scispace - formally typeset
Search or ask a question

What are the current advancements in understanding the role of the tumor microenvironment in neuroblastoma development and progression? 


Best insight from top research papers

Advancements in understanding the role of the tumor microenvironment (TME) in neuroblastoma (NB) development and progression have been made. The TME in NB consists of various components, including tumor-associated macrophages (TAMs) and stromal cells . TAMs, particularly those with an M2-like phenotype, play a crucial role in mediating NB development and immune evasion . The immune infiltrative profiles of NB have been analyzed, and high immune cell infiltration (ICI) subtypes have been associated with better overall survival and potential responsiveness to immunotherapy . The TME also influences the efficacy of immunotherapeutic approaches, such as chimeric antigen receptor (CAR) T cell therapy, due to its immunosuppressive nature . Strategies aimed at enhancing innate immunity, particularly natural killer (NK) cells and macrophages, are being explored to overcome these challenges . Additionally, 3D bioprinting and NB spheroid technologies have been integrated to create in vitro models that can study the role of the NB microenvironment in tumor progression . These advancements provide insights into the complex interactions within the TME and offer potential targets for precision immunotherapy in NB.

Answers from top 4 papers

More filters
Papers (4)Insight
The paper discusses the immunosuppressive nature of the tumor microenvironment (TME) in neuroblastoma and the role of various immunoregulatory mechanisms in tumor development and progression.
The provided paper does not specifically mention neuroblastoma or its role in tumor microenvironment signaling and therapeutics.
The paper discusses the use of CIBERSORT and ESTIMATE algorithms to analyze immune cell infiltration in the neuroblastoma tumor microenvironment, and how differences in immune cell subtypes are related to individual heterogeneity and treatment efficacy. However, it does not specifically mention current advancements in understanding the role of the tumor microenvironment in neuroblastoma development and progression.
The paper discusses the role of tumor-associated macrophages (TAMs) in neuroblastoma development and immune evasion, as well as strategies to target and reprogram TAMs to enhance natural killer (NK) cell functions.

Related Questions

What are the current survival rates for childhood neuroblastoma in the UK?5 answersThe current survival rates for childhood neuroblastoma in the UK vary based on risk factors and treatment modalities. Studies have shown that high-risk neuroblastoma patients diagnosed after 2000 had a median post-relapse overall survival (PROS) of 8.4 months, with a median post-relapse progression-free survival (PRPFS) of 4.7 months. Additionally, unresectable non-MYC amplified intermediate-risk patients had a median PROS of 11.8 months, with a 5-year survival rate of 24%. Furthermore, the long-term survival of neuroblastoma patients has been influenced by treatment choices, with surgery improving overall survival, while chemotherapy and radiotherapy may have unfavorable impacts on patient outcomes. These findings underscore the importance of tailored treatment strategies and close monitoring to improve survival rates for childhood neuroblastoma patients in the UK.
How has anti-GD2 immunotherapy contributed to advances in treatment for neuroblastoma?4 answersAnti-GD2 immunotherapy has significantly advanced neuroblastoma treatment by offering promising therapeutic strategies. Research has shown that anti-GD2 antibodies, such as IgA3.0 ch14.18 and anti-GD2-MSCs, exhibit potent antitumor effects through mechanisms like antibody-dependent cellular cytotoxicity (ADCC) and antibody secretion. Additionally, combining anti-GD2 antibodies with immune checkpoint blockade therapies like TIGIT and PD-L1 inhibitors has demonstrated enhanced tumor growth inhibition and improved survival rates in high-risk neuroblastoma models. Clinical studies have further highlighted the efficacy and safety of anti-GD2 immunotherapy, like dinutuximab beta, in pediatric patients, leading to increased event-free survival rates and manageable toxicity profiles, allowing for outpatient management. These findings underscore the significant contributions of anti-GD2 immunotherapy in revolutionizing neuroblastoma treatment approaches.
What components does the microenvironment of glioblastoma include?5 answersThe microenvironment of glioblastoma (GBM) includes a complex network of cellular and non-cellular components that play crucial roles in tumor progression and therapy resistance. Cellular components consist of resident brain cells, immune cells, marrow-derived immune cells, lymphocytes, macrophages, and cancer-associated fibroblasts. Non-cellular components involve the extracellular matrix, which undergoes alterations in GBM, leading to upregulated collagen gene expression and aligned fibrillary ECM networks. Additionally, the microenvironment features chemical factors like cytokines, growth factors, and neurotransmitter-based interactions that influence GBM cell behavior and treatment response. Understanding the diverse components of the GBM microenvironment is crucial for developing effective therapeutic strategies to target these interactions and improve patient outcomes.
What is the immune microenvironment of neuroblastoma?4 answersThe immune microenvironment of neuroblastoma is characterized by a complex composition of immune cells, including T cells, macrophages, myeloid-derived suppressor cells (MDSCs), and cancer-associated fibroblasts (CAFs). Tumor-infiltrating T cells in neuroblastoma exhibit features of exhaustion, with increased expression of inhibitory receptors and low expression of effector cytokines. MDSCs isolated from neuroblastoma tumors have immunosuppressive properties, impairing the proliferation of T lymphocytes. The myeloid cell population in the neuroblastoma microenvironment includes polymorphonuclear leukocytes (PMNs), monocytes, and macrophages, which are present in both the stroma and tumor parenchyma. The immune microenvironment of neuroblastoma is influenced by molecular subtypes, such as MYCN amplification, and can change dynamically with chemotherapy. These findings provide insights into the immunocompromised nature of the neuroblastoma microenvironment and suggest potential therapeutic strategies targeting both tumor cells and components of the immune microenvironment.
How do tumor cells and the tumor microenvironment interact?3 answersTumor cells interact with the tumor microenvironment (TME) through various mechanisms. The TME consists of stromal cells, immune cells, and extracellular matrix components that influence tumor biology and response to therapy. Co-culture models have been developed to study these interactions in detail. Tumors adapt to unfavorable environments by communicating with neighboring cells, such as stromal cells, immune cells, and fibroblasts, through secreted molecules and cellular plasticity. The TME plays a crucial role in tumor progression and repression, with immune cells and fibroblasts having both pro- and anti-tumorigenic functions. The tumor and its microenvironment communicate through membrane interactions, soluble factors, and transporter interactions, which activate signaling pathways involved in tumor promotion and progression. In liver cancer, tumor-initiating cells interact with the TME, which is immunosuppressive and tumor-promoting, contributing to tumor heterogeneity, metastasis, and chemoresistance.
How can 3D modeling be used to improve neuroblastoma surgery?4 answersThree-dimensional (3D) modeling can be used to improve neuroblastoma surgery by providing preoperative assessment and surgical planning. High-quality preoperative images are used to create 3D reconstructions, which help visualize and precisely locate vessels within tumors and preserve parenchyma during surgery. These 3D models allow surgeons to practice on the model before the actual procedure, raising questions regarding surgical approach, feasibility, and potential complications in advance. By using 3D prototypes of tumors encasing major vessels, surgeons can refine their surgical management and achieve greater and safer resections, which is prognostic in a significant percentage of tumor types. Overall, 3D modeling provides a valuable tool for surgical planning and improving outcomes in neuroblastoma surgery.