header logo image


Page 38«..1020..37383940..50..»

Archive for the ‘Immune System’ Category

Injecting the flu vaccine into a tumor gets the immune system to attack it – Ars Technica

Monday, January 6th, 2020

picture alliance/Getty Images

A number of years back, there was a great deal of excitement about using viruses to target cancer. A number of viruses explode the cells that they've infected in order to spread to new ones. Engineering those viruses so that they could only grow in cancer cells would seem to provide a way of selectively killing these cells. And some preliminary tests were promising, showing massive tumors nearly disappearing.

But the results were inconsistent, and there were complications. The immune system would respond to the virus, limiting our ability to use it more than once. And some of the tumor killing seemed to be the result of the immune system, rather than the virus.

Now, some researchers have focused on the immune response, inducing it at the site of the tumor. And they do so by a remarkably simple method: injecting the tumor with the flu vaccine. As a bonus, the mice it was tested on were successfully immunized, too.

This is one of those ideas that seems nuts but had so many earlier results pointing toward it working that it was really just a matter of time before someone tried it. To understand it, you have to overcome the idea that the immune system is always diffuse, composed of cells that wander the blood stream. Instead, immune cells organize at the sites of infections (or tumors), where they communicate with each other to both organize an attack and limit that attack so that healthy tissue isn't also targeted.

From this perspective, the immune system's inability to eliminate tumor cells isn't only the product of their similarities to healthy cells. It's also the product of the signaling networks that help restrain the immune system to prevent it from attacking normal cells. A number of recently developed drugs help release this self-imposed limit, winning their developers Nobel Prizes in the process. These drugs convert a "cold" immune response, dominated by signaling that shuts things down, into a "hot" one that is able to attack a tumor.

But not everyone has a response to these drugs, raising the question of whether there are other ways to activate the immune system at the site of a tumor. One potential option is simply the things that normally rev up the immune system: infectious agents. The immune response to cancer-targeting viruses mentioned above would provide an indication that this does occur. Others have targeted a variety of pathogens to the sites of tumors and found that this increases the immune response to the tumor as well.

To check whether something similar might be happening in humans, the researchers identified over 30,000 people being treated for lung cancer and found those who also received an influenza diagnosis. You might expect that the combination of the flu and cancer would be very difficult for those patients, but instead, they had lower mortality than the patients who didn't get the flu.

For more detailed tests, the researchers moved to mice, using melanoma cells that can form tumors when transplanted into the lungs of the mice. These model systems often respond to treatments that don't end up working in humans, so the results have to be treated with appropriate caution. Still, they can be a valuable way of understanding the biology of the immune response here.

The use of melanoma cells is informative, as these cells cannot be infected by the influenza virus. So this system also provides a test of whether the tumor cells themselves have to be infected in order to increase the immune response to them. Apparently they do not. Having an active influenza virus infection reduced the ability of the melanoma cells to establish themselves in the lung. The effect isn't limited to the location of the infection, though, as tumors in the lung that wasn't infected were also inhibited. The effects were similar when breast cancer cells were placed into the lung, as well.

All of this is consistent with the immune stimulation provided by a pathogen. The stimulation causes a general activation of the immune system that releases it from limits on its activity that prevent it from attacking tumor cells. But does it require an actual infection? To find out, the researchers used a flu virus that had been inactivated by heat treatment. Normally, heat treating a virus is used to create a control for an effect that needs an active virus. But here, it turned out to be another experiment, as the heat-treated virus was also able to work just as effectively as the live virus.

This isn't entirely surprising, given that inactive viruses are often used as vaccines and thus clearly can stimulate the immune system. But that, in turn, suggested another experiment: would vaccines actually work? To find out, the researchers obtained this year's flu vaccine and injected it into the sites of tumors. Not only was tumor growth slowed, but the mice ended up immune to the flu virus.

Oddly, this wasn't true for every flu vaccine. Some vaccines contain chemicals that enhance the immune system's memory, promoting the formation of a long-term response to pathogens (called adjuvants). When a vaccine containing one of these chemicals was used, the immune system wasn't stimulated to limit the tumors' growth.

This suggests that it's less a matter of stimulating the immune system and more an issue of triggering it to attack immediately. But this is one of the things that will need to be sorted out with further study. The location of the stimulation will also need to be sorted out, too. Here, stimulation in one lung increases activity in both. But injection into muscles didn't work at all, and earlier work by some of the same team had indicated a heavy infection outside the lungs enhanced tumor growth by diverting immune cells elsewhere.

But the story does fit in well with the general consensus that the immune system can be a powerful tool against cancer, provided it can be mobilized properly. And, in at least some cases, a flu vaccine just might do the trick.

PNAS, 2019. DOI: 10.1073/pnas.1904022116 (About DOIs).

Read the original here:
Injecting the flu vaccine into a tumor gets the immune system to attack it - Ars Technica

Read More...

How to Keep Your Immune System Strong During Winter – The Free Press of the University of Southern Maine

Monday, January 6th, 2020

The importance of a healthy immune system cant be overstatedespecially during the cold winter months. Common colds and the full-on flu are at their worst throughout the winter, so youll want to do whatever you can to boost your immune system and stay as healthy as possible. Follow this guide onhow to keep your immune system strong during winterto avoid untimely sickness.

Stress can do all kinds of terrible and negative things to your body. If your stress levels are through the roof, take a moment to find a release for relaxation. Without stress to weigh your body down, it can focus on building your immune system and keeping you at tip-top shape to face the harsh winter.

Drinking enough water is crucial to supporting your immune health; a healthy body requires eight cups of water a day. Unfortunately, many people dont like the way water tastes and consider it an unfulfilling drink. As a result, they dont drink itor they dont drink enough of it. One way to combat these issues is to jazz up plain water with some additional goodness. Heat up water and add lemonsfora delicious and nutritious wayto increase your H2O intake.

Vitamin C and zinc are common ingredients in many over-the-counter medicines because they help prevent and fight sicknesses. You dont have to rely on medicine, though. Many common foods contain vitamin C, zinc, or both: potatoes, broccoli, chicken, and tomatoes are some of the most popular items. Simply increasing your daily intake of these essentials can slowly and surely build extra immune system health and strength.

As you sleep, your body slows down and recharges. Consequently, your immune system can heal from the many battles it fought over the course of your day. While there are some exceptions to this rule, most of us need at least eight hours of sleep at nightthis give our bodies enough time to fully recover. Enough sleep is equally as important during the spring and summer seasons, too, as it gives your body time to fend off any germs it encountered during the day.

Exercise helps your body maintain health and can even contribute to building a stronger immune system. If youd like to go to the gym, thats great! However, even though exercise helps boost your immune systemand the gym is a wonderful place to exercisegyms are home to many different types of germs. Here are a few ways you can avoid germs before, during, and after your work out.

The winter is chilly, and keeping your body warm will help your immune system avoid fights and become stronger. Wear a hat when outdoors, as most of your bodys heat actually escapes through the top of the head. You should also wear a coat, proper footwear, gloves, and a scarf.

Read this article:
How to Keep Your Immune System Strong During Winter - The Free Press of the University of Southern Maine

Read More...

Investigating the Immune System’s Connection to Blood Clots | University of Michigan – Michigan Medicine

Monday, January 6th, 2020

Serious infections increase the risk of life-threatening conditions such as deep vein thrombosis (DVT), a blood clot that can form in deep veins, and pulmonary embolisms (PE), a condition where an artery in the lung becomes blocked by a blood clot. These conditions affect 1 in 1000 adults and lead to approximately 200,000-300,000 deaths per year.

In an effort to better understand blood clots, Andrea Obi, M.D., a vascular surgeon and investigator at Michigan Medicines Frankel Cardiovascular Center, is exploring the condition further through new research in her lab.

Her team is establishing a link between infection, thrombosis and changes in the bone marrow programming of immune cells and working on understanding the interplay between the immune system and thrombosis to help identify new, non-blood thinning techniques for preventing and treating DVT in the future.

LISTEN UP: Add the new Michigan Medicine News Break to your Alexa-enabled device, or subscribe to our daily updates on iTunes, Google Play and Stitcher.

Obi, also the winner of the 2019 Wylie award from Vascular Cures, takes a deeper dive into her work here with questions from the Michigan Health Lab:

Whats the focus of your research?

My research focuses on the intersection between innate immunity and the coagulation system, specifically evaluating the role immune cells play in forming and breaking down blood clots in the venous system.

What sparked your interest in this?

My current work, like many surgeons, was the result of observing human suffering. During the 2009-2010 H1N1 influenza outbreak, I spent much time in the surgical intensive care unit caring for young individuals, some in their 20s and 30s, with severe pulmonary disease requiring advanced life support. We noticed that a large number of these patients developed severe venous clotting, leading to a very high mortality. Blood thinners helped us decrease the risk of death but led to a host of other bleeding complications for these ill patients.

When I had a chance to review data from large population studies, it became evident that DVT was associated with all types of infections to varying degrees, and the risk persisted over the course of the persons next year of life, even if he or she made a rapid recovery.

What discoveries has your team already made?

So far weve discovered that even with a remote infection, such as a pneumonia, the endothelium (inner lining of the blood vessel) in a remote location changes the proteins expressed on the cell surface and talks to circulating leukocytes differently.

How will patients benefit from this research?

My hope is that by using both animal models and human tissue we can identify some of the changes that occur in innate immune cell memory that predisposes individuals to form a DVT after suffering from an infection.

MORE FROM MICHIGAN: Sign up for our weekly newsletter

In recent years, the pharmacology for a wide range of diseases such as rheumatoid arthritis, psoriasis, and melanoma have been transformed by the ability to target the immune system. Despite the fact that its never been tried in DVT, doesnt mean that it is impossible, rather that we just dont understand enough about the crosstalk between the immune system and coagulation (blood clotting) system to identify a protein or molecule that we can manipulate to change the course of human disease.

I hope that in the course of my work and in my lifetime we can make a discovery that decreases the need for dangerous blood thinning medication and improves the lives of individuals who are at high risk or whom have suffered from a DVT.

Read more from Dr. Obi about her research goals and progress in this blog post from the Michigan Medicine Department of Surgery.

More:
Investigating the Immune System's Connection to Blood Clots | University of Michigan - Michigan Medicine

Read More...

Tips to protect yourself from getting the flu – CBS46 News Atlanta

Monday, January 6th, 2020

'); $("#expandable-weather-block .modal-body #mrd-alert"+ alertCount).append(""+val.title+""); // if (window.location.hostname == "www.kmov.com" || window.location.hostname == "www.kctv5.com" || window.location.hostname == "www.azfamily.com" || window.location.hostname == "www.kptv.com" || window.location.hostname == "www.fox5vegas.com" || window.location.hostname == "www.wfsb.com") { if (val.poly != "" && val.polyimg != "") { $("#expandable-weather-block .modal-body #mrd-alert"+ alertCount).append('"+val.ihtml+""); $("#expandable-weather-block .weather-index-alerts").show(); $("#expandable-weather-block .modal-body h2").css({"font-family":"'Fira Sans', sans-serif", "font-weight":"500", "padding-bottom":"10px"}); $("#expandable-weather-block .modal-body p").css({"font-size":"14px", "line-height":"24px"}); $("#expandable-weather-block .modal-body span.wxalertnum").css({"float":"left", "width":"40px", "height":"40px", "color":"#ffffff", "line-height":"40px", "background-color":"#888888", "border-radius":"40px", "text-align":"center", "margin-right":"12px"}); $("#expandable-weather-block .modal-body b").css("font-size", "18px"); $("#expandable-weather-block .modal-body li").css({"font-size":"14px", "line-height":"18px", "margin-bottom":"10px"}); $("#expandable-weather-block .modal-body ul").css({"margin-bottom":"24px"}); $("#expandable-weather-block .modal-body pre").css({"margin-bottom":"24px"}); $("#expandable-weather-block .modal-body img").css({"width":"100%", "margin-bottom":"20px", "borderWidth":"1px", "border-style":"solid", "border-color":"#aaaaaa"}); $("#expandable-weather-block .modal-body #mrd-alert"+ alertCount).css({"borderWidth":"0", "border-bottom-width":"1px", "border-style":"dashed", "border-color":"#aaaaaa", "padding-bottom":"10px", "margin-bottom":"40px"}); }); } function parseAlertJSON(json) { console.log(json); alertCount = 0; if (Object.keys(json.alerts).length > 0) { $("#mrd-wx-alerts .modal-body ").empty(); } $.each(json.alerts, function(key, val) { alertCount++; $("#mrd-wx-alerts .alert_count").text(alertCount); $("#mrd-wx-alerts .modal-body ").append(''); $("#mrd-wx-alerts .modal-body #mrd-alert"+ alertCount).append(""+val.title+""); // if (window.location.hostname == "www.kmov.com" || window.location.hostname == "www.kctv5.com" || window.location.hostname == "www.azfamily.com" || window.location.hostname == "www.kptv.com" || window.location.hostname == "www.fox5vegas.com" || window.location.hostname == "www.wfsb.com") { if (val.poly != "" && val.polyimg != "") { $("#mrd-wx-alerts .modal-body #mrd-alert"+ alertCount).append(''); } else if (val.fips != "" && val.fipsimg != "") { // $("#mrd-wx-alerts .modal-body #mrd-alert"+ alertCount).append(''); } // } //val.instr = val.instr.replace(/[W_]+/g," "); $("#mrd-wx-alerts .modal-body #mrd-alert"+ alertCount).append(val.dhtml+"

Instruction

See more here:
Tips to protect yourself from getting the flu - CBS46 News Atlanta

Read More...

Trabectedin Targets Leukemic Cells and Restores Immune Cell Function in Models of Chronic Lymphocytic Leukemia – Cancer Therapy Advisor

Monday, January 6th, 2020

The marine-derived compound trabectedin depletes both human primary leukemic cells and myeloid-derived suppressor cells, according to a new study published in Cancer Immunology Research.1 The researchers think their findings could lead to a new therapy that targets both leukemic cells and the protumor microenvironment, repairing the immune dysfunction that is characteristic of chronic lymphocytic leukemia (CLL).

CLLis characterized by lymphocyte accumulation in the blood, bone marrow, andlymphoid tissues.2 Recent advances in CLL therapy have come fromfinding and targeting the appropriate molecular pathways of the disease,explained Kanti R. Rai, MD, a professor of medicine and molecular medicine atthe Donald and Barbara Zucker School of Medicine at Hofstra/Northwell whowasnt involved in the study. Dr Rai said that, for instance, the Brutontyrosine kinase inhibitor ibrutinib binds to the receptor and affects B-cellreceptorsignaling. Another drug, venetoclax, an antagonist to BCL2, can effectivelyinduce apoptosis in CLL cells. However, treatment of this disease remainschallenging due to its immunosuppressive nature. If we [are] to attain a cure,newer compounds have to be identified which have a different mechanism ofcontrolling CLL, he said.

Patientswith CLL have dysfunctional T cells, noted Maria Teresa Bertilaccio, PhD, whois an assistant professor in the department of experimental therapeutics at TheUniversity of Texas MD Anderson Cancer Center in Houston, and the correspondingauthor of the study. Patients [with CLL] have immunosuppression features, sothey might develop an infection because their immune system is not working,she told Cancer Therapy Advisor. Our approach is not only to eradicateleukemia, but also to rearm the immune system to give patients a better qualityof life.

Trabectedintargets tumor-associated macrophages (TAMs); TAMs are thought to support CLLgrowth. A previous study by the Bertilaccio group showed that depleting TAMs byblocking CSF1R signaling reprograms the tumor microenvironment toward anantitumor phenotype.3 This led them to hypothesize that trabectedincould simultaneously target both leukemic cells and nonmalignant cells in thetumor microenvironment.

See the rest here:
Trabectedin Targets Leukemic Cells and Restores Immune Cell Function in Models of Chronic Lymphocytic Leukemia - Cancer Therapy Advisor

Read More...

Yisheng Biopharma and Tavotek Biotherapeutics Announce Strategic Research Alliance for Development of YS-ON-001/002 and Tavo-301/303 Combination…

Monday, January 6th, 2020

GAITHERSBURG, Md. andAMBLER, Pa., Jan. 6, 2020 /PRNewswire/ --Yisheng Biopharma Co., Ltd.("Yisheng Biopharma"), a biopharmaceutical company focusing on research, development, manufacturing, sales and marketing of immunological biologics and vaccines, and Tavotek Biotherapeutics, a biotech company focusing on novel multi-specific antibodies in immuno-oncology and autoimmune diseases, announced today that the companies have entered into a strategic research alliance and collaborate in the development involving their lead assets in oncology. The objective of the alliance is to co-develop a combination therapy of YS-ON-001/002 and Tavo-301/303 for cancer treatment. The two companies are also in discussions regarding additional research and development collaborations beyond oncology.

"We are excited to collaborate with Tavotek, which has a rich pipeline of multi-specific antibodies and advanced technology platforms in oncology and other therapeutic areas," commentedDr. David Shao, President and Chief Executive Officer of Yisheng Biopharma. "As potent agonists of TLR3, MDA5 and RIG-I pathways, YS-ON-001 and YS-ON-002 have demonstrated promising effects in activating the innate and adaptive immune systems and modulating the tumor microenvironment, and YS-ON-001 has shown an excellent safety profile with clinical data to date. By combining YS-ON-001/002 with multi-specific antibodies directed against tumors, we are well positioned in developing a first-in-class immunotherapy with potentially higher response rates. We look forward to working with the Tavotek team to explore the potential synergy of YS-ON-001/002 and Tavo-301/303."

"The collaboration with Yisheng aligns with Tavotek's mission to develop life-changing therapies for patients with significant unmet medical need," saidDr. Mann Fung, Chief Executive Officer of Tavotek. "Current immuno-oncology approaches such as PD1/PDL1 antibody achieve only approximately 20 to 30 percent response rates in clinical settings. A majority of cancer patients still are not seeing a benefit from current therapies. We believe a combination regimen of YS-ON-001/002 with multi-specific IO antibodies, such as Tavo-301/303, will have beneficial impact on cancer care."

YS-ON-001 and YS-ON-002areimmunotherapeutic productsbased on the TLR3/RIG-I/MDA-5 signaling pathways of PIKA immunomodulating technology. They arecapable of both reducing the immunosuppressive effect of tumor microenvironment and enhancing the anti-tumor function of the immune system to tumor cells. YS-ON-001 is currently in clinical development in China and Singapore, and has received orphan drug designations from the U.S. FDA for treatment of pancreatic and liver cancers. The product has been approved in Cambodia for the treatment of advanced solid tumors. YS-ON-002 is a clinical candidate ready for IND submission. YS-ON-001/002 can be an integral immunotherapy component with standard of oncology care, such as chemotherapies, targeted therapies and checkpoint inhibitors or with emerging immunotherapies for additive or synergistic treatment benefits.

Tavo-301/303 is a series of novel multi-specific antibody-based Immuno-Oncology assets that was developed using Tavotek's proprietary TavoSelect Platform. Novel protein engineering employing TavoSelect technology generates multi-specific biologics with optimal molecular profiles for targeting multiple relevant epitopes simultaneously to manage difficult-to-treat solid tumors. These biologics modulate the immunosuppressive pathway by multiple mechanisms of action to promote anti-tumor response and efficacy. The TavoSelec technology also provides biologic molecules with favorable pharmacokinetic profiles and stable formats for ease of development and manufacturing.

About Yisheng Biopharma Co., Ltd.

Yisheng Biopharma is a fully integrated biopharmaceutical company with a global footprint that is discovering, developing and commercializing innovative biotherapeutics for cancer and infectious disease using its novel PIKA immunomodulating technology. PIKA technology augments both innate and adaptive immune responses through the TLR3, RIG-I and MDA5 pathways. Products in clinical development include YS-ON-001 for the treatment of advanced solid tumors, YS-HBV-001 hepatitis B vaccine, and the PIKA rabies vaccine for accelerated protection against rabies infection. The Company has two marketed products in China and South Asia market. Yisheng Biopharma is headquartered in Beijing and has approximately 500 employees in China, U.S., Singapore and other countries in Asia. For more information on Yisheng, please visit http://www.yishengbio.com.

About PIKA Technology

Proprietary PIKA technology originated from research in a class of well-defined synthetic dsRNA molecules. Endosomal dsRNA can be recognized by TLR3 while cytosolic dsRNA can be sensed by the RIG-I-like receptor family which include RIG-I and MDA-5. Through TLR3, RIG-I and MDA-5 signaling, PIKA technology can induce prompt production of interferon, cytokines, chemokines and costimulatory factors. The antiviral and antitumor effects of interferon have been well established. Production of type I interferon upon PIKA administration facilitates antigen cross-presentation by dendritic cells and augments CD4+ T-cell, CD8+ T-cell and natural killer cell responses, which makes PIKA-based therapeutics suitable for both antiviral and antitumor applications.

About Tavotek

Tavotek is a biopharmaceutical company focused on discovering, acquiring, developing, and commercializing therapeutic medicines for patients suffering from debilitating diseases with significant unmet medical need. Tavotek has a rich pipeline of product candidates in various stages of development, focused on cancers, autoimmune conditions, and infectious diseases. For more information, please visit http://www.tavotek.com

About TavoSelectPlatform

The TavoSelect Platform offers highly diversified human antibody sequences that can be utilized in different formats: VHH, scFv, IgG, multi-specific antibodies, fusion proteins, and the engineering of host defense cell surface receptors.TavoSelect of leads emphasize creative screening procedures, NGS analyses, and utilization of proprietary AI software to capture and optimize the best candidates rapidly.The efficiency of the TavoSelect Platform generates novel diverse therapeutic biologic molecules and medical diagnostics for patients.

Yisheng Contact:

Solebury TroutRich Allan (media)Tel: +1 646-378-2958Email: rallan@soleburytrout.com

Bob Ai (investors)Tel +1 646-378-2926Email: bai@soleburytrout.com

Tavotek Contact:

Wei Zhang, Investors and MediaTel: +1 267-405-9426Email: info@tavotek.com

View original content:http://www.prnewswire.com/news-releases/yisheng-biopharma-and-tavotek-biotherapeutics-announce-strategic-research-alliance-for-development-of-ys-on-001002-and-tavo-301303-combination-therapy-for-cancer-treatment-300980756.html

SOURCE Yisheng Biopharma Co., Ltd.

Here is the original post:
Yisheng Biopharma and Tavotek Biotherapeutics Announce Strategic Research Alliance for Development of YS-ON-001/002 and Tavo-301/303 Combination...

Read More...

HOOKIPA’s Gilead Sciences Collaboration for HIV and HBV Therapeutic Vaccines Advancing Towards Clinical Entry – Associated Press

Monday, January 6th, 2020

NEW YORK and VIENNA, Austria, Jan. 06, 2020 (GLOBE NEWSWIRE) -- HOOKIPA Pharma Inc. (NASDAQ: HOOK, HOOKIPA), a company developing a new class of immunotherapeutics targeting infectious diseases and cancers based on its proprietary arenavirus platform, today announced that HOOKIPA has made strong progress in its collaboration with Gilead for novel arenavirus-based therapeutics intended to support functional cures for chronic Hepatitis B virus (HBV) and human immunodeficiency virus (HIV) infections.

HOOKIPA and Gilead Sciences designed and tested multiple arenaviral vectors expressing HIV and HBV immunogens, optimizing each for potential preclinical immunogenicity, safety and manufacturability. In 2019, HOOKIPA earned multiple Gilead milestone payments for the delivery of research vectors and advancing the programs closer to clinical studies. On the basis of promising preclinical data, Gilead has committed to preparations to advance the HBV and HIV vectors toward development, with the HBV development decision triggering an additional milestone payment to HOOKIPA. To enable the development activities and expanded research programs, Gilead has agreed to reserve manufacturing capacity and expanded the HOOKIPA resources allocated to the Gilead collaboration.

About HOOKIPA

HOOKIPA Pharma Inc. (NASDAQ: HOOK) is a clinical stage biopharmaceutical company developing a new class of immunotherapeutics, targeting infectious diseases and cancers based on its proprietary arenavirus platform that is designed to reprogram the bodys immune system.

HOOKIPAs proprietary arenavirus-based technologies, VaxWave*, a replication-deficient viral vector, and TheraT*, a replication-attenuated viral vector, are designed to induce robust antigen specific CD8+ T cells and pathogen-neutralizing antibodies. Both technologies are designed to allow for repeat administration to augment and refresh immune responses. TheraT has the potential to induce CD8+ T cell response levels previously not achieved by other immuno-therapy approaches. HOOKIPAs off-the-shelf viral vectors target dendritic cells in vivo to activate the immune system.

HOOKIPAs VaxWave-based prophylactic Cytomegalovirus vaccine candidate is currently in a Phase 2 clinical trial in patients awaiting kidney transplantation from living Cytomegalovirus-positive donors. To expand its infectious disease portfolio, HOOKIPA has entered into a collaboration and licensing agreement with Gilead Sciences, Inc. to jointly research and develop functional cures for HIV and Hepatitis B infections.

In addition, HOOKIPA is building a proprietary immuno-oncology pipeline by targeting virally mediated cancer antigens, self-antigens and next-generation antigens. The TheraT based lead oncology product candidates, HB-201 and HB-202, are in development for the treatment of Human Papilloma Virus16-positive cancers. The Phase 1/2 clinical trial for HB-201 was initiated in December 2019. The HB-202 IND filing is intended for the first half of 2020.

Find out more about HOOKIPA online at http://www.hookipapharma.com.

*Registered in Europe; Pending in the US.

HOOKIPA Forward Looking StatementsCertain statements set forth in this press release constitute forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995, as amended. Forward-looking statements can be identified by terms such as believes, expects, plans, potential, would or similar expressions and the negative of those terms. Such forward-looking statements involve substantial risks and uncertainties that could cause HOOKIPAs research and clinical development programs, future results, performance or achievements to differ significantly from those expressed or implied by the forward-looking statements. Such risks and uncertainties include, among others, the uncertainties inherent in the drug development process, including HOOKIPAs programs early stage of development, the process of designing and conducting preclinical and clinical trials, the regulatory approval processes, the timing of regulatory filings, the challenges associated with manufacturing drug products, HOOKIPAs ability to successfully establish, protect and defend its intellectual property and other matters that could affect the sufficiency of existing cash to fund operations and HOOKIPAs ability to achieve the milestones under the agreement with Gilead. HOOKIPA undertakes no obligation to update or revise any forward-looking statements. For a further description of the risks and uncertainties that could cause actual results to differ from those expressed in these forward-looking statements, as well as risks relating to the business of the company in general, see HOOKIPAs quarterly report on Form 10-Q for the quarter ended September 30, 2019 which is available on the Security and Exchange Commissions website at http://www.sec.gov and HOOKIPAs website at http://www.hookipapharma.com.

Investors and others should note that we announce material financial information to our investors using our investor relations website ( https://ir.hookipapharma.com/ ), SEC filings, press releases, public conference calls and webcasts. We use these channels, as well as social media, to communicate with our members and the public about our company, our services and other issues. It is possible that the information we post on social media could be deemed to be material information. Therefore, we encourage investors, the media, and others interested in our company to review the information we post on the U.S. social media channels listed on our investor relations website.

For further information, please contact:

Media Investors Nina Waibel Matt Beck Senior Director - Communications Executive Director Investor Relations Nina.Waibel@HookipaPharma.com Matthew.Beck@HookipaPharma.com Media enquiries Ashley Tapp Instinctif Partners Hookipa@Instinctif.com +44 (0)20 7457 2020

Continue reading here:
HOOKIPA's Gilead Sciences Collaboration for HIV and HBV Therapeutic Vaccines Advancing Towards Clinical Entry - Associated Press

Read More...

Forty Seven: Early Indications Suggest Magrolimab Could Be A Winner – Seeking Alpha

Monday, January 6th, 2020

Investment Thesis

Forty Seven (FTSV) management must be pleased with the progress they made in December '19.

First of all, early results from clinical trials of the company's flagship drug magrolimab were positive. In a trial evaluating magrolimab in combination with azacitidine for the treatment of myelodysplastic syndrome ("MDS") and acute myeloid leukaemia ("AML"), Complete Response ("CR") rates of 50% and Overall Response Rates ("ORR") of 92% were observed in untreated patients with higher risk MDS.

In Patients with Untreated AML who are ineligible for induction chemotherapy CR and ORR rates were 55% and 64% respectively. Furthermore, the combination of magrolimab and azacitidine was well tolerated, meaning the treatment may be safe for fragile, sicker and older patients.

The strong results appeared to take the market by surprise and Forty Seven's share price accelerated immediately. The stock gained 84% on 28x average volume in a single day to reach $39 and continued its ascent to reach an all-time high of over $44 (price at the time of writing is a little lower at just over $38).

Secondly, after releasing the results Forty Seven management wisely decided to issue a public offering of 5.59m shares at $35 per share, successfully raising $195.6m. (Source: Bloomberg). Given that the last fundraising, in July of last year, raised $86m at an offer price of just $8, the latest raise must be a cause of satisfaction. It is also a clear indication that investors are starting to see Forty Seven and magrolimab as the frontrunner amongst a plethora of biotechs focused on CD-47 directed therapies, in my view.

The company reported cash, cash equivalent and short-term investments of $166.7m on its Q319 earnings call which includes proceeds from the July raise and a $15.7m upfront licence payment from a collaboration with Ono Pharmaceuticals (Source: Globenewswire) which will see the Japanese firm develop, market and commercialise magrolimab across Japan and the ASEAN region.

Management stated this funding would be sufficient for Forty Seven to support its operations - which include up to ten clinical trials of magrolimab plus pre-clinical trials of anti-SIRPa antibody FSI 189 and anti-cKIT antibody FSI-174 - through to the first quarter of 2021.

Factor in December's raise and we can see that Forty Seven is now in a strong position to pursue and meet its stated goal of being the first company to release an approved therapy targeting the CD47 checkpoint of the innate immune system.

This being biotech, there are many reasons why Forty Seven's best efforts may fall short of winning approval for commercialisation from the FDA. Magrolimab is still in the early stages of being tested and its good results to date will count for nothing should Phase II or III trials reveal safety concerns or a failure to meet the primary endpoint.

Forty Seven does not have a strong pipeline to fall back on should magrolimab ultimately fail to secure commercialisation, meaning investing at this time comes with a high chance of making a loss.

A rival company could produce a CD47-directed treatment that proves to be more effective in which case Fifty Seven will struggle to sell magrolimab even if it is approved. Clinical tests could go on for longer than expected requiring further funding and there is no guarantee the company will be able to raise enough cash. Or, an alternative therapy, such as gene editing or RNAi could outperform all other treatments, rendering the company's development efforts fruitless.

Despite these concerns, however, if I were to pick a CD47 focused immunology company to back today, it would be Forty Seven. With no current concerns on the funding front and with such impressive early trial results from its lead candidate the near-term future certainly looks bright.

There are further reasons for optimism. The company owns exclusive rights to magrolimab which means should the drug be approved Forty Seven will retain the bulk of the profits from its sale. If results continue to impress Forty Seven represents an attractive acquisition target for a big pharma firm. And perhaps most importantly, besides MDS and AML magrolimab has the potential to be approved for numerous indications. Non Hodgkin's Lymphoma, for example, as well as ovarian cancer, colorectal cancer and bladder cancer.

In other words, magrolimab has blockbuster potential, and therefore, despite the obvious risks - one bad trial result could decimate the current share price - in my view Forty Seven should be carefully considered as an investment due to its upside potential. There has not been a new treatment available for Myelodysplastic syndromes ("MDS") in over a decade. Some investors may feel the rewards on offer for a successful treatment are significant enough to justify the risks.

Forty Seven was founded in 2014 in Menlo Park, California by a group of Stanford scientists, most notably Irv Weissman. Weissman played an instrumental role in identifying and developing CD47 as a potential cancer treatment.

Forty Seven went public in June 2018. The company raised $112m at a price of $16 giving it a valuation just shy of $480m. Today, thanks to the recent share price gain, Forty Seven's market cap stands at over $1.5bn.

Forty Seven's lead drug candidate magrolimab is an anti-CD47 antibody formerly known as 5f9. 5f9 has the ability to "switch off" the "don't eat me" signalling pathway used by cancerous cells to avoid detection by macrophages.

Macrophages are the innate immune system's first line of defence against abnormal cells. CD47 is expressed by healthy cells as a means of sending a "don't eat me" signal to macrophages, thereby exempting themselves from a process known as phagocytosis whereby a macrophage consumes abnormal cells to protect the body.

Nearly all cancerous cells over-express CD47 as a means of disguising themselves against macrophages to avoid being swallowed up and eliminated. The "don't eat me" message is sent when the cancerous cell binds to a receptor on macrophages known as SIRP-alpha.

Weissman's research at Stanford demonstrated three things. That blocking the "don't eat me" signalling pathway leads to elimination of many types of tumours and increases a patient's chances of survival. That boosting "eat me" signals found on cancer cells using therapeutic antibodies can work in conjunction with blocking CD47. And that, besides phagocytosis, macrophages activate tumor specific antigens that can activate T-cells against the cancerous cells, meaning that blocking CD47 can also work in conjunction with T-cell based therapies. (Source: FTSV Fundraising prospectus Dec '19)

FTSV 3-fold strategy. Source: FTSV Website

This has led directly to Forty Seven's three pronged development strategy. Monotherapy, e.g. facilitating phagocytosis, synergizing with other tumor targeting antibodies and T-cell activation, and using pro-phagocytic signals on tumor cells in conjunction with chemotherapy.

5F9 is a humanized IgG4 subclass monoclonal antibody that Forty Seven say is designed to combine with a proprietary dosing regimen to help overcome the toxicity limitations of rival anti-CD47 therapies developed by other companies.

Besides 5F9 / magrolimab, Forty Seven are also advancing FSI-189, an anti-SIRPa antibody, and FSI-174, an anti-cKIT antibody. FSI-189 is expected to enter solid-tumor trials this year, whilst cKIT - an antibody targeting stem cell growth factor inhibitors and issuing an "eat me signal" - may prove effective in treating leukemia, melanoma and gastrointestinal stroma tumors.

Forty Seven has 6 clinical trials of magrolimab ongoing that have progressed beyond the pre-clinical stage.

The trial that has produced the most positive results to date (referred to in the introduction of this article) is evaluating magrolimab both as a monotherapy and in conjunction with azacitidine as a treatment for MDS and AML in patients with haemotological malignancies.

Trial investigator David Sallman, M.D., H Lee Moffit Cancer Center and Research Institute had this to say on the publication of the encouraging early data:

The data that continue to emerge from this clinical trial are incredibly exciting, suggesting that the combination of magrolimab and azacitidine may offer the first new therapeutic regimen in over a decade, with the potential to induce meaningful and lasting responses in patients with higher-risk disease. Importantly, these results also support magrolimabs tolerability profile, further differentiating it as a safe treatment that may be used even in more fragile, sicker, and older patients.

Forty Seven have subsequently entered discussions with the FDA with regard to initiating a registration-enabling program with the goal of securing accelerated approval, and also hope to submit a biologics license application ("BLA") in Q421.

MDS is regarded by Forty Seven management as one of its most important treatment targets given its high incidence in the US (as illustrated in the chart below) and due to the paucity of treatment options available to patients.

Source: FTSV Investor Presentation Dec '19

Research suggests that 75% of MDS patients receive only supportive care with the only other options: chemotherapy drugs Vidaza (the brand name version of azacitidine), revlimid and Dacogen, or allogeneic stem cell therapy, being ineffective.

As we can also see from the above chart, diffuse large B-cell lymphoma ("DLBCL") represents another target for Forty Seven and it is the subject of a second planned clinical trial of magrolimab, this time in conjunction with rituximab, a monoclonal antibody that targets a protein known as CD20.

The trial will enrol 100 patients who have failed at least two prior lines of therapy, and will begin, management say, in Q120 with the earliest interim efficacy data slated to be made available in Q420. On the Q319 earnings call Forty Seven CEO Mark McCamish stated his desire to advance into earlier lines of treatment as early as possible referring to a "substantial unmet need" in treatment of DLBCL.

McCamish also updated investors and analysts concerning the phase 1b solid tumor trials. Results from both magrolimab combined with avelumab to treat patients with brain cancer, and in combination with cetuximab in patients with colo-rectal cancer will be made available in meetings scheduled for Q120 with abstracts of early data having already been submitted.

McCamish also announced a collaboration with gene therapy specialists Bluebird Bio. Forty Seven intend to leverage Bluebird's LentiGlobin platform to evaluate FSI-174 and move forward the cKIT program with a focus on pretransplantation and avoiding the need for chemotherapy or radiation toxicities or secondary malignancies when performing stem cell transplants.

Targeting blood-forming stem cells that express cKIT with FSI-174 releases macrophages to clear the steam cells, and, used in conjunction with magrolimab could, McCamish says:

massively expand the number of patients eligible for transplantation and therefore enable many more people to benefit from the curative potential transplantation.

During Q3 Forty Seven had an R&D spend of $27.1m - up from $18m in Q318, ascribed to the advancement of clinical trials and contract manufacturing costs for the proposed BLA.

In total, the company made a loss of $15.1m, down from $21.7m the previous year. In the first nine months of 2019 losses totalled $61.2m. As mentioned previously, Forty Seven should have more than enough funding to complete its trials and submit the BLA without having to dilute investors further - but it would be wise not to rule anything out. One failed trial could set the whole process back by years. (Source: FTSV 10Q Submission Q319).

There is no doubt that Forty Seven faces stiff competition. Amongst the companies competing in the CD47 antibody space are Surf therapeutics (SURF), Trillium Therapeutics (TRIL), Celgene (CELG), China based biotech Innovent Biologics, and Netherlands Based Aurigene and Synthon. (Source: PM Live)

All are worth studying in more detail and both SURF and TRIL represent a far cheaper investment opportunity, with shares priced at just $1.94 and $1.26 respectively. Neither have experienced a "Forty Seven moment", delivering outstanding results from early stage trials, but the price of Trillium recently spiked as Morgan Stanley reported a 5% holding. (Source: Benzinga)

For my money, however, Forty Seven is the frontrunner, and although it is priced at a premium to some of its rivals, there are good reasons for this, as I have discussed above. Another reason Forty Seven is at a competitive advantage is the 187+ patents it owns protecting magrolimab and FSI-189.

Additionally, although it was painful at the time, in 2018 Forty Seven agreed to make $47m of milestone payments to Synthon to secure non-exclusive rights to several CD-47- and SIRPa- directed antibodies, including rituximab. Other companies will need to make similar agreements if they want to develop their drugs with the same freedom that Forty Seven now has. Further analysis can be found in this informative recent SA article.

The average analyst price target for Forty Seven at time of writing is $39.25 (Source: Nasdaq) with a high of $48 and a low of $35 with the majority of analysts issuing "buy" ratings for the stock.

In my view, provided trial results remain positive, each new development can move the share price higher. Given the size of the addressable market (the global market for MDS treatment alone is set to reach $2.4bn by 2022, at a CAGR of 9.7% according to research from Grand View) and the urgent need for new and better treatments for diseases such as NHL, MDS, AML and DLBCL, the potential upside here is substantial.

If Forty Seven were to perform as well as, for example, gene-silencing treatment developer Arrowhead (ARWR) has done in 2019, buoyed by positive data, the share price could easily double as Arrowhead's has done. That is a big "if", however.

Biotech investing is inherently risky and it is all too easy to get sucked into a "next big thing" such as CD47 antibodies. In Forty Seven's case, however, the exciting premise is backed by years of research and real clinical data. Importantly, the FDA has issued Forty Seven with accelerated approval status both for magrolimab as a treatment for MDS, AML and DLBCL, as well as follicular lymphoma.

The company has treated over 190 relapsed or refractory cancer patients with magrolimab and will shortly enter a pivotal phase III trial, ENHANCE, enrolling 90 new patients to evaluate the combination of azacitidine and magrolimab together, plus it has the BLA scheduled for submission before the end of 2021.

The management team are experienced with big pharma backgrounds including Abbott Laboratories, Amgen, Genentech, Gilead, Janssen Global Services, LLC, PDL Biopharma, Inc. and Sandoz Inc.

Furthermore, Forty Seven has agreed collaborations with big pharma companies Merck and Genentech, a subsidiary of the Roche group to explore opportunities within ovarian and bladder cancer.

On balance, I think there are enough positive signals to make Forty Seven are worthwhile, if speculative investment.

Disclosure: I/we have no positions in any stocks mentioned, and no plans to initiate any positions within the next 72 hours. I wrote this article myself, and it expresses my own opinions. I am not receiving compensation for it (other than from Seeking Alpha). I have no business relationship with any company whose stock is mentioned in this article.

Originally posted here:
Forty Seven: Early Indications Suggest Magrolimab Could Be A Winner - Seeking Alpha

Read More...

Inflammation is body’s response to a threat – NewmarketToday.ca

Monday, January 6th, 2020

Hi Nonie,I consistently follow your articles and I havea follow-up question about a recent article. You state, Systemic inflammation can almost always be traced back at least in good part to malnutrition or toxicity related to dietary imbalances. Inflammation is an issue I deal with as I have Lupus, arthritis and sinusitis. Could you elaborate further on areas of malnutrition that contribute to inflammation? Would you also be more specific in regards to toxicity? What should I be looking for or doing to fix these two possible issues? And lastly, is there anywhere you can recommend to have a reliable food intolerance test?Thanks,Confused

Dear Confused,

Thank you for your question, and for reading regularly.With your specific conditions Lupus, arthritis (RA?), and sinusitis you do need to address inflammation on a systemic level. Each of these conditions is inherently inflammatory in nature. Doing so will be the cornerstone to improving your health.

Yours is a very complex health picture and obviously outside of the scope of my column to address fully, but holistic treatment should include addressing gut dysbiosis and permeability, which is certainly a factor in your inflammation. Ill try to unpack this a bit, as it relates to toxins and malnutrition. But first I want to clarify the meaning of malnutrition.

The root mal means disordered (Latin) or badly (le francais). We tend to take malnutrition to mean a lack of proper nutrition from not getting enough to eat. But it can also mean a lack of nutrition from not eating enough of the right things or from not getting adequate nutrients from the food one does eat. I will explain how this is relevant.

Healthy intestines dont just absorb nutrients and channel waste, their mucosa and structure also act as a barrier to pathogens and an informant to the immune system. When theres permeability in this barrier (think of it as tiny holes or gaps in a tube shaped, tightly woven screen covered in little finger like projections) food, various toxins, and microorganisms weve been exposed to escape or leak into the bloodstream to circulate. This is not supposed to happen in a healthy body!

In a healthy body, only beneficial, fully digested nutrients are given entry into the bloodstream by specific transport molecules via approved pathways to strategic sites where they are unloaded and used for specific manufacturing purposes. And all the dangerous things are locked inside the tube and swept along to be excreted. So foreign molecules (even nutrients) arent supposed to get into our bloodstream indiscriminately.

But our bodies are wonderfully designed! Our immune systems are set up to tag antigens on the surface of all potentially harmful substances that get into our bloodstream, creating antibodies to them forevermore so once we have come into contact with an offender, we are more quickly able to identify and remove it in the future. Antibodies are a signal to obliterate: a sort of seek and destroy function, if you will. Toxins, viruses, fungi, bacteria, chemicals, drugs, foreign debris, and proteins all have surface antigens that identify them.

So do many of our own cells, it turns out. But with an immune system that is functioning at normal levels, these are clearly recognized.

In a healthy body the antigens are limited and of a specific duration under normal circumstances (a flu virus, for example), after which the body recuperates and the immune response dies down. But, in the case of a leaky gut, the invaders are continual every time anything is passing through the gut there is a slow leak of antigens from various sources. The immune system goes to work tagging them all as a threat - even proteins we need for survival because they are escaping in an unrecognized form. Now theyre seen as offenders and a food reaction (sensitivity) is borne. Of course, the response is system-wide because the gaps allow these antigens directly into the bloodstream to circulate around.

Now, the immune system response doesnt stop there. Its super sophisticated.It has a backup to that backup, called inflammation. When the body gets the memo that the seek-and-destroy mission isnt doing the job and there are still circulating offenders getting in like gangbusters, it mounts an inflammatory response that is sort of like a system lockdown.

Inflammation can be localized when a tissue is damaged, for example, and we get swelling, heat, redness, and pain as chemicals are released to bring blood to the area to facilitate healing and clean up. Or inflammation can be non-specific and systemic, depending on the perceived threat.

Systemic inflammation looks like this:

Seek and destroy botched it again.Not surprising. Snort. Okay, lets get this done right. Initiate operation overkill.Heat or fever - lets torch these guys.Mucous - lets trap these guys.Tissue swelling - lets lock these guys in.Call in the Mr. Ts. (T-cells) - lets attack these guys.Okay, but were running low on energy.Right. Shut down the brain and draw all the energy reserves.

For someone with Lupus, these symptoms will sound very familiar.

The current medical paradigm is that systemic inflammation is a response in the body that doesnt understand there is no threat a spontaneously overactive or delusional immune system, if you will. In this paradigm its recognized that people with chronic systemic inflammation are more likely to develop autoimmune disease, although its not really understood how. Its perceived that the body just randomly attacks its own tissues. And these people, in turn, develop multiple chemical and food sensitivities, by unknown mechanisms.

But what if the leaky gut sets all this in motion? When antigens are appearing several times a day the immune system naturally goes on high alert. This isnt a dysfunctional immune system its a smart one. And when it does this, it has to become hypervigilant in attacking antigens. Since the bodys cells have their own antigens, its easy to see how they could get accidentally tagged in sites where thereis a perceived threat.

When quality control in any factory isdealing with threeor four timesthe normal maximum capacity, there are mistakes. And in this factory, tags are a forever thing. Any food with any protein component that escapes into the bloodstream via this mechanism would also be tagged and become something the body reacts to. Ditto chemicals. When this is happening daily, of course the backup inflammatory system is going to kick in to try to protect against the perceived invasion.

BOOM! Continual systemic inflammation with multiple food and chemical intolerances and an immune system on overdrive that is now busy attacking not only outside offenders, but itself. This is a holistic understanding of autoimmune disease. And it all starts in the gut.

And leaky gut is often linked to gluten intolerance. With your symptom set I feel safe saying its likely that youre gluten intolerant, if not celiac. I would recommend testing for celiac immediately. If that does not return a positive test, I would recommend non-celiac gluten sensitivity testing. You can get antibody, saliva and blood testing, but they are not always accurate in the absence of symptomatology and a knowledgeable practitioner. Its still new territory. I prefer my food intolerance test, antibody testing, and intestinal permeability testing, working collaboratively with your physician. In complex cases its always best to have a collaborative approach if you can.

Why do I feel you are gluten intolerant? Gluten, in particular, damages the gut lining in sensitive individuals. It burns the microvilli and causes gaps in the junctions of the intestinal lining. Remember, these gaps are how antigens leak into the bloodstream to initiate and fuel the immune system mayhem.

Undiagnosed non-celiac gluten sensitivity has been shown to masquerade as Lupus.

As for toxins, mycotoxins (molds), parasites, candida, bacteria from root canals, heavy metals, synthetic fragrances, chemical cleaners, chemical body care products, and environmental toxins should all be considered and eliminated. These can precipitate the problem or just add to it. Remember, if there are regular antigens the immune system mayhem starts. Regular antigens can come from a food intolerance (gluten), from environmental toxins (mould), from endogenous toxins (bacterial imbalance or candida), or from any number of sources. So if you are gluten intolerant, that is also acting as a toxin.

The person with an autoimmune disease will, as a result of all this, have an overburdened liver because it is trying to deal with all these toxins -and this means any subsequent toxin will have an exaggerated impact compared to the impact they would have on a healthy host. Something as benign as perfume can even trigger migraines or seizures in such people. Their livers just cant handle any more! Discerning and eliminating these toxins can make a big impact on the burden of the body. Removing common toxins like sugar, alcohol, drugs, cigarettes, and processed foods are also imperative for managing symptoms and discomfort.

It may be helpful to think of toxicity as anything that adds to the burden of the body. It only makes sense that if the burden is increased chronically, the body is going to need copious nutrients to do that work. Unfortunately, when we are chronically ill the first thing to go is often our diet. We eat convenience food instead of quality, nutrient dense foods. These foods are full of toxic chemicals and additives. Too, many healthy molecules are mistakenly tagged as toxins (above). The body struggles in the presence of so many substances to protect itself from and is overwhelmed by the burden of it.

But how does malnutrition play in? When the digestive system cant contain nutrients in the lumen until they are properly broken down and delivered via appropriate pathways, deficiencies develop. At the same time, the body requires more nutrients than normal to sustain its protective detail. These deficiencies, in turn, make it difficult for tissues (like the damaged gut tissue) to self repair. This happens even when a person is eating well - because the food is not being absorbed properly. This is why I say malnutrition plays a large role.

As such, I believe healing the gut and adding strategic supplements to a nutrient dense, easy to digest diet that identifies and avoids or rotates intolerances is essential in starting to address chronic inflammation and autoimmunity.

I hope this helps clarify the role of inflammation, toxicity, and deficiency, particular to your concerns, and of food intolerances. I hope it gives you some clear actions you can take to start to improve your health. If you want further guidance you can come see me in my clinic. I offer comprehensive testing for food intolerances and gut biome health, which would tell us a lot about the state of your digestive system to create a strategic plan. At the very least, I hope you are less confused.

As always, if readers have their own questions I encourage them to write to me at nonienutritionista@gmail.com. For more health news, recipes, and nutrition related events in the community, readers can go to my website at nonienutritonista.com and sign up for the newsletter. Its nutritionist certified and 100 per cent gluten and sugar free!

Namaste!Nonie Nutritionista

Nonie De Long is a registered orthomolecular nutritionist with a clinic in Bradford West Gwillimbury, where she offers holistic, integrative health care for physical and mental health issues. Check out her website here.

Do you have a question about health and wellness? Emailnonienutritionista@gmail.com

Read more:
Inflammation is body's response to a threat - NewmarketToday.ca

Read More...

Magenta Therapeutics to Present at the 38th Annual J.P. Morgan Healthcare Conference on Wednesday, January 15th in San Francisco – Business Wire

Monday, January 6th, 2020

CAMBRIDGE, Mass.--(BUSINESS WIRE)--Magenta Therapeutics (NASDAQ: MGTA), a clinical-stage biotechnology company developing novel medicines to bring the curative power of immune reset to more patients, today announced that the company is scheduled to present at the 38th Annual J.P. Morgan Healthcare Conference in San Francisco on Wednesday, January 15th, 2020 at 11:30 a.m. PT (2:30 p.m. ET), immediately followed by a Q&A session.

A live webcast of the presentation and Q&A session can be accessed on the Magenta Therapeutics website at https://investor.magentatx.com/events-and-presentations. The webcast replay will be available for 90 days following the event.

About Magenta Therapeutics

Magenta Therapeutics is a clinical-stage biotechnology company developing medicines to bring the curative power of immune system reset through stem cell transplant to more patients with autoimmune diseases, genetic diseases and blood cancers. Magenta is combining leadership in stem cell biology and biotherapeutics development with clinical and regulatory expertise, a unique business model and broad networks in the stem cell transplant world to revolutionize immune reset for more patients.

Magenta is based in Cambridge, Mass. For more information, please visit http://www.magentatx.com.

Follow Magenta on Twitter: @magentatx.

Forward-Looking Statement

This press release may contain forward-looking statements and information within the meaning of The Private Securities Litigation Reform Act of 1995 and other federal securities laws. The use of words such as may, will, could, should, expects, intends, plans, anticipates, believes, estimates, predicts, projects, seeks, endeavor, potential, continue or the negative of such words or other similar expressions can be used to identify forward-looking statements. The express or implied forward-looking statements included in this press release are only predictions and are subject to a number of risks, uncertainties and assumptions, including, without limitation risks set forth under the caption Risk Factors in Magentas Registration Statement on Form S-1, as updated by Magentas most recent Quarterly Report on Form 10-Q and its other filings with the Securities and Exchange Commission. In light of these risks, uncertainties and assumptions, the forward-looking events and circumstances discussed in this press release may not occur and actual results could differ materially and adversely from those anticipated or implied in the forward-looking statements. You should not rely upon forward-looking statements as predictions of future events. Although Magenta believes that the expectations reflected in the forward-looking statements are reasonable, it cannot guarantee that the future results, levels of activity, performance or events and circumstances reflected in the forward-looking statements will be achieved or occur. Moreover, except as required by law, neither Magenta nor any other person assumes responsibility for the accuracy and completeness of the forward-looking statements included in this press release. Any forward-looking statement included in this press release speaks only as of the date on which it was made. We undertake no obligation to publicly update or revise any forward-looking statement, whether as a result of new information, future events or otherwise, except as required by law.

Read more from the original source:
Magenta Therapeutics to Present at the 38th Annual J.P. Morgan Healthcare Conference on Wednesday, January 15th in San Francisco - Business Wire

Read More...

Cellectis: An Expert Review on Allogeneic CAR-T for Cancer Published in Nature Reviews Drug Discovery – Yahoo Finance

Monday, January 6th, 2020

Cellectis and World Experts Review New Avenue of Allogeneic CAR T-cells, Optimization and Promises in Oncology

Cellectis (Paris:ALCLS) (NASDAQ:CLLS) (Euronext Growth:ALCLS; Nasdaq:CLLS), a biopharmaceutical company focused on developing immunotherapies based on gene-edited allogeneic CAR T-cells (UCART), announced today the publication of a review in Nature Reviews Drug Discovery by Prof. Stphane Depil1*, Dr. Philippe Duchateau2, Prof. Stephan Grupp3, Prof. Ghulam Mufti4 and Dr. Laurent Poirot2. The authors review the opportunities and challenges presented by universal allogeneic CAR T-cell therapies.

One of the most promising approaches in cancer treatment is chimeric antigen receptor (CAR) T-cell therapy, in which part of the bodys own immunological defendors, T-cells, are redirected against cancerous cells after being engineered to express CARs. Since their initial development in the early 90s, CAR T-cells have evolved through several generations. The use of autologous (patient-derived) CAR T-cells has proven to be successful in treating people with certain blood cancers such as B-cell malignancies. However, autologous CAR T-cell therapy is not suitable for all patients, and it often requires a long and expensive manufacturing process since each treatment must be made individually for each patient.

Cellectis was the first company to develop and test an allogeneic CAR T-cell therapy in patients, where T-cells are derived from healthy donors. This gives rise to off-the-shelf product candidates which aim to be suitable for many patients as opposed to only a single person.

"We realized early on that refined gene-editing techniques were what was needed to take an allogeneic approach to CAR T-cell therapy," said Dr. Laurent Poirot, VP, Immunology Division, Cellectis. "Despite the complexity of this approach, we decided to follow this route because we are confident that it can provide the most impact for a maximum number of people living with severe cancers. This comprehensive review underlines just how much this technology has evolved in very little time. It also gives us exciting areas to explore as we continue to improve our product candidates."

One of the major challenges in the allogeneic approach involves mitigating the risk of graft-versus-host-disease (GvHD) a medical complication that can present itself in people that have received tissues or cells from another person. The review examines aspects of this challenge and helps weigh the pros and cons associated with the different methods used to create allogeneic CAR T-cells. It also outlines some of the gene-editing work that Cellectis has done in this area along with complementary approaches being taken by others in the field, such as using cells other than conventional T-cells, also known as alpha beta T-cells.

"Our immune system, including our T-cells, is incredibly sophisticated. We know that T-cells can now be retasked to successfully fight cancer. There are amazing approaches to gene editing that are driving progress towards the most safe and efficacious versions of allogeneic products. It is exciting to see these approaches applied to off the shelf CAR T-cell products," said Prof. Stephan Grupp, Chief of Cell Therapy and Transplant Section at the Childrens Hospital of Philadelphia, Professor of Pediatrics at the Perelman School of Medicine, and a member of Cellectis Clinical Advisory Board. "Im looking forward to seeing emerging clinical data as well as even newer approaches, as Cellectis expertise in gene-editing technology continues to transform CAR-T."

Off-the-shelf allogeneic CAR T cells: new development and current challenges

Stphane Depil1*, Philippe Duchateau2, Stephan Grupp3, Ghulam Mufti4, Laurent Poirot2

1Formerly Cellectis, now Centre Lon Brard and Centre de Recherche en Cancrologie de Lyon, 28 rue Laennec, 69008 Lyon, France2Cellectis, 8 rue de la Croix Jarry, 75013, Paris, France3Childrens Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, 3401 Civic Center Blvd Philadelphia, PA 10104, USA4Kings College London and Kings College Hospital, Denmark Hill, London, SE5 9RS, United Kingdom

About CellectisCellectis is developing the first of its kind allogeneic approach for CAR-T therapies, pioneering the concept of off-the-shelf and ready-to-use gene-edited CAR-T cells to treat patients. As a clinical-stage biopharmaceutical company with over 20 years of expertise in gene editing, we are developing game-changer product candidates in immune-oncology. Utilizing TALEN, our gene editing technology, and PulseAgile, our pioneering electroporation system, we are harnessing the power of the immune system to target and eradicate cancer cells.

Story continues

As part of our commitment to a cure, Cellectis remains dedicated to its goal of providing life-saving UCART product candidates to address unmet need for multiple cancers including B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphoma (NHL) and multiple myeloma (MM). Cellectis is listed on the Nasdaq (ticker: CLLS) and on Euronext Growth (ticker: ALCLS).

Cellectis headquarters are in Paris, France, with additional locations in New York, New York and Raleigh, North Carolina. For more information, visit http://www.cellectis.com.

Follow Cellectis on social media: @Cellectis, LinkedIn and YouTube.

TALEN is a registered trademark owned by Cellectis.

DisclaimerThis press release contains "forward-looking" statements that are based on our managements current expectations and assumptions and on information currently available to management. Forward-looking statements involve known and unknown risks, uncertainties and other factors that may cause our actual results, performance or achievements to be materially different from any future results, performance or achievements expressed or implied by the forward-looking statements. Further information on the risk factors that may affect company business and financial performance is included in Cellectis Annual Report on Form 20-F and the financial report (including the management report) for the year ended December 31, 2018 and subsequent filings Cellectis makes with the Securities Exchange Commission from time to time. Except as required by law, we assume no obligation to update these forward-looking statements publicly, or to update the reasons why actual results could differ materially from those anticipated in the forward-looking statements, even if new information becomes available in the future.

View source version on businesswire.com: https://www.businesswire.com/news/home/20200106005969/en/

Contacts

Media:Jennifer Moore, VP of Communications, 917-580-1088, media@cellectis.comCaitlin Kasunich, KCSA Strategic Communications, 212-896-1241, ckasunich@kcsa.com

IR:Simon Harnest, VP of Corporate Strategy and Finance, 646-385-9008, simon.harnest@cellectis.com

More:
Cellectis: An Expert Review on Allogeneic CAR-T for Cancer Published in Nature Reviews Drug Discovery - Yahoo Finance

Read More...

Human T cell response to CD1a and contact dermatitis allergens in botanical extracts and commercial skin care products – Science

Monday, January 6th, 2020

Oily skin allergens hole up inside CD1a

Contact dermatitis induced by allergens in personal care products is a common cause of skin rashes, but the molecular mechanisms leading to T cell activation are poorly understood. Nicolai et al. tested known contact allergens for their ability to boost IFN- production by human T cells autoreactive to the CD1a antigen presentation molecule. Several hydrophobic chemicals came up as hits, including farnesol, a compound often used as a fragrance. Structural analysis of CD1a-farnesol complexes revealed that farnesol is buried deep within CD1as antigen-binding cleft beyond the reach of T cell receptor chains. These findings suggest that several hydrophobic contact allergens elicit T cellmediated hypersensitivity reactions through displacement of self-lipids normally bound to CD1a, thereby exposing T cellstimulatory surface regions of CD1a that are normally hidden.

During industrialization, humans have been exposed to increasing numbers of foreign chemicals. Failure of the immune system to tolerate drugs, cosmetics, and other skin products causes allergic contact dermatitis, a T cellmediated disease with rising prevalence. Models of T cell response emphasize T cell receptor (TCR) contact with peptide-MHC complexes, but this model cannot readily explain activation by most contact dermatitis allergens, which are nonpeptidic molecules. We tested whether CD1a, an abundant MHC Ilike protein in human skin, mediates contact allergen recognition. Using CD1a-autoreactive human T cell clones to screen clinically important allergens present in skin patch testing kits, we identified responses to balsam of Peru, a tree oil widely used in cosmetics and toothpaste. Additional purification identified benzyl benzoate and benzyl cinnamate as antigenic compounds within balsam of Peru. Screening of structurally related compounds revealed additional stimulants of CD1a-restricted T cells, including farnesol and coenzyme Q2. Certain general chemical features controlled response: small size, extreme hydrophobicity, and chemical constraint from rings and unsaturations. Unlike lipid antigens that protrude to form epitopes and contact TCRs, the small size of farnesol allows sequestration deeply within CD1a, where it displaces self-lipids and unmasks the CD1a surface. These studies identify molecular connections between CD1a and hypersensitivity to consumer products, defining a mechanism that could plausibly explain the many known T cell responses to oily substances.

The human immune system evolved to respond to foreign microbial antigens but must also tolerate foreign compounds present in the environment, such as plants and foods. Over the past two centuries, industrialization has introduced the widespread use of chemical extraction techniques and synthetic chemistry methods. Industrial development has greatly increased the range of synthetic or purified botanical compounds to which humans are commonly exposed through pollution or the intentional use of drugs, fragrances, cosmetics, and other consumer products, especially those applied at high concentrations directly on the skin. Accordingly, the incidence of contact dermatitis has risen, especially in industrialized countries (1). Lifetime incidence currently exceeds 50%, making contact dermatitis the most common occupational skin disease (2). The essential pathophysiological feature of contact dermatitis is the allergen-specific nature of immune hypersensitivity reactions. Diagnosis relies on identifying the specific allergens to which a patient was exposed. Physicians measure local skin inflammation to a grid network of allergen patches applied to the skin as a diagnostic test. The mainstay of treatment is avoidance of exposure to named allergens.

Considerable evidence documents a role for T cells in contact dermatitis, which is caused by delayed-type hypersensitivity reactions. Gell and Coombs (3) defined type IV reactions as delayed-type hypersensitivity because they appear after 72 hours. Type IV reactions are T cell mediated and are worsened after repeated exposure to allergens. During the sensitization phase, naive T cells are activated in a process that involves Langerhans cells and dermal dendritic cells (2). In the elicitation phase, T cells cause inflammatory manifestations in the skin. Biologists views of T cell response are strongly influenced by the known mechanisms by which T cell receptors (TCRs) recognize peptide antigens bound to major histocompatibility complex I (MHC I) and MHC II proteins (46). Yet, most known contact allergens are nonpeptidic small molecules, cations, or metals that are typically delivered to skin as drugs, oils, cosmetics, skin creams, or fragrances (1, 2). Thus, the chemical nature of contact allergens does not match the chemical structures of most antigens commonly recognized within the TCR-peptideMHC axis.

This apparent disconnect, which represents a core question regarding the origin of delayed-type hypersensitivity, might be explained if MHC proteins use atypical binding interactions to display nonpeptidic antigens to TCRs. For example, the antiretroviral drug abacavir binds within the human leucocyte antigen (HLA)B*57:01 groove to alter the seating of self-peptides, creating neo-self epitopes (7). Similarly, the MHC class II protein encoded by HLA-DP2 can bind beryllium, thereby plausibly altering the MHC-peptide complex shape to enable binding of an autoreactive TCR (8). Here, autoimmune response to nonpeptidic compounds still involves peptides in some way and is linked to a specific HLA allomorph that uses a defined structural mechanism. A second general model is that nonpeptidic allergens form covalent bonds with peptides in vivo. Such haptenation reactions might create hybrid molecules with peptide-based MHC binding moieties and TCR epitopes formed from the haptenizing drug or chemical. This concept derived from Landsteiners landmark studies with 2,4-dinitrophenols (9) and evolved into broader predictions that drugs could haptenate peptides or innate receptors (10). Some evidence indicates that drugs can generate immune hypersensitivity reactions via haptenation. For example, sulfamethoxazole, lidocaine, penicillins, lamotrigine, carbamazepine, p-phenylenediamine, or gadolinium can bind peptides, MHC proteins, or TCRs (1116). Although the haptenation hypothesis is broadly taught to physicians, the extent to which it accounts for the larger spectrum of contact allergens remains unknown (17).

Both of these models derive from the premise that T cell responses are mediated by MHC-encoded proteins and emphasize atypical modes of peptide presentation. Putting aside this premise, we tested a straightforward model whereby drugs and other nonpeptidic contact allergens are presented by a system that evolved to present nonpeptidic antigens to T cells (18). CD1 proteins are MHC Ilike molecules that fold to form an antigen binding cleft composed of two pockets, A and F, which are larger and more hydrophobic than the clefts present in MHC I and MHC II proteins (19, 20). Most published studies of human CD1 proteins (CD1a, CD1b, CD1c, and CD1d) emphasize display of amphipathic membrane phospholipids and sphingolipids. The alkyl chains bind within and fill up the cleft of CD1, and the polar head groups, composed of carbohydrates or phosphate esters, protrude through a small portal (F portal) to lie on the outer surface of CD1, where they are presented to TCRs (21).

Whereas most known CD1-presented antigens are amphipathic lipids, some evidence suggests that CD1 proteins mediate recognition of nonlipidic, drug-like molecules. For example, CD1d mediates T cell response to phenyl pentamethyldihydrobenzofuran sulfonates (PPBFs) (22), and chemically reactive small molecules can influence CD1-restricted T cell response by an unknown mechanism that might involve induced lipid autoantigen synthesis (23). PPBFs lack aliphatic hydrocarbon chains that define lipids, and they are instead ringed, sulfated small molecules that chemically resemble allergenic drugs, such as sulfonamide antibiotics and furosemide. However, PPBF antigens are much smaller than the known volume of CD1d cleft. Unlike amphipathic lipids, they lack a defined lipid anchor and hydrophilic head group (22), raising questions about how PPBFs could bind within CD1d and yet protrude in some way for TCR contact.

Among human CD1 isoforms, we focused on CD1a because it is abundantly expressed on epidermal Langerhans cells and dermal dendritic cells, which are implicated in contact dermatitis (24). In addition, CD1a-autoreactive T cells home to the skin, and polyclonal autoreactive T cells derived from blood and skin show higher responses to CD1a as compared with other CD1 proteins (25, 26). In addition, surface CD1a proteins can rapidly capture extracellular antigens using mechanisms that do not require complex mechanisms of antigen processing within the endosomal network (27, 28). Recently, transfer of the human CD1a gene into mice (29) was found to augment intradermal T cell responses to the natural, plant-derived compound, urushiol (30). Actual CD1a-mediated T cell responses to commonly used drugs or contact allergens in consumer goods are, to our knowledge, unknown.

As a screen for the most common and clinically important contact dermatitis antigens, we tested for human T cell response to compounds embedded in the thin-layer rapid use epicutaneous (T.R.U.E.) test (or Truetest), which is broadly used in dermatology and allergy clinics to screen patients for contact dermatitis allergens that are most commonly encountered in medical practice. This approach identified a human T cell response to a tree oilderived contact allergen known as balsam of Peru. Larger-scale screens defined the general chemical requirements for a T cell response to oily substances and discovered additional contact allergens presented by CD1a, including farnesol. The crystal structure of the CD1a-farnesol complex and study of the self-lipids bound to CD1a provided evidence for a molecular mechanism for recognition of a contact allergen, explaining how small antigens sequestered fully within CD1a can lead to T cell responses through the absence of interference with CD1a-TCR contact.

To determine whether CD1a can present contact allergens to T cells, we initially used the CD1a-restricted T cell line known as BC2 for testing response to the T.R.U.E. test panel 1 (Truetest 1) (fig. S1). BC2 is a T cell line derived from peripheral blood T cells of a blood bank donor and has previously been shown to be activated by CD1a loaded with small hydrophobic self-lipids (31). Normally, the Truetest panel consists of compounds arrayed on sterile matrix, which is placed on patient skin. Localized erythema occurring in vivo on skin 2 to 5 days after exposure is considered a positive test, allowing allergen identification based on position in the grid. For testing in vitro, individual allergen patches and untreated patch matrix (control patch) were cut apart with sterile technique. Patches were soaked in media and removed (soaking method) or inserted into wells to contact (contact method) CD1a-transfected K562 (K562-CD1a) antigen-presenting cells (APCs). We saw a modest response to K562-CD1a in the absence of added patch material using interferon- (IFN-) enzyme-linked immunosorbent assay (ELISA), as expected on the basis of the known CD1a autoreactivity of the BC2 T cell line (fig. S1A).

Compared with the control patch, most of the antigen-containing patches, including nickel, potassium dichromate, colophony, lanolin, and paraben, showed no effect. A combination of molecules known as fragrance mix 1 showed slight suppression of cytokine release, consistent with toxicity to cells (fig. S1A). Cobalt, neomycin, and ethylenediamine dihydrochloride showed small increases in IFN- at some doses tested but not reproducibly in subsequent assays. In contrast, balsam of Peru showed a significant response above background (fig. S1A), which also repeated in subsequent assays (fig. S1B and Fig. 1A). Response to balsam of Peru was not seen with patch soaking (fig. S1B), indicating that the stimulatory factor(s) was not physically released from the patch. Overall, the screen suggested a T cell response to balsam of Peru embedded in Truetest patches, leading to focused studies of this natural botanical extract.

(A to E) T cell lines with CD1a autoreactivity (BC2 and Bgp) or foreign antigen reactivity (CD8-2) were tested for activation to lipids using IFN- ELISA in cellular assays with CD1a-transfected K562 cells (K562-CD1a) or mock-transfected K562 cells (K562-mock) (A, B, and E) or on streptavidin plates coated with biotinylated CD1 proteins (C and D). Data are representative of three or more experiments each with the mean of triplicate measurements shown with SD. The significance of lipid concentration on IFN- release was tested by one-way ANOVA (A and C). Relevant pairwise comparisons were tested using Welchs t test (B). Post hoc comparison of marginal means after adjustment by the Sidak method was used to group treatments at the specified significance level after a significant result by two-way ANOVA (D). Post hoc comparison by least squares means after adjustment by the Sidak method was used to group treatments with nonoverlapping marginal means and 95% confidence levels into a, b, or c at the specified significance level after a significant result by two-way ANOVA (E). IgG, immunoglobulin G.

Balsam of Peru is a resin from the South American tree Myroxylon balsamum, which has a vanilla scent and is used as a fragrance and flavor in many personal care products such as skin creams and toothpaste. Balsam of Peru is a common contact allergen seen in medical practice, where it causes severe skin rash in allergic individuals (32, 33). We tested balsam of Peru extract and oily substances derived therefrom, which is known as balsam of Peru oil. Both preparations are commonly used in consumer products. BC2 T cells were activated by both preparations, establishing a T cell dose response to a common botanical extract used in consumer goods (Fig. 1A).

Given the unusual chemical nature of oily substances found in Balsam of Peru oil, we considered candidate mechanisms of T cell activation other than antigen display by CD1a. In theory, compounds might undergo peptide haptenation reactions for presentation by MHC proteins, but this possibility was less favored because K562 cells express very low or undetectable MHC I and MHC II (25). Oily mixtures might influence cellular lipid production (23) or contain mitogens that cross-link CD3 complexes or broadly activate lymphocytes via TCR-independent mechanisms (34). To determine the cellular and molecular mechanisms of T cell stimulation, we measured T cell activation by K562 APCs and by biotinylated CD1a proteins bound to avidin-coated plates. As assessed with anti-CD1a blocking antibodies and K562 cells lacking CD1a, CD1a was required for the BC2 response to crude balsam of Peru and oils derived therefrom (Fig. 1, B and C). Treating plate-bound CD1a protein with balsam of Peru was sufficient to activate the BC2 response, albeit at higher doses than with antigen in the presence of CD1a-expressing cells (Fig. 1C). Thus, APCs facilitate some aspects of T cell response, but clear activation in APC-free systems ruled out that antigen processing is required. As a specificity control, BC2 did not respond to a structurally unrelated lipid, sphingomyelin, which is a known ligand for CD1a (Fig. 1D) (35). These results were most consistent with CD1a forming complexes with some molecule in these antigen preparations. Further specificity controls showed that balsam of Peru preparations did not activate a CD1a-restricted T cell clone, CD8-2, that recognizes CD1a presenting a mycobacterial antigen (Fig. 1D) (18, 36). This finding, along with the absolute requirement for CD1a in all recognition events, strongly indicated that these substances are not mitogens. However, both balsam of Peru and balsam of Peru oil did activate another CD1a-autoreactive T cell line, Bgp (31). This indicates that balsam of Peru response was not limited to the BC2 T cell line (Fig. 1E).

Next, we sought to pinpoint chemical structures of the antigenic substances. Balsam of Peru is a complex botanical extract, with the most abundant components previously reported to be benzyl cinnamate and benzyl benzoate (37). Silica thin-layer chromatography (TLC) showed that crude balsam of Peru contained hydrophilic compounds that remained near the origin, as well as two dark spots that comigrate with synthetic benzyl benzoate and benzyl cinnamate standards (Fig. 2A). As expected, oils extracted from balsam of Peru lacked the hydrophilic compounds that adhered at the origin. Balsam of Peru oil generated one dark spot that comigrated with benzyl benzoate. More sensitive methods of positive-mode nanoelectrospray ionization mass spectrometry (MS) (Fig. 2B) detected sodium adducts [M+Na]+ of benzyl cinnamate [mass/charge ratio (m/z) 261.3] and benzyl benzoate (m/z 235.3) in both preparations. The signal for benzyl benzoate was ~10-fold stronger than for benzyl cinnamate in balsam of Peru oil. Thus, benzyl cinnamate was present in both preparations, but its concentration was below the threshold of detection by TLC.

(A) Normal-phase silica TLC plate resolves balsam of Peru oil (BPO), crude balsam of Peru (BP), synthetic benzyl cinnamate (BC), and synthetic benzyl benzoate (BB). (B) Structures of benzyl cinnamate and benzyl benzoate are shown with the expected mass of sodium adducts [M+Na]+, which were detected in positive-mode nanoelectrospray ionization MS. (C to E) T cell clones that are autoreactive to CD1a (BC2) or foreign antigen (CD8-2) were tested for response to antigens (g/ml) or SM (sphingomy) by IFN- ELISA in cellular (E) or CD1a-coated plate (C and D) assays. Data are representative of three or more experiments, each shown as the mean of triplicate samples SD. The significance of lipid concentration on IFN- release was tested by one-way ANOVA (C). The significance of benzyl cinnamate and benzyl benzoate concentration on IFN- release and of the effects of CD1b or CD8-2 T cells were tested by two-way ANOVA (D and E).

False-positive results from trace contaminants in natural preparations occur, so we tested whether benzyl benzoate and benzyl cinnamate, provided as purified synthetic molecules, activated CD1a-restricted T cells. We observed T cell activation in response to both synthetic molecules, and the response was dependent on precoating the plate with CD1a. We observed a stronger and more potent response to benzyl cinnamate (Fig. 2C), which was then used for further mechanistic studies. Detailed testing of BC2 and CD8-2 activation by benzyl cinnamate confirmed the dose dependence, CD1a dependence, and TCR specificity of the T cell response to benzyl cinnamate (Fig. 2D). Sphingomyelin, a known CD1a ligand (31), which has a bulky polar head group, did not activate T cells. Responses to benzyl cinnamate were seen in two T cell lines, BC2 and Bgp (31). Benzyl cinnamate and benzyl benzoate were efficiently presented by plate-bound CD1a proteins after a short coincubation, demonstrating the lack of a cellular processing requirement (Fig. 2, C and D). These findings are most consistent with the formation of CD1abenzyl cinnamate complexes as the target of T cell response. Thus, tree oils that are known to act as potent contact hypersensitivity agents also function as T cell stimulants that act via CD1a.

The dual benzyl rings present in benzyl cinnamate and benzyl benzoate (Fig. 2B) are chemically different from the alkyl chains present in most CD1-presented antigens. However, they are notably similar to the dually ringed structure present in the unusual nonlipidic antigen presented by CD1d known as PPBF (22). All three nonlipidic T cell stimulants are smaller (212 to 345 Da) than most previously known CD1-presented lipid antigens (~700 to 1500 Da) (21). Prior CD1-lipid structures (21) established a widely accepted mechanism whereby the acyl chains rest inside the hydrophobic clefts of CD1 proteins, so that hydrophilic head groups protrude outside CD1 and form epitopes that specifically contact TCRs (Fig. 3A) (38). In contrast, the antigenic tree oils identified here lack any identifiable polar group that could function as a TCR epitope (Fig. 3B). Further, the size of the carbon skeletons of benzyl benzoate and benzyl cinnamate (C14 to C16) are substantially smaller than other CD1 antigens (C20 to C40) and the estimated capacity of the CD1a cleft (~C36) (19, 39, 40). Because tree oils are apparently too small to fill the CD1a cleft we hypothesized that they might not form TCR epitopes and so function outside the main CD1 antigen display paradigm. For example, interactions within the CD1a cleft might alter the shape of CD1-lipid complexes from the inside (41). Alternatively, similar to recent studies of CD1a (31, 35) and CD1c (42), tree oils might displace endogenous lipids, whose large head groups interfere with TCR contact with CD1a. This emerging model is known as the absence of interference because carried lipids do not contact TCRs directly but instead bind CD1 in a manner that allows direct contact between CD1 and the TCR (31, 35).

(A) Using PC as an example, CD1 ligands are often composed of head groups and lipid anchors, but (B) recently identified CD1a presented antigens are oils. (C) BC2 T cells were tested for cytokine release in response to small hydrophobic molecules pulsed on plate-bound CD1a pretreated with acidic citrate buffer to strip ligands (31). Tested compounds are classified into groups based on the presence of branched-chain unsaturated lipids structurally related to squalene, (D) ringed lipids structurally related to benzyl cinnamate, or (E) molecules that show branched, polyunsaturated, and ringed structures, such as coenzyme Q2. Results of triplicate analyses are shown as means SD with each compound tested two or more times. Post hoc comparison by marginal means of the interaction term between lipid and concentration after adjustment by the Sidak method was used to group treatments by nonoverlapping 95% confidence levels at the specified significance level after a significant result by two-way ANOVA. (F) The size of all tested antigens is shown on the basis of the number of carbon atoms (C) or mass [atomic mass units (u)], as compared with the volume of the CD1a cleft, which has been measured at 1650 3, and can accommodate ~36 methylene units (C36) (19, 40). (G) Purified T cells (CD4 and CD4+) were incubated overnight with plate-bound CD1a, either mock treated or pretreated with the indicated antigens (50 g/ml). Real-time PCR of IFN- mRNA relative to -actin.*P < 0.05, two-sided Students t test, antigen-treated compared with mock-treated CD1a.

The approach to testing chemical features was guided by the observation that squalene, benzyl benzoate, and benzyl cinnamate have ringed or unsaturated structures that chemically constrain molecules, rendering them bulky and rigid. Using tree oils and skin oils as lead compounds (Fig. 3B) to generate a larger test panel (Fig. 3, C to E, and fig. S2), we surveyed 29 structurally related molecules that differed in size, saturation, branching patterns, or ringed structures. Fifteen compounds, including examples among branched (Fig. 3C), ringed (Fig. 3, D and E), and saturated or unsaturated fatty acyl compounds (fig. S2), were recognized. This moderately promiscuous pattern was markedly different from T cell responses to glycolipids such as -galactosyl ceramide or glucose monomycolate, where altering a single stereocenter on the carbohydrate epitope abolished recognition (43, 44). However, not every oily substance was sufficient to activate T cells.

Considering the particular chemical structures that control response, squalene is a C30 polyunsaturated branched-chain lipid antigen (Fig. 3B) (31). We found cross-reactivity to structurally related C20 geranylgeraniol and C23 geranylgeranylactone, as well as C15 farnesol, but not smaller geraniol-based compounds (Fig. 3C). The farnesol response is notable because it is also a contact allergen in Truetest panel 2 (45) (Fig. 1) and so represents another link between contact allergens and CD1a antigens. Further, considering molecules with branched and ringed structures related to benzyl cinnamate, we identified a new antigen, coenzyme Q2 (Fig. 3D). Although coenzyme Q2 has not been described as a contact allergen, idebenone, which has an identical head group (2,3-dimethoxy, 5-methyl, 1,4-benzoquinone) but a less hydrophobic lipid tail, composed of a 10-carbon alkyl chain with a hydroxyl group, is a well-known skin allergen (4648). In addition, in our CD1a plate assays, idebenone stimulated a dose-dependent T cell response, supporting a link between coenzyme Q2related structures and contact allergens (fig. S3). Vitamin E, a known skin allergen, did not induce a response in this BC2-based screening. However, this does not exclude the existence of CD1a-restricted T cells to this hydrophobic compound within a polyclonal T cell repertoire.

The identification of a strong stimulatory response to coenzyme Q2 prompted screening of coenzyme Q length analogs, finding optimal response to coenzyme Q2 but not larger or smaller chain length analogs (Fig. 3, D and E). Last, comparison of 12 fatty acyl analogs consistently showed stronger response when the normally charged carboxylate group was capped by a methyl, alkyl, or other structure to generate a nonpolar molecule (fig. S2). A weak effect was seen in some cases, where potency was increased by cis unsaturation.

In summary, compared with highly flexible lipids with saturated alkyl chains, an unsaturated, ringed, or branched structure correlated with higher response. However, very highly constrained or bulky structures, such as vitamin A, vitamin D, and vitamin E, were not recognized. Considering molecular size, response was optimal with compounds (222 to 410 Da, C15 to C30) that were near the middle of the size range tested (154 to 862 Da, C9 to C59) (Fig. 3F). These optima were considerably smaller than known CD1 antigens (~700 to 1500 Da). Even the largest stimulatory compound, squalene (C30, 410 Da), was substantially smaller than the predicted number of methylene units (~C36) that would fill the CD1a cleft (1650 3) (19, 40). Unlike molecules that form antigenic epitopes for TCRs, no single molecular variant could be assigned as essential for T cell activation.

Last, to determine whether the identified link between CD1a and contact allergens is generalizable to polyclonal T cells and among genetically unrelated human donors, we screened purified polyclonal T cells (CD4+ and CD4) from blood bank donors and determined their response to plate-bound CD1a preloaded with either farnesol or coenzyme Q2. As also seen in clinical evaluation of contact dermatitis patients, not all patients responded to every antigen, but we observed polyclonal responses to both antigens in two or more subjects using sensitive real-time quantitative polymerase chain reaction (qPCR) testing of IFN- response (Fig. 3G). Responses were seen in the CD4+ T cell fractions but were stronger in the CD4 T cell fraction (Fig. 3G). This suggests that the normal T cell repertoire contains T cells that respond to CD1acontact allergen complexes. Similarly, in a different set of donors, T cell responses were detected to benzyl cinnamateloaded CD1a (fig. S4). Together, these results support the broader relevance of these CD1a allergens beyond the specificity of two T cell lines.

Farnesol is a common additive to cosmetics and skin creams, where its use requires precaution labeling, based on its recognized role as a contact allergen (45). Farnesol testing is routine in clinical practice, where it is present in the fragrance mix 2 in Truetest patches. Farnesol can also be tested as a pure compound, generating responses in ~1% of people with suspected contact dermatitis (45). After the screen identified a farnesol response (Fig. 3C), we observed reproducible and dose-dependent response for BC2 in the CD1a-coated plate assay (Fig. 4A). Thus, farnesol was unlikely to be modified before recognition and was likely recognized by the BC2 TCR as a CD1a-farnesol complex.

(A) IFN- release by BC2 T cells in response to CD1a-coated plates treated with farnesol was measured. Asterisk (*) indicates that the significance of lipid concentration on IFN- release was assessed by marginal means with adjustment by the Sidak method after a significant result by ANOVA, treating experiments 1 and 2 as blocks. At the highest concentration of farnesol in both experiments, nonoverlapping 95% confidence intervals were observed at P < 0.001. (B) Affinity measurements (KD) by SPR in response to the recombinant BC2 TCR binding biotinylated CD1a directly isolated from cells (CD1a-endo), CD1a pretreated with farnesol (CD1a-farnesol), or CD1a treated with buffer (CD1a-mock). Positive-mode HPLC-MS analysis of a farnesol standard (C) and eluents from farnesol-treated CD1a (D) demonstrated ions that matched the expected mass (m/z 205.195) of an indicated dehydration product with a retention time of 2.9 min. (E and F) Lipid eluents from CD1a-endo and CD1a-farnesol were analyzed by positive normal-phase HPLC-QToF-MS. Ion chromatograms were generated at the nominal mass values of DAG, PC, SM, and PI, which are shown as CX:Y, where X is the number of methylene units in the combined lipid chains, and Y is the total number of unsaturations. (G) Compound identifications were based on the unknown matching of the retention time and mass of standards. Further, one compound in the PC, SM, and PI families (shown in color) underwent collision-induced dissociation MS analysis to generate the indicated diagnostic fragments. RU, resonance units.

To test this hypothesis, we loaded farnesol onto biotinylated CD1a monomers, generated fluorescent tetramers, and stained the BC2 T cell line and a control line. In several attempts with differing protocols, we failed to detect staining with CD1a-farnesol tetramers above background levels seen with farnesol-treated CD1b tetramers (fig. S5). Turning to surface plasmon resonance (SPR), we produced the BC2 TCR heterodimer in vitro and measured its binding to untreated CD1a carrying mixed endogenous lipids (CD1a-endo), CD1a that was treated with media (CD1a-mock), and CD1a treated with farnesol (CD1a-farnesol) after coupling to SPR chips. The BC2 TCR bound to all three complexes with low but measurable binding affinities for CD1a-endo [dissociation constant (KD) = 123 M], CD1a-mock (KD = 144 M), and CD1a-farnesol (KD = 123 M) (Fig. 4B). SPR is known to be more sensitive than tetramer staining (49), so the relatively low affinity interactions likely explained the absent tetramer staining. Yet, interactions are still in the physiological range, demonstrating direct binding between the BC2 TCR and CD1a. However, the cross-reactivity of the BC2 TCR to three forms of CD1a left unclear the role of farnesol or other carried lipids in mediating CD1a-TCR interactions.

A recently proposed but unproven hypothesis is small hydrophobic lipids could fully sequester within CD1a (31, 50), displacing larger endogenous self-lipids that cover TCR epitopes on the outer surface of CD1a. Therefore, we undertook direct biochemical analysis of CD1a-lipid complexes formed in vitro with detergents and stimulatory substances, analyzing elutable lipids using high-performance liquid chromatographymass spectrometry (HPLC-MS). First, we addressed the trivial possibility that the lack of effect of farnesol treatment on TCR binding to CD1a resulted from the lack of farnesol loading onto CD1a. Analysis of eluents from farnesol-treated CD1a monomers was initially inconclusive because farnesol is a nonpolar alcohol and does not readily adduct the cations or anions needed for MS detection. However, building on the fortuitous detection of a positively charged dehydration fragment [M-H2O + H]+ generated in the MS source (31), we could reliably detect the equivalent product (C15H25+; m/z 205.196) from a farnesol standard. Subsequently, we detected strong signal for this product from farnesol-treated CD1a proteins but not CD1a-endo, directly documenting farnesol in CD1a complexes (Fig. 4D).

Further, the HPLC-MSbased platform allowed broader analysis of the lipid ligands carried in CD1a-endo and CD1a-farnesol complexes. Similar to prior reports (31, 35), we could detect many ions in CD1a-endo eluents, which were self-lipids captured during protein expression in cells. Focusing on specific classes of lipids, including neutral lipids, phospholipids, and sphingolipids, we could identify many self-ligands. CD1a-endo complexes carried at least three molecular species of diacylglycerol (DAG), six phosphatidylcholine (PC), six sphingomyelin (SM), and two phosphatidylinositol (PI) species. Initially, these identifications were based on the expected early (DAG) or later (PI, PC, and SM) retention time, as well as match of the detected m/z value with the expected mass of these ligands (Fig. 4, E to F). For one lead compound in each class, we confirmed the identification using collision-induced dissociation MS, which demonstrated the characteristic phosphocholine, phosphoinositol, sphingolipid, or DAG fragments (Fig. 4G).

Elution analysis of farnesol-treated CD1a directly demonstrated farnesol loading (Fig. 4D). The comparison of CD1a-endo and CD1a-farnesol eluents showed complete or nearly complete suppression of ion chromatogram signals corresponding to all the 17 tested self-lipids (Fig. 4E, blue). Although the conditions used to load farnesol in vitro are not the same as those in immunological assays, these findings suggest high occupancy of CD1a proteins by farnesol and that farnesol and self-lipids are not simultaneously bound. Together, these data support a simple model for the cross-reactivity, where the TCR binds CD1a carrying either farnesol or certain self-lipids that permit recognition. Treatment of CD1a with farnesol displaces lipids with hydrophilic head groups to generate more homogeneously liganded CD1a proteins (Fig. 4, D and E).

To determine the structural basis of farnesol response, we solved the CD1a-farnesol crystal structure at 2.2- resolution (table S1). The electron density for the bound farnesol and surrounding CD1a residues were unambiguous (fig. S6), allowing determination of the position and orientation of farnesol within the cleft (Fig. 5, A and B). Unlike covalent binding of vitamin B metabolites to major histocompatibility complex class Irelated protein (MR1) (51) and the predictions of haptenation models, we find no evidence for haptenation of CD1a residues by farnesol.

(A) Overview of the binary crystal structure of CD1a (gray)farnesol (purple)/2m (cyan). (B) Molecular interactions of farnesol (purple) with the hydrophobic residues within CD1a binding cleft (gray surface). The side chains of the residues within a 4- distance from the lipid are shown. A diagram of trans,trans-farnesol with carbon numbering is shown. The A pole formed by V12-F70 interaction in the context of oleic acidbound CD1a pocket [Protein Data Bank (PDB) ID: 4X6D] is highlighted in the inset. (C to E) Superimposition of CD1a bound to farnesol and SM [PDB ID: 4X6F (35)] (C), lipopeptide [PDB ID: 1XZ0 (40)] (D), and urushiol [PDB ID: 5J1A (30)] (E).

Instead, the notable finding is that farnesol is deeply sequestered within the CD1a cleft, where it is fully inaccessible to TCRs. Most known amphipathic membrane lipids, such as sulfatide or SM (19), occupy nearly all of the CD1a cleft and then extend their head groups through a portal (F portal) onto the external surface of CD1a (Fig. 5C). In contrast, farnesol occupies only 36% of the cleft. Accordingly, this relatively small ligand could have been seated in many ways within the larger cavity or potentially bound with lipid:CD1 stoichiometry of 2:1 or 3:1 (52). Instead, a preferred seating and orientation of a single molecule is observed at the junction of the A and F pockets. Unlike CD1b structures in which two lipids bind simultaneously within the cleft (53, 54), electron density corresponding to a second lipid or spacer in the cleft was not observed (Fig. 5, A and B). This finding agreed with elution experiments showing substantial exclusion of the measured self-lipids from CD1a complexes (Fig. 4E). Together, the biochemical and structural data indicated that farnesol itself was sufficient to stabilize a partially occupied CD1a cleft.

In previously solved CD1a structures in complex with oleic acid (35) or an acyl peptide (40), the flexible fatty acyl chains take a C-shaped conformation around the margin of the curved A pocket (Fig. 5D) (19, 35, 40). These lipids encircle a vertical structure known as the A pole, which is formed by an interaction of Phe70 and Val12, located in the ceiling and floor of the A pocket, respectively (Fig. 5B, inset) (19, 35, 40). The semirigid and branched structure of farnesol does not allow the C-shaped peripheral conformation seen with other lipids and instead lies in the center of the A pocket, disrupting the A pole. The orientation of farnesol is discernible: The terminal methyl and hydroxyl groups point toward the A and F pocket, respectively (Fig. 5B). The polar hydroxyl group is situated nearer the solvent-exposed F portal of CD1a with ~15% of its surface water exposed. Farnesol made van der Waals contacts with Phe10, Trp14, Phe70, Val98, Leu161, Leu162, and Phe169 from CD1a (Fig. 5B and table S2). Here, Trp14 stacked against the unsaturated hydrocarbons C12 and C14 of farnesol, further stabilizing the bound lipid within the cleft. The same Trp14 residue maintains hydrophobic contacts with sphingosine and acyl chain moieties in the CD1a-SM and CD1a-sulfatide structures (19), respectively. Collectively, this positioning mechanism appears to be driven by unsaturations in farnesol, which limit its ability to bend and provide van der Waals interactions with the inner surface of CD1a.

Parallels in the positioning of CD1a-urushiol and CD1a-farnesol (Fig. 5E) highlight how the positioning of bulky and constrained lipids differs from the seating of acyl chaincontaining ligands (Fig. 5D). Although farnesol and urushiol are not located in the same position, they are both situated near the junction of the A and F pockets (Fig. 5E) and do not take the deep and curved positioning at the rim of the A toroid (Fig. 5D). Whereas oleate and acyl peptide wrap around the intact A pole [Fig. 5B (inset) and D], farnesol and urushiol complexes show a marked repositioning of Phe70, which disrupts the A pole (Fig. 5, B and E). Urushiol extends substantially into the F-pocket so that it approaches the F portal of CD1a. It is unknown whether TCRs contact urushiol, but the molecule is adjacent to the surface portal (30), and TCRs can contact lipids located just within the portal (55). In contrast, farnesol is ~8 more deeply positioned, so that it is unequivocally separated from the F portal and the TCR contact surface (Fig. 5E).

Overall, the structure-activity relationships (Fig. 3) indicated that many small, hydrophobic, bulky lipids from consumer goods are recognized by T cells. The biochemical (Fig. 4) and structural (Fig. 5) analyses of CD1a-lipid complexes demonstrate that farnesols small size and unsaturated structure allow it to interact specifically, but not covalently, within CD1a. This binding interaction stabilizes the CD1a cleft and positions farnesol out of the reach of the TCR, largely or fully displacing lipids that normally emerge to the outer surface of CD1a (19, 35, 40).

In 1963, Gell and Coombs (3) classified human diseaserelated immune manifestations into four types of hypersensitivity reactions. Despite the early development and descriptive nature of this scheme, the classification system is still widely taught in clinical immunology and medicine. Type I, II, and III reactions are rapid and mediated by B cells, whereas the delayed type IV response is mediated by T cells. Our study sought molecular mechanisms underpinning type IV hypersensitivity to the most common contact dermatitis allergens in consumer products. Our data provide specific molecular connections between CD1a-reactive T cells and four structurally related contact dermatitis allergens: benzyl benzoate, benzyl cinnamate, farnesol, and coenzyme Qrelated compounds. Whereas haptens (9), drugs (7), or cations (8) can influence MHC-peptide display, here, we detail a straightforward mechanism for T cell activation by small molecules that noncovalently bind CD1a.

In the MHC and CD1 systems, the most common recognition mechanism involves TCR cocontact with an epitope on the carried peptide or lipid and the antigen-presenting molecule (21, 5658). Here, we show evidence that the key active components of balsam of Peru and farnesol activate T cells by binding to CD1a without cellular processing. However, both the structural and biochemical data strongly point to a new model of recognition that does not involve TCR contact with epitopes present on the stimulatory small molecules. Antigenic tree oils, PPBF, farnesol, coenzyme Q2, and the other 14 oily stimulants identified here all lack carbohydrate, phosphate, or peptidic groups that normally serve as TCR epitopes. We show that the BC2 TCR can cross-react among at least 16 stimulatory compounds, which do not share any single chemical structure that would be a candidate cross-reactive epitope. More conclusively, farnesol resides deeply within the CD1a cleft, essentially ruling out direct contact with the TCR. Sequestration of molecules of a small size could be a general mechanism of their recognition, because all of the stimulatory molecules are smaller than the CD1a cleft (21, 40, 57).

Prior studies of CD1-lipid complexes have emphasized head group positioning, where the seating of amphipathic lipids in the cleft is guided by carbohydrates or charged moieties that interact near the F portal. Alkyl chains have a bland repetitive structure and have been described as sliding within CD1 allowing diversely positioning in the groove (54, 59). On the basis of this concept, we expected that the small hydrophobic ligands studied here might slide freely or adopt multiple positions in the CD1a cleft. Also, because many of the lipids have a molecular size that is less than half the volume of the CD1a cleft, they might have bound in pairs or together with spacer lipids (52, 53, 60, 61). However, farnesol shows one defined position in the CD1a groove. Both MS and crystallographic analysis failed to detect cobinding spacer lipids, indicating that partial occupancy by one small lipid is sufficient to stabilize the CD1a cleft.

Comparison of CD1a-farnesol with previously solved CD1a-lipid structures provides insight into the roles of steric hindrance and interior pocket remodeling. CD1a-oleate (35), CD1amycobactin-like lipopeptide (40), CD1a-sulfatide (19), and CD1a-SM (35) complexes involve lipids with flexible alkyl chains. These alkyl chains insert deeply into CD1a by curling along the outer wall of the A pocket and wrapping around the A pole to insert fully within the cleft (40). In contrast, farnesol is chemically hindered and bulky, on the basis of polyunsaturation and methyl branching. The rigid and bulky moiety in urushiol derives from a substituted catechol ring. These two molecules cannot curl to trace the outer wall of the A pocket and so do not penetrate deeply, and both sit in a central position within the A pocket that prevents the A pole from forming. Farnesol is anchored in a specific position by a series of van der Waals interactions with named pocket residues formed by its polyunsaturated and branched structure. Although the roles of benzyl rings in benzyl benzoate and benzyl cinnamate are not studied structurally, they also constrain the chemical structure in ways that are also expected to prevent the side wall curvature (19, 35, 40). More generally, many of the stimulatory lipids identified here and in a recent study (31)including farnesol, squalene, geranylgeraniol, geranylgeranylacetone, and coenzyme Q2are polyunsaturated or branched isoprenoid lipids that could plausibly anchor in CD1a by similar mechanisms.

Lipid antigen binding wholly within CD1a could trigger T cell responses by remodeling the three-dimensional structure of CD1a, as previously reported for CD1d (62, 63), CD1b (54), and CD1c (41, 64). However, comparing CD1a-farnesol with all CD1a-lipid structures solved to date (19, 35, 40) does not demonstrate a broad or obvious change in CD1a conformation. Also, binding of the BC2 TCR to both CD1a-farnesol and CD1a-endo points away from this explanation. Instead, biochemical analysis of CD1a-endo complexes and the CD1a-farnesol structure both indicate that farnesol displaces endogenous ligands from the cleft. Whereas farnesol can be considered a headless ligand, some amphipathic self-lipid ligands in CD1a-endo structures have head groups composed of phosphates or sugars that normally cover the exposed surface of CD1a (35). In the case of the SM, it blocks autoreactive T cells by interfering with TCR contact with CD1a (31, 35). Our experimental observations rule in key aspects of the absence of interference model, where activating substances are sequestered within the CD1a cleft, so that recognition occurs by ejecting self-lipids and freeing up epitopes on the surface of CD1a itself.

As contrasted with MHC I and MHC II, where peptides are broadly exposed over the lateral dimension of the platform, human CD1 proteins have a large roof-like structure above their clefts and a small antigen exit portal at the margin of the platform (65). This creates a potentially large, ligand-free TCR contact surface on CD1 proteins. Evidence for the predominant contact of TCRs with the surface of CD1 proteins in preference to carried lipids, including the extreme case in which TCRs contact CD1 only, is becoming a central theme in CD1 research (65). Recent studies have shown direct TCR contact with the unliganded surface of CD1a and CD1c by autoreactive clones and polyclonal T cells (31, 35, 42). Thus, the stimulatory compounds identified here, which are small and internally sequestered, provide a molecular link to polyclonal autoreactive T cell responses, which are specific for the surface of CD1 rather than the carried lipid.

The presence of CD1a in all individuals prompts the question of why allergic contact dermatitis does not universally develop in everyone. However, interindividual differences that may play a role include permeability of the skin barrier (66), dose and number of chemical exposures to allergens, regulatory T cell activity (6769), and interindividual differences in T cell repertoires. Prior studies show that there is interindividual variability in the frequency of CD1a-restricted T cells in the blood and skin of healthy individuals and differences in CD1a-autoreactive response rates in skin (25, 66, 70, 71). Increased CD1a-restricted T cells responses were observed in allergic individuals and those with inflammatory skin disease (66, 70, 72), which may be a factor in susceptibility to development of CD1a-mediated allergic contact dermatitis in certain individuals. Consistent with these known patterns of antigen response, our small study of 11 humans demonstrates differing patterns of polyclonal response in each individual rather than a universal response to one antigen, which might be expected from an innate receptor.

Overall, the molecular analysis of tree oils and isoprenoid lipids presented in this manuscript invites focused consideration of the role of CD1a in T cellmediated skin diseases. In this new view, the pattern of high-density CD1a on the Langerhans cell network present throughout the skin could mediate responses to oils naturally produced within the skin or oils that contact the skin through application of commercial skin products containing botanical extracts, synthetic lipids, or oils. Other immunogenic oils used in human patients or for experimental biology include the adjuvant MF-59 (squalene) and incomplete Freunds adjuvant (mineral oil). These immunogens, as well as drug-like small molecules resembling PPBF or sulfonamide antibiotics, could plausibly act through the CD1 system.

The goal of this study was to determine whether known contact allergens can bind to CD1a and stimulate a CD1a-dependent T cell response. This study involved in vitro T cell assays using both CD1a-restricted T cell lines and polyclonal purified T cells from healthy blood bank donors. For T cell recognition, either cell-based assays using CD1a-expressing APCs or CD1a plate assays using recombinant plate-bound CD1a were performed. Cytokine release was measured by ELISA, and/or cytokine transcription was measured by real-time qPCR. Complex lipid mixtures, such as balsam of Peru, were purified by TLC and analyzed by nanoelectrospray ionization MS. Lipid eluents from CD1a, after displacement by contact allergens, were analyzed by positive normal-phase HPLC-quadrupole time-of-flight (QToF)MS. Structural insights into CD1a complexed with the contact allergen farnesol were obtained by x-ray crystallography.

T.R.U.E. Test panel 1 (Truetest 1) is a patch test routinely used in clinic to diagnose contact dermatitis in response to the most common allergens (SmartPractice, Phoenix, AZ). The system consists of surgical tape (5.2 cm by 13.0 cm) that is embedded with antigen patches of 0.81 cm2 with each coated with a polyester film that contains uniformly dispersed specific allergen. Using sterile technique, individual allergen patches were cut and placed directly in the assay wells containing ~106 APCs and 1 ml of T cell media in 24-well plates (contact method) or first extracted by soaking patch in 1 ml of media (2 hours, 37C), followed by removing the patch and transferring 100 l of media to T cell assays. Antigen dose was normalized to square millimeters of patch exposure. Antigens or extracts were cocultured with 50,000 CD1a- or mock-transfected K562 cells (25) and a CD1a-dependent T cell line in a 96-well plate. Activation was measured by IFN- ELISA (Thermo Fisher Scientific).

Balsam of Peru, balsam of Peru oil, benzyl cinnamate, and benzyl benzoate or other isolated antigens were dried in clean glass, subjected to water bath sonication in T cell media for 120 s, cultured with 50,000 CD1a- or mock-transfected K562 cells for 3 hours at 37C, and then cocultured with 50,000 to 200,000 cells per well of an autoreactive T cell line (BC2 or Bgp) (31) or foreign antigen reactive T cells (CD8-2) (18) for 24 hours at 37C in 96-well plates as previously described (31). Activation was measured using IFN- ELISA (Thermo Fisher Scientific). For blocking experiments, CD1a-transfected K562 cells were preincubated for 1 hour at 37C with CD1a-blocking antibody (OKT-6) or isotype-matched control immunoglobulin G (P3) (10 g/ml) before the addition of T cells. For plate assays, 96-well streptavidin plates (Thermo Fisher Scientific) were incubated for 24 hours at room temperature with biotinylated CD1a or CD1b protein [10 g/ml; National Institutes of Health (NIH) Tetramer Core Facility] and anti-CD11a (2.5 g/ml) in phosphate-buffered saline (PBS) (pH 7.4) as previously described (31). For the acid-stripping protocol (Figs. 4 and 5A and fig. S2), after 24 hours of coating with protein, plates were washed three times with PBS, followed by washing twice with citrate buffer at pH 3.4 for 10 min, followed by three washes in PBS before the addition of lipid antigens (30). Peripheral blood mononuclear cells (PBMC) were isolated from buffycoats obtained from the New York Blood Center, as approved by the Institutional Review Board of Columbia University Irving Medical Center. Polyclonal T cell assays were performed using FACS (fluorescence-activated cell sorting)sorted T cells from PBMCs (CD4 and CD4+) and CD1a-coated 96-well plates as described above. Plate-coated CD1a was either treated with buffer only (0.05% CHAPS in PBS) or lipid antigens sonicated in buffer and incubated overnight at 37C. Plates were washed three times, and then purified T cells were added to the wells and incubated overnight at 37C. RNA was extracted using RNeasy (Qiagen), and first-strand complementary DNA synthesis was performed using iScript (Bio-Rad).

Balsam of Peru (W211613), balsam of Peru oil (W211710), benzyl cinnamate (234214), benzyl benzoate (B9550), geranylgeraniol (G3278), farnesol (277541), geranylgeranyl acetone (G5048), geraniol (163333), squalene (S3626), geranyl acetone (250716), vitamin K1 (V3501), vitamin K2 (V9378), vitamin A (R7632), vitamin E (T3251), vitamin D3 (C9756), coenzyme Q2 (C8081), coenzyme Q0 (D9150), coenzyme Q4 (C2470), coenzyme Q6 (C9504), coenzyme Q10 (C9538), palmitoleic acid (P9417), methyl palmitoleate (P9667), cis-11-hexadecenal (249084), palmityl acetate (P0260), palmitoleyl alcohol (P1547), lauryl palmitoleate (P1642), oleamide (O2136), palmitoyl ethanolamide (P0359), tetradecanoic acid ethylamide (R425567), N-oleoyl glycin (O9762), N,N-dimethyl tetradecanamide (S347388), and 1-dodecyl-2-pyrrolidinone (335673) were obtained from Sigma-Aldrich (St. Louis, MO). Coenzyme Q1 (270-294-M002) was obtained from Alexis Biochemicals.

Silica-coated glass TLC plates (10 cm by 20 cm; Scientific Adsorbents Incorporated) were precleared in chloroform-methanol-water (60:30:6, v/v/v). Samples (10 to 20 g) were developed with a solvent system-hexane/diethyl ether/acetic acid (70/30/1, v/v/v). For visualization, plates were sprayed with a solution of 3% (w/v) of cupric acetate in 8% (v/v) phosphoric acid, followed by heating for 20 to 30 min at 150C.

Methanol solution (2 g/ml) was prepared for each reagent, and then, 10 l was loaded onto a glass nanospray tip for positive-mode electrospray ionization MS performed on an LXQ (Thermo Scientific), two-dimensional ion trap mass spectrometer. The spray voltage and capillary temperature were set to 0.8 kV and 200C.

CD1a-endo (200 g) and CD1a-farnesol (200 g) were transferred to 15-ml glass tubes and treated with chloroform, methanol, and water for lipid extraction according to the method of Bligh and Dyer (73). The lipid-containing organic solvent layer was separated from the top aqueous layer by centrifugation at 850g for 10 min. For HPLC-MS analysis, the samples were normalized on the basis of the input proteins (20 M), and 20 l of eluent was injected to an Agilent 6530 Accurate-Mass Q-TOF spectrometer equipped with a 1260 series HPLC system using a normal-phase Inertsil diol column (150 mm by 2.1 mm, 3 m; GL Sciences) with a guard column (10 mm by 3 mm, 3 m; GL Sciences), running at 0.15 ml/min according to a published method (74).

The glycoprotein CD1a was expressed in human embryonic kidney (HEK) 293S GnTI cells and purified as previously described (35). After an endoglycosidase H (New England BioLabs) and thrombin treatment, the purified CD1a was first loaded with the ganglioside GD3 (GD3) (Avanti) that was dissolved in a solution containing 2.5% dimethyl sulfoxide (DMSO) and 0.5% tyloxapol (Sigma-Aldrich). CD1a was first incubated overnight with GD3 at room temperature at a molar ratio of 1:8. The CD1a sample loaded with GD3 was further purified using ion exchange chromatography (MonoQ 10/100 GL, GE Healthcare). Trans,trans-farnesol (Sigma-Aldrich) was dissolved in a solution containing 2.5% DMSO and 0.5% tyloxapol (Sigma-Aldrich). The CD1a-GD3 sample was then incubated overnight with farnesol at a 1:100 molar ratio and at room temperature. A subsequent ion exchange chromatography (MonoQ 10/100 GL) was performed to remove the excess of farnesol, CD1a-GD3, and tyloxapol.

The BC2 TCR was produced using a previously described method (31). Briefly, individual and chains of the TCR, with an engineered disulfide bond between the TCR and TCR constant domains were expressed in BL21 Escherichia coli cells as inclusion bodies and solubilized in 8 M urea buffer containing 10 mM tris-HCl (pH 8), 0.5 mM Na-EDTA, and 1 mM dithiothreitol. The TCR was then refolded in buffer that was composed of 5 M urea, 100 mM tris-HCl (pH 8), 2 mM Na-EDTA, 400 mM l-Arg-HCl, 0.5 mM oxidized glutathione, and 5 mM reduced glutathione. The refolded solution was dialyzed twice against 10 mM tris-HCl (pH 8.0) overnight. The dialyzed samples were then purified through DEAE cellulose, size exclusion, and anion exchange HiTrap Q chromatography approaches. The quality and purity of the samples were analyzed via SDSpolyacrylamide gel electrophoresis.

Seeds obtained from previous binary CD1a antigen crystals (30) were used to grow crystals of the CD1a-farnesol binary complex in 20 to 25% polyethylene glycol 1500/10% dl-Malic acid, MES monohydrate, Tris (MMT) buffer (pH 5 to 6). The crystals were flash-frozen, and data were collected at the MX2 beamline (Australian Synchrotron) to a resolution of 2.2 . All the data were processed with the program XDS (75) and were scaled with SCALA from the CCP4 programs suite (76). Upon successful phasing by molecular replacement using the program PHASER (77) and the CD1a-urushiol structure as the search model (30), the farnesol electron density was evident in the unbiased electron density maps in addition to some very weak residual density. An initial run of rigid body refinement was performed using phenix.refine (78). Iterative model improvement was performed using with the program COOT (79) and phenix.refine. The final refinement led to an R/R-free (%) of 20/25. The quality of the structure was confirmed at the Research Collaboratory for Structural Bioinformatics Protein Data Bank Data Validation and Deposition Services website. All presentations of molecular graphics were created with UCSF Chimera (80).

Biotinylated CD1a-endogenous lipids derived from HEK293 cells was incubated overnight with 30-fold molar excess of farnesol solubilized in 2.5% DMSO/0.5% tyloxapol (CD1a-farnesol) or with solvent only (CD1a-mock). The sample was coupled onto research-grade streptavidin-coated chips to a mass concentration of ~3000 resonance units. Increasing concentrations of the BC2 TCR (0 to 200 M) were injected over all flow cells for 30 s at a rate of 5 l/min on a Biacore 3000 in 10 mM tris-HCl (pH 8) and 150 mM NaCl buffer. The final response was calculated by subtraction of the response for CD1a-endogenous minus a flow cell containing an unrelated protein. The data were fitted to a 1:1 Langmuir binding model using BIAevaluation version 3.1 software (Biacore AB) and the equilibrium data analyzed using Prism program for biostatistics, curve fitting, and scientific graphing (GraphPad).

All statistical analyses were performed in R (www.R-project.org/). Pairwise t tests, analysis of variance (ANOVA) post hoc testing, and adjustments of P values for multiple hypothesis testing used base R and the package emmeans (https://CRAN.R-project.org/package=emmeans). Dose-response analyses used the package drc to fit log normal or logistic curves to the data and to test fitted models against simplified, pooled models (81). R code is available on request.

immunology.sciencemag.org/cgi/content/full/5/43/eaax5430/DC1

Fig. S1. Screening human T cells for responses to known contact allergens.

Fig. S2. CD1a-dependent T cell response to small hydrophobic molecules.

Fig. S3. Idebenone is recognized by CD1a-restricted T cell line BC2.

Fig. S4. CD1a-dependent polyclonal T cell responses to contact allergens.

Fig. S5. CD1a tetramer staining of CD1a-autoreactive T cell line.

Fig. S6. Electron density for farnesol in CD1a-farnesol binary complex.

Table S1. Supporting data CD1a-farnesol binary complex.

Table S2. Van der Waals bonds between CD1a and farnesol.

Table S3. Raw data sets for main figures (Excel spreadsheet).

Acknowledgments: We thank A. G. Kasmar, M. C. Castells, and P. Brennan for advice or critical comments on the manuscript. We thank the staff at the Australian Synchrotron for assistance with data collection and the NIH Tetramer Core Facility for recombinant biotinylated CD1 protein. Funding: S.N. was supported by an NIH training grant (T32 AI007306) and is currently employed by HealthPartners, St. Paul, Minnesota. A.d.J. is supported by a K01 award from the NIH (K01 AR068475) and an Irving Scholarship from the Irving Institute for Clinical and Translational Research at Columbia University. D.B.M. is supported by the NIH (R01 AR048632) and the Wellcome Trust Collaborative Award. This work was supported by the National Health and Medical Research Council of Australia (NHMRC) and the Australian Research Council (ARC) (CE140100011). J.L.N. is supported by an ARC Future Fellowship (FT160100074); J.R. is supported by an Australian ARC Laureate Fellowship and the Wellcome Trust Collaborative Award. Research reported in this publication was performed in the CCTI Flow Cytometry Core, supported, in part, by the Office of the Director, NIH under award S10OD020056. Author contributions: The indicated individuals carried out project oversight and direction (A.d.J., D.B.M., and J.R.); T cell assays (S.N., T.-Y.C., E.A.B., R.N.C., I.V.R., G.C.M., and A.d.J.); protein chemistry, structure, and SPR (M.W. and J.L.N.); and manuscript preparation (S.N., A.d.J., D.B.M., and J.R.) with input from all authors. Competing interests: The authors declare that they have no competing interests. Data and materials availability: Reagents are available to qualified scientists subject to the limitation that cells from primary T cell lines can be limited in number. The data and refined coordinates for the CD1a-farnesol structure were deposited in the Protein Data Bank under accession code 6NUX. All other data needed to evaluate the conclusions in the paper are present in the paper or the Supplementary Materials.

Here is the original post:
Human T cell response to CD1a and contact dermatitis allergens in botanical extracts and commercial skin care products - Science

Read More...

Recession proofing the supply chain – Supply Chain Digital – The Procurement & Supply Chain Platform

Monday, January 6th, 2020

Many organisations have enjoyed unquestionable success during the recent economic expansion, including higher margins, product portfolio expansion and a renaissance in talent acquisition, with millions of new jobs created over the past several years. However, signals from multiple sources point to a coming recession within the next 18 to 24 months. A recent Grant Thornton survey of more than 250 C-level executives and business owners found more than three-quarters of respondents expect a recession to occur within the next two years, with the potential to impact a number of critical supply chain attributes, including physical infrastructure planning, new-equipment procurement and critical research and development investments.Faced with these potential challenges, many organizations are embracing the pathway to recession preparedness by adopting a number of strategies designed to preserve recent performance gains and minimize supply chain disruptions.

Organisations can prepare for the uncertain times ahead by adopting a recession mitigation framework that targets the most high-risk supply base, process and delivery capabilities. With such a framework in place, they can quickly develop what can be termed a ready-for-deployment supply chain immune system to minimize any disruptions and preserve the competitive advantage of their supply chain. While some leaders are resigned to acceptance of inevitable recessionary effects, savvy, proactive executives can translate the noise of a recession into actionable, deployment-ready mitigation strategies to capture the highest level of supply chain resilience and agility possible.

Identifying the signs of recession

While knowledge of an impending recession is important, paying close attention and preparing for the potential impact on your operation is mission critical. Grant Thorntons survey found that nearly three in 10 (29%) respondents have a supply chain resiliency strategy in place. The balance of respondents, 71%, are in various stages of considering, planning or implementing such a strategy.

Based on our research, Grant Thornton has identified seven key recession indicators which, when combined, can provide a useful framework to assess your risk preparedness for a possible economic slowdown. These indicators, when viewed across the enterprise, can also inform your supply chain mitigation strategies:

Global dashboard deterioration outside of normal control limits (e.g. growing inventories, demand drop)

Supply partner financial issues including market contraction and growing backlogs

Internal and trading partner budget contractions and budget freezes

Diminishing margin capture against long-term forecasts

Talent pool shrinkage across core functions due to functional budget issues

Cross-functional continuous improvement investment shrinkage and funding shortfalls

Shrinking cross-functional cooperation and synchronization (local survival focus)

SEE ALSO:

Taken together, these seven indicators should prompt a number of introspective questions regarding an organizations supply chain health, including:

Could diminishing supply partner financial performance potentially transition from periodic issue management to a quantifiable supply risk?

Will our promised market-known business process improvements lapse because of internal funding choke-holds?

Can talent attrition become a highly visible concern to key customers?

When will my growing inventory pipeline reach the critical stage of unbudgeted obsolescence, disposal and excess storage costs?

While each question individually poses a tactical risk with (hopefully) only a localized performance impact, the effects of multiple scenarios in parallel pose significant risks to supply chain flexibility, adaptability and ultimately profitability. For those executives who see the signs of a recession as an opportunity to further refine and strengthen their existing supply chains, there are several practical strategies leaders can rapidly deploy as part of an offensive recessionary response.

Building up organisational supply chain immunity

Organisations have a clear opportunity to benefit from a supply chain recession preparedness framework designed to address the seven indicators outlined across the end-to-end supply chain value stream. Holistically evaluating the complete supply chain value stream, an effective supply chain immunity assessment should catalog the most significant supply chain risks, including the magnitude of the possible fallout of each identified risk along with the individual risk trigger point. Armed with priority risks and their associated trigger points, the assessment must then translate into ready-for-deployment mitigation strategies to lessen quantified recessionary impacts, preserve competitive advantages, and lessen market fears. Once in place, the immune system can provide the tailwinds to preserve market confidence and sound financial footing. Key aspects of a successful immune system include:

Contingency SKU rationalization planning to drive out unnecessary cost and complexity when laser focus on higher-margin products become a true recessionary lifeboat

Highly targeted supply contract temporary modifications, including advanced buys for high-risk components, tighter backlog management and improved risk-sharing across the relationship

Customer segmentation refinement to pre-select and plan for service level adjustments should supply constraints become a reality

Cross-functional preparedness to harness available enterprise-wide cerebral horsepower and prevent isolated, siloed recession response plans (e.g. expand the power of S&OP as a functional integrator)

Advanced automation efforts prior to major recessionary impacts to better prepare for, and ethically manage, labor reallocations

Close coordination with strategic suppliers to improve transparency to core supplier cost drivers and appropriately share recessionary concerns and deploy a partnership approach to proactively manage down costs where possible

Ultimately, the assessment, along with the adoption of targeted risk mitigation strategies, can proactively reduce an organizations overall recession risk profile and the magnitude of any possible disruption. Known and documented risks, proactive mitigation strategies and deployment-ready action plans are the necessary resilience tools for the next recession. Long before competitors are scrambling to adapt, proactive business leaders can take the steps necessary today to set their organisations on the right path. Theyll face down a coming recession with confidence, greater precision and highly ethical responses to challenging supply chain design and resourcing questions.

By Bob Hawkey, Director, Operations Transformation, Advisory Services, Grant Thornton LLP

For more information on all topics for Procurement, Supply Chain & Logistics - please take a look at the latest edition ofSupply Chain Digital magazine.

Follow us onLinkedInandTwitter.

Read more:
Recession proofing the supply chain - Supply Chain Digital - The Procurement & Supply Chain Platform

Read More...

Belgian researcher aims to turn body’s own defences against cancer – The Brussels Times

Monday, December 30th, 2019

A Belgian researcher has made important steps in developing a new technique that would recruit the bodys own defences against the presence of cancer cells.

Ultimately, the technique would be much less damaging than conventional techniques such as radio- and chemotherapy.

Sophie Lucas is a researcher at the Duve Institute, linked with the Catholic University of Louvain-la-Neuve (UCL). Her work concerns a type of cell known as Regulatory T lymphocytes, or Tregs cells that are part of the lymph system which makes up a large part of the bodys immune system.

Some reactions of the immune system can cause auto-immune conditions, where the bodys defences attack healthy tissues, causing numerous conditions including Type 1 diabetes, lupus, psoriasis and coeliac disease. Even simple hay fever is a sort of auto-immune condition, where the body has a violent reaction to something which is not in itself dangerous, i.e. tree or grass pollen.

The body has equipped itself with Treg cells to suppress excessive immune reactions, but these cause a problem in the case of cancer. In cancer patients, they may favour tumour progression by suppressing immune responses against tumour cells: Tregs inhibit anti-tumour T cells, Lucas writes on the Institutes website. Our group studies how human Tregs inhibit anti-tumour responses.

Cancer itself can be seen as a consequence of the inhibitory action of Tregs. When cancer cells begin to grow in the body, the immune system attacks them as foreign bodies. But as the cancer grows large enough to be detectable, the immune system is not able to keep up with its growth, and this is one probably result of Treg action: the Tregs have stopped the immune system from working full-out.

The idea is to somehow harness the power of Tregs to turn them against not the bodys own immune response, but the presence of the cancerous cells themselves. The Tregs and the bodys normal immune system would then work together against the invader, instead of being in opposition to each other.

The Institutes work is firstly to study how Tregs work, to allow researchers to develop ways of making them work as desired. But while Tregs can be difficult to differentiate from ordinary white blood cells, the team has found a work-around: We circumvented this problem by deriving clones of human Treg cells. Clones are pure populations of cells that can be kept in culture for long periods of time, and provide very stable material to perform repeated experiments and derive robust results. We used our human Treg clones to identify functional features present in Tregs, but not in other types of lymphocytes, that could therefore mediate the Treg immune suppressive functions, Lucas writes.

So far, the team has found that Tregs produce a protein called TGF-, which has an important role in immune system suppression. They regulate TGF- by using another protein attached to their surface, known as GARP. Now, if the exact roles of the two proteins separately and apart can be decoded, the possibility of ordering Tregs to stand down when the immune system is fighting cancer cells but not other malign cells, the battle against cancer could enter a new era.In the long term, the objective is to try to manipulate immune responses to make them more effective, and to permit the patient to reject their own tumour cells.

Alan HopeThe Brussels Times

Continued here:
Belgian researcher aims to turn body's own defences against cancer - The Brussels Times

Read More...

How to stay healthy while traveling this winter – The Boston Globe

Monday, December 30th, 2019

Acts of kindness can keep us healthy by releasing chemicals in the pleasure center of our brains, such as serotonin and oxytocin, says Dr. Sara Whatley-Dustin, family medicine physician at Dignity Health. Anxiety lowers our immune system and therefore makes us more susceptible to illness.

Mother knows best

Talia Segal Fidler, nutritionist at The Lodge at Woodloch, a destination spa resort in Pennsylvania, suggests taking a page from Mother Natures recipe box to beat a winter cold or virus. If you eat with the seasons, you will be giving your body exactly what it needs, she says. This includes winter greens like chard, kale, cabbage, and collards for fiber, antioxidants, phytonutrients, vitamins, and minerals. Also, root veggies like beets, sweet potatoes, and turnips provide immune system-loving minerals and Vitamin A. And, citrus fruits from warmer climes in the winter months are Vitamin C-rich and hydrating, help your body absorb other nutrients, and are anti-bacterial, too.

Cover up

New York City-based Kemi Adewumi, founder and CEO of Go Galavant, a group trip travel platform for travelers who hate group travel, just returned from an eight-month travel stint and swears by covering your face holes on flights, long train rides, and long bus rides. Yes, youll look like patient zero, but youll avoid actually being patient zero, she says.

She started out with scarves, and now uses the mouth masks, like the kind that doctors use during surgery (you can find the masks on Amazon or in some drug stores). You can also buy fun-looking reusable ones, says Adewumi, which, of course, require washing after use.

Keep hydrated

Your nose, throat, and lungs use a special part of the immune system called IgA to fight infections on moist surfaces, says Dr. Jacob Teitelbaum, board-certified internal medicine physician and author of Real Cause, Real Cure. You can think of it as the bodys Navy. Just like the Navy, it works very poorly on dry surfaces. So, drinking plenty of water (not sodas, as the sugar in one can of soda suppresses immune function by 30 percent) or hot tea (which loosens the secretions so they can be coughed out) can be very helpful in preventing and fighting respiratory infections.

Strike a pose

Liz Zabel, co-manager of the Emerson Spa at Emerson Resort & Spa in the Catskills (named for Ralph Waldo Emerson), is a certified yoga instructor who has practiced for more than a decade. Zabel endorses easy, quick yoga poses to ward off winter travel bugs. To change the blood flow and redirect pranic energy, lay on the floor for a few moments with your legs lifted over your head and against a wall, says Zabel. This simple pose, Legs Up the Wall, will help relieve stress and spark the bodys inner ability to heal itself. The pose is especially helpful because it does not add any stress to the body, it calms the nervous system and helps regulate blood flow.

Just breathe

Breathing, including yawning, can also help the immune system bully germs. If you find yourself holding back any yawns (or sighs) throughout the day, says Zabel, try letting them go. Yawning and sighing can help to cool your brain down from clotting and is a natural way for your body to wake itself up. It also improves the circulation to your brain by carrying more oxygen through the bloodstream and moving more carbon dioxide out.

Rest up

One of the most powerful ways to immune suppress an animal is to sleep deprive it, says Dr. Teitelbaum. Humans are no different. The average nights sleep in the United States until light bulbs were invented 140 years ago was nine hours a night. We are now down to six and three-quarter hours. This is a contributing factor in the increasing immune dysfunction being seen today.

Dr. Rand McClain, chief medical officer at Santa Monica, Calif.-based LCR Health, also emphasizes sleep. The most important immune supporting tool that I see most in the Western world eschew is adequate sleep, says Dr. McClain. He recommends seven to nine hours a night and routinely at the same time period, such as 11 p.m. to 7 a.m. nightly, rather than at varying times. So, if not always, then before traveling awhile on the road, pay attention to getting adequate sleep, he says.

Laurie Wilson can be reached at laurieheather@yahoo.com.

Here is the original post:
How to stay healthy while traveling this winter - The Boston Globe

Read More...

Solving the puzzle of IgG4-related disease, the elusive autoimmune disorder – QS WOW News

Monday, December 30th, 2019

Scientists piece together the inflammation mechanism in IgG4-related disease, an autoimmune condition with no current cure, revealing possible therapeutic targets

IgG4-related disease is an autoimmune disorder affecting millions and has no established cure. Previous research indicates that T cells, a major component of the immune system, and the immunoglobulin IgG4 itself are key causative factors, but the mechanism of action of these components is unclear. Now, Scientists from Tokyo University of Science have meticulously explored this pathway in their experiments, and their research brings to light new targets for therapy.

Autoimmune diseases are a medical conundrum. In people with these conditions, the immune system of the body, the designated defense system, starts attacking the cells or organs of its own body, mistaking the self-cells for invading disease-causing cells. Often, the cause for this spontaneous dysfunction is not clear, and hence, treatment of these diseases presents a major and ongoing challenge.

One recently discovered autoimmune disease is the IgG4-related disease (or IgG4-RD), which involves the infiltration of plasma cells that are specific to the immunoglobulin (antibody) IgG4 into the body tissue, resulting in irreversible tissue damage in multiple organs. In most patients with IgG4-RD, the blood levels of IgG4 also tend to be higher than those in healthy individuals. Previous studies show that T cellswhich are white blood cells charged with duties of the immune responseplay a key role in the disease mechanism. In particular, special T cells called cytotoxic T lymphocytes, or CTLs, were found in abundance from the inflamed or affected pancreas of patients, along with IgG4. But what was the exact role of CTLs?

In a new study published in International Immunology, a team of scientists from Tokyo University of Science decided to find the answer to this question. Prof. Masato Kubo, a member of this team, states that their aim was twofold. We planned to explore how IgG4 Abs contributes to the CTL-mediated pancreas tissue damage in IgG4-RD, and also to evaluate the pathogenic function of human IgG4 Abs using the mouse model that we have established. The latter is especially important, as IgG4 is not naturally present in mice, meaning that there is a severe lack of adequate animal models to explore this disease.

With these aims, they selected mice that have been genetically programmed to express a protein called ovalbumin (the major protein in egg white) in their pancreas. Then, they injected IgG4 that specifically targets ovalbumin into the mice. Their assumption was that IgG4 would target the pancreas and bring about IgG-4-RD-like symptoms. However, what they found was surprising. No inflammation or any other symptom typical of IgG4-RD appeared. This convinced the researchers that IgG4 alone was not the causative factor of IgG4-RD.

Next, to check if it was the CTLs that were perhaps the villain of the story, the scientists injected both IgG4 specific against ovalbumin as well as CTLs. Now, the pancreas of the mice showed tissue damage and inflammation. Thus, it was established that the presence of CTLs and IgG4 was necessary for pancreatic inflammation.

When they probed further, they found that another variation of T cells, known as T follicular helper or TFH cells, which develop from the natural T cells of the mice, produce self-reactive antibodies like IgG4, which induce inflammation in combination with CTLs.

Once the puzzle was pieced together, the scientists now had the opportunity to zero in on the target step for intervention; after all, if one of these steps is disrupted, the inflammation can be prevented. After much deliberation, they propose that Janus kinase, or JAK, can be a suitable target. JAK is a key component of the JAK-STAT cellular signaling pathway, and this pathway is an integral step in the conversion of natural T cells of the mice to TFH cells. If this JAK is inhibited, this conversion will not take place, meaning that even the presence of CTLs will not be able to induce inflammation.

Prof. Kubo also suggests a broader outlook, not limited to the therapeutic option explored in the study. He states, based on our findings, the therapeutic targets for IgG4-related diseases can be the reduction of TFH cell responses and the auto-antigen specific CTL responses. These can also provide the fundamental basis for developing new therapeutic applications.

These proposed therapeutic targets need further exploration, but once developed, they have the potential to improve the lives of millions of patients with IgG4-RD worldwide.

###

Reference

Journal:

International Immunology

About The Tokyo University of Science

Tokyo University of Science (TUS) is a well-known and respected university, and the largest science-specialized private research university in Japan, with four campuses in central Tokyo and its suburbs and in Hokkaido. Established in 1881, the university has continually contributed to Japans development in science through inculcating the love for science in researchers, technicians, and educators.

With a mission of Creating science and technology for the harmonious development of nature, human beings, and society, TUS has undertaken a wide range of research from basic to applied science. TUS has embraced a multidisciplinary approach to research and undertaken intensive study in some of todays most vital fields. TUS is a meritocracy where the best in science is recognized and nurtured. It is the only private university in Japan that has produced a Nobel Prize winner and the only private university in Asia to produce Nobel Prize winners within the natural sciences field.Website: https://www.tus.ac.jp/en/mediarelations/

About Professor Masato Kubo from Tokyo University of Science

Dr Masato Kubo is a Professor at the Tokyo University of Science. A respected and senior researcher in his field, he has more than 226 publications to his credit. He is also the corresponding author of this study. His research interests include Immunology and Allergology. He is the team leader at the Laboratory for Cytokine Regulation, RIKEN Center for Integrative Medical Sciences.

Funding information

This study was supported by grants from JSPS KAKENHI (grant no. 19H03491), Japan Agency for Medical Research and Development (AMED), AMED-CREST, and Toppan Printing CO., LTD.

Read more:
Solving the puzzle of IgG4-related disease, the elusive autoimmune disorder - QS WOW News

Read More...

Have you tried immunotherapy to treat cancer? Calling All Readers – cleveland.com

Monday, December 30th, 2019

CLEVELAND, Ohio Immunotherapy, which boosts a patients immune system to destroy cancer cells, is widely seen as the future of cancer care.

This kind of therapy uses substances made by the body, or in a laboratory, to improve or restore the immune system. It can slow the growth of cancer cells, help the immune system do a better job of destroying tumors and slow the spread of cancer to other parts of the body.

There are several kinds of immunotherapy, including CAR-T cell therapy and immune checkpoint inhibitors. In CAR-T, white blood cells called T cells are genetically modified to activate the immune system to recognize and destroy certain cancers.

Immune checkpoint inhibitors are drugs that block proteins made by some immune system cells. When these proteins are blocked, T cells are better able to kill cancer cells.

Cancer vaccines, which are used to prevent and treat cancer, also are a kind of immunotherapy.

Have you or a family member experienced immunotherapy treatment for cancer? Was the treatment effective or ineffective? We want to hear your story.

Please write a short email, no more than 500 words, about your experiences with immunotherapy. Include your full name, age, city and daytime phone number. Your name, age and city will be published with your comment, but your phone number will be kept private.

Comments must be received by Friday, Jan. 10.

Read more from the original source:
Have you tried immunotherapy to treat cancer? Calling All Readers - cleveland.com

Read More...

These biomedical breakthroughs of the decade saved lives and reduced suffering – CNBC

Monday, December 30th, 2019

When it came to biomedical breakthroughs, this past decade represented a lot more hype than substance. We were promised a new era of "precision medicine," where every patient would receive highly personalized treatments to target specifics fault in their genes. But in recent months, reports surfaced that these experimental therapies were failing most of the time.

Vas Bailey, a biotech investor with Artis Ventures, acknowledges that it might have seemed like a disappointing decade, barring a few important breakthroughs in the fields of gene editing and therapeutics. But Bailey thinks that there's a lot more to the story.

In recent years, he notes, scientists have focused on the "tools and building blocks" that will drive the breakthroughs in next decade.

"With HIV, we made progresses in life expectancy but didn't cure it; with gene-editing, we achieved one approval that could lay the foundation for others; with AI we learned how to develop it, but haven't yet applied it; and with the microbiome, we have learned more about the relationships (between organisms) but we still haven't designed next generation drugs."

So with the help of a team of biotech experts, here's our list of the most important advancements in biomedicine, many of are likely to propel great advancements going forward.

CAR-T sounds like the stuff of science fiction: Take blood from a cancer patient, re-engineer the blood cells to target and fight cancer cells, and re-infuse the cells in the human body.

But it's real, with the U.S. Food and Drug Administration granting approval in 2017 to the first two drugs that use this approach: Novartis's Kymriah for acute lymphoblastic leukemia (ALL) and Gilead Sciences' Yescarta for certain types of non-Hodgkins lymphoma.

But this therapeutic approach has faced roadblocks, including price -- the drugs cost hundreds of thousands of dollars -- and the logistical complexity of making a treatment out of the patient's own T cells. It can take up to a month to manufacture the cells while the patient's health might be getting progressively worse from the cancer. But this decade also saw progress in companies making off-the-shelf T cells, which are made from healthy donor cells and used for multiple patients.

The next ten years could see advances in using these donor cells -- in particular, reducing the likelihood that patients' immune systems will attack them -- by applying technologies and tools like CRISPR/Cas9.

And that brings us to...

Jennifer Doudna, inventor of the revolutionary gene-editing tool CRISPR photographed in the Li Ka Shing Center on the Campus of the University of California, Berkeley.

Nick Otto | The Washington Post | Getty Images

CRISPR has been hailed as one of the most important breakthroughs of all time. It's essentially a pair of molecular scissors, or a technique to make precise edits in DNA. It was discovered by scientists while exploring the immune system of bacteria.

Scientists have subsequently learned that there are risks to making these modifications. There are also ethical considerations, which were brought to the fore this decade when a scientist in China called He Jiankui reported that he had used the technology to create the first human babies with CRISPR-edited genes.

But this decade ended on a positive note. NPR reported that the first patient in the United States with a genetic disease -- a blood disorder called sickle cell anemia -- was treated with the CRISPR technique. The director of the National Institutes of Health Dr. Francis Collins, told NPR it could give patients a "chance for a new life."

After decades of research into better treatments for Hepatitis C, a debilitating liver disease that could affect as many as 4 million Americans, the U.S. Food and Drug Administration approved a new treatment in 2017 that can reverse the disease in as little as 8 weeks.

The drug from AbbVie followed approvals for similar drugs from fellow drug makers Gilead and Merck, providing a welcome alternative to the prior regimen of shots and pills that didn't always work and came with many side effects. Gilead, which saw its first drug for Hepatitis C approved in 2013, provided a cure in 12 weeks for about 90 percent of patients.

The drugs are extremely expensive, and led to a big spike in revenues for Gilead, but new entrants will provide more competition, potentially leading to lower prices.

A man checks in with a program manager after driving nearly three hours to get to his appointment at Open Arms Healthcare Center on Wednesday, January 23, 2019, in Jackson, MS. Open Arms provides PrEP, or Pre-exposure prophylaxis, which is "a way for people who do not have HIV but who are at substantial risk of getting it to prevent HIV infection by taking a pill every day," according to the CDC.

Jahi Chikwendiu | The Washington Post | Getty Images

This decade, scientists did not cure HIV, the virus that causes AIDS, but they did make some major breakthroughs.

In 2012, the Food and Drug Administration approved Truvada, the first drug to reduce the risk of HIV infection in uninfected individuals who might be at high risk. The drug, which can be taken daily, is used for pre-exposure prophylaxis (PrEP) to lower chances of becoming infected with HIV if exposed.

Unlike some of the other breakthroughs on this list, Truvada has become more pervasive and widespread throughout the decade as costs have come down. The Trump Administration recently unveiled a plan to make these drugs free for people who don't have insurance coverage, and virtual medicine companies like Nurx and Plushcare have sprung up to prescribe PrEP online without a lab visit.

A young boy is handed a certificate confirming he is Ebola-free, outside the Ebola treatment centre in Beni, eastern Democratic Republic of the Congo.

Sally Hayden | SOPA Images | LightRocket | Getty Images

In November of 2019, European regulators finally approved an immunization against Ebola. That decision means that Merck can market its vaccine and distribute it beyond Africa. It has also been approved by regulators in the U.S.

Several vaccines are in development to prevent the outbreak of the fever, which causes such symptoms as diarrhea and bleeding, but Merck's is the only one that was tested during a real outbreak from 2014 to 2016. Ebola has killed more than 2,000 people in the Congo since the middle of this year alone.

Merck has said that it expects to start manufacturing licensed doses of the vaccine in the third quarter of 2020, and that it is working closely with the U.S. government, public health organizations and other groups.

These are just five of the breakthroughs from the past decade, which also saw developments in new "biologic" drugs, which contain or are produced from living organisms, advancements in prosthetics, and in a heartwarming twist, a new medicine for a single patient.

But as Bailey reminds, the best might still be to come. In the past ten years, scientists also made great strides to map out all the cells in the human bodyand to understand contributors to disease encoded in DNA. Teams of researchers also sought to create reference databases for the collections of microbes living in our bodies, and computer scientists teamed up with biologists to extract meaning from huge volumes of medical information, including X-ray imaging and pathology slides.

With all this in mind, Bailey thinks we could see in the next ten years regulatory approvals for the first blood test to screen for cancer at the earliest stages, new therapies that take advantage of our understanding of RNA (the intermediary between DNA and the proteins it instructs the body to make), drugs based on our ever-growing knowledge of the human microbiome, a cure for HIV, and novel approaches to ending multi-drug resistance.

What we've built in recent years are "enabling systems," said Bailey, "and we will benefit from it in the next decade."

Follow @CNBCtech on Twitter for the latest tech industry news.

Excerpt from:
These biomedical breakthroughs of the decade saved lives and reduced suffering - CNBC

Read More...

These are the most-read Science News stories of 2019 – Science News

Monday, December 30th, 2019

Science News drew more than 15 million visitors to our website this year. Heres a rundown of the most-read news stories of 2019 that didnt make our Top 10 list, as well as the most popular longer reads.

1. A chip made with carbon nanotubes, not silicon, marks a computing milestoneResearchers built a new kind of computer chip with thousands of carbon nanotube transistors. Though the prototype cant yet compete with silicon chips, carbon nanotube computing technology could lead to faster electronics (SN: 9/28/19, p. 7).

2. People can sense Earths magnetic field, brain waves suggestPeoples brain waves showed a distinct pattern when exposed to an Earth-strength magnetic field pointing in a specific direction in the lab. That finding hints that humans may have magnetoreception, similar to birds and certain other organisms .

Headlines and summaries of the latest Science News articles, delivered to your inbox

3. In a first, scientists took the temperature of a sonic black holeThe temperature of a lab-made black hole that traps sound instead of light agrees with a prediction by cosmologist Stephen Hawking: that black holes emit a small stream of particles called Hawking radiation (SN: 6/22/19, p. 14).

4. Why kids may be at risk from vinyl floors and fire-resistant couchesChildren from homes with all vinyl flooring and flame-retardant couches had higher levels of some potentially harmful chemicals in blood and urine than other kids did, one study found. The finding suggests that these home furnishings release the chemicals quickly enough for them to build up in residents bodies (SN: 3/16/19, p. 14).

5. Archaeologists tie ancient bones to a revolt chronicled on the Rosetta StoneAn ancient soldiers skeleton unearthed from Egypts Nile Delta may be physical proof of a revolt around 2,200 years ago. The Rosetta Stone describes the victory of pharaoh Ptolemy V, from a Greek dynasty, over a faction of the native Egyptian revolt. But archaeological evidence of the uprising is scarce.

1. Vitamin D supplements arent living up to their hype Vitamin Ds popularity soared after findings hinted that it could protect against multiple sclerosis, asthma, depression, heart disease, cancer and other ailments. But a series of studies has cast doubt on these supposed benefits (SN: 2/2/19, p. 16).

2. Measles erases the immune systems memoryMeasles wipes away the immune systems memories of germs it has previously fought. This immune amnesia can leave people at risk of infections from harmful viruses and bacteria for months to years (SN: 6/8/19, p. 20).

3. With its burning grip, shingles can do lasting damageThe virus responsible for chicken pox can lay dormant for decades, only to reemerge later in life as shingles. The disease is more than just a painful rash. Shingles can damage arteries and may raise the risk of stroke and dementia, scientists are finding (SN: 3/2/19, p. 22).

4. The CBD boom is way ahead of the scienceFood, health and wellness products infused with cannabidiol, also known as CBD, are becoming increasingly popular. The substance, derived from cannabis plants, is sold as a remedy for pain, anxiety, insomnia and other conditions without getting the user high. But most health benefits attributed to CBD dont yet have scientific backing (SN: 3/30/19, p. 14).

5. How the periodic table went from a sketch to an enduring masterpieceScience News kicked off its coverage of the periodic tables 150th anniversary with a look at Russian chemist Dmitrii Mendeleev, whose original table had just 63 elements. Scientists have since added many more elements to the table, one of the most important tools in chemistry (SN: 1/19/19, p. 14).

Read more:
These are the most-read Science News stories of 2019 - Science News

Read More...

Autoimmune disorders when the body attacks itself – Times of Malta

Monday, December 30th, 2019

The immune system coordinates a complex array of components consisting of specialised organs, cells and proteins. On the surface, the primary role of the immune system in defending the body against infectious pathogens might appear straightforward. This begs the question: why is the immune system so complicated?

There are a number of reasons for this degree of complexity. The first is that the immune system evolved to fight pathogenic infection while minimising collateral damage to healthy tissue. For sake of argument, if the lungs of a patient were infected with a pathogenic microorganism, the immune system could release molecules that would kill the pathogen. However, if these molecules were to be released in an uncontrolled and directionless fashion, healthy tissue would be destroyed too.

This analogy raises another important feature of the immune system, namely, the recognition of differences between healthy cells and pathogens referred to as self and non-self respectively.

Each one of us has a personal army of immune cells that are programmed to harmoniously communicate with one another ensuring discrimination of self from non-self and efficiently respond to infection. Inflammation is a key process that ensures the recruitment of immune cells and proteins to the site of infection.

Among these are killer immune cells and antibodies that specifically neutralise pathogen-associated molecules called antigens. Once the pathogen is eliminated, other immune cells play a key role in restoring normal conditions by promoting tissue repair processes.

In autoimmune disorders, rogue immune cells fail to recognise healthy tissue as self and attack it. Among such disorders are psoriasis, lupus, rheumatoid arthritis, multiple sclerosis, Crohns disease and type I diabetes. Overall, they affect 5 per cent of the population in industrialised countries.

Autoimmune responses resemble normal immune responses but are initiated by self-antigens rather than pathogen-associated antigens. The human body has evolved a multitude of checkpoints that prevent the immune system from attacking healthy tissue. Together, these checkpoints must act concertedly to strike a delicate balance of dampening autoimmune responses while maintaining the ability of the immune system to strike against pathogenic infection. Indeed, it is quite common to observe isolated breakdown of one of these checkpoints in healthy individuals without any clear consequences.

Autoimmunity occurs when there is a collapse of enough checkpoints resulting in sustained reaction to self-antigens ultimatelyleading to tissue damage. In autoimmunity, self-antigens are not eliminated by the immune system. The constant presence of self-antigen leads to a salient feature of autoimmune disorders called chronic (sustained) inflammation, which leads to a continuing self-destructive process. Both auto-antibodies and killer cells directed at self-antigens participate in this immune mediated tissue damage.

The focus of my research is, therefore, directed at understanding how immune cells recognise antigens and communicate effectively with one another. Understanding these events is relevant in developing future therapies where the immune system plays a central role and intervention can help balance its response to disease.

David Saliba is a senior lecturer in the Department of Applied Biomedical Sciences, Faculty of Health Sciences at the University of Malta and also holds an honorary research fellow position at the Kennedy Institute of Rheumatology at the University of Oxford.

Identifying the cause of auto-immune disorders. While we understand the complex mechanisms that result in autoimmunity, intense research by the scientific community is directed at identifying the root cause of each of these disorders. In recent years, it has become clear that both genetic and environmental factors contribute to the development of autoimmune disorders. Genetic studies have identified multiple genes that contribute to increased susceptibility to autoimmune diseases. On the other hand, it is much more difficult to identify environmental influences. This is partly due to the chronic nature of these disorders. It is hard to pinpoint exactly when the autoimmune response began and therefore challenging to determine the single or combination of environmental triggers.

Imbalance in gut flora linked to Rheumatoid arthritis. The intestinal gut flora is an intricate world of microbes that has important implications in proper development of the immune system. Alterations of non-pathogenic microbial populations are often associated with immune disorders, particularly of the autoimmune kind. Rheumatoid arthritis is a debilitating disease since it primarily affects the hands, wrists and synovial joints. Scientists have compared faecal microbes of healthy individuals to rheumatoid arth-ritis patients. These studies have evidenced a clear shift in microbial populations demonstrating that specific microbial species may be responsible for the onset and progression of rheumatoid arthritis. It will be exciting to see how this field shapes the development of novel therapeutic approaches and biological markers of this disease.

For more soundbites listen to Radio Mocha every Saturday at 7.30pm on Radju Malta and the following Monday at 9pm on Radju Malta 2 https://www.fb.com/RadioMochaMalta/

To test the first barcode design, IBM employees hired a softball pitcher to fling one attached to a bean bag as fast as he could past a scanner.

Word of the Day: Cryptophasia when twins develop a language that only they can understand.

The board game The Campaign for North Africa is famously complex. It has 1,800 pieces, three volumes of rules and takes teams of five 1,500 hours to complete.

Jingle Bells was originally a Thanksgiving song.

Online porn has the same carbon footprint as the whole of Belgium.

Drinking alcohol with a diet mixer gets you 18 per cent more drunk than regular mixer.

For more trivia see: http://www.um.edu.mt/think

See the original post here:
Autoimmune disorders when the body attacks itself - Times of Malta

Read More...

Page 38«..1020..37383940..50..»


2025 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick