×

Resistance exercise could be a new ‘prescription’

A new study by Griffith University’s Menzies Health Institute Queensland has linked resistance exercise with boosting you immune system and aiding in injury health.

Until this study was undertaken, little was known about the impact of resistance exercise.

Published in Immunology Letters, the researchers examined 16 previous studies undertaken during1989-2016 that investigated participants undertaking a single session of resistance exercise encompassing various exercises.

“We combined the data from all relevant scientific publications, including two of our own original articles, to conduct a stringent systematic analysis of the resistance exercise research,” said Dr Adam Szlezak from Griffith’s Menzies Health Institute Queensland.

The study found that both high and low dosages of resistance exercise increased the immune system’s surveillance potential in the participants in a similar way to that of aerobic exercise, much like drugs can.

“We found that both high and low dosages of resistance exercise increased the immune system’s surveillance potential in the participants in a similar way to that of aerobic exercise. Even a low dose of thumb resistance exercise increased the number of key white blood cells in the circulation,” Dr Szlezak said.

The research suggested that resistance exercise appears to improve immuno-surveillance similar to that of moderate intensity aerobic exercise, regular moderate intensity 20-45 min work-outs in the gym may provide similar protection against upper respiratory tract infections (URTI).

Now that we know that different resistance exercise doses can result in distinct biological responses, much like drugs can, we now need to see if these responses can be used to reduce risk of URTI, as well as improve recovery from illness and injury, said Dr Szlezak.

The research also suggests that GPs should recommend that their patients abstain from all forms of exercise in the hours prior to blood collection for requesting full blood counts due to its impact on white cell count.

Meredith Horne

[Series] The evolution of modern respiratory care for preterm infants

Preterm birth rates are rising, and many preterm infants have breathing difficulty after birth. Treatments for infants with prolonged breathing difficulty include oxygen therapy, exogenous surfactant, various modes of respiratory support, and postnatal corticosteroids. In this Series paper, we review the history of neonatal respiratory care and its effect on long-term outcomes, and we outline the future direction of the research field. The delivery and monitoring of oxygen therapy remains controversial, despite being in use for more than 50 years.

[Correspondence] Prolonged glucocorticoid treatment in acute respiratory distress syndrome

We were disappointed that Rob Mac Sweeney and Daniel F McAuley’s Seminar (Nov 12, p 2416)1 on acute respiratory distress syndrome overlooked much of the evidence for prolonged glucocorticoid treatment. The authors reference two outdated meta-analyses2,3 that have contradictory results. The basis for the inconsistency between these two meta-analyses can be explained and current evidence suggests a net benefit for glucocorticoids in acute respiratory distress syndrome.4

[Correspondence] Prolonged glucocorticoid treatment in acute respiratory distress syndrome – Authors’ reply

G Umberto Meduri and Reed A C Siemieniuk incorrectly suggest our Seminar1 misinterprets the literature regarding corticosteroids in acute respiratory distress syndrome. Although more recent studies might have refined the research question, older studies still remain the best evidence to address the question such studies originally asked, and so are not outdated. These older studies are the reason high-dose steroids are not used today.

[Correspondence] Health systems resilience: meaningful construct or catchphrase?

Resilience is an emerging concept in the health systems discourse, further highlighted by infectious disease outbreaks including Ebola virus disease, Zika virus disease, and Middle East respiratory syndrome. However, the definition and exploration of resilience within health systems research remains a source of debate, as underscored at the recent 4th Global Symposium on Health Systems Research; Vancouver, BC, Canada; Nov 14–18, 2016.

[Obituary] Lars Olof Lennart Nilsson

Innovative biomedical photographer. Born in Strängnäs, Sweden, on Aug 24, 1922, he died after a respiratory infection in Stockholm, Sweden, on Jan 28, 2017, aged 94 years.

News briefs

Hidden risk population for thunderstorm asthma

Research presented at the Thoracic Society for Australia and New Zealand (TSANZ) Annual Scientific Meeting in Canberra last month identified “a potentially hidden and significant population susceptible to thunderstorm asthma”. “This is a wake-up call for all of Australia, but particularly Victoria as it prepares for its next pollen season,” said Professor Peter Gibson, president of TSANZ. “Many more people than previously thought are at risk of sudden, unforeseen asthma attack. It is essential that we invest more research into this phenomenon and educate our health services and public to take preventative and preparedness measures.” Nine people died in Victoria late last year and over 8500 required emergency hospital care when a freak weather event combining high pollen count with hot winds and sudden downpour led to the release of thousands of tiny allergen particles triggering sudden and severe asthma attacks. Those most seriously affected were people who were unaware they were at risk of asthma and therefore had no medication to hand. In the study of over 500 health care workers, led by the Department of Respiratory and Sleep Medicine, Eastern Health, Victoria, almost half the respondents with asthma experienced symptoms during the thunderstorm event. Most took their own treatment, a few sought medical attention and one was hospitalised. More alarming was the 37% of respondents with no prior history of asthma who reported symptoms such as hayfever, shortness of breath, cough, chest tightness and wheeze during the storms. The study also found that people with a history of sensitivity to environmental aeroallergens (eg, ryegrass or mould) were far more likely to report symptoms than those with a history of either no allergy or allergy to dust mite/cats. Physical location, described as predominantly indoors versus outdoors, was not a risk factor. “This study gives us an indication of the proportion of our population that might be at risk of thunderstorm asthma, but are unaware of it as they have no history of asthma. It also suggests that a history of hayfever is one of the greatest risk factors,” said lead researcher Dr Daniel Clayton-Chubb. “The key message from our work is that anyone with hayfever should ensure that they have ready access to quick-acting asthma treatments such as bronchodilators at all times, but particularly in pollen season or if thunderstorms are predicted. Severe thunderstorm asthma symptoms can strike rapidly and without warning.”

New genetic causes of ovarian cancer identified

A major international collaboration has identified new genetic drivers of ovarian cancer, findings which have been published in Nature Genetics. The study involved 418 researchers from both the Ovarian Cancer Association Consortium, led by Dr Andrew Berchuck from the United States, and the Consortium of Investigators of Modifiers of BRCA1/2, led by Professor Georgia Chenevix-Trench from QIMR Berghofer Medical Research Institute. Professor Chenevix-Trench said it was known that a woman’s genetic make-up accounts for about one-third of her overall risk of developing ovarian cancer. “This is the inherited component of the disease risk,” Professor Chenevix-Trench said. “Inherited faults in genes such as BRCA1 and BRCA2 account for about 40% of that genetic risk. Other variants that are more common in the population (carried by more than one in 100 people) are believed to account for most of the rest of the inherited component of risk. We’re less certain of environmental factors that increase the risk, but we do know that several factors reduce the risk of ovarian cancer, including taking the oral contraceptive pill, having your tubes tied and having children. In this study, we trawled through the DNA of nearly 100 000 people, including patients with the most common types of ovarian cancer and healthy controls. We have identified 12 new genetic variants that increase a woman’s risk of developing the cancer. We have also confirmed that 18 variants that had been previously identified do increase the risk. As a result of this study, we now know about a total of 30 genetic variants in addition to BRCA1 and BRCA2 that increase a woman’s risk of developing ovarian cancer. Together, these 30 variants account for another 6.5% of the genetic component of ovarian cancer risk.”

Targeted therapy for chronic respiratory disease: a new paradigm

Targeted therapy represents a new treatment paradigm that shows great promise in treating serious chronic respiratory diseases for which we presently have limited treatment options.1 Pharmacotherapy in the 20th century was best characterised by the use of antibiotics for infectious diseases. An antimicrobial drug targets a specific pathogen which is the cause of an infectious disease, leading to cure. This approach has its basis in Paul Ehrlich’s magic bullet (zauberkugel) theory and his success with a series of specific antimicrobials for syphilis.2 Although highly effective for acute infectious diseases, this approach cannot be usefully applied to non-communicable chronic diseases (NCCDs) because a single aetiological agent cannot be identified.

Step therapy has emerged as the dominant treatment paradigm for NCCDs such as cardiovascular disease (hypertension, congestive cardiac failure), type 2 diabetes mellitus, cancer and chronic airway disease (asthma, chronic obstructive pulmonary disease). In step therapy, drugs are added (step up) or withdrawn (step down) based on the level of responsiveness to treatment. This has led to improved outcomes but is limited. A key limitation is that individual variability in treatment response is ignored in this one-size-fits-all paradigm.

A new concept is to take individual variability into account when making management decisions. This approach, termed targeted therapy, represents a new treatment paradigm for NCCDs and is defined as “treatments targeted to the needs of individual patients on the basis of genetic, biomarker, phenotypic, or psychosocial characteristics that distinguish a given patient from other patients with similar clinical presentations”.1

In targeted therapy for NCCD, a drug targets a specific pathogenic pathway, leading to major improvements. The corollary is that the treatment is ineffective if the pathway does not operate. This approach is made possible by developments in pathway identification (eg, biomarkers and omics technologies) and specific therapies to target components of pathways (eg, monoclonal antibodies). Targeted therapy is now being successfully applied to chronic respiratory diseases that previously had dismal outcomes. Conditions such as lung cancer, cystic fibrosis (CF) and severe refractory asthma have all seen advances based on the targeted therapy concept. These conditions are associated with premature death, significant quality of life impairment, and a poor response to current therapy. This review highlights these advances (Box 1) using randomised controlled trials published in high impact journals.

Severe asthma

By global standards, Australia suffers a disproportionate burden from asthma. Both the prevalence of and mortality from asthma are among the highest in the world. While major improvements were made in outcomes for mild to moderate asthma in the latter part of the 20th century, these gains have now stalled. Calls for a different approach to asthma care have emerged, and advances have been made in the use of targeted therapy for severe asthma.3

Severe refractory asthma is characterised by severe symptoms, significant lung function impairment and/or asthma exacerbations that occur despite maximal therapy with high dose inhaled corticosteroids and long-acting β-2 agonists.4 It is a high burden disease that affects 3–5% of people with asthma, and up to half of these have eosinophilic disease.5,6 Most patients are unable to work because of their illness,7 and the condition is responsible for almost 50% of direct health care costs for asthma.7 Oral corticosteroids are frequently used to manage severe asthma, however they are only partially effective and are associated with a high prevalence of treatment-related side effects such as type 2 diabetes, osteopenia and osteoporosis, dyspepsia, obesity, hypertension, cataracts, and obstructive sleep apnoea.8

The pathogenesis of severe asthma is heterogeneous. One endotype — severe refractory asthma with eosinophilia (SREA) — is associated with an interleukin (IL)-5 mediated eosinophil influx into the blood and airway (Box 2). Other endotypes include severe refractory allergic asthma, which can be treated with the anti-IgE monoclonal antibody omalizumab, and non-eosinophilic asthma, for which targeted therapy is currently lacking. IL-5 plays a fundamental role in eosinophil growth, differentiation and survival. Human eosinophils express comparatively high levels of IL-5 receptor subunit alpha compared with other cells. The major cellular sources of IL-5 in asthma are T helper type 2 (Th2) cells, mast cells, group 2 innate lymphoid cells, and eosinophils themselves. Monoclonal antibodies directed against IL-5 (mepolizumab, reslizumab) or its receptor (benralizumab) have been developed to treat SREA. Studies consistently show that mepolizumab reduces blood and airway eosinophils in asthma.9

The clinical efficacy of mepolizumab has now been demonstrated in several randomised controlled trials (RCTs). The key effects are a highly significant reduction in asthma exacerbations10,11and an oral steroid-sparing effect.12 Some studies also show improvement in lung function and quality of life.11 These trials recruited adults and adolescents with severe asthma, persistent asthma exacerbations despite inhaled therapy, and evidence of eosinophilia from blood eosinophil counts > 0.3 cells × 109/L, sputum eosinophil counts ≥ 3%, or an elevated fraction of exhaled nitric oxide (a marker of Th2-mediated inflammation). The DREAM study10 was a dose-ranging, parallel group, multicentre, double-blind RCT of mepolizumab in which 621 participants were randomised to one of three doses of intravenous mepolizumab or placebo every 4 weeks for a year. The results showed a highly significant reduction in asthma exacerbations between 39% and 52%, which have been replicated in subsequent RCTs.11,12 Mepolizumab treatment of patients with severe oral corticosteroid-dependent asthma was associated with a median 50% reduction in maintenance corticosteroid dose.12 The safety profile was similar to placebo. Mepolizumab is licensed for use by the United States Food and Drug Administration (FDA) and has been approved in Australia by the Therapeutic Goods Administration (TGA) but is not funded for clinical use at this time.

The studies show preferential effects of anti-eosinophil therapy on asthma exacerbations consistent with studies that target airway eosinophilia in severe asthma.13 The level of blood eosinophils used to select participants for targeted therapy in severe asthma is within the quoted normal range; however, studies of normal values that exclude people with atopic disorders have found a lower level for the upper normal range.14 Values of blood eosinophils > 0.3 cells × 109/L have been associated with airway eosinophilia in corticosteroid-treated asthma.15

Therapy targeted at the IL-5/eosinophil endotype of asthma is highly effective. In adults and adolescents with severe refractory eosinophilic asthma recognised by a persistence of blood eosinophils despite corticosteroid therapy, the addition of targeted therapy against IL-5 or its receptor may reduce asthma exacerbations by almost 50% and reduce oral corticosteroid requirements. Administration and access require a systematic assessment of asthma and management of potentially reversible factors, including non-adherence to maintenance corticosteroid therapy.

Cystic fibrosis

CF is a multisystem genetic disorder that primarily affects the lungs causing bronchiectasis, recurrent infections, and premature death. In CF, there is a defective or deficient CF transmembrane conductance regulator (CFTR) protein that functions as an anion channel predominantly in epithelial cell membranes. There are about 3300 patients with CF in Australia, most of whom are adults. The CFTR gene was discovered in 198916 and has over 2000 different mutations17 that may cause either an inadequate amount of CFTR protein or deficient function of CFTR protein at the epithelial cell membrane, or a combination of these abnormalities (Box 3). Deficient function may be due to a reduced open probability of the CFTR channel (gating) or abnormal conductance of the channel. The most common CFTR mutation is Phe508del (formerly F508del); about 45% of patients are homozygous for this class II mutation worldwide.18 Therapies targeted at specific defects in CFTR function have been developed, and new therapies are currently in development.

In 2004, ivacaftor, the first CFTR modulator approved for clinical use, commenced development. Ivacaftor targets the deficient function of CFTR protein at the epithelial membrane and increases the open probability (gating) of the CFTR channel. It is referred to as a CFTR potentiator (Box 3). The most common CFTR gating mutation is Gly551Asp (formerly G551D), a class III mutation which accounts for about 4% of CFTR alleles; it is associated with an adequate amount of protein at the epithelial membrane but the protein has no CFTR function. Two landmark phase 3 trials of ivacaftor in patients aged 6–11 years and 12 years and older carrying at least one copy of the Gly551Asp mutation19,20 demonstrated improvements in lung function (mean absolute improvement in the percentage of predicted forced expiratory volume in 1 second [FEV1] of about 10.6%) and nutritional status, and reported a marked reduction in sweat chloride measurement confirming the effect of restoration of CFTR function. These clinical benefits persisted in a 2-year open-label extension study.21 In 2012, the US FDA approved the use of ivacaftor for patients aged 6 years and older with CF carrying at least one Gly551Asp mutation. Successful clinical trials of ivacaftor permitted extension of approval to eight other non-Gly551Asp gating mutations,22 and to patients carrying a residual function Arg117His (formerly R117H) mutation.23 In Australia, ivacaftor is Pharmaceutical Benefits Scheme (PBS)-listed on the high cost drug program for patients aged 6 years and older with CF carrying at least one Gly551Asp mutation or one of the other eight gating mutations.

The Phe508del mutation results in a processing defect with almost no protein available at the epithelial membrane. Where small amounts of the protein are present or can be rescued, it nonetheless has a gating defect and is unstable and more rapidly cleared. A combination of therapies aimed at both rescuing and potentiating Phe508del was therefore considered for patients with CF and the Phe508del mutation. Lumacaftor, a CFTR corrector that targets class II defects, was shown in vitro to correct p.Phe508del CFTR misprocessing and increase the amount of available protein.24 Ivacaftor was shown in vitro to potentiate p.Phe508del,25 and the combination of the two drugs in vitro resulted in greater chloride transport than either drug alone.24 The in vitro experience was confirmed in clinical trials, as monotherapy with each of these agents was found not to have clinical benefit.26,27 Combination therapy with lumacaftor and ivacaftor was shown in a large phase 3 trial in patients aged 12 years and older with CF and who were homozygous for the Phe508del mutation to provide modest improvement in lung function (mean absolute improvement in FEV1, 2.6–4%) and reduce pulmonary exacerbations as well as provide nutritional benefit.28 Combination therapy with lumacaftor and ivacaftor was approved by the FDA in July 2015 for patients aged 12 years and older who are homozygous for Phe508del. In Australia, lumacaftor–ivacaftor combination therapy has been approved by the TGA but is not funded for clinical use at this time.

There has been less success to date with CFTR modulator therapies for class 1 mutations,29 and no drugs are currently approved for patients with such mutations. The targeted therapy approach has shown that it is possible to correct the underlying defect in some patients with CF and achieve clinically important benefit.30 Specific approaches are required for different CFTR mutations and differing clinical benefits have been observed. The potential for long term disease modification over time and in early life will need to be taken into account when considering the overall clinical and health economic benefit of these therapies. In addition, maintaining optimal outcomes from the use of ivacaftor, lumacaftor–ivacaftor combination or other CFTR modulator therapies as they become available in clinical use will require careful assessment of patient CF genotype and assessment of the potential for clinical benefit as well as education and support of patients and families to ensure these medicines are taken correctly and appropriately with long term monitoring for adverse events and drug interactions.

Lung cancer

Lung cancer is the most common cause of cancer mortality in Australia, with nearly 9500 deaths annually.31 Tumours with similar pathological appearance may have different genetic abnormalities. Specific mutations can be used to guide optimal treatment. In lung cancer, epidermal growth factor (EGF) binds to its specific receptor causing tumour growth. In early trials that recruited unselected patients, EGF receptor tyrosine kinase inhibitors (EGFR-TKIs) gefitinib and erlotinib were disappointing until it was recognised that the dramatic responses seen in a subset of patients32 were in patients whose tumours carried specific EGFR gene mutations: deletions within exon 19 or a specific point mutation in exon 21. EGFR-TKI treatment is superior to cytotoxic chemotherapy only when these specific mutations are present (Box 4). EGFR mutation testing should be performed on all advanced non-squamous non-small cell lung cancer (NSCLC). This requires larger tissue biopsy33,34 but expertise to meet this need exists and blood testing for tumour DNA may soon be available. Typical time before progressive disease emerges is 2–3 years, which well exceeds that of previously standard cytotoxic chemotherapy.

Additional targets have been identified such as the EML4-ALK and ROS-1 fusion genes. Crizotinib is an available targeted treatment with a high response rate for EML4-ALK-mutated tumours. It is also effective for tumours that carry a ROS-1 fusion gene but it is not presently subsidised under the PBS for this purpose. Tumour progression after a good initial response to EGFR-TKIs and EML4-ALK inhibitors is eventually seen. Newer agents such as osmertinib in the case of EGFR mutations and alectinib for EML4-ALK mutations are effective for the common additional resistance mechanisms but none is presently available via the PBS.35 These oral targeted treatments have side effects and a treatment benefit versus toxicity trade-off must be made. A distinctive rash is common with EGFR-TKIs and should be aggressively managed. Crizotinib has gastrointestinal effects and some visual symptoms are common. Adverse effects are less than with cytotoxic chemotherapy but may not be trivial and persist for as long as treatment continues.

Immune checkpoint inhibitors are a class of treatments with similar promise (Box 4). They prevent interactions between the programmed death 1 (PD1) protein and its ligand that suppress host versus tumour immunity. Unlike the tumour mutations described, PD1 expression is not a great predictor of the effectiveness of such treatments. The response rate is not as high as with EGFR-TKIs but there may be more enduring responses.

Between 500 and 1000 Australians annually may be suitable for current targeted treatment — more than the total incidence of many common cancers. If second line TKI treatments and checkpoint inhibitors are as effective as initial data suggest, careful consideration of cost, benefit and equity of access will be required. Oral treatments do provide some offset by reducing day-stay infusion costs. Refusal of funding for checkpoint inhibitors will be ethically challenging if the effect is as great in lung cancer as it is in melanoma, where access is already provided.

Related issues

Study design

Targeted therapy raises several issues (Box 5) regarding study design that require a re-examination of the traditional parallel group RCT used to establish drug efficacy. Large parallel group RCTs use representative samples of the population in order to test the efficacy of a therapy. However, targeted treatments are only effective in a subset of patients, specifically those in whom the targeted pathway or mutation is active. This means that accurate subject selection is a key part of the evaluation of a targeted treatment, and failure to do this can result in the rejection of a potentially effective therapy. There are examples of this in both severe asthma and NSCLC trials. A study of mepolizumab in unselected asthma patients was negative,36 indicating that while the targeted therapy may be highly effective in patients with an active eosinophil pathway,1012 it may not be effective in those without this pathway operating. Similarly, initial trials of EGFR-TKIs involving unselected populations with NSCLC found no effect on survival.37 Consequently, privileging RCTs when trying to assess the effects of targeted treatment has been called into question.38

A further study design problem that occurs with targeted therapy relates to clinical trial access. Trials are more challenging in infants and young children, patients with more severe illness and patients with rarer disease subtypes, such as those involving rarer CFTR mutations. New approaches will be needed to ensure equitable access to new therapies for all patients with diseases undergoing a targeted therapy approach. As more patients start to use targeted therapy, the pool of patients naive to targeted treatment will decline, which will make placebo-controlled trials less possible. Comparative effectiveness studies will likely be used.

Maintaining adherence remains a challenge for all therapies. For example, CFTR modulator therapies require oral administration with fatty food to maximise absorption and adherence can be suboptimal.39 Patients may also feel healthier taking targeted therapies and there is a risk of reduced adherence with other maintenance therapies. How this may affect the long term benefits of treatment is unknown, although the existing clinical trials of targeted therapy have been undertaken with careful support of patients to maintain their therapeutic regimens.

Cost

The financial costs of currently available targeted therapies are high, with costs estimated at up to $300 000 per patient per year for some therapies. Currently, relatively small numbers of patients with lung cancer, CF or severe asthma benefit from the personalised medicines available. As a society, we will need to discuss and plan how we can manage the costs of personalised medicine into the future to enable equitable access as new medicines become available and the overall numbers of patients using highly expensive medicines grow. Australia has developed a framework to address aspects of this developing treatment paradigm.40

Therapies are currently approved based on an average improvement in a clinical outcome, yet in reality they only help a small proportion of the people who receive them. For example, the top ten highest-grossing drugs in the US help between one in 25 and one in four people who use them.41 This has led to calls for greater implementation of personalised medicine as a means to better use resources based on nationally consistent collection of data.1,3,38

Conclusion

Targeted therapy is a new treatment paradigm that has demonstrated improved efficacy for serious chronic respiratory diseases such as severe asthma, CF and lung cancer. Implementing targeted therapy offers hope to many patients with chronic respiratory diseases but also creates challenges for study design and equitable delivery of new medications. Health economic considerations require that clinicians cease the therapy if the expected response is not seen in individual patients.

Box 1 –
Targeted therapies for chronic respiratory diseases

Disease

Target

Therapy

Biomarker

Bio-effect

Clinical effect


Severe asthma

IL-5

Mepolizumab; reslizumab

Eosinophils (blood)

Reduced eosinophils

Reduced asthma exacerbations

Cystic fibrosis

CFTR

Ivacaftor

CFTR gating mutations: eg, Gly551Asp

Reduced sweat chloride

Increased lung function, improved nutrition and reduced exacerbations

Lumacaftor–ivacaftor combination

Phe508del-CFTR mutation

Reduced sweat chloride

Increased lung function, improved nutrition and reduced exacerbations

Lung cancer

EGFR-TKI; EML4-ALK; ROS-1

Gefitinib; erlotinib; crizotinib*

EGFR mutation; gene fusion

Tumour regression

Increased progression-free survival


IL-5 = interleukin-5. CFTR = cystic fibrosis transmembrane conductance regulator. EGFR-TKI = epidermal growth factor receptor tyrosine kinase inhibitor. * Crizotinib is approved by the Therapeutic Goods Administration but is not listed on the Pharmaceutical Benefits Scheme.

Box 2 –
Targeted therapy in asthma blocks the interleukin (IL)-5 pathway in severe refractory eosinophilic asthma


IL-5 is produced by T helper type 2 lymphocytes (Th2), mast cells and innate lymphoid cells type 2 (ILC2), which leads to eosinophil influx into the blood and airway. Anti-IL-5 monoclonal antibody blocks the effects of IL-5 on eosinophils and reduces asthma exacerbations.

Box 3 –
CFTR mutational classes and molecular consequences in cystic fibrosis


A: Class III mutations cause reduced cystic fibrosis transmembrane conductance regulator (CFTR) chloride (Cl) channel opening owing to defective channel gating or regulation. Ivacaftor increases CFTR channel opening. B: Class II mutations cause CFTR processing defects owing to misfolding of CFTR and degradation by the proteasome. Lumacaftor rescues the misfolded protein.

Box 4 –
Targeted therapy in non-small cell lung cancer


A: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) block EGFR-TK and lead to reduced cancer cell proliferation and metastasis. B: Blockade of programmed death 1 (PD1) or its ligand (PDL1) increases host anti-tumour responses and causes tumour cell death. Host immune surveillance via the T cell that uses T cell receptor (TCR) recognition of tumour major histocompatibility complex (MHC) antigens can be subverted by tumour expression of PD1; PD1 or PDL1 blockade increases host anti-tumour responses and causes tumour cell death.

Box 5 –
Targeted medicine: unresolved questions


  • What is the best name for this approach? Targeted therapy, personalised medicine or precision medicine?
  • What are the implications for study design, drug regulation, health economics and companion diagnostics?
  • Moving beyond targeted pharmacotherapy: can this approach be generalised to problem-based management of chronic disease?
  • Translating into practice: what are the best models to implement targeted therapy in practice?

[Correspondence] Chronic obstructive pulmonary disease: time to discuss new concepts

The adoption of precise concepts, decades ago, is one of the landmarks that has allowed enhancement of the knowledge about chronic obstructive pulmonary disease (COPD).1,2 Chronic bronchitis is clinically characterised, emphysema is pathologically defined, and COPD is defined as a respiratory function abnormality. These classic definitions are still the foundations for appropriate clinical communication and scientific discussions.3

[Comment] Zoonotic tuberculosis in Africa: challenges and ways forward

Outbreaks of Ebola virus, Middle East respiratory syndrome coronavirus, and Zika virus have highlighted the importance of the One Health initiative, and have challenged the field of public health to think about disease management and surveillance within multispecies host–pathogen ecosystems. The threat of zoonotic tuberculosis presents another case in point: millions of people exposed to unregulated mobile livestock—especially in Africa—are at risk of zoonotic tuberculosis from a range of mycobacterium species from these animals.