Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Hum Vaccin Immunother ; 20(1): 2303799, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38346926

ABSTRACT

Efficacy of cancer immunotherapies relies on correct recognition of tumor antigens by lymphocytes, eliciting thus functional responses capable of eliminating tumor cells. Therefore, important efforts have been carried out in antigen identification, with the aim of understanding mechanisms of response to immunotherapy and to design safer and more efficient strategies. In addition to classical tumor-associated antigens identified during the last decades, implementation of next-generation sequencing methodologies is enabling the identification of neoantigens (neoAgs) arising from mutations, leading to the development of new neoAg-directed therapies. Moreover, there are numerous non-classical tumor antigens originated from other sources and identified by new methodologies. Here, we review the relevance of neoAgs in different immunotherapies and the results obtained by applying neoAg-based strategies. In addition, the different types of non-classical tumor antigens and the best approaches for their identification are described. This will help to increase the spectrum of targetable molecules useful in cancer immunotherapies.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Antigens, Neoplasm/genetics , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , Mutation
4.
Front Immunol ; 13: 985886, 2022.
Article in English | MEDLINE | ID: mdl-36405725

ABSTRACT

Immune checkpoint inhibitor (ICI)-based immunotherapy in triple negative breast cancer (TNBC) is achieving limited therapeutic results, requiring the development of more potent strategies. Combination of ICI with vaccination strategies would enhance antitumor immunity and response rates to ICI in patients having poorly infiltrated tumors. In heavily mutated tumors, neoantigens (neoAgs) resulting from tumor mutations have induced potent responses when used as vaccines. Thus, our aim was the identification of immunogenic neoAgs suitable as vaccines in TNBC patients. By using whole exome sequencing, RNAseq and HLA binding algorithms of tumor samples from a cohort of eight TNBC patients, we identified a median of 60 mutations/patient, which originated a putative median number of 98 HLA class I-restricted neoAgs. Considering a group of 27 predicted neoAgs presented by HLA-A*02:01 allele in two patients, peptide binding to HLA was experimentally confirmed in 63% of them, whereas 55% were immunogenic in vivo in HLA-A*02:01+ transgenic mice, inducing T-cells against the mutated but not the wild-type peptide sequence. Vaccination with peptide pools or DNA plasmids expressing these neoAgs induced polyepitopic T-cell responses, which recognized neoAg-expressing tumor cells. These results suggest that TNBC tumors harbor neoAgs potentially useful in therapeutic vaccines, opening the way for new combined immunotherapies.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Mice , Animals , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/therapy , Immunotherapy/methods , Antigens, Neoplasm , HLA-A2 Antigen , Peptides , Mice, Transgenic
5.
Front Immunol ; 13: 991311, 2022.
Article in English | MEDLINE | ID: mdl-36300124

ABSTRACT

Vaccination using optimized strategies may increase response rates to immune checkpoint inhibitors (ICI) in some tumors. To enhance vaccine potency and improve thus responses to ICI, we analyzed the gene expression profile of an immunosuppressive dendritic cell (DC) population induced during vaccination, with the goal of identifying druggable inhibitory mechanisms. RNAseq studies revealed targetable genes, but their inhibition did not result in improved vaccines. However, we proved that immunosuppressive DC had a monocytic origin. Thus, monocyte depletion by gemcitabine administration reduced the generation of these DC and increased vaccine-induced immunity, which rejected about 20% of LLC-OVA and B16-OVA tumors, which are non-responders to anti-PD-1. This improved efficacy was associated with higher tumor T-cell infiltration and overexpression of PD-1/PD-L1. Therefore, the combination of vaccine + gemcitabine with anti-PD-1 was superior to anti-PD-1 monotherapy in both models. B16-OVA tumors benefited from a synergistic effect, reaching 75% of tumor rejection, but higher levels of exhausted T-cells in LLC-OVA tumors co-expressing PD-1, LAG3 and TIM3 precluded similar levels of efficacy. Our results indicate that gemcitabine is a suitable combination therapy with vaccines aimed at enhancing PD-1 therapies by targeting vaccine-induced immunosuppressive DC.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , B7-H1 Antigen , Hepatitis A Virus Cellular Receptor 2 , Immune Checkpoint Inhibitors , Vaccination , Neoplasms/drug therapy , Dendritic Cells , Gemcitabine
6.
Int J Mol Sci ; 23(4)2022 Feb 11.
Article in English | MEDLINE | ID: mdl-35216137

ABSTRACT

Immune checkpoint inhibitors (ICI) have been used as immunotherapy for hepatocellular carcinoma (HCC) with promising but still limited results. Identification of immune elements in the tumor microenvironment of individual HCC patients may help to understand the correlations of responses, as well as to design personalized therapies for non-responder patients. Immune-enhancing strategies, such as vaccination, would complement ICI in those individuals with poorly infiltrated tumors. The prominent role of responses against mutated tumor antigens (neoAgs) in ICI-based therapies suggests that boosting responses against these epitopes may specifically target tumor cells. In this review we summarize clinical vaccination trials carried out in HCC, the available information on potentially immunogenic neoAgs in HCC patients, and the most recent results of neoAg-based vaccines in other tumors. Despite the low/intermediate mutational burden observed in HCC, data obtained from neoAg-based vaccines in other tumors indicate that vaccines directed against these tumor-specific antigens would complement ICI in a subset of HCC patients.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Immune Checkpoint Inhibitors/pharmacology , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Humans , Immunotherapy/methods
7.
J Immunother Cancer ; 10(2)2022 02.
Article in English | MEDLINE | ID: mdl-35193931

ABSTRACT

BACKGROUND: Neoantigens, new immunogenic sequences arising from tumor mutations, have been associated with response to immunotherapy and are considered potential targets for vaccination. Hepatocellular carcinoma (HCC) is a moderately mutated tumor, where the neoantigen repertoire has not been investigated. Our aim was to analyze whether tumors in HCC patients contain immunogenic neoantigens suitable for future use in therapeutic vaccination. METHODS: Whole-exome sequencing and RNAseq were performed in a cohort of fourteen HCC patients submitted to surgery or liver transplant. To identify mutations, single-nucleotide variants (SNV) originating non-synonymous changes that were confirmed at the RNA level were analyzed. Immunogenicity of putative neoAgs predicted by HLA binding algorithms was confirmed by using in vitro HLA binding assays and T-cell stimulation experiments, the latter in vivo, by immunizing HLA-A*02.01/HLA-DRB1*01 (HHD-DR1) transgenic mice, and in in vitro, using human lymphocytes. RESULTS: Sequencing led to the identification of a median of 1217 missense somatic SNV per patient, narrowed to 30 when filtering by using RNAseq data. A median of 13 and 5 peptides per patient were predicted as potential binders to HLA class I and class II molecules, respectively. Considering only HLA-A*02.01- and HLA-DRB1*01-predicted binders, 70% demonstrated HLA-binding capacity and about 50% were immunogenic when tested in HHD-DR1 mice. These peptides induced polyfunctional T cells that specifically recognized the mutated but not the wild-type sequence as well as neoantigen-expressing cells. Moreover, coimmunization experiments combining CD8 and CD4 neoantigen epitopes resulted in stronger CD8 T cell responses. Finally, responses against neoantigens were also induced in vitro using human cells. CONCLUSION: These results show that mutations in HCC tumors may generate immunogenic neoantigens with potential applicability for future combinatorial therapeutic strategies.


Subject(s)
Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Animals , Antigens, Neoplasm/pharmacology , Cancer Vaccines/pharmacology , Humans , Mice
8.
Front Immunol ; 13: 1044025, 2022.
Article in English | MEDLINE | ID: mdl-36761163

ABSTRACT

Current vaccines against SARS-CoV-2, based on the original Wuhan sequence, induce antibodies with different degrees of cross-recognition of new viral variants of concern. Despite potent responses generated in vaccinated and infected individuals, the Omicron (B.1.1.529) variant causes breakthrough infections, facilitating viral transmission. We previously reported a vaccine based on a cyclic peptide containing the 446-488 S1 sequence (446-488cc) of the SARS-CoV-2 spike (S) protein from Wuhan isolate. To provide the best immunity against Omicron, here we compared Omicron-specific immunity induced by a Wuhan-based 446-488cc peptide, by a Wuhan-based recombinant receptor-binding domain (RBD) vaccine and by a new 446-488cc peptide vaccine based on the Omicron sequence. Antibodies induced by Wuhan peptide 446-488cc in three murine strains not only recognized the Wuhan and Omicron 446-488 peptides similarly, but also Wuhan and Omicron RBD protein variants. By contrast, antibodies induced by the Wuhan recombinant RBD vaccine showed a much poorer cross-reactivity for the Omicron RBD despite similar recognition of Wuhan and Omicron peptide variants. Finally, although the Omicron-based 446-488cc peptide vaccine was poorly immunogenic in mice due to the loss of T cell epitopes, co-immunization with Omicron peptide 446-488cc and exogenous T cell epitopes induced strong cross-reactive antibodies that neutralized Omicron SARS-CoV-2 virus. Since mutations occurring within this sequence do not alter T cell epitopes in humans, these results indicate the robust immunogenicity of 446-488cc-based peptide vaccines that induce antibodies with a high cross-recognition capacity against Omicron, and suggest that this sequence could be included in future vaccines targeting the Omicron variant.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Animals , Mice , COVID-19 Vaccines , Epitopes, T-Lymphocyte , COVID-19/prevention & control , Vaccines, Subunit , Antibodies
9.
Emerg Microbes Infect ; 10(1): 1931-1946, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34538222

ABSTRACT

Identification of relevant epitopes is crucial for the development of subunit peptide vaccines inducing neutralizing and cellular immunity against SARS-CoV-2. Our aim was the characterization of epitopes in the receptor-binding domain (RBD) of SARS-CoV-2 spike (S) protein to generate a peptide vaccine. Epitope mapping using a panel of 10 amino acid overlapped 15-mer peptides covering region 401-515 from RBD did not identify linear epitopes when tested with sera from infected individuals or from RBD-immunized mice. However, immunization of mice with these 15-mer peptides identified four peptides located at region 446-480 that induced antibodies recognizing the peptides and RBD/S1 proteins. Immunization with peptide 446-480 from S protein formulated with Freund's adjuvant or with CpG oligodeoxinucleotide/Alum induced polyepitopic antibody responses in BALB/c and C56BL/6J mice, recognizing RBD (titres of 3 × 104-3 × 105, depending on the adjuvant) and displaying neutralizing capacity (80-95% inhibition capacity; p < 0.05) against SARS-CoV-2. Murine CD4 and CD8T-cell epitopes were identified in region 446-480 and vaccination experiments using HLA transgenic mice suggested the presence of multiple human T-cell epitopes. Antibodies induced by peptide 446-480 showed broad recognition of S proteins and S-derived peptides belonging to SARS-CoV-2 variants of concern. Importantly, vaccination with peptide 446-480 or with a cyclic version of peptide 446-488 containing a disulphide bridge between cysteines 480 and 488, protected humanized K18-hACE2 mice from a lethal dose of SARS-CoV-2 (62.5 and 75% of protection; p < 0.01 and p < 0.001, respectively). This region could be the basis for a peptide vaccine or other vaccine platforms against Covid-19.


Subject(s)
Antibodies, Neutralizing/immunology , COVID-19 Vaccines/immunology , COVID-19/prevention & control , Immunity, Cellular , Immunity, Humoral , SARS-CoV-2/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , COVID-19 Vaccines/standards , Cross Reactions/immunology , Epitope Mapping , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte/immunology , Humans , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
10.
Cancer Lett ; 499: 279-289, 2021 02 28.
Article in English | MEDLINE | ID: mdl-33232788

ABSTRACT

Analyzing immunomodulatory elements operating during antitumor vaccination in prostate cancer patients and murine models we identified IL-10-producing DC as a subset with poorer immunogenicity and clinical efficacy. Inhibitory TAM receptors MER and AXL were upregulated on murine IL-10+ DC. Thus, we analyzed conditions inducing these molecules and the potential benefit of their blockade during vaccination. MER and AXL upregulation was more efficiently induced by a vaccine containing Imiquimod than by a poly(I:C)-containing vaccine. Interestingly, MER expression was found on monocyte-derived DC, and was dependent on IL-10. TAM blockade improved Imiquimod-induced DC activation in vitro and in vivo, resulting in increased vaccine-induced T-cell responses, which were further reinforced by concomitant IL-10 inhibition. In different tumor models, a triple therapy (including vaccination, TAM inhibition and IL-10 blockade) provided the strongest therapeutic effect, associated with enhanced T-cell immunity and enhanced CD8+ T cell tumor infiltration. Finally, MER levels in DC used for vaccination in cancer patients correlated with IL-10 expression, showing an inverse association with vaccine-induced clinical response. These results suggest that TAM receptors upregulated during vaccination may constitute an additional target in combinatorial therapeutic vaccination strategies.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Melanoma, Experimental/therapy , Prostatic Neoplasms/therapy , Adjuvants, Immunologic/administration & dosage , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Imiquimod/administration & dosage , Immunogenicity, Vaccine/drug effects , Interleukin-10/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Poly I-C/administration & dosage , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrimidines , Quinolines , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Up-Regulation/drug effects , Up-Regulation/immunology , c-Mer Tyrosine Kinase/antagonists & inhibitors , c-Mer Tyrosine Kinase/genetics , Axl Receptor Tyrosine Kinase
11.
Cancers (Basel) ; 12(11)2020 Nov 16.
Article in English | MEDLINE | ID: mdl-33207844

ABSTRACT

Therapies based on immune checkpoint inhibitors (ICPI) have yielded promising albeit limited results in patients with hepatocellular carcinoma (HCC). Vaccines have been proposed as combination partners to enhance response rates to ICPI. Thus, we analyzed the combined effect of a vaccine based on the TLR4 ligand cold-inducible RNA binding protein (CIRP) plus ICPI. Mice were immunized with vaccines containing ovalbumin linked to CIRP (OVA-CIRP), with or without ICPI, and antigen-specific responses and therapeutic efficacy were tested in subcutaneous and orthotopic mouse models of liver cancer. OVA-CIRP elicited polyepitopic T-cell responses, which were further enhanced when combined with ICPI (anti-PD-1 and anti-CTLA-4). Combination of OVA-CIRP with ICPI enhanced ICPI-induced therapeutic responses when tested in subcutaneous and intrahepatic B16-OVA tumors, as well as in the orthotopic PM299L HCC model. This effect was associated with higher OVA-specific T-cell responses in the periphery, although many tumor-infiltrating lymphocytes still displayed an exhausted phenotype. Finally, a new vaccine containing human glypican-3 linked to CIRP (GPC3-CIRP) induced clear responses in humanized HLA-A2.01 transgenic mice, which increased upon combination with ICPI. Therefore, CIRP-based vaccines may generate anti-tumor immunity to enhance ICPI efficacy in HCC, although blockade of additional checkpoint molecules and immunosuppressive targets should be also considered.

12.
Fertil Steril ; 102(6): 1567-75.e1, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25305728

ABSTRACT

OBJECTIVE: To determine the effect of removing presumptive apoptotic sperm cells from samples from unselected males by means of magnetic activated cell sorting (MACS) on live-birth delivery rates after intracytoplasmic sperm injection (ICSI) in couples undergoing ovum donation (OD). DESIGN: Prospective, randomized, triple-blinded, and controlled study. SETTING: Private university-affiliated IVF center. PATIENT(S): A total of 237 infertile couples undergoing ICSI as part of an OD program. INTERVENTION(S): Semen specimens from the control group were prepared by swim-up. Samples from the study group were prepared by swim-up followed by MACS and incubation with annexin V-conjugated microbeads to remove annexin V-positive (AV+) sperm cells. MAIN OUTCOME MEASURE(S): Fertilization rates, morphological features of early embryo development, implantation rates, ongoing pregnancy rates, and live-birth rates. RESULT(S): Similar results were obtained between groups for all the parameters compared: fertilization rates of 75.3% (95% confidence interval [CI], 71.6-78.9) versus 72.1% (95% CI, 68.6-75.7); percentage of good-quality embryos on day 2 of 53.7% (95% CI, 50.3-57.1) versus 51.8% (95% CI, 48.3-55.3) and on day 3 of 54.2% (95% CI, 50.7-57.6) versus 48.9% (95% CI, 45.3-52.4); implantation rates of 42.2% (95% CI, 33.8-48.1) versus 40.1% (95% CI, 34.8-49.6); positive beta-hCG tests of 63.2% (95% CI, 54.7-71.6) versus 68.6% (95% CI, 60.2-76.9), and live-birth rates of 48.4% (95% CI, 39.6-57.1) versus 56.4% (95% CI, 47.3-65.5) in the MACS versus control group. None of the differences reached statistical significance. CONCLUSION(S): Applying MACS technology to remove AV+ sperm cells from unselected males does not improve the reproductive outcome of ICSI in OD.


Subject(s)
Annexin A5/metabolism , Cell Separation/methods , Sperm Injections, Intracytoplasmic/methods , Spermatozoa/immunology , Adult , Apoptosis , Birth Rate , Female , Fertilization in Vitro , Humans , Magnetics , Male , Oocyte Donation , Phosphatidylserines/metabolism , Pregnancy , Sperm Maturation/physiology , Spermatozoa/pathology
13.
J Assist Reprod Genet ; 30(8): 1043-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23820799

ABSTRACT

PURPOSE: To determine the survival and subsequent in vitro development of human cleavage stage embryos and hatched blastocysts following varying periods of short-term storage at 4 °C, using tripronucleated human embryos (TPN) as a model. METHODS: TPN cleavage embryos and hatched blastocysts short-term stored at 4 °C for 0 h (control), 24 h and 48 h. The main outcome measures were: survival rates (SR) and in vitro developmental ability (blastocyst rate and blastocyst-re-expansion rate) in each of the groups after storage. RESULTS: Cleavage-stage TPN survived at comparable rates to controls, regardless of storage time (average: 97.3 %). The in vitro development of cleavage-stage TPN stored for 24 h was comparable to that of controls (average 64.7 %), but was significantly impaired when storage lasted 48-h (20.8 %). After artificial shrinkage, SR was comparable in 24-h-stored and non-stored hatched blastocysts (85.7 %; p > 0.05), but was significantly impaired in the 48-h-stored group (20.0 %). Following 24-h storage, the re-expansion rate of hatched blastocysts was similar to that of controls (average: 57.1 %; p > 0.05), but was higher than that of the 48-h-stored group (15.0 %; p < 0.05). CONCLUSIONS: TPN human cleavage embryos and blastocysts can be successfully stored short-term for up to 24 h at 4 °C without using cryoprotectants without any significant negative impact on survival or subsequent in vitro development.


Subject(s)
Blastocyst/cytology , Embryo Culture Techniques , Embryonic Development , Blastocyst/ultrastructure , Humans , Reproductive Techniques, Assisted , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...