Skip to main content

Main menu

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • COVID-19 submission information
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • COVID-19 submission information
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions

The impact of personalised therapies on respiratory medicine

J. Stuart Elborn
European Respiratory Review 2013 22: 72-74; DOI: 10.1183/09059180.00008212
J. Stuart Elborn
Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queens University, Belfast, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: s.elborn@qub.ac.uk
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Stratified approaches to treating disease are very attractive, as efficacy is maximised by identifying responders using a companion diagnostic or by careful phenotyping. This approach will spare non-responders form potential side-effects. This has been pioneered in oncology where single genes or gene signatures indicate tumours that will respond to specific chemotherapies. Stratified approaches to the treatment of asthma with biological therapies are currently being extensively studied. In cystic fibrosis (CF), therapies have been developed that are targeted at specific functional classes of mutations. Ivacaftor, the first of such therapies, potentiates dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) protein Class III mutations and is now available in the USA and some European countries. Pivotal studies in patients with a G551D mutation, the most common Class III mutation, have demonstrated significant improvements in clinically important outcomes such as spirometry and exacerbations. Sweat chloride was significantly reduced demonstrating a functional effect on the dysfunctional CFTR protein produced by the G551D mutation. Symptom scores are also greatly improved to a level that indicates that this is a transformational treatment for many patients. This stratified approach to the development of therapies based on the functional class of the mutations in CF is likely to lead to new drugs or combinations that will correct the basic defect in many patients with CF.

  • Cystic fibrosis
  • personalised medicine
  • potentiator therapy

Treatment approaches for respiratory diseases have generally involved all patients with a given condition receiving the same therapy. This assumes that the underlying disease pathophysiology is similar for all patients and that all patients will have the same treatment response to a therapy. As this is invariably not the case, there are increased efforts to develop personalised therapies. The series of articles in the current issue of the European Respiratory Review has explored this approach in some detail, describing its application to respiratory diseases, in general, and cystic fibrosis, in particular [1–4].

Early examples of personalised or stratified medicine mostly stem from the field of oncology [2]. An archetypal example is trastuzumab, better known as Herceptin®, for adjunctive treatment of patients with human epidermal growth factor receptor 2 (HER2) gene-amplified breast cancer [5]. Two clinical trials have shown that trastuzumab was associated with a 33% reduction in the risk of death (p=0.015) [6] and improved 3-yr recurrence-free survival [7], compared to patients who did not receive therapy. In the field of endocrinology, Pearson et al. [8] reported that diabetic patients with mutations in the gene encoding a subunit of the pancreatic ATP-sensitive channel responded better to sulfonylurea therapy treatment than insulin.

Examples of personalised medicine in the field of respiratory medicine are also mostly in the area of oncology, where knowledge of tumour-related mutations has been used to refine the selection of chemotherapy. The kinase inhibitor crizotinib was specifically developed to treat nonsmall cell lung cancer (NSCLC) patients who have a chromosomal transformation that activates the anaplastic lymphoma kinase (ALK) gene [9]. Updated results of a phase I trial assessing crizotinib in ALK-positive NSCLC patients showed that 87 (60.8%) out of 143 patients had an objective response (95% CI 52.3–68.9%) and median progression-free survival was 9.7 months (95% CI 7.7–12.8 months) [10].

Extending these approaches to non-malignant respiratory disease presents exciting possibilities, and has already led to new approaches to phenotyping in areas such as chronic obstructive pulmonary disease, asthma, lung fibrosis or cancer using, for example, serum markers, blood count, clinical phenotypes, somatic genotype, germinal genotype and co-morbidities. Not all of these approaches will necessarily end up being applicable in practice, but they are worthy of investigation. Multiple genome-wide association studies (GWAS) have been published in respiratory medicine, mostly relating to genes associated with asthma development or susceptibility to chronic obstructive pulmonary disease [11]. Recently, there has been increased interest in the role of interleukin (IL)-33 in asthma. IL-33 is associated with promoting a type-1 T-helper cell response and may play a role in chronic inflammation and airway remodelling [12]. Moreover, IL-33 gene polymorphisms have been identified in various asthma GWAS analyses [2, 13, 14].

The recent identification of the ability of ivacaftor, a cystic fibrosis transmembrane conductance regulator (CFTR) modulator, to treat some patients with cystic fibrosis (CF) is the first example in which a stratified medicine approach has resulted in a major change in therapy for patients with this disease who carry the gating channel mutation, G551D [15].

CFTR MODULATORS: A POTENTIAL PARADIGM FOR “PERSONALISED RESPIRATORY MEDICINE”

Ivacaftor, formerly known as VX-770, was identified from a library of 228,000 diverse drugs using high-throughput screening [13]. The candidate agent was subsequently shown to increase the channel opening probability of G551D CFTR by approximately six-fold and to partially restore fluid regulation and cilia beating in cultured CF human bronchial epithelia with the G551D/F508del genotype [3, 16]. These results provided convincing evidence and rapidly advanced into clinical trials.

The efficacy and safety of ivacaftor was evaluated in two 48-week, randomised, double-blind, placebo-controlled trials in patients aged 6–11 yrs (ENVISION study) and patients aged ≥12 yrs (STRIVE study) who have at least one copy of the G551D-CFTR mutation [4].

Ivacaftor was shown to result in a rapid and sustained improvement in lung function through to week 24 and this effect continued through to week 48. Sweat chloride concentration significantly decreased in both trials to levels below the diagnostic cut-off point for CF. Moreover, treated patients in the ENVISION and STRIVE studies demonstrated significant improvements in body weight. The most common reported adverse events were abdominal pain, diarrhoea, dizziness, rash, upper respiratory tract reactions, headache and bacteria in sputum.

A separate phase II trial, DISCOVER, assessed the efficacy of ivacaftor in patients homozygous for the F508del-CFTR mutation [17]. No observed clinical effect was measured between the treatment and placebo arms, suggesting that CFTR potentiation with ivacaftor alone will not benefit CF patients who are homozygous for F508del-CFTR. The reason for this is clear; ivacaftor is a CFTR potentiator that targets gating defects in G551D-CFTR channels. F508del mutation defects, however, can only be repaired through use of a CFTR corrector, such as VX-808 [3].

Emerging CFTR modulators

Ivacaftor, the first CFTR modulator to be approved for clinical use, can be only prescribed to approximately 4–5% of CF patients who have the G551D mutation. If successful, other emerging CFTR therapies will hold greater promise to treat many more CF patients (table 1).

View this table:
  • View inline
  • View popup
Table 1. Mechanism of action and targets of emerging cystic fibrosis transmembrane conductance regulator (CFTR) modulators

The read through agent, ataluren, is undergoing clinical trials to treat CF in patients with mutations caused by premature stop codons. Ataluren was linked to improvement in cough over 3 months and increases in CFTR activity, as measured by the nasal potential difference [18, 19]. Results of a phase III pivotal trial are expected soon and are eagerly awaited.

VX-809 (or lumicaftor) is a CFTR corrector that has been assessed in a phase IIa study of patients homozygous for the F508del-CFTR mutation. The drug, which was well tolerated with no significant safety signals, caused a moderate reduction in sweat chloride compared to placebo [20]. Surprisingly, no other clinically significant improvements were measured. Nevertheless, the trial provided a basis for investigation into the safety and efficacy of lumicaftor plus ivacaftor combination therapy in CF patients homozygous or heterozygous for the F508del-CFTR mutations [21].

Impact of personalised medicine on CF care

Based on experience with ivacaftor, the potential for CFTR modulators (alone or combined) to transform the lives of a significant number of CF patients is exciting. This optimism must also be guarded: many trials of promising drug candidates are still in their infancy and the long-term effect of these therapies is still unknown. Nevertheless, as personalised therapies, CFTR modulators truly represent a milestone in CF treatment.

CONCLUSION

The theoretical benefit of personalised respiratory medicine is almost beyond reproof. However, despite an increasing number of success stories, such as crizotinib, trastuzumab, imatinib and ivacaftor, the development of personalised therapies remains a significant challenge; one that is perhaps above and beyond those typically faced by the pharmaceutical industry, which also has to consider how to recoup the not-insignificant development costs in a specialised and limited market. Overcoming these challenges will involve input and consensus from funders, scientists, providers, patients and regulatory authorities. Nevertheless, personalised medicine has the genuine potential to revolutionise healthcare as we know it.

Footnotes

  • Provenance

    Publication of this peer-reviewed article was supported by Vertex Pharmaceuticals Inc., USA (principal sponsor, European Respiratory Review issue 127).

  • Statement of Interest

    J.S. Elborn has received fees for consultancy and speaking from Vertex. He received payment as a grant for three clinical trials (€50,000).

  • Received December 20, 2012.
  • Accepted January 15, 2013.
  • ©ERS 2013

REFERENCES

  1. ↵
    1. Elborn JS
    . Personalised medicine for cystic fibrosis: treating the basic defect. Eur Respir Rev 2013; 22: 3–5.
    OpenUrlFREE Full Text
  2. ↵
    1. Hall IP
    . Stratified medicine: drugs meet genetics. Eur Respir Rev 2013; 22: 53–57.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Derichs N
    . Targeting a genetic defect: cystic fibrosis transmembrane conductance regulator modulators modulators in cystic fibrosis. Eur Respir Rev 2013; 22: 58–65.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Sermet-Gaudelus I
    . Ivacaftor treatment in patients with cystic fibrosis and the G551D-CFTR mutation. Eur Respir Rev 2013; 22: 66–71.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Slamon DJ,
    2. Leyland-Jones B,
    3. Shak S,
    4. et al
    . Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344: 783–792.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Romond EH,
    2. Perez EA,
    3. Bryant J,
    4. et al
    . Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005; 353: 1673–1684.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Joensuu H,
    2. Kellokumpu-Lehtinen PL,
    3. Bono P,
    4. et al
    . Adjuvant docetaxel or vinorelbine with or without trastuzumab for breast cancer. N Engl J Med 2006; 354: 809–820.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Pearson ER,
    2. Flechtner I,
    3. Njolstad PR,
    4. et al
    . Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 2006; 355: 467–477.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Ou SH,
    2. Bartlett CH,
    3. Mino-Kenudson M,
    4. et al
    . Crizotinib for the treatment of ALK-rearranged non-small cell lung cancer: a success story to usher in the second decade of molecular targeted therapy in oncology. Oncologist 2012; 17: 1351–1375.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Camidge DR,
    2. Bang YJ,
    3. Kwak EL,
    4. et al
    . Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol 2012; 13: 1011–1019.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Todd JL,
    2. Goldstein DB,
    3. Ge D,
    4. et al
    . The state of genome-wide association studies in pulmonary disease: a new perspective. Am J Respir Crit Care Med 2011; 184: 873–880.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Prefontaine D,
    2. Nadigel J,
    3. Chouiali F,
    4. et al
    . Increased IL-33 expression by epithelial cells in bronchial asthma. J Allergy Clin Immunol 2010; 125: 752–754.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Smith DE
    . IL-33: a tissue derived cytokine pathway involved in allergic inflammation and asthma. Clin Exp Allergy 2010; 40: 200–208.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Ober C,
    2. Yao TC
    . The genetics of asthma and allergic disease: a 21st century perspective. Immunol Rev 2011; 242: 10–30.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Clancy JP,
    2. Jain M
    . Personalized medicine in cystic fibrosis: dawning of a new era. Am J Respir Crit Care Med 2012; 186: 593–597.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Van Goor F,
    2. Hadida S,
    3. Grootenhuis PD,
    4. et al
    . Correction of the F508del-CFTR protein processing defect in vitro by the investigational drug VX-809. Proc Nat Acad Sci USA 2011; 108: 18843–18848.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Flume PA,
    2. Liou TG,
    3. Borowitz DS,
    4. et al
    . Ivacaftor in subjects with cystic fibrosis who are homozygous for the F508del-CFTR mutation. Chest 2012; 142: 718–724.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Kerem E,
    2. Hirawat S,
    3. Armoni S,
    4. et al
    . Effectiveness of PTC124 treatment of cystic fibrosis caused by nonsense mutations: a prospective phase II trial. Lancet 2008; 372: 719–727.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Sermet-Gaudelus I,
    2. Boeck KD,
    3. Casimir GJ,
    4. et al
    . Ataluren (PTC124) induces cystic fibrosis transmembrane conductance regulator protein expression and activity in children with nonsense mutation cystic fibrosis. Am J Respir Crit Care Med 2010; 182: 1262–1272.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Clancy JP,
    2. Rowe SM,
    3. Accurso FJ,
    4. et al
    . Results of a phase IIa study of VX-809, an investigational CFTR corrector compound, in subjects with cystic fibrosis homozygous for the F508del-CFTR mutation. Thorax 2012; 67: 12–18.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Pettit RS
    . Cystic fibrosis transmembrane conductance regulator-modifying medications: the future of cystic fibrosis treatment. Ann Pharmacother 2012; 46: 1065–1075.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
View this article with LENS
Vol 22 Issue 127 Table of Contents
  • Table of Contents
  • Table of Contents (PDF)
  • Cover (PDF)
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
The impact of personalised therapies on respiratory medicine
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
The impact of personalised therapies on respiratory medicine
J. Stuart Elborn
European Respiratory Review Mar 2013, 22 (127) 72-74; DOI: 10.1183/09059180.00008212

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
The impact of personalised therapies on respiratory medicine
J. Stuart Elborn
European Respiratory Review Mar 2013, 22 (127) 72-74; DOI: 10.1183/09059180.00008212
del.icio.us logo Digg logo Reddit logo Technorati logo Twitter logo CiteULike logo Connotea logo Facebook logo Google logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • CFTR MODULATORS: A POTENTIAL PARADIGM FOR “PERSONALISED RESPIRATORY MEDICINE”
    • CONCLUSION
    • Footnotes
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

Subjects

  • CF and non-CF bronchiectasis
  • Genetics
  • Interstitial and orphan lung disease
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

  • Role of air pollutants in airway epithelial barrier dysfunction
  • E-cigarettes and nicotine abstinence
  • Lung imaging in cystic fibrosis
Show more Review

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERR

  • Journal information
  • Editorial board
  • Reviewers
  • Press
  • Permissions and reprints
  • Advertising
  • Sponsorship

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN: 0905-9180
Online ISSN: 1600-0617

Copyright © 2023 by the European Respiratory Society