Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Immunology ; 171(2): 198-211, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37884280

ABSTRACT

Glioblastoma, isocitrate dehydrogenase-wildtype (GB), is the most common and aggressive primary brain malignancy with poor outcome. Immune checkpoint inhibitors (ICIs) have been tested in GB and, despite disappointing results, the identification of a small subgroup of responders underlies the need to improve our understanding of the tumour microenvironment (TME) immunity. This study aimed to determine whether the expression of selected immune checkpoints on tissue-resident memory T cells (Trm) may predict patient outcome. We conducted a single cohort observational study. Tumour samples were collected from 45 patients with histologically confirmed GB (WHO grade 4) and processed to obtain single-cell suspensions. Patients were assessed for the correlation of Trm phenotype with overall survival (OS) or progression-free survival (PFS) using multiparametric flow cytometry and uni/multivariate analyses. Levels of Trm expressing programmed cell death protein 1 (PD1) and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3) were found to be linked to clinical outcome. Low frequency of Trm expressing PD1 or TIM3 or both markers defined subgroups as independent positive prognostic factors for patient survival. On multivariate analysis, low CD8+CD103+PD1+TIM3+ Trm and Karnofsky performance status (KPS) ≥70 were confirmed to be the most predictive independent factors associated with longer OS (hazard ratios-HR [95%CI]: 0.14 [0.04-0.52] p < 0.001, 0.39 [0.16-0.96] p = 0.04, respectively). The CD8+CD103+ Trm subgroups were also age-related predictors for survival in GB.


Subject(s)
Glioblastoma , Hepatitis A Virus Cellular Receptor 2 , Humans , Programmed Cell Death 1 Receptor/metabolism , Prognosis , CD8-Positive T-Lymphocytes , Tumor Microenvironment
2.
Int J Cancer ; 153(5): 1080-1095, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37293858

ABSTRACT

BRAFV600 mutations are the most common oncogenic alterations in melanoma cells, supporting proliferation, invasion, metastasis and immune evasion. In patients, these aberrantly activated cellular pathways are inhibited by BRAFi whose potent antitumor effect and therapeutic potential are dampened by the development of resistance. Here, by using primary melanoma cell lines, generated from lymph node lesions of metastatic patients, we show that the combination of two FDA-approved drugs, the histone deacetylate inhibitor (HDCAi) romidepsin and the immunomodulatory agent IFN-α2b, reduces melanoma proliferation, long-term survival and invasiveness and overcomes acquired resistance to the BRAFi vemurafenib (VEM). Targeted resequencing revealed that each VEM-resistant melanoma cell line and the parental counterpart are characterized by a distinctive and similar genetic fingerprint, shaping the differential and specific antitumor modulation of MAPK/AKT pathways by combined drug treatment. By using RNA-sequencing and functional in vitro assays, we further report that romidepsin-IFN-α2b treatment restores epigenetically silenced immune signals, modulates MITF and AXL expression and induces both apoptosis and necroptosis in sensitive and VEM-resistant primary melanoma cells. Moreover, the immunogenic potential of drug-treated VEM-resistant melanoma cells results significantly enhanced, given the increased phagocytosis rate of these cells by dendritic cells, which in turn exhibit also a selective down-modulation of the immune checkpoint TIM-3. Overall, our results provide evidence that combined epigenetic-immune drugs can overcome VEM resistance of primary melanoma cells by oncogenic and immune pathways reprogramming, and pave the way for rapidly exploiting this combination to improve BRAFi-resistant metastatic melanoma treatment, also via reinforcement of immune checkpoint inhibitor therapy.


Subject(s)
Interferon Type I , Melanoma , Humans , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Cell Line, Tumor
3.
J Transl Med ; 13: 139, 2015 May 02.
Article in English | MEDLINE | ID: mdl-25933939

ABSTRACT

BACKGROUND: Advanced melanoma patients have an extremely poor long term prognosis and are in strong need of new therapies. The recently developed targeted therapies have resulted in a marked antitumor effect, but most responses are partial and some degree of toxicity remain the major concerns. Dendritic cells play a key role in the activation of the immune system and have been typically used as ex vivo antigen-loaded cell drugs for cancer immunotherapy. Another approach consists in intratumoral injection of unloaded DCs that can exploit the uptake of a wider array of tumor-specific and individual unique antigens. However, intratumoral immunization requires DCs endowed at the same time with properties typically belonging to both immature and mature DCs (i.e. antigen uptake and T cell priming). DCs generated in presence of interferon-alpha (IFN-DCs), due to their features of partially mature DCs, capable of efficiently up-taking, processing and cross-presenting antigens to T cells, could successfully carry out this task. Combining intratumoral immunization with tumor-destructing therapies can induce antigen release in situ, facilitating the injected DCs in triggering an antitumor immune response. METHODS: We tested in a phase I clinical study in advanced melanoma a chemo-immunotherapy approach based on unloaded IFN-DCs injected intratumorally one day after administration of dacarbazine. Primary endpoint of the study was treatment safety and tolerability. Secondary endpoints were immune and clinical responses of patients. RESULTS: Six patients were enrolled, and only three completed the treatment. The chemo-immunotherapy was well tolerated with no major side effects. Three patients showed temporary disease stabilization and two of them showed induction of T cells specific for tyrosinase, NY-ESO-1 and gp100. Of interest, one patient showing a remarkable long-term disease stabilization kept showing presence of tyrosinase specific T cells in PBMC and high infiltration of memory T cells in the tumor lesion at 21 months. CONCLUSION: We tested a chemo-immunotherapeutic approach based on IFN-DCs injected intratumorally one day after DTIC in advanced melanoma. The treatment was well tolerated, and clinical and immunological responses, including development of vitiligo, were observed, therefore warranting additional clinical studies aimed at evaluating efficacy of this approach. TRIAL REGISTRATION: Trial Registration Number not publicly available due to EudraCT regulations: https://www.clinicaltrialsregister.eu/doc/EU_CTR_FAQ.pdf.


Subject(s)
Dacarbazine/chemistry , Dendritic Cells/cytology , Drug Therapy/methods , Immunotherapy/methods , Injections, Intralesional , Interferon-alpha/metabolism , Melanoma/therapy , Adult , Aged , Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Combined Modality Therapy/methods , Female , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/cytology , Male , Membrane Proteins/metabolism , Microscopy, Confocal , Middle Aged , Monocytes/metabolism , Monophenol Monooxygenase/metabolism , Vitiligo/chemically induced , gp100 Melanoma Antigen/metabolism
4.
Cytokine Growth Factor Rev ; 19(1): 93-107, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18054517

ABSTRACT

Dendritic cells (DCs) play a crucial role in linking innate and adaptive immunity and, thus, in the generation of a protective immune response against both infectious diseases and tumors. The ability of DCs to prime and expand an immune response is regulated by signals acting through soluble mediators, mainly cytokines and chemokines. Understanding how cytokines influence DC functions and orchestrate the interactions of DCs with other immune cells is strictly instrumental to the progress in cancer immunotherapy. Herein, we will illustrate how certain cytokines and immune stimulating molecules can induce and sustain the antitumor immune response by acting on DCs. We will also discuss these cytokine-DC interactions in the light of clinical results in cancer patients.


Subject(s)
Antigens, Neoplasm/immunology , Cytokines/physiology , Dendritic Cells/immunology , Neoplasms/immunology , Adjuvants, Immunologic/physiology , Aminoquinolines/therapeutic use , Animals , Antigen Presentation , Cancer Vaccines/therapeutic use , Cells, Cultured , Clinical Trials as Topic , CpG Islands/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Imiquimod , Immunotherapy , Interferon-alpha/therapeutic use , Neoplasms/prevention & control , Oligodeoxyribonucleotides/therapeutic use , Toll-Like Receptor 9/agonists
5.
Cytokine Growth Factor Rev ; 58: 66-74, 2021 04.
Article in English | MEDLINE | ID: mdl-33071044

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic, induces severe pneumonia mainly in elderly males. Epidemiological data clearly indicate sex-based differences in disease outcomes, with men accounting for about 70 % of deaths, despite similar susceptibility to infection. It is well known that females are endowed with higher capacity to produce antibodies, which correlates with viral clearance and disease resolution in the context of SARS-Cov-2 infection. Many X-linked immune genes escape X inactivation showing biallelic expression in female immune cells, particularly in plasmacytoid dendritic cells (pDCs). PDCs are more active in females and endowed with high capability to induce IFN-α-mediated B cell activation and differentiation into antibody-producing plasma cells throughout epigenetic mechanisms linked to trained immunity. Thus, we hypothesize that following SARS-CoV-2 infection, epigenetic modifications of X-linked genes involved in pDC-mediated type I IFN (IFN-I) signaling occurs more effectively in females, for inducing neutralizing antibody response as an immune correlate driving sex-biased disease outcome.


Subject(s)
Antibody Formation , COVID-19/diagnosis , COVID-19/immunology , Interferon Type I/physiology , SARS-CoV-2/immunology , COVID-19/epidemiology , Female , Humans , Male , Pandemics , Prognosis , Sex Characteristics
6.
Front Oncol ; 10: 202, 2020.
Article in English | MEDLINE | ID: mdl-32211314

ABSTRACT

Clinical studies based on novel rationales and mechanisms of action of chemotherapy agents and cytokines can contribute to the development of new concepts and strategies of antitumor combination therapies. In previous studies, we investigated the paradoxical immunostimulating effects of some chemotherapeutics and the immunoadjuvant activity of interferon alpha (IFN-α) in preclinical and clinical models, thus unraveling novel rationales and mechanisms of action of chemotherapy agents and cytokines for cancer immunotherapy. Here, we carried out a randomized, phase II clinical trial, in which we analyzed the relapse-free (RFS) and overall survival (OS) of 34 completely resected stage III-IV melanoma patients, treated with peptide-based vaccination (Melan-A/MART-1 and NY-ESO-1) in combination with IFN-α2b, with (arm 2) or without (arm 1) dacarbazine preconditioning. All patients were included in the intention-to-treat analysis. At a median follow-up of 4.5 years (interquartile range, 15.4-81.0 months), the rates of RFS were 52.9 and 35.3% in arms 1 and 2, respectively. The 4.5-year OS rates were 68.8% in arm 1 and 62.7% in arm 2. No significant differences were observed between the two arms for both RFS and OS. Interestingly, the RFS and OS curves remained stable starting from 18 and 42 months, respectively. Grade 3 adverse events occurred in 5.9% of patients, whereas grade 4 events were not observed. Both treatments induced a significant expansion of vaccine-specific CD8+ T cells, with no correlation with the clinical outcome. However, treatment-induced increase of polyfunctionality and of interleukin 2 production by Melan-A-specific CD8+ T cells and expansion/activation of natural killer cells correlated with RFS, being observed only in nonrelapsing patients. Despite the recent availability of different therapeutic options, low-cost, low-toxic therapies with long-lasting clinical effects are still needed in patients with high-risk resected stage III/IV melanoma. The combination of peptide vaccination with IFN-α2b showed a minimal toxicity profile and resulted in encouraging RFS and OS rates, justifying further evaluation in clinical trials, which may include the use of checkpoint inhibitors to further expand the antitumor immune response and the clinical outcome. Clinical Trial Registration: https://www.clinicaltrialsregister.eu/ctr-search/search, identifier: 2008-008211-26.

7.
Int J Cancer ; 124(1): 130-9, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18839429

ABSTRACT

Combination of chemotherapy with cancer vaccines is currently regarded as a potentially valuable therapeutic approach for the treatment of some metastatic tumors, but optimal modalities remain unknown. We designed a phase I/II pilot study for evaluating the effects of dacarbazine (DTIC) on the immune response in HLA-A2(+) disease-free melanoma patients who received anticancer vaccination 1 day following chemotherapy (800 mg/mq i.v.). The vaccine, consisting of a combination of HLA-A2 restricted melanoma antigen A (Melan-A/MART-1) and gp100 analog peptides (250 microg each, i.d.), was administered in combination or not with DTIC to 2 patient groups. The combined treatment is nontoxic. The comparative immune monitoring demonstrates that patients receiving DTIC 1 day before the vaccination have a significantly improved long-lasting memory CD8(+) T cell response. Of relevance, these CD8(+) T cells recognize and lyse HLA-A2(+)/Melan-A(+) tumor cell lines. Global transcriptional analysis of peripheral blood mononuclear cells (PBMC) revealed a DTIC-induced activation of genes involved in cytokine production, leukocyte activation, immune response and cell motility that can favorably condition tumor antigen-specific CD8(+) T cell responses. This study represents a proof in humans of a chemotherapy-induced enhancement of CD8(+) memory T cell response to cancer vaccines, which opens new opportunities to design novel effective combined therapies improving cancer vaccination effectiveness.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/therapeutic use , Melanoma/drug therapy , Melanoma/immunology , Adult , CD8-Positive T-Lymphocytes/metabolism , Female , Humans , Immunotherapy/methods , Interferon-alpha/metabolism , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Neoplasm Metastasis , Pilot Projects , Treatment Outcome
8.
Cancers (Basel) ; 11(12)2019 Dec 04.
Article in English | MEDLINE | ID: mdl-31817234

ABSTRACT

The first report on the antitumor effects of interferon α/ß (IFN-I) in mice was published 50 years ago. IFN-α were the first immunotherapeutic drugs approved by the FDA for clinical use in cancer. However, their clinical use occurred at a time when most of their mechanisms of action were still unknown. These cytokines were being used as either conventional cytostatic drugs or non-specific biological response modifiers. Specific biological activities subsequently ascribed to IFN-I were poorly considered for their clinical use. Notably, a lot of the data in humans and mice underlines the importance of endogenous IFN-I, produced by both immune and tumor cells, in the control of tumor growth and in the response to antitumor therapies. While many oncologists consider IFN-I as "dead drugs", recent studies reveal new mechanisms of action with potential implications in cancer control and immunotherapy response or resistance, suggesting novel rationales for their usage in target and personalized anti-cancer treatments. In this Perspectives Article, we focus on the following aspects: (1) the added value of IFN-I for enhancing the antitumor impact of standard anticancer treatments (chemotherapy and radiotherapy) and new therapeutic approaches, such as check point inhibitors and epigenetic drugs; (2) the role of IFN-I in the control of cancer stem cells growth and its possible implications for the development of novel antitumor therapies; and (3) the role of IFN-I in the development of cancer vaccines and the intriguing therapeutic possibilities offered by in situ delivery of ex vivo IFN-stimulated dendritic cells.

9.
Clin Cancer Res ; 25(17): 5231-5241, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31171545

ABSTRACT

PURPOSE: This study was aimed at evaluating the feasibility, safety, immunologic and clinical responses in patients with follicular lymphoma treated with monocyte-derived dendritic cells generated in the presence of IFNα and GM-CSF (IFN-DC) in combination with low doses of rituximab. PATIENTS AND METHODS: Firstly, we analyzed in vitro and in vivo the immunologic properties of IFN-DC against follicular lymphoma. Thus, we performed a phase I trial in 8 patients with refractory and relapsed follicular lymphoma based on sequential intranodal injections of low-dose of rituximab and unloaded IFN-DC and report the safety, clinical, and immunologic results of the enrolled patients. RESULTS: Preclinical studies indicated that IFN-DC can synergize with rituximab leading to increased cytotoxicity and T-cell tumor infiltration. The clinical evaluation showed that the combined treatment was totally safe. The overall response rate was 50%, PET-negative complete response rate 37%, and remission is still ongoing in 2/4 of responding patients (median follow-up 26 months, range 11-47). Notably, following the combined therapy all patients showed induction/enhancement of T-cell responses by CD107 degranulation or IFNγ ELISPOT assay against patient-specific tumor IGHV sequences. CONCLUSIONS: These results represent the proof-of-principle on the effectiveness of unloaded IFN-DC in inducing durable clinical responses and promoting induction of tumor-specific peripheral T cells, thus suggesting the occurrence of an effective endogenous antitumor vaccination. The overall findings indicate that some unique properties of IFN-DC can be successfully exploited to induce/enhance antitumor responses, thus representing a valuable antitumor strategy for novel and more effective combination therapies in patients with cancer.


Subject(s)
Dendritic Cells/transplantation , Immunotherapy, Adoptive/methods , Lymphoma, Follicular/therapy , Neoplasm Recurrence, Local/therapy , Rituximab/administration & dosage , Adult , Aged , Animals , Antineoplastic Agents, Immunological/administration & dosage , Combined Modality Therapy , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Resistance, Neoplasm , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Injections, Intralymphatic , Interferon-alpha/pharmacology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Remission Induction , Salvage Therapy , Xenograft Model Antitumor Assays
10.
Cancer Res ; 66(9): 4943-51, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651452

ABSTRACT

The use of IFN-alpha in clinical oncology has generally been based on the rationale of exploiting its antiproliferative and antiangiogenic activities. However, IFN-alpha also exhibits enhancing effects on T-cell and dendritic cell functions, which may suggest a novel use as a vaccine adjuvant. We have carried out a pilot phase I-II trial to determine the effects of IFN-alpha, administered as an adjuvant of Melan-A/MART-1:26-35(27L) and gp100:209-217(210M) peptides, on immune responses in stage IV melanoma patients. In five of the seven evaluable patients, a consistent enhancement of CD8(+) T cells recognizing modified and native MART-1 and gp100 peptides and MART-1(+)gp100(+) melanoma cells was observed. Moreover, vaccination induced an increase in CD8(+) T-cell binding to HLA tetramers containing the relevant peptides and an increased frequency of CD45RA(+)CCR7(-) (terminally differentiated effectors) and CD45RA(-)CCR7(-) (effector memory) cells. In all patients, treatment augmented significantly the percentage of CD14(+) monocytes and particularly of the CD14(+)CD16(+) cell fraction. An increased expression of CD40 and CD86 costimulatory molecules in monocytes was also observed. Notably, postvaccination monocytes from two of the three patients showing stable disease or long disease-free survival showed an enhanced antigen-presenting cell function and capability to secrete IP10/CXCL10 when tested in mixed leukocyte reaction assays, associated to a boost of antigen and melanoma-specific CD8(+) T cells. Although further clinical studies are needed to show the adjuvant activity of IFN-alpha, the present data represent an important starting point for considering a new clinical use of IFN-alpha and new immunologic end points, potentially predictive of clinical response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Interferon-alpha/therapeutic use , Melanoma/therapy , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Adjuvants, Immunologic/therapeutic use , Antigen Presentation , Antigens, Neoplasm , Cancer Vaccines/immunology , Dendritic Cells/cytology , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Immunophenotyping , Lymphocyte Activation , MART-1 Antigen , Melanoma/immunology , Melanoma/pathology , Monocytes/cytology , Monocytes/immunology , Neoplasm Staging , Pilot Projects , gp100 Melanoma Antigen
11.
Front Immunol ; 9: 552, 2018.
Article in English | MEDLINE | ID: mdl-29619026

ABSTRACT

Nowadays, several types of tumors can benefit from the new frontier of immunotherapy, due to the recent increasing knowledge of the role of the immune system in cancer control. Among the new therapeutic strategies, there is the immune checkpoint blockade (ICB), able to restore an efficacious antitumor immunity and significantly prolong the overall survival (OS) of patients with advanced tumors such as melanoma and non-small cell lung cancer (NSCLC). Despite the impressive efficacy of these agents in some patients, treatment failure and resistance are frequently observed. In this regard, the signaling governed by IFN type I (IFN-I) has emerged as pivotal in orchestrating host defense. This pathway displays different activation between sexes, thus potentially contributing to sexual dimorphic differences in the immune responses to immunotherapy. This perspective article aims to critically consider the immune signals, with particular attention to IFN-I, that may differently affect female and male antitumor responses upon immunotherapy.


Subject(s)
Immunotherapy , Neoplasms/therapy , Sex Characteristics , Humans , Neoplasms/immunology
12.
Biochimie ; 89(6-7): 884-93, 2007.
Article in English | MEDLINE | ID: mdl-17532550

ABSTRACT

Interferons-alpha (IFN-alpha) are pleiotropic cytokines belonging to type I IFNs, extensively used in the treatment of patients with some types of cancer and viral disease. IFN-alpha can affect tumor cell functions by multiple mechanisms. In addition, these cytokines can promote the differentiation and activity of host immune cells. Early studies in mouse tumor models showed the importance of host immune mechanisms in the generation of a long-lasting antitumor response after treatment of the animals with IFN-alpha/beta. Subsequently, an ensemble of studies based on the use of genetically modified tumor cells expressing specific IFN molecules provided important information on the host-mediated antitumor mechanisms induced by the local production of IFN-alpha. Of note, several studies have then underscored new immunomodulatory effects of IFN-alpha, including activities on T cells and dendritic cells, which may lead to IFN-induced antitumor immunity. In addition, recent reports on new immune correlates in cancer patients responding to IFN-alpha represent additional evidence on the importance of the interactions of IFN-alpha with the immune system for the generation of a durable antitumor response. On the whole, this knowledge suggests the advantage of using these cytokines as adjuvants of cancer vaccines and for the in vitro generation of highly active dendritic cells to be utilized for therapeutic vaccination of cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Immunotherapy/methods , Interferon-alpha/physiology , Neoplasms/metabolism , Animals , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Transfer Techniques , Humans , Interferon-alpha/metabolism , Models, Biological , Monocytes/metabolism , Neoplasms/therapy
13.
Cancer Immunol Res ; 5(7): 604-616, 2017 07.
Article in English | MEDLINE | ID: mdl-28615266

ABSTRACT

Colorectal cancer results from the progressive accumulation of genetic and epigenetic alterations. IFN signaling defects play an important role in the carcinogenesis process, in which the inability of IFN transcription regulatory factors (IRF) to access regulatory sequences in IFN-stimulated genes (ISG) in tumors and in immune cells may be pivotal. We reported that low-dose combination of two FDA-approved epidrugs, azacytidine (A) and romidepsin (R), with IFNα2 (ARI) hampers the aggressiveness of both colorectal cancer metastatic and stem cells in vivo and triggers immunogenic cell death signals that stimulate dendritic cell (DC) function. Here, we investigated the molecular signals induced by ARI treatment and found that this drug combination increased the accessibility to regulatory sequences of ISGs and IRFs that were epigenetically silenced in both colorectal cancer cells and DCs. Likewise, specific ARI-induced histone methylation and acetylation changes marked epigenetically affected ISG promoters in both metastatic cancer cells and DCs. Analysis by ChIP-seq confirmed such ARI-induced epigenetically regulated IFN signature. The activation of this signal endowed DCs with a marked migratory capability. Our results establish a direct correlation between reexpression of silenced ISGs by epigenetic control and ARI anticancer activity and provide new knowledge for the development of innovative combined therapeutic strategies for colorectal cancer. Cancer Immunol Res; 5(7); 604-16. ©2017 AACR.


Subject(s)
Carcinogenesis/drug effects , Colorectal Neoplasms/drug therapy , Dendritic Cells/drug effects , Interferon-alpha/immunology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/immunology , Azacitidine/administration & dosage , Carcinogenesis/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dendritic Cells/immunology , Depsipeptides/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factors/genetics , Interferon-alpha/genetics , Interferon-gamma/genetics , Nitric Oxide Synthase Type II/genetics , Receptors, Cytokine/genetics , Receptors, Interferon , Signal Transduction/drug effects
14.
Oncotarget ; 7(18): 26361-73, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27028869

ABSTRACT

Epigenetic alterations, including dysregulated DNA methylation and histone modifications, govern the progression of colorectal cancer (CRC). Cancer cells exploit epigenetic regulation to control cellular pathways, including apoptotic and metastatic signals. Since aberrations in epigenome can be pharmacologically reversed by DNA methyltransferase and histone deacetylase inhibitors, epigenetics in combination with standard agents are currently envisaged as a new therapeutic frontier in cancer, expected to overcome drug resistance associated with current treatments. In this study, we challenged this idea and demonstrated that the combination of azacitidine and romidepsin with IFN-α owns a high therapeutic potential, targeting the most aggressive cellular components of CRC, such as metastatic cells and cancer stem cells (CSCs), via tight control of key survival and death pathways. Moreover, the antitumor efficacy of this novel pharmacological approach is associated with induction of signals of immunogenic cell death. Of note, a previously undisclosed key role of IFN-α in inducing both antiproliferative and pro-apoptotic effects on CSCs of CRC was also found. Overall, these findings open a new frontier on the suitability of IFN-α in association with epigenetics as a novel and promising therapeutic approach for CRC management.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/pathology , Interferon-alpha/pharmacology , Neoplastic Stem Cells/drug effects , Animals , Apoptosis/drug effects , Azacitidine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Depsipeptides/pharmacology , Epigenesis, Genetic/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays
15.
Cytokine Growth Factor Rev ; 26(2): 195-201, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466627

ABSTRACT

Type I IFNs (IFN-I) are antiviral cytokines endowed with many biological effects, including antitumor activity. Over the last 15 years, an ensemble of studies has revealed that these cytokines play a crucial role in the induction of a protective antitumor immune response. Early in vivo studies in mouse models have been instrumental for understanding the IFN-I-induced host-mediated mechanisms. IFN-α is currently recognized as a powerful inducer of the differentiation/activation of dendritic cells (DCs) and today IFN-α-conditioned DCs represent promising DC candidates for the development of therapeutic cancer vaccines. Moreover, data from pilot clinical trials support the concept of using IFN-α as an enhancer of the response of patients to cancer vaccines. Notably, endogenous IFN-I production does also play a critical role in the antitumor response to some chemotherapeutic agents. Thus, we can now envisage new strategies of clinical use of IFN-α, based on the injection of IFN-conditioned cells as well as the usage of these cytokines as cancer vaccine adjuvants, alone or in combination with other treatments (including epigenetic drugs) to induce an immunogenic cell death and a long lasting antitumor response.


Subject(s)
Interferon Type I/immunology , Interferon Type I/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Adaptive Immunity , Adjuvants, Immunologic , Animals , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Humans , Immunity, Innate , Interferon-alpha/biosynthesis , Interferon-alpha/therapeutic use , Mice , Viral Vaccines/immunology
16.
Expert Rev Vaccines ; 12(10): 1195-210, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24090117

ABSTRACT

Due to their central role in priming and modulating the immune response, dendritic cells (DCs) represent an ideal instrument for the design of effective immunotherapeutic strategies for cancer patients. Recent advancement on the knowledge of the numerous DC subtypes, their functions and T-cell polarizing abilities has led to the development of several protocols for the ex vivo differentiation of autologous DCs and their loading with tumor-associated antigens. Moreover, novel strategies for the in vivo targeting of tumor antigens and adjuvants to natural DC subsets have been developed. Despite the large number of clinical studies carried out in cancer patients, a consensus on the optimal treatment modalities has not been reached yet. In this review, we summarize our current knowledge on DC biology and on DC use in clinical trials. Special attention is given to the many open issues regarding DC-based vaccination to sensitize researchers in the field to the compelling need of conducting comparative studies systematically addressing the still unresolved problems.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Neoplasms/therapy , Cancer Vaccines/administration & dosage , Clinical Trials as Topic , Drug Discovery/methods , Drug Discovery/trends , Humans
17.
Mol Oncol ; 6(1): 1-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22177803

ABSTRACT

Since the early clinical studies of cancer immunotherapy, the question arose as to whether it was possible to combine it with standard cancer treatments, mostly chemotherapy. The answer, now, is past history. The combined use of immunotherapy and chemotherapy is not only possible but, in certain cases, can be advantageous, depending on the drug, the dose and the combination modalities. In order to find the best synergisms between the two treatments and to turn weak immunotherapeutic interventions into potent anticancer instruments, it is mandatory to understand the complex mechanisms responsible for the positive interactions between chemotherapy and immunotherapy. In this article, we review the current knowledge on mechanisms involved in the immunostimulating activity of chemotherapy and summarize the main studies in both mouse models and patients aimed at exploiting such mechanisms for enhancing the response to cancer immunotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Animals , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Humans , Immunization , Immunomodulation/drug effects
18.
Expert Rev Vaccines ; 10(4): 487-98, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21506646

ABSTRACT

The IFN-α family are pleiotropic cytokines with the longest record of clinical use. Over the last decade, new biological effects of IFN-α on immune cells, including dendritic cells, have been described, supporting the concept that these cytokines can act as effective vaccine adjuvants. Recently, an important advance in our understanding of the mechanisms of interferon adjuvant activity has been achieved. Some clinical studies have been performed to assess the adjuvant activity in individuals immunized with preventive vaccines, showing variable results depending on interferon/vaccine formulation and vaccinated subjects. In spite of many data in animal models, little information is available on the possible advantage of utilizing IFN-α as an adjuvant for cancer vaccines in humans. Further clinical trials specifically designed to explore vaccine adjuvant activity are needed in order to define the best conditions for using IFN-α or IFN-α-conditioned dendritic cells for the development of therapeutic vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interferon-alpha/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Communicable Disease Control , Humans , Neoplasms/prevention & control , Neoplasms/therapy , Vaccines/administration & dosage , Vaccines/immunology
19.
Ann N Y Acad Sci ; 1194: 169-78, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20536466

ABSTRACT

A major challenge in cancer immunotherapy is the identification of effective strategies for enhancing its clinical efficacy. One approach is based on adjuvants capable of breaking tolerance against tumor-associated antigens. Interferon-alpha(IFN-alpha), an antiviral cytokine with a long record of clinical use, has recently been shown to act as an effective adjuvant in cancer patients. Notably, a special interest is currently focused on the use of dendritic cells (DC) generated in the presence of IFN-alpha (IFN-DC) for the preparation of anticancer vaccines. An additional approach for enhancing the response to immunotherapy relies on its combination with chemotherapy. In fact, an ensemble of results from both studies in animal models and pilot clinical trials suggest that certain chemotherapeutic agents can act, under defined conditions, as strong adjuvants for enhancing the efficacy of immunotherapy. These results open new opportunities for designing mechanism-based combination therapies involving both chemotherapy and new-generation cancer vaccines, including IFN-DC-based vaccines.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Interferon-alpha/therapeutic use , Neoplasms , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , Antineoplastic Agents/immunology , Antiviral Agents/immunology , Antiviral Agents/therapeutic use , Cancer Vaccines/immunology , Clinical Trials as Topic , Cytokines/immunology , Cytokines/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/physiology , Humans , Interferon-alpha/immunology , Male , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/therapy , Patients , Vaccines/immunology , Vaccines/therapeutic use
20.
Vaccine ; 27(41): 5654-60, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19635606

ABSTRACT

The use of adjuvants capable of improving the deficient immune response to hepatitis B virus (HBV) vaccine in haemodialysis patients is highly needed. Among potential adjuvants, type I interferons deserve a special attention in view of their known effects promoting cellular and humoral immune responses. The aim of the present trial was to evaluate the effects of recombinant interferon-alpha2b (IFN) administered as an adjuvant of HBV vaccine in unvaccinated haemodialysis patients. A significant and early enhancing effect on the antibody response was observed in patients receiving IFN. In addition, a predominance of IgG1 anti-HBs along with a transient normalization of circulating Th1 lymphocytes was only found in patients receiving IFN who achieved an early seroprotection. However, 6 months after the last vaccine dose, no significant differences were observed in the seroprotection rate achieved in patients vaccinated with IFN compared to that in patients receiving HBV vaccine alone. Mild to moderate fever, asthenia, and arthromyalgia were the most common reactions that occurred in vaccinees given IFN. In conclusion, addition of IFN to HBV vaccine, under the conditions used in this trial, is safe and achieves an earlier and higher seroprotection rate improving Th1-dependent immune response in haemodialysis patients.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Hepatitis B Vaccines/immunology , Hepatitis B/prevention & control , Interferon-alpha/administration & dosage , Adjuvants, Immunologic/adverse effects , Aged , Arthralgia/chemically induced , Asthenia/chemically induced , Drug-Related Side Effects and Adverse Reactions , Female , Fever/chemically induced , Hepatitis B Antibodies/blood , Hepatitis B Vaccines/adverse effects , Humans , Immunoglobulin G/blood , Interferon alpha-2 , Interferon-alpha/adverse effects , Male , Middle Aged , Recombinant Proteins/administration & dosage , Renal Dialysis , Renal Insufficiency/therapy , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL