Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Clin Lab Anal ; 35(5): e23746, 2021 May.
Article in English | MEDLINE | ID: mdl-33826163

ABSTRACT

BACKGROUND: Mismatch repair deficiency (dMMR) status induced by MLH1 protein deficiency plays a pivotal role in therapeutic decision-making for cancer patients. Appropriate quality control (QC) materials are necessary for monitoring the accuracy of MLH1 protein deficiency assays used in clinical laboratories. METHODS: CRISPR/Cas9 technology was used to edit the MLH1 gene of GM12878Cas9 cells to establish MLH1 protein-deficient cell lines. The positive cell lines were screened and validated by Sanger sequencing, Western blot (WB), and next-generation sequencing (NGS) and were then used to prepare formalin-fixed, paraffin-embedded (FFPE) samples through xenografting. These FFPE samples were tested by hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) for suitability as novel QC materials for MLH1 protein deficiency testing. RESULTS: We successfully cultured 358 monoclonal cells, with a survival rate of 37.3% (358/960) of the sorted monoclonal cells. Through Sanger sequencing, cell lines with MLH1 gene mutation were identified. Subsequently, two cell lines with MLH1 protein deficiency were identified by WB and named as GM12878Cas9_6 and GM12878Cas9_10. The NGS results further confirmed that the MLH1 gene mutation in these two cell lines would cause the formation of stop codons and terminate the expression of the MLH1 protein. The H&E staining and IHC results also verified the deficiency of the MLH1 protein, and FFPE samples from xenografts proved their similarity and consistency with clinical samples. CONCLUSIONS: We successfully established MLH1 protein-deficient cell lines. Followed by xenografting, we developed novel FFPE QC materials with homogenous, sustainable, and typical histological structures advantages that are suitable for the standardization of clinical IHC methods.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/standards , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/genetics , Xenograft Model Antitumor Assays , Animals , Base Sequence , Cell Line , Humans , Mice, Inbred NOD , Mice, SCID , Mutation/genetics , Quality Control
2.
Anticancer Res ; 41(2): 975-982, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33517304

ABSTRACT

BACKGROUND/AIM: DNA mismatch repair (MMR) deficiency has received increasing attention as a biomarker of anti-PD-1 treatments of solid tumors including gastric cancer (GC). However, efficient screening has not been established. PATIENTS AND METHODS: A total of 513 patients were tested for the expression of MMR proteins by immunohistochemistry to identify MMR deficient GC. Development of a prediction model was attempted using the common clinicopathological features. RESULTS: In total, 11% (57/513) of the patients showed loss of expression of either one or more MMR proteins (MMR protein deficiency; MMR-D). Multivariate analysis demonstrated that age (≥70 years), sex (female), tumor location (lower 1/3), depth invasion (low, T1/T2/T3), and absence of distant metastasis were significantly independent predictive factors of MMR-D GCs. The MMR-D GC probability estimated by the prediction model ranged from 0.4% to 62.2%, and the area under the curve of the receiver operating characteristics curve was 0.82 (95% confidence interval=0.75-0.87). CONCLUSION: Our prediction model can sufficiently and efficiently identify MMR-D GCs using clinical features.


Subject(s)
DNA Mismatch Repair , DNA-Binding Proteins/deficiency , Mismatch Repair Endonuclease PMS2/deficiency , MutL Protein Homolog 1/deficiency , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Area Under Curve , Biomarkers, Tumor/metabolism , Female , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Sex Characteristics , Stomach Neoplasms/metabolism , Tumor Burden , Young Adult
3.
Cancer Cell ; 39(1): 96-108.e6, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33338425

ABSTRACT

Increased neoantigens in hypermutated cancers with DNA mismatch repair deficiency (dMMR) are proposed as the major contributor to the high objective response rate in anti-PD-1 therapy. However, the mechanism of drug resistance is not fully understood. Using tumor models defective in the MMR gene Mlh1 (dMLH1), we show that dMLH1 tumor cells accumulate cytosolic DNA and produce IFN-ß in a cGAS-STING-dependent manner, which renders dMLH1 tumors slowly progressive and highly sensitive to checkpoint blockade. In neoantigen-fixed models, dMLH1 tumors potently induce T cell priming and lose resistance to checkpoint therapy independent of tumor mutational burden. Accordingly, loss of STING or cGAS in tumor cells decreases tumor infiltration of T cells and endows resistance to checkpoint blockade. Clinically, downregulation of cGAS/STING in human dMMR cancers correlates with poor prognosis. We conclude that DNA sensing within tumor cells is essential for dMMR-triggered anti-tumor immunity. This study provides new mechanisms and biomarkers for anti-dMMR-cancer immunotherapy.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Membrane Proteins/genetics , MutL Protein Homolog 1/deficiency , Neoplasms/genetics , Nucleotidyltransferases/genetics , Animals , Cell Line, Tumor , DNA Mismatch Repair , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interferon-beta/metabolism , Membrane Proteins/metabolism , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Nucleotidyltransferases/metabolism , Prognosis , Signal Transduction/drug effects
4.
Cancer Cell ; 39(1): 109-121.e5, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33338427

ABSTRACT

Tumors with defective mismatch repair (dMMR) are responsive to immunotherapy because of dMMR-induced neoantigens and activation of the cGAS-STING pathway. While neoantigens result from the hypermutable nature of dMMR, it is unknown how dMMR activates the cGAS-STING pathway. We show here that loss of the MutLα subunit MLH1, whose defect is responsible for ~50% of dMMR cancers, results in loss of MutLα-specific regulation of exonuclease 1 (Exo1) during DNA repair. This leads to unrestrained DNA excision by Exo1, which causes increased single-strand DNA formation, RPA exhaustion, DNA breaks, and aberrant DNA repair intermediates. Ultimately, this generates chromosomal abnormalities and the release of nuclear DNA into the cytoplasm, activating the cGAS-STING pathway. In this study, we discovered a hitherto unknown MMR mechanism that modulates genome stability and has implications for cancer therapy.


Subject(s)
Chromosome Aberrations , DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/metabolism , MutL Protein Homolog 1/deficiency , Neoplasms/genetics , Signal Transduction , Animals , Cell Line, Tumor , DNA Breaks, Single-Stranded , DNA Mismatch Repair , DNA Repair , DNA, Single-Stranded/metabolism , HeLa Cells , Humans , Membrane Proteins/metabolism , Mice , MutL Protein Homolog 1/metabolism , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Replication Protein A/metabolism
5.
Int J Cancer ; 148(3): 759-768, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33105024

ABSTRACT

Lymphocyte activation gene 3 (LAG-3) is a transmembrane immune checkpoint that facilitates immune escape via suppressing T-cell-mediated anti-tumor immunity. The role of LAG-3 in gastric cancer is little known. Consequently, we assessed the clinical significance of LAG-3 in gastric cancer. In our study, patients with gastric cancer from Zhongshan Hospital (n = 464) and data from the Asian Cancer Research Group (n = 300) were analyzed. LAG-3+ cell infiltration and other immune contexture in gastric cancer were detected by immunohistochemistry. Kaplan-Meier curves and log-rank test were used for survival analyses. Intratumoral LAG-3+ cells mainly accumulated in Epstein-Barr virus (EBV)-positive (EBV subtype) and MLH1-defective (dMLH1 subtype) gastric cancer. Furthermore, LAG-3+ cell infiltration was strongly associated with inferior clinical outcomes in patients with these two subtypes of gastric cancer. Moreover, we found intratumoral LAG-3+ cell high infiltration was associated with an immunoevasive contexture featured by decreased IFN-γ+ cells and perforin-1+ cells, but increased regulatory T cells and M2-like macrophages in EBV/dMLH1 subtype of gastric cancer. LAG-3 was a poor prognostic factor and might be a potential immunotherapeutic target in EBV-positive and MLH1-defective gastric cancer.


Subject(s)
Antigens, CD/metabolism , Epstein-Barr Virus Infections/genetics , MutL Protein Homolog 1/deficiency , Stomach Neoplasms/virology , Cohort Studies , Epstein-Barr Virus Infections/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Analysis , Lymphocyte Activation Gene 3 Protein
6.
Int J Gynecol Cancer ; 30(12): 1951-1958, 2020 12.
Article in English | MEDLINE | ID: mdl-33082239

ABSTRACT

OBJECTIVES: For synchronous endometrial and ovarian cancers, most centers rely on mismatch repair testing of the endometrial cancer to identify Lynch syndrome, and neglect the ovarian tumor site completely. We examined the mismatch repair immunohistochemistry and microsatellite instability results from the endometrium and ovary to assess discordance between the tumor sites and between tests. METHODS: 30 women with newly diagnosed synchronous endometrial and ovarian cancer were prospectively recruited from three cancer centers in Ontario, Canada. Both tumor sites were assessed for mismatch repair deficiency by immunohistochemistry and microsatellite instability test; discordance in results between tumor sites and discordance between test results at each site was examined. Cases with discordant results had tumors sequenced with a targeted panel in order to reconcile the findings. All women underwent mismatch repair gene germline testing. RESULTS: Of 30 patients, 11 (37%) were mismatch repair deficient or microsatellite instable at either tumor site, with 5 (17%) testing positive for Lynch syndrome. Mismatch repair immunohistochemistry expression was discordant between endometrial and ovarian tumor sites in 2 of 27 patients (7%) while microsatellite instability results were discordant in 2 of 25 patients (8%). Relying on immunohistochemistry or microsatellite instability alone on the endometrial tumor would have missed one and three cases of Lynch syndrome, respectively. One patient with Lynch syndrome with a PMS2 pathogenic variant was not detected by either immunohistochemistry or microsatellite instability testing. The rate of discordance between immunohistochemistry and microsatellite instability test was 3.8% in the ovary and 12% in the endometrium. CONCLUSIONS: There was discordance in immunohistochemistry and microsatellite instability results between tumor sites and between tests within each site. Endometrial tumor testing with mismatch repair immunohistochemistry performed well, but missed one case of Lynch syndrome. Given the high incidence of Lynch syndrome (17%), consideration may be given to germline testing in all patients with synchronous endometrial and ovarian cancers.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , DNA Mismatch Repair , Endometrial Neoplasms/genetics , Neoplasms, Multiple Primary/genetics , Ovarian Neoplasms/genetics , Adult , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Microsatellite Instability , Middle Aged , Mismatch Repair Endonuclease PMS2/deficiency , Mismatch Repair Endonuclease PMS2/genetics , Mismatch Repair Endonuclease PMS2/metabolism , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , Neoplasms, Multiple Primary/metabolism , Neoplasms, Multiple Primary/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
7.
Gynecol Oncol ; 159(3): 712-720, 2020 12.
Article in English | MEDLINE | ID: mdl-33046272

ABSTRACT

OBJECTIVE: To evaluate clinicopathologic characteristics and survival impact associated with mismatch repair (MMR) deficient subgroups of endometrial cancer (EC) in patients undergoing universal screening for Lynch Syndrome. METHODS: A retrospective cohort study using a prospectively maintained gynecologic oncology registry of patients who underwent surgery for EC was conducted. All pathology specimens underwent tumor testing using immunohistochemistry for MMR deficiency with reflex MLH1 promotor methylation testing. Tumors were classified as MMR-I (intact MMR expression), MMR-DM (MMR deficient due to MLH1 methylation), and MMR-DU (MMR deficient without MLH1 methylation). Univariate and multivariate analysis performed to determine factors associated with MMR-DM. Progression-free survival (PFS) and overall survival (OS) analyzed by stage and endometrioid subgroup. RESULTS: From 2012 to 2016, 1018 EC patients were identified and screened. Overall, 71.6% were classified as MMR-I, 23.8% MMR-DM, and 4.6% MMR-DU. In comparison to MMR-DU, MMR-DM tumors were associated with older age, postmenopausal status, lymphovascular space invasion, and pure endometrioid histology. Compared to MMR-I, MMR-DM tumors were associated with older age, endometrioid histology, lymphovascular space invasion, and higher grade on multivariable analysis. There was no difference in PFS and OS between the three groups overall. In patients with endometrioid EC, MMR-DM tumors were associated with lower PFS vs. MMR-I (HR:2.51, CI:1.54, 4.10, P < 0.001). This effect persisted for stage I/II endometrioid EC (HR 2.66, CI:1.34, 5.26 p = 0.005). No difference in PFS or OS was noted among stage III/IV endometrioid tumors. CONCLUSION: MMR deficiency is associated with adverse prognostic factors and worse PFS among endometrioid tumors, particularly in early stage EC. MMR testing outside of LS screening has prognostic value, warranting consideration for inclusion as a biomarker in prospective clinical trials.


Subject(s)
Carcinoma, Endometrioid/mortality , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , DNA Mismatch Repair , Early Detection of Cancer/statistics & numerical data , Endometrial Neoplasms/mortality , Age Factors , Aged , Carcinoma, Endometrioid/diagnosis , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/surgery , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Methylation , Early Detection of Cancer/methods , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/surgery , Endometrium/pathology , Endometrium/surgery , Female , Humans , Hysterectomy , Middle Aged , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/genetics , Mutation , Neoplasm Grading , Neoplasm Invasiveness/genetics , Neoplasm Staging , Prognosis , Progression-Free Survival , Promoter Regions, Genetic/genetics , Prospective Studies , Registries/statistics & numerical data , Retrospective Studies , Risk Factors
8.
Anticancer Res ; 40(8): 4379-4385, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32727766

ABSTRACT

BACKGROUND/AIM: Identifying patients with DNA mismatch repair-deficient (dMMR) colorectal cancer (CRC) is vital to improve treatment and identify patients with Lynch syndrome (LS). We developed a prediction model for dMMR CRC using clinicopathologic features. PATIENTS AND METHODS: We reviewed the medical records of 1,147 patients who underwent resection of stage I-IV CRC in whom universal screening for LS using immunohistochemistry for MMR proteins had performed. Univariate and multivariate logistic regression analyses were used to build a prediction model of dMMR CRC. RESULTS: The prevalence of dMMR CRC was 5.2%. Age (≥75 years), tumor location (right-sided colon), main histologic features (poor differentiation), maximum tumor size (≥65 mm), and stage (I/II) were independent significant variables related to dMMR. We created a formula for predicting the likelihood of dMMR, and the probability ranged from 0.2% to 83%. CONCLUSION: dMMR CRC can be identified efficiently using clinicopathologic features obtained in daily clinical practice.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , DNA Repair Enzymes/deficiency , Adult , Age Factors , Aged , Aged, 80 and over , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/deficiency , Female , Humans , Logistic Models , Male , Middle Aged , Mismatch Repair Endonuclease PMS2/deficiency , Models, Genetic , MutL Protein Homolog 1/deficiency , MutS Homolog 2 Protein/deficiency , Neoplasm Staging , Prevalence , Risk Factors
9.
Eur J Hum Genet ; 28(11): 1555-1562, 2020 11.
Article in English | MEDLINE | ID: mdl-32661327

ABSTRACT

BRAF and MLH1 promoter methylation testings have been proven effective prescreens for Lynch Syndrome. We aimed to compare different screening strategies for Lynch Syndrome in patients with MLH1(-) CRC. Patients with MLH1(-) CRC who had been tested for BRAF mutation and germline variants of DNA mismatch repair genes were included. We compared the sensitivities and specificities for identifying Lynch Syndrome and the cost-effectiveness of four screening approaches that used the following tests as prescreens: BRAF testing, MLH1 methylation testing, MLH1 methylation & BRAF testing, and MLH1 methylation testing & Revised Bethesda Criteria. Of 109 patients included, 23 (21.1%) were Lynch Syndrome patients. BRAF mutation and MLH1 methylation occurred in 6 (5.5%) and 40 (36.7%) patients, respectively. The sensitivity for identifying Lynch syndrome of BRAF testing was 100%, but the specificity was only 7%. MLH1 methylation testing had a lower sensitivity than BRAF testing (97.5% vs 100%), but had a markedly higher specificity (45.3% vs 7%). The combination of the two testings had a slightly higher specificity than MLH1 methylation testing alone (47.7% vs 45.3%). The MLH1 methylation testing approach had a 10% lower cost of identifying MLH1(-) Lynch syndrome carriers per case than universal genetic testing, but it missed 4.5% of patients. BRAF and MLH1 promoter methylation testings as prescreens for Lynch syndrome are less effective in Chinese patients with MLH1(-) CRC than in their Western counterparts. Universal genetic testing could be considered an up-front option for this population.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Genetic Testing/methods , MutL Protein Homolog 1/deficiency , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Costs and Cost Analysis , DNA Methylation , Female , Genetic Testing/economics , Genetic Testing/standards , Humans , Male , Middle Aged , MutL Protein Homolog 1/genetics , Mutation , Proto-Oncogene Proteins B-raf
10.
Int J Radiat Oncol Biol Phys ; 108(4): 1091-1102, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32629081

ABSTRACT

PURPOSE: Humans are exposed to charged particles in different scenarios. The use of protons and high-linear energy transfer (LET) in cancer treatment is steadily growing. In outer space, astronauts will be exposed to a mixed radiation field composed of both protons and heavy ions, in particularly the long-term space missions outside of earth's magnetosphere. Thus, understanding the radiobiology and transforming potential of these types of ionizing radiation are of paramount importance. METHODS AND MATERIALS: We examined the effect of 10 or 100 cGy of whole-body doses of protons or 28Si ions on the hematopoietic system of a genetic model of aging based on recent studies that showed selective loss of the MLH1 protein in human hematopoietic stems with age. RESULTS: We found that Mlh1 deficient animals are highly prone to develop lymphomas when exposed to either low doses of protons or 28Si ions. The lymphomas that develop are genetically indistinguishable, in spite of different types of damage elicited by low- and high-LET radiation. RNA sequencing analyses reveal similar gene expression patterns, similar numbers of altered genes, similar numbers of single nucleotide variants and insertions and deletions, and similar activation of known leukemogenic loci. CONCLUSIONS: Although the incidence of malignancy is related to radiation quality, and increased due to loss of Mlh1, the phenotype of the tumors is independent of LET.


Subject(s)
Hematopoietic System/radiation effects , Linear Energy Transfer , Lymphoma/genetics , MutL Protein Homolog 1/deficiency , Neoplasms, Radiation-Induced/genetics , Protons/adverse effects , Silicon/adverse effects , Aging , Animals , DNA Mismatch Repair , Disease Models, Animal , Female , Gene Expression Profiling , Hematopoietic System/physiology , Humans , Lymphoma/pathology , Male , Mice , MutL Protein Homolog 1/genetics , Neoplasms, Radiation-Induced/pathology , Penetrance , Radiation Exposure/adverse effects , Sequence Analysis, RNA/methods , Space Flight , Whole-Body Irradiation/adverse effects , Whole-Body Irradiation/methods
11.
Fam Cancer ; 19(4): 323-336, 2020 10.
Article in English | MEDLINE | ID: mdl-32363481

ABSTRACT

Germline pathogenic variants in the DNA mismatch repair genes (MMR): MLH1, MSH2, MSH6, and PMS2, are causative of Lynch syndrome (LS). However, many of the variants mapping outside the invariant splice site positions (IVS ± 1, IVS ± 2) are classified as variants of unknown significance (VUS). Three such variants (MLH1 c.588+5G>C, c.588+5G>T and c.677+5G>A) were identified in 8 unrelated LS families from Argentina, Brazil and Chile. Herein, we collected clinical information on these families and performed segregation analysis and RNA splicing studies to assess the implication of these VUS in LS etiology. Pedigrees showed a clear pattern of variant co-segregation with colorectal cancer and/or other LS-associated malignancies. Tumors presented deficient expression of MLH1-PMS2 proteins in 7/7 of the LS families, and MSI-high status in 3/3 cases. Moreover, RNA analyses revealed that c.588+5G>C and c.588+5G>T induce skipping of exon 7 whereas c.677+5G>A causes skipping of exon 8. In sum, we report that the combined clinical findings in the families and the molecular studies provided the evidences needed to demonstrate that the three MLH1 variants are causative of LS and to classify c.588+5G>C and c.677+5G>A as class 5 (pathogenic), and c.588+5G>T as class 4 (likely-pathogenic). Our findings underline the importance of performing clinical and family analyses, as well as RNA splicing assays in order to determine the clinical significance of intronic variants, and contribute to the genetic counseling and clinical management of patients and their relatives.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Introns , MutL Protein Homolog 1/genetics , RNA Splice Sites , RNA Splicing , Adult , Argentina , Brazil , Chile , Colorectal Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , DNA Mismatch Repair , Exons , Female , Genetic Counseling , Humans , Male , Middle Aged , Mismatch Repair Endonuclease PMS2/deficiency , Mismatch Repair Endonuclease PMS2/genetics , Mismatch Repair Endonuclease PMS2/metabolism , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/metabolism , Pedigree , Protein Isoforms
12.
Genes Chromosomes Cancer ; 59(10): 562-568, 2020 10.
Article in English | MEDLINE | ID: mdl-32427409

ABSTRACT

Oncogenic gene fusions represent attractive targets for therapy of cancer. However, the frequency of actionable genomic rearrangements in colorectal cancer (CRC) is very low, and universal screening for these alterations seems to be impractical and costly. To address this problem, several large scale studies retrospectivelly showed that CRC with gene fusions are highly enriched in groups of tumors defined by MLH1 DNA mismatch repair protein deficiency (MLH1d), and hypermethylation of MLH1 promoter (MLH1ph), and/or the presence of microsatellite instability, and BRAF/KRAS wild-type status (BRAFwt/KRASwt). In this study, we used targeted next generation sequencing (NGS) to explore the occurence of potentially therapeutically targetable gene fusions in an unselected series of BRAFwt/KRASwt CRC cases that displayed MLH1d/MLH1ph. From the initially identified group of 173 MLH1d CRC cases, 141 cases (81.5%) displayed MLH1ph. BRAFwt/RASwt genotype was confirmed in 23 of 141 (~16%) of MLH1d/MLH1ph cases. Targeted NGS of these 23 cases identified oncogenic gene fusions in nine patients (39.1%; CI95: 20.5%-61.2%). Detected fusions involved NTRK (four cases), ALK (two cases), and BRAF genes (three cases). As a secondary outcome of NGS testing, we identified PIK3K-AKT-mTOR pathway alterations in two CRC cases, which displayed PIK3CA mutation. Altogether, 11 of 23 (~48%) MLH1d/MLH1ph/BRAFwt/RASwt tumors showed genetic alterations that could induce resistance to anti-EGFR therapy. Our study confirms that targeted NGS of MLH1d/MLH1ph and BRAFwt/RASwt CRCs could be a cost-effective strategy in detecting patients with potentially druggable oncogenic kinase fusions.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Genetic Testing/methods , High-Throughput Nucleotide Sequencing/methods , MutL Protein Homolog 1/deficiency , Oncogene Proteins, Fusion/genetics , Adenocarcinoma/diagnosis , Adult , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase/genetics , Colorectal Neoplasms/diagnosis , DNA Methylation , Female , Genetic Testing/standards , High-Throughput Nucleotide Sequencing/standards , Humans , Male , Middle Aged , MutL Protein Homolog 1/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptor Protein-Tyrosine Kinases/genetics
13.
Endocr Pathol ; 31(4): 411-417, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32388775

ABSTRACT

Pancreatic neuroendocrine carcinomas (NECs) are rare and very aggressive neoplasms with dismal prognosis, especially when metastatic or with negative prognostic factors, such as vascular invasion. To the best of our knowledge, no case of pancreatic NEC with mismatch repair deficiency has been reported to date. We describe a 62-year-old patient who underwent pancreaticoduodenectomy for a NEC located in the pancreatic head, with peripancreatic lymph node metastases. Tumor necrosis was prominent and the Ki67 proliferative index was 60%. One year after the diagnosis, the patient experienced recurrence with a left supraclavicular lymph node metastasis, which was surgically removed, followed by standard cisplatin-etoposide chemotherapy. Neoplastic cells showed combined loss of expression of MLH1 and PMS2 in both primary tumor and lymph node metastasis. Microsatellite instability (MSI) test using a mononucleotide repeats pentaplex PCR (BAT-25, BAT-26, NR-21, NR-22, and NR-24) revealed minimal mononucleotide shifts showing deletion of less than 3 bp at NR-21, BAT-26, NR-24, and NR-22 loci. MLH1 methylation analysis revealed absence of the gene promoter methylation. BRAF and KRAS mutations were not detected. In gut, NECs' mismatch repair deficiency phenotype has been reported in about 10% of cases, and it represents an independent factor of more favorable outcome. Likewise, our patient is currently alive with a follow-up of more than 12 years after pancreaticoduodenectomy, by itself an unexpected finding for such an aggressive neoplasm.


Subject(s)
Carcinoma, Neuroendocrine , Mismatch Repair Endonuclease PMS2/deficiency , MutL Protein Homolog 1/deficiency , Pancreatic Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/therapy , Cisplatin/therapeutic use , Combined Modality Therapy , DNA Mismatch Repair/physiology , Etoposide/therapeutic use , Humans , Male , Microsatellite Instability , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Pancreaticoduodenectomy/methods , Remission Induction , Pancreatic Neoplasms
14.
Gynecol Oncol ; 157(1): 245-251, 2020 04.
Article in English | MEDLINE | ID: mdl-31980219

ABSTRACT

OBJECTIVES: To apply the Proactive Molecular Risk Classifier for Endometrial Cancer (ProMisE) to a consecutive series of endometrial cancer (EC) patients diagnosed at a tertiary referral center and assign EC specimens to one of four molecular subgroups using immunohistochemistry (IHC) for p53/mismatch repair protein expression and sequencing for Polymerase Epsilon Exonuclease Domain Mutations (POLE-EDM). Mismatch Repair Deficient (MMR-D) cases were more thoroughly investigated to identify underlying somatic or germline genetic defects. METHODS: Hundred-and eight consecutive endometrial cancer patients, diagnosed between March 2017 and April 2019, were subjected to immunohistochemical and molecular analysis, according to ProMisE. IHC for p53 and the mismatch repair proteins (MLH1, PMS2, MSH6 and PMS2) was performed. All patients were also tested for POLE-EDM by Sanger sequencing. In addition, tumor and corresponding normal tissue of cases with abnormal MMR IHC were tested by PCR for microsatellite instability (MSI) (MSI analysis system, Promega). Hypermethylation of MLH1 promotor was tested with (methylation specific) multiplex ligation dependent probe amplification. MMR-D cases were subjected to germline mutation analysis of the mismatch repair genes, using next generation sequencing on MiSeq (Illumina) with the BRCA Hereditary Cancer MASTR Plus, (Multiplicom/Agilent), RNA mutation analysis and MLPA. RESULTS: FIGO classification was stage IA (n = 54), IB (n = 22) II(n = 8), III(n = 18) and IV(n = 6). Of the 33 patients with MMR-D on IHC (31%), 26 showed MLH1 promotor hypermethylation as the probable cause of MMR-D. The remaining 7 patients without MLH1 promotor hypermethylation were referred for germline analysis of Lynch syndrome. Six patients carried a pathogenic germline mutation in one of the mismatch repair genes: MSH6(n = 3), PMS2(n = 1), MLH1(n = 1) and MSH2 (n = 1). Pathogenic POLE-EDM were identified in 7 (6%) patients. Multiple molecular features (POLE-EDM + MMR-D or POLE-EDM + p53 abnormal) were observed in 4 patients (4%). A high concordance between MMR-D and microsatellite instability was observed in our cohort. In cases of a genetic defect in the MMR genes, we do note a large proportion of cases exhibiting microsatellite instability. On the contrary a hypermutation state, as seen in POLE EDM, does not result in accompanied phenotypic changes in MSI status. CONCLUSION: The ProMisE classification proved to be an efficient and easily implementable system. Future research should elucidate the precise biological and prognostic meaning of the cases with multiple molecular markers.


Subject(s)
DNA Mismatch Repair , DNA Repair Enzymes/genetics , Endometrial Neoplasms/classification , Aged , Aged, 80 and over , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Microsatellite Instability , Middle Aged , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , Neoplasm Staging , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Tumor Suppressor Protein p53/genetics
15.
Gynecol Oncol ; 156(3): 669-675, 2020 03.
Article in English | MEDLINE | ID: mdl-31924330

ABSTRACT

OBJECTIVE: Mismatch repair (MMR) deficiency and Bethesda panel microsatellite instability (MSI) are increasingly analyzed to identify tumors that might benefit from immune checkpoint inhibitors, but tumor heterogeneity is a potential obstacle for such analyses. In ovarian cancer, data on intratumoral heterogeneity of MMR deficiency/MSI are lacking. METHODS: N = 582 ovarian cancers were screened for MMR deficiency by immunohistochemistry (IHC) on a tissue microarray. 10 cases suspect for MMR deficiency were identified among 478 interpretable cancers and repeated IHC on large sections combined with polymerase chain reaction (PCR)-based MSI analysis validated MMR deficiency/MSI in 9 of these tumors. RESULTS: MMR deficiency/MSI was predominantly seen in endmetrioid cancers (8 of 35, 23%) and also in 1 of 358 serous carcinomas (0.3%), but was absent in 34 mucinous carcinomas, 23 clear cell carcinomas, 17 malignant mixed Mullerian tumors (carcinosarcomas), and 11 mixed carcinomas. MMR deficiency involed protein loss of PMS2/MLH1 in 6 cases and of MSH2 and/or MSH6 in 3 cases. 7 MMR deficient cancers were MSI-high (all endometrioid), one was MSI-low (endometrioid) and one cancer with unequivocal MMR protein loss exhibited microsatellite stability (serous). MLH1 promotor methylation was observed in 4 of 5 endometrioid cancers with MLH1 protein loss. Immunostaining of all available cancer-containing tissue blocks (n = 114) of tumors with confirmed MMR deficiency/MSI revealed uniform MMR status throughout the entire tumor mass. CONCLUSIONS: Our data show that MSI is present in a substantial proportion of endometrioid ovarian cancers but can also occur in other tumor subtypes. MMR deficiency/MSI typically involves the entire tumor mass, suggesting that MMR inactivation occurs early in tumorigenesis in a subset of ovarian cancers.


Subject(s)
DNA Mismatch Repair , DNA Repair Enzymes/deficiency , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Endometrioid/genetics , DNA Repair Enzymes/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Humans , Immunohistochemistry , Microsatellite Instability , Middle Aged , Mismatch Repair Endonuclease PMS2/deficiency , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/deficiency , MutS Homolog 2 Protein/genetics , Tissue Array Analysis , Young Adult
16.
Cell Death Dis ; 10(11): 795, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31641109

ABSTRACT

The DNA mismatch repair (MMR) pathway is responsible for the repair of base-base mismatches and insertion/deletion loops that arise during DNA replication. MMR deficiency is currently estimated to be present in 15-17% of colorectal cancer cases and 30% of endometrial cancers. MLH1 is one of the key proteins involved in the MMR pathway. Inhibition of a number of mitochondrial genes, including POLG and PINK1 can induce synthetic lethality in MLH1-deficient cells. Here we demonstrate for the first time that loss of MLH1 is associated with a deregulated mitochondrial metabolism, with reduced basal oxygen consumption rate and reduced spare respiratory capacity. Furthermore, MLH1-deficient cells display a significant reduction in activity of the respiratory chain Complex I. As a functional consequence of this perturbed mitochondrial metabolism, MLH1-deficient cells have a reduced anti-oxidant response and show increased sensitivity to reactive oxidative species (ROS)-inducing drugs. Taken together, our results provide evidence for an intrinsic mitochondrial dysfunction in MLH1-deficient cells and a requirement for MLH1 in the regulation of mitochondrial function.


Subject(s)
Mitochondria/metabolism , MutL Protein Homolog 1/deficiency , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Mismatch Repair , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , HCT116 Cells , HT29 Cells , Humans , Male , Mitochondria/genetics , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Rotenone/pharmacology , Transfection
17.
SLAS Discov ; 24(5): 548-562, 2019 06.
Article in English | MEDLINE | ID: mdl-30897027

ABSTRACT

DNA mismatch repair (MMR) deficiency has been associated with a higher risk of developing colorectal, endometrial, and ovarian cancer, and confers resistance in conventional chemotherapy. In addition to the lack of treatment options that work efficaciously on these MMR-deficient cancer patients, there is a great need to discover new drug leads for this purpose. In this study, we screened through a library of commercial and semisynthetic natural compounds to identify potential synthetic lethal drugs that may selectively target MLH1 mutants using MLH1 isogenic colorectal cancer cell lines and various cancer cell lines with known MLH1 status. We identified a novel diarylpentanoid analogue, 2-benzoyl-6-(2,3-dimethoxybenzylidene)-cyclohexenol, coded as AS13, that demonstrated selective toxicity toward MLH1-deficient cancer cells. Subsequent analysis suggested AS13 induced elevated levels of oxidative stress, resulting in DNA damage where only the proficient MLH1 cells were able to be repaired and hence escaping cellular death. While AS13 is modest in potency and selectivity, this discovery has the potential to lead to further drug development that may offer better treatment options for cancer patients with MLH1 deficiency.


Subject(s)
Benzylidene Compounds/pharmacology , Colorectal Neoplasms/drug therapy , Cyclohexenes/pharmacology , MutL Protein Homolog 1/genetics , Oxidative Stress/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclohexenes/chemistry , DNA Damage/drug effects , DNA Mismatch Repair/drug effects , Drug Discovery , HT29 Cells , Humans , MutL Protein Homolog 1/deficiency , Synthetic Lethal Mutations/drug effects , Synthetic Lethal Mutations/genetics
18.
PLoS One ; 14(3): e0213411, 2019.
Article in English | MEDLINE | ID: mdl-30856214

ABSTRACT

INTRODUCTION: Colorectal cancers (CRCs) deficient in the DNA mismatch repair protein MutL homolog 1 (MLH1) display distinct clinicopathological features and require a different therapeutic approach compared to CRCs with MLH1 proficiency. However, the molecular basis of this fundamental difference remains elusive. Here, we report that MLH1-deficient CRCs exhibit reduced levels of the cytoskeletal scaffolding protein non-erythroid spectrin αII (SPTAN1), and that tumor progression and metastasis of CRCs correlate with SPTAN1 levels. METHODS AND RESULTS: To investigate the link between MLH1 and SPTAN1 in cancer progression, a cohort of 189 patients with CRC was analyzed by immunohistochemistry. Compared with the surrounding normal mucosa, SPTAN1 expression was reduced in MLH1-deficient CRCs, whereas MLH1-proficient CRCs showed a significant upregulation of SPTAN1. Overall, we identified a strong correlation between MLH1 status and SPTAN1 expression. When comparing TNM classification and SPTAN1 levels, we found higher SPTAN1 levels in stage I CRCs, while stages II to IV showed a gradual reduction of SPTAN1 expression. In addition, SPTAN1 expression was lower in metastatic compared with non-metastatic CRCs. Knockdown of SPTAN1 in CRC cell lines demonstrated decreased cell viability, impaired cellular mobility and reduced cell-cell contact formation, indicating that SPTAN1 plays an important role in cell growth and cell attachment. The observed weakened cell-cell contact of SPTAN1 knockdown cells might indicate that tumor cells expressing low levels of SPTAN1 detach from their primary tumor and metastasize more easily. CONCLUSION: Taken together, we demonstrate that MLH1 deficiency, low SPTAN1 expression, and tumor progression and metastasis are in close relation. We conclude that SPTAN1 is a candidate molecule explaining the tumor progression and metastasis of MLH1-deficient CRCs. The detailed analysis of SPTAN1 is now mandatory to substantiate its relevance and its potential value as a candidate protein for targeted therapy, and as a predictive marker of cancer aggressiveness.


Subject(s)
Carrier Proteins/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Microfilament Proteins/metabolism , MutL Protein Homolog 1/deficiency , Aged , Aged, 80 and over , Caco-2 Cells , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Survival , Cohort Studies , Colorectal Neoplasms/genetics , Disease Progression , Down-Regulation , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology
19.
Leukemia ; 33(5): 1135-1147, 2019 05.
Article in English | MEDLINE | ID: mdl-30275527

ABSTRACT

Cancer-causing genome instability is a major concern during space travel due to exposure of astronauts to potent sources of high-linear energy transfer (LET) ionizing radiation. Hematopoietic stem cells (HSCs) are particularly susceptible to genotoxic stress, and accumulation of damage can lead to HSC dysfunction and oncogenesis. Our group recently demonstrated that aging human HSCs accumulate microsatellite instability coincident with loss of MLH1, a DNA Mismatch Repair (MMR) protein, which could reasonably predispose to radiation-induced HSC malignancies. Therefore, in an effort to reduce risk uncertainty for cancer development during deep space travel, we employed an Mlh1+/- mouse model to study the effects high-LET 56Fe ion space-like radiation. Irradiated Mlh1+/- mice showed a significantly higher incidence of lymphomagenesis with 56Fe ions compared to γ-rays and unirradiated mice, and malignancy correlated with increased MSI in the tumors. In addition, whole-exome sequencing analysis revealed high SNVs and INDELs in lymphomas being driven by loss of Mlh1 and frequently mutated genes had a strong correlation with human leukemias. Therefore, the data suggest that age-related MMR deficiencies could lead to HSC malignancies after space radiation, and that countermeasure strategies will be required to adequately protect the astronaut population on the journey to Mars.


Subject(s)
Environmental Exposure/adverse effects , Hematologic Neoplasms/etiology , MutL Protein Homolog 1/deficiency , Radiation Exposure/adverse effects , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/radiation effects , Disease Models, Animal , Disease Susceptibility , Female , Heterozygote , Humans , INDEL Mutation , Immunohistochemistry , Incidence , Male , Mice , Mice, Knockout , Microsatellite Instability , Polymorphism, Single Nucleotide , Risk Assessment , Risk Factors , Exome Sequencing
20.
Anticancer Res ; 38(11): 6399-6404, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30396964

ABSTRACT

BACKGROUND/AIM: The benefit of IFL (irinotecan, fluorouracil and leucovorin) regimen for metastatic colorectal cancer patients (mCRCs) with high levels of microsatellite instability (MSI-H) or loss of mismatch repair (dMMR) protein expression, is uncertain. This study investigated the association of tumour MMR-status and VEGF-1 expression with response to first-line IFL regimen in mCRCs. PATIENTS AND METHODS: This prospective study analyzed patients diagnosed with mCRC between August 1st, 1998, and August 30th, 2003, at the Turku University Hospital, Finland. All patients received postoperative IFL regimen. Tumour expression of the MMR proteins, hMLH1 and hMSH2, and VEGF-1 expression were assessed by immunohistochemistry (IHC). Tumours with dMMR were those demonstrating loss of MMR protein expression, and tumours with high VEGF-1 expression were those showing moderate or strong cytoplasmic staining. The primary endpoint was the association between tumour hMLH1 or/and hMSH2-deficient and VEGF-1 expression; the relation between tumour MMR-status and IFL response rate was the secondary endpoint. RESULTS: Of the 67 mCRCs patients, 29 (43%) were hMLH1 or/and hMSH2-deficient and 15 (22%) were pMMR mCRCs. At diagnosis, patients with hMLH1 or/and hMSH2-deficient tumours expressed lower levels of VEGF-1 compared to pMMR tumour patients (p=0.01). More than half (n=17, 59%) of those with dMMR were chemosensitive to first-line IFL regimen, while just one-fifth (n=3, 20%) of those with pMMR were chemosensitive to the IFL regimen (p=0.045). CONCLUSION: Association between MMR-status and VEGF-1 expression predicts clinical outcome in mCRC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , MutL Protein Homolog 1/deficiency , MutS Homolog 2 Protein/deficiency , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Colorectal Neoplasms/metabolism , Female , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Humans , Leucovorin/administration & dosage , Leucovorin/therapeutic use , Male , Middle Aged , Neoplasm Metastasis , Prospective Studies , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...