Tumgik
#oncogenic pathways
cancer-researcher · 7 days
Text
youtube
0 notes
willowcrowned · 2 years
Note
Lmao I’m dying over your tags about fruit fly gene names. I listened to an interview with a geneticist who works with C. elegans a while ago and he had major beef with fruit fly geneticists over naming conventions. 😂
you know, I’m beginning to wonder if this isn’t just a normal thing for them. my professor for the developmental biology part of this course is a big drosophila guy and two weeks ago he waxed lyrical about gene naming conventions for it, which was somehow still better than the solid twenty minutes he spent today trying to convince us of drosophila’s superiority as a means of studying development and cancer (presumably as a preface to Thursday’s lesson, where he actually has to talk about c elegans organogeneis experiments). at one point he had to admit that c elegans has a shorter generation time, but he quickly alleviated his own distress by following up that harrowing admission with three minutes of describing how wonderful the Nüsslein-Volhard and Wieschaus experiments were for the study of embryonic development
13 notes · View notes
medicomunicare · 1 month
Text
Though "poker faces", psoriasis and skin diseases show their last CARD: 1 4 Myc should be enough
Our skin, the body’s largest organ–provides the first line of defense against infections and many other threats to our health. Decades of research has shown that a wide range of diseases can occur, or become worse, when the skin cannot form an effective barrier. Millions of people are affected by eczema, psoriasis and other inflammatory skin disorders. Now, experts in human genetics and asthma…
0 notes
lifepulse · 10 months
Text
Drug resistance in multiple myeloma: When cancer cells say "NO" to treatment
Tumblr media
Drug resistance is like a game of cat and mouse. Cancer cells are the cat, and researchers are the mouse. The cat is always trying to find new ways to catch the mouse, but the mouse is always trying to find new ways to avoid getting caught.
Read More
0 notes
delveinsight12 · 10 days
Text
Understanding c-MET Non-Small Cell Lung Cancer Market: Causes, Symptoms, Diagnosis, and Treatment Options
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, comprising approximately 85% of all lung cancer cases. Within the realm of NSCLC, c-MET-positive tumors represent a significant subset associated with specific molecular and clinical characteristics. This article provides a detailed exploration of c-MET-positive NSCLC, including its causes, signs and symptoms, diagnostic approaches, and treatment options, drawing insights from the latest market research and developments.
c-MET, also known as hepatocyte growth factor receptor (HGFR), is a proto-oncogene that plays a crucial role in cell growth, migration, and invasion. In NSCLC, the c-MET gene can become dysregulated, leading to aberrant signaling pathways that promote tumor growth and metastasis. Overexpression or mutation of the c-MET gene has been implicated in aggressive tumor behavior and resistance to standard therapies.
c-MET-positive NSCLC often presents with distinct clinical features and may require specialized treatment approaches to effectively manage the disease. Identifying and targeting c-MET alterations is essential for optimizing therapeutic strategies and improving patient outcomes.
Causes of c-MET Non-Small Cell Lung Cancer
The exact causes of c-MET-positive NSCLC are multifaceted and involve a combination of genetic and environmental factors:
1. Genetic Mutations: Mutations or amplifications in the c-MET gene can lead to the overexpression of the c-MET protein, driving tumor growth and progression. These genetic alterations can result from inherited genetic predispositions or acquired mutations.
2. Environmental Factors: Exposure to carcinogens, such as tobacco smoke, asbestos, and environmental pollutants, is a well-established risk factor for lung cancer. These environmental factors can contribute to the development of genetic mutations and aberrant signaling pathways, including those involving c-MET.
3. Preexisting Lung Conditions: Chronic lung conditions, such as chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis, can increase the risk of developing lung cancer. These conditions may contribute to the dysregulation of c-MET signaling.
4. Family History: A family history of lung cancer or other cancers can increase the risk of developing NSCLC, including c-MET-positive tumors. Genetic predisposition and familial cancer syndromes may play a role in c-MET dysregulation.
Signs and Symptoms
The symptoms of c-MET-positive NSCLC are similar to those of other NSCLC subtypes, though the presence of c-MET alterations may influence the disease's clinical behavior. Common signs and symptoms include:
1. Persistent Cough: A chronic cough that does not improve with treatment may be an early sign of lung cancer. In c-MET-positive NSCLC, the cough may become more severe and persistent.
2. Shortness of Breath: Difficulty breathing or shortness of breath can occur as the tumor grows and obstructs the airways or lung tissue.
3. Chest Pain: Patients may experience localized chest pain or discomfort, which can be caused by the tumor's growth or invasion into surrounding tissues.
4. Hemoptysis: Coughing up blood or blood-streaked sputum may indicate advanced disease or tumor invasion.
5. Weight Loss and Fatigue: Unexplained weight loss and persistent fatigue are common systemic symptoms of cancer, including c-MET-positive NSCLC.
6. Recurrent Infections: Frequent respiratory infections or pneumonia may occur as the tumor obstructs or damages the lung tissue.
To know more about c-MET NSCLC Treatment market, visit: https://www.delveinsight.com/report-store/cmet-mnsclc-market
Diagnosis of c-MET Non-Small Cell Lung Cancer
The diagnosis of c-MET-positive NSCLC involves a combination of imaging studies, biopsy, and molecular testing:
1. Imaging Studies:
   - Chest X-Ray: A preliminary imaging test that can reveal abnormalities in the lungs, such as masses or nodules.
   - Computed Tomography (CT) Scan: Provides detailed cross-sectional images of the chest and can help identify the size, location, and extent of the tumor. CT scans are often used for staging and assessing tumor spread.
   - Positron Emission Tomography (PET) Scan: PET scans can detect areas of increased metabolic activity associated with cancer, aiding in the detection of metastases.
2. Biopsy:
   - Bronchoscopy: A procedure in which a flexible tube with a camera is inserted through the airways to obtain tissue samples from the tumor.
   - Needle Biopsy: A needle is used to extract tissue samples from the tumor, often guided by imaging techniques.
3. Molecular Testing:
   - c-MET Gene Testing: Identifies mutations or amplifications in the c-MET gene that are associated with tumor growth and progression. This testing is essential for determining the appropriate targeted therapy.
   - Next-Generation Sequencing (NGS): Provides comprehensive genetic analysis to identify various mutations and alterations in the tumor, including c-MET.
Treatment Options for c-MET Non-Small Cell Lung Cancer
Treatment for c-MET-positive NSCLC often involves a combination of standard therapies and targeted approaches to address the specific molecular characteristics of the tumor:
1. Surgery: Surgical resection is often the first-line treatment for localized NSCLC. The goal is to remove the tumor and affected lung tissue. However, surgery may not be suitable for all patients, particularly those with advanced or metastatic disease.
2. Radiation Therapy: Radiation therapy uses high-energy rays to target and destroy cancer cells. It may be used as an adjuvant treatment following surgery or as a primary treatment for inoperable tumors.
3. Chemotherapy: Systemic chemotherapy involves the use of drugs to kill cancer cells throughout the body. It is often used for advanced or metastatic NSCLC and may be combined with other treatments.
4. Targeted Therapy:
   - c-MET Inhibitors: Specific drugs targeting the c-MET pathway, such as crizotinib, cabozantinib, and tepotinib, are used to inhibit abnormal c-MET signaling and reduce tumor growth. These therapies are tailored to patients with c-MET gene mutations or amplifications.
   - Combination Therapies: Combining c-MET inhibitors with other targeted agents or immunotherapies may enhance treatment efficacy and overcome resistance.
5. Immunotherapy: Immune checkpoint inhibitors, such as pembrolizumab and nivolumab, can help stimulate the immune system to recognize and attack cancer cells. While not specific to c-MET, these therapies may be used in conjunction with targeted treatments.
6. Clinical Trials: Participation in clinical trials may provide access to new and experimental treatments that are not yet widely available. Clinical trials are essential for advancing treatment options and improving outcomes for patients with c-MET-positive NSCLC.
Market Insights
The market for c-MET-positive NSCLC treatments reflects the growing demand for targeted therapies and advancements in personalized medicine:
- Market Size and Growth: The global market for c-MET-positive NSCLC treatments is expanding, driven by increasing prevalence, advances in molecular diagnostics, and the development of targeted therapies. Key market segments include pharmaceuticals, diagnostic tools, and personalized treatment solutions.
- Key Players: Leading companies involved in the c-MET NSCLC market include:
   - Pfizer Inc.: Known for its development of targeted therapies and involvement in the treatment of lung cancer.
   - Roche Holdings: Engaged in the development of targeted therapies and molecular diagnostics for NSCLC.
   - Novartis Pharmaceuticals: Focuses on innovative treatments and research in oncology, including c-MET-targeted therapies.
   - AstraZeneca: Contributes to research and development of targeted and combination therapies for lung cancer.
- Research and Development: Ongoing research aims to improve understanding of c-MET signaling, develop new targeted therapies, and enhance diagnostic capabilities. Innovations in drug development and molecular diagnostics are contributing to better management and treatment outcomes for patients with c-MET-positive NSCLC.
c-MET-positive NSCLC represents a significant subset of non-small cell lung cancer with distinct molecular and clinical characteristics. Understanding the causes, symptoms, and treatment options is crucial for effective management of this challenging condition. Advances in targeted therapies and molecular diagnostics offer hope for improved outcomes and personalized treatment strategies. The growing market for c-MET NSCLC treatments underscores the importance of continued research and innovation in addressing this complex and impactful form of lung cancer.
Download report @ https://www.delveinsight.com/sample-request/cmet-mnsclc-market
0 notes
Text
Tumblr media
Adagrasib is an advanced investigational anti-cancer agent, specifically designed as a potent and selective inhibitor of the KRAS G12C mutation, which is prevalent in various cancers such as non-small cell lung cancer (NSCLC). This API (Active Pharmaceutical Ingredient) works by irreversibly binding to the mutant KRAS protein, thereby preventing its activation and subsequent oncogenic signaling pathways. This document delves into the pharmacodynamics, detailed mechanism of action, and emerging clinical applications of Adagrasib, offering insights into its potential role in targeted cancer therapies.
0 notes
Deciphering the Ras/MAPK Signaling Pathway in the Progression and Treatment of Hepatocellular Carcinoma_Crimson Publishers
Tumblr media
Abstract
Hepatocellular Carcinoma (HCC) is a serious health issue and its frequency is rapidly escalating throughout the world therefore researchers have focused more attention to the Ras/MAPK signaling pathway. The signaling pathways are linked to develop tumors and the Ras/MAPK pathway is one of these pathways, activated in 60% of HCCs with poor prognosis. A number of different proteins causes the abnormal regulation of the MAPK pathway in HCC. Ras, a small GTPase and Raf are the most commonly mutated oncogene supports the critical function of this pathway in oncogenesis. The genetic mutations leading to effector molecule to permanently activated in the Ras/MAPK signaling cascades. The inappropriate activation of this pathway is primarily due to the downregulation of various Ras/MAPK pathway inhibitors including RASSF proteins, GAPs, DUSP1, Spred and Sprouty proteins. The post-transcriptional or epigenetic processes downregulate these cancer suppressor genes. The aim of current study on the primary mutations resulting in aberrant activation of Ras/MAPK pathway and their role on the initiation and progression of HCC. It also offers an update on the various inhibitors to target this central signaling pathway including various Ras, Raf, MEK inhibitors in the context of HCC. Finally, we evaluate the available options for treatment in this context.
Read more about this article:
For more articles in Novel Approaches in Cancer Study
0 notes
healthcarehubhh · 2 months
Text
Neurofibromatosis Treatment Drugs Industry Growth
Tumblr media
Neurofibromatosis (NF) is a genetic disorder that causes tumors to form on nerve tissues. There are two main types - NF1 and NF2. NF1, also known as von Recklinghausen disease, is the more common form affecting around 1 in 3000 people. The main symptoms include light brown spots on the skin, tumors on or under the skin (neurofibromas), and Lisch nodules on the iris. NF2 is rarer and causes bilateral vestibular schwannomas (tumors on the eighth cranial nerve), which can lead to hearing loss and balance problems if not treated. Other features may include meningiomas (tumors of the meninges) and ependymomas (tumors of the central nervous system). Medical Management of NF1 For NF1, the treatment approach depends on the symptoms. If neurofibromas are small and cause no problems, only monitoring is needed. However, larger or painful neurofibromas may require surgery to remove them. Magnetic resonance imaging (MRI) scans are useful to monitor the growth of tumors. Children with NF1 are closely followed to watch for the development of optic pathway gliomas, which can affect vision if not treated. Medical therapy focuses on managing complications like high blood pressure, learning disabilities, and bone abnormalities. Targeted Neurofibromatosis Treatment Drugs Therapies In recent years, research has led to the development of targeted drug therapies that interfere with molecular pathways driving tumor growth in NF. One such pathway involves the RAS family of oncogenes, which are mutated in a high percentage of NF1 tumors. Selumetinib (Koselugo) is a MEK inhibitor drug approved by the FDA to treat inoperable plexiform neurofibromas in patients with NF1. By blocking the MEK protein, it helps control tumor growth. Another RAS pathway drug, sotorasib (Lumakras), showed efficacy against KRAS G12C mutant solid tumors in a clinical trial and may offer an option for NF patients with specific mutations. Several other MEK and RAF inhibitors are under investigation for NF. Medical Management of NF2 For NF2, treatment goals are to halt tumor growth and preserve hearing and neurological function as long as possible. Surgery continues to play a major role by removing tumors causing symptoms. Stereotactic radiosurgery uses focused beams of radiation to control residual or growing tumors without the risks of open surgery. Monitoring with serial MRIs helps determine when intervention is needed. The multikinase inhibitor sorafenib was shown to slow tumor growth in an NF2 clinical trial and represents a potential medical option. However, effective drug therapies are still quite limited for systemic treatment of NF2. Research Directions Ongoing research aims to discover new drugs that more specifically target signaling pathways driving NF tumor formation and growth. Candidate pathways include PI3K-AKT-mTOR, Hedgehog, Notch, Wnt, and Hippo signaling. Therapies modulating these cascades are in preclinical testing. Immunotherapies are another area of investigation since NF tumors can express tumor antigens that may stimulate anti-tumor immune responses. Combining targeted drugs with immunotherapy is a strategy to make treatments more effective. Advances in gene therapy also offer hope that someday, mutations causing NF could be directly corrected. Progress is being made, but more work is still required to develop curative options for these currently incurable genetic tumor predisposition syndromes.
0 notes
divyabodhwani23 · 5 months
Text
Accelerating Advancement: Navigating the Landscape of the EZH2 Inhibitors Market from 2024 to 2030
The EZH2 Inhibitors Market is a burgeoning sector within the pharmaceutical industry, propelled by advancements in cancer research and targeted therapy development. EZH2, or enhancer of zeste homolog 2, is a histone methyltransferase enzyme involved in epigenetic regulation, specifically in the addition of methyl groups to histone proteins. Dysregulation of EZH2 activity has been implicated in various cancers, making it an attractive target for therapeutic intervention. In this market analysis, we explore the key drivers, challenges, trends, and opportunities shaping the EZH2 Inhibitors Market.
𝐆𝐞𝐭 𝐟𝐫𝐞𝐞 𝐒𝐚𝐦𝐩𝐥𝐞: https://www.marketdigits.com/request/sample/4673
One of the primary drivers of the EZH2 Inhibitors Market is the increasing understanding of epigenetic mechanisms in cancer biology. Epigenetic alterations, including aberrant histone methylation mediated by EZH2, play a critical role in cancer initiation, progression, and metastasis. Targeting EZH2 with selective inhibitors offers a promising strategy for disrupting these epigenetic alterations and inhibiting tumor growth. As researchers unravel the complexities of EZH2 signaling pathways and their implications for cancer pathogenesis, the demand for EZH2 inhibitors as potential anticancer agents continues to grow.
The EZH2 Inhibitors Market is valued at USD 492.3 million in 2024 and projected to reach USD 3,244.7 million by 2030, experiencing a Compound Annual Growth Rate (CAGR) of 26.6% during the forecast period spanning 2024-2032.
Moreover, the EZH2 Inhibitors Market benefits from the growing prevalence of EZH2-altered cancers across various malignancies. EZH2 overexpression or gain-of-function mutations have been identified in several cancer types, including lymphoma, leukemia, prostate cancer, breast cancer, and solid tumors. These genetic alterations drive oncogenic processes, such as cell proliferation, survival, invasion, and metastasis, making EZH2 an attractive therapeutic target for precision medicine approaches. The expanding patient population with EZH2-altered cancers fuels the demand for effective EZH2 inhibitors that can provide therapeutic benefits and improve clinical outcomes.
Major vendors in the global EZH2 Inhibitors Market are Daichi Sankyo Co. Ltd., Epizyme Inc., Merck KGaA, Pfizer Inc., Eternity Bioscience, Kainos Medicine, Morphosys AG, Jiangsu Hengrui Pharmaceuticals Co. Ltd, Others Prominent Players.
In addition to targeting EZH2 directly, combination therapies involving EZH2 inhibitors and other anticancer agents are emerging as a promising approach to enhance treatment efficacy and overcome drug resistance. Preclinical and clinical studies have demonstrated synergistic effects when EZH2 inhibitors are combined with chemotherapy, targeted therapies, immunotherapy, or other epigenetic modifiers. By exploiting complementary mechanisms of action and therapeutic vulnerabilities in cancer cells, combination therapies hold potential for achieving durable responses and overcoming therapeutic resistance in EZH2-altered cancers.
However, the EZH2 Inhibitors Market also faces challenges and limitations that may hinder its growth and adoption. One of the main challenges is the development of selective and potent EZH2 inhibitors with favorable pharmacokinetic properties and acceptable safety profiles. Designing small molecules that specifically target the catalytic activity of EZH2 while sparing other methyltransferases and off-target effects remains a significant hurdle in drug discovery and development. Additionally, addressing acquired resistance mechanisms and optimizing treatment regimens to maximize clinical benefits represent ongoing challenges in the clinical translation of EZH2 inhibitors.
Furthermore, regulatory approval and market access for EZH2 inhibitors require robust clinical evidence demonstrating their safety, efficacy, and therapeutic utility in EZH2-altered cancers. Conducting clinical trials with appropriate patient selection criteria, biomarker validation, and endpoint assessment is essential for establishing the clinical value of EZH2 inhibitors and obtaining regulatory approvals. Moreover, reimbursement considerations, pricing strategies, and market dynamics influence the commercialization and accessibility of EZH2 inhibitors for patients with EZH2-altered cancers.
In conclusion, the EZH2 Inhibitors Market represents a promising frontier in cancer therapeutics, driven by advances in understanding the role of EZH2 in cancer biology and the development of targeted therapy approaches. While challenges and limitations exist, ongoing research efforts, collaborative partnerships, and technological innovations hold promise for overcoming these hurdles and advancing the clinical development and commercialization of EZH2 inhibitors. By harnessing the potential of EZH2 inhibitors as precision medicine agents, stakeholders in the oncology community can contribute to improving patient outcomes and addressing unmet medical needs in EZH2-altered cancers.
0 notes
Tumblr media Tumblr media Tumblr media Tumblr media
All credit to William Makis MD
NEW ARTICLE: Thymoquinone (Black Seed Oil / Nigella Sativa) and CANCER - New Research - 5 papers reviewed Black seed oil (Nigella Sativa) has a phytochemical compound with advanced anti-cancer properties called Thymoquinone.
I review 5 recent peer-reviewed papers about Thymoquinone's anti-cancer properties- inducing apoptosis- inhibiting proliferation- inhibiting angiogenesis (VEGF)- inhibiting cancer signaling pathways (NFkB)- inhibiting metastasis (MMP)Black Seed Oil & Thymoquinone have very unique properties, that may ALSO be relevant to people who have COVID-19 mRNA Vaccine Induced Turbo Cancer.1. Pfizer & Moderna mRNA Vaccine Spike protein can damage tumor suppressor p53 which may lead to cancer.
Black Seed Oil upregulates p53 and several other tumor suppressor genes like p21 and PTEN.2. COVID-19 Vaccines may contain oncogenic microRNAs or ONCOMIRS (anagrams include "Omicron" and "Moronic", which I'm sure is just a coincidence noticed only by conspiracy theorists) Black Seed Oil & Thymoquinone regulate a large number of microRNAs:- downregulate Oncogenic miR-21- downregulate Oncogenic miR-10- upregulate tumor suppressor miR-34a Wikipedia: "miR-21 is considered to be a typical 'onco-miR', miR-21 is one of the most frequently upregulated miRNAs in solid tumours, miR-21 is associated with a wide variety of cancers including: breast, ovaries, cervix, colon, lung, liver, brain, esophagus, prostate, pancreas and thyroid"
The big question is: are microRNAs involved in COVID-19 mRNA Vaccine Induced Turbo Cancer? Sadly, many who develop Turbo Cancer will not live long enough to find out.Pfizer = Co-miRNA-ty (miRNA is microRNA) Much more in my article...don't miss this forbidden topic! Article Link in photo to avoid shadowban, just re-type the URL in the 1st photo at the top, into your browser to access
@HighWireTalk
@ClaytonMorris
@TheChiefNerd
@VigilantFox
@joerogan
@TuckerCarlson
1 note · View note
Text
The Basics of DNA Methylation
DNA methylation is a fundamental epigenetic mechanism that plays a pivotal role in regulating gene expression and maintaining cellular function. Understanding the basics of DNA methylation is essential for comprehending its significance in various biological processes and its potential implications in health and disease.
What is DNA Methylation?
DNA methylation entails the attachment of a methyl cluster (CH3) to the cytosine constituent of DNA, predominantly happening at CpG dyads, where cytosine is succeeded by guanine. This alteration is facilitated by proteins termed DNA methyltransferases (DNMTs). Methylation at CpG positions has the potential to impact gene behavior by either encouraging gene suppression or aiding gene activation, contingent upon the site and surroundings within the genetic material.
Regulation of Gene Expression
DNA methylation serves as a critical mechanism for regulating gene expression patterns without altering the underlying DNA sequence. Hypermethylation of promoter regions typically results in gene repression by blocking the binding of transcription factors, RNA polymerase, and other regulatory proteins essential for gene activation. Conversely, hypomethylation of gene promoters often correlates with increased gene expression.
Tumblr media
Inheritance and Stability
DNA methylation patterns are established during early development and are typically faithfully maintained through cell divisions, contributing to cellular identity and function. However, DNA methylation is also subject to dynamic changes influenced by various factors such as environmental exposures, aging, and disease processes. These alterations can have profound effects on gene expression profiles and cellular phenotypes.
Epigenetic Regulation
DNA methylation is a key player in the broader landscape of epigenetic regulation, which encompasses heritable changes in gene expression that do not involve alterations to the DNA sequence itself. Alongside other epigenetic modifications such as histone modifications and non-coding RNAs, DNA methylation orchestrates complex regulatory networks governing diverse biological processes, including development, differentiation, and disease susceptibility.
Clinical Implications and Genetic Methylation Tests
Understanding aberrant DNA methylation patterns is crucial for unraveling the molecular mechanisms underlying various diseases, including cancer, neurodevelopmental disorders, and autoimmune conditions. Genetic methylation tests, which analyze the methylation status of specific genomic regions, have emerged as valuable diagnostic and prognostic tools in clinical settings.
Cancer and DNA Methylation
In cancer, widespread changes in DNA methylation patterns contribute to tumorigenesis by disrupting the normal regulation of gene expression. Hypermethylation of tumor suppressor genes and hypomethylation of oncogenes are common events associated with the development and progression of cancer. Genetic methylation tests enable the detection of these aberrant methylation signatures, aiding in cancer diagnosis, prognosis, and treatment selection.
Neurodevelopmental Disorders
Aberrant DNA methylation patterns have also been implicated in neurodevelopmental disorders such as autism spectrum disorders and intellectual disabilities. Altered methylation profiles of genes involved in neuronal development, synaptic function, and neurotransmitter signaling pathways may contribute to the pathogenesis of these conditions. Genetic methylation tests offer insights into the epigenetic signatures associated with neurodevelopmental disorders, potentially informing personalized treatment strategies.
Conclusion
DNA methylation is a fundamental mechanism of epigenetic regulation with profound implications for gene expression, cellular function, and disease pathogenesis. Advances in genetic methylation tests have revolutionized our ability to interrogate DNA methylation patterns, providing valuable insights into health and disease states. Continued research in this field promises to uncover new therapeutic targets and diagnostic biomarkers, ultimately improving patient care and outcomes.
0 notes
Text
Biomarker driven segments of NSCLC - Yet to saturate with new approvals
Industry experts think that NSCLC space is rushed with so many approvals, yet it is a hot space for researchers, as NSCLC market has huge potential and even 1% market share is a piece of hot cake for the investors.
Currently, there are a total of 5 FDA-approved TKIs for frontline treatment in EGFR- positive NSCLC.
Though, recent data on frontline Osimertinib confers the greatest progression-free survival (PFS) advantage for patients with EGFR-positive non–small cell lung cancer (NSCLC)
Experts indicated that they would look forward to the novel approaches that are trying to enhance the activity of Osimertinib by targeting other oncogenic pathways in combination with anti-EGFR therapy.
Write to us at [email protected] to learn how GRG Health is helping clients gather more in-depth market-level information on such topics.
Chemotherapy is backbone to treat many types of cancers, and research continues to find new chemotherapy regimens in combination with novel drugs. Over a decade, NSCLC space has evolved immensely.
Even though NSCLC space has been bifurcated into many biomarkers driven patients’ segment with specific approval in these segments, still in upcoming years we will see more approvals in the space. Industry experts think that this space is rushed with so many drugs, yet it is a hot space for researchers, as NSCLC is a huge market and even a single percent market share is a piece of hot cake for the investors.
Currently there are a total of 5 FDA-approved TKIs for frontline treatment of EGFR- positive NSCLC. These include erlotinib (Tarceva), gefitinib (Iressa), afatinib (Gilotrif), Dacomitinib (Vizimpro), and Osimertinib (Tagrisso).
We have seen a lot of exciting data over the past couple years with regard to these agents as monotherapy or in combination. Despite the sequential efficacy of these FDA-approved EGFR TKIs, frontline Osimertinib confers the greatest progression-free survival (PFS) advantage for patients with EGFR-positive non–small cell lung cancer (NSCLC) recently.
Therefore, we still expect to see several studies moving forward with results reported out in the coming years. We expect an emergence of new biomarker driven segments also (for e.g., Novel combinations, certainly targeting VEGF or MET).
There are exciting ongoing studies targeting MET following progression on Osimertinib. Experts indicated that they would look forward to the novel approaches that are trying to enhance the activity of Osimertinib by targeting other oncogenic pathways in combination with anti-EGFR therapy.
Visit our website now: https://www.grgonline.com/
0 notes
persephoneflouwers · 1 year
Note
hello, can you please help me explain the entire pathophysiology of breast cancer? it just doesn’t makes sense to me 🥲 even though i’ve watched videos on youtube but they do not talk about what happened to the cells or tissues or pathways or mechanisms for cancer metastasis
Hi! How deep should I go with this? What is not making sense to you? It’s a huge topic and kinda hard to summarise. If I were you, I would start with the basics of a cancer cell and what happens to any neoplastic cell in human body. I can send you a few papers I just downloaded for you (just send me an email and I’ll give it to you).
I would start with definition of tumor. A cancer cell is a cell that has compromised its normal, physiological process to die and keeps growing and reproducing uncontrollably. The genetic changes that contribute to cancer tend to affect three main types of genes—proto-oncogenes, tumor suppressor genes, and DNA repair genes. These changes are sometimes called “drivers” of cancer.
Breast cancer basically evolves like this. Cancer cells develop genetic mutations - that being familiarity or acquired mutations - that basically help them proliferate possibly forever.
Breast cancer is the most common cancer in women and used to be the main type of cancer leading to death in women (i think lately lung cancer has become pretty prevalent for women too, but im not aure).
There are different types of breast cancer: BC expressing Hormone Receptor (estrogen ER+ or progesterone PR+) BC expressing Human epidermal receptor (HER2+) and the triple negative (ER- , PR-, HER2-). The treatment is based on the molecular and histological type today.
The metastasis is a whole other chapter too. Metastasis is the dissemination of cancer cells from a primary lesion to distal organs that involves a variety of cellular mechanisms: invading through, or colluding with, stroma, escaping immune surveillance by inhibiting or co-opting their anti-tumorigenic processes, evading and modulating the tissue microenvironment, and evolving resistance to therapeutic intervention. These processe happens because inside the tumor cells genetic or epigenetic processes happens. This will bring to the alteration of many intracellular signalling pathways that control autonomous cellular progression to metastasis.
But you can come back for more questions. It’s just a huge topic, I don’t know what’s your level and how many infos you need
0 notes
tumimmtxpapers · 1 year
Text
β2-AR inhibition enhances EGFR antibody efficacy hampering the oxidative stress response machinery
The β2-Adrenergic receptor (β2-ARs) is a cell membrane-spanning G protein-coupled receptors (GPCRs) physiologically involved in stress-related response. In many cancers, the β2-ARs signaling drives the tumor development and transformation, also promoting the resistance to the treatments. In HNSCC cell lines, the β2-AR selective inhibition synergistically amplifies the cytotoxic effect of the MEK 1/2 by affecting the p38/NF-kB oncogenic pathway and contemporary reducing the NRF-2 mediated... http://dlvr.it/SwNJ8t
0 notes
rnomics · 1 year
Text
Cells, Vol. 12, Pages 2313: Let-7g Upregulation Attenuated the KRAS–PI3K–Rac1–Akt Axis-Mediated Bioenergetic Functions
The aberrant activation of signaling pathways contributes to #cancer cells with metabolic reprogramming. Thus, targeting signaling modulators is considered a potential therapeutic strategy for #cancer. Subcellular fractionation, coimmunoprecipitation, biochemical analysis, and gene manipulation experiments revealed that decreasing the interaction of kirsten rat sarcoma viral oncogene homolog (KRAS) with p110α in lipid rafts with the use of naringenin (NGN), a citrus flavonoid, causes lipid raft-associated phosphatidylinositol 3-kinase (PI3K)GTP-ras-related C3 botulinum toxin substrate 1 (Rac1)protein kinase B (Akt)-regulated metabolic dysfunction of glycolysis and mitochondrial oxidative phosphorylation (OXPHOS), leading to apoptosis in human nasopharyngeal carcinoma (NPC) cells. The use of lethal-7g (let-7g) mimic and let-7g inhibitor confirmed that elevated let-7g resulted in a decrease in KRAS expression, which attenuated the PI3KRac1AktBCL-2/BCL-xL-modulated mitochondrial energy metabolic functions. Increased let-7g depends on the suppression of the #RNA-specificity of monocyte chemoattractant protein-induced protein-1 (MCPIP1) ribonuclease since NGN specifically blocks the degradation of pre-let-7g by NPC cell-derived immunoprecipitated MCPIP1. Converging lines of evidence indicate that the inhibition of MCPIP1 by NGN leads to let-7g upregulation, suppressing oncogenic KRAS-modulated PI3K–Rac1–Akt signaling and thereby impeding the metabolic activities of aerobic glycolysis and mitochondrial OXPHOS. https://www.mdpi.com/2073-4409/12/18/2313?utm_source=dlvr.it&utm_medium=tumblr
0 notes
michellem123 · 1 year
Text
Kinases & Phosphatases
Protein phosphorylation is a reversible post-translational modification (PTM) that acts as a molecular switch for many cellular events. The activation and deactivation of signaling pathways through phosphorylation by kinases or dephosphorylation by phosphatases are under delicate control.
Approximately one-third of proteins in the human proteome are presumed to be phosphorylated during their life cycle, accounting for an estimated 100,000 different phosphorylation sites. Protein phosphorylation often leads to structural changes which then affect protein functions by modulating protein folding, substrate affinity, stability, localization and activity. Notably, the majority of oncogenes identified so far are protein kinases, whose activity when dysregulated is related to the development of cancers.
CD BioSciences offers a complete portfolio of solutions to study protein phosphorylation including phosphorylation profiling, kinase/phosphatase identification, substrate identification, and inhibitor/activator screening.
0 notes