Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Clin Pathol ; 159(1): 69-80, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36317501

RESUMO

OBJECTIVES: With a substantial number of patients with multiple myeloma (MM) experiencing disease relapse, the quest for more sensitive methods to assess deeper responses indicative of cure continues. METHODS: In this prospective analysis of 170 patients with MM at day 100 after autologous stem cell transplant, we evaluated the predictive value of conventional response, measurable residual disease (MRDTOTAL: the aberrant percentage of plasma cells [PC%] among total bone marrow cells), and neoplastic plasma cell index scores (NPCI: the aberrant PC% of total PCs). RESULTS: Significantly better progression-free survival (PFS) and overall survival (OS) were observed with deepening conventional response. Conventional response-based stratification within the MRD-positive and MRD-negative subgroups showed a significantly higher PFS (hazard ratio [HR], 3.11; P < .005) and OS (HR, 3.08; P = .01) in the conventional response-positive/MRD-positive group compared with the conventional response-negative/MRD-positive group. Using K-adaptive partitioning to find the optimum threshold for MRD, patients achieving less than 0.001% MRDTOTAL had superior PFS (MRDTOTAL 0.001% to <0.1%: HR, 6.66, P < .005; MRDTOTAL ≥0.1%: HR, 11.52, P < .005) and OS (MRDTOTAL 0.001% to <0.1%: HR, 5.3, P < .05; MRDTOTAL ≥0.1%: HR = 9.21, P < .005). The C index and Akaike information criterion metrics demonstrated the superior performance of the NPCI compared with MRDTOTAL in predicting treatment outcome. CONCLUSIONS: Progressive deepening of response, conventional as well as MRD, correlates with superior survival outcomes. The NPCI proved to be a superior determinant of survival and can be explored as a better statistic than MRD.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/terapia , Plasmócitos , Citometria de Fluxo/métodos , Recidiva Local de Neoplasia , Transplante de Células-Tronco/métodos , Resultado do Tratamento , Neoplasia Residual , Transplante de Células-Tronco Hematopoéticas/métodos
2.
Comput Biol Med ; 149: 106048, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36113255

RESUMO

In this study, we present an efficient Graph Convolutional Network based Risk Stratification system (GCRS) for cancer risk-stage prediction of newly diagnosed multiple myeloma (NDMM) patients. GCRS is a hybrid graph convolutional network consisting of a fusion of multiple connectivity graphs that are used to learn the latent representation of topological structures among patients. This proposed risk stratification system integrates these connectivity graphs prepared from the clinical and laboratory characteristics of NDMM cancer patients for partitioning them into three cancer risk groups: low, intermediate, and high. Extensive experiments demonstrate that GCRS outperforms the existing state-of-the-art methods in terms of C-index and hazard ratio on two publicly available datasets of NDMM patients. We have statistically validated our results using the Cox Proportional-Hazards model, Kaplan-Meier analysis, and log-rank test on progression-free survival (PFS) and overall survival (OS). We have also evaluated the contribution of various clinical parameters as utilized by the GCRS risk stratification system using the SHapley Additive exPlanations (SHAP) analysis, an interpretability algorithm for validating AI methods. Our study reveals the utility of the deep learning approach in building a robust system for cancer risk stage prediction.


Assuntos
Mieloma Múltiplo , Algoritmos , Humanos , Estadiamento de Neoplasias , Modelos de Riscos Proporcionais , Medição de Risco
3.
Transl Oncol ; 23: 101472, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35777247

RESUMO

Multiple myeloma (MM) is a heterogeneous plasma cell proliferative disorder that arises from its premalignant precursor stages through a complex cascade of interactions between clonal mutations and co-evolving microenvironment. The temporo-spatial evolutionary trajectories of MM are established early during myelomatogenesis in precursor stages and retained in MM. Such molecular events impact subsequent disease progression and clinical outcomes. Identification of clonal sweeps of actionable gene targets in MM could reveal potential vulnerabilities that may exist in early stages and thus potentiate prognostication and customization of early therapeutic interventions. We have evaluated clonal evolution at multiple time points in 76 MM patients enrolled in the MMRF CoMMpass study. The major findings of this study are (a) MM progresses predominantly through branching evolution, (b) there is a heterogeneous spectrum of mutational landscapes that include unique actionable gene targets at diagnosis compared to progression, (c) unique clonal gains/ losses of mutant driver genes can be identified in patients with different cytogenetic aberrations, (d) there is a significant correlation between co-occurring oncogenic mutations/ co-occurring subclones e.g., with mutated TP53+SYNE1, NRAS+MAGI3, and anticorrelative dependencies between FAT3+FCGBP gene pairs. Such co-trajectories may synchronize molecular events of drug response, myelomatogenesis and warrant future studies to explore their potential for early prognostication and development of risk stratified personalized therapies in MM.

4.
Am J Cancer Res ; 12(4): 1919-1933, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35530275

RESUMO

Mutational Signatures and Tumor mutational burden (TMB) have emerged as prognostic biomarkers in cancer genomics. However, the association of TMB with overall survival (OS) is still unknown in newly diagnosed multiple myeloma (NDMM) patients. Further, the change in the mutational spectrum involving both synonymous and non-synonymous mutations as MGUS progresses to MM is unexplored. This study addresses both these aspects via extensive evaluation of the mutations in MGUS and NDMM. WES data of 1018 NDMM patients and 61 MGUS patients collected from three different global regions were analyzed in this study. Single base substitutions, mutational signatures and TMB were inferred from the variants identified in MGUS and MM patients. The cutoff value for TMB was estimated to divide patients into low TMB and high TMB (hypermutators) groups. This study finds a change in the mutational spectrum with a statistically significant increase from MGUS to MM. There was a statistically significant increase in the frequency of all the three categories of variants, non-synonymous (NS), synonymous (SYN), and others (OTH), from MGUS to MM (P<0.05). However, there was a statistically significant rise in the TMB values for TMB_NS and TMB_SYN only. We also observed that 3' and 5'UTR mutations were more frequent in MM and might be responsible for driving MGUS to MM via regulatory binding sites. NDMM patients were also examined separately along with their survival outcomes. The frequency of hypermutators was low in MM with poor OS and PFS outcome. We observed a statistically significant rise in the frequency of C>A and C>T substitutions and a statistically significant decline in T>G substitutions in the MM patients with poor outcomes. Additionally, there was a statistically significant increase in the TMB of the patients with poor outcome compared to patients with a superior outcome. A statistically significant association between the APOBEC activity and poor overall survival in MM was discovered. These findings have potential clinical relevance and can assist in designing risk-adapted therapies to inhibit the progression of MGUS to MM and prolong the overall survival in high-risk MM patients.

5.
J Biomed Inform ; 129: 104055, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35337943

RESUMO

Tumor heterogeneity, marked by the presence of divergent clonal subpopulations of tumor cells, impedes the treatment response in cancer patients. Single-cell sequencing technology provides substantial prospects to gain an in-depth understanding of the cellular phenotypic variability driving tumor progression. A comprehensive insight into the intra-tumor heterogeneity may further assist in dealing with the treatment-resistant clones in cancer patients, thereby improving their overall survival. However, this task is hampered due to the challenges associated with the single-cell data, such as false positives, false negatives and missing bases, and the increase in their size. As a result, the computational cost of the existing methods increases, thereby limiting their usage. In this work, we propose a robust graph learning-based method, ARCANE-ROG (Algorithm for Reconstruction of CANcer Evolution via RObust Graph learning), for inferring clonal evolution from single-cell datasets. The first step of the proposed method is a joint framework of denoising with data imputation for the noisy and incomplete matrix while simultaneously learning an adjacency graph. Both the operations in the joint framework boost each other such that the overall performance of the denoising algorithm is improved. In the second step, an optimal number of clusters are identified via the Leiden method. In the last step, clonal evolution trees are inferred via a minimum spanning tree algorithm. The method has been benchmarked against a state-of-the-art method, RobustClone, using simulated datasets of varying sizes and five real datasets. The performance of our proposed method is found to be significantly superior (p-value < 0.05) in terms of reconstruction error, False Positive to False Negative (FPFN) ratio, tree distance error and V-measure compared to the other method. Overall, the proposed method is an improvement over the existing methods as it enhances cluster assignment and inference on clonal hierarchies.


Assuntos
Evolução Clonal , Neoplasias , Algoritmos , Humanos , Neoplasias/genética
6.
Front Oncol ; 11: 720932, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858811

RESUMO

INTRODUCTION: Current risk predictors of multiple myeloma do not integrate ethnicity-specific information. However, the impact of ethnicity on disease biology cannot be overlooked. In this study, we have investigated the impact of ethnicity in multiple myeloma risk prediction. In addition, an efficient and robust artificial intelligence (AI)-enabled risk-stratification system is developed for newly diagnosed multiple myeloma (NDMM) patients that utilizes ethnicity-specific cutoffs of key prognostic parameters. METHODS: K-adaptive partitioning is used to propose new cutoffs of parameters for two different datasets-the MMIn (MM Indian dataset) dataset and the MMRF (Multiple Myeloma Research Foundation) dataset belonging to two different ethnicities. The Consensus-based Risk-Stratification System (CRSS) is designed using the Gaussian mixture model (GMM) and agglomerative clustering. CRSS is validated via Cox hazard proportional methods, Kaplan-Meier analysis, and log-rank tests on progression-free survival (PFS) and overall survival (OS). SHAP (SHapley Additive exPlanations) is utilized to establish the biological relevance of the risk prediction by CRSS. RESULTS: There is a significant variation in the key prognostic parameters of the two datasets belonging to two different ethnicities. CRSS demonstrates superior performance as compared with the R-ISS in terms of C-index and hazard ratios on both the MMIn and MMRF datasets. An online calculator has been built that can predict the risk stage of a multiple myeloma (MM) patient based on the values of parameters and ethnicity. CONCLUSION: Our methodology discovers changes in the cutoffs with ethnicities from the established cutoffs of prognostic features. The best predictor model for both cohorts was obtained with the new ethnicity-specific cutoffs of clinical parameters. Our study also revealed the efficacy of AI in building a deployable risk prediction system for MM. In the future, it is suggested to use the CRSS risk calculator on a large dataset as the cohort size of the present study is 25% of the cohort used in the R-ISS reported in 2015.

7.
Am J Cancer Res ; 11(11): 5659-5679, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34873486

RESUMO

Multiple Myeloma (MM) arises from malignant transformation and deregulated proliferation of clonal plasma cells (PCs) harbouring heterogeneous molecular anomalies. The effect of evolving mutations on clone fitness and their cellular prevalence shapes the progressing myeloma genome and impacts clinical outcomes. Although clonal heterogeneity in MM is well established, which subclonal mutations emerge/persist/perish with progression in MM and which of these can be targeted therapeutically remains an open question. In line with this, we have sequenced pairwise whole exomes of 62 MM patients collected at two time points, i.e., at diagnosis and on progression. Somatic variants were called using a novel ensemble approach where a consensus was deduced from four variant callers (Illumina's Dragen, Strelka2, SomaticSniper and SpeedSeq) and actionable/druggable gene targets were identified. A marked intraclonal heterogeneity was observed. Branching evolution was observed among 72.58% patients, of whom 64.51% had low TMBs (<10) and 61.29% had 2 or more founder clones. The hypermutator patients (with high TMB levels ≥10 to ≤100) showed a significant decrease in their TMBs from diagnosis (median TMB 77.11) to progression (median TMB 31.22). A distinct temporal fall in subclonal driver mutations was identified recurrently across diagnosis to progression e.g., in PABPC1, BRAF, KRAS, CR1, DIS3 and ATM genes in 3 or more patients suggesting such patients could be treated early with target specific drugs like Vemurafenib/Cobimetinib. An analogous rise in driver mutations was observed in KMT2C, FOXD4L1, SP140, NRAS and other genes. A few drivers such as FAT4, IGLL5 and CDKN1A retained consistent distribution patterns at two time points. These findings are clinically relevant and point at consideration of evaluating multi time point subclonal mutational landscapes for designing better risk stratification strategies and tailoring time to time risk adapted combination therapies in future.

8.
Transl Oncol ; 14(9): 101157, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34247136

RESUMO

INTRODUCTION: An efficient readily employable risk prognostication method is desirable for MM in settings where genomics tests cannot be performed owing to geographical/economical constraints. In this work, a new Modified Risk Staging (MRS) has been proposed for newly diagnosed Multiple Myeloma (NDMM) that exploits six easy-to-acquire clinical parameters i.e. age, albumin, ß2-microglobulin (ß2M), calcium, estimated glomerular filtration rate (eGFR) and hemoglobin. MATERIALS AND METHODS: MRS was designed using a training cohort of 716 NDMM patients of our inhouse MM Indian (MMIn) cohort and validated on MMIn (n=354) cohort and MMRF (n=900) cohort. K-adaptive partitioning (KAP) was used to find new thresholds for the parameters. Risk staging rules, obtained via training a J48 classifier, were used to build MRS. RESULTS: New thresholds were identified for albumin (3.6 g/dL), ß2M (4.8 mg/L), calcium (11.13 mg/dL), eGFR (48.1 mL/min), and hemoglobin (12.3 g/dL) using KAP on the MMIn dataset. On the MMIn dataset, MRS outperformed ISS for OS prediction in terms of C-index, hazard ratios, and its corresponding p-values, but performs comparable in prediction of PFS. On both MMIn and MMRF datasets, MRS performed better than RISS in terms of C-index and p-values. A simple online tool was also designed to allow automated calculation of MRS based on the values of the parameters. DISCUSSION: Our proposed ML-derived yet simple staging system, MRS, although does not employ genetic features, outperforms RISS as confirmed by better separability in KM survival curves and higher values of C-index on both MMIn and MMRF datasets. FUNDING: Grant: BT/MED/30/SP11006/2015 (Department of Biotechnology, Govt. of India), Grant: DST/ICPS/CPS-Individual/2018/279(G) (Department of Science and Technology, Govt. of India), UGC-Senior Research Fellowship.

9.
Front Oncol ; 9: 1442, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31970084

RESUMO

Purpose: Gene expression data generated from microarray technology is often analyzed for disease diagnostics and treatment. However, this data suffers with missing values that may lead to inaccurate findings. Since data capture is expensive, time consuming, and is required to be collected from subjects, it is worthwhile to recover missing values instead of re-collecting the data. In this paper, a novel but simple method, namely, DSNN (Doubly Sparse DCT domain with Nuclear Norm minimization) has been proposed for imputing missing values in microarray data. Extensive experiments including pathway enrichment have been carried out on four blood cancer dataset to validate the method as well as to establish the significance of imputation. Methods: A new method, namely, DSNN, was proposed for missing value imputation on gene expression data. Method was validated on four dataset, CLL, AML, MM (Spanish data), and MM (Indian data). All the dataset were downloaded from GEO repository. Missing values were introduced in the original data from 10 to 90% in steps of 10% because method validation requires ground truth. Quantitative results on normalized mean square error (NMSE) between the ground truth and imputed data were computed. To further validate and establish the significance of the proposed imputation method, two experiments were carried out on the data imputed with the proposed method, data imputed with the state-of-art methods, and data with missing values. In the first experiment, classification of normal vs. cancer subjects was carried out. In the second experiment, biological significance of imputation was ascertained by identifying top candidate tumor drivers using the existing state-of-the-art SPARROW algorithm, followed by gene list enrichment analysis on top candidate drivers. Results: Quantitative NMSE results of the DSNN method were compared with three state-of-the-art imputation methods. DSNN method was observed to perform better compared to these other methods both at high as well as low observable data. Experiment-1 demonstrated superior results on classification with imputation compared to that performed on missing data matrix as well as compared to classification on imputed data with existing methods. In experiment-2, cancer affected pathways were discovered with higher significance in the data imputed with the proposed method compared to those discovered with the missing data matrix. Conclusion: Missing value problem in microarray data is a serious problem and can adversely influence downstream analysis. A novel method, namely, DSNN is proposed for missing value imputation. The method is validated quantitatively on the application of classification and biologically by performing pathway enrichment analysis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA