3,646
Views
29
CrossRef citations to date
0
Altmetric
Review

Biosafety considerations for attenuated measles virus vectors used in virotherapy and vaccination

, , &
Pages 1102-1116 | Received 08 Sep 2015, Accepted 14 Nov 2015, Published online: 22 Mar 2016

ABSTRACT

Attenuated measles virus (MV) is one of the most effective and safe vaccines available, making it attractive candidate vector to prevent infectious diseases. Attenuated MV have acquired the ability to use the complement regulator CD46 as a major receptor to mediate virus entry and intercellular fusion. Therefore, attenuated MV strains preferentially infect and destroy a wide variety of cancer cells making them also attractive oncolytic vectors. The use of recombinant MV vector has to comply with various regulatory requirements, particularly relating to the assessment of potential risks for human health and the environment. The present article highlights the main characteristics of MV and recombinant MV vectors used for vaccination and virotherapy and discusses these features from a biosafety point of view.

Introduction

Attenuated measles virus (MV) represents an attractive vector candidate for virotherapy and vaccination. This is due to several features including: (i) impressive track record of safety and efficacy in human population, (ii) lack of genomic integration in the host cells due to their cytoplasmic replication, (iii) high immunogenicity as vaccine, (iv) compared to other RNA viruses MV maintain high levels of genetic stability,Citation1 (v) large foreign gene(s) insertion capacity (>6 kb)2 and (vi) selectivity to tumor cells in case of virotherapy applications.

Many of the attenuated strains in use are derived from the MV Edmonston strain (MV-Edm) isolated in 1954 from the throat washings and blood of a child with measles, in a primary culture of human kidney cells.Citation3 This isolate was subsequently adapted to various types of cultured cells, giving rise to attenuated Edmonston A and B seeds.Citation4 Further passages of Edmonston A and B seeds on chicken embryo fibroblasts (CEF) produced the more attenuated Schwarz and Moraten viruses.Citation5 summarizes the characteristics of the different measles vaccine strains derived from the Edmonston strain. Administration of these vaccines has dramatically reduced the incidence of measles. These vaccines are among the most effective and safe human vaccines in use providing long-lasting protection.Citation17 These characteristics make measles vaccine attractive as a viral vector backbone for the development of recombinant vaccines against other viral infections such as human immunodeficiency virus (HIV), SARS coronavirus (SARS-CoV) and flavivirus infections.Citation18-20 Nucleotide differences between the different attenuated vaccine strains used as vectors and MV-Edm are indicated in . Attenuation is not determined by the absolute number of substitutions, because the Zagreb strain which is more attenuated than Edmonston B strain has fewer substitutions, as compared with MV-Edm. Schwarz and Moraten strains have identical nucleotide sequences, despite their divergent passage histories. Because both strains have been passaged in CEF at reduced temperatures, it is possible that similar cell culture conditions may have resulted in similar nucleotide substitutions. Despite the diverse geographic origins of the progenitors and the variations in cell culture systems, incubation temperatures, and passage numbers, the genome of vaccines demonstrates sequence similarity.Citation5 The choice of a strain for the design of the recombinant vector is based on the attenuation of the strain and its safe use as vaccine rather than nucleotide sequence analysis.

Table 1. Main characteristics of the wild-type MV strain and the live-attenuated measles vaccines derived from the Edmonston virus.

Other measles vaccines were derived from wild-type progenitors isolated independently in Russia (Leningrad-4), Japan (CAM-70), and China (Shangai-191).Citation5

The wild-type (wt) MV enters cells predominantly via the signaling lymphocyte activation molecule (SLAM also known as CD150), mainly expressed on subsets of lymphocytes, thymocytes, macrophages and mature dendritic cells (DCs). In contrast all laboratory adapted attenuated MV-Edm strains have acquired the ability to use, besides CD150, the complement regulator CD46 (also known as membrane cofactor protein; MCP) as receptor to mediate virus entry and intercellular fusion. This receptor is overexpressed on the surface of malignant cells protecting them against complement mediated cell lysis.Citation21 Another cellular receptor, Nectin-4 (also known as poliovirus receptor-like protein 4; PVRL4), expressed on primary airway epithelial cells and also overexpressed in many tumor types, has also been identified as a receptor for MV viral entry.Citation22,23 Therefore, attenuated MV strains preferentially infect and destroy a wide variety of cancer cells making them attractive oncolytic vectors.Citation24

Recombinant, attenuated MV strains are currently being tested in several phase I clinical trials as vaccine against HIVCitation25 or chikungunya virusCitation26 and as oncolytic vector in ovarian cancer, glioblastoma multiforme, multiple myeloma, head and neck cancer, and mesothelioma.Citation24,27-29

The use of recombinant, genetically modified (GM) viral vectors for pre-clinical and clinical trials must comply with several European Union legislations including the legal provisions on biosafety aiming at protecting public health and the environment against potentially adverse effects of genetically modified organisms. Activities involving manipulation of GM viral vectors in contained conditions (e.g. laboratories, animal husbandries, production facilities, hospital rooms) may comply with Directive 2009/41/EC.Citation30 Activities involving their deliberate release into the environment require that a case-by-case environmental risk assessment (ERA) should be carried out before release according the principles defined in annex II of Directive 2001/18/EC.Citation31 The ERA is also part of the procedure for marketing authorization.Citation32 The general steps underlying an ERA of viral vectors have been discussed in Baldo et al.Citation33

This article focuses on biosafety issues in the European Union when performing clinical trials with recombinant attenuated MV vectors. The risk related to research and development activities and large scale production of these vectors are not developed in the present review.

Hazard related to the molecular and biological characteristics of measles viruses

Wild-type measles virus

Measles virus is an enveloped negative-strand RNA virus of the genus Morbillivirus within the Paramyxoviridae family and is the causative agent of the acute, exanthemous, infectious measles disease.Citation34 The MV genome encodes 8 proteins: 2 non-structural proteins (V and C), a phosphoprotein (P), the large polymerase protein (L) and the nucleoprotein (N) forming the viral nucleocapsid which contains the viral RNA genome, the matrix protein (M), and 2 envelop glycoproteins, the hemagglutinin protein (H) and the fusion protein (F) responsible for receptor binding and membrane fusion respectively.Citation35 Upon infection of susceptible cells, MV causes cell-cell fusion producing multinucleated giant cells, the typical cytopathic effect of MV infection.Citation36

Infection through the natural route is initiated in the respiratory tract via infection of CD150+ alveolar macrophages and/or DCs in the airway lumen or just below the epithelial cell layer by receptor-mediated recognition of MV.Citation36 Measles virus is captured by the DCs, which are widely distributed throughout the respiratory epithelium. Instead of being degraded, MV is protected and transported into the regional lymph nodes where it is efficiently transmitted to CD150+ lymphocytes which amplify the virus establishing primary viremia.Citation36,37 After amplification of the virus in lymph nodes, lymphocytes and monocytes carry the virus to various organs throughout the body including the skin, kidneys, gastrointestinal tract and liver, and throughout the respiratory tract.Citation23 MV-infected lymphocytes migrate into the lung tissue from the blood vessels where they may induce degradation of the basement membrane. They travel between adjacent epithelial cells and transmit the virus to epithelial cells via the receptor Nectin-4 expressed in adherens junctions.Citation22,23 This causes extensive epithelial damage and apical shedding of MV resulting in transmission to the next host.

Humans are the only known natural host for MV.Citation23 Non-human primates can also be infected and are commonly used as model of infection. To date there is no known animal reservoir and no asymptomatic carrier state has been documented.

Measles (Rubeola) is one of the most contagious viral diseases known. Measles virus is efficiently transmitted by aerosols entering the respiratory tract or by direct contact with respiratory secretions.Citation36,38 Its capacity of spreading is high, one person infected with MV can infect 15 to 20 others (R0 = 11–18),Citation39 meaning that the interruption of endemic transmission in population requires that more than 95% of the population is immune.Citation40 The infectious dose for wt MV is 0.2 units by intranasal spray.Citation41

Measles virus has a cytoplasmic replication cycle eliminating the possibility of integration into the host cell DNA and then a possible insertional mutagenesis.

Measles virus is an enveloped virus unstable in the environment surviving less than 2 hours on surfaces or objects.Citation42 Respiratory droplets can remain infective for at least one hour in confined spaces.Citation38 MV is susceptible to a variety of disinfectants (e.g., povidone iodine, 1% sodium hypochlorite, peracetic acid, hydrogen peroxide, 70% Ethanol).Citation41

The World Health Organization (WHO) has defined criteria for the classification of microorganisms into 4 Risks Groups, taking into account the severity of the disease that pathogens may cause in humans or animals, their ability to spread among the population, and the availability of prophylaxis or efficient treatment.Citation43 In Belgium, classification lists for human, animal, or plant pathogens provide a tool for identifying biological hazards associated with the contained useFootnote of wild-type pathogenic organisms.Citation44

MV is classified as a biological agent of class of risk 2 for humans: it can cause human disease and might be a hazard for directly exposed persons; it is however unlikely to spread to the community because an effective prophylactic vaccine against measles is available.Citation43-45

Attenuated measles virus generation

Attenuation of MV strains is the result of adaptation of the virus to growth conditions in non-permissive cell culture, especially avian cell lines.Citation5 MV isolation and passages in CD150 negative cells such as human kidney cells and Vero cells may have selected the viruses capable of using CD46 as a receptor for viral entry into host cells.Citation7,37,46 The use of CD46 by some MV strains may be considered as an in vitro adaptation rather than in vivo property of those strains.Citation7 Likewise, the vaccine strains must have adapted to chicken embryo fibroblasts by using an unknown receptor present on them.Citation7 CD46 is a complement regulatory protein that plays an important role in protecting autologous cells from complement attack. CD46 is ubiquitously expressed at low density by all normal human cell types except erythrocytes. CD46 level on tumor cells can be up to 7-10 fold higher compared with normal cells and protects tumor cells from complement mediated lysis.Citation47 High CD46 receptor density on tumor cells is a key determinant of oncolytic specificity of attenuated MV strains. Whereas virus entry increases progressively with CD46 density, there is a threshold number of CD46 receptors required for cell-to-cell fusion, which leads to death of all the cells incorporated into the syncytia. The differential expression of CD46 in tumor cells versus normal cells significantly increases the susceptibility of tumor cells to the oncolytic activity of MV attenuated strains. The in vivo tropism of attenuated MV and wt MV was compared using an animal model: in cynomoglus macaques experimentally infected via intratracheal or aerosol route, only the wt MV caused significant viremia and viral dissemination to the skin and the submucosa of respiratory epithelia.Citation48 MV strains that have acquired the ability to use CD46 as a receptor might gain a growth advantage in vitro in human and monkey cells because distribution of CD46 is ubiquitous, unlike that of CD150. However, in vivo, MV attenuated strains down-regulate CD46 in infected cells, which are then subject to complement-mediated cell lysis that may limits the spread of MV infection.Citation49

In vitro, attenuated MV has a much wider tropism than wt MV as it can use both CD46 and CD150 as cellular receptors to enter host cells. Nectin-4, the third MV receptor is expressed abundantly in placental trophoblasts, glandular cells of the stomach, and adenocarcinomas of the lung, breast and ovary. Moderate amounts are expressed in the epithelium of tonsils, oral mucosa, esophagus, and the epithelial cells of the nasopharynx and the trachea. Nectin-4 is expressed in the adherens junctions, on the basolateral side of epithelial cells in close contact to infected MV lymphocytes and dendritic cells and it is necessary for infection of human airway epithelia.Citation50 Smaller amounts are expressed in the lung macrophages and neuronal cells of the cerebral cortex. In cancer cells, Nectin-4 is highly up-regulated, and expressed on both apical and basolateral surfaces.Citation50 Reverse genetics technology using a helper-cell-based rescue systemCitation2,51 allowed the rescue of replicating measles viruses from cloned DNA able to stably express heterologous antigens. This technology enables the rescue of clinically approved and genetically relevant measles vaccine strains.Citation12,16,20,52 Edmonston B strain has been used for the construction of essentially all genetically modified oncolytic MV-Edm derivatives described until now and for all those which entered the clinical trial phase (). Oncolytic activities of MV have been demonstrated for at least 12 different cancer types.Citation60 Furthermore attenuated MV has been used worldwide to vaccinate children with an excellent safety profile and with no reversion to the wt MV making it attractive as oncolytic vector compared to other oncolytic viruses that are not used in vaccination.Citation60

Table 2. Clinical trials using oncolytic measles virus derived vectors. Legend: CEA: carcinoembryonic antigen, NIS: sodium iodide symporter, TCID50: medium tissue culture infective dose. The MV vectors have been produced as reported by Radecke et al.Citation51

Attenuated oncolytic MV vectors retain some characteristics enabling them to replicate in the human host. Compared to replication defective viral vectors, the likelihood of exposure of the environment around the patient is increased.Citation61 However, dissemination of the viral vector from the patient into the environment is not an adverse event per se. Its impact will mostly depend on the characteristics of the recombinant vector itself, such as its pathogenicity, its infectious dose, its transmission mode, the availability of effective prophylaxis or treatment, its susceptibility to disinfection.Citation33

The vectors used to develop attenuated recombinant MV for vaccination against infectious diseases and which entered the clinical trial phase have been engineered by reverse genetics from the cDNA of different measles vaccine strains as described by Radecke et al.Citation51 Additional transcription units (ATU) were introduced in the viral genome in order to construct the vector expressing the sequences corresponding to foreign antigens. summarizes the clinical and pre-clinical studies performed using recombinant MV vector as prophylaxis vaccine candidates against infectious diseases.

Table 3. Clinical and preclinical studies using recombinant attenuated measles virus as prophylaxis vaccine candidates against infectious diseases. Legend: intramuscular, i.m; intraperitoneal, i.p; subcutaneous, s.c; TCID50: medium tissue culture infective dose; pfu, plaque forming units.

History of safe use of attenuated measles virus

Although the wt MV can result in potentially serious infectious disease, the attenuated MV strains have a significant safety record, with millions of vaccine doses having been safely administrated in more than 40 y of use.Citation70 However, fatal infections have been documented in immunodeficient vaccinated childrenCitation71-74 and in a vaccinated adult with AIDS.Citation75

Administration of a high-titer (HT) Edmonston-Zagreb vaccine in areas with a high incidence of measles in children younger than 9 months shown an increased incidence of female mortality.Citation76,77 Investigators suggested that the HT measles vaccine had caused immune suppression similar to that of measles infection.Citation78-80 A review summarizing HT studies suggests that the HT vaccine itself is unlikely to be the cause of immunosuppression, indeed the effect was not found in all studies. Moreover, the HT studies with excess mortality rates showed increased female mortality rates that could be due to environmental or contextual conditions.Citation81

According to their high attenuation profiles and their history of safe use, MV attenuated strains currently used as measles vaccines could be classified as agents of class of risk 1 because these strains are non-pathogenic for human and not harmful for the environment and present a negligible risk during contained use.Citation43 MV-Edm B strain should also be classified in class of risk 1. MV-Edm strain is reactogenic and can cause fever and rash in measles-naïve children.Citation82 However, this strain is not able to cause a disease and is not transmissible to other persons.Citation70

Reconversion to wild-type

In addition and as already mentioned, the MV genome is very stable and reversion of measles vaccine strains to pathogenicity and subsequent transmission to other individuals have to date not be reported.Citation70

Hazards related to the recombinant MV vectors

In addition to its use as vaccine against measles, attenuated MV is used as a backbone for the development of prophylactic vaccine and as recombinant oncolytic vector. MV vector has been shown to stably express large, heterologous antigen-coding sequences up to 6 kb long.Citation2 Unlike primary strains, attenuated vaccine strains cause no immunosuppression.Citation18 In addition, MV infects cells of the immune system, including macrophages and DCs, thus providing an opportunity to deliver antigens directly to the most effective antigen presenting cells, a major advantage for a vaccine vector. Several studies provide good indications of the clinical safety and efficacy of these vectors. Preclinical studies have tested recombinant MV vaccine for prophylaxis against HIV-1,Citation20 SARS-CoV,Citation19 Chikungunya virus,Citation83 Dengue virus,Citation66 West Nile virusCitation64 and other infectious disease or cancer (). MV recombinant vector elicits strong and long-term HIV-specific neutralizing antibodies and cellular immune responses, even in the presence of preexisting immunity to MVCitation20,84-86 arboviruses such as West Nile virus,Citation18 Dengue virusCitation66 or SARS-CoVCitation19 in animal models. The strong capacity to raise T-cell responses that persist on long term in lymphoid organs is a hallmark of live attenuated vaccines. MV is particularly efficient at generating live long memory CD4 and CD8 T cells that help to maintain neutralizing antibodies and to prevent from reinfection.Citation87 The long-term protective immunity in humans against expressed foreign transgenes should still be evaluated.

A phase I clinical study with recombinant MV (MV1-F4) used to immunize healthy people against HIV-1 is ongoingCitation25 and the results of a phase I clinical trial with the vaccine candidate (MV-CHIK) used to immunize people against chikungunya virus are now available.Citation26

Hazard related to the transgene

When recombinant MV vector is used, risk assessment should also take into account the potential risk associated with the transgene.

The vaccine candidate MV1-F4 has been developed by reverse genetics from the cDNA of the Schwarz strain and has been genetically modified to express the F4 HIV antigen.Citation20 F4 is a single fusion protein comprising 4 HIV-1 clade antigens, p17 and p24 encoded by gag, reverse transcriptase encoded by pol and the regulatory protein Nef.Citation88 P24 is a structural protein present on the capsid. Its association with the viral protein p17 results in a matrix protein between the envelope and the capsid, ensuring the integrity of the virion. Nef acts both to increase the infectivity of viral particles and to reduce the expression of the CD4 receptor molecule on the cell surface.Citation89 And finally, reverse transcriptase is the enzyme used to generate cDNA from viral RNA. The F4 fusion protein alone has been administered as vaccine candidate by intramuscular route to healthy HIV seronegative adults and no toxic or allergic effects have been observed in a phase I clinical trial.Citation88,90

The vaccine candidate MV-CHIK has also been developed with the Schwarz strain as vector backbone and modified to express structural genes from chikungunya virus.Citation26 The structural proteins capsid (C), the envelope E1 and E2 glycoproteins and 2 small peptides, E3 and 6K allows the formation of self-assembling virus particles (VLP) that mimic the alphavirus external structure.Citation91 These structural proteins have been administered as vaccine candidate in healthy adults in a phase I trial and has been shown to be safe and well tolerated.Citation92 Furthermore, the results of a phase I trial showed that MV-CHIK is safe and had an overall acceptable tolerability profile when administrated in healthy humans.Citation26,92

The MV vector pTM-MV-Schw that was used to construct MV1-F4 and MV-CHIK contains an infectious cDNA corresponding to the antigenome of the Schwarz vaccine strain.Citation93 Additional transcription units (ATU) were introduced in the viral genome in order to construct the vector expressing the sequence corresponding to the F4 fusion protein for MV-F4 or to the structural protein from CHIK for MV-CHIK. The ATU was introduced into the plasmid backbone by site-directed mutagenesis between the MV P and M genes. Rescue of recombinant virus from the plasmid was performed using helper-cell-based rescue system cells.Citation51

MV Edm B strain has been used in virotherapy and modified to express reporter proteins. MV-CEA expresses the soluble N-terminal domain of human carcinoembryonic antigen (CEA). CEA is a biological inert tumor marker and has only minimal immunogenicity. In a phase I clinical trial, MV-CEA has been administrated by intraperitoneal route in 21 patients and no dose-limiting toxicity or treatment-induced immunosuppression was observed.Citation27 CEA expression allows non-invasive monitoring of viral gene expression but does not provide any information about the anatomic localization of virally infected cells and does not enhance the oncolytic effect of the viral vector. Therefore, MV has been modified to express another reporter protein, the sodium iodide symporter (NIS). The NIS protein is a membrane ion channel that is normally expressed in the thyroid, mammary glands, stomach and salivary tissue. Expression of NIS allows cells to actively transport ions into the cell. It may be used as therapeutic transgene capable of further increasing the oncolytic potency of MV-NIS by facilitating the intracellular entry of radioisotopes, which can cause direct radiation damage to tumor cells, thereby enhancing the therapeutic efficacy (radiovirotherapy). In a phase I clinical trial, MV-NIS has been administered by intraperitoneal route in 16 patients and no dose-limiting toxicity was observed.Citation28 MV-CEA and MV-NIS have been achieved via the introduction of the transgene as additional transcription units upstream of the viral N gene for MV-CEA and downstream of the viral H gene between H and L gene for MV-NIS.Citation24

In preclinical studies, MV-NIS was administered intravenously in measles-naïve squirrel monkeys and measles-susceptible transgenic mice and no toxicity was observed.Citation94 There were no premature or unscheduled deaths during the study. No adverse clinical signs or meaningful effects on body weight or temperature were seen. No MV-NIS treatment-related effects were observed for any hematology, clinical chemistry, thyroid hormone, or cytokine parameter, but reticulocyte counts were transiently suppressed in the cyclophosphamide-treated animals. No treatment-related lesions were seen in any of the animals at the time of necropsy, and no histopathological changes were observed in any of the organs tested.Citation94 A phase I clinical trial was therefore initiated to determine the maximum tolerated dose of intravenously administrated MV-NIS in patients with advanced refractory multiple myeloma.Citation56 Preliminary data on 2 patients from this study has been reported.Citation29 These patients who were seronegative for prior measles exposure received the highest feasible dose level (1011 TCID50, 50% tissue culture infectious dose). They had secondary effects, they became febrile, tachycardic and hypotensive with severe nausea and vomiting. Toxicity resolved within the first week after therapy and was probably due to the high dose of recombinant virus administrated combined with the absence of pre-existing immunity.

These inserts (F4, VLP, CEA and NIS) are currently being used in phase I/II clinical trials. Other inserts used in combination with oncolytic MV are tested in pre-clinical studies. For a review see Msaouel et al.Citation24 Their safety profile has still to be characterized, taking into account that some of the expressed gene product may have intrinsic hazardous properties such as toxic or allergenic properties. Gene products that may be considered as potentially hazardous in the particular context of gene therapy using viral vectors have been reviewed by Bergmans et al.Citation95 and Van den Akker et al.Citation61 have also discussed inserts with unknown or novel characteristics produced by synthetic biology that could be considered as potentially hazardous. Finally, the hazard related to the transgene also depends on the conditions of use (dose, administration route, expression level of the transgene).

Some foreign genes could potentially lead to retargeting oncolytic MV vectors to other cells to increase specificity against tumor cells. MV vector can be retargeted to specific tumor cells by linking a single-chain antibody (single chain fragment variable, scFv) or naturally occurring ligand to the virus attachment H glycoprotein displayed on the virus surface. The ablation of receptor CD46 and CD150 binding sites of some vectors allows entry only into cells expressing the receptor for the scFv or ligand linked to H. A variety of scFv's have been displayed on H against different receptor (). Broadening viral tropism to molecules expressed on the luminal endothelial surface of tumor neovascularization may increase targeted viral delivery to tumor sites following systemic administration.Citation97,98,103 Retargeted attenuated MV Edm vector lack neurotoxic activity even when administered directly to the CNS of measles-naïve IFN type I receptor deficient (IFNARKO) CD46 Ge transgenic mice, a very sensitive model of viral neurotoxicity.Citation99-101 However, toxicity of the retargeted vector should be evaluated in non-human primates before initiation of a clinical study because transgenic mice do not express human receptors.Citation104

Table 4. Preclinical studies using retargeted attenuated measles virus.

Finally, the recombinant MV shows an excellent stability of transgene expression. Various genes or combinations of genes were expressed stably in MV vectors for more than 12 passages.Citation18,93

Possibility of recombination with other viruses and genetic stability of MV strains

There has been no conclusive evidence to date of any genetic recombination events between MV vaccine and wt strains in people co-infected with both viruses.Citation24 Recombination, which is an efficient means for rapid genetic change for many viruses, does not occur in paramyxoviruses. No recombinant viruses have been isolated from natural infections. Unlike other RNA viruses such as influenza and HIV, MV vaccine strains demonstrate high genetic stability even after prolonged replication in human host.Citation105 Consequently, the probability of generating mutant strains is very low.Citation18,93

Influence of pre-existing immunity

Like all viral vectors, the MV vector efficacy in inducing a protecting immune answer could be affected by the pre-existing immunity among the human population.Citation106 However, numerous studies have shown that revaccinating already immunized individuals results in a boost of anti-MV antibodies, indicating that the live vaccine replicates despite pre-existing immunity.Citation107,108 Likewise, the presence of maternal anti-MV antibodies has been shown to limit the induction of humoral but not CMI responses in immunized infants.Citation109 Moreover, vaccination of pre-immunized mice and primates with recombinant MV expressing HIV antigens has shown induction of anti-HIV env antibodies in the presence of anti-MV antibodies.Citation20 In another study, mice immunized with a recombinant MV-SARS vector developed SARS-specific immune responses in the presence of anti-MV immunity.Citation110

The route of administration of the vector could also influence the immune response. The vaccine strain MVb and the MV vector developed from this vaccine strain could be administrated by aerosol and it could therefore circumvent systemic MV pre-immunity more efficiently than parenteral administration.Citation106 Studies evaluating the safety of aerosolized measles vaccine did not identify severe side effects. However, aerosolized measles vaccine could trigger or exacerbate asthma in young childrenCitation111 as it was observed for intranasal administration of attenuated influenza vaccine.Citation112 Anti-measles immunity enhances the safety of oncolytic MV and has been a prerequisite in the majority of the MV virotherapy trials.Citation24 However, neutralizing antibodies may limit infection of tumor cells by MV. Indeed, cell-associated viruses are protected from antibody neutralization and have been tested to overcome antiviral pre-existing immunity.Citation113 Mesenchymal stem cells (MSC) transduced with MV-NIS were injected into ovarian tumor xenografts. The MV-NIS transduced MSC significantly extended the survival of measles immune tumor bearing mice in contrast to MV-NIS injected alone.Citation113 Mesenchymal stem cells transduced with MV-NIS are currently being tested in a phase I/II clinical trial.Citation54

Risk classification of the recombinant vector

The risk classification of the recombinant MV vectors depends on the nature of the vector but has also to take into account any potential risk associated with the inserted gene(s). We recommend classifying MV1-F4 and MV-CHIK in class of risk 1. Indeed, the MV strain backbone derived from the attenuated measles vaccine strains, and the F4 protein and VLP have no known toxic or allergic effects when administrated to humans.Citation92,93

The recombinant vectors MV-CEA and MV-NIS should also be classified in class of risk 1, based on the class of risk of the Edm B strain used as vector backbone. There is no reasons to think that the presence of CEA and NIS could change the safety profile of the recombinant virus.

Hazard related to exposure pathways

An important step in the risk assessment of viral vectors is the evaluation of exposure pathways whereby personnel, non-patients and/or the environment may be exposed to recombinant MV administrated to patients.

Biodistribution and shedding

Biodistribution is defined as the dispersion of the vector within the patient's body from the site of administration. Indeed, the presence of viral vectors in organs might be indicative of potential shedding which corresponds to the dissemination of viral vector in any form into the environment via excreta (urine, faeces, sweat, saliva, nasopharyngal fluids), blood and semen from the treated patient.Citation114 Biodistribution and shedding of MV vector depends on the dose, the route of administration and the MV strain used.

Wild-type MV strains use primarily the CD150 receptor expressed on lymphocytes, thymocytes, macrophages and mature DCs cells and Nectin-4 receptor expressed on epithelial cells for viral entry to host cells. Infection with wt MV has been thought to begin by infection of DCs in the respiratory tract followed by lymphoid organs.Citation115 This phase of localized replication is followed by primary and secondary viremia, with viral spread to multiple epithelial tissues including the skin, kidneys, gastrointestinal tract, liver and the respiratory tract. Shedding of the virus occurs from the nasopharynx.Citation36 In natural infection, MV can also be isolated from urine up to 10 d after rash onset.Citation116 MV RNA persists at multiple sites for many months after resolution of the rash and apparent recovery in a proportion of children. Experimental infection of rhesus macaques with MV showed that persistence of viral RNA in blood, respiratory tract, or lymph nodes is characteristic of primary MV infection.Citation117

In addition to the receptors used by wt MV, attenuated MV strains can use the CD46 receptor for viral entry into host cells and were therefore expected to have a wider tropism than the wt MV strains. The biodistribution of attenuated MV (Edm B strain) was compared to that of wt MV in cynomoglus macaques experimentally infected via intratracheal or aerosol route.Citation48 Both viruses predominantly infected alveolar macrophages and DCs in the lungs. However, strikingly, only the wt MV caused viremia and was disseminated to lymphoid tissues, the respiratory submucosa, and the skin. Some mechanism must operate in vivo to suppress the growth of the virus capable of using CD46 as receptor.Citation7 It is also important to take into account that the standard route of administration of live-attenuated MV vaccine, the subcutaneous route has not been evaluated in this study. Intramuscular vaccination of macaques with a MV Edmonston-Zagreb strain expressing GFP demonstrated that muscle cells were not infected and dendritic cells and macrophages were the predominantly the target cells.Citation118

Published data on shedding of MV vaccine strains are limited. After Measles vaccination in humans, MV RNA could be detected in urine as late as 14 d post immunizationCitation119 and was rarely detected in throat or nasopharyngal secretions. Morfin et al.Citation120 reported a case of Measles Schwarz virus isolation in throat swabs of a child, 12 d after vaccination and in a study realized until 2002 and 2006, 9 children presented a symptomatic post vaccinal excretion of MV in nasopharynxCitation121 showing that subcutaneous injection of the attenuated MV Schwarz strain can result in respiratory excretion of this virus. However, person to person transmission of measles vaccine has never been reported.Citation17

The biodistribution of the HIV-1 candidate vaccine MV1-F4 was similar to that of the parental MV Schwarz strain in monkeys immunized by intramuscular route, with both vaccines replicating preferentially in secondary lymphoid organs and epithelium-rich tissues.Citation20

To assess the potential shedding of the parental Schwarz vaccine strain and MV1-F4 vaccine candidate, excretions and body fluids of monkeys immunized with these vaccine strains were tested for the presence of MV viral sequences by RT-qPCR assay. Of the 8 time points at which shedding analysis was performed, MV viral RNA was only detected at day 11, the expected peak of viremia, in some biological fluids samples from few immunized monkeys. However, none of these samples contained infectious virus, indicating that no shedding of infectious virus particles was observed for either of the 2 vaccines.Citation20 The introduction of HIV F4 fusion protein did not alter the tropism of the parental strain or its shedding capacity.

No data are available regarding the biodistribution and shedding of MV-CHIK. However, no alteration in tropism is expected for the same vector containing the genes coding for structural proteins from chikungunya virus.

With regards to the 2 recombinant oncolytic MV vectors described before, the following data are available:

MV-CEA and MV-NIS: In order to maximize viral infection of the tumor cells, MV-CEA and MV-NIS were administrated by intraperitoneal route to patients with recurrent ovarian cancer.Citation21,27,28. In pre-clinical studies performed in measles naïve IFNARKO CD46 Ge mice, there was no evidence of viral replication outside the peritoneal cavity (i.e., the brain, heart and skeletal muscle).Citation122 In phase I clinical studies, 21 women were administered and low levels of MV-CEA genomes were found by RT-qPCR in peripheral blood mononuclear cells (PBMCs) of 4 asymptomatic patients.Citation27 Sixteen women received MV-NIS and no detection of viral genome in peripheral blood was observed.Citation28

Shedding of oncolytic MV has also been evaluated in pre-clinical and clinical studies. In phase I clinical studies, no viral RNA in urine or saliva was observed after intraperitoneal administration of MV-CEA or MV-NIS.Citation27,28

In one of the phase I clinical trialCitation56 listed in Table 2, MV-NIS is administrated by intravenous route with or without pretreatment with cyclophosphamide. In addition to its chemotherapeutic activity, cyclophosphamide is an immunosuppressive agent. Therefore, it can potentially prolong and enhance viral dissemination and replication in the tumor.Citation123 To evaluate the biodistribution of intravenous MV-NIS administration, IFNARKO CD46 Ge mice pre-treated with cyclophosphamide demonstrated alterations in the biodistribution of MV-NIS-infected cells and the kinetics of MV-NIS elimination.Citation94 Virus was sporadically detected by RT-qPCR in the brains of cyclophosphamide-pretreated mice whereas those not treated with cyclophosphamide remained negative.

In two patients with advanced refractory multiple myeloma treated with intravenous infusion of MV-NIS, radioiodine SPECT-CT scan provided clear evidence of tumor-targeted MV-NIS infection and propagation of the MV-NIS infection in tumor cells. There was no evidence for spread of MV-NIS from the tumor to adjacent normal tissues.Citation29

Shedding of MV-NIS administered by intravenous route alone or with pretreatment with cyclophosphamide has been evaluated by RT-qPCR.Citation123 Viral RNA was detected in buccal swabs of monkeys after intravenous administration of MV-NIS.Citation94 A higher level of virus RNA was detected in the buccal swabs in the cyclophosphamide treated monkey and viral RNA remained detectable for a longer period of time indicating that this drug enhance the propagation of the vector in animals.Citation94

Measles virus transcripts were still detectable in the circulating cells of one patient at 6 weeks after intravenous infusion of MV-NIS.Citation29

During the course of a clinical trial, the possible consequences of leakage of the vector outside the patient's body may include, for example, adverse effects associated with the infection of personnel or people in general coming into contact with the vaccinated individuals. However, it is important to take into account that most individuals in industrialized countries are immune to the WT measles virus as a result of natural infection or vaccination. Moreover, as mentioned before, measles virus is very rapidly degraded in the environment. The consequences of dissemination will also depend on the characteristics of the recombinant vector itself, in particular the safety profile of the transgene which have been shown to be safe.

Direct exposure

Some manipulations might lead to exposure to the recombinant MV vector, e.g. the preparation of the viral vector, its administration to the patient and waste disposal. Direct exposure may also result from accidental inoculation of personnel during the treatment of the patient via droplets or aerosols contacting mucous membrane, non-intact skin or eyes. The risk of piercing by needle or injury due to sharps, cutting with broken vials is the greatest for the personnel handling vials and syringes. Medical and paramedical staff and the people visiting the treated patient could also be exposed to contaminated material, waste material or spoiled surfaces. These exposure pathways can be reduced or eliminated by application of appropriate risk management strategies.

Considerations for risk management (containment, workers protection measures, waste treatment)

When attenuated MV recombinant vectors are used in the clinical setting (under contained conditions), appropriate containment and other measures to protect human health and the environment should be implemented as a result of a risk assessment taking into account the characteristics of the biological agent manipulated, the nature of the transgene and the nature of the activity. Four containment levels (CL-1 to CL-4, CL-4 being the most stringent) are defined in the EU legislation and consist in a combination of technical characteristics of the facility, safety equipment, laboratory practices and operational procedures such as waste management procedures.Citation30

The attenuation in MV1-F4 and MV-CHIK vaccine candidates and the history of safe use of MV Schwarz strain allows handling this recombinant virus under CL-1 in the clinical setting. As mentioned before the addition of the transgenes does not change its safety profile.

A CL-1 should also be recommended for handling MV-CEA and MV-NIS oncolytic vectors.

Transduction of MSC cells by recombinant MV-NIS should be carried out in CL-2. When manipulating mesenchymal stem cells which are primary cell cultures obtained directly from organs or tissues,Citation113 the presence of adventitious contaminating agents constitutes the main hazard. As they are characterized by a finite life span, the time available for characterization and detection of contaminating agents remains limited. Therefore, primary cell cultures derived from human must be transduced in a CL-2 and manipulated in a class II biosafety cabinet guaranteeing the product sterility and personnel should wear gloves.

To prevent or manage risks associated with dissemination of recombinant MV vectors into the environment during viral vector administration, application of specific work practices should be applied and personnel should wear personal protective equipment to prevent or manage risks:

Operations producing aerosols such as the puncture of a vial should be limited or strictly contained during preparation and administration of the recombinant MV vector. Consequently, the preparation of the GM vector should preferably be performed in a class II biosafety cabinet. Personnel administrating the vector should wear adequate protective clothing such as lab coats, gloves, goggles and masks. Work with needles and other sharp objects should be strictly limited and workers should never recap nor remove needles from syringes.

Spills should be inactivated by an appropriate disinfectant (e.g., 1% sodium hypochlorite extemporaneously prepared), allowing sufficient contact time before disposal.

Contaminated waste and personal protective equipment should be inactivated using an appropriate method (autoclave or incineration) before disposal and potentially contaminated non-disposable materials (e.g., the material used for the transport, preparation or administration of the GM vector, instruments, surfaces) need to be properly decontaminated.

If an incident occurs that could lead to infection (e.g., breakage of a vial containing the vector, piercing with a needle), applicable first aid should be performed (i.e., flushing eyes for ocular exposure, placing an absorbent tissue on the affected area in order to absorb viral particles and apply disinfectant directly to the tissue and after removing this tissue washing the skin thoroughly), followed by reporting to the supervisor.

Personnel manipulating the recombinant vector and persons in close contact with the treated patient should be vaccinated against measles.

After injection of a recombinant MV, the treated patient should avoid contacts with immunosuppressed individuals or individuals lacking protective immunity for a minimum of 8 d or until saliva, urine and blood PBMC test negative for the virus (whichever is longer). As the number of patients treated with different MV strains for specific clinical indications increases, and clinical information regarding the likelihood and duration of shedding accumulates, biosafety guidelines are expected to evolve.

As MV is a human virus with no known animal reservoir, there is no risk for animals and plants.

Conclusion

Attenuated MV is one of the most effective and safe human vaccine in use providing long-lasting protection. Attenuated MV is now widely used as recombinant vector for vaccination against various infectious diseases or cancers and for virotherapy. Measles virus has a cytoplasmic replication cycle eliminating the possibility of integration into the host cells DNA avoiding a possible insertional mutagenesis. Furthermore, the MV genome is very stable and MV vectors have been shown to stably express large sizes of foreign inserted genetic sequences. We have presented and discussed in this paper several biosafety issues which need to be considered carefully when performing clinical trials with recombinant attenuated MV vectors. After having performed a comprehensive risk assessment of the MV vectors currently used in clinical trials as prophylactic vaccine or oncolytic vector we can conclude that these MV vectors belong to class of risk 1 for humans. In order to maximize risk mitigation and avoid dissemination into the environment, these MV vectors should be manipulated under containment level 1.

Note

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgements

The authors thank Dr. Amaya Leunda, Dr. Katia Pauwels, Dr. Didier Breyer, Dr. Martine Goossens and Dr. Alexandre Dobly [Scientific Institute of Public Health (ISP-WIV), Brussels, Belgium] for their helpful contribution to this document.

Funding

This work received support from the Brussels-Capital Region (IBGE-BIM), the Flemish Region (LNE), and Wallonia (DGARNE).

Notes

1. Any activity in which micro-organisms are genetically modified or in which GMM pathogenic or not are used and for which specific containment measures are used to limit their contact with, and to provide a high level of safety for, the general population and the environment.

References

  • Zhang X, Rennick LJ, Duprex WP, Rima BK. Determination of spontaneous mutation frequencies in measles virus under nonselective conditions. J Virol 2013; 87:2686-92; PMID:23255805; http://dx.doi.org/10.1128/JVI.02146-12
  • Billeter MA, Naim HY, Udem SA. Reverse genetics of measles virus and resulting multivalent recombinant vaccines: applications of recombinant measles viruses. Curr Top Microbiol Immunol 2009; 329:129-62; PMID:19198565
  • Enders JF, Peebles TC. Propagation in tissue cultures of cytopathogenic agents from patients with measles. Proc Soc Exp Biol Med 1954; 86:277-86; PMID:13177653; http://dx.doi.org/10.3181/00379727-86-21073
  • Enders JF, Katz SL, Milovanovic MV, Holloway A. Studies on an attenuated measles-virus vaccine. I. Development and preparations of the vaccine: technics for assay of effects of vaccination. N Engl J Med 1960; 263:153-9; PMID:13820246; http://dx.doi.org/10.1056/NEJM196007282630401
  • Bankamp B, Takeda M, Zhang Y, Xu W, Rota PA. Genetic characterization of measles vaccine strains. J Infect Dis 2011; 204 (Suppl 1):S533-48; PMID:21666210; http://dx.doi.org/10.1093/infdis/jir097
  • Erlenhöfer C, Duprex WP, Rima BK, ter Meulen V, Schneider-Schaulies J. Analysis of receptor (CD46, CD150) usage by measles virus. J Gen Virol 2002; 83:1431-6; PMID:12029158; http://dx.doi.org/10.1099/0022-1317-83-6-1431
  • Yanagi Y, Ono N, Tatsuo H, Hashimoto K, Minagawa H. Measles virus receptor SLAM (CD150). Virology 2002; 299:155-61; PMID:12202217; http://dx.doi.org/10.1006/viro.2002.1471
  • Zuniga A, Wang Z, Liniger M, Hangartner L, Caballero M, Pavlovic J, Wild P, Viret JF, Glueck R, Billeter MA, et al. Attenuated measles virus as a vaccine vector. Vaccine 2007; 25:2974-83; PMID:17303293; http://dx.doi.org/10.1016/j.vaccine.2007.01.064
  • Ikic D, Juzbasic M, Beck M, Hrabar A, Cimbur-Schreiber T. Attenuation and characterisation of Edmonston-Zagreb measles virus. Ann Immunol Hung 1972; 16:175-81; PMID:4679646
  • Ikic D. Edmonston-Zagreb strain of measles vaccine: epidemiologic evaluation in Yugoslavia. Rev Infect Dis 1983; 5:558-63; PMID:6879014; http://dx.doi.org/10.1093/clinids/5.3.558
  • Beck M, Smerdel S, Dedic I, Delimar N, Rajninger-Miholic M, Juzbasic M, Manhalter T, Vlatkovic R, Borcic B, Mihajic Z. Immune response to Edmonston-Zagreb measles virus strain in monovalent and combined MMR vaccine. Dev Biol Stand 1986; 65:95-100; PMID:3556780
  • Bolton DL, Santra S, Sweet C, Custers J, Song K, Balachandran H, Kozlowski PA, Letvin N, Roeder M, Radosevic K. Priming T-cell responses with recombinant measles vaccine vector in a heterologous prime-boost setting in non-human primates. Vaccine 2012; 30:5991-98; PMID:22732429; http://dx.doi.org/10.1016/j.vaccine.2012.06.029
  • Mori T, Sasaki K, Hashimoto H, Makino S. Molecular cloning and complete nucleotide sequence of genomic RNA of the AIC-K strain of attenuated measles virus. Virus Genes 1993; 7:67-81; PMID:8470368; http://dx.doi.org/10.1007/BF01702349
  • Mirchamsy H, Shafyi A, Bahrami S, Kamali M, Nazari P. Use of human diploid cell MRC-5 for production of measles and rubella virus vaccines. Dev Biol Stand 1976; 37:297-300; PMID:1031692
  • Makino S. Development and characteristics of live AIC-K measles virus vaccine: a brief report. Rev Infect Dis 1983; 5:504-5; PMID:6410489; http://dx.doi.org/10.1093/clinids/5.3.504
  • Zuniga A, Liniger M, Morin TN, Marty RR, Wiegand M, Ilter O, Weibel S, Billeter MA, Knuchel MC, Naim HY. Sequence and immunogenicity of a clinically approved novel measles virus vaccine vector. Hum Vaccin Immunother 2013; 9:607-13; PMID:23324616; http://dx.doi.org/10.4161/hv.23242
  • WHO position on measles vaccines. Vaccine 2009; 27:7219-21; PMID:19833246; http://dx.doi.org/10.1016/j.vaccine.2009.09.116
  • Brandler S, Tangy F. Recombinant vector derived from live attenuated measles virus: potential for flavivirus vaccines. Comp Immunol Microbiol Infect Dis 2008; 31:271-91; PMID:17869338; http://dx.doi.org/10.1016/j.cimid.2007.07.012
  • Escriou N, Callendret B, Lorin V, Combredet C, Marianneau P, Février M, Tangy F. Protection from SARS coronavirus conferred by live measles vaccine expressing the spike glycoprotein. Virology 2014; 452-453:32-41
  • Lorin C, Segal L, Mols J, Morelle J, Bourguignon P, Rovira O, Mettens P, Silvano J, Dumey N, Le Goff F, et al. Toxicology, biodistribution and shedding profile of a recombinant measles vaccine vector expressing HIV-1 antigens, in cynomolgus macaques. Naunyn Schmiedebergs Arch Pharmacol 2012; 385:1211-25; PMID:22983013; http://dx.doi.org/10.1007/s00210-012-0793-4
  • Msaouel P, Dispenzieri A, Galanis E. Clinical testing of engineered oncolytic measles virus strains in the treatment of cancer: an overview. Curr Opin Mol Ther 2009; 11:43-53; PMID:19169959
  • Mühlebach MD, Mateo M, Sinn PL, Prüfer S, Uhlig KM, Leonard VH, Navaratnarajah CK, Frenzke M, Wong XX, Sawatsky B, et al. Adherens junction protein nectin-4 is the epithelial receptor for measles virus. Nature 2011; 480:530-3; PMID:22048310
  • Noyce RS, Richardson CD. Nectin 4 is the epithelial cell receptor for measles virus. Trends Microbiol 2012; 20:429-39; PMID:22721863; http://dx.doi.org/10.1016/j.tim.2012.05.006
  • Msaouel P, Opyrchal M, Domingo ME, Galanis E. Oncolytic measles virus strains as novel anticancer agents. Expert Opin Biol Ther 2013; 13:483-502; PMID:23289598; http://dx.doi.org/10.1517/14712598.2013.749851
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. Study to Evaluate the Dosage and Safety of Two Intramuscular Injections of an Investigational Clade B HIV Vaccine. Official title: An open-label, Phase I, Dose-escalation and safety study of two intramuscular injections of a Dose of 2.9 Log or 4 Log CCID50 of the Recombinant HIV I clade B Measles Vaccine Vector in Healthy Adults. ClinicalTrials.gov Identifier, NCT01320176; 2011 Mar 21 [updated 2012 Feb 13; cited 2016 Jan 26]. Availaible from: http://clinicaltrials.gov/ct2/show/NCT01320176
  • Ramsauer K, Schwameis M, Firbas C, Müllner M, Putnak RJ, Thomas SJ, Desprès P, Tauber E, Jilma B, Tangy F. Immunogenicity, safety and tolerability of a recombinant measles virus-based chikungunya vaccine: an observer and subject blinded, block randomised, active and placebo-controlled first in man trial. Lancet Infect Dis 2015; 15:519-527; PMID:25739878; http://dx.doi.org/10.1016/S1473-3099(15)70043-5
  • Galanis E, Hartmann LC, Cliby WA, Long HJ, Peethambaram PP, Barrette BA, Kaur JS, Haluska PJ Jr, Aderca I, Zollman PJ, et al. Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer Res 2010; 70:875-82; PMID:20103634; http://dx.doi.org/10.1158/0008-5472.CAN-09-2762
  • Galanis E, Atherton PJ, Maurer MJ, Knutson KL, Dowdy SC, Cliby WA, Haluska PJ Jr, Long HJ, Oberg A, Aderca I, et al. Oncolytic measles virus expressing the sodium iodide symporter to treat drug-resistant ovarian cancer. Cancer Res 2015; 75:22-30; PMID:25398436; http://dx.doi.org/10.1158/0008-5472.CAN-14-2533
  • Russell SJ, Federspiel MJ, Peng KW, Tong C, Dingli D, Morice WG, Lowe V, O'Connor MK, Kyle RA, Leung N, et al. Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clin Proc 2014; 89:926-33; PMID:24835528; http://dx.doi.org/10.1016/j.mayocp.2014.04.003
  • Directive 2009/41/EC of the European Parliament and of the Council of 6 May 2009 on the contained use of genetically modified micro-organisms (Recast). Off J 21.05.2009; L 125:0075
  • Directive 2001/18/EC of the European Parliament and of the Council of 12 March 2001 on the deliberate release into the environment of genetically modified organisms and repealing Council Directive 90/220/EEC. Off J 17.04.2001; L106:1
  • Regulation (EC) No. 726/2004 of the European Parliament and of the Council of 31 March 2004 laying down Community procedures for the authorisation and supervision of medicinal products for human and veterinary use and establishing a European Medicines Agency. Off J 30.04.2004; L 136:1
  • Baldo A, van den Akker E, Bergmans HE, Lim F, Pauwels K. General Considerations on the Biosafety of Virus-derived Vectors Used in Gene Therapy and Vaccination. Curr Gene Ther 2013; 13:385-94; PMID:24195604; http://dx.doi.org/10.2174/15665232113136660005
  • Griffin D. Measles Virus. In: Knipe D, Howley P, eds. Field's virology. 4th ed. Philadelphia, Lippincott-Raven publishers, 2001: pp. 1401-41.
  • Yanagi Y, Takeda M, Ohno S. Measles virus: cellular receptors, tropism and pathogenesis. J Gen Virol 2006; 87:2767-79; PMID:16963735; http://dx.doi.org/10.1099/vir.0.82221-0
  • de Vries RD, Mesman AW, Geijtenbeek TB, Duprex WP, de Swart RL. The pathogenesis of measles. Curr Opin Virol 2012; 2:248-55; PMID:22483507; http://dx.doi.org/10.1016/j.coviro.2012.03.005
  • de Witte L, de Vries RD, van der Vlist M, Yüksel S, Litjens M, de Swart RL, Geitenbeek TB. DC-SIGN and CD150 have distinct roles in transmission of measles virus from dendritic cells to T-lymphocytes. PLoS Pathog 2008; 4(4):e1000049; PMID:18421379; http://dx.doi.org/10.1371/journal.ppat.1000049
  • Bloch AB, Orenstein WA, Ewing WM, Spain WH, Mallison GF, Herrmann KL, Hinman AR. Measles outbreak in a pediatric practice: airborne transmission in an office setting. Pediatrics 1985; 75:676-83; PMID:3982900
  • Anderson RM, May RM. infectious diseases of humans. Dynamics and control. 11th ed. 2006.
  • Griffin DE, Pan CH, Moss MJ. Measles vaccine. Front Biosci 2008; 13:1352-70; PMID:17981635; http://dx.doi.org/10.2741/2767
  • Public Health Agency of Canada [Internet]. Laboratory Biosafety and Biosecurity. Pathogen Safety Data Sheets and Risk Assessment - Measles virus; [updated 2011 Sep 08; cited 2016 Jan 26]. Available from: http://www.phac-aspc.gc.ca/lab-bio/res/psds-ftss/measles-rougeole-eng.php
  • Atkinson W, Wolfe S, Hamborsky J, McIntyre L. Epidemiology and prevention of vaccine-preventable diseases. 11th ed. Washington (DC): Public Health Foundation; 2009.
  • World Health Organization. Laboratory Biosafety Manual. 3rd ed. Geneva; 2004, 178p. [online] Available from: http://www.who.int/csr/resources/publications/biosafety/Biosafety7.pdf?ua=1 Access date 26-01-2016.
  • Belgium Biosafety Server [Internet]. Belgian classifications for micro-organisms based on their biological risks - Definitions; 2006 Feb 28 [cited 2016 Jan 26]. Available from: http://www.biosafety.be/RA/Class/ClassBELdef.html
  • Directive 2000/54/EC of the European Parliament and of the Council of 18 September 2000 on the protection of workers from risks related to exposure to biological agents at work. Official Journal of the European Communities. 17.10.2000. L262/21.
  • Naniche D, Varior-Krishnan G, Cervoni F, Wild TF, Rossi B, Rabourdin-Combe C, Gerlier D. Human membrane cofactor protein (CD46) acts as a cellular receptor for measles virus. J Virol 1993; 67:6025-32; PMID:8371352
  • Anderson BD, Nakamura T, Russell SJ, Peng KW. High CD46 receptor density determines preferential killing of tumor cells by oncolytic measles virus. Cancer Res 2004; 64:4919-26; PMID:15256464; http://dx.doi.org/10.1158/0008-5472.CAN-04-0884
  • de Vries RD, Lemon K, Ludlow M, McQuaid S, Yüksel S, van Amerongen G, Rennick LJ, Rima BK, Osterhaus AD, de Swart RL, et al. In vivo tropism of attenuated and pathogenic measles virus expressing green fluorescent protein in macaques. J Virol 2010; 84:4714-24; PMID:20181691; http://dx.doi.org/10.1128/JVI.02633-09
  • Schnorr J, Dunster L, Nanan J, Schneider-Schaulies J, Schneider Schaulies S, ter Meulen V. Measles virus-induced down-regulation of CD46 is associated with enhanced sensitivity to complement-mediated lysis of infected cells. Eur J Immunol 1995; 25:976-984; PMID:7737301; http://dx.doi.org/10.1002/eji.1830250418
  • Noyce RS, Bondre DG, Ha MN, Lin L-T, Sisson G, Tsao M-S, Richardson CD. Tumor cell marker PVRL4 (Nectin 4) is an epithelial cell receptor for measles virus. PloS Pathog 2011; 7:e1002240; PMID:21901103; http://dx.doi.org/10.1371/journal.ppat.1002240
  • Radecke F, Spielhofer P, Schneider H, Kaelin K, Huber M, Dötsch C, Christiansen G, Billeter MA. Rescue of measles viruses from cloned DNA. EMBO J 1995; 14:5773-84; PMID:8846771
  • Zuniga A, Wang Z, Liniger M, Hangartner L, Caballero M, Pavlovic J, Wild P, Viret JF, Glueck R, Billeter MA, Naim HY. Attenuated measles virus as a vaccine vector. Vaccine 2007; 25:2974-83; PMID:17303293; http://dx.doi.org/10.1016/j.vaccine.2007.01.064
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. Recombinant Measles Virus Vaccine Therapy and Oncolytic virus therapy in Treating Patients With Progressive, Recurrent, or Refractory Ovarian Epithelial Cancer or Primary Peritoneal Cancer. Official title: Phase I Trial of Intraperitoneal Administration of a) a CEA-Expressing Derivative, and b) a NIS-Expressing Derivative Manufactured From a Genetically Engineered Strain of Measles Virus in Patients With Recurrent Ovarian Cancer. ClinicalTrials.gov Identifier, NCT00408590; 2006 Dec 6 [updated 2015 Mar 23; cited 2016 Jan 26]. Available from: http://clinicaltrials.gov/ct2/show/NCT00408590
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. MV-NIS Infected Mesenchymal Stem Cells in Treating Patients With Recurrent Ovarian Cancer. Official title: Phase I/II trial of intraperitoneal administration of adipose tissue derived mesenchymal stem cells infected with a NIS-expressing derivative manufactured from a genetically engineered strain of Measles virus in patients with recurrent ovarian cancer. ClinicalTrials.gov Identifier, NCT02068794; 2014 Feb 19 [updated 2015 Nov 16; cited 2016 Janv 26]. Available from: http://clinicaltrials.gov/ct2/show/NCT02068794
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. Viral Therapy in Treating Patients With Recurrent Glioblastoma Multiforme. Official title: Phase I trial of Measles virus derivative producing CEA (MV-CEA) in patients with recurrent Glioblastoma Multiforme (GBM). ClinicalTrials.gov Identifier, NCT00390299; 2006 Oct 18 [updated 2015 oct 19; cited 2016 Jan 26]. Available from: http://clinicaltrials.gov/ct2/show/NCT00390299
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. Vaccine Therapy With or Without Cyclophosphamide in Treating Patients With Recurrent or Refractory Multiple Myeloma. Official title: Phase I Trial of Systemic Administration of Edmonston Strain of Measles Virus, Genetically Engineered to Express NIS, With or Without Cyclophosphamide, in Patients With Recurrent or Refractory Multiple Myeloma. ClinicalTrials.gov Identifier, NCT00450814; 2007 Mar 20 [updated 2015 Jul 25; cited 2016 Jan 26]. Available from: http://clinicaltrials.gov/ct2/show/NCT00450814
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. UARK 2014-21 A Phase II Trial of Oncolytic Virotherapy by Systemic Administration of Edmonston Strain of Measles Virus. Official title: A phase II trial of oncolytic virotherapy by systemic administration of Edmonston strain of Measles virus, genetically engineered to express NIS, with cyclophosphamide, in patients with recurrent of refractory multiple myeloma. ClinicalTrials.gov Identifier, NCT02192775; 2014 Jul 2 [updated 2015 Sep 23; cited 2016 Jan 26]. Available from: http://clinicaltrials.gov/ct2/show/NCT02192775
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. Intrapleural Measles Virus Therapy in Patients With Malignant Pleural Mesothelioma. Official title: A phase I Trial of oncolytic Measles virotherapy in Mesothelioma. ClinicalTrials.gov Identifier, NCT01503177; 2011 Dec 16 [updated 2016 Jan 7; cited 2016 Jan 26]. Available from: http://clinicaltrials.gov/ct2/show/NCT01503177
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. Viral Therapy in Treating Patients With Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck Cancer. Official title: Phase I Trial of intratumoral administration of a NIS-expressing derivative manufactured from a genetically engineered strain of Measles virus in patients with recurrent/metastatic squamous cell carcinoma of Head and Neck. ClinicalTrials.gov Identifier, NCT01846091; 2013 May 1 [updated 2016 Jan 4; cited 2016 Jan 26]. Available from: https://clinicaltrials.gov/ct2/show/NCT01846091
  • Guillerme JB, Gregoire M, Tangy F, Fonteneau JF. Antitumor Virotherapy by Attenuated Measles Virus (MV). Biology 2013; 2:587-602; PMID:24832799; http://dx.doi.org/10.3390/biology2020587
  • van den Akker E, van der Vlugt CJ, Bleijs DA, Bergmans HE. Environmental risk assessment of replication competent viral vectors applied in clinical trials: potential effects of inserted sequences. Curr Gene Ther 2013; 13:395-412; PMID:24397527; http://dx.doi.org/10.2174/156652321306140103221621
  • Liniger M, Zuniga A, Morin TM, Combardiere D, Marty R, Wiegand M, Iter O, Knuchel M, Naim HY. Recombinant measles viruses expressing single or multiple antigens of human immunodeficiency virus (HIV-1) induce cellular and humoral responses. Vaccine 2009; 27:3299-305; PMID:19200842; http://dx.doi.org/10.1016/j.vaccine.2009.01.057
  • Despres P, Combredet C, Frenkiel MP, Lorin C, Brahic M, Tangy F. Live measles vaccine expressing the secreted form of the West Nile virus envelope glycoprotein protects against West Nile virus encephalitis. J Infect Dis 2005; 191:207-14; PMID:15609230; http://dx.doi.org/10.1086/426824
  • Brandler S, Marianneau P, Loth P, Lacôte S, Combredet C, Frenkiel M-P, Desprès P, Contamin H, Tangy F. Measles vaccine expressing the secreted form of West Nile virus envelope glycoprotein induces protective immunity in squirrel monkeys, a new model of West Nile virus infection. J Infect Dis 2012; 206:212-9; PMID:22551814; http://dx.doi.org/10.1093/infdis/jis328
  • Yoneda M, Georges-Courbot MC, Ikeda F, Ishii M, Nagata N, Jacquot F, Raoul H, Sato H, Kai C. Recombinant measles virus vaccine expressing the Nipah virus glycoprotein protects against lethal Nipah virus challenge. PLoS One 2013; 8:e58414; PMID:23516477; http://dx.doi.org/10.1371/journal.pone.0058414
  • Brandler S, Ruffie C, Najburg V, Frenkiel M-P, Bedouelle H, Desprès P, Tangy F. Pediatric measles vaccine expressing a dengue tetravalent antigen elicits neutralizing antibodies against all four dengue viruses. Vaccine 2010; 28:6730-9; PMID:20688034; http://dx.doi.org/10.1016/j.vaccine.2010.07.073
  • del Valle JR, Devaux P, Hodge G, Wegner NJ, McChesney MB, Cattaneo R. A vectored measles virus induces hepatitis B surface antigen antibodies while protecting macaques against measles virus challenge. J Virol 2007; 81:10597-605; PMID:17634218; http://dx.doi.org/10.1128/JVI.00923-07
  • Yamaji Y, Nakayama T. Recombinant measles viruses expressing respiratory syncytial virus proteins induced virus-specific CTL responses in cotton rats. Vaccine 2014; 32:4529-36; PMID:24951869; http://dx.doi.org/10.1016/j.vaccine.2014.06.024
  • Cantarella G, Liniger M, Zuniga A, Schiller JT, Billeter M, Naim HY, Glueck R. Recombinant measles virus-HPV vaccine candidates for prevention of cervical carcinoma. Vaccine 2009; 27:3385-90; PMID:19200837; http://dx.doi.org/10.1016/j.vaccine.2009.01.061
  • Griffin DE, Pan CH. Measles: old vaccines, new vaccines. Curr Top Microbiol Immunol 2009; 330:191-212; PMID:19203111
  • Mawhinney H, Allen IV, Beare JM, Bridges JM, Connolly JH, Haire M, Nevin NC, Neill DW, Hobbs JR. Dysgammaglobulinaemia complicated by disseminated measles. Br Med J 1971; 2:380-1; PMID:5575976; http://dx.doi.org/10.1136/bmj.2.5758.380
  • Mitus A, Holloway A, Evans AE, Enders JF. Attenuated measles vaccine in children with acute leukemia. Am J Dis Child 1962; 103:413-8; PMID:14474744
  • Monafo WJ, Haslam DB, Roberts RL, Zaki SR, Bellini WJ, Coffin CM. Disseminated measles infection after vaccination in a child with a congenital immunodeficiency. J Pediatr 1994; 124:273-6; PMID:8301437; http://dx.doi.org/10.1016/S0022-3476(94)70318-3
  • Moss WJ, Clements CJ, Halsey NA. Immunization of children at risk of infection with human immunodeficiendy virus. Bulletin World Health Organization 2003; 81:61-70; PMID:12640478
  • Angel JB, Walpita P, Lerch RA, Sidhu MS, Masurekar M, DeLellis RA, Noble JT, Syndman DR, Udem SA. Vaccine-associated measles pneumonitis in an adult with AIDS. Ann Intern Med 1998; 192:104-106; PMID:NOT_FOUND; http://dx.doi.org/10.7326/0003-4819-129-2-199807150-00007
  • Aaby P, Knudsen K, Whittle H, Lisse IM, Thaarup J, Poulsen A, Sodemann M, Jakobsen M, Brink K, Gansted U, et al. Long-term survival after Edmonston-Zagreb measles vaccination: Increased female mortality. J Pediatr 1993; 122:904-908; PMID:8501567; http://dx.doi.org/10.1016/S0022-3476(09)90015-4
  • Aaby P, Samb B, Simondon F, Knudsen K, Seck AM, Bennett J, Markowitz L, Rhodes P, Whittle H. Sex specific mortality after high titre measles vaccines in rural Senegal. Bull World Health Organ 1994; 72:761-70; PMID:7955026
  • Halsey. Increased mortality following high titre measles vaccines: Too much of a good thing. Pediatr Infect Dis J 1993; 12:462-465; PMID:8345977; http://dx.doi.org/10.1097/00006454-199306000-00002
  • National Institute of Health (NIH). National Institute of Allergy and Infectious Diseases (NIAID). The Jordan Report 2000: Accelerated Development of Vaccines.
  • Hussey GD, Goddard EA, Hugues J, Ryon JJ, Kerran M, Carelse E, Strebel PM, Markowitz LE, Moodie J, Barron P, et al. The effect of Edmonston-Zagreb and Schwarz measles vaccine on immune responses in infants. J Infect Dis 1996; 173:1320-1326; PMID:8648203; http://dx.doi.org/10.1093/infdis/173.6.1320
  • Aaby P, Jensen H, Simondon F, Whittle H. High-titer measles vaccination before 9 months of age and increased female mortality: do we have an explanation? Semin Pediat Infect Dis 2003; 14:220-232; PMID:NOT_FOUND; http://dx.doi.org/10.1016/S1045-1870(03)00037-2
  • Katz SL. Immunization with live attenuated measles virus vaccines: five years'experience. Arch Gesamte Virusforsch 1965; 16, 222-30; PMID:14322868; http://dx.doi.org/10.1007/BF01253813
  • Brandler S, Ruffié C, Combredet C, Brault J-B, Najburg V, Prevost M-C, Habel A, Tauber E, Desprès P, Tangy F. A recombinant measles vaccine expressing chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine 2013; 31:3718-25; PMID:23742993; http://dx.doi.org/10.1016/j.vaccine.2013.05.086
  • Stebbings R, Février M, Li B, Lorin C, Koutsoukos M, Mee E, Rose N, Hall J, Page M, Almond N, Voss G, Tangy F. Immunogenicity of a recombinant measles-HIV-1 clade B candidate vaccine. PLoS One 2012; 7(11):e50397; PMID:23226275; http://dx.doi.org/10.1371/journal.pone.0050397
  • Stebbings R, Li B, Lorin C, Koutsoukos M, Février M, Mee ET, Page M, Almond N, Tangy F, Voss G. Immunogenicity of a recombinant measles HIV-1 subtype C vaccine. Vaccine 2013; 31:6079-86; PMID:24161574; http://dx.doi.org/10.1016/j.vaccine.2013.09.072
  • Guerbois M, Moris A, Combredet C, Najburg V, Ruffié C, Février M. Live attenuated measles vaccine expressing HIV-1 Gag virus like particles covered with gp160ΔV1V2 is strongly immunogenic. Virology 2009; 388:191-203; PMID:19345390; http://dx.doi.org/10.1016/j.virol.2009.02.047
  • Ovsyannikova IG, Dhiman N, Jacobson RM, Vierkant RA, Poland GA. Frequency of measles virus-specific CD4+ and CD8+ T cells in subjects seronegative or highly seropositive for measles vaccine. Clin Diagn Lab Immunol 2003; 10:411-6; PMID:12738640
  • Van Braeckel E, Bourguignon P, Koutsoukos M, Clement F, Janssens M, Carletti I, Collard A, Demoitié MA, Voss G, Leroux-Roels G, et al. An adjuvanted polyprotein HIV-1 vaccine induces polyfunctional cross-reactive CD4+ T cell responses in seronegative volunteers. Clin Infect Dis 2011; 52:522-31; PMID:21208909; http://dx.doi.org/10.1093/cid/ciq160
  • Miller MD, Feinberg MB, Greene WC. The HIV-1 nef gene acts as a positive viral infectivity factor. Trends Microbiol 1994; 2:294-8; PMID:7981973; http://dx.doi.org/10.1016/0966-842X(94)90007-8
  • U.S. National Institutes of Health. ClinicalTrials.gov [Internet]. Dose-ranging Study to Evaluate the Safety & Immunogenicity of a HIV Vaccine 732461 in Healthy HIV Seronegative Volunteers. Official title: Dose-ranging Study to Compare the Safety and Immunogenicity of a Candidate HIV Vaccine 732461, Adjuvanted or Not, Administered According to a 0, 1 Month Schedule to Healthy Adult HIV Seronegative Volunteers. ClinicalTrials.gov Identifier, NCT00434512; 2007 Fev 12 [updated 2011 Oct 27; cited 2016 Jan 26] Available from: http://clinicaltrials.gov/ct2/show/NCT00434512
  • Akahata W, Yang ZY, Andersen H, Sun S, Holdaway HA, Kong WP, Lewis MG, Higgs S, Rossmann MG, Rao S, et al. A virus-like particle vaccine for epidemic Chikungunya virus protects nonhuman primates against infection. Nat Med 2010; 16:334-338; PMID:20111039; http://dx.doi.org/10.1038/nm.2105
  • Chang L-J, Dowd KA, Mendoza FH, Saunders JG, Sitar S, Plummer SH, Yamshchikov G, Sarwar UN, Hu Z, Enama ME, et al. Safety and tolerability of chikungunya virus-like particle vaccine in healthy adults: a phase 1 dose-escalation. Lancet 2014; 384:2046-2052; PMID:25132507; http://dx.doi.org/10.1016/S0140-6736(14)61185-5
  • Combredet C, Labrousse V, Mollet L, Lorin C, Delebecque F, Hurtrel B, McClure H, Feinberg MB, Brahic M, Tangy F. A molecularly cloned Schwarz strain of measles virus vaccine induces strong immune responses in macaques and transgenic mice. J Virol 2003; 77:11546-54; PMID:14557640; http://dx.doi.org/10.1128/JVI.77.21.11546-11554.2003
  • Myers RM, Greiner SM, Harvey ME, Griesmann G, Kuffel MJ, Buhrow SA, Reid JM, Federspiel M, Ames MM, Dingli D, et al. Preclinical pharmacology and toxicology of intravenous MV-NIS, an oncolytic measles virus administered with or without cyclophosphamide. Clin Pharmacol Ther 2007; 82:700-10; PMID:17971816; http://dx.doi.org/10.1038/sj.clpt.6100409
  • Bergmans H, Logie C, Van Maanen K, Hermsen H, Meredyth M, Van der Vlugt C. Identification of potentially hazardous human gene products in GMO risk assessment. Env Biosafety Res 2008; 7:1-9; PMID:18384725; http://dx.doi.org/10.1051/ebr:2008001
  • Hallak LK, Merchan JR, Storgard CM, Loftus JC, Russel SJ. Targeted measles virus vector displaying echistatin infects endothelial cells via alpha(v)beta3 and leads to tumor regression. Cancer Res 2005; 65:5292-300; PMID:15958576; http://dx.doi.org/10.1158/0008-5472.CAN-04-2879
  • Nakamura T, Peng KW, Harvey M, Greiner S, Lorimer IA, James CD, Russel SJ. Rescue and propagation of fully retargeted oncolytic measles virus. Nat Biotechnol 2005; 23:209-214; PMID:15685166; http://dx.doi.org/10.1038/nbt1060
  • Jing Y, Tong C, Zhang J, Nakamura T, Iankov I, Russel SJ, Merchan JR. Tumor and vascular targeting of a novel oncolytic measles virus retargeted against the urokinase receptor. Cancer Res 2009; 69:1459-1468; PMID:19208845; http://dx.doi.org/10.1158/0008-5472.CAN-08-2628
  • Ungerechts G, Springfeld C, Frenzke ME, Lampe J, Johnston PB, Parker WB, Sorscher EJ, Cattaneo R. Lymphoma chemovirotherapy: CD20-targeted and convertase-armed measles virus can synergize with fludarabine. Cancer Res 2007; 67:10939-47; PMID:18006839; http://dx.doi.org/10.1158/0008-5472.CAN-07-1252
  • Allen C, Paraskevakou G, Iankov I, Giannini C, Schroeder M, Sarkaria J, Puri RK, Russell SJ, Galanis E. Interleukin-13 displaying retargeted oncolytic measles virus strains have significant activity against gliomas with improved specificity. Mol Ther 2008; 16:1556-64; PMID:18665158; http://dx.doi.org/10.1038/mt.2008.152
  • Paraskevakou G, Allen C, Nakamura T, Zollman P, James CD, Peng KW, Schroeder M, Russel SJ, Galanis E. Epidermal growth factor receptor (EGFR)-retargeted measles virus strains effectively target EGFR- or EGFRvIII expressing gliomas; Mol Ther 2007; 15:677-86; PMID:17299404
  • Nakatsu Y, Takeda M, Kidokoro M, Kohara M, Yanagi Y. Rescue system for measles virus from cloned cDNA driven by vaccinia virus Lister vaccine strain. J Virolog Methods 2006; 137:152-155; PMID:NOT_FOUND; http://dx.doi.org/10.1016/j.jviromet.2006.05.029
  • Ong HT, Trejo TR, Pharm LD, Oberg AL, Russel SJ, Peng KW. Intravascularly administred RGD-displaying measles viruses bind to and infect neovessel endothelial cells in vivo. Mol Ther 2009. 17:1012-21; PMID:19277014; http://dx.doi.org/10.1038/mt.2009.39
  • Hasegawa K, Nakamura T, Harvey M, Ikeda Y, Oberg A, Figini M, Canevari S, Hartmann LC, Peng K-W. The use of a tropism-modified measles virus in folate receptor-targeted virotherapy of ovarian cancer. Clin Cancer Res 2006; 12:6170-8; PMID:17062694; http://dx.doi.org/10.1158/1078-0432.CCR-06-0992
  • Bellini WJ, Rota PA. Genetic diversity of wild-type measles viruses: implications for global measles elimination programs. Emerg Infect Dis 1998; 4:29-35; PMID:9452396; http://dx.doi.org/10.3201/eid0401.980105
  • Knuchel MC, Marty RR, Azzouz Morin TN, Iter O, Zuniga A, Naim HY. Relevance of pre-existing measles immunity prior immunization with a recombinant measles virus vector. Hum Vaccines Immunother 2013; 9:599-606; PMID:23324399; http://dx.doi.org/10.4161/hv.23241
  • Rager-Zisman B, Bazarsky E, Skibin A, Chamney S, Belmaker I, Shai I, Kordysh E, Griffin DE. The effect of measles-mumps-rubella (MMR) immunization on the immune responses of previously immunized primary school children. Vaccine 2003; 21:2580-2588; PMID:12744894; http://dx.doi.org/10.1016/S0264-410X(03)00053-7
  • Wong-Chew RM, Beeler JA, Audet S, Santos JI. Cellular and humoral immune responses to measles in immune adults re-immunized with measles vaccine. J Med Virol 2003; 70:276-280; PMID:12696117; http://dx.doi.org/10.1002/jmv.10390
  • Gans H, DeHovitz R, Forghani B, Beeler J, Maldonado Y, Arvin AM. Measles and mumps vaccination as a model to investigate the developing immune system: passive and active immunity during the first year of live. Vaccine 2003; 3398-305; PMID:12850348; http://dx.doi.org/10.1016/S0264-410X(03)00341-4
  • Liniger M, Zuniga A, Tamin A, Azzouz-Morin TN, Knuchel M, Marty R, Wiegand M, Weibel S, Kelvin D, Rota PA, Naim HY. Induction of neutralising antibodies and cellular immune responses against SARS coronavirus by recombinant measles virus. Vaccine 2008; 26:2164-74; PMID:18346823
  • Low N, Kraemer S, Schneider M, Restrepo AMH. Immunogenicity and safety of aerosolized measles vaccine: systematic review and meta-analysis. Vaccine 2008; 26:383-98; PMID:18082295; http://dx.doi.org/10.1016/j.vaccine.2007.11.010
  • Harper SA, Fukuda K, Cox NJ, Bridges CB, Advisory Committee on Immunization Practices. Using live, attenuated influenza vaccine for prevention and control of influenza: supplemental recommendations of the advisory committee on influenza practices (ACIP). MMWR Recomm Rep 2003; 52:1-8; PMID:AMBIGUOUS
  • Mader EK, Maeyama Y, Lin Y, Butler GW, Russel HM, Galanis E, Russel SJ, Dietz AB, Peng KW. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res 2009; 15:7246-55; PMID:19934299; http://dx.doi.org/10.1158/1078-0432.CCR-09-1292
  • Schenk-Braat EA, van Mierlo MM, Wagemaker G, Bangma CH, Kaptein LC. An inventory of shedding data from clinical gene therapy trials. J Gene Med 2007; 9:910-21; PMID:17880045; http://dx.doi.org/10.1002/jgm.1096
  • Ludlow M, de Vries RD, Lemon K, McQuaid S, Millar E, van Amerongen G, Yuksel S, Verburgh RJ, Osterhaus ADME, de Swart RL, Duprex WP. Infection of lymphoid tissues in the macaque upper respiratory tract contributes to the emergence of transmissible measles virus. J Gen Virol 2013; 94, 1933-44; PMID:23784446; http://dx.doi.org/10.1099/vir.0.054650-0
  • Gresser I, Katz SL. Isolation of measles virus from urine. N Engl J Med 1960; 263:452-4; PMID:13829216; http://dx.doi.org/10.1056/NEJM196009012630907
  • Lin W-H, Kouyos RD, Adams RJ, Grenfell BT, Griffin DE. Prolonged persistence of measles virus RNA is characteristic of primary infection dynamics. PNAS 2012; 109:14989-14994; PMID:22872860; http://dx.doi.org/10.1073/pnas.1211138109
  • Rennick L, de Vries RD, Carsillo TJ, Lemon K, van Amerongen G, Ludlow M, Nguyen DT, Yuksel S, Verburgh RJ, Haddock P, McQuaid S, Duprex WP, de Swart RL. Live-attenuated measles virus vaccine targets dendritic cells and macrophages in muscle of nonhuman primates. J Virol 2015; 89:2192-2200; PMID:25473055; http://dx.doi.org/10.1128/JVI.02924-14
  • Rota PA, Khan AS, Durigon E, Yuran T, Villamarzo YS, Bellini WJ. Detection of measles virus RNA in urine specimens from vaccine recipients. J Clin Microbiol 1995; 33:2485-8; PMID:7494055.
  • Morfin F, Beguin A, Lina B, Thouvenot D. Detection of measles vaccine in the throat of a vaccinated child. Vaccine 2002; 20:1541-3; PMID:11858860; http://dx.doi.org/10.1016/S0264-410X(01)00495-9
  • Vergison A, Lina B, Van Beers D., Van Ranst M, Stévart O, Lepage P, et al., editors. Symptomatic post vaccinal excretion of Measles in the nasopharynx: a report of nine cases. Proceedings of the 44th Annual Meeting of the Infectious Diseases Society of America (IDSA), 2006 Oct 12-15. Toronto, ON; Poster session: Pediatrics I, P605.
  • Peng KW, Frenzke M, Myers R, Soeffker D, Harvey M, Greiner S, Galanis E, Cattaneo R, Fderspiel MJ, Russel SJ. Biodistribution of oncolytic measles virus after intraperitoneal administration into Ifnar-CD46Ge transgenic mice. Hum Gene Ther 2003; 14:1565-77; PMID:14577918; http://dx.doi.org/10.1089/104303403322495070
  • Kambara H, Saeki Y, Chiocca EA. Cyclophosphamide allows for in vivo dose reduction of a potent oncolytic virus. Cancer Res 2005; 65:11255-8; PMID:16357128; http://dx.doi.org/10.1158/0008-5472.CAN-05-2278