Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Rep Med ; 4(3): 100939, 2023 03 21.
Article de Anglais | MEDLINE | ID: mdl-36796366

RÉSUMÉ

Immune checkpoint therapy (ICT) has the power to eradicate cancer, but the mechanisms that determine effective therapy-induced immune responses are not fully understood. Here, using high-dimensional single-cell profiling, we interrogate whether the landscape of T cell states in the peripheral blood predict responses to combinatorial targeting of the OX40 costimulatory and PD-1 inhibitory pathways. Single-cell RNA sequencing and mass cytometry expose systemic and dynamic activation states of therapy-responsive CD4+ and CD8+ T cells in tumor-bearing mice with expression of distinct natural killer (NK) cell receptors, granzymes, and chemokines/chemokine receptors. Moreover, similar NK cell receptor-expressing CD8+ T cells are also detected in the blood of immunotherapy-responsive cancer patients. Targeting the NK cell and chemokine receptors in tumor-bearing mice shows the functional importance of these receptors for therapy-induced anti-tumor immunity. These findings provide a better understanding of ICT and highlight the use and targeting of dynamic biomarkers on T cells to improve cancer immunotherapy.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs , Animaux , Souris , Antigène CD274 , Différenciation cellulaire , Tumeurs/anatomopathologie , Récepteurs aux chimiokines
2.
Nat Commun ; 13(1): 3966, 2022 07 08.
Article de Anglais | MEDLINE | ID: mdl-35803932

RÉSUMÉ

Understanding the mechanisms and impact of booster vaccinations are essential in the design and delivery of vaccination programs. Here we show that a three dose regimen of a synthetic peptide vaccine elicits an accruing CD8+ T cell response against one SARS-CoV-2 Spike epitope. We see protection against lethal SARS-CoV-2 infection in the K18-hACE2 transgenic mouse model in the absence of neutralizing antibodies, but two dose approaches are insufficient to confer protection. The third vaccine dose of the single T cell epitope peptide results in superior generation of effector-memory T cells and tissue-resident memory T cells, and these tertiary vaccine-specific CD8+ T cells are characterized by enhanced polyfunctional cytokine production. Moreover, fate mapping shows that a substantial fraction of the tertiary CD8+ effector-memory T cells develop from re-migrated tissue-resident memory T cells. Thus, repeated booster vaccinations quantitatively and qualitatively improve the CD8+ T cell response leading to protection against otherwise lethal SARS-CoV-2 infection.


Sujet(s)
COVID-19 , Déterminants antigéniques des lymphocytes T , Animaux , Anticorps neutralisants , Anticorps antiviraux , Lymphocytes T CD8+ , COVID-19/prévention et contrôle , Modèles animaux de maladie humaine , Mémoire immunologique , Souris , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Vaccination , Vaccins synthétiques
3.
J Immunother Cancer ; 9(4)2021 04.
Article de Anglais | MEDLINE | ID: mdl-33879600

RÉSUMÉ

BACKGROUND: High serum interleukin (IL-6) levels may cause resistance to immunotherapy by modulation of myeloid cells in the tumor microenvironment. IL-6 signaling blockade is tested in cancer, but as this inflammatory cytokine has pleiotropic effects, this treatment is not always effective. METHODS: IL-6 and IL-6R blockade was applied in an IL-6-mediated immunotherapy-resistant TC-1 tumor model (TC-1.IL-6) and immunotherapy-sensitive TC-1. CONTROL: Effects on therapeutic vaccination-induced tumor regression, recurrence and survival as well on T cells and myeloid cells in the tumor microenvironment were studied. The effects of IL-6 signaling in macrophages under therapy conditions were studied in Il6rafl/fl×LysMcre+ mice. RESULTS: Our therapeutic vaccination protocol elicits a strong tumor-specific CD8+ T-cell response, leading to enhanced intratumoral T-cell infiltration and recruitment of tumoricidal macrophages. Blockade of IL-6 signaling exacerbated tumor outgrowth, reflected by fewer complete regressions and more recurrences after therapeutic vaccination, especially in TC-1.IL-6 tumor-bearing mice. Early IL-6 signaling blockade partly inhibited the development of the vaccine-induced CD8+ T-cell response. However, the main mechanism was the malfunction of macrophages during therapy-induced tumor regression. Therapy efficacy was impaired in Il6rafl/fl×LysMcre+ but not cre-negative control mice, while no differences in the vaccine-induced CD8+ T-cell response were found between these mice. IL-6 signaling blockade resulted in decreased expression of suppressor of cytokine signaling 3, essential for effective M1-type function in macrophages, and increased expression of the phagocytic checkpoint molecule signal-regulatory protein alpha by macrophages. CONCLUSION: IL-6 signaling is critical for macrophage function under circumstances of immunotherapy-induced tumor tissue destruction, in line with the acute inflammatory functions of IL-6 signaling described in infections.


Sujet(s)
Vaccins anticancéreux/administration et posologie , Interleukine-6/métabolisme , Tumeurs/traitement médicamenteux , Oligodésoxyribonucléotides/administration et posologie , Protéines E7 de papillomavirus/administration et posologie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Animaux , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Vaccins anticancéreux/immunologie , Lignée cellulaire tumorale , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Femelle , Injections sous-cutanées , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Souris de lignée C57BL , Souris knockout , Tumeurs/immunologie , Tumeurs/métabolisme , Oligodésoxyribonucléotides/immunologie , Protéines E7 de papillomavirus/immunologie , Phénotype , Récepteurs à l'interleukine-6/génétique , Récepteurs à l'interleukine-6/métabolisme , Transduction du signal , Charge tumorale/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme
4.
Int J Cancer ; 148(1): 211-225, 2021 01 01.
Article de Anglais | MEDLINE | ID: mdl-32875568

RÉSUMÉ

High serum levels of interleukin-6 (IL-6) correlate with poor prognosis and chemotherapy resistance in several cancers. The underlying mechanisms and its effects on immunotherapy are largely unknown. To address this, we developed a human papillomavirus type 16 (HPV16)-associated tumor model expressing IL-6 to investigate the impact of tumor-expressed IL-6 during cisplatin chemotherapy and HPV16 synthetic long peptide vaccination as immunotherapy. The effects of tumor-produced IL-6 on tumor growth, survival and the tumor microenvironment were analyzed. Our data demonstrated that tumor-produced IL-6 conferred resistance to cisplatin and therapeutic vaccination. This was not caused by a changed in vitro or in vivo growth rate of tumor cells, or a changed sensitivity of tumor cells to chemotherapy or T-cell-mediated killing. Furthermore, no overt differences in the frequencies of tumor-infiltrating subsets of T cells or CD11b+ myeloid cells were observed. IL-6, however, affected the systemic and local function of myeloid cells, reflected by a strong reduction of major histocompatibility complex (MHC) class II expression on all major myeloid cell subtypes. Resistance to both therapies was associated with a changed intratumoral influx of MHC class II+ myeloid cells toward myeloid cells with no or lower MHC class II expression. Importantly, while these IL-6-mediated effects provided resistance to the immunotherapy and chemotherapy as single therapies, their combination still successfully mediated tumor control. In conclusion, IL-6-mediated therapy resistance is caused by an extrinsic mechanism involving an impaired function of intratumoral myeloid cells. The fact that resistance can be overcome by combination therapies provides direction to more effective therapies for cancer.


Sujet(s)
Vaccins anticancéreux/administration et posologie , Interleukine-6/métabolisme , Cellules myéloïdes/immunologie , Tumeurs/thérapie , Infections à papillomavirus/thérapie , Animaux , Vaccins anticancéreux/immunologie , Lignée cellulaire tumorale/transplantation , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/immunologie , Femelle , Antigènes d'histocompatibilité de classe II/métabolisme , Papillomavirus humain de type 16/immunologie , Humains , Immunothérapie/méthodes , Interleukine-6/génétique , Souris , Cellules myéloïdes/métabolisme , Tumeurs/immunologie , Tumeurs/anatomopathologie , Tumeurs/virologie , Infections à papillomavirus/immunologie , Infections à papillomavirus/anatomopathologie , Infections à papillomavirus/virologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Vaccins sous-unitaires/immunologie
5.
J Immunother Cancer ; 8(2)2020 09.
Article de Anglais | MEDLINE | ID: mdl-32873723

RÉSUMÉ

BACKGROUND: Immunotherapy of cancer is successful but tumor regression often is incomplete and followed by escape. Understanding the mechanisms underlying this acquired resistance will aid the development of more effective treatments. METHODS: We exploited a mouse model where tumor-specific therapeutic vaccination results in tumor regression, followed by local recurrence and resistance. In depth studies on systemic, local and tumor intrinsic changes were performed with flow and mass cytometry, immunohistochemistry, transcriptomics and several perturbation studies with inhibitors or agonistic antibodies in mice. Main findings were recapitulated in vaccinated patients. RESULTS: Full tumor regression and cure of tumor-bearing mice is dependent on the magnitude of the vaccine-induced T-cell response. Recurrence of tumors did not involve classical immune escape mechanisms, such as antigen-presentation alterations, immune checkpoint expression, resistance to killing or local immune suppression. However, the recurrent tumors displayed a changed transcriptome with alterations in p53, tumor necrosis factor-α and transforming growth factor-ß signaling pathways and they became immunologically cold. Remarkably, ex vivo cell-sorted recurrent tumors, directly reinjected in naïve hosts retained their resistance to vaccination despite a strong infiltration with tumor-specific CD8+ T cells, similar to that of vaccine-responsive tumors. The influx of inflammatory mature myeloid effector cells in the resistant tumors, however, was impaired and this turned out to be the underlying mechanisms as restoration of inflammatory myeloid cell infiltration reinstated the sensitivity of these refractory tumors to vaccination. Notably, impaired myeloid cell infiltration after vaccination was also associated with vaccine resistance in patients. CONCLUSION: An immunotherapy-induced disability of tumor cells to attract innate myeloid effector cells formed a major mechanism underlying immune escape and acquired resistance. These data not only stresses the importance of myeloid effector cells during immunotherapy but also demands for new studies to harness their tumoricidal activities.


Sujet(s)
Résistance aux médicaments antinéoplasiques/immunologie , Immunothérapie/méthodes , Cellules myéloïdes/immunologie , Animaux , Modèles animaux de maladie humaine , Humains , Souris
6.
J Immunother Cancer ; 7(1): 25, 2019 01 31.
Article de Anglais | MEDLINE | ID: mdl-30704520

RÉSUMÉ

BACKGROUND: The capacity of cytomegalovirus (CMV) to elicit long-lasting strong T cell responses, and the ability to engineer the genome of this DNA virus positions CMV-based vaccine vectors highly suitable as a cancer vaccine platform. Defined immune thresholds for tumor protection and the factors affecting such thresholds have not well been investigated in cancer immunotherapy. We here determined using CMV as a vaccine platform whether critical thresholds of vaccine-specific T cell responses can be established that relate to tumor protection, and which factors control such thresholds. METHODS: We generated CMV-based vaccine vectors expressing the E7 epitope and tested these in preclinical models of HPV16-induced cancer. Vaccination was applied via different doses and routes (intraperitoneal (IP), subcutaneous (SC) and intranasal (IN)). The magnitude, kinetics and phenotype of the circulating tumor-specific CD8+ T cell response were determined. Mice were subsequently challenged with tumor cells, and the tumor protection was monitored. RESULTS: Immunization with CMV-based vaccines via the IP or SC route eliciting vaccine-induced CD8+ T cell responses of > 0.3% of the total circulating CD8 T cell population fully protects mice against lethal tumor challenge. However, low dose inoculations via the IP or SC route or IN vaccination elicited vaccine-induced CD8+ T cell responses that did not reach protective thresholds for tumor protection. In addition, whereas weak pre-existing immunity did not alter the protective thresholds of the vaccine-specific T cell response following subsequent immunization with CMV-based vaccine vectors, strong pre-existing immunity inhibited the development of vaccine-induced T cells and their control on tumor progression. CONCLUSIONS: This study highlight the effectiveness of CMV-based vaccine vectors, and shows that demarcated thresholds of vaccine-specific T cells could be defined that correlate to tumor protection. Together, these results may hold importance for cancer vaccine development to achieve high efficacy in vaccine recipients.


Sujet(s)
Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/administration et posologie , Cytomegalovirus , Épitopes/immunologie , Tumeurs/thérapie , Protéines E7 de papillomavirus/immunologie , Infections à papillomavirus/complications , Animaux , Lignée cellulaire tumorale , Épitopes/génétique , Papillomavirus humain de type 16 , Souris de lignée C57BL , Tumeurs/étiologie , Tumeurs/immunologie , Protéines E7 de papillomavirus/génétique , Infections à papillomavirus/immunologie
7.
Expert Opin Biol Ther ; 17(1): 87-103, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27802061

RÉSUMÉ

INTRODUCTION: The treatment options for cancer-surgery, radiotherapy and chemotherapy-are now supplemented with immunotherapy. Previously underappreciated but now gaining strong interest are the immune modulatory properties of the three conventional modalities. Moreover, there is a better understanding of the needs and potential of the different immune therapeutic platforms. Key to improved treatment will be the combinations of modalities that complete each other's shortcomings. Area covered: Tumor-specific T-cells are required for optimal immunotherapy. In this review, the authors focus on the correct timing of different types of chemotherapeutic agents or immune modulators and immunotherapeutic drugs, not only for the activation and expansion of tumor-specific T-cells but also to support and enhance their anti-tumor efficacy. Expert opinion: At an early phase of disease, clinical success can be obtained using single treatment modalities but at later disease stages, combinations of several modalities are required. The gain in success is determined by a thorough understanding of the direct and indirect immune effects of the modalities used. Profound knowledge of these effects requires optimal tuning of immunomonitoring. This will guide the appropriate combination of treatments and allow for correct sequencing the order and interval of the different therapeutic modalities.


Sujet(s)
Immunothérapie/méthodes , Tumeurs/immunologie , Tumeurs/thérapie , Animaux , Antinéoplasiques/immunologie , Antinéoplasiques/usage thérapeutique , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Humains , Facteurs immunologiques/immunologie , Facteurs immunologiques/usage thérapeutique , Tumeurs/diagnostic , Lymphocytes T/immunologie , Facteurs temps , Vaccination/méthodes
8.
Cancer Res ; 76(20): 6017-6029, 2016 10 15.
Article de Anglais | MEDLINE | ID: mdl-27569212

RÉSUMÉ

Certain cytotoxic chemotherapeutic drugs are immunogenic, stimulating tumor immunity through mechanisms that are not completely understood. Here we show how the DNA-damaging drug cisplatin modulates tumor immunity. At the maximum tolerated dose (MTD), cisplatin cured 50% of mice with established murine TC-1 or C3 tumors, which are preclinical models of human papillomavirus (HPV)-associated cancer. Notably, the curative benefit of cisplatin relied entirely upon induction of tumor-specific CD8+ T cells. Mechanistic investigations showed that cisplatin stimulated tumor infiltration of inflammatory antigen-presenting cells (APC) expressing relatively higher levels of the T-cell costimulatory ligands CD70, CD80, and CD86. Cell death triggered by cisplatin was associated with the release of at least 19 proteins in the tumor environment that could act as damage-associated molecular patterns and upregulate costimulatory molecules, either alone or in concert, but the responsible proteins remain unknown. Essentially, the curative effect of cisplatin was abrogated in mice lacking expression of CD80 and CD86 on APCs. Furthermore, cisplatin treatment was improved by CTLA-4 blockade, which increases the availability of CD80/86 to bind to CD28. In contrast, there was no effect of CD27 stimulation, which replaces CD70 interaction. At the cisplatin MTD, cure rates could also be increased by vaccination with synthetic long peptides, whereas cures could also be achieved at similar rates at 80% of the MTD with reduced side effects. Our findings reveal an essential basis for the immunogenic properties of cisplatin, which are mediated by the induction of costimulatory signals for CD8+ T-cell-dependent tumor destruction. Cancer Res; 76(20); 6017-29. ©2016 AACR.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Antigène CD80/physiologie , Antigène CD86/physiologie , Lymphocytes T CD8+/immunologie , Cisplatine/usage thérapeutique , Tumeurs expérimentales/traitement médicamenteux , Animaux , Cellules présentatrices d'antigène/physiologie , Antigènes CD70/physiologie , Interféron gamma/biosynthèse , Activation des lymphocytes , Souris , Souris de lignée C57BL , Cellules myéloïdes suppressives/physiologie , Tumeurs expérimentales/immunologie , Vaccination
9.
Clin Cancer Res ; 21(4): 781-94, 2015 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-25501579

RÉSUMÉ

PURPOSE: Cancer immunotherapy, such as vaccination, is an increasingly successful treatment modality, but its interaction with chemotherapy remains largely undefined. Therefore, we explored the mechanism of synergy between vaccination with synthetic long peptides (SLP) of human papillomavirus type 16 (HPV16) and cisplatin in a preclinical tumor model for HPV16. EXPERIMENTAL DESIGN: SLP vaccination in this preclinical tumor model allowed the elucidation of novel mechanisms of synergy between chemo- and immunotherapy. By analyzing the tumor immune infiltrate, we focused on the local intratumoral effects of chemotherapy, vaccination, or the combination. RESULTS: Of several chemotherapeutic agents, cisplatin synergized best with SLP vaccination in tumor eradication, without requirement for the maximum-tolerated dose (MTD). Upon SLP vaccination, tumors were highly infiltrated with HPV-specific, tumor necrosis factor-α (TNFα)- and interferon-γ (IFNγ)-producing T cells. Upon combined treatment, tumor cell proliferation was significantly decreased compared with single treated and untreated tumors. Furthermore, we showed that TNFα strongly enhanced cisplatin-induced apoptotic tumor cell death in a JNK-dependent manner. This is consistent with upregulation of proapoptotic molecules and with enhanced cell death in vivo upon combined SLP vaccination and cisplatin treatment. In vivo neutralization of TNFα significantly reduced the antitumor responses induced by the combined treatment. CONCLUSION: Taken together, our data show that peptide vaccination with cisplatin treatment leads to decreased tumor cell proliferation and TNFα-induced enhanced cisplatin-mediated killing of tumor cells, together resulting in superior tumor eradication.


Sujet(s)
Antinéoplasiques/pharmacologie , Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/pharmacologie , Cisplatine/pharmacologie , Tumeurs du col de l'utérus/anatomopathologie , Transfert adoptif , Animaux , Mort cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Femelle , Papillomavirus humain de type 16 , Humains , Lymphocytes TIL , Souris , Souris de lignée C57BL , Infections à papillomavirus/complications , Peptides , Réaction de polymérisation en chaine en temps réel , Facteur de nécrose tumorale alpha/biosynthèse , Tumeurs du col de l'utérus/immunologie , Tumeurs du col de l'utérus/virologie , Protéines virales/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE