Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Clin Invest ; 131(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34907910

ABSTRACT

BACKGROUNDMEK inhibitors have limited activity in biliary tract cancers (BTCs) as monotherapy but are hypothesized to enhance responses to programmed death ligand 1 (PD-L1) inhibition.METHODSThis open-label phase II study randomized patients with BTC to atezolizumab (anti-PD-L1) as monotherapy or in combination with cobimetinib (MEK inhibitor). Eligible patients had unresectable BTC with 1 to 2 lines of prior therapy in the metastatic setting, measurable disease, and Eastern Cooperative Oncology Group (ECOG) performance status less than or equal to 1. The primary endpoint was progression-free survival (PFS).RESULTSSeventy-seven patients were randomized and received study therapy. The trial met its primary endpoint, with a median PFS of 3.65 months in the combination arm versus 1.87 months in the monotherapy arm (HR 0.58, 90% CI 0.35-0.93, 1-tail P = 0.027). One patient in the combination arm (3.3%) and 1 patient in the monotherapy arm (2.8%) had a partial response. Combination therapy was associated with more rash, gastrointestinal events, CPK elevations, and thrombocytopenia. Exploratory analysis of tumor biopsies revealed enhanced expression of antigen processing and presentation genes and an increase in CD8/FoxP3 ratios with combination treatment. Patients with higher baseline or lower fold changes in expression of certain inhibitory ligands (LAG3, BTLA, VISTA) on circulating T cells had evidence of greater clinical benefit from the combination.CONCLUSIONThe combination of atezolizumab plus cobimetinib prolonged PFS as compared with atezolizumab monotherapy, but the low response rate in both arms highlights the immune-resistant nature of BTCs.TRIAL REGISTRATIONClinicalTrials.gov NCT03201458.FUNDINGNational Cancer Institute (NCI) Experimental Therapeutics Clinical Trials Network (ETCTN); F. Hoffmann-La Roche, Ltd.; NCI, NIH (R01 CA228414-01 and UM1CA186691); NCI's Specialized Program of Research Excellence (SPORE) in Gastrointestinal Cancers (P50 CA062924); NIH Center Core Grant (P30 CA006973); and the Passano Foundation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biliary Tract Neoplasms/drug therapy , Biliary Tract Neoplasms/mortality , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azetidines/administration & dosage , Azetidines/adverse effects , Disease-Free Survival , Female , Humans , Male , Middle Aged , Piperidines/administration & dosage , Piperidines/adverse effects , Progression-Free Survival
2.
Cancer Immunol Res ; 9(10): 1187-1201, 2021 10.
Article in English | MEDLINE | ID: mdl-34389557

ABSTRACT

MEK inhibition (MEKi) is proposed to enhance antitumor immunity but has demonstrated mixed results as an immunomodulatory strategy in human clinical trials. MEKi exerts direct immunomodulatory effects on tumor cells and tumor-infiltrating lymphocytes (TIL), but these effects have not been independently investigated. Here we modeled tumor-specific MEKi through CRISPR/Cas-mediated genome editing of tumor cells [MEK1 knockout (KO)] and pharmacologic MEKi with cobimetinib in a RAS-driven model of colorectal cancer. This approach allowed us to distinguish tumor-mediated and tumor-independent mechanisms of MEKi immunomodulation. MEK1 KO tumors demonstrated upregulation of JAK/STAT signaling, enhanced MHCI expression, CD8+ T-cell infiltration and T-cell activation, and impaired tumor growth that is immune dependent. Pharmacologic MEKi recapitulated tumor-intrinsic effects but simultaneously impaired T-cell activation in the tumor microenvironment. We confirmed a reduction in human peripheral-lymphocyte activation from a clinical trial of anti-PD-L1 (atezolizumab) with or without cobimetinib in biliary tract cancers. Impaired activation of TILs treated with pharmacologic MEKi was reversible and was rescued with the addition of a 4-1BB agonist. Collectively, these data underscore the ability of MEKi to induce context-dependent immunomodulatory effects and suggest that T cell-agonist therapy maximizes the beneficial effects of MEKi on the antitumor immune response.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Immunomodulation/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Protein Kinase Inhibitors/pharmacology , Animals , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
3.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33529175

ABSTRACT

Intratumor heterogeneity is an important mediator of poor outcomes in many cancers, including breast cancer. Genetic subclones frequently contribute to this heterogeneity; however, their growth dynamics and interactions remain poorly understood. PIK3CA and HER2 alterations are known to coexist in breast and other cancers. Herein, we present data that describe the ability of oncogenic PIK3CA mutant cells to induce the proliferation of quiescent HER2 mutant cells through a cell contact-mediated mechanism. Interestingly, the HER2 cells proliferated to become the major subclone over PIK3CA counterparts both in vitro and in vivo. Furthermore, this phenotype was observed in both hormone receptor-positive and -negative cell lines, and was dependent on the expression of fibronectin from mutant PIK3CA cells. Analysis of human tumors demonstrated similar HER2:PIK3CA clonal dynamics and fibronectin expression. Our study provides insight into nonrandom subclonal architecture of heterogenous tumors, which may aid the understanding of tumor evolution and inform future strategies for personalized medicine.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Communication/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Coculture Techniques , Female , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Gene Frequency , Gene Knockout Techniques , Humans , Immunohistochemistry , MCF-7 Cells , Mutation , Phenotype , Receptor, ErbB-2/genetics
4.
Curr Oncol Rep ; 23(2): 23, 2021 02 06.
Article in English | MEDLINE | ID: mdl-33547983

ABSTRACT

PURPOSE OF REVIEW: Mitogen-activated protein kinase (MAPK) kinase (MEK) is an integral component of the RAS signaling pathway, one of the most frequently mutated pathways in cancer biology. MEK inhibitors were initially developed to directly target oncogenic signaling, but are recognized to have pleiotropic effects on both tumor cells and lymphocytes. Here, we review the preclinical and clinical evidence that MEK inhibition is immunomodulatory and discuss the potential rationale for combining MEK inhibitors with systemic immunotherapies. RECENT FINDINGS: MEK inhibition may modulate the tumor microenvironment (TME) through direct effects on both tumor cells and immune cells. Despite encouraging evidence that MEK inhibition can reprogram the tumor microenvironment (TME) and augment anti-tumor immunity regardless of KRAS/BRAF status, recent clinical outcome studies combining MEK inhibition with systemic immunotherapy have yielded mixed results. The combination of MEK inhibitors plus systemic immunotherapies has been tolerable, but has thus far failed to demonstrate clear evidence of synergistic clinical activity. These results underscore the need to understand the appropriate therapeutic context for this combination. MEK inhibitors have the potential to inhibit oncogenic signaling and reprogram the tumor immune microenvironment, representing an attractive therapy to combine with systemic immunotherapies. Ongoing preclinical and clinical studies will further clarify the immunomodulatory effects of MEK inhibitors to inform the design of rational therapeutic combinations.


Subject(s)
Immunotherapy/methods , MAP Kinase Kinase 1/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasms/immunology , Treatment Outcome , Tumor Microenvironment/drug effects
5.
JCI Insight ; 5(10)2020 05 21.
Article in English | MEDLINE | ID: mdl-32434989

ABSTRACT

Immunotherapies that modulate T cell function have been firmly established as a pillar of cancer therapy, whereas the potential for B cells in the antitumor immune response is less established. B cell-activating factor (BAFF) is a B cell-activating cytokine belonging to the TNF ligand family that has been associated with autoimmunity, but little is known about its effects on cancer immunity. We find that BAFF upregulates multiple B cell costimulatory molecules; augments IL-12a expression, consistent with Be-1 lineage commitment; and enhances B cell antigen-presentation to CD4+ Th cells in vitro. In a syngeneic mouse model of melanoma, BAFF upregulates B cell CD40 and PD-L1 expression; it also modulates T cell function through increased T cell activation and TH1 polarization, enhanced expression of the proinflammatory leukocyte trafficking chemokine CCR6, and promotion of a memory phenotype, leading to enhanced antitumor immunity. Similarly, adjuvant BAFF promotes a memory phenotype of T cells in vaccine-draining lymph nodes and augments the antitumor efficacy of whole cell vaccines. BAFF also has distinct immunoregulatory functions, promoting the expansion of CD4+Foxp3+ Tregs in the spleen and tumor microenvironment (TME). Human melanoma data from The Cancer Genome Atlas (TCGA) demonstrate that BAFF expression is positively associated with overall survival and a TH1/IFN-γ gene signature. These data support a potential role for BAFF signaling as a cancer immunotherapy.


Subject(s)
B-Cell Activating Factor/immunology , Immunity, Cellular , Interleukin-12 Subunit p35/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , B-Cell Activating Factor/genetics , Interferon-gamma/immunology , Interleukin-12 Subunit p35/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice
6.
Breast Cancer Res Treat ; 179(3): 631-642, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31823098

ABSTRACT

BACKGROUND/PURPOSE: TrkA overexpression occurs in over 20% of breast cancers, including triple-negative breast cancers (TNBC), and has recently been recognized as a potential driver of carcinogenesis. Recent clinical trials of pan-Trk inhibitors have demonstrated targeted activity against tumors harboring NTRK fusions, a relatively rare alteration across human cancers. Despite this success, current clinical trials have not investigated TrkA overexpression as an additional therapeutic target for pan-Trk inhibitors. Here, we evaluate the cancerous phenotypes of TrkA overexpression relative to NTRK1 fusions in human cells and assess response to pharmacologic Trk inhibition. EXPERIMENTAL DESIGN/METHODS: To evaluate the clinical utility of TrkA overexpression, a panel of TrkA overexpressing cells were developed via stable transfection of an NTRK1 vector into the non-tumorigenic breast cell lines, MCF10A and hTERT-IMEC. A panel of positive controls was generated via stable transfection with a CD74-NTRK1 fusion vector into MCF10A cells. Cells were assessed via various in vitro and in vivo analyses to determine the transformative potential and targetability of TrkA overexpression. RESULTS: TrkA overexpressing cells demonstrated transformative phenotypes similar to Trk fusions, indicating increased oncogenic potential. TrkA overexpressing cells demonstrated growth factor-independent proliferation, increased PI3Kinase and MAPKinase pathway activation, anchorage-independent growth, and increased migratory capacity. These phenotypes were abrogated by the addition of the pan-Trk inhibitor, larotrectinib. In vivo analysis demonstrated increased tumorgenicity and metastatic potential of TrkA overexpressing breast cancer cells. CONCLUSIONS: Herein, we demonstrate TrkA overexpressing cells show increased tumorgenicity and are sensitive to pan-Trk inhibitors. These data suggest that TrkA overexpression may be an additional target for pan-Trk inhibitors and provide a targeted therapy for breast cancer patients.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression , Oncogenes , Receptor, trkA/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
7.
PLoS One ; 14(10): e0224600, 2019.
Article in English | MEDLINE | ID: mdl-31671149

ABSTRACT

Mitogen-activated protein kinase (MAPK) kinase (MEK) is an integral component of the RAS pathway and a therapeutic target in RAS-driven cancers. Although tumor responses to MEK inhibition are rarely durable, MEK inhibitors have shown substantial activity and durable tumor regressions when combined with systemic immunotherapies in preclinical models of RAS-driven tumors. MEK inhibitors have been shown to potentiate anti-tumor T cell immunity, but little is known about the effects of MEK inhibition on other immune subsets, including B cells. We show here that treatment with a MEK inhibitor reduces B regulatory cells (Bregs) in vitro, and reduces the number of Bregs in tumor draining lymph nodes in a colorectal cancer model in vivo. MEK inhibition does not impede anti-tumor humoral immunity, and B cells contribute meaningfully to anti-tumor immunity in the context of MEK inhibitor therapy. Treatment with a MEK inhibitor is associated with improved T cell infiltration and an enhanced response to anti-PD1 immunotherapy. Together these data indicate that MEK inhibition may reduce Bregs while sparing anti-tumor B cell function, resulting in enhanced anti-tumor immunity.


Subject(s)
B-Lymphocytes, Regulatory/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Animals , Azetidines/pharmacology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, ras/drug effects , Humans , Immunotherapy , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Xenograft Model Antitumor Assays
8.
PLoS One ; 14(5): e0217604, 2019.
Article in English | MEDLINE | ID: mdl-31120998

ABSTRACT

C/EBPα plays a key role in specifying myeloid lineage development. HoxA9 is expressed in myeloid progenitors, with its level diminishing during myeloid maturation, and HOXA9 is over-expressed in a majority of acute myeloid leukemia cases, including those expressing NUP98-HOXD13. The objective of this study was to determine whether HoxA9 directly represses Cebpa gene expression. We find 4-fold increased HoxA9 and 5-fold reduced Cebpa in marrow common myeloid and LSK progenitors from Vav-NUP98-HOXD13 transgenic mice. Conversely, HoxA9 decreases 5-fold while Cebpa increases during granulocytic differentiation of 32Dcl3 myeloid cells. Activation of exogenous HoxA9-ER in 32Dcl3 cells reduces Cebpa mRNA even in the presence of cycloheximide, suggesting direct repression. Cebpa transcription in murine myeloid cells is regulated by a hematopoietic-specific +37 kb enhancer and by a more widely active +8 kb enhancer. ChIP-Seq analysis of primary myeloid progenitor cells expressing exogenous HoxA9 or HoxA9-ER demonstrates that HoxA9 localizes to both the +8 kb and +37 kb Cebpa enhancers. Gel shift analysis demonstrates HoxA9 binding to three consensus sites in the +8 kb enhancer, but no affinity for the single near-consensus site present in the +37 kb enhancer. Activity of a Cebpa +8 kb enhancer/promoter-luciferase reporter in 32Dcl3 or MOLM14 myeloid cells is increased ~2-fold by mutation of its three HOXA9-binding sites, suggesting that endogenous HoxA9 represses +8 kb Cebpa enhancer activity. In contrast, mutation of five C/EBPα-binding sites in the +8 kb enhancer reduces activity 3-fold. Finally, expression of a +37 kb enhancer/promoter-hCD4 transgene reporter is reduced ~2-fold in marrow common myeloid progenitors when the Vav-NUP98-HOXD13 transgene is introduced. Overall, these data support the conclusion that HoxA9 represses Cebpa expression, at least in part via inhibition of its +8 kb enhancer, potentially allowing normal myeloid progenitors to maintain immaturity and contributing to the pathogenesis of acute myeloid leukemia associated with increased HOXA9.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid Cells/metabolism , Animals , Binding Sites/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Enhancer Elements, Genetic/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Myeloid Cells/pathology , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myelopoiesis/genetics , Nuclear Pore Complex Proteins/genetics , Proto-Oncogene Proteins c-vav/genetics , Transcription Factors/genetics
9.
Breast Cancer Res Treat ; 174(2): 401-412, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30560461

ABSTRACT

PURPOSE: Estrogen receptor-alpha (ER) is a therapeutic target of ER-positive (ER+) breast cancers. Although ER signaling is complex, many mediators of this pathway have been identified. Specifically, phosphorylation of ER at serine 118 affects responses to estrogen and therapeutic ligands and has been correlated with clinical outcomes in ER+ breast cancer patients. We hypothesized that a newly described germline variant (S118P) at this residue would drive cellular changes consistent with breast cancer development and/or hormone resistance. METHODS: Isogenic human breast epithelial cell line models harboring ER S118P were developed via genome editing and characterized to determine the functional effects of this variant. We also examined the frequency of ER S118P in a case-control study (N = 536) of women with and without breast cancer with a familial risk. RESULTS: In heterozygous knock-in models, the S118P variant demonstrated no significant change in proliferation, migration, MAP Kinase pathway signaling, or response to the endocrine therapies tamoxifen and fulvestrant. Further, there was no difference in the prevalence of S118P between women with and without cancer relative to population registry databases. CONCLUSIONS: This study suggests that the ER S118P variant does not affect risk for breast cancer or hormone therapy resistance. Germline screening and modification of treatments for patients harboring this variant are likely not warranted.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/epidemiology , Estrogen Receptor alpha/genetics , Germ-Line Mutation , Adult , Aged , Breast Neoplasms/genetics , Case-Control Studies , Cell Line, Tumor , Cell Movement , Cell Proliferation , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/therapeutic use , Genetic Variation , Humans , Incidence , MCF-7 Cells , Middle Aged , Phosphorylation , Survival Analysis , Tamoxifen/therapeutic use , Treatment Outcome
10.
Nat Rev Microbiol ; 14(11): 669-675, 2016 11.
Article in English | MEDLINE | ID: mdl-27615028

ABSTRACT

Parasitic unicellular eukaryotes use extracellular vesicles (EVs) as vehicles for intercellular communication and host manipulation. By using various mechanisms to generate EVs and by transferring a wide range of molecules through EVs, pathogenic protozoans are able to establish infective niches, modulate the immune system of the host and cause disease. In addition to effects on the host, EVs are able to transfer virulence factors, drug-resistance genes and differentiation factors between parasites. In this Progress article, we explore recent insights into the biology of EVs from human infectious protozoan parasites, including Trichomonas vaginalis, Plasmodium spp. and kinetoplastids, such as Trypanosoma spp. and Leishmania spp.


Subject(s)
Cell Communication , Extracellular Vesicles/physiology , Parasites/physiology , Animals , Biological Transport , Host-Parasite Interactions , Humans , Leishmania/immunology , Leishmania/pathogenicity , Leishmania/physiology , Parasites/immunology , Parasites/pathogenicity , Plasmodium/immunology , Plasmodium/pathogenicity , Plasmodium/physiology , Trichomonas vaginalis/immunology , Trichomonas vaginalis/pathogenicity , Trichomonas vaginalis/physiology , Trypanosoma/immunology , Trypanosoma/pathogenicity , Trypanosoma/physiology , Virulence Factors/physiology
11.
Cell ; 164(1-2): 246-257, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26771494

ABSTRACT

Intercellular communication between parasites and with host cells provides mechanisms for parasite development, immune evasion, and disease pathology. Bloodstream African trypanosomes produce membranous nanotubes that originate from the flagellar membrane and disassociate into free extracellular vesicles (EVs). Trypanosome EVs contain several flagellar proteins that contribute to virulence, and Trypanosoma brucei rhodesiense EVs contain the serum resistance-associated protein (SRA) necessary for human infectivity. T. b. rhodesiense EVs transfer SRA to non-human infectious trypanosomes, allowing evasion of human innate immunity. Trypanosome EVs can also fuse with mammalian erythrocytes, resulting in rapid erythrocyte clearance and anemia. These data indicate that trypanosome EVs are organelles mediating non-hereditary virulence factor transfer and causing host erythrocyte remodeling, inducing anemia.


Subject(s)
Extracellular Vesicles/metabolism , Membrane Glycoproteins/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei rhodesiense/cytology , Trypanosoma brucei rhodesiense/immunology , Trypanosomiasis, African/pathology , Trypanosomiasis, African/parasitology , Virulence Factors/metabolism , Anemia/pathology , Animals , Erythrocytes/parasitology , Flagella/metabolism , Humans , Immune Evasion , Mice , Proteome/metabolism , Rhodamines/analysis , Trypanosoma brucei rhodesiense/metabolism , Trypanosoma brucei rhodesiense/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...