Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Mol Med ; 12(6): e10979, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32419365

ABSTRACT

Drugs that mobilise the immune system against cancer are dramatically improving care for many people. Dying cancer cells play an active role in inducing anti-tumour immunity but not every form of death can elicit an immune response. Moreover, resistance to apoptosis is a major problem in cancer treatment and disease control. While the term "immunogenic cell death" is not fully defined, activation of receptor-interacting serine/threonine-protein kinase 1 (RIPK1) can induce a type of death that mobilises the immune system against cancer. However, no clinical treatment protocols have yet been established that would harness the immunogenic potential of RIPK1. Here, we report the first pre-clinical application of an in vivo treatment protocol for soft-tissue sarcoma that directly engages RIPK1-mediated immunogenic cell death. We find that RIPK1-mediated cell death significantly improves local disease control, increases activation of CD8+ T cells as well as NK cells, and enhances the survival benefit of immune checkpoint blockade. Our findings warrant a clinical trial to assess the survival benefit of RIPK1-induced cell death in patients with advanced disease at limb extremities.


Subject(s)
Immunogenic Cell Death , Sarcoma , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Humans , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Sarcoma/therapy , Signal Transduction , Tumor Necrosis Factor-alpha
2.
Clin Cancer Res ; 25(11): 3443-3454, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30885937

ABSTRACT

PURPOSE: The prevention and treatment of metastatic sarcoma are areas of significant unmet need. Immune checkpoint inhibitor monotherapy has shown little activity in sarcoma and there is great interest in identifying novel treatment combinations that may augment responses. In vitro and in vivo, we investigated the potential for an oncolytic vaccinia virus (GLV-1h68) delivered using isolated limb perfusion (ILP) to promote antitumor immune responses and augment response to PD-1 blockade in sarcoma.Experimental Design: In an established animal model of extremity sarcoma, we evaluated the potential of locoregional delivery of a vaccinia virus (GLV-1h68) alongside biochemotherapy (melphalan/TNFα) in ILP. Complementary in vitro assays for markers of immunogenic cell death were performed in sarcoma cell lines. RESULTS: PD-1 monotherapy had minimal efficacy in vivo, mimicking the clinical scenario. Pretreatment with GLV-1h68 delivered by ILP (viral ILP) significantly improved responses. Furthermore, when performed prior to surgery and radiotherapy, viral ILP and PD-1 blockade prevented both local and distant relapse, curing a previously treatment-refractory model. Enhanced therapy was associated with marked modulation of the tumor microenvironment, with an increase in the number and penetrance of intratumoral CD8+ T cells and expansion and activation of dendritic cells. GLV-1h68 was capable of inducing markers of immunogenic cell death in human sarcoma cell lines. CONCLUSIONS: Viral ILP augments the response to PD-1 blockade, transforming this locoregional therapy into a potentially effective systemic treatment for sarcoma and warrants translational evaluation.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Genetic Therapy , Humans , Immunohistochemistry , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Sarcoma/etiology , Sarcoma/metabolism , Sarcoma/pathology , Sarcoma/therapy , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Oncotarget ; 7(49): 81208-81222, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27783991

ABSTRACT

Advanced extremity melanoma and sarcoma present a significant therapeutic challenge, requiring multimodality therapy to treat or even palliate disease. These aggressive tumours are relatively chemo-resistant, therefore new treatment approaches are urgently required. We have previously reported on the efficacy of oncolytic virotherapy (OV) delivered by isolated limb perfusion. In this report, we have improved therapeutic outcomes by combining OV with radiotherapy. In vitro, the combination of oncolytic vaccinia virus (GLV-1h68) and radiotherapy demonstrated synergistic cytotoxicity. This effect was not due to increased viral replication, but mediated through induction of intrinsic apoptosis. GLV-1h68 therapy downregulated the anti-apoptotic BCL-2 proteins (MCL-1 and BCL-XL) and the downstream inhibitors of apoptosis, resulting in cleavage of effector caspases 3 and 7. In an in vivo ILP model, the combination of OV and radiotherapy significantly delayed tumour growth and prolonged survival compared to single agent therapy. These data suggest that the virally-mediated down-regulation of anti-apoptotic proteins may increase the sensitivity of tumour cells to the cytotoxic effects of ionizing radiation. Oncolytic virotherapy represents an exciting candidate for clinical development when delivered by ILP. Its ability to overcome anti-apoptotic signals within tumour cells points the way to further development in combination with conventional anti-cancer therapies.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/radiation effects , Fibrosarcoma/therapy , Oncolytic Virotherapy , Oncolytic Viruses/pathogenicity , Vaccinia virus/pathogenicity , Animals , Apoptosis Regulatory Proteins/genetics , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Dose-Response Relationship, Radiation , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Fibrosarcoma/virology , Gene Expression Regulation, Neoplastic , Host-Pathogen Interactions , Humans , Male , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Radiotherapy, Adjuvant , Rats, Inbred BN , Signal Transduction/radiation effects , Time Factors , bcl-X Protein/metabolism
4.
Int J Cancer ; 139(6): 1414-22, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27116656

ABSTRACT

The management of locally advanced or recurrent extremity sarcoma often necessitates multimodal therapy to preserve a limb, of which isolated limb perfusion (ILP) is a key component. However, with standard chemotherapeutic agents used in ILP, the duration of response is limited. Novel agents or treatment combinations are urgently needed to improve outcomes. Previous work in an animal model has demonstrated the efficacy of oncolytic virotherapy when delivered by ILP and, in this study, we report further improvements from combining ILP-delivered oncolytic virotherapy with radiation and surgical resection. In vitro, the combination of radiation with an oncolytic vaccinia virus (GLV-1h68) and melphalan demonstrated increased cytotoxicity in a panel of sarcoma cell lines. The effects were mediated through activation of the intrinsic apoptotic pathway. In vivo, combinations of radiation, oncolytic virotherapy and standard ILP resulted in delayed tumour growth and prolonged survival when compared with standard ILP alone. However, local disease control could only be secured when such treatment was combined with surgical resection, the timing of which was crucial in determining outcome. Combinations of oncolytic virotherapy with surgical resection and radiation have direct clinical relevance in extremity sarcoma and represent an exciting prospect for improving outcomes in this pathology.


Subject(s)
Antineoplastic Agents/administration & dosage , Chemotherapy, Cancer, Regional Perfusion , Combined Modality Therapy , Oncolytic Virotherapy , Radiotherapy , Sarcoma/pathology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Disease Models, Animal , Extremities , Genetic Vectors/genetics , Humans , Male , Melphalan/administration & dosage , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Proton Therapy , Radiotherapy/methods , Rats , Recurrence , Sarcoma/genetics , Sarcoma/mortality , Sarcoma/therapy , Transduction, Genetic , Tumor Burden/drug effects , Tumor Burden/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...