Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.008
Filtrer
1.
Cancer Immunol Immunother ; 73(9): 178, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954031

RÉSUMÉ

Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.


Sujet(s)
Tumeurs du cerveau , Vaccins anticancéreux , Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Vaccins à ADN , Animaux , Souris , Vaccins à ADN/immunologie , Vaccins à ADN/usage thérapeutique , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Humains , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Souris de lignée C57BL , Femelle , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Immunothérapie/méthodes , Glioblastome/immunologie , Glioblastome/thérapie , Glioblastome/traitement médicamenteux , Lignée cellulaire tumorale , Intramolecular oxidoreductases
2.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38955422

RÉSUMÉ

BACKGROUND: Despite advances in checkpoint inhibitor (CPI) therapy for cancer treatment, many cancers remain resistant. Tumors deemed "cold" based on lack of T cell infiltration show reduced potential for CPI therapy. Cancer vaccines may overcome the inadequacy of existing T cells by inducing the needed antitumor T cell response to synergize with CPIs and overcome resistance. METHODS: CT26 and TC1 tumor cells were injected subcutaneously into mice. Mice were treated with combinations of CPIs alone or a cancer vaccine specific to the tumor antigen E7 present in TC1 cells. CPIs for the TC1 model were selected because of immunophenotyping TC1 tumors. Antitumor and protumor immunity, tumor size and survival, sequence and timing of vaccine and CPI administration, and efficacy of treatment in young and aged mice were probed. RESULTS: While "hot" CT26 tumors are treatable with combinations of second-generation CPIs alone or with anti-TGFß, "cold" TC1 tumor reduction requires the synergy of a tumor-antigen-specific vaccine in combination with two CPIs, anti-TIGIT and anti-PD-L1, predicted by tumor microenvironment (TME) characterization. The synergistic triple combination delays tumor growth better than any pairwise combination and improves survival in a CD8+T cell-dependent manner. Depletion of CD4+T cells improved the treatment response, and depleting regulatory T cells (Treg) revealed Tregs to be inhibiting the response as also predicted from TME analysis. We found the sequence of CPI and vaccine administration dictates the success of the treatment, and the triple combination administered concurrently induces the highest E7-specific T cell response. Contrary to young mice, in aged mice, the cancer vaccine alone is ineffective, requiring the CPIs to delay tumor growth. CONCLUSIONS: These findings show how pre-existing or vaccine-mediated de novo T cell responses can both be amplified by and facilitate synergistic CPIs and Treg depletion that together lead to greater survival, and how analysis of the TME can help rationally design combination therapies and precision medicine to enhance clinical response to CPI and cancer vaccine therapy.


Sujet(s)
Vaccins anticancéreux , Inhibiteurs de points de contrôle immunitaires , Lymphocytes T régulateurs , Microenvironnement tumoral , Animaux , Vaccins anticancéreux/pharmacologie , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/immunologie , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Microenvironnement tumoral/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Femelle , Lignée cellulaire tumorale , Humains
3.
Theranostics ; 14(9): 3674-3692, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948057

RÉSUMÉ

Trophoblast cell surface antigen 2 (Trop2) is overexpressed in a range of solid tumors and participants in multiple oncogenic signaling pathways, making it an attractive therapeutic target. In the past decade, the rapid development of various Trop2-targeted therapies, notably marked by the advent of the antibody-drug conjugate (ADC), revolutionized the outcome for patients facing Trop2-positive tumors with limited treatment opinions, such as triple-negative breast cancer (TNBC). This review provides a comprehensive summary of advances in Trop2-targeted therapies, including ADCs, antibodies, multispecific agents, immunotherapy, cancer vaccines, and small molecular inhibitors, along with in-depth discussions on their designs, mechanisms of action (MOAs), and limitations. Additionally, we emphasize the clinical research progress of these emerging Trop2-targeted agents, focusing on their clinical application and therapeutic efficacy against tumors. Furthermore, we propose directions for future research, such as enhancing our understanding of Trop2's structure and biology, exploring the best combination strategies, and tailoring precision treatment based on Trop2 testing methodologies.


Sujet(s)
Antigènes néoplasiques , Molécules d'adhérence cellulaire , Immunoconjugués , Thérapie moléculaire ciblée , Tumeurs , Humains , Antigènes néoplasiques/immunologie , Molécules d'adhérence cellulaire/antagonistes et inhibiteurs , Molécules d'adhérence cellulaire/métabolisme , Immunoconjugués/usage thérapeutique , Immunoconjugués/pharmacologie , Thérapie moléculaire ciblée/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Immunothérapie/méthodes , Animaux , Vaccins anticancéreux/usage thérapeutique
4.
Expert Opin Biol Ther ; 24(6): 415-423, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38861054

RÉSUMÉ

INTRODUCTION: BCG-unresponsive non-muscle invasive bladder cancer (NMIBC) represent a significant therapeutic challenge in the treatment of bladder cancer. Nadofaragene firadenovec, represents a breakthrough in this area, offering a novel approach for the treatment of BCG-unresponsive NMIBC. AREAS COVERED: This overview explores the historical development of nadofaragene firadenovec, assessing its efficacy and safety, and discusses future NMIBC therapy directions. EXPERT OPINION: Patients with high grade NMIBC who are BCG unresponsive will have a growing number of treatment alternatives to bladder removal. Nadofaragene firadenovec offers good short-term efficacy but lacks significant durability for most patients. Its strengths include ease of administration and low risk of adverse events. This will need to balance with risk of progression and cost. Furthermore, the likely approval of other agents will require consideration of which therapy to use and for which patient. The need for biomarkers to tailor treatment choices to individual patient needs is becoming more critical. The treatment field is rapidly advancing, with several Phase 3 single-arm trials underway, indicating a potential broader range of treatment options for NMIBC. Further research will be necessary to determine the optimal choice for patients.


Sujet(s)
Vaccin BCG , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Humains , Vaccin BCG/usage thérapeutique , Invasion tumorale , Vaccins anticancéreux/usage thérapeutique , Tumeurs de la vessie n'infiltrant pas le muscle
5.
Curr Oncol ; 31(6): 3040-3063, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38920716

RÉSUMÉ

Breast cancer is the most commonly diagnosed cancer in women and is a leading cause of cancer death in women worldwide. Despite the implementation of multiple treatment options, including immunotherapy, breast cancer treatment remains a challenge. In this review, we aim to summarize present challenges in breast cancer immunotherapy and recent advancements in overcoming treatment resistance. We elaborate on the inhibition of signaling cascades, such as the Notch, Hedgehog, Hippo, and WNT signaling pathways, which regulate the self-renewal and differentiation of breast cancer stem cells and, consequently, disease progression and survival. Cancer stem cells represent a rare population of cancer cells, likely originating from non-malignant stem or progenitor cells, with the ability to evade immune surveillance and develop resistance to immunotherapeutic treatments. We also discuss the interactions between breast cancer stem cells and the immune system, including potential agents targeting breast cancer stem cell-associated signaling pathways, and provide an overview of the emerging approaches to breast cancer stem cell-targeted immunotherapy. Finally, we consider the development of breast cancer vaccines and adoptive cellular therapies, which train the immune system to recognize tumor-associated antigens, for eliciting T cell-mediated responses to target breast cancer stem cells.


Sujet(s)
Tumeurs du sein , Immunothérapie , Cellules souches tumorales , Humains , Tumeurs du sein/immunologie , Tumeurs du sein/thérapie , Cellules souches tumorales/immunologie , Immunothérapie/méthodes , Femelle , Transduction du signal , Vaccins anticancéreux/usage thérapeutique
7.
Blood Cancer Discov ; 5(4): 234-248, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38904305

RÉSUMÉ

Despite advancements, acute myeloid leukemia (AML) remains unconquered by current therapies. Evidence of immune evasion during AML progression, such as HLA loss and T-cell exhaustion, suggests that antileukemic immune responses contribute to disease control and could be harnessed by immunotherapy. In this review, we discuss a spectrum of AML immunotherapy targets, encompassing cancer cell-intrinsic and surface antigens as well as targeting in the leukemic milieu, and how they can be tailored for personalized approaches. These targets are overviewed across major immunotherapy modalities applied to AML: immune checkpoint inhibitors, antibody-drug conjugates, therapeutic vaccines, bispecific/trispecific antibodies, and chimeric antigen receptor (CAR)-T and CAR-NK cells. Significance: Immune therapies in AML treatment show evolving promise. Ongoing research aims to customize approaches for varied patient profiles and clinical scenarios. This review covers immune surveillance mechanisms, therapy options like checkpoint inhibitors, antibodies, CAR-T/NK cells, and vaccines, as well as resistance mechanisms and microenvironment considerations.


Sujet(s)
Immunothérapie , Leucémie aigüe myéloïde , Humains , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/thérapie , Immunothérapie/méthodes , Immunothérapie/tendances , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/immunologie , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie
8.
Science ; 384(6703): eadh4567, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38935717

RÉSUMÉ

Inflammation and tissue damage associated with pancreatitis can precede or occur concurrently with pancreatic ductal adenocarcinoma (PDAC). We demonstrate that in PDAC coupled with pancreatitis (ptPDAC), antigen-presenting type I conventional dendritic cells (cDC1s) are specifically activated. Immune checkpoint blockade therapy (iCBT) leads to cytotoxic CD8+ T cell activation and elimination of ptPDAC with restoration of life span even upon PDAC rechallenge. Using PDAC antigen-loaded cDC1s as a vaccine, immunotherapy-resistant PDAC was rendered sensitive to iCBT with elimination of tumors. cDC1 vaccination coupled with iCBT identified specific CDR3 sequences in the tumor-infiltrating CD8+ T cells with potential therapeutic importance. This study identifies a fundamental difference in the immune microenvironment in PDAC concurrent with, or without, pancreatitis and provides a rationale for combining cDC1 vaccination with iCBT as a potential treatment option.


Sujet(s)
Carcinome du canal pancréatique , Cellules dendritiques , Immunothérapie , Tumeurs du pancréas , Microenvironnement tumoral , Animaux , Souris , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/immunologie , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Souris de lignée C57BL , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Pancréatite/immunologie , Pancréatite/thérapie , Microenvironnement tumoral/immunologie
10.
Cancer Lett ; 595: 216989, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-38825162

RÉSUMÉ

Exosomes, a subset of extracellular vesicles, are released by all active cells and play a crucial role in intercellular communications. Exosomes could facilitate the transfer of various biologically active molecules, such as DNA, non-coding RNAs, and proteins, from donor to recipient cells, thereby participating in diverse biological and pathological processes. Besides, exosomes possess unique characteristics, including non-toxicity, low-immunogenicity, and stability within biological systems, rendering them highly advantageous for cancer drug development. Meanwhile, accumulating evidence suggests that exosomes originating from tumor cells and immune cells possess distinct composition profiles that play a direct role in anticancer immunotherapy. Of note, exosomes can transport their contents to specific cells, thereby exerting an impact on the phenotype and immune-regulatory functions of targeted cells. Therapeutic cancer vaccines, an emerging therapeutics of immunotherapy, could enhance antitumor immune responses by delivering a large number of tumor antigens, thereby augmenting the immune response against tumor cells. Therefore, the therapeutic rationale of cancer vaccines and exosome-based immunotherapy are almost similar to some extent, but some challenges have hindered their application in the clinical setting. Here, in this review, we first summarized the biogenesis, structure, compositions, and biological functions of exosomes. Then we described the roles of exosomes in cancer biology, particularly in tumor immunity. We also comprehensively reviewed current exosome-based anticancer vaccine development and we divided them into three types. Finally, we give some insights into clinical translation and clinical trial progress of exosome-based anticancer vaccines for future direction.


Sujet(s)
Vaccins anticancéreux , Exosomes , Immunothérapie , Tumeurs , Humains , Exosomes/immunologie , Exosomes/métabolisme , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Tumeurs/immunologie , Tumeurs/thérapie , Immunothérapie/méthodes , Animaux
11.
World J Gastroenterol ; 30(21): 2734-2739, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38899328

RÉSUMÉ

In this editorial we comment on the article by Li published in the recent issue of the World Journal of Gastroenterology. We focus specifically on the application of immune checkpoint inhibitors (ICIs) and microsatellite instability (MSI) in gastric cancer (GC). The four pillars of GC management have long been considered, including surgery, chemotherapy, radiotherapy and targeted therapy. However, immunotherapy has recently emerged as a "fifth pillar", and its use is rapidly expanding. There are four principal strategies for tumor immunotherapy: ICIs, tumor vaccines, adoptive immunotherapy and nonspecific immunomodulators. Of them, ICIs are the most advanced and widespread type of cancer immunotherapy for GC. Recent breakthrough results for ICIs have paved the way to a new era of cancer immunotherapy. In particular, inhibition of the PD-1/PD-L1 axis with ICIs, including nivolumab and pembrolizumab, has emerged as a novel treatment strategy for advanced GC. Unfortunately, these therapies are sometimes associated with often subtle, potentially fatal immune-related adverse events (irAEs), including dermatitis, diarrhea, colitis, endocrinopathy, hepatotoxicity, neuropathy and pneumonitis. We must be aware of these irAEs and improve the detection of these processes to prevent inappropriate discharges, emergency department revisits, and downstream complications. Recent studies have revealed that MSI-high or mismatch- repair-deficient tumors, regardless of their primary site, have a promising response to ICIs. So, it is important to detect MSI before applying ICIs for treatment of GC.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Instabilité des microsatellites , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/thérapie , Tumeurs de l'estomac/anatomopathologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Antigène CD274/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/immunologie , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Immunothérapie/méthodes , Immunothérapie/effets indésirables , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/effets indésirables , Vaccins anticancéreux/immunologie
13.
J Hematol Oncol ; 17(1): 40, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38835055

RÉSUMÉ

Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.


Sujet(s)
Immunothérapie , Tumeurs du pancréas , Microenvironnement tumoral , Humains , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Immunothérapie/méthodes , Microenvironnement tumoral/immunologie , Vaccins anticancéreux/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Animaux , Immunothérapie adoptive/méthodes
16.
Front Immunol ; 15: 1414737, 2024.
Article de Anglais | MEDLINE | ID: mdl-38938562

RÉSUMÉ

This report details a case of pancreatic cancer with liver metastasis that exhibited a positive immune response to personalized immunization therapy. Our study involved the identification of neoantigens and their corresponding immunogenic peptides using an in-house bioinformatic pipeline. This process included the identification of somatic mutations through DNA/RNA sequencing of solid tumor tissue and blood liquid biopsy. Computational prediction techniques were then employed to identify novel epitopes, followed by the design and manufacture of patient-specific immunization peptides. In combination with standard-of-care chemotherapy, the patient received a sequence of 5 biweekly prime injections followed by 2 boost injections 2 and 5 months later. The peptides were emulsified in Montanide and the injection-site was conditioned with nivolumab and imiquimod. The combined regimen of peptide immunization and chemotherapy resulted in a notable decline in CA19-9 tumor marker levels following both prime and boost applications. Subsequent MRI assessments revealed a reduction in the size of liver metastases several months post-immunization initiation. Importantly, the patient showed and improved overall survival and reported an improved quality of life without experiencing significant treatment-related adverse effects. This case underscores the potential benefits of personalized peptide-based immunization as an adjunctive therapy in the treatment of advanced pancreatic cancer, showcasing promising outcomes in tumor marker reduction, tumor shrinkage, and enhanced patient well-being.


Sujet(s)
Antigènes néoplasiques , Tumeurs du pancréas , Médecine de précision , Humains , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Antigènes néoplasiques/immunologie , Biopsie liquide/méthodes , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/usage thérapeutique , Tumeurs du foie/secondaire , Tumeurs du foie/thérapie , Tumeurs du foie/immunologie , Mâle , Peptides/immunologie , Peptides/administration et posologie , Adulte d'âge moyen , Vaccins sous-unitaires/administration et posologie , Immunisation , Femelle , Marqueurs biologiques tumoraux
17.
Curr Oncol Rep ; 26(7): 826-839, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38789670

RÉSUMÉ

PURPOSE OF REVIEW: This review provides a comprehensive update on recent advancements in melanoma treatment by highlighting promising therapeutics with an aim to increase awareness of novel interventions currently in development. RECENT FINDINGS: Over the last decade there has been considerable expansion of the previously available treatment options for patients with melanoma. In particular, novel immunotherapeutics have been developed to expand on the clinical advancements brought by BRAF targeting and immune checkpoint inhibitors. Despite the success of checkpoint inhibitors there remains an unmet need for patients that do not respond to treatment. This review delves into the latest advancements in novel checkpoint inhibitors, cytokines, oncolytic viruses, vaccines, bispecific antibodies, and adoptive cell therapy. Preclinical experiments and early-stage clinical trials studies have demonstrated promising results for these therapies, many of which have moved into pivotal, phase 3 studies.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Mélanome , Humains , Mélanome/traitement médicamenteux , Mélanome/immunologie , Mélanome/thérapie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Vaccins anticancéreux/usage thérapeutique , Virus oncolytiques/immunologie , Anticorps bispécifiques/usage thérapeutique , Cytokines/antagonistes et inhibiteurs , Cytokines/métabolisme , Thérapie virale de cancers/méthodes , Thérapie moléculaire ciblée/méthodes
18.
Cell ; 187(10): 2521-2535.e21, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38697107

RÉSUMÉ

Cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Here, we create "onion-like" multi-lamellar RNA lipid particle aggregates (LPAs) to substantially enhance the payload packaging and immunogenicity of tumor mRNA antigens. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for Toll-like receptor engagement in immune cells, systemically administered RNA-LPAs activate RIG-I in stromal cells, eliciting massive cytokine/chemokine response and dendritic cell/lymphocyte trafficking that provokes cancer immunogenicity and mediates rejection of both early- and late-stage murine tumor models. In client-owned canines with terminal gliomas, RNA-LPAs improved survivorship and reprogrammed the TME, which became "hot" within days of a single infusion. In a first-in-human trial, RNA-LPAs elicited rapid cytokine/chemokine release, immune activation/trafficking, tissue-confirmed pseudoprogression, and glioma-specific immune responses in glioblastoma patients. These data support RNA-LPAs as a new technology that simultaneously reprograms the TME while eliciting rapid and enduring cancer immunotherapy.


Sujet(s)
Immunothérapie , Lipides , ARN , Microenvironnement tumoral , Animaux , Chiens , Femelle , Humains , Souris , Antigènes néoplasiques/immunologie , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/immunologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Lignée cellulaire tumorale , Cytokines/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Glioblastome/thérapie , Glioblastome/immunologie , Gliome/thérapie , Gliome/immunologie , Immunothérapie/méthodes , Souris de lignée C57BL , Tumeurs/thérapie , Tumeurs/immunologie , ARN/composition chimique , ARN/usage thérapeutique , ARN messager/métabolisme , ARN messager/génétique , Lipides/composition chimique
19.
Nat Commun ; 15(1): 3882, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38719809

RÉSUMÉ

In this randomized phase II clinical trial, we evaluated the effectiveness of adding the TLR agonists, poly-ICLC or resiquimod, to autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination in patients with newly-diagnosed or recurrent WHO Grade III-IV malignant gliomas. The primary endpoints were to assess the most effective combination of vaccine and adjuvant in order to enhance the immune potency, along with safety. The combination of ATL-DC vaccination and TLR agonist was safe and found to enhance systemic immune responses, as indicated by increased interferon gene expression and changes in immune cell activation. Specifically, PD-1 expression increases on CD4+ T-cells, while CD38 and CD39 expression are reduced on CD8+ T cells, alongside an increase in monocytes. Poly-ICLC treatment amplifies the induction of interferon-induced genes in monocytes and T lymphocytes. Patients that exhibit higher interferon response gene expression demonstrate prolonged survival and delayed disease progression. These findings suggest that combining ATL-DC with poly-ICLC can induce a polarized interferon response in circulating monocytes and CD8+ T cells, which may represent an important blood biomarker for immunotherapy in this patient population.Trial Registration: ClinicalTrials.gov Identifier: NCT01204684.


Sujet(s)
Lymphocytes T CD8+ , Vaccins anticancéreux , Carboxyméthylcellulose de sodium/analogues et dérivés , Cellules dendritiques , Gliome , Interférons , Poly I-C , Polylysine/analogues et dérivés , Humains , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Gliome/immunologie , Gliome/thérapie , Femelle , Mâle , Adulte d'âge moyen , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/usage thérapeutique , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Poly I-C/administration et posologie , Poly I-C/pharmacologie , Adulte , Récepteurs de type Toll/agonistes , Imidazoles/pharmacologie , Imidazoles/usage thérapeutique , Sujet âgé , Vaccination , Monocytes/immunologie , Monocytes/effets des médicaments et des substances chimiques , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/traitement médicamenteux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Immunothérapie/méthodes ,
20.
Am Soc Clin Oncol Educ Book ; 44(3): e433330, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38718318

RÉSUMÉ

The treatment for recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) with immune checkpoint inhibitors (anti-PD1) with or without chemotherapy has led to an improvement in survival. Yet, despite this therapeutic advancement, only 15%-19% of patients remain alive at four years, highlighting the poor survival and unmet need for improved therapies for this patient population. Some of the key evolving novel therapeutics beyond anti-PD1 in R/M HNSCC have included therapeutic vaccine therapies, bispecific antibodies/fusion proteins and multitargeted kinase inhibitors, and antibody-drug conjugates (ADCs). Multiple concurrent investigations of novel therapeutics for patients with R/M HNSCC beyond anti-PD(L)1 inhibition are currently underway with some promising early results. Beyond immune checkpoint inhibition, novel immunotherapeutic strategies including therapeutic vaccines ranging from targeting human papillomavirus-specific epitopes to personalized neoantigen vaccines are ongoing with some early efficacy signals and large, randomized trials. Other novel weapons including bispecific antibodies, fusion proteins, and multitargeted kinase inhibitors leverage multiple concurrent targets and modulation of the tumor microenvironment to harness antitumor immunity and inhibition of protumorigenic signaling pathways with emerging promising results. Finally, as with other solid tumors, ADCs remain a promising therapeutic intervention either alone or in combination with immunotherapy for patients with R/M HNSCC. With early enthusiasm across novel therapies in R/M HNSCC, results of larger randomized trials in R/M HNSCC are eagerly awaited.


Sujet(s)
Immunothérapie , Carcinome épidermoïde de la tête et du cou , Humains , Carcinome épidermoïde de la tête et du cou/thérapie , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Immunothérapie/méthodes , Tumeurs de la tête et du cou/thérapie , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/traitement médicamenteux , Antigène CD274/antagonistes et inhibiteurs , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Thérapie moléculaire ciblée , Métastase tumorale , Récidive tumorale locale , Vaccins anticancéreux/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...