Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
South Asian J Cancer ; 13(2): 132-141, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38919665

ABSTRACT

Atreye MajumdarSambit K. MohantyObjective This article identifies and evaluates the frequency of mutations in the BCR-ABL1 kinase domain (KD) of chronic myeloid leukemia (CML) patients who showed suboptimal response to their current tyrosine kinase inhibitor (TKI) regime and assesses their clinical value in further treatment decisions. Materials and Methods Peripheral and/or bone marrow were collected from 791 CML patients. Ribonucleic acid was extracted, reverse transcribed, and Sanger sequencing method was utilized to detect single-nucleotide variants (SNVs) in BCR-ABL1 KD. Results Thirty-eight different SNVs were identified in 29.8% ( n = 236/791) patients. T315I, E255K, and M244V were among the most frequent mutations detected. In addition, one patient harbored a novel L298P mutation. A subset of patients from the abovementioned harbored compound mutations (13.3%, n = 33/236). Follow-up data was available in 28 patients that demonstrated the efficacy of TKIs in correlation with mutation analysis and BCR-ABL1 quantitation. Molecular response was attained in 50% patients following an appropriate TKI shift. A dismal survival rate of 40% was observed in T315I-harboring patients on follow-up. Conclusion This study shows the incidence and pattern of mutations in one of the largest sets of Indian CML patients. In addition, our findings strengthen the prognostic value of KD mutation analysis among strategies to overcome TKI resistance.

2.
Pathol Res Pract ; 253: 155013, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38096714

ABSTRACT

BACKGROUND: The understanding of molecular mechanisms involved in non-small cell lung carcinoma (NSCLC) has revolutionized significantly in the recent years. These have helped to develop personalized management strategies by identifying specific molecular alterations such as mutations in EGFR, ROS1, BRAF, ERBB2, MET, ALK, and KRAS genes. These mutations are targetable ensuring a better clinical outcome. Next-generation sequencing (NGS) methodology is the recommended technique for the identification of driver mutations in the five hot-spot genes (EGFR, ALK, ROS1, MET, and BRAF) involved in the NSCLC. NGS has numerous advantages including multiplexing, tissue conservation, identification of rare and novel variants, and reduced cost over the sequential single gene testing. Herein, we sought to demonstrate the mutational profile in NSCLC and their clinicopathologic correlation in a contemporary cohort of Indian NSCLC patients. Additionally, we studied the correlation of oncogenic driver mutations with PD-L1 status in these patients. MATERIALS AND METHODS: Five fifty-two stage IV NSCLC patients (adenocarcinoma=490; squamous cell carcinoma=51; adenosquamous carcinoma=5; large cell carcinoma=2; sarcomatoid carcinoma=3; spindle cell carcinoma=1) underwent broad molecular profiling by a custom-made, targeted DNA- and RNA-based five hot-spot genes lung cancer panel (EGFR, ALK, ROS1, BRAF, and MET), compatible with the NGS Ion S5 system. The mutations were correlated with the clinicopathologic characteristics. Additionally, PD-L1 expression status, available on 252 tumors, was correlated with the oncogenic drivers. RESULTS: Validation of the 5 gene panel yielded the following results: a) specificity of 99.74%; b) sensitivity of 100% for single nucleotide variants (SNVs) (>5% variant allele frequency, VAF), indels (>10% VAF) and fusions; c) 100% intra- and inter-run reproducibility; d) 88% inter-laboratory agreement. Validated panel was then used to analyze clinical samples. Sixty percentage tumors harbored either one (54.71%) or multiple (3.26%) mutations. EGFR and BRAF V600E mutations, ALK and ROS1 rearrangements, and MET exon 14 skipping mutation were observed in 38.41% (n = 212) and 2.72% (n = 15) patients, 12.14% (n = 67) and 3.62% (n = 20) patients, and 1.09% (n = 6) patients, respectively. EGFR exon 19 deletion accounted for 52.83% of all mutations, followed by L858R (35.85%), T790M (5.19%), exon 20 insertions (6.6%), and other rare mutations (G719X, L861Q, S768I) (9.91%). Concurrent EGFR with ALK, EGFR with ROS1, EGFR with MET, and EGFR with BRAF were observed in 10, 4, 1, and 3 patients, respectively. PD-L1 was expressed in 134 patients (53.2%). Exon 19 deletion was more prevalent in PD-L1 negative tumors whereas exon 21 substitution (L858R) was seen more in PD-L1 positive tumors. CONCLUSIONS: This is one of the largest cohorts of NSCLC for comprehensive targeted mutational profiling and correlation with the PD-L1 expression. The mutations are more prevalent in non-smoker females for all genes, except ALK (non-smoker males). MET and BRAF mutations are more common in elderly population whereas EGFR mutations, and ALK and ROS1 genes rearrangements are more prevalent in younger population. The most common histopathologic subtype/feature associated with various mutations was as follows: acinar with EGFR, solid with ALK, macronucleoli with ROS1, signet ring with MET, and micropapillary with BRAF.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma , Lung Neoplasms , Male , Female , Humans , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins B-raf/genetics , B7-H1 Antigen/genetics , Reproducibility of Results , Mutation , Proto-Oncogene Proteins/genetics , Protein Kinase Inhibitors
3.
Pathol Res Pract ; 220: 153394, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33706124

ABSTRACT

OBJECTIVES: Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene of non-small cell lung carcinomas (NSCLC) can be targeted by the tyrosine kinase inhibitors. A number of molecular diagnostic platforms are used to detect actionable targets in the exon(s) 18, 19, 20, and 21 of the EGFR gene. The Idylla™ system (Biocartis, Mechelen, Belgium) is a relatively novel technique and is unique in integrating both sample processing and real-time polymerase chain reaction (RT-PCR) in a single cartridge. We sought to conduct this study to compare the turnaround time (TAT) and concordance of Idylla™ system with the conventional RT-PCR and next-generation sequencing (NGS) for EGFR mutation detection. METHODS: In this retrospective analysis, 38 formalin-fixed, paraffin-embedded NSCLC tissue blocks with known NGS results by Ion Torrent™ S5 NGS platform were retested by the RT-PCR and Idylla™ platforms. RESULTS: A total of 15 of 38 (39.4 %) tumors that showed various EGFR mutations by NGS and conventional RT-PCR techniques were subjected to the Idylla™testing. These cases satisfied the specimen adequacy criteria of at least 10 % tumor cells for the testing. The mutations detected by the NGS were also detected by the Idylla™ testing. However, NGS identified additional 3 mutations in 3 cases, involving T709 V (exon 18, n = 1) and V774 M (exon 20, n = 2). The tumors with wild type EGFR on NGS did not have any actionable mutation detected by the Idylla™. Average EGFR testing TAT by Idylla™ was only 7.2 h (4-12 hours), as compared to conventional RT-PCR taking 54 h (31-79 hours) and NGS requiring 10.7 days (7.1-14 days). The actual procedure time by conventional RT-PCR was 24 h, NGS was 6.5 days, and Idylla™ was only 3 h. CONCLUSIONS: In summary, the Idylla™EGFR testing is an efficient, rapid, and fairly simple tool that can be used in the routine molecular laboratory with limited expertise and infrastructure and using the lowest amount of tissue material.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , DNA Mutational Analysis , High-Throughput Nucleotide Sequencing , Lung Neoplasms/genetics , Mutation , Real-Time Polymerase Chain Reaction , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Exons , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Time Factors , Workflow
4.
Pathol Res Pract ; 213(7): 868-871, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28554758

ABSTRACT

Dura-based intracranial neoplasms include a wide range of primary and metastatic tumors, varying in their clinical, radiologic, morphologic, and immunophenotypic characteristics. At this anatomic location, sarcomas are rare, however, they exhibit close morphologic resemblances to meningioma. Herein we describe the third case of primary synovial sarcoma of the parafalcine region in a50-years-old female, who presented with left-sided hemiplegia. The radiologic survey revealed a 5.5cm×5.8cm contrast enhancing dura-based mass at the right parafalcine region with meningeal enhancement and edema in the surrounding areas. Morphologic evaluation exhibited a high-grade spindle cell neoplasm, with focal hemangiopericytomatous pattern. The tumor cells were diffusely immunoreactive for CD99, Bcl2, TLE-1, and vimentin. The Ki-67 proliferation index was 40%. Pancytokeratin was focally positive. Epithelial membrane antigen, progesterone receptor, CD34, S-100, and glial fibrillary acidic protein were negative. Fluorescence in situ hybridization confirmed tumor specific translocation t(X;18)(p11.2;q11.2). Hence, final diagnosis of synovial sarcoma was rendered. Primary meningeal synovial sarcoma should be considered in the differential of aggressive and high-grade dura-based tumors in view of their relative chemosensitivity and future prospect of a molecular target-based therapy. The index case highlights the importance of an extensive pathologic analysis of high-grade mesenchymal lesions of the meninges to arrive at a definitive diagnosis and differentiate such tumors from other usual dura-based tumors, which has important therapeutic and prognostic implications.


Subject(s)
Brain Neoplasms/pathology , Dura Mater/pathology , Sarcoma, Synovial/pathology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Brain Edema/etiology , Brain Neoplasms/chemistry , Brain Neoplasms/complications , Brain Neoplasms/genetics , Cell Proliferation , Chromosomes, Human, Pair 18 , Chromosomes, Human, X , Diagnosis, Differential , Dura Mater/chemistry , Female , Hemiplegia/etiology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Middle Aged , Neoplasm Grading , Predictive Value of Tests , Sarcoma, Synovial/chemistry , Sarcoma, Synovial/complications , Sarcoma, Synovial/genetics , Translocation, Genetic
5.
Pathol Res Pract ; 213(1): 72-74, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27894622

ABSTRACT

The Philadelphia (Ph) chromosome is infrequently found in acute lymphoblastic leukemia and is associated with poor prognosis. We present a case of Ph chromosome positive B cell-acute lymphoblastic leukemia with the partial trisomy of chromosome 1q involving chromosome 13 as the acceptor which has never been reported in the English literature. Jumping translocation (JT) of chromosome 1 is rare and is associated with disease progression and poor prognosis. Herein, we report the first case of Ph chromosome positive B cell-acute lymphoblastic leukemia with coexisting jumping translocation of chromosome 1 leading to trisomy of chromosome 1q. Dismal prognosis associated with synchronous presence of a Ph chromosome and JT leading to a partial trisomy of chromosome 1q may carry significant prognostic and therapeutic implications. This may be an incidental finding and further studies with large patient cohorts and clinical outcomes are needed to definitively determine the predictive value of this cytogenetic finding.


Subject(s)
Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 1 , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic , Trisomy , Adult , Cytogenetics , Female , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
6.
Biol Chem ; 391(9): 1031-40, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20536385

ABSTRACT

Mutations in the tumor suppressor gene p53 are frequent in human glioblastomas. Similarly cathepsin L, a lysosomal cysteine protease, is overexpressed and secreted by most human tumors including glioblastomas. However, hitherto there is no information on whether or not the mutation(s) in the p53 gene affect(s) expression of this protease. Using human glioblastoma cell lines harboring wild type and mutant p53, we demonstrate here for the first time that only the wild type but not the mutant p53 upregulates cathepsin L expression. By transfection of promoter reporter constructs, site-directed mutagenesis and chip assays we have established that wild type p53 elevates the levels of cathepsin L in these cells. It does so directly by binding to the cathepsin L promoter and also indirectly by inducing the expression of C/EBPα, which is crucial for the transcription of this protease. In view of its role in tumorigenesis, angiogenesis and tumor cell invasion, increased expression of cathepsin L in glioblastoma cells harboring wild type p53 might confer invasive ability and growth advantage to these cells. Therefore, use of cathepsin L inhibitors could prove useful in the management of these tumors.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cathepsin L/genetics , Glioblastoma/genetics , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/genetics , Glioblastoma/pathology , Humans , Reverse Transcriptase Polymerase Chain Reaction
7.
Mol Cell Biochem ; 322(1-2): 119-25, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19009237

ABSTRACT

In this study, we demonstrate that pGEM-4Z can be used as a mammalian expression vector. Western blotting and Immunocytochemical analyses revealed that transfection of pGEM-4Z-containing human cathepsin L cDNA under T-7 but not under SP-6 promoter into NIH 3T3 cells resulted in a high-level expression of cathepsin L. Expression of proteins using this vector in mammalian cells was further confirmed by using luciferase reporter gene. Furthermore, NIH 3T3 cells after stable or transient transfection with pGEM-4Z containing the first exon, first intron, and rest of the human cathepsin L cDNA downstream to its T-7 promoter synthesized and secreted large quantities of cathepsin L. RNase protection assays and 5' RACE established that the cloned cathepsin L cDNA is transcribed from a cryptic promoter present in the backbone of this vector upstream to T-7 sequence. This promoter was active in cell lines derived from four different mammalian species. In NIH 3T3 cells, this cryptic promoter could transcribe structural part of the genomic DNA into a primary transcript, which was efficiently spliced into mature mRNA and translated into protein. Thus this vector is equally useful for expressing proteins from genomic DNA. This hitherto unknown property of pGEM-4Z may be useful for expression of proteins in mammalian cells besides its use in synthesis of riboprobes, DNA sequencing, and in vitro transcription coupled translation assays.


Subject(s)
DNA, Complementary/chemistry , Genetic Vectors/genetics , Promoter Regions, Genetic , Animals , Base Sequence , CHO Cells , Cathepsin L , Cathepsins/genetics , Cathepsins/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , Cricetulus , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , DNA, Complementary/metabolism , Genetic Vectors/metabolism , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , RNA, Messenger/metabolism , Transcription Initiation Site , Transfection , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL