Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cancer Metab ; 9(1): 31, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34399819

ABSTRACT

BACKGROUND: Metabolic reprogramming is a central feature in many cancer subtypes and a hallmark of cancer. Many therapeutic strategies attempt to exploit this feature, often having unintended side effects on normal metabolic programs and limited efficacy due to integrative nature of metabolic substrate sourcing. Although the initiating oncogenic lesion may vary, tumor cells in lymphoid malignancies often share similar environments and potentially similar metabolic profiles. We examined cells from mouse models of MYC-, RAS-, and BCR-ABL-driven lymphoid malignancies and find a convergence on de novo lipogenesis. We explore the potential role of MYC in mediating lipogenesis by 13C glucose tracing and untargeted metabolic profiling. Inhibition of lipogenesis leads to cell death both in vitro and in vivo and does not induce cell death of normal splenocytes. METHODS: We analyzed RNA-seq data sets for common metabolic convergence in lymphoma and leukemia. Using in vitro cell lines derived in from conditional MYC, RAS, and BCR-ABL transgenic murine models and oncogene-driven human cell lines, we determined gene regulation, metabolic profiles, and sensitivity to inhibition of lipogenesis in lymphoid malignancies. We utilize preclinical murine models and transgenic primary model of T-ALL to determine the effect of lipogenesis blockade across BCR-ABL-, RAS-, and c-MYC-driven lymphoid malignancies. Statistical significance was calculated using unpaired t-tests and one-way ANOVA. RESULTS: This study illustrates that de novo lipid biogenesis is a shared feature of several lymphoma subtypes. Using cell lines derived from conditional MYC, RAS, and BCR-ABL transgenic murine models, we demonstrate shared responses to inhibition of lipogenesis by the acetyl-coA carboxylase inhibitor 5-(tetradecloxy)-2-furic acid (TOFA), and other lipogenesis inhibitors. We performed metabolic tracing studies to confirm the influence of c-MYC and TOFA on lipogenesis. We identify specific cell death responses to TOFA in vitro and in vivo and demonstrate delayed engraftment and progression in vivo in transplanted lymphoma cell lines. We also observe delayed progression of T-ALL in a primary transgenic mouse model upon TOFA administration. In a panel of human cell lines, we demonstrate sensitivity to TOFA treatment as a metabolic liability due to the general convergence on de novo lipogenesis in lymphoid malignancies driven by MYC, RAS, or BCR-ABL. Importantly, cell death was not significantly observed in non-malignant cells in vivo. CONCLUSIONS: These studies suggest that de novo lipogenesis may be a common survival strategy for many lymphoid malignancies and may be a clinically exploitable metabolic liability. TRIAL REGISTRATION: This study does not include any clinical interventions on human subjects.

2.
Front Immunol ; 11: 832, 2020.
Article in English | MEDLINE | ID: mdl-32457754

ABSTRACT

Inhibitors that block the programmed cell death-1 (PD-1) pathway can potentiate endogenous antitumor immunity and have markedly improved cancer survival rates across a broad range of indications. However, these treatments work for only a minority of patients. The efficacy of anti-PD-1 inhibitors may be extended by cytokines, however, the incorporation of cytokines into therapeutic regimens has significant challenges. In their natural form when administered as recombinant proteins, cytokine treatments are often associated with low response rates. Most cytokines have a short half-life which limits their exposure and efficacy. In addition, cytokines can activate counterregulatory pathways, in the case of immune-potentiating cytokines this can lead to immune suppression and thereby diminish their potential efficacy. Improving the drug-like properties of natural cytokines using protein engineering can yield synthetic cytokines with improved bioavailability and tissue targeting, allowing for enhanced efficacy and reduced off-target effects. Using structure guided engineering we have designed a novel class of antibody-cytokine fusion proteins consisting of a PD-1 targeting antibody fused together with an interleukin-21 (IL-21) cytokine mutein. Our bifunctional fusion proteins can block PD-1/programmed death-ligand 1 (PD-L1) interaction whilst simultaneously delivering IL-21 cytokine to PD-1 expressing T cells. Targeted delivery of IL-21 can improve T cell function in a manner that is superior to anti-PD-1 monotherapy. Fusion of engineered IL-21 variants to anti-PD1 antibodies can improve the drug-like properties of IL-21 cytokine leading to improved cytokine serum half-life allowing for less frequent dosing. In addition, we show that targeted delivery of IL-21 can minimize any potential detrimental effect on local antigen-presenting cells. A highly attenuated IL-21 mutein variant (R9E:R76A) fused to a PD-1 antibody provides protection in a humanized mouse model of cancer that is refractory to anti-PD-1 monotherapy. Collectively, our preclinical data demonstrate that this approach may improve upon and extend the utility of anti-PD-1 therapeutics currently in the clinic.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Interleukins/therapeutic use , Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , B7-H1 Antigen/immunology , Disease Models, Animal , Female , Humans , Interleukins/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasms/immunology , Protein Engineering , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use
3.
Blood ; 131(18): 2007-2015, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29514782

ABSTRACT

The MYC proto-oncogene is a gene product that coordinates the transcriptional regulation of a multitude of genes that are essential to cellular programs required for normal as well as neoplastic cellular growth and proliferation, including cell cycle, self-renewal, survival, cell growth, metabolism, protein and ribosomal biogenesis, and differentiation. Here, we propose that MYC regulates these programs in a manner that is coordinated with a global influence on the host immune response. MYC had been presumed to contribute to tumorigenesis through tumor cell-intrinsic influences. More recently, MYC expression in tumor cells has been shown to regulate the tumor microenvironment through effects on both innate and adaptive immune effector cells and immune regulatory cytokines. Then, MYC was shown to regulate the expression of the immune checkpoint gene products CD47 and programmed death-ligand 1. Similarly, other oncogenes, which are known to modulate MYC, have been shown to regulate immune checkpoints. Hence, MYC may generally prevent highly proliferative cells from eliciting an immune response. MYC-driven neoplastic cells have coopted this mechanism to bypass immune detection. Thus, MYC inactivation can restore the immune response against a tumor. MYC-induced tumors may be particularly sensitive to immuno-oncology therapeutic interventions.


Subject(s)
Gene Expression Regulation , Immunity/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Humans , Immunomodulation/genetics , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Oncogenes , Proto-Oncogene Mas
4.
Trends Immunol ; 38(4): 298-305, 2017 04.
Article in English | MEDLINE | ID: mdl-28233639

ABSTRACT

Cancers are often initiated by genetic events that activate proto-oncogenes or inactivate tumor-suppressor genes. These events are also crucial for sustained tumor cell proliferation and survival, a phenomenon described as oncogene addiction. In addition to this cell-intrinsic role, recent evidence indicates that oncogenes also directly regulate immune responses, leading to immunosuppression. Expression of many oncogenes or loss of tumor suppressors induces the expression of immune checkpoints that regulate the immune response, such as PD-L1. We discuss here how oncogenes, and in particular MYC, suppress immune surveillance, and how oncogene-targeted therapies may restore the immune response against tumors.


Subject(s)
Carcinogenesis , Immune Tolerance , Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/immunology , Tumor Suppressor Proteins/immunology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinogenesis/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Immunologic Surveillance , Immunomodulation , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Escape
5.
Science ; 352(6282): 227-31, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26966191

ABSTRACT

The MYC oncogene codes for a transcription factor that is overexpressed in many human cancers. Here we show that MYC regulates the expression of two immune checkpoint proteins on the tumor cell surface: the innate immune regulator CD47 (cluster of differentiation 47) and the adaptive immune checkpoint PD-L1 (programmed death-ligand 1). Suppression of MYC in mouse tumors and human tumor cells caused a reduction in the levels of CD47 and PD-L1 messenger RNA and protein. MYC was found to bind directly to the promoters of the Cd47 and Pd-l1 genes. MYC inactivation in mouse tumors down-regulated CD47 and PD-L1 expression and enhanced the antitumor immune response. In contrast, when MYC was inactivated in tumors with enforced expression of CD47 or PD-L1, the immune response was suppressed, and tumors continued to grow. Thus, MYC appears to initiate and maintain tumorigenesis, in part, through the modulation of immune regulatory molecules.


Subject(s)
B7-H1 Antigen/genetics , CD47 Antigen/genetics , Cell Transformation, Neoplastic/immunology , Gene Expression Regulation, Neoplastic , Immune Tolerance/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Down-Regulation , Gene Knockdown Techniques , Humans , Jurkat Cells , Lymphoma/genetics , Lymphoma/immunology , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , RNA, Small Interfering/genetics
6.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26590477

ABSTRACT

Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.


Subject(s)
Genetic Heterogeneity , Molecular Targeted Therapy , Neoplasms/therapy , Precision Medicine , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/prevention & control , Signal Transduction , Tumor Microenvironment/genetics
7.
Regul Toxicol Pharmacol ; 73(3): 1007-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26239693

ABSTRACT

We present a detailed response to the critique of "State of the Science of Endocrine Disrupting Chemicals 2012" (UNEP/WHO, 2013) by financial stakeholders, authored by Lamb et al. (2014). Lamb et al.'s claim that UNEP/WHO (2013) does not provide a balanced perspective on endocrine disruption is based on incomplete and misleading quoting of the report through omission of qualifying statements and inaccurate description of study objectives, results and conclusions. Lamb et al. define extremely narrow standards for synthesizing evidence which are then used to dismiss the UNEP/WHO 2013 report as flawed. We show that Lamb et al. misuse conceptual frameworks for assessing causality, especially the Bradford-Hill criteria, by ignoring the fundamental problems that exist with inferring causality from empirical observations. We conclude that Lamb et al.'s attempt of deconstructing the UNEP/WHO (2013) report is not particularly erudite and that their critique is not intended to be convincing to the scientific community, but to confuse the scientific data. Consequently, it promotes misinterpretation of the UNEP/WHO (2013) report by non-specialists, bureaucrats, politicians and other decision makers not intimately familiar with the topic of endocrine disruption and therefore susceptible to false generalizations of bias and subjectivity.


Subject(s)
Endocrine Disruptors/toxicity , Animals , Humans
8.
Carcinogenesis ; 36 Suppl 1: S160-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106136

ABSTRACT

Potentially carcinogenic compounds may cause cancer through direct DNA damage or through indirect cellular or physiological effects. To study possible carcinogens, the fields of endocrinology, genetics, epigenetics, medicine, environmental health, toxicology, pharmacology and oncology must be considered. Disruptive chemicals may also contribute to multiple stages of tumor development through effects on the tumor microenvironment. In turn, the tumor microenvironment consists of a complex interaction among blood vessels that feed the tumor, the extracellular matrix that provides structural and biochemical support, signaling molecules that send messages and soluble factors such as cytokines. The tumor microenvironment also consists of many host cellular effectors including multipotent stromal cells/mesenchymal stem cells, fibroblasts, endothelial cell precursors, antigen-presenting cells, lymphocytes and innate immune cells. Carcinogens can influence the tumor microenvironment through effects on epithelial cells, the most common origin of cancer, as well as on stromal cells, extracellular matrix components and immune cells. Here, we review how environmental exposures can perturb the tumor microenvironment. We suggest a role for disrupting chemicals such as nickel chloride, Bisphenol A, butyltins, methylmercury and paraquat as well as more traditional carcinogens, such as radiation, and pharmaceuticals, such as diabetes medications, in the disruption of the tumor microenvironment. Further studies interrogating the role of chemicals and their mixtures in dose-dependent effects on the tumor microenvironment could have important general mechanistic implications for the etiology and prevention of tumorigenesis.


Subject(s)
Environmental Exposure/adverse effects , Hazardous Substances/adverse effects , Tumor Microenvironment/drug effects , Animals , Carcinogenesis/chemically induced , Humans , Neoplasms/chemically induced
9.
Proc Natl Acad Sci U S A ; 112(21): 6539-44, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25964345

ABSTRACT

The MYC oncogene is frequently mutated and overexpressed in human renal cell carcinoma (RCC). However, there have been no studies on the causative role of MYC or any other oncogene in the initiation or maintenance of kidney tumorigenesis. Here, we show through a conditional transgenic mouse model that the MYC oncogene, but not the RAS oncogene, initiates and maintains RCC. Desorption electrospray ionization-mass-spectrometric imaging was used to obtain chemical maps of metabolites and lipids in the mouse RCC samples. Gene expression analysis revealed that the mouse tumors mimicked human RCC. The data suggested that MYC-induced RCC up-regulated the glutaminolytic pathway instead of the glycolytic pathway. The pharmacologic inhibition of glutamine metabolism with bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide impeded MYC-mediated RCC tumor progression. Our studies demonstrate that MYC overexpression causes RCC and points to the inhibition of glutamine metabolism as a potential therapeutic approach for the treatment of this disease.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Genes, myc , Glutamine/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Enzyme Inhibitors/pharmacology , Genes, ras , Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Humans , Kidney Neoplasms/pathology , Lipid Metabolism , Mice , Mice, SCID , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Spectrometry, Mass, Electrospray Ionization , Sulfides/pharmacology , Thiadiazoles/pharmacology , Up-Regulation
10.
Semin Cancer Biol ; 35 Suppl: S199-S223, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25865775

ABSTRACT

Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.


Subject(s)
Carcinogenesis/drug effects , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment/genetics , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Cell Proliferation/drug effects , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/prevention & control , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Signal Transduction , Tumor Microenvironment/drug effects
11.
PLoS One ; 10(3): e0119177, 2015.
Article in English | MEDLINE | ID: mdl-25749104

ABSTRACT

Steroid and xenobiotic receptor (SXR) and its murine ortholog, pregnane X receptor (PXR), are nuclear receptors that are expressed at high levels in the liver and the intestine where they function as xenobiotic sensors that induce expression of genes involved in detoxification and drug excretion. Recent evidence showed that SXR and PXR are also expressed in bone tissue where they mediate bone metabolism. Here we report that systemic deletion of PXR results in aging-dependent wearing of articular cartilage of knee joints. Histomorphometrical analysis showed remarkable reduction of width and an enlarged gap between femoral and tibial articular cartilage in PXR knockout mice. We hypothesized that genes induced by SXR in chondrocytes have a protective effect on articular cartilage and identified Fam20a (family with sequence similarity 20a) as an SXR-dependent gene induced by the known SXR ligands, rifampicin and vitamin K2. Lastly, we demonstrated the biological significance of Fam20a expression in chondrocytes by evaluating osteoarthritis-related gene expression of primary articular chondrocytes. Consistent with epidemiological findings, our results indicate that SXR/PXR protects against aging-dependent wearing of articular cartilage and that ligands for SXR/PXR have potential role in preventing osteoarthritis caused by aging.


Subject(s)
Aging/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/metabolism , Receptors, Steroid/deficiency , Aging/genetics , Aging/pathology , Animals , Cartilage, Articular/pathology , Chondrocytes/pathology , Dental Enamel Proteins , Knee Joint/metabolism , Knee Joint/pathology , Mice , Mice, Knockout , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Pregnane X Receptor , Proteins/genetics , Proteins/metabolism
12.
Cancer Res ; 74(21): 6205-15, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25217519

ABSTRACT

Cancers due to germline mutations in the BRCA1 gene tend to lack targets for approved chemoprevention agents. This study aimed at a targeted chemoprevention strategy for BRCA1-associated malignancies. Mutant BRCA1 limits the base-excision DNA repair activity that addresses oxidative DNA damage, the accumulation of which heightens one's risk for cancer. Therefore, we conducted a high-throughput chemical screen to identify drug candidates that could attenuate the inhibitory effects of mutant BRCA1 on this repair activity, thereby describing a new class of DNA repair-activating chemopreventive agents. In the screen design, such drugs functioned by enhancing base-excision DNA repair of oxidative DNA damage in the presence of mutant BRCA1, with minimal cytotoxicity. We identified at least one new agent that decreased malignant properties associated with tumorigenesis, including anchorage-independent growth and tumor progression. This work offers a preclinical proof-of-concept for a wholly new approach to chemoprevention in carriers of BRCA1 mutations as a strategy to reduce the prevalence of BRCA1-associated malignancy.


Subject(s)
BRCA1 Protein/antagonists & inhibitors , Breast Neoplasms/genetics , Molecular Targeted Therapy , Small Molecule Libraries/administration & dosage , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , DNA Repair/drug effects , DNA Repair/genetics , Female , Germ-Line Mutation , Humans , MCF-7 Cells , Small Molecule Libraries/chemistry
13.
PLoS One ; 9(9): e107589, 2014.
Article in English | MEDLINE | ID: mdl-25208064

ABSTRACT

The Cre/loxP system is a powerful tool for generating conditional genomic recombination and is often used to examine the mechanistic role of specific genes in tumorigenesis. However, Cre toxicity due to its non-specific endonuclease activity has been a concern. Here, we report that tamoxifen-mediated Cre activation in vivo induced the regression of primary lymphomas in p53-/- mice. Our findings illustrate that Cre activation alone can induce the regression of established tumors.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Integrases/genetics , Lymphoma/drug therapy , Lymphoma/genetics , Tamoxifen/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/drug effects , Carcinogenesis/genetics , Enzyme Activation/drug effects , Genetic Therapy , Integrases/metabolism , Lymphoma/metabolism , Lymphoma/pathology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Promoter Regions, Genetic , Tumor Burden/drug effects , Tumor Suppressor Protein p53/deficiency , Ubiquitin C/genetics , Ubiquitin C/metabolism
14.
Cancer Cell ; 26(2): 262-72, 2014 Aug 11.
Article in English | MEDLINE | ID: mdl-25117713

ABSTRACT

The MYC oncogene regulates gene expression through multiple mechanisms, and its overexpression culminates in tumorigenesis. MYC inactivation reverses turmorigenesis through the loss of distinguishing features of cancer, including autonomous proliferation and survival. Here we report that MYC via miR-17-92 maintains a neoplastic state through the suppression of chromatin regulatory genes Sin3b, Hbp1, Suv420h1, and Btg1, as well as the apoptosis regulator Bim. The enforced expression of miR-17-92 prevents MYC suppression from inducing proliferative arrest, senescence, and apoptosis and abrogates sustained tumor regression. Knockdown of the five miR-17-92 target genes blocks senescence and apoptosis while it modestly delays proliferative arrest, thus partially recapitulating miR-17-92 function. We conclude that MYC, via miR-17-92, maintains a neoplastic state by suppressing specific target genes.


Subject(s)
Cell Proliferation , Cell Survival , Lymphoma/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/physiology , Animals , Apoptosis , Gene Expression Regulation, Neoplastic , Lymphoma/genetics , Lymphoma/pathology , Mice , Multigene Family , Neoplasm Transplantation , RNA Interference , Tumor Burden , Tumor Cells, Cultured
15.
J Immunother Cancer ; 2: 24, 2014.
Article in English | MEDLINE | ID: mdl-25089198

ABSTRACT

The targeted inactivation of a single oncogene can induce dramatic tumor regression, suggesting that cancers are "oncogene addicted." Tumor regression following oncogene inactivation has been thought to be a consequence of restoration of normal physiological programs that induce proliferative arrest, apoptosis, differentiation, and cellular senescence. However, recent observations illustrate that oncogene addiction is highly dependent upon the host immune cells. In particular, CD4(+) helper T cells were shown to be essential to the mechanism by which MYC or BCR-ABL inactivation elicits "oncogene withdrawal." Hence, immune mediators contribute in multiple ways to the pathogenesis, prevention, and treatment of cancer, including mechanisms of tumor initiation, progression, and surveillance, but also oncogene inactivation-mediated tumor regression. Data from both the bench and the bedside illustrates that the inactivation of a driver oncogene can induce activation of the immune system that appears to be essential for sustained tumor regression.

16.
Immunol Res ; 58(2-3): 282-91, 2014 May.
Article in English | MEDLINE | ID: mdl-24791942

ABSTRACT

Tumors are genetically complex and can have a multitude of mutations. Consequently, it is surprising that the suppression of a single oncogene can result in rapid and sustained tumor regression, illustrating the concept that cancers are often "oncogene addicted." The mechanism of oncogene addiction has been presumed to be largely cell autonomous as a consequence of the restoration of normal physiological programs that induce proliferative arrest, apoptosis, differentiation, and/or cellular senescence. Interestingly, it has recently become apparent that upon oncogene inactivation, the immune response is critical in mediating the phenotypic consequences of oncogene addiction. In particular, CD4(+) T cells have been suggested to be essential to the remodeling of the tumor microenvironment, including the shutdown of host angiogenesis and the induction of cellular senescence in the tumor. However, adaptive and innate immune cells are likely involved. Thus, the effectors of the immune system are involved not only in tumor initiation, tumor progression, and immunosurveillance, but also in the mechanism of tumor regression upon targeted oncogene inactivation. Hence, oncogene inactivation may be an effective therapeutic approach because it both reverses the neoplastic state within a cancer cell and reactivates the host immune response that remodels the tumor microenvironment.


Subject(s)
Immune System , Neoplasms/genetics , Neoplasms/immunology , Oncogene Proteins/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Humans , Immunologic Surveillance , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , Oncogene Proteins/antagonists & inhibitors , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
17.
Cancer Cell ; 25(3): 350-65, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24651014

ABSTRACT

Tumor endothelial cells (ECs) promote cancer progression in ways beyond their role as conduits supporting metabolism. However, it is unknown how vascular niche-derived paracrine factors, defined as angiocrine factors, provoke tumor aggressiveness. Here, we show that FGF4 produced by B cell lymphoma cells (LCs) through activating FGFR1 upregulates the Notch ligand Jagged1 (Jag1) on neighboring ECs. In turn, upregulation of Jag1 on ECs reciprocally induces Notch2-Hey1 in LCs. This crosstalk enforces aggressive CD44(+)IGF1R(+)CSF1R(+) LC phenotypes, including extranodal invasion and chemoresistance. Inducible EC-selective deletion of Fgfr1 or Jag1 in the Eµ-Myc lymphoma model or impairing Notch2 signaling in mouse and human LCs diminished lymphoma aggressiveness and prolonged mouse survival. Thus, targeting the angiocrine FGF4-FGFR1/Jag1-Notch2 loop inhibits LC aggressiveness and enhances chemosensitivity.


Subject(s)
Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Calcium-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Fibroblast Growth Factor 4/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Notch2/metabolism , Animals , Burkitt Lymphoma/genetics , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Endothelial Cells/metabolism , Enzyme Activation , Genes, myc , Humans , Hyaluronan Receptors/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neoplasm Invasiveness , RNA Interference , RNA, Small Interfering , Receptor, IGF Type 1/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Serrate-Jagged Proteins , Signal Transduction/genetics , Tumor Cells, Cultured , Up-Regulation
18.
Mol Cell Oncol ; 1(4): e970092, 2014.
Article in English | MEDLINE | ID: mdl-27308380

ABSTRACT

MYC regulates tumorigenesis by coordinating the expression of thousands of genes. We found that MYC appears to regulate the decisions between cell survival versus death and self-renewal versus senescence through the microRNA miR-17-92 cluster. Addiction to the MYC oncogene may therefore in fact be an addiction to miR-17-92.

19.
Environ Health ; 12: 69, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23981490

ABSTRACT

The "common sense" intervention by toxicology journal editors regarding proposed European Union endocrine disrupter regulations ignores scientific evidence and well-established principles of chemical risk assessment. In this commentary, endocrine disrupter experts express their concerns about a recently published, and is in our considered opinion inaccurate and factually incorrect, editorial that has appeared in several journals in toxicology. Some of the shortcomings of the editorial are discussed in detail. We call for a better founded scientific debate which may help to overcome a polarisation of views detrimental to reaching a consensus about scientific foundations for endocrine disrupter regulation in the EU.


Subject(s)
Endocrine Disruptors/toxicity , Environmental Exposure , Environmental Pollutants/toxicity , Periodicals as Topic , Toxicology/standards , European Union , Government Regulation , Health Policy , Humans
20.
Curr Opin Immunol ; 25(2): 246-58, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23571026

ABSTRACT

Cancer is highly complex. The magnitude of this complexity makes it highly surprising that even the brief suppression of an oncogene can sometimes result in rapid and sustained tumor regression, illustrating that cancers can be 'oncogene addicted' [1-10]. The essential implication is that oncogenes may not only fuel the initiation of tumorigenesis, but in some cases must be excessively activated to maintain a neoplastic state [11]. Oncogene suppression acutely restores normal physiological programs that effectively overrides secondary genetic events and a cancer collapses [12,13]. Oncogene addiction is the description of the dramatic and sustained regression of some cancers upon the specific inactivation of a single oncogene [1-13,14(••),15,16(••)], that can occur through tumor intrinsic [1,2,4,12], but also host immune mechanisms [17-23]. Notably, oncogene inactivation elicits a host immune response that involves specific immune effectors and cytokines that facilitate a remodeling of the tumor microenvironment including the shut down of angiogenesis and the induction of cellular senescence of tumor cells [16(••)]. Hence, immune effectors are not only critically involved in tumor prevention, initiation [17-19], and progression [20], but also appear to be essential to tumor regression upon oncogene inactivation [21,22(••),23(••)]. Understanding how the inactivation of an oncogene elicits a systemic signal in the host that prompts a deconstruction of a tumor could have important implications. The combination of oncogene-targeted therapy together with immunomodulatory therapy may be ideal for the development of both robust tumor intrinsic and immunological responses, effectively leading to sustained tumor regression.


Subject(s)
Neoplasms/genetics , Neoplasms/immunology , Oncogenes/genetics , Animals , Humans , Models, Biological , Oncogenes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...