#RNA PCR Test
Explore tagged Tumblr posts
dshseodelhi · 1 year ago
Text
Tumblr media
Book RNA PCR Test For HIV 2 By Drsafehands . Get more info RNA PCR Test For HIV Please Call / WhatsApp : 9013161616 .
0 notes
studychemistrytopics · 11 months ago
Text
2 notes · View notes
pathologylab · 28 days ago
Text
Tumblr media
On #World_AIDS_Day, we're proud to introduce the #HIV-Q Real-Time RT-PCR #Kit. This advanced test detects and quantifies HIV-1 #RNA with high sensitivity and specificity, ensuring precise diagnostics for healthcare professionals. Compatible with most real-time #PCR instruments, it offers reliable, cross-reactivity-free results to support better patient outcomes.
Let’s join hands in the fight against HIV with innovative #solutions!
#WorldAIDSDay2024 #HIVTesting #DiagnosticsInnovation #RealTimePCR #HealthcareInnovation #Diagnostics #Genes2Me #g2m #awareness
0 notes
covid-safer-hotties · 2 months ago
Text
Reference saved on our archive
By Amanda Blum
PCR tests are far superior to rapid antigen tests—and now you can get them for home use.
Last week, I was about to go on a date, and because I'm severely immunocompromised, we agreed he would take a COVID test using one of my rapid home PCR tests. It was a courtesy—he felt perfectly fine— but he tested positive. By the next day, he was sick as a dog. And, by the way, the rapid antigen test he took when he got home that night was negative.
Regardless of how you much of a health risk you see in COVID, it is still, at best, an inconvenience that costs you days off work. A simple home PCR test saved me from that inconvenience (and worse), and if I'd relied on the common rapid antigen test or done nothing at all, I would probably be sick right now.
While the world has desperately attempted to move on from COVID, this summer saw the highest case loads since 2022, with a winter surge just around the corner. Almost 300,000 people died from COVID in the US over the last three months alone, so while the pandemic has transitioned into endemic, according to the CDC, there are still risks to be aware of. Around 400 million people worldwide have long COVID, where symptoms can range from annoying to absolutely debilitating, regardless of your age, pre-COVID health, or fitness levels. Cases of long COVID are crushing our medical system, too. The two best tools to avoid getting COVID continue to be masking and testing. Unfortunately, the PCR testing centers that used to be available in each city have long closed, and obtaining a PCR has become expensive and hard to locate. This is why home testing kits are so important.
While you may be used to thinking of COVID tests as interchangeable, there’s a big difference between the standard at-home antigen test and a PCR (molecular) test. Almost five years in, it’s important to understand why PCR tests are the ones you want when accurate testing is important.
The difference between a PCR and a Rapid Antigen Test What you normally think of as a home COVID test—like the kind you can order for free from the government—is a rapid antigen test. When these at-home COVID tests became available, they were a powerful tool to help people know they were positive so they could isolate themselves from others. Almost all at-home tests were lateral flow tests, also known as rapid antigen tests (RATs). They measure for proteins on the outside of SARS-C0V-2, but they have a major flaw: They can only detect active virus. If you’re asymptomatic or don’t have a high viral load yet, the RAT may show negative results while you have an active and contagious infection.
This is why, if you already have symptoms, a negative antigen test isn't conclusive. You may need to test a number of times to confirm you have COVID. When you first get sick, you may go a number of days (as many as five) without enough virus to set off a positive RAT test. RATs were designed to be taken multiple times in sequence.
A PCR, also known as a NAAT or molecular test, measures RNA and can detect even small amounts of the virus. This is why it has always been considered the “gold standard” of COVID testing. These tests are generally considered accurate starting one to three days before you experience symptoms. Until last year, you needed to get a PCR from a testing center, but home tests have evolved and there are now four rapid, at-home molecular COVID tests, meaning you test and get a result within 30 minutes.
Why we still need COVID testing The world is now divided into people who view COVID as part of regular life and those who, due to chronic illness, immune issues, previous infections, or age, cannot afford to get infected. For a long time, we viewed COVID testing as something you do for your own health, but home PCR testing represents a way you can easily protect those vulnerable people in your life without cutting them off from society.
But even if you're not concerned about others, you should still care about protecting yourself from multiple infections. While the likelihood you will die of COVID has gone down dramatically due to vaccines, medical interventions, and natural immunity from infection, the news has not done a great job talking about long COVID. As people get infected two, three, four, and more times, they are playing against the odds. It’s estimated that one in 10—or even as many as one in five—infections leads to long COVID, and to explain how much it’s not “just the flu,” COVID is now considered to be a vascular illness. That means it affects the blood vessels in your body, which go everywhere. Thinking of COVID as a vascular illness helps explain why long COVID is everything from extreme fatigue to migraines to numbness in your extremities, loss of smell and taste, extreme fatigue, and neurological and cardiovascular conditions.
While lots of people no longer even test to see if they have COVID, there are a few reasons to get a definitive answer. First, you can only get the intervention Paxlovid within the first five days of symptoms. Anti-virals like Paxlovid knock down your viral load, one of the things we think helps prevent long COVID. Second, no one knows who will get long COVID, and you might need proof of that positive test in the future for insurance or benefits or even to justify sick days.
Lastly, you need to get tested because it is hard to know when you have COVID. Symptoms of COVID include headache, body ache, fever, sniffles, congestion, fatigue, sore throat, vomiting, diarrhea, and loss of smell or taste. In other words, absolutely anything out of the ordinary. While a RAT is unreliable for safe socializing with people for the reasons explained above, a molecular test can pretty reliably clear someone to come in your house that day, or be in close proximity. In that way, these molecular tests can be a tool to help immunocompromised people back into the world and make multigenerational celebrations safer.
How to get a molecular/PCR test Outside of your home, your main options now are urgent care clinics and places that do testing for travel. In both cases, they’ll be expensive. In the case of urgent care, they’ll put you in the same space as all the sick people, who are now no longer required to mask in healthcare settings, so if you don't already have COVID, you might pick it up there. Fortunately, there are molecular (PCR quality) tests you can take at home.
Rapid molecular tests require a similar effort on your part as a RAT test. You’ll swab yourself and then insert that swab into a machine that gives you a result. There are currently just four brands of these tests available: Lucira, Metrix, 3EO, and PlusLife. Unlike RAT tests, you have to order them, although Metrix and Lucira tests are available on Amazon, and Walgreens stocks Lucira tests in select stores. For a long time, they were just too expensive for most people, so they were relegated to the likes of movie sets, law firms, and Google employees. Prices have gone down, so now they’re more accessible—as low as $10 a test. Here are your options.
Follow the link to see the full review with relevant links!
97 notes · View notes
darkmaga-returns · 14 days ago
Text
This is no small question. It’s huge.
COVID proved that. On the basis of the test alone, millions of people were falsely diagnosed with the disease.
Let me back up. 99.99999999999999 of medical professionals believe the fairy tale called VIRUSES is real. They’ll always believe it, all the evidence to the contrary notwithstanding.
So this article operates within that insane context, because that’s where the pros live and breathe and work.
The PCR test searches for a piece of RNA which is part of a virus. When it finds that piece, it blows it up to a relatively enormous size. A size that can be observed.
Because in its original size, it was far too tiny to detect.
That’s called a clue. If the piece of RNA was so “tiny and alone,” why would anyone think it could cause a disease?
Traditional medical research asserts you need a whole crowd, a whole large mob of a particular virus to create disease.
So the PCR test is mortally flawed at the outset.
On top of this, the test can be adjusted to become even more sensitive. Meaning it will find not just a tiny, tiny remote RNA fragment, but a much, much, much TINIER fragment.
Such an adjustment is made by most testing labs. The PCR is tuned up to become more sensitive. Therefore, the useless test becomes even more useless.
4 notes · View notes
allthebrazilianpolitics · 1 month ago
Text
Retrospective epidemiologic and genomic surveillance of arboviruses in 2023 in Brazil reveals high co-circulation of chikungunya and dengue viruses
Tumblr media
Background
The rapid spread and increase of chikungunya (CHIKV) and dengue (DENV) cases in Brazilian regions in 2023 has raised concerns about the impact of arboviruses on public health. Epidemiological and genomic surveillance was performed to estimate the introduction and spread of CHIKV and DENV in Brazil.
Methods
This study obtained results from the Hermes Pardini (HP), a private medical laboratory, and the Health Department of Minas Gerais state (SES-MG). We investigated the positivity rates of CHIKV and DENV by analyzing the results of 139,457 samples tested for CHIKV (44,029 in 2022 and 95,428 in 2023) and 491,528 samples tested for DENV (163,674 in 2022 and 327,854 in 2023) across the five representative geographical regions of Brazil. Genome sequencing was performed on 80 CHIKV and 153 DENV samples that had been positive for RT-PCR tests.
Results
In our sampling, the data from CHIKV tests indicated that the Northeast region had the highest regional positivity rate in 2022 (58.1%). However, in 2023, the Southeast region recorded the highest positivity rate (40.5%). With regard to DENV, the South region exhibited the highest regional positivity rate in both 2022 (40.8%) and 2023 (22.7%), followed by the Southeast region in both years (34.8% in 2022; 21.4% in 2023). During the first 30 epidemiological weeks of 2023 in the state of Minas Gerais (MG), there was a 5.8-fold increase in CHIKV cases and a 3.5-fold increase in DENV compared to the same period in 2022. Analysis of 151 new DENV-1 and 80 CHIKV genomes revealed the presence of three main clusters of CHIKV and circulation of several DENV lineages in MG. All CHIKV clades are closely related to genomes from previous Brazilian outbreaks in the Northeast, suggesting importation events from this region to MG. We detected the RNA of both viruses in approximately 12.75% of the confirmed positive cases, suggesting an increase of co-infection with DENV and CHIKV during the period of analysis.
Conclusions
These high rates of re-emergence and co-infection with both arboviruses provide useful data for implementing control measures of Aedes vectors and the urgent implementation of public health politics to reduce the numbers of CHIKV and DENV cases in the country.
Read the paper.
2 notes · View notes
monopeptide · 2 years ago
Text
to do list
- review first manuscript for editor meeting tomorrow
- read papers about RQC/surveillance
- bsu rna prep (gDNA / rRNA)
- J&J library prep (cup pcr & excision / gup test)
4 notes · View notes
kristinahertzz · 3 days ago
Text
Hantavirus Diagnostics Market
Hantavirus Diagnostics Market Size, Share, Trends: Roche Diagnostics Lead
Advancements in Molecular Diagnostics Driving Market Growth Worldwide
Market Overview
The Hantavirus Diagnostics Market is experiencing steady growth, with a projected CAGR of 5.8% from 2024 to 2031. North America currently leads the market, driven by a higher incidence of hantavirus cases, advanced healthcare infrastructure, and increased awareness about the disease. Key metrics include rising investments in infectious disease diagnostics, growing demand for rapid and accurate testing methods, and increasing government initiatives for disease surveillance and control.
The market is expanding due to the rising prevalence of hantavirus infections worldwide, particularly in rural and suburban areas. The COVID-19 pandemic has also increased awareness of zoonotic infections, resulting in a renewed emphasis on improving diagnostic skills for viruses such as hantavirus.
DOWNLOAD FREE SAMPLE
Market Trends
Significant advances in molecular diagnostic techniques are transforming the field of hantavirus diagnostics, especially RT-PCR (Reverse Transcription Polymerase Chain Reaction) technology. These advancements enhance the sensitivity and specificity of hantavirus detection, enabling more timely and accurate diagnoses. Recently, multiplex PCR techniques capable of detecting multiple hantavirus strains simultaneously have been developed, reducing diagnostic time and costs.
There is also a growing interest in developing portable and point-of-care molecular diagnostic tools for hantavirus detection. These devices provide rapid and accurate testing capabilities in remote or resource-limited settings where hantavirus infections are more prevalent. For instance, a novel isothermal amplification method for hantavirus detection, performed on a portable device, can produce results in less than an hour.
Market Segmentation
The RT-PCR (Reverse Transcription Polymerase Chain Reaction) segment is expected to dominate the Hantavirus Diagnostics market during the forecast period. This segment's growth is largely due to the high sensitivity and specificity of RT-PCR testing in detecting hantavirus RNA, facilitating early and accurate diagnoses.
Recent advancements in the RT-PCR sector have focused on improving testing speed and efficiency. For example, several diagnostic companies have developed automated RT-PCR systems capable of handling multiple samples simultaneously, significantly reducing turnaround time. These advancements are particularly beneficial during outbreaks, where rapid testing of a large number of samples is crucial.
The COVID-19 pandemic has indirectly benefited the RT-PCR segment. The widespread use of RT-PCR testing for SARS-CoV-2 has led to increased investment in PCR infrastructure and expertise, which can be leveraged for hantavirus testing. The Association of Public Health Laboratories estimated a 25% increase in molecular testing capacity across public health labs in 2020, enhancing the availability of RT-PCR testing for other infectious diseases, including hantavirus.
Market Key Players
Prominent players in the Hantavirus Diagnostics Market include:
Roche Diagnostics
Abbott Laboratories
Thermo Fisher Scientific
bioMérieux SA
Qiagen N.V.
Siemens Healthineers
DiaSorin S.p.A.
Hologic, Inc.
Becton, Dickinson and Company
Bio-Rad Laboratories, Inc.
These leading companies are investing in technological advancements, strategic collaborations, and expanding their product portfolios to maintain their competitive edge.
Contact Us:
Name: Hari Krishna
Website: https://aurorawaveintellects.com/
0 notes
testing-services · 10 days ago
Text
How Advanced Technology is Transforming Virus Testing Labs
In recent years, the demand for accurate, efficient, and timely virus testing has skyrocketed, largely due to global health challenges such as the COVID-19 pandemic. Virus testing labs are at the forefront of combating infectious diseases, ensuring the safety of populations, and helping public health authorities control outbreaks. However, with advancements in technology, these labs are undergoing significant transformations that are enhancing their capabilities. In this blog, we will explore how advanced technology is reshaping virus testing lab, improving efficiency, and contributing to better health outcomes.
1. High-Throughput Testing Systems
One of the most notable technological advancements in virus testing labs is the development of high-throughput testing systems. These systems allow labs to process large volumes of samples in a short amount of time, making them ideal for managing widespread outbreaks. Traditional virus testing methods can be time-consuming and labor-intensive, often resulting in long wait times for results. High-throughput systems, on the other hand, enable labs to analyze hundreds or even thousands of samples simultaneously.
By automating much of the process, high-throughput testing reduces the burden on lab technicians and accelerates the time it takes to return test results. This is especially important during outbreaks, where quick results are crucial for isolating infected individuals and preventing further spread. Additionally, high-throughput systems increase the overall efficiency of virus testing labs, making them more equipped to handle surges in demand.
2. PCR Technology and Advancements
Polymerase Chain Reaction (PCR) testing remains one of the gold standards for virus detection. Over the years, PCR technology has become faster, more accurate, and more accessible. The continuous evolution of PCR-based testing technologies has improved virus detection in several key areas.
For instance, real-time PCR, or quantitative PCR (qPCR), enables virus testing labs to detect viral RNA more accurately and in real-time. This innovation has become vital in detecting viruses such as SARS-CoV-2, the virus responsible for COVID-19. With real-time PCR, labs can quantify the viral load, which not only helps in diagnosing active infections but also in determining the stage of the infection. Furthermore, multiplex PCR technology allows labs to test for multiple viruses simultaneously, reducing the time and cost associated with separate tests.
These advancements in PCR technology enable faster, more reliable testing, which is essential in controlling the spread of infectious diseases and managing public health crises.
3. Artificial Intelligence (AI) and Machine Learning
Artificial Intelligence (AI) and Machine Learning (ML) have become invaluable tools in virus testing labs, revolutionizing how data is analyzed and interpreted. AI algorithms can process vast amounts of data much faster and more accurately than humans, helping lab technicians identify trends, make predictions, and detect anomalies that may otherwise go unnoticed.
In the context of virus testing, AI and ML can analyze test results, cross-reference data from multiple sources, and even predict the likelihood of an outbreak based on current trends. AI systems can also be used to improve the design of diagnostic tests, optimizing parameters such as sensitivity, specificity, and cost-effectiveness.
Moreover, AI-based diagnostic tools are being integrated with PCR and other molecular testing methods to automate the process of reading results, reducing human error and increasing the speed of diagnosis. These technologies enable labs to deliver faster, more accurate results, which is especially crucial during outbreaks or emergencies.
4. Point-of-Care Testing Devices
Point-of-care (POC) testing devices are transforming the virus testing landscape by enabling rapid testing outside of traditional laboratory settings. These portable devices are used to detect viruses such as flu, COVID-19, and even HIV at the point of care, meaning that tests can be conducted in clinics, airports, or other public spaces.
Advancements in POC technology have made these devices more accurate, easier to use, and quicker to produce results. With results available within minutes, POC testing allows for quicker interventions, reducing the time it takes to isolate infected individuals and prevent further transmission.
In addition to speed, POC testing devices have become more cost-effective, which makes them accessible to underserved or remote populations. This is particularly important in ensuring equitable access to virus testing, especially in regions with limited healthcare infrastructure. These devices are also less reliant on specialized lab equipment, which makes them ideal for use in areas with limited resources.
5. Next-Generation Sequencing (NGS)
Next-generation sequencing (NGS) is a powerful tool used in virus testing labs to analyze viral genomes. This technology enables labs to sequence the genetic material of viruses with high accuracy and efficiency. NGS plays a crucial role in understanding how viruses evolve, mutate, and spread, providing vital information for researchers and healthcare providers.
For example, during the COVID-19 pandemic, NGS was used to track variants of the virus by sequencing its genome and identifying mutations. By understanding how the virus is changing, scientists and public health authorities can adapt public health strategies, modify vaccines, and update diagnostic tests.
NGS also helps in detecting viral infections that may not be captured by traditional testing methods. It can identify novel viruses or previously undiagnosed pathogens, which is essential for preventing future outbreaks. As NGS becomes more affordable and accessible, it is expected to play an even larger role in the diagnosis and surveillance of viral diseases worldwide.
6. Blockchain Technology for Data Security
As virus testing labs generate vast amounts of sensitive patient data, the importance of data security cannot be overstated. Blockchain technology is increasingly being used to secure test results and ensure the integrity of lab data.
Blockchain’s decentralized nature means that test results can be stored securely and remain tamper-proof. It provides a transparent and immutable record of each test result, which is crucial in maintaining trust and accountability in the testing process. In addition, blockchain can ensure that data is shared only with authorized individuals or organizations, preventing unauthorized access to sensitive health information.
By implementing blockchain technology, virus testing labs can strengthen data security, improve privacy compliance, and create a more transparent system for tracking and reporting test results.
7. Robotics in Virus Testing Labs
Robotics is another transformative technology in virus testing labs. Robotic systems are used to automate repetitive tasks, such as sample handling, extraction, and preparation. These systems can operate continuously without breaks, improving throughput and reducing human error.
In addition, robotics can help labs scale their operations during times of increased demand, such as during an outbreak or pandemic. By automating routine tasks, robotic systems allow lab technicians to focus on more complex aspects of testing and analysis, improving the overall efficiency of the lab.
The use of robotics in virus testing labs is not only limited to sample processing but also includes the delivery of test results, packaging, and even logistics. This reduces the manual workload and enhances the speed and accuracy of testing, which ultimately benefits patients and healthcare systems.
8. Telemedicine Integration for Remote Consultations
Telemedicine has become an essential part of healthcare delivery, especially in the wake of the COVID-19 pandemic. Virus testing labs are integrating telemedicine capabilities to allow patients to consult healthcare providers remotely and receive results without having to visit a clinic or hospital.
Telemedicine platforms can link patients to healthcare professionals who can advise them on the next steps after receiving their test results. Additionally, remote consultations reduce the risk of spreading infections in healthcare settings, allowing people to receive care from the safety of their homes.
As virus testing labs continue to integrate telemedicine, patients will have easier access to care and results, improving their overall experience and reducing the burden on healthcare facilities.
9. AI-Powered Prediction Tools
Predicting the future course of viral outbreaks is a critical aspect of public health preparedness. Advanced technology is enabling virus testing labs to harness the power of predictive analytics powered by AI. These tools can analyze large datasets, such as infection rates, social behaviors, and environmental factors, to forecast future outbreaks and identify areas at risk.
By using predictive analytics, virus testing labs can provide valuable insights to governments, healthcare providers, and businesses, helping them make informed decisions regarding public health measures, vaccination campaigns, and resource allocation.
Conclusion
The future of virus testing labs is undoubtedly tied to the continued integration of advanced technologies. From high-throughput testing systems to AI-driven data analysis, these innovations are enhancing the capabilities of virus testing labs and enabling faster, more accurate diagnoses. As the world continues to face new health threats, technology will play an increasingly vital role in preventing the spread of infectious diseases and ensuring the safety of populations globally. With these advancements, virus testing labs are becoming more efficient, accessible, and impactful, ultimately contributing to better health outcomes for all.
0 notes
unhonestlymirror · 12 days ago
Text
My fellow medics, this is must-read for you:
"The House Select Subcommittee on the Coronavirus Pandemic has completed its two-year investigation and released its final report.
The team prepared more than 100 investigative letters, conducted more than 30 transcripts of interviews and depositions, and held 25 hearings and meetings, reviewing more than a million pages of documents.
Key Points:
1. The virus has biological characteristics that are not found in nature. The first people to become ill were the lab workers themselves.
2. The publication “The Proximal Origin of SARS-CoV-2,” which has been repeatedly used by officials to discredit the lab leak theory, was called out by Dr. Fauci to promote the preferred theory that COVID-19 originated in nature.
3. EcoHealth, led by Dr. Peter Daszak, used U.S. taxpayer dollars to support dangerous research in China.
4. The NIH’s procedures for funding and overseeing potentially dangerous research are flawed, unreliable, and pose a serious threat to both public health and national security.
5. The Paycheck Protection Program, which offered Americans significant relief in the form of forgivable loans, was rife with fraud, resulting in at least $64 billion in lost taxpayer money.
6. Fraudsters cost American taxpayers more than $191 billion; at least half of the money lost in relief programs was stolen by international fraudsters.
7. The COVID-19 vaccine did not stop the spread or transmission of the virus as promised.
8. The FDA rushed to approve a COVID-19 vaccine. Two top FDA scientists have warned their colleagues about the dangers of rushing the vaccine approval process and the potential for side effects. They were ignored, and mandatory vaccinations were introduced within days. (Remember, these same scientists resigned from the FDA COVID-19 vaccine task force.)
9. Vaccine requirements were not supported by science and did more harm than good.
10. Public health officials made a concerted effort to ignore natural immunity from COVID-19 when developing vaccination recommendations and requirements, and they often spread misinformation through conflicting messages and lack of transparency.
11. Vaccine injury reporting systems have created confusion, failed to adequately inform the American public, and undermined public trust in vaccine safety during the covid-19 pandemic.
12. As a result of school closures, students have faced historic learning loss, higher rates of psychological distress, and worse physical well-being. Standardized test results show that children have lost decades of academic progress, and mental and physical health problems have also increased dramatically, with suicide attempts among girls aged 12-17 up 51%.
Full report here (520 pages): - https://oversight.house.gov/wp-content/uploads/2024/12/12.04.2024-SSCP-FINAL-REPORT.pdf
It's worth remembering how some of our so-called “Doctors” (Ukrainian) called in intensive care units (!) not to provide medical care to the unvaccinated... I’d better keep silent about the rest of the bacchanalia, the fruits of which we are still reaping, so as not to open the wounds.
"There have been previous reports of the long-term presence of s-protein in the body after immunization and large amounts of foreign DNA in the drug, since the end of 2022 this is a new study.
"We analyzed the contents of RNA and DNA in the vials and detected large amounts of DNA after RNAse treatment in all batches with concentrations ranging from 32.7 ng to 43.4 ng per clinical dose. This far exceeds the maximum permissible concentration of 10 ng per clinical dose established by international regulatory authorities.
Genetic analysis with selected pairs of PCR primers proved that the residual DNA is not only fragments of DNA templates encoding the s-protein, but also all genes from the plasmid, including the sv40 promoter/enhancer (unfortunately, highly oncogenic) and the antibiotic resistance gene."
https://publichealthpolicyjournal.com/biontech-rna-based-covid-19-injections-contain-large-amounts-of-residual-dna-including-an-sv40-promoter-enhancer-sequence/
In Germany, the Institute for Molecular Diagnostics (Inmodia GmbH) offers tests to determine the presence of vaccine proteins or DNA plasmids in the human body:
"We focus on detecting spike proteins in tissue samples (including tumor material), measuring the concentration of spike proteins in blood or cerebrospinal fluid (including immune cells), and detecting "vaccine mRNA" and residual DNA (plasmid DNA).
We focus on finding out whether the sample contains spike proteins and whether they come from a COVID-19 injection or from a natural infection."
https://inmodia.de/en/
0 notes
dshseodelhi · 1 year ago
Text
https://drsafehands.com/hiv-profile
DrSafeHands offers HIV 1 & 2 tests, HIV NAT tests, HIV RNA PCR tests, and HIV PCR tests.Testing at DrSafehands is highly affordable as our HIV tests are reasonably priced . affordable as our HIV tests are reasonably priced .
0 notes
stevecarell600 · 12 days ago
Text
Cancer Diagnostics Market Size, Share, Industry Growth and Emerging Trends Analysis by 2032
In 2023, the global cancer diagnostics market was worth $15.13 billion. It's expected to grow steadily, reaching $16.12 billion in 2024 and climbing to $31 billion by 2032, with an average annual growth rate of 8.5% over this period. North America led the market in 2023, holding a significant 35.89% share. 
Informational Source:
Major Key Companies Covered in Cancer Diagnostics Market are:
F. Hoffmann-La Roche Ltd (Switzerland)
Thermo Fisher Scientific Inc. (U.S.)
Abbott (U.S.)
Illumina, Inc. (U.S.)
GE Healthcare (U.S.)
BD (U.S.)
bioMérieux SA (France)
Myriad Genetics, Inc (U.S.)
Bio-Rad Laboratories, Inc. (U.S.)
QIAGEN (Germany)
Advancements and Trends in Cancer Diagnostics
Cancer diagnostics play a critical role in detecting, monitoring, and managing cancer at various stages. With advancements in technology and ongoing research, the field has witnessed transformative changes, offering new hope for early detection and improved patient outcomes. Below, we delve into the latest innovations and trends shaping cancer diagnostics today.
1. The Role of Liquid Biopsies
Liquid biopsy technology has revolutionized cancer diagnostics by offering a non-invasive method to detect cancer-related biomarkers, such as circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes, in blood or other bodily fluids. Unlike traditional biopsies, liquid biopsies can be performed with minimal discomfort and provide real-time insights into tumor dynamics.
Key Applications:
Early Detection: Screening for cancers like lung, colorectal, and breast cancers before symptoms appear.
Monitoring: Tracking tumor progression and response to treatments.
Personalized Treatment: Identifying genetic mutations to guide targeted therapies.
Recent Innovations:
Multi-Cancer Early Detection (MCED): Tests like GRAIL’s Galleri aim to detect multiple cancers simultaneously by analyzing ctDNA.
High Sensitivity Platforms: Techniques like next-generation sequencing (NGS) enhance the precision of biomarker detection.
2. Artificial Intelligence (AI) in Cancer Diagnostics
AI and machine learning (ML) are increasingly being integrated into cancer diagnostics to analyze vast amounts of data, identify patterns, and improve diagnostic accuracy. These technologies augment traditional methods by reducing human error and speeding up the diagnostic process.
Applications:
Image Analysis: AI algorithms analyze imaging data from MRI, CT, and mammography to detect anomalies indicative of cancer.
Pathology: Digital pathology solutions powered by AI can evaluate tissue samples for malignant changes with high precision.
Risk Prediction Models: AI systems can predict a patient’s risk of developing cancer based on their medical history, genetics, and lifestyle factors.
Notable Examples:
Google Health’s AI: Demonstrated higher accuracy than human radiologists in detecting breast cancer in mammograms.
PathAI: Utilizes deep learning to assist pathologists in diagnosing cancer from biopsy samples.
3. Advances in Molecular Diagnostics
Molecular diagnostics has seen significant advancements, allowing for the precise identification of genetic and molecular markers associated with different cancer types.
Technologies Driving Innovation:
Next-Generation Sequencing (NGS): Enables comprehensive genomic profiling to identify mutations, fusions, and other alterations that drive cancer.
Polymerase Chain Reaction (PCR): Used to amplify and detect specific DNA or RNA sequences linked to cancer.
CRISPR-based Detection: CRISPR technology is being developed for rapid and highly specific cancer biomarker detection.
Impact on Personalized Medicine:
Molecular diagnostics forms the backbone of personalized medicine by guiding therapies tailored to the genetic profile of a patient’s tumor. For instance:
EGFR mutations in lung cancer guide the use of tyrosine kinase inhibitors.
BRCA mutations in breast and ovarian cancer inform the use of PARP inhibitors.
4. Imaging Technologies in Cancer Detection
Imaging remains a cornerstone of cancer diagnostics, and advancements in this field have significantly improved the ability to detect and monitor tumors.
Innovations in Imaging:
Positron Emission Tomography (PET): Combined with CT or MRI, PET scans provide detailed information about tumor metabolism and structure.
Multiparametric MRI (mpMRI): Offers a more accurate assessment of prostate cancer compared to traditional methods.
AI-Enhanced Imaging: Machine learning algorithms improve the resolution and interpretation of imaging data, aiding in early detection and reducing false positives.
Emerging Modalities:
Optical Imaging: Techniques like fluorescence and bioluminescence imaging allow for the visualization of cancer at the cellular level.
Theranostic Imaging: Combines diagnostic imaging with therapy, enabling real-time monitoring of treatment efficacy.
5. Biomarker Discovery and Utilization
Biomarkers are critical for early detection, diagnosis, and prognosis in cancer care. Advances in proteomics, genomics, and metabolomics have expanded the pool of potential biomarkers.
Breakthroughs in Biomarker Research:
Proteomics: Identifying protein signatures unique to cancer cells.
Epigenetics: Analyzing DNA methylation and histone modifications as cancer-specific markers.
Metabolomics: Profiling metabolic changes associated with cancer progression.
Clinical Utility:
Predictive Biomarkers: EGFR, HER2, and PD-L1 guide targeted and immunotherapies.
Prognostic Biomarkers: Help estimate disease progression and survival rates.
Companion Diagnostics: Ensure that patients receive the most effective therapy based on their biomarker profile.
6. Point-of-Care (POC) Diagnostics
Point-of-care testing is transforming cancer diagnostics by bringing testing capabilities closer to patients, reducing the time to diagnosis and enabling quicker interventions.
Examples of POC Diagnostics:
Portable Devices: Handheld devices for detecting specific biomarkers in blood or saliva.
Lab-on-a-Chip Technology: Integrates multiple diagnostic processes on a microchip for rapid results.
Immunoassays: Quick tests for detecting cancer antigens, such as PSA for prostate cancer.
Impact on Low-Resource Settings:
POC diagnostics are particularly valuable in remote or underserved areas, where access to advanced diagnostic facilities may be limited.
7. Role of Genomics and Epigenomics
Genomic and epigenomic approaches are uncovering the complexities of cancer, enabling highly personalized diagnostic and therapeutic strategies.
Key Areas of Progress:
Whole Genome Sequencing (WGS): Offers a complete view of genetic alterations driving cancer.
Epigenetic Markers: Identifying changes in gene expression regulation without altering DNA sequences.
RNA Sequencing: Provides insights into gene expression changes specific to cancer.
Implications for Clinical Practice:
These techniques are helping identify rare and aggressive cancers, paving the way for novel treatments and clinical trials.
8. Emerging Diagnostic Technologies
Several groundbreaking technologies are poised to redefine cancer diagnostics in the coming years:
Nanotechnology:
Nanoparticles: Used for targeted imaging and detection of cancer cells.
Nanosensors: Detect minute changes in biomarker levels with high sensitivity.
Single-Cell Analysis:
Examines individual cancer cells, providing insights into tumor heterogeneity and resistance mechanisms.
Microbiome Analysis:
Studies suggest that changes in the gut microbiome may be linked to cancer development, offering a new avenue for diagnostics.
9. Challenges and Future Directions
Despite significant progress, challenges remain in the widespread adoption and implementation of advanced cancer diagnostics.
Key Challenges:
Cost: Many advanced diagnostic tools are expensive and inaccessible to a large population.
Regulatory Hurdles: Approvals for new diagnostics can be lengthy and complex.
Integration: Combining diverse diagnostic data into a cohesive patient profile.
Future Focus Areas:
Affordable Solutions: Development of cost-effective diagnostic tools for global accessibility.
Precision Diagnostics: Further integration of genomics, proteomics, and AI for more accurate and personalized care.
Global Collaboration: Sharing data and resources to accelerate innovation and standardize best practices.
Conclusion
The field of cancer diagnostics is undergoing a transformative era, fueled by technological innovations and a deeper understanding of cancer biology. From liquid biopsies and AI-driven imaging to molecular diagnostics and epigenomics, these advancements are paving the way for earlier detection, improved accuracy, and personalized treatment.
0 notes
pathologylab · 1 year ago
Text
Tumblr media
#Genes2Me's Blood #DNA Purification Kits allow #rapid and efficient #purification of genomic DNA from small volumes of human blood. #Kits are available in both #spin-column and #magnetic bead based formats. The purified genomic DNA is suitable for use in downstream applications including #PCR, restriction #enzyme digestion, and Southern blotting.
Visit our #website for more information. https://www.genes2me.com/nucleic-acid-extraction-solutions
For more details, Call us at +91-8800821778 or drop us an email at [email protected]
#forensic #rna #blood #hair #extraction #nucleicacid #ivd #madeinindia #molecular #diagnostics #g2m #diseases #moleculardiagnostics #moleculardiagnosticslab #risk #testing #testingsolutions
0 notes
covid-safer-hotties · 3 months ago
Text
Reference archived on our website
Published in 2023. Proof positive that just breathing spreads covid over a large area. Mask up. Ventilate. Clean the air.
Summary Background Effectively implementing strategies to curb SARS-CoV-2 transmission requires understanding who is contagious and when. Although viral load on upper respiratory swabs has commonly been used to infer contagiousness, measuring viral emissions might be more accurate to indicate the chance of onward transmission and identify likely routes. We aimed to correlate viral emissions, viral load in the upper respiratory tract, and symptoms, longitudinally, in participants who were experimentally infected with SARS-CoV-2.
Methods In this phase 1, open label, first-in-human SARS-CoV-2 experimental infection study at quarantine unit at the Royal Free London NHS Foundation Trust, London, UK, healthy adults aged 18–30 years who were unvaccinated for SARS-CoV-2, not previously known to have been infected with SARS-CoV-2, and seronegative at screening were recruited. Participants were inoculated with 10 50% tissue culture infectious dose of pre-alpha wild-type SARS-CoV-2 (Asp614Gly) by intranasal drops and remained in individual negative pressure rooms for a minimum of 14 days. Nose and throat swabs were collected daily. Emissions were collected daily from the air (using a Coriolis μ air sampler and directly into facemasks) and the surrounding environment (via surface and hand swabs). All samples were collected by researchers, and tested by using PCR, plaque assay, or lateral flow antigen test. Symptom scores were collected using self-reported symptom diaries three times daily. The study is registered with ClinicalTrials.gov, NCT04865237.
Findings Between March 6 and July 8, 2021, 36 participants (ten female and 26 male) were recruited and 18 (53%) of 34 participants became infected, resulting in protracted high viral loads in the nose and throat following a short incubation period, with mild-to-moderate symptoms. Two participants were excluded from the per-protocol analysis owing to seroconversion between screening and inoculation, identified post hoc. Viral RNA was detected in 63 (25%) of 252 Coriolis air samples from 16 participants, 109 (43%) of 252 mask samples from 17 participants, 67 (27%) of 252 hand swabs from 16 participants, and 371 (29%) of 1260 surface swabs from 18 participants. Viable SARS-CoV-2 was collected from breath captured in 16 masks and from 13 surfaces, including four small frequently touched surfaces and nine larger surfaces where airborne virus could deposit. Viral emissions correlated more strongly with viral load in nasal swabs than throat swabs. Two individuals emitted 86% of airborne virus, and the majority of airborne virus collected was released on 3 days. Individuals who reported the highest total symptom scores were not those who emitted most virus. Very few emissions occurred before the first reported symptom (7%) and hardly any before the first positive lateral flow antigen test (2%).
Interpretation After controlled experimental inoculation, the timing, extent, and routes of viral emissions was heterogeneous. We observed that a minority of participants were high airborne virus emitters, giving support to the notion of superspreading individuals or events. Our data implicates the nose as the most important source of emissions. Frequent self-testing coupled with isolation upon awareness of first symptoms could reduce onward transmissions.
48 notes · View notes
geneticteacher123 · 13 days ago
Text
Biological Labs supplies are: 1- Refrigerator (- 20) use to tore variety of samples at a very specific temperature range, 2- Ultra Low Temperature freezer (- 80) use to store samples for long period of time, 3- Distillation unit use for remove impurities from water by converting water into steam, 4- Liquid Nitrogen Containers are best for storage and transportation, 5- Electronic Balance use for obtaining the weight or mass of objects, 6- Laminar Flow Cabinet use for safely working with biosafety level by providing personal, environment and working protection as a contamination free work environment, 7- pH Meter use to measure hydrogen ion activity in water base solutions indicating its acidity or alkalinity which expressed as pH, 8- Water Bath use to incubate samples in water at a constant temperature over long period of time, 9- Hot Plate use to stir and heat solution simultaneously, 10- Microwave use for heating samples and solution for diverse experiments, 11- Incubator use in tissue culture and microbial genetic labs purposes, 12- Autoclave uses steam under pressure to sterilize labs apparatus, 13- Hot Air Oven use dry heat to sterilize labs apparatus, 14- Mechanical Shaker use to mix, blend or agitate substances in bottles or flasks by shaking, 15- Vortex use for mixing or agitate labs samples in test tubes for homogenization with high degree of precision , 16- Centrifuge is a device uses centrifugal force to separate various components of fluid on the basis of densities at controlled temperature, 17- Light Microscope use to view objects which are too small to see or explore with eye, 18- Mixer Mill is a grinding small amounts of samples use to mix and homogenize powders and suspensions in seconds for DNA, RNA and Proteins extractions, 19- Spectrophotometer measures the light intensity as a function of wavelength and use to measure the concentration of compound in aqueous solution, 20- Nanodrop is a quickly and easily quantify concentration of samples containing protein and nucleic acid even with a very small quantity of samples (2 microliters), 21- Real-Time PCR system use for detection and quantification nucleic acid sequences by measuring cycle accumulation, 22- Agarose Gel Electrophoresis use to separate and analyze macromolecules DNA and RNA on the basis of molecular size and charge, 23- SDS- PAGE use to separate and analyze protein on the basis of molecule size, 24- UV Illuminator use to view nucleic acids and protein suspended within polyacrylamide or agarose gels, 25- Gel Documentation system offer high image quality for analyzing Agarose or polyacrylamide gels #geneticteacher
0 notes
allthebrazilianpolitics · 2 months ago
Text
Re-emergence of Oropouche virus between 2023 and 2024 in Brazil: an observational epidemiological study
Tumblr media
Background
Oropouche virus is an arthropod-borne virus that has caused outbreaks of Oropouche fever in central and South America since the 1950s. This study investigates virological factors contributing to the re-emergence of Oropouche fever in Brazil between 2023 and 2024.
Methods
In this observational epidemiological study, we combined multiple data sources for Oropouche virus infections in Brazil and conducted in-vitro and in-vivo characterisation. We collected serum samples obtained in Manaus City, Amazonas state, Brazil, from patients with acute febrile illnesses aged 18 years or older who tested negative for malaria and samples from people with previous Oropouche virus infection from Coari municipality, Amazonas state, Brazil. Basic clinical and demographic data were collected from the Brazilian Laboratory Environment Management System. We calculated the incidence of Oropouche fever cases with data from the Brazilian Ministry of Health and the 2022 Brazilian population census and conducted age–sex analyses. We used reverse transcription quantitative PCR to test for Oropouche virus RNA in samples and subsequently performed sequencing and phylogenetic analysis of viral isolates. We compared the phenotype of the 2023–24 epidemic isolate (AM0088) with the historical prototype strain BeAn19991 through assessment of titre, plaque number, and plaque size. We used a plaque reduction neutralisation test (PRNT50) to assess the susceptibility of the novel isolate and BeAn19991 isolate to antibody neutralisation, both in serum samples from people previously infected with Oropouche virus and in blood collected from mice that were inoculated with either of the strains.
Findings
8639 (81·8%) of 10 557 laboratory-confirmed Oropouche fever cases from Jan 4, 2015, to Aug 10, 2024, occurred in 2024, which is 58·8 times the annual median of 147 cases (IQR 73–325). Oropouche virus infections were reported in all 27 federal units, with 8182 (77·5%) of 10 557 infections occurring in North Brazil. We detected Oropouche virus RNA in ten (11%) of 93 patients with acute febrile illness between Jan 1 and Feb 4, 2024, in Amazonas state. AM0088 had a significantly higher replication at 12 h and 24 h after infection in mammalian cells than the prototype strain. AM0088 had a more virulent phenotype than the prototype in mammalian cells, characterised by earlier plaque formation, between 27% and 65% increase in plaque number, and plaques between 2·4-times and 2·6-times larger. Furthermore, serum collected on May 2 and May 20, 2016, from individuals previously infected with Oropouche virus showed at least a 32-fold reduction in neutralising capacity (ie, median PRNT50 titre of 640 [IQR 320–640] for BeAn19991 vs <20 [ie, below the limit of detection] for AM0088) against the reassortant strain compared with the prototype.
Interpretation
These findings provide a comprehensive assessment of Oropouche fever in Brazil and contribute to an improved understanding of the 2023–24 Oropouche virus re-emergence. Our exploratory in-vitro data suggest that the increased incidence might be related to a higher replication efficiency of a new Oropouche virus reassortant for which previous immunity shows lower neutralising capacity.
Read the paper.
2 notes · View notes