Article Text
Abstract
Messenger RNA (mRNA) vaccines as a novel vaccine platform offer new tools to effectively combat both emerging and existing pathogens which were previously not possible. The ‘plug and play’ feature of mRNA vaccines enables swift design and production of vaccines targeting complex antigens and rapid incorporation of new vaccine constituents as needed. This feature makes them likely to be adopted for widespread clinical use in the future.
Currently approved mRNA vaccines include only those against SARS-CoV-2 virus. These vaccines demonstrate robust immunogenicity and offer substantial protection against severe disease. Numerous mRNA vaccines against viral pathogens are in the early to late phase of development. Several mRNA vaccines for influenza are tested in clinical trials, with some already in phase 3 studies. Other vaccines in the early and late phases of development include those targeting Cytomegalovirus, varicella zoster virus, respiratory syncytial virus and Epstein-Barr virus. Many of these vaccines will likely be indicated for immunosuppressed populations including those with autoimmune inflammatory rheumatic diseases (AIIRD). This review focuses on the mechanism, safety and efficacy of mRNA in general and summarises the status of mRNA vaccines in development for common infectious diseases of particular interest for patients with AIIRD.
- Vaccination
- Autoimmune Diseases
- Therapeutics
Statistics from Altmetric.com
WHAT IS ALREADY KNOWN
Patients with autoimmune inflammatory rheumatic diseases (AIIRD) are at higher risk for serious infections and require vaccines against preventable diseases.
Messenger RNA (mRNA) vaccines consisting of antigen-encoding mRNA in a nanoparticle carrier can be swiftly designed, produced and adapted to evolving pathogens.
The COVID-19 mRNA vaccine has demonstrated remarkable efficacy and acceptable safety concerns of this novel mRNA vaccine platform.
WHAT ARE THE NEW FINDINGS
Advancement in mRNA design and optimisation allows for the development of vaccines against diseases that present significant technical challenges from a vaccine standpoint, such as respiratory syncytial virus, Epstein-Barr virus and Cytomegalovirus; as well as enhancing efficacy against other targets (eg, influenza vaccine).
Several mRNA or mRNA targeting multiple epitopes can be integrated into a single carrier, leading to the development of combination vaccines or multivalent vaccines with enhanced efficacy.
Several mRNA vaccines targeting common infections such as influenza are currently in the various phases of development and clinical trials.
The efficacy and safety of novel mRNA vaccines in patients with AIIRD need to be examined in robust clinical trials.
Introduction
Vaccination in rheumatic diseases
Patients with autoimmune inflammatory rheumatic diseases (AIIRD) are at higher risk for serious infections due to underlying immune dysfunction, treatment-associated immunosuppression or both.1–3 Although these patients should receive the recommended vaccines, the vaccine coverage in this population remains suboptimal.4 5 Factors contributing to vaccination hesitance include concerns about disseminated infections from live-attenuated vaccines, the potential risk of exacerbating underlying autoimmune diseases and uncertainties surrounding vaccine efficacy in patients with immunosuppressed AIIRD.6–10 In this context, the novel mRNA vaccine platform has emerged as a promising solution, providing new vaccines with potentially improved efficacy and safety.11
Structure of mRNA vaccines
Instead of using preformed antigens, messenger RNA (mRNA) vaccines introduce mRNA-encoding antigenic proteins into host cells12 (figure 1). Within these cells, the target antigens are synthesised, processed and presented to immune cells, closely mimicking the immune response triggered by a viral infection.13 14 mRNA vaccines comprise two key components: antigen-encoding mRNA and a delivery system. Non-self mRNAs, like viral RNAs, are detected by intracellular nucleotide sensors, such as toll-like receptor (TLR) 7, retinoic acid-inducible gene-I (RIG-1) and anti-melanoma differentiation-associated gene (MDA)-5, initiating a cellular antiviral response that can hinder protein synthesis and degrade nucleic acids to suppress the production of viral particles.15 16 Consequently, vaccine mRNA, being non-self, is eliminated before it can be translated into antigenic proteins. Modifying mRNA such as substitution of uridine with pseudouridine reduces TLR activation, thereby bypassing host antiviral response.17 18 This modification also enhances the biological stability and translational capacity of mRNA, marking a pivotal milestone in the feasibility of mRNA vaccines and mRNA-based cancer and gene therapy, which was acknowledged with a Nobel Prize in Physiology and Medicine in 2023.19 20 Further mRNA editing techniques, including methylpseudouridine, 5’ capping, tailing and codon optimisation (eg, point mutations), have significantly improved mRNA stability and the expression of target antigens.21 22
Mechanism of mRNA vaccines. (A) mRNA vaccines consist of lipid nanoparticle as a vehicle that carries mRNA. mRNA vaccine is given as an intramuscular injection; (1) after injection, mRNA is taken up by host cells via endocytosis. (2) mRNA is released from the endosome into the cytoplasm; (3) mRNA is translated by ribosomes into antigenic proteins; (4) antigenic proteins are secreted and then be taken up by to local antigen-presenting cells, including dendritic cells (DCs). (5) DCs are trafficked to lymph nodes where they activate CD4+ and CD8+ T cells; (6) CD4+T cells differentiate into Tfh cells or Th1 cells; CD8+T cells become circulating cytotoxic T cells; Tfh cells prime B cells to differentiate into antibody-secreting plasma cells. Vaccine mRNAs are recognised by intracellular nucleotide sensing receptors, such as (a) TLR and (b) RIG-I/MDA-5, triggering a type I IFN response. (B) Antibody response following mRNA vaccines. IFN, interferon; IRF, interferon regulatory factor; LPN, lipid nanoparticle; MAVS, mitochondrial antiviral-signalling protein; MDA, melanoma differentiation-associated gene; MHC, major histocompatibility complex; mRNA, messenger RNA; RIG-I, retinoic acid-inducible gene-I; Tfh, follicular helper T cell; Th, helper T cell; TLR, toll like receptor.
An efficient delivery vehicle protects mRNA from degradation by ubiquitous RNases and ensures effective cellular entry and release.23 The most advanced delivery system is lipid nanoparticles (LNPs).24 LNPs consist of an ionisable or cationic lipid that interacts with the polyanionic RNA, a helper phospholipid supporting the bilayer structure, a cholesterol analogue adjusting the fluidity of the lipid bilayer and a polyethylene glycol (PEG)-lipid enhancing colloidal stability while reducing opsonisation. LNPs are engineered to fuse with the target cell surface and to form endosomes. A pH change within the acidic endosome facilitates the release of mRNA into the cytosol, where they are translated into antigenic proteins by cytosolic ribosomes.25
Advantages of mRNA vaccine
mRNA vaccines present several key advantages over other vaccine classes (box 1). As non-live vaccine, they do not produce infectious particles, eliminating the possibility of vaccine-induced infection, even in severely immunocompromised patients. Live-attenuated vaccines including Measles, Mumps and Rubella, varicella, yellow fever and polio are contraindicated for immunosuppressed patients due to the potential risk of life-threatening disseminated infections, while Zostavax, a live-attenuated herpes zoster (HZ) vaccine can be administered to patients receiving low-to-moderate immunosuppression.4 26 However, determining the thresholds for moderate and high immunosuppression remains arbitrary, largely based on expert opinions due to the lack of robust clinical trial data. This is further complicated by the emergence of novel biological and synthetic disease-modifying antirheumatic drugs such as Janus kinase (JAK) inhibitors, Tyk inhibitors and granulocyte macrophage-colony stimulating factor inhibitors, with uncertain extent of immunosuppressive effect.27 28 In this regard, mRNA vaccines with no risk of infection would provide a safer alternative than live vaccines for patients taking any immunosuppressants.
Properties of messenger RNA (mRNA) vaccines.
Simple structure: mRNA vaccines consist of antigen-encoding mRNA in lipid nanoparticles as a delivery system.
Rapid mRNA design and synthesis: Once the target sequence is identified, mRNA can be swiftly designed and synthesised.
mRNA modification and optimisation: mRNA can be modified to optimise antigenicity (and presumed efficacy) to accommodate new evolving variants of pathogens.
Combination vaccines: Multiple mRNA sequences targeting the same or different pathogens can be packaged together for improved efficacy or combination vaccines.
Inbuilt adjuvant: mRNA activates the immune system as an adjuvant. However, this may lead to undesired reactogenicity as a side effect.
No vaccine-induced infection: mRNA vaccines do not produce infectious particles, eliminating the risk of vaccine-induced infection.
Rapid manufacturing: The cost-effective large-scale production enables swift response to public health emergencies.
mRNA vaccines elicit an immune response closely resembling natural viral infections, inducing robust and durable cellular and humoral immunity.13 14 The manipulation of nucleotide sequence allows for fine-tuning of vaccine immunogenicity. Self-amplifying RNAs, which are replicated by host cellular machinery, enhance the production of antigenic proteins, allowing for reduced vaccine dosing and so vaccine side effects.29 30 The limited biological half-life of mRNA ensures complete clearance from the body after sufficient antigen expression, minimising any long-term effects.31 Unlike viral-vector vaccines, LNP carrier do not induce a host immune response against the carrier itself. Adenoviral vector-based COVID-19 vaccines, where host immune response could potentially neutralise the vectors during subsequent vaccinations, can reduce mRNA delivery and accordingly vaccine efficacy.32 Importantly, mRNA does not integrate with the host DNA, eliminating the risk of insertional mutagenesis and cancer risk.
The most significant advantage of mRNA vaccines is the concept of ‘plug and play’. Once the genetic sequence of any target antigen is known, mRNA can be swiftly designed, synthesised and assembled into LNP vehicles. This enables the timely development of new vaccines against emerging infectious diseases, such as Zika infection,33 or adapting current vaccines to rapidly evolving pathogens like COVID-19 and influenza viruses. Multiple mRNAs can be combined in a single combination vaccine to target various pathogens or create multiple antigens from a single pathogen, enhancing vaccine efficacy.24 Finally, mRNA vaccines offer remarkable manufacturing advantages. They can be rapidly and efficiently produced in a cell-free environment, facilitating swift, scalable and cost-effective production.34 Therefore, the rapid response potential of mRNA vaccine platforms along with cost-effect production is well-suited to efficiently respond to future epidemics and pandemics.
Inbuilt adjuvants and immune activation
Both mRNA and LNP possess inherent adjuvant properties.35 36 mRNA is detected by intracellular innate immune sensors, such as cytosolic RIG-1/MDA-5 and endosomal TLR, which stimulate the production of type 1 interferon and proinflammatory cytokines interleukin (IL)-1β and IL-614 (figure 1). These cytokines boost vaccine response while also triggering local and systemic inflammation, observed in 70–80% of COVID-19 vaccine recipients in a dose-dependent manner.37–39 Fine-tuning innate immune activation through modification of mRNA and carrier may optimise the efficacy and safety of mRNA vaccines.18
Lessons learnt from COVID-19 pandemics
The COVID-19 pandemic accelerated the realisation of a long conceptual vaccine platform. Pfizer and Moderna’s mRNA vaccines emerged as the recommended strategy against SARS-CoV-2, demonstrating remarkable efficacy and safety for hundreds of millions globally over the past 2 years.40 First-generation SARS-CoV-2-COVID-19 vaccines were developed within weeks of publication of the full-length Wuhan strain spike protein sequence with the first clinical trial initiated just 4 months after virus isolation.41 42 Within 11 months of the RNA sequence publication, emergency use authorisation was granted for Pfizer and Moderna vaccines, with approximately 90% efficacy demonstrated in phase 3 studies. This efficacy was shown to fade over time, leading to the recommendations for booster vaccinations roughly every 6 months for 2 years.43 However, these vaccines have been shown to produce robust long-term humoral and cell-mediated immunity and provide protection against severe COVID-19 despite waning neutralising antibody titres.44
The emergence of COVID-19 variants posed challenges to vaccine efficacy. Bivalent or multivalent vaccines like mRNA-1273 (Moderna) and BNT162b2 (Pfizer-BioNTech), targeting both Omicron BA.4–BA.5 and the ancestral wild-type spike, as well as the new monovalent mRNA vaccines targeting Omicron XBB1.5 subvariant, theoretically could have enhanced vaccine effectiveness. This rapid adaptation underscores the advantage of mRNA vaccine technology in combating rapidly changing pathogens.45–47
Risks of mRNA vaccines
A large-scale vaccination campaign during the COVID-19 pandemic demonstrated a good safety profile. Nevertheless, sporadic reports of infrequent yet significant adverse events have contributed to vaccine hesitancy (table 1). Mild anaphylactic reactions have been documented in 2.5 cases per million vaccinations with the Moderna vaccine and 2.2 cases per million for the Pfizer-BioNTech vaccine.37 48 This incidence appears to exceed that associated with traditional vaccines.49 PEG allergy has been estimated at 0.15 cases per million person-years in the USA and Canada, implying that other vaccine components contribute to anaphylaxis.50 51 Interestingly, pre-existing antibodies against PEGylated lipids, are present in approximately 40% of the general population, suggesting that anti-PEG antibodies are not the sole causative factor.52 The Centers for Disease Control and Prevention recommends that individuals with a history of anaphylactic reactions to mRNA vaccines avoid them.
Concerns about vaccination in patients with autoimmune inflammatory rheumatic diseases
A small fraction (0.125%) of mRNA-vaccinated individuals experienced severe immune-mediated adverse effects, encompassing a range of conditions such as acute myocardial infarction, Bell’s palsy, cerebral venous sinus thrombosis, Guillain-Barré syndrome, myocarditis/pericarditis (more prevalent among younger age groups), pulmonary embolism, stroke, thrombosis with thrombocytopenia syndrome, lymphadenopathy, appendicitis, HZ reactivation, neurological complications and autoimmunity.53
mRNA vaccines in patients with AIIRD
COVID-19 vaccination in autoimmune diseases study, an international collaborative study involving 167 collaborators from 110 countries, demonstrated a safety profile for COVID-19 vaccines in patients with AIIRD comparable to that of the general population.54–56 However, the flare rate of AIIRD following vaccination ranged from 10% to 37%.57 58 In the COVAX physician-reported registry including 5121 participants across 30 countries, flares were reported in 4.4% of cases, with 0.6% of severe cases.59 In summary, although disease flare is not uncommon, severe flares are rare. Immunosuppressant medications (corticosteroids, methotrexate, abatacept, mycophenolate mofetil and JAK inhibitors) can diminish vaccine immunogenicity and presumably efficacy.60 Further optimisation of mRNA and LNP formulation may mitigate the flare risk and simultaneously enhance vaccine efficacy in patients with AIIRD.
During the COVID-19 pandemic, new AIIRD cases, including inflammatory arthritis, connective tissue disease, systemic vasculitis, were reported after vaccination or infection.61–64 Further investigation is warranted to determine whether preclinical autoimmune diseases manifest clinically or whether they are triggered by the COVID-19 vaccine or infection.
mRNA vaccines in development
This mRNA vaccine technology, validated through the success of the COVID-19 experience, drives the development of mRNA vaccines for other indications. Dozens of new vaccines are currently in phase 1–3 clinical trials, most notably targeting easily communicable respiratory pathogens. Here, we summarised the current mRNA vaccine development tailored to the needs of patients with AIIRD (tables 2 and 3).
Clinical trials on monovalent mRNA vaccines (registered with ClinicalTrials.gov)
Combination/multivalent mRNA vaccines
Influenza
Inactivated, live-attenuated influenza and haemagglutinin (HA) protein subunit influenza vaccines target the highly immunogenic HA protein of the four endemic subtypes A(H1N1), A(H3N2), B-Victoria lineage and B-Yamagata lineage.65 The continuous antigenic drift in the HA protein due to viral mutations warrants annual vaccination with unpredictable vaccine efficacy, often falling below 50%.66 Avian cell cultures, the primary source for inactivated influenza vaccine production, are time-consuming, taking approximately 6 months from strain selection to vaccine distribution. The challenges in predicting virus variants and adhering to production timelines hamper swift adaptation to emerging dominant variants. Repeat vaccinations may diminish cell-mediated vaccine responses.67 68 Universal vaccines using new mRNA vaccines encoding cross-protective T-cell responses to conserved HA stalk or non-HA proteins, such as nucleoprotein, would offer a potential solution, eliminating the need for annual immunisation.69 70 In a study, vaccination with mRNA encoding 20 HAs representing all A and B subtypes induced humoral responses against all antigens, providing protection against influenza in mice and ferrets, even with mismatched strains.71 Moderna is developing at least five mRNA vaccines for influenza.72 The first, using a traditional approach, encodes mRNA for HA proteins from the four dominant strains (H3N2, H1N1, B/Yamagata and B/Victoria), showing mixed results in phase 3, particularly for influenza B strains.73 Other vaccines in early development target a broader range of HA and/or neuraminidase proteins, potentially offering a more comprehensive ‘pan-flu’ vaccine.
Respiratory syncytial virus
Respiratory syncytial virus (RSV) infection can progress into lower respiratory tract disease with a significant morbidity in older adults and particularly those who are immunosuppressed or with other comorbidities (eg, cardiovascular or pulmonary disease).74 It reached epidemic proportions in 2022.75
RSV uses the highly conserved fusion (F) protein to enter the host cells. On encountering a target cell, the prefusion F protein transforms rapidly into elongated postfusion conformation, concealing the primary target of neutralising antibodies. Therefore, neutralising antibodies must bind to the prefusion F protein to block cell entry. The 1960s formalin-inactivated RSV vaccine proved unsuccessful because formalin inactivation triggers postfusion conformation change; the vaccine generated antibodies failed to neutralise the infectious prefusion F protein.76 mRNA engineering enables the synthesis of more stable F protein in the prefusion state.77 Recently two subunit RSV vaccines targeting prefusion F proteins have been approved. AS01E-adjuvant (Arexvy) and the non-adjuvanted (Abrysvo) RSV had an efficacy of 82.6% and 67%, compared with placebo, respectively.78 79 Currently, there are several mRNA RSV vaccines in development. In the ConquerRSV trial, mRNA-1345 (Moderna) vaccine, targeting prefusion F glycoprotein, showed vaccine efficacy of 83.7% against RSV lower respiratory tract disease in older adults.80 At least one has shown the ability to produce robust neutralising antibody and cell-mediated responses,81 and safety data published for another suggests a profile that is comparable to COVID-19 mRNA vaccines in that a majority of the patients have non-serious local or systemic reactogenicity.82 These mRNA vaccines, as well as the two recently approved protein-based vaccines, need to be evaluated in patients with AIIRD and other immunosuppressed populations.
Of note, the novel bivalent RSV prefusion F vaccine has shown a vaccine efficacy of 66.7% (96% CI 28.5% to 85.8%).79 In a recent study, mRNA-based RSV preF exhibited a vaccine efficacy of 83.7% (96% CI 66.0% to 92.2%).83 To further clarify the comparative effectiveness between mRNA and preF protein-based vaccines, a head-to-head trial is warranted.
Zoster
HZ is the reactivation of latent varicella zoster (VZV), which manifests by a vesicular rash along dermatomes and pain.84 Incidence of shingles is increased in patients with AIIRD.85 The protein-adjuvanted HZ subunit (HZ/su) vaccine (Shingrix, GSK) has received little study in patients with AIIRD with a paucity of efficacy or safety data in this setting, despite its proven long-lasting efficacy in the general population.86 In a recent non-randomised clinical trial involving 82 patients with rheumatoid arthritis (RA) treated with Janus kinase inhibitors, the HZ/su vaccine induced a humoral response with an acceptable safety profile. However, the serological response was diminished in patients with RA compared with healthy controls (80.5% vs 98.0%). Cell-mediated immunity was not assessed.87 An mRNA-based shingles vaccine is currently in phase 1/2 clinical trial. A key benefit of mRNA technology is to provide robust immunity to pathogens that are rapidly changing pathogens, which may not be crucial for a latent varicella infection like VZV. Therefore, whether the mRNA vaccine would provide additional benefit to the existing protein-based vaccine remains to be seen.
Cytomegalovirus
Cytomegalovirus (CMV) infection in immunocompromised patients results from reactivation of latent CMV reinfection.88 Approximately 1 in 200 babies are born with CMV infection, and 1 in 5 of them will experience devastating sequelae including hearing loss, seizure and blindness.89 Controlling CMV infection might be crucial, especially in childbearing patients with AIIRD with dysfunctional T-cell immunity such as women with systemic lupus erythematosus (SLE).90–92
For effective immunity against CMV, targeting six distinct proteins, including a glycoprotein B (one mRNA sequence) and the glycoprotein subunits of the pentameric complex (five mRNA sequences) is essential. The ability of mRNA vaccines, allowing the incorporation of six different mRNAs, each encoding a specific protein, has greatly facilitated the development of a vaccine against this pathogen.93 It is a crowning example of ‘plug and play’ aspect of mRNA vaccines. Moderna’s mRNA-1647 has been tested in diverse clinical scenarios including post-haematopoietic stem cell transplantation. Early phase 2 data suggest typical levels of reactogenicity for an mRNA vaccine and a robust antibody response.94
Epstein-Barr virus
Epstein-Barr virus (EBV) establishes a lifelong infection in over 95% of individuals by their 40s. While often asymptomatic in childhood, its clinical implications become more significant later in life, with approximately 1% of patients experiencing serious complications, such as hepatitis and neurological issues.95 EBV has been implicated in the development of various cancers, including Burkitt and Hodgkin’s lymphomas, as well as several autoimmune disorders like RA, SLE and multiple sclerosis.96 97 As such, a prophylactic vaccine holds the potential to reduce the severe EBV infection and potentially the associated risks of malignancies and autoimmune diseases. Multiple vaccine classes, including subunit vaccines, epitope vaccines and DNA vaccines, are currently in development. Moderna’s mRNA-1189, designed to target glycoproteins gp-350, gB, gH/gL and gp42 is currently in phase 1 clinical study.98 99
Pneumococcal vaccines
The risk for invasive infection with Streptococcal pneumoniae is increased in patients with AIIRD. Currently, the 23-valent pneumococcal polysaccharide vaccine, the 13-valent and 20-valent pneumococcal conjugate vaccine are available. To date, there is no mRNA vaccine for pneumococcal pneumonia.
Multivalent and combination vaccines
mRNA technology can encode epitopes from one or more pathogens simultaneously. Moderna’s mRNA-1045 vaccine targets both RSV and influenza, whereas mRNA-1083 targets influenza and COVID-19. Furthermore, mRNA-1230 aims to tackle all three pathogens: influenza, RSV and COVID-19 infection. These vaccines are currently in the early phases of clinical trials. Combining existing and novel vaccines to prevent several common respiratory infections such as influenza, RSV and COVID-19 can postulate the strategy of a ‘pan-respiratory vaccine’. Numerous trials are underway to investigate potential interactions or interference of mRNA vaccines with other vaccines concerning efficacy and safety in clinical settings (table 3). mRNA vaccines as immunosuppressive therapy
The selective inhibition of autoreactive immune cells, as opposed to a broad suppression of the entire immune system, has become increasingly realistic. The transfection of antigen-presenting cells (APCs) with nucleoside-modified, single-strand non-inflammatory mRNA encoding auto-antigens ensures that transfected APCs present auto-antigens with no to low-level expression of co-stimulatory molecules, generating regulatory T cells for peripheral tolerance.100 In experimental autoimmune encephalitis in mice, the introduction of modified N1-methylpresduouridine mRNA, encoding myeline oligodendrocyte glycoprotein, in dendritic cells in lymphoid tissue was effective in mitigating the autoimmune response. The innovative approach to induce a peripheral tolerance holds promise to treat autoimmune diseases characterised by well-defined antigens, such as antiphospholipid syndrome.
Role mRNA vaccines in cancer treatment
By comparing tumour and normal sequences, mRNA encoding tumour-specific or tumour-associated antigens are synthesised and delivered to dendritic cells for presentation to T cells.101 Vaccination with cancer-specific mRNA elicits a robust adaptive immune response with long-term immune memory against cancer antigens, outperforming peptide-based cancer vaccines.102 Combining cancer vaccines with immune checkpoint inhibitors enhances anticancer immunity, showing promising results in the treatment of various cancers, such as lung cancer, melanoma and pancreatic cancer, as evidenced in early-stage clinical trials.103
Conclusions
In summary, mRNA vaccines are reshaping the vaccination landscape, providing new powerful tools not only to combat existing and emerging infectious pathogens but also to potentially treat autoimmune diseases. Even though they are not without safety concerns including reactogenicity, flares of underlying autoimmune diseases and a potential for causing autoimmune disease, the benefit clearly outweighs these potential side effects. Further research is imperative to thoroughly assess the efficacy and safety of new mRNA vaccines in patients with AIIRD.
Ethics statements
Patient consent for publication
Ethics approval
Not applicable.
References
Footnotes
Handling editor Josef S Smolen
Contributors KLW designed and supervised the review. All authors actively participated in data acquisition and analysis, manuscript preparation and approved the final version.
Funding The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for-profit sectors.
Competing interests None declared.
Provenance and peer review Not commissioned; externally peer reviewed.