scispace - formally typeset
Search or ask a question

Does metformin marker related to CAR-T therapy? 


Best insight from top research papers

The marker related to CAR-T therapy is not directly associated with metformin in the provided contexts. However, the contexts discuss the use of metformin in modulating the biological functions of CAR-T cells. Specifically, metformin has been shown to inhibit CD19-CAR T cell proliferation and cytotoxicity, induce apoptosis, and activate AMPK while suppressing mTOR and HIF1α expression . Additionally, metformin has been found to promote the differentiation of memory CD8+ T cells, enhance their antiapoptotic abilities, and reprogram their cytotoxic functions, potentially benefiting cancer therapy . Although the contexts do not mention a direct link between metformin and a marker for CAR-T therapy, they provide valuable insights into how metformin affects the functions of CAR-T cells in cancer treatment.

Answers from top 5 papers

More filters
Papers (5)Insight
Not addressed in the paper.
Not addressed in the paper.
Not addressed in the paper.
Metformin affects CD19-CAR T cells via the AMPK pathway, inhibiting proliferation, cytotoxicity, and inducing apoptosis, suggesting a potential role in CAR-T therapy.
Metformin enhances CAR-T therapy by promoting memory CD8+ T cell differentiation via the AMPK-miR-107-Eomes-PD-1 pathway, leading to increased cytotoxicity and reduced tumor growth.

Related Questions

What are the latest clinical trials with CAR-T therapy?5 answersThe latest clinical trials with CAR-T therapy encompass a wide range of hematological malignancies. These trials focus on enhancing treatment outcomes, addressing limitations like high relapse rates and adverse effects, and exploring novel targets for CAR-T cell therapy. Various phase I trial designs are being employed to determine the maximum tolerated dose and ensure safety. While CAR-T cells have shown remarkable efficacy in hematological malignancies, efforts are ongoing to overcome limitations in solid tumors, such as an immunosuppressive tumor microenvironment and insufficient tumor infiltration, by exploring other immune cells like CAR-NK and CAR-M cells. Despite the hurdles like limited efficacy in solid tumors and high costs, CAR-T therapy has revolutionized cancer treatment, offering promising therapeutic alternatives for patients with blood malignancies.
What are the current clinical trials evaluating the effectiveness of metformin in cancer treatment?4 answersMetformin is being evaluated in several clinical trials for its effectiveness in cancer treatment. One trial is investigating the use of metformin in combination with standard neo-adjuvant chemotherapy for improving pathological complete response rates in breast cancer patients. Another trial is exploring the potential of metformin in the treatment of hepatocellular carcinoma (HCC), the most common type of liver cancer. The study aims to determine the efficacy of metformin as an antineoplastic drug in HCC and its potential for enhancing treatment outcomes when combined with immune checkpoint inhibitors. Additionally, a trial is assessing the use of metformin in offsetting the adverse effects of adriamycin-cyclophosphamide plus paclitaxel (AC-T) chemotherapy in non-diabetic breast cancer patients. The study aims to determine whether metformin can reduce the incidence and severity of peripheral neuropathy, oral mucositis, fatigue, and cardiac dysfunction caused by AC-T chemotherapy. These clinical trials are investigating the potential benefits of metformin in different cancer types and treatment settings.
Metformin use in cancer?5 answersMetformin use in cancer has been studied in several papers. Wang et al. found that metformin use was associated with decreased all-cause mortality among esophageal cancer patients. Jo et al. analyzed a cohort of prostate cancer patients and found that the duration of metformin administration was inversely associated with prostate cancer risk. Mu et al. provided a comprehensive perspective of metformin as an anticancer drug, highlighting its potential therapeutic effects and modulation of tumor cell response to treatments. Another study by Wang et al. conducted a meta-analysis and found that metformin usage was associated with improved overall survival in cancer patients, particularly in those with diabetes. However, a cohort study by the American Association for Cancer Research found no evidence that metformin use affects overall lung cancer risk, although there was variation in the association by smoking history and histology. Overall, these studies suggest that metformin may have potential benefits in cancer treatment and prevention, particularly in certain patient populations.
What are the latest findings on the use of metformin?5 answersMetformin has been extensively studied for its potential in cancer prevention and treatment. Observational studies initially reported a reduction in cancer incidence among metformin users, but these findings were later found to be affected by biases such as immortal time bias. Meta-analyses of randomized controlled trials and cohort studies did not find a significant association between metformin use and skin cancer risk, although there was suggestive evidence of modestly reduced risks. In terms of cancer therapy, metformin has shown potential as an adjuvant in chemoradiotherapy, particularly for diabetic individuals. Metformin use has also been associated with a lower risk of immune-mediated skin diseases but does not have a preventive effect on androgen-mediated skin diseases. In the context of COVID-19, metformin has been found to reduce mortality among diabetic patients. However, further assessment of risks versus benefits is needed before implementing metformin at a larger scale for COVID-19 patients.
What is car-t therapy used for?5 answersCAR-T therapy is used for the treatment of hematologic malignancies, including acute lymphoblastic leukemia and large B-cell lymphomas. It involves the use of a patient's own immune cells that are engineered to target and kill cancer cells. CAR-T therapy has shown success in hematological malignancies due to the almost universal expression of the CD19 antigen. However, its effectiveness in solid tumors is limited due to challenges such as variable expression of target antigens, difficulty in penetrating solid tumors, and hostile microenvironment. Despite these challenges, there have been breakthroughs and potential solutions proposed to improve CAR-T therapy for cancer treatment. CAR-T therapy has generated significant excitement and is being explored as a potential treatment option for both hematologic and solid-organ malignancies.
How metformin can affect immune cells during colorectal cancer?2 answersMetformin has been shown to affect immune cells during colorectal cancer (CRC) by reprogramming the tumor immune microenvironment and enhancing the anti-tumor immune response. One study found that metformin disrupted the dense tumor stroma, promoting drug delivery and remodeling the tumor immune microenvironment. Another study demonstrated that metformin decreased the expression of programmed cell death ligand 1 (PD-L1), a key suppressor of the cytotoxic immune response, in CRC cells. Additionally, metformin was found to activate the AMP-activated protein kinase (AMPK) pathway and suppress the mevalonate (MVA) pathway, leading to a decrease in cancer stem cells (CSCs) in CRC. Furthermore, a high throughput functional screen identified microRNAs that sensitize CRC cells to metformin, suggesting that microRNAs can modulate the anti-proliferative effects of metformin on CRC cells. These findings highlight the potential of metformin as an immunomodulatory agent in CRC therapy.

See what other people are reading

What are the beneficial mechanisms of metformin?
4 answers
Metformin, a widely used antidiabetic drug, exerts its beneficial effects through various mechanisms. It primarily activates AMPK, reducing hepatic glucose output and regulating glucose and lipid metabolism in cardiomyocytes, thus lowering cardiovascular risks. Metformin also impacts redox balance, mitochondrial function, gut microbiome, and various signaling pathways like FBP1, PP2A, FGF21, SIRT1, and mTOR. These mechanisms contribute to metformin's efficacy in glycemic control, cardiovascular protection, cancer intervention, anti-inflammation, antiaging, and weight control. Additionally, metformin's AMPK activation corrects endothelial dysfunction, reduces oxidative stress, and improves inflammatory responses, leading to cardiovascular benefits in diabetic and non-diabetic patients. Overall, metformin's multifaceted mechanisms make it a valuable therapeutic agent for various conditions beyond just diabetes.
How does nutrition impact the immune system in obesity?
5 answers
Nutrition plays a crucial role in modulating the immune system in obesity. The Western diet and sedentary lifestyle contribute to the rising obesity rates, leading to chronic inflammation and altered tissue functions. Obesity-related inflammation is linked to cellular senescence, characterized by irreversible cell cycle arrest and the secretion of pro-inflammatory factors. Both undernutrition and overnutrition have significant impacts on immunity, with undernutrition causing immunosuppression and overnutrition leading to chronic inflammation, increasing the risk of metabolic and cardiovascular diseases. An anti-inflammatory diet can reduce markers of inflammation and improve immune status in individuals with obesity, highlighting the importance of dietary interventions in managing immune responses in obesity.
How does IL-4 contribute to the immune response during viral infections?
5 answers
IL-4 plays a multifaceted role in the immune response during viral infections. It can influence the activation and polarization of macrophages, leading to either anti-inflammatory or proinflammatory responses depending on the context. Additionally, IL-4 can regulate the frequency, function, and reactivity of CD8+ T cells, impacting their ability to respond to viral infections. Furthermore, IL-4 has been shown to decrease susceptibility to secondary bacterial infections post-influenza by suppressing immune cell death and bacterial burden. These findings highlight the diverse effects of IL-4 on different immune cells, suggesting its potential in modulating immune responses to enhance protection against viral infections and associated complications.
What are the specific roles of IL-4 in the immune response to viral infections?
5 answers
IL-4 plays crucial roles in the immune response to viral infections by exerting diverse functions. In the context of respiratory viruses like human metapneumovirus (hMPV) and Streptococcus pneumoniae co-infection post influenza, IL-4 has been found to promote viral replication and decrease susceptibility to secondary bacterial infections, respectively. Additionally, IL-4 can induce an alternative activation state in macrophages, leading to anti-inflammatory responses and tissue repair, but it can also mediate proinflammatory responses in conditions like asthma and allergic inflammation. Furthermore, IL-4 is essential for regulating antibody responses through TFH cells, controlling class-switch recombination to IgG1 and IgE, and contributing to germinal center formation during humoral immune responses. Overall, IL-4's multifaceted roles highlight its significance in modulating immune responses to viral infections.
What are the benefits of trial transcripts?
5 answers
Trial transcripts offer various advantages in different contexts. They can enhance precision oncology trials by integrating transcriptomics to predict patient responses to targeted therapies. Additionally, real-world data (RWD) integrated with transcriptomics can provide insights into disease pathways and clinical outcomes, aiding in personalized medicine. Moreover, trial transcripts can improve the accuracy of audio data recognition, facilitating the processing of trial information for multiple terminals. Overall, trial transcripts play a crucial role in advancing medical research, optimizing patient selection for treatments, and improving the efficiency of clinical trials through data integration and analysis.
Dose of Camptothecin for treatment of colorectal cancer
5 answers
The effective dose of a camptothecin derivative for the treatment of colorectal cancer is approximately 0.05 to 1 mg/kg body weight/day when dissolved in a non-aqueous solvent, as suggested in a study focusing on an oral pharmaceutical composition. Camptothecins, including irinotecan, are potent topoisomerase I inhibitors used in colorectal cancer treatment, but their high toxicity and administration challenges have been noted. Research has explored the upregulation of DNA damage response-related molecules and apoptosis proteins by a novel topoisomerase inhibitor, PCC0208037, showing promising anti-tumor efficacy in colorectal cancer cells and xenograft models, potentially offering a comparative advantage over standard treatments like irinotecan. Additionally, combining topoisomerase inhibitors like camptothecin with immune-checkpoint inhibitors has been suggested to enhance treatment outcomes in colon cancer by modulating immunoregulatory responses.
Is PDL1 expressed on B lymphoma cells?
4 answers
Yes, programmed cell death ligand 1 (PD-L1) is expressed on B lymphoma cells. Research has shown that PD-L1 is expressed in various aggressive B-cell lymphomas, including nodular sclerosis and mixed cellularity classical Hodgkin lymphoma, primary mediastinal large B-cell lymphoma, T-cell/histiocyte-rich B-cell lymphoma, EBV-positive and -negative posttransplant lymphoproliferative disorders, and diffuse large B-cell lymphoma (DLBCL) among others. Additionally, PD-L1 expression has been observed in anaplastic large cell lymphoma (ALCL) cell lines and a subset of DLBCL cases with activated B-cell features. Furthermore, a study on diffuse large B-cell lymphoma (DLBCL) patients found that PD-L1 expression could predict event-free survival, indicating a poor prognosis when PD-L1 is present on tumor cells.
Is there any FDA approved small molecule for cancer immunetherapy targeting PDL-1?
5 answers
Yes, there are FDA-approved small molecules for cancer immunotherapy targeting PD-L1. While several therapeutic monoclonal antibodies against the PD-1/PD-L1 immune checkpoint have been successful in cancer treatment, small molecule inhibitors have emerged as alternatives. These small molecules offer advantages such as improved tissue penetration, low immunogenicity, and lower manufacturing costs compared to monoclonal antibodies. Recent studies have focused on screening natural compounds to identify potent PD-L1 inhibitors, with some compounds showing promising results in computational investigations and molecular dynamics simulations. The development of small-molecule inhibitors targeting PD-L1 is seen as a complementary approach to monoclonal antibodies in cancer immunotherapy, although further validation of their safety and efficacy in clinical settings is still needed.
Does signalling through ADRA1A lead to mtor activation?
9 answers
The signaling through ADRA2A, not ADRA1A, has been specifically investigated in the context of cervical cancer, where it was found to have a significant impact on the PI3K/Akt/mTOR pathway. ADRA2A overexpression led to a notable decrease in the phosphorylation levels of PI3K, AKT, and mTOR, indicating that ADRA2A signaling can suppress the activation of the mTOR pathway in cervical cancer cells. This suggests that while the question pertains to ADRA1A, the available data on ADRA2A provides insight into how adrenergic receptor signaling can influence mTOR activity, albeit through a mechanism of suppression rather than activation. Further exploration into the regulation of mTOR signaling reveals various factors and pathways that can modulate its activity. For instance, AKAP13 has been identified as a crucial regulator of mTORC1 inhibition, acting through GPCR signaling to phosphorylate Raptor and inhibit mTORC1. Additionally, genetic mutations, such as those in ADAR1, have been shown to activate the mTOR/p70S6K/S6 ribosomal protein signaling axis, contributing to the development and progression of gastric cancer. Resistance exercise has also been shown to activate mTOR signaling, indicating the complexity and multifactorial nature of mTOR regulation. Moreover, the involvement of mTOR signaling in various diseases and conditions, including autism spectrum disorder (ASD), cancer, and fibrodysplasia ossificans progressiva (FOP), underscores its critical role in cellular processes and disease pathogenesis. The activation of mTORC1 by amino acids and its regulation by cellular nutrient and energy status further highlight the intricate control mechanisms governing mTOR activity. In summary, while the specific question regarding ADRA1A was not directly addressed in the provided contexts, the information on ADRA2A and other regulatory mechanisms of mTOR signaling offers valuable insights into how adrenergic signaling might interact with the mTOR pathway, suggesting a broader landscape of regulation involving various receptors and signaling molecules.
How do t cells recognise foreign tissue?
4 answers
T cells recognize foreign tissue through the interaction of their T cell receptors (TCRs) with peptides presented on major histocompatibility complex (MHC) molecules on the surface of target cells. This recognition process involves the systematic scanning of cell surfaces by T cells, which express numerous antigen receptors, enabling them to detect even minute quantities of the appropriate antigenic peptide bound to MHC molecules. The ability of T cells to distinguish between foreign antigens and self-antigens is crucial for immune responses. Studies have shown that T cells can reliably differentiate between foreign and self-antigens based on rare stochastic events, allowing for effective immune responses against pathogens or malignancies while avoiding autoreactivity. Additionally, the recognition of foreign MHC antigens by T cells can occur through direct and indirect pathways, with xenogeneic antigens potentially inducing more robust immune responses compared to allogeneic antigens.
How the immune system works?
5 answers
The immune system functions by distinguishing between self and non-self entities, aiming to eliminate the latter. It comprises the innate immune system, offering a rapid response against common pathogens, and the adaptive immune system, evolving to combat new invaders. Specialized cells present antigens to T cells, activating them for defense. T cells orchestrate immune responses, while B cells produce antibodies for protection. The immune system maintains self-tolerance and memory for efficient defense. It can be modulated by agents like azathioprine and monoclonal antibodies, enhancing responses or treating diseases. Understanding the immune system is crucial for pharmacists due to the increasing use of biotechnological products in healthcare.