Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ludmila Danilova is active.

Publication


Featured researches published by Ludmila Danilova.


Science | 2017

Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade

Dung T. Le; Jennifer N. Durham; Kellie Nicole Smith; Hao Wang; Bjarne Bartlett; Laveet K. Aulakh; Steve Lu; Holly Kemberling; Cara Wilt; Brandon Luber; Fay Wong; Nilofer Saba Azad; Agnieszka A. Rucki; Daniel A. Laheru; Ross C. Donehower; Atif Zaheer; George A. Fisher; Todd S. Crocenzi; James J. Lee; Tim F. Greten; Austin Duffy; Kristen K. Ciombor; Aleksandra Eyring; Bao H. Lam; Andrew K. Joe; S. Peter Kang; Matthias Holdhoff; Ludmila Danilova; Leslie Cope; Christian Meyer

Predicting responses to immunotherapy Colon cancers with loss-of-function mutations in the mismatch repair (MMR) pathway have favorable responses to PD-1 blockade immunotherapy. In a phase 2 clinical trial, Le et al. showed that treatment success is not just limited to colon cancer (see the Perspective by Goswami and Sharma). They found that a wide range of different cancer types with MMR deficiency also responded to PD-1 blockade. The trial included some patients with pancreatic cancer, which is one of the deadliest forms of cancer. The clinical trial is still ongoing, and around 20% of patients have so far achieved a complete response. MMR deficiency appears to be a biomarker for predicting successful treatment outcomes for several solid tumors and indicates a new therapeutic option for patients harboring MMR-deficient cancers. Science, this issue p. 409; see also p. 358 A pan-cancer biomarker is identified that can predict successful response to cancer immunotherapy in human patients. The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor–1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair–deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair–deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers’ tissue of origin.


Science | 2018

Detection and localization of surgically resectable cancers with a multi-analyte blood test

Joshua D. Cohen; Lu Li; Yuxuan Wang; Christopher J. Thoburn; Bahman Afsari; Ludmila Danilova; Christopher Douville; Ammar A. Javed; Fay Wong; Austin Mattox; Ralph H. Hruban; Christopher L. Wolfgang; Michael Goggins; Marco Dal Molin; Tian Li Wang; Richard Roden; Alison P. Klein; Janine Ptak; Lisa Dobbyn; Joy Schaefer; Natalie Silliman; Maria Popoli; Joshua T. Vogelstein; James Browne; Robert E. Schoen; Randall E. Brand; Jeanne Tie; Peter Gibbs; Hui-Li Wong; Aaron S. Mansfield

SEEK and you may find cancer earlier Many cancers can be cured by surgery and/or systemic therapies when detected before they have metastasized. This clinical reality, coupled with the growing appreciation that cancers rapid genetic evolution limits its response to drugs, have fueled interest in methodologies for earlier detection of the disease. Cohen et al. developed a noninvasive blood test, called CancerSEEK that can detect eight common human cancer types (see the Perspective by Kalinich and Haber). The test assesses eight circulating protein biomarkers and tumor-specific mutations in circulating DNA. In a study of 1000 patients previously diagnosed with cancer and 850 healthy control individuals, CancerSEEK detected cancer with a sensitivity of 69 to 98% (depending on cancer type) and 99% specificity. Science, this issue p. 926; see also p. 866 A blood test that combines protein and DNA markers may allow earlier detection of eight common cancer types. Earlier detection is key to reducing cancer deaths. Here, we describe a blood test that can detect eight common cancer types through assessment of the levels of circulating proteins and mutations in cell-free DNA. We applied this test, called CancerSEEK, to 1005 patients with nonmetastatic, clinically detected cancers of the ovary, liver, stomach, pancreas, esophagus, colorectum, lung, or breast. CancerSEEK tests were positive in a median of 70% of the eight cancer types. The sensitivities ranged from 69 to 98% for the detection of five cancer types (ovary, liver, stomach, pancreas, and esophagus) for which there are no screening tests available for average-risk individuals. The specificity of CancerSEEK was greater than 99%: only 7 of 812 healthy controls scored positive. In addition, CancerSEEK localized the cancer to a small number of anatomic sites in a median of 83% of the patients.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Characterization of HPV and host genome interactions in primary head and neck cancers

Michael Parfenov; Chandra Sekhar Pedamallu; Nils Gehlenborg; Samuel S. Freeman; Ludmila Danilova; Christopher A. Bristow; Semin Lee; Angela Hadjipanayis; Elena Ivanova; Matthew D. Wilkerson; Alexei Protopopov; Lixing Yang; Sahil Seth; Xingzhi Song; Jiabin Tang; Xiaojia Ren; Jianhua Zhang; Angeliki Pantazi; Netty Santoso; Andrew W. Xu; Harshad S. Mahadeshwar; David A. Wheeler; Robert I. Haddad; Joonil Jung; Akinyemi I. Ojesina; Natalia Issaeva; Wendell G. Yarbrough; D. Neil Hayes; Jennifer R. Grandism; Adel K. El-Naggar

Significance A significant proportion of head and neck cancer is driven by human papillomavirus (HPV) infection, and the expression of viral oncogenes is involved in the development of these tumors. However, the role of HPV integration in primary tumors beyond increasing the expression of viral oncoproteins is not understood. Here, we describe how HPV integration impacts the host genome by amplification of oncogenes and disruption of tumor suppressors as well as driving inter- and intrachromosomal rearrangements. Tumors that do and do not have HPV integrants display distinct gene expression profiles and DNA methylation patterns, which further support the view that the mechanisms by which tumors with integrated and nonintegrated HPV arise are distinct. Previous studies have established that a subset of head and neck tumors contains human papillomavirus (HPV) sequences and that HPV-driven head and neck cancers display distinct biological and clinical features. HPV is known to drive cancer by the actions of the E6 and E7 oncoproteins, but the molecular architecture of HPV infection and its interaction with the host genome in head and neck cancers have not been comprehensively described. We profiled a cohort of 279 head and neck cancers with next generation RNA and DNA sequencing and show that 35 (12.5%) tumors displayed evidence of high-risk HPV types 16, 33, or 35. Twenty-five cases had integration of the viral genome into one or more locations in the human genome with statistical enrichment for genic regions. Integrations had a marked impact on the human genome and were associated with alterations in DNA copy number, mRNA transcript abundance and splicing, and both inter- and intrachromosomal rearrangements. Many of these events involved genes with documented roles in cancer. Cancers with integrated vs. nonintegrated HPV displayed different patterns of DNA methylation and both human and viral gene expressions. Together, these data provide insight into the mechanisms by which HPV interacts with the human genome beyond expression of viral oncoproteins and suggest that specific integration events are an integral component of viral oncogenesis.


Cancer Cell | 2016

Comprehensive Pan-Genomic Characterization of Adrenocortical Carcinoma

Siyuan Zheng; Andrew D. Cherniack; Ninad Dewal; Richard A. Moffitt; Ludmila Danilova; Bradley A. Murray; Antonio M. Lerario; Tobias Else; Theo Knijnenburg; Giovanni Ciriello; Seungchan Kim; Guillaume Assié; Olena Morozova; Rehan Akbani; Juliann Shih; Katherine A. Hoadley; Toni K. Choueiri; Jens Waldmann; Ozgur Mete; Robertson Ag; Hsin-Ta Wu; Benjamin J. Raphael; Shao L; Matthew Meyerson; Michael J. Demeure; Felix Beuschlein; Anthony J. Gill; Stan B. Sidhu; Madson Q. Almeida; Maria Candida Barisson Villares Fragoso

We describe a comprehensive genomic characterization of adrenocortical carcinoma (ACC). Using this dataset, we expand the catalogue of known ACC driver genes to include PRKAR1A, RPL22, TERF2, CCNE1, and NF1. Genome wide DNA copy-number analysis revealed frequent occurrence of massive DNA loss followed by whole-genome doubling (WGD), which was associated with aggressive clinical course, suggesting WGD is a hallmark of disease progression. Corroborating this hypothesis were increased TERT expression, decreased telomere length, and activation of cell-cycle programs. Integrated subtype analysis identified three ACC subtypes with distinct clinical outcome and molecular alterations which could be captured by a 68-CpG probe DNA-methylation signature, proposing a strategy for clinical stratification of patients based on molecular markers.


Nature Methods | 2016

Inferring causal molecular networks: empirical assessment through a community-based effort

Steven M. Hill; Laura M. Heiser; Thomas Cokelaer; Michael Unger; Nicole K. Nesser; Daniel E. Carlin; Yang Zhang; Artem Sokolov; Evan O. Paull; Christopher K. Wong; Kiley Graim; Adrian Bivol; Haizhou Wang; Fan Zhu; Bahman Afsari; Ludmila Danilova; Alexander V. Favorov; Wai Shing Lee; Dane Taylor; Chenyue W. Hu; Byron L. Long; David P. Noren; Alexander J Bisberg; Gordon B. Mills; Joe W. Gray; Michael R. Kellen; Thea Norman; Stephen H. Friend; Amina A. Qutub; Elana J. Fertig

It remains unclear whether causal, rather than merely correlational, relationships in molecular networks can be inferred in complex biological settings. Here we describe the HPN-DREAM network inference challenge, which focused on learning causal influences in signaling networks. We used phosphoprotein data from cancer cell lines as well as in silico data from a nonlinear dynamical model. Using the phosphoprotein data, we scored more than 2,000 networks submitted by challenge participants. The networks spanned 32 biological contexts and were scored in terms of causal validity with respect to unseen interventional data. A number of approaches were effective, and incorporating known biology was generally advantageous. Additional sub-challenges considered time-course prediction and visualization. Our results suggest that learning causal relationships may be feasible in complex settings such as disease states. Furthermore, our scoring approach provides a practical way to empirically assess inferred molecular networks in a causal sense.


Journal of Bioinformatics and Computational Biology | 2006

RNAKinetics: a web server that models secondary structure kinetics of an elongating RNA.

Ludmila Danilova; Dmitri D. Pervouchine; Alexander V. Favorov; Andrei A. Mironov

The RNAKinetics server (http://www.ig-msk.ru/RNA/kinetics) is a web interface for the newly developed RNAKinetics software. The software models the dynamics of RNA secondary structure by the means of kinetic analysis of folding transitions of a growing RNA molecule. The result of the modeling is a kinetic ensemble, i.e. a collection of RNA structures that are endowed with probabilities, which depend on time. This approach gives comprehensive probabilistic description of RNA folding pathways, revealing important kinetic details that are not captured by the traditional structure prediction methods. The access to the RNAKinetics server is free.


Genome Biology | 2016

Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures.

Yasin Şenbabaoğlu; Ron S. Gejman; Andrew G. Winer; Ming Liu; Eliezer M. Van Allen; Guillermo Velasco; Diana Miao; Irina Ostrovnaya; Esther Drill; Augustin Luna; Nils Weinhold; William R. Lee; Brandon J. Manley; Danny N. Khalil; Samuel D. Kaffenberger; Ying-Bei Chen; Ludmila Danilova; Martin H. Voss; Jonathan A. Coleman; Paul Russo; Victor E. Reuter; Timothy A. Chan; Emily H. Cheng; David A. Scheinberg; Ming O. Li; Toni K. Choueiri; James J. Hsieh; Chris Sander; A. Ari Hakimi

BackgroundTumor-infiltrating immune cells have been linked to prognosis and response to immunotherapy; however, the levels of distinct immune cell subsets and the signals that draw them into a tumor, such as the expression of antigen presenting machinery genes, remain poorly characterized. Here, we employ a gene expression-based computational method to profile the infiltration levels of 24 immune cell populations in 19 cancer types.ResultsWe compare cancer types using an immune infiltration score and a T cell infiltration score and find that clear cell renal cell carcinoma (ccRCC) is among the highest for both scores. Using immune infiltration profiles as well as transcriptomic and proteomic datasets, we characterize three groups of ccRCC tumors: T cell enriched, heterogeneously infiltrated, and non-infiltrated. We observe that the immunogenicity of ccRCC tumors cannot be explained by mutation load or neo-antigen load, but is highly correlated with MHC class I antigen presenting machinery expression (APM). We explore the prognostic value of distinct T cell subsets and show in two cohorts that Th17 cells and CD8+ T/Treg ratio are associated with improved survival, whereas Th2 cells and Tregs are associated with negative outcomes. Investigation of the association of immune infiltration patterns with the subclonal architecture of tumors shows that both APM and T cell levels are negatively associated with subclone number.ConclusionsOur analysis sheds light on the immune infiltration patterns of 19 human cancers and unravels mRNA signatures with prognostic utility and immunotherapeutic biomarker potential in ccRCC.Tumor-infiltrating immune cells have been linked to prognosis and response to immunotherapy; however, the levels of distinct immune cell subsets and the signals that draw them into a tumor, such as the expression of antigen presenting machinery genes, remain poorly characterized. Here, we employ a gene expression-based computational method to profile the infiltration levels of 24 immune cell populations in 19 cancer types. We compare cancer types using an immune infiltration score and a T cell infiltration score and find that clear cell renal cell carcinoma (ccRCC) is among the highest for both scores. Using immune infiltration profiles as well as transcriptomic and proteomic datasets, we characterize three groups of ccRCC tumors: T cell enriched, heterogeneously infiltrated, and non-infiltrated. We observe that the immunogenicity of ccRCC tumors cannot be explained by mutation load or neo-antigen load, but is highly correlated with MHC class I antigen presenting machinery expression (APM). We explore the prognostic value of distinct T cell subsets and show in two cohorts that Th17 cells and CD8+ T/Treg ratio are associated with improved survival, whereas Th2 cells and Tregs are associated with negative outcomes. Investigation of the association of immune infiltration patterns with the subclonal architecture of tumors shows that both APM and T cell levels are negatively associated with subclone number. Our analysis sheds light on the immune infiltration patterns of 19 human cancers and unravels mRNA signatures with prognostic utility and immunotherapeutic biomarker potential in ccRCC.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Association of PD-1/PD-L axis expression with cytolytic activity, mutational load, and prognosis in melanoma and other solid tumors

Ludmila Danilova; Hao Wang; Joel C. Sunshine; Genevieve J. Kaunitz; Tricia R. Cottrell; Haiying Xu; Jessica Esandrio; Robert A. Anders; Leslie Cope; Drew M. Pardoll; Charles G. Drake; Janis M. Taube

Significance The Cancer Genome Atlas datasets were used in the current study to explore the relationship of programmed death ligand-1 (PD-L1) expression, a cytotoxic T-cell gene signature, and mutational load to each other, to immunoactive factors, such as programmed cell death protein-1 (PD-1), PD-L2, and other checkpoint molecules, and to survival across multiple solid tumor types. We found that PD-L2 expression is more closely related to an ongoing host immune response in certain tumor types than PD-L1. Notably, mutational load was not immediately related to inflammation in any tumor type studied, and was inferior to an inflamed tumor microenvironment for predicting survival in patients with metastatic melanoma. Our findings also indicate the need for biomarker assays that are tumor-type–specific and include both expression studies and genomic profiling. Programmed cell death protein-1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint blockade has led to remarkable and durable objective responses in a number of different tumor types. A better understanding of factors associated with the PD-1/PD-L axis expression is desirable, as it informs their potential role as prognostic and predictive biomarkers and may suggest rational treatment combinations. In the current study, we analyzed PD-L1, PD-L2, PD-1, and cytolytic activity (CYT) expression, as well as mutational density from melanoma and eight other solid tumor types using The Cancer Genome Atlas database. We found that in some tumor types, PD-L2 expression is more closely linked to Th1/IFNG expression and PD-1 and CD8 signaling than PD-L1. In contrast, mutational load was not correlated with a Th1/IFNG gene signature in any tumor type. PD-L1, PD-L2, PD-1, CYT expression, and mutational density are all positive prognostic features in melanoma, and conditional inference modeling revealed PD-1/CYT expression (i.e., an inflamed tumor microenvironment) as the most impactful feature, followed by mutational density. This study elucidates the highly interdependent nature of these parameters, and also indicates that future biomarkers for anti–PD-1/PD-L1 will benefit from tumor-type–specific, integrated, mRNA, protein, and genomic approaches.


Clinical Cancer Research | 2013

Frequent inactivation of cysteine dioxygenase type 1 contributes to survival of breast cancer cells and resistance to anthracyclines.

Jana Jeschke; Heather M. O'Hagan; Wei Zhang; Rajita Vatapalli; Marilia Freitas Calmon; Ludmila Danilova; Claudia Nelkenbrecher; Leander Van Neste; Ingrid T.G.W. Bijsmans; Manon van Engeland; Edward Gabrielson; Kornel E. Schuebel; Andreas Winterpacht; Stephen B. Baylin; James G. Herman; Nita Ahuja

Purpose: Genome-wide DNA methylation analyses have identified hundreds of candidate DNA-hypermethylated genes in cancer. Comprehensive functional analyses provide an understanding of the biologic significance of this vast amount of DNA methylation data that may allow the determination of key epigenetic events associated with tumorigenesis. Experimental Design: To study mechanisms of cysteine dioxygenase type 1 (CDO1) inactivation and its functional significance in breast cancer in a comprehensive manner, we screened for DNA methylation and gene mutations in primary breast cancers and analyzed growth, survival, and reactive oxygen species (ROS) production in breast cancer cells with restored CDO1 function in the context of anthracycline treatment. Results: DNA methylation-associated silencing of CDO1 in breast cancer is frequent (60%), cancer specific, and correlates with disease progression and outcome. CDO1 function can alternatively be silenced by repressive chromatin, and we describe protein-damaging missense mutations in 7% of tumors without DNA methylation. Restoration of CDO1 function in breast cancer cells increases levels of ROS and leads to reduced viability and growth, as well as sensitization to anthracycline treatment. Priming with 5-azacytidine of breast cancer cells with epigenetically silenced CDO1 resulted in restored expression and increased sensitivity to anthracyclines. Conclusion: We report that silencing of CDO1 is a critical epigenetic event that contributes to the survival of oxidative-stressed breast cancer cells through increased detoxification of ROS and thus leads to the resistance to ROS-generating chemotherapeutics including anthracyclines. Our study shows the importance of CDO1 inactivation in breast cancer and its clinical potential as a biomarker and therapeutic target to overcome resistance to anthracyclines. Clin Cancer Res; 19(12); 3201–11. ©2013 AACR.


PLOS ONE | 2014

Novel insight into mutational landscape of head and neck squamous cell carcinoma

Daria A. Gaykalova; Elizabeth Mambo; Ashish Choudhary; Jeffery Houghton; Kalyan Buddavarapu; Tiffany Sanford; Will Darden; Alex Adai; Andrew Hadd; Gary J. Latham; Ludmila Danilova; Justin A. Bishop; Ryan J. Li; William H. Westra; Patrick T. Hennessey; Wayne M. Koch; Michael F. Ochs; Joseph A. Califano; Wenyue Sun

Development of head and neck squamous cell carcinoma (HNSCC) is characterized by accumulation of mutations in several oncogenes and tumor suppressor genes. We have formerly described the mutation pattern of HNSCC and described NOTCH signaling pathway alterations. Given the complexity of the HNSCC, here we extend the previous study to understand the overall HNSCC mutation context and to discover additional genetic alterations. We performed high depth targeted exon sequencing of 51 highly actionable cancer-related genes with a high frequency of mutation across many cancer types, including head and neck. DNA from primary tumor tissues and matched normal tissues was analyzed for 37 HNSCC patients. We identified 26 non-synonymous or stop-gained mutations targeting 11 of 51 selected genes. These genes were mutated in 17 out of 37 (46%) studied HNSCC patients. Smokers harbored 3.2-fold more mutations than non-smokers. Importantly, TP53 was mutated in 30%, NOTCH1 in 8% and FGFR3 in 5% of HNSCC. HPV negative patients harbored 4-fold more TP53 mutations than HPV positive patients. These data confirm prior reports of the HNSCC mutational profile. Additionally, we detected mutations in two new genes, CEBPA and FES, which have not been previously reported in HNSCC. These data extend the spectrum of HNSCC mutations and define novel mutation targets in HNSCC carcinogenesis, especially for smokers and HNSCC without HPV infection.

Collaboration


Dive into the Ludmila Danilova's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elana J. Fertig

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Theresa Guo

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Emily Flam

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Justin A. Bishop

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Bahman Afsari

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge