scispace - formally typeset
Search or ask a question

What is popular immunocytokine? 


Best insight from top research papers

A popular immunocytokine involves the fusion of antibodies with cytokines to enhance targeted cancer therapy . These immunocytokines aim to improve the therapeutic index of cytokines by utilizing antibodies as vehicles for delivering immunomodulatory agents specifically within the tumor microenvironment. Various molecular formats and cytokine payloads have been explored to develop effective immunocytokines, with the goal of increasing efficacy while minimizing off-target toxicities associated with systemic cytokine administration . For instance, a notable immunocytokine combines IL-15 and IL-15Ra fused to an antibody, effectively targeting the tumor microenvironment and activating immune cells to achieve specific tumor killing while preventing systemic immunotoxicity. Combination treatments involving immunocytokines with conventional chemotherapy or immune-stimulatory agents have shown promise in enhancing therapeutic outcomes in cancer models.

Answers from top 5 papers

More filters
Papers (5)Insight
The immunocytokine L19-TNF, a fusion of L19 antibody and tumor necrosis factor, shows promise in eradicating sarcomas when combined with chemotherapy agents or immune check-point inhibitors.
The popular immunocytokine discussed in the paper is an antibody fusing IL-15 and IL-15Ra, targeting tumor microenvironments to activate immune cells for specific tumor killing while preventing immunotoxicity.
Popular immunocytokines include those utilizing IL-2 for activating cytotoxic effector cells, and CmAb(IL-10)2 combining IL-10 with Cetuximab for enhanced tumor growth reduction, especially in combination with checkpoint inhibitors.
Immunocytokines, utilizing antibodies for targeted cytokine delivery in cancer therapy, are promising to enhance efficacy and reduce toxicity, offering hope for improved cancer immunotherapy.
Immunocytokines, utilizing antibodies for targeted cytokine delivery in the tumor microenvironment, are promising for cancer immunotherapy, aiming to enhance efficacy and reduce systemic toxicity compared to natural cytokines.

Related Questions

What are the different types of immunomodulation?3 answersImmunomodulation refers to the modification of the immune response using various agents to achieve a desired level of immune activity. Different types of immunomodulators include prebiotics, probiotics, vitamins, adjuvants, polysaccharides, and herbs. Synthetic, natural, and recombinant preparations are also used for immunomodulation. Immunomodulation can be achieved through the modulation of soluble mediators, immune cells, neurotransmitters, hormones, and other signaling molecules. Additionally, immunomodulation can be induced by external beam radiation therapy (EBRT) and targeted radionuclide therapy (TRT), both of which have been shown to promote an antitumor immune response. These different types of immunomodulation have been explored for their potential in treating autoimmune diseases, enhancing disease resistance, improving immune function, and preventing inflammatory diseases.
What are examples of immunotherapy?5 answersImmunotherapy examples include monoclonal antibodies, cancer vaccines, tumor-infecting viruses, cytokines, adjuvants, and autologous T cells carrying chimeric antigen receptors (CARs). Other examples of immunotherapy are interleukins, interferons, bacilli, and cancer vaccines. Cancer immunotherapy also includes immune checkpoint inhibitors, tumor infiltrating lymphocytes, TCR T cells, and CAR T cells. Additionally, immunotherapy can involve vaccines that mobilize the patient's own immune system to fight cancer, as well as passive immunotherapy that administers antibodies or T cells.
What are the most recent and relevant systematic review on immunology?4 answersImmunology research has seen recent progress in various areas. One systematic review focused on the immunology of osteochondral allograft (OCA) transplantation and highlighted the role of host immune responses in graft survival and outcomes. Another review explored the complex role of immune therapy in triple-negative breast cancer (TNBC) and discussed the importance of tumor-infiltrating lymphocytes (TILs) and tumor-associated macrophages (TAMs) in the TNBC microenvironment. Additionally, a review examined the historiography of immunology since 1999 and concluded that the field has become a bustling and pluripotent discipline. While these reviews provide valuable insights, there is no specific mention of a recent and relevant systematic review on immunology in the abstracts provided.
What is immunizaton?5 answersImmunization is a process of modifying the immune response by administering drugs or compounds to positively or negatively regulate the immune system. It is a therapeutic strategy used to combat cancer, autoimmune diseases, and infectious diseases by inducing, enhancing, or suppressing the immune system. Immunotherapy has revolutionized modern therapeutics by making medicine personalized, accurate, and precise. It relies on the host's immune system and can be categorized as activation or suppression immunotherapies. Immunotherapy is now the primary or adjunct treatment for many diseases and is also used for diagnostic purposes. Immunization information systems (IISs) are computerized databases that record immunization doses administered to individuals and can generate complete immunization histories. IISs have the potential to reduce missed vaccination opportunities, prevent over-vaccination, and support public health efforts by generating accurate jurisdiction-level reports.
What is natural immunotherapy?3 answers
Who gives immunotherapy?5 answers

See what other people are reading

How have tumor-on-a-chip platforms evolved in recent years to facilitate the study of cancer-immune system crosstalk?
5 answers
Tumor-on-a-chip platforms have evolved significantly in recent years to enhance the study of cancer-immune system crosstalk. These platforms utilize microfluidic technologies to create 3D cell co-cultures that mimic the tumor microenvironment, enabling researchers to investigate immune cell recruitment, tumor progression, and response to immunotherapies in a patient-specific manner. By incorporating various cell types, such as cancer cells, immune cells, and stromal cells, these platforms can replicate the complex interactions within the tumor ecosystem. Additionally, advanced imaging techniques and computational algorithms allow for real-time monitoring of cellular behaviors and responses to treatments. Overall, tumor-on-a-chip platforms offer a promising approach to advancing our understanding of cancer-immune system interactions and improving the efficacy of immunotherapies through personalized medicine applications.
How is bioavailability measured for subcutaneously injected monoclonal antibodies?
4 answers
Bioavailability of subcutaneously injected monoclonal antibodies (mAbs) can be measured using predictive models based on physicochemical and pharmacokinetic properties. These models utilize factors like human intravenous clearance (CL), isoelectric point (pI), and allometric scaling from non-human primate data to predict human subcutaneous bioavailability. Specifically, an inverse linear correlation exists between human subcutaneous bioavailability and factors like CL and pI, with mAbs showing a stronger correlation with CL and fusion proteins with pI. Multivariate regression models incorporating these parameters have shown high predictive accuracy, with up to 82% of predictions falling within 0.8-to 1.2-fold deviations from observed bioavailability. Additionally, an integrated physiologically based modeling framework has been developed to predict bioavailability based on antibody-specific characteristics and mechanisms governing subcutaneous absorption and disposition.
What are the Clinical and preclinical states of CAR T cell therapy in vivo for solid tumors cancer?
5 answers
Clinical and preclinical states of CAR T-cell therapy for solid tumors cancer are evolving. Challenges like antigen heterogeneity and immunosuppressive tumor microenvironment hinder CAR T-cell efficacy in solid tumors. Strategies to enhance CAR T-cell function include incorporating co-stimulatory domains, developing armored CAR-T cells, and exploring other immune cells like CAR-NK and CAR-M cells. Novel CAR T-cell products like YTB323 show enhanced in vivo expansion and antitumor activity in preclinical models, with promising safety and efficacy profiles in clinical trials for r/r DLBCL. To overcome limitations such as restricted trafficking, hypoxic TME, antigen escape, and exhaustion, researchers are advancing CAR designs and combining therapies to optimize clinical efficacy in solid tumors.
What is the mechanism of reverse cholesterol transport pathway?
5 answers
The reverse cholesterol transport (RCT) pathway involves the transport of excess peripheral cholesterol to the liver for conversion into bile acids and subsequent excretion, reducing hyperlipidemia and cardiovascular disease risks. Key proteins involved in RCT include ATP-binding cassette sub-family A1 (ABCA1), scavenger receptor class B type I (SR-BI), apolipoprotein A-I (ApoA-I), and apolipoprotein A-II (ApoA-II). Additionally, liver X receptor (LXR) and its downstream regulators ABCA1 and ATP-binding cassette subfamily G member 1 (ABCG1) genes play a crucial role in regulating cholesterol metabolism and the RCT pathway. Furthermore, the ATP-binding cassette transporter A1 (ABCA1) is central in mediating the biosynthesis of high-density lipoprotein (HDL), facilitating cholesterol efflux from macrophages and preventing lipid deposition, thus contributing to RCT.
What are the potential mechanisms underlying the association between ICIP and TMB, and how might these influence treatment decisions?
5 answers
The association between immune checkpoint inhibitor therapy (ICI) and tumor mutational burden (TMB) involves several key mechanisms. High TMB has been linked to improved survival with ICI treatment, but not all high-TMB patients respond well. Specific gene alterations impact outcomes differently in TMB-high and TMB-low patients, influencing response to ICI. For instance, mutations in genes like STK11 and E2F3 are associated with worse outcomes in TMB-high tumors, while mutations in genes like NTRK and PTPRD are linked to better outcomes. Additionally, the immune-excluded phenotype, characterized by high stromal TIL and low intratumoral TIL density, is correlated with high TGFBs and poor response to immunotherapy. These insights can guide treatment decisions by helping identify patients likely to benefit from ICI based on their TMB status and specific gene alterations.
What is the shifting-mosaic steady-state equilibrium?
5 answers
The shifting-mosaic steady state (SMSS) equilibrium refers to a dynamic landscape pattern where patches within an area undergo continuous change while the total area of each landscape component remains relatively constant over time. This concept challenges the traditional view of landscape patterns as spatially stationary, highlighting the dynamic nature of ecosystems. Similarly, in the context of tumor-immune interactions, an equilibrium is established between effector and regulatory T cells within the tumor microenvironment, impacting the outcome of immune control over tumors. Furthermore, in tree population dynamics, factors like growth-dependent thinning, changing mortality risks, and episodic recruitment contribute to the dynamic equilibrium within forest ecosystems. Understanding such dynamic equilibria is crucial for environmental conservation, immune response modulation in cancer therapy, and managing chronic virus infections.
WHAT IS sex differences in cancer incidence IN SHIZOPHRENIA SPECTRUM DISORDER?
10 answers
The exploration of sex differences in cancer incidence, particularly within the context of schizophrenia spectrum disorder, necessitates a nuanced understanding of the broader landscape of sex disparities in cancer rates and the biological, environmental, and social determinants that may influence these patterns. While the provided contexts do not directly address schizophrenia spectrum disorder, they offer valuable insights into the mechanisms and patterns of sex differences in cancer incidence that could have implications for this specific population. Research has consistently shown that cancer incidence is statistically significantly higher in men than in women for a majority of non-reproductive cancer sites, with disparities exceeding 2-fold for several cancers. This male predominance in cancer incidence is echoed across various studies, indicating that men have a 20% higher chance of developing cancer over their lifetime compared to women. These differences are partly attributed to biological factors, including sex-specific genetic, epigenetic, and hormonal influences that affect cell cycle regulation, immunity, and metabolism. For instance, sex hormones and genetic differences play a role in modulating cancer risk and progression in a sex-specific manner. Environmental and lifestyle factors, alongside social determinants, also significantly contribute to sex disparities in cancer incidence. Studies have highlighted the importance of considering gender as a critical factor in understanding cancer development, pointing to the predominant environmental origin of cancer and its relationship with social determinants. Furthermore, the variability in the male-to-female incidence ratio across different regions suggests the influence of both biological factors and carcinogenic exposure differences. Given the complex interplay of genetic, hormonal, environmental, and social factors in determining cancer risk and incidence, individuals with schizophrenia spectrum disorder may exhibit unique patterns of sex differences in cancer incidence. However, specific research into cancer incidence within this population, taking into account these multifaceted determinants, is necessary to draw definitive conclusions.
How does the high mutation rate associated with POLE mutations contribute to the resistance to standard therapeutic approaches?
5 answers
The high mutation rate linked to POLE mutations contributes to resistance to standard therapies through various mechanisms. POLE mutations, particularly in the exonuclease domain, lead to a significantly increased tumor mutation burden (TMB). These mutations can result in a hypermutated phenotype, impacting immunological markers like TMB and microsatellite instability (MSI). Additionally, POLE mutant tumors may exhibit decreased fidelity of DNA replication, potentially affecting response to treatments. Notably, the presence of POLE mutations, even in microsatellite stable (MSS) colorectal tumors, can still make them effective targets for immunotherapy. Overall, the unique genomic characteristics of POLE mutant tumors, including high TMB and specific co-alterations, suggest a potential role in therapy resistance and the need for further investigation into tailored treatment approaches.
Do PD-L1 inhibitors slow TNBC xenograft growth in nude mice?
5 answers
PD-L1 inhibitors have shown efficacy in slowing TNBC xenograft growth in nude mice. Studies using a murine breast cancer model demonstrated that anti-PD-L1 treatment significantly suppressed primary tumor growth and extended survival. Additionally, in a study involving a syngeneic mouse tumor model, Dato-DXd, an ADC targeting TROP2, enhanced antitumor activity when combined with PD-1/PD-L1 inhibitors, suggesting a potential synergy in inhibiting tumor growth. Furthermore, PD-1 blockade in nude mice resulted in decreased tumor growth, indicating the importance of PD-1 signaling in promoting tumor progression. These findings collectively support the notion that PD-L1 inhibitors can effectively slow TNBC xenograft growth in nude mice, highlighting their potential as a therapeutic strategy for TNBC.
What are downsides of ethanol treatment on cancer cells?
5 answers
Ethanol treatment poses several downsides on cancer cells. Firstly, ethanol induces oxidative stress in cancer-associated fibroblasts, promoting myofibroblastic differentiation and autophagy in the tumor microenvironment. This oxidative stress leads to ketone production in fibroblasts and ketone re-utilization in epithelial cancer cells, fueling tumor growth via mitochondrial metabolism. Additionally, heavy alcohol consumption increases cancer stem cell (CSC) populations in various cancers, potentially enhancing tumor promotion and aggressiveness. Ethanol's metabolism generates free radicals that interfere with essential cellular functions, affecting growth and differentiation processes, including disturbances in CSCs. Furthermore, ethanol treatment alters protein expression in fibroblast cells, resembling patterns seen in nasopharyngeal carcinoma-associated stromal fibroblasts, hinting at potential cancer-related pathways being targeted by ethanol.
How does TNBC spread?
5 answers
TNBC, or Triple Negative Breast Cancer, spreads through a complex process involving various factors. Studies have shown that TNBC cells can induce IL-8 secretion from stromal cells, promoting cancer cell proliferation, migration, and metastasis. Additionally, TNBC's aggressive nature is fueled by enhanced cell motility, with specific genes like PRPF4B, BUD31, and BPTF playing crucial roles in driving TNBC cell migration and metastasis. Furthermore, the expression of Nodal protein in TNBC cells has been linked to promoting cancer cell survival and resistance to genotoxic insults, highlighting Nodal as a potential therapeutic target to inhibit TNBC spread. Understanding these mechanisms of TNBC spread involving cytokine signaling, gene regulation, and protein expression is vital for developing targeted interventions to combat the metastatic progression of this aggressive breast cancer subtype.