Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Cancer Res ; 84(7): 1115-1132, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38294344

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease that lacks effective treatment options, highlighting the need for developing new therapeutic interventions. Here, we assessed the response to pharmacologic inhibition of KRAS, the central oncogenic driver of PDAC. In a panel of PDAC cell lines, inhibition of KRASG12D with MRTX1133 yielded variable efficacy in suppressing cell growth and downstream gene expression programs in 2D cultures. On the basis of CRISPR-Cas9 loss-of-function screens, ITGB1 was identified as a target to enhance the therapeutic response to MRTX1133 by regulating mechanotransduction signaling and YAP/TAZ expression, which was confirmed by gene-specific knockdown and combinatorial drug synergy. Interestingly, MRTX1133 was considerably more efficacious in 3D cell cultures. Moreover, MRTX1133 elicited a pronounced cytostatic effect in vivo and controlled tumor growth in PDAC patient-derived xenografts. In syngeneic models, KRASG12D inhibition led to tumor regression that did not occur in immune-deficient hosts. Digital spatial profiling on tumor tissues indicated that MRTX1133-mediated KRAS inhibition enhanced IFNγ signaling and induced antigen presentation that modulated the tumor microenvironment. Further investigation of the immunologic response using single-cell sequencing and multispectral imaging revealed that tumor regression was associated with suppression of neutrophils and influx of effector CD8+ T cells. Together, these findings demonstrate that both tumor cell-intrinsic and -extrinsic events contribute to response to MRTX1133 and credential KRASG12D inhibition as a promising therapeutic strategy for a large percentage of patients with PDAC. SIGNIFICANCE: Pharmacologic inhibition of KRAS elicits varied responses in pancreatic cancer 2D cell lines, 3D organoid cultures, and xenografts, underscoring the importance of mechanotransduction and the tumor microenvironment in regulating therapeutic responses.


Carcinoma, Pancreatic Ductal , Heterocyclic Compounds, 2-Ring , Naphthalenes , Pancreatic Neoplasms , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Microenvironment , Mechanotransduction, Cellular , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor
2.
Int J Mol Sci ; 24(13)2023 Jun 29.
Article En | MEDLINE | ID: mdl-37446025

The prevalence of obesity, defined as the body mass index (BMI) ≥ 30 kg/m2, has reached epidemic levels. Obesity is associated with an increased risk of various cancers, including gastrointestinal ones. Recent evidence has suggested that obesity disproportionately impacts males and females with cancer, resulting in varied transcriptional and metabolic dysregulation. This study aimed to elucidate the differences in the metabolic milieu of adenocarcinomas of the gastrointestinal (GI) tract both related and unrelated to sex in obesity. To demonstrate these obesity and sex-related effects, we utilized three primary data sources: serum metabolomics from obese and non-obese patients assessed via the Biocrates MxP Quant 500 mass spectrometry-based kit, the ORIEN tumor RNA-sequencing data for all adenocarcinoma cases to assess the impacts of obesity, and publicly available TCGA transcriptional analysis to assess GI cancers and sex-related differences in GI cancers specifically. We applied and integrated our unique transcriptional metabolic pipeline in combination with our metabolomics data to reveal how obesity and sex can dictate differential metabolism in patients. Differentially expressed genes (DEG) analysis of ORIEN obese adenocarcinoma as compared to normal-weight adenocarcinoma patients resulted in large-scale transcriptional reprogramming (4029 DEGs, adj. p < 0.05 and |logFC| > 0.58). Gene Set Enrichment and metabolic pipeline analysis showed genes enriched for pathways relating to immunity (inflammation, and CD40 signaling, among others) and metabolism. Specifically, we found alterations to steroid metabolism and tryptophan/kynurenine metabolism in obese patients, both of which are highly associated with disease severity and immune cell dysfunction. These findings were further confirmed using the TCGA colorectal adenocarcinoma (CRC) and esophageal adenocarcinoma (ESCA) data, which showed similar patterns of increased tryptophan catabolism for kynurenine production in obese patients. These patients further showed disparate alterations between males and females when comparing obese to non-obese patient populations. Alterations to immune and metabolic pathways were validated in six patients (two obese and four normal weight) via CD8+/CD4+ peripheral blood mononuclear cell RNA-sequencing and paired serum metabolomics, which showed differential kynurenine and lipid metabolism, which corresponded with altered T-cell transcriptome in obese populations. Overall, obesity is associated with differential transcriptional and metabolic programs in various disease sites. Further, these alterations, such as kynurenine and tryptophan metabolism, which impact both metabolism and immune phenotype, vary with sex and obesity together. This study warrants further in-depth investigation into obesity and sex-related alterations in cancers that may better define biomarkers of response to immunotherapy.


Adenocarcinoma , Gastrointestinal Neoplasms , Male , Female , Humans , Kynurenine , Tryptophan , Leukocytes, Mononuclear , Obesity/genetics , Gastrointestinal Neoplasms/genetics
3.
bioRxiv ; 2023 Apr 20.
Article En | MEDLINE | ID: mdl-37162905

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease for which new therapeutic interventions are needed. Here we assessed the cellular response to pharmacological KRAS inhibition, which target the central oncogenic factor in PDAC. In a panel of PDAC cell lines, pharmaceutical inhibition of KRAS G12D allele, with MRTX1133 yields variable efficacy in the suppression of cell growth and downstream gene expression programs in 2D culture. CRISPR screens identify new drivers for enhanced therapeutic response that regulate focal adhesion and signaling cascades, which were confirmed by gene specific knockdowns and combinatorial drug synergy. Interestingly, MRTX1133 is considerably more efficacious in the context of 3D cell cultures and in vivo PDAC patient-derived xenografts. In syngeneic models, KRAS G12D inhibition elicits potent tumor regression that did not occur in immune-deficient hosts. Digital spatial profiling on tumor tissues indicates that MRTX1133 activates interferon-γ signaling and induces antigen presentation that modulate the tumor microenvironment. Further investigation on the immunological response using single cell sequencing and multispectral imaging reveals that tumor regression is associated with suppression of neutrophils and influx of effector CD8 + T-cells. Thus, both tumor cell intrinsic and extrinsic events contribute to response and credential KRAS G12D inhibition as promising strategy for a large percentage of PDAC tumors.

4.
Nat Cancer ; 4(1): 62-80, 2023 01.
Article En | MEDLINE | ID: mdl-36585453

Pancreatic ductal adenocarcinoma (PDAC) is considered non-immunogenic, with trials showing its recalcitrance to PD1 and CTLA4 immune checkpoint therapies (ICTs). Here, we sought to systematically characterize the mechanisms underlying de novo ICT resistance and to identify effective therapeutic options for PDAC. We report that agonist 41BB and antagonist LAG3 ICT alone and in combination, increased survival and antitumor immunity, characterized by modulating T cell subsets with antitumor activity, increased T cell clonality and diversification, decreased immunosuppressive myeloid cells and increased antigen presentation/decreased immunosuppressive capability of myeloid cells. Translational analyses confirmed the expression of 41BB and LAG3 in human PDAC. Since single and dual ICTs were not curative, T cell-activating ICTs were combined with a CXCR1/2 inhibitor targeting immunosuppressive myeloid cells. Triple therapy resulted in durable complete responses. Given similar profiles in human PDAC and the availability of these agents for clinical testing, our findings provide a testable hypothesis for this lethal disease.


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Carcinoma, Pancreatic Ductal/drug therapy , Myeloid Cells/pathology , Pancreatic Neoplasms/drug therapy , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Receptors, Interleukin-8A/immunology , Pancreatic Neoplasms
5.
STAR Protoc ; 3(3): 101644, 2022 09 16.
Article En | MEDLINE | ID: mdl-36052343

Intratumor and gut mycobiome is linked to pancreatic ductal adenocarcinoma (PDAC) tumorigenesis; however, an optimal approach to culture and transplant fungus into mouse for in vivo studies is missing. This protocol describes culture steps of Alternaria alternata and Malassezia globosa and their subsequent transplantation into a PDAC mouse model via oral gavage. The utilization of the fungal culture method will allow for consistent growth and expansion of specific fungal species for downstream processing. For complete details on the use and execution of this protocol, please refer to Alam et al. (2022).


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/surgery , Mice , Pancreatic Ducts/pathology , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms
6.
STAR Protoc ; 3(3): 101563, 2022 09 16.
Article En | MEDLINE | ID: mdl-35874471

Innate lymphoid cells 2 (ILC2) play a significant role in the tumorigenesis of pancreatic ductal adenocarcinoma (PDAC). An important aspect of ILC2-mediated tumorigenesis is the expansion of the resident ILC2 and simultaneous recruitment of the peripheral ILC2. Here, we describe a protocol for isolation, enrichment, and DiD labeling of ILC2 for in vivo tracking of ILC2s in the mouse. Further, we describe steps for the adoptive transfer of ILC2 to a recipient mouse model of PDAC. For complete details on the use and execution of this protocol, please refer to Alam et al. (2022).


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Adoptive Transfer , Animals , Carcinogenesis , Carcinoma, Pancreatic Ductal/therapy , Disease Models, Animal , Immunity, Innate , Lymphocytes , Mice
7.
Oncogene ; 41(31): 3821-3829, 2022 07.
Article En | MEDLINE | ID: mdl-35817840

Activated oncogenes and disrupted tumor suppressor genes (TSGs) not only endow aspiring cancer cells with new biological capabilities but also influence the composition and function of host cells in the tumor microenvironment (TME). These non-cancer host cells can in turn provide cancer cells with growth support and protection from the anti-tumor immune response. In this ecosystem, geospatially heterogenous "subTME" adds to the complexity of the "global" TME which bestows tumors with increased tumorigenic ability and resistance to therapy. This review highlights how specific genetic alterations in cancer cells establish various symbiotic co-dependencies with surrounding host cells and details the cooperative role of the host cells in tumor biology. These essential interactions expand the repertoire of targets for the development of precision cancer treatments.


Neoplasms , Tumor Microenvironment , Carcinogenesis/genetics , Ecosystem , Humans , Neoplasms/pathology , Oncogenes , Tumor Microenvironment/genetics
8.
Cancer Cell ; 40(2): 153-167.e11, 2022 02 14.
Article En | MEDLINE | ID: mdl-35120601

TH2 cells and innate lymphoid cells 2 (ILC2) can stimulate tumor growth by secreting pro-tumorigenic cytokines such as interleukin-4 (IL-4), IL-5, and IL-13. However, the mechanisms by which type 2 immune cells traffic to the tumor microenvironment are unknown. Here, we show that oncogenic KrasG12D increases IL-33 expression in pancreatic ductal adenocarcinoma (PDAC) cells, which recruits and activates TH2 and ILC2 cells. Correspondingly, cancer-cell-specific deletion of IL-33 reduces TH2 and ILC2 recruitment and promotes tumor regression. Unexpectedly, IL-33 secretion is dependent on the intratumoral fungal mycobiome. Genetic deletion of IL-33 or anti-fungal treatment decreases TH2 and ILC2 infiltration and increases survival. Consistently, high IL-33 expression is observed in approximately 20% of human PDAC, and expression is mainly restricted to cancer cells. These data expand our knowledge of the mechanisms driving PDAC tumor progression and identify therapeutically targetable pathways involving intratumoral mycobiome-driven secretion of IL-33.


Immunity, Innate , Interleukin-33/biosynthesis , Mycobiome , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , Disease Models, Animal , Disease Progression , Disease Susceptibility , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Models, Biological , Mycobiome/immunology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Pancreatic Neoplasms
9.
Phys Fluids (1994) ; 33(11): 113310, 2021 Nov.
Article En | MEDLINE | ID: mdl-34803363

We have performed a three-dimensional numerical simulation to determine the effect of local atmospheric pollution level on the spreading characteristics of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus through ejected droplets during sneezing and coughing in an open space. Utilizing a finite volume-based numerical method, we have performed computations for various ranges of droplet diameters and sneezing speeds. The interactions between the droplets and the suspended particles are considered by taking both hydrophobic and hydrophilic wettability characteristics into account. Our computational results show that the virus-containing droplets partially affect aerosols during the path of their transmission. With the progression of time, the droplet distribution shows an asymmetric pattern. The maximum dispersion of these droplets is found for higher sneezing velocities. The droplets with a diameter of 50 µm travel a larger distance than the larger diameter droplets. We have found that an aerosol with hydrophilic wettability undergoes complete wetting by the disease-containing droplets and therefore is conducive to disease propagation. The droplet engagement duration with aerosol decreases with increase in the sneezing velocity. Our study recommends against using physical exercise centers in a closed environment such as gymnasium and indoor games during the COVID pandemic, especially in a polluted environment. The results from our work will help in deciding proper social distancing guidelines based on the local atmospheric pollution level. They may act as a precursor in controlling further spread of diseases during this unprecedented situation of the COVID pandemic.

10.
Science ; 373(6561): eabj0486, 2021 Sep 17.
Article En | MEDLINE | ID: mdl-34529467

Inflammation is a major risk factor for pancreatic ductal adenocarcinoma (PDAC). When occurring in the context of pancreatitis, KRAS mutations accelerate tumor development in mouse models. We report that long after its complete resolution, a transient inflammatory event primes pancreatic epithelial cells to subsequent transformation by oncogenic KRAS. Upon recovery from acute inflammation, pancreatic epithelial cells display an enduring adaptive response associated with sustained transcriptional and epigenetic reprogramming. Such adaptation enables the reactivation of acinar-to-ductal metaplasia (ADM) upon subsequent inflammatory events, thereby limiting tissue damage through a rapid decrease of zymogen production. We propose that because activating mutations of KRAS maintain an irreversible ADM, they may be beneficial and under strong positive selection in the context of recurrent pancreatitis.


Acinar Cells/pathology , Carcinogenesis , Carcinoma, Pancreatic Ductal/pathology , Genes, ras , Pancreas/pathology , Pancreatitis/physiopathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/physiopathology , Cell Transformation, Neoplastic , Cells, Cultured , Cellular Reprogramming , Chromatin/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Enzyme Precursors/metabolism , Epigenesis, Genetic , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , MAP Kinase Signaling System , Male , Metaplasia , Mice , Mutation , Pancreas/metabolism , Pancreatitis/genetics , Pancreatitis/immunology , Spheroids, Cellular , Transcriptome
11.
Cancer Discov ; 11(5): 1067-1081, 2021 05.
Article En | MEDLINE | ID: mdl-33504580

Metabolic reprogramming enables cancer cell growth, proliferation, and survival. This reprogramming is driven by the combined actions of oncogenic alterations in cancer cells and host cell factors acting on cancer cells in the tumor microenvironment. Cancer cell-intrinsic mechanisms activate signal transduction components that either directly enhance metabolic enzyme activity or upregulate transcription factors that in turn increase expression of metabolic regulators. Extrinsic signaling mechanisms involve host-derived factors that further promote and amplify metabolic reprogramming in cancer cells. This review describes intrinsic and extrinsic mechanisms driving cancer metabolism in the tumor microenvironment and how such mechanisms may be targeted therapeutically. SIGNIFICANCE: Cancer cell metabolic reprogramming is a consequence of the converging signals originating from both intrinsic and extrinsic factors. Intrinsic signaling maintains the baseline metabolic state, whereas extrinsic signals fine-tune the metabolic processes based on the availability of metabolites and the requirements of the cells. Therefore, successful targeting of metabolic pathways will require a nuanced approach based on the cancer's genotype, tumor microenvironment composition, and tissue location.


Cell Transformation, Neoplastic , Neoplasms/metabolism , Tumor Microenvironment , Humans , Metabolic Networks and Pathways , Neoplasms/pathology
12.
Nat Commun ; 11(1): 4766, 2020 09 21.
Article En | MEDLINE | ID: mdl-32958778

Germline telomere maintenance defects are associated with an increased incidence of inflammatory diseases in humans, yet whether and how telomere dysfunction causes inflammation are not known. Here, we show that telomere dysfunction drives pATM/c-ABL-mediated activation of the YAP1 transcription factor, up-regulating the major pro-inflammatory factor, pro-IL-18. The colonic microbiome stimulates cytosolic receptors activating caspase-1 which cleaves pro-IL-18 into mature IL-18, leading to recruitment of interferon (IFN)-γ-secreting T cells and intestinal inflammation. Correspondingly, patients with germline telomere maintenance defects exhibit DNA damage (γH2AX) signaling together with elevated YAP1 and IL-18 expression. In mice with telomere dysfunction, telomerase reactivation in the intestinal epithelium or pharmacological inhibition of ATM, YAP1, or caspase-1 as well as antibiotic treatment, dramatically reduces IL-18 and intestinal inflammation. Thus, telomere dysfunction-induced activation of the ATM-YAP1-pro-IL-18 pathway in epithelium is a key instigator of tissue inflammation.


Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Inflammation/pathology , Telomere/pathology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Anti-Bacterial Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Caspase 1/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Child , Colon/metabolism , Colon/microbiology , Colon/pathology , Gastrointestinal Diseases/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/microbiology , Interleukin-18/genetics , Interleukin-18/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Mutant Strains , Phosphorylation , Protein Precursors/genetics , Protein Precursors/metabolism , Signal Transduction , Telomerase/genetics , Telomerase/metabolism , YAP-Signaling Proteins
13.
Langmuir ; 36(28): 8111-8122, 2020 Jul 21.
Article En | MEDLINE | ID: mdl-32589848

Surface nanostructure, either designed or generated as an artifact of the fabrication procedure, is known to influence interfacial phenomena intriguingly. While surface roughness-wettability coupling over nanometer scales has been addressed to some extent, the explicit interplay of hydrodynamics and confinement toward dictating the underlying characteristics for practically relevant material interfaces remains unexplored. Here, we bring out unique roles of surface nanostructures toward altering flow of water in a copper nanochannel, by capturing an exclusive interplay of confinement, roughness, wettability and flow dynamics. Toward this, non-equilibrium molecular dynamics (NEMD) simulations are performed to examine the effect of nanoscale triangular roughness. The width and height of the triangular microgroove are varied along with different driving forces at the channel inlet, and the results are compared with those corresponding to smooth-walled nanochannels. We also unveil the nontrivial characteristics of the interfacial topology as a consequence of spontaneous phase separation at the fluid-solid interface. For a constant driving force, we show that the interface may exhibit concave or convex topology, depending on the nanogroove geometry. Our results provide new vistas on how designed nanoscale roughness structures can be harnessed toward controlling the transport of water in a practically engineered nanosystem, as demanded by the specific application on hand.

14.
Cancer Discov ; 10(4): 608-625, 2020 04.
Article En | MEDLINE | ID: mdl-32046984

A hallmark of pancreatic ductal adenocarcinoma (PDAC) is an exuberant stroma comprised of diverse cell types that enable or suppress tumor progression. Here, we explored the role of oncogenic KRAS in protumorigenic signaling interactions between cancer cells and host cells. We show that KRAS mutation (KRAS*) drives cell-autonomous expression of type I cytokine receptor complexes (IL2rγ-IL4rα and IL2rγ-IL13rα1) in cancer cells that in turn are capable of receiving cytokine growth signals (IL4 or IL13) provided by invading Th2 cells in the microenvironment. Early neoplastic lesions show close proximity of cancer cells harboring KRAS* and Th2 cells producing IL4 and IL13. Activated IL2rγ-IL4rα and IL2rγ-IL13rα1 receptors signal primarily via JAK1-STAT6. Integrated transcriptomic, chromatin occupancy, and metabolomic studies identified MYC as a direct target of activated STAT6 and that MYC drives glycolysis. Thus, paracrine signaling in the tumor microenvironment plays a key role in the KRAS*-driven metabolic reprogramming of PDAC. SIGNIFICANCE: Type II cytokines, secreted by Th2 cells in the tumor microenvironment, can stimulate cancer cell-intrinsic MYC transcriptional upregulation to drive glycolysis. This KRAS*-driven heterotypic signaling circuit in the early and advanced tumor microenvironment enables cooperative protumorigenic interactions, providing candidate therapeutic targets in the KRAS* pathway for this intractable disease.


Cytokines/metabolism , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cellular Reprogramming/genetics , Humans , Mice , Oncogenes , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Transfection , Tumor Microenvironment
15.
Rev Sci Instrum ; 90(6): 063901, 2019 Jun.
Article En | MEDLINE | ID: mdl-31255018

We performed nonlinear optical two-dimensional Fourier transform spectroscopy measurements using an optical resistive high-field magnet on GaAs quantum wells. Magnetic fields up to 25 T can be achieved using the split helix resistive magnet. Two-dimensional spectroscopy measurements based on the coherent four-wave mixing signal require phase stability. Therefore, these measurements are difficult to perform in environments prone to mechanical vibrations. Large resistive magnets use extensive quantities of cooling water, which causes mechanical vibrations, making two-dimensional Fourier transform spectroscopy very challenging. Here, we report on the strategies we used to overcome these challenges and maintain the required phase-stability throughout the measurement. A self-contained portable platform was used to set up the experiments within the time frame provided by a user facility. Furthermore, this platform was floated above the optical table in order to isolate it from vibrations originating from the resistive magnet. Finally, we present two-dimensional Fourier transform spectra obtained from GaAs quantum wells at magnetic fields up to 25 T and demonstrate the utility of this technique in providing important details, which are obscured in one dimensional spectroscopy.

16.
Cancer Cell ; 35(4): 559-572.e7, 2019 04 15.
Article En | MEDLINE | ID: mdl-30905761

The biological functions and mechanisms of oncogenic KRASG12D (KRAS∗) in resistance to immune checkpoint blockade (ICB) therapy are not fully understood. We demonstrate that KRAS∗ represses the expression of interferon regulatory factor 2 (IRF2), which in turn directly represses CXCL3 expression. KRAS∗-mediated repression of IRF2 results in high expression of CXCL3, which binds to CXCR2 on myeloid-derived suppressor cells and promotes their migration to the tumor microenvironment. Anti-PD-1 resistance of KRAS∗-expressing tumors can be overcome by enforced IRF2 expression or by inhibition of CXCR2. Colorectal cancer (CRC) showing higher IRF2 expression exhibited increased responsiveness to anti-PD-1 therapy. The KRAS∗-IRF2-CXCL3-CXCR2 axis provides a framework for patient selection and combination therapies to enhance the effectiveness of ICB therapy in CRC.


Antineoplastic Agents, Immunological/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Interferon Regulatory Factor-2/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Escape , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Adult , Aged , Animals , Cell Line, Tumor , Cell Movement , Chemokines, CXC/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-2/genetics , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Middle Aged , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Young Adult
17.
Cancer Cell ; 35(2): 204-220.e9, 2019 02 11.
Article En | MEDLINE | ID: mdl-30753823

Alterations in chromatin remodeling genes have been increasingly implicated in human oncogenesis. Specifically, the biallelic inactivation of the SWI/SNF subunit SMARCB1 results in the emergence of extremely aggressive pediatric malignancies. Here, we developed embryonic mosaic mouse models of malignant rhabdoid tumors (MRTs) that faithfully recapitulate the clinical-pathological features of the human disease. We demonstrated that SMARCB1-deficient malignancies exhibit dramatic activation of the unfolded protein response (UPR) and ER stress response via a genetically intact MYC-p19ARF-p53 axis. As a consequence, these tumors display an exquisite sensitivity to agents inducing proteotoxic stress and inhibition of the autophagic machinery. In conclusion, our findings provide a rationale for drug repositioning trials investigating combinations of agents targeting the UPR and autophagy in SMARCB1-deficient MRTs.


Autophagy , Endoplasmic Reticulum Stress , Proteostasis , Rhabdoid Tumor/metabolism , SMARCB1 Protein/deficiency , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endoplasmic Reticulum Stress/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Proteasome Inhibitors/pharmacology , Proteostasis/drug effects , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , SMARCB1 Protein/genetics , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Unfolded Protein Response
18.
Oncotarget ; 9(15): 12201-12211, 2018 Feb 23.
Article En | MEDLINE | ID: mdl-29552303

Triple negative breast cancer (TNBC) still remains a challenge to treat in the clinic due to a lack of good targets for treatment. Although TNBC lacks expression of ERα, the expression of ERß and its variants are detected quite frequently in this cancer type and can represent an avenue for treatment. We show that two of the variants of ERß, namely ERß2 and ERß5, control aggressiveness of TNBC by regulating hypoxic signaling through stabilization of HIF-1α. RNA-seq of patient derived xenografts (PDX) from TNBC shows expression of ERß2, ERß4 and ERß5 variants in more than half of the samples. Furthermore, expression of ERß4 in the immortalized, normal mammary epithelial cell line MCF-10A that is resistant to tumorsphere formation caused transformation and development of tumorspheres. By contrast, ERß1, ERß2 or ERß5 were unable to support tumorsphere formation. We have previously shown that all variants except ERß1 stabilize HIF-1α but only ERß4 appears to have the ability to transform normal mammary epithelial cells, pointing towards a unique property of ERß4. We propose that ERß variants may be good diagnostic tools and also serve as novel targets for treatment of breast cancer.

19.
Genes Dev ; 31(11): 1109-1121, 2017 06 01.
Article En | MEDLINE | ID: mdl-28698296

A key feature of high-grade serous ovarian carcinoma (HGSOC) is frequent amplification of the 3q26 locus harboring PRKC-ι (PRKCI). Here, we show that PRKCI is also expressed in early fallopian tube lesions, called serous tubal intraepithelial carcinoma. Transgenic mouse studies establish PRKCI as an ovarian cancer-specific oncogene. Mechanistically, we show that the oncogenic activity of PRKCI relates in part to the up-regulation of TNFα to promote an immune-suppressive tumor microenvironment characterized by an abundance of myeloid-derived suppressor cells and inhibition of cytotoxic T-cell infiltration. Furthermore, system-level and functional analyses identify YAP1 as a downstream effector in tumor progression. In human ovarian cancers, high PRKCI expression also correlates with high expression of TNFα and YAP1 and low infiltration of cytotoxic T cells. The PRKCI-YAP1 regulation of the tumor immunity provides a therapeutic strategy for highly lethal ovarian cancer.


Gene Expression Regulation, Neoplastic/genetics , Immune Tolerance/genetics , Isoenzymes/genetics , Isoenzymes/immunology , Ovarian Neoplasms/genetics , Protein Kinase C/genetics , Protein Kinase C/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Movement/genetics , Cytokines/genetics , Female , Humans , Isoenzymes/metabolism , Mice , Mice, Transgenic , Ovarian Neoplasms/immunology , Ovarian Neoplasms/physiopathology , Phosphoproteins/metabolism , Protein Kinase C/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/metabolism , YAP-Signaling Proteins
20.
Nature ; 542(7642): 484-488, 2017 02 23.
Article En | MEDLINE | ID: mdl-28166537

Synthetic lethality and collateral lethality are two well-validated conceptual strategies for identifying therapeutic targets in cancers with tumour-suppressor gene deletions. Here, we explore an approach to identify potential synthetic-lethal interactions by screening mutually exclusive deletion patterns in cancer genomes. We sought to identify 'synthetic-essential' genes: those that are occasionally deleted in some cancers but are almost always retained in the context of a specific tumour-suppressor deficiency. We also posited that such synthetic-essential genes would be therapeutic targets in cancers that harbour specific tumour-suppressor deficiencies. In addition to known synthetic-lethal interactions, this approach uncovered the chromatin helicase DNA-binding factor CHD1 as a putative synthetic-essential gene in PTEN-deficient cancers. In PTEN-deficient prostate and breast cancers, CHD1 depletion profoundly and specifically suppressed cell proliferation, cell survival and tumorigenic potential. Mechanistically, functional PTEN stimulates the GSK3ß-mediated phosphorylation of CHD1 degron domains, which promotes CHD1 degradation via the ß-TrCP-mediated ubiquitination-proteasome pathway. Conversely, PTEN deficiency results in stabilization of CHD1, which in turn engages the trimethyl lysine-4 histone H3 modification to activate transcription of the pro-tumorigenic TNF-NF-κB gene network. This study identifies a novel PTEN pathway in cancer and provides a framework for the discovery of 'trackable' targets in cancers that harbour specific tumour-suppressor deficiencies.


Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Genes, Essential/genetics , Neoplasms/metabolism , Neoplasms/pathology , PTEN Phosphohydrolase/deficiency , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , DNA Helicases/chemistry , DNA Helicases/deficiency , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Male , Methylation , Molecular Targeted Therapy , NF-kappa B/metabolism , Neoplasms/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination , beta-Transducin Repeat-Containing Proteins/metabolism
...