#tumor antigen release
Explore tagged Tumblr posts
cancer-researcher · 18 hours ago
Text
youtube
0 notes
transgenderer · 1 year ago
Text
Tumblr media
Virus latency (or viral latency) is the ability of a pathogenic virus to lie dormant (latent) within a cell, denoted as the lysogenic part of the viral life cycle.[1] A latent viral infection is a type of persistent viral infection which is distinguished from a chronic viral infection. Latency is the phase in certain viruses' life cycles in which, after initial infection, proliferation of virus particles ceases. However, the viral genome is not eradicated. The virus can reactivate and begin producing large amounts of viral progeny (the lytic part of the viral life cycle) without the host becoming reinfected by new outside virus, and stays within the host indefinitely.[2]
Episomal latency refers to the use of genetic episomes during latency. In this latency type, viral genes are stabilized, floating in the cytoplasm or nucleus as distinct objects, either as linear or lariat structures. Episomal latency is more vulnerable to ribozymes or host foreign gene degradation than proviral latency (see below).
Advantages of episomal latency include the fact that the virus may not need to enter the cell nucleus, and hence may avoid nuclear domain 10 (ND10) from activating interferon via that pathway. Disadvantages include more exposure to cellular defenses, leading to possible degradation of viral gene via cellular enzymes.[12]
Proviral latency: A provirus is a virus genome that is integrated into the DNA of a host cell
All interferons share several common effects: they are antiviral agents and they modulate functions of the immune system. Administration of Type I IFN has been shown experimentally to inhibit tumor growth in animals, but the beneficial action in human tumors has not been widely documented. A virus-infected cell releases viral particles that can infect nearby cells. However, the infected cell can protect neighboring cells against a potential infection of the virus by releasing interferons. In response to interferon, cells produce large amounts of an enzyme known as protein kinase R (PKR). This enzyme phosphorylates a protein known as eIF-2 in response to new viral infections; the phosphorylated eIF-2 forms an inactive complex with another protein, called eIF2B, to reduce protein synthesis within the cell. Another cellular enzyme, RNAse L—also induced by interferon action—destroys RNA within the cells to further reduce protein synthesis of both viral and host genes. Inhibited protein synthesis impairs both virus replication and infected host cells. In addition, interferons induce production of hundreds of other proteins—known collectively as interferon-stimulated genes (ISGs)—that have roles in combating viruses and other actions produced by interferon.[13][14] They also limit viral spread by increasing p53 activity, which kills virus-infected cells by promoting apoptosis.[15][16] The effect of IFN on p53 is also linked to its protective role against certain cancers.[15]
Another function of interferons is to up-regulate major histocompatibility complex molecules, MHC I and MHC II, and increase immunoproteasome activity. All interferons significantly enhance the presentation of MHC I dependent antigens. Interferon gamma (IFN-gamma) also significantly stimulates the MHC II-dependent presentation of antigens. Higher MHC I expression increases presentation of viral and abnormal peptides from cancer cells to cytotoxic T cells, while the immunoproteasome processes these peptides for loading onto the MHC I molecule, thereby increasing the recognition and killing of infected or malignant cells. Higher MHC II expression increases presentation of these peptides to helper T cells; these cells release cytokines (such as more interferons and interleukins, among others) that signal to and co-ordinate the activity of other immune cells.[17][18][19]
Epstein–Barr virus lytic reactivation (which can be due to chemotherapy or radiation) can result in genome instability and cancer.[5]
HSV reactivates upon even minor chromatin loosening with stress,[7] although the chromatin compacts (becomes latent) upon oxygen and nutrient deprivation.[8]
Cytomegalovirus (CMV) establishes latency in myeloid progenitor cells, and is reactivated by inflammation.[9] Immunosuppression and critical illness (sepsis in particular) often results in CMV reactivation.[10] CMV reactivation is commonly seen in patients with severe colitis.[11]
viral latency is so fucked up
11 notes · View notes
science-sculpt · 11 months ago
Text
A Dive into CAR-T Cell Therapy
Imagine training your own soldiers to fight cancer. Not just any soldiers, but elite warriors genetically modified to recognize and demolish the enemy with laser-like precision. That's the essence of CAR-T cell therapy, a revolutionary approach turning heads in the medical world. In the fight against cancer, CAR-T cell therapy embodies this very concept, harnessing the body's own immune system to wage war against malignant cells. T cells are the special forces of your immune system, constantly scanning for and eliminating threats. But sometimes, cancer cells outsmart them, hiding in plain sight. CAR-T cell therapy steps in, equipping these T-cells with a unique weapon: a Chimeric Antigen Receptor (CAR). Think of it as a GPS that locks onto a specific protein on cancer cells, guiding the T-cells straight to their target.
How does it work? Here's the simplified version:
Recruitment: First, T cells are extracted from your blood. Modification Station: In the lab, scientists use a virus or other tools to insert the CAR gene into the T cells' DNA. This equips them with the cancer-targeting GPS. Bootcamp Boost: The modified T cells are grown in a special environment, multiplying into a powerful army. Redeployment: The CAR-T cell troops are infused back into your bloodstream, ready to seek and destroy.
Sounds amazing, right? But like any powerful technology, CAR-T comes with its own set of challenges. The treatment process is complex and expensive, and there can be serious side effects like cytokine release syndrome, where the immune system goes into overdrive. So, is CAR-T a miracle cure? Not yet. But for some patients with aggressive blood cancers like leukemia and lymphoma, it has shown remarkable results, offering hope where other treatments have failed. Researchers are constantly working to improve the safety and efficacy of CAR-T, making it a potential game-changer for even more cancers in the future. CAR-T cell therapy has demonstrated remarkable efficacy in the treatment of certain types of hematologic malignancies, including acute lymphoblastic leukemia (ALL) and certain subtypes of non-Hodgkin lymphoma. Clinical trials have shown unprecedented response rates and durable remissions in patients who have exhausted all other treatment options. Furthermore, ongoing research is exploring the potential of CAR-T cell therapy in treating solid tumors, extending its therapeutic reach to a broader spectrum of cancers. The future is bright for CAR-T. It's a testament to the power of human ingenuity and our ongoing quest to conquer one of humanity's greatest foes. While there's still a way to go, this groundbreaking therapy is a beacon of hope, reminding us that even the seemingly impossible can become reality.
3 notes · View notes
jerrycret68 · 28 days ago
Text
Creative Biolabs: Creative Thoughts to Shake Up mRNA R&D
Creative Biolabs offers top-notch mRNA technology support to fuel emerging therapeutic discoveries.
Recent headlines have been dominated by the transformative potential of mRNA-based therapies, extending beyond infectious diseases to encompass cancer, genetic disorders, and autoimmune conditions. This surge in interest underscores the importance of reliable and innovative partners in mRNA research and development. Creative Biolabs stands out as a pivotal player, providing comprehensive solutions that streamline the drug development process and enhance the efficacy of mRNA therapeutics.
One of the key offerings of Creative Biolabs is the mRNA technology platform, which provides a kaleidoscopic toolbox for mRNA research and development. The platform integrates the lipoplyplex (LPR) technique to produce mRNA modified by specific histidylated liposomes and polymers and encoding a wide variety of tumor antigens, ensuring a streamlined development workflow, improved mRNA delivery, and reduced overall cost.
A decisive component of successful mRNA therapeutics is the delivery system. Recognizing that one size does not fit all, Creative Biolabs offers customized mRNA delivery vehicles tailored to the unique requirements of each project. Their ability to customize delivery vehicles enables researchers to optimize the therapeutic index and overcome the specific challenges associated with the candidate mRNA drugs, whether they be targeted delivery, controlled release, or enhanced cellular uptake.
"We help scheme LNP formulations to make sure that the desired therapeutic mRNA reaches its intended destination within the body, maximizing therapeutic potential while minimizing side effects. Researchers can rely on our lipid nanoparticle technology to advance their mRNA-based treatments from the laboratory to next stage trials with confidence." According to a scientist at Creative Biolabs.
Furthermore, Creative Biolabs excels in developing lipid nanoparticles, one of the prevalent options of a successful delivery system, as an essential carrier to transport mRNA therapeutics into cells with their LNP technology designed to optimize the delivery efficiency and stability of mRNA molecules, allowing for one-stop, customized LNP preparation and scalable proprietary manufacturing.
Overall, Creative Biolabs is dedicated to pushing the boundaries of mRNA technology, empowering researchers to advance their mRNA-related research projects, and playing a crucial role in driving innovation in the field of mRNA therapeutics.
To learn more about Creative Biolabs' mRNA technology services, please visit: https://mrna.creative-biolabs.com.
About In an era where precision medicine and rapid drug development are paramount, Creative Biolabs' contributions are not just relevant—they are revolutionary. Their comprehensive mRNA technology platform, coupled with pioneering lipid nanoparticle and delivery system solutions, positions them as a leader in the mRNA therapeutics arena. By partnering with Creative Biolabs, scientists and pharmaceutical companies can accelerate their research, bringing life-changing therapies to patients faster and more efficiently than ever before.
0 notes
globalinsightblog · 1 month ago
Text
CAR T-Cell Therapy Market to Reach $20.3B by 2033, CAGR 14.7%
CAR T-Cell therapy Market: CAR T-cell therapy is revolutionizing oncology by offering a cutting-edge approach to treating certain cancers. This personalized immunotherapy involves modifying a patient’s T-cells to express chimeric antigen receptors (CARs) that specifically target and destroy cancer cells. It has shown remarkable success in treating blood cancers such as leukemia and lymphoma, providing hope for patients with otherwise limited options. As clinical trials expand, researchers are exploring its potential to combat solid tumors, pushing the boundaries of cancer treatment and transforming outcomes for countless individuals.
To Request Sample Report : https://www.globalinsightservices.com/request-sample/?id=GIS31609 &utm_source=SnehaPatil&utm_medium=Article
The rapid advancements in CAR T-cell therapy highlight its promise and challenges. Efforts to enhance efficacy, reduce side effects like cytokine release syndrome, and make the therapy more accessible are shaping the next generation of treatments. With increasing approvals and innovations in manufacturing processes, this therapy is poised to become a cornerstone of personalized medicine. Its success exemplifies the power of biotechnology to turn groundbreaking research into life-saving solutions, heralding a new era in the fight against cancer.
#CARTCellTherapy #CancerTreatment #Immunotherapy #PersonalizedMedicine #BiotechInnovation #BloodCancerCare #RevolutionInOncology #CancerBreakthroughs #FutureOfMedicine #CancerResearch #TCellEngineering #AdvancedTherapies #HopeForCancerPatients #BiotechRevolution #OncologyAdvancements
0 notes
fuhdubai · 3 months ago
Text
What is a Urology Test?
A urology test refers to any diagnostic procedure or medical examination performed to evaluate the function and health of the urinary tract system and male reproductive organs. These tests are used to diagnose conditions related to the kidneys, bladder, urethra, ureters, and in men, the prostate and testicles.
يشير اختبار المسالك البولية إلى أي إجراء تشخيصي أو فحص طبي يتم إجراؤه لتقييم وظيفة وصحة الجهاز البولي والأعضاء التناسلية الذكرية. تُستخدم هذه الاختبارات لتشخيص الحالات المتعلقة بالكلى والمثانة والإحليل والحالب، وفي الرجال البروستاتا والخصيتين.
Common urology tests include:
Urinalysis: A basic test that examines the urine for abnormalities, such as infection, blood, or protein.
Ultrasound: Imaging test used to visualize the kidneys, bladder, and reproductive organs.
Cystoscopy: A procedure where a thin tube with a camera is inserted through the urethra to inspect the bladder and urethra.
Urodynamic Testing: Evaluates how well the bladder and urethra are storing and releasing urine.
Prostate-Specific Antigen (PSA) Test: Blood test used to screen for prostate cancer.
CT Scan or MRI: Advanced imaging to look for kidney stones, tumors, or other structural abnormalities.
Tumblr media
What is URS in Urology?
URS (Ureteroscopy) is a medical procedure used in urology to examine the inside of the ureters (the tubes that connect the kidneys to the bladder) and, in some cases, the kidneys. URS is typically used to diagnose and treat conditions like kidney stones, tumors, or strictures within the ureters.
URS (تنظير الحالب) هو إجراء طبي يستخدم في طب المسالك البولية لفحص داخل الحالب (الأنابيب التي تربط الكلى بالمثانة)، وفي بعض الحالات، الكلى. يُستخدم URS عادةً لتشخيص وعلاج حالات مثل حصوات الكلى أو الأورام أو التضيقات داخل الحالب.
During URS:
A thin, flexible tube called a ureteroscope is inserted into the bladder and then guided up into the ureter and kidney.
The ureteroscope has a light and a camera, allowing the urologist to view the inside of the ureter and kidney.
If stones are found, they can be removed or broken up using specialized tools.
The procedure is usually done under general anesthesia, and patients can often go home the same day.
Tumblr media
URS is minimally invasive and is often preferred over more invasive surgical options for treating kidney stones and other urological issues.
The Urology and Neurourology Department at Fakeeh University Hospital treats patients with problems with the bladder, prostate, kidneys, and reproductive organs in men. The latter may include problems with erections, ejaculation, and infertility.
يُعالج قسم المسالك البولية والأعصاب في مستشفى فقيه الجامعي المرضى الذين يعانون من مشكلات في المثانة والبروستاتا والكلى والأعضاء التناسلية للذكور. وقد تتضمن مشكلات الأعضاء التناسلية هذه مشكلات الانتصاب، والقذف، والعقم.
يتم إيلاء اهتمام خاص بالجوانب الوظيفية لطب المسالك البولية، مثل الاختلال الوظيفي في الجهاز البولي (تسرب البول، صعوبة التبول) وآلا�� الحوض المزمنة (ألم في منطقة الأعضاء التناسلية والشرج). كما تُوفَّر أعلى مستويات الخدمات لتشخيص مشكلات المسالك البولية وعلاجها
Particular attention is given to the functional aspects of urology, like urinary dysfunctions (leak of urine, difficulty to pass urine) and chronic pelvic pain (pain in the genital and anal area). A high standard of diagnosis and treatment of urological problems is provided.
One of the standout departments at Fakeeh University Hospital Dubai مستشفى فقيه الجامعي دبي is the Urology and Andrology Department. Whether you’re dealing with kidney stones, urinary tract infections, prostate issues, or other urological conditions, the expert team at Fakeeh University Hospital is equipped with the latest technology and expertise to provide effective treatment. They offer a range of diagnostic and therapeutic services, including advanced procedures like URS (Ureteroscopy).
For more information on their urology services, you can visit their dedicated page here.
Trust your health with the specialists at Fakeeh University Hospital Dubai مستشفى فقيه الجامعي دبي, where patient care and medical excellence come first.
Tumblr media
Why Choose Fakeeh University Hospital?
State-of-the-Art Facility: The hospital is equipped with cutting-edge technology and modern infrastructure, ensuring that patients receive top-tier medical care in a comfortable and safe environment.
Expert Medical Team: The hospital boasts a highly qualified team of doctors, surgeons, nurses, and support staff, all dedicated to delivering personalized care. The medical professionals here are not only experts in their fields but also committed to staying updated with the latest medical advancements.
Comprehensive Care: Fakeeh University Hospital offers a wide range of specialties under one roof, making it a convenient choice for all your healthcare needs. From routine check-ups to complex surgeries, the hospital is designed to cater to patients with varying medical conditions.
🔗Click Here To Learn More
Spotlight on the Urology and Andrology Department
The Urology and Andrology Department at Fakeeh University Hospital Dubai مستشفى فقيه الجامعي دبي is one of the most advanced in the region. Here’s why it stands out:
Comprehensive Urological Services: The department provides a full spectrum of urological care, addressing conditions such as kidney stones, urinary tract infections, prostate disorders, bladder issues, and male infertility.
Advanced Diagnostic Tools: The hospital utilizes the latest diagnostic technologies, such as high-definition imaging and minimally invasive techniques, to ensure accurate diagnosis and effective treatment plans.
Minimally Invasive Procedures: The Urology Department is skilled in performing minimally invasive procedures like URS (Ureteroscopy). This procedure allows for the treatment of kidney stones and other urological conditions with minimal discomfort and quicker recovery times compared to traditional surgery.
Patient-Centered Approach: The team at Fakeeh University Hospital understands that urological conditions can be sensitive and sometimes challenging to discuss. They prioritize patient comfort, privacy, and clear communication throughout the treatment process.
Tumblr media
Why Trust Your Urological Care to Fakeeh University Hospital?
Holistic Approach: The hospital doesn’t just treat the symptoms but looks at the overall well-being of the patient, ensuring a holistic approach to healthcare.
Collaborative Care: The Urology Department works closely with other specialties within the hospital, such as nephrology and oncology, to provide coordinated care for patients with complex conditions.
Continuous Support: From diagnosis through treatment and follow-up, the team at Fakeeh University Hospital is dedicated to providing continuous support to their patients, ensuring the best possible outcomes.
Tumblr media
If you’re facing urological issues or need expert advice, don’t hesitate to reach out to the Urology and Andrology Department at Fakeeh University Hospital. For more detailed information, including how to book an appointment, visit their Urology and Andrology page.
Choosing Fakeeh University Hospital Dubai مستشفى فقيه الجامعي دبي means entrusting your health to one of Dubai’s most respected medical institutions, where your well-being is the top priority.
🏥 Click Here To Book An Appointment
0 notes
sunaleisocial · 3 months ago
Text
A new way to reprogram immune cells and direct them toward anti-tumor immunity
New Post has been published on https://sunalei.org/news/a-new-way-to-reprogram-immune-cells-and-direct-them-toward-anti-tumor-immunity/
A new way to reprogram immune cells and direct them toward anti-tumor immunity
Tumblr media
A collaboration between four MIT groups, led by principal investigators Laura L. Kiessling, Jeremiah A. Johnson, Alex K. Shalek, and Darrell J. Irvine, in conjunction with a group at Georgia Tech led by M.G. Finn, has revealed a new strategy for enabling immune system mobilization against cancer cells. The work, which appears today in ACS Nano, produces exactly the type of anti-tumor immunity needed to function as a tumor vaccine — both prophylactically and therapeutically.
Cancer cells can look very similar to the human cells from which they are derived. In contrast, viruses, bacteria, and fungi carry carbohydrates on their surfaces that are markedly different from those of human carbohydrates. Dendritic cells — the immune system’s best antigen-presenting cells — carry proteins on their surfaces that help them recognize these atypical carbohydrates and bring those antigens inside of them. The antigens are then processed into smaller peptides and presented to the immune system for a response. Intriguingly, some of these carbohydrate proteins can also collaborate to direct immune responses. This work presents a strategy for targeting those antigens to the dendritic cells that results in a more activated, stronger immune response.
Tackling tumors’ tenacity
The researchers’ new strategy shrouds the tumor antigens with foreign carbohydrates and co-delivers them with single-stranded RNA so that the dendritic cells can be programmed to recognize the tumor antigens as a potential threat. The researchers targeted the lectin (carbohydrate-binding protein) DC-SIGN because of its ability to serve as an activator of dendritic cell immunity. They decorated a virus-like particle (a particle composed of virus proteins assembled onto a piece of RNA that is noninfectious because its internal RNA is not from the virus) with DC-binding carbohydrate derivatives. The resulting glycan-costumed virus-like particles display unique sugars; therefore, the dendritic cells recognize them as something they need to attack.
“On the surface of the dendritic cells are carbohydrate binding proteins called lectins that combine to the sugars on the surface of bacteria or viruses, and when they do that they penetrate the membrane,” explains Kiessling, the paper’s senior author. “On the cell, the DC-SIGN gets clustered upon binding the virus or bacteria and that promotes internalization. When a virus-like particle gets internalized, it starts to fall apart and releases its RNA.” The toll-like receptor (bound to RNA) and DC-SIGN (bound to the sugar decoration) can both signal to activate the immune response.
Once the dendritic cells have sounded the alarm of a foreign invasion, a robust immune response is triggered that is significantly stronger than the immune response that would be expected with a typical untargeted vaccine. When an antigen is encountered by the dendritic cells, they send signals to T cells, the next cell in the immune system, to give different responses depending on what pathways have been activated in the dendritic cells.
Advancing cancer vaccine development
The activity of a potential vaccine developed in line with this new research is twofold. First, the vaccine glycan coat binds to lectins, providing a primary signal. Then, binding to toll-like receptors elicits potent immune activation.
The Kiessling, Finn, and Johnson groups had previously identified a synthetic DC-SIGN binding group that directed cellular immune responses when used to decorate virus-like particles. But it was unclear whether this method could be utilized as an anticancer vaccine. Collaboration between researchers in the labs at MIT and Georgia Tech demonstrated that in fact, it could.
Valerie Lensch, a chemistry PhD student from MIT’s Program in Polymers and Soft Matter and a joint member of the Kiessling and Johnson labs, took the preexisting strategy and tested it as an anticancer vaccine, learning a great deal about immunology in order to do so.
“We have developed a modular vaccine platform designed to drive antigen-specific cellular immune responses,” says Lensch. “This platform is not only pivotal in the fight against cancer, but also offers significant potential for combating challenging intracellular pathogens, including malaria parasites, HIV, and Mycobacterium tuberculosis. This technology holds promise for tackling a range of diseases where vaccine development has been particularly challenging.”
Lensch and her fellow researchers conducted in vitro experiments with extensive iterations of these glycan-costumed virus-like particles before identifying a design that demonstrated potential for success. Once that was achieved, the researchers were able to move on to an in vivo model, an exciting milestone for their research.
Adele Gabba, a postdoc in the Kiessling Lab, conducted the in vivo experiments with Lensch, and Robert Hincapie, who conducted his PhD studies with Professor M.G. Finn at Georgia Tech, built and decorated the virus-like particles with a series of glycans that were sent to him from the researchers at MIT.
“We are discovering that carbohydrates act like a language that cells use to communicate and direct the immune system,” says Gabba. “It’s thrilling that we have begun to decode this language and can now harness it to reshape immune responses.”
“The design principles behind this vaccine are rooted in extensive fundamental research conducted by previous graduate student and postdoctoral researchers over many years, focusing on optimizing lectin engagement and understanding the roles of lectins in immunity,” says Lensch. “It has been exciting to witness the translation of these concepts into therapeutic platforms across various applications.”
0 notes
jcmarchi · 3 months ago
Text
A new way to reprogram immune cells and direct them toward anti-tumor immunity
New Post has been published on https://thedigitalinsider.com/a-new-way-to-reprogram-immune-cells-and-direct-them-toward-anti-tumor-immunity/
A new way to reprogram immune cells and direct them toward anti-tumor immunity
Tumblr media Tumblr media
A collaboration between four MIT groups, led by principal investigators Laura L. Kiessling, Jeremiah A. Johnson, Alex K. Shalek, and Darrell J. Irvine, in conjunction with a group at Georgia Tech led by M.G. Finn, has revealed a new strategy for enabling immune system mobilization against cancer cells. The work, which appears today in ACS Nano, produces exactly the type of anti-tumor immunity needed to function as a tumor vaccine — both prophylactically and therapeutically.
Cancer cells can look very similar to the human cells from which they are derived. In contrast, viruses, bacteria, and fungi carry carbohydrates on their surfaces that are markedly different from those of human carbohydrates. Dendritic cells — the immune system’s best antigen-presenting cells — carry proteins on their surfaces that help them recognize these atypical carbohydrates and bring those antigens inside of them. The antigens are then processed into smaller peptides and presented to the immune system for a response. Intriguingly, some of these carbohydrate proteins can also collaborate to direct immune responses. This work presents a strategy for targeting those antigens to the dendritic cells that results in a more activated, stronger immune response.
Tackling tumors’ tenacity
The researchers’ new strategy shrouds the tumor antigens with foreign carbohydrates and co-delivers them with single-stranded RNA so that the dendritic cells can be programmed to recognize the tumor antigens as a potential threat. The researchers targeted the lectin (carbohydrate-binding protein) DC-SIGN because of its ability to serve as an activator of dendritic cell immunity. They decorated a virus-like particle (a particle composed of virus proteins assembled onto a piece of RNA that is noninfectious because its internal RNA is not from the virus) with DC-binding carbohydrate derivatives. The resulting glycan-costumed virus-like particles display unique sugars; therefore, the dendritic cells recognize them as something they need to attack.
“On the surface of the dendritic cells are carbohydrate binding proteins called lectins that combine to the sugars on the surface of bacteria or viruses, and when they do that they penetrate the membrane,” explains Kiessling, the paper’s senior author. “On the cell, the DC-SIGN gets clustered upon binding the virus or bacteria and that promotes internalization. When a virus-like particle gets internalized, it starts to fall apart and releases its RNA.” The toll-like receptor (bound to RNA) and DC-SIGN (bound to the sugar decoration) can both signal to activate the immune response.
Once the dendritic cells have sounded the alarm of a foreign invasion, a robust immune response is triggered that is significantly stronger than the immune response that would be expected with a typical untargeted vaccine. When an antigen is encountered by the dendritic cells, they send signals to T cells, the next cell in the immune system, to give different responses depending on what pathways have been activated in the dendritic cells.
Advancing cancer vaccine development
The activity of a potential vaccine developed in line with this new research is twofold. First, the vaccine glycan coat binds to lectins, providing a primary signal. Then, binding to toll-like receptors elicits potent immune activation.
The Kiessling, Finn, and Johnson groups had previously identified a synthetic DC-SIGN binding group that directed cellular immune responses when used to decorate virus-like particles. But it was unclear whether this method could be utilized as an anticancer vaccine. Collaboration between researchers in the labs at MIT and Georgia Tech demonstrated that in fact, it could.
Valerie Lensch, a chemistry PhD student from MIT’s Program in Polymers and Soft Matter and a joint member of the Kiessling and Johnson labs, took the preexisting strategy and tested it as an anticancer vaccine, learning a great deal about immunology in order to do so.
“We have developed a modular vaccine platform designed to drive antigen-specific cellular immune responses,” says Lensch. “This platform is not only pivotal in the fight against cancer, but also offers significant potential for combating challenging intracellular pathogens, including malaria parasites, HIV, and Mycobacterium tuberculosis. This technology holds promise for tackling a range of diseases where vaccine development has been particularly challenging.”
Lensch and her fellow researchers conducted in vitro experiments with extensive iterations of these glycan-costumed virus-like particles before identifying a design that demonstrated potential for success. Once that was achieved, the researchers were able to move on to an in vivo model, an exciting milestone for their research.
Adele Gabba, a postdoc in the Kiessling Lab, conducted the in vivo experiments with Lensch, and Robert Hincapie, who conducted his PhD studies with Professor M.G. Finn at Georgia Tech, built and decorated the virus-like particles with a series of glycans that were sent to him from the researchers at MIT.
“We are discovering that carbohydrates act like a language that cells use to communicate and direct the immune system,” says Gabba. “It’s thrilling that we have begun to decode this language and can now harness it to reshape immune responses.”
“The design principles behind this vaccine are rooted in extensive fundamental research conducted by previous graduate student and postdoctoral researchers over many years, focusing on optimizing lectin engagement and understanding the roles of lectins in immunity,” says Lensch. “It has been exciting to witness the translation of these concepts into therapeutic platforms across various applications.”
1 note · View note
healthcarehubhh · 4 months ago
Text
Growing Antibody-Drug Conjugates Market Owing to Rising Demand for Targeted Cancer Therapy.
Tumblr media
Antibody-drug conjugates (ADCs) are a type of bioconjugate consisting of monoclonal antibodies that are attached by chemical linkers to highly potent anti-cancer payloads. ADCs selectively target antigens that are highly expressed on tumor cells while sparing normal tissues through the use of antibodies. Linkers attached between the antibody and cytotoxic drug allow for the drug to be delivered unchanged until it reaches the intended tumor site, minimizing harm to healthy cells. ADCs have demonstrated clinical efficacy in treating various cancers including lymphoid malignancies, breast cancer, and solid tumors.
The Global Antibody-Drug Conjugates Market is estimated to be valued at US$ 5.38 Bn in 2024 and is expected to exhibit a CAGR of 14% over the forecast period 2023 to 2030. Key Takeaways: Key players operating in the Antibody-Drug Conjugates are AstraZeneca PLC, Daiichi Sankyo Company, Limited, Novasep, ADC Therapeutics SA, Alentis Therapeutics AG, F. Hoffmann-La Roche, Gilead Sciences, Inc., AbbVie Inc., Biosion USA, Inc., Astellas Pharma Inc., Duality Biologics (Suzhou) Co. Ltd., BioNTech SE, LaNova Medicines Ltd., Bliss Biopharmaceutical, Eisai Co., Ltd., ProfoundBio, Pfizer, Inc., ImmunoGen Inc., Mersana Therapeutics Inc., Sorrento Therapeutics Inc., Oxford BioTherapeutics Ltd, and Takeda Pharmaceutical Company Ltd. Growing demand for targeted cancer therapy with minimal side effects is expected to drive significant growth of the ADC market over the forecast period. Additionally, ongoing technological advancements in linker chemistry, increasing pipeline products and approvals are further fueling the market growth. Market Trends: The ADC market is witnessing increasing adoption of cleavable linkers that are stable in circulation but rapidly release the drug payload intracellularly upon internalization into target tumor cells. Additionally, the development of novel conjugation technologies such as DBCO-azide click chemistry is allowing for site-specific conjugation without effect on bioactivity and efficacy of ADCs. Market Opportunities: The significant opportunities in the ADC market are in developing ADCs for liquid and solid tumor indications with unmet medical needs. Additionally, optimization of physiochemical properties of molecules to improve pharmacokinetics is another key area that ADC developers are increasingly focusing on to enhance therapeutic index and efficacy of ADCs.
0 notes
credenceresearchdotblog · 4 months ago
Text
The demand for antibody drug conjugate was valued at USD 9984.20 million in 2023 and is expected to reach USD 26594.21 million in 2032, growing at a CAGR of 11.50% between 2024 and 2032.The Antibody-Drug Conjugate (ADC) market represents a groundbreaking intersection of biotechnology and pharmacology, offering a novel approach to cancer treatment. ADCs are sophisticated biopharmaceuticals designed to deliver cytotoxic drugs directly to cancer cells, minimizing damage to healthy tissues. This targeted therapy approach has gained substantial attention in recent years due to its potential to enhance the efficacy and safety profiles of cancer treatments. As the ADC market continues to expand, it is poised to revolutionize oncology and drive significant advancements in personalized medicine.
Browse the full report at https://www.credenceresearch.com/report/antibody-drug-conjugate-market
Market Overview
The global ADC market has witnessed rapid growth, driven by increasing cancer prevalence, technological advancements in drug development, and rising demand for targeted therapies. According to recent market research, the ADC market was valued at approximately USD 4.8 billion in 2022 and is expected to reach over USD 12 billion by 2028, growing at a compound annual growth rate (CAGR) of 16.7% during the forecast period.
This growth is fueled by the rising incidence of cancer worldwide, coupled with the limitations of conventional cancer therapies, such as chemotherapy and radiation, which often result in severe side effects. ADCs, by contrast, offer a more precise mechanism of action, delivering cytotoxic agents directly to cancer cells while sparing healthy tissues. This specificity reduces adverse effects and improves patient outcomes, making ADCs a promising option in the oncology landscape.
Technological Advancements
Advancements in ADC technology have been pivotal in driving market growth. Early-generation ADCs faced challenges such as low therapeutic indices, off-target toxicities, and limited efficacy. However, recent innovations have addressed these issues, leading to the development of more stable linkers, improved antibody engineering, and the use of highly potent cytotoxic agents.
Modern ADCs utilize cleavable linkers that release the drug payload only in the presence of specific enzymes or conditions within the cancer cell, ensuring targeted drug delivery. Additionally, advancements in monoclonal antibody engineering have enhanced the ability of ADCs to bind selectively to tumor-specific antigens, further improving their therapeutic potential.
Key Market Players
Several pharmaceutical companies are at the forefront of the ADC market, investing heavily in research and development to bring new ADCs to market. Notable players include:
1. Seagen Inc.: Known for its pioneering work in ADCs, Seagen’s ADC technology has led to the successful development of drugs like Adcetris, used in the treatment of Hodgkin lymphoma and other CD30-expressing lymphomas.
2. Roche: A global leader in oncology, Roche has developed Kadcyla, an ADC used in the treatment of HER2-positive breast cancer. Kadcyla combines the HER2-targeting properties of trastuzumab with a cytotoxic agent, offering a targeted approach to treating this aggressive form of breast cancer.
3. AstraZeneca: With the development of Enhertu, an ADC for HER2-positive breast cancer, AstraZeneca has further solidified its position in the ADC market. Enhertu’s unique design allows it to deliver a higher drug-to-antibody ratio, enhancing its therapeutic efficacy.
Challenges and Opportunities
Despite the promising outlook, the ADC market faces several challenges. The complexity of ADC development, high production costs, and stringent regulatory requirements can hinder market growth. Additionally, issues related to drug resistance and the need for personalized approaches to treatment pose ongoing challenges.
However, these challenges also present opportunities for innovation. Companies are exploring new ADC technologies, such as bispecific ADCs that target multiple antigens, and the use of alternative payloads to overcome drug resistance. Furthermore, ongoing research into biomarker-driven patient selection is expected to enhance the precision of ADC therapies, aligning them with the principles of personalized medicine.
Future Outlook
The future of the ADC market looks promising, with continued advancements in technology and a growing pipeline of ADC candidates in clinical trials. As more ADCs receive regulatory approval and enter the market, the adoption of this targeted therapy is expected to increase, offering new hope to cancer patients worldwide.
Moreover, the expanding application of ADCs beyond oncology, such as in the treatment of autoimmune diseases, presents additional growth opportunities. As research in this area progresses, ADCs may become a cornerstone of targeted therapy across various therapeutic areas.
Key Players
Seagen Inc.
Takeda Pharmaceutical Company Ltd.
AstraZeneca Plc.
F. Hoffmann-La Roche Ltd.
Pfizer Inc.
ImmunoGen Inc.
Gilead Sciences Inc.
Daiichi Sankyo Company Ltd.
Segmentation
By Product
Kadcyla
Enhertu
Adcetris
Padcev
Trodelvy
Polivy
Others
By Disease Type
Breast Cancer
Blood Cancer
Others
By Linker Type
Non-Cleavable
Cleavable
By Target
HER2
CD22
CD30
Others
By Payload Type
MMAE/Auristatin
Calicheamicin
Maytansinoids
Others
By Region
North America
The U.S
Canada
Mexico
Europe
Germany
France
The U.K.
Italy
Spain
Rest of Europe
Asia Pacific
China
Japan
India
South Korea
South-east Asia
Rest of Asia Pacific
Latin America
Brazil
Argentina
Rest of Latin America
Middle East & Africa
GCC Countries
South Africa
Rest of the Middle East and Africa
Browse the full report at https://www.credenceresearch.com/report/antibody-drug-conjugate-market
About Us:
Credence Research is committed to employee well-being and productivity. Following the COVID-19 pandemic, we have implemented a permanent work-from-home policy for all employees.
Contact:
Credence Research
Please contact us at +91 6232 49 3207
Website: www.credenceresearch.com
0 notes
tushar38 · 5 months ago
Text
CAR T-Cell Therapy: Challenges and Future Directions
Tumblr media
CAR T-cell therapy, short for Chimeric Antigen Receptor T-cell therapy, represents a groundbreaking advancement in cancer treatment. This innovative approach harnesses the power of a patient's own immune system to target and destroy cancer cells, offering new hope for patients with certain types of cancer, particularly those who have not responded to traditional therapies.
The process begins with the extraction of T-cells, a type of white blood cell crucial to the immune system, from the patient’s blood. These T-cells are then genetically modified in a laboratory to express a chimeric antigen receptor (CAR) on their surface. This receptor is designed to specifically recognize and bind to antigens on the surface of cancer cells. Once the T-cells are engineered and multiplied, they are infused back into the patient’s bloodstream, where they seek out and attack the cancer cells.
CAR T-cell therapy has shown remarkable success, particularly in treating hematologic malignancies such as certain types of leukemia and lymphoma. For instance, it has been approved for the treatment of relapsed or refractory B-cell acute lymphoblastic leukemia (ALL) in children and young adults, as well as certain types of non-Hodgkin lymphoma in adults. Clinical trials and real-world studies have demonstrated impressive remission rates, offering a lifeline to patients with otherwise limited treatment options.
However, CAR T-cell therapy is not without challenges. One of the most significant is the potential for severe side effects. Cytokine release syndrome (CRS) is a common and potentially life-threatening reaction, resulting from the rapid activation and proliferation of CAR T-cells and the subsequent release of large amounts of cytokines. Symptoms can range from high fever and flu-like symptoms to more severe complications affecting multiple organs. Another concern is neurotoxicity, which can cause confusion, seizures, and other neurological issues.
Additionally, the therapy’s high cost and complex manufacturing process pose barriers to widespread accessibility. The personalized nature of CAR T-cell therapy means that it must be tailored to each individual patient, which is both time-consuming and expensive. Efforts are ongoing to streamline production and reduce costs, as well as to expand the therapy’s applicability to solid tumors and other cancer types.
Despite these challenges, CAR T-cell therapy remains a beacon of hope in the oncology field. Continued research and clinical advancements are likely to enhance its efficacy, safety, and accessibility, potentially transforming the landscape of cancer treatment and providing new avenues for patient care.
0 notes
delveinsight12 · 5 months ago
Text
Transforming Cancer Treatment: The Rise of CAR T-Cell Therapy
In recent years, Chimeric Antigen Receptor (CAR) T-cell therapy has emerged as a revolutionary approach in the treatment of certain types of cancer. This innovative therapy involves reprogramming a patient's own immune cells to recognize and attack cancer cells, offering new hope where conventional treatments have fallen short.
Understanding CAR T-Cell Therapy
CAR T-cell therapy harnesses the power of the immune system, particularly T cells, to combat cancer effectively. The process begins by extracting a patient's T cells from their blood. These cells are then genetically engineered in a laboratory to express CARs on their surface. CARs are synthetic receptors that enable T cells to recognize specific antigens present on cancer cells.
Mechanism of Action
Once engineered, these CAR T cells are multiplied in number and infused back into the patient. Upon encountering cancer cells that express the targeted antigen, CAR T cells become activated, proliferate, and mount a targeted attack against the cancerous cells. This targeted approach minimizes damage to healthy tissues, unlike traditional chemotherapy or radiation therapy.
Download latest CAR-T Market Report @ https://www.delveinsight.com/sample-request/chimeric-antigen-receptor-car-t-cell-therapy-market
Clinical Success and Applications
CAR T-cell therapy has demonstrated remarkable success in treating certain types of blood cancers, such as acute lymphoblastic leukemia (ALL) and certain types of non-Hodgkin lymphomas. The therapy has shown durable responses in patients who have not responded to other treatments, including chemotherapy and stem cell transplants.
CAR-T Market Landscape and Growth
The CAR T-cell therapy market has been expanding rapidly, driven by increasing investment in research and development, as well as approvals of new therapies by regulatory authorities. According to industry insights, the market for CAR T-cell therapies is expected to grow significantly in the coming years, fueled by advancements in technology and expanding applications across various types of cancer.
Challenges and Future Directions
Despite its promising efficacy, CAR T-cell therapy is not without challenges. These include managing potential side effects, such as cytokine release syndrome and neurotoxicity, and optimizing manufacturing processes to meet the growing demand for personalized cell therapies.
Looking ahead, ongoing clinical trials are exploring the potential of CAR T-cell therapy in solid tumors and other challenging cancers. Researchers are also investigating ways to enhance the therapy's effectiveness and broaden its applicability across different cancer types.
CAR-T Leading Companies and Innovations
Several pharmaceutical and biotechnology companies are at the forefront of CAR T-cell therapy development. These include Novartis with Kymriah (tisagenlecleucel) and Gilead Sciences with Yescarta (axicabtagene ciloleucel), among others. These therapies have set new standards in oncology treatment and paved the way for future innovations in cellular immunotherapy.
In conclusion, CAR T-cell therapy represents a transformative approach in cancer treatment, offering personalized and targeted therapy options for patients with refractory or relapsed cancers. As research continues to advance and clinical outcomes improve, CAR T-cell therapy is poised to redefine the landscape of oncology and provide new hope for patients facing challenging diagnosis.
The journey from laboratory discovery to clinical application has been rapid, and the future promises further breakthroughs that may extend the benefits of CAR T-cell therapy to more cancer patients worldwide.
Request for a sample page @ https://www.delveinsight.com/sample-request/chimeric-antigen-receptor-car-t-cell-therapy-market
0 notes
internationalbiotherapy · 6 months ago
Text
Unveiling the Power of Oncolytic Viruses in Cancer Therapy: A Promising Frontier
Oncolytic viruses represent a promising frontier in cancer therapy, leveraging the innate ability of viruses to selectively target and destroy cancer cells while sparing healthy tissue. This innovative approach harnesses the dual power of direct cytotoxicity and immune system activation, offering new hope in the fight against various forms of cancer.
Historical Perspective and Evolution
The concept of using viruses to combat cancer dates back to the 1960s, when initial experiments explored the potential of viruses like poliovirus and adenovirus to induce tumor regression. These early efforts, though promising, faced significant challenges due to the potential for uncontrollable systemic infections. This led to a temporary halt in research until advances in DNA and RNA mapping technology enabled scientists to engineer safer, more targeted oncolytic viruses.
Among the most promising oncolytic viruses is the Newcastle Disease Virus (NDV). NDV specifically targets and kills cancer cells, inducing immune responses that further aid in eliminating tumors. Its unique properties make it an invaluable tool in developing personalized cancer therapies, enhancing treatment efficacy while minimizing side effects. Alongside NDV, other oncolytic viruses such as herpes simplex virus (HSV), reovirus, and vaccinia virus are being extensively studied and show significant potential in the realm of cancer treatment. Each of these viruses offers unique mechanisms of action and therapeutic benefits, broadening the scope and effectiveness of oncolytic virotherapy.
How Oncolytic Viruses Operate
Oncolytic viruses are adept at identifying and attaching themselves to cancer cells, exploiting specific receptors that distinguish them from healthy cells. Once inside the cancer cell, these viruses replicate, triggering a process known as apoptosis—programmed cell death. As infected cancer cells break down, they release new viral particles, which then proceed to infect neighboring cancer cells. This cycle continues, effectively amplifying the treatment's impact within the tumor.
Moreover, oncolytic viruses initiate an immune response against cancer cells by exposing viral antigens. This dual mechanism—direct cell destruction and immune activation—enhances the body's natural defenses against cancer, potentially eliminating residual cancer cells that conventional therapies might miss.
Enhancing Therapeutic Efficacy
To optimize the efficiency of oncolytic viruses, researchers are exploring various strategies. One approach involves combining oncolytic virotherapy with existing treatments such as radiation or chemotherapy. These therapies not only complement each other but also help mitigate immune responses that could prematurely neutralize the virus. By weakening the immune system's vigilance around the tumor site, these treatments create a more conducive environment for the oncolytic viruses to exert their effects.
Furthermore, scientists are investigating ways to augment the immune response triggered by oncolytic viruses. This includes integrating viral antigens into personalized cancer vaccines, which educate the immune system to recognize and attack cancer cells displaying these antigens. Such approaches transform "cold" tumors—those previously resistant to immune attacks—into "hot" targets for immune-mediated destruction.
Administration and Future Directions
Currently, oncolytic viruses are primarily administered intravenously or directly into localized tumors that are accessible. Intravenous administration allows for widespread dissemination of the virus throughout the body, targeting metastatic cancer cells that may have spread beyond the primary tumor site. Alternatively, direct injection into tumors converts them into sites of ongoing viral replication, essentially turning the tumor itself into an internal factory for generating anti-cancer agents.
Looking Ahead
The future of oncolytic virotherapy holds promise for further advancements. Ongoing research aims to refine virus engineering techniques to enhance tumor specificity and reduce potential side effects. Additionally, regulatory approvals for systemic administration are pending, suggesting broader accessibility and adoption of this cutting-edge treatment modality.
In conclusion, oncolytic viruses represent a paradigm shift in cancer treatment, merging virology with immunotherapy to combat malignancies in novel ways. As research continues to unravel the complexities of viral interactions with cancer cells and the immune system, the potential for personalized and potent cancer therapies continues to grow. With each breakthrough, the prospect of turning viruses once considered harmful into potent allies in the fight against cancer becomes increasingly tangible.
0 notes
tritonmarketresearchamey · 6 months ago
Text
Nanotech in Drug Delivery: Transforming Therapeutic Approaches
Tumblr media
The widely known methods of drug delivery include oral, parenteral (injected), topical, transdermal, nasal, ocular, etc. With current advancements in the medical field, nanotechnology is gaining significant traction due to its potential application in targeted drug delivery. As nanoparticles can penetrate biological membranes to deliver medications, they are crucial in drug therapy and developing methods for early diagnosis of diseases.
What are nanoparticles made of?
Defined as particles between 1 and 100 nanometers in size, nanoparticles exhibit unique properties that make them ideal for medical applications. These properties include a high surface area-to-volume ratio and customizable surface characteristics. Nanoparticles are made from various materials, including lipids, natural and synthetic polymers, metals like gold and silver, carbon-based materials, etc.
What is a nanoparticle drug delivery system?
A nanoparticle drug delivery system is a technology that uses tiny particles to deliver therapeutic agents like drugs, proteins, and genes. These nanoparticles can be engineered to specifically target diseased tissues, minimizing damage to healthy tissues. To achieve this, they are often coated with molecules that help them recognize and bind to diseased cells.
How Nanoparticles Function in Delivering Drugs?
Nanoparticles can enter the human body through three main routes: direct injection, inhalation, and oral intake. The mechanism of action of nanoparticles in drug delivery primarily involves their ability to improve the bioavailability of drugs and reduce side effects.
Drugs are encapsulated or attached to nanoparticles through methods like covalent bonding, adsorption, or encapsulation in liposomes or polymeric nanoparticles. In the bloodstream, these nanoparticles bind to specific markers on target cells. Once inside the target cell, they release the drug in response to conditions like acidic pH or specific enzymes, ensuring the drug is activated directly within the cell.
Key Applications of Nanoparticles in drug delivery
From vaccines to gene therapy, nanoparticles are utilized in treating cardiovascular diseases, infectious diseases, and neurological disorders. These particles can deliver nucleic acids (DNA, RNA) to specific cells, offering potential treatments for genetic disorders and cancers.
In recent years, an increasing number of scholars have been developing nano-drug carrier systems for the diagnosis and treatment of cardiovascular diseases (CVDs). The treatment strategies of these carriers include reducing inflammation, regulating lipoprotein levels, and preventing coagulation. Such strategies are used as interventions that help in managing atherosclerosis (the most common type of CVD leading to stroke).
For the treatment of hyper tension, nano-drug carriers provide certain benefits like lowering the blood pressure effect, longer maintenance time, and manage to produce dose reduction by three times. Additionally, they also serve as delivery vehicles in vaccines, improving immune response and stability of antigens. Their application can also enhance the contrast in imaging techniques like MRI, aiding in early and precise diagnosis.
Prominent Role of Nanotechnology in Cancer Therapy
In spite of being the most preferred cancer treatment, chemotherapy drugs meet limitations such as inconsistent penetration through physiological barriers, drug resistance, and toxicity to healthy, normal cells.
In order to tackle limited cancer cell targeting and the adverse effects on normal cells, nanotechnology plays a crucial role in sorting, imaging, and characterizing immune cells while minimizing side effects. Thus, nanoparticles enhance cancer treatments by overcoming drug resistance mechanisms, improving drug accumulation in tumors, and delivering vaccines that boost T-cell responses.
Let us have a look at the case study in San Diego, where nanoparticles were developed to treat tumors.
In October 2023, engineers at the University of California created customizable modular nanoparticles for targeting tumors, viruses, and toxins. Their modularity is enabled by two synthetic proteins, SpyCatcherand SpyTag, which spontaneously bind to each other.
As a proof of concept, these nanoparticles were tested on mice with ovarian tumors, which effectively suppressed tumor growth and improved survival rates. The researchers further aim to explore the nanoparticle drug delivery system, which holds potential for vaccine development.
Efficacy of Gold Nanoparticles in Treating Cancer
Another interesting application of nanotech in cancer treatments is using gold nanoparticles. Their unique properties enhance the efficacy of chemotherapy by delivering higher concentrations of the drug directly to the tumor, potentially allowing for reduced dosage and fewer associated side effects.
In this regard, the University of Texas at Dallas and UT Southwestern Medical Center published their research findings in 2023 regarding the development of a treatment method for glioblastoma, a common brain cancer. This technique used gold nanoparticles injected into the bloodstream, co-delivered with the medication.
Latest Developments in Nanotechnology in Drug Delivery
Research on nanoparticle drug delivery systems focuses on several key areas:
selecting and combining carrier materials to achieve optimal drug release speed
modifying the surface of nanoparticles to enhance their targeting ability
optimizing nanoparticle preparation to improve drug delivery efficiency
Recently, researchers at the University of Sydney have developed an oral insulin based on nanotechnology. It uses a nano-carrier to deliver insulin via tablets and release it only when blood sugar levels are high.
The nanomaterial is 1/10,000th the width of a human hair, which enhances insulin absorption in the gut. Preclinical trials show effective blood glucose management without hypoglycemia or weight gain. Human trials, led by Endo Axiom Pty Ltd, are set to begin in 2025.
Looking Forward:
The advent of nanotechnology has revolutionized the healthcare sector, particularly medicine being primary beneficiaries. One of the most promising applications of nanotechnology is evident in drug delivery systems, where nanoparticles are engineered to enhance the delivery and efficacy of therapeutic agents. Thus, the exploration and latest developments in nanotechnology imply a widening scope of nanoparticles in drug delivery.
0 notes
edenmartinez · 6 months ago
Text
Performed felled
Durability/performance/ safety/Se
Related infections
Result
transmission rates
• prove the case
• Me bought is posible in what store Instrumentive 90% in the intuiticion Patient injury
I Endoscopy design
small convention sgap expected contamination In 0%
Sign and sinthomy
Risk of contamination
For not cime constipation new scope 10%
now one solution what is 5the future hold
Señas de proves
Novel washing modalitié Table top
& Some remove biofil
other modalities Capaple o esterilazation long lumens, in develoments
*pinzas *electrolisis
*
sponge
*Refrigator
*Pale Cleaner
China Karten
*Desinfection termica- baño de maria
*alcohol
*Miltrex Instrument
*Surgery cleaner
*desinfection for methods chemystre
*evaporation
*Germicide[ Glutaraldehido; peroxido de hidrogeno y acido peritico
Cloruro mg
Sales de sodio
Silicato ppm
Valor ph
Acido citrico
Fosforicos
Orto-phalardehido
Peroxido de hidrogeno
Acido peracético
Miltex instrument
Surgery Jeaner
Destinacion por metodo
Circulo de Sinner
resistence corrosive presencia el agua
potable
Desalinizacion unsesfully del ag
Sal regenerator dragged ion exchangers
Comprobar con Norma Iso 10993 -3 Evaluacion Biologica de productos sanitarios
Level Antidrug concentracion high Concentration can result increase toxicity Low result in efficary biologics immunogenicity non- I ENF
Commonly performen other situation in
ransomized of considered dosie an adition
of therapy drug monitorizing
Ciclosporine an tacroliti-mas user organ
trasplation
Dancomycin angentamycin in sepsia
The presentation regarder show the monotherapy and study How is needly the cortecosteroide and others transition
considerations in the treatment inclusive options treatment
Conmeeting have the wormen virgin period its premenospause
Maintenance drops, for induction and derivates of quimiotherapy of that list of medicament is necesary for the study and benefict of patient pharmaeconomic benefict.
Potencial TDM and STRC
Redused in drug concentration
Monitorizing Drug with inmunohilatores to antibodys of immunogenocity..
Certolizumab pegol >20
Golimumab post induction > 10
Vedolizumab > 3
Ustekinomab > 5
Cledolizumab week >2,4 10 Cledoligimen. week 2,8 post induction, 8, 3, 5
of week 14 levels prediting duabiling
Sustained
PIFX Dose Dashboard
For Symphony of reflux electrodo-up the next quipement
One consicered
GERD = Infectious System To prevent rejection of organ transplant recipients
Hermozone: Cream with metamethazone conicoids: gel fusicor
Gel fusico GERD = Infectious System To prevent rejection of organ transplant recipients
What are antibodys - Drug congugates monoclonal antibodys Linked to a cytotoxic drug des to -n the therapeulic window by focusing a delivery to specifics cells.
*Tumor antigen *binding site
*monoclonal antibody
* selective for antigeno
with high copy numbers in target tumor celk with minimal inmyngenic response.
Linkers Can be cleavable (a temo Cells, Linkers Can be cleabank via tumor-
associated factors) or non-cleavable (via tumor associated factor of monel cavable (Lysosomalal degration
Payload:enhances cytoxicity all Thoughes and septicity although antibody raty afecct and clearance
Antibody bind target antigeno. Internalized → → payload releas +
bystander gel
ADCS may circulate as dynamic mixture of contact conjugate, naked antibodys,and
free payload
ACDS Reach tumors via capiliares releasing somepayload in micro
enviroment ass diffuse toward target
Any's independing tacles
Antibody engagement payload -
independent antitumor activity
Disruption o receptor dimerization and
funtion leads to disruption ofdownstream signaling
Antibody Drug congugates: Mechanism of action
• what are antibodys - Drug Congutgates + Monoclonal antidays Linked To a
CYTOTOXIC drog designes to ovide the therapevtic window by focusing a delivery To specific cells
Turmor antigen the hy
•Antibody binds target antigen Internalized 7 Payload release ADCS may circulate as dynamic mixture of context congujate, naked antibodys,
Conjugate, naked antibody, and free payload
ADCS reach tumors via capiliares releasing some payload in tumor micro enviroment aa diffuse toward target
ADCS: More than the Sum of their parts
Efective in heavily pretreated patient of those who have resistance to treatment antibody or chemotherapy components may be due to a superior therapeutic index Components vary, eg, FDM1 vs I-Dxd! vs Additive benefit from best. bystander Effect
Identifying the mechanist of effacey is challenging given multiple contributing part
ACDS toxicityies
Grade 3/4 Neutropenia by ADC on targeted ADC pivatuzumab off target an off tumor more difusse payload release inadverted
cellular uptake.
Anemia, neutropenia seen with many ADCS unknow pulmonaty toxicitys with many ADCS unknow pulmonaty with t-Dxd
THC imunohistochemistry
predictive biomarkers for ADCs hard to establish
•Where will ADCs for within the current
treatment Porading for lung cancer?
• Consideration of ADCs in combination with agent thar increase APC delivery modulate target antigen any or for internalization Promove antitumor immunity,
in comon, up Be complex bypass infysion guideline Opideline (ACD proteins)
by an antibody drug conjugate 's wely expressed IGFR mutation with con monosiclive tumor with adquired resistance To EGFR Targeted is Antigen is the ADC ressistention to antibodies
monociclive from tumor cell via endoson recicly prior to payload release Minimum issues chemotherapy or quimiotherapy
Halganior promore antllemor
is Antigen is the ADC. ressistention to Antibodies monactive From Tumor cell vier end es gon feciding, prior to praylord Teles
Lover is the thored meal for cance
example Urothelia Camer
Whal I describe of Her 3 itsel
Lmmunohibidor
HER3 DX d
operacion (Surgery 2 operasion (Surgery)
By pas asi minth
The atmen
I operacion (Sunger) operasion (Surgery,
By pas asiaminib
fuimia her afte -reatmen is overall 3 Mon
inimum issues
Insugnificance issues
Chemotherapy or Quimiotherapy Mechanism of resistance, to osimetris
Toaclina
Water
Witch GI is no have phrase
Hiz incidence of lineage plasticity including both small cell and squamous transformation by Frequent adquired gene alteration (eg, gene fusion)
Anticipate a role for non biomarker select in therapies in setting of osirmertinib resistance
Progresion Oh Osimertinib subpectos
• obtein brain MRI and chest/abdomen/ pelvis Ct
•Radiographic progressing lesion and submit for biomarker testing consider Liquid biopsy if tissue biopsy not feasible
but less sensitive for fusion/
transformations.
• Oligome ometastatic continue TX with addition of log ablative
•Slows and/or asymptomatic. Continue to beyond progression
CNS only. If Limited, continue tx with addition
if diffuse transition to CNS-penetrant Systemic
Progression on Osimerinibs. Supped to
Obtein brain MRI and Chess/aboomen, peluk- Pabgraphy of progresing confirmed! Consider whether special circumstances are obtein tissue present
obiopsy of progresing lesion and Submit for biomarker testing: consider liquid biopsy of tissue biopsy nor feaside, but
less sensitive for fusion/ transformation
bypass pathways
⚫met amplification
⚫clinical trial of Combined EGFR/MET
Inhibition
• Osimetinib + Met inhibitor (eg, crizotinibtinib, capmatinib
Fusion clinical trial
RET: OsimerTinibt
Ret Inhibitor (eg, Pralsetinib selpercatinib
ARK: Osimertive +ALK TIKE
Ros1 osimertinibt ROSTRI
Anexo.
maintenance Therapy observation
Osimetinib is almost in clinical
Considerated
is the standar of care Chemo Therapy (Histology, driven, platinum-based
Long Cancer
Anemia >
EGFR
PHASE III
Pembrolizumab - Carboplatin /
permetraxes.co alone in Advanced NSCLC Vs Chemotherapy add EGFR mutations with L858R or del19 EFRG mutations after EGFR IKI Therapy
mechanism or fusion of therapy
transformation of histology teatretment, follow up with, X Ray, abdomen of confirmed Progress
1First chemotherapy
2 on therapy I will aproximing
3 hemoestasis
4 Metastasis
trombotic Nervious sistem
Adjuvant treatmen in deumatic diseases o Spookify your armets, Juvenile reumatoid arthritis, post-traumatic Osteoarthritis, etc.) or in exacerbation of rheumatoid psoriatic arthrit.
Peace dose gradually avoid disease High doses decrease song every 1-2 days Weeks up to 18mg gay 19 Average dose 20-60mg day decreasdz by 5mg by 5.g
Starting at 10 ming day, decrease by 2,5 mg every 1-2 weeks.
mild dermal and Join Outbreaks Can be enough Short Courses of Cortecosteroids evidence how to be the construction in future of organ trasplnt in benefic of surge
What Phase no Have metatasis.
Ciclofosfamida
Classic Indications
Vasculitis Sistemicas con afectation and organs remopulmonary syndrome
Nefropatia Lupica Tipo III, IV Alveolitis fibrosane of multiple sclerosis sistemica
In monarche patient with hormone receptor (HR)-positive/HER2-mode- positive high risk EBC and previous
surgery and radiotherapy an indicated and adjuvant chemotherapy were randomize to receive adjuvant endocrine Therapy with or without adjuvant abemaceclir for two year. After a median follow of 19.1 month
treatmen with aberacidib up plus endocrine therapy significantly reduced the risk of an invasive desease-free
survival (IDFS) event hazan ratio 0,71, 95% Cl: 0,58-082; P=01) The benefit was even greater in patient with a Ki-67=20% (Harzard ratio 0.69, 95% CI: 052-092
Lon biopsy confirms invasive ductal carcinome estrogen receptor (ER) Positive, progesterone keeptor PR positive and HER2 negative Ki-67- Scome was 60% Germline mutation testing confirms no patogeny Vananita She plans to underjo Meoadjuvant Chemotherapy followed by
surgery
Guideline Recommended Biomaker testing For Breast Cancer
[on biopsy confirms invasive ductal carcinome estrogen receptor (ER) positive, progesterone keeptor positive and HER2 negative K1-67- Scone was 60% Germline noutation testing confins no patogeny Varianita She plans to undergo
Medadjuvant Chemotherapy followed by
surgery
Guideline Recommended Biomarker testing For Breast Cancer
Breast cancer receptor status Should be determinated as part of the diagnostic Workup for patient worth newly aliagnosed or current breast cancer, including Here statue by inmonohistochemistry with or witho Fluorescence in situ hybridization if needed and HR Status including ER and PR F3. several gene expres assays are avalaible that can help predict risk of recurrence and response to systemic therapy More recently germine mutation resting has been incorporated"
manregements of £BC, especif booking for BREA mutation To quide at uvant Herapy Select For patient with Her 2 negative breast cancer. "Las
me asurement of the Ki-67 antigen can be helpful when Considery specific targeted the rapies Such as aberma why the begen of breast tumlr like drive to cancer
Among pation with metas
breast cancer approximately 50% will desselop brain To F
metastaser managing brain metastases is a critical Before the last year
part of caring the population - There were no agent that Where no feven to be to treat central nervous metastases and result improved outcome me
Optimising systemic therapy for patient with Herz Pos. The early breast cancer
based den deseas charac-
terist and risk
0 notes
vermablogs · 9 months ago
Text
Revolutionizing Cancer Treatment: The Rise of CAR T-Cell Therapy
The global CAR T-Cell Therapy Market projected to surge to over USD 22.2 billion by 2032, from a modest USD 2.1 billion in 2023, it’s clear that this innovative approach is reshaping the landscape of cancer treatment at an unprecedented pace.
Unprecedented Demand: Transforming Cancer Care
The exponential growth in demand for CAR T-cell therapy is driven by its remarkable success in treating certain blood cancers, particularly leukemia. As of 2022, over 487,294 individuals worldwide were diagnosed with leukemia, underscoring the urgent need for effective treatment modalities. CAR T-cell therapy has emerged as a game-changer, boasting an impressive 80% cure rate for some leukemias, catapulting it to the forefront of cancer care.
Get Free Sample Report Here: https://www.astuteanalytica.com/request-sample/car-t-cell-therapy-market
Pioneering Research: Expanding the Therapeutic Horizon
While CAR T-cell therapies have demonstrated unparalleled efficacy in targeting B-cell CD19 antigens, researchers are actively exploring a multitude of other antigens across various cancer types. From hematologic malignancies to solid tumors, the potential applications of CAR T-cell therapy are vast and diverse. Clinical trials worldwide are exploring innovative approaches to enhance efficacy and broaden the scope of treatable cancers, offering renewed hope to patients facing previously insurmountable odds.
Overcoming Challenges: Navigating the Roadblocks
Despite its transformative potential, CAR T-cell therapy faces significant challenges on the path to widespread adoption. The staggering cost of treatment, exceeding $450,000 per patient, poses a formidable barrier to accessibility, limiting its reach to a select few. Moreover, the complexity of manufacturing CAR T-cell therapies presents logistical hurdles, hindering scalability and affordability on a global scale. Additionally, the occurrence of severe side effects such as cytokine release syndrome (CRS) and neurotoxicity underscores the critical need for further research and development to optimize safety and efficacy profiles.
Charting the Future: Collaborative Innovation
As we navigate the complexities of CAR T-cell therapy, collaboration emerges as a cornerstone of progress. By fostering partnerships between academia, industry, and healthcare providers, we can accelerate advancements in research, streamline manufacturing processes, and mitigate treatment-related risks. Through collective efforts, we can overcome existing challenges, expand access to CAR T-cell therapy, and empower patients with new avenues for conquering cancer.
Conclusion: A New Era in Cancer Treatment
In conclusion, the ascent of CAR T-cell therapy heralds a new era in cancer treatment — one characterized by innovation, optimism, and transformative outcomes. As we confront the formidable challenges ahead, let us remain steadfast in our commitment to advancing science, improving patient care, and ultimately, conquering cancer together.
0 notes