < 0.05). dMMR was noted in 14 (35%) right-sided and no (0%) left-sided CRC. When stratified according to MMR status, dMMR cases showed a lower frequency of CD133 expression (42.9%) and CD133 score (mean ± SD = 2.5 ± 3.6 units) compared with pMMR tumors on the right (frequency = 84.6%; mean score ± SD = 9.2 ± 5.0 units) as well as pMMR tumors on the left (frequency = 90.0%; mean score ± SD = 9.6 ± 5.3 units) (p < 0.05). Interestingly, frequencies of CD133 immunoreactivity and CD133 scores did not differ between pMMR CRC on the right versus the left (p > 0.05). Conclusion Proficient MMR correlated with high levels of CD133-marked putative cancer stem cells in both right- and left-sided tumors, whereas significantly lower levels of CD133-marked putative cancer stem cells were associated with deficient MMR status in colorectal carcinomas found on the right.
Collapse
Affiliation(s)
- Phaik-Leng Cheah
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jing Li
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Lai-Meng Looi
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kean-Hooi Teoh
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Diana Bee-Lan Ong
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mark J Arends
- Division of Pathology, Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
224
|
Ho V, Chung L, Singh A, Lea V, Abubakar A, Lim SH, Ng W, Lee M, de Souza P, Shin JS, Lee CS. Overexpression of the MRE11-RAD50-NBS1 (MRN) complex in rectal cancer correlates with poor response to neoadjuvant radiotherapy and prognosis. BMC Cancer 2018; 18:869. [PMID: 30176843 PMCID: PMC6122630 DOI: 10.1186/s12885-018-4776-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The MRE11/RAD50/NBS1 (MRN) complex plays an essential role in detecting and repairing double-stranded breaks, and thus the potential roles of MRE11, RAD50 and NBS1 proteins in the pathogenesis of various cancers is the subject of investigation. This study was aimed at assessing the three-protein panel of MRN complex subunits as a potential radiosensitivity marker and evaluating the prognostic and clinicopathological implications of MRN expression in rectal cancer. METHODS Samples from 265 rectal cancer patients treated with surgery and adjuvant chemoradiotherapy, including samples from 55 patients who were treated with neoadjuvant radiotherapy between 2000 and 2011, were analyzed. Expression of MRN complex proteins in tissue samples was determined by immunohistochemistry. Univariate and multivariate analyses were carried out to identify clinicopathological characteristics that are associated with the MRN three-protein panel expression in rectal cancer samples. RESULTS In Kaplan-Meier survival analyses, we found that high level expression of MRN complex proteins in postoperative samples was associated with poor disease-free (p = 0.021) and overall (P = 0.002) survival. Interestingly, high MRN expression also correlated with poor disease-free (P = 0.047) and overall (P = 0.024) survival in the neoadjuvant radiotherapy subgroup. In multivariate analysis, combined MRN expression (hazard ratio = 2.114, 95% confidence interval 1.096-4.078, P = 0.026) and perineural invasion (hazard ratio = 2.160, 95% confidence interval 1.209-3.859, P = 0.009) were significantly associated with a worse disease-free survival. CONCLUSIONS Expression levels of MRN complex proteins significantly predict disease-free survival in rectal cancer patients, including those treated with neoadjuvant radiotherapy, and may have value in the management of these patients.
Collapse
Affiliation(s)
- Vincent Ho
- MBBS FRACP, School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751 Australia
| | - Liping Chung
- MBBS FRACP, School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751 Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170 Australia
| | - Amandeep Singh
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170 Australia
| | - Vivienne Lea
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170 Australia
| | - Askar Abubakar
- MBBS FRACP, School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751 Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170 Australia
| | - Stephanie H. Lim
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170 Australia
- Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, NSW 2560 Australia
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170 Australia
| | - Weng Ng
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170 Australia
| | - Mark Lee
- Department of Radiation Oncology, Liverpool Hospital, Liverpool, NSW 2170 Australia
| | - Paul de Souza
- MBBS FRACP, School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751 Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170 Australia
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170 Australia
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170 Australia
| | - Joo-Shik Shin
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050 Australia
| | - Cheok Soon Lee
- MBBS FRACP, School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751 Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170 Australia
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170 Australia
- Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW 2560 Australia
- Faculty of Medicine, South Western Sydney Clinical School, The University of New South Wales, Liverpool, NSW 2170 Australia
| |
Collapse
|
225
|
Affiliation(s)
- Frank A Sinicrope
- From the Divisions of Oncology and of Gastroenterology and Hepatology, Mayo Clinic and Mayo Comprehensive Cancer Center, Rochester, MN
| |
Collapse
|
226
|
Abstract
GOALS The aim of this study was to assess the histopathologic characteristics of colorectal carcinomas (CRC) in patients with Crohn's disease (CD). BACKGROUND A higher frequency of microsatellite instability (MSI) is seen in mucinous compared with nonmucinous CRC which suggests that its pathogenesis involves distinct molecular pathways. Several publications reported a higher percentage of mucinous adenocarcinoma in CD patients with CRC. So far, there has been no investigation of MSI in CD patients with mucinous CRC. STUDY The medical records of patients who underwent surgery for CRC were reviewed and those with a history of CD identified. The data of histologic classification and MSI status of the tumor were investigated. RESULTS Fourteen patients with CD-associated CRC were identified (5 female, 9 male) resulting in 20 CRC in total. Histologic investigation revealed 7 adenocarcinomas without a mucinous or signet ring cell component. All other CRCs harbored a mucinous (n=11) and/or signet ring cell (n=6) component. All tumors assessed for MSI were found to be microsatellite stable. CONCLUSIONS Our data indicate that CRCs with signet ring cell and mucinous components were much more common in patients with CD than in patients with sporadic CRC. This observation suggests that CRC in CD represent an own entity with distinct histopathologic and molecular features. This may implicate potential consequences for diagnosis and therapy of CRC in CD in the future as well as new factors to identify patients with an increased risk for developing CRC in CD.
Collapse
|
227
|
Halldorsson MO, Hauptmann M, Snaebjornsson P, Haraldsdóttir KH, Aspelund T, Gudmundsson EF, Gudnason V, Jonasson JG, Haraldsdottir S. The risk of developing a mismatch repair deficient colorectal cancer after undergoing cholecystectomy. Scand J Gastroenterol 2018; 53:972-975. [PMID: 30010450 DOI: 10.1080/00365521.2018.1481997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Mismatch repair deficient (dMMR) colorectal cancer (CRC) is caused by inactivation of the MMR DNA repair system, most commonly via epigenetic inactivation of the MLH1 gene, and these tumors occur most frequently in the right colon. The objective was to determine whether cholecystectomy (CCY) increases the risk of a dMMR CRC by comparing CCY incidence in patients with dMMR CRC and proficient MMR (pMMR) CRC to unaffected controls. MATERIALS AND METHODS All patients diagnosed with CRC in Iceland from 2000 to 2009 (n = 1171) were included. They had previously been screened for dMMR by immunohistochemistry (n = 129 were dMMR). Unaffected age- and sex-matched controls (n = 17,460) were obtained from large Icelandic cohort studies. Subjects were cross-referenced with all pathology databases in Iceland to establish who had undergone CCY. Odds ratios were calculated using unconditional logistic regression. RESULTS Eighteen (13.7%) dMMR CRC cases and 90 (8.7%) pMMR CRC cases had undergone CCY compared to 1532 (8.8%) controls. CCY-related odds ratios (OR) were 1.06 (95% CI 0.90-1.26, p = .577) for all CRC, 1.16 (95% CI 0.66-2.05 p = .602) for dMMR CRCand 1.04 (95% CI 0.83-1.29, p = .744) for pMMR CRC. Furthermore, OR for dMMR CRC was 0.51 (95% CI 0.16-1.67, p = .266), 2.04 (95% CI 0.92-4.50, p = .080) and 1.08 (95% CI 0.40-2.89, p = .875) <10 years, 10-20 years and >20 years after a CCY, respectively. CONCLUSIONS There was no evidence of increased risk of developing dMMR CRC after CCY although a borderline significantly increased 2-fold risk was observed 10-20 years after CCY. Larger studies are warranted to examine this further.
Collapse
Affiliation(s)
| | - Michael Hauptmann
- b Department of Epidemiology and Biostatistics , The Netherlands Cancer Institute , Amsterdam , The Netherlands
| | - Petur Snaebjornsson
- c Department of Pathology , The Netherlands Cancer Institute , Amsterdam , The Netherlands
| | | | - Thor Aspelund
- e University of Iceland , Reykjavík , Iceland.,f Icelandic Heart Association , Kópavogur , Iceland
| | | | - Vilmundur Gudnason
- a Faculty of Medicine , University of Iceland , Reykjavík , Iceland.,f Icelandic Heart Association , Kópavogur , Iceland
| | - Jon Gunnlaugur Jonasson
- a Faculty of Medicine , University of Iceland , Reykjavík , Iceland.,d Landspitali University Hospital Iceland , Reykjavík , Iceland.,g Department of Pathology , Landspitali-University Hospital , Iceland
| | | |
Collapse
|
228
|
De Nicola F, Goeman F, Pallocca M, Sperati F, Pizzuti L, Melucci E, Casini B, Amoreo CA, Gallo E, Diodoro MG, Buglioni S, Mazzotta M, Vici P, Sergi D, Di Lauro L, Barba M, Pescarmona E, Ciliberto G, De Maria R, Fanciulli M, Maugeri-Saccà M. Deep sequencing and pathway-focused analysis revealed multigene oncodriver signatures predicting survival outcomes in advanced colorectal cancer. Oncogenesis 2018; 7:55. [PMID: 30032163 PMCID: PMC6054833 DOI: 10.1038/s41389-018-0066-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/26/2018] [Accepted: 06/25/2018] [Indexed: 12/31/2022] Open
Abstract
Genomic technologies are reshaping the molecular landscape of colorectal cancer (CRC), revealing that oncogenic driver mutations (APC and TP53) coexist with still underappreciated genetic events. We hypothesized that mutational analysis of CRC-linked genes may provide novel information on the connection between genetically-deregulated pathways and clinical outcomes. We performed next-generation sequencing (NGS) analysis of 16 recurrently mutated genes in CRC exploiting tissue specimens from 98 advanced CRC patients. Multiple correspondence analysis (MCA) was used to identify gene sets characterizing negative and positive outliers (patients in the lowest and highest quartile of progression-free survival, PFS). Variables potentially affecting PFS and overall survival (OS) were tested in univariate and multivariate Cox proportional hazard models. Sensitivity analyses and resampling were used to assess the robustness of genomic predictors. MCA revealed that APC and TP53 mutations were close to the negative outlier group, whereas mutations in other WNT pathway genes were in proximity of the positive outliers. Reasoning that genetic alterations interact epistatically, producing greater or weaker consequences in combination than when individually considered, we tested whether patients whose tumors carried a genetic background characterized by APC and TP53 mutations without coexisting mutations in other WNT genes (AMER1, FBXW7, TCF7L2, CTNNB1, SOX9) had adverse survival outcomes. With this approach, we identified two oncodriver signatures (ODS1 and ODS2) associated with shorter PFS (ODS1 multivariate Cox for PFS: HR 2.16, 95%CI: 1.28–3.64, p = 0.004; ODS2 multivariate Cox for PFS: HR 2.61, 95%CI: 1.49–4.58, p = 0.001). Clinically-focused and molecularly-focused sensitivity analyses, resampling, and reclassification of mutations confirmed the stability of ODS1/2. Moreover, ODS1/2 negatively impacted OS. Collectively, our results point to co-occurring driver mutations as an adverse molecular factor in advanced CRC. This relationship depends on a broader genetic context highlighting the importance of genetic interactions.
Collapse
Affiliation(s)
- Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Frauke Goeman
- Oncogenomic and Epigenetic Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Matteo Pallocca
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Francesca Sperati
- Biostatistics-Scientific Direction, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Elisa Melucci
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Beatrice Casini
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Carla Azzurra Amoreo
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Enzo Gallo
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Maria Grazia Diodoro
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Marco Mazzotta
- Medical Oncology Unit, Policlinico Sant'Andrea, Via Di Grotta Rossa 1035/1039, 00189, Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 10, 00168, Rome, Italy.
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
229
|
Augustus GJ, Roe DJ, Jacobs ET, Lance P, Ellis NA. Is increased colorectal screening effective in preventing distant disease? PLoS One 2018; 13:e0200462. [PMID: 30001362 PMCID: PMC6042755 DOI: 10.1371/journal.pone.0200462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/30/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Screening in the average risk population for colorectal cancer (CRC) is expected to reduce the incidence of distant (i.e., metastatic) CRCs at least as much as less advanced CRCs. Indeed, since 2000, during which time colonoscopy became widely used as a screening tool, the overall incidence of CRC has been reduced by 29%. OBJECTIVE The purpose of the current study was to determine whether the reduction of incidence rates is the same for all stages of disease. METHODS We evaluated incidence data from the Surveillance, Epidemiology, and End Results (SEER) program from 2000-2014 for Localized, Regional, and Distant disease. Joinpoint models were compared to assess parallelism of trends. Data were stratified by race, age, tumor location, and sex to determine whether these subgroupings could explain overall trends. RESULTS Inconsistent with the expectations of a successful screening program, the reduction in incidence rates of distant CRCs from 2000-2014 has been slower than the reductions in incidence rates of both regional and localized CRCs. This trend is evident even when the data are stratified by age at diagnosis, sex, race, or tumor location. CONCLUSIONS The slower decrease in the incidence rate of distant disease is not consistent with a screening effect, that is, CRC screening may not be effective in preventing many distant CRCs. As a consequence, distant CRCs represent an increasing fraction of all CRCs, accounting for 21% of all CRCs in 2014. The analysis indicates that inadequate screening does not explain the slower decrease in incidence of distant CRCs. Consequently, we suggest that a subtype of CRC exists that advances rapidly, evading detection because screening intervals are too long to prevent it. Microsatellite unstable tumors represent a known subtype that advances more rapidly, and we suggest that another rapidly advancing subtype very likely exists that is microsatellite stable.
Collapse
Affiliation(s)
- Gaius Julian Augustus
- Cancer Biology Graduate Interdisciplinary Program, Tucson, AZ
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, Tucson, AZ
| | - Denise J. Roe
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, Tucson, AZ
| | - Elizabeth T. Jacobs
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, Tucson, AZ
| | - Peter Lance
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, Tucson, AZ
| | - Nathan A. Ellis
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, Tucson, AZ
| |
Collapse
|
230
|
Jonchere V, Marisa L, Greene M, Virouleau A, Buhard O, Bertrand R, Svrcek M, Cervera P, Goloudina A, Guillerm E, Coulet F, Landman S, Ratovomanana T, Job S, Ayadi M, Elarouci N, Armenoult L, Merabtene F, Dumont S, Parc Y, Lefèvre JH, André T, Fléjou JF, Guilloux A, Collura A, de Reyniès A, Duval A. Identification of Positively and Negatively Selected Driver Gene Mutations Associated With Colorectal Cancer With Microsatellite Instability. Cell Mol Gastroenterol Hepatol 2018; 6:277-300. [PMID: 30116770 PMCID: PMC6089198 DOI: 10.1016/j.jcmgh.2018.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/05/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Recent studies have shown that cancers arise as a result of the positive selection of driver somatic events in tumor DNA, with negative selection playing only a minor role, if any. However, these investigations were concerned with alterations at nonrepetitive sequences and did not take into account mutations in repetitive sequences that have very high pathophysiological relevance in the tumors showing microsatellite instability (MSI) resulting from mismatch repair deficiency investigated in the present study. METHODS We performed whole-exome sequencing of 47 MSI colorectal cancers (CRCs) and confirmed results in an independent cohort of 53 MSI CRCs. We used a probabilistic model of mutational events within microsatellites, while adapting pre-existing models to analyze nonrepetitive DNA sequences. Negatively selected coding alterations in MSI CRCs were investigated for their functional and clinical impact in CRC cell lines and in a third cohort of 164 MSI CRC patients. RESULTS Both positive and negative selection of somatic mutations in DNA repeats was observed, leading us to identify the expected true driver genes associated with the MSI-driven tumorigenic process. Several coding negatively selected MSI-related mutational events (n = 5) were shown to have deleterious effects on tumor cells. In the tumors in which deleterious MSI mutations were observed despite the negative selection, they were associated with worse survival in MSI CRC patients (hazard ratio, 3; 95% CI, 1.1-7.9; P = .03), suggesting their anticancer impact should be offset by other as yet unknown oncogenic processes that contribute to a poor prognosis. CONCLUSIONS The present results identify the positive and negative driver somatic mutations acting in MSI-driven tumorigenesis, suggesting that genomic instability in MSI CRC plays a dual role in achieving tumor cell transformation. Exome sequencing data have been deposited in the European genome-phenome archive (accession: EGAS00001002477).
Collapse
Key Words
- CRC, colorectal cancer
- Colorectal Cancer
- Driver Gene Mutations
- HR, hazard ratio
- MLH1, MutL Homolog 1
- MMR, mismatch repair
- MSH, MutS Homolog
- MSI, microsatellite instability
- Microsatellite Instability
- NR, nonrepetitive
- PBS, phosphate-buffered saline
- PCR, polymerase chain reaction
- Positive and Negative Selection
- R, repetitive
- RFS, relapse-free survival
- RTCA, Real-Time Cell Analyzer
- Tumorigenic Process
- UTR, untranslated region
- WES, whole-exome sequencing
- WGA, whole-genome amplification
- bp, base pair
- indel, insertion/deletion
- mRNA, messenger RNA
- shRNA, short hairpin RNA
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Vincent Jonchere
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Laetitia Marisa
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Malorie Greene
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France
| | - Alain Virouleau
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Laboratoire de Mathématiques et Modélisation d’Évry, University Évry, Évry, France,Centre National de la Recherche Scientifique, Université Paris-Saclay, Evry, France
| | - Olivier Buhard
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France
| | - Romane Bertrand
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France
| | - Magali Svrcek
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Service d’Anatomie et Cytologie Pathologiques, Assistance publique - Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Pascale Cervera
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Service d’Anatomie et Cytologie Pathologiques, Assistance publique - Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Anastasia Goloudina
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Inovarion, Collaborative research Department Paris, France
| | - Erell Guillerm
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Genetics Department, Assistance publique - Hôpitaux de Paris, Pitié Salpêtrière Hôpital, Paris, France
| | - Florence Coulet
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Genetics Department, Assistance publique - Hôpitaux de Paris, Pitié Salpêtrière Hôpital, Paris, France
| | - Samuel Landman
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France
| | - Toky Ratovomanana
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France
| | - Sylvie Job
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Mira Ayadi
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Nabila Elarouci
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Lucile Armenoult
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Fatiha Merabtene
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,University Pierre and Marie CURIE Paris 06, Unité Mixte de Service 30 L'Unité Mixte de service Imagerie Cytométrie, Plateforme d’Histomorphologie, Sorbonne Université Paris, France
| | - Sylvie Dumont
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,University Pierre and Marie CURIE Paris 06, Unité Mixte de Service 30 L'Unité Mixte de service Imagerie Cytométrie, Plateforme d’Histomorphologie, Sorbonne Université Paris, France
| | - Yann Parc
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Service de Chirurgie Générale et Digestive, Assistance publique - Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Jérémie H. Lefèvre
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Service de Chirurgie Générale et Digestive, Assistance publique - Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Thierry André
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Department of Oncology, Assistance publique - Hôpitaux de Paris, Hôpital Saint Antoine, Paris, France
| | - Jean-François Fléjou
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Service d’Anatomie et Cytologie Pathologiques, Assistance publique - Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Agathe Guilloux
- Laboratoire de Mathématiques et Modélisation d’Évry, University Évry, Évry, France,Centre National de la Recherche Scientifique, Université Paris-Saclay, Evry, France
| | - Ada Collura
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Alex Duval
- Sorbonne Université, University Pierre and Marie CURIE Paris 06, INSERM, Unité Mixte de Recherche938, Equipe Instabilité des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France,Correspondence Address correspondence to: Alex Duval, MD, PhD, Sorbonne Universite, UPMC Univ Paris 06, Inserm, UMR938, Equipe Instabilite Des Microsatellites et Cancer, Centre de Recherche Saint Antoine, Paris, France. fax: (33) 149284603.
| |
Collapse
|
231
|
Nguyen HT, Duong HQ. The molecular characteristics of colorectal cancer: Implications for diagnosis and therapy. Oncol Lett 2018; 16:9-18. [PMID: 29928381 DOI: 10.3892/ol.2018.8679] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) results from the progressive accumulation of multiple genetic and epigenetic aberrations within cells. The progression from colorectal adenoma to carcinoma is caused by three major pathways: Microsatellite instability, chromosomal instability and CpG island methylator phenotype. A growing body of scientific evidences suggests that CRC is a heterogeneous disease, and genetic characteristics of the tumors determine their prognostic outcome and response to targeted therapies. Early diagnosis and effective targeted therapies based on a current knowledge of the molecular characteristics of CRC are essential to the successful treatment of CRC. Therefore, the present review summarized the current understanding of the molecular characteristics of CRC, and discussed its implications for diagnosis and targeted therapy.
Collapse
Affiliation(s)
- Ha Thi Nguyen
- Center for Molecular Biology, Institute of Research and Development, Duy Tan University, Danang 550000, Vietnam
| | - Hong-Quan Duong
- Department of Cancer Research, Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi 100000, Vietnam
| |
Collapse
|
232
|
Boland PM, Yurgelun MB, Boland CR. Recent progress in Lynch syndrome and other familial colorectal cancer syndromes. CA Cancer J Clin 2018; 68:217-231. [PMID: 29485237 PMCID: PMC5980692 DOI: 10.3322/caac.21448] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/08/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022] Open
Abstract
The current understanding of familial colorectal cancer was limited to descriptions of affected pedigrees until the early 1990s. A series of landscape-altering discoveries revealed that there were distinct forms of familial cancer, and most were related to genes previously not known to be involved in human disease. This review largely focuses on advances in our understanding of Lynch syndrome because of the unique relationship of this disease to defective DNA mismatch repair and the clinical implications this has for diagnostics, prevention, and therapy. Recent advances have occurred in our understanding of the epidemiology of this disease, and the advent of broad genetic panels has altered the approach to germline and somatic diagnoses for all of the familial colorectal cancer syndromes. Important advances have been made toward a more complete mechanistic understanding of the pathogenesis of neoplasia in the setting of Lynch syndrome, and these advances have important implications for prevention. Finally, paradigm-shifting approaches to treatment of Lynch-syndrome and related tumors have occurred through the development of immune checkpoint therapies for hypermutated cancers. CA Cancer J Clin 2018;68:217-231. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Patrick M Boland
- Assistant Professor, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY
| | - Matthew B Yurgelun
- Assistant Professor of Medicine, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - C Richard Boland
- Professor, Department of Medicine, University of California at San Diego School of Medicine, San Diego, CA
| |
Collapse
|
233
|
Buglioni S, Melucci E, Sperati F, Pallocca M, Terrenato I, De Nicola F, Goeman F, Casini B, Amoreo CA, Gallo E, Diodoro MG, Pescarmona E, Vici P, Sergi D, Pizzuti L, Di Lauro L, Mazzotta M, Barba M, Fanciulli M, Vitale I, De Maria R, Ciliberto G, Maugeri-Saccà M. The clinical significance of PD-L1 in advanced gastric cancer is dependent on ARID1A mutations and ATM expression. Oncoimmunology 2018; 7:e1457602. [PMID: 30221053 DOI: 10.1080/2162402x.2018.1457602] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 01/05/2023] Open
Abstract
Whether PD-L1 expression is associated with survival outcomes in gastric cancer (GC) is controversial. The inhibition of the PD-1/PD-L1 pathway is effective against genomically unstable tumors. Hypothesizing that also the clinical significance of PD-L1 might be dependent on the activation of molecular circuits ensuring genomic stability, we evaluated PD-L1 expression in tissue samples from 72 advanced GC patients treated with first-line chemotherapy. Samples were already characterized for DNA damage repair (DDR) component expression (pATM, pChk1, pWee1, γ-H2AX and pRPA2) along with mutations in DDR-linked genes (TP53 and ARID1A). Overall, PD-L1 expression was not associated with progression-free survival (PFS) and overall survival (OS), independently on whether we considered its expression in tumor cells (PD-L1-TCs) or in the immune infiltrate (PD-L1-TILs). In subgroup analysis, positive PD-L1-TC immunostaining was associated with better PFS in patients whose tumors did not carry DDR activation (multivariate Cox: HR 0.34, 95%CI: 0.15-0.76, p = 0.008). This subset (DDRoff) was characterized by negative pATM expression or the presence of ARID1A mutations. Conversely, the relationship between PD-L1-TC expression and PFS was lost in a molecular scenario denoting DDR activation (DDRon), as defined by concomitant pATM expression and ARID1A wild-type form. Surprisingly, while PD-L1-TC expression was associated with better OS in the DDRoff subset (multivariate Cox: HR 0.41, 95% CI: 0.17-0.96, p = 0.039), in the DDRon subgroup we observed an opposite impact on OS (multivariate Cox: HR 2.56, 95%CI: 1.06-6.16, p = 0.036). Thus, PD-L1-TC expression may impact survival outcomes in GC on the basis of the activation/inactivation of genome-safeguarding pathways.
Collapse
Affiliation(s)
- Simonetta Buglioni
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Elisa Melucci
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Francesca Sperati
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Matteo Pallocca
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Irene Terrenato
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Frauke Goeman
- Department of Diagnostic Research and Technological Innovation, Oncogenomic and Epigenetic Unit, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Beatrice Casini
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Carla Azzurra Amoreo
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Enzo Gallo
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Maria Grazia Diodoro
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Marco Mazzotta
- Department of Medical Oncology, Medical Oncology Unit, Policlinico Sant'Andrea, Via Di Grotta Rossa 1035/1039, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Ilio Vitale
- Cellular Networks and Molecular Therapeutic Targets Unit, Department of Research, Advanced Diagnostics and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, Rome, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Catholic University of the Sacred Heart and Fondazione Policlinico Universitario Agostino Gemelli, Largo Agostino Gemelli, 10, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| |
Collapse
|
234
|
Cama A, Genuardi M, Guanti G, Radice P, Varesco L. Molecular Genetics of Hereditary Non-Polyposis Colorectal Cancer (HNPCC). TUMORI JOURNAL 2018; 82:122-35. [PMID: 8644374 DOI: 10.1177/030089169608200206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The story of the molecular genetics of HNPCC is one of astonishingly rapid achievements. In just 16 months, from May 1993 to September 1994, four different genes, namely hMSH2, hMLH1, hPMS1 and hPMS2 have been identified and demonstrated to be associated with the disease. Their cloning was facilitated by the finding that tumor cells in HNPCC patients display a hypermutability of DNA short tandem repeats (microsatellite instability). In fact, HNPCC associated genes are the human counterparts of genetic elements known to control the fidelity of DNA replication in lower organisms. So far, more than 50 germline mutations of hMSH2 and hMLH1 genes have been reported in HNPCC kindreds. In addition, somatic mutations have been documented in hereditary as well as sporadic cancers. Unfortunately, the molecular diagnosis of HNPCC is hampered by the lack of mutational “hot spots” and of clearly defined genotype-phenotype correlations and different screening methods are to be employed for the analysis of affected and at-risk individuals.
Collapse
Affiliation(s)
- A Cama
- Cattedra di Patologia Generale, Università Gabriele D'Annuzio, Chieti,Italy
| | | | | | | | | |
Collapse
|
235
|
Abstract
Starting from a survey of the studies on familial aggregation of colorectal cancer, we introduce the aims of genetic epidemiology. One of its main goals is to assess population frequency of cancer susceptibility genes and to determine the age-specific risks for carriers with respect to non-carriers. In section two, segregation analysis investigations are reviewed, and inferences on the relevance of genetic components of susceptibility to colorectal cancer are drawn. In section three, the HNPCC paradigm is discussed in the light of the Knudson model of tumorigenesis and recent advances of molecular research. In the last section we show an example of genotype/environment interaction in the etiology of a particular cancer and present a conceptual framework for studies on cancer genetic epidemiology in terms of attributable and relative risk.
Collapse
Affiliation(s)
- S Presciuttini
- Dipartimento di Scienze dell'Ambiente e del Territorio, Pisa, Italy
| | | |
Collapse
|
236
|
Jin Z, Sanhueza CT, Johnson B, Nagorney DM, Larson DW, Mara KC, Harmsen WC, Smyrk TC, Grothey A, Hubbard JM. Outcome of Mismatch Repair-Deficient Metastatic Colorectal Cancer: The Mayo Clinic Experience. Oncologist 2018; 23:1083-1091. [PMID: 29674439 DOI: 10.1634/theoncologist.2017-0289] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 02/12/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Deficiencies in the DNA mismatch repair system cause errors during DNA replication, which in turn give rise to microsatellite instability (MSI). The impact of MSI on survival in metastatic colorectal cancer (mCRC) is unclear. This cohort study aims to investigate the prognostic and predictive value of MSI in mCRC prior to the immune therapy era. MATERIALS AND METHODS A total of 75 MSI-high (MSI-H) mCRC patients (pts) and 75 matched (age, gender, disease sidedness, metachronous/synchronous) microsatellite-stable (MSS) mCRC pts were identified from 1,268 mCRC pts who had MSI/mismatch repair test results at Mayo Clinic Rochester between January 1992 and July 2016. A retrospective review was conducted by using data from electronic medical records. Statistical analyses utilized the Kaplan-Meier method, log-rank test, and Cox proportional hazards models. RESULTS The MSS group was well matched to the MSI-H group based on age, gender, location, and chronicity of metastatic disease. MSI-H mCRC pts had earlier disease recurrence (median time from initial diagnosis to metastatic disease diagnosis, MSI-H group 12.9 vs. MSS group 20.9 months, p = .034). Median overall survival (OS) was 28.1 and 37.4 months for MSI-H and MSS pts, respectively (p = .99). In total, 94.7% of MSI-H pts and 98.7% of MSS pts had fluoropyrimidine-based chemotherapy for metastatic disease, and there was no difference in OS between these two groups (32.3 vs. 37.4 months, p = .91). Forty-three MSI-H and thirty-nine MSS pts had metastasectomy and/or ablation of metastases (p = .51) with longer median OS compared with pts without metastasectomy (MSI-H: 82.0 vs. 13.9, p < .001; MSS: 69.9 vs. 19.7, p < .001). Age <65 years, BRAF wild type, and metastasectomy were associated with better OS in univariate analysis. Only metastasectomy remained statistically significant in multivariate analysis (p < .001). CONCLUSION In mCRC, patients with MSI-H tumors have similar, but numerically shorter, median overall survival compared with those with MSS tumors. In both groups, metastasectomy and ablation of metastatic disease should be considered to optimize OS. IMPLICATIONS FOR PRACTICE This study clearly demonstrated the survival benefits that aggressive metastasectomy provides in selected microsatellite instability-high metastatic colorectal cancer patients. This could be meaningful practice-changing information that has been long awaited.
Collapse
Affiliation(s)
- Zhaohui Jin
- Department of Medical Oncology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - Cristobal T Sanhueza
- Department of Medical Oncology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - Benny Johnson
- Department of Medical Oncology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - David M Nagorney
- Department of Surgery, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - David W Larson
- Department of Surgery, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - Kristin C Mara
- Department of Statistics, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - William C Harmsen
- Department of Statistics, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - Thomas C Smyrk
- Department of Pathology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - Axel Grothey
- Department of Medical Oncology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | - Joleen M Hubbard
- Department of Medical Oncology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| |
Collapse
|
237
|
Kim ST, Kim HK, Lee J, Park SH, Lim HY, Park YS, Kang WK, Park JO. The impact of microsatellite instability status and sidedness of the primary tumor on the effect of bevacizumab-containing chemotherapy in patients with metastatic colorectal cancer. J Cancer 2018; 9:1791-1796. [PMID: 29805705 PMCID: PMC5968767 DOI: 10.7150/jca.25132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/02/2018] [Indexed: 01/21/2023] Open
Abstract
Purpose: We aimed to evaluate the effect of bevacizumab in metastatic CRC (colorectal cancer) regarding to microsatellite instability (MSI) and the sidedness of the primary tumor. Materials and Methods: A total of 140 CRC patients were retrospectively analyzed, who received bevacizumab-containing chemotherapy between April 2008 and January 2013. MSI status and Kirsten RSAS (KRAS) mutational status were available in all 140 patients, but BRAF (the gene for the B-type Raf kinase) mutational status was only available in 74 patients (52.9%). Results: MSI-high (MSI-H) was detected in 4.3% of analyzed patients. Characteristics of patients, with the exception of BRAF mutational status, were generally similar between those with right- (RC) and left-sided colon cancer (LC). Right-sided tumors were significantly associated with a BRAF mutation (p=0.025). In addition, patient characteristics with a microsatellite stable (MSS) tumor were not different from those with an MSI-H tumor. For all 140 patients, the most commonly used regimen with bevacizumab was capecitabine plus oxaliplain (XELOX), irrespective of treatment line, followed by 5-fluorouracil, leucovorin, and irinotecan (FOLFIRI), 5-fluorouracil, leucovorin, and oxaliplatin (FOLFOX), intravenous 5-fluorouracil (5-FU) and capecitabine plus irinotecan (XELIRI). There was no significant difference between the MSI-H and MSS groups in treatment efficacy, including response rate (RR) and disease control rate (DCR). There was also no difference in RR and DCR according to the sidedness of the primary tumor. No significant difference in progression-free survival (PFS) was observed between MSI-H and MSS groups (5.93 months vs. 7.37 months; p=0.801) or between LC and RC groups (7.37 months vs. 5.83 months; p=0.801). Conclusions: The effect of bevacizumab was not different between LC and RC and between MSS and the MSI-H tumors.
Collapse
Affiliation(s)
- Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Kyung Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Division of Oncology, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se Hoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ho Yeong Lim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Suk Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
238
|
Gray PN, Tsai P, Chen D, Wu S, Hoo J, Mu W, Li B, Vuong H, Lu HM, Batth N, Willett S, Uyeda L, Shah S, Gau CL, Umali M, Espenschied C, Janicek M, Brown S, Margileth D, Dobrea L, Wagman L, Rana H, Hall MJ, Ross T, Terdiman J, Cullinane C, Ries S, Totten E, Elliott AM. TumorNext-Lynch-MMR: a comprehensive next generation sequencing assay for the detection of germline and somatic mutations in genes associated with mismatch repair deficiency and Lynch syndrome. Oncotarget 2018; 9:20304-20322. [PMID: 29755653 PMCID: PMC5945525 DOI: 10.18632/oncotarget.24854] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
The current algorithm for Lynch syndrome diagnosis is highly complex with multiple steps which can result in an extended time to diagnosis while depleting precious tumor specimens. Here we describe the analytical validation of a custom probe-based NGS tumor panel, TumorNext-Lynch-MMR, which generates a comprehensive genetic profile of both germline and somatic mutations that can accelerate and streamline the time to diagnosis and preserve specimen. TumorNext-Lynch-MMR can detect single nucleotide variants, small insertions and deletions in 39 genes that are frequently mutated in Lynch syndrome and colorectal cancer. Moreover, the panel provides microsatellite instability status and detects loss of heterozygosity in the five Lynch genes; MSH2, MSH6, MLH1, PMS2 and EPCAM. Clinical cases are described that highlight the assays ability to differentiate between somatic and germline mutations, precisely classify variants and resolve discordant cases.
Collapse
Affiliation(s)
- Phillip N Gray
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Pei Tsai
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Daniel Chen
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Sitao Wu
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Jayne Hoo
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Wenbo Mu
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Bing Li
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Huy Vuong
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Hsiao-Mei Lu
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Navanjot Batth
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Sara Willett
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Lisa Uyeda
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Swati Shah
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Chia-Ling Gau
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Monalyn Umali
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Carin Espenschied
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Mike Janicek
- Cancer Genetic Risk Assessment Program, Arizona Oncology, Scottsdale, AZ 85258, USA
| | - Sandra Brown
- Cancer Genetics Program, Saint Joseph of Orange, Orange, CA 92868, USA
| | - David Margileth
- Cancer Genetics Program, Saint Joseph of Orange, Orange, CA 92868, USA
| | - Lavinia Dobrea
- Oncology Research and Biospecimen Program, Saint Joseph of Orange, Orange, CA 92868, USA
| | - Lawrence Wagman
- The Center for Cancer Prevention and Treatment, Saint Joseph of Orange, Orange, CA 92868, USA
| | - Huma Rana
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02461, USA
| | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia PA 19111, USA
| | - Theodora Ross
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan Terdiman
- Department of Medicine - Gastroenterology, University of California San Francisco, San Francisco, CA 94115, USA
| | - Carey Cullinane
- Department of Pathology, Long Beach Memorial Medical Center, Long Beach, CA 90801, USA
| | - Savita Ries
- Department of Pathology, Long Beach Memorial Medical Center, Long Beach, CA 90801, USA
| | - Ellen Totten
- Advocate Medical Group, Park Ridge, Illinois 60068, USA
| | - Aaron M Elliott
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| |
Collapse
|
239
|
Abstract
Microsatellite instability (MSI) refers to the hypermutator phenotype secondary to frequent polymorphism in short repetitive DNA sequences and single nucleotide substitution, as consequence of DNA mismatch repair (MMR) deficiency. MSI secondary to germline mutation in DNA MMR proteins is the molecular fingerprint of Lynch syndrome (LS), while epigenetic inactivation of these genes is more commonly found in sporadic MSI tumors. MSI occurs at different frequencies across malignancies, although original methods to assess MSI or MMR deficiency have been developed mostly in LS related cancers. Here we will discuss the current methods to detect MSI/MMR deficiency with a focus of new tools which are emerging as highly sensitive detector for MSI across multiple tumor types. Due to high frequencies of non-synonymous mutations, the presence of frameshift-mutated neoantigens, which can trigger a more robust and long-lasting immune response and strong TIL infiltration with tumor eradication, MSI has emerged as an important predictor of sensitivity for immunotherapy-based strategies, as showed by the recent FDA's first histology agnostic-accelerated approval to immune checkpoint inhibitors for refractory, adult and pediatric, MMR deficient (dMMR) or MSI high (MSI-H) tumors. Moreover, it is known that MSI status may predict cancer response/resistance to certain chemotherapies. Here we will describe the complex interplay between the genetic and clinical-pathological features of MSI/dMMR tumors and the cancer immunotherapy, with a focus on the predictive and prognostic role of MMR status for immune checkpoint inhibitors (ICIs) and providing some suggestions on how to conceive better predictive markers for immunotherapy in the next future.
Collapse
Affiliation(s)
- Marina Baretti
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Hospital, United States
| | - Dung T Le
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Hospital, United States.
| |
Collapse
|
240
|
Knijnenburg TA, Wang L, Zimmermann MT, Chambwe N, Gao GF, Cherniack AD, Fan H, Shen H, Way GP, Greene CS, Liu Y, Akbani R, Feng B, Donehower LA, Miller C, Shen Y, Karimi M, Chen H, Kim P, Jia P, Shinbrot E, Zhang S, Liu J, Hu H, Bailey MH, Yau C, Wolf D, Zhao Z, Weinstein JN, Li L, Ding L, Mills GB, Laird PW, Wheeler DA, Shmulevich I, Monnat RJ, Xiao Y, Wang C. Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep 2018; 23:239-254.e6. [PMID: 29617664 PMCID: PMC5961503 DOI: 10.1016/j.celrep.2018.03.076] [Citation(s) in RCA: 746] [Impact Index Per Article: 106.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/07/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
DNA damage repair (DDR) pathways modulate cancer risk, progression, and therapeutic response. We systematically analyzed somatic alterations to provide a comprehensive view of DDR deficiency across 33 cancer types. Mutations with accompanying loss of heterozygosity were observed in over 1/3 of DDR genes, including TP53 and BRCA1/2. Other prevalent alterations included epigenetic silencing of the direct repair genes EXO5, MGMT, and ALKBH3 in ∼20% of samples. Homologous recombination deficiency (HRD) was present at varying frequency in many cancer types, most notably ovarian cancer. However, in contrast to ovarian cancer, HRD was associated with worse outcomes in several other cancers. Protein structure-based analyses allowed us to predict functional consequences of rare, recurrent DDR mutations. A new machine-learning-based classifier developed from gene expression data allowed us to identify alterations that phenocopy deleterious TP53 mutations. These frequent DDR gene alterations in many human cancers have functional consequences that may determine cancer progression and guide therapy.
Collapse
Affiliation(s)
| | - Linghua Wang
- Department of Genomic Medicine, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael T Zimmermann
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226-0509, USA; Department of Health Sciences Research, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | - Galen F Gao
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Andrew D Cherniack
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Huihui Fan
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Hui Shen
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Gregory P Way
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19103, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19103, USA
| | - Yuexin Liu
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bin Feng
- TESARO Inc., Waltham, MA 02451, USA
| | - Lawrence A Donehower
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chase Miller
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang Shen
- Department of Electrical and Computer Engineering, 3128 TAMU, Texas A&M University, College Station, TX 77843, USA
| | - Mostafa Karimi
- Department of Electrical and Computer Engineering, 3128 TAMU, Texas A&M University, College Station, TX 77843, USA
| | - Haoran Chen
- Department of Electrical and Computer Engineering, 3128 TAMU, Texas A&M University, College Station, TX 77843, USA
| | - Pora Kim
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Eve Shinbrot
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shaojun Zhang
- Department of Genomic Medicine, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jianfang Liu
- Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA 15963, USA
| | - Hai Hu
- Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA 15963, USA
| | - Matthew H Bailey
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63110, USA
| | - Christina Yau
- University of California, San Francisco, San Francisco, CA 94115, USA; Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Denise Wolf
- University of California, San Francisco, San Francisco, CA 94115, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lei Li
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Li Ding
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63110, USA; Department of Genetics, Washington University, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University, St. Louis, MO 63110, USA
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peter W Laird
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - David A Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Raymond J Monnat
- Departments of Pathology & Genome Sciences, University of Washington, Seattle, WA 98195-7705, USA.
| | | | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA; Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
241
|
Long non-coding RNA DILC suppresses cell proliferation and metastasis in colorectal cancer. Gene 2018; 666:18-26. [PMID: 29621586 DOI: 10.1016/j.gene.2018.03.100] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 03/07/2018] [Accepted: 03/29/2018] [Indexed: 12/26/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors and one of the leading causes of cancer-related death in both men and women. The prognosis of CRC remains poor due to the advanced stage and cancer metastasis at the time of diagnosis. However, the exact mechanism of tumorigenesis in CRC remains unclear. Long non-coding RNAs (lncRNAs), which refer to transcripts longer than 200 nucleotides that are not translated into protein, are known to play important roles in multiple human cancers. Lnc-DILC is reported to be an important tumor suppressor gene and its inactivation is closely associated with liver cancer stem cells. However, the role of lnc-DILC in CRC remains to be elucidated. In the present study, we observed that lnc-DILC overexpression inhibited the growth and metastasis of CRC cells. Consistently, lnc-DILC knockdown facilitated the proliferation and metastasis of CRC cells. Mechanically, lnc-DILC suppressed CRC cell progression via IL-6/STAT3 signaling inactivation. More importantly, the specific STAT3 inhibitor S3I-201 and IL-6R inhibitor tocilizumab abolished the discrepancy of growth and metastasis capacity between lnc-DILC-interference CRC cells and control cells, which further confirmed that IL-6/STAT3 signaling was required in lnc-DILC-disrupted CRC cell growth and metastasis. Taken together, our results suggest that lnc-DILC is a novel CRC suppressor and may prove to be an inhibitor of CRC progression by inactivating IL-6/STAT3 signaling.
Collapse
|
242
|
Lerner L, Winn R, Hulbert A. Lung cancer early detection and health disparities: the intersection of epigenetics and ethnicity. J Thorac Dis 2018; 10:2498-2507. [PMID: 29850158 DOI: 10.21037/jtd.2018.04.07] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lung cancer is the most prominent cause of cancer-related mortality. Significant disparities in incidence and outcome characterize the disease's manifestations among ethnically and racially diverse populations. Complete surgical resection is the most effective curative treatment. However, success relies on early tumor detection. The National Lung Cancer Screening trial showed that lung cancer related mortality can be reduced by the use of low-dose CT (LDCT) screening. However, this test is plagued by a high false positive rate of 97% and the device itself is limited to designated cancer centers due to its expense and size. This restriction makes it difficult for underserved groups to access LDCT screening, the current standard of care. Highly sensitive and specific epigenetic DNA methylation-based biomarkers have the potential to work independently or in conjunction with LDCT screening to identify early-stage tumors. These tests could reduce unnecessary invasive confirmatory diagnostic tests and their associated morbidity and mortality. These tests also have the opportunity to bring lung cancer screening to the community thereby reducing unequal accessibility. However, epigenetic alterations are closely linked to the interplay between hereditary and environmental factors such as diet, lifestyle, ethnic ancestry, toxin exposure, residential segregation, and disparate community support structures. Despite this, the overwhelming number of early detection DNA methylation biomarker studies to date have either failed to control for ethnicity or have employed heavily Caucasian-biased patient cohorts. This review seeks to summarize the literature related to the early detection of lung cancer through molecular biomarkers among different ethnicities. Ethnical specific epigenetic biomarkers have the potential to be the first step towards an accessible, available personalized medicine approach to cancer through liquid biopsy.
Collapse
Affiliation(s)
- Lane Lerner
- 1University of Illinois at Chicago Cancer Center, 2Department of Surgery/Cancer Center University of Illinois at Chicago Cancer Center, University of Illinois Hospital and Health Sciences System, Chicago, USA
| | - Robert Winn
- 1University of Illinois at Chicago Cancer Center, 2Department of Surgery/Cancer Center University of Illinois at Chicago Cancer Center, University of Illinois Hospital and Health Sciences System, Chicago, USA
| | - Alicia Hulbert
- 1University of Illinois at Chicago Cancer Center, 2Department of Surgery/Cancer Center University of Illinois at Chicago Cancer Center, University of Illinois Hospital and Health Sciences System, Chicago, USA
| |
Collapse
|
243
|
Cuceu C, Hempel WM, Sabatier L, Bosq J, Carde P, M'kacher R. Chromosomal Instability in Hodgkin Lymphoma: An In-Depth Review and Perspectives. Cancers (Basel) 2018; 10:cancers10040091. [PMID: 29587466 PMCID: PMC5923346 DOI: 10.3390/cancers10040091] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/20/2018] [Accepted: 03/23/2018] [Indexed: 12/22/2022] Open
Abstract
The study of Hodgkin lymphoma (HL), with its unique microenvironment and long-term follow-up, has provided exceptional insights into several areas of tumor biology. Findings in HL have not only improved our understanding of human carcinogenesis, but have also pioneered its translation into the clinics. HL is a successful paradigm of modern treatment strategies. Nonetheless, approximately 15–20% of patients with advanced stage HL still die following relapse or progressive disease and a similar proportion of patients are over-treated, leading to treatment-related late sequelae, including solid tumors and organ dysfunction. The malignant cells in HL are characterized by a highly altered genomic landscape with a wide spectrum of genomic alterations, including somatic mutations, copy number alterations, complex chromosomal rearrangements, and aneuploidy. Here, we review the chromosomal instability mechanisms in HL, starting with the cellular origin of neoplastic cells and the mechanisms supporting HL pathogenesis, focusing particularly on the role of the microenvironment, including the influence of viruses and macrophages on the induction of chromosomal instability in HL. We discuss the emerging possibilities to exploit these aberrations as prognostic biomarkers and guides for personalized patient management.
Collapse
Affiliation(s)
- Corina Cuceu
- Laboratory of Radiobiology and Oncology and PROCyTOX, DRF, CEA, 91534 Paris-Saclay, France.
| | - William M Hempel
- Laboratory of Radiobiology and Oncology and PROCyTOX, DRF, CEA, 91534 Paris-Saclay, France.
| | - Laure Sabatier
- Laboratory of Radiobiology and Oncology and PROCyTOX, DRF, CEA, 91534 Paris-Saclay, France.
| | - Jacques Bosq
- Departement of Anapathology, Gustave Roussy Cancer Campus, 94805 Villejuif, France.
| | - Patrice Carde
- Department of Hematology Gustave Roussy Cancer Campus, 94800 Villejuif, France.
| | - Radhia M'kacher
- Laboratory of Radiobiology and Oncology and PROCyTOX, DRF, CEA, 91534 Paris-Saclay, France.
- Cell Environment, DNA damages R&D, Oncology section, 75020 Paris, France.
| |
Collapse
|
244
|
Miniati P, Sourvinos G, Michalodimitrakis M, Spandidos DA. Loss of Heterozygosity on Chromosomes 1, 2, 8, 9 and 17 in Cerebral Atherosclerotic plaques. Int J Biol Markers 2018; 16:167-71. [PMID: 11605728 DOI: 10.1177/172460080101600302] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective Atherosclerosis is a fibroproliferative disease which has been attributed to several factors including genetic and molecular alterations. Initial studies have shown genetic alterations at the microsatellite level in the DNA of atherosclerotic plaques. Extending our initial findings, we performed a microsatellite analysis on cerebral atherosclerotic plaques. Methods Twenty-seven cerebral atherosclerotic plaques were assessed for loss of heterozygosity (LOH) and microsatellite instability (MI) using 25 microsatellite markers located on chromosomes 2, 8, 9 and 17. DNA was extracted from the vessels as well as the respective blood from each patient and subjected to polymerase chain reaction. Results Our analyses revealed that specific loci on chromosomes 2, 8, 9 and 17 exhibited a significant incidence of LOH. Forty-six percent of the specimens showed loss of heterozygosity at 2p13–p21, 48% exhibited LOH at 8p12–q11.2, while allelic imbalance was detected in 47% of the cases. The LOH incidence was 39%, 31% and 27% at 17q21, 9q31–34 and 17p13, respectively. Genetic alterations were detected at a higher rate as compared to the corresponding alterations observed in plaques from other vessels. Discussion This is the first microsatellite analysis using atherosclerotic plaques obtained from cerebral vessels. Our results indicate an elevated mutational rate on specific chromosomal loci, suggesting a potential implication of these regions in atherogenesis.
Collapse
MESH Headings
- Chromosomes, Human, Pair 1
- Chromosomes, Human, Pair 17
- Chromosomes, Human, Pair 2
- Chromosomes, Human, Pair 8
- Chromosomes, Human, Pair 9
- Genetic Markers
- Humans
- Intracranial Arteriosclerosis/genetics
- Loss of Heterozygosity
- Microsatellite Repeats
Collapse
Affiliation(s)
- P Miniati
- Department of Forensic Sciences, University of Crete, Heraklion, Greece
| | | | | | | |
Collapse
|
245
|
Salbe C, Trevisiol C, Ferruzzi E, Mancuso T, Nascimbeni R, Di Fabio F, Salerni B, Dittadi R. Molecular Detection of Codon 12 K-RAS Mutations in Circulating DNA from Serum of Colorectal Cancer Patients. Int J Biol Markers 2018; 15:300-7. [PMID: 11192825 DOI: 10.1177/172460080001500404] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Point mutations of the K-RAS gene at codon 12 are found in about 40% of cases with colorectal cancer. The diagnostic implications of the detection of these mutations and their clinical utility are still unclear. The aim of this study was to test both the feasibility of the detection of the mutated K-RAS gene in serum and its potential role in colorectal cancer detection and monitoring. Codon 12 K-RAS mutations were examined in DNA extracted from the serum of 35 patients with colorectal cancer and were compared with the K-RAS status in the corresponding primary tumor. Molecular detection was performed by the mutant-enriched PCR (ME-PCR) assay, a sensitive method capable of distinguishing a small quantity of mutated DNA in the presence of abundant wild-type DNA. The occurrence of mutations was compared with clinicopathological parameters as well as CEA and CA19.9 serum levels. We found codon 12 K-RAS mutations in the tissue of 13/35 (37%) patients. Serum mutations were detected in 5/13 (38.5%) patients with mutated K-RAS in the tissue. 26/35 (74%) patients showed an identical K-RAS pattern in tissue and serum. No codon 12 K-RAS alterations were found in serum samples of 22 patients with benign gastrointestinal diseases. Elevated serum CEA levels were detected in 16 patients, four of whom also presented serum RAS mutations. Our results confirm that K-RAS mutations can be found in circulating DNA extracted from serum samples of patients with colorectal cancer and show that there is a correspondence between serum and tissue K-RAS patterns.
Collapse
Affiliation(s)
- C Salbe
- Center for Biological Markers of Malignancy, Regional Hospital ULSS 12, Venice, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Itatani Y, Kawada K, Sakai Y. Treatment of Elderly Patients with Colorectal Cancer. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2176056. [PMID: 29713641 PMCID: PMC5866880 DOI: 10.1155/2018/2176056] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/11/2018] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. As society ages, the number of elderly patients with CRC will increase. The percentage of patients with right-sided colon cancer and the incidence of microsatellite instability are higher in elderly than in younger patients with CRC. Moreover, the higher incidence of comorbid diseases in elderly patients indicates the need for less invasive treatment strategies. For example, care should be taken in performing additional surgery after endoscopic submucosal dissection for elderly patients with high-risk T1 CRC. Minimally invasive surgery, such as laparoscopic colectomy, would be preferable for elderly patients with CRC. Chemotherapy for elderly patients requires careful monitoring for adverse events. The aim of this review is to summarize the clinicopathological features of CRC in elderly patients, optical surgical strategies, including endoscopic and laparoscopic resection, and chemotherapeutic strategies, including postoperative adjuvant chemotherapy and systemic chemotherapy for unresectable CRC.
Collapse
Affiliation(s)
- Yoshiro Itatani
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
247
|
Mehrvarz Sarshekeh A, Overman MJ, Kopetz S. Nivolumab in the treatment of microsatellite instability high metastatic colorectal cancer. Future Oncol 2018; 14:1869-1874. [PMID: 29473436 DOI: 10.2217/fon-2017-0696] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Nivolumab is a PD-1 inhibitor approved for the use in treatment of multiple tumor types (such as melanoma, non-small-cell lung cancer, renal cell carcinoma, urothelial cancer and Hodgkin's lymphoma). In July 2017, the US FDA granted accelerated approval of this agent for the treatment of metastatic colorectal cancer patients whose tumor harbors deficient mismatch repair, or microsatellite-instability high and have progressed on conventional chemotherapy. In this review, we will discuss the use, efficacy, and safety of this agent in microsatellite-instability high metastatic colorectal cancer.
Collapse
Affiliation(s)
- Amir Mehrvarz Sarshekeh
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX 77030, USA
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX 77030, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
248
|
Zeinalian M, Hashemzadeh-Chaleshtori M, Salehi R, Emami MH. Clinical Aspects of Microsatellite Instability Testing in Colorectal Cancer. Adv Biomed Res 2018. [PMID: 29531926 PMCID: PMC5841008 DOI: 10.4103/abr.abr_185_16] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Microsatellite instability (MSI) is a molecular hallmark for some colorectal cancers (CRCs) in which short tandem repeats are prone to mutations along with DNA sequences. It is due to DNA-mismatch-repair system deficiency because of a germline/somatic mutation in mismatch-repair (MMR) genes. The germline mutations lead to Lynch syndrome (LS) while epigenetic gene silencing results in sporadic CRC tumors. We discuss in our paper the most important clinical aspects of MSI testing in CRCs. We reviewed the most reliable relevant studies and clinical trials according to their high-quality methods, particularly within two recent decades. MSI testing is used to classify CRC tumors as MSI-high (MSI-H), MSI-low, and microsatellite stable tumors. MSI-H or MMR deficient tumors have shown the best prognosis among all CRCs, so MSI testing is considered as a good prognostic marker. Moreover, it is used to identify LS among familial CRC patients. There is a diagnostic mutation in BRAF gene (V600E) by which sporadic CRCs could be distinguished from LS associated CRCs, due to its concordance with sporadic CRCs not LS. Although, some previous studies had demonstrated a predictive role for MSI testing in chemotherapy process, emerging some controversial findings in recent studies has not convinced many authors to recommend it as a routine examination to evaluate therapeutic response. Though emerging new molecular findings have opened novel windows to develop clinical management of CRC, MSI testing has remained as an excellent prognostic and diagnostic tool for CRC tumors.
Collapse
Affiliation(s)
- Mehrdad Zeinalian
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hassan Emami
- Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Poursina-Hakim Gastrointestinal Research Center, Isfahan, Iran
| |
Collapse
|
249
|
Akbari MR, Zhang S, Cragun D, Lee JH, Coppola D, McLaughlin J, Risch HA, Rosen B, Shaw P, Sellers TA, Schildkraut J, Narod SA, Pal T. Correlation between germline mutations in MMR genes and microsatellite instability in ovarian cancer specimens. Fam Cancer 2018; 16:351-355. [PMID: 28176205 DOI: 10.1007/s10689-017-9973-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A high proportion of ovarian cancers from women who carry germline mutations in mismatch repair (MMR) genes demonstrate microsatellite instability (MSI). The utility of pre-screening ovarian cancer specimens for MSI to identify potential patients for germline screening for MMR mutations is uncertain. 656 women with malignant ovarian cancer underwent both MSI testing and germline mutation testing for large rearrangements in three MMR genes, MLH1, MSH2 and MSH6. Germline DNA sequencing data for the same genes was available. Among the 656 women, only four (0.6%) carried a clearly pathogenic MMR mutation. All four cancers from patients with mutations had loss of two or more microsatellite markers (MSI-high). Eighty-four of 652 (13.0%) women without a mutation had MSI-high ovarian cancers. Using MSI-high as a prescreening criterion, the sensitivity of MSI testing to identify germline MMR gene mutations was 100% and the positive predictive value was 4.5%. Germline mutations in MLH1, MSH2 and MSH6 are rare among unselected cases of ovarian cancer. Patients with germline mutations often will have MSI-positive cancers and pre-screening of ovarian cancer specimens may be an efficient way of identifying patients with Lynch syndrome.
Collapse
Affiliation(s)
- Mohammad R Akbari
- Women's College Research Institute, Women's College Hospital, University of Toronto, 790 Bay Street, 7th Floor, Toronto, ON, M5G 1N8, Canada
| | - Shiyu Zhang
- Women's College Research Institute, Women's College Hospital, University of Toronto, 790 Bay Street, 7th Floor, Toronto, ON, M5G 1N8, Canada
| | - Deborah Cragun
- Departments of Cancer Epidemiology, Moffitt Cancer Center, Biostatistics, Anatomic Pathology, and Experimental Therapeutics, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Ji-Hyun Lee
- Departments of Cancer Epidemiology, Moffitt Cancer Center, Biostatistics, Anatomic Pathology, and Experimental Therapeutics, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Domenico Coppola
- Departments of Cancer Epidemiology, Moffitt Cancer Center, Biostatistics, Anatomic Pathology, and Experimental Therapeutics, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - John McLaughlin
- Samuel Lunenfeld Research Institute, and Dalla Lana School of Public Health, University of Toronto, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College St., New Haven, CT, 06510, USA
| | - Barry Rosen
- Department of Gynecology-Oncology, Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, ON, M5T 2M9, Canada.,Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toronto, 610 University Avenue, Toronto, ON, M5T 2M9, Canada
| | - Patricia Shaw
- Department of Pathology, Princess Margaret Hospital, 610 University Avenue, Toronto, ON, M5T 2M9, Canada
| | - Thomas A Sellers
- Departments of Cancer Epidemiology, Moffitt Cancer Center, Biostatistics, Anatomic Pathology, and Experimental Therapeutics, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Joellen Schildkraut
- Department of Community and Family Medicine, Duke Comprehensive Cancer Center, Duke University Medical Center, Box 2949, Durham, NC, 27710, USA
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, University of Toronto, 790 Bay Street, 7th Floor, Toronto, ON, M5G 1N8, Canada.
| | - Tuya Pal
- Departments of Cancer Epidemiology, Moffitt Cancer Center, Biostatistics, Anatomic Pathology, and Experimental Therapeutics, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| |
Collapse
|
250
|
Zheng J, Huang B, Nie X, Zhu Y, Han N, Li Y. The clinicopathological features and prognosis of tumor MSI in East Asian colorectal cancer patients using NCI panel. Future Oncol 2018; 14:1355-1364. [PMID: 29366338 DOI: 10.2217/fon-2017-0662] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
AIM To compare test results obtained from a PCR assay for the National Cancer Institute (NCI) five loci criteria for detecting microsatellite instability (MSI) with those obtained from immunohistochemistry of mismatch repair and a five-mononucleotide site amplification system in East Asian patients with colorectal cancer. PATIENTS & METHODS A total of 245 East Asian patients with colorectal cancer were studied retrospectively at our institution. RESULTS The consistency of the NCI panel PCR method compared with detection of mismatch repair protein expression by immunohistochemistry was 0.898. High level MSI (MSI-H) status was correlated with the Tumor, Node, Metastasis stage, tumor location site, metastasis, tumor grade, mucinous histological type and BRAF-type mutations. CONCLUSION The NCI panel PCR assay has excellent sensitivity and specificity for detecting MSI in an East Asian population.
Collapse
Affiliation(s)
- Jianmin Zheng
- Department of Pathology, Changhai Hospital of Shanghai, 168 Changhai Road, Shanghai 200433, PR China
| | - Bangxing Huang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| | - Xiu Nie
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| | - Yan Zhu
- Department of Pathology, Changhai Hospital of Shanghai, 168 Changhai Road, Shanghai 200433, PR China
| | - Ningning Han
- Department of Clinical Medicine, Shanghai Tongshu Biotech Co. Ltd, Shanghai 200120, PR China
| | - Yan Li
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| |
Collapse
|