Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
Nature ; 629(8010): 211-218, 2024 May.
Article in English | MEDLINE | ID: mdl-38600391

ABSTRACT

A major limitation of chimeric antigen receptor (CAR) T cell therapies is the poor persistence of these cells in vivo1. The expression of memory-associated genes in CAR T cells is linked to their long-term persistence in patients and clinical efficacy2-6, suggesting that memory programs may underpin durable CAR T cell function. Here we show that the transcription factor FOXO1 is responsible for promoting memory and restraining exhaustion in human CAR T cells. Pharmacological inhibition or gene editing of endogenous FOXO1 diminished the expression of memory-associated genes, promoted an exhaustion-like phenotype and impaired the antitumour activity of CAR T cells. Overexpression of FOXO1 induced a gene-expression program consistent with T cell memory and increased chromatin accessibility at FOXO1-binding motifs. CAR T cells that overexpressed FOXO1 retained their function, memory potential and metabolic fitness in settings of chronic stimulation, and exhibited enhanced persistence and tumour control in vivo. By contrast, overexpression of TCF1 (encoded by TCF7) did not enforce canonical memory programs or enhance the potency of CAR T cells. Notably, FOXO1 activity correlated with positive clinical outcomes of patients treated with CAR T cells or tumour-infiltrating lymphocytes, underscoring the clinical relevance of FOXO1 in cancer immunotherapy. Our results show that overexpressing FOXO1 can increase the antitumour activity of human CAR T cells, and highlight memory reprogramming as a broadly applicable approach for optimizing therapeutic T cell states.


Subject(s)
Forkhead Box Protein O1 , Immunologic Memory , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , T-Lymphocytes , Animals , Humans , Mice , Cell Line, Tumor , Chromatin/metabolism , Chromatin/genetics , Forkhead Box Protein O1/metabolism , Gene Editing , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/cytology
3.
Cancer Immunol Res ; 11(1): 13-19, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36255409

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 has been a clinical breakthrough for pediatric B-cell acute lymphoblastic leukemia (B-ALL), and loss of the CD19 target antigen on leukemic cells represents a major mechanism of relapse. Previous studies have observed CD19 mutations specific to CD19- relapses, and we sought to clarify and strengthen this relationship using deep whole-exome sequencing in leukemic cells expanded in a patient-derived xenograft. By assessing pre-treatment and relapse cells from 13 patients treated with CAR T-cell therapy, 8 of whom developed CD19- relapse and 5 of whom developed CD19+ relapse, we demonstrate that relapse-specific single-nucleotide variants and small indels with high allele frequency combined with deletions in the CD19 gene in a manner specific to those patients with CD19- relapse. Before CAR T-cell infusion, one patient was found to harbor a pre-existing CD19 deletion in the context of genomic instability, which likely represented the first hit leading to the patient's subsequent CD19- relapse. Across patients, preexisting mutations and genomic instability were not significant predictors of subsequent CD19- relapse across patients, with sample size as a potential limiting factor. Together, our results clarify and strengthen the relationship between genomic events and CD19- relapse, demonstrating this intriguing mechanism of resistance to a targeted cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive , T-Lymphocytes , Humans , Child , Immunotherapy, Adoptive/methods , Immunotherapy , Recurrence , Antigens, CD19 , Adaptor Proteins, Signal Transducing , Genomics , Receptors, Antigen, T-Cell
5.
Sci Transl Med ; 14(650): eabn3353, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35731887

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapies targeting CD19 and CD22 have been successful for treating B cell cancers, but CAR T cells targeting non-B cell cancers remain unsuccessful. We propose that rather than being strictly a side effect of therapy, the large number of CAR interactions with normal B cells may be a key contributor to clinical CAR T cell responses.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Immunotherapy, Adoptive , Antigens, CD19 , B-Lymphocytes , Humans , Immunotherapy, Adoptive/adverse effects , Receptors, Antigen, T-Cell , T-Lymphocytes
6.
Nature ; 602(7897): 503-509, 2022 02.
Article in English | MEDLINE | ID: mdl-35110735

ABSTRACT

The adoptive transfer of T lymphocytes reprogrammed to target tumour cells has demonstrated potential for treatment of various cancers1-7. However, little is known about the long-term potential and clonal stability of the infused cells. Here we studied long-lasting CD19-redirected chimeric antigen receptor (CAR) T cells in two patients with chronic lymphocytic leukaemia1-4 who achieved a complete remission in 2010. CAR T cells remained detectable more than ten years after infusion, with sustained remission in both patients. Notably, a highly activated CD4+ population emerged in both patients, dominating the CAR T cell population at the later time points. This transition was reflected in the stabilization of the clonal make-up of CAR T cells with a repertoire dominated by a small number of clones. Single-cell profiling demonstrated that these long-persisting CD4+ CAR T cells exhibited cytotoxic characteristics along with ongoing functional activation and proliferation. In addition, longitudinal profiling revealed a population of gamma delta CAR T cells that prominently expanded in one patient concomitant with CD8+ CAR T cells during the initial response phase. Our identification and characterization of these unexpected CAR T cell populations provide novel insight into the CAR T cell characteristics associated with anti-cancer response and long-term remission in leukaemia.


Subject(s)
CD4-Positive T-Lymphocytes , Immunotherapy, Adoptive , Leukemia , Receptors, Chimeric Antigen , Antigens, CD19/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Humans , Leukemia/immunology , Leukemia/therapy , Receptors, Chimeric Antigen/immunology , Time Factors
7.
Clin Cancer Res ; 27(18): 5109-5122, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34210682

ABSTRACT

PURPOSE: Systems biology approaches can identify critical targets in complex cancer signaling networks to inform new therapy combinations that may overcome conventional treatment resistance. EXPERIMENTAL DESIGN: We performed integrated analysis of 1,046 childhood B-ALL cases and developed a data-driven network controllability-based approach to identify synergistic key regulator targets in Philadelphia chromosome-like B-acute lymphoblastic leukemia (Ph-like B-ALL), a common high-risk leukemia subtype associated with hyperactive signal transduction and chemoresistance. RESULTS: We identified 14 dysregulated network nodes in Ph-like ALL involved in aberrant JAK/STAT, Ras/MAPK, and apoptosis pathways and other critical processes. Genetic cotargeting of the synergistic key regulator pair STAT5B and BCL2-associated athanogene 1 (BAG1) significantly reduced leukemia cell viability in vitro. Pharmacologic inhibition with dual small molecule inhibitor therapy targeting this pair of key nodes further demonstrated enhanced antileukemia efficacy of combining the BCL-2 inhibitor venetoclax with the tyrosine kinase inhibitors ruxolitinib or dasatinib in vitro in human Ph-like ALL cell lines and in vivo in multiple childhood Ph-like ALL patient-derived xenograft models. Consistent with network controllability theory, co-inhibitor treatment also shifted the transcriptomic state of Ph-like ALL cells to become less like kinase-activated BCR-ABL1-rearranged (Ph+) B-ALL and more similar to prognostically favorable childhood B-ALL subtypes. CONCLUSIONS: Our study represents a powerful conceptual framework for combinatorial drug discovery based on systematic interrogation of synergistic vulnerability pathways with pharmacologic inhibitor validation in preclinical human leukemia models.


Subject(s)
Antineoplastic Agents , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Antineoplastic Agents/therapeutic use , Child , Dasatinib/pharmacology , Dasatinib/therapeutic use , Humans , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
8.
Cancer Discov ; 11(9): 2186-2199, 2021 09.
Article in English | MEDLINE | ID: mdl-33820778

ABSTRACT

The adoptive transfer of chimeric antigen receptor (CAR) T cells represents a breakthrough in clinical oncology, yet both between- and within-patient differences in autologously derived T cells are a major contributor to therapy failure. To interrogate the molecular determinants of clinical CAR T-cell persistence, we extensively characterized the premanufacture T cells of 71 patients with B-cell malignancies on trial to receive anti-CD19 CAR T-cell therapy. We performed RNA-sequencing analysis on sorted T-cell subsets from all 71 patients, followed by paired Cellular Indexing of Transcriptomes and Epitopes (CITE) sequencing and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) on T cells from six of these patients. We found that chronic IFN signaling regulated by IRF7 was associated with poor CAR T-cell persistence across T-cell subsets, and that the TCF7 regulon not only associates with the favorable naïve T-cell state, but is maintained in effector T cells among patients with long-term CAR T-cell persistence. These findings provide key insights into the underlying molecular determinants of clinical CAR T-cell function. SIGNIFICANCE: To improve clinical outcomes for CAR T-cell therapy, there is a need to understand the molecular determinants of CAR T-cell persistence. These data represent the largest clinically annotated molecular atlas in CAR T-cell therapy to date, and significantly advance our understanding of the mechanisms underlying therapeutic efficacy.This article is highlighted in the In This Issue feature, p. 2113.


Subject(s)
Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/transplantation , Adolescent , Child , Disease-Free Survival , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Philadelphia , T-Lymphocytes/immunology
9.
Clin Cancer Res ; 26(14): 3505-3513, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32127393

ABSTRACT

The adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells has opened a new frontier in cancer therapy. Unlike the paradigm of targeted therapies, the efficacy of CAR T-cell therapy depends not only on the choice of target but also on a complex interplay of tumor, immune, and stromal cell communication. This presents both challenges and opportunities from a discovery standpoint. Whereas cancer consortia have traditionally focused on the genomic, transcriptomic, epigenomic, and proteomic landscape of cancer cells, there is an increasing need to expand studies to analyze the interactions between tumor, immune, and stromal cell populations in their relevant anatomical and functional compartments. Here, we focus on the promising application of systems biology to address key challenges in CAR T-cell therapy, from understanding the mechanisms of therapeutic resistance in hematologic and solid tumors to addressing important clinical challenges in biomarker discovery and therapeutic toxicity. We propose a systems biology view of key clinical objectives in CAR T-cell therapy and suggest a path forward for a biomedical discovery process that leverages modern technological approaches in systems biology.


Subject(s)
Biomedical Research/methods , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Systems Biology , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/immunology , Disease Models, Animal , Humans , Immunotherapy, Adoptive/adverse effects , Models, Immunological , Neoplasms/diagnosis , Neoplasms/immunology , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
10.
Sci Rep ; 9(1): 8770, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217513

ABSTRACT

A wealth of transcriptomic and clinical data on solid tumours are under-utilized due to unharmonized data storage and format. We have developed the MetaGxData package compendium, which includes manually-curated and standardized clinical, pathological, survival, and treatment metadata across breast, ovarian, and pancreatic cancer data. MetaGxData is the largest compendium of curated transcriptomic data for these cancer types to date, spanning 86 datasets and encompassing 15,249 samples. Open access to standardized metadata across cancer types promotes use of their transcriptomic and clinical data in a variety of cross-tumour analyses, including identification of common biomarkers, and assessing the validity of prognostic signatures. Here, we demonstrate that MetaGxData is a flexible framework that facilitates meta-analyses by using it to identify common prognostic genes in ovarian and breast cancer. Furthermore, we use the data compendium to create the first gene signature that is prognostic in a meta-analysis across 3 cancer types. These findings demonstrate the potential of MetaGxData to serve as an important resource in oncology research, and provide a foundation for future development of cancer-specific compendia.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , Databases, Nucleic Acid , Ovarian Neoplasms , Pancreatic Neoplasms , Transcriptome , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Humans , Male , Metadata , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism
11.
JAMA Netw Open ; 2(3): e191083, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30901051

ABSTRACT

Importance: The language of medical research appears to be intrinsically tied to the culture of medical research and provides a unique window into broader trends in the culture of medicine. Objective: To analyze medical language from 5 premier medical journals and investigate broader changes in the culture of clinical investigation during the last 40 years. Design, Setting, and Participants: In this qualitative study using a data-driven analysis, 302 293 PubMed records were extracted from JAMA, The Lancet, Annals of Internal Medicine, the BMJ, and New England Journal of Medicine from January 1, 1976, through December 31, 2015, to identify key trends in medical language. A frequency analysis was applied across the 40-year time frame in JAMA to assess the major trends in all publication types. Patient-centered language was analyzed in clinical trials in the flanking time periods (1976-1980 and 2011-2015) across the 5 journals. Data were analyzed from November 16, 2016, through November 9, 2018. Main Outcomes and Measures: Increasing or decreasing frequency of words (monograms) and word pairs (bigrams) and the proportion of patient-centric words in journal article titles. Results: In JAMA, 50 277 articles of all publication types were included. In the frequency analysis, the most increased terms were reflective of the language of epidemiological research. The bigram analysis revealed a decline in causal language (-2.42/100 000 words to -2.03/100 000 words; false discovery rate [FDR], <0.01) and an increased description of patients in the plural form (6.92/100 000 words to 11.4/100 000 words; FDR, <0.01). A trend to separate patient from disease was observed; for example, there was a decrease in describing a patient as a diabetic (-2.21/100 000 words; FDR, <0.01) compared with a patient with diabetes. In the analysis of clinical trials in all 5 journals, 3125 titles were identified (range, 193-932 per journal). In 4 of the 5 journals, use of patient-centric keywords increased significantly (absolute increase, 18.9%-34.3%; P < .001 for 3 journals; P = .01 for 1 journal), with the New England Journal of Medicine as the exception. This finding reflects a change from shorter disease-centric titles to longer titles that describe patients with a disease. Conclusions and Relevance: Trends in medical language reflect the rise of evidence-based medicine, a shift in focus from individuals to populations, and a separation of patient and disease. Data-driven analysis of medical language provides a unique window into the changing landscape of medical culture.


Subject(s)
Biomedical Research/organization & administration , Language , Medical Writing , Periodicals as Topic/trends , Humans , Organizational Culture , Qualitative Research , Terminology as Topic
12.
BMC Bioinformatics ; 20(1): 42, 2019 Jan 21.
Article in English | MEDLINE | ID: mdl-30665349

ABSTRACT

BACKGROUND: We introduce BPG, a framework for generating publication-quality, highly-customizable plots in the R statistical environment. RESULTS: This open-source package includes multiple methods of displaying high-dimensional datasets and facilitates generation of complex multi-panel figures, making it suitable for complex datasets. A web-based interactive tool allows online figure customization, from which R code can be downloaded for integration with computational pipelines. CONCLUSION: BPG provides a new approach for linking interactive and scripted data visualization and is available at http://labs.oicr.on.ca/boutros-lab/software/bpg or via CRAN at https://cran.r-project.org/web/packages/BoutrosLab.plotting.general.


Subject(s)
Data Analysis , Simulation Training/methods , Humans , Software
13.
Clin Cancer Res ; 24(20): 5037-5047, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30084834

ABSTRACT

Purpose: The majority of ovarian carcinomas are of high-grade serous histology, which is associated with poor prognosis. Surgery and chemotherapy are the mainstay of treatment, and molecular characterization is necessary to lead the way to targeted therapeutic options. To this end, various computational methods for gene expression-based subtyping of high-grade serous ovarian carcinoma (HGSOC) have been proposed, but their overlap and robustness remain unknown.Experimental Design: We assess three major subtype classifiers by meta-analysis of publicly available expression data, and assess statistical criteria of subtype robustness and classifier concordance. We develop a consensus classifier that represents the subtype classifications of tumors based on the consensus of multiple methods, and outputs a confidence score. Using our compendium of expression data, we examine the possibility that a subset of tumors is unclassifiable based on currently proposed subtypes.Results: HGSOC subtyping classifiers exhibit moderate pairwise concordance across our data compendium (58.9%-70.9%; P < 10-5) and are associated with overall survival in a meta-analysis across datasets (P < 10-5). Current subtypes do not meet statistical criteria for robustness to reclustering across multiple datasets (prediction strength < 0.6). A new subtype classifier is trained on concordantly classified samples to yield a consensus classification of patient tumors that correlates with patient age, survival, tumor purity, and lymphocyte infiltration.Conclusions: A new consensus ovarian subtype classifier represents the consensus of methods and demonstrates the importance of classification approaches for cancer that do not require all tumors to be assigned to a distinct subtype. Clin Cancer Res; 24(20); 5037-47. ©2018 AACR.


Subject(s)
Biomarkers, Tumor , Cystadenocarcinoma, Serous/diagnosis , Cystadenocarcinoma, Serous/etiology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/etiology , Algorithms , Clinical Decision-Making , Consensus , Cystadenocarcinoma, Serous/mortality , Disease Management , Disease Susceptibility , Female , Gene Expression Profiling , Humans , Neoplasm Grading , Ovarian Neoplasms/mortality , Prognosis , ROC Curve , Reproducibility of Results
14.
J Pathol ; 241(3): 375-391, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27861902

ABSTRACT

The histopathological evaluation of morphological features in breast tumours provides prognostic information to guide therapy. Adjunct molecular analyses provide further diagnostic, prognostic and predictive information. However, there is limited knowledge of the molecular basis of morphological phenotypes in invasive breast cancer. This study integrated genomic, transcriptomic and protein data to provide a comprehensive molecular profiling of morphological features in breast cancer. Fifteen pathologists assessed 850 invasive breast cancer cases from The Cancer Genome Atlas (TCGA). Morphological features were significantly associated with genomic alteration, DNA methylation subtype, PAM50 and microRNA subtypes, proliferation scores, gene expression and/or reverse-phase protein assay subtype. Marked nuclear pleomorphism, necrosis, inflammation and a high mitotic count were associated with the basal-like subtype, and had a similar molecular basis. Omics-based signatures were constructed to predict morphological features. The association of morphology transcriptome signatures with overall survival in oestrogen receptor (ER)-positive and ER-negative breast cancer was first assessed by use of the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset; signatures that remained prognostic in the METABRIC multivariate analysis were further evaluated in five additional datasets. The transcriptomic signature of poorly differentiated epithelial tubules was prognostic in ER-positive breast cancer. No signature was prognostic in ER-negative breast cancer. This study provided new insights into the molecular basis of breast cancer morphological phenotypes. The integration of morphological with molecular data has the potential to refine breast cancer classification, predict response to therapy, enhance our understanding of breast cancer biology, and improve clinical management. This work is publicly accessible at www.dx.ai/tcga_breast. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Databases, Genetic , Female , Gene Expression Profiling , Genomics , Humans , Neoplasm Invasiveness , Phenotype , Receptors, Estrogen/metabolism
15.
Nat Chem Biol ; 12(12): 1007-1014, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27694801

ABSTRACT

Polyketides (PKs) and nonribosomal peptides (NRPs) are profoundly important natural products, forming the foundations of many therapeutic regimes. Decades of research have revealed over 11,000 PK and NRP structures, and genome sequencing is uncovering new PK and NRP gene clusters at an unprecedented rate. However, only ∼10% of PK and NRPs are currently associated with gene clusters, and it is unclear how many of these orphan gene clusters encode previously isolated molecules. Therefore, to efficiently guide the discovery of new molecules, we must first systematically de-orphan emergent gene clusters from genomes. Here we provide to our knowledge the first comprehensive retro-biosynthetic program, generalized retro-biosynthetic assembly prediction engine (GRAPE), for PK and NRP families and introduce a computational pipeline, global alignment for natural products cheminformatics (GARLIC), to uncover how observed biosynthetic gene clusters relate to known molecules, leading to the identification of gene clusters that encode new molecules.


Subject(s)
Multigene Family , Peptide Biosynthesis, Nucleic Acid-Independent , Peptides/metabolism , Polyketides/metabolism , Algorithms , Multigene Family/genetics , Peptide Biosynthesis, Nucleic Acid-Independent/genetics , Peptides/chemistry , Peptides/genetics , Polyketides/chemistry
16.
PLoS Comput Biol ; 12(6): e1004890, 2016 06.
Article in English | MEDLINE | ID: mdl-27351836

ABSTRACT

Acute Myeloid Leukemia (AML) is a fatal hematological cancer. The genetic abnormalities underlying AML are extremely heterogeneous among patients, making prognosis and treatment selection very difficult. While clinical proteomics data has the potential to improve prognosis accuracy, thus far, the quantitative means to do so have yet to be developed. Here we report the results and insights gained from the DREAM 9 Acute Myeloid Prediction Outcome Prediction Challenge (AML-OPC), a crowdsourcing effort designed to promote the development of quantitative methods for AML prognosis prediction. We identify the most accurate and robust models in predicting patient response to therapy, remission duration, and overall survival. We further investigate patient response to therapy, a clinically actionable prediction, and find that patients that are classified as resistant to therapy are harder to predict than responsive patients across the 31 models submitted to the challenge. The top two performing models, which held a high sensitivity to these patients, substantially utilized the proteomics data to make predictions. Using these models, we also identify which signaling proteins were useful in predicting patient therapeutic response.


Subject(s)
Algorithms , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/therapy , Crowdsourcing/methods , Outcome and Process Assessment, Health Care/methods , Proteome/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Biomarkers/metabolism , Humans , Reproducibility of Results , Risk Assessment , Sensitivity and Specificity , Treatment Outcome
17.
Nat Chem Biol ; 12(4): 233-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26829473

ABSTRACT

Antibiotics are essential for numerous medical procedures, including the treatment of bacterial infections, but their widespread use has led to the accumulation of resistance, prompting calls for the discovery of antibacterial agents with new targets. A majority of clinically approved antibacterial scaffolds are derived from microbial natural products, but these valuable molecules are not well annotated or organized, limiting the efficacy of modern informatic analyses. Here, we provide a comprehensive resource defining the targets, chemical origins and families of the natural antibacterial collective through a retrobiosynthetic algorithm. From this we also detail the directed mining of biosynthetic scaffolds and resistance determinants to reveal structures with a high likelihood of having previously unknown modes of action. Implementing this pipeline led to investigations of the telomycin family of natural products from Streptomyces canus, revealing that these bactericidal molecules possess a new antibacterial mode of action dependent on the bacterial phospholipid cardiolipin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biological Products/pharmacology , Cardiolipins/biosynthesis , Gram-Positive Bacteria/drug effects , Peptides/pharmacology , Streptomyces/metabolism , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/isolation & purification , Biological Products/isolation & purification , Biosynthetic Pathways , Cardiolipins/genetics , Colony Count, Microbial , Databases, Genetic , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/growth & development , Gram-Positive Bacteria/metabolism , Microbial Sensitivity Tests , Multigene Family , Peptides/genetics , Peptides/isolation & purification , Web Browser
18.
Cancer Res ; 75(21): 4494-503, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26363007

ABSTRACT

The cell surface nucleotidase CD73 is an immunosuppressive enzyme involved in tumor progression and metastasis. Although preclinical studies suggest that CD73 can be targeted for cancer treatment, the clinical impact of CD73 in ovarian cancer remains unclear. In this study, we investigated the prognostic value of CD73 in high-grade serous (HGS) ovarian cancer using gene and protein expression analyses. Our results demonstrate that high levels of CD73 are significantly associated with shorter disease-free survival and overall survival in patients with HGS ovarian cancer. Furthermore, high levels of CD73 expression in ovarian tumor cells abolished the good prognosis associated with intraepithelial CD8(+) cells. Notably, CD73 gene expression was highest in the C1/stromal molecular subtype of HGS ovarian cancer and positively correlated with an epithelial-to-mesenchymal transition gene signature. Moreover, in vitro studies revealed that CD73 and extracellular adenosine enhance ovarian tumor cell growth as well as expression of antiapoptotic BCL-2 family members. Finally, in vivo coinjection of ID8 mouse ovarian tumor cells with mouse embryonic fibroblasts showed that CD73 expression in fibroblasts promotes tumor immune escape and thereby tumor growth. In conclusion, our study highlights a role for CD73 as a prognostic marker of patient survival and also as a candidate therapeutic target in HGS ovarian cancers.


Subject(s)
5'-Nucleotidase/metabolism , Biomarkers, Tumor/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , 5'-Nucleotidase/genetics , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation/genetics , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Female , Fibroblasts/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering , Tumor Escape/immunology
19.
Nat Genet ; 47(7): 736-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26005866

ABSTRACT

Herein we provide a detailed molecular analysis of the spatial heterogeneity of clinically localized, multifocal prostate cancer to delineate new oncogenes or tumor suppressors. We initially determined the copy number aberration (CNA) profiles of 74 patients with index tumors of Gleason score 7. Of these, 5 patients were subjected to whole-genome sequencing using DNA quantities achievable in diagnostic biopsies, with detailed spatial sampling of 23 distinct tumor regions to assess intraprostatic heterogeneity in focal genomics. Multifocal tumors are highly heterogeneous for single-nucleotide variants (SNVs), CNAs and genomic rearrangements. We identified and validated a new recurrent amplification of MYCL, which is associated with TP53 deletion and unique profiles of DNA damage and transcriptional dysregulation. Moreover, we demonstrate divergent tumor evolution in multifocal cancer and, in some cases, tumors of independent clonal origin. These data represent the first systematic relation of intraprostatic genomic heterogeneity to predicted clinical outcome and inform the development of novel biomarkers that reflect individual prognosis.


Subject(s)
Prostatic Neoplasms/genetics , Cell Line, Tumor , DNA Copy Number Variations , Genetic Association Studies , Genetic Heterogeneity , Genome, Human , Humans , Male , Middle Aged , Neoplasm Grading , Point Mutation , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...