Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 41
1.
Bioconjug Chem ; 35(4): 489-498, 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38591251

The role of the intestinal microbiota in host health is increasingly revealed in its contributions to disease states. The host-microbiome interaction is multifactorial and dynamic. One of the factors that has recently been strongly associated with host physiological responses is peptidoglycan from bacterial cell walls. Peptidoglycan from gut commensal bacteria activates peptidoglycan sensors in human cells, including the nucleotide-binding oligomerization domain-containing protein 2. When present in the gastrointestinal tract, both the polymeric form (sacculi) and depolymerized fragments can modulate host physiology, including checkpoint anticancer therapy efficacy, body temperature and appetite, and postnatal growth. To utilize this growing area of biology toward therapeutic prescriptions, it will be critical to directly analyze a key feature of the host-microbiome interaction from living hosts in a reproducible and noninvasive way. Here we show that metabolically labeled peptidoglycan/sacculi can be readily isolated from fecal samples collected from both mice and humans. Analysis of fecal samples provided a noninvasive route to probe the gut commensal community including the metabolic synchronicity with the host circadian clock. Together, these results pave the way for noninvasive diagnostic tools to interrogate the causal nature of peptidoglycan in host health and disease.


Gastrointestinal Microbiome , Microbiota , Humans , Animals , Mice , Peptidoglycan , Bacteria/metabolism
2.
bioRxiv ; 2023 Sep 02.
Article En | MEDLINE | ID: mdl-37693563

The role of the intestinal microbiota in host health is increasingly revealed in its contributions to disease states. The host-microbiome interaction is multifactorial and dynamic. One of the factors that has recently been strongly associated with host physiological responses is peptidoglycan from bacterial cell walls. Peptidoglycan from gut commensal bacteria activate peptidoglycan sensors in human cells, including the Nucleotide-binding oligomerization domain containing protein 2 (NOD2). When present in the gastrointestinal tract, both the polymeric form (sacculi) and de-polymerized fragments can modulate host physiology, including checkpoint anticancer therapy efficacy, body temperature and appetite, and postnatal growth. To leverage this growing area of biology towards therapeutic prescriptions, it will be critical to directly analyze a key feature of the host-microbiome interaction from living hosts in a reproducible and non-invasive way. Here we show that metabolically labeled peptidoglycan/sacculi can be readily isolated from fecal samples collected from both mice and humans. Analysis of fecal samples provided a non-invasive route to probe the gut commensal community including the metabolic synchronicity with the host circadian clock. Together, these results pave the way for non-invasive diagnostic tools to interrogate the causal nature of peptidoglycan in host health and disease.

3.
Cell Chem Biol ; 2022 Dec 05.
Article En | MEDLINE | ID: mdl-36516833

In mammals, gut commensal microbiota interact extensively with the host, and the same interactions can be dysregulated in diseased states. Animal imaging is a powerful technique that is widely used to diagnose, measure, and track biological changes in model organisms such as laboratory mice. Several imaging techniques have been discovered and adopted by the research community that provide dynamic, non-invasive assessment of live animals, but these gains have not been universal across all fields of biology. Herein, we describe a method to non-invasively image commensal bacteria based on the specific metabolic labeling of bacterial cell walls to illuminate the gut bacteria of live mice. This tagging strategy may additionally provide unprecedented insight into cell wall turnover of gut commensals, which has implications for bacterial cellular growth and division, in a live animal.

4.
Cancer Immunol Res ; 10(11): 1309-1325, 2022 11 02.
Article En | MEDLINE | ID: mdl-36040846

Establishing commensal dysbiosis, defined as an inflammatory gut microbiome with low biodiversity, before breast tumor initiation, enhances early dissemination of hormone receptor-positive (HR+) mammary tumor cells. Here, we sought to determine whether cellular changes occurring in normal mammary tissues, before tumor initiation and in response to dysbiosis, enhanced dissemination of HR+ tumors. Commensal dysbiosis increased both the frequency and profibrogenicity of mast cells in normal, non-tumor-bearing mammary tissues, a phenotypic change that persisted after tumor implantation. Pharmacological and adoptive transfer approaches demonstrated that profibrogenic mammary tissue mast cells from dysbiotic animals were sufficient to enhance dissemination of HR+ tumor cells. Using archival HR+ patient samples, we determined that enhanced collagen levels in tumor-adjacent mammary tissue positively correlated with mast cell abundance and HR+ breast cancer recurrence. Together, these data demonstrate that mast cells programmed by commensal dysbiosis activate mammary tissue fibroblasts and orchestrate early dissemination of HR+ breast tumors.


Gastrointestinal Microbiome , Mammary Neoplasms, Animal , Animals , Dysbiosis , Mast Cells/pathology , Neoplasm Recurrence, Local , Cell Transformation, Neoplastic
5.
iScience ; 24(9): 103012, 2021 Sep 24.
Article En | MEDLINE | ID: mdl-34522855

The gut microbiota's function in regulating health has seen it linked to disease progression in several cancers. However, there is limited research detailing its influence in breast cancer (BrCa). This study found that antibiotic-induced perturbation of the gut microbiota significantly increases tumor progression in multiple BrCa mouse models. Metagenomics highlights the common loss of several bacterial species following antibiotic administration. One such bacteria, Faecalibaculum rodentium, rescued this increased tumor growth. Single-cell transcriptomics identified an increased number of cells with a stromal signature in tumors, and subsequent histology revealed an increased abundance of mast cells in the tumor stromal regions. We show that administration of a mast cell stabilizer, cromolyn, rescues increased tumor growth in antibiotic treated animals but has no influence on tumors from control cohorts. These findings highlight that BrCa-microbiota interactions are different from other cancers studied to date and suggest new research avenues for therapy development.

6.
Cancer Immunol Res ; 9(5): 583-597, 2021 05.
Article En | MEDLINE | ID: mdl-33619119

CD8+ T-cell infiltration and effector activity in tumors are correlated with better overall survival of patients, suggesting that the ability of T cells to enter and remain in contact with tumor cells supports tumor control. CD8+ T cells express the collagen-binding integrins CD49a and CD49b, but little is known about their function or how their expression is regulated in the tumor microenvironment (TME). Here, we found that tumor-infiltrating CD8+ T cells initially expressed CD49b, gained CD49a, and then lost CD49b over the course of tumor outgrowth. This differentiation sequence was driven by antigen-independent elements in the TME, although T-cell receptor (TCR) stimulation further increased CD49a expression. Expression of exhaustion markers and CD49a associated temporally but not mechanistically. Intratumoral CD49a-expressing CD8+ T cells failed to upregulate TCR-dependent Nur77 expression, whereas CD69 was constitutively expressed, consistent with both a lack of productive antigen engagement and a tissue-resident memory-like phenotype. Imaging T cells in live tumor slices revealed that CD49a increased their motility, especially of those in close proximity to tumor cells, suggesting that it may interfere with T-cell recognition of tumor cells by distracting them from productive engagement, although we were not able to augment productive engagement by short-term CD49a blockade. CD49b also promoted relocalization of T cells at a greater distance from tumor cells. Thus, our results demonstrate that expression of these integrins affects T-cell trafficking and localization in tumors via distinct mechanisms, and suggests a new way in which the TME, and likely collagen, could promote tumor-infiltrating CD8+ T-cell dysfunction.


CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/genetics , Integrin alpha1/metabolism , Integrin alpha2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Humans , Male , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Tumor Microenvironment
7.
Article En | MEDLINE | ID: mdl-32923409

The obligate intracellular bacterium Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections. Once internalized in host cells, C. trachomatis undergoes a biphasic developmental cycle within a membrane-bound compartment, known as the inclusion. Successful establishment of the intracellular niche relies on bacterial Type III effector proteins, such as Inc proteins. In vitro and in vivo systems have contributed to elucidating the intracellular lifestyle of C. trachomatis, but additional models combining the archetypal environment of infection with the advantages of in vitro systems are needed. Organoids are three-dimensional structures that recapitulate the microanatomy of an organ's epithelial layer, bridging the gap between in vitro and in vivo systems. Organoids are emerging as relevant model systems to study interactions between bacterial pathogens and their hosts. Here, we took advantage of recently developed murine endometrial organoids (EMOs) and present a C. trachomatis-murine EMO infection model system. Confocal microscopy of EMOs infected with fluorescent protein-expressing bacteria revealed that inclusions are formed within the cytosol of epithelial cells. Moreover, infection with a C. trachomatis strain that allows for the tracking of RB to EB transition indicated that the bacteria undergo a full developmental cycle, which was confirmed by harvesting infectious bacteria from infected EMOs. Finally, the inducible gene expression and cellular localization of a Chlamydia Inc protein within infected EMOs further demonstrated that this model is compatible with the study of Type III secreted effectors. Altogether, we describe a novel and relevant system for the study of Chlamydia-host interactions.


Chlamydia Infections , Organoids , Animals , Bacterial Proteins , Chlamydia trachomatis , Female , HeLa Cells , Humans , Mice
8.
Front Immunol ; 10: 1942, 2019.
Article En | MEDLINE | ID: mdl-31555258

Ductal carcinoma in situ (DCIS) is a non-obligate precursor of breast cancer, and it only progresses to invasive breast cancer in around 40% of patients. While immune infiltrates have been observed in these early cancer lesions, their potential prognostic value is still unclear. Regulatory T (Treg) cells accumulate in advanced breast cancers, and predict poor outcome. We have shown before that ablation of Treg cells in established tumors leads to significant decrease in primary and metastatic tumor burden. In this work, we sought to investigate Treg cell function in the progression from non-invasive to invasive breast cancer lesions. To this end, we used the murine mammary tumor virus polyoma middle T (MMTV-PyMT) murine model of spontaneous, stage-wise breast carcinogenesis crossed to Foxp3 DTR knock in mice, allowing Treg cell ablation by administration of diphtheria toxin. Transient targeting of Treg cells at the in situ carcinoma stage resulted in a significant increase in the number of tumor-bearing mammary glands and size of growing tumors compared with control mice. Whole mammary gland mounts and histological examination confirmed larger emergent tumor area in Treg cell-ablated mice, and revealed that these tumors were characterized by a more advanced tumor staging, with presence of early invasion, increased desmoplasia and collagen deposition. Furthermore, Treg cell ablation increased the percentage of cancer stem/progenitor cells in the mammary compartment. Interestingly, Treg cell ablation resulted in increased inflammatory cytokines IL-4 and IL-5 with a concomitant reduction in classically activated tumor associated macrophages. This TH2-biased immune regulatory mammary inflammation was consistent with the enhancement in tumor promotion that we observed. Overall, our study demonstrates that Treg cells oppose breast cancer progression at early stages, raising a cautionary note regarding the consideration of immune intervention targeted at boosting immune responses for DCIS.


Breast Neoplasms/immunology , Carcinoma, Intraductal, Noninfiltrating/immunology , Mammary Neoplasms, Animal/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cytokines/immunology , Disease Models, Animal , Disease Progression , Female , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/physiology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred C57BL , Neoplasm Staging/methods , T-Lymphocytes, Regulatory/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Tumor Burden/immunology
9.
Adv Cancer Res ; 143: 255-294, 2019.
Article En | MEDLINE | ID: mdl-31202360

Humans are a colonized with trillions of commensal microorganisms which exert a profound effect on normal host physiology and immune function through an abundance of genetic and metabolic by-products. Although the commensal microbiome has beneficial functions to host physiology, perturbations of the composition of the commensal microbiome or the homeostatic mucosal environment can lead to the induction of immune pathology and systemic inflammation. In the context of cancer progression or response to immune therapy, this inflammation can be detrimental, resulting in tumor growth and the promotion of immune suppression. On the other hand, significant associations have been identified whereby certain commensal microorganisms are able to enhance T cell function or are required for tumor control in cancer patients treated with certain immune therapies and chemotherapies. The focus of this chapter is to highlight the role of the commensal microbiome during tumor progression and in response to immune therapies.


Antineoplastic Agents/therapeutic use , Immune System/immunology , Immunotherapy/methods , Inflammation/immunology , Microbiota/immunology , Neoplasms/drug therapy , T-Lymphocytes/immunology , Animals , Homeostasis , Humans , Immunomodulation , Neoplasms/immunology , Neoplasms/microbiology , Symbiosis , T-Lymphocytes/microbiology
10.
Cancer Res ; 79(14): 3662-3675, 2019 Jul 15.
Article En | MEDLINE | ID: mdl-31064848

It is unknown why some patients with hormone receptor-positive (HR+) breast cancer present with more aggressive and invasive disease. Metastatic dissemination occurs early in disease and is facilitated by cross-talk between the tumor and tissue environment, suggesting that undefined host-intrinsic factors enhance early dissemination and the probability of developing metastatic disease. Here, we have identified commensal dysbiosis as a host-intrinsic factor associated with metastatic dissemination. Using a mouse model of HR+ mammary cancer, we demonstrate that a preestablished disruption of commensal homeostasis results in enhanced circulating tumor cells and subsequent dissemination to the tumor-draining lymph nodes and lungs. Commensal dysbiosis promoted early inflammation within the mammary gland that was sustained during HR+ mammary tumor progression. Furthermore, dysbiosis enhanced fibrosis and collagen deposition both systemically and locally within the tumor microenvironment and induced significant myeloid infiltration into the mammary gland and breast tumor. These effects were recapitulated both by directly targeting gut microbes using nonabsorbable antibiotics and by fecal microbiota transplantation of dysbiotic cecal contents, demonstrating the direct impact of gut dysbiosis on mammary tumor dissemination. This study identifies dysbiosis as a preexisting, host-intrinsic regulator of tissue inflammation, myeloid recruitment, fibrosis, and dissemination of tumor cells in HR+ breast cancer. SIGNIFICANCE: Identification of commensal dysbiosis as a host-intrinsic factor mediating evolution of metastatic breast cancer allows for development of interventions or diagnostic tools for patients at highest risk for developing metastatic disease.See related commentary by Ingman, p. 3539.


Breast Neoplasms , Gastrointestinal Microbiome , Dysbiosis , Humans , Inflammation , Symbiosis , Tumor Microenvironment
11.
Nature ; 562(7727): 423-428, 2018 10.
Article En | MEDLINE | ID: mdl-30305738

Tumours evade immune control by creating hostile microenvironments that perturb T cell metabolism and effector function1-4. However, it remains unclear how intra-tumoral T cells integrate and interpret metabolic stress signals. Here we report that ovarian cancer-an aggressive malignancy that is refractory to standard treatments and current immunotherapies5-8-induces endoplasmic reticulum stress and activates the IRE1α-XBP1 arm of the unfolded protein response9,10 in T cells to control their mitochondrial respiration and anti-tumour function. In T cells isolated from specimens collected from patients with ovarian cancer, upregulation of XBP1 was associated with decreased infiltration of T cells into tumours and with reduced IFNG mRNA expression. Malignant ascites fluid obtained from patients with ovarian cancer inhibited glucose uptake and caused N-linked protein glycosylation defects in T cells, which triggered IRE1α-XBP1 activation that suppressed mitochondrial activity and IFNγ production. Mechanistically, induction of XBP1 regulated the abundance of glutamine carriers and thus limited the influx of glutamine that is necessary to sustain mitochondrial respiration in T cells under glucose-deprived conditions. Restoring N-linked protein glycosylation, abrogating IRE1α-XBP1 activation or enforcing expression of glutamine transporters enhanced mitochondrial respiration in human T cells exposed to ovarian cancer ascites. XBP1-deficient T cells in the metastatic ovarian cancer milieu exhibited global transcriptional reprogramming and improved effector capacity. Accordingly, mice that bear ovarian cancer and lack XBP1 selectively in T cells demonstrate superior anti-tumour immunity, delayed malignant progression and increased overall survival. Controlling endoplasmic reticulum stress or targeting IRE1α-XBP1 signalling may help to restore the metabolic fitness and anti-tumour capacity of T cells in cancer hosts.


Endoribonucleases/metabolism , Mitochondria/metabolism , Ovarian Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , X-Box Binding Protein 1/metabolism , Amino Acid Transport Systems, Basic , Animals , Ascites/metabolism , Cell Respiration , Disease Progression , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glutamine/metabolism , Glycosylation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Signal Transduction , Survival Rate , T-Lymphocytes/metabolism , Tumor Escape/immunology , Unfolded Protein Response , X-Box Binding Protein 1/biosynthesis , X-Box Binding Protein 1/deficiency
12.
J Leukoc Biol ; 103(5): 799-805, 2018 05.
Article En | MEDLINE | ID: mdl-29537705

Due to their cytotoxic activities, many anticancer drugs cause extensive damage to the intestinal mucosa and have antibiotic activities. Here, we show that cisplatin induces significant changes in the repertoire of intestinal commensal bacteria that exacerbate mucosal damage. Restoration of the microbiota through fecal-pellet gavage drives healing of cisplatin-induced intestinal damage. Bacterial translocation to the blood stream is correspondingly abrogated, resulting in a significant reduction in systemic inflammation, as evidenced by decreased serum IL-6 and reduced mobilization of granulocytes. Mechanistically, reversal of dysbiosis in response to fecal gavage results in the production of protective mucins and mobilization of CD11b+ myeloid cells to the intestinal mucosa, which promotes angiogenesis. Administration of Ruminococcus gnavus, a bacterial strain selectively depleted by cisplatin treatment, could only partially restore the integrity of the intestinal mucosa and reduce systemic inflammation, without measurable increases in the accumulation of mucin proteins. Together, our results indicate that reconstitution of the full repertoire of intestinal bacteria altered by cisplatin treatment accelerates healing of the intestinal epithelium and ameliorates systemic inflammation. Therefore, fecal microbiota transplant could paradoxically prevent life-threatening bacteremia in cancer patients treated with chemotherapy.


Cisplatin/pharmacology , Dysbiosis/therapy , Fecal Microbiota Transplantation , Intestines/microbiology , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Dysbiosis/mortality , Dysbiosis/pathology , Female , Gastrointestinal Microbiome , Intestines/drug effects , Intestines/pathology , Ovarian Neoplasms/microbiology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/microbiology , Peritoneal Neoplasms/pathology , Tumor Cells, Cultured
13.
Semin Immunol ; 32: 62-73, 2017 08.
Article En | MEDLINE | ID: mdl-28687194

Commensal microbes inhabit barrier surfaces, providing a first line of defense against invading pathogens, aiding in metabolic function of the host, and playing a vital role in immune development and function. Several recent studies have demonstrated that commensal microbes influence systemic immune function and homeostasis. For patients with extramucosal cancers, or cancers occurring distal to barrier surfaces, the role of commensal microbes in influencing tumor progression is beginning to be appreciated. Extrinsic factors such as chronic inflammation, antibiotics, and chemotherapy dysregulate commensal homeostasis and drive tumor-promoting systemic inflammation through a variety of mechanisms, including disruption of barrier function and bacterial translocation, release of soluble inflammatory mediators, and systemic changes in metabolic output. Conversely, it has also been demonstrated that certain immune therapies, immunogenic chemotherapies, and checkpoint inhibitors rely on the commensal microbiota to facilitate anti-tumor immune responses. Thus, it is evident that the mechanisms associated with commensal microbe facilitation of both pro- and anti-tumor immune responses are context dependent and rely upon a variety of factors present within the tumor microenvironment and systemic periphery. The goal of this review is to highlight the various contexts during which commensal microbes orchestrate systemic immune function with a focus on describing possible scenarios where the loss of microbial homeostasis enhances tumor progression.


Gastrointestinal Microbiome/immunology , Inflammation/immunology , Microbiota/immunology , Neoplasms/microbiology , Animals , Cell Transformation, Neoplastic , Dysbiosis , Homeostasis , Humans , Neoplasms/immunology , Symbiosis , Tumor Microenvironment
14.
Clin Cancer Res ; 23(2): 441-453, 2017 Jan 15.
Article En | MEDLINE | ID: mdl-27435394

PURPOSE: To define the safety and effectiveness of T cells redirected against follicle-stimulating hormone receptor (FSHR)-expressing ovarian cancer cells. EXPERIMENTAL DESIGN: FSHR expression was determined by Western blotting, immunohistochemistry, and qPCR in 77 human ovarian cancer specimens from 6 different histologic subtypes and 20 human healthy tissues. The effectiveness of human T cells targeted with full-length FSH in vivo was determined against a panel of patient-derived xenografts. Safety and effectiveness were confirmed in immunocompetent tumor-bearing mice, using constructs targeting murine FSHR and syngeneic T cells. RESULTS: FSHR is expressed in gynecologic malignancies of different histologic types but not in nonovarian healthy tissues. Accordingly, T cells expressing full-length FSHR-redirected chimeric receptors mediate significant therapeutic effects (including tumor rejection) against a panel of patient-derived tumors in vivo In immunocompetent mice growing syngeneic, orthotopic, and aggressive ovarian tumors, fully murine FSHR-targeted T cells also increased survival without any measurable toxicity. Notably, chimeric receptors enhanced the ability of endogenous tumor-reactive T cells to abrogate malignant progression upon adoptive transfer into naïve recipients subsequently challenged with the same tumor. Interestingly, FSHR-targeted T cells persisted as memory lymphocytes without noticeable PD-1-dependent exhaustion during end-stage disease, in the absence of tumor cell immunoediting. However, exosomes in advanced tumor ascites diverted the effector activity of this and other chimeric receptor-transduced T cells away from targeted tumor cells. CONCLUSIONS: T cells redirected against FSHR+ tumor cells with full-length FSH represent a promising therapeutic alternative against a broad range of ovarian malignancies, with negligible toxicity even in the presence of cognate targets in tumor-free ovaries. Clin Cancer Res; 23(2); 441-53. ©2016 AACR.


Immunotherapy , Ovarian Neoplasms/therapy , Receptors, FSH/immunology , T-Lymphocytes/immunology , Animals , Ascites/immunology , Ascites/pathology , Exosomes/immunology , Exosomes/pathology , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunohistochemistry , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Receptors, Antigen, T-Cell/immunology , Receptors, FSH/genetics , Xenograft Model Antitumor Assays
15.
Cancer Discov ; 7(1): 72-85, 2017 01.
Article En | MEDLINE | ID: mdl-27694385

The role of estrogens in antitumor immunity remains poorly understood. Here, we show that estrogen signaling accelerates the progression of different estrogen-insensitive tumor models by contributing to deregulated myelopoiesis by both driving the mobilization of myeloid-derived suppressor cells (MDSC) and enhancing their intrinsic immunosuppressive activity in vivo Differences in tumor growth are dependent on blunted antitumor immunity and, correspondingly, disappear in immunodeficient hosts and upon MDSC depletion. Mechanistically, estrogen receptor alpha activates the STAT3 pathway in human and mouse bone marrow myeloid precursors by enhancing JAK2 and SRC activity. Therefore, estrogen signaling is a crucial mechanism underlying pathologic myelopoiesis in cancer. Our work suggests that new antiestrogen drugs that have no agonistic effects may have benefits in a wide range of cancers, independently of the expression of estrogen receptors in tumor cells, and may synergize with immunotherapies to significantly extend survival. SIGNIFICANCE: Ablating estrogenic activity delays malignant progression independently of the tumor cell responsiveness, owing to a decrease in the mobilization and immunosuppressive activity of MDSCs, which boosts T-cell-dependent antitumor immunity. Our results provide a mechanistic rationale to block estrogen signaling with newer antagonists to boost the effectiveness of anticancer immunotherapies. Cancer Discov; 7(1); 72-85. ©2016 AACR.See related commentary by Welte et al., p. 17This article is highlighted in the In This Issue feature, p. 1.


Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , Janus Kinase 2/metabolism , MCF-7 Cells , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Transplantation , Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , src-Family Kinases/metabolism
16.
Cancer Res ; 76(21): 6253-6265, 2016 11 01.
Article En | MEDLINE | ID: mdl-27803104

Targeted therapies elicit seemingly paradoxical and poorly understood effects on tumor immunity. Here, we show that the MEK inhibitor trametinib abrogates cytokine-driven expansion of monocytic myeloid-derived suppressor cells (mMDSC) from human or mouse myeloid progenitors. MEK inhibition also reduced the production of the mMDSC chemotactic factor osteopontin by tumor cells. Together, these effects reduced mMDSC accumulation in tumor-bearing hosts, limiting the outgrowth of KRas-driven breast tumors, even though trametinib largely failed to directly inhibit tumor cell proliferation. Accordingly, trametinib impeded tumor progression in vivo through a mechanism requiring CD8+ T cells, which was paradoxical given the drug's reported ability to inhibit effector lymphocytes. Confirming our observations, adoptive transfer of tumor-derived mMDSC reversed the ability of trametinib to control tumor growth. Overall, our work showed how the effects of trametinib on immune cells could partly explain its effectiveness, distinct from its activity on tumor cells themselves. More broadly, by providing a more incisive view into how MEK inhibitors may act against tumors, our findings expand their potential uses to generally block mMDSC expansion, which occurs widely in cancers to drive their growth and progression. Cancer Res; 76(21); 6253-65. ©2016 AACR.


Antineoplastic Agents/pharmacology , Mutation , Myelopoiesis/drug effects , Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/genetics , Pyridones/pharmacology , Pyrimidinones/pharmacology , T-Lymphocytes/physiology , Animals , Cell Line, Tumor , Female , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/physiology , Neoplasms/genetics , Neoplasms/physiopathology , Osteopontin/biosynthesis
17.
Cell Rep ; 16(11): 2829-2837, 2016 09 13.
Article En | MEDLINE | ID: mdl-27626654

Restoration of anti-tumor immunity by blocking PD-L1 signaling through the use of antibodies has proven to be beneficial in cancer therapy. Here, we show that BET bromodomain inhibition suppresses PD-L1 expression and limits tumor progression in ovarian cancer. CD274 (encoding PD-L1) is a direct target of BRD4-mediated gene transcription. In mouse models, treatment with the BET inhibitor JQ1 significantly reduced PD-L1 expression on tumor cells and tumor-associated dendritic cells and macrophages, which correlated with an increase in the activity of anti-tumor cytotoxic T cells. The BET inhibitor limited tumor progression in a cytotoxic T-cell-dependent manner. Together, these data demonstrate a small-molecule approach to block PD-L1 signaling. Given the fact that BET inhibitors have been proven to be safe with manageable reversible toxicity in clinical trials, our findings indicate that pharmacological BET inhibitors represent a treatment strategy for targeting PD-L1 expression.


Azepines/pharmacology , B7-H1 Antigen/genetics , Immunity/drug effects , Neoplasms/immunology , Neoplasms/metabolism , Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , B7-H1 Antigen/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Disease Progression , Female , Humans , Mice, Inbred C57BL , Neoplasms/pathology , Nuclear Proteins/metabolism , Proteins/metabolism , T-Lymphocytes, Cytotoxic/drug effects , Time Factors , Transcription Factors/metabolism , Transcription, Genetic/drug effects
18.
Pharmacol Ther ; 164: 97-104, 2016 08.
Article En | MEDLINE | ID: mdl-27118338

Dendritic cells (DCs) with robust immunosuppressive activity are commonly found in the microenvironment of advanced solid tumors. These innate immune cells are generically termed regulatory DCs and include various subsets such as plasmacytoid, conventional and monocyte-derived/inflammatory populations whose normal function is subverted by tumor-derived signals. This review summarizes recent findings on the nature and function of regulatory DCs, their relationship with other myeloid subsets and unique therapeutic opportunities to abrogate malignant progression through their targeting.


Dendritic Cells/metabolism , Neoplasms/physiopathology , Animals , Dendritic Cells/immunology , Dinoprostone/metabolism , Humans , Immune Tolerance/immunology , Macrophages/metabolism , Matrix Attachment Region Binding Proteins/biosynthesis , Myelopoiesis/immunology , Nanostructures , Neoplasms/immunology , Transforming Growth Factor beta/metabolism
19.
Cancer Res ; 76(9): 2561-72, 2016 05 01.
Article En | MEDLINE | ID: mdl-26980764

Many signal transduction inhibitors are being developed for cancer therapy target pathways that are also important for the proper function of antitumor lymphocytes, possibly weakening their therapeutic effects. Here we show that most inhibitors targeting multiple signaling pathways have especially strong negative effects on T-cell activation at their active doses on cancer cells. In particular, we found that recently approved MEK inhibitors displayed potent suppressive effects on T cells in vitro However, these effects could be attenuated by certain cytokines that can be administered to cancer patients. Among them, clinically available IL15 superagonists, which can activate PI3K selectively in T lymphocytes, synergized with MEK inhibitors in vivo to elicit potent and durable antitumor responses, including by a vaccine-like effect that generated resistance to tumor rechallenge. Our work identifies a clinically actionable approach to overcome the T-cell-suppressive effects of MEK inhibitors and illustrates how to reconcile the deficiencies of signal transduction inhibitors, which impede desired immunologic effects in vivo Cancer Res; 76(9); 2561-72. ©2016 AACR.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Neoplasms, Experimental/pathology , Proteins/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Flow Cytometry , High-Throughput Screening Assays , Humans , Interleukin-15 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Recombinant Fusion Proteins , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
20.
Cell Rep ; 14(7): 1774-1786, 2016 Feb 23.
Article En | MEDLINE | ID: mdl-26876172

Special AT-rich sequence-binding protein 1 (Satb1) governs genome-wide transcriptional programs. Using a conditional knockout mouse, we find that Satb1 is required for normal differentiation of conventional dendritic cells (DCs). Furthermore, Satb1 governs the differentiation of inflammatory DCs by regulating major histocompatibility complex class II (MHC II) expression through Notch1 signaling. Mechanistically, Satb1 binds to the Notch1 promoter, activating Notch expression and driving RBPJ occupancy of the H2-Ab1 promoter, which activates MHC II transcription. However, tumor-driven, unremitting expression of Satb1 in activated Zbtb46(+) inflammatory DCs that infiltrate ovarian tumors results in an immunosuppressive phenotype characterized by increased secretion of tumor-promoting Galectin-1 and IL-6. In vivo silencing of Satb1 in tumor-associated DCs reverses their tumorigenic activity and boosts protective immunity. Therefore, dynamic fluctuations in Satb1 expression govern the generation and immunostimulatory activity of steady-state and inflammatory DCs, but continuous Satb1 overexpression in differentiated DCs converts them into tolerogenic/pro-inflammatory cells that contribute to malignant progression.


Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/immunology , Matrix Attachment Region Binding Proteins/immunology , Ovarian Neoplasms/immunology , Animals , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , Dendritic Cells/pathology , Female , Galectin 1/genetics , Galectin 1/immunology , Histocompatibility Antigens Class II/genetics , Histones/genetics , Histones/immunology , Humans , Immune Tolerance , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Knockout , Neoplasm Transplantation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/immunology
...