Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Epigenomics ; 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530086

ABSTRACT

Aim: Castration-resistant prostate cancer (CRPC) eventually becomes resistant to androgen receptor pathway inhibitors like enzalutamide. Immunotherapy also fails in CRPC. We propose a new approach to simultaneously revert enzalutamide resistance and rewire anti-tumor immunity. Methods: We investigated in vitro and in subcutaneous and spontaneous mouse models the effects of combining enzalutamide and GSK-126, a drug inhibiting the epigenetic modulator EZH2. Results: Enzalutamide and GSK-126 synergized to reduce CRPC growth, also restraining tumor neuroendocrine differentiation. The anti-tumor activity was lost in immunodeficient mice. Indeed, the combination treatment awoke cytotoxic activity and IFN-γ production of tumor-specific CD8+ T lymphocytes. Conclusion: These results promote the combination of enzalutamide and GSK-126 in CRPC, also offering new avenues for immunotherapy in prostate cancer.


Prostate cancer depends on hormones called androgens for its growth. Therefore, hormonal therapies are commonly used. However, the tumor often does not respond to these treatments and new therapeutic approaches are needed. Here, using cell and mouse models, we have tested a new combination between hormone therapy and a drug that restrains an enzyme regulating gene expression. Our results have shown that this combination therapy not only reduces the growth of the tumor but also stops it from becoming more aggressive. This is really important because aggressive prostate cancer is much harder to treat. We have also found that this approach helps the immune system recognizing and attacking cancer cells. More research is needed to identify the mechanism of action of this treatment. However, our findings suggest that this approach could pave the way for new therapeutic strategies, including using immunotherapy, typically unsuccessful in treating prostate cancer.

2.
Pharmaceutics ; 15(6)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37376140

ABSTRACT

Tumor development, progression, and resistance to therapies are influenced by the interactions between tumor cells and the surrounding microenvironment, comprising fibroblasts, immune cells, and extracellular matrix proteins. In this context, mast cells (MCs) have recently emerged as important players. Yet, their role is still controversial, as MCs can exert pro- or anti-tumor functions in different tumor types depending on their location within or around the tumor mass and their interaction with other components of the tumor microenvironment. In this review, we describe the main aspects of MC biology and the different contribution of MCs in promoting or inhibiting cancer growth. We then discuss possible therapeutic strategies aimed at targeting MCs for cancer immunotherapy, which include: (1) targeting c-Kit signaling; (2) stabilizing MC degranulation; (3) triggering activating/inhibiting receptors; (4) modulating MC recruitment; (5) harnessing MC mediators; (6) adoptive transferring of MCs. Such strategies should aim to either restrain or sustain MC activity according to specific contexts. Further investigation would allow us to better dissect the multifaceted roles of MCs in cancer and tailor novel approaches for an "MC-guided" personalized medicine to be used in combination with conventional anti-cancer therapies.

3.
Expert Rev Anticancer Ther ; 23(2): 135-145, 2023 02.
Article in English | MEDLINE | ID: mdl-36803369

ABSTRACT

INTRODUCTION: Oncological treatments are changing rapidly due to the advent of several targeted anticancer drugs and regimens. The primary new area of research in oncological medicine is the implementation of a combination of novel therapies and standard care. In this scenario, radioimmunotherapy is one of the most promising fields, as proven by the exponential growth of publications in this context during the last decade. AREAS COVERED: This review provides an overview of the synergistic use of radiotherapy and immunotherapy and addresses questions like the importance of this subject, aspects clinicians look for in patients to administer this combined therapy, individuals who would benefit the most from this treatment, how to achieve abscopal effect and when does radio-immunotherapy become standard clinical practice. EXPERT OPINION: Answers to these queries generate further issues that need to be addressed and solved. The abscopal and bystander effects are not utopia, rather physiological phenomena that occur in our bodies. Nevertheless, substantial evidence regarding the combination of radioimmunotherapy is lacking. In conclusion, joining forces and finding answers to all these open questions is of paramount importance.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Neoplasms/radiotherapy , Immunotherapy , Radioimmunotherapy , Combined Modality Therapy
4.
Cancer Res ; 83(1): 117-129, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36318106

ABSTRACT

Cancer is a systemic disease able to reprogram the bone marrow (BM) niche towards a protumorigenic state. The impact of cancer on specific BM subpopulations can qualitatively differ according to the signals released by the tumor, which can vary on the basis of the tissue of origin. Using a spontaneous model of mammary carcinoma, we identified BM mesenchymal stem cells (MSC) as the first sensors of distal cancer cells and key mediators of BM reprogramming. Through the release of IL1B, BM MSCs induced transcriptional upregulation and nuclear translocation of the activating transcription factor 3 (ATF3) in hematopoietic stem cells. ATF3 in turn promoted the formation of myeloid progenitor clusters and sustained myeloid cell differentiation. Deletion of Atf3 specifically in the myeloid compartment reduced circulating monocytes and blocked their differentiation into tumor-associated macrophages. In the peripheral blood, the association of ATF3 expression in CD14+ mononuclear cells with the expansion CD11b+ population was able to discriminate between women with malignant or benign conditions at early diagnosis. Overall, this study identifies the IL1B/ATF3 signaling pathway in the BM as a functional step toward the establishment of a tumor-promoting emergency myelopoiesis, suggesting that ATF3 could be tested in a clinical setting as a circulating marker of early transformation and offering the rationale for testing the therapeutic benefits of IL1B inhibition in patients with breast cancer. Significance: Bone marrow mesenchymal stem cells respond to early breast tumorigenesis by upregulating IL1B to promote ATF3 expression in hematopoietic stem cells and to induce myeloid cell differentiation that supports tumor development.


Subject(s)
Bone Marrow , Breast Neoplasms , Humans , Female , Bone Marrow/pathology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Breast Neoplasms/pathology , Hematopoietic Stem Cells/metabolism , Cell Transformation, Neoplastic/metabolism , Bone Marrow Cells/metabolism
5.
Front Immunol ; 13: 987639, 2022.
Article in English | MEDLINE | ID: mdl-36203609

ABSTRACT

PD-L1 in tumor cells is the only used biomarker for anti PD1/PD-L1 immune-checkpoints inhibitors (ICI) in Non Small Cell Lung Cancer (NSCLC) patients. However, this parameter is inaccurate to predict response, especially in patients with low tumor PD-L1. Here, we evaluated circulating EVs as possible biomarkers for ICI in advanced NSCLC patients with low tumoral PD-L1. EVs were isolated from plasma of 64 PD-L1 low, ICI-treated NSCLC patients, classified either as responders (R; complete or partial response by RECIST 1.1) or non-responders (NR). EVs were characterized following MISEV guidelines and by flow cytometry. T cells from healthy donors were triggered in vitro using patients' EVs. Unsupervised statistical approach was applied to correlate EVs' and patients' features to clinical response. R-EVs showed higher levels of tetraspanins (CD9, CD81, CD63) than NR-EVs, significantly associated to better overall response rate (ORR). In multivariable analysis CD81-EVs correlated with ORR. Unsupervised analysis revealed a cluster of variables on EVs, including tetraspanins, significantly associated with ORR and improved survival. R-EVs expressed more costimulatory molecules than NR-EVs although both increased T cell proliferation and partially, activation. Tetraspanins levels on EVs could represent promising biomarkers for ICI response in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Extracellular Vesicles , Lung Neoplasms , B7-H1 Antigen , Biomarkers , Carcinoma, Non-Small-Cell Lung/pathology , Extracellular Vesicles/pathology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/pathology , Tetraspanin 28 , Tetraspanins
6.
Elife ; 112022 04 26.
Article in English | MEDLINE | ID: mdl-35471185

ABSTRACT

Neutrophil extracellular traps (NETs) are web-like chromatin structures composed by dsDNA and histones, decorated with antimicrobial proteins. Their interaction with dendritic cells (DCs) allows DC activation and maturation toward presentation of NET-associated antigens. Differently from other types of cell death that imply protein denaturation, NETosis preserves the proteins localized onto the DNA threads for proper enzymatic activity and conformational status, including immunogenic epitopes. Besides neutrophils, leukemic cells can release extracellular traps displaying leukemia-associated antigens, prototypically mutant nucleophosmin (NPMc+) that upon mutation translocates from nucleolus to the cytoplasm localizing onto NET threads. We tested NPMc+ immunogenicity through a NET/DC vaccine to treat NPMc-driven myeloproliferation in transgenic and transplantable models. Vaccination with DC loaded with NPMc+ NET (NPMc+ NET/DC) reduced myeloproliferation in transgenic mice, favoring the development of antibodies to mutant NPMc and the induction of a CD8+ T-cell response. The efficacy of this vaccine was also tested in mixed NPMc/WT bone marrow (BM) chimeras in a competitive BM transplantation setting, where the NPMc+ NET/DC vaccination impaired the expansion of NPMc+ in favor of WT myeloid compartment. NPMc+ NET/DC vaccination also achieved control of an aggressive leukemia transduced with mutant NPMc, effectively inducing an antileukemia CD8 T-cell memory response.


Subject(s)
Extracellular Traps , Leukemia , Animals , Extracellular Traps/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Vaccination
7.
Cancers (Basel) ; 13(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34439233

ABSTRACT

BACKGROUND: A combination of TLR9 agonists and an anti-PD-1 antibody has been reported to be effective in immunocompetent mice but the role of innate immunity has not yet been completely elucidated. Therefore, we investigated the contribution of the innate immune system to this combinatorial immunotherapeutic regimens using an immunodeficient mouse model in which the effector functions of innate immunity can clearly emerge without any interference from T lymphocytes. METHODS: Athymic mice xenografted with IGROV-1 human ovarian cells, reported to be sensitive to TLR9 agonist therapy, were treated with cytosine-guanine (CpG)-oligodeoxynucleotides (ODNs), an anti-PD-1 antibody or their combination. RESULTS: We found that PD-1 blockade dampened CpG-ODN antitumor activity. In vitro studies indicated that the interaction between the anti-PD-1 antibody fragment crystallizable (Fc) domain and macrophage Fc receptors caused these immune cells to acquire an immunoregulatory phenotype, contributing to a decrease in the efficacy of CpG-ODNs. Accordingly, in vivo macrophage depletion abrogated the detrimental effect exerted by the anti-PD-1 antibody. CONCLUSION: Our data suggest that if TLR signaling is active in macrophages, coadministration of an anti-PD-1 antibody can reprogram these immune cells towards a polarization state able to negatively affect the immune response and eventually promote tumor growth.

8.
Cancer Res ; 81(16): 4257-4274, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34185677

ABSTRACT

Fatal neuroendocrine differentiation (NED) of castration-resistant prostate cancer is a recurrent mechanism of resistance to androgen deprivation therapies (ADT) and antiandrogen receptor pathway inhibitors (ARPI) in patients. The design of effective therapies for neuroendocrine prostate cancer (NEPC) is complicated by limited knowledge of the molecular mechanisms governing NED. The paucity of acquired genomic alterations and the deregulation of epigenetic and transcription factors suggest a potential contribution from the microenvironment. In this context, whether ADT/ARPI induces stromal cells to release NED-promoting molecules and the underlying molecular networks are unestablished. Here, we utilized transgenic and transplantable mouse models and coculture experiments to unveil a novel tumor-stroma cross-talk that is able to induce NED under the pressure of androgen deprivation. Castration induced upregulation of GRP78 in tumor cells, which triggers miR29-b-mediated downregulation of the matricellular protein SPARC in the nearby stroma. SPARC downregulation enabled stromal cells to release IL6, a known inducer of NED. A drug that targets GRP78 blocked NED in castrated mice. A public, human NEPC gene expression dataset showed that Hspa5 (encoding for GRP78) positively correlates with hallmarks of NED. Finally, prostate cancer specimens from patients developing local NED after ADT showed GRP78 upregulation in tumor cells and SPARC downregulation in the stroma. These results point to GRP78 as a potential therapeutic target and to SPARC downregulation in stromal cells as a potential early biomarker of tumors undergoing NED. SIGNIFICANCE: Tumor-stroma cross-talk promotes neuroendocrine differentiation in prostate cancer in response to hormone therapy via a GRP78/SPARC/IL6 axis, providing potential therapeutic targets and biomarkers for neuroendocrine prostate cancer.


Subject(s)
Down-Regulation , Osteonectin/biosynthesis , Prostatic Neoplasms/metabolism , Stromal Cells/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Line, Tumor , Coculture Techniques , Endoplasmic Reticulum Chaperone BiP/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Neuroendocrine Cells/metabolism , Transgenes , Tumor Microenvironment
9.
Front Immunol ; 12: 662048, 2021.
Article in English | MEDLINE | ID: mdl-34084166

ABSTRACT

Background: Within the bone marrow (BM), mature T cells are maintained under homeostatic conditions to facilitate proper hematopoietic development. This homeostasis depends upon a peculiar elevated frequency of regulatory T cells (Tregs) and immune regulatory activities from BM-mesenchymal stem cells (BM-MSCs). In response to BM transplantation (BMT), the conditioning regimen exposes the BM to a dramatic induction of inflammatory cytokines and causes an unbalanced T-effector (Teff) and Treg ratio. This imbalance negatively impacts hematopoiesis, particularly in regard to B-cell lymphopoiesis that requires an intact cross-talk between BM-MSCs and Tregs. The mechanisms underlying the ability of BM-MSCs to restore Treg homeostasis and proper B-cell development are currently unknown. Methods: We studied the role of host radio-resistant cell-derived CD40 in restoring Teff/Treg homeostasis and proper B-cell development in a murine model of BMT. We characterized the host cellular source of CD40 and performed radiation chimera analyses by transplanting WT or Cd40-KO with WT BM in the presence of T-reg and co-infusing WT or - Cd40-KO BM-MSCs. Residual host and donor T cell expansion and activation (cytokine production) and also the expression of Treg fitness markers and conversion to Th17 were analyzed. The presence of Cd40+ BM-MSCs was analyzed in a human setting in correlation with the frequency of B-cell precursors in patients who underwent HSCT and variably developed acute graft-versus-host (aGVDH) disease. Results: CD40 expression is nearly undetectable in the BM, yet a Cd40-KO recipient of WT donor chimera exhibited impaired B-cell lymphopoiesis and Treg development. Lethal irradiation promotes CD40 and OX40L expression in radio-resistant BM-MSCs through the induction of pro-inflammatory cytokines. OX40L favors Teff expansion and activation at the expense of Tregs; however, the expression of CD40 dampens OX40L expression and restores Treg homeostasis, thus facilitating proper B-cell development. Indeed, in contrast to dendritic cells in secondary lymphoid organs that require CD40 triggers to express OX40L, BM-MSCs require CD40 to inhibit OX40L expression. Conclusions: CD40+ BM-MSCs are immune regulatory elements within BM. Loss of CD40 results in uncontrolled T cell activation due to a reduced number of Tregs, and B-cell development is consequently impaired. GVHD provides an example of how a loss of CD40+ BM-MSCs and a reduction in B-cell precursors may occur in a human setting.


Subject(s)
Bone Marrow Transplantation , Bone Marrow/immunology , CD40 Antigens/genetics , Gene Expression Regulation/immunology , Homeostasis/immunology , Mesenchymal Stem Cells/immunology , OX40 Ligand/genetics , Stress, Physiological/immunology , Adult , Aged , Animals , Bone Marrow/physiology , Bone Marrow Cells/immunology , CD40 Antigens/immunology , Female , Homeostasis/genetics , Humans , Lymphocyte Activation/immunology , Male , Mesenchymal Stem Cell Transplantation , Mice , Middle Aged , OX40 Ligand/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Conditioning , Young Adult
10.
Mol Oncol ; 15(7): 1921-1941, 2021 07.
Article in English | MEDLINE | ID: mdl-33793068

ABSTRACT

Metastatic neuroendocrine prostate cancer (NEPC) is a highly aggressive disease, whose incidence is rising. Long noncoding RNAs (lncRNAs) represent a large family of disease- and tissue-specific transcripts, most of which are still functionally uncharacterized. Thus, we set out to identify the highly conserved lncRNAs that play a central role in NEPC pathogenesis. To this end, we performed transcriptomic analyses of donor-matched patient-derived xenograft models (PDXs) with immunohistologic features of prostate adenocarcinoma (AR+ /PSA+ ) or NEPC (AR- /SYN+ /CHGA+ ) and through differential expression analyses identified lncRNAs that were upregulated upon neuroendocrine transdifferentiation. These genes were prioritized for functional assessment based on the level of conservation in vertebrates. Here, LINC00261 emerged as the top gene with over 3229-fold upregulation in NEPC. Consistently, LINC00261 expression was significantly upregulated in NEPC specimens in multiple patient cohorts. Knockdown of LINC00261 in PC-3 cells dramatically attenuated its proliferative and metastatic abilities, which are explained by parallel downregulation of CBX2 and FOXA2 through distinct molecular mechanisms. In the cell cytoplasm, LINC00261 binds to and sequesters miR-8485 from targeting the CBX2 mRNA, while inside the nucleus, LINC00261 functions as a transcriptional scaffold to induce SMAD-driven expression of the FOXA2 gene. For the first time, these results demonstrate hyperactivation of the LINC00261-CBX2-FOXA2 axes in NEPC to drive proliferation and metastasis, and that LINC00261 may be utilized as a therapeutic target and a biomarker for this incurable disease.


Subject(s)
Prostatic Neoplasms , RNA, Long Noncoding , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cytoplasm/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
11.
Front Immunol ; 12: 622001, 2021.
Article in English | MEDLINE | ID: mdl-33737929

ABSTRACT

A relevant fraction of castration-resistant prostate cancers (CRPC) evolve into fatal neuroendocrine (NEPC) tumors in resistance to androgen deprivation and/or inhibitors of androgen receptor pathway. Therefore, effective drugs against both CRPC and NEPC are needed. We have previously described a dual role of mast cells (MCs) in prostate cancer, being capable to promote adenocarcinoma but also to restrain NEPC. This finding suggests that a molecule targeting both MCs and NEPC cells could be effective against prostate cancer. Using an in silico drug repurposing approach, here we identify the antiepileptic drug levetiracetam as a potential candidate for this purpose. We found that the protein target of levetiracetam, SV2A, is highly expressed by both NEPC cells and MCs infiltrating prostate adenocarcinoma, while it is low or negligible in adenocarcinoma cells. In vitro, levetiracetam inhibited the proliferation of NEPC cells and the degranulation of MCs. In mice bearing subcutaneous tumors levetiracetam was partially active on both NEPC and adenocarcinoma, the latter effect due to the inhibition of MMP9 release by MCs. Notably, in TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mice subjected to surgical castration to mimic androgen deprivation therapy, levetiracetam reduced onset and frequency of both high grade prostatic intraepithelial neoplasia, adenocarcinoma and NEPC, thus increasing the number of cured mice showing only signs of tumor regression. Our results demonstrate that levetiracetam can directly restrain NEPC development after androgen deprivation, and that it can also block adenocarcinoma progression through the inhibition of some MCs functions. These findings open the possibility of further testing levetiracetam for the therapy of prostate cancer or of MC-mediated diseases.


Subject(s)
Anticonvulsants/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Levetiracetam/therapeutic use , Mast Cells/immunology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Prostatic Neoplasms/drug therapy , Animals , Cell Degranulation/drug effects , Cell Differentiation , Cell Proliferation/drug effects , Drug Repositioning , Gene Expression Regulation, Neoplastic , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental , Tumor Cells, Cultured
12.
Cell Immunol ; 362: 104301, 2021 04.
Article in English | MEDLINE | ID: mdl-33588246

ABSTRACT

Immuno checkpoint blockade (ICB) targeting the PD-1/PD-L1 axis is the main breakthrough for the treatment of several cancers. Nevertheless, not all patients benefit from this treatment and clinical response not always correlates with PD-L1 expression by tumor cells. The tumor microenvironment, including myeloid derived suppressor cells (MDSCs), can influence therapeutic resistance to ICB. MDSCs also express PD-L1, which contributes to their suppressive activity. Moreover, anticancer therapies including chemotherapy, radiotherapy, hormone- and targeted- therapies can modulate MDSCs recruitment, activity and PD-L1 expression. Such effects can be induced also by innovative anticancer treatments targeting metabolism and lifestyle. The outcome on cancer progression can be either positive or negative, depending on tumor type, treatment schedule and possible combination with ICB. Further studies are needed to better understand the effects of cancer therapies on the PD-1/PD-L1 axis, to identify patients that could benefit from combinatorial regimens including ICB or that rather should avoid it.


Subject(s)
B7-H1 Antigen/metabolism , Myeloid-Derived Suppressor Cells/immunology , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/immunology , B7-H1 Antigen/physiology , Cell Line, Tumor , Humans , Immunotherapy , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment
13.
Sci Rep ; 11(1): 651, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436685

ABSTRACT

We xeno-transplanted human neural precursor cells derived from induced pluripotent stem cells into the cerebellum and brainstem of mice and rats during prenatal development or the first postnatal week. The transplants survived and started to differentiate up to 1 month after birth when they were rejected by both species. Extended survival and differentiation of the same cells were obtained only when they were transplanted in NOD-SCID mice. Transplants of human neural precursor cells mixed with the same cells after partial in vitro differentiation or with a cellular extract obtained from adult rat cerebellum increased survival of the xeno-graft beyond one month. These findings are consistent with the hypothesis that the slower pace of differentiation of human neural precursors compared to that of rodents restricts induction of immune-tolerance to human antigens expressed before completion of maturation of the immune system. With further maturation the transplanted neural precursors expressed more mature antigens before the graft were rejected. Supplementation of the immature cells suspensions with more mature antigens may help to induce immune-tolerance for those antigens expressed only later by the engrafted cells.


Subject(s)
Cell Differentiation , Cerebellum/immunology , Graft Survival , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/transplantation , Stem Cell Transplantation/methods , Animals , Cells, Cultured , Cerebellum/growth & development , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neurons/cytology , Rats , Rats, Wistar , Species Specificity , Transplantation, Heterologous
14.
Front Immunol ; 11: 1820, 2020.
Article in English | MEDLINE | ID: mdl-33013832

ABSTRACT

Galectin-3 (Gal-3) is an extracellular matrix glycan-binding protein with several immunosuppressive and pro-tumor functions. The role of Galectin-3 in cancer stem-like cells (CSCs) is poorly investigated. Here, we show that prostate CSCs also colonizing prostate-draining lymph nodes of transgenic adenocarcinoma of the mouse prostate (TRAMP) mice overexpress Gal-3. Gal-3 contributes to prostate CSC-mediated immune suppression because either Gal-3 silencing in CSCs, or co-culture of CSCs and T cells in the presence of the Gal-3 inhibitor N-Acetyl-D-lactosamine rescued T cell proliferation. N-Acetyl-D-lactosamine also rescued the proliferation of T cells in prostate-draining lymph nodes of TRAMP mice affected by prostate intraepithelial neoplasia. Additionally, Gal-3 impacted prostate CSC tumorigenic and metastatic potential in vivo, as Gal-3 silencing in prostate CSCs reduced both primary tumor growth and secondary invasion. Gal-3 was also found expressed in more differentiated prostate cancer cells, but with different intracellular distribution as compared to CSCs, which suggests different functions of Gal-3 in the two cell populations. In fact, the prevalent nuclear and cytoplasmic distribution of Gal-3 in prostate CSCs made them less susceptible to apoptosis, when compared to more differentiated prostate cancer cells, in which Gal-3 was predominantly intra-cytoplasmic. Finally, we found Gal-3 expressed in human and mouse prostate intraepithelial neoplasia lesions and in metastatic lymph nodes. All together, these findings identify Gal-3 as a key molecule and a potential therapeutic target already in the early phases of prostate cancer progression and metastasis.


Subject(s)
Adenocarcinoma/metabolism , Galectin 3/metabolism , Neoplastic Stem Cells/metabolism , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , Tumor Escape , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Animals , Blood Proteins , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Galectin 3/genetics , Galectins , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neoplastic Stem Cells/immunology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/immunology , Prostatic Intraepithelial Neoplasia/secondary , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Signal Transduction , Tumor Microenvironment
16.
J Mol Med (Berl) ; 98(4): 595-612, 2020 04.
Article in English | MEDLINE | ID: mdl-32338310

ABSTRACT

Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) is increased in inflammatory bowel disease (IBD) patients, and its serum levels correlate with a worse prognosis. In the present manuscript, we show that eNAMPT serum levels are increased in IBD patients that fail to respond to anti-TNFα therapy (infliximab or adalimumab) and that its levels drop in patients that are responsive to these therapies, with values comparable with healthy subjects. Furthermore, eNAMPT administration in dinitrobenzene sulfonic acid (DNBS)-treated mice exacerbates the symptoms of colitis, suggesting a causative role of this protein in IBD. To determine the druggability of this cytokine, we developed a novel monoclonal antibody (C269) that neutralizes in vitro the cytokine-like action of eNAMPT and that reduces its serum levels in rodents. Of note, this newly generated antibody is able to significantly reduce acute and chronic colitis in both DNBS- and dextran sulfate sodium (DSS)-induced colitis. Importantly, C269 ameliorates the symptoms by reducing pro-inflammatory cytokines. Specifically, in the lamina propria, a reduced number of inflammatory monocytes, neutrophils, Th1, and cytotoxic T lymphocytes are found upon C269 treatment. Our data demonstrate that eNAMPT participates in IBD and, more importantly, that eNAMPT-neutralizing antibodies are endowed with a therapeutic potential in IBD. KEY MESSAGES: What are the new findings? Higher serum eNAMPT levels in IBD patients might decrease response to anti-TNF therapy. The cytokine-like activity of eNAMPT may be neutralized with a monoclonal antibody. Neutralization of eNAMPT ameliorates acute and chronic experimental colitis. Neutralization of eNAMPT limits the expression of IBD inflammatory signature. Neutralization of eNAMPT impairs immune cell infiltration in lamina propria.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Colitis/etiology , Cytokines/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Animals , Biomarkers , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , Cytokines/metabolism , Disease Models, Animal , Extracellular Space/metabolism , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Mice , Mucous Membrane/immunology , Mucous Membrane/metabolism
17.
Cancer Res ; 80(11): 2311-2324, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32179512

ABSTRACT

Tumor growth and development is determined by both cancer cell-autonomous and microenvironmental mechanisms, including the contribution of infiltrating immune cells. Because the role of mast cells (MC) in this process is poorly characterized and even controversial, we investigated their part in breast cancer. Crossing C57BL/6 MMTV-PyMT mice, which spontaneously develop mammary carcinomas, with MC-deficient C57BL/6-KitW-sh/W-sh (Wsh) mice, showed that MCs promote tumor growth and prevent the development of basal CK5-positive areas in favor of a luminal gene program. When cocultured with breast cancer cells in vitro, MCs hindered activation of cMET, a master regulator of the basal program, and simultaneously promoted expression and activation of estrogen receptor (ESR1/ER) and its target genes (PGR, KRT8/CK8, BCL2), which are all luminal markers. Moreover, MCs reduced ERBB2/HER2 levels, whose inhibition further increased ESR1 expression. In vivo and in silico analysis of patients with breast cancer revealed a direct correlation between MC density and ESR1 expression. In mice engrafted with HER2-positive breast cancer tumors, coinjection of MCs increased tumor engraftment and outgrowth, supporting the link between MCs and increased risk of relapse in patients with breast cancer. Together, our findings support the notion that MCs influence the phenotype of breast cancer cells by stimulating a luminal phenotype and ultimately modifying the outcome of the disease. SIGNIFICANCE: Mast cells impact breast cancer outcome by directly affecting the phenotype of tumor cells through stimulation of the estrogen receptor pathway.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Mast Cells/pathology , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/genetics , Cell Communication/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Humans , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism
18.
Int J Mol Sci ; 21(3)2020 Feb 06.
Article in English | MEDLINE | ID: mdl-32041153

ABSTRACT

Neuroendocrine prostate cancer (NEPC) can arise de novo, but much more commonly occurs as a consequence of a selective pressure from androgen deprivation therapy or androgen receptor antagonists used for prostate cancer (PCa) treatment. The process is known as neuroendocrine transdifferentiation. There is little molecular characterization of NEPCs and consequently there is no standard treatment for this kind of tumors, characterized by highly metastases rates and poor survival. For this purpose, we profiled 54 PCa samples with more than 10-years follow-up for gene and miRNA expression. We divided samples into two groups (NE-like vs. AdenoPCa), according to their clinical and molecular features. NE-like tumors were characterized by a neuroendocrine fingerprint made of known neuroendocrine markers and novel molecules, including long non-coding RNAs and components of the estrogen receptor signaling. A gene expression signature able to predict NEPC was built and tested on independently published datasets. This study identified molecular features (protein-coding, long non-coding, and microRNAs), at the time of surgery, that may anticipate the NE transformation process of prostate adenocarcinoma. Our results may contribute to improving the diagnosis and treatment of this subgroup of tumors for which traditional therapy regimens do not show beneficial effects.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Neuroendocrine/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Adenocarcinoma/drug therapy , Aged , Androgen Receptor Antagonists/adverse effects , Androgen Receptor Antagonists/therapeutic use , Androgens/metabolism , Carcinoma, Neuroendocrine/drug therapy , Cell Transdifferentiation/physiology , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Prostate/pathology , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Signal Transduction
19.
Future Oncol ; 16(6): 147-149, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31964195

ABSTRACT

The Open University's first one-day symposium on treatment-emergent neuroendocrine prostate cancer attracted world-leading figures, early career researchers and industry colleagues. The symposium proved insightful into the 'real-world' impact and current problems faced in the diagnosis and treatment of neuroendocrine prostate cancer. It was important for this meeting to take place as the incidence of neuroendocrine prostate cancer is increasing due to the widespread use of next-generation androgen deprivation drugs. The symposium discussions proposed new molecularly driven deadlines to accelerate research and improved the treatment of this deadly and poorly recognized malignancy.


Subject(s)
Neuroendocrine Tumors/therapy , Prostatic Neoplasms, Castration-Resistant/therapy , Androgen Antagonists/adverse effects , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Male , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/immunology , Neuroendocrine Tumors/pathology , Practice Guidelines as Topic , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/diagnosis , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Tumor Microenvironment/immunology
20.
Circulation ; 140(25): 2089-2107, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31661975

ABSTRACT

BACKGROUND: Inflammation is a key component of cardiac disease, with macrophages and T lymphocytes mediating essential roles in the progression to heart failure. Nonetheless, little insight exists on other immune subsets involved in the cardiotoxic response. METHODS: Here, we used single-cell RNA sequencing to map the cardiac immune composition in the standard murine nonischemic, pressure-overload heart failure model. By focusing our analysis on CD45+ cells, we obtained a higher resolution identification of the immune cell subsets in the heart, at early and late stages of disease and in controls. We then integrated our findings using multiparameter flow cytometry, immunohistochemistry, and tissue clarification immunofluorescence in mouse and human. RESULTS: We found that most major immune cell subpopulations, including macrophages, B cells, T cells and regulatory T cells, dendritic cells, Natural Killer cells, neutrophils, and mast cells are present in both healthy and diseased hearts. Most cell subsets are found within the myocardium, whereas mast cells are found also in the epicardium. Upon induction of pressure overload, immune activation occurs across the entire range of immune cell types. Activation led to upregulation of key subset-specific molecules, such as oncostatin M in proinflammatory macrophages and PD-1 in regulatory T cells, that may help explain clinical findings such as the refractivity of patients with heart failure to anti-tumor necrosis factor therapy and cardiac toxicity during anti-PD-1 cancer immunotherapy, respectively. CONCLUSIONS: Despite the absence of infectious agents or an autoimmune trigger, induction of disease leads to immune activation that involves far more cell types than previously thought, including neutrophils, B cells, Natural Killer cells, and mast cells. This opens up the field of cardioimmunology to further investigation by using toolkits that have already been developed to study the aforementioned immune subsets. The subset-specific molecules that mediate their activation may thus become useful targets for the diagnostics or therapy of heart failure.


Subject(s)
Heart Failure/immunology , Immunity, Cellular/physiology , Myocardium/immunology , Single-Cell Analysis/methods , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry/methods , Heart Failure/blood , Heart Failure/pathology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Neutrophils/immunology , Neutrophils/metabolism , Sequence Analysis, RNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...