Next Article in Journal
Optimizing the Extraction Process of Value-Added Products from Olive Cake Using Neuro-Fuzzy Models
Previous Article in Journal
Current Status and Economic Analysis of Green Hydrogen Energy Industry Chain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Review of the Potential of Probiotics in Disease Treatment: Mechanisms, Engineering, and Applications

1
Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
2
Department of Chemical Engineering, University of Waterloo, 200 University Avenue West Waterloo, Waterloo, ON N2L 3G1, Canada
*
Authors to whom correspondence should be addressed.
Processes 2024, 12(2), 316; https://doi.org/10.3390/pr12020316
Submission received: 10 January 2024 / Revised: 23 January 2024 / Accepted: 29 January 2024 / Published: 2 February 2024
(This article belongs to the Section Biological Processes and Systems)

Abstract

:
Probiotics, living microorganisms with demonstrated health benefits when administered in sufficient quantities, have a rich history as dietary supplements to benefit human health. Recently, understanding of their mechanisms in the gastrointestinal tract has prompted exploration of probiotics in treating human diseases. However, the effective and precise delivery of probiotics remains a significant challenge in therapeutic applications. Here, we review the mechanisms of action of probiotics in human health and the most advanced strategies for efficient probiotic delivery. We also discuss the potential applications of engineered probiotics in disease treatment. This review contributes insights into the evolving landscape of probiotic research for therapeutic applications.

1. Introduction

Probiotics have played a pivotal role in advancing human health, from their initial discovery to their contemporary integration into healthcare practices. The foundational understanding of probiotics can be attributed to Élie Metchnikoff, recognized as the progenitor of Lactobacillus and a Nobel laureate for his pioneering work on phagocytes [1]. In 2001, the United Nations’ Food and Agriculture Organization (FAO) and the World Health Organization (WHO) formally defined probiotics as “live microorganisms which, when administered in adequate amounts, confer a health benefit on the host” [2]. This definition underscores the transformative potential of probiotics in promoting host well-being by modulating microbial balance, particularly within the gastrointestinal tract.
In recent years, the utilization of probiotics in disease treatment has developed into a dynamic and multidimensional field. Probiotics exhibit considerable promise in addressing disorders associated with dysbiosis, encompassing allergic, digestive, and metabolic conditions, as well as cancers and cardiovascular diseases [3]. The mechanisms underlying these functions are diverse and heterogeneous and have not yet been fully elucidated. Moreover, the delivery of adequate probiotics into the gastrointestinal tract poses several challenges, stemming from the harsh conditions of the digestive system. Advancements in chemistry, biology, and research on membrane vesicles in the last decade have significantly enhanced the efficiency and targeted delivery of engineered probiotics, thereby extending their applicability to the treatment of a broader spectrum of diseases [4,5,6].
This review comprehensively explores the intrinsic mechanisms through which probiotics influence overall host health. Subsequently, we discuss probiotic-based delivery systems, offering an overview of the predominant current delivery methods. These methods encompass not only chemical engineering modifications, but also genetic engineering strategies designed to augment the efficiency of probiotic delivery to crucial anatomical sites. The innovative application of membrane vesicles derived from probiotics is also discussed. Finally, this review outlines the applications of functional probiotics in disease treatment, discusses the outcomes of probiotic interventions in preclinical experiments, and offers insights into the prospective role of probiotic therapy in future clinical applications.

2. Probiotic Mechanisms of Action for Disease Treatment

As of now, the food and drug regulatory agencies in most countries, excluding the European Food Safety Authority, recognize the positive impact of probiotics on human health. However, they are typically recommended as dietary supplements rather than used as pharmaceutical interventions for disease treatment [7]. The accessibility and nonprescriptive nature of dietary supplements contribute to the widespread adoption of probiotics. However, our limited understanding of probiotic action mechanisms constrains their effective use in disease treatment. Recognizing the imperative to comprehend these mechanisms becomes essential, as it has the potential to enhance the engineering of probiotics for more effective disease treatment, optimizing their impact on human health through informed and targeted applications. This encouraged us to initially explore and outline the primary mechanisms of probiotic action (Figure 1): (1) production of bioactive compounds; (2) regulation of the gastrointestinal microbiome; and (3) modulation of the immune and nervous systems.

2.1. Production of Bioactive Compounds

Probiotics generate a diverse range of bioactive compounds, including vitamins, short-chain fatty acids (SCFAs), amino acids, peptides, enzymes, and exopolysaccharides (EPSs), thereby exhibiting beneficial health effects on the host [8]. For example, probiotic bacteria are able to produce B-group vitamins (B6, B12, folate, and thiamine) and K-group vitamins (K1 and K2), effectively addressing vitamin deficiencies and supporting the treatment of inflammatory bowel disease (IBD) and carcinogenesis [9]. SCFAs (mainly lactate, acetate, and butyrate) are essential metabolic products of probiotics. Zhang et al. discovered that Lactobacillus plantarum-derived indole-3-lactic acid enhanced the antitumor immunity of CD8+ T cells through epigenetic regulation, thereby ameliorating colorectal tumorigenesis [10]. SCFAs play a crucial role in various physiological functions, including energy expenditure, mucosal barrier maintenance, and immune system modulation. Administering probiotics producing SCFAs holds promise as an adjunctive therapy for conditions like obesity, diabetes, Alzheimer’s, tumors, and cancer [11,12]. Probiotics also secrete specific amino acids, small peptides, or proteins to fight pathogens and to activate host immunity. Recently, Zhou et al. genetically engineered E. coli Nissle 1917 (ECN) to express antioxidant enzymes (catalase and superoxide dismutase) for the treatment of IBD [13]. The EPSs produced by probiotics have also been reported along with antitumor and anti-inflammatory activities in melanoma mice [14]. Finally, Piewngam et al. discovered that fengycin, a peptide secreted by Bacillus subtilis, competitively inhibits the AgrC receptor in the quorum-sensing system of Staphylococcus aureus, thereby suppressing the proliferation of S. aureus [15]. These bioactive compounds can exert their effects by directly interacting with host cells. However, the majority of these compounds influence the host by interacting with other bacteria and modulating the composition of microbiota.

2.2. Regulation of Gastrointestinal Microbiota

The human gastrointestinal (GIT) system, crucial for health, accommodates more than 1014 microorganisms of over 1000 species. Changes in the GIT microbial composition have been associated with a wide spectrum of human diseases, including cancer, obesity, inflammatory disorders, and even mental illness and neurological disorders [16,17].
Probiotics contribute to the regulation of the gastrointestinal microbiota through diverse mechanisms. For example, the probiotics of Lactobacillus, Bacillus, and Saccharomyces have shown potential in reducing the Firmicutes/Bacteroidetes (F/B) ratio which indicates therapy applications in obesity and IBD [18]. Osbelt et al. observed nutrient competition, where commensal Klebsiella oxytoca strains inhibited the growth of multidrug-resistant Klebsiella pneumoniae by outcompeting it through beta-glucoside utilization, thereby reducing the risk of bloodstream infections [19]. Zou et al. found that sortase A-anchored SDPs in Lactobacillus gasseri Kx110A1 competitively exclude Helicobacter pylori through steric hindrance, reducing the initial colonization of H. pylori in mouse stomachs, a key risk factor for stomach cancer development [20]. Furthermore, Zhou et al. discovered that the administration of engineered ECN enhances the abundance of the beneficial innate probiotics, Lachnospiraceae_NK4A136 and Odoribacter, thereby promoting intestinal homeostasis and mitigating IBD [13]. These studies highlight the profound impact of probiotics on gut microbiota composition, prompting a deeper exploration of their effects on modulating the immune and nervous systems given their close interactions [21].

2.3. Modulation of Immune and Nervous Systems

Probiotics exhibit significant effects in modulating both immune and nervous systems, offering promising avenues for the therapy of related diseases.
Probiotics exhibit the capability to bolster both innate and adaptive immune responses by interacting with intestinal epithelial cells (IECs), immune cells, or other gastrointestinal microbiota. They reinforce the intestinal barrier by increasing mucins and tight junction proteins and supporting the function of goblet and Paneth cells. Probiotics also stimulate the production of various cytokines and chemokines through toll-like receptors, initiating a cascade of signaling events. These cytokines, in turn, facilitate the stimulation of adaptive immune responses and establish a network of communication among different immune cells. La Fata et al. [22] and Galdeano et al. [23] systematically reviewed these.
In recent years, scientific studies have increasingly centered around investigating the impact of probiotics on various neurological conditions in humans. This exploration has given rise to a new category of probiotics termed “psychobiotics”, which have the ability to generate or stimulate the production of SCFAs, neurotransmitters, enteroendocrine hormones, and anti-inflammatory cytokines. As a result, they exhibit potential benefits for a spectrum of neurological diseases like anxiety, depression, Parkinson’s disease, autism spectrum disorders, and Alzheimer’s disease [24,25]. In addition, probiotics show therapeutic potential for aging-related cognitive decline. For instance, Yang et al. found that Probiotic-4 administration improves cognitive function in aging through association with the inhibition of both the TLR4- and RIG-I-mediated NF-κB signaling pathway and inflammatory responses [26].
Probiotics exert a significant and far-reaching influence on human health through the production of functional compounds and modulation of the microbiome–gut–brain axis. However, their effects vary extensively based on factors such as the strain species, dosage, and individual differences. Therefore, conducting in-depth molecular mechanism studies, animal experiments, and clinical trials is crucial to refining their applications and targeting of specific diseases more effectively.

3. Probiotic Engineering Strategies

Probiotics hold promising clinical potential for treating various diseases. Currently, there are two main challenges in utilizing probiotics for disease treatment. On the one hand, probiotics face numerous environmental challenges, such as low gastric pH, enzymatic degradation, the antimicrobial effects of bile salts, and competition with other bacteria, before reaching their intended destinations. To ensure their efficacy, overcoming these obstacles is crucial, requiring probiotics to resist these barriers and reach specific sites within the body in sufficient quantities. On the other hand, the interactions between probiotics and their host, as well as probiotics and other microorganisms, are complex and challenging to precisely control. Therefore, conferring specific functions to probiotics proactively contributes to their precise application in therapeutic contexts. Recent preclinical studies have reported various strategies (Figure 2), including genetic and metabolic engineering, surface modification, and encapsulation, which have been employed to design probiotics, enhancing their efficiency and targeted delivery capabilities. Furthermore, the development of nanotechnology has sparked considerable interest in extracellular vesicles derived from probiotics.

3.1. Genetic and Metabolic Engineering

Progress in synthetic biology offers expanded avenues for the genetic and metabolic engineering of probiotics, including exogenous gene expression, genome editing, and metabolic regulation. These advancements fortify probiotics against digestive challenges, enhance the production of beneficial substances, and diminish their immunogenicity, thereby bolstering their potential in disease treatment.
Many researchers have suggested that engineered probiotics harboring specific genes or pathways show programmable functions of disease treatment. For example, Yan et al. inserted a 3-hydroxybutyrate (3HB) pathway into an ECN genome and deleted the competitive branch pathways, which significantly increased the production of bioactive compounds, 3HB and SCFAs, and thus ameliorated colitis in mice [27]. Praveschotinunt et al. genetically modified ECN to generate a curly fiber matrix composed of curli nanofibers displaying trefoil factors (TFFs), promoting intestinal barrier function and epithelial repair, and leading to enhanced gut epithelial integrity and alleviated colitis in a mouse model [28]. Isabella et al. engineered ECN to express Phe-metabolizing enzymes that achieved a live bacterial therapeutic for metabolic disease phenylketonuria in mice and primates [29]. The developed genetic and metabolic engineering technologies, together with chemical methods, have also facilitated the development of surface modification and encapsulation techniques of probiotics.

3.2. Surface Modification

Altering the surface properties of probiotics can enhance their adherence to specific target areas in the body or improve their interaction with other substances. Surface engineering techniques include modifying the surface charge or roughness or adding functional groups to facilitate specific interactions.
The surface modification of probiotics encompasses a range of techniques, including chemical, physical, and biological approaches. For instance, Song et al. pioneered a click chemistry-driven approach to fortify Clostridium butyricum’s intestinal presence. By introducing azido-modified D-alanine (N3-DAA) to generate azide groups on gut microbiota surfaces and subsequently surface-modifying probiotics with dibenzocyclooctyne (DBCO), they enabled engineered probiotics to engage in precise click chemistry reactions upon oral administration. This innovative strategy ensured prolonged intestinal residency and notably alleviated disease in a colitis mouse model, underscoring the potential of click chemistry in enhancing probiotic colonization efficacy [30]. Pan et al. utilized a dual approach: they genetically enhanced E. coli MG1655 by overexpressing cytolysin A (ClyA) within cells and chemically adorned Bi2S3 nanoparticles (BNPs) onto the cell surface. This modification resulted in remarkable tumor-targeting capabilities and improved sensitivity to radiotherapy for efficient tumor ablation, while mitigating damage to the surrounding normal tissues [31]. Recently, Wang et al. and Wu et al. have systematically reviewed diverse physicochemical and biological technologies for cell surface decoration, such as covalent conjugation with existing functional groups like amine, thiol, carboxyl, diol, and artificially introduced groups, electrostatic and/or hydrophobic interactions, receptor–ligand interactions, avidin–biotin interactions, and others [32,33].

3.3. Encapsulation

Unlike surface modification, probiotic encapsulation envelops either bulk or individual cells within protective wall materials, shielding them from harsh environments, enabling controlled release, and enhancing their bioavailability.
Conventional probiotic encapsulation techniques include extrusion, emulsion, and spray-drying that use polysaccharides, lipids, and proteins as protective matrix materials [34]. Advancements in nanotechnology have led to the creation of nanofibers and nanoparticles for bulk encapsulation, alongside the innovation of biofilms, biomembranes, and nanocoating techniques for individual probiotic nanoencapsulation. These developments aim to ensure high cell viability, resistance to gastric conditions and temperature, and extended shelf life [35]. For example, Harimoto et al. utilized inducible synthetic gene circuits to generate surface capsular polysaccharides, enabling the self-encapsulation of engineered ECN. This increased the fraction of microbial translocation among mouse tumors, resulting in therapeutic efficacy in distal tumors [36]. Cao et al. employed red blood cell membranes to encapsulate engineered probiotics, accomplished through a straightforward process of extruding erythrocyte membranes with bacteria. This innovative method effectively shields bacteria from triggering a high inflammatory response or being eliminated by macrophages, preserving their biological activity [37].

3.4. Membrane Vesicles (MVs)

In recent years, the therapeutic potential of membrane vesicles (MVs) derived from probiotics has garnered significant attention alongside the engineered probiotics. These MVs, ranging from 20 to 400 nm in diameter, encapsulate a variety of macromolecules and play pivotal roles in microbial activities, facilitating inter-microbial and host–microbe communication, metabolite efflux, and gene transfer [38]. Their cost-effective production and facile molecular manipulation for antigenic display make bacterial MVs promising for therapeutic development. Their nanoparticle size enables broad tissue dissemination, their innate immunogenicity aids in vaccine design, and their potential targeted delivery via surface receptors hints at precise therapeutic applications [39].
A type of nanoprobiotic was created by Li et al., encapsulating manganese dioxide (MnO2) with MVs from ECN, and combining this with the anti-inflammatory drug metformin (Met) for treating IBD. The MnO2 reduced oxidative stress induced by IBD, while the ECN-derived MVs regulated the gut microbiome, and Met further improved the inflammatory environment. In a colitis model, this approach reduced inflammatory markers, improving gut health without disrupting the healthy microbial balance [40]. Wang et al. explored the therapeutic potential of MVs from Akkermansia muciniphila in addressing IBD and colorectal cancer. They found that these MVs aided in the growth of beneficial bacteria, helping to rebalance gut microbes linked with these diseases. Additionally, the MVs could interact with immune cells, triggering increased production of immunoglobulin A (IgA) and improving intestinal cell adhesion and mucus secretion, crucial for maintaining gut barrier integrity [41]. Moreover, since MVs can penetrate the central nervous system, they have the potential to provide novel noninvasive therapies against blood–brain barrier infections [42].
Despite numerous articles highlighting the benefits of MVs in disease treatment, such as their non-self-replicating nature which enhances safety in contrast to probiotics, the broader utilization of MVs encounters specific challenges. Firstly, one of the most critical issues is that MVs, like other biological membranes, have very low yields, resulting in relatively high production costs for large-scale applications. Liu et al. reported a method using synthetic biology to induce bacterial peptidoglycan hydrolase to increase MV production, but its industrial application requires more assessment [43]. Secondly, the mechanism of MV formation remains to be elucidated in detail and necessitates further research. Lastly, as a biological product, it is essential to explore the relationship between the suitable storage conditions for MVs and their effective functionality.

4. Engineered Probiotics for Living Therapeutics

Using the various engineering methods mentioned earlier opens an exciting path for creating probiotics designed specifically for living therapeutics. By harnessing probiotics’ natural abilities or introducing novel traits, engineered probiotics hold immense potential for targeted disease treatment. Their precise manipulation allows for customization, enabling them to address diverse health challenges, combating metabolic, immune, and nervous-related diseases. Apart from validating the therapeutic efficacy of probiotics in mice, numerous clinical trials associated with probiotics have achieved initial success (Table 1). In recent years, significant progress has been made in the research on engineered probiotics in the treatment of IBD, cancer, neurological disorders, and other diseases.

4.1. IBD

IBD is a noninfectious chronic gastrointestinal inflammatory disorder, primarily encompassing two main conditions, Crohn’s disease (CD) and ulcerative colitis (UC) [44]. Since 1990, the global number of IBD patients has steadily increased. It is estimated that more than 3 million people in the United States and Europe suffer from IBD. In many countries across North America, Oceania, and Europe, the prevalence of IBD is estimated to exceed 0.3%. Furthermore, the incidence of IBD is on the rise in several emerging industrialized nations [45]. In clinical practice, traditional treatment methods involve the use of 5-aminosalicylic acid, tofacitinib, or biologics such as antitumor necrosis factor (anti-TNF), anti-integrins, and anti-interleukins [5,46,47]. It is worth noting that IBD patients often exhibit disruptions in their gut microbiota, accompanied by a high level of reactive oxygen species (ROS) in the affected areas [48,49,50]. Therefore, the development of engineered probiotics with the ability to scavenge ROS is a feasible therapeutic approach.
The primary challenge that needs to be overcome when using probiotics to treat IBD is the high sensitivity of probiotics to the harsh environment of the GIT. Therefore, it is essential to employ suitable methods to protect probiotics, enabling them to overcome the challenges during delivery and establish colonization at the affected sites [51]. Liu et al. reported an engineered probiotic system [52]. In this system, poly(propylene sulfide) (PPS) and hyaluronic acid (HA) self-assemble to form HA–PPS nanoparticles (HPNs). They then coat the probiotic’s surface with norepinephrine (NE) to create HPN–NE–EcN for treating IBD. PPS can be oxidized into sulfone by ROS, thereby mitigating the high ROS environment in the gut. HA, as a highly biocompatible material, resolves the challenge of applying PPS in the body due to its high hydrophobicity. Norepinephrine assists ECN in withstanding harsh gastrointestinal conditions and enhances its adhesion properties, prolonging ECN’s retention in the gut, and ultimately facilitating its modulation of gut microbiota.
Cao et al. developed an engineered Bifidobacterium longum probiotic that also possesses the capability to scavenge ROS and modulate gut microbiota [53]. Notably, their work not only demonstrated superior therapeutic effects in a murine IBD model but also exhibited promising IBD alleviation capabilities in large animals, such as beagle dogs. This bodes well for potential clinical applications in the future.

4.2. Cancer

Cancer is a prominent global cause of mortality, with data from the WHO indicating that it accounted for nearly 10 million deaths in 2020. The primary clinical modalities for treatment include surgical intervention and the application of radiotherapy and chemotherapy. Radiotherapy operates by either activating signaling pathways that lead to cell death or inducing cellular damage, thereby triggering cellular defense mechanisms, ultimately culminating in the demise of cancer cells [54]. Nonetheless, radiotherapy is often accompanied by challenges, including a scarcity of highly specialized medical professionals, elevated costs associated with radiopharmaceuticals, and the potential harm to nontumor tissues caused by radiation [55]. Chemotherapy involves the use of chemical agents to eradicate cancer cells. However, there are currently no chemotherapeutic drugs specifically targeting cancer cells, and as a result, drug toxicity remains one of the limiting factors for the widespread application of chemotherapy. Furthermore, chemotherapy drugs are primarily administered via intravenous injection, which may lead to patient resistance and diminish treatment efficacy upon repeated injections [56]. Some studies have reported the oral delivery of chemotherapy drugs; however, oral administration entails the passage of chemotherapy drugs through the harsh GIT environment, which raises the possibility of drug inactivation [57]. As the body of literature related to engineered bacteria continues to expand, the utilization of bacteria for drug delivery has garnered widespread research attention.
Wang et al. created an engineered probiotic hybrid material, designated as EcN@HPB, by combining the anticancer drug paclitaxel and BAY-876 bound human serum albumin nanodrugs with ECN [58]. Due to the hypoxic conditions within tumor tissues as compared to normal tissues [59], ECN actively targets tumor tissues, thereby purposefully delivering loaded drugs to the desired locations. At the tumor site, ECN competes with tumor cells for glucose uptake, while BAY-876 further reduces tumor cell glucose uptake via inhibiting glucose transporter 1. Through their synergistic actions, they activate the AMPK signaling pathway in tumor cells, enhancing macropinocytosis in tumor cells. This leads to increased internalization of HPB, raising the concentration of paclitaxel within tumor cells and consequently achieving the goal of antitumor therapy.
With the advancements in immunology, immunotherapy for cancer is gradually emerging as one of the treatment modalities for cancer. The primary goal of immunotherapy is to harness the patient’s immune system to combat malignant tumors [60]. Immunotherapy encompasses various approaches, including immune checkpoint blockade therapy [61], adoptive T cell therapies [62], and cancer vaccines [63]. Similar to chemotherapy, immunotherapy still faces the challenge of nonspecifically affecting tumor tissues, resulting in systemic toxicity. Therefore, leveraging specific probiotics for tumor hypoxia targeting can effectively address the targeting issue in immunotherapy, further expanding the application of immunotherapy in cancer treatment.
Savage et al. utilized synthetic biology techniques to engineer ECN, enabling it to express human chemokine CXCL16 [64]. The primary function of this cytokine is to promote the migration of lymphocytes, particularly T cells, to inflammatory sites, thereby activating the immune response. Furthermore, researchers designed strains expressing CCL20 to target the presentation of tumor-derived antigens by dendritic cells. The synergistic action of these two engineered probiotics effectively restrained tumor progression in MC38 tumor model mice. Chimeric antigen receptor (CAR)–T cell therapy has shown promising results in the treatment of hematologic malignancies, but its effectiveness in solid tumors has been disappointing [65,66]. Vincent et al. developed engineered ECN cells that express specific targets to induce CAR–T cell enrichment and destruction of tumor cells [67]. ECN can target the hypoxic tumor microenvironment, and given the clever design of a synchronized lysis circuit, when the number of ECN cells reaches a critical threshold, they actively lyse to release the target. Subsequently, CAR–T cells recognize these antigen targets released by these probiotic bacteria, effectively killing these tumor cells in situ. To enhance T cell enrichment, researchers combined this system with engineered ECN that were reported earlier in their research to express CXCL16, and the results showed that it effectively suppressed tumor growth in mouse models of human and mouse cancer. This provides a new approach for the application of CAR–T cell therapy in solid tumors.

4.3. Others

Engineered probiotics, in addition to achieving promising results in the treatment of IBD and cancer, have also been reported in the literature for the treatment of various other diseases. Ventilator-associated pneumonia (VAP) is an acute pulmonary infection, typically caused by pathogens such as Pseudomonas aeruginosa or Staphylococcus aureus [68]. Zhang et al. developed a microrobot for VAP treatment by modifying Chlamydomonas reinhardtii with antibiotic nanoparticles encapsulated within neutrophil membranes through click chemistry reactions [69]. Due to Chlamydomonas reinhardtii’s advantageous mobility, it can, to some extent, evade phagocytosis by macrophages. Furthermore, with the assistance of a neutrophil membrane, this microrobot can reduce the chances of clearance. In a mouse model of pneumonia induced by Pseudomonas aeruginosa infection, the authors observed that this system exhibited substantial antibacterial capabilities, leading to increased mouse survival rates.
Candidal vaginitis, a commonly encountered inflammatory fungal infection of the female genital tract, is typically caused by Candida albicans [70]. Statistics indicate that approximately 75% of women worldwide suffer from this disease, which significantly impairs their quality of life [71]. Currently employed clinical treatments often result in damage to normal vaginal cells and tissues, exacerbating the situation [72]. Wei et al. developed a novel approach for treating Candida infection by combining a nanozyme capable of catalyzing hydrogen peroxide into OH radicals with lactobacilli [73]. These lactobacilli were encapsulated within a hyaluronic acid hydrogel. Hyaluronidase secreted by Candida albicans degrades the hydrogel, releasing the composite probiotics. Lactobacilli produce lactic acid, restoring the vaginal environment to its normal acidic state. Simultaneously, the nanozyme breaks down the hydrogen peroxide produced by lactobacilli, generating harmful free radicals detrimental to pathogenic microorganisms. Remarkably, researchers observed satisfying therapeutic effects in a murine disease model. Furthermore, this system exhibited a superior treatment capacity compared to clinical medications, with no significant impact on the vaginal microbiota.
Probiotics also play an important role in neurodevelopment and various brain functions by affecting the gut–brain axis through the production of bioactive compounds and modulation of gut microbiota. Guo et al. utilized a simple biointerface supramolecular self-assembly approach, using a methacrylic acid- and ethyl acrylate-based pH-responsive anionic copolymer to encapsulate Lactobacillus plantarum, enhancing the probiotic’s survival capabilities under extreme conditions [74]. Approximately 2 h after reaching the intestines, these probiotics began synthesizing biologically active substances. The researchers observed significant symptom relief in a mouse model of Parkinson’s disease. This study provides a novel approach to the clinical treatment of neurological disorders.
Engineered probiotics can be endowed with multiple functions, such as resistance to extreme pH environments, drug delivery for therapeutic purposes, and the recruitment of immune cells to activate the host’s immune response. As living entities capable of adapting to their environment, engineered probiotics stand at the forefront of innovative medical interventions, heralding a new era of personalized and precise therapeutics.

5. Conclusions and Outlook

Certainly, probiotics have been demonstrated to exert positive effects on host health. However, commercial probiotics still face various challenges. For instance, probiotics, while exhibiting lower toxicity compared to other microorganisms, still possess immunogenicity, and adverse outcomes like inflammatory reactions can be anticipated during treatment. Moreover, there remains a risk of sepsis when probiotics are administered intravenously. While oral ingestion significantly reduces the risk of sepsis, the extreme pH conditions in the GIT and the presence of various enzymes pose significant barriers to whether probiotics can reach specific sites.
The engineering of probiotics using chemical and biological approaches offers a promising avenue to overcoming the limitations mentioned above and expanding their clinical applications. Several engineered probiotics have seen successful commercialization. For instance, the FDA has sanctioned a genetically engineered probiotic, Bacillus subtilis ZB183, developed and commercialized by ZBiotics (https://zbiotics.com/ accessed on 8 July 2019). This engineered probiotic, hosting an acetaldehyde dehydrogenase enzyme, transforms acetaldehyde into acetic acid, effectively reducing post-alcohol consumption acetaldehyde accumulation and thus relieving the alcohol hangover [75]. Despite the known safety of the parent strain, the modified variant underwent thorough safety assessments before gaining recognition as safe for general use [76]. This marks the world’s pioneering commercialized engineered probiotic, heralding a promising future for engineered probiotics in various commercial applications. In addition, Synlogic is an established biotechnology company specializing in the development of engineered probiotics with therapeutic applications. Among their products are the strains SYNB1618 and SYNB1934, which have been engineered to combat phenylketonuria (PKU). These strains employ ECN as the genetic backbone. However, it is important to acknowledge that, despite the advancements in this domain, the project is currently in phase 2 of clinical trials, as outlined by Vockley et al. [77].
Engineered probiotics currently face stringent regulatory challenges, requiring extensive safety and efficacy testing that is time-consuming and costly. Ensuring consistent effectiveness among diverse individuals, long-term safety, and monitoring for potential side effects adds complexity. Public acceptance and competition with natural probiotics further complicate their adoption. Ethical considerations around genetic modification are also pivotal. For engineered probiotics to advance in clinical applications, advancements in science must align with adjustments in regulatory frameworks to facilitate their utilization. Simultaneously, all modifications made to probiotics should revolve around clinical experiments to ensure practical clinical translational value.

Author Contributions

Conceptualization, M.L. and H.L.; validation, Y.L.; supervision, Y.L. and H.L.; visualization, J.C. and I.P.W.D.; writing—original draft, M.L.; writing—review and editing, M.L., J.C., I.P.W.D. and S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Shenzhen Science and Technology Innovation Commission, China (Grant No. JCYJ20220530114409020 and 20231115134555001).

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shortt, C. The probiotic century: Historical and current perspectives. Trends Food Sci. Technol. 1999, 10, 411–417. [Google Scholar] [CrossRef]
  2. Who, F. Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria. In Proceedings of the Report of a Joint FAO/WHO Expert Consultation, Córdoba, Argentina, 1–4 October 2001. [Google Scholar]
  3. Li, Z.; Wang, Y.; Liu, J.; Rawding, P.; Bu, J.; Hong, S.; Hu, Q. Chemically and biologically engineered bacteria-based delivery systems for emerging diagnosis and advanced therapy. Adv. Mater. 2021, 33, 2102580. [Google Scholar] [CrossRef] [PubMed]
  4. Singh, T.P.; Natraj, B.H. Next-generation probiotics: A promising approach towards designing personalized medicine. Crit. Rev. Microbiol. 2021, 47, 479–498. [Google Scholar] [CrossRef]
  5. Lin, S.; Wu, F.; Zhang, Y.; Chen, H.; Guo, H.; Chen, Y.; Liu, J. Surface-modified bacteria: Synthesis, functionalization and biomedical applications. Chem. Soc. Rev. 2023, 52, 6617–6643. [Google Scholar] [CrossRef]
  6. Hahn, J.; Ding, S.; Im, J.; Harimoto, T.; Leong, K.W.; Danino, T. Bacterial therapies at the interface of synthetic biology and nanomedicine. Nat. Rev. Bioeng. 2023, 1–6. [Google Scholar] [CrossRef]
  7. Feng, Q.; Chen, W.D.; Wang, Y.D. Gut microbiota: An integral moderator in health and disease. Front. Microbiol. 2018, 9, 151. [Google Scholar] [CrossRef] [PubMed]
  8. Chugh, B.; Kamal-Eldin, A. Bioactive compounds produced by probiotics in food products. Curr. Opin. Food Sci. 2020, 32, 76–82. [Google Scholar] [CrossRef]
  9. Zhai, Z.; Dong, W.; Sun, Y.; Gu, Y.; Ma, J.; Wang, B.; Cao, H. Vitamin-microbiota crosstalk in intestinal inflammation and carcinogenesis. Nutrients 2022, 14, 3383. [Google Scholar] [CrossRef]
  10. Zhang, Q.; Zhao, Q.; Li, T.; Lu, L.; Wang, F.; Zhang, H.; Liu, Z.; Ma, H.; Zhu, Q.; Wang, J.; et al. Lactobacillus plantarum-derived indole-3-lactic acid ameliorates colorectal tumorigenesis via epigenetic regulation of CD8+ T cell immunity. Cell Metab. 2023, 35, 943–960.e949. [Google Scholar] [CrossRef]
  11. Wiciński, M.; Gębalski, J.; Gołębiewski, J.; Malinowski, B. Probiotics for the treatment of overweight and obesity in humans—A review of clinical trials. Microorganisms 2020, 8, 1148. [Google Scholar] [CrossRef]
  12. Naomi, R.; Embong, H.; Othman, F.; Ghazi, H.F.; Maruthey, N.; Bahari, H. Probiotics for Alzheimer’s Disease: A Systematic Review. Nutrients 2022, 14, 20. [Google Scholar] [CrossRef]
  13. Zhou, J.; Li, M.; Chen, Q.; Li, X.; Chen, L.; Dong, Z.; Zhu, W.; Yang, Y.; Liu, Z.; Chen, Q. Programmable probiotics modulate inflammation and gut microbiota for inflammatory bowel disease treatment after effective oral delivery. Nat. Commun. 2022, 13, 3432. [Google Scholar] [CrossRef] [PubMed]
  14. Jiang, B.; Wei, M.; He, Y.; Wang, Y.; Zhang, Q.; Wei, H.; Tao, X. Antitumor effect of exopolysaccharide from Lactiplantibacillus plantarum WLPL09 on melanoma mice via regulating immunity and gut microbiota. Int. J. Biol. Macromol. 2024, 254, 127624. [Google Scholar] [CrossRef]
  15. Piewngam, P.; Zheng, Y.; Nguyen, T.H.; Dickey, S.W.; Joo, H.-S.; Villaruz, A.E.; Glose, K.A.; Fisher, E.L.; Hunt, R.L.; Li, B.; et al. Pathogen elimination by probiotic Bacillus via signalling interference. Nature 2018, 562, 532–537. [Google Scholar] [CrossRef] [PubMed]
  16. Lee, J.-Y.; Tsolis, R.M.; Bäumler, A.J. The microbiome and gut homeostasis. Science 2022, 377, eabp9960. [Google Scholar] [CrossRef]
  17. Chin Fatt, C.R.; Asbury, S.; Jha, M.K.; Minhajuddin, A.; Sethuram, S.; Mayes, T.; Kennedy, S.H.; Foster, J.A.; Trivedi, M.H. Leveraging the microbiome to understand clinical heterogeneity in depression: Findings from the T-RAD study. Transl. Psychiatry 2023, 13, 139. [Google Scholar] [CrossRef] [PubMed]
  18. Stojanov, S.; Berlec, A.; Štrukelj, B. The influence of probiotics on the firmicutes/bacteroidetes ratio in the treatment of obesity and inflammatory bowel disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef] [PubMed]
  19. Osbelt, L.; Wende, M.; Almási, É.; Derksen, E.; Muthukumarasamy, U.; Lesker, T.R.; Galvez, E.J.C.; Pils, M.C.; Schalk, E.; Chhatwal, P.; et al. Klebsiella oxytoca causes colonization resistance against multidrug-resistant K. pneumoniae in the gut via cooperative carbohydrate competition. Cell Host Microbe 2021, 29, 1663–1679.e1667. [Google Scholar] [CrossRef]
  20. Zuo, F.; Appaswamy, A.; Gebremariam, H.G.; Jonsson, A.-B. Role of sortase a in Lactobacillus gasseri Kx110A1 adhesion to gastric epithelial cells and competitive exclusion of Helicobacter pylori. Front. Microbiol. 2019, 10, 2770. [Google Scholar] [CrossRef]
  21. Fung, T.C.; Olson, C.A.; Hsiao, E.Y. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017, 20, 145–155. [Google Scholar] [CrossRef]
  22. La Fata, G.; Weber, P.; Mohajeri, M.H. Probiotics and the gut immune system: Indirect regulation. Probiotics Antimicrob. Proteins 2018, 10, 11–21. [Google Scholar] [CrossRef] [PubMed]
  23. Maldonado Galdeano, C.; Cazorla, S.I.; Lemme Dumit, J.M.; Vélez, E.; Perdigón, G. Beneficial effects of probiotic consumption on the immune system. Ann. Nutr. Metab. 2019, 74, 115–124. [Google Scholar] [CrossRef]
  24. Dinan, T.G.; Stanton, C.; Cryan, J.F. Psychobiotics: A novel class of psychotropic. Biol. Psychiatry 2013, 74, 720–726. [Google Scholar] [CrossRef] [PubMed]
  25. Ross, K. Psychobiotics: Are they the future intervention for managing depression and anxiety? A literature review. Explore 2023, 19, 669–680. [Google Scholar] [CrossRef] [PubMed]
  26. Yang, X.; Yu, D.; Xue, L.; Li, H.; Du, J. Probiotics modulate the microbiota–gut–brain axis and improve memory deficits in aged SAMP8 mice. Acta Pharm. Sin. B 2020, 10, 475–487. [Google Scholar] [CrossRef] [PubMed]
  27. Yan, X.; Liu, X.-Y.; Zhang, D.; Zhang, Y.-D.; Li, Z.-H.; Liu, X.; Wu, F.; Chen, G.-Q. Construction of a sustainable 3-hydroxybutyrate-producing probiotic Escherichia coli for treatment of colitis. Cell. Mol. Immunol. 2021, 18, 2344–2357. [Google Scholar] [CrossRef] [PubMed]
  28. Praveschotinunt, P.; Duraj-Thatte, A.M.; Gelfat, I.; Bahl, F.; Chou, D.B.; Joshi, N.S. Engineered E. coli Nissle 1917 for the delivery of matrix-tethered therapeutic domains to the gut. Nat. Commun. 2019, 10, 5580. [Google Scholar] [CrossRef] [PubMed]
  29. Isabella, V.M.; Ha, B.N.; Castillo, M.J.; Lubkowicz, D.J.; Rowe, S.E.; Millet, Y.A.; Anderson, C.L.; Li, N.; Fisher, A.B.; West, K.A.; et al. Development of a synthetic live bacterial therapeutic for the human metabolic disease phenylketonuria. Nat. Biotechnol. 2018, 36, 857–864. [Google Scholar] [CrossRef]
  30. Song, W.-F.; Yao, W.-Q.; Chen, Q.-W.; Zheng, D.; Han, Z.-Y.; Zhang, X.-Z. In situ bioorthogonal conjugation of delivered bacteria with gut inhabitants for enhancing probiotics colonization. ACS Cent. Sci. 2022, 8, 1306–1317. [Google Scholar] [CrossRef]
  31. Pan, P.; Dong, X.; Chen, Y.; Zeng, X.; Zhang, X.-Z. Engineered bacteria for enhanced radiotherapy against breast carcinoma. ACS Nano 2022, 16, 801–812. [Google Scholar] [CrossRef]
  32. Wang, Y.; Li, Z.; Mo, F.; Chen-Mayfield, T.-J.; Saini, A.; LaMere, A.M.; Hu, Q. Chemically engineering cells for precision medicine. Chem. Soc. Rev. 2023, 52, 1068–1102. [Google Scholar] [CrossRef]
  33. Wu, F.; Liu, J. Decorated bacteria and the application in drug delivery. Adv. Drug Deliv. Rev. 2022, 188, 114443. [Google Scholar] [CrossRef] [PubMed]
  34. Reque, P.M.; Brandelli, A. Encapsulation of probiotics and nutraceuticals: Applications in functional food industry. Trends Food Sci. Technol. 2021, 114, 1–10. [Google Scholar] [CrossRef]
  35. Centurion, F.; Basit, A.W.; Liu, J.; Gaisford, S.; Rahim, M.A.; Kalantar-Zadeh, K. Nanoencapsulation for probiotic delivery. ACS Nano 2021, 15, 18653–18660. [Google Scholar] [CrossRef]
  36. Harimoto, T.; Hahn, J.; Chen, Y.-Y.; Im, J.; Zhang, J.; Hou, N.; Li, F.; Coker, C.; Gray, K.; Harr, N.; et al. A programmable encapsulation system improves delivery of therapeutic bacteria in mice. Nat. Biotechnol. 2022, 40, 1259–1269. [Google Scholar] [CrossRef] [PubMed]
  37. Cao, Z.; Cheng, S.; Wang, X.; Pang, Y.; Liu, J. Camouflaging bacteria by wrapping with cell membranes. Nat. Commun. 2019, 10, 3452. [Google Scholar] [CrossRef] [PubMed]
  38. Toyofuku, M.; Nomura, N.; Eberl, L. Types and origins of bacterial membrane vesicles. Nat. Rev. Microbiol. 2019, 17, 13–24. [Google Scholar] [CrossRef] [PubMed]
  39. Bitto, N.J.; Kaparakis-Liaskos, M. The therapeutic benefit of bacterial membrane vesicles. Int. J. Mol. Sci. 2017, 18, 1287. [Google Scholar] [CrossRef] [PubMed]
  40. Li, J.; Sun, M.; Liu, L.; Yang, W.; Sun, A.; Yu, J.; Liu, D.; Zhao, W.; Cheng, M.; He, Z.; et al. Nanoprobiotics for remolding the pro-inflammatory microenvironment and microbiome in the treatment of colitis. Nano Lett. 2023, 23, 8593–8601. [Google Scholar] [CrossRef]
  41. Wang, X.; Lin, S.; Wang, L.; Cao, Z.; Zhang, M.; Zhang, Y.; Liu, R.; Liu, J. Versatility of bacterial outer membrane vesicles in regulating intestinal homeostasis. Sci. Adv. 2023, 9, eade5079. [Google Scholar] [CrossRef]
  42. Srivastava, P.; Kim, K.-s. Membrane vesicles derived from gut microbiota and probiotics: Cutting-edge therapeutic approaches for multidrug-resistant superbugs linked to neurological anomalies. Pharmaceutics 2022, 14, 2370. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, Y.; Chen, J.; Raj, K.; Baerg, L.; Nathan, N.; Philpott, D.J.; Mahadevan, R. A universal strategy to promote secretion of G+/G− bacterial extracellular vesicles and its application in host innate immune responses. ACS Synth. Biol. 2023, 12, 319–328. [Google Scholar] [CrossRef] [PubMed]
  44. Schirmer, M.; Garner, A.; Vlamakis, H.; Xavier, R.J. Microbial genes and pathways in inflammatory bowel disease. Nat. Rev. Microbiol. 2019, 17, 497–511. [Google Scholar] [CrossRef] [PubMed]
  45. Alatab, S.; Sepanlou, S.G.; Ikuta, K.; Vahedi, H.; Bisignano, C.; Safiri, S.; Sadeghi, A.; Nixon, M.R.; Abdoli, A.; Abolhassani, H.; et al. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990–2017: A systematic analysis for the global burden of disease study 2017. Lancet Gastroenterol. Hepatol. 2020, 5, 17–30. [Google Scholar] [CrossRef] [PubMed]
  46. Al-Bawardy, B.; Shivashankar, R.; Proctor, D.D. Novel and emerging therapies for inflammatory bowel disease. Front. Pharmacol. 2021, 12, 651415. [Google Scholar] [CrossRef]
  47. Baumgart, D.C.; Le Berre, C. Newer biologic and small-molecule therapies for inflammatory bowel disease. N. Engl. J. Med. 2021, 385, 1302–1315. [Google Scholar] [CrossRef] [PubMed]
  48. Jostins, L.; Ripke, S.; Weersma, R.K.; Duerr, R.H.; McGovern, D.P.; Hui, K.Y.; Lee, J.C.; Philip Schumm, L.; Sharma, Y.; Anderson, C.A.; et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012, 491, 119–124. [Google Scholar] [CrossRef] [PubMed]
  49. Investigators, I.; Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  50. Plichta, D.R.; Graham, D.B.; Subramanian, S.; Xavier, R.J. Therapeutic opportunities in inflammatory bowel disease: Mechanistic dissection of host-microbiome relationships. Cell 2019, 178, 1041–1056. [Google Scholar] [CrossRef]
  51. Anselmo, A.C.; McHugh, K.J.; Webster, J.; Langer, R.; Jaklenec, A. Layer-by-layer encapsulation of probiotics for delivery to the microbiome. Adv. Mater. 2016, 28, 9486–9490. [Google Scholar] [CrossRef]
  52. Liu, J.; Wang, Y.; Heelan, W.J.; Chen, Y.; Li, Z.; Hu, Q. Mucoadhesive probiotic backpacks with ROS nanoscavengers enhance the bacteriotherapy for inflammatory bowel diseases. Sci. Adv. 2022, 8, eabp8798. [Google Scholar] [CrossRef] [PubMed]
  53. Cao, F.; Jin, L.; Gao, Y.; Ding, Y.; Wen, H.; Qian, Z.; Zhang, C.; Hong, L.; Yang, H.; Zhang, J.; et al. Artificial-enzymes-armed Bifidobacterium longum probiotics for alleviating intestinal inflammation and microbiota dysbiosis. Nat. Nanotechnol. 2023, 18, 617–627. [Google Scholar] [CrossRef] [PubMed]
  54. Petroni, G.; Cantley, L.C.; Santambrogio, L.; Formenti, S.C.; Galluzzi, L. Radiotherapy as a tool to elicit clinically actionable signalling pathways in cancer. Nat. Rev. Clin. Oncol. 2022, 19, 114–131. [Google Scholar] [CrossRef] [PubMed]
  55. Bodei, L.; Herrmann, K.; Schöder, H.; Scott, A.M.; Lewis, J.S. Radiotheranostics in oncology: Current challenges and emerging opportunities. Nat. Rev. Clin. Oncol. 2022, 19, 534–550. [Google Scholar] [CrossRef] [PubMed]
  56. Chi, Y.H.; Lee, J.H.; Kim, J.H.; Tan, H.K.; Kim, S.L.; Lee, J.Y.; Rim, H.-K.; Paik, S.H.; Lee, K.-T. Pharmacological characterization of BR-A-657, a highly potent nonpeptide angiotensin II receptor antagonist. Biol. Pharm. Bull. 2013, 36, 1208–1215. [Google Scholar] [CrossRef] [PubMed]
  57. Pridgen, E.M.; Alexis, F.; Farokhzad, O.C. Polymeric nanoparticle technologies for oral drug delivery. Clin. Gastroenterol. Hepatol. 2014, 12, 1605–1610. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, J.W.; Chen, Q.W.; Luo, G.F.; Ji, P.; Han, Z.Y.; Song, W.F.; Chen, W.H.; Zhang, X.Z. Interference of glucose bioavailability of tumor by engineered biohybrids for potentiating targeting and uptake of antitumor nanodrugs. Nano Lett 2022, 22, 8735–8743. [Google Scholar] [CrossRef]
  59. Wang, J.J.; Lei, K.F.; Han, F. Tumor microenvironment: Recent advances in various cancer treatments. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3855–3864. [Google Scholar] [CrossRef]
  60. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
  61. Sharma, P.; Goswami, S.; Raychaudhuri, D.; Siddiqui, B.A.; Singh, P.; Nagarajan, A.; Liu, J.; Subudhi, S.K.; Poon, C.; Gant, K.L.; et al. Immune checkpoint therapy-current perspectives and future directions. Cell 2023, 186, 1652–1669. [Google Scholar] [CrossRef]
  62. Cascone, T.; McKenzie, J.A.; Mbofung, R.M.; Punt, S.; Wang, Z.; Xu, C.; Williams, L.J.; Wang, Z.; Bristow, C.A.; Carugo, A.; et al. Increased tumor glycolysis characterizes immune resistance to adoptive T cell therapy. Cell Metab. 2018, 27, 977–987.e974. [Google Scholar] [CrossRef] [PubMed]
  63. Goldman, B.; DeFrancesco, L. The cancer vaccine roller coaster. Nat. Biotechnol. 2009, 27, 129–139. [Google Scholar] [CrossRef] [PubMed]
  64. Savage, T.M.; Vincent, R.L.; Rae, S.S.; Huang, L.H.; Ahn, A.; Pu, K.; Li, F.; De Los Santos-Alexis, K.; Coker, C.; Danino, T.; et al. Chemokines expressed by engineered bacteria recruit and orchestrate antitumor immunity. Sci. Adv. 2023, 9, eadc9436. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, N.; Li, X.; Chintala, N.K.; Tano, Z.E.; Adusumilli, P.S. Driving CARs on the uneven road of antigen heterogeneity in solid tumors. Curr. Opin. Immunol. 2018, 51, 103–110. [Google Scholar] [CrossRef]
  66. Hyrenius-Wittsten, A.; Roybal, K.T. Paving new roads for CARs. Trends Cancer 2019, 5, 583–592. [Google Scholar] [CrossRef]
  67. Vincent, R.L.; Gurbatri, C.R.; Li, F.; Vardoshvili, A.; Coker, C.; Im, J.; Ballister, E.R.; Rouanne, M.; Savage, T.; De Los Santos-Alexis, K.; et al. Probiotic-guided CAR-T cells for solid tumor targeting. Science 2023, 382, 211–218. [Google Scholar] [CrossRef] [PubMed]
  68. Lisboa, T.; Diaz, E.; Sa-Borges, M.; Socias, A.; Sole-Violan, J.; Rodríguez, A.; Rello, J. The ventilator-associated pneumonia PIRO score. Chest 2008, 134, 1208–1216. [Google Scholar] [CrossRef]
  69. Zhang, F.; Zhuang, J.; Li, Z.; Gong, H.; de Ávila, B.E.-F.; Duan, Y.; Zhang, Q.; Zhou, J.; Yin, L.; Karshalev, E.; et al. Nanoparticle-modified microrobots for in vivo antibiotic delivery to treat acute bacterial pneumonia. Nat. Mater. 2022, 21, 1324–1332. [Google Scholar] [CrossRef]
  70. Ilkit, M.; Guzel, A.B. The epidemiology, pathogenesis, and diagnosis of vulvovaginal candidosis: A mycological perspective. Crit. Rev. Microbiol. 2011, 37, 250–261. [Google Scholar] [CrossRef]
  71. Watson, M.C.; Grimshaw, J.M.; Bond, C.M.; Mollison, J.; Ludbrook, A. Oral versus intra-vaginal imidazole and triazole anti-fungal agents for the treatment of uncomplicated vulvovaginal candidiasis (thrush): A systematic review. BJOG 2002, 109, 85–95. [Google Scholar] [CrossRef]
  72. Denning, D.W.; Kneale, M.; Sobel, J.D.; Rautemaa-Richardson, R. Global burden of recurrent vulvovaginal candidiasis: A systematic review. Lancet Infect. Dis. 2018, 18, e339–e347. [Google Scholar] [CrossRef] [PubMed]
  73. Wei, G.; Liu, Q.; Wang, X.; Zhou, Z.; Zhao, X.; Zhou, W.; Liu, W.; Zhang, Y.; Liu, S.; Zhu, C.; et al. A probiotic nanozyme hydrogel regulates vaginal microenvironment for Candida vaginitis therapy. Sci. Adv. 2023, 9, eadg0949. [Google Scholar] [CrossRef] [PubMed]
  74. Guo, M.; Yang, C.; Li, B.; Cheng, S.-X.; Guo, Q.; Ming, D.; Zheng, B. Bionic dormant body of timed wake-up for bacteriotherapy in vivo. ACS Nano 2022, 16, 823–836. [Google Scholar] [CrossRef] [PubMed]
  75. Merlo, A.; Abbott, Z.; Alford, C.; Balikji, S.; Bruce, G.; Gunn, C.; Iversen, J.; Iversen, J.; Johnson, S.J.; Kruisselbrink, L.D.; et al. Proceedings of the 10th Alcohol Hangover Research Group Meeting in Utrecht, The Netherlands. Proceedings 2020, 43, 4. [Google Scholar] [CrossRef]
  76. Appala Naidu, B.; Kannan, K.; Santhosh Kumar, D.P.; Oliver, J.W.K.; Abbott, Z.D. Lyophilized B. subtilis ZB183 spores: 90-day repeat dose oral (Gavage) toxicity study in wistar rats. J. Toxicol. 2019, 2019, 3042108. [Google Scholar] [CrossRef] [PubMed]
  77. Vockley, J.; Sondheimer, N.; Puurunen, M.; Diaz, G.A.; Ginevic, I.; Grange, D.K.; Harding, C.; Northrup, H.; Phillips, J.A., 3rd; Searle, S.; et al. Efficacy and safety of a synthetic biotic for treatment of phenylketonuria: A phase 2 clinical trial. Nat. Metab. 2023, 5, 1685–1690. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of the primary mechanisms of probiotic action for disease treatment.
Figure 1. Schematic diagram of the primary mechanisms of probiotic action for disease treatment.
Processes 12 00316 g001
Figure 2. Engineering strategies of probiotics. (A) Genetic and metabolic engineering; (B) surface modification; (C) encapsulation; (D) probiotic-derived membrane vesicles.
Figure 2. Engineering strategies of probiotics. (A) Genetic and metabolic engineering; (B) surface modification; (C) encapsulation; (D) probiotic-derived membrane vesicles.
Processes 12 00316 g002
Table 1. Partial clinical trials related to probiotics.
Table 1. Partial clinical trials related to probiotics.
PhaseTrade NameDiseasePrimary SponsorMain ID
N/ALactobacillus rhamnosus LRa05Type 2 diabetesThe first affiliated hospital of Harbin Medical UniversityChiCTR2300073308
2–3SlimBiotics probioticPrediabetesShahid Beheshti University of Medical SciencesIRCT20221120056553N1
1Lactobacillus rhamnosus SD11Dental cariesThai health promotion foundationTCTR20170525002
1/2VivomixxBone lossTel Aviv Sourasky Medical CenterNCT03518268
3Lactic acid bacteriaGastrointestinal complicationsAlliance for Clinical Trials in OncologyNCT01473290
2Bifidobacterium longum BL21Radiation intestinal injuryThe second affiliated hospital of Soochow UniversityChiCTR2300069881
2LactobacillusDrug-resistant epilepsyTanta UniversityNCT05539287
4Lactobacillus sakei proBio65Chronic rhinosinusitisUniversity of Illinois at ChicagoNCT05427695
2Lactobacillus rhamnosus GGAlcohol use disorderUniversity of LouisvilleNCT05178069
3Lactobacillus acidophilusRectal cancerVice Chancellor of Research of Iran University of Medical SciencesIRCT2016061118745N8
1/2LactobacillusPsoriasisDr. Soetomo General HospitalNCT05254249
1/2BB-12Autism spectrum disorderThe University of Texas Health Science CenterNCT03514784
1Lactobacillus plantarum, PS128Children with autism spectrum disorderYan HaoNCT04942522
1/2VSL#3Nonalcoholic fatty liver DiseaseNorthwell HealthNCT03511365
3FamiLact AsthmaSabzevar University of Medical SciencesIRCT20180115038378N1
2MultiprobioticParkinson’s diseaseThe third affiliated hospital of Sun Yat-Sen UniversityChiCTR2100049412
4Mutaflor®Chronic constipationKeimyung University Dongsan Medical CenterNCT02726295
2LABTHERA-001VaginosisAtoGen Co., Ltd., Daejeon, Republic of KoreaCTRI/2022/10/046723
2MultiprobioticEpilepsyKashan University of Medical SciencesIRCT20170615034549N2
3/4resB® Lung SupportPneumoniaResBiotic NutritionCTRI/2022/04/041687
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liu, M.; Chen, J.; Dharmasiddhi, I.P.W.; Chen, S.; Liu, Y.; Liu, H. Review of the Potential of Probiotics in Disease Treatment: Mechanisms, Engineering, and Applications. Processes 2024, 12, 316. https://doi.org/10.3390/pr12020316

AMA Style

Liu M, Chen J, Dharmasiddhi IPW, Chen S, Liu Y, Liu H. Review of the Potential of Probiotics in Disease Treatment: Mechanisms, Engineering, and Applications. Processes. 2024; 12(2):316. https://doi.org/10.3390/pr12020316

Chicago/Turabian Style

Liu, Mingkang, Jinjin Chen, Ida Putu Wiweka Dharmasiddhi, Shiyi Chen, Yilan Liu, and Hongmei Liu. 2024. "Review of the Potential of Probiotics in Disease Treatment: Mechanisms, Engineering, and Applications" Processes 12, no. 2: 316. https://doi.org/10.3390/pr12020316

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop