Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Cell ; 184(11): 2988-3005.e16, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34019793

ABSTRACT

Clear cell renal carcinoma (ccRCC) is a heterogeneous disease with a variable post-surgical course. To assemble a comprehensive ccRCC tumor microenvironment (TME) atlas, we performed single-cell RNA sequencing (scRNA-seq) of hematopoietic and non-hematopoietic subpopulations from tumor and tumor-adjacent tissue of treatment-naive ccRCC resections. We leveraged the VIPER algorithm to quantitate single-cell protein activity and validated this approach by comparison to flow cytometry. The analysis identified key TME subpopulations, as well as their master regulators and candidate cell-cell interactions, revealing clinically relevant populations, undetectable by gene-expression analysis. Specifically, we uncovered a tumor-specific macrophage subpopulation characterized by upregulation of TREM2/APOE/C1Q, validated by spatially resolved, quantitative multispectral immunofluorescence. In a large clinical validation cohort, these markers were significantly enriched in tumors from patients who recurred following surgery. The study thus identifies TREM2/APOE/C1Q-positive macrophage infiltration as a potential prognostic biomarker for ccRCC recurrence, as well as a candidate therapeutic target.


Subject(s)
Carcinoma, Renal Cell/metabolism , Neoplasm Recurrence, Local/genetics , Tumor-Associated Macrophages/metabolism , Adult , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cohort Studies , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Kidney/metabolism , Kidney Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Middle Aged , Neoplasm Recurrence, Local/metabolism , Prognosis , Receptors, Complement/genetics , Receptors, Complement/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Tumor Microenvironment , Tumor-Associated Macrophages/physiology
2.
Nat Cancer ; 2(8): 803-818, 2021 08.
Article in English | MEDLINE | ID: mdl-35122025

ABSTRACT

Unlike several other tumor types, prostate cancer rarely responds to immune checkpoint blockade (ICB). To define tumor cell intrinsic factors that contribute to prostate cancer progression and resistance to ICB, we analyzed prostate cancer epithelial cells from castration-sensitive and -resistant samples using implanted tumors, cell lines, transgenic models and human tissue. We found that castration resulted in increased expression of interleukin-8 (IL-8) and its probable murine homolog Cxcl15 in prostate epithelial cells. We showed that these chemokines drove subsequent intratumoral infiltration of tumor-promoting polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), which was largely abrogated when IL-8 signaling was blocked genetically or pharmacologically. Targeting IL-8 signaling in combination with ICB delayed the onset of castration resistance and increased the density of polyfunctional CD8 T cells in tumors. Our findings establish a novel mechanism by which castration mediates IL-8 secretion and subsequent PMN-MDSC infiltration, and highlight blockade of the IL-8/CXCR2 axis as a potential therapeutic intervention.


Subject(s)
Myeloid-Derived Suppressor Cells , Prostatic Neoplasms , Animals , Castration , Humans , Interleukin-8/genetics , Male , Mice , Prostate , Prostatic Neoplasms/genetics
3.
Clin Cancer Res ; 27(2): 608-621, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33148676

ABSTRACT

PURPOSE: Intratumoral immunosuppression mediated by myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) represents a potential mechanism of immune checkpoint inhibitor (ICI) resistance in solid tumors. By promoting TAM and MDSC infiltration, IL1ß may drive adaptive and innate immune resistance in renal cell carcinoma (RCC) and in other tumor types. EXPERIMENTAL DESIGN: Using the RENCA model of RCC, we evaluated clinically relevant combinations of anti-IL1ß plus either anti-PD-1 or the multitargeted tyrosine kinase inhibitor (TKI), cabozantinib. We performed comprehensive immune profiling of established RENCA tumors via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA sequencing (RNA-seq). Similar analyses were extended to the MC38 tumor model. RESULTS: Analyses via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA-seq showed that anti-IL1ß reduces infiltration of polymorphonuclear MDSCs and TAMs. Combination treatment with anti-IL1ß plus anti-PD-1 or cabozantinib showed increased antitumor activity that was associated with decreases in immunosuppressive MDSCs and increases in M1-like TAMs. CONCLUSIONS: Single-cell RNA-seq analyses show that IL1ß blockade and ICI or TKI remodel the myeloid compartment through nonredundant, relatively T-cell-independent mechanisms. IL1ß is an upstream mediator of adaptive myeloid resistance and represents a potential target for kidney cancer immunotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Renal Cell/drug therapy , Disease Models, Animal , Interleukin-1beta/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Anilides/administration & dosage , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Checkpoint Inhibitors/administration & dosage , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/metabolism , Pyridines/administration & dosage , RNA-Seq/methods , Single-Cell Analysis/methods , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor-Associated Macrophages/classification , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism
4.
Urol Clin North Am ; 47(4): 419-431, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33008493

ABSTRACT

The management of metastatic renal cell carcinoma (RCC) has evolved rapidly in recent years with several immunotherapy-based combinations of strategies approved as first-line therapies. Targeted strategies, including systemic antiangiogenesis agents and immune checkpoint blockade, form the basis of a therapeutic approach. With rising rates of recurrence after first-line treatment, it is increasingly important to not only adopt a personalized treatment plan with minimal adverse events but also develop predictive biomarkers for response. This review discusses currently available first-line and second-line therapies in RCC and their pivotal data, with specific focus on ongoing clinical trials in the adjuvant setting, including those involving novel agents.


Subject(s)
Biological Products/therapeutic use , Carcinoma, Renal Cell/drug therapy , Immunotherapy/methods , Kidney Neoplasms/drug therapy , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/surgery , Clinical Trials, Phase II as Topic , Combined Modality Therapy , Drug Therapy, Combination , Female , Humans , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Neoplasm Invasiveness/pathology , Neoplasm Staging , Nephrectomy/methods , Prognosis , Survival Analysis , Treatment Outcome , United States
5.
J Clin Oncol ; 34(36): 4390-4397, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27998227

ABSTRACT

Purpose Gene expression profiling assays are frequently used to guide adjuvant chemotherapy decisions in hormone receptor-positive, lymph node-negative breast cancer. We hypothesized that the clinical value of these new tools would be more fully realized when appropriately integrated with high-quality clinicopathologic data. Hence, we developed a model that uses routine pathologic parameters to estimate Oncotype DX recurrence score (ODX RS) and independently tested its ability to predict ODX RS in clinical samples. Patients and Methods We retrospectively reviewed ordered ODX RS and pathology reports from five institutions (n = 1,113) between 2006 and 2013. We used locally performed histopathologic markers (estrogen receptor, progesterone receptor, Ki-67, human epidermal growth factor receptor 2, and Elston grade) to develop models that predict RS-based risk categories. Ordering patterns at one site were evaluated under an integrated decision-making model incorporating clinical treatment guidelines, immunohistochemistry markers, and ODX. Final locked models were independently tested (n = 472). Results Distribution of RS was similar across sites and to reported clinical practice experience and stable over time. Histopathologic markers alone determined risk category with > 95% confidence in > 55% (616 of 1,113) of cases. Application of the integrated decision model to one site indicated that the frequency of testing would not have changed overall, although ordering patterns would have changed substantially with less testing of estimated clinical risk-high or clinical risk-low cases and more testing of clinical risk-intermediate cases. In the validation set, the model correctly predicted risk category in 52.5% (248 of 472). Conclusion The proposed model accurately predicts high- and low-risk RS categories (> 25 or ≤ 25) in a majority of cases. Integrating histopathologic and molecular information into the decision-making process allows refocusing the use of new molecular tools to cases with uncertain risk.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Profiling/statistics & numerical data , Adult , Aged , Breast Neoplasms/therapy , Cohort Studies , Female , Humans , Immunohistochemistry , Ki-67 Antigen/genetics , Linear Models , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis , Receptor, ErbB-2/genetics , Receptors, Progesterone/genetics , Retrospective Studies
6.
Mol Genet Metab ; 111(4): 522-532, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24503136

ABSTRACT

Zellweger spectrum disorder (ZSD) is a disease continuum that results from inherited defects in PEX genes essential for normal peroxisome assembly. These autosomal recessive disorders impact brain development and also cause postnatal liver, adrenal, and kidney dysfunction, as well as loss of vision and hearing. The hypomorphic PEX1-G843D missense allele, observed in approximately 30% of ZSD patients, is associated with milder clinical and biochemical phenotypes, with some homozygous individuals surviving into early adulthood. Nonetheless, affected children with the PEX1-G843D allele have intellectual disability, failure to thrive, and significant sensory deficits. To enhance our ability to test candidate therapies that improve human PEX1-G843D function, we created the novel Pex1-G844D knock-in mouse model that represents the murine equivalent of the common human mutation. We show that Pex1-G844D homozygous mice recapitulate many classic features of mild ZSD cases, including growth retardation and fatty livers with cholestasis. In addition, electrophysiology, histology, and gene expression studies provide evidence that these animals develop a retinopathy similar to that observed in human patients, with evidence of cone photoreceptor cell death. Similar to skin fibroblasts obtained from ZSD patients with a PEX1-G843D allele, we demonstrate that murine cells homozygous for the Pex1-G844D allele respond to chaperone-like compounds, which normalizes peroxisomal ß-oxidation. Thus, the Pex1-G844D mouse provides a powerful model system for testing candidate therapies that address the most common genetic cause of ZSD. In addition, this murine model will enhance studies focused on mechanisms of pathogenesis.


Subject(s)
Adenosine Triphosphatases/genetics , Disease Models, Animal , Mutation, Missense/genetics , Zellweger Syndrome/pathology , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , Animals , Animals, Newborn , Bile Acids and Salts/metabolism , Fatty Acids/blood , Female , Fibroblasts/metabolism , Gene Expression Profiling , Growth and Development , Hearing , Heterozygote , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Molecular Chaperones/metabolism , Phenotype , Retina/pathology , Retina/physiopathology , Sexual Behavior, Animal , Skin/pathology , Survival Analysis , Vision, Ocular , Zellweger Syndrome/blood , Zellweger Syndrome/genetics , Zellweger Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL