Discover CAR T Therapy from Creative Biolabs
CAR T Therapy from Creative Biolabs

CAR T Therapy from Creative Biolabs
Author: Bella Smith
Subscribed: 10Played: 238Subscribe
Share
Description
As a global company, Creative Biolabs has more than 200 talented and well-trained scientists located in different continents working closely with partners from the entire world to develop and produces medicines of tomorrow. Specifically, we are the established leading expert in TCR and CAR T&NK cell immune therapy development, as we offer the one-stop custom services that cover the entire new drug development pipeline. Additionally, we also offer an exclusive line of ready-to-use TCR and CAR T&NK cell construction products, such as virus packaging, purification, expansion and titer determination kits. Furthermore, we have built up a unique unparalleled CAR construction and production platform for all four CAR generations.
35 Episodes
Reverse
The target tumor cells are labeled with specific fluorescent cytoplasmic reagent, the live cells can be imaged and monitored at real-time level to measure the tumor cell lysis. The reagent is safe for cells at optimized concentration, and fluorescence measurements can be accurately performed for short-term experiments, as the reagent will be diluted with cell proliferation. In addition, the ratio of target tumor cells and CAR-T cells can be optimized in the assay.
Altering the specificity of T cell receptor (TCR) is one of the popular strategies to genetically modify T cells to enhance the tumor-killing activity of T cells. From a tumor-reactive T cell or active anti-tumor T-cell antigens, the appropriate target sequence is introduced to modify T cells to target a broad range of tumors with improved specificity. https://www.creative-biolabs.com/car-t/cellrapeutics-tcr-technology.htm
In patient's T Cell Isolation, flow cytometry analysis of isolated blood products ensures that a sufficient number of healthy and functional T cells being used for processing and re-infusion.
T cell infiltration, accumulation, and survival of chimeric antigen receptor T (CAR-T) cells in solid tumors are crucial for tumor clearance. T cell infiltration assays ensure the functional T cells are located in tumor microenvironment to display humoral or cellular immunity both in vitro and in vivo.
CAR-T cell preclinical in vivo animal testing performed in xenograft murine models derived by well-defined cell lines or primary human tumor cells. Immunodeficient mice, lacking of effective innate immunity for the absence of B cells, T cells, or NK cells, are the optimal animal models for engraftment of tumor cells and evaluation of CAR-T cells.
Electroporation transfer is emerged as a powerful tool for the genetic modification of diverse cell types based on the transient disruption of cell membrane via exposure to an electric field, which allows charged molecules to enter the cell. For instance, the square-wave pulse-based new electroporation devices, such as Lonza Nucleofector II electroporation system, manifests a high efficiency in the genetic modification of T cells with proprietary electroporation buffers and electric parameters.
Notably, several CAR-T libraries have been generated not only for blood cancer antigen CD19, but also for solid tumors against different antigens, such as Her2, Her3, EGFR, FGFR1, VEGFR, etc. The selected stable clones can be used in clinic trials immediately, thus making this technology more powerful and attractive in chimeric antigen receptor t cell therapy.
In a narrow sense of cancer biomarker, it is limited to proteins the most used to challenge in the clinical applications, especially in the CAR-T therapy. Specifically, a cancer biomarker of the CAR-T provides the most prominent signal of cancer cells for distinguishing from normal cells and the most effective CAR T target for immune recognition and destruction. https://www.creative-biolabs.com/car-t/biomarker-identification-selection.htm
The typical structure of a chimeric antigen receptor molecule includes a single chain variable fragment (scFv), a spacer, a transmembrane domain (TM) and an intracellular signaling domain. The scFv is derived from monoclonal antibody (mAb), which can specifically recognize the target protein on tumor surface and subsequently transduct activation signal into CAR-T cell.
Due to the linkage between the T cell activation and the expression and function of the CARs, we would obtain functional CARs with high affinities ranging from 10 pmol to nmol. Notably, we have generated several CAR-T libraries not only for blood cancer antigen CD19, but also for solid tumors against different antigens, such as Her2, Her3, EGFR, FGFR1, VEGFR, etc. The selected stable clones can be used in clinic trials immediately, thus making this CAR T technology more powerful and attractive in CAR-T cell immunotherapy.
We have high qualified groups and advanced technologies to perform TCR engineering services covering every unit of manufacturing and delivery pipeline. From a tumor-reactive T cell or active anti-tumor T-cell antigens, the appropriate target sequence is introduced to modify T cells to target a broad range of tumors with improved specificity. With engineering TCR construction and a series of assay, such as transgene expression, TCR/CD3 stability, lymphocyte antigen reactivity and cytotoxicity, animal experiments, etc., we provide cGMP-compliant TCR products for preclinical and clinical trials. Learn more about TCR T cells.
We have high qualified groups and advanced technologies to perform TCR engineering services covering every unit of manufacturing and delivery pipeline. From a tumor-reactive T cell or active anti-tumor T-cell antigens, the appropriate target sequence is introduced to modify T cells to target a broad range of tumors with improved specificity. With engineering TCR construction and a series of assay, such as transgene expression, TCR/CD3 stability, lymphocyte antigen reactivity and cytotoxicity, animal experiments, etc., we provide cGMP-compliant TCR products for preclinical and clinical trials. Learn more about TCR T cells.
Since the introduction of CAR, this field has developed greatly. The first CAR T generation contains a portion of the CD3 molecule that is responsible for signaling that the receptor has been bound by the outside world.
CAR T design can use any BCR to detect any known antigen and use it to activate T cells to kill the target. This is a method that many companies are currently exploring to fight cancer with CAR. First, T cells are isolated from the patient, and the patient's T cells are designed with a CAR specific to their cancer, which is then expanded and reintroduced into the patient.
In May of this year, the researchers reported that one patient developed symptoms on the 50th day after receiving CAR T treatment, which is obviously a special case for CAR-T therapy, either fast-acting or completely ineffective
Retrospective studies have shown that a gene called TET2 is accidentally destroyed during gene editing of T cells, and it itself inhibits the CAR T cell proliferation. In other words, this allows CAR-T cells to proliferate rapidly and eliminate cancer. Up to now, this patient has been cancer-free for five years.
CAR-T therapy is designed to target the CD19 protein on the surface of cancer cells. Once the cancer cells themselves acquire the CAR sequence, they can bind their own chimeric antigen receptor to the CD19 protein without triggering an immune response, equivalent to hiding CD19 and making the original effective CAR-T therapy lose its tracking goal.
CAR T therapy is a revolutionary anti-cancer therapy that isolates immune T cells from patients with blood cancers such as leukemia, genetically engineers them, and adds CAR to the surface proteins of cancer cells (chimeric antigen receptors), enabling them to attack cancer cells. These cells are then infused back into the patient to treat cancer.
Chimeric antigen receptor (CAR)-engineered T cell therapy has been the most promising approach up to date. The main advantage of CAR-T cell is that it bypasses MHC restriction, which allows for direct activation of effector cells for the treatment of various types of tumors.
To fully validate the technology, which is different from CAR-T technology, the trial is intended to recruit approximately 330 patients and statistical results are expected to be released in 2023. Leading researcher Rosenberg said: "Because this new immunotherapy relies on mutations rather than cancer types, in a sense, we can use this therapy to treat many types of cancer.
What is CAR-T?
CAR T is a T cell modified by genetic recombination technology. CAR in CAR-T is a recombinant receptor, and CAR T cell structure mainly includes three parts: extracellular antigen binding domain, transmembrane domain and intracellular signal domain.
As an effector cell of the human immune system, T cells are the only cells known to specifically kill tumors. However, before killing tumor cells, T cells must first "transform" into activated T cells, and the "transformation" process requires activation of both signals:
The first signal activation mode is that T cell receptors (TCR) bind to major histocompatibility class (MHC) on antigen presenting cells.
With the increasing use of CAR T treatment, treatment guidelines, comprehensive training for multidisciplinary staff, and other measures should help to properly manage the toxicity of this new treatment.
To this end, MD Anderson's CAR-T Cell Therapy-Related Toxicity (CARTOX) project, in collaboration with PALISI and its Hematopoietic Stem Cell Transplant (HSCT) team, provides a comprehensive guide for children receiving CAR T solution. The guide brings together experts from a wide range of fields, including pediatric intensive care physicians, pharmacy specialists, neurologists, and translational immunotherapeutics research experts, to provide important knowledge designed to help improve patient outcomes and outcomes.
Last August, the FDA approved CAR-T therapy for the treatment of acute lymphoblastic leukemia (ALL). A year later, scientists at the University of Texas MD Anderson Cancer Center, in conjunction with the Pediatric Acute Lung Injury and Sepsis Researcher Network (PALISI), published a comprehensive consensus guide on children receiving CAR-T cell therapy. The guide brings together the lessons learned by top experts in identifying early signs and symptoms of treatment-related toxicity and details how to respond to these symptoms.
CAR-T therapy is a genetic modification of a specific protein (chimeric antigen receptor, CAR) into a T cell after extraction of a patient's T cells. After reinjection of the modified T cells into the human body, the antigen (CD19) on the surface of leukemia cells can be specifically identified and killed. The data showed that the success rate of ALL patients after CAR-T clinical cell immunotherapy reached 90%. The ongoing research is aimed at expanding the use of this therapy in other cancers.
All cancers have mutations, and this is exactly what the immunotherapy is attacking. Ironically, the unique mutations that cause cancer are probably the best targets for treating cancer." ACT therapy used by researchers is different from chimeric antigen receptor T cell (CAR-T) therapy, the most promising immunotherapy for cancer treatment, which has shown remarkable ability to eliminate various kinds of tumors, especially for B cell malignancies, with up to 95% response rates and durable complete remission. CAR-T has been approved for use in hematological tumors, but it has not been shown to be effective against solid tumors.
ACT does not involve genetic modification of T cells to target cancer cell antigens, but whether this has an impact on its safety or efficacy remains to be seen. Like CAR-T, ACT technology requires the use of high-dose chemotherapy to destroy existing immune cells, which is itself a dangerous process.






