Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Appl Microbiol Biotechnol

DOI 10.1007/s00253-016-7557-x

MINI-REVIEW

Lactic acid bacteria as mucosal delivery vehicles: a realistic


therapeutic option
Miao Wang 1 & Zeqian Gao 1 & Yongguang Zhang 1,2 & Li Pan 1,2

Received: 10 March 2016 / Revised: 12 April 2016 / Accepted: 14 April 2016


# Springer-Verlag Berlin Heidelberg 2016

Abstract Recombinant lactic acid bacteria (LAB), in partic- Keywords Lactic acid bacteria . Mucosal immunity . Delivery
ular lactococci and lactobacilli, have gained increasing interest vehicles . Therapeutic molecules . Mucosal vaccines .
as mucosal delivery vehicles in recent years. With the devel- DNA vaccines
opment of mucosal vaccines, studies on LAB expression sys-
tems have been mainly focused on the generation of genetic
tools for antigen expression in different locations. Introduction
Recombinant LAB show advantages in a wide range of as-
pects over other mucosal delivery systems and represent an Lactic acid bacteria (LAB), which include species of
attractive candidate for the delivery of therapeutic and prophy- Lactobacillus, Leuconostoc, Lactococcus, Pediococcus, and
lactic molecules in different applications. Here, we review the Streptococcus, are a heterogeneous group of non-sporulating
recent data on the use of recombinant LAB as mucosal deliv- and gram-positive bacteria of high relevance for the produc-
ery vectors and the associated health benefits, including the tion of fermented food and beverages and for human and
prevention and treatment of inflammatory bowel diseases animal health (Gareau et al. 2010). LAB have been used for
(IBDs), autoimmune disorders, and infections by pathogenic centuries to produce and preserve food, such as cheese (spe-
microorganisms from mucosal surfaces. In addition, we dis- cies of Lactococcus), pickled vegetables (species of
cuss the use of LAB as vehicles to deliver DNA directly to Lactobacillus and Lactococcus), and yoghurt (species of
eukaryotic cells. Researches from the last 5 years demonstrate Lactobacillus and Streptococcus); they are non-pathogenic
that LAB as vectors for mucosal delivery of therapeutic mol- and are generally regarded as safe (GRAS) according to the
ecules seem to be a realistic therapeutic option both in human US Food and Drug Administration (FDA) (Bermudez-
and animal diseases. Humaran et al. 2013; Wells and Mercenier 2008). LAB reside
in a diverse range of niches including milk, the surface of
plants, the oral cavity, gastrointestinal tract (GIT), and vagina
of humans and animals (Bermudez-Humaran et al. 2013).
Specific strains of LAB, Lactobacillus, Streptococcus, and
* Yongguang Zhang Bifidobacterium, are used as probiotics for human and animal
yongguang.zhang@163.com health, as they play a crucial role in maintaining gastrointesti-
* Li Pan nal homeostasis and influence other organs by producing bio-
panli@caas.cn active metabolites and by modulating immunological param-
eters and intestinal permeability (de Vrese and Schrezenmeir
1
State Key Laboratory of Veterinary Etiological Biology, National 2008; Tsai et al. 2012). In the past two decades, LAB strains,
Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary
Research Institute, Chinese Academy of Agricultural Sciences, 1
in particular Lactococcus lactic and Lactobacillus plantarum
Xujiaping, Yanchangbu, Lanzhou, Gansu 730046, China (Cano-Garrido et al. 2015), have generated increasing inter-
2
Jiangsu Co-innovation Center for Prevention and Control of
ests as vectors for the delivery of therapeutic and prophylactic
Important Animal Infectious Diseases and Zoonoses, molecules via the intranasal, oral, or genital mucosal surfaces
Yangzhou, Jiangsu, China (Dimitrijevic et al. 2014; Hiramatsu et al. 2014; Li et al.
Appl Microbiol Biotechnol

2015a; Wang et al. 2014a; Xu et al. 2015a). Recombinant and enzymes, which could advance the development of multi-
LAB-mediated delivery of mucosal vaccines, particularly via valent vaccines (Lei et al. 2015a; Mobergslien et al. 2015;
intranasal or oral immunization, is more applicable and con- Saraiva et al. 2015; Yang et al. 2015). Dietary LAB can survive
venient than the intravenous or intramuscular injection of tra- exposure to gastric acid and contact with bile, allowing uptake
ditional systemic vaccines, because mucosal vaccination can into the inductive sites of the mucosal immune system such as
save both time and labor, for instance, oral vaccine could be Peyer’s patches (Wang et al. 2014b). In addition, the mucosal
made into powder and stirred into the feed, especially appli- administration of LAB by oral, intranasal, or genital routes can
cable for scaled livestock and poultry, and meanwhile, muco- stimulate both mucosal and systemic immune responses. Their
sal immunization can reduce the pain compared to intravenous ability to elicit the production of secretory IgA is one major
or intramuscular injection. Moreover, mucosal delivery of advantage of LAB as mucosal vaccines (Macpherson et al.
therapeutics and prophylactic molecules for chronic diseases 2015). LAB are not only used as live bacteria for mucosal
and infections associated with mucosal tissues may increase immunization, but certain killed recombinant strains have
their efficacy and specificity while minimizing the side effects shown efficacy as mucosal vehicles, mainly by intranasal vac-
of systemic vaccine administration (LeBlanc et al. 2013; Pan cination (Almada et al. 2015). LAB expression systems (espe-
et al. 2014; Wang et al. 2011). Many LAB strains have the cially the NICE and the Self-Immobilizing Plasmid (pSIP) sys-
capacity to survive transit through the GIT, and most of them tems) enable the targeted expression of foreign antigens at spe-
are present into the intestinal tract and eventually remain in the cific locations (Fig. 1), such as the cytoplasm, cell wall, or the
lumen or are trapped in the mucus, which facilitates mucosal medium, which prevents protein degradation under harsh con-
targeting of therapeutic molecules (Daniel et al. 2011). ditions associated with the administration route or through in-
Because of these positive features, LAB have emerged as an teraction with the environment that may affect immunogenicity
attractive potential alternative to other mucosal delivery sys- (Nguyen et al. 2015; Zhou et al. 2015).
tems, such as microspheres, liposomes, nanoparticles,
immunostimulating complexes, and attenuated pathogens
(Daudel et al. 2007; Hu et al. 2001; Pabreja et al. 2014; Prevention and treatment of inflammatory bowel
Wendorf et al. 2008). In this review, we summarize recent data diseases
on recombinant LAB as mucosal delivery vehicles and discuss
their potential medical applications in animal studies and hu- Inflammatory bowel disease (IBD) affects millions of people
man clinical trials. worldwide and refers to two chronic diseases that cause inflam-
mation of the intestines: Crohn’s disease (CD) and ulcerative
colitis (UC). IBD symptoms include abdominal pain, cramps,
LAB delivery systems and advantages as mucosal diarrhea, fatigue, fever, weight loss, and poor appetite
delivery vehicles (Bermudez-Humaran et al. 2015). The first reported use of
recombinant LAB for the treatment of IBD was published
Mucosal vaccines have attracted significant attention in recent 15 years ago, in a study that described the use of secreted IL-
years because of their remarkable properties and potential ap- 10 in two mouse models of colitis (Steidler et al. 2000). This
plications against a wide range of microbial pathogens. They was followed by several studies reporting on the use of recom-
confer protection against infections at mucosal surfaces, pre- binant LAB for human IL-10 delivery (Bermudez-Humaran
vent antigen degradation, and promote uptake in the GIT, in et al. 2015; Martin et al. 2014; Schotte et al. 2000; Steidler
addition to activating adaptive immune responses. Despite et al. 2003). The effect of recombinant Lactococcus lactis
efforts to develop attenuated bacterial pathogens as mucosal MG1363 secreting IL-10 generated using a stress-inducible
vaccines based on their ability to elicit host immune responses controlled expression (SICE) system was tested in a murine
against foreign antigens, there are limitations to the use of model of chronic low inflammation in response to repeated
attenuated live vaccines, such as the potential risk of reversion dinitrobenzene sulfonic acid challenge (Martin et al. 2014).
to a virulent phenotype, side effects of varying severity, and The results showed that oral administration of L. lactis secret-
imbalances between immunogenicity and reactogenicity ing active IL-10 (LL-IL10) decreased gut hyperpermeability,
(Roland et al. 2005; Wang et al. 2013). LAB can overcome improved dysfunctional parameters, and enhanced immune ac-
some of the limitations of attenuated vaccines as mucosal tivation in immunized mice. Lactococci strains containing the
delivery vehicles and fulfill the requirements of delivery sys- recombinant plasmid and the empty plasmid both
tems for mucosal administration. counterbalanced cytokine levels and LL-IL10 T cell levels,
LAB are non-pathogenic microorganisms, and they are con- confirming previous data in which the immunomodulatory ca-
sidered safer than some attenuated vectors. Recombinant LAB pacities of wild-type L. lactis were observed in in vitro and
strains have the capacity to express numerous heterologous in vivo assays (Smelt et al. 2012). The protective effects of
proteins, such as chimeric or non-chimeric antigens, cytokines, IL-10 delivered by recombinant L. lactis MG1363 were further
Appl Microbiol Biotechnol

Fig. 1 Four approaches based on the use of lactic acid bacteria (LAB) as antigens into the lumen (c). The strains harboring a eukaryotic
delivery vehicles for protein antigens or DNA in the gastrointestinal tract. expression system transfer DNA directly to eukaryotes and transduce
Cell wall-anchored proteins produced by LAB interact with cells in the host cells to express and release proteins into the lumen (d). DC
intestinal tract (a). Cytoplasmic proteins expressed by LAB are released dendritic cell, M cell microfold cell, LAB lactic acid bacteria
into the lumen after lysis of LAB (b). LAB directly secrete protein

tested using two expression systems, the SICE system and a results highlight the importance of other anti-inflammatory
complementary DNA (cDNA) delivery system named pValac, molecules (especially serine protease inhibitors) expressed by
in a murine colitis model mimicking the relapse nature of IBD recombinant LAB for the treatment of IBD in addition to the
(del Carmen et al. 2014). The beneficial effects of non-invasive L. lactis IL-10 recombinant approach (de LeBlanc et al. 2015).
recombinant L. lactis strains through two different ways be- Taken together, these studies indicate that the use of recom-
tween DNA and protein delivery were compared, which binant LAB for mucosal therapy could be valuable for the
showed that both approaches had a similar anti-inflammatory treatment of IBD in humans in the future.
effect in a chronic 2,4,6-trinitrobenzenesulfonic acid-induced
colitis model, underscoring the relevance of L. lactis delivered
IL-10 for the treatment of IBD. The efficacy of LAB-mediated Preventing infections by pathogenic microorganisms
delivery to the gut lumen was tested using other anti- and parasites at mucosal surfaces
inflammatory molecules. A recent study compared the efficacy
of recombinant L. lactis, MG1363 and NZ9000, secreting two Over the past 10 years, several anti-infective bioshield strate-
types of anti-inflammatory molecules, namely cytokines and gies with recombinant LAB have been applied for preventing
serine protease inhibitors, to identify the best strategy for the infection at the mucosal surface (Table 1). The first report on
treatment of IBD in a dextran sulfate sodium-induced colitis the use of live L. lactis for the delivery of a vaccinal antigen
mouse model (Bermudez-Humaran et al. 2015). The results was published in 1993 (Wells et al. 1993). In that study, the
showed that treatment with recombinant L. lactis strains deliv- protective role of recombinant L. lactis expressing the tetanus
ering the serine protease inhibitors elafin and secretory leuko- fragment C (TTFC) was initially tested via subcutaneous in-
cyte protease inhibitor (SLPI) was more efficient than treatment jection in mice after tetanus toxin challenge and later via oral
with anti-inflammatory cytokines (IL-10 and TGF-β1) for al- and nasal administration using live L. lactis producing TTFC
leviating the symptoms of colitis. Elafin and SLPI released in (Norton et al. 1997; Robinson et al. 1997; Wells et al. 1993).
the lumen by LAB attenuated the effects of proteases produced Although the protective efficacy was the same, nasal immu-
by inflammatory cells, downregulated pro-inflammatory tran- nization elicited higher levels of serum IgG and mucosal IgA
scription factors, restored barrier functions, and promoted anti- than oral delivery. Since then, numerous studies have reported
microbial activity (Butler et al. 2006; Henriksen et al. 2004; on the use of recombinant LAB as mucosal vaccines express-
Motta et al. 2012), indicating that they exerted anti- ing antigens derived from viral, bacterial, fungal, or parasitic
inflammatory effects more efficiently than cytokines. These pathogens.
Appl Microbiol Biotechnol

Table 1 Current anti-infective applications of lactic acid bacteria (LAB) delivery

Application Vehicle Recombinant protein Model Route References

Bacillus Lactobacillus paracasei scFv antibody Mouse Oral Andersen et al. (2011)
anthracis
Candida albicans Lactobacillus casei Enolase 1 protein Mouse Oral Shibasaki et al. (2014)
Clostridium Lactococcus lactis Cytotoxins Mouse Oral Guo et al. (2015)
difficile
Cryptosporidium L. casei Surface adhesion protein Mouse Oral Geriletu et al. (2011)
parvum P23
CSFV Lactobacillus plantarum E2 protein with Pig Oral Xu et al. (2015b)
thymosin -1
EHEC L. lactis EspB Mouse Oral Ahmed et al. (2014)
Eimeria tenella L. lactis 3-1E protein Chicken Oral Ma et al. (2013)
ETEC L. casei F41 Mouse Oral and Liu et al. (2014b)
intranasal
Helicobacter Lactobacillus acidophilus, Adhesin Hp0410, CTB- Mouse Oral Hongying et al. (2014), Li et al. (2014a),
pylori L. lactis UE, UreB with IL-2 Zhang et al. (2014)
Hepatitis E virus L. lactis ORF2 protein Mouse Oral Gao et al. (2015)
HPAI L. lactis NA, HA with LTB Chicken, Oral and Lei et al. (2015b, c, d)
ferret Intranasal
HPV L. lactis, L. casei E7 protein, E7 protein Mouse, Intranasal Kawana et al. (2014), Li et al. (2014b),
with IL-12 human and oral Ribelles et al. (2013)
IBV L. lactis EpiC peptide with SPA Chicken Oral and Cao et al. (2013)
Intranasal
Influenza viruses L. lactis, L. casei, NA, HA, NA with CTB, Mouse, Oral and Chowdhury et al. (2014), Lei et al. (2015a, e),
Lactobacillus rhamnosus, sM2 with CTA1, sM2 human intranasal Li et al. (2015b), Shi et al. (2014), Song
L. plantarum, and HA2 with CTA1 et al. (2014), Waki et al. (2014)
Lactobacillus brevis
PRRSV L. lactis ORF6 Mouse Intranasal Wang et al. (2014c)
Rotavirus L. rhamnosus, L. acidophilus Protein G, ARP1, oral Mouse, Oral Alvarez et al. (2015), Gunaydin et al. (2014),
attenuated human pig Liu et al. (2014a)
rotavirus
Salmonella L. acidophilus HMLABs Caco-2, Oral Chen et al. (2013)
mouse
Staphylococcus L. lactis SEB Mouse Oral Asensi et al. (2013)
aureus
Streptococcus Lactobacillus zeae scFv antibody Mouse Oral Andersen et al. (2011)
mutans
TGEV L. casei Muramyl dipeptide and Mouse Oral Jiang et al. (2014)
tuftsin fusion protein

scFv antibody single-chain fragment variable antibody, CSFV classical swine fever virus, EHEC entero-hemorrhagic E. coli, EspB E. coli secreted
protein B, ETEC entero-toxigenic Escherichia coli, CTB-UE cholera toxin B subunit and urease subunits, UreB urease B, HPAI highly pathogenic H5N1
avian influenza virus, NA neuraminidase, HA hemagglutinin, LTB heat-labile toxin B subunit, HPV human papillomavirus, IL interleukin, IBV infectious
bronchitis virus, SPA Staphylococcus aureus protein A, CTA1 cholera toxin subunit A1, sM2 consensus matrix protein-2, PRRSV porcine reproductive
and respiratory syndrome virus, ARP1 anti-rotavirus protein 1, HMLABs the heat-killed multispecies combination of LAB, TGEV transmissible
gastroenteritis virus, SEB Staphylococcal enterotoxin B

The health benefits of recombinant LAB in the prevention systemic responses stimulated by these two strains were eval-
of human immunodeficiency virus (HIV)-1 infection at the uated following oral administration in mice (Wang et al.
mucosa have been clearly demonstrated; however, in recent 2012). Both recombinant strains improved the production of
years, research focus has shifted toward their effects on influ- specific anti-HA1 IgA antibody in the mucosa and IgG in
enza virus, rotavirus, and human papillomavirus infections serum, as well as splenic T lymphocyte proliferative re-
(Lei et al. 2015a, c, d; Li et al. 2014b, 2015b; Wells and sponses; however, DLD17-pH was more effective in inducing
Mercenier 2008). Two recombinant Lactobacillus strains mucosal and systemic immune responses than LA4356-pH,
(LA4356-pH and DLD17-pH) expressing the highly patho- promoting higher anti-HA1-specific IgA and IgG levels. In a
genic avian influenza (HPAI) virus protein hemagglutinin trial testing oral vaccines against avian influenza virus (AIV),
(HA)-1 were successfully generated, and the mucosal and a recombinant L. plantarum NC8 (CCUG 61730) strain
Appl Microbiol Biotechnol

expressing the HA gene of H9N2 AIV via the pSIP system Nowadays, it is feasible to express antibody fragments by
was orally administered in mice (Shi et al. 2014). The recom- LAB against infectious agents. The single-chain fragment var-
binant NC8 strain induced mucosal and systemic immune iable (scFv) antibodies can stop pathogen from reaching the
responses, eliciting the production of sIgA, IgG, and hemag- epithelial cells by preventing pathogen adherence or neutral-
glutination inhibition antibodies and conferring complete pro- izing bacteria toxin. Based on those characteristics, scFv se-
tection against challenge with mouse-adapted H9N2 virus. In creted by L. casei was used to block cell-associated HIV-1
a different approach to develop a safer and more effective transmission across a cervical epithelial monolayer in vitro
mucosal vaccine against AIV, a Lactobacillus casei strain (Chancey et al. 2006). For in vivo immunotherapy, a recom-
L525 was designed to express a conserved matrix protein with binant scFv antibody fragment was constructed and expressed
the cholera toxin subunit (pgsA-CTA1-sM2/L. casei) on the by Lactobacillus zeae, and it recognized streptococcal antigen
surface and the sM2 protein without CTA1 as a control I/II. The transformed lactobacilli exerted the therapeutic action
(Chowdhury et al. 2014). After verification of the surface on experimental animals by decreasing the quantity of
localization, the effects of the vaccine were tested in mice in Streptococcus mutans bacteria and reducing the development
response to oral or nasal inoculation. The recombinant L. casei of caries (Kruger et al. 2002). The recombinant LAB were
expressing CTA1-conjugated sM2 conferred better protection also used to deliver antibody fragments against surface epi-
against divergent influenza virus subtypes than pgsA-sM2/ topes of many pathogens, such as Bacillus anthracis,
L. casei but less potent mucosal and systemic immune re- Porphyromonas gingivalis, and rotavirus (Andersen et al.
sponses. A further study aimed at improving the efficacy of 2011; Marcotte et al. 2006; Pant et al. 2006). This strategy
this system led to the induction of strong mucosal immune could be considered for immunotherapy or prophylaxis and
responses with CTA1 using L. casei as a delivery vehicle (Li will overcome the need for daily administration of purified
et al. 2015b). In this latter strategy, the HA2 gene from H5N1 antibodies or antibody fragments.
strain was used as an additional component of the vaccine as Other recent anti-infective strategies are based on LAB
well as conserved sM2. Both oral and nasal administrations of prototype vaccines against different viral infections including
pgs-CTAsM2HA2/L. casei in mice generated high levels of human papillomavirus (Kawana et al. 2014; Li et al. 2014a),
serum IgG (IgG1 and IgG2a) and mucosal IgA and enhanced rotavirus (Alvarez et al. 2015; Liu et al. 2014a), and hepatitis
sM2- or HA2-specific cell-mediated immunity, with high E virus (Gao et al. 2015); bacterial infections including
levels of IFN-γ and IL-4. HA2 is one of the highly conserved Staphylococcus aureus (Veloso et al. 2015), Candida albicans
subunit of HA which was investigated and found to be effec- (Shibasaki et al. 2014), and Clostridium difficile (Guo et al.
tive for cross-protective antibodies against several subtypes of 2015); and parasitic infections including Babesia bovis
viruses in mice, and therefore, HA2-based recombinant vac- (Bautista-Garfias et al. 2015), Eimeria tenella (Ma et al.
cines demonstrated better effects against AIV. Furthermore, 2013), and Cryptosporidium parvum (Geriletu et al. 2011).
the recombinant pgs-CTAsM2HA2/L. casei showed protec- Despite the promising features of mucosal vaccination as an
tive effects and elicited long-lasting immunity against diver- anti-infective approach against numerous antigens, most of
gent influenza subtypes. Intranasal administration induced these vaccines are still in the experimental stage and only a
stronger immune responses and better protection than oral handful are currently approved for human and animal use
administration despite shorter immunization times and lower (Holmgren and Czerkinsky 2005; Wang et al. 2015).
dosages, supporting the greater efficacy of intranasal inocula-
tion for this mucosal vaccine. Although the HA protein is
the target of conventional influenza vaccines, neuramini- Prevention and treatment of autoimmune disorders
dase (NA), the other major glycoprotein on the surface
of the influenza virus, is attracting increasing attention. Recombinant LAB have attracted attention for their potential
Recombinant L. lactis (NZ3000)/pNZ2103-NA adminis- in the treatment of autoimmune disorders, such as allergy,
tered orally in the absence of adjuvant to evaluate the diabetes, and obesity (Walters et al. 2014). The potential of
protective immunity against homologous H5N1 virus chal- LAB as an oral allergy vaccine has been researched extensive-
lenge elicited significant humoral and mucosal immune ly because of their safety as food microorganisms and their
responses in a chicken model (Lei et al. 2015c). In anoth- potent adjuvant activity in triggering anti-allergic immune re-
er approach, the immunogenicity of an L. lactis (NZ9000) sponses (Cano-Garrido et al. 2015). In a recent study, the
vectored vaccine expressing nucleoprotein (NP) was en- prophylactic effect of recombinant L. plantarum NCL21 ex-
hanced using the cholera toxin B (CTB) subunit as a pressing a common Japanese cedar pollen allergen, Cry j 1
mucosal adjuvant (Lei et al. 2015a). Oral administration (Cry j 1-LAB), was evaluated in vivo (Ohkouchi et al. 2012).
of L. lactis/pNZ8008-NP with CTB in mice induced ef- Oral vaccination with Cry j 1-LAB ameliorated cedar
fective humoral and mucosal responses, providing good pollinosis-like clinical symptoms and allergen-specific IgE
protection against divergent influenza viruses. responses in a murine model of cedar pollinosis, providing a
Appl Microbiol Biotechnol

new platform of safe and effective oral immunotherapy normoglycemia in non-obese diabetic (NOD) mice when used
against Japanese cedar pollinosis. Another engineered in combination with low-dose systemic anti-CD3 (Robert
L. plantarum WCFS1 (CCUG 61730) was generated by co- et al. 2014; Takiishi et al. 2012). L. lactis-mediated delivery
valent attachment of the Fes p1 allergen (from Festuca of T1D autoantigens, such as GAD65 and tyrosine
pratensis) to the bacterial cell wall and consequent surface phosphatase-like protein ICA512, in combination with human
display (Minic et al. 2015). Assessment of its effects in a IL-10 has been successfully used to treat T1D (Robert et al.
mouse model showed that oral administration of recombinant 2015; Vandenbroucke et al. 2004). Heat shock proteins (Hsps)
and non-recombinant live L. plantarum reduced peripheral as autoantigens are delivered by LAB-mediated mucosal ad-
blood eosinophilia and increased serum IgG2A levels, which ministration, and oral administration of L. lactis expressing
was attributed to L. plantarum itself, whereas mice treated Hsp shows potential for enhancing immune tolerance in
with the recombinant LAB construct showed increased spe- T1D. Nasal administration of the fusion protein Hsp65 with
cific serum IgA levels, indicating that cell surface expression tandem repeats of P227 (Hsp65-6P277) decreases the inci-
of the Fes p1 allergen potentiated the beneficial therapeutic dence of T1D in NOD mice (Jin et al. 2010). Recombinant
effect of orally administered L. plantarum in an allergen- LAB producing Hsp65-6P277 are used to deliver large
specific manner. Another important anti-allergic application amounts of antigen and enhance long-term immune tolerance.
of antigen-producing LAB is in the treatment of celiac disease Oral administration of recombinant L. lactis NZ9000 express-
(CD), an often-undiagnosed autoimmune disease caused by ing the fusion protein prevents hyperglycemia, increases glu-
gluten in wheat-containing foods. Prolyl endopeptidases cose tolerance, and reduces insulitis (Ma et al. 2014).
(PEPs) have the capacity to hydrolyze the peptide bond on In summary, the use of recombinant LAB strains is a po-
the carboxyl side of an internal proline residue and can de- tentially effective strategy for the treatment of autoimmune
grade immunotoxic peptides responsible for CD, such as a 33- disorders, and novel therapeutic approaches for mucosally
residue gluten peptide (33-mer). Modified LAB provide an induced immunological tolerance are urgently needed.
effective approach to the local delivery of PEP, which catalyze
the cleavage of the gliadin fragment leading to the detoxifica-
tion of dietary gluten. Oral administration of PEP is therefore a DNA vaccines
potential therapeutic approach for the treatment of CD
(Alvarez-Sieiro et al. 2014). Two food-grade L. casei strains Among recent strategies based on genetically modified LAB
(L. casei IPLA1503 and L. casei IPLA1506) were engineered as live delivery vectors, the direct delivery of DNA into eu-
to deliver PEP in an in vitro model mimicking the environ- karyotic cells has several advantages, including the induction
ment of the small intestine: One strain was designed to secrete of potent cellular and humoral immune responses and the
PEP into the extracellular medium and the other one for potential to express multiple antigens or epitopes in a single
retaining the enzyme in the intracellular environment. The DNA vector. LAB are more attractive than other vectors such
results showed that the former was more effective at degrading as Salmonella spp., Listeria monocytogenes, and Escherichia
the immunotoxic 33-mer peptide than the latter and showed coli for the delivery of DNA to eukaryotic cells because LAB
resistance against simulated gastrointestinal stress while main- are GRAS and considered as transiting and non-invasive bac-
taining the ability to secrete PEP, suggesting its potential for teria (Daudel et al. 2007; Grillot-Courvalin et al. 1998).
the delivery of PEP to the duodenum of celiac patients. A new invasive L. lactis strain (NZ9000) was engineered to
Type 1 diabetes (T1D) is a prominent example of a chronic produce a mutant form of internalin A (mInlA) of
autoimmune disease characterized by the selective disruption L. monocytogenes (LL-InlA+) that allows the binding of
of insulin-producing pancreatic ß-cells by pathogenic self- mInlA to murine E-cadherin (de Azevedo et al. 2012). The
reactive T cells. Although antibody-based immunotherapies mInlA protein was successfully expressed on the surface of
and broad immunosuppressive drugs have been developed L. lactis, as validated by fluorescence microscopy. The LL-
to control this disease, these treatments are accompanied by mInlA+ strain attached to Caco-2 cells with an intracellular
short- and long-term toxic effects and failure of discrimination localization and increased the invasiveness of the cell by
between autoreactive and non-autoimmune effectors (Luo 1000-fold and the plasmid containing the cDNA of bovine
et al. 2010; Petrovsky 2010; Van Belle et al. 2011). Antigen- ß-lactoglobulin (BLG) transfer 10 times in vitro, whereas in
based immunotherapies are therefore becoming the focus of vivo, the proportion of mice producing BLG increased only
research because of their ability to selectively target disease- slightly. One possible interpretation of this result is that a
relevant T cells while preserving the function of the immune reduction in the direct interaction of L. lactis with the mouse
system. Mucosal delivery of modified L. lactis MG1363 se- intestinal epithelium inhibited the binding of the recombinant
creting the proinsulin autoantigen or glutamic acid decarbox- mInlA to the corresponding receptors. Another hypothesis is
ylase (GAD) along with IL-10 resulted in the stabilization of the release of the plasmid induced by lysed Lactococcus under
insulitis, preserved functional β-cell mass, and restored the physicochemical conditions of the gut lumen. Considering
Appl Microbiol Biotechnol

these factors, Lactobacillus could be a better option because of expressed antigens (cytoplasmic, secreted, or cell wall-
its longer persistence in the gut. The third probable reason is anchored) and the effect of localization on their immunogenic
that epithelial cells saturate with bacteria, even in the presence capacity. LAB expressing antigens on the surface may show
of invasin (Zelmer et al. 2005). To test the roles of dendritic more pronounced immunological effects than those express-
cells (DCs) in the process of LAB DNA vaccination, mouse ing cytoplasmic antigens, since cell surface antigens have the
bone marrow-derived DCs (BMDCs) were incubated with ability to directly interact with the environment and avoid
invasive L. lactis strains expressing LL-mIlA+ and S. aureus proteolytic degradation (Nouaille et al. 2003; Pontes et al.
fibronectin-binding protein A (LL-FnBPA+), both carrying a 2011). However, in some cases, intracellular antigens are more
plasmid DNA vaccine (pValac) encoding for BLG (de immunogenic than those expressed in other locations because
Azevedo et al. 2015). In this experiment, both recombinant intracellular proteins avoid exposure to harsh environmental
invasive strains were able to transfer DNA directly to conditions (Shaw et al. 2000; Wells and Mercenier 2008). On
BMDCs, resulting in the secretion of IL-12 by the cells, inde- the other hand, biologically active molecules, such as hor-
pendent of the invasive status. The recombinant strains were mones, enzymes, and cytokines, require an extracellular local-
approximately 100 times more effective at invading the Caco- ization to produce a functional protein. In addition, different
2 monolayer than the wild-type strains. Furthermore, data mucosal administration routes elicit different immune effects.
from a co-culture model indicated that pValac:BLG could be Intranasal administration is more effective than the intragastric
transferred to DCs across a monolayer of epithelial cells by the route for inducing systemic responses, and killed recombinant
LL-mInlA strain, suggesting that DNA vaccines can cross the LAB are more suitable for intranasal vaccination (Almada
epithelial barrier and express antigens in DCs. et al. 2015; Hanniffy et al. 2007; Vintini and Medina 2011).
In a previous study, the Lactobacillus acidophilus SW1 However, intragastric immunization is more feasible, econom-
(LASW1) strain derived from swine was shown to act as an ic, and applicable for the animal feed business. Despite broad
immunoadjuvant when used as a live vector for DNA vacci- prospects in application of recombinant LAB, there are still
nation against foot-and-mouth disease virus (FMDV) (Li et al. some issues existing which need to be solved, such as safety,
2007). Immune responses to the DNA vaccine were only test- stability, and persistence. The foreign genes have a potential to
ed after injection; however, mucosal immunization could affect safety status of LAB and even have unsuspected harm-
stimulate a specific immune response. Therefore, further in- ful effects on the host because of producing poisonous metab-
vestigation should be aimed at making LAB effective for mu- olizing substances. To address this issue, lots of animal exper-
cosal delivery as DNA vaccines and at demonstrating their iments and clinical trials need to be tested frequently and
immune efficacy in animal models. Another current study repeatedly. Although many protection studies have been per-
was published that LASW1 was used as an oral adjuvant for formed in small animals, determining doses and side effects in
a DNA vaccine (pRC/CMV-vp1) harboring the FMDV VP1 humans requires vaccination studies in larger animals. In most
gene (Su et al. 2014). Unlike the former study, the strain was experiments, plasmids are used as vectors to express foreign
orally administered rather than as a DNA vaccine by intramus- genes, but the transgenes could potentially be transferred to
cular routes, avoiding the unstable replication of recombinant other individuals. A strategy to solve the above problem is the
DNA plasmid in LASW1. The results showed that oral ad- genome recombination of LAB, which has been adopted in
ministration of this strain significantly enhanced FMDV- the first human trial with recombinant L. lactis producing IL-
specific humoral and cellular responses and improved the pro- 10 to treat CD (Braat et al. 2006). The thymidylate synthase
duction of IFN rather than IL-4 and IL-10. These results sug- gene of genetically modified L. lactis (LL-Thy12) was re-
gested that LASW1 is a promising immune adjuvant for DNA placed with synthetic mature human IL-10, which is biologi-
vaccination via oral administration and may become a candi- cally contained and avoids systemic side effects (Steidler et al.
date DNA delivery vector. 2003). Meanwhile, many emerging technologies can increase
To increase the efficiency of LAB DNA vaccination, efforts the efficiency of genome recombination, such as CRIPR/
should focus on improving the transfer efficacy of the plasmid Cas9. Most LAB cannot persist in animals or humans for a
from the cytoplasm to the nucleus; furthermore, in vivo ex- long time, but Lactobacillus has longer persistence in the gut
periments are necessary to test the effects of bacterial physi- than other genera, exerting therapeutic effects for a few days
ology and select LAB species that persist for long periods in after administration (Wells and Mercenier 2008). To improve
the gut along with invasin inserted via adaptive plasmids. the design of therapies using LAB as vehicles, the following
issues should be addressed: (i) co-expression of different ther-
apeutic molecules in recombinant LAB; (ii) selection of per-
Conclusion sistent LAB species, especially in the genus Lactobacillus;
(iii) development of appropriate mucosal delivery systems,
The development of LAB expression systems has stimulated including ideal antigen delivery systems and specific adju-
studies aimed at determining the cellular localization of vants; and (iv) design of highly efficient expression systems.
Appl Microbiol Biotechnol

Additional studies and clinical trials will facilitate the use of Bermudez-Humaran LG, Motta JP, Aubry C, Kharrat P, Rous-Martin L,
Sallenave JM, Deraison C, Vergnolle N, Langella P (2015) Serine
recombinant LAB as a novel therapeutic approach to the treat-
protease inhibitors protect better than IL-10 and TGF-beta anti-in-
ment of a wide variety of human diseases in the near future. flammatory cytokines against mouse colitis when delivered by re-
combinant lactococci. Microbial Cell Factories 14:26. doi:10.1186/
Acknowledgments This study was financially supported by the s12934-015-0198-4
Chinese B863^ National Programs for High Technology Research and Braat H, Rottiers P, Hommes DW, Huyghebaert N, Remaut E, Remon JP,
Development (grant no 2011AA10A211), the National Pig Industrial van Deventer SJ, Neirynck S, Peppelenbosch MP, Steidler L (2006)
System (CARS-36-06B), and the Special Fund for Agro-scientific A phase I trial with transgenic bacteria expressing interleukin-10 in
Research in the Public Interest (201203039). Crohn’s disease. Clin Gastroenterol Hepatol 4(6):754–759. doi:10.
1016/j.cgh.2006.03.028
Butler MW, Robertson I, Greene CM, O’Neill SJ, Taggart CC,
Compliance with ethical standards McElvaney NG (2006) Elafin prevents lipopolysaccharide-induced
AP-1 and NF-kappaB activation via an effect on the ubiquitin-
Conflict of interest The authors declare no conflicts of interests. proteasome pathway. J Biol Chem 281(46):34730–34735. doi:10.
1074/jbc.M604844200
Ethical statement This article does not contain any studies with human Cano-Garrido O, Seras-Franzoso J, Garcia-Fruitos E (2015) Lactic acid
participants or animals performed by any of the authors. bacteria: reviewing the potential of a promising delivery live vector
for biomedical purposes. Microb Cell Factories 14:137. doi:10.
1186/s12934-015-0313-6
Cao HP, Wang HN, Yang X, Zhang AY, Li X, Ding MD, Liu ST, Zhang
ZK, Yang F (2013) Lactococcus lactis anchoring avian infectious
bronchitis virus multi-epitope peptide EpiC induced specific im-
References mune responses in chickens. Biosci Biotechnol Biochem 77(7):
1499–1504. doi:10.1271/bbb.130157
Ahmed B, Loos M, Vanrompay D, Cox E (2014) Oral immunization with Chancey CJ, Khanna KV, Seegers JF, Zhang GW, Hildreth J, Langan A,
Lactococcus lactis-expressing EspB induces protective immune re- Markham RB (2006) Lactobacilli-expressed single-chain variable
sponses against Escherichia coli O157:H7 in a murine model of fragment (scFv) specific for intercellular adhesion molecule 1
colonization. Vaccine 32(31):3909–3916. doi:10.1016/j.vaccine. (ICAM-1) blocks cell-associated HIV-1 transmission across a cervi-
2014.05.054 cal epithelial monolayer. J Immunol 176(9):5627–5636
Chen CY, Tsen HY, Lin CL, Lin CK, Chuang LT, Chen CS, Chiang YC
Almada G, Haro C, Vintini E, Medina M (2015) Safety of a nasal vaccine
(2013) Enhancement of the immune response against Salmonella
against Streptococcus pneumoniae using heat-killed Lactobacillus
infection of mice by heat-killed multispecies combinations of lactic
casei as adjuvant. Immunobiology 220(1):109–116. doi:10.1016/j.
acid bacteria. J Med Microbiol 62(Pt 11):1657–1664. doi:10.1099/
imbio.2014.08.013
jmm.0.061010-0
Alvarez B, Krogh-Andersen K, Tellgren-Roth C, Martinez N, Gunaydin
Chowdhury MY, Li R, Kim JH, Park ME, Kim TH, Pathinayake P,
G, Lin Y, Martin MC, Alvarez MA, Hammarstrom L, Marcotte H
Weeratunga P, Song MK, Son HY, Hong SP, Sung MH, Lee JS,
(2015) An exopolysaccharide-deficient mutant of Lactobacillus
Kim CJ (2014) Mucosal vaccination with recombinant
rhamnosus GG efficiently displays a protective llama antibody frag-
Lactobacillus casei-displayed CTA1-conjugated consensus matrix
ment against rotavirus on its surface. Appl Environ Microbiol
protein-2 (sM2) induces broad protection against divergent influen-
81(17):5784–5793. doi:10.1128/AEM.00945-15
za subtypes in BALB/c mice. PLoS One 9(4):e94051. doi:10.1371/
Alvarez-Sieiro P, Martin MC, Redruello B, Del Rio B, Ladero V, Palanski journal.pone.0094051
BA, Khosla C, Fernandez M, Alvarez MA (2014) Generation of Daniel C, Roussel Y, Kleerebezem M, Pot B (2011) Recombinant lactic
food-grade recombinant Lactobacillus casei delivering acid bacteria as mucosal biotherapeutic agents. Trends Biotechnol
Myxococcus xanthus prolyl endopeptidase. Appl Microbiol 29(10):499–508. doi:10.1016/j.tibtech.2011.05.002
Biotechnol 98(15):6689–6700. doi:10.1007/s00253-014-5730-7 Daudel D, Weidinger G, Spreng S (2007) Use of attenuated bacteria as
Andersen KK, Marcotte H, Alvarez B, Boyaka PN, Hammarstrom L delivery vectors for DNA vaccines. Expert Rev Vaccines 6(1):97–
(2011) In situ gastrointestinal protection against anthrax edema toxin 110. doi:10.1586/14760584.6.1.97
by single-chain antibody fragment producing lactobacilli. BMC de Azevedo M, Karczewski J, Lefevre F, Azevedo V, Miyoshi A, Wells
Biotechnol 11:126. doi:10.1186/1472-6750-11-126 JM, Langella P, Chatel JM (2012) In vitro and in vivo characteriza-
Asensi GF, de Sales NF, Dutra FF, Feijo DF, Bozza MT, Ulrich RG, tion of DNA delivery using recombinant Lactococcus lactis express-
Miyoshi A, de Morais K, Azevedo VA, Silva JT, Le Loir Y, ing a mutated form of L. monocytogenes Internalin A. Bmc
Paschoalin VM (2013) Oral immunization with Lactococcus lactis Microbiol 12:299. doi:10.1186/1471-2180-12-299
secreting attenuated recombinant staphylococcal enterotoxin B in- de Azevedo M, Meijerink M, Taverne N, Pereira VB, LeBlanc JG,
duces a protective immune response in a murine model. Microb Cell Azevedo V, Miyoshi A, Langella P, Wells JM, Chatel JM (2015)
Factories 12:32. doi:10.1186/1475-2859-12-32 Recombinant invasive Lactococcus lactis can transfer DNA vac-
Bautista-Garfias CR, Lozano AR, Martinez CR, Martinez JA, Millan JV, cines either directly to dendritic cells or across an epithelial cell
Garcia GR, Castaneda-Arriola R, Aguilar-Figueroa BR (2015) Co- monolayer. Vaccine 33(38):4807–4812. doi:10.1016/j.vaccine.
immunization of cattle with a vaccine against babesiosis and 2015.07.077
Lactobacillus casei increases specific IgG1 levels to Babesia bovis de LeBlanc AD, del Carmen S, Chatel JM, Miyoshi A, Azevedo V,
and B. bigemina. Parasitol Int 64(5):319–323. doi:10.1016/j.parint. Langella P, Bermudez-Humaran LG, LeBlanc JG (2015) Current
2015.04.005 review of genetically modified lactic acid bacteria for the prevention
Bermudez-Humaran LG, Aubry C, Motta JP, Deraison C, Steidler L, and treatment of colitis using murine models. Gastroenterol Res
Vergnolle N, Chatel JM, Langella P (2013) Engineering lactococci Pract 2015:146972. doi:10.1155/2015/146972
and lactobacilli for human health. Curr Opin Microbiol 16(3):278– de Vrese M, Schrezenmeir J (2008) Probiotics, prebiotics, and synbiotics.
283. doi:10.1016/j.mib.2013.06.002 Adv Biochem Eng Biotechnol 111:1–66. doi:10.1007/10_2008_097
Appl Microbiol Biotechnol

del Carmen S, Rosique RM, Saraiva T, Zurita-Turk M, Miyoshi A, against anti-transmissible gastroenteritis virus. Appl Microbiol
Azevedo V, de LeBlanc AD, Langella P, Bermudez-Humaran LG, Biotechnol 98(19):8301–8312. doi:10.1007/s00253-014-5893-2
LeBlanc JG (2014) Protective effects of Lactococci strains deliver- Jin L, Zhu AH, Wang Y, Lu Y, Liu JJ (2010) HSP65 serves as an immu-
ing either IL-10 protein or cDNA in a TNBS-induced chronic colitis nogenic carrier for a diabetogenic peptide P277 inducing anti-
model. J Clin Gastroenterol 48:S12–S17 inflammatory immune response in NOD mice by nasal administra-
Dimitrijevic R, Ivanovic N, Mathiesen G, Petrusic V, Zivkovic I, tion. Vaccine 28(19):3312–3317. doi:10.1016/j.vaccine.2010.02.100
Djordjevic B, Dimitrijevic L (2014) Effects of Lactobacillus Kawana K, Adachi K, Kojima S, Taguchi A, Tomio K, Yamashita A,
rhamnosus LA68 on the immune system of C57BL/6 mice upon Nishida H, Nagasaka K, Arimoto T, Yokoyama T, Wada-Hiraike O,
oral administration. J Dairy Res 81(2):202–207. doi:10.1017/ Oda K, Sewaki T, Osuga Y, Fujii T (2014) Oral vaccination against
S0022029914000028 HPV E7 for treatment of cervical intraepithelial neoplasia grade 3
Gao S, Li D, Liu Y, Zha E, Zhou T, Yue X (2015) Oral immunization with (CIN3) elicits E7-specific mucosal immunity in the cervix of CIN3
recombinant hepatitis E virus antigen displayed on the Lactococcus patients. Vaccine 32(47):6233–6239. doi:10.1016/j.vaccine.2014.
lactis surface enhances ORF2-specific mucosal and systemic im- 09.020
mune responses in mice. Int Immunopharmacol 24(1):140–145. Kruger C, Hu Y, Pan Q, Marcotte H, Hultberg A, Delwar D, van Dalen
doi:10.1016/j.intimp.2014.10.032 PJ, Pouwels PH, Leer RJ, Kelly CG, van Dollenweerd C, Ma JK,
Gareau MG, Sherman PM, Walker WA (2010) Probiotics and the gut Hammarstrom L (2002) In situ delivery of passive immunity by
microbiota in intestinal health and disease. Nat Rev Gastroenterol lactobacilli producing single-chain antibodies. Nat Biotechnol
Hepatol 7(9):503–514. doi:10.1038/nrgastro.2010.117 20(7):702–706. doi:10.1038/nbt0702-702
Geriletu XR, Jia H, Terkawi MA, Xuan X, Zhang H (2011) LeBlanc JG, Aubry C, Cortes-Perez NG, de LeBlanc AD, Vergnolle N,
Immunogenicity of orally administrated recombinant Lactobacillus Langella P, Azevedo V, Chatel JM, Miyoshi A, Bermudez-Humaran
casei Zhang expressing cryptosporidium parvum surface adhesion LG (2013) Mucosal targeting of therapeutic molecules using genet-
protein P23 in mice. Curr Microbiol 62(5):1573–1580. doi:10.1007/ ically modified lactic acid bacteria: an update. FEMS Microbiol Lett
s00284-011-9894-4 344(1):1–9. doi:10.1111/1574-6968.12159
Grillot-Courvalin C, Goussard S, Huetz F, Ojcius DM, Courvalin P (1998) Lei H, Peng X, Ouyang J, Zhao D, Jiao H, Shu H, Ge X (2015a)
Functional gene transfer from intracellular bacteria to mammalian Intranasal immunization of recombinant Lactococcus lactis induces
cells. Nat Biotechnol 16(9):862–866. doi:10.1038/Nbt0998-862 protection against H5N1 virus in ferrets. Virus Res 196:56–59. doi:
Gunaydin G, Zhang R, Hammarstrom L, Marcotte H (2014) Engineered 10.1016/j.virusres.2014.11.009
Lactobacillus rhamnosus GG expressing IgG-binding domains of Lei H, Peng X, Jiao H, Zhao D, Ouyang J (2015b) Broadly protective
protein G: capture of hyperimmune bovine colostrum antibodies and immunity against divergent influenza viruses by oral co-
protection against diarrhea in a mouse pup rotavirus infection model. administration of Lactococcus lactis expressing nucleoprotein
Vaccine 32(4):470–477. doi:10.1016/j.vaccine.2013.11.057 adjuvanted with cholera toxin B subunit in mice. Microb Cell
Guo S, Yan W, McDonough SP, Lin N, Wu KJ, He H, Xiang H, Yang M, Factories 14:111. doi:10.1186/s12934-015-0287-4
Moreira MA, Chang YF (2015) The recombinant Lactococcus lactis Lei H, Peng X, Ouyang J, Zhao D, Jiao H, Shu H, Ge X (2015c)
oral vaccine induces protection against C. Difficile spore challenge Protective immunity against influenza H5N1 virus challenge in
in a mouse model. Vaccine 33(13):1586–1595. doi:10.1016/j. chickens by oral administration of recombinant Lactococcus lactis
vaccine.2015.02.006 expressing neuraminidase. BMC Vet Res 11:85. doi:10.1186/
Hanniffy SB, Carter AT, Hitchin E, Wells JM (2007) Mucosal delivery of s12917-015-0399-4
a pneumococcal vaccine using Lactococcus lactis affords protection Lei H, Peng X, Shu H, Zhao D (2015d) Intranasal immunization with live
against respiratory infection. J Infect Dis 195(2):185–193. doi:10. recombinant Lactococcus lactis combined with heat-labile toxin B
1086/509807 subunit protects chickens from highly pathogenic avian influenza
Henriksen PA, Hitt M, Xing Z, Wang J, Haslett C, Riemersma RA, Webb H5N1 virus. J Med Virol 87(1):39–44. doi:10.1002/jmv.23983
DJ, Kotelevtsev YV, Sallenave JM (2004) Adenoviral gene delivery Lei H, Peng X, Zhao D, Ouyang J, Jiao H, Shu H, Ge X (2015e)
of elafin and secretory leukocyte protease inhibitor attenuates NF- Lactococcus lactis displayed neuraminidase confers cross protective
kappa B-dependent inflammatory responses of human endothelial immunity against influenza a viruses in mice. Virology 476:189–
cells and macrophages to atherogenic stimuli. J Immunol 172(7): 195. doi:10.1016/j.virol.2014.12.017
4535–4544 Li YG, Tian FL, Gao FS, Tang XS, Xia C (2007) Immune responses
Hiramatsu Y, Yamamoto M, Satho T, Irie K, Kai A, Uyeda S, Fukumitsu generated by Lactobacillus as a carrier in DNA immunization
Y, Toda A, Miyata T, Miake F, Arakawa T, Kashige N (2014) against foot-and-mouth disease virus. Vaccine 25(5):902–911. doi:
Recombinant fusion protein of cholera toxin B subunit with 10.1016/j.vaccine.2006.09.034
Y VA D s e c r e t e d b y L a c t o b a c i l l u s c a s e i i n h i b i t s Li Y, Li X, Liu H, Zhuang S, Yang J, Zhang F (2014a) Intranasal immu-
lipopolysaccharide-induced caspase-1 activation and subsequent nization with recombinant Lactococci carrying human papillomavi-
IL-1 beta secretion in Caco-2 cells. Bmc Biotechnol 14:38. doi:10. rus E7 protein and mouse interleukin-12 DNA induces E7-specific
1186/1472-6750-14-38 antitumor effects in C57BL/6 mice. Oncol Lett 7(2):576–582. doi:
Holmgren J, Czerkinsky C (2005) Mucosal immunity and vaccines. Nat 10.3892/ol.2013.1743
Med 11(4 Suppl):S45–S53. doi:10.1038/nm1213 Li X, Xing Y, Guo L, Lv X, Song H, Xi T (2014b) Oral immunization
Hongying F, Xianbo W, Fang Y, Yang B, Beiguo L (2014) Oral immu- with recombinant Lactococcus lactis delivering a multi-epitope an-
nization with recombinant Lactobacillus acidophilus expressing the tigen CTB-UE attenuates Helicobacter pylori infection in mice.
adhesin Hp0410 of Helicobacter pylori induces mucosal and sys- Pathog Dis 72(1):78–86. doi:10.1111/2049-632X.12173
temic immune responses. Clin Vaccine Immunol 21(2):126–132. Li HS, Piao DC, Jiang T, Bok JD, Cho CS, Lee YS, Kang SK, Choi YJ
doi:10.1128/CVI.00434-13 (2015a) Recombinant interleukin 6 with M cell-targeting moiety
Hu KF, Lovgren-Bengtsson K, Morein B (2001) Immunostimulating produced in Lactococcus lactis IL1403 as a potent mucosal adjuvant
complexes (ISCOMs) for nasal vaccination. Adv Drug Deliv Rev for peroral immunization. Vaccine 33(16):1959–1967. doi:10.1016/
51(1–3):149–159. doi:10.1016/S0169-409x(01)00165-X j.vaccine.2015.02.061
Jiang XP, Yu ML, Qiao XY, Liu M, Tang LJ, Jiang YP, Cui W, Li YJ Li R, Chowdhury MY, Kim JH, Kim TH, Pathinayake P, Koo WS, Park
(2014) Up-regulation of MDP and tuftsin gene expression in Th1 ME, Yoon JE, Roh JB, Hong SP, Sung MH, Lee JS, Kim CJ (2015b)
and Th17 cells as an adjuvant for an oral Lactobacillus casei vaccine Mucosally administered Lactobacillus surface-displayed influenza
Appl Microbiol Biotechnol

antigens (sM2 and HA2) with cholera toxin subunit A1 (CTA1) Ohkouchi K, Kawamoto S, Tatsugawa K, Yoshikawa N, Takaoka Y,
induce broadly protective immune responses against divergent in- Miyauchi S, Aki T, Yamashita M, Murooka Y, Ono K (2012)
fluenza subtypes. Vet Microbiol 179(3–4):250–263. doi:10.1016/j. Prophylactic effect of Lactobacillus oral vaccine expressing a
vetmic.2015.07.020 Japanese cedar pollen allergen. J Biosci Bioeng 113(4):536–541.
Liu FN, Wen K, Li GH, Yang XD, Kocher J, Bui T, Jones D, Pelzer K, doi:10.1016/j.jbiosc.2011.11.025
Clark-Deener S, Yuan LJ (2014a) Dual functions of Lactobacillus Pabreja S, Garg T, Rath G, Goyal AK (2014) Mucosal vaccination against
acidophilus NCFM as protection against rotavirus diarrhea. J Pediatr tuberculosis using Ag85A-loaded immunostimulating complexes.
Gastroenterol Nutr 58(2):169–176. doi:10.1097/Mpg. Artif Cells Nanomed Biotechnol:1–8 doi:10.3109/
0000000000000197 21691401.2014.966195
Liu JK, Wei CH, Hou XL, Yu LY (2014b) Passive protection of mice Pan L, Zhang Z, Lv J, Zhou P, Hu W, Fang Y, Chen H, Liu X, Shao J,
pups through oral or intranasal immunization of dams with recom- Zhao F, Ding Y, Lin T, Chang H, Zhang J, Zhang Y, Wang Y (2014)
binant Lactobacillus casei vaccine against ETEC F41. Res Vet Sci Induction of mucosal immune responses and protection of cattle
96(2):283–287. doi:10.1016/j.rysc.2014.01.010 against direct-contact challenge by intranasal delivery with foot-
Luo XR, Herold KC, Miller SD (2010) Immunotherapy of type 1 diabe- and-mouth disease virus antigen mediated by nanoparticles. Int J
tes: where are we and where should we be going? Immunity 32(4): Nanomedicine 9:5603–5618. doi:10.2147/IJN.S72318
488–499. doi:10.1016/j.immuni.2010.04.002 Pant N, Hultberg A, Zhao Y, Svensson L, Pan-Hammarstrom Q, Johansen
Ma D, Gao M, Dalloul RA, Ge J, Ma C, Li J (2013) Protective effects of K, Pouwels PH, Ruggeri FM, Hermans P, Frenken L, Boren T,
oral immunization with live Lactococcus lactis expressing Eimeria Marcotte H, Hammarstrom L (2006) Lactobacilli expressing vari-
tenella 3-1E protein. Parasitol Res 112(12):4161–4167. doi:10. able domain of llama heavy-chain antibody fragments (lactobodies)
1007/s00436-013-3607-9 confer protection against rotavirus-induced diarrhea. J Infect Dis
Ma YJ, Liu JJ, Hou J, Dong YK, Lu Y, Jin L, Cao R, Li TM, Wu J (2014) 194(11):1580–1588. doi:10.1086/508747
Oral administration of recombinant Lactococcus lactis expressing Petrovsky N (2010) Immunomodulation with microbial vaccines to pre-
HSP65 and tandemly repeated P277 reduces the incidence of type vent type 1 diabetes mellitus. Nat Rev Endocrinol 6(3):131–138.
I diabetes in non-obese diabetic mice. Plos One 9(8):e105701. doi: doi:10.1038/nrendo.2009.273
10.1371/journal.pone.0105701 Pontes DS, de Azevedo MS, Chatel JM, Langella P, Azevedo V, Miyoshi
Macpherson AJ, Koller Y, Mccoy KD (2015) The bilateral responsive- A (2011) Lactococcus lactis as a live vector: heterologous protein
ness between intestinal microbes and IgA. Trends Immunol 36(8): production and DNA delivery systems. Protein Expr Purif 79(2):
460–470. doi:10.1016/j.it.2015.06.006 165–175. doi:10.1016/j.pep.2011.06.005
Marcotte H, Koll-Klais P, Hultberg A, Zhao Y, Gmur R, Mandar R,
Ribelles P, Benbouziane B, Langella P, Suarez JE, Bermudez-Humaran
Mikelsaar M, Hammarstrom L (2006) Expression of single-chain
LG, Riazi A (2013) Protection against human papillomavirus type
antibody against RgpA protease of Porphyromonas gingivalis in
16-induced tumors in mice using non-genetically modified lactic
Lactobacillus. J Appl Microbiol 100(2):256–263. doi:10.1111/j.
acid bacteria displaying E7 antigen at its surface. Appl Microbiol
1365-2672.2005.02786.x
Biotechnol 97(3):1231–1239. doi:10.1007/s00253-012-4575-1
Martin R, Chain F, Miquel S, Natividad JM, Sokol H, Verdu EF, Langella
Robert S, Gysemans C, Takiishi T, Korf H, Spagnuolo I, Sebastiani G,
P, Bermudez-Humaran LG (2014) Effects in the use of a genetically
Van Huynegem K, Steidler L, Caluwaerts S, Demetter P, Wasserfall
engineered strain of Lactococcus lactis delivering in situ IL-10 as a
CH, Atkinson MA, Dotta F, Rottiers P, Van Belle TL, Mathieu C
therapy to treat low-grade colon inflammation. Human Vaccines
(2014) Oral delivery of glutamic acid decarboxylase (GAD)-65 and
Immunotherapeutics 10(6):1611–1621. doi:10.4161/hv.28549
IL10 by Lactococcus lactis reverses diabetes in recent-onset NOD
Minic R, Gavrovic-Jankulovic M, Petrusic V, Zivkovic I, Eijsink VG,
mice. Diabetes 63(8):2876–2887. doi:10.2337/db13-1236
Dimitrijevic L, Mathiesen G (2015) Effects of orally applied Fes
p1-displaying L. plantarum WCFS1 on Fes p1 induced allergy in Robert S, Van Huynegem K, Gysemans C, Mathieu C, Rottiers P, Steidler
mice. J Biotechnol 199:23–28. doi:10.1016/j.jbiotec.2015.01.028 L (2015) Trimming of two major type 1 diabetes driving antigens,
Mobergslien A, Vasovic V, Mathiesen G, Fredriksen L, Westby P, Eijsink GAD65 and IA-2, allows for successful expression in Lactococcus
VG, Peng Q, Sioud M (2015) Recombinant Lactobacillus lactis. Benef Microbes 6(4):591–601. doi:10.3920/BM2014.0083
plantarum induces immune responses to cancer testis antigen NY- Robinson K, Chamberlain LM, Schofield KM, Wells JM, Le Page RW
ESO-1 and maturation of dendritic cells. Hum Vaccin Immunother: (1997) Oral vaccination of mice against tetanus with recombinant
1–10 doi:10.1080/21645515.2015.1056952 Lactococcus lactis. Nat Biotechnol 15(7):653–657. doi:10.1038/
Motta JP, Bermudez-Humaran LG, Deraison C, Martin L, Rolland C, nbt0797-653
Rousset P, Boue J, Dietrich G, Chapman K, Kharrat P, Vinel JP, Roland KL, Tinge SA, Killeen KP, Kochi SK (2005) Recent advances in
Alric L, Mas E, Sallenave JM, Langella P, Vergnolle N (2012) the development of live, attenuated bacterial vectors. Curr Opin Mol
Food-grade bacteria expressing elafin protect against inflammation Ther 7(1):62–72
and restore colon homeostasis. Sci Transl Med 4(158):158ra144. Saraiva TD, Morais K, Pereira VB, de Azevedo M, Rocha CS, Prosperi
doi:10.1126/scitranslmed.3004212 CC, Gomes-Santos AC, Bermudez-Humaran L, Faria AM, Blottiere
Nguyen T, Nguyen M, Geiger B, Mathiesen G, Eijsink VGH, Peterbauer HM, Langella P, Miyoshi A, de LeBlanc AM, LeBlanc JG, Azevedo
CK, Haltrich D, Nguyen TH (2015) Heterologous expression of a V (2015) Milk fermented with a 15-lipoxygenase-1-producing
recombinant lactobacillal beta-galactosidase in Lactobacillus Lactococcus lactis alleviates symptoms of colitis in a murine model.
plantarum: effect of different parameters on the sakacin P-based Curr Pharm Biotechnol 16(5):424–429
expression system. Microbial Cell Factories 14:30. doi:10.1186/ Schotte L, Steidler L, Vandekerckhove J, Remaut E (2000) Secretion of
s12934-015-0214-8 biologically active murine interleukin-10 by Lactococcus lactis.
Norton PM, Wells JM, Brown HW, Macpherson AM, Le Page RW Enzym Microb Technol 27(10):761–765
(1997) Protection against tetanus toxin in mice nasally immunized Shaw DM, Gaerthe B, Leer RJ, Van Der Stap JG, Smittenaar C, Heijne
with recombinant Lactococcus lactis expressing tetanus toxin frag- Den Bak-Glashouwer M, Thole JE, Tielen FJ, Pouwels PH,
ment C. Vaccine 15(6–7):616–619 Havenith CE (2000) Engineering the microflora to vaccinate the
Nouaille S, Ribeiro LA, Miyoshi A, Pontes D, Le Loir Y, Oliveira SC, mucosa: serum immunoglobulin G responses and activated draining
Langella P, Azevedo V (2003) Heterologous protein production and cervical lymph nodes following mucosal application of tetanus toxin
delivery systems for Lactococcus lactis. Genet Mol Res 2(1):102–111 fragment C-expressing lactobacilli. Immunology 100(4):510–518
Appl Microbiol Biotechnol

Shi SH, Yang WT, Yang GL, Cong YL, Huang HB, Wang Q, Cai RP, Ye Wang G, Pan L, Zhang YG, Wang YL, Zhang ZW, Lu JL, Zhou P, Fang
LP, Hu JT, Zhou JY, Wang CF, Li Y (2014) Immunoprotection YZ, Jiang ST (2011) Intranasal delivery of cationic PLGA
against influenza virus H9N2 by the oral administration of recom- nano/microparticles-loaded FMDV DNA vaccine encoding IL-6
binant Lactobacillus plantarumNC8 expressing hemagglutinin in elicited protective immunity against FMDV challenge. Plos One
BALB/c mice. Virology 464-465:166–176. doi:10.1016/j.virol. 6(11):e27605. doi:10.1371/journal.pone.0027605
2014.07.011 Wang Z, Yu Q, Gao J, Yang Q (2012) Mucosal and systemic immune
Shibasaki S, Karasaki M, Tafuku S, Aoki W, Sewaki T, Ueda M (2014) responses induced by recombinant Lactobacillus spp. expressing the
Oral immunization against candidiasis using Lactobacillus casei hemagglutinin of the avian influenza virus H5N1. Clin Vaccine
displaying Enolase 1 from Candida albicans. Sci Pharm 82(3): Immunol 19(2):174–179. doi:10.1128/CVI.05618-11
697–708. doi:10.3797/scipharm.1404-07 Wang X, Zhang X, Zhou D, Yang R (2013) Live-attenuated Yersinia
Smelt MJ, de Haan BJ, Bron PA, van Swam I, Meijerink M, Wells JM, pestis vaccines. Expert Rev Vaccines 12(6):677–686. doi:10.1586/
Faas MM, de Vos P (2012) L. plantarum, L. salivarius, and L. lactis erv.13.42
attenuate Th2 responses and increase Treg frequencies in healthy Wang DY, Xu SY, Lin Y, Fang ZF, Che LQ, Xue B, Wu D (2014a)
mice in a strain dependent manner. Plos One 7(10):e47244. doi:10. Recombinant porcine epidermal growth factor-secreting Lactococcus
1371/journal.pone.0047244 lactis promotes the growth performance of early-weaned piglets. Bmc
Song JA, Kim HJ, Hong SK, Lee DH, Lee SW, Song CS, Kim KT, Choi IS, Vet Res 10:171. doi:10.1186/S12917-014-0171-1
Lee JB, Park SY (2014) Oral intake of Lactobacillus rhamnosus M21 Wang M, Gao Z, Zhang Z, Pan L, Zhang Y (2014b) Roles of M cells in
enhances the survival rate of mice lethally infected with influenza infection and mucosal vaccines. Hum Vaccin Immunother 10(12):
virus. J Microbiol Immunol Infect. doi:10.1016/j.jmii.2014.07.011 3544–3551. doi:10.4161/hv.36174
Steidler L, Hans W, Schotte L, Neirynck S, Obermeier F, Falk W, Fiers W, Wang ZH, Cao XH, Du XG, Feng HB, Di W, He S, Zeng XY (2014c)
Remaut E (2000) Treatment of murine colitis by Lactococcus lactis Mucosal and systemic immunity in mice after intranasal immunization
secreting interleukin-10. Science 289(5483):1352–1355 with recombinant Lactococcus lactis expressing ORF6 of PRRSV.
Steidler L, Neirynck S, Huyghebaert N, Snoeck V, Vermeire A, Cell Immunol 287(2):69–73. doi:10.1016/j.cellimm.2013.12.004
Goddeeris B, Cox E, Remon JP, Remaut E (2003) Biological con- Wang M, Pan L, Zhou P, Lv J, Zhang Z, Wang Y, Zhang Y (2015)
tainment of genetically modified Lactococcus lactis for intestinal Protection against foot-and-mouth disease virus in Guinea pigs via
delivery of human interleukin 10. Nat Biotechnol 21(7):785–789. oral Administration of Recombinant Lactobacillus plantarum ex-
doi:10.1038/nbt840 pressing VP1. PLoS One 10(12):e0143750. doi:10.1371/journal.
Su J, Li J, Zheng H, You Y, Luo X, Li Y, Li X, Ma X, Li J, Dou Y, Cai X pone.0143750
(2014) Adjuvant effects of L. acidophilus LW1 on immune responses
Wells JM, Mercenier A (2008) Mucosal delivery of therapeutic and pro-
to the foot-and-mouth disease virus DNA vaccine in mice. PLoS One
phylactic molecules using lactic acid bacteria. Nat Rev Microbiol
9(8):e104446. doi:10.1371/journal.pone.0104446
6(5):349–362. doi:10.1038/nrmicro1840
Takiishi T, Korf H, Van Belle TL, Robert S, Grieco FA, Caluwaerts S,
Wells JM, Wilson PW, Norton PM, Gasson MJ, Le Page RW (1993)
Galleri L, Spagnuolo I, Steidler L, Van Huynegem K, Demetter P,
Lactococcus lactis: high-level expression of tetanus toxin fragment C
Wasserfall C, Atkinson MA, Dotta F, Rottiers P, Gysemans C,
and protection against lethal challenge. Mol Microbiol 8(6):1155–1162
Mathieu C (2012) Reversal of autoimmune diabetes by restoration
of antigen-specific tolerance using genetically modified Wendorf J, Chesko J, Kazzaz J, Ugozzoli M, Vajdy M, O’Hagan D, Singh
Lactococcus lactis in mice. J Clin Investig 122(5):1717–1725. doi: M (2008) A comparison of anionic nanoparticles and microparticles
10.1172/JCI60530 as vaccine delivery systems. Human Vaccines 4(1):44–49
Tsai YT, Cheng PC, Pan TM (2012) The immunomodulatory effects of Xu S, Wang D, Zhang P, Lin Y, Fang Z, Che L, Wu D (2015a) Oral
lactic acid bacteria for improving immune functions and benefits. administration of Lactococcus lactis-expressed recombinant porcine
Appl Microbiol Biotechnol 96(4):853–862. doi:10.1007/s00253- epidermal growth factor stimulates the development and promotes
012-4407-3 the health of small intestines in early-weaned piglets. J Appl
Van Belle TL, Coppieters KT, Von Herrath MG (2011) Type 1 diabetes: Microbiol 119(1):225–235. doi:10.1111/jam.12833
etiology, immunology, and therapeutic strategies. Physiol Rev Xu YG, Guan XT, Liu ZM, Tian CY, Cui LC (2015b) Immunogenicity in
91(1):79–118. doi:10.1152/physrev.00003.2010 swine of orally administered recombinant Lactobacillus plantarum
Vandenbroucke K, Hans W, Van Huysse J, Neirynck S, Demetter P, expressing classical swine fever virus E2 protein in conjunction with
Remaut E, Rottiers P, Steidler L (2004) Active delivery of trefoil thymosin alpha-1 as an adjuvant. Appl Environ Microbiol 81(11):
factors by genetically modified Lactococcus lactis prevents and 3745–3752. doi:10.1128/Aem.00127-15
heals acute colitis in mice. Gastroenterology 127(2):502–513 Yang Y, Kang Z, Zhou J, Chen J, Du G (2015) High-level expression and
Veloso TR, Mancini S, Giddey M, Vouillamoz J, Que YA, Moreillon P, characterization of recombinant acid urease for enzymatic degrada-
Entenza JM (2015) Vaccination against Staphylococcus aureus ex- tion of urea in rice wine. Appl Microbiol Biotechnol 99(1):301–308.
perimental endocarditis using recombinant Lactococcus lactis ex- doi:10.1007/s00253-014-5916-z
pressing ClfA or FnbpA. Vaccine 33(30):3512–3517. doi:10.1016/ Zelmer A, Krusch S, Koschinski A, Rohde M, Repp H, Chakraborty T,
j.vaccine.2015.05.060 Weiss S (2005) Functional transfer of eukaryotic expression plas-
Vintini EO, Medina MS (2011) Host immunity in the protective response mids to mammalian cells by Listeria monocytogenes: a mechanistic
to nasal immunization with a pneumococcal antigen associated to approach. J Gene Med 7(8):1097–1112. doi:10.1002/jgm.764
live and heat-killed Lactobacillus casei. BMC Immunol 12:46. doi: Zhang HX, Qiu YY, Zhao YH, Liu XT, Liu M, Yu AL (2014)
10.1186/1471-2172-12-46 Immunogenicity of oral vaccination with Lactococcus lactis derived
Waki N, Matsumoto M, Fukui Y, Suganuma H (2014) Effects of probiotic vaccine candidate antigen (UreB) of Helicobacter pylon fused with
Lactobacillus brevis KB290 on incidence of influenza infection the human interleukin 2 as adjuvant. Mol Cell Probes 28(1):25–30.
among schoolchildren: an open-label pilot study. Lett Appl doi:10.1016/j.mcp.2013.08.003
Microbiol 59(6):565–571. doi:10.1111/lam.12340 Zhou H, Gao Y, Gao G, Lou Y (2015) Oral Administration of
Walters WA, Xu Z, Knight R (2014) Meta-analyses of human gut mi- Recombinant Lactococcus lactis expressing the Cellulase Gene in-
crobes associated with obesity and IBD. FEBS Lett 588(22):4223– creases digestibility of fiber in geese. Curr Microbiol. doi:10.1007/
4233. doi:10.1016/j.febslet.2014.09.039 s00284-015-0904-9

You might also like