Category Archives: Immunology

Combining an HIV vaccine with immunotherapy may reduce the need for daily medication – The Conversation US

The Research Brief is a short take about interesting academic work.

A new combination treatment for HIV can strengthen a patients immune response against the virus even after they stop taking traditional medications, according to a study published in the journal Science Immunology we co-led at the Amara Lab at Emory Univeristy.

People with HIV take a combination of HIV medications to reduce the amount of virus they have in their body. When taken as prescribed, these medications, collectively called antiretroviral therapy, can reduce the amount of virus in the body to undetectable levels. Antiretroviral therapy must be taken daily so the virus is less likely to mutate and become resistant to the drugs.

While reducing the amount of virus in the body to undetectable levels means it can no longer be transmitted, however, the most effective antiretroviral therapy drugs are unable to completely eliminate the virus. This is because HIV hides in immune-privileged areas of the body, such as certain parts of the lymphoid tissue, that are less accessible to the immune system to protect them from damage. Killer T cells, which search for and eliminate infected cells, are unable to patrol these viral reservoirs that harbor HIV.

Constant exposure to the virus can push killer T cells into a state of exhaustion in which they dont work as well. Exhausted killer T cells display more of a protein called PD-1, which functions as an off switch to its killing activity.

One way to reverse killer T cell exhaustion is to block the PD-1 off switch, but this does not boost the immune systems response to the virus. Conversely, an HIV vaccine can significantly boost immunity against the virus.

So we tested whether combining these two tactics could enhance HIV infection control. We administered a vaccine for SIV, a close cousin to HIV, with a drug that blocks PD-1 in SIV-infected rhesus monkeys treated with antiretroviral therapy.

We found that our approach generated robust anti-viral response in multiple parts of the body, including immune-privileged sites in the lymph nodes, and allowed killer T cells to infiltrate and purge viral reservoirs. Most importantly, the monkeys maintained strong immunity against the virus even after they stopped antiretroviral therapy and significantly improved their survival. None of the seven monkeys in the combination treatment group developed AIDS through our six-month follow-up period, compared with half of the monkeys who received only the vaccine or antiretroviral therapy alone.

Around 38 million people worldwide were living with HIV in 2020. If left untreated, HIV can cripple the immune system and leave the body vulnerable to normally harmless infections.

There are accessibility issues with the treatment that must be diligently taken every day for life. A 2015 study estimated that the lifetime antiretroviral therapy cost for someone who acquires HIV at age 35 is US$358,380. And many people dont have access to daily antiretroviral therapy. Around three-quarters of adults with HIV in sub-Saharan Africa do not reach persistent viral suppression due to lack of treatment availability.

Finally, even though antiretroviral therapy can thoroughly suppress HIV infection, it does not cure it. There is always a risk that the virus may mutate to become resistant to existing drugs.

Completely wiping out HIV from the body is one way to eliminate the need for daily antiretroviral therapy. But a more achievable strategy is to put the infected cells in check.

Currently, only 0.5% of HIV positive individuals are considered elite controllers who are able to suppress infection without medication.

While our study showed a potential pathway to control HIV, it is still in development and not ready for human patients. More research is necessary to understand how viral reservoirs form and why certain cells respond differently to different immunotherapies.

A single form of therapy may not result in complete HIV remission. Our team is currently testing other drug combinations to unleash the full potential of the immune system and overcome barriers to a cure.

Read this article:
Combining an HIV vaccine with immunotherapy may reduce the need for daily medication - The Conversation US

NIH awards Brown $10.8M to expand data-informed research to fight human disease – Brown University

PROVIDENCE, R.I. [Brown University] Five years after an $11.5 million federal grant launched the COBRE Center for Computational Biology of Human Disease at Brown University, the National Institutes of Health has awarded $10.8M in new funds to Brown to build on the centers early success.

The center a federal Center of Biomedical Research Excellence funded by the NIHs National Institute of General Medical Sciences uses sophisticated computer analyses to advance research aimed at understanding and fighting human diseases.

Director David Rand, a professor of biology at Brown, said the renewal funds will enhance the centers research infrastructure, enable strengthened collaboration among scientists working with computational and bioinformatics tools, and support four new research projects. Rand said there is a computational revolution happening in the biomedical sciences, as researchers need computational analyses to help them make sense of massive amounts of available data.

Even those working in wet labs or clinics who dont use computers in their daily work will at some point need assistance in analyzing complex data sets, he said.

Rand compared the current moment to the molecular biology revolution thats been changing science since the 1970s, when DNA cloning and sequencing became standard tools used by researchers across diverse fields. Computational analysis is bringing groups together today in a similar way, he said. For example, people working in engineering, computer science, basic biology and medicine will face situations where they need to convert data sets into information that can help them find solutions and answer questions. While their research projects are highly distinct, he said, the data analysis work shares common themes.

In addition to helping researchers with individual projects, we view the Center for Computational Biology of Human Disease as a vehicle for raising the level of computational ability for researchers in the community overall, Rand said.

To provide that service to COBRE project leaders and researchers across Brown, the center is home to a Computational Biology Core a group of four scientists, data analysts and software engineers who support data-intensive research. With the renewal grant, center leaders will work to build sustainable support for the group through continued funding to its scientists and support to ensure that four members are at the Ph.D.-level (past budget included support for two Ph.D.s and two masters-level scientists).

Everyone has large data sets and needs to convert these into useful information, and we aim to help people achieve that goal, Rand said. The center brings together researchers in the lab and clinic with exceptionally skilled and creative data scientists to turn data into information.

Funds from the grant will also support the research of junior faculty investigators and help position them to earn additional, longer-term funding for their work enabling them, Rand said, to build upon discoveries and continue their research while freeing up center funds to seed innovative new projects. With the initial $11.5 million from the NIH in the centers first phase, faculty projects at the Center for Computational Biology of Human Disease generated an additional $17.9 million in grants in areas of research such as human genomics, immunology and infectious disease, microbiome and machine learning approaches to complex genetics.

More here:
NIH awards Brown $10.8M to expand data-informed research to fight human disease - Brown University

Society for Immunotherapy of Cancer Publishes Clinical Practice Guideline on Immunotherapy for the Treatm – Benzinga

The Society for Immunotherapy of Cancer (SITC), the world's leading member-driven organization dedicated to improving cancer patient outcomes by advancing the science and application of cancer immunotherapy, is pleased to announce the publication of the first clinical practice guideline (CPG) focused on immunotherapy for the treatment of hepatocellular carcinoma (HCC).

MILWAUKEE (PRWEB) October 05, 2021

The Society for Immunotherapy of Cancer (SITC), the world's leading member-driven organization dedicated to improving cancer patient outcomes by advancing the science and application of cancer immunotherapy, is pleased to announce the publication of the first clinical practice guideline (CPG) focused on immunotherapy for the treatment of hepatocellular carcinoma (HCC).

The new manuscript, "Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of hepatocellular carcinoma", was published in the Journal for ImmunoTherapy of Cancer (JITC), SITC's peer-reviewed online journal, and it will be available in SITC's mobile guidelines app.

"Immunotherapy has become the standard of care for advanced HCC" said Ignacio M. Melero, co-Chair of the SITC HCC Immunotherapy Guideline Expert Panel. "Patients with advanced HCC have more options than ever and this new guideline will be an essential resource for clinicians to navigate the landscape to develop immunotherapy treatment plans."

In 2020, the checkpoint inhibitor immunotherapy atezolizumab was approved by the U.S. Food and Drug Administration in combination with bevacizumab as a first-line treatment for HCC, representing the first new regimen to improve overall survival for patients in more than a decade. Additional immune checkpoint inhibitors are also approved in the second-line setting as well, and ongoing trials are evaluating additional immunotherapeutic strategies. To provide guidance to clinicians treating HCC on how to incorporate these new options into the evolving therapeutic landscape, SITC convened a panel of leading experts with perspectives from oncology, hepatology, surgery, interventional radiology, nursing and patient advocacy.

"HCC always develops as disease on top of a disease including both the cancer and underlying liver damage," said Tim F. Greten, co-chair of the SITC HCC Immunotherapy Guideline Expert Panel. "Our expert panel included leading voices from hepatology and oncology to provide clinicians with the recommendations they need on both liver-specific and the immunotherapy-specific aspects of HCC treatment."

To support clinicians in their decision-making with their patients with HCC, the Hepatocellular Carcinoma Immunotherapy Guideline Expert Panel developed evidence- and consensus-based recommendations on topics including the selection of appropriate immunotherapeutic regimens, recognition and management of immune-related adverse events, and patient quality of life considerations.

"SITC's clinical practice guidelines are the authoritative resource for recommendations on the optimal use of immunotherapy to improve patient outcomes." said SITC President Patrick Hwu, MD. "The society is proud to have brought together this expert panel of international leaders in HCC to develop this timely and important guideline."

In addition to the published manuscript, SITC is also offering a number of different opportunities to help clinicians understand and implement the guidelines into their practice. One such resource is free live webinars and on-demand modules hosted on the SITC website that will focus on this published manuscript and provide attendees with the opportunity to ask questions of expert faculty:

Click here to view past SITC Cancer Immunotherapy Guidelines webinars on-demand.

About the SITC Cancer Immunotherapy Guidelines

The hepatocellular carcinoma immunotherapy guideline is part of the broader SITC Cancer Immunotherapy Guidelines program that has produced a collection of CPGs developed by multi-disciplinary panels of experts who draw from their own practical experience as well as evidence in the published literature and clinical trial data to develop evidence- and consensus-based recommendations on when and how to use immunotherapy to help improve outcomes for patients with cancer. SITC uses as a model, the Institute of Medicine's 2011 "Standards for Developing Trustworthy Clinical Practice Guidelines" to ensure the recommendations are unbiased, transparent and balanced and aids oncologists in effective clinical decision-making concerning patient selection, toxicity management, response evaluation, and the sequencing or combination of therapies, among other topics.

About SITC

Established in 1984, the Society for Immunotherapy of Cancer (SITC) is a nonprofit organization of medical professionals dedicated to improving cancer patient outcomes by advancing the development, science and application of cancer immunotherapy and tumor immunology. SITC is comprised of influential basic and translational scientists, practitioners, health care professionals, government leaders and industry professionals around the globe. Through educational initiatives that foster scientific exchange and collaboration among leaders in the field, SITC aims to one day make the word "cure" a reality for cancer patients everywhere. Learn more about SITC, our educational offerings and other resources at sitcancer.org and follow us on Twitter, LinkedIn, Facebook and YouTube.

About JITC

Journal for ImmunoTherapy of Cancer (JITC) is the official open access, peer reviewed journal of the Society for Immunotherapy of Cancer. The journal publishes high-quality articles on all aspects of tumor immunology and cancer immunotherapy, on subjects across the basic science-translational-clinical spectrum. JITC publishes original research articles, position papers and practice guidelines, and case reports; invited and pre-vetted reviews and commentaries are also considered by the journal editors. These articles make JITC the leading forum dedicated to tumor immunology and cancer immunotherapy research.

For the original version on PRWeb visit: https://www.prweb.com/releases/society_for_immunotherapy_of_cancer_publishes_clinical_practice_guideline_on_immunotherapy_for_the_treatment_of_hepatocellular_carcinoma/prweb18242472.htm

See the article here:
Society for Immunotherapy of Cancer Publishes Clinical Practice Guideline on Immunotherapy for the Treatm - Benzinga

New discovery reveals direct link between opioids and gut inflammation – New Atlas

Intriguing new preclinical research from a team of Japanese scientists has found targeting opioid receptors in the gut could be a new treatment for inflammatory bowel disease. The research builds on early evidence showing a relationship between opioids and immune cell function.

For several years scientists have noticed links between opioid use and immune suppression. Observational studies have noted some hospital patients treated with opioids were more vulnerable to viral infections, while in vitro research has demonstrated opioids can influence immune cell function. But exactly what is going has been unclear.

There has long been a noted connection between opioids and the gut, but only relatively recently have researchers discovered opioid receptors are not isolated to the brain. This new research set out to better understand how opioids interact with the immune system by focusing on a mouse model of inflammatory bowel disease.

The researchers focused on a previously developed drug called KNT-127, which is designed to selectively stimulate delta opioid receptors only. Drugs specifically targeting delta opioid receptors have recently been receiving plenty of research attention in the hope they work effectively as pain relievers without negative side effects such as respiratory depression or dependency often seen with currently available opioid drugs.

The new study, published in the journal Frontiers in Immunology, first revealed KNT-127 effectively reduced the severity of colon inflammation in several mouse models. A number of different experiments showed the experimental opioid directly suppressing immune cell activity.

Nagata et al (2021), Frontiers in Immunology

But a key question remained was the drug exerting its anti-inflammatory effects by directly activating delta opioid receptors in the gut, or were receptors in the brain playing a role?

To answer this question the researchers looked to a different version of KNT-127 designed to be unable to cross the blood-brain barrier. Similar results were seen in the mouse models, affirming the anti-inflammatory activity of the drug was independent of any central nervous system (CNS) actions.

"Several people around the world suffer from diseases related to colon inflammation, and so far, optimal treatment strategies are lacking, says Hiroshi Nagase, an author on the new study. Our findings show that KNT-127 and other activators of opioid receptors could be promising therapeutic options for such diseases.

Nagase does stress there is plenty more work needed before these findings can be translated into a clinical treatment for human patients. However, the study offers some of the first clear evidence of opioid receptors in the gut being involved directly in inflammatory responses.

Chiharu Nishiyama, from the Tokyo University of Science and lead researcher on the new study, says these findings offer compelling insights into gut-brain connections.

"Today, we know that poor mental health has physical manifestations, explains Nishiyama. For example, stress worsens inflammation in the gut, which in turn affects the health of the brain. Our results on the immune-related effects of opioids, which commonly act on the brain, is a step toward unraveling the biological mechanisms that govern the reciprocative relationship of gut health and the immune system with the CNS.

The new study was published in the journal Frontiers in Immunology.

Source: Tokyo University of Science

View post:
New discovery reveals direct link between opioids and gut inflammation - New Atlas

COVID-triggered autoimmunity may be mostly temporary – Emory Health Now

In people with severe COVID-19, the immune system goes temporarily berserk and generates a wide variety of autoantibodies: proteins that are tools for defense, but turned against the bodys own tissues.

During acute infection, COVID-19 patients immune systems resemble those of people with diseases such as lupus or rheumatoid arthritis. However, after the storm passes, the autoantibodies decay and are mostly removed from the body over time, according to a study of a small number of patients who were hospitalized and then recovered.

In apreprint posted on medRxiv, Emory immunologists provide a view of the spectrum of what COVID-generated autoantibodies react against, both during acute infection and later. Note: the results have not yet been published in a peer-reviewed journal.

The findings on COVID-19-triggered autoimmunity may have implications for both the treatment of acute infection and for long-haulers, in whom autoantibodies are suspected of contributing to persistent symptoms such as fatigue, skin rashes and joint pain.

During acute infection, testing for autoantibodies may enable identification of some patients who need early intervention to head off problems later. In addition, attenuation of autoantibody activity by giving intravenous immunoglobulin (IVIG) an approach that has been tested on a small scale may help resolve persistent symptoms, the Emory investigators suggest.

Researchers led by Ignacio Sanz, MD and Frances Eun-Hyung Lee, MD, isolated thousands of antibody-secreting cells from 7 COVID-19 patients who were in ICUs at Emory hospitals. They also looked for markers of autoimmunity in a larger group of 52 COVID-19 ICU patients.

Sanz is head of the division of rheumatology in the Department of Medicine, director of the Lowance Center for Human Immunology, and a Georgia Research Alliance Eminent Scholar. Lee is associate professor of medicine and director of Emorys Asthma/Allergy Immunology program.

The co-first authors are Matthew Woodruff, PhD, an instructor in Sanzs lab, and Richard Ramonell, MD, a fellow in pulmonary and critical care medicine at Emory University Hospital. In some of the experiments, Emory researchers collaborated with Exagen, using technology developed by the company to probe clinical autoimmune profiles.

We were trying to address the question: what is the origin of the autoantibodies? Sanz says.

That is, are they generated during the course of infection? Or were immune cells capable of making the observed autoantibodies already existing in the body, and let loose by the infection?

These possibilities are not mutually exclusive, but it appears that new generation of autoreactive clones from nave cells is a dominant mechanism in acute severe infection, he says. What we see is a broad breakdown of tolerance, at least temporarily.

The researchers characterized the antibodies the cells produced and their DNA, looking for signs of the editing processes the immune system usually applies to ensure antibodies dont react against the body itself.

The editing processes appeared to have weakened during infection. Germinal centers, the anatomical structures within lymph nodes where antibody-secreting cells mature, can collapse during intense inflammation, Sanz says. As a result, antibody-secreting cells develop in a disorganized, uncontrolled way outside highly regulated germinal centers.

In the hospitalized patients, the majority of their antibody-secreting cells produced antibodies directed against parts of the coronavirus, but some of them were also reactive against different targets in the body. The broad pattern resembled that seen in lupus, but was different in that some types of autoantibodies, such as those against DNA, were not observed. One patients immune cells were producing antibodies against glomerular basement membrane, which may contribute to lung and kidney damage.

Six months after acute infection, when some of the patients had survived and recovered, most of the indiscriminately generated antibody-secreting cells had disappeared from their blood. However, the antibodies themselves what the cells produce remain in the blood and decay gradually over time.Approaches such as IVIG could encourage recycling of lingering autoantibodies, the authors write.

Sanz says that the question of whether autoantibodies or perhaps, lingering autoreactive cells contribute to persistent symptoms needs to be tested in a larger group of COVID-19 patients. His and Lees laboratories continue to examine people with severe COVID-19 and those who recovered, as well as outpatients, including those with long haul symptoms. Their expanded study now includes an additional 150 patients, with the aim of correlating clinical features with the presence of autoantibodies.

This is something we are going to have to follow, he says. We also need to compare this to other viral infections that have been associated with autoimmunity.

The research was supported by the National Cancer Institute (SeroNet, U54CA260563), the National Institute of Allergy and Infectious Diseases (U19AI110483 Emory Autoimmunity Center of Excellence, P01AI125180, R37AI049660, R01AI121252, U01AI141993), and the National Heart Lung and Blood Institute (T32HL116271).

Additional links:

2020 Nature Immunology paper on B cell responses in severe COVID-19

Matthew Woodruffs explainer in The Conversation

Emory Health Digest article on long COVID

Emory expanding post-COVID care

Panel discussion (scroll down) on long COVID

Science Writer, Research Communicationsqeastma@emory.edu404-727-7829 Office

More:
COVID-triggered autoimmunity may be mostly temporary - Emory Health Now

EXTENDED INTERVIEW: Marc Jenkins Director of Immunology at the University of Minnesota Medical School – KARE11.com

Two studies show evidence that natural immunity can last at least a year against the Alpha variant, but the strength of immunity is unclear against other variants.

Author: kare11.com

Published: 7:32 PM CDT July 25, 2021

Updated: 7:32 PM CDT July 25, 2021

Follow this link:
EXTENDED INTERVIEW: Marc Jenkins Director of Immunology at the University of Minnesota Medical School - KARE11.com

Infinitus Co-Organizes the 11th National Conference on Immunology of Traditional Chinese Medicine – Yahoo Finance

GUANGZHOU, China, July 28, 2021 /PRNewswire/ -- The 11th National Conference on Immunology of Traditional Chinese Medicine (TCM) organized by the Immunology of Traditional Chinese Medicine Branch of the Chinese Society for Immunology (CSI) in partnership with Shandong University of Traditional Chinese Medicine was held in China's Shandong Province from July 21 to July 23, 2021. Co-organizer Infinitus (China) Company Ltd. attended the events held during the conference.

Dr. Zhu Ying, senior R&D engineer of the health food department of Infinitus, delivering a keynote speech at the event

As a national professional academic event in the field of immunology of TCM, the biennial conference plays a key role in TCM immunology research. As a company which has always been committed to research into and collaboration around the immunology of Chinese herbal medicines, Infinitus has co-organized several consecutive editions of the conference.

During the conference, immunology experts from across China gathered together to discuss a wide range of topics including immunopharmacology and pharmacodynamics of TCM, research on prevention and treatment of COVID-19 through incorporation of immunology of TCM into the process, immunology and TCM theory, immunology and acupuncture, as well as the technological development of immunology and its applications in TCM research.

At the conference, Dr. Zhu Ying, senior R&D engineer of the health food department of Infinitus, delivered a keynote speech entitled "R&D Strategy for Immunoregulatory Functional Food". During her speech, Dr. Zhu shared Infinitus' strategy and experience in developing health foods, which covered the areas of the elements of high quality products, the methods of R&D, the analyses of consumer expectations, as well as the selection and optimization of efficacy evaluation models.

Infinitus, the health industry player, plans to continue its efforts on the development of immunology of TCM, keep digging into the research on the Chinese herbal polysaccharides which have immunoregulatory functions, keep providing consumers with high quality health products, and help accelerate the growth of the health industry.

SOURCE Infinitus (China) Company Ltd.

Read the rest here:
Infinitus Co-Organizes the 11th National Conference on Immunology of Traditional Chinese Medicine - Yahoo Finance

Macomics, the Macrophage-based Drug Discovery company, secures 4.24m expansion financing, and appoints CEO, CSO and VP Immunology – Business Wire

EDINBURGH, Scotland & CAMBRIDGE, England--(BUSINESS WIRE)--Macomics Ltd, an immuno-oncology company with world-leading expertise in macrophage biology, announces today that it has closed a follow-on financing of 4.24 million from its 2020 Seed round, bringing the total amount raised to 7.44m.

The additional investment brings in new investor Caribou Property Limited alongside existing investors Epidarex Capital, who led the round, and Scottish Enterprise. It will be used to accelerate the companys growth, including expansion of the team.

Macomics is developing precision medicines to modulate macrophages for the treatment of cancer. The company was co-founded in 2019 by Prof Jeffrey Pollard and Dr Luca Cassetta, University of Edinburgh, internationally recognised leaders in macrophage biology. It is progressing a diversified portfolio of therapies targeting disease specific tumour associated macrophages (TAMs) towards the clinic. Its target discovery platform enables identification and validation of novel macrophage therapeutic targets and is based on its deep understanding of macrophage biology. The new investment and planned Series A will enable the company to accelerate progress of its antibody programs towards the clinic, expand its portfolio, and further invest in its target discovery technology.

Cancer cells are known to be able to evade destruction by the immune system and TAMs are a key component of this immuno-suppressive and pro-tumoral ecosystem. Modulating TAMs will alter the tumour microenvironment enhancing the bodys ability to fight cancer. The tumour microenvironment changes macrophage behaviour, and the company is exploiting disease specific TAM biology to selectively target these immunosuppressive cells.

Alongside the financing, the company announces the expansion of its leadership team with the appointment of Dr Myatt as Chief Executive Officer and Dr Ries as Chief Scientific Officer, as well as Dr Cassetta who will join as VP Immunology on 1 August.

Dr Steve Myatt, CEO who joined Macomics in February 2021 said:

Macomics has made great progress since formation, and I am excited to lead Macomics in driving forward its vision to become a leading immuno-oncology company pioneering macrophage-based therapies for the treatment of cancer. Our diversified portfolio of antibody programs combined with our proprietary target identification approach and world-class team uniquely positions us to deliver on this vision.

Dr Carola Ries, who joins as Chief Scientific Officer from 16 years at Roche added:

It is an exciting time to be joining Macomics. In recognising disease specific TAM populations Macomics is taking a new approach to macrophage-based drug discovery, one that I believe has potential to deliver significant benefit to patients. By combining analysis of human disease tissue, in silico data mining, and functional data from our state-of-the-art cellular models we are well placed to identify and validate new drug targets in diseases where macrophages play a central role. I look forward to applying my extensive industry experience gained at Roche to driving our programs towards the clinic.

Commenting on his full-time move into the company, Dr Luca Cassetta, said:

Macomics was founded based on leading academic research around macrophage biology, particularly in cancer, from the laboratory of Professor Jeffrey Pollard, Director of the MRC Centre for Reproductive Health at the University of Edinburgh, as well as my own research studying TAMs. I am delighted to be joining the company full-time to exploit the potential of macrophage-based approaches to developing novel immuno-oncology therapies that have the potential to change the lives of patients with cancer.

Dr Elizabeth Roper, Partner at Epidarex Capital and Investor Director at Macomics said:

We are delighted with the progress that Macomics has made since we made our initial investment last year. The company has demonstrated the power of its platform and approach and has already moved two of its programs into antibody discovery and identified a series of additional targets. We are excited about its potential, reflected in our increased commitment to the company.

She added On behalf of the board, I am pleased to welcome Steve, Carola and Luca to the team. Their experience in biotech, pharma and academia are highly complementary and broaden and deepen the skills of the leadership team.

Georges Aboud, Chief Investment Officer at Caribou Property, commented: "We are pleased to join the investment round, having seen the progress Macomics has made in just over 12 months. Our goal is to back exciting and innovative companies that can make an impact on patients' lives. Macomics has shown that its knowledge of macrophage biology can deliver innovative approaches to treat cancer and it has assembled an experienced team to maximise its potential."

Jan Robertson, Interim Director of Growth Investments at Scottish Enterprise, added: This funding round secured by Macomics is testament to the companys pioneering macrophage-based therapies for the treatment of cancer as well as the talent in Scotland. Scottish Enterprise is pleased to support this leading biotech firm that is now set to accelerate its research and development programmes, and expand its facilities in Edinburgh.

Macomics has expanded its R&D and office facilities on the Cambridge Science Park and has taken additional laboratory and cell culture space within Edinburgh University, and will be expanding its scientific team to support its accelerated R&D.

-Ends-

About Macomics http://www.macomics.com

Macomics Ltd is an immuno-oncology company with world-leading expertise in macrophage biology, developing precision medicines to modulate macrophages for the treatment of cancer. The company is progressing a diversified portfolio of therapies targeting disease specific tumour associated macrophages (TAMs) towards the clinic. Its target discovery platform enables identification and validation of novel macrophage therapeutic targets and is based on its deep understanding of macrophage biology.

Macomics was co-founded in 2019 by Prof. Jeffrey Pollard and Dr. Luca Cassetta, University of Edinburgh, internationally recognised leaders in macrophage biology. It has R&D and office facilities in Edinburgh and Cambridge, UK and is backed by Epidarex Capital, Scottish Enterprise, and Caribou Property Limited.

Follow us on LinkedIn and https://twitter.com/MacomicsL

About the new appointments

Dr Steve Myatt, Chief Executive Officer

Dr Myatt joined Macomics in February 2021 and previously served as Chief Executive Officer at Azeria Therapeutics, where he led the companys $44mn Series B financing. Prior, he was Partner at venture capital firm Sixth Element Capital, managing a $95mn oncology venture fund focussed on building early-stage companies and supporting the progression of therapeutic programs from discovery to early clinic. Successful fund exits include transactions with Novartis, Johnson & Johnson Innovation, Sierra Oncology, Boston Pharmaceuticals, Stemline Therapeutics, and Achilles Therapeutics (NASDAQ). Companies in which Sixth Element Capital invested as a founding investor have to date raised over $325mn. Steve began his career at Cancer Research Technology, responsible for identifying new academic drug discovery opportunities in Cambridge UK and was latterly Head of Alliances, Business Development, establishing multiple drug discovery partnerships between Pharma, biotech and academia. Steve has a first-class degree in Pharmacology and a PhD in paediatric oncology.

Dr Carola Ries, Chief Scientific Officer

Dr Ries joined Macomics in July 2021 and has over 20 years of immunology and drug discovery experience and is an internationally recognised expert in macrophage immunology. She formally led Roches innate immunity department in cancer immunotherapy and was a member of the pRED immunotherapy discovery leadership team. Dr Ries led the discovery of emactuzumab (CSF1R program) and contributed to its clinical translation, identifying a clinical population in which emactuzumab provided significant clinical benefit. Earlier in her career Dr Ries was a postdoctoral fellow in the lab of Dr. McCormick at UCSF, where she discovered her passion for oncology. Dr Ries has over 35 publications and is a named in inventor on over 10 patents.

Dr Luca Cassetta, VP Immunology and Founder

Dr. Cassetta is a co-Founder of Macomics and is an immunologist with in-depth knowledge of human myeloid cell biology applied to different human diseases. He obtained a PhD in immunology at the S.Raffaele Institute in Milan studying the role of macrophage polarization in HIV pathogenesis; he then moved to NY at the Albert Einstein Medical college in the lab of Prof. Jeffrey Pollard where he studied Tumor Associated Macrophages in breast cancer. He then moved to the University of Edinburgh continuing his studies on TAMs where he established his own lab as principal investigator. His extensive experience in human macrophage biology contributed to the development of the Macomics screening platform. Dr Cassetta is an author of multiple publications in high-impact international scientific journals including Cancer Cell, Nature Communications, Journal of Experimental Medicine, Blood, PNAS, Nature Reviews Drug Discovery. Dr Cassetta joins Macomics in August 2021.

Original post:
Macomics, the Macrophage-based Drug Discovery company, secures 4.24m expansion financing, and appoints CEO, CSO and VP Immunology - Business Wire

Study Shows Improved Tolerance with Second COVID-19 Vaccine – MD Magazine

Ever since the COVID-19 vaccines were approved for the public population, conversations have persisted regarding their efficacy, especially regarding people who had immediate allergic reactions with the first dose.

Despite this, a multicenter, retrospective study confirmed that most patients with immediate and potentially allergic reactions to the COVID-19 vaccines tolerated the 2nd dose.

In an interview with HCPLive, 2 of the leading authors of the study, Kimberly Blumenthal, MD, MSc, Quality and Safety Officer for Allergy, Massachusetts General Hospital, Co-Director, Clinical Epidemiology Program, Division of Rheumatology Allergy and Immunology and Matthew Krantz, MD, Clinical Fellow, Allergy/Immunology, Vanderbilt University Medical Center spoke of the genesis of the study as well as the data gathered on reactions from participants.

Prior to the study, the team targeted healthcare workers who had received some of the first doses of the Pfizer-BioNTech and Moderna vaccines.

One of the unexpected observations early on after emergency use authorization in December of 2020 where we primarily targeted vaccinating healthcare workers was we did observe a higher rate of immediate potentially allergic reactions and anaphylaxis than with traditional vaccines, Krantz said.

Krantz, Blumenthal and colleagues set out to find if participants in the study were truly allergic to the COVID-19 vaccines.

If you're allergic to something, you have an IgE antibody, [so] you have to avoid it, Blumenthal said. Thats really important for mRNA vaccines: were people forming true, classic allergy to these vaccines?

From January 1, 2021-March 31, 2021, the investigators enrolled 189 patients from participating centers, all of whom recorded an immediate reaction to their first Pfizer-BioNTech or Moderna vaccine.

Patient population demographics showed that the mean age of all patients was 43 years, and that most of the total population (86%) were women. Of the vaccine first-dose reactions, 130 (69%) were Moderna and 59 (31%) were Pfizer-BioNTech.

Allergic reactions were defined as symptom onset within 4 hours of dose 1, at least 1 allergic symptom, and referral for an allergy/immunology consultation with in-clinic or tele-heath assessment.

Anaphylaxis was scored using the Brighton and the National Institute of Allergy and Infectious Diseases/Food Allergy and Anaphylaxis Network Criteria.

Blumenthal, Krantz and colleagues defined second dose tolerance as 1 of the following:

1. No immediate symptoms after second dose administration

2. Symptoms that were mild, self-limited and/or resolved with antihistamines

Phone calls regarding clinical details were made for individuals who did not have their second dose observed by allergy/immunology departments.

The investigators noted that the most frequently reported first-dose reactions in patients were flushing or erythema (28%), dizziness or lightheadedness (26%), tingling (24%), throat tightness (22%), and hives (21%), and 32 participants (21%) met the anaphylaxis criteria.

Encouragingly, 159 patients as well as 19 individuals who experienced first-dose anaphylaxis tolerated the second dose of the COVID-19 vaccine. Only 20% of participants reported immediate and potentially allergic symptoms associated with the second dose, which were either self-limited, mild, or resolved with antihistamines alone.

Additionally, most of the participants in the study (89%) received the second dose, and 47 patients (30%) were given an antihistamine premedication before the second dose.

It's important to note that in terms of classic IgE allergy, we do not expect premedications to abort a reaction, Krantz said. But, in the case of a non-IgE mediated reaction, antihistamines can also blunt or make these reactions (milder).

The effectiveness of the drugs on some patients supported the hypothesis that the participants did not have to avoid the COVID-19 vaccines, as they did not have whats considered a classic allergy response.

Blumenthal also noted the effectiveness of the premedication and clarified that the study was not intended to recommend a specific medication, as each patient was treated with individual methods depending on what their allergist felt was appropriate.

Everybody had a different [recommendation], Blumenthal said. This study wasn't designed to test how best to get from a dose 1 immediate reaction to safe dose 2 completion. But it gives us a hint that it's largely possible.

Blumenthal noted that further research might explore the possibilities of a specific premedication or treatment methods for patients who had a reaction to the COVID-19 vaccines, especially considering the possibilities of COVID-19 vaccine boosters in the coming fall season.

What we really need is a prospective study when it comes to the booster doses, Blumenthal said. So, prospective, meaning that we enroll people who had reactions before (and) who didn't have reactions, and we have them diary their medication so that we can piece out what premedication regimen might work.

Blumenthal and Krantz both noted the potential for improvement in identifying allergic reactions in patients regarding the COVID-19 vaccines and general vaccines.

The study, Safety Evaluation of the Second Dose of Messenger RNA COVID-19 Vaccines in Patients With Immediate Reactions to the First Dose, was published online in JAMA Internal Medicine.

Continued here:
Study Shows Improved Tolerance with Second COVID-19 Vaccine - MD Magazine

Ireland getting "very close to the end of the pandemic", says Immunology Professor – JOE.ie

Immunology Professor Paul Moynagh has said that Ireland is getting "very close to the end of the pandemic" and closer to moving to an "endemic phase".

Moynagh, who is Professor of Immunology at Maynooth University, said that while the virus will still be "circulating in the background", most people will be "protected" via vaccination or natural immunity.

Moynaghtold Newstalk Breakfast that things could be taking a positive turn in spite of increasing case numbers over the last number of weeks.

A number of weeks ago, the projections were not very good, we were hearing very large numbers," he said.

Now what we seem to be seeing is a decrease in the day-to-day increase we had been seeing. The percentage of those cases ending up in hospital is going down the duration of hospital stays is going down.

He added that Ireland and other countries in the UK and Europe could be getting close to the "end of the pandemic phase".

I think were getting to the stage where a number of countries like ourselves, the UK and most countries in Europe - are getting very close to the end of the pandemic phase, and will probably move more into an endemic phase," he added.

The virus will be circulating in the background, but thankfully most of us will be protected by either vaccination or natural immunity through infection.

Moynagh's comments come as 1,408 cases of Covid-19 were reported across Ireland on Wednesday, just 2% of which were hospitalised cases.

Chief Medical Officer (CMO) Dr Tony Holohan said on Wednesday that Ireland might be in a position "in the coming weeks" to move away from some economic and social Covid-19 restrictions.

During a Department of Health press briefing he said: "If we keep pushing on with the kinds of uptake rates we've seen in some of the older age groups, which by international standards are some of the best in the world, that gives us a lot of reason for optimism that the conditions that we think will need to be satisfied to allow us to move away from some of the restrictions that still remain in place could be met."

Read more from the original source:
Ireland getting "very close to the end of the pandemic", says Immunology Professor - JOE.ie