Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cell ; 156(4): 744-58, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24529377

ABSTRACT

The clinical benefit conferred by vascular endothelial growth factors (VEGF)-targeted therapies is variable, and tumors from treated patients eventually reinitiate growth. Here, we identify a glycosylation-dependent pathway that compensates for the absence of cognate ligand and preserves angiogenesis in response to VEGF blockade. Remodeling of the endothelial cell (EC) surface glycome selectively regulated binding of galectin-1 (Gal1), which upon recognition of complex N-glycans on VEGFR2, activated VEGF-like signaling. Vessels within anti-VEGF-sensitive tumors exhibited high levels of α2-6-linked sialic acid, which prevented Gal1 binding. In contrast, anti-VEGF refractory tumors secreted increased Gal1 and their associated vasculature displayed glycosylation patterns that facilitated Gal1-EC interactions. Interruption of ß1-6GlcNAc branching in ECs or silencing of tumor-derived Gal1 converted refractory into anti-VEGF-sensitive tumors, whereas elimination of α2-6-linked sialic acid conferred resistance to anti-VEGF. Disruption of the Gal1-N-glycan axis promoted vascular remodeling, immune cell influx and tumor growth inhibition. Thus, targeting glycosylation-dependent lectin-receptor interactions may increase the efficacy of anti-VEGF treatment.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic , Vascular Endothelial Growth Factors/antagonists & inhibitors , Animals , Endothelial Cells/metabolism , Galectin 1/genetics , Galectin 1/metabolism , Glycosylation , Humans , Hypoxia , Mice , Receptors, Mitogen/metabolism
2.
Immunity ; 51(1): 119-130.e5, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31231034

ABSTRACT

Tissue-resident macrophages require specific milieus for the maintenance of defining gene-expression programs. Expression of the transcription factor GATA6 is required for the homeostasis, function and localization of peritoneal cavity-resident macrophages. Gata6 expression is maintained in a non-cell autonomous manner and is elicited by the vitamin A metabolite, retinoic acid. Here, we found that the GATA6 transcriptional program is a common feature of macrophages residing in all visceral body cavities. Retinoic acid-dependent and -independent hallmark genes of GATA6+ macrophages were induced by mesothelial and fibroblastic stromal cells that express the transcription factor Wilms' Tumor 1 (WT1), which drives the expression of two rate-limiting enzymes in retinol metabolism. Depletion of Wt1+ stromal cells reduced the frequency of GATA6+ macrophages in the peritoneal, pleural and pericardial cavities. Thus, Wt1+ mesothelial and fibroblastic stromal cells constitute essential niche components supporting the tissue-specifying transcriptional landscape and homeostasis of cavity-resident macrophages.


Subject(s)
GATA6 Transcription Factor/metabolism , Macrophages/physiology , Pericardium/immunology , Peritoneal Cavity/physiology , Pleural Cavity/immunology , Repressor Proteins/metabolism , Stromal Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , GATA6 Transcription Factor/genetics , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Tretinoin/metabolism , WT1 Proteins
3.
Proc Natl Acad Sci U S A ; 114(51): E10947-E10955, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203670

ABSTRACT

KRAS mutant tumors are largely recalcitrant to targeted therapies. Genetically engineered mouse models (GEMMs) of Kras mutant cancer recapitulate critical aspects of this disease and are widely used for preclinical validation of targets and therapies. Through comprehensive profiling of exomes and matched transcriptomes of >200 KrasG12D-initiated GEMM tumors from one lung and two pancreatic cancer models, we discover that significant intratumoral and intertumoral genomic heterogeneity evolves during tumorigenesis. Known oncogenes and tumor suppressor genes, beyond those engineered, are mutated, amplified, and deleted. Unlike human tumors, the GEMM genomic landscapes are dominated by copy number alterations, while protein-altering mutations are rare. However, interspecies comparative analyses of the genomic landscapes demonstrate fidelity between genes altered in KRAS mutant human and murine tumors. Genes that are spontaneously altered during murine tumorigenesis are also among the most prevalent found in human indications. Using targeted therapies, we also demonstrate that this inherent tumor heterogeneity can be exploited preclinically to discover cancer-specific and genotype-specific therapeutic vulnerabilities. Focusing on Kras allelic imbalance, a feature shared by all three models, we discover that MAPK pathway inhibition impinges uniquely on this event, indicating distinct susceptibility and fitness advantage of Kras-mutant cells. These data reveal previously unknown genomic diversity among KrasG12D-initiated GEMM tumors, places them in context of human patients, and demonstrates how to exploit this inherent tumor heterogeneity to discover therapeutic vulnerabilities.


Subject(s)
Genes, ras , Genetic Heterogeneity , Neoplasms/genetics , Neoplasms/pathology , Alleles , Animals , Carcinogenesis/genetics , Cell Line, Tumor , DNA Mutational Analysis , Disease Models, Animal , Gene Expression Profiling , Genomics/methods , Humans , Lung Neoplasms/genetics , MAP Kinase Signaling System , Mice , Mutation , Neoplasms/metabolism , Neoplasms/mortality , Prognosis , Selection, Genetic , Transcriptome
4.
Proc Natl Acad Sci U S A ; 114(15): 3903-3908, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28351980

ABSTRACT

Alveolar type II (AT2) cell dysfunction contributes to a number of significant human pathologies including respiratory distress syndrome, lung adenocarcinoma, and debilitating fibrotic diseases, but the critical transcription factors that maintain AT2 cell identity are unknown. Here we show that the E26 transformation-specific (ETS) family transcription factor Etv5 is essential to maintain AT2 cell identity. Deletion of Etv5 from AT2 cells produced gene and protein signatures characteristic of differentiated alveolar type I (AT1) cells. Consistent with a defect in the AT2 stem cell population, Etv5 deficiency markedly reduced recovery following bleomycin-induced lung injury. Lung tumorigenesis driven by mutant KrasG12D was also compromised by Etv5 deficiency. ERK activation downstream of Ras was found to stabilize Etv5 through inactivation of the cullin-RING ubiquitin ligase CRL4COP1/DET1 that targets Etv5 for proteasomal degradation. These findings identify Etv5 as a critical output of Ras signaling in AT2 cells, contributing to both lung homeostasis and tumor initiation.


Subject(s)
DNA-Binding Proteins/metabolism , Lung Neoplasms/pathology , Pulmonary Alveoli/cytology , Transcription Factors/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Bleomycin , Cell Proliferation , DNA-Binding Proteins/genetics , Gene Expression Regulation , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Mice, Mutant Strains , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Protein Stability , Proto-Oncogene Proteins p21(ras)/genetics , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Nature ; 501(7467): 346-54, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24048067

ABSTRACT

Tumour formation involves the co-evolution of neoplastic cells together with extracellular matrix, tumour vasculature and immune cells. Successful outgrowth of tumours and eventual metastasis is not determined solely by genetic alterations in tumour cells, but also by the fitness advantage such mutations confer in a given environment. As fitness is context dependent, evaluating tumours as complete organs, and not simply as masses of transformed epithelial cells, becomes paramount. The dynamic tumour topography varies drastically even throughout the same lesion. Heterologous cell types within tumours can actively influence therapeutic response and shape resistance.


Subject(s)
Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment , Animals , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Disease Progression , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Neoplasms/blood supply , Neoplasms/immunology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
6.
Nature ; 468(7323): 567-71, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-21107427

ABSTRACT

Non-small cell lung carcinoma (NSCLC) is the leading cause of cancer-related death worldwide, with an overall 5-year survival rate of only 10-15%. Deregulation of the Ras pathway is a frequent hallmark of NSCLC, often through mutations that directly activate Kras. p53 is also frequently inactivated in NSCLC and, because oncogenic Ras can be a potent trigger of p53 (ref. 3), it seems likely that oncogenic Ras signalling has a major and persistent role in driving the selection against p53. Hence, pharmacological restoration of p53 is an appealing therapeutic strategy for treating this disease. Here we model the probable therapeutic impact of p53 restoration in a spontaneously evolving mouse model of NSCLC initiated by sporadic oncogenic activation of endogenous Kras. Surprisingly, p53 restoration failed to induce significant regression of established tumours, although it did result in a significant decrease in the relative proportion of high-grade tumours. This is due to selective activation of p53 only in the more aggressive tumour cells within each tumour. Such selective activation of p53 correlates with marked upregulation in Ras signal intensity and induction of the oncogenic signalling sensor p19(ARF)( )(ref. 6). Our data indicate that p53-mediated tumour suppression is triggered only when oncogenic Ras signal flux exceeds a critical threshold. Importantly, the failure of low-level oncogenic Kras to engage p53 reveals inherent limits in the capacity of p53 to restrain early tumour evolution and in the efficacy of therapeutic p53 restoration to eradicate cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/physiopathology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/physiopathology , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation , Disease Models, Animal , Lung Neoplasms/metabolism , Mice , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/genetics , ras Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 110(15): 6079-84, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23530240

ABSTRACT

Granulocyte-colony stimulating factor (G-CSF) promotes mobilization of CD11b(+)Gr1(+) myeloid cells and has been implicated in resistance to anti-VEGF therapy in mouse models. High G-CSF production has been associated with a poor prognosis in cancer patients. Here we show that activation of the RAS/MEK/ERK pathway regulates G-CSF expression through the Ets transcription factor. Several growth factors induced G-CSF expression by a MEK-dependent mechanism. Inhibition of G-CSF release with a MEK inhibitor markedly reduced G-CSF production in vitro and synergized with anti-VEGF antibodies to reduce CD11b(+)Ly6G(+) neutrophil mobilization and tumor growth and led to increased survival in animal models of cancer, including a genetically engineered mouse model of pancreatic adenocarcinoma. Analysis of biopsies from pancreatic cancer patients revealed increased phospho-MEK, G-CSF, and Ets expression and enhanced neutrophil recruitment compared with normal pancreata. These results provide insights into G-CSF regulation and on the mechanism of action of MEK inhibitors and point to unique anticancer strategies.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , MAP Kinase Signaling System , Neutrophils/cytology , Proto-Oncogene Protein c-ets-2/metabolism , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Binding Sites , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Mice, Transgenic , Neoplasms/metabolism , Neovascularization, Pathologic , Neutrophil Infiltration , Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
8.
Clin Cancer Res ; 30(6): 1111-1120, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38226958

ABSTRACT

PURPOSE: Increased glucocorticoid receptor (GR) signaling is a proposed compensatory mechanism of resistance to androgen receptor (AR) inhibition in metastatic castration-resistant prostate cancer (mCRPC). ORIC-101 is a potent and selective orally-bioavailable GR antagonist. PATIENTS AND METHODS: Safety, pharmacokinetic/pharmacodynamic, and antitumor activity of ORIC-101 in combination with enzalutamide were studied in patients with mCRPC progressing on enzalutamide. ORIC-101 doses ranging from 80 to 240 mg once daily were tested in combination with enzalutamide 160 mg once daily. Pharmacokinetics/pharmacodynamics was assessed after a single dose and at steady state. Disease control rate (DCR) at 12 weeks was evaluated at the recommended phase 2 dose (RP2D). RESULTS: A total of 41 patients were enrolled. There were no dose-limiting toxicities and the RP2D was selected as 240 mg of ORIC-101 and 160 mg of enzalutamide daily. At the RP2D, the most common treatment-related adverse events were fatigue (38.7%), nausea (29.0%), decreased appetite (19.4%), and constipation (12.9%). Pharmacokinetic/pharmacodynamic data confirmed ORIC-101 achieved exposures necessary for GR target engagement. Overall, for 31 patients treated at the RP2D, there was insufficient clinical benefit based on DCR (25.8%; 80% confidence interval: 15.65-38.52) which did not meet the prespecified target rate, leading to termination of the study. Exploratory subgroup analyses based on baseline GR expression, presence of AR resistance variants, and molecular features of aggressive variant prostate cancer suggested possible benefit in patients with high GR expression and no other resistance markers, although this would require confirmation. CONCLUSIONS: Although the combination of ORIC-101 and enzalutamide demonstrated an acceptable tolerability profile, GR target inhibition with ORIC-101 did not produce clinical benefit in men with metastatic prostate cancer resistant to enzalutamide.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Glucocorticoid , Phenylthiohydantoin , Benzamides/therapeutic use , Nitriles/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use
9.
Cancer Res Commun ; 4(9): 2415-2426, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39177285

ABSTRACT

PURPOSE: In preclinical models, glucocorticoid receptor (GR) signaling drives resistance to taxane chemotherapy in multiple solid tumors via upregulation of antiapoptotic pathways. ORIC-101 is a potent and selective GR antagonist that was investigated in combination with taxane chemotherapy as an anticancer regimen preclinically and in a phase 1 clinical trial. PATIENTS AND METHODS: The ability of ORIC-101 to reverse taxane resistance was assessed in cell lines and xenograft models, and a phase 1 study (NCT03928314) was conducted in patients with advanced solid tumors to determine the dose, safety, and antitumor activity of ORIC-101 with nab-paclitaxel. RESULTS: ORIC-101 reversed chemoprotection induced by glucocorticoids in vitro and achieved tumor regressions when combined with paclitaxel in both taxane-naïve and -resistant xenograft models. In the phase 1 study, 21 patients were treated in dose escalation and 62 patients were treated in dose expansion. All patients in dose expansion had previously progressed on a taxane-based regimen. In dose escalation, five objective responses were observed. A preplanned futility analysis in dose expansion showed a 3.2% (95% confidence interval, 0.4-11.2) objective response rate with a median progression-free survival of 2 months (95% confidence interval, 1.8-2.8) across all four cohorts, leading to study termination. Pharmacodynamic analysis of tissue and plasma showed GR pathway downregulation in most patients in cycle 1. CONCLUSIONS: ORIC-101 with nab-paclitaxel showed limited clinical activity in taxane-resistant solid tumors. Despite clear inhibition of GR pathway signaling, the insufficient clinical signal underscores the challenges of targeting a single resistance pathway when multiple mechanisms of resistance may be in play. SIGNIFICANCE: Glucocorticoid receptor (GR) upregulation is a mechanism of resistance to taxane chemotherapy in preclinical cancer models. ORIC-101 is a small molecule GR inhibitor. In this phase 1 study, ORIC-101 plus nab-paclitaxel did not show meaningful clinical benefit in patients who previously progressed on taxanes despite successful GR pathway downregulation.


Subject(s)
Albumins , Antineoplastic Combined Chemotherapy Protocols , Neoplasms , Paclitaxel , Receptors, Glucocorticoid , Humans , Paclitaxel/therapeutic use , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Female , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Male , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aged , Albumins/administration & dosage , Albumins/therapeutic use , Albumins/pharmacology , Animals , Adult , Mice , Xenograft Model Antitumor Assays , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor
10.
Cancer Res Commun ; 3(9): 1788-1799, 2023 09.
Article in English | MEDLINE | ID: mdl-37691854

ABSTRACT

The FOXA1 pioneer factor is an essential mediator of steroid receptor function in multiple hormone-dependent cancers, including breast and prostate cancers, enabling nuclear receptors such as estrogen receptor (ER) and androgen receptor (AR) to activate lineage-specific growth programs. FOXA1 is also highly expressed in non-small cell lung cancer (NSCLC), but whether and how it regulates tumor growth in this context is not known. Analyzing data from loss-of-function screens, we identified a subset of NSCLC tumor lines where proliferation is FOXA1 dependent. Using rapid immunoprecipitation and mass spectrometry of endogenous protein, we identified chromatin-localized interactions between FOXA1 and glucocorticoid receptor (GR) in these tumor cells. Knockdown of GR inhibited proliferation of FOXA1-dependent, but not FOXA1-independent NSCLC cells. In these FOXA1-dependent models, FOXA1 and GR cooperate to regulate gene targets involved in EGF signaling and G1-S cell-cycle progression. To investigate the therapeutic potential for targeting this complex, we examined the effects of highly selective inhibitors of the GR ligand-binding pocket and found that GR antagonism with ORIC-101 suppressed FOXA1/GR target expression, activation of EGF signaling, entry into the S-phase, and attendant proliferation in vitro and in vivo. Taken together, our findings point to a subset of NSCLCs harboring a dependence on the FOXA1/GR growth program and provide rationale for its therapeutic targeting. Significance: NSCLC is the leading cause of cancer deaths worldwide. There is a need to identify novel druggable dependencies. We identify a subset of NSCLCs dependent on FOXA1-GR and sensitive to GR antagonism.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Hepatocyte Nuclear Factor 3-alpha , Lung Neoplasms , Receptors, Glucocorticoid , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Epidermal Growth Factor , Lung Neoplasms/drug therapy , Receptors, Glucocorticoid/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics
11.
Nat Rev Drug Discov ; 21(2): 99-114, 2022 02.
Article in English | MEDLINE | ID: mdl-34702990

ABSTRACT

Recent progress in understanding the molecular basis of cellular processes, identification of promising therapeutic targets and evolution of the regulatory landscape makes this an exciting and unprecedented time to be in the field of oncology drug development. However, high costs, long development timelines and steep rates of attrition continue to afflict the drug development process. Lack of predictive preclinical models is considered one of the key reasons for the high rate of attrition in oncology. Generating meaningful and predictive results preclinically requires a firm grasp of the relevant biological questions and alignment of the model systems that mirror the patient context. In doing so, the ability to conduct both forward translation, the process of implementing basic research discoveries into practice, as well as reverse translation, the process of elucidating the mechanistic basis of clinical observations, greatly enhances our ability to develop effective anticancer treatments. In this Review, we outline issues in preclinical-to-clinical translatability of molecularly targeted cancer therapies, present concepts and examples of successful reverse translation, and highlight the need to better align tumour biology in patients with preclinical model systems including tracking of strengths and weaknesses of preclinical models throughout programme development.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Drug Development , Drug Evaluation, Preclinical/methods , Molecular Targeted Therapy , Neoplasms/drug therapy , Animals , Biomarkers, Tumor/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
12.
iScience ; 25(12): 105712, 2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36582483

ABSTRACT

Here, we have developed an automated image processing algorithm for segmenting lungs and individual lung tumors in in vivo micro-computed tomography (micro-CT) scans of mouse models of non-small cell lung cancer and lung fibrosis. Over 3000 scans acquired across multiple studies were used to train/validate a 3D U-net lung segmentation model and a Support Vector Machine (SVM) classifier to segment individual lung tumors. The U-net lung segmentation algorithm can be used to estimate changes in soft tissue volume within lungs (primarily tumors and blood vessels), whereas the trained SVM is able to discriminate between tumors and blood vessels and identify individual tumors. The trained segmentation algorithms (1) significantly reduce time required for lung and tumor segmentation, (2) reduce bias and error associated with manual image segmentation, and (3) facilitate identification of individual lung tumors and objective assessment of changes in lung and individual tumor volumes under different experimental conditions.

13.
Elife ; 112022 08 19.
Article in English | MEDLINE | ID: mdl-35983994

ABSTRACT

Lung development, integrity and repair rely on precise Wnt signaling, which is corrupted in diverse diseases, including cancer. Here, we discover that EHMT2 methyltransferase regulates Wnt signaling in the lung by controlling the transcriptional activity of chromatin-bound ß-catenin, through a non-histone substrate in mouse lung. Inhibition of EHMT2 induces transcriptional, morphologic, and molecular changes consistent with alveolar type 2 (AT2) lineage commitment. Mechanistically, EHMT2 activity functions to support regenerative properties of KrasG12D tumors and normal AT2 cells-the predominant cell of origin of this cancer. Consequently, EHMT2 inhibition prevents KrasG12D lung adenocarcinoma (LUAD) tumor formation and propagation and disrupts normal AT2 cell differentiation. Consistent with these findings, low gene EHMT2 expression in human LUAD correlates with enhanced AT2 gene expression and improved prognosis. These data reveal EHMT2 as a critical regulator of Wnt signaling, implicating Ehmt2 as a potential target in lung cancer and other AT2-mediated lung pathologies.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/genetics , Animals , Genes, ras , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Methyltransferases/metabolism , Mice , Proto-Oncogene Proteins p21(ras)/metabolism
14.
Cell Rep ; 38(6): 110351, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139374

ABSTRACT

KRAS, which is mutated in ∼30% of all cancers, activates the RAF-MEK-ERK signaling cascade. CRAF is required for growth of KRAS mutant lung tumors, but the requirement for CRAF kinase activity is unknown. Here, we show that subsets of KRAS mutant tumors are dependent on CRAF for growth. Kinase-dead but not dimer-defective CRAF rescues growth inhibition, suggesting that dimerization but not kinase activity is required. Quantitative proteomics demonstrates increased levels of CRAF:ARAF dimers in KRAS mutant cells, and depletion of both CRAF and ARAF rescues the CRAF-loss phenotype. Mechanistically, CRAF depletion causes sustained ERK activation and induction of cell-cycle arrest, while treatment with low-dose MEK or ERK inhibitor rescues the CRAF-loss phenotype. Our studies highlight the role of CRAF in regulating MAPK signal intensity to promote tumorigenesis downstream of mutant KRAS and suggest that disrupting CRAF dimerization or degrading CRAF may have therapeutic benefit.


Subject(s)
Carcinogenesis/metabolism , Dimerization , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Mice , Phosphorylation/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , ras Proteins/genetics
15.
Clin Cancer Res ; 27(4): 1162-1173, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33023953

ABSTRACT

PURPOSE: Lung adenocarcinomas comprise the largest fraction of non-small cell lung cancer, which is the leading cause of cancer-related deaths. Seventy-five percent of adenocarcinomas lack targeted therapies because of scarcity of druggable drivers. Here, we classified tumors on the basis of signaling similarities and discovered subgroups within this unmet patient population. EXPERIMENTAL DESIGN: We leveraged transcriptional data from >800 early- and advanced-stage patients. RESULTS: We identified three robust subtypes dubbed mucinous, proliferative, and mesenchymal with respective pathway phenotypes. These transcriptional states lack discrete and causative mutational etiology as evidenced by similarly distributed oncogenic drivers, including KRAS and EGFR. The subtypes capture heterogeneity even among tumors lacking known oncogenic drivers. Paired multi-regional intratumoral biopsies demonstrated unified subtypes despite divergently evolved prooncogenic mutations, indicating subtype stability during selective pressure. Heterogeneity among in vitro and in vivo preclinical models is expounded by the human lung adenocarcinoma subtypes and can be leveraged to discover subtype-specific vulnerabilities. As proof of concept, we identified differential subtype response to MEK pathway inhibition in a chemical library screen of 89 lung cancer cell lines, which reproduces across model systems and a clinical trial. CONCLUSIONS: Our findings support forward translational relevance of transcriptional subtypes, where further exploration therein may improve lung adenocarcinoma treatment.See related commentary by Skoulidis, p. 913.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Biomarkers, Tumor/genetics , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Cell Line, Tumor , Clinical Trials as Topic , Datasets as Topic , Female , Genetic Heterogeneity , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Neoplasm Staging , Protein Kinase Inhibitors/pharmacology , RNA-Seq , Transcriptome/genetics , Xenograft Model Antitumor Assays
16.
Nat Commun ; 12(1): 6967, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845227

ABSTRACT

Breast cancer is now globally the most frequent cancer and leading cause of women's death. Two thirds of breast cancers express the luminal estrogen receptor-positive (ERα + ) phenotype that is initially responsive to antihormonal therapies, but drug resistance emerges. A major barrier to the understanding of the ERα-pathway biology and therapeutic discoveries is the restricted repertoire of luminal ERα + breast cancer models. The ERα + phenotype is not stable in cultured cells for reasons not fully understood. We examine 400 patient-derived breast epithelial and breast cancer explant cultures (PDECs) grown in various three-dimensional matrix scaffolds, finding that ERα is primarily regulated by the matrix stiffness. Matrix stiffness upregulates the ERα signaling via stress-mediated p38 activation and H3K27me3-mediated epigenetic regulation. The finding that the matrix stiffness is a central cue to the ERα phenotype reveals a mechanobiological component in breast tissue hormonal signaling and enables the development of novel therapeutic interventions. Subject terms: ER-positive (ER + ), breast cancer, ex vivo model, preclinical model, PDEC, stiffness, p38 SAPK.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Mechanotransduction, Cellular/genetics , Transcriptome , p38 Mitogen-Activated Protein Kinases/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Cinnamates/pharmacology , Collagen/chemistry , Collagen/pharmacology , Drug Combinations , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Indazoles/pharmacology , Laminin/chemistry , Laminin/pharmacology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Phenotype , Proteoglycans/chemistry , Proteoglycans/pharmacology , Tamoxifen/pharmacology , Tissue Culture Techniques , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Cancer Discov ; 10(2): 232-253, 2020 02.
Article in English | MEDLINE | ID: mdl-31699795

ABSTRACT

With only a fraction of patients responding to cancer immunotherapy, a better understanding of the entire tumor microenvironment is needed. Using single-cell transcriptomics, we chart the fibroblastic landscape during pancreatic ductal adenocarcinoma (PDAC) progression in animal models. We identify a population of carcinoma-associated fibroblasts (CAF) that are programmed by TGFß and express the leucine-rich repeat containing 15 (LRRC15) protein. These LRRC15+ CAFs surround tumor islets and are absent from normal pancreatic tissue. The presence of LRRC15+ CAFs in human patients was confirmed in >80,000 single cells from 22 patients with PDAC as well as by using IHC on samples from 70 patients. Furthermore, immunotherapy clinical trials comprising more than 600 patients across six cancer types revealed elevated levels of the LRRC15+ CAF signature correlated with poor response to anti-PD-L1 therapy. This work has important implications for targeting nonimmune elements of the tumor microenvironment to boost responses of patients with cancer to immune checkpoint blockade therapy. SIGNIFICANCE: This study describes the single-cell landscape of CAFs in pancreatic cancer during in vivo tumor evolution. A TGFß-driven, LRRC15+ CAF lineage is associated with poor outcome in immunotherapy trial data comprising multiple solid-tumor entities and represents a target for combinatorial therapy.This article is highlighted in the In This Issue feature, p. 161.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm/genetics , Immune Checkpoint Inhibitors/pharmacology , Membrane Proteins/metabolism , Myofibroblasts/immunology , Pancreatic Neoplasms/drug therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Cell Lineage/genetics , Cell Lineage/immunology , Clinical Trials as Topic , Computational Biology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Mice , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , RNA-Seq , Single-Cell Analysis , Transforming Growth Factor beta/metabolism , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
18.
J Med Chem ; 63(18): 10433-10459, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32865411

ABSTRACT

The adenosinergic pathway represents an attractive new therapeutic approach in cancer immunotherapy. In this pathway, ecto-5-nucleotidase CD73 has the unique function of regulating production of immunosuppressive adenosine (ADO) through the hydrolysis of AMP. CD73 is overexpressed in many cancers, resulting in elevated levels of ADO that correspond to poor patient prognosis. Therefore, reducing the level of ADO via inhibition of CD73 is a potential strategy for treating cancers. Based on the binding mode of adenosine 5'-(α,ß-methylene)diphosphate (AOPCP) with human CD73, we designed a series of novel monophosphonate small-molecule CD73 inhibitors. Among them, OP-5244 (35) proved to be a highly potent and orally bioavailable CD73 inhibitor. In preclinical studies, 35 completely inhibited ADO production in both human cancer cells and CD8+ T cells. Furthermore, 35 lowered the ratio of ADO/AMP significantly and reversed immunosuppression in mouse models, indicating its potential as an in vivo tool compound for further development.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Adenosine/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Immunologic Factors/pharmacology , Nucleosides/pharmacology , Organophosphonates/pharmacology , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Dogs , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , GPI-Linked Proteins/antagonists & inhibitors , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacokinetics , Macaca fascicularis , Mice, Inbred BALB C , Molecular Structure , Nucleosides/administration & dosage , Nucleosides/chemical synthesis , Nucleosides/pharmacokinetics , Organophosphonates/administration & dosage , Organophosphonates/chemical synthesis , Organophosphonates/pharmacokinetics , Rats , Stereoisomerism , Structure-Activity Relationship
19.
JCI Insight ; 5(7)2020 04 09.
Article in English | MEDLINE | ID: mdl-32271166

ABSTRACT

Systemic cytokine release and on-target/off-tumor toxicity to normal tissues are the main adverse effects limiting the clinical utility of T cell-redirecting therapies. This study was designed to determine how binding affinity for CD3 and tumor target HER2 impact the efficacy and nonclinical safety of anti-HER2/CD3 T cell-dependent antibodies (TDBs). Affinity was found to be a major determinant for the overall tolerability. Higher affinity for CD3 associated with rapidly elevated peripheral cytokine concentrations, weight loss in mice, and poor tolerability in cynomolgus monkeys. A TDB with lower CD3 affinity was better tolerated in cynomolgus monkeys compared with a higher CD3-affinity TDB. In contrast to tolerability, T cell binding affinity had only limited impact on in vitro and in vivo antitumor activity. High affinity for HER2 was critical for the tumor-killing activity of anti-HER2/CD3 TDBs, but higher HER2 affinity also associated with a more severe toxicity profile, including cytokine release and damage to HER2-expressing tissues. The tolerability of the anti-HER2/CD3 was improved by implementing a dose-fractionation strategy. Fine-tuning the affinities for both the tumor target and CD3 is likely a valuable strategy for achieving maximal therapeutic index of CD3 bispecific antibodies.


Subject(s)
Antibodies, Bispecific/immunology , Antibody Affinity , Antineoplastic Agents, Immunological/immunology , Receptor, ErbB-2/immunology , Animals , Antibodies, Bispecific/chemistry , Antineoplastic Agents, Immunological/chemistry , CD3 Complex/chemistry , CHO Cells , Cricetulus , Drug Evaluation, Preclinical , Humans , Macaca fascicularis , Receptor, ErbB-2/chemistry
20.
Cell Death Differ ; 27(1): 161-175, 2020 01.
Article in English | MEDLINE | ID: mdl-31101885

ABSTRACT

The kinase RIP1 acts in multiple signaling pathways to regulate inflammatory responses and it can trigger both apoptosis and necroptosis. Its kinase activity has been implicated in a range of inflammatory, neurodegenerative, and oncogenic diseases. Here, we explore the effect of inhibiting RIP1 genetically, using knock-in mice that express catalytically inactive RIP1 D138N, or pharmacologically, using the murine-potent inhibitor GNE684. Inhibition of RIP1 reduced collagen antibody-induced arthritis, and prevented skin inflammation caused by mutation of Sharpin, or colitis caused by deletion of Nemo from intestinal epithelial cells. Conversely, inhibition of RIP1 had no effect on tumor growth or survival in pancreatic tumor models driven by mutant Kras, nor did it reduce lung metastases in a B16 melanoma model. Collectively, our data emphasize a role for the kinase activity of RIP1 in certain inflammatory disease models, but question its relevance to tumor progression and metastases.


Subject(s)
Inflammation/enzymology , Neoplasms/enzymology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Arthritis/enzymology , Cell Death , Cell Line , Cell Line, Tumor , Colitis/etiology , Colitis/prevention & control , Dermatitis/enzymology , Female , Gene Knock-In Techniques , Humans , Ileitis/etiology , Ileitis/prevention & control , Intracellular Signaling Peptides and Proteins/genetics , Male , Melanoma, Experimental/pathology , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL