Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Ther Adv Med Oncol ; 15: 17588359231193732, 2023.
Article in English | MEDLINE | ID: mdl-37720495

ABSTRACT

Background: Systemic inflammatory markers draw great interest as potential blood-based prognostic factors in several oncological settings. Objectives: The aim of this study is to evaluate whether neutrophil-to-lymphocyte ratio (NLR) and pan-immune-inflammation value (PIV) predict nodal pathologic complete response (pCR) after neoadjuvant chemotherapy (NAC) in node-positive (cN+) breast cancer (BC) patients. Design: Clinically, cN+ BC patients undergoing NAC followed by breast and axillary surgery were enrolled in a multicentric study from 11 Breast Units. Methods: Pretreatment blood counts were collected for the analysis and used to calculate NLR and PIV. Logistic regression analyses were performed to evaluate independent predictors of nodal pCR. Results: A total of 1274 cN+ BC patients were included. Nodal pCR was achieved in 586 (46%) patients. At multivariate analysis, low NLR [odds ratio (OR) = 0.71; 95% CI, 0.51-0.98; p = 0.04] and low PIV (OR = 0.63; 95% CI, 0.44-0.90; p = 0.01) were independently predictive of increased likelihood of nodal pCR. A sub-analysis on cN1 patients (n = 1075) confirmed the statistical significance of these variables. PIV was significantly associated with axillary pCR in estrogen receptor (ER)-/human epidermal growth factor receptor 2 (HER2)+ (OR = 0.31; 95% CI, 0.12-0.83; p = 0.02) and ER-/HER2- (OR = 0.41; 95% CI, 0.17-0.97; p = 0.04) BC patients. Conclusion: This study found that low NLR and PIV levels predict axillary pCR in patients with BC undergoing NAC. Registration: Eudract number NCT05798806.

2.
Nat Cancer ; 4(8): 1102-1121, 2023 08.
Article in English | MEDLINE | ID: mdl-37460872

ABSTRACT

Cancer is highly infiltrated by myeloid-derived suppressor cells (MDSCs). Currently available immunotherapies do not completely eradicate MDSCs. Through a genome-wide analysis of the translatome of prostate cancers driven by different genetic alterations, we demonstrate that prostate cancer rewires its secretome at the translational level to recruit MDSCs. Among different secreted proteins released by prostate tumor cells, we identified Hgf, Spp1 and Bgn as the key factors that regulate MDSC migration. Mechanistically, we found that the coordinated loss of Pdcd4 and activation of the MNK/eIF4E pathways regulate the mRNAs translation of Hgf, Spp1 and Bgn. MDSC infiltration and tumor growth were dampened in prostate cancer treated with the MNK1/2 inhibitor eFT508 and/or the AKT inhibitor ipatasertib, either alone or in combination with a clinically available MDSC-targeting immunotherapy. This work provides a therapeutic strategy that combines translation inhibition with available immunotherapies to restore immune surveillance in prostate cancer.


Subject(s)
Prostatic Neoplasms , Protein Serine-Threonine Kinases , Male , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , TOR Serine-Threonine Kinases/metabolism , Prostatic Neoplasms/genetics , Myeloid Cells/metabolism , Hepatocyte Growth Factor/metabolism , Osteopontin/metabolism , Biglycan/metabolism
3.
Cancer Cell ; 41(3): 602-619.e11, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36868226

ABSTRACT

Tumor cells promote the recruitment of immunosuppressive neutrophils, a subset of myeloid cells driving immune suppression, tumor proliferation, and treatment resistance. Physiologically, neutrophils are known to have a short half-life. Here, we report the identification of a subset of neutrophils that have upregulated expression of cellular senescence markers and persist in the tumor microenvironment. Senescent-like neutrophils express the triggering receptor expressed on myeloid cells 2 (TREM2) and are more immunosuppressive and tumor-promoting than canonical immunosuppressive neutrophils. Genetic and pharmacological elimination of senescent-like neutrophils decreases tumor progression in different mouse models of prostate cancer. Mechanistically, we have found that apolipoprotein E (APOE) secreted by prostate tumor cells binds TREM2 on neutrophils, promoting their senescence. APOE and TREM2 expression increases in prostate cancers and correlates with poor prognosis. Collectively, these results reveal an alternative mechanism of tumor immune evasion and support the development of immune senolytics targeting senescent-like neutrophils for cancer therapy.


Subject(s)
Apolipoproteins E , Prostatic Neoplasms , Animals , Humans , Male , Mice , Apolipoproteins E/metabolism , Cellular Senescence/genetics , Membrane Glycoproteins/genetics , Myeloid Cells/metabolism , Prostatic Neoplasms/metabolism , Receptors, Immunologic/metabolism , Tumor Microenvironment
4.
Nat Commun ; 12(1): 7033, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857732

ABSTRACT

Comprehensive genomic studies have delineated key driver mutations linked to disease progression for most cancers. However, corresponding transcriptional changes remain largely elusive because of the bias associated with cross-study analysis. Here, we overcome these hurdles and generate a comprehensive prostate cancer transcriptome atlas that describes the roadmap to tumor progression in a qualitative and quantitative manner. Most cancers follow a uniform trajectory characterized by upregulation of polycomb-repressive-complex-2, G2-M checkpoints, and M2 macrophage polarization. Using patient-derived xenograft models, we functionally validate our observations and add single-cell resolution. Thereby, we show that tumor progression occurs through transcriptional adaption rather than a selection of pre-existing cancer cell clusters. Moreover, we determine at the single-cell level how inhibition of EZH2 - the top upregulated gene along the trajectory - reverts tumor progression and macrophage polarization. Finally, a user-friendly web-resource is provided enabling the investigation of dynamic transcriptional perturbations linked to disease progression.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Transcriptome , Animals , Atlases as Topic , Cell Line, Tumor , Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Neoplasm Proteins/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Principal Component Analysis , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Single-Cell Analysis
5.
Front Oncol ; 11: 730131, 2021.
Article in English | MEDLINE | ID: mdl-34707988

ABSTRACT

AIM: In a consecutive series of cancer patients tested for SARS-CoV-2 infection, this retrospective population-based study investigates the risks of viral infection and death. METHODS: Malignancies were distinguished as incident or prevalent (active or inactive). Cancer management and vital status were retrieved from institutional regional databases. Comorbidities were recorded, based on Adjusted Clinical Groups (ACG). Six Resource Utilization Bands (RUBs) were also considered. Independent risk factors for SARS-CoV-2 infection and death were identified using multivariable logistic regression, considering sex, age, comorbidities and RUBs, cancer status (active versus prevalent), primary cancer site, and treatments (chemotherapy and/or radiotherapy). RESULTS: Among 34,929 cancer patients, 1,090 (3.1%) tested positive for SARS-CoV-2 infection (CoV2+ve). The risk of infection was associated with age (OR per 1-year increase=1.012; 95%CI=1.007-1.017), prevalent-inactive disease, hematologic malignancies (OR=1.33; 95%CI=1.03-1.72) and RUB (OR per 1-level increase=1.14; 95%CI=1.05-1.24). Among CoV2+ve cancer patients, the risk of death was doubled for males, and increased with age (OR per 1-year increase=1.07; 95%CI=1.06-1.09) and comorbidities (renal [OR=3.18; 95%CI=1.58-6.49], hematological [OR=3.08; 95%CI=1.49-6.50], respiratory [OR=2.87; 95%CI=1.61-5.14], endocrine [OR=2.09; 95%CI=1.25-3.51]). Lung and blood malignancies raised the mortality risk (OR=3.55; 95%CI=1.56-8.33, and OR=1.81; 95%CI=1.01-3.25 respectively). Incident or prevalent-active disease and recent chemotherapy and radiotherapy (OR=4.34; 95%CI=1.85-10.50) increased the risk of death. CONCLUSION: In a large cohort of cancer patients, the risk of SARS-CoV-2 infection was higher for those with inactive disease than in incident or prevalent-active cases. Among CoV2+ve cancer patients, active malignancies and recent multimodal therapy both significantly raised the risk of death, which increased particularly for lung cancer.

6.
Science ; 374(6564): 216-224, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34618582

ABSTRACT

The microbiota comprises the microorganisms that live in close contact with the host, with mutual benefit for both counterparts. The contribution of the gut microbiota to the emergence of castration-resistant prostate cancer (CRPC) has not yet been addressed. We found that androgen deprivation in mice and humans promotes the expansion of defined commensal microbiota that contributes to the onset of castration resistance in mice. Specifically, the intestinal microbial community in mice and patients with CRPC was enriched for species capable of converting androgen precursors into active androgens. Ablation of the gut microbiota by antibiotic therapy delayed the emergence of castration resistance even in immunodeficient mice. Fecal microbiota transplantation (FMT) from CRPC mice and patients rendered mice harboring prostate cancer resistant to castration. In contrast, tumor growth was controlled by FMT from hormone-sensitive prostate cancer patients and Prevotella stercorea administration. These results reveal that the commensal gut microbiota contributes to endocrine resistance in CRPC by providing an alternative source of androgens.


Subject(s)
Androgens/biosynthesis , Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Host Microbial Interactions , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/microbiology , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/genetics , Cell Line, Tumor , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasms, Experimental , Prevotella/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Symbiosis , Xenograft Model Antitumor Assays
7.
Blood Adv ; 5(14): 2817-2828, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34269799

ABSTRACT

Chronic lymphocytic leukemia (CLL) is caused by the progressive accumulation of mature CD5+ B cells in secondary lymphoid organs. In vitro data suggest that CD4+ T lymphocytes also sustain survival and proliferation of CLL clones through CD40L/CD40 interactions. In vivo data in animal models are conflicting. To clarify this clinically relevant biological issue, we generated genetically modified Eµ-TCL1 mice lacking CD4+ T cells (TCL1+/+AB0), CD40 (TCL1+/+CD40-/-), or CD8+ T cells (TCL1+/+TAP-/-), and we monitored the appearance and progression of a disease that mimics aggressive human CLL by flow cytometry and immunohistochemical analyses. Findings were confirmed by adoptive transfer of leukemic cells into mice lacking CD4+ T cells or CD40L or mice treated with antibodies depleting CD4 T cells or blocking CD40L/CD40 interactions. CLL clones did not proliferate in mice lacking or depleted of CD4+ T cells, thus confirming that CD4+ T cells are essential for CLL development. By contrast, CD8+ T cells exerted an antitumor activity, as indicated by the accelerated disease progression in TCL1+/+TAP-/- mice. Antigen specificity of CD4+ T cells was marginal for CLL development, because CLL clones efficiently proliferated in transgenic mice whose CD4 T cells had a T-cell receptor with CLL-unrelated specificities. Leukemic clones also proliferated when transferred into wild-type mice treated with monoclonal antibodies blocking CD40 or into CD40L-/- mice, and TCL1+/+CD40-/- mice developed frank CLL. Our data demonstrate that CD8+ T cells restrain CLL progression, whereas CD4+ T cells support the growth of leukemic clones in TCL1 mice through CD40-independent and apparently noncognate mechanisms.


Subject(s)
Dromaiidae , Leukemia, Lymphocytic, Chronic, B-Cell , Animals , CD4-Positive T-Lymphocytes , CD40 Ligand/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins
8.
Swiss Med Wkly ; 151: w20483, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33872378

ABSTRACT

Tumour-infiltrating myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid cells. The main feature of MDSCs is their ability to suppress T-cell activation and function, which leads to immunosuppressive activity in the tumour microenvironment. Higher numbers of circulating and tumour-infiltrating MDSCs have been observed in a large number of patients with various types of tumour, and are linked to poor prognosis, especially in hormone-driven tumours. Recently, it has been demonstrated that the recruitment of MDSCs in prostate cancer confers resistance to canonical endocrine therapies, opening a new approach to the treatment of hormone-driven cancer patients.


Subject(s)
Myeloid-Derived Suppressor Cells , Prostatic Neoplasms , Hormones , Humans , Male , Myeloid Cells , Tumor Microenvironment
9.
J Clin Invest ; 130(5): 2435-2450, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32250342

ABSTRACT

The mechanisms by which prostate cancer shifts from an indolent castration-sensitive phenotype to lethal castration-resistant prostate cancer (CRPC) are poorly understood. Identification of clinically relevant genetic alterations leading to CRPC may reveal potential vulnerabilities for cancer therapy. Here we find that CUB domain-containing protein 1 (CDCP1), a transmembrane protein that acts as a substrate for SRC family kinases (SFKs), is overexpressed in a subset of CRPC. Notably, CDCP1 cooperates with the loss of the tumor suppressor gene PTEN to promote the emergence of metastatic prostate cancer. Mechanistically, we find that androgens suppress CDCP1 expression and that androgen deprivation in combination with loss of PTEN promotes the upregulation of CDCP1 and the subsequent activation of the SRC/MAPK pathway. Moreover, we demonstrate that anti-CDCP1 immunoliposomes (anti-CDCP1 ILs) loaded with chemotherapy suppress prostate cancer growth when administered in combination with enzalutamide. Thus, our study identifies CDCP1 as a powerful driver of prostate cancer progression and uncovers different potential therapeutic strategies for the treatment of metastatic prostate tumors.


Subject(s)
Antigens, Neoplasm/biosynthesis , Cell Adhesion Molecules/biosynthesis , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Prostatic Neoplasms/metabolism , Up-Regulation , Animals , Antigens, Neoplasm/genetics , Benzamides , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Drosophila melanogaster , Humans , Liposomes , Male , Nitriles , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
10.
Cell Rep ; 28(8): 2156-2168.e5, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433989

ABSTRACT

Tumor-associated macrophages (TAMs) represent a major component of the tumor microenvironment supporting tumorigenesis. TAMs re-education has been proposed as a strategy to promote tumor inhibition. However, whether this approach may work in prostate cancer is unknown. Here we find that Pten-null prostate tumors are strongly infiltrated by TAMs expressing C-X-C chemokine receptor type 2 (CXCR2), and activation of this receptor through CXCL2 polarizes macrophages toward an anti-inflammatory phenotype. Notably, pharmacological blockade of CXCR2 receptor by a selective antagonist promoted the re-education of TAMs toward a pro-inflammatory phenotype. Strikingly, CXCR2 knockout monocytes infused in Ptenpc-/-; Trp53pc-/- mice differentiated in tumor necrosis factor alpha (TNF-α)-releasing pro-inflammatory macrophages, leading to senescence and tumor inhibition. Mechanistically, PTEN-deficient tumor cells are vulnerable to TNF-α-induced senescence, because of an increase of TNFR1. Our results identify TAMs as targets in prostate cancer and describe a therapeutic strategy based on CXCR2 blockade to harness anti-tumorigenic potential of macrophages against this disease.


Subject(s)
Cellular Senescence , Macrophages/pathology , Prostatic Neoplasms/pathology , Receptors, Interleukin-8B/antagonists & inhibitors , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Polarity , Chemokine CXCL2/administration & dosage , Chemokine CXCL2/pharmacology , Humans , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Staging , Neutralization Tests , PTEN Phosphohydrolase/metabolism , Receptors, Interleukin-8B/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
11.
Physiol Rev ; 99(2): 1047-1078, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30648461

ABSTRACT

Cellular senescence is a permanent state of cell cycle arrest that occurs in proliferating cells subjected to different stresses. Senescence is, therefore, a cellular defense mechanism that prevents the cells to acquire an unnecessary damage. The senescent state is accompanied by a failure to re-enter the cell cycle in response to mitogenic stimuli, an enhanced secretory phenotype and resistance to cell death. Senescence takes place in several tissues during different physiological and pathological processes such as tissue remodeling, injury, cancer, and aging. Although senescence is one of the causative processes of aging and it is responsible of aging-related disorders, senescent cells can also play a positive role. In embryogenesis and tissue remodeling, senescent cells are required for the proper development of the embryo and tissue repair. In cancer, senescence works as a potent barrier to prevent tumorigenesis. Therefore, the identification and characterization of key features of senescence, the induction of senescence in cancer cells, or the elimination of senescent cells by pharmacological interventions in aging tissues is gaining consideration in several fields of research. Here, we describe the known key features of senescence, the cell-autonomous, and noncell-autonomous regulators of senescence, and we attempt to discuss the functional role of this fundamental process in different contexts in light of the development of novel therapeutic targets.


Subject(s)
Aging/physiology , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/physiology , Neoplasms/metabolism , Wound Healing/physiology , Aging/metabolism , Animals , Cell Proliferation/physiology , Humans
12.
Nat Commun ; 9(1): 4832, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30510245

ABSTRACT

The gut microbiota has been causally linked to cancer, yet how intestinal microbes influence progression of extramucosal tumors is poorly understood. Here we provide evidence implying that Prevotella heparinolytica promotes the differentiation of Th17 cells colonizing the gut and migrating to the bone marrow (BM) of transgenic Vk*MYC mice, where they favor progression of multiple myeloma (MM). Lack of IL-17 in Vk*MYC mice, or disturbance of their microbiome delayed MM appearance. Similarly, in smoldering MM patients, higher levels of BM IL-17 predicted faster disease progression. IL-17 induced STAT3 phosphorylation in murine plasma cells, and activated eosinophils. Treatment of Vk*MYC mice with antibodies blocking IL-17, IL-17RA, and IL-5 reduced BM accumulation of Th17 cells and eosinophils and delayed disease progression. Thus, in Vk*MYC mice, commensal bacteria appear to unleash a paracrine signaling network between adaptive and innate immunity that accelerates progression to MM, and can be targeted by already available therapies.


Subject(s)
Eosinophils/immunology , Gastrointestinal Microbiome/immunology , Interleukin-17/immunology , Multiple Myeloma/immunology , Th17 Cells/immunology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Disease Progression , Eosinophils/metabolism , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Prevotella/immunology , Th17 Cells/metabolism
13.
Nature ; 559(7714): 363-369, 2018 07.
Article in English | MEDLINE | ID: mdl-29950727

ABSTRACT

Patients with prostate cancer frequently show resistance to androgen-deprivation therapy, a condition known as castration-resistant prostate cancer (CRPC). Acquiring a better understanding of the mechanisms that control the development of CRPC remains an unmet clinical need. The well-established dependency of cancer cells on the tumour microenvironment indicates that the microenvironment might control the emergence of CRPC. Here we identify IL-23 produced by myeloid-derived suppressor cells (MDSCs) as a driver of CRPC in mice and patients with CRPC. Mechanistically, IL-23 secreted by MDSCs can activate the androgen receptor pathway in prostate tumour cells, promoting cell survival and proliferation in androgen-deprived conditions. Intra-tumour MDSC infiltration and IL-23 concentration are increased in blood and tumour samples from patients with CRPC. Antibody-mediated inactivation of IL-23 restored sensitivity to androgen-deprivation therapy in mice. Taken together, these results reveal that MDSCs promote CRPC by acting in a non-cell autonomous manner. Treatments that block IL-23 can oppose MDSC-mediated resistance to castration in prostate cancer and synergize with standard therapies.


Subject(s)
Interleukin-23/antagonists & inhibitors , Interleukin-23/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Androgens/deficiency , Animals , Benzamides , Cell Proliferation , Cell Survival , Humans , Interleukin-23/blood , Interleukin-23/immunology , Male , Mice , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/immunology , Nitriles , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Receptors, Interleukin/metabolism , Signal Transduction
14.
Cell Rep ; 22(11): 3006-3020, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29539427

ABSTRACT

Heterotypic cellular and molecular interactions in the tumor microenvironment (TME) control cancer progression. Here, we show that CD1d-restricted invariant natural killer (iNKT) cells control prostate cancer (PCa) progression by sculpting the TME. In a mouse PCa model, iNKT cells restrained the pro-angiogenic and immunosuppressive capabilities of tumor-infiltrating immune cells by reducing pro-angiogenic TIE2+, M2-like macrophages (TEMs), and sustaining pro-inflammatory M1-like macrophages. iNKT cells directly contacted macrophages in the PCa stroma, and iNKT cell transfer into tumor-bearing mice abated TEMs, delaying tumor progression. iNKT cells modulated macrophages through the cooperative engagement of CD1d, Fas, and CD40, which promoted selective killing of M2-like and survival of M1-like macrophages. Human PCa aggressiveness associate with reduced intra-tumoral iNKT cells, increased TEMs, and expression of pro-angiogenic genes, underscoring the clinical significance of this crosstalk. Therefore, iNKT cells may control PCa through mechanisms involving differential macrophage modulation, which may be harnessed for therapeutically reprogramming the TME.


Subject(s)
CD40 Antigens/metabolism , Macrophages/metabolism , Natural Killer T-Cells/immunology , Prostatic Neoplasms/genetics , Animals , Disease Progression , Humans , Male , Mice , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
15.
Swiss Med Wkly ; 147: w14367, 2017.
Article in English | MEDLINE | ID: mdl-28102876

ABSTRACT

Robust scientific evidence demonstrates that senes-cence induction in cancer works as a potent weapon to eradicate tumorigenesis. Therapies that enhance senescence not only promote a stable cell growth arrest but also work as a strong stimulus for the acti-vation of the antitumour immune response. However, recent advances suggest that if senescent tumour cells are not cleared from the tumours, they may promote tumour progression and metastasis. In this article, we focus on concepts that are relevant to a pro-senescence therapeutic approach, including caveats, and we propose therapeutic strategies that involve the combined use of pro-senescence therapies with im-munotherapies to promote the clearance of senescent tumour cells. In our opinion, these approaches may avoid potential negative effects of pro-senescence therapies and may also enhance the efficacy of cur-rently available immunotherapies.


Subject(s)
Cellular Senescence , Neoplasms/immunology , Neoplasms/therapy , Cell Proliferation , Humans , Neoplastic Cells, Circulating/immunology
16.
Nat Med ; 22(12): 1411-1420, 2016 12.
Article in English | MEDLINE | ID: mdl-27841872

ABSTRACT

The cellular inhibitors of apoptosis (cIAP) 1 and 2 are amplified in about 3% of cancers and have been identified in multiple malignancies as being potential therapeutic targets as a result of their role in the evasion of apoptosis. Consequently, small-molecule IAP antagonists, such as LCL161, have entered clinical trials for their ability to induce tumor necrosis factor (TNF)-mediated apoptosis of cancer cells. However, cIAP1 and cIAP2 are recurrently homozygously deleted in multiple myeloma (MM), resulting in constitutive activation of the noncanonical nuclear factor (NF)-κB pathway. To our surprise, we observed robust in vivo anti-myeloma activity of LCL161 in a transgenic myeloma mouse model and in patients with relapsed-refractory MM, where the addition of cyclophosphamide resulted in a median progression-free-survival of 10 months. This effect was not a result of direct induction of tumor cell death, but rather of upregulation of tumor-cell-autonomous type I interferon (IFN) signaling and a strong inflammatory response that resulted in the activation of macrophages and dendritic cells, leading to phagocytosis of tumor cells. Treatment of a MM mouse model with LCL161 established long-term anti-tumor protection and induced regression in a fraction of the mice. Notably, combination of LCL161 with the immune-checkpoint inhibitor anti-PD1 was curative in all of the treated mice.


Subject(s)
Antineoplastic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Thiazoles/therapeutic use , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclophosphamide/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Female , Gene Expression/drug effects , Humans , Interferon Type I/drug effects , Interferon Type I/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Transgenic , Middle Aged , Multiple Myeloma/immunology , Neoplasm Recurrence, Local/immunology , Phagocytosis/drug effects , Phagocytosis/immunology , Thiazoles/pharmacology
17.
Cancer Res ; 76(7): 1781-91, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26869462

ABSTRACT

Angiogenesis has been postulated to be critical for the pathogenesis of multiple myeloma, a neoplastic disease characterized by abnormal proliferation of malignant plasma cells in the bone marrow (BM). Cleavage of the N- and C-terminal regions of circulating chromogranin A (CgA, CHGA), classically an antiangiogenic protein, can activate latent antiangiogenic and proangiogenic sites, respectively. In this study, we investigated the distribution of CgA-derived polypeptides in multiple myeloma patients and the subsequent implications for disease progression. We show that the ratio of pro/antiangiogenic forms of CgA is altered in multiple myeloma patients compared with healthy subjects and that this ratio is higher in BM plasma compared with peripheral plasma, suggesting enhanced local cleavage of the CgA C-terminal region. Enhanced cleavage correlated with increased VEGF and FGF2 BM plasma levels and BM microvascular density. Using the Vk*MYC mouse model of multiple myeloma, we further demonstrate that exogenously administered CgA was cleaved in favor of the proangiogenic form and was associated with increased microvessel density. Mechanistic studies revealed that multiple myeloma and proliferating endothelial cells can promote CgA C-terminal cleavage by activating the plasminogen activator/plasmin system. Moreover, cleaved and full-length forms could also counter balance the pro/antiangiogenic activity of each other in in vitro angiogenesis assays. These findings suggest that the CgA-angiogenic switch is activated in the BM of multiple myeloma patients and prompt further investigation of this CgA imbalance as a prognostic or therapeutic target. Cancer Res; 76(7); 1781-91. ©2016 AACR.


Subject(s)
Bone Marrow/pathology , Chromogranin A/genetics , Multiple Myeloma/genetics , Peptides/genetics , Animals , Female , Humans , Male , Mice , Middle Aged , Multiple Myeloma/pathology , Neovascularization, Pathologic
18.
Oncoimmunology ; 4(6): e1008850, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26155424

ABSTRACT

While multiple myeloma (MM) is almost invariably preceded by asymptomatic monoclonal gammopathy of undetermined significance (MGUS) and/or smoldering MM (SMM), the alterations of the bone marrow (BM) microenvironment that establish progression to symptomatic disease are circumstantial. Here we show that in Vk*MYC mice harboring oncogene-driven plasma cell proliferative disorder, disease appearance associated with substantial modifications of the BM microenvironment, including a progressive accumulation of both CD8+ and CD4+ T cells with a dominant T helper type 1 (Th1) response. Progression from asymptomatic to symptomatic MM was characterized by further BM accrual of T cells with reduced Th1 and persistently increased Th2 cytokine production, which associated with accumulation of CD206+Tie2+ macrophages, and increased pro-angiogenic cytokines and microvessel density (MVD). Notably, MVD was also increased at diagnosis in the BM of MGUS and SMM patients that subsequently progressed to MM when compared with MGUS and SMM that remained quiescent. These findings suggest a multistep pathogenic process in MM, in which the immune system may contribute to angiogenesis and disease progression. They also suggest initiating a large multicenter study to investigate MVD in asymptomatic patients as prognostic factor for the progression and outcome of this disease.

19.
Cancer Res ; 75(10): 2095-108, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25808872

ABSTRACT

Precociously disseminated cancer cells may seed quiescent sites of future metastasis if they can protect themselves from immune surveillance. However, there is little knowledge about how such sites might be achieved. Here, we present evidence that prostate cancer stem-like cells (CSC) can be found in histopathologically negative prostate draining lymph nodes (PDLN) in mice harboring oncogene-driven prostate intraepithelial neoplasia (mPIN). PDLN-derived CSCs were phenotypically and functionally identical to CSC obtained from mPIN lesions, but distinct from CSCs obtained from frank prostate tumors. CSC derived from either PDLN or mPIN used the extracellular matrix protein Tenascin-C (TNC) to inhibit T-cell receptor-dependent T-cell activation, proliferation, and cytokine production. Mechanistically, TNC interacted with α5ß1 integrin on the cell surface of T cells, inhibiting reorganization of the actin-based cytoskeleton therein required for proper T-cell activation. CSC from both PDLN and mPIN lesions also expressed CXCR4 and migrated in response to its ligand CXCL12, which was overexpressed in PDLN upon mPIN development. CXCR4 was critical for the development of PDLN-derived CSC, as in vivo administration of CXCR4 inhibitors prevented establishment in PDLN of an immunosuppressive microenvironment. Taken together, our work establishes a pivotal role for TNC in tuning the local immune response to establish equilibrium between disseminated nodal CSC and the immune system.


Subject(s)
Neoplastic Stem Cells/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes/immunology , Tenascin/physiology , Tumor Escape , Animals , Cell Movement , Cell Proliferation , Humans , Integrin alpha5beta1/metabolism , Lymphatic Metastasis , Lymphocyte Activation , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Prostatic Neoplasms/pathology , Stress Fibers/metabolism , Tumor Cells, Cultured
20.
Oncoimmunology ; 2(7): e25032, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24073378

ABSTRACT

Using both transplantable and oncogene-driven autochthonous tumor models challenged with dendritic cell-based vaccines, we have recently found that boosting provides a clear advantage in prophylactic settings, unless performed on an excessively tight schedule, which causes the loss of central memory T cells. In therapeutic settings, boosting turned out to be always detrimental.

SELECTION OF CITATIONS
SEARCH DETAIL
...