scispace - formally typeset
Search or ask a question

What is the potential safety risk of PD-L1/CD3ε Bispecific T-cell engager (BiTE) ? 


Best insight from top research papers

PD-L1/CD3ε Bispecific T-cell engagers (BiTEs) have shown potential safety risks. BiTE therapy has been associated with adverse events (AEs) such as cytokine release syndrome, neurotoxicity, and febrile neutropenia . In addition, the immunosuppression via the PD-1/PD-L1 axis has been a challenge for BiTEs in solid tumors . However, a study on a Protease-Activated PSTAGylated BiTE showed promising results in solid tumor immunotherapy . Another study on a bispecific antibody targeting TIM-3 and PD-L1 reported unexpected immunogenicity targeting both arms of the bispecific antibody, leading to early study termination . These findings highlight the importance of thorough analyses for preexisting antibodies and immunogenicity risk assessment in the development of BiTE molecules . Overall, while BiTEs have shown potential in cancer immunotherapy, their safety profiles and immunosuppressive effects need to be carefully evaluated and monitored in clinical trials .

Answers from top 5 papers

More filters
Papers (5)Insight
The potential safety risk of PD-L1/CD3ε Bispecific T-cell engager (BiTE) is not mentioned in the provided paper.
The potential safety risk of PD-L1/CD3ε Bispecific T-cell engager (BiTE) is not mentioned in the provided paper.
The provided paper is about the safety and efficacy of LY3415244, a TIM-3/PD-L1 bispecific antibody, in patients with advanced solid tumors. It does not provide information about the safety risk of PD-L1/CD3ε Bispecific T-cell engager (BiTE).
The paper does not mention any potential safety risks associated with PD-L1/CD3ε Bispecific T-cell engager (BiTE).
The paper does not mention the safety risk of PD-L1/CD3ε BiTE specifically. The paper focuses on the safety profile of BiTE therapy in general, including adverse effects such as cytokine release syndrome, neurotoxicity, and febrile neutropenia.

Related Questions

What is the difference between cell engagers and bispecific antibodies?5 answersBispecific antibodies and cell engagers are both innovative immunotherapies with distinct mechanisms of action. Bispecific antibodies, as demonstrated in Context_1 and Context_4, are designed to simultaneously bind to two different targets, such as tumor antigens and immune cells, activating effector mechanisms like complement-dependent cytotoxicity or T cell-mediated killing. On the other hand, cell engagers, exemplified in Context_2 and Context_3, specifically recruit immune cells like T cells or NK cells to target tumor cells, inducing cell activation and tumor cell lysis. While bispecific antibodies can activate complement systems and have versatile applications, cell engagers, particularly NK-cell engagers, are highlighted for their safer profile and effectiveness in reducing proinflammatory cytokine production. Overall, the key distinction lies in the direct targeting of immune cells by cell engagers versus the dual binding capacity of bispecific antibodies to engage both immune cells and tumor cells simultaneously.
How does PD-L1 contribute to prognosis in hematological malignancies?5 answersPD-L1 expression plays a crucial role in predicting prognosis in various hematological malignancies. In multiple myeloma (MM), high PD-L1 expression is linked to poor overall survival (OS) and progression-free survival, especially in patients not undergoing autologous stem cell transplantation (ASCT). In acute myeloid leukemia (AML), high levels of the PD-L1 transcription variant v1/v2 are associated with worse OS, particularly in AML subgroups with specific genetic mutations. Additionally, PD-L1 expression in extranodal natural killer/T-cell lymphoma (ENKTL) serves as an independent predictor of poor prognosis, with lower expression levels correlating with a higher risk of relapse and refractoriness. Overall, PD-L1 expression in hematological malignancies like diffuse large B-cell lymphoma (DLBCL) is indicative of poor OS and adverse clinicopathological features, emphasizing its significance as a prognostic biomarker.
Is death receptor signaling involved in the drug efficacy of CD3 bispecific antibodies?5 answersDeath receptor signaling is not directly mentioned in the provided contexts regarding the drug efficacy of CD3 bispecific antibodies. However, the contexts highlight crucial factors influencing the efficacy of CD3 bispecific antibodies. Studies reveal that CD8+ T cells rely on CD4+ T cell-derived signaling factors for sustained killing in vitro. Additionally, resistance mechanisms to CD3-bispecific antibodies involve IFNγ signaling and core fucosylation pathways, impacting the therapeutic response. These findings emphasize the intricate interplay of various signaling pathways and molecular mechanisms in modulating the effectiveness of CD3 bispecific antibodies, shedding light on potential targets for enhancing their clinical potential.
How CD3 cause T cell exhaution through PD-L1 expression and PD-1 checkpoint pathway in cancer?5 answersCD8 positive lymphocytes expressing CD3 can lead to T cell exhaustion through the PD-L1 expression and PD-1 checkpoint pathway in cancer. PD-L1, synthesized in the endoplasmic reticulum, interacts with PD-1 on T cells, delivering coinhibitory signals that suppress T cell function, aiding in immune evasion by tumor cells. PD-L1 expression is found in various human tumor types and correlates with the presence of intratumoral CD8 positive lymphocytes, suggesting that the antitumor immune response may induce tumoral PD-L1 expression. In fibroblastic tumors, the expression of PD-L1 and PD-1 is limited, but TIM3 is widely expressed, with high TIM3 levels correlating with TIL density, indicating a potential role in immunosuppression. Therefore, targeting PD-1/PD-L1 and TIM3 in fibroblastic tumors with high expression levels may benefit patients through combination therapy.
What are the potential drawbacks of using PD-L1 in the treatment of melanoma?5 answersPD-L1 expression in melanoma has been used as a potential biomarker for predicting response to immune checkpoint blockade (ICB) therapy. However, there are potential drawbacks associated with using PD-L1 in the treatment of melanoma. One drawback is that PD-L1 expression is not consistently correlated with clinical outcomes in melanoma patients treated with ICB therapy. Another drawback is the heterogeneity of PD-L1 expression within tumors, which can lead to discordant results and challenges in accurately assessing PD-L1 status. Additionally, the use of different PD-L1 antibodies in immunohistochemistry assays can result in variations in staining properties, further complicating the interpretation of PD-L1 expression. These drawbacks highlight the need for further research and refinement in the use of PD-L1 as a biomarker in melanoma treatment.
Is TCR-T cell therapy generally safe?3 answersTCR-T cell therapy has shown promise as a treatment for cancer patients, but safety concerns remain. While some TCRs possess cross-reactivity to unrelated MHC molecules, TCR chain centricity can be exploited to enhance desired antitumor TCR reactivity and eliminate unwanted TCR cross-reactivity. Personalized TCR-T cell therapy, using TCRs from naturally occurring tumor antigen-specific T cells, has shown therapeutic effects on autologous patient-derived xenograft tumors. TCR-engineered cellular therapy targeting tumor-specific neoantigens has demonstrated significant antitumor effects in preclinical models. Further fine-tuning is necessary to improve functionality and safety for the treatment of solid tumors. The AFNT-111 cell therapy, targeting the KRAS G12V mutation, aims to limit tumor escape and promote increased T cell activity and persistence. While these studies show promising results, careful monitoring for possible harmful toxicities caused by adoptive transfer of T cells redirected by TCRs is still mandatory.

See what other people are reading

What are the primary target cells and organs of Perfringolysin O?
5 answers
Perfringolysin O (PFO), a cholesterol-dependent pore-forming toxin secreted by Clostridium perfringens, primarily targets human epithelial and endothelial cells, inducing profound cytoskeletal reorganization and plasma membrane damage. PFO binds to cholesterol-rich membranes, forming large pores that lead to cell lysis. The cholesterol distribution in the plasma membrane affects PFO binding, with a threshold of 35 mol% of total lipids required for PFO accessibility. PFO plays a crucial role in the progression of gas gangrene and necrohemorrhagic enteritis in humans and animals, acting synergistically with other toxins. The immunogenicity and protective potential of PFO have been explored, with the non-toxic C-terminal domain showing promise as a vaccine candidate.
How does immunohistochemistry aid in the diagnosis of Alzheimer's disease and Parkinson's disease?
5 answers
Immunohistochemistry plays a crucial role in the diagnosis of Alzheimer's disease (AD) and Parkinson's disease (PD) by enabling the identification of specific markers associated with these neurodegenerative conditions. In the case of AD, in silico immunohistochemistry (IHC) can computationally identify key features like neurofibrillary tangles and β-amyloid plaques with high accuracy. Similarly, multiplexed fluorescence-based immunohistochemistry (MP-IHC) allows for the simultaneous screening of numerous markers in brain tissue sections, aiding in the characterization of AD and PD pathology. Additionally, immunohistochemical analysis of skin biopsies has shown promise in detecting alpha-synuclein aggregates, a potential biomarker for PD, providing insights for early disease diagnosis. These techniques enhance the understanding and diagnosis of AD and PD through the precise visualization of pathological hallmarks.
What is the limit of detection for a COVID-19 paper lateral flow test?
4 answers
The limit of detection for COVID-19 lateral flow tests varies across different studies. One study developed a rapid lateral flow screening tool with a limit of detection of 4 ng/mL of antibodies against the SARS-CoV-2 spike protein. Another study achieved a detection limit of 5 × 10^4 copies/mL for cultured SARS-CoV-2 using a colorimetric lateral flow immunoassay. Additionally, an open-access lateral flow assay demonstrated a limit of detection of 4 TCID50/swab of gamma-irradiated SARS-CoV-2 virus, meeting the World Health Organization's acceptable analytical sensitivity. These variations in the limit of detection highlight the diverse approaches and sensitivities of lateral flow tests for COVID-19 detection.
How do exosomes contribute to the epigenetic changes that occur during prostate cancer progression and metastasis?
5 answers
Exosomes play a crucial role in the epigenetic changes observed during prostate cancer progression and metastasis. They act as messengers carrying various biomolecules, including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), which regulate gene expression. Exosomes derived from prostate cancer cells can promote androgen resistance in other cells, affecting proliferation and invasion abilities. Additionally, exosomes modulate cancer cell metastasis by regulating cell motility, extracellular matrix remodeling, and interactions with nerves through miRNA cargo influenced by the mechanical forces of the tumor microenvironment. Furthermore, exosomes are implicated in regulating tumor cell metastasis by modulating signaling pathways associated with epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs). These findings highlight the significant impact of exosomes on epigenetic changes driving prostate cancer progression and metastasis.
How do tumor derived exosomes contribute to the epigenetic changes that occur during prostate cancer progression and metastasis?
5 answers
Tumor-derived exosomes play a crucial role in prostate cancer progression and metastasis by contributing to epigenetic changes. These exosomes carry genetic and molecular cargo, including microRNAs (miRNAs), which can modulate various processes in the tumor microenvironment. Exosome-mediated transfer of miRNAs can regulate cancer cell behavior, such as viability, proliferation, and migration. Additionally, exosomes can influence androgen resistance in prostate cancer cells through specific miRNA interactions. Furthermore, exosomes can transmit mechanical forces from the extracellular matrix, affecting miRNA cargo and promoting cancer cell metastasis. Overall, tumor-derived exosomes act as messengers that facilitate epigenetic regulation, impacting the progression and metastatic potential of prostate cancer cells.
What are the recent aptamers used for diagnosis of small cell lung cancer discovered in 2022?
5 answers
Recent aptamers used for the diagnosis of small cell lung cancer (SCLC) discovered in 2022 include the apHAT610 aptamer, which targets Histone acetyltransferase 1 (HAT1) and has shown high specificity and affinity for HAT1, inhibiting its acetyltransferase activity in vitro. Another aptamer, TEPAP, designed for SCLC detection, utilizes an extension polymerization approach and has demonstrated high sensitivity and specificity in recognizing SCLC cells, with potential application in clinical samples. Aptamers, such as those targeting cancer biomarkers, offer promising diagnostic applications in cancer, including SCLC, due to their high specificity, binding affinity, and low immunogenicity, making them valuable tools for early detection and accurate diagnosis of the disease. These aptamers represent innovative approaches in the field of SCLC diagnosis, showing potential for improving patient outcomes through early and precise detection.
Assessing the Effectiveness of Canine Vaccination Protocols in Preventing Blood Parasites infections?
5 answers
Assessing the effectiveness of canine vaccination protocols in preventing blood parasite infections is crucial in combating diseases like visceral leishmaniasis. Various studies have highlighted the significance of vaccines in reducing the risk of active infection and symptomatic disease in dogs infected with Leishmania infantum. Additionally, therapeutic vaccines have shown promising results in restoring biochemical and hematological parameters, reducing clinical signs, and lowering parasite burden in symptomatic dogs naturally infected with L. infantum. Furthermore, prophylactic vaccines like the rA2 protein-saponin vaccine have demonstrated immunogenicity and a reduction in the cumulative incidence of infection in endemic areas, although further optimization may be needed for enhanced efficacy. These findings collectively emphasize the importance of vaccination strategies in preventing blood parasite infections in canines.
What is the role of glutamine in cancer metabolism?
4 answers
Glutamine plays a crucial role in cancer metabolism by serving as a carbon source for lipid and metabolite synthesis, a nitrogen source for amino acids and nucleotides, and maintaining redox homeostasis. It is essential for supporting cell growth, activating signal transduction, and regulating redox balance in cancer cells, particularly in non-small cell lung cancer (NSCLC) and prostate cancer (PCa). Glutamine metabolism also influences cancer stem cell (CSC) fate by contributing to various metabolic pathways, including ATP production, amino acid biosynthesis, and epigenetic modifications within the CSC population. Furthermore, glutamine metabolic reprogramming in tumors can activate multiple signaling pathways, promote oncogene expression, regulate tumor immunity, and contribute to drug resistance, highlighting its significance in cancer development and progression.
What factors contribute to a poor immune response after vaccination?
5 answers
Various factors contribute to a poor immune response after vaccination. Metabolic dysregulation, such as in malnourished, obese, and elderly individuals, can lead to suboptimal immune responses, impacting vaccine efficacy. Gender, body mass index (BMI), and vaccine type play significant roles in post-vaccination weak immune responses, with males, individuals with higher BMI, and those vaccinated with Sinopharm being more vulnerable. In myeloma patients, predictors of poor immune responses include male gender, specific myeloma therapies, and receiving the Pfizer-BioNTech vaccine. Comorbidities like cardiovascular diseases, chronic obstructive pulmonary disease, end-stage renal disease, depression, dementia, and malnutrition can also impair vaccine immunogenicity and immune function in older individuals. Understanding these diverse factors is crucial for improving vaccination outcomes in vulnerable populations.
Does vaccination with 23-valent pneumococcal polysaccharide vaccine (PPSV23) increase the risk of cardiovascular, neurological, and immunological adverse events?
5 answers
Vaccination with the 23-valent pneumococcal polysaccharide vaccine (PPSV23) does not significantly increase the risk of cardiovascular, neurological, or immunological adverse events. Studies on patients with Chronic Lymphocytic Leukemia (CLL) and systemic lupus erythematosus (SLE) show that adverse events post-PPSV23 are generally well-tolerated, with low rates of serious events. Additionally, a study on children living with HIV found that a sequential administration of a 15-valent pneumococcal conjugate vaccine followed by PPSV23 was well-tolerated and induced immune responses without significant adverse events. Real-life data from Puglia, Italy, also supports the safety profile of PPSV23, with adverse events occurring in less than 0.1‰ of patients, mostly being mild and self-limiting.
How important is longevity of protective immunity in livestock vaccine?
5 answers
The longevity of protective immunity in livestock vaccines is crucial for designing effective control and eradication programs. Studies on vaccines for diseases like lumpy skin disease (LSD) and bovine herpesvirus-1 (BHV-1) highlight the significance of sustained immunity. Research on poultry vaccinations against avian respiratory diseases emphasizes the need for long-lasting immunity to protect against multiple pathogens. Understanding the duration of vaccine-induced protection is essential, especially in the context of diseases like COVID-19, where successful vaccination is pivotal for societal health. Efforts to enhance vaccine longevity focus on mechanisms like long-lived plasma cells and memory B cells, as well as strategies to improve immunogenicity and prime-boost schemes. Overall, ensuring durable immunity through vaccines is critical for effective disease control in livestock populations.