Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Cancers (Basel) ; 15(24)2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38136335

ABSTRACT

Glioblastoma (GBM) is the most common primary malignant brain tumour, and it confers a dismal prognosis despite intensive multimodal treatments. Whilst historically, research has focussed on the evolution of GBM tumour cells themselves, there is growing recognition of the importance of studying the tumour microenvironment (TME). Improved characterisation of the interaction between GBM cells and the TME has led to a better understanding of therapeutic resistance and the identification of potential targets to block these escape mechanisms. This review describes the network of cells within the TME and proposes treatment strategies for simultaneously targeting GBM cells, the surrounding immune cells, and the crosstalk between them.

3.
Praxis (Bern 1994) ; 112(3): 189-193, 2023.
Article in English | MEDLINE | ID: mdl-36855881

ABSTRACT

Oncology has been rapidly evolving over the past decade with tremendous therapeutic development. Engineered cell therapies such as chimeric antigen receptor (CAR)-T cells are increasingly used in daily practice, and provided a paradigm change especially for hematological malignancies. Their development is a scientific and technological achievement, but their toxicities can be life-threatening. As their utilization expands, better understanding of pathophysiology leads to better management. In this article we present a general overview of cell-therapy toxicities and their management.


Subject(s)
Hematologic Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/therapeutic use , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/therapy , Cell- and Tissue-Based Therapy , Hematologic Neoplasms/therapy
4.
Praxis (Bern 1994) ; 112(3): 160-171, 2023.
Article in English | MEDLINE | ID: mdl-36855889

ABSTRACT

Immunotherapy with immune checkpoint inhibitors (ICI) is administered in different cancer types and can lead to a wide range of immune-related adverse events including toxicity in vital organs such as the lungs, the kidneys, and the heart. The main hypothesis suggests an overactivation of the immune cells in the different organs. Whereas immune-related cardiotoxicity is very rare but life threatening, ICI-induced acute kidney injury and pneumonitis are more frequent but in general less severe. Renal toxicity corresponds in more than 90% to an acute tubulo-interstitial nephritis. Checkpoint inhibitors pneumonitis is diagnosed mainly on respiratory symptoms with new radiological features, especially under the form of a cryptogenic organising pneumonia. Cardiotoxicity is predominantly marked by myocarditis but also pericarditis and arrhythmias, among others. Early recognition, temporary or definitive cessation of ICI therapy and rapid initiation of high-dose corticosteroids are the cornerstones of the management, which must to be multidisciplinary in a specialised center.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Cardiotoxicity/etiology , Neoplasms/drug therapy , Kidney , Lung/diagnostic imaging
5.
Front Neurol ; 14: 1108297, 2023.
Article in English | MEDLINE | ID: mdl-36970518

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy represents a scientific breakthrough in the treatment of advanced hematological malignancies. It relies on cell engineering to direct the powerful cytotoxic T-cell activity toward tumor cells. Nevertheless, these highly powerful cell therapies can trigger substantial toxicities such as cytokine release syndrome (CRS) and immune cell-associated neurological syndrome (ICANS). These potentially fatal side effects are now better understood and managed in the clinic but still require intensive patient follow-up and management. Some specific mechanisms seem associated with the development of ICANS, such as cytokine surge caused by activated CAR-T cells, off-tumor targeting of CD19, and vascular leak. Therapeutic tools are being developed aiming at obtaining better control of toxicity. In this review, we focus on the current understanding of ICANS, novel findings, and current gaps.

6.
JCI Insight ; 6(18)2021 09 22.
Article in English | MEDLINE | ID: mdl-34403371

ABSTRACT

Glioblastoma is a highly malignant brain tumor with no curative treatment options, and immune checkpoint blockade has not yet shown major impact. We hypothesized that drugs targeting mitosis might affect the tumor microenvironment and sensitize cancer cells to immunotherapy. We used 2 glioblastoma mouse models with different immunogenicity profiles, GL261 and SB28, to test the efficacy of antineoplastic and immunotherapy combinations. The spindle assembly checkpoint activator BAL101553 (lisavanbulin), agonistic anti-CD40 antibody, and double immune checkpoint blockade (anti-programmed cell death 1 and anti-cytotoxic T lymphocyte-associated protein 4; anti-PD-1 and anti-CTLA-4) were evaluated individually or in combination for treating orthotopic GL261 and SB28 tumors. Genomic and immunological analyses were used to predict and interpret therapy responsiveness. BAL101553 monotherapy increased survival in immune checkpoint blockade-resistant SB28 glioblastoma tumors and synergized with anti-CD40 antibody, in a T cell-independent manner. In contrast, the more immunogenic and highly mutated GL261 model responded best to anti-PD-1 and anti-CTLA-4 therapy and more modestly to BAL101553 and anti-CD40 combination. Our results show that BAL101553 is a promising therapeutic agent for glioblastoma and could synergize with innate immune stimulation. Overall, these data strongly support immune profiling of glioblastoma patients and preclinical testing of combination therapies with appropriate models for particular patient groups.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Benzimidazoles/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Mitosis/drug effects , Oxadiazoles/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Benzimidazoles/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , CD40 Antigens/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Expression/drug effects , Glioblastoma/genetics , Glioblastoma/immunology , HMGB1 Protein/metabolism , Interferon-gamma/genetics , Mice , Neoplasm Transplantation , Oxadiazoles/pharmacology , Programmed Cell Death 1 Receptor/immunology , Survival Rate , Temozolomide/therapeutic use , Tumor Microenvironment/drug effects
7.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34083417

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is refractory to immune checkpoint inhibitor (ICI) therapy. We sought to determine to what extent this immune evasion is due to intrinsic properties of the tumor cells versus the specialized immune context of the brain, and if it can be reversed. METHODS: We used CyTOF mass cytometry to compare the tumor immune microenvironments (TIME) of human tumors that are generally ICI-refractory (GBM and sarcoma) or ICI-responsive (renal cell carcinoma), as well as mouse models of GBM that are ICI-responsive (GL261) or ICI-refractory (SB28). We further compared SB28 tumors grown intracerebrally versus subcutaneously to determine how tumor site affects TIME and responsiveness to dual CTLA-4/PD-1 blockade. Informed by these data, we explored rational immunotherapeutic combinations. RESULTS: ICI-sensitivity in human and mouse tumors was associated with increased T cells and dendritic cells (DCs), and fewer myeloid cells, in particular PD-L1+ tumor-associated macrophages. The SB28 mouse model of GBM responded to ICI when grown subcutaneously but not intracerebrally, providing a system to explore mechanisms underlying ICI resistance in GBM. The response to ICI in the subcutaneous SB28 model required CD4 T cells and NK cells, but not CD8 T cells. Recombinant FLT3L expanded DCs, improved antigen-specific T cell priming, and prolonged survival of mice with intracerebral SB28 tumors, but at the cost of increased Tregs. Targeting PD-L1 also prolonged survival, especially when combined with stereotactic radiation. CONCLUSIONS: Our data suggest that a major obstacle for effective immunotherapy of GBM is poor antigen presentation in the brain, rather than intrinsic immunosuppressive properties of GBM tumor cells. Deep immune profiling identified DCs and PD-L1+ tumor-associated macrophages as promising targetable cell populations, which was confirmed using therapeutic interventions in vivo.


Subject(s)
Brain Neoplasms/therapy , CTLA-4 Antigen/metabolism , Glioblastoma/therapy , Immune Checkpoint Inhibitors/administration & dosage , Membrane Proteins/administration & dosage , Programmed Cell Death 1 Receptor/metabolism , Animals , Brain Neoplasms/immunology , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Glioblastoma/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Membrane Proteins/pharmacology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory/metabolism , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
8.
Int J Mol Sci ; 22(7)2021 Mar 28.
Article in English | MEDLINE | ID: mdl-33800593

ABSTRACT

Glioblastoma is the most frequent primary neoplasm of the central nervous system and still suffers from very poor therapeutic impact. No clear improvements over current standard of care have been made in the last decade. For other cancers, but also for brain metastasis, which harbors a very distinct biology from glioblastoma, immunotherapy has already proven its efficacy. Efforts have been pursued to allow glioblastoma patients to benefit from these new approaches, but the road is still long for broad application. Here, we aim to review key glioblastoma immune related characteristics, current immunotherapeutic strategies being explored, their potential caveats, and future directions.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , Glioblastoma/immunology , Glioblastoma/therapy , Immunotherapy/methods , Cancer Vaccines/therapeutic use , Central Nervous System/pathology , Clinical Trials as Topic , Humans , Immune System , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/trends , Medical Oncology/methods , Medical Oncology/trends , Neoplasm Metastasis , Oncolytic Viruses
9.
Oncoimmunology ; 7(12): e1501137, 2018.
Article in English | MEDLINE | ID: mdl-30524896

ABSTRACT

Immune checkpoint blockade (ICB) is currently evaluated in patients with glioblastoma (GBM), based on encouraging clinical data in other cancers, and results from studies with the methylcholanthrene-induced GL261 mouse glioma. In this paper, we describe a novel model faithfully recapitulating some key human GBM characteristics, including low mutational load, a factor reported as a prognostic indicator of ICB response. Consistent with this observation, SB28 is completely resistant to ICB, contrasting with treatment sensitivity of the more highly mutated GL261. Moreover, SB28 shows features of a poorly immunogenic tumor, with low MHC-I expression and modest CD8+ T-cell infiltration, suggesting that it may present similar challenges for immunotherapy as human GBM. Based on these key features for immune reactivity, SB28 may represent a treatment-resistant malignancy likely to mirror responses of many human tumors. We therefore propose that SB28 is a particularly suitable model for optimization of GBM immunotherapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...