Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Publication year range
1.
Cell ; 184(21): 5482-5496.e28, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34597583

ABSTRACT

Determining how cells vary with their local signaling environment and organize into distinct cellular communities is critical for understanding processes as diverse as development, aging, and cancer. Here we introduce EcoTyper, a machine learning framework for large-scale identification and validation of cell states and multicellular communities from bulk, single-cell, and spatially resolved gene expression data. When applied to 12 major cell lineages across 16 types of human carcinoma, EcoTyper identified 69 transcriptionally defined cell states. Most states were specific to neoplastic tissue, ubiquitous across tumor types, and significantly prognostic. By analyzing cell-state co-occurrence patterns, we discovered ten clinically distinct multicellular communities with unexpectedly strong conservation, including three with myeloid and stromal elements linked to adverse survival, one enriched in normal tissue, and two associated with early cancer development. This study elucidates fundamental units of cellular organization in human carcinoma and provides a framework for large-scale profiling of cellular ecosystems in any tissue.


Subject(s)
Neoplasms/pathology , Tumor Microenvironment , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Inflammation/pathology , Ligands , Neoplasms/genetics , Phenotype , Prognosis , Transcription, Genetic
2.
Nat Immunol ; 23(2): 318-329, 2022 02.
Article in English | MEDLINE | ID: mdl-35058616

ABSTRACT

Tuberculosis (TB) in humans is characterized by formation of immune-rich granulomas in infected tissues, the architecture and composition of which are thought to affect disease outcome. However, our understanding of the spatial relationships that control human granulomas is limited. Here, we used multiplexed ion beam imaging by time of flight (MIBI-TOF) to image 37 proteins in tissues from patients with active TB. We constructed a comprehensive atlas that maps 19 cell subsets across 8 spatial microenvironments. This atlas shows an IFN-γ-depleted microenvironment enriched for TGF-ß, regulatory T cells and IDO1+ PD-L1+ myeloid cells. In a further transcriptomic meta-analysis of peripheral blood from patients with TB, immunoregulatory trends mirror those identified by granuloma imaging. Notably, PD-L1 expression is associated with progression to active TB and treatment response. These data indicate that in TB granulomas, there are local spatially coordinated immunoregulatory programs with systemic manifestations that define active TB.


Subject(s)
Granuloma/immunology , Tuberculosis/immunology , B7-H1 Antigen/immunology , Cells, Cultured , Cytokines/immunology , Gene Expression Profiling/methods , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , Myeloid Cells/immunology
4.
Nature ; 619(7970): 595-605, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37468587

ABSTRACT

Beginning in the first trimester, fetally derived extravillous trophoblasts (EVTs) invade the uterus and remodel its spiral arteries, transforming them into large, dilated blood vessels. Several mechanisms have been proposed to explain how EVTs coordinate with the maternal decidua to promote a tissue microenvironment conducive to spiral artery remodelling (SAR)1-3. However, it remains a matter of debate regarding which immune and stromal cells participate in these interactions and how this evolves with respect to gestational age. Here we used a multiomics approach, combining the strengths of spatial proteomics and transcriptomics, to construct a spatiotemporal atlas of the human maternal-fetal interface in the first half of pregnancy. We used multiplexed ion beam imaging by time-of-flight and a 37-plex antibody panel to analyse around 500,000 cells and 588 arteries within intact decidua from 66 individuals between 6 and 20 weeks of gestation, integrating this dataset with co-registered transcriptomics profiles. Gestational age substantially influenced the frequency of maternal immune and stromal cells, with tolerogenic subsets expressing CD206, CD163, TIM-3, galectin-9 and IDO-1 becoming increasingly enriched and colocalized at later time points. By contrast, SAR progression preferentially correlated with EVT invasion and was transcriptionally defined by 78 gene ontology pathways exhibiting distinct monotonic and biphasic trends. Last, we developed an integrated model of SAR whereby invasion is accompanied by the upregulation of pro-angiogenic, immunoregulatory EVT programmes that promote interactions with the vascular endothelium while avoiding the activation of maternal immune cells.


Subject(s)
Maternal-Fetal Exchange , Trophoblasts , Uterus , Female , Humans , Pregnancy , Arteries/physiology , Decidua/blood supply , Decidua/cytology , Decidua/immunology , Decidua/physiology , Pregnancy Trimester, First/genetics , Pregnancy Trimester, First/metabolism , Pregnancy Trimester, First/physiology , Trophoblasts/cytology , Trophoblasts/immunology , Trophoblasts/physiology , Uterus/blood supply , Uterus/cytology , Uterus/immunology , Uterus/physiology , Maternal-Fetal Exchange/genetics , Maternal-Fetal Exchange/immunology , Maternal-Fetal Exchange/physiology , Time Factors , Proteomics , Gene Expression Profiling , Datasets as Topic , Gestational Age
5.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34789568

ABSTRACT

Cancer precision medicine implies identification of tumor-specific vulnerabilities associated with defined oncogenic pathways. Desmoid tumors are soft-tissue neoplasms strictly driven by Wnt signaling network hyperactivation. Despite this clearly defined genetic etiology and the strict and unique implication of the Wnt/ß-catenin pathway, no specific molecular targets for these tumors have been identified. To address this caveat, we developed fast, efficient, and penetrant genetic Xenopus tropicalis desmoid tumor models to identify and characterize drug targets. We used multiplexed CRISPR/Cas9 genome editing in these models to simultaneously target a tumor suppressor gene (apc) and candidate dependency genes. Our methodology CRISPR/Cas9 selection-mediated identification of dependencies (CRISPR-SID) uses calculated deviations between experimentally observed gene editing outcomes and deep-learning-predicted double-strand break repair patterns to identify genes under negative selection during tumorigenesis. This revealed EZH2 and SUZ12, both encoding polycomb repressive complex 2 components, and the transcription factor CREB3L1 as genetic dependencies for desmoid tumors. In vivo EZH2 inhibition by Tazemetostat induced partial regression of established autochthonous tumors. In vitro models of patient desmoid tumor cells revealed a direct effect of Tazemetostat on Wnt pathway activity. CRISPR-SID represents a potent approach for in vivo mapping of tumor vulnerabilities and drug target identification.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/isolation & purification , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Editing/methods , Abdominal Neoplasms/genetics , Adenomatous Polyposis Coli/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein , Fibromatosis, Aggressive/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins , Oncogenes , Polycomb Repressive Complex 2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway , Xenopus , beta Catenin
6.
Lab Invest ; 102(7): 762-770, 2022 07.
Article in English | MEDLINE | ID: mdl-35351966

ABSTRACT

Multiplexed ion beam imaging by time-of-flight (MIBI-TOF) is a form of mass spectrometry imaging that uses metal labeled antibodies and secondary ion mass spectrometry to image dozens of proteins simultaneously in the same tissue section. Working with the National Cancer Institute's (NCI) Cancer Immune Monitoring and Analysis Centers (CIMAC), we undertook a validation study, assessing concordance across a dozen serial sections of a tissue microarray of 21 samples that were independently processed and imaged by MIBI-TOF or single-plex immunohistochemistry (IHC) over 12 days. Pixel-level features were highly concordant across all 16 targets assessed in both staining intensity (R2 = 0.94 ± 0.04) and frequency (R2 = 0.95 ± 0.04). Comparison to digitized, single-plex IHC on adjacent serial sections revealed similar concordance (R2 = 0.85 ± 0.08) as well. Lastly, automated segmentation and clustering of eight cell populations found that cell frequencies between serial sections yielded an average correlation of R2 = 0.94 ± 0.05. Taken together, we demonstrate that MIBI-TOF, with well-vetted reagents and automated analysis, can generate consistent and quantitative annotations of clinically relevant cell states in archival human tissue, and more broadly, present a scalable framework for benchmarking multiplexed IHC approaches.


Subject(s)
Diagnostic Imaging , Neoplasms , Antibodies , Diagnostic Imaging/methods , Humans , Immunohistochemistry , Ions , Mass Spectrometry/methods
7.
Cancer ; 127(15): 2666-2673, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33788262

ABSTRACT

BACKGROUND: Leiomyosarcoma (LMS) is the most common soft tissue and uterine sarcoma, but no standard therapy is available for recurrent or metastatic LMS. TP53, p16/RB1, and PI3K/mTOR pathway dysregulations are recurrent events, and some LMS express estrogen receptor (ER) and/or progesterone receptor (PR). To characterize relationships between these pathway perturbations, the authors evaluated protein expression in soft tissue and uterine nonprimary leiomyosarcoma (np-LMS), including local recurrences and distant metastases. METHODS: TP53, RB1, p16, and PTEN expression aberrations were determined by immunohistochemistry (IHC) in tissue microarrays (TMAs) from 227 np-LMS and a comparison group of 262 primary leiomyosarcomas (p-LMS). Thirty-five of the np-LMS had a matched p-LMS specimen in the TMAs. Correlative studies included differentiation scoring, ER and PR IHC, and CDKN2A/p16 fluorescence in situ hybridization. RESULTS: Dysregulation of TP53, p16/RB1, and PTEN was demonstrated in 90%, 95%, and 41% of np-LMS, respectively. PTEN inactivation was more common in soft tissue np-LMS than uterine np-LMS (55% vs 31%; P = .0005). Moderate-strong ER expression was more common in uterine np-LMS than soft tissue np-LMS (50% vs 7%; P < .0001). Co-inactivation of TP53 and RB1 was found in 81% of np-LMS and was common in both soft tissue and uterine np-LMS (90% and 74%, respectively). RB1, p16, and PTEN aberrations were nearly always conserved in p-LMS and np-LMS from the same patients. CONCLUSIONS: These studies show that nearly all np-LMS have TP53 and/or RB1 aberrations. Therefore, therapies targeting cell cycle and DNA damage checkpoint vulnerabilities should be prioritized for evaluations in LMS.


Subject(s)
Genes, p53 , Leiomyosarcoma , Retinoblastoma Binding Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Uterine Neoplasms , Female , Genes, p16 , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , PTEN Phosphohydrolase/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
8.
N Engl J Med ; 374(3): 211-22, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26789870

ABSTRACT

Background The identification of high-risk stage II colon cancers is key to the selection of patients who require adjuvant treatment after surgery. Microarray-based multigene-expression signatures derived from stem cells and progenitor cells hold promise, but they are difficult to use in clinical practice. Methods We used a new bioinformatics approach to search for biomarkers of colon epithelial differentiation across gene-expression arrays and then ranked candidate genes according to the availability of clinical-grade diagnostic assays. With the use of subgroup analysis involving independent and retrospective cohorts of patients with stage II or stage III colon cancer, the top candidate gene was tested for its association with disease-free survival and a benefit from adjuvant chemotherapy. Results The transcription factor CDX2 ranked first in our screening test. A group of 87 of 2115 tumor samples (4.1%) lacked CDX2 expression. In the discovery data set, which included 466 patients, the rate of 5-year disease-free survival was lower among the 32 patients (6.9%) with CDX2-negative colon cancers than among the 434 (93.1%) with CDX2-positive colon cancers (hazard ratio for disease recurrence, 3.44; 95% confidence interval [CI], 1.60 to 7.38; P=0.002). In the validation data set, which included 314 patients, the rate of 5-year disease-free survival was lower among the 38 patients (12.1%) with CDX2 protein-negative colon cancers than among the 276 (87.9%) with CDX2 protein-positive colon cancers (hazard ratio, 2.42; 95% CI, 1.36 to 4.29; P=0.003). In both these groups, these findings were independent of the patient's age, sex, and tumor stage and grade. Among patients with stage II cancer, the difference in 5-year disease-free survival was significant both in the discovery data set (49% among 15 patients with CDX2-negative tumors vs. 87% among 191 patients with CDX2-positive tumors, P=0.003) and in the validation data set (51% among 15 patients with CDX2-negative tumors vs. 80% among 106 patients with CDX2-positive tumors, P=0.004). In a pooled database of all patient cohorts, the rate of 5-year disease-free survival was higher among 23 patients with stage II CDX2-negative tumors who were treated with adjuvant chemotherapy than among 25 who were not treated with adjuvant chemotherapy (91% vs. 56%, P=0.006). Conclusions Lack of CDX2 expression identified a subgroup of patients with high-risk stage II colon cancer who appeared to benefit from adjuvant chemotherapy. (Funded by the National Comprehensive Cancer Network, the National Institutes of Health, and others.).


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Colonic Neoplasms/genetics , Gene Expression , Homeodomain Proteins/metabolism , Analysis of Variance , Biomarkers, Tumor/genetics , CDX2 Transcription Factor , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Computational Biology , Databases, Genetic , Disease-Free Survival , Female , Homeodomain Proteins/genetics , Humans , Male , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/metabolism , Retrospective Studies
9.
Development ; 143(19): 3632-3637, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27702788

ABSTRACT

In situ hybridization methods are used across the biological sciences to map mRNA expression within intact specimens. Multiplexed experiments, in which multiple target mRNAs are mapped in a single sample, are essential for studying regulatory interactions, but remain cumbersome in most model organisms. Programmable in situ amplifiers based on the mechanism of hybridization chain reaction (HCR) overcome this longstanding challenge by operating independently within a sample, enabling multiplexed experiments to be performed with an experimental timeline independent of the number of target mRNAs. To assist biologists working across a broad spectrum of organisms, we demonstrate multiplexed in situ HCR in diverse imaging settings: bacteria, whole-mount nematode larvae, whole-mount fruit fly embryos, whole-mount sea urchin embryos, whole-mount zebrafish larvae, whole-mount chicken embryos, whole-mount mouse embryos and formalin-fixed paraffin-embedded human tissue sections. In addition to straightforward multiplexing, in situ HCR enables deep sample penetration, high contrast and subcellular resolution, providing an incisive tool for the study of interlaced and overlapping expression patterns, with implications for research communities across the biological sciences.


Subject(s)
In Situ Hybridization/methods , RNA, Messenger/metabolism , Animals , Drosophila , Embryo, Nonmammalian/metabolism , Humans , Zebrafish
10.
N Engl J Med ; 373(5): 428-37, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26222558

ABSTRACT

BACKGROUND: Expression of the colony-stimulating factor 1 (CSF1) gene is elevated in most tenosynovial giant-cell tumors. This observation has led to the discovery and clinical development of therapy targeting the CSF1 receptor (CSF1R). METHODS: Using x-ray co-crystallography to guide our drug-discovery research, we generated a potent, selective CSF1R inhibitor, PLX3397, that traps the kinase in the autoinhibited conformation. We then conducted a multicenter, phase 1 trial in two parts to analyze this compound. In the first part, we evaluated escalations in the dose of PLX3397 that was administered orally in patients with solid tumors (dose-escalation study). In the second part, we evaluated PLX3397 at the chosen phase 2 dose in an extension cohort of patients with tenosynovial giant-cell tumors (extension study). Pharmacokinetic and tumor responses in the enrolled patients were assessed, and CSF1 in situ hybridization was performed to confirm the mechanism of action of PLX3397 and that the pattern of CSF1 expression was consistent with the pathological features of tenosynovial giant-cell tumor. RESULTS: A total of 41 patients were enrolled in the dose-escalation study, and an additional 23 patients were enrolled in the extension study. The chosen phase 2 dose of PLX3397 was 1000 mg per day. In the extension study, 12 patients with tenosynovial giant-cell tumors had a partial response and 7 patients had stable disease. Responses usually occurred within the first 4 months of treatment, and the median duration of response exceeded 8 months. The most common adverse events included fatigue, change in hair color, nausea, dysgeusia, and periorbital edema; adverse events rarely led to discontinuation of treatment. CONCLUSIONS: Treatment of tenosynovial giant-cell tumors with PLX3397 resulted in a prolonged regression in tumor volume in most patients. (Funded by Plexxikon; ClinicalTrials.gov number, NCT01004861.).


Subject(s)
Aminopyridines/administration & dosage , Giant Cell Tumors/drug therapy , Pyrroles/administration & dosage , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Soft Tissue Neoplasms/drug therapy , Adult , Aged , Aminopyridines/adverse effects , Aminopyridines/pharmacokinetics , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Discovery , Female , Giant Cell Tumors/pathology , Humans , Male , Middle Aged , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Soft Tissue Neoplasms/pathology , Tendons/pathology , Tumor Burden
11.
Gynecol Oncol ; 149(2): 388-393, 2018 05.
Article in English | MEDLINE | ID: mdl-29544705

ABSTRACT

OBJECTIVE: Low-grade endometrial stromal sarcomas (LGESS) harbor chromosomal translocations that affect proteins associated with chromatin remodeling Polycomb Repressive Complex 2 (PRC2), including SUZ12, PHF1 and EPC1. Roughly half of LGESS also demonstrate nuclear accumulation of ß-catenin, which is a hallmark of Wnt signaling activation. However, the targets affected by the fusion proteins and the role of Wnt signaling in the pathogenesis of these tumors remain largely unknown. METHODS: Here we report the results of a meta-analysis of three independent gene expression profiling studies on LGESS and immunohistochemical evaluation of nuclear expression of ß-catenin and Lef1 in 112 uterine sarcoma specimens obtained from 20 LGESS and 89 LMS patients. RESULTS: Our results demonstrate that 143 out of 310 genes overexpressed in LGESS are known to be directly regulated by SUZ12. In addition, our gene expression meta-analysis shows activation of multiple genes implicated in Wnt signaling. We further emphasize the role of the Wnt signaling pathway by demonstrating concordant nuclear expression of ß-catenin and Lef1 in 7/16 LGESS. CONCLUSIONS: Based on our findings, we suggest that LGESS-specific fusion proteins disrupt the repressive function of the PRC2 complex similar to the mechanism seen in synovial sarcoma, where the SS18-SSX fusion proteins disrupt the mSWI/SNF (BAF) chromatin remodeling complex. We propose that these fusion proteins in LGESS contribute to overexpression of Wnt ligands with subsequent activation of Wnt signaling pathway and formation of an active ß-catenin/Lef1 transcriptional complex. These observations could lead to novel therapeutic approaches that focus on the Wnt pathway in LGESS.


Subject(s)
Endometrial Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Sarcoma, Endometrial Stromal/genetics , Wnt Signaling Pathway/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Lymphoid Enhancer-Binding Factor 1/genetics , Neoplasm Grading , Neoplasm Proteins , Oncogene Proteins, Fusion/metabolism , Polycomb Repressive Complex 2/biosynthesis , Polycomb Repressive Complex 2/genetics , Sarcoma, Endometrial Stromal/metabolism , Sarcoma, Endometrial Stromal/pathology , Tissue Array Analysis , Transcription Factors , beta Catenin/biosynthesis , beta Catenin/genetics
12.
Mod Pathol ; 30(9): 1312-1320, 2017 09.
Article in English | MEDLINE | ID: mdl-28643791

ABSTRACT

PAX7 is a paired-box transcription factor that is required for the developmental specification of adult skeletal muscle progenitors in mice. We previously demonstrated PAX7 expression as a marker of skeletal muscle differentiation in rhabdomyosarcoma. Here, using analyses of published whole-genome gene expression microarray data, we identify PAX7 as a gene with significantly increased expression in Ewing sarcoma in comparison to CIC-DUX4 round cell sarcoma. Analysis of PAX7 in a large cohort of 103 Ewing sarcoma cases by immunohistochemistry revealed expression in 99.0% of cases (102/103). PAX7 expression was noted in cases demonstrating three distinct Ewing sarcoma EWSR1 translocations involving FLI1, ERG, and NFATc2. No PAX7 expression was observed in any of 27 cases of CIC-DUX4 sarcoma by immunohistochemistry (0%; 0/27). Exploring the mechanism of PAX7 expression in Ewing sarcoma using curated RNA- and ChIP-sequencing data, we demonstrate that the EWSR1 fusion protein is required for PAX7 expression in Ewing sarcoma and identify a candidate EWSR1-FLI1-bound PAX7 enhancer that coincides with both a consensus GGAA repeat-containing binding site and a peak of regulatory H3K27 acetylation. Taken together, our findings provide mechanistic support for the utility of PAX7 immunohistochemistry in the diagnosis of Ewing sarcoma, while linking this sarcoma of uncertain histogenesis to a key transcriptional regulator of mammalian muscle progenitor cells.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , PAX7 Transcription Factor/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/metabolism , Acetylation , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Computational Biology , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Gene Fusion , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , PAX7 Transcription Factor/genetics , Protein Binding , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Transcription, Genetic , United States
13.
Gastroenterology ; 149(3): 705-17.e2, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26026391

ABSTRACT

BACKGROUND & AIMS: Receptor tyrosine kinase (RTK) inhibitors have advanced colon cancer treatment. We investigated the role of the RTK KIT in development of human colon cancer. METHODS: An array of 137 patient-derived colon tumors and their associated xenografts were analyzed by immunohistochemistry to measure levels of KIT and its ligand KITLG. KIT and/or KITLG was stably knocked down by expression of small hairpin RNAs from lentiviral vectors in DLD1, HT29, LS174T, and COLO320 DM colon cancer cell lines, and in UM-COLON#8 and POP77 xenografts; cells transduced with only vector were used as controls. Cells were analyzed by real-time quantitative reverse transcription polymerase chain reaction, single-cell gene expression analysis, flow cytometry, and immunohistochemical, immunoblot, and functional assays. Xenograft tumors were grown from control and KIT-knockdown DLD1 and UM-COLON#8 cells in immunocompromised mice and compared. Some mice were given the RTK inhibitor imatinib after injection of cancer cells; tumor growth was measured based on bioluminescence. We assessed tumorigenicity using limiting dilution analysis. RESULTS: KIT and KITLG were expressed heterogeneously by a subset of human colon tumors. Knockdown of KIT decreased proliferation of colon cancer cell lines and growth of xenograft tumors in mice compared with control cells. KIT knockdown cells had increased expression of enterocyte markers, decreased expression of cycling genes, and, unexpectedly, increased expression of LGR5 associated genes. No activating mutations in KIT were detected in DLD1, POP77, or UM-COLON#8 cells. However, KITLG-knockdown DLD1 cells formed smaller xenograft tumors than control cells. Gene expression analysis of single CD44(+) cells indicated that KIT can promote growth via KITLG autocrine and/or paracrine signaling. Imatinib inhibited growth of KIT(+) colon cancer organoids in culture and growth of xenograft tumors in mice. Cancer cells with endogenous KIT expression were more tumorigenic in mice. CONCLUSIONS: KIT and KITLG are expressed by a subset of human colon tumors. KIT signaling promotes growth of colon cancer cells and organoids in culture and xenograft tumors in mice via its ligand, KITLG, in an autocrine or paracrine manner. Patients with KIT-expressing colon tumors can benefit from KIT RTK inhibitors.


Subject(s)
Cell Proliferation , Colonic Neoplasms/enzymology , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Stem Cell Factor/metabolism , Animals , Autocrine Communication , Caco-2 Cells , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Inbred NOD , Paracrine Communication , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , RNA Interference , Signal Transduction/drug effects , Stem Cell Factor/genetics , Time Factors , Transcription, Genetic , Transfection , Tumor Burden , Up-Regulation , Xenograft Model Antitumor Assays
14.
Mod Pathol ; 29(6): 582-90, 2016 06.
Article in English | MEDLINE | ID: mdl-26990975

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas that can show overlapping features with benign neurofibromas as well as high-grade sarcomas. Additional diagnostic markers are needed to aid in this often challenging differential diagnosis. Recently mutations in two critical components of the polycomb repressor 2 (PRC2) complex, SUZ12 and EED, were reported to occur specifically in MPNSTs while such mutations are absent in neurofibromas, both in the setting of neurofibromatosis (NF) and sporadic cases. Furthermore, both SUZ12 and EED mutations in MPNSTs were associated with loss of H3K27 tri-methylation, a downstream target of PRC2. Therefore, we tested whether H3K27me3 immunohistochemistry is useful as a diagnostic and prognostic marker for MPNSTs. We performed H3K27me3 immunohistochemistry in 162 primary MPNSTs, 97 neurofibromas and 341 other tumors using tissue microarray. We observed loss of H3K27me3 in 34% (55/162) of all MPNSTs while expression was retained in all neurofibromas including atypical (n=8) and plexiform subtypes (n=24). Within other tumors we detected loss of H3K27me3 in only 7% (24/341). Surprisingly, 60% (9/15) of synovial sarcomas and 38% (3/8) of fibrosarcomatous dermatofibrosarcoma protuberans (DFSP) showed loss of H3K27 trimethylation. Only 1 out of 44 schwannomas showed loss of H3K27me3 and all 4 perineuriomas showed intact H3K27me3. Furthermore, MPNSTs with loss of H3K27 tri-methylation showed inferior survival compared with MPNSTs with intact H3K27 tri-methylation, which was validated in two independent cohorts. Our results indicate that H3K27me3 immunohistochemistry is useful as a diagnostic marker, in which loss of H3K27me3 favors MPNST above neurofibroma. However, H3K27me3 immunohistochemistry is not suitable to distinguish MPNST from its morphological mimicker synovial sarcoma or fibrosarcomatous DFSP. Since loss of H3K27 tri-methylation was related to poorer survival in MPNST, chromatin modification mediated by this specific histone seems to orchestrate more aggressive tumour biology.


Subject(s)
DNA Methylation , Histones/analysis , Immunohistochemistry , Neurilemmoma/chemistry , Adolescent , Adult , Aged , California , Child , Child, Preschool , Diagnosis, Differential , Female , Humans , Kaplan-Meier Estimate , Lysine , Male , Middle Aged , Netherlands , Neurilemmoma/genetics , Neurilemmoma/mortality , Neurilemmoma/pathology , Predictive Value of Tests , Prognosis , Texas , Time Factors , Tissue Array Analysis , Young Adult
15.
Nature ; 466(7302): 133-7, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20596026

ABSTRACT

The question of whether tumorigenic cancer stem cells exist in human melanomas has arisen in the last few years. Here we show that in melanomas, tumour stem cells (MTSCs, for melanoma tumour stem cells) can be isolated prospectively as a highly enriched CD271(+) MTSC population using a process that maximizes viable cell transplantation. The tumours sampled in this study were taken from a broad spectrum of sites and stages. High-viability cells isolated by fluorescence-activated cell sorting and re-suspended in a matrigel vehicle were implanted into T-, B- and natural-killer-deficient Rag2(-/-)gammac(-/-) mice. The CD271(+) subset of cells was the tumour-initiating population in 90% (nine out of ten) of melanomas tested. Transplantation of isolated CD271(+) melanoma cells into engrafted human skin or bone in Rag2(-/-)gammac(-/-) mice resulted in melanoma; however, melanoma did not develop after transplantation of isolated CD271(-) cells. We also show that in mice, tumours derived from transplanted human CD271(+) melanoma cells were capable of metastatsis in vivo. CD271(+) melanoma cells lacked expression of TYR, MART1 and MAGE in 86%, 69% and 68% of melanoma patients, respectively, which helps to explain why T-cell therapies directed at these antigens usually result in only temporary tumour shrinkage.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Antigens, Neoplasm/analysis , Antigens, Neoplasm/metabolism , Bone Transplantation , Bone and Bones/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , Lung Neoplasms/secondary , Melanoma-Specific Antigens , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/transplantation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Crest/cytology , Neural Crest/pathology , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Skin/pathology , Skin Transplantation , Transplantation, Heterologous/pathology
16.
Proc Natl Acad Sci U S A ; 110(9): 3501-6, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23382202

ABSTRACT

Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract and arises from the interstitial cells of Cajal. It is characterized by expression of the receptor tyrosine kinase CD117 (KIT). In 70-80% of GIST cases, oncogenic mutations in KIT are present, leading to constitutive activation of the receptor, which drives the proliferation of these tumors. Treatment of GIST with imatinib, a small-molecule tyrosine kinase inhibitor, inhibits KIT-mediated signaling and initially results in disease control in 70-85% of patients with KIT-positive GIST. However, the vast majority of patients eventually develop resistance to imatinib treatment, leading to disease progression and posing a significant challenge in the clinical management of these tumors. Here, we show that an anti-KIT monoclonal antibody (mAb), SR1, is able to slow the growth of three human GIST cell lines in vitro. Importantly, these reductions in cell growth were equivalent between imatinib-resistant and imatinib-sensitive GIST cell lines. Treatment of GIST cell lines with SR1 reduces cell-surface KIT expression, suggesting that mAb-induced KIT down-regulation may be a mechanism by which SR1 inhibits GIST growth. Furthermore, we also show that SR1 treatment enhances phagocytosis of GIST cells by macrophages, indicating that treatment with SR1 may enhance immune cell-mediated tumor clearance. Finally, using two xenotransplantation models of imatinib-sensitive and imatinib-resistant GIST, we demonstrate that SR1 is able to strongly inhibit tumor growth in vivo. These results suggest that treatment with mAbs targeting KIT may represent an alternative, or complementary, approach for treating GIST.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Drug Resistance, Neoplasm , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Piperazines/therapeutic use , Proto-Oncogene Proteins c-kit/immunology , Pyrimidines/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Benzamides , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Imatinib Mesylate , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
17.
PLoS Genet ; 9(4): e1003464, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23637631

ABSTRACT

Gene fusions, like BCR/ABL1 in chronic myelogenous leukemia, have long been recognized in hematologic and mesenchymal malignancies. The recent finding of gene fusions in prostate and lung cancers has motivated the search for pathogenic gene fusions in other malignancies. Here, we developed a "breakpoint analysis" pipeline to discover candidate gene fusions by tell-tale transcript level or genomic DNA copy number transitions occurring within genes. Mining data from 974 diverse cancer samples, we identified 198 candidate fusions involving annotated cancer genes. From these, we validated and further characterized novel gene fusions involving ROS1 tyrosine kinase in angiosarcoma (CEP85L/ROS1), SLC1A2 glutamate transporter in colon cancer (APIP/SLC1A2), RAF1 kinase in pancreatic cancer (ATG7/RAF1) and anaplastic astrocytoma (BCL6/RAF1), EWSR1 in melanoma (EWSR1/CREM), CDK6 kinase in T-cell acute lymphoblastic leukemia (FAM133B/CDK6), and CLTC in breast cancer (CLTC/VMP1). Notably, while these fusions involved known cancer genes, all occurred with novel fusion partners and in previously unreported cancer types. Moreover, several constituted druggable targets (including kinases), with therapeutic implications for their respective malignancies. Lastly, breakpoint analysis identified new cell line models for known rearrangements, including EGFRvIII and FIP1L1/PDGFRA. Taken together, we provide a robust approach for gene fusion discovery, and our results highlight a more widespread role of fusion genes in cancer pathogenesis.


Subject(s)
Gene Fusion , Protein-Tyrosine Kinases , Genomics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics
18.
J Biol Chem ; 289(49): 34089-103, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25320080

ABSTRACT

Leiomyosarcoma (LMS) is a mesenchymal cancer that occurs throughout the body. Although LMS is easily recognized histopathologically, the cause of the disease remains unknown. Versican, an extracellular matrix proteoglycan, increases in LMS. Microarray analyses of 80 LMSs and 24 leiomyomas showed a significant elevated expression of versican in human LMS versus benign leiomyomas. To explore the importance of versican in this smooth muscle cell tumor, we used versican-directed siRNA to knock down versican expression in a LMS human cell line, SK-LMS-1. Decreased versican expression was accompanied by slower rates of LMS cell proliferation and migration, increased adhesion, and decreased accumulation of the extracellular matrix macromolecule hyaluronan. Addition of purified versican to cells expressing versican siRNA restored cell proliferation to the level of LMS controls, increased the pericellular coat and the retention of hyaluronan, and decreased cell adhesion in a dose-dependent manner. The presence of versican was not only synergistic with hyaluronan in increasing cell proliferation, but the depletion of versican decreased hyaluronan synthase expression and decreased the retention of hyaluronan. When LMS cells stably expressing versican siRNA were injected into nude mice, the resulting tumors displayed significantly less versican and hyaluronan staining, had lower volumes, and had reduced levels of mitosis as compared with controls. Collectively, these results suggest a role for using versican as a point of control in the management and treatment of LMS.


Subject(s)
Gene Expression Regulation, Neoplastic , Hyaluronic Acid/metabolism , Leiomyosarcoma/genetics , Muscle Neoplasms/genetics , Versicans/genetics , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Profiling , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , Leiomyosarcoma/metabolism , Leiomyosarcoma/pathology , Mice , Mice, Nude , Muscle Neoplasms/metabolism , Muscle Neoplasms/pathology , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tissue Array Analysis , Versicans/antagonists & inhibitors , Versicans/metabolism , Versicans/pharmacology
19.
Histopathology ; 66(5): 627-38, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24889065

ABSTRACT

AIMS: Well-differentiated leiomyosarcomas show morphologically recognizable smooth muscle differentiation, whereas poorly differentiated tumours may form a spectrum with a subset of undifferentiated pleomorphic sarcomas. The expression of certain muscle markers has been reported to have prognostic impact. We investigated the correlation between the morphological spectrum and the muscle marker expression profile of leiomyosarcoma, and the impact of these factors on patient outcomes. METHODS AND RESULTS: Tissue microarrays including 202 non-uterine and 181 uterine leiomyosarcomas with a spectrum of tumour morphologies were evaluated for expression of immunohistochemical markers of muscle differentiation. Poorly differentiated tumours frequently lost one or more conventional smooth muscle markers [smooth muscle actin, desmin, h-caldesmon, and smooth muscle myosin (P < 0.0001)], as well as the more recently described markers SLMAP, MYLK, and ACTG2 (P < 0.0001). In primary tumours, both desmin and CFL2 expression predicted improved overall survival in multivariate analyses (P = 0.0111 and P = 0.043, respectively). Patients with muscle marker-enriched tumours (expressing all four conventional markers or any three of ACTG2, CFL2, CASQ2, MYLK, and SLMAP) had improved overall survival (P < 0.05) in univariate analyses. CONCLUSIONS: Morphologically and immunohistochemically, poorly differentiated leiomyosarcomas can masquerade as undifferentiated pleomorphic sarcomas with progressive loss of muscle markers. The expression of muscle markers has prognostic significance in primary leiomyosarcomas independently of tumour morphology.


Subject(s)
Leiomyosarcoma/diagnosis , Muscle, Smooth/pathology , Retroperitoneal Neoplasms/diagnosis , Uterine Neoplasms/diagnosis , Biomarkers, Tumor/metabolism , Cell Differentiation , Cofilin 2/metabolism , Desmin/metabolism , Female , Humans , Immunohistochemistry , Leiomyosarcoma/classification , Leiomyosarcoma/mortality , Male , Muscle, Smooth/metabolism , Prognosis , Retroperitoneal Neoplasms/classification , Retroperitoneal Neoplasms/mortality , Tissue Array Analysis/methods , Uterine Neoplasms/classification , Uterine Neoplasms/mortality
20.
J Surg Oncol ; 111(7): 808-12, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25920434

ABSTRACT

BACKGROUND: Leiomyosarcoma (LMS) can originate from the retroperitoneum, uterus, extremity, and trunk. It is unclear whether tumors of different origin represent discrete entities. We compared clinicopathologic features and outcomes following surgical resection of LMS stratified by site of origin. METHODS: Patients with LMS undergoing resection at a single institution were retrospectively reviewed. Clinicopathologic variables were compared across sites. Survival was calculated using the Kaplan-Meier method and compared using log-rank and Cox regression analyses. RESULTS: From 1983 to 2011, 138 patients underwent surgical resection for LMS. Retroperitoneal and uterine LMS were larger, higher grade, and more commonly associated with synchronous metastases. However, disease-specific survival, recurrence-free survival, and recurrence patterns were not significantly different across the four sites. Synchronous metastases (HR 3.20, P < 0.001), but not site of origin, size, grade, or margin status, were independently associated with worse DSS. A significant number of recurrences and disease-related deaths were noted beyond 5 years. CONCLUSIONS: Although larger and higher grade, retroperitoneal and uterine LMS share similar survival and recurrence patterns with their trunk and extremity counterparts. LMS of various anatomic sites may not represent distinct disease processes based on clinical outcomes. The presence of metastatic disease remains the most important prognostic factor for LMS.


Subject(s)
Extremities/pathology , Leiomyosarcoma/pathology , Neoplasm Recurrence, Local/pathology , Retroperitoneal Neoplasms/pathology , Uterine Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Extremities/surgery , Female , Follow-Up Studies , Humans , Leiomyosarcoma/mortality , Leiomyosarcoma/surgery , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Prognosis , Retroperitoneal Neoplasms/mortality , Retroperitoneal Neoplasms/surgery , Retrospective Studies , Survival Rate , Uterine Neoplasms/mortality , Uterine Neoplasms/surgery , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL