Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Int Rev Immunol ; 41(6): 582-605, 2022.
Article in English | MEDLINE | ID: mdl-35938932

ABSTRACT

The aggressive and recurrent nature of glioblastoma is multifactorial and has been attributed to its biological heterogeneity, dysfunctional metabolic signaling pathways, rigid blood-brain barrier, inherent resistance to standard therapy due to the stemness property of the gliomas cells, immunosuppressive tumor microenvironment, hypoxia and neoangiogenesis which are very well orchestrated and create the tumor's own highly pro-tumorigenic milieu. Once the relay of events starts amongst these components, eventually it becomes difficult to control the cascade using only the balanced contemporary care of treatment consisting of maximal resection, radiotherapy and chemotherapy with temozolamide. Over the past few decades, implementation of contemporary treatment modalities has shown benefit to some extent, but no significant overall survival benefit is achieved. Therefore, there is an unmet need for advanced multifaceted combinatorial strategies. Recent advances in molecular biology, development of innovative therapeutics and novel delivery platforms over the years has resulted in a paradigm shift in gliomas therapeutics. Decades of research has led to emergence of several treatment molecules, including immunotherapies such as immune checkpoint blockade, oncolytic virotherapy, adoptive cell therapy, nanoparticles, CED and BNCT, each with the unique proficiency to overcome the mentioned challenges, present research. Recent years are seeing innovative combinatorial strategies to overcome the multifactorial resistance put forth by the GBM cell and its TME. This review discusses the contemporary and the investigational combinatorial strategies being employed to treat GBM and summarizes the evidence accumulated till date.


Glioblastoma is a form of brain tumor which typically leads to death in almost all patients. Over the last two decades, traditional treatment strategies such as surgery, radiotherapy and chemotherapy have been combined as standard therapy. Together, these aggressive treatment strategies have provided modest survival benefit with acceptable toxicity. However, relapse is the invariable norm resulting in death in the overwhelming majority of patients. Relapse occurs due to multiple factors such as inability of drugs to cross blood­brain barrier, immunosuppressive tumor microenvironment, stemness nature of glioma cells, tumor heterogeneity and enhanced hypoxia and angiogenic factors. Therefore, there is an urgent need to develop an innovative treatment approach to treat glioblastoma. Recently, several treatment strategies known as immunotherapies including CAR T cell therapy, dendritic cell vaccines, immune checkpoints blockade and oncolytic virus, nano particles and gene editing/silencing technology have demonstrated promising results in preclinical and few clinical trials. Furthermore, to increase the efficacy of these novel strategies, combinatorial approaches are being implemented for the treatment. This includes CAR T cell therapy in combination with small molecules, immune checkpoint inhibitors and oncolytic virus and nanoparticles plus gene editing, silencing or immune checkpoints inhibitors. These treatments have shown exciting results in preclinical settings and few of these trials are in progress. The review summarizes these combinatorial novel approaches and discusses them in detail.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Oncolytic Virotherapy , Humans , Glioblastoma/drug therapy , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Immune Checkpoint Inhibitors , Oncolytic Virotherapy/methods , Immunotherapy/methods , Glioma/drug therapy , Tumor Microenvironment
2.
Front Immunol ; 13: 886546, 2022.
Article in English | MEDLINE | ID: mdl-35677038

ABSTRACT

Chimeric antigen receptor T cell (CAR-T) therapy demonstrated remarkable success in long-term remission of cancers and other autoimmune diseases. Currently, six products (Kymriah, Yescarta, Tecartus, Breyanzi, Abecma, and Carvykti) are approved by the US-FDA for treatment of a few hematological malignancies. All the six products are autologous CAR-T cell therapies, where delivery of CAR, which comprises of scFv (single-chain variable fragment) derived from monoclonal antibodies for tumor target antigen recognition is through a lentiviral vector. Although available CAR-T therapies yielded impressive response rates in a large number of patients in comparison to conventional treatment strategies, there are potential challenges in the field which limit their efficacy. One of the major challenges is the induction of humoral and/or cellular immune response in patients elicited due to scFv domain of CAR construct, which is of non-human origin in majority of the commercially available products. Generation of anti-CAR antibodies may lead to the clearance of the therapeutic CAR-T cells, increasing the likelihood of tumor relapse and lower the CAR-T cells efficacy upon reinfusion. These immune responses influence CAR-T cell expansion and persistence, that might affect the overall clinical response. In this review, we will discuss the impact of immunogenicity of the CAR transgene on treatment outcomes. Finally, this review will highlight the mitigation strategies to limit the immunogenic potential of CARs and improve the therapeutic outcome.


Subject(s)
Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Neoplasm Recurrence, Local , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
3.
Mol Cancer Ther ; 20(5): 846-858, 2021 05.
Article in English | MEDLINE | ID: mdl-33632869

ABSTRACT

Recent studies have described the remarkable clinical outcome of anti-CD19 chimeric antigen receptor (CAR) T cells in treating B-cell malignancies. However, over 50% of patients develop life-threatening toxicities associated with cytokine release syndrome which may limit its utilization in low-resource settings. To mitigate the toxicity, we designed a novel humanized anti-CD19 CAR T cells by humanizing the framework region of single-chain variable fragment (scFv) derived from a murine FMC63 mAb and combining it with CD8α transmembrane domain, 4-1BB costimulatory domain, and CD3ζ signaling domain (h1CAR19-8BBζ). Docking studies followed by molecular dynamics simulation revealed that the humanized anti-CD19 scFv (h1CAR19) establishes higher binding affinity and has a flexible molecular structure with CD19 antigen compared with murine scFv (mCAR19). Ex vivo studies with CAR T cells generated from healthy donors and patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) expressing either h1CAR19 or mCAR19 showed comparable antitumor activity and proliferation. More importantly, h1CAR19-8BBζ T cells produced lower levels of cytokines (IFNγ, TNFα) upon antigen encounter and reduced the induction of IL6 cytokine from monocytes than mCAR19-8BBζ T cells. There was a comparable proliferation of h1CAR19-8BBζ T cells and mCAR19-8BBζ T cells upon repeated antigen encounter. Finally, h1CAR19-8BBζ T cells efficiently eliminated NALM6 tumor cells in a preclinical model. In conclusion, the distinct structural modification in CAR design confers the novel humanized anti-CD19 CAR with a favorable balance of efficacy to toxicity providing a rationale to test this construct in a phase I trial.


Subject(s)
Antigens, CD19/metabolism , Cytokines/metabolism , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL