This website is intended for healthcare professionals only.

Hospital Healthcare Europe
Hospital Pharmacy Europe     Newsletter       

Press Releases

Take a look at a selection of our recent media coverage:

New technology could lead to quicker diagnoses of infectious diseases

15th January 2019

 
Late diagnosis of blood-borne viruses (BBV), such as HIV and hepatitis, remains high in the UK despite efforts to increase testing among those at risk of infection.
 
Experts at Teesside University and South Tees Hospitals NHS Foundation Trust have developed a new software application to assist GPs and health-care workers to make decisions about BBV- testing in real time.
 
The technology is currently undergoing pilot testing in several GP surgeries and, if successful, could ultimately lead to quicker diagnosis and treatment, improved patient outcomes and also reduce transmission of BBVs.
 
There is often a reluctance for GPs to offer BBV-testing due to issues around the process of consent. It is also a time consuming process as GPs have to examine each patient’s individual medical records before being able to make an informed decision.
 
Concerns have been raised that when patients arrive at hospitals, there are regularly cases where BBV-testing should have been picked up earlier.
 
The team of researchers and experts, led by Dr David Chadwick, Consultant in Infectious Diseases at South Tees Hospitals NHS Foundation Trust, Professor Paul Van Schaik, Professor of Psychology at Teesside University and Dr Gareth Forbes, a GP at Leadgate Surgery in Consett, have developed a software module which can be applied to the database of existing medical records. It provides an automatic prompt to GPs and other health-care workers each time a BBV test should be recommended for a patient, speeding up the process and potentially enabling quicker diagnosis and treatment.
 
The decision to test for BBV is still in the hands of the GP, but the technology provides an automatic prompt based on existing medical information, rather than having to read through individual patient records.
 
The technology has recently won the Medipex NHS Innovation Award for Primary Care, as well as the Bright Ideas in Health Award for development of an innovative device or technology.
 
Professor Van Schaik said: “We are trialling the technology at the moment and will then need to compare the results for BBV-testing with and without this technology. The rationale is that it has the potential to speed up the testing and diagnosis of infectious diseases – improving patient outcomes and potentially saving lives.”
 
Professor Van Schaik has researched and published in areas around technology acceptance for over 20 years. His main role in this project is to test the acceptance of the new system from patients and health-care workers.
 
The two main factors with any new technology are usability and usefulness”, explained Professor Van Schaik.
 
Is it easy to use? Will GPs be able to make better and more informed decisions? Will it improve patient outcomes? The data from the pilot testing should provide the answers to these kinds of questions.
 
The impact, if successful, would be the take-up of this technology across the country and improved outcomes for patients. It would be a relatively small addition to the existing system that has the potential to make a significant difference.”
 
Dr Chadwick is Research Lead for Infectious Diseases at South Tees Hospitals NHS Foundation Trust and has an interest in HIV and approaches to reduce late diagnosis of BBVs.
 
He said: “We hope this innovative technology will reduce late diagnosis of HIV, along with viral hepatitis. As well as being a great accolade for South Tees, it also recognises the important contribution of my research colleagues at The James Cook University Hospital, in primary care and at Teesside University.”

Could the gut microbiome help prevent the development of cow’s milk allergy?

Scientists at the University of Chicago have found that gut microbes from healthy human infant donors transplanted into mice protected animals exposed to milk from experiencing allergic reactions, while gut microbes transplanted from infants allergic to milk did not.

The study, published in the journal Nature Medicine, was supported in part by NIH’s National Institute of Allergy and Infectious Diseases.

Researchers transplanted gut microbes from each of eight infant donors into groups of mice raised in a sterile environment and sensitised to milk protein — meaning the animals’ immune systems created allergic antibodies to milk.

When later exposed to milk, mice receiving no microbes or microbes from milk-allergic children produced allergic antibodies and experienced anaphylaxis. Mice receiving gut microbes from non-allergic infants had no reactions.

Investigators then analysed microbes in infant stool samples, finding many differences between the stool of infants who were allergic to milk and those who were not.

Mice transplanted with microbes from non-allergic infants also harboured a family of microbes previously found to protect against developing food allergies. Further experiments identified one microbe, Anaerostipes caccae, that prevented the development of milk allergy when transplanted alone into groups of mice.

Researchers then sampled cells along the mice’s gut linings — where food allergies in mice and humans begin to develop. They found that mice that received microbes from non-allergic infants expressed different genes compared to those that did not, suggesting that microbes residing in the gut impact the host’s immune system.

The researchers conclude that intestinal microbes play a critical role in regulating allergic responses to food and suggest that further research could lead to microbiome-modifying therapies to prevent or treat food allergy.

Fulfilling a promise to Mosul with point-of-care ultrasound

8th January 2019

Dr Henryk Pich spent time travelling in the Middle East as a young man. Not surprisingly, he feels a personal connection to the region and, after seeing the devastation caused by years of war to cities such as Mosul, wanted to offer whatever help he could.

He contacted CADUS, an independent German aid organisation building mobile clinics for areas with significant need and, once the fighting had ended, joined a highly skilled, inter-disciplinary team of paramedics, nurses, doctors, technicians and project managers working in Mosul.

His first trip centred on a refugee camp outside of Mosul providing primary healthcare to the hundreds of thousands of refugees displaced from their homes, from where the team visited the specialist women and children’s teaching hospital in Mosul, the Al Khansaa Hospital.

The experience formed a lasting impact on Dr Pich: “The Al Khansaa used to be one of the biggest hospitals in Northern Iraq – with 300 beds for women and children’s services and a high quality reputation in the wider region – yet 85 % of the hospital and its resources had been destroyed or stolen. The staff are very well trained, but there is a lack of equipment. There is a lot of improvisation and many things have to run at a rudimentary level. I naturally expressed my concern and desire to help as much as I could and pledged to return.”

Dr Pich is passionate about ultrasound, particularly in the specialties of regional anaesthesia and emergency medicine, and uses his skills to educate others whenever possible. He explained: “Point-of-care ultrasound is a vital resource for intensivists and emergency physicians around the world. It enables a quick and accurate diagnosis, guides life-saving interventions, and operates as the ‘eyes and hands’ of modern-day doctors. The reliability, durability and portability of some ultrasound systems make them ideally suited to use in war-torn destinations, and it made sense to me to return with an appropriate ultrasound machine and training resources.”

The potential impact of a new point-of-care ultrasound system in the Mosul hospital could be far-reaching, and so Dr Pich approached FUJIFILM SonoSite for support for his return mission and a SonoSite M-Turbo system was donated. Dr Pich explained: “I know this machine well and was very happy to take it to Mosul. It is small, light and easy to carry in one hand. It has excellent probes of various types that are resilient and reliable, and the whole system is intuitive to use and easy to teach to others. In a critical care setting, time is of the essence and this ultrasound machine boots up within a few seconds and is ready for use straightaway.”

With the ultrasound system arranged, Dr Pich had clear objectives for his next visit – to hand over the machine and, alongside an Arabic translator, deliver a training course in regional anaesthesia and emergency ultrasound. “In Germany I carry out a lot of FAST (focused assessment with sonography for trauma) scans, and planned to teach this technique to my Iraqi colleagues, as well as lung scans and guidance for nerve blocks.”

Dr Pich was eager to see how life had changed for the people of Mosul since his visit the previous year: “There is some life on the streets again, security has greatly improved and, although there is still a lack of basic materials in the hospital, staff continue to work there with enthusiasm and dedication. It was a pleasure to hand over the M-Turbo and deliver the training course to surgeons and radiologists. I talked about regional anaesthesia, guiding needle insertion, and how to interpret views of structures and identify nerves; I had a practice pad and needles with me, so they could each experience a simulated exercise. Then one radiologist on the course was very keen to use it to guide her in taking biopsies of breast tissue, which was an application of the device that I hadn’t foreseen. As soon as the course was over, the Iraqi attendees took the M-Turbo to the emergency room to assess a woman with a hernia in the abdominal wall – it was very satisfying to see it being used immediately and effectively to raise the level of care they could provide.”

Dr Pich continues to be involved with CADUS and is now working with them to plan ongoing support for Al Khansaa in the future: “It is very important for us to keep the connection with our colleagues in Mosul, not only to encourage more donations but also to transfer knowledge and skills. I am very grateful to SonoSite for its generosity and think the M-Turbo was just the right piece of equipment they needed. I felt a large sense of responsibility transporting this valuable machine to a war-stricken country, for people who really needed it, but I am very pleased with what we were able to accomplish. The most satisfying aspect of this experience is knowing that I fulfilled my promise to the dedicated staff at Al Khansaa, to bring resources and education that could help them deliver, and raise, the quality of care the women and children in Mosul so desperately need.”

New plan for NHS tackling ‘major killer conditions’ to save up to 500,000 lives

The NHS Long Term Plan, published today (7 January) outlines how over three million people will be treated from new stroke, respiratory and cardiac services over the next decade.

The measures, supported by £4.5bn in new service model funding, will help prevent over 150,000 heart attacks, strokes and cases of dementia, according to the plan.

Measures in the plan will also help over three million people through improved stroke, respiratory and cardiac services over the next 10 years, it said.

Around 30,000 people with dangerously high inherited cholesterol levels will be genetically tested –around a quarter of those affected.

Roughly 80,000 hospital admissions could be avoided through investing in earlier detection and better treatment of respiratory conditions. Smart inhalers will also be piloted, allowing patients to monitor their condition at all times.

Every hospital with a major A&E department will offer same day emergency care, meaning that patents will be treated and discharged in the same day, avoiding the need for overnight stays.

NOVESA Phase IIa trial in patients with systemic sclerosis initiated

7th January 2019

Galapagos NV has expanded its clinical study program with GLPG1690 in systemic sclerosis (SSc), following the recent start of the ISABELA Phase III program with ’1690 in idiopathic pulmonary fibrosis (IPF).
 
NOVESA is a double-blind, placebo-controlled Phase IIa trial evaluating the efficacy, safety and PK/PD of ’1690 in patients with systemic sclerosis. NOVESA is planned to recruit 30 patients with diffuse cutaneous SSc, an autoimmune disease involving multiorgan fibrosis, which has one of the highest mortality rates among rheumatic diseases. One of the most visible manifestions is hardening of the skin. In diffuse cutaneous SSc, skin thickening affects several body areas, and patients have a higher risk of developing fibrosis of various internal organs, such as the lung. Currently, there are no approved drugs for this disease. SSc affects approximately 90,000 patients in the US and Europe, with a predominance of female patients (75%).
 
The primary endpoint of NOVESA is the modified Rodnan skin score (mRSS) at 24 weeks. mRRS measures the skin thickness as a surrogate measure of disease severity and mortality, with an increase in thickness associated with involvement of internal organs and increased mortality. Secondary objectives and exploratory endpoints include FVC, HRCT, quality of life as measured by QoL-Q (SHAQ), and CRISS, a SSc disease composite score.
 
In addition to our Phase III program in IPF, we are excited to broaden our development program with ’1690 to a second indication,” said Dr Walid Abi-Saab, Chief Medical Officer at Galapagos. “Moreover, SSc is particularly interesting, as this disease straddles our expertise in autoimmune diseases as well as in fibrosis. Thanks to the broad mode of action of ’1690, which is both anti-inflammatory and anti-fibrotic, this compound has the potential to address the important unmet medical need in SSc.”

Janssen seeks EMA approval of Stelara for adults with ulcerative colitis

Janssen has announced the submission of a Group Type II Variation Application to the European Medicines Agency (EMA) seeking approval of Stelara® (ustekinumab) for the treatment of adults with moderately to severely active ulcerative colitis (UC).

Ustekinumab is a human monoclonal antibody that targets the interleukin (IL)-12 and IL-23 cytokines, which are believed to play an important role in the immune and inflammatory responses seen in immune-mediated diseases, such as UC and Crohn’s disease.1

This submission follows a supplemental Biologics License Application (sBLA) made to the United States’ Food and Drug Administration (FDA) on December 20, 2018, which also seeks approval of ustekinumab for the treatment of adults with moderately to severely active UC.

“Ulcerative colitis (UC) is a chronic, painful and debilitating condition that has a significant impact on quality of life. UC affects up to one million people across Europe, and some of these patients struggle to achieve and maintain high levels of clinical response with currently available therapies. This submission for ustekinumab in UC brings us one step closer to providing a new treatment option to help address this important unmet need,” said Jaime Oliver, MD, Janssen Therapeutic Area Lead, Immunology, Europe, Middle East & Africa, Cilag GmbH International. “We look forward to working with the European Medicines Agency (EMA) as the application process progresses.”

This submission is based on data from the Phase III UNIFI global clinical development programme, which includes two studies (one induction and one maintenance study) evaluating the efficacy and safety of ustekinumab for the treatment of moderately to severely active UC in adults. Data from the Phase III induction study were recently presented at the 2018 American College of Gastroenterology (ACG) and United European Gastroenterology Week (UEGW) annual meetings, indicating that treatment with a single intravenous (IV) dose of ustekinumab induces clinical remission and response in adults with moderately to severely active UC who previously experienced an inadequate response or intolerance to conventional or biologic therapies.2 Results from the Phase III maintenance study will be presented at future scientific meetings.

“We’re excited to bring this innovative therapy, with a proven track record in Crohn’s and other immune diseases, one step closer to being available for people living with ulcerative colitis,” said Scott E Plevy, MD, Gastroenterology Disease Area and IL-23 Pathway Leader, Janssen Research & Development, LLC. “This submission builds upon our 20-year legacy of research and development to address unmet needs of people living with inflammatory bowel diseases.”

The common (in ≥1% of patients) adverse reactions reported in controlled periods of the adult psoriasis, psoriatic arthritis and Crohn’s disease clinical studies with ustekinumab as well as in the post-marketing experience are: arthralgia, back pain, diarrhoea, dizziness, fatigue, headache, injection site erythema, infection site pain, myalgia, nasopharyngitis, nausea, oropharyngeal pain, pruritus, upper respiratory tract infection and vomiting.3

References

  1. Croxford A et al. IL-23 and IL-23 in health and disease. Cytokine Growth Factor Rev. 2014;25(4):415–21.
  2. Sands B et al. Safety and efficacy of ustekinumab induction therapy in patients with moderate to severe ulcerative colitis: results from the Phase 3 UNIFI study. United Europe Gastroenterology Week (UEGW) 2018, Vienna, Austria. 20–24 Oct 2018: LB01.
  3. European Medicines Agency. Ustekinumab Summary of Product Characteristics. https://www.ema.europa.eu/en/medicines/human/EPAR/stelara. Last updated December 2018; last accessed December 2018.

Scientists recognised for transformative lupus research projects

21st December 2018

The Lupus Research Alliance has announced the 2018 recipients of the Dr. William E. Paul Distinguished Innovator Award in Lupus and Autoimmunity: namely, Nir Hacohen, PhD and Vijay Kuchroo, DVM, PhD.

Dr. Hacohen is seeking better ways to treat lupus kidney disease, the major cause of illness and death among patients with lupus. Dr. Kuchroo is looking at ways to harness regulatory T and B cells as a new approach to lupus treatment. Both projects have the potential to stimulate innovative strategies for prevention, treatment, and cure of lupus.

Dr. Hacohen serves as Director of the Center for Cell Circuits and Center for Cancer Immunology, Massachusetts General Hospital and Broad Institute, and Professor of Medicine at Harvard Medical School. Dr. Kuchroo is a Professor of Neurology at Harvard Medical School and Brigham and Women’s Hospital. 

The immune response in lupus nephritis

Lupus nephritis (inflammation of the kidneys) is a major cause of illness and death among patients with lupus. The failure to stop the harmful effects caused by lupus nephritis is thought by many researchers to be due to an incomplete understanding of the immune system. 

Dr. Hacohen is seeking to understand why the immune response (to tumors, bacteria or self) varies so dramatically across individuals. According to Dr. Hacohen, “The results of these studies will generate new hypotheses for how immune cells work together to cause tissue damage in lupus nephritis patient kidneys, lead to new drug targets and better predictors of disease, and guide researchers in the improvement of mouse models to understand human lupus nephritis.”

The role proteins play in regulating the body’s immune system in lupus

T cells and B cells are the primary types of lymphocytes (a subtype of white blood cells) that determine the body’s immune response to foreign substances in the body,” noted Dr. Kuchroo. “Once activated to mount an immune response, T and B cells need to be turned off by another class of regulatory cells. In patients with lupus, these regulatory cells are not able to properly function. This study will examine how to induce and promote the function of these regulatory T and B cells for treating lupus.”

European Commission approval for self-administration of Xolair® across all indications

17th December 2018

Novartis has announced that the European Commission (EC) has approved Xolair® (omalizumab) prefilled syringe (PFS) for self-administration, allowing patients with severe allergic asthma (SAA) and chronic spontaneous urticaria (CSU) to administer their own treatment.
 
With this approval, Xolair is the first and only biologic to offer the option of self-administration for SAA and CSU.
 
Xolair, which targets immunoglobulin E (IgE), is the first and only biologic to be approved in the European Union, Iceland, Norway, and Liechtenstein for self-administration (or administration by a trained caregiver) for the treatment of SAA in patients 6 years of age and older that have difficulty in controlling their asthma symptoms and for CSU in patients 12 years of age and older who continue to have hives that are not controlled by H1 antihistamines. Studies in severe allergic asthma and chronic spontaneous urticaria have shown that appropriately trained patients can effectively self-administer Xolair at home.1-3
 
The efficacy of Xolair has been demonstrated in large-scale clinical trials and real world studies. Xolair has been shown to reduce severe exacerbations and corticosteroid use in SAA1, as well as rapidly reduce symptoms in CSU.4
 
The EC approval will allow patients with no known history of anaphylaxis to self-inject Xolair PFS, or be injected by a trained lay-caregiver, from the fourth dose onwards, if a physician determines that this is appropriate.5 The patient or the caregiver must have been trained in the correct sub-cutaneous injection technique and the recognition of the early signs and symptoms of serious allergic reactions.5
 
Today’s positive news is a big step forward for patients living with immunoglobulin E- mediated asthma and chronic spontaneous urticaria. Decreasing the number of regular clinic visits allows patients the flexibility to fit their treatment around their lives and helps to reduce the burden of these diseases. It also allows physicians a greater capacity for patients who need extra care, which is important” said Professor Dr Karl-Christian Bergmann, Allergy Center Charité, Berlin.
 
Administered via injection every two or four weeks, Xolair is widely used and well tolerated.6 With 13 years of physician experience in Europe and one million patient years of exposure, use of Xolair in SAA and CSU is supported by a wealth of evidence from randomised clinical trials and real-world studies.1-3 Anaphylactic reactions were rare in clinical trials (≥ 1/10,000 to < 1/1000)5 and via post-marketing reports (approximately 0.2%).5
 
References
  1. Liebhaber M, Dyer Z. J Asthma 2007;44(3):195-196.
  2. Ghazanfar M,Thomsen S. J Dermatolog Treat 2018;29(2):196.
  3. Denman S et al. Br J Dermatol 2016;175(6):1405-1407.
  4. Maurer M et al. N Engl J Med 2013;368(10):924-935.
  5. Xolair® Summary of Product Characteristics. Novartis Europharm Limited. Last accessed: December 2018.
  6. Humbert M et al. Allergy 2005;60(3):309-316.

Expert analysis: Challenges in diagnosing connective tissue diseases

 

Connective tissue diseases (CTD) are a group of disorders involving the protein-rich tissue that supports organs and other parts of the body. However, more commonly the acronym CTD identifies systemic autoimmune rheumatic diseases such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and SLE-like diseases, Sjogren syndrome (SS), scleroderma (SSc), inflammatory autoimmune myopathies (AM), systemic autoimmune vasculitides (AV), mixed or undifferentiated connective tissue disease (MCTD/UCTD), and anti-phospholipid syndrome (APS).

All these disorders are also defined as systemic autoimmune rheumatic diseases (SARD) and are characterised by an autoimmune response against self-antigens that ends in chronic inflammation and irreversible tissue damage if untreated, with high direct and indirect costs for the social/health systems. SARD can affect several organs and tissues of the body; as a consequence, different specialists may see these patients but ultimately, rheumatologists or clinical immunologists are the main physicians providing care for SARD.

Classification and diagnosis 

The classification and the diagnosis of SARD are based on clinical signs and symptoms, however laboratory and instrumental parameters are also required.1 These tools improved our classification/diagnostic power; this is the case, for example, for RA with the new anti-citrullinated peptide antibodies and joint magnetic resonance and/or ultrasound Doppler evaluation.2 In addition, the histology or immune-phenotyping of the damaged tissues help in diagnosis but, unfortunately, are not always feasible or specific because of overlapping pathogenic pathways.3

Although each disease displays peculiar clinical pictures, some manifestations are not specific at all (such as thrombosis in APS) or frequently overlapping among different diseases (for example, inflammatory arthritis). Moreover, the clinical manifestations are non-specific particularly at the beginning of the clinical history making difficult the correct diagnosis. Consequently, biological biomarkers play a crucial role in directing physicians towards the right diagnosis. In particular, autoantibodies are offering the most reliable tools nowadays (Table 1). Additional tools, such as cytokines or genetic biomarkers (including non-coding RNA/DNA) are quite promising but not useful in practice at the moment.

Table 1. Autoantibodies in SARD

Antibody/abbreviation Disease
Rheumatoid factor (RF)

Anti-citrullinated protein antibodies (ACPA)

Rheumatoid arthritis
Anti-nuclear antibodies (ANA)

Anti-double stranded DNA antibodies (anti-dsDNA)

Anti-Sm antibodies (anti-Sm)

Lupus anticoagulant (LA)

Anti-cardiolipin antibodies (anti-CL)

Systemic lupus erythematosus
ANA

Anti-topoisomerase I

Anti-centromere A,B,C (anti-CENP A,B,C)

Anti-RNA polymerase III

Systemic sclerosis
Anti-SSA/Ro

Anti-SSB/La

RF

ANA

Sjogren’s syndrome
Anti-U1 ribonucleoprotein (anti-U1 RNP) Mixed connective tissue disease
ANA Undifferentiated connective tissue disease
LA

Anti-CL

Anti-beta2-glycoprotein I antibodies (anti-b2GPI)

Antiphospholipid syndrome
ANCA

Proteinase 3 (PR3)

Myeloperoxidase (MPO)

ANCA-associated vasculitides

 

Autoantibodies represent well-accepted classification criteria and, in some cases, they are also entry classification criteria; the most recent examples are represented by anti-phospholipid antibodies (aPL) for APS and anti-nuclear antibodies detectable by indirect immunofluorescence or equivalent solid phase assays for SLE.4,5 From this point of view, they represent mandatory inclusion criteria for clinical trials.

Autoantibodies in diagnosis                    

SARD are chronically evolving disorders characterised by the production of autoantibodies, even years before the appearance of the clinical manifestations. Because of the non-specific clinical symptoms at the beginning of the SARD, the detection of autoantibodies is a useful diagnostic tool. By contrast, early diagnosis is mandatory in order to start treatment before irreversible tissue damage takes place. This approach results in a better prognosis (that is, less organ damage) and in the reduction of the costs of the disease. The most common approach is the use of screening assays (for example, ANA detection) for supporting the suspect of the autoimmune origin of the disease and then the request for autoantibody profiles for identifying autoantibodies associated to SLE-, SSc-, SS- or AM-like disorders6 (Table 2).

Table 2. Autoantibodies and their associations with clinical manifestations in SARD

Autoantibody Disease association Clinical manifestations
anti-topoisomerase I SSc ILD, digital ulcers, diffuse skin involvement, heart involvement
anti-CENP A, B, C SSc PAH, bowel involvement, digital ulcers, limited skin involvement
anti-RNA polymerase III SSc SRC, tendon friction rubs, severe diffuse skin involvement
anti-U3 RNP SSc PAH, myositis, heart involvement, diffuse skin involvement
anti-U1 RNP SSc, SSc/PM overlap, SLE, MCTD Myositis, PAH, arthritis, limited skin involvement
anti-PM-Scl PM/DM, SSc/myositis overlap, SSc Myositis, limited skin involvement
anti-Th/To SSc PAH, ILD, bowel involvement, limited skin involvement
anti-U11/U12 SSc ILD
anti-mitochondria PBC/lcSSc and PBC/SS overlap Cholestatic liver disease
anti-Ku SLE, SSc, SLE/PM/SSc and PM/SSc overlap Myositis, arthritis
anti-dsDNA SLE Nephritis, disease activity
anti-Ro/SSA SLE, SS, SCLE Haematologic disorder, photosensitivity, neonatal lupus, C2 deficiency
anti-aminoacyl-tRNA synthetases

(anti-Jo1, -PL7,

-PL12, -EJ,  -OJ,

-KS, -Zo, -YRS)

PM/DM Myositis, ILD, mechanic’s hands
anti-ribosomal P protein SLE Neuropsychiatric manifestations
anti-histones SLE Drug-induced lupus
anti-nucleosomes SLE, APS Drug-induced lupus, nephritis, disease activity
anti-C1q SLE Active renal disease
anti-citrullinated proteins RA Severe disease
LA, anti-CL, anti-b2GPI APS Thrombosis, miscarriages
 
ILD, interstitial lung disease; SRC, scleroderma renal crisis; PAH, pulmonary arterial hypertension; PM, polymyositis; DM, dermatomyositis; lcSSc, limited cutaneous systemic sclerosis; SCLE, sub-acute cutaneous lupus erythematosus
 

Autoantibodies are also widely used for disease subgrouping. For example, SSc patients sub grouped according to the presence of anti-topoisomerase or anti-centromere autoantibodies display different clinical picture and evolution.7,8 The same is also the case for the autoantibodies detectable in a different AM.9

The presence of different autoantibody profiles is useful for risk stratification in some diseases. The best example is represented by APS, in which the number of tests positive for aPL (that is, lupus anticoagulant, anti-cardiolipin, anti-b2 glycoprotein) or the aPL titres are risk factors. In other words, the higher the number of positive tests (triple or double versus single positivity) or the antibody titres, the higher the risk for thrombosis or miscarriages.10

SARD subgrouping and risk profiling allow a better disease characterisation and ultimately the best treatment approach according to precision (or personalised) medicine.

In addition to their defined role in the diagnosis of AID, a number of autoantibodies represent biomarkers for damage or involvement of particular tissues or organs, making them important in defining relevant comorbidities. This is the case for anti-C1q antibodies closely associated with active renal disease in SLE11 (Table 2).

The presence of an autoantibody might be the determining factor for starting primary prophylactic therapy, even in the absence of overt clinical signs, in order to reduce the risk of the clinical manifestations. Anti-phospholipid antibodies are the most popular example, justifying antiplatelet therapy even in asymptomatic aPL-positive carriers or low-dose aspirin and heparin in pregnant women at high risk for miscarriages.10

Conclusions                     

Nowadays, autoantibody detection is performed by automated assays in high-throughput routine service laboratories. The availability of the new assays has uncovered many problems, including quality control, quality assurance, standardisation, analytical sensitivity/specificity, within and between laboratory reproducibility and clinical sensitivity. These aspects are all relevant to the overall clinical interpretation of the tests and several international standardisation initiatives are currently ongoing.12,13

References

  1. Meroni PL et al. Standardization of autoantibody testing: a paradigm for serology in rheumatic diseases. Nat Rev Rheumatol 2014;10(1):35–43.
  2. Aletaha D et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum 2010;62(9):2569–81.
  3.  Barturen G et al. Moving towards a molecular taxonomy of autoimmune rheumatic diseases. Nat Rev Rheumatol 2018;14(3):180.
  4. Miyakis S et al. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost 2006;4(2):295–306.
  5. Aringer M et al. Classification criteria for systemic lupus erythematosus (in press).
  6. Meroni PL, Borghi MO. Diagnostic laboratory tests for systemic autoimmune rheumatic diseases: unmet needs towards harmonization. Clin Chem Lab Med 2018;56(10):1743–8.
  7. Didier K et al. Autoantibodies associated with connective tissue diseases: What meaning for clinicians? Front Immunol 2018;9:541.
  8. Choi MY, Fritzler MJ. Progress in understanding the diagnostic and pathogenic  role of autoantibodies associated with systemic sclerosis. Curr Opin Rheumatol 2016;28(6):586–94.
  9. Palterer B et al. Bench to bedside review of myositis autoantibodies. Clin Mol Allergy 2018;16:5.
  10. Chighizola CB et al. The treatment of anti-phospholipid syndrome: A comprehensive clinical approach. J Autoimmun 2018;90:1–27.
  11. Gensous N et al; FHU ACRONIM. Predictive biological markers of systemic lupus erythematosus flares: a systematic literature review. Arthritis Res Ther 2017;19(1):238.
  12. Monogioudi E et al. Standardization of autoimmune testing – is it feasible? Clin Chem Lab Med 2018;56(10):1734–42.
  13. Conrad K et al. From autoantibody research to standardized diagnostic assays in the management of human diseases – report of the 12th Dresden Symposium on Autoantibodies. Lupus 2016;25(8):787–96.

Most accurate tool yet developed to predict asthma in young children

Scientists at Cincinnati Children’s Hospital Medical Center in the US have created and tested a decision tool that appears to be the most accurate, non-invasive method yet developed to predict asthma in young children.
 
The researchers hope the Pediatric Asthma Risk Score (PARS) will become the most common tool used by medical practitioners to predict asthma and help prevent the common airway disease from developing.
 
PARS is superior to the Asthma Predictive Index (API) in its ability to predict asthma in children with mild to moderate asthma risk, with an 11% increase in sensitivity,” says Gurjit Khurana Hershey, MD, PhD, director of Asthma Research at Cincinnati Children’s and senior author of the study. “Children with mild to moderate risk may be the most likely asthma patients to respond favourably to prevention strategies.”
 
The study, published online in the Journal of Allergy and Clinical Immunology, found that the API missed 43% of asthmatic children identified by PARS as mild to moderate risk. PARS and the API equally predicted asthma risk for children with the most risk factors.
 
The API has been seen as the gold standard to which other predictive models have been compared. While useful for predicting which children will not develop asthma, it “leaves much room for improvement in terms of identifying children who will,” says Jocelyn Biagini Myers, PhD, a researcher in the division of Asthma Research and lead author of the study. A notable achievement of the PARS over the API is that it delivers a personalised asthma risk score to the patient, she says.
 
The PARS tool included new and less invasive criteria than previous tools. Additions included demographic data and clinical factors routinely collected during an asthma or allergy assessment in a doctor’s office.
 
Dr Khurana Hershey and her colleagues devised the tool using data from the Cincinnati Childhood Allergy and Air Pollution study – a group of 762 infants born between 2001 and 2003 in Cincinnati and northern Kentucky. These were children of parents who had at least one allergy symptom. The children were examined annually at the ages of 1, 2, 3, 4 and 7 for the development of allergic disease. They were skin tested for 15 airborne and food allergens including cat, dog, cockroach, dust mites, trees, mould, weeds, grass, cow’s milk and hen’s egg.
 
Of the 762 infants, 589 were evaluated for asthma development at the age of 7 using objective measures of lung function. Sixteen percent had asthma. The researchers also queried parents for numerous factors that contribute to asthma risk.
 
The children with asthma at age 7 were more likely to have at least one parent with asthma, two or more positive skin tests to airborne or food allergens, eczema at a young age, wheezing apart from colds, frequent wheezing at a young age, a diagnosis of allergic rhinitis in the first three years of life, and to be African-American.
 
The researchers compared the PARS model to the original API and found it to be 11% more sensitive than the API.
 
Our PARS model either outperforms and/or is less invasive than 30 existing models intended to predict asthma development,” says Dr Hershey. “The PARS also may be more clinically useful and applicable in an office setting.”
 
To facilitate easy implementation of PARS in clinical and research settings, the study contains a PARS scoring sheet that includes the decision tool and clinical interpretations. A PARS web application, which provides fast and easy calculation, is accessible at https://pars.research.cchmc.org. Smartphone apps are being developed for the iPhone and for android phones.
 
Reference
  1. Biagini Myers J et al. A Pediatric Asthma Risk Score to better predict asthma development in young children. J Allergy Clin Immune 2018;DOI: 10.1016/j.jaci.2018.09.037.

x