Skip to main content

Bacterial derivatives mediated drug delivery in cancer therapy: a new generation strategy

Abstract

Cancer is measured as a major threat to human life and is a leading cause of death. Millions of cancer patients die every year, although a burgeoning number of researchers have been making tremendous efforts to develop cancer medicine to fight against cancer. Owing to the complexity and heterogeneity of cancer, lack of ability to treat deep tumor tissues, and high toxicity to the normal cells, it complicates the therapy of cancer. However, bacterial derivative-mediated drug delivery has raised the interest of researchers in overcoming the restrictions of conventional cancer chemotherapy. In this review, we show various examples of tumor-targeting bacteria and bacterial derivatives for the delivery of anticancer drugs. This review also describes the advantages and limitations of delivering anticancer treatment drugs under regulated conditions employing these tumor-targeting bacteria and their membrane vesicles. This study highlights the substantial potential for clinical translation of bacterial-based drug carriers, improve their ability to work with other treatment modalities, and provide a more powerful, dependable, and distinctive tumor therapy.

Graphical Abstract

Introduction

Cancer is a complex, devastating disease that invades surrounding tissues and organs, leading to serious health complications. Cancer is a major global health challenge, encompassing various different cancer types. According to the WHO, cancer is a leading cause of death worldwide, with an estimated nearly 10 million deaths in 2020, 20 million new cancer cases, and 9.7 million deaths in 2022 [1]. The most common cancer types include lung, breast, prostate, stomach, and colorectal cancers. In 2022, nearly half of all cases (49.2%) and most cancer-related deaths (56.1%) worldwide were predicted to occur in Asia [1, 2]. Cancers of the lung, prostate, and colorectum in males. The lung, breast, and colorectum in women account for the majority of deaths [2, 3]. Due to limited resources and infrastructure, cancer is also a significant health concern in low- and middle-income countries. Early detection and treatment of cancer are vital to improving outcomes for cancer patients.

Researchers have been developing various new methods to improve the outcomes of tumor patients, among that, drug delivery methods have been raised to the forefront of studies due to the efficiency in recent years. Numerous therapeutic agent delivery systems, such as liposomes and nanoparticles, have been developed and extensively tested in animal models and clinical trials because they may increase the effectiveness of treatment by lowering toxic side effects, extending circulation time, and enhancing tumor specificity. Few therapeutic drugs truly reach the cancer site using these kinds of drug delivery systems due to inevitable instability, leakage, inadequate local targeting [4]. As a result, it's critical to create novel drug delivery methods that can get past biological and physical obstacles to transport and reach malignant tissue in sufficiently high quantities.

Bacterial-based drug delivery for cancer is an innovative approach that harnesses the unique properties of bacteria to target and deliver therapeutic agents specifically to tumor cells. By utilizing the natural ability of certain bacteria to selectively accumulate in tumors, this method offers a promising strategy for enhancing the efficacy and reducing the side effects of cancer treatment [4,5,6].

Bacteria show great promise as medicinal, bioimaging, and diagnostic agents due to their distinct features. First of all, under the correct circumstances, bacteria can be easily genetically altered and grown in vast quantities. It's interesting to note that different bacterial species can colonize different parts of mammalian hosts' bodies, such the skin or the stomach, as well as disease locations like tumor tissues and abscesses. Furthermore, bacteria are naturally able to interact with living things [4, 5, 7]. For example, they can adhere to host epithelial cells and decorate their surface with synthetic materials like different kinds of nanoparticles and medications that carry cargo. Furthermore, bacteria have the capacity to produce outer membrane vesicles (MVs) either actively or passively, and these MVs have demonstrated a great deal of promise as a form of effective and safe drug carrier [5, 6, 8]. Recently, bacteria-derived membrane vesicles have been successfully used to load different kinds of diagnosis, imaging, treatment and vaccine agents with various structures, hydrophobicity, charges and solubility, for cancer therapy (Scheme 1).

Scheme 1.
scheme 1

MVs-mediated delivery of diagnosis, imaging, treatment and vaccine agents for cancer therapy

The use of bacteria and bacterial derivatives bacteria has been shown to significantly influence the fabrication of controlled targeted medication delivery systems to combat the spread of cancer. Bacteria and their derivatives are unique medication carriers for cancer therapy compared to most other conventional drug delivery methods. They have the ability to penetrate physical barriers, gather in tumor tissues, and trigger immune responses that are anti-tumorous. Moreover, they may be chemically and genetically altered to generate and transport anticancer drugs into tumor tissues, improving the safety and effectiveness of cancer treatment while reducing the harmful effects on healthy cells.

This article reviews recent developments of drugs delivery systems mediated by bacteria, and it also discusses the advantages and present obstacles of using these to treat cancer. In particular, concentrating on several therapeutic approaches within the framework of bacterial-mediated drug delivery systems, which fuse bacteria with nanoparticles to combat cancer. This work also describes the genetic engineering used to genetically modify a variety of pathogenic and non-pathogenic microorganisms in order to promote tumor regression. Drug delivery systems based on microorganisms have proven to be a reliable and efficient approach for tumor theranostics. Finally, we discuss the clinical translation and future prospects of bacterial derivatives-based cancer treatments.

Bacterial-mediated anticancer drug carriage

Since the late nineteenth century, researchers have been exploring the potential use of bacteria and spores as agents to combat tumors, inspired by observations that bacteria could infiltrate tumor cells and impede their growth [9,10,11,12]. This pioneering approach involves injecting various therapeutic agents, including potent proteins and low-molecular-weight drugs, directly into tumors. Several anaerobic bacterial species have demonstrated the capacity to overcome physiological barriers that often impede the effectiveness of conventional chemotherapeutics. They are well-suited for precise localized tumor targeting due to their ability to thrive in the hypoxic conditions inside tumors [13, 14]. However, challenges persist, including dosage-dependent bacterial side effects and issues related to limited therapeutic efficacy, as the reticuloendothelial system can clear bacteria before they reach their specific targets [15, 16]. A cooled charge-coupled device detector detects and tracks the light released by Lux-expressing bacteria. The administration of these microorganisms to animals offers an extremely useful method for producing sensitive whole-body pictures with a very low backdrop. With many mouse tumor models, this imaging method should allow for the robust measurement of bacterial migration into primary and metastatic tumors in real-time [16].

To mitigate these challenges, extensive research has focused on reducing systemic toxicity through genetic and chemical modifications of microorganisms. For instance, the deletion of crucial virulence factor genes has been utilized to attenuate various bacterial species, including Salmonella, Clostridium, Escherichia coli, and Listeria (Table 1) [17,18,19,20,21,22,23]. In recent years, Phase I/II clinical studies have shown progress in various bacterial treatments. These treatments typically involve the use of modified microbes containing cytotoxic proteins, cytokines, angiogenesis inhibitors, antigens, and antibodies as anticancer agents [24,25,26]. Modified bacteria have leveraged their intricate sensory systems to enable self-guided movement in response to gradients of oxygen, pH, temperature, and various attractive chemical compounds. Because of their biological functions and biocompatibility, modified bacteria can administer anticancer treatments within in vivo settings, capitalizing on their unique capabilities [27]. For instance, their chemotactic migratory abilities propel bacteria toward preferred nutritional environments, while their anaerobic and hypoxia tropism traits direct them to the hypoxic regions of disease lesions (Fig. 1).

Table 1 The delivery of anticancer medications through bacterial mediation is an innovative approach that holds promise in cancer treatment
Fig. 1
figure 1

A Genetic engineering. Through the bacterial cycle, plasmid-transfected bacteria may generate the medicine constantly and accomplish pulsed release of the medication. B Grafting of linkers. Biotin antibody-modified and streptavidin-modified NP “drug-linker-bacteria” is formed by salmonella. Reproduced from ref. [9]. MDPI, Copyright 2023. C The killing of tumor cells produced by photothermal treatment based on pDA-VNP enhances bacteria-mediated biotherapy much further, Reproduced from ref. [166]. ACS Nano, Copyright 2018

Furthermore, substantial efforts are dedicated to expanding bacteria-powered biohybrid microswimmers. These biohybrids enhance the capabilities of microswimmers designed to transport synthetic vehicles, such as liposomes, NPs, and hydrogels for drug delivery. They achieve this by incorporating the self-directed biotic momentum and detection skills of microbes. An illustrative example is the utilization of magneto-aerotactic bacteria in a micromotor system to deliver drug-loaded liposomes to tumor locations, as demonstrated by Felfoul et al. [28] Bacteriabots, with synthetic microparticles attached to bacterial surfaces through biotin-streptavidin linkages, exhibit increased adherence to gastrointestinal and urinary system epithelial cells due to the lectin's preference for mannose molecules on cell surfaces. As shown in Table 1, the delivery of anticancer medications through bacterial mediation is an innovative approach that shows potential in cancer treatment.

Bacterial surface modification strategies and advantages

Bacteria's distinct qualities, including their capacity to manipulate DNA, grow quickly, colonize well, and exhibit targeted behaviors, have drawn a lot of interest among the biomedical community. But there are several barriers that hinder therapeutic implementation, including the unavoidably harmful adverse reactions of bacteria and inadequate colonization in disease locations [9, 29]. Using the numerous groups of chemicals on the surface of bacteria, surface alterations have been carried out to modify the structure and composition of the bacteria's surface, reduce their toxicity, or add unique therapeutic components to modify the biological characteristics of the bacteria and give them novel roles. The advancement of such methods will show the prospective role that bacteria could have in medicine, particularly in tumor immunotherapy. Generally, bacterial methods for modifying surfaces can be classified into three categories: chemical, physical, and biological (Fig. 2).

Fig. 2
figure 2

Reproduced from ref. [30] Wiley, Copyright 2024

Diagram illustrating the use of chemical, physical, and biological reactions to modify the bacterial surface. 1) Chemical modification contains the formation of covalent bonds by the many chemical locations on the surface of bacteria, as well as other techniques such metal ion chelation, in situ deposition, biotin-streptavidin conjugation, and in-situ polymerization. 2) Electrostatic contact, physical extrusion, self-assembly, and layer-by-layer techniques are the principal examples of physical interaction. 3) The two primary biological interactions that result in outcomes that are visible on the surface are genetic engineering and biosynthesis.

The bacterial surface consists of polysaccharides, proteins, and lipids which make up the complex biological structure. As an essential part of the bacterial cell wall, peptididoglycan is mostly made up of small peptide chains, nacetylmuramic acid, and n-acetylglucosamine. Furthermore, a multitude of chemical locations, including free thiols, amines, hydroxyls, and carboxyl groups are found. These sites have the potential to form permanent chemical interactions with diverse substances.

Mostly in outer membranes of bacteria peptidoglycans and teichoic acids having plenty of hydroxyl groups. These molecules provide the membranes a negative charge that permits surface alteration by electrostatic interactions. At the moment, bacterial surface modification is accomplished using physical techniques including mechanical extrusion and electrostatic adsorption [30].

Biological modification of the surface uses genetic engineering, biosynthesis, and numerous other methods to change the surface structure of bacteria, giving them additional capabilities and improved biocompatibility, physical and chemical methods of modification improve the surface structure of bacteria artificially.

Drug-carrying bacteria’s cellular envelopes

When compared to bacterial cells, cellular envelopes for drug administration offer several advantages, with a key benefit being their inability to colonize vital organs. By genetically modifying living bacterial cells to customize both their inner and outer surfaces, we can harness the potential of cellular envelopes. Importantly, cellular envelopes retain their immunomodulatory qualities by preserving their surface features [31]. Bacterial ghosts (BGs) represent a common type of cellular envelope, essentially the empty cell envelopes of gram-negative bacteria altered by the lysis gene E from bacteriophage X174. Gene E codes for a 91-aa polypeptide. When the simulated gene E is expressed under controlled conditions in gram-positive bacteria, these bacteria quickly perish without undergoing lysis [32]. The expression of protein E, a membrane protein capable of oligomerizing and forming transmembrane tunnel structures, induces a differential in osmotic pressure between the cytoplasm and its adjoining environment. This leads to the expulsion of cytoplasmic content, microbial cell lysis, ultimately transforming the cell into a lifeless covering, justifying the term “bacterial ghost.” The remarkable stability of these cellular envelopes, which can even be lyophilized, is a noteworthy attribute [33,34,35,36].

For over 20 years, these non-denatured cellular envelopes have served predominantly as non-living delivery vehicles for medicines, antigens, nucleic acids, and various physiologically active substances, capitalizing on their intrinsic cellular features [37]. BGs, derivatives of E. coli NM522, were biologically modified in 1999 to facilitate the passage of biotinylated substances through their cytoplasmic membrane. To achieve this, the BG creation process commenced before the inner side of the bacterial cytoplasmic membrane produced a streptavidin fusion protein. Exploiting the strong binding affinity between streptavidin and biotinylated substances, DNA of the plasmid combined with biotinylated poly-l-lysine and biotinylated fluorescent dextran could adhere to the surface of the BG crust. This study demonstrated that BGs, acting alongside streptavidin on the cytoplasmic membrane, might serve as drug delivery vehicles for the site-specific transportation of medications that have been biotinylated [38].

A companion study by Haslberger et al. investigated the immune system activation effects of various bacterial ghost (BG) platforms through in vitro absorption. The findings revealed remarkably effective BG acceptance and suggested that BGs, capable of triggering immunological responses, may hold value as in situ immunotherapy delivery systems. Measurement of IL-12(p70) production and IL-12(p40) mRNA accumulation indicates activation of IL-12. Particular significance lies in this interleukin in the induction of cellular TH1 immunological responses. Electron microscopy could verify the quick absorption of bacterial ghosts in macrophages, occurring within a half-hour to an hour. These sites can change the bacterial surface functionally by forming long-lasting chemical interactions with various substances [39]. Due to the immunosuppressive milieu often present in tumor-draining lymph nodes (TDLNs), oncolytic microorganisms struggle to efficiently cross-prime tumor-specific T lymphocytes via antigen-presenting cells such as dendritic cells (DCs) in TDLNs. In this context, Escherichia coli (EcP) overexpressing pyranose oxidase (P2O) were used, encapsulated in low-concentration photosensitizer NPs and PEGylated mannose, to create a micro-to-nano oncolytic microbial therapy. P2O generated hazardous hydrogen peroxide upon treatment, leading to tumor regression and the release of tumor antigens. By promoting DC maturation and influencing the TDLNs' immuno-microenvironment, the enhanced TDLNs distribution by OMVs improved tumor antigen-specific T cell immune responses. This micro-to-nano oncolytic bacterium shows promise in altering TDLNs and eliminating tumors, as illustrated in (Fig. 3) [40].

Fig. 3
figure 3

Reproduced from ref. [40] with permission from Wiley, Copyright 2023

A Laser irradiation-induced OMV release and immune activation effects in vitro. EcP@TAPP NPs-PMAN preparation diagram. B Diagram illustrating the timetable for creating and treating a bilateral tumor model. C TDLNs remolding for improved immunotherapy and EcP@TAPP NPs-PMAN for cancer carnage. Following an intravenous injection of EcL and EcL@TAPP NPs-PMAN, the primary organs and tumor-bearing 4T1 mice's fluorescent pictures (T: tumor, H: heart, Li: liver, S: spleen, Lu: lung, K: kidney).

By encapsulating the systemically delivered anticancer drug DOX within bacterial ghosts (BGs) derived from Mannheimia haemolytica, they significantly alleviated its severe side effects, highlighting the potential of these engineered BGs for controlled and prolonged drug administration [41]. Through in vivo investigations, these BG systems also demonstrated effectiveness in delivering medications to sites of ocular surface disorders [42]. In a separate study, resveratrol, a well-known polyphenolic substance with immunomodulatory properties, was incorporated into E. coli NM522 BGs. These resveratrol-loaded BGs exhibited an enhanced impact on macrophage cells when cultured through murine macrophage cells, emphasizing the potential of BGs as efficient delivery carriers for substances like resveratrol [43].

Bacteria that cause cancer may initiate innate immunity. Attenuated live bacteria provide significant safety hazards, whereas inactivated microorganisms have limited antitumor effectiveness. In this instance, Wang et al. demonstrate that manganese dioxide-coated paraformaldehyde-fixed bacteria administered intratumorally have a capacity to stimulate innate immune reactions, alter the immunosuppressive tumor microenvironment, and elicit tumor-specific and abscopal antitumor responses. In mice, rabbits, and tree shrews, one single intratumoural injection of mineralized Salmonella typhimurium inhibited the development of several subcutaneous and orthotopic tumor types and shielded the treated animals against tumor recurrence [44]. Additionally, they demonstrate that orthotopic liver cancer in rabbits may be treated with mineralized bacteria delivered by arterial embolism (Fig. 4). The results of the study encourage the use of oncolytic calcified bacteria as effective and secure antitumor immunotherapeutics in more translational studies.

Fig. 4
figure 4

Reproduced from ref. [44] Nature, Copyright 2024

Mineralized bacteria produce immunological protection and activate the systemic immune response. a Building and managing the B16F10 bilateral tumor model for melanoma. The following treatments are shown in the graphs b, c: normal saline (blank), 15 μg anti-PD-1 (i.v.), 20 μg mineralized FS (i.t.), and 15 μg anti-PD-1 (i.v.) + 20 μg mineralized FS (i.t.) (n = 5). Growth curves of primary tumors and distant tumors are shown in the graphs (b) and the survival rates of mice are shown in the graph (c). d The mineralized bacteria's successive immune activation and modulation methods against tumors.

Minicells represent a distinct type of non-chromosomal cellular envelope that holds promise as a drug delivery system. Typically ranging in size from 100 to 400 nm, these minicells result from aberrant cell division. They contain minimal to no chromosomal DNA but retain all the RNA and membrane proteins of the parent cell in their molecular form [45, 46]. While minicells can undergo plasmid-directed protein synthesis, they lack the capacity for sexual reproduction. Notably, their ability to express therapeutic compounds encoded by recombinant plasmid DNA at specific target sites is a remarkable feature [47, 48]. In practical applications, siRNA-encoding plasmids have been effectively loaded into minicells, enabling RNA interference to inhibit the expression of proteins that support tumor development or enhance drug efflux mechanisms [49, 50].

Several envisioned applications underscore the diverse utility of non-chromosomal cellular envelopes in drug delivery. Their production is both straightforward and cost-effective, allowing manufacturing at various scales. Notably, they are characterized by extended shelf life and an inability to revert to pathogenic forms [51]. However, it is crucial to recognize that these cellular envelopes bear the same surface antigens as living bacteria, potentially inducing immunological responses. Despite this, these envelopes provide a pragmatic avenue for designing drug delivery systems, leveraging their internal storage capacity and exceptional internalization properties. Similar to living bacteria, a major challenge for drug carriage methods based on cellular envelopes is ensuring accurate spatiotemporal distribution. Incorporating artificial elements, such as magnetic NPs, into these enclosures may further enhance this controllability.

Generation of bacterial membrane vesicles

In a natural process, bacteria bud their membranes, releasing substances into their surrounding environment. Once separated and purified, bacterial membrane vesicles (BMVs) structures enveloped by lipid bilayers that remarkably resemble eukaryotic extracellular vesicles (EVs). BMVs may be spontaneously released by bacteria, both Gram-positive and Gram-negative, typically ranging in size from 20 to 400 nm [52,53,54]. The environment in which these vesicles are released significantly influences various biological activities, including pathogenicity, horizontal gene transfer, metabolite export, phage infection, and intercellular communication [55, 56]. In this section, we discuss various types of BMVs formed by microbes, such as OMV, IMV, and DMV (Fig. 5) [167].

Fig. 5
figure 5

Reproduced from ref. [167] with permission from Elsevier, Copyright 2022

Diagrammatic depiction of the several BMV types that are manufactured from either Gram-positive or Gram-negative bacteria, as well as their applications in the treatment of infections and cancer. Gram-negative bacteria's outer membranes give rise to OMVs. DMVs (double-membrane vesicles) and IMVs (inner membrane vesicles) are made by physical or biological methods. Furthermore, BMVs are made using genetically altered Gram-positive and Gram-negative bacteria.

Differentiating between Gram-positive and Gram-negative bacteria is facilitated by their structural differences. Gram-positive bacteria possess a thick peptidoglycan coat but lack an outer lipid membrane, whereas Gram-negative bacteria have both an outer lipid membrane and a thin peptidoglycan layer [57]. Consequently, the type of bacteria influences the construction and development of bacterial membrane vesicles (BMVs) (Fig. 5) [167].

BMVs created by Gram-negative bacteria

Outer membrane vesicles (OMV)

Numerous studies have investigated bacterial membrane vesicles (BMVs) produced by Gram-negative bacteria [56, 57]. The distinctive structure of gram-negative bacteria is characterized by an outer lipid membrane and the presence of lipopolysaccharides (LPS). The outer lipid membrane of these bacteria spontaneously separates, leading to the formation of outer membrane vesicles (OMVs) [58, 59].

Initially, membrane proteins play a crucial role in connecting the outer lipid membrane to peptidoglycan, forming a stable Gram-negative envelope. However, as outer membrane vesicles (OMVs) begin to form, these proteins may become less stable. This instability can result from either the movement of connecting proteins causing a disruption in the linkage between peptidoglycan and the outer lipid membrane or a direct break in the connection. The disruption of these connecting proteins leads to the release of OMVs [56, 60]. It is important to note that local environmental factors significantly influence the generation of OMVs. Consequently, OMVs can carry a variety of cargoes, including proteins and genetic materials. Additionally, they may contain proteins that facilitate the interaction between peptidoglycans and the outer lipid membrane.

The intricate processes involved in OMV generation result in the production of vesicles with variable sizes and compositions [61]. Among the materials found in these vesicles are phospholipids, proteins, nucleic acids, virulence factors (such as LPS), and occasionally metal ions, signaling molecules, and metabolites [62]. The study of OMV proteins is crucial for understanding tissue targeting and signal transduction, and proteomics has played a vital role in this regard. Various techniques have been employed in proteomic investigations, including direct trypsin digestion of electrophoresis gels followed by liquid chromatography (LC)-MS/MS, 2D electrophoresis followed by in situ mass spectrometry (MS), and sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) [63, 64]. These investigations consistently support the notion that OMVs are predominantly composed of proteins associated with the outer membrane, further substantiating the natural formation of OMVs [65, 66]. Local signals, including stressors and stimulants, significantly influence the formation of OMVs in Gram-negative bacteria. Enhancing the culture medium and adjusting environmental conditions can lead to improvements in OMV production and enable better control over their compositions [67,68,69,70].

Inner membrane vesicles (IMVs)

Gram-negative bacteria encounter challenges in spontaneously generating membrane vesicles due to the protective outer membrane and peptidoglycan layer surrounding the inner membrane. Moreover, concerns arise regarding potential immunological toxicity when utilizing outer membrane vesicles (OMVs) in medical applications, primarily due to the presence of LPS in OMVs. To address these challenges, Kim et al. introduced a technique for producing bacterial protoplast-derived nanovesicles (PDNVs), also known as inner membrane vesicles, from Gram-negative microbes [71].

Protoplasts, lacking the peptidoglycan layer (cell wall) and the toxic outer membrane, are achieved through the action of lysozyme, representing a bacterial state devoid of these components. Subsequently, protoplasts give rise to inner membrane vesicles (IMVs) once the outer membrane has been eliminated. The utilization of a serial extrusion method has proven effective in the production of IMVs, which have found application in the development of a universal adjuvant-free vaccine. The remarkable aspect is that IMVs have demonstrated superior effectiveness and safety compared to vaccinations using outer membrane vesicles (OMVs). Strong humoral and cellular immune responses specific to antigens have been effectively elicited by IMVs. This breakthrough opens new avenues for the development of vaccines that are not only safer but also more effective [72].

Double membrane vesicles (DMVs)

When used as drug delivery systems, both outer membrane vesicles (OMVs) and inner membrane vesicles (IMVs) may face stability and cargo loading challenges as they share the same lipid membrane. Addressing these issues, especially in the context of medicinal applications, necessitates the creation of vesicles that incorporate both membrane linkers made of peptidoglycans and lipid membranes of the bacterium. This is particularly crucial for the development of vaccines [73, 74].

This study has confirmed that double membrane vesicles (DMVs) indeed encompass the complete bacterial membrane and possess the unique property of containing multiple crucial antigens essential for vaccine production. This achievement was realized through the application of cryogenic transmission electron microscopy (cryo-TEM), biochemistry, and proteomics. In comparison to outer membrane vesicles (OMVs) generated from the same bacteria, DMVs significantly increased animal survival in a sepsis mouse model caused by Pseudomonas aeruginosa. This superior survival is attributed to DMVs' enhanced adaptive immunity and distinct biodistribution, most likely resulting from the presence of more pathogen-associated molecular patterns (PAMPs) on DMVs [55].

A study used double membrane vesicles (DMVs) to target various cells within tumor microenvironments and highlights the potential of the nitrogen cavitation method for generating DMVs from diverse bacterial sources. The endogenous targeting ligands were produced, and arginine-glycine-aspartate (RGD) peptides were expressed by Escherichia coli to construct DMVs. Within tumor microenvironments, these DMVs demonstrated a spontaneous attraction to neutrophils, monocytes, and endothelial cells. Particularly noteworthy is the finding that DOX can be loaded with remarkable efficiency into DMVs using a pH gradient (12% w/w). DMVs loaded with Dox significantly reduced tumor diameters in a mouse model of melanoma compared to DMVs without the expression of targeting ligands, indicating the innovative and efficacious potential of DMVs as a platform for targeted drug delivery in cancer therapy [75].

BMVs formed by Gram-positive bacteria

Gram-positive bacterial membrane vesicles (BMVs) have emerged as a unique and captivating research area, recently gaining significant attention and evolving into a thriving field of study [76]. BMVs have been observed in Gram-positive bacteria from over 30 different species (Table 2) [77]. These vesicles carry a wide range of cargo molecules, including proteins, lipids, enzymes, poisons, and nucleic acids. Notably, Gram-positive BMVs exhibit substantial differences from Gram-negative outer membrane vesicles (OMVs), primarily in their absence of periplasmic components and lipopolysaccharides (LPS) [78].

Table 2 Anticancer medication delivery facilitated by bacterial MVs

The exact process controlling the biogenesis of Gram-positive bacterial membrane vesicles (BMVs) is still under investigation. In contrast to Gram-negative bacteria, which release outer membrane vesicles (OMVs) from their outer membrane, Gram-positive bacteria possess a robust cell wall composed of peptidoglycans, potentially hindering the formation of lipid membranes [79, 80]. According to a prevailing theory, enzymes could potentially degrade the peptidoglycan layer, exposing the lipid membrane and facilitating the development of BMVs [81, 82]. Gram-positive BMVs serve two essential functions, namely the transfer of various chemicals and the promotion of bacterial survival [83, 84]. Studies have documented the movement of surface receptors in BMVs and the transfer of bacterial chromosomal DNA, particularly in Ruminococcin species. Remarkably, Gram-positive bacteria release BMVs that contain factors promoting nutrient uptake, antibiotic-degrading enzymes such as β-lactamase, and even harmful agents designed to enhance bacterial survival [85]. The size of Gram-positive BMVs typically ranges from 10 to 400 nm [86]. As shown in (Table 2), facilitation of anticancer medication delivery by bacterial membrane vesicles (MVs).

Bacteria-driven biohybrid drug delivery system

The primary objective of a drug delivery system is to transport and release medication precisely at the intended location in the body. This process aims to shield the drug from adverse conditions, including potential immune reactions and encounters with low pH levels, during its journey from the administration site to the site of action. Furthermore, the system should offer protection to healthy tissues against potential drug side effects (Table 3).

Table 3 An overview of biohybrid nanocarriers based on bacteria for the treatment of cancer

Bacterial cells not only function as efficient microswimmers but also act as microsensors, capable of perceiving alterations in the physicochemical properties of their environment, such as pH, oxygen levels, glucose levels, and temperature. They respond accordingly to these changes [87]. However, the effectiveness of bacterial sensing often relies on placing bacterial cells close to the target action site. Additionally, bacterial sensing is typically most efficient at short distances. Therefore, achieving active control over the placement of the medication delivery device within the body is considered ideal [88,89,90].

Synthetic mobile microrobots hold the potential to address challenges related to long-range communication and precise control. However, they encounter various technological limitations when operated at small sizes. The biohybrid approach, involving the integration of microbial cells with micro/NPs, aims to overcome these limitations [91]. Through this integration, biohybrids can collaborate and carry out advanced functions that neither the biological nor the synthetic component could perform independently. For instance, when in proximity to the target action site, bacteria can utilize chemotactic sensing and steering. When farther from the target, remote magnetic steering can be employed to guide microbes configured as an imitation microrobot body toward the desired location. This biohybrid technique accelerates the arrival of drug delivery systems at the target location, reducing the threat of sanction by the reticuloendothelial mechanism [92].

One major benefit of the biohybrid method is the “division of labor” between biological and non-biological systems, which reduces the need for major genetic alterations to bacterial cells [93]. Concerns about the potential reversibility of genetic alterations in microorganisms used within the body have prompted challenges related to control and containment. Therefore, when incorporating bacteria into biohybrid systems, it is preferable to choose non-pathogenic, ideally food-grade, or commensal bacteria with few or no mutations. This choice aligns with the fundamental need for bacteria to actively respond to environmental gradients. While this approach may not fully harness all the advantageous features of bacteria, it simplifies the concept of bacterial drug delivery systems, making it more practical for implementation. Subsequent sections will delve into the essential design criteria for the improvement of biohybrid medication delivery systems [93]. Here, we focus on different routes of administration for bacteria-based drugs, including intra-tumoral injection, oral administration, intravenous injection, and intranasal administration. As shown in (Table 3), the summary of biohybrid nanocarriers based on various bacteria for the treatment of cancer.

Nanoparticle-mediated anticancer drug carriage

Nanoparticles (NPs) are extensively employed in tumor treatment due to their outstanding drug-loading capacity, ease of fabrication, and biocompatibility. Specifically, organic NPs are favored for drug delivery because of their numerous advantageous characteristics, including biocompatibility, biodegradability, and adaptability. The rapid advancements in nanotechnology have facilitated the incorporation of various therapeutic substances into NPs, such as liposomes, silica-based porous constituents, polymeric structures, and micelles [94]. A pivotal milestone in cancer treatment was the introduction of the first nano-delivery system, a liposome containing doxorubicin (DOX) with a diameter of approximately 100 nm. This innovation enhanced pharmacokinetics and drug distribution, thanks to the passive accumulation of nanomaterials within tumors facilitated by the EPR effect. Drug-loaded NPs exhibited a higher tumor uptake rate and reduced systemic toxicity compared to drug-free NPs [95, 96] (Fig. 6).

Fig. 6
figure 6

Reproduced from ref. [7] Elsevier, Copyright 2019

An example of a liposome-based smart medication delivery system for cancer treatment in steps.

Nanoparticle drug delivery systems have revealed great potential in cancer therapy, but they also come with certain drawbacks [8]. Nanoparticles may face challenges in penetrating deep into tumor tissues, which can affect the distribution and efficacy of the drugs. Nanoparticles can be recognized and cleared by the immune system, reducing their circulation time and effectiveness in delivering drugs to cancer cells. Some nanoparticles may exhibit toxicity or induce inflammatory responses in healthy tissues, leading to adverse effects [97]. Controlling the release of drugs from nanoparticles can be challenging, affecting the therapeutic efficacy and potential side effects. Nanoparticles may require specific storage conditions and have limited stability, which can impact their shelf life and practicality for clinical use [98]. It is important to address these disadvantages through further research and development to optimize nanoparticle drug delivery systems for effective and safe cancer treatment [99].

Although each of these variables possess a significant impact upon the effectiveness of the nano-drug delivery process and, consequently, govern the efficacy of therapy, the NPs employed in medical therapy often have certain sizes, shapes, and surface properties. Nanoparticles (NPs) with diameters in the range of 10 to 100 nm are commonly deemed appropriate for cancer therapy due to their capacity to efficiently transport medications and provide an increased EPR effect. NPs larger than 100 nm will probably to be removed from circulation through phagocytes, while smaller NPs may escape from the normal blood vessels and are swiftly filtrated through kidneys [99, 100].

Bacterially mediated drug delivery for cancer therapy does offer certain advantages compared to other nanoparticle-based drug delivery systems [100]. Bacteria have the ability to actively target and penetrate deep into tumor tissues, which can enhance the efficacy of drug delivery to cancer cells. Additionally, bacteria can be engineered to release therapeutic agents specifically within the tumor microenvironment, minimizing off-target effects and reducing systemic toxicity [101]. The inhibition of cancer mediated by bacteria involves several pathways, including immune system activation. Furthermore, some bacteria have inherent tumor-seeking properties, making them ideal vehicles for targeted drug delivery in cancer therapy. Overall, the unique capabilities of bacteria in targeting and delivering drugs to cancer cells make them a promising approach in cancer therapy [102].

Administration routes of bacteria-based drugs

Bacteria, whether wild-type or genetically modified, can enter the body through various routes. The choice of the administration route is influenced by several variables, including the target site, pharmacological properties, and ease of application. Apart from considerations related to patient comfort, the selected route of administration significantly impacts the efficacy of the treatment approach and the likelihood of adverse effects [103,104,105]. It is essential to understand that the design of the medication delivery system is strongly influenced by the chosen method of administration.

Intra-tumor injection

The most effective method for delivering medications to the targeted site is through direct injection into or in proximity to the affected region. Anaerobic bacteria such as Clostridium novyi, well-known for their exceptional capacity to target tumors, do not demonstrate the same predilection when given systemically in larger animals with a substantial blood volume [9]. In contrast, compared to systemic injection, intratumor injection of S. typhimurium results in a significant improvement in tumor suppression with fewer side effects [106].

For anatomical regions that are challenging to access via the circulatory system, such as the blood–brain barrier (BBB)-protected central nervous system, direct injection is the preferred technique [107, 108]. Bacterial targeting through direct injection into brain tumors has shown considerable promise [109]. In addition to enhanced efficacy, intratumoral injection of therapeutic microorganisms leads to reduced systemic toxicity. Germs introduced via this method can be eliminated with antibiotics after treatment, thereby reducing the risk of genetically altered organisms being released into the environment. It is crucial to recognize that intratumoral injection is typically highly invasive and, as a result, extra multifarious.

Intravenous injection

Intravenous injection is an extremely effective method of medication delivery, especially for tumors with a richer blood supply compared to surrounding tissues [110, 111]. Although systemic injection of bacteria may result in severe systemic infections, experiments conducted in vivo with several strains of bacteria, including S. typhimurium and Bifidobacterium bifidum, have not been discouraged [22, 112]. Although systemic injection of bacteria may pose a risk of severe systemic infections, several strategies have been developed to mitigate these risks and harness the therapeutic potential of bacteria, particularly in cancer therapy. Bacteria can penetrate and thrive in hypoxic and necrotic regions of tumors where traditional therapies are less effective. In most cases, bacteria were able to traverse from the circulatory systems of mice to tumors and establish themselves within the core of the tumor. However, the feasibility of applying this approach in larger animals is currently under investigation [113, 114].

Oral administration

Oral administration is a widely adopted drug delivery method due to its convenience, adaptability, non-invasiveness, and greater patient amenability. It proves particularly beneficial for treating specific gastrointestinal (GI) tract diseases, such as inflammatory bowel syndrome. Researchers have shown a keen interest in oral medication administration, especially for oral immunizations delivered through the intestinal mucosa [115, 116]. To be effective in this role, the drug delivery system must traverse the intestinal epithelial barrier and interact effectively with the complex GI environment, which includes factors like significant pH changes, digestive enzymes, and the commensal bacteria population [117, 118].

One approach to address these challenges is using gastrointestinal (GI) commensal bacteria, such as lactic acid bacteria (LAB). Notably, Lactococcus and Bifidobacterium, commensal gut bacteria, have garnered significant attention [119]. For example, systemic IL-27 injection revealed to be less successful than oral administration of genetically modified Lactobacillus lactis, which produces immunosuppressive interleukin-27 (IL-27), to mice for the treatment of colitis [120, 121]. Additionally, there has been promise in treating type 1 diabetes with this approach, reducing diarrhea caused by C. difficile, and avoiding hemolytic-uremic syndrome caused by E. coli O157:H7 [122, 123]. In another application, orally administered genetically engineered B. longum expressing alpha-melanocyte-stimulating hormone has been employed to treat ulcerative colitis, develop a vaccine against the Hepatitis C virus, and manage myocarditis using the anti-inflammatory cytokine IL-12 [124, 125]. Other bacterial strains under investigation for oral medication administration include S. typhimurium, E. coli, and L. casei [126,127,128]. Another method for the oral administration of medicinal microorganisms is encapsulation in protective materials [129]. This approach has been shown to significantly enhance the acid survival time of various bacterial strains, including Lactobacilli, Bifidobacterium, and E. coli [130, 131]. While oral bacterial treatment has demonstrated promising results in delivering bacteria to non-GI tract malignancies in mice [132, 133], it is essential to note that delivery efficiency may vary in humans, where bacterial escape from the stomach into the circulation is less common [134,135,136]. Additionally, it's worth mentioning that confining orally delivered microorganisms may pose challenges regarding potential environmental discharge [137,138,139].

It is important to remember that most lactic acid bacteria (LAB) employed as delivery vectors are food-grade, innocuous strains of bacteria obtained from fermented foods rather than true commensal bacteria. The physiology of these food-grade bacteria could make it difficult for them to survive in the gastrointestinal system of humans. Therefore, greater research into human commensal bacteria is becoming more important to develop drug delivery vectors that should last and function effectively within the gastrointestinal tract for an extended period of time [140, 141].

Intranasal administration

The nasal route was employed in earlier applications of bacteria-based medicine carriage, specifically in fecal microbiota transplants. However, due to the accessibility of mucosal surfaces, the intranasal administration of bacteria-based treatments is primarily utilized for immunization [142]. For instance, Streptococcus gordonii recombinant strains expressing specific antigens from Mycobacterium tuberculosis have been administered intranasally to activate CD4 + and CD8 + T cells and provide immunization against Neisseria meningitidis, a major cause of meningitis [143]. Lactobacillus pentosus has demonstrated the ability to stimulate an immune response against the influenza virus in the respiratory system [144]. Notably, several lactic acid bacteria elicited significantly stronger immune responses when administered intranasally as opposed to intragastrically.

Drug loading inside bacteria

Direct drug introduction into bacteria is also an option, as is the use of gene editing methods that make use of nucleic acids to help integrate pharmaceuticals into bacterial cells. Anticancer medications can be produced constantly by genetically engineered bacteria, whereas the direct loading technique allows for a single drug release after bacterial lysis. Advances in genetic engineering and synthetic biology have made it possible to design highly controlled and safe bacterial therapies. Researchers can precisely tailor bacterial strains to maximize their therapeutic benefits while minimizing risks.

Electroporation

A method employed to enhance cell membrane permeability is electroporation, which involves delivering brief, high-voltage electrical pulses to cells. This process induces temporary holes in the cell membrane surface, enabling the movement of substances that would otherwise struggle to pass through [145]. Reversible electroporation, characterized by brief electric pulses to facilitate the absorption of medicines or liposomes by bacteria, is utilized to maintain the biological activity of the bacteria. It is important to note, however, that electroporation does cause some damage to the bacteria. Electroporated bacteria may exhibit varying degrees of decreased biological activity compared to untreated control microorganisms.

Zoaby et al. investigated the incubation procedures of electroporation and direct incubation to enhance the delivery of DOX liposomes into S. typhimurium [146]. The findings revealed that the liposome absorption rate by bacteria was less than 5% when treated directly for over 4 h, while electroporation resulted in 62% of the bacteria absorbing the liposomes. Treated bacteria, compared to untreated controls, showed approximately a 20% reduction in growth. In a separate study, Xie et al. utilized gold nanorods to modify the surface of E. coli and employed electroporation to introduce 5-fluorouracil (5-FU) and zoledronic acid (Fig. 7) [147]. These modified bacteria exhibited a lowered existence ratio and crusade hustle to 87% and 88%, respectively, while loading 8.8% 5-FU and 10.5% ZOL. Upon exposure to near-infrared (NIR) light, gold nanorods generated heat, leading to the destruction of both bacteria and tumor cells. Subsequently, the medication was released from the deceased bacteria, enhancing its efficacy against tumor cells.

Fig. 7
figure 7

Reproduced from ref. [147] Elsevier Copyright 2021

EcNZ/F@Au/NIR preparation and treatment mechanism. a EcNZ/F@Au are created by electroporating FU and ZOL into EcN and decorating the EcN surface with Au NRs. b EcN's self-guided motility encourages EcNZ/F@Au to extravasate blood vessels, accumulate in tumor tissues, and engage with tumor cells. Tumor cells have photothermal effects under NIR light, which triggers their conversion to BGs. While ZOL's local release of BGs increases TAM polarization toward the M1 phenotype for immunotherapy, FU's release from BGs has a chemotherapeutic impact on tumor cells.

Genetic engineering

One technique used to modify bacteria's genes is the transfection of DNA fragments carrying anticancer medicines in the form of plasmids. Through genetic alteration, bacteria gain the ability to continuously produce various compounds associated with cancer prevention, including immunological factors, tumor antigens, cytotoxic agents, and more. This approach ensures that bacteria within the tumor can sustain their therapeutic actions. For instance, Nguyen et al. developed a tempered strain of S. typhimurium capable of expressing cytolysin A (ClyA) [148]. An l-arabinose is integrated into the bacterial ClyA gene, which specifically activates ClyA in the presence of L-arabinose, preventing damage to normal tissue cells. Upon introducing l-arabinose to the tumor site, the promoter activates, allowing the bacteria to consistently produce ClyA, targeting and destroying tumor cells. In another study, Chou et al. suggested that to overcome the immune tolerance to auto-antigens prevalent in liver cancer cells, a plasmid containing the Alpha-fetoprotein (AFP) gene was introduced into an attenuated strain of S. typhimurium [149]. Genetically modified bacteria expressing unique AFP associated with liver cancer triggered an immune response, involving T cells, to eradicate and eliminate the tumor (Fig. 8).

Fig. 8
figure 8

Reproduced from ref. [148] with permission from Cancer Research, Copyright 2010

A The biological mechanisms that bacteria use to perform these tasks include machinery for gene translation, which produces anticancer proteins (dark blue); flagella, which performs chemotaxation; specific gene promoter regions, which respond to molecular signals (red squares); chemotactic receptors, which produce blue; and machinery, which produces red molecules. B Bioengineering of S. typhimurium. C ClyA-expressing S. typhimurium: imaging and therapeutic implications in tumor-bearing mice. Hep3B2.1-7 or CT-26 cells were administered subcutaneously (s.c.) into five mice per group. Tumor-bearing mice were given PBS, untransformed S. typhimurium (S.t.Lux), or transformed S. typhimurium [S.t.Lux + pBC (Ara−)] once tumors reached 130 mm3 in volume. After day 4 of injection with transformed S. typhimurium [S.t.Lux + pBC (Ara +)], 60 mg of l-arabinose was given intraperitoneally (i.v.) daily into a different set of tumor-bearing mice (n = 5). D Pictures of typical mice with subcutaneous tumors. Bacterial bioluminescence in vivo imaging using noninvasive methods in the typical animals.

Yoon et al. developed a Salmonella strain incorporating IFN-γ to combat tumors in the context of utilizing bacteria for cytokine production in tumor therapy [150]. When administered subcutaneously, this genetically modified S. typhimurium significantly inhibited tumor development compared to unaltered phosphate-buffered saline, enhancing the survival of mice with tumors. In a distinct approach, Din et al. introduced an innovative bacterial drug delivery system employing genetic manipulation to synchronize, pulse, and repeat drug release [151]. This represents a novel method for periodic drug delivery, deviating from the conventional engineering of bacteria for the continuous expression of antitumor substances. The system's three essential components, regulatory protein LuxR, AHL synthesis protein LuxI, and the pointer molecule Acyl-homoserine lactone (AHL), collaborate to control the microbial cycle.

When the quantity of bacteria is low, most of the AHL that the bacteria make exits the cell and accumulates relatively little. Conversely, intracellular AHL reaches a threshold level when population density rises, significantly enhancing lysin protein synthesis. This, in turn, results in the release of medicine through the lysis of a substantial percentage of bacteria. The periodic cycling technique has the potential to take advantage of circadian rhythms in host-microbe interactions by controlling the recurrence rate with the generosity of these inhabitant cycles. This might lead to a more efficient administration of bacterial medicine [147].

Bacteria are capable of being genetically modified to generate powerful therapeutic gadgets, including cytotoxic drugs, immunomodulators, cytokines, prodrug converting enzymes, small interfering RNAs, and nanobodies, while acting as a tumor-targeting drug carriage. Through their interactions with immune cells, tumor cells, and other TME constituents, these cargoes and the bacteria cooperate to rewire the TME. Immune cells are brought in, activated, and their cytokines and chemokines produced in order to complete this conversion [152]. As a result, the bacteria anticancer effect is higher (Fig. 9).

Fig. 9
figure 9

Reproduced from ref. [152] Nature, Copyright 2023

a, b When chemical compounds like Doxy or L-arabinose are present, bacteria release ClyA, which kills cancer cells, or FlaB, which reprogrammes the TME and increases the recruitment of M1-type macrophages that fight tumors by activating TLR-4 and TLR-5 signaling. c Other possible strategies include the use of physical stimuli like heat, light, or targeted ultrasound to encourage the delivery of immunotherapeutics into the TME by microorganisms. d, e The QS system, which in this instance is based on the AHL autoinducer, has been used to genetically modify bacteria. This system produces AHL through the luxI promoter and regulated lysE expression, which leads to quorum-mediated lysis and intratumoral release of therapeutics like PD-L1 and CTLA-4-blocking or CD47-blocking nanobodies. f Cloned neoantigens activate tumor-infiltrating T lymphocytes, which then identify and eliminate tumor cells. g, h by secreting cytokines, chemokines, or other immunomodulatory payloads to attract and activate TILs in the TME, engineered bacteria can also elicit adaptive immune responses against malignancies.

Challenges and limitations of using bacteria

Using bacteria for cancer therapy, though promising, comes with several challenges and limitations that need to be addressed to ensure safety and efficacy. Systemic injection of bacteria can lead to severe systemic infections, posing a significant risk to patients. This necessitates careful control and monitoring of bacterial growth and spread within the body. Certain bacteria can produce toxins that may cause harm to normal tissues and organs. Managing these toxic effects is crucial to ensure patient safety. The human immune system is designed to eliminate bacterial infections. This immune response can reduce the effectiveness of bacterial therapy by rapidly clearing the bacteria before they can exert their therapeutic effects. Bacterial therapies can trigger inflammation, which, while potentially aiding in tumor destruction, can also cause collateral damage to normal tissues and exacerbate adverse effects. Tumors are heterogeneous, and not all cancer cells may be equally susceptible to bacterial infection or the effects of bacterial toxins. Ensuring that bacteria selectively target and kill cancer cells without affecting healthy cells is challenging. Ensuring that bacteria localize to the tumor site and do not spread to other parts of the body is essential to prevent unwanted infections and side effects. This requires advanced delivery systems and precise control mechanisms. While bacterial therapy for cancer holds significant promise, addressing these challenges and limitations is essential for its successful development and clinical application. Advances in genetic engineering, immune modulation, and targeted delivery systems are helping to overcome some of these hurdles. Ongoing research, rigorous clinical trials, and collaboration between scientists, clinicians, and regulatory bodies are critical to unlocking the full potential of bacterial therapies for cancer treatment.

Clinical translation

Several significant challenges impede the practical implementation of modified bacterial treatments. Firstly, the creation of aesthetically pleasing bacteria is a crucial prerequisite. These modified bacteria should not only possess the ability to detect latent tumors and metastases in patients but also enable real-time tracking of bacterial development and spread throughout the body. Another major challenge involves enhancing the precision of genetically modified microorganisms in accurately targeting tumors. Overcoming this obstacle is pivotal for obtaining regulatory approval and ensuring patient safety [153]. Ineffective tumor targeting can result in robust bacterial growth in healthy tissues and the potential dissemination of infection, posing a particular challenge for immune-compromised patients with advanced cancer. Despite the utilization of nonpathogenic bacterial agents in clinical studies, therapeutic benefits have often been negligible or nonexistent. Fortunately, viable solutions exist to address these issues through the integration of synthetic biology and nanotechnology. These advancements hold the potential to significantly improve the safety and effectiveness of modified bacterial medicines. In this context, we highlight real-time detection, improved tumor targeting, and enhanced therapeutic outcomes [154].

Facilitating real-time detection

Monitoring microbial settlement over time is essential for the advancement of therapeutic applications, as it serves two main purposes: first, to monitor proliferation in target locations and other organs to prevent confrontational events or damage to healthy tissue; second, to evaluate the efficacy of bacteria-based cancer therapy in localizing and growing within the tumor. To detect bacterial colonization in tumors, various techniques have been employed, including bioluminescence, fluorescence, magnetic resonance imaging (MRI), and positron emission tomography (PET) [153, 154]. Plasmids carrying the luxCDABE operon from Photobacterium leiognathi are utilized to create bioluminescent bacteria such as E. coli and S. typhimurium [155]. GFP-carrying plasmids transform bacteria to emit fluorescence. Additionally, fluorescence imaging can be conducted using bacteria loaded with NPs, even in the absence of GFP. These optical imaging techniques, demonstrated through whole-body imaging, have shown excellent effectiveness in detecting cancers in animal models. However, their clinical use is challenging due to the limited penetration of visible light in human tissue. PET and MRI techniques are anticipated to replace optical imaging due to their greater sensitivity and deeper tissue penetration. Magnetic NPs can be employed to directly modify bacterial strains for use in MRI, or they can be genetically transformed to produce magnetosomes [156]. For instance, an external magnetic field can be applied to precisely image tumor locations using Magnetococcus marinus strains laden with magnetosomes. Additionally, the use of metabolic 2-nitroimidazole-based PET and 18F-fluorodeoxysorbitol enables the imaging of E. coli to observe their colonization within tumors [157].

There is evidence to support the great therapeutic effectiveness of bacteria-initiated cancer treatment. Nevertheless, the therapeutic impact and result are compromised by the unintended therapeutic efficaciousness and the systemically produced inflammatory maelstrom. Wang et al. presents the rational design and engineering of a thermally-activated living nanomedicine, Sa@FeS, based on reactive biohybrid. The goal is to improve hydrogen sulfide (H2S)-combined chemodynamic oncotherapy by biomineralizing ferrous sulfide nanoparticles (FeS NPs) onto the surface of a Salmonella typhimurium strain (Sa) without lowering microbial activity. Owing to Sa extensive penetration capacity, FeS NPs promote a photothermally-enhanced catalytic Fenton reaction, which, when exposed to near-infrared light, breaks down endogenous H2O2 into deadly hydroxyl radicals within tumor tissues [158]. In the meantime, Sa bacteria continue to generate H2S continuously inside the tumor, resulting in H2S-induced intracellular acidosis which encourages the cooperative production of reactive oxygen species (Fig. 10).

Fig. 10
figure 10

Reproduced from ref. [158] Wiley, Copyright 2024

Systemic demonstration of Sa@FeS, a living nanomedicine, as comprehensive anticancer therapy. a The Sa@FeS preparing process. Ferrous iron and exogenous thiosulfate (S2O32-) were added into liquid media containing bacteria and sterilized at a high temperature using an inductive cooker. To create the live nanomedicine Sa@FeS, the bacterial colony was subsequently merged with the medium and cultivated anaerobically for a full day. b Sa@FeS for all-in-one tumor treatment using targeted therapy. I Intrathecal administration of H2S gas generated by bacteria has the potential to stimulate CDT by inhibiting CAT activity, which raises H2O2 levels, and building up lactic acid, which causes intracellular acidosis, which lowers TME pH. II Cytotoxic ROS against tumor cells may be produced via the Fenton reaction, which is mediated by released Fe2 + /Fe3 +. III FeS nanoparticles on the surface of bacteria may enable substantially improved CDT and photothermal treatment following exposure to a 1064 nm laser. The above-mentioned treatment routes operate in concert to induce tumor cancer cells' mitochondrial respiratory failure, which in turn blocks the flow of ATP and ultimately causes cellular death or necrosis to prevent cancer.

Improving tumor targeting

Tumor-targeting capabilities aim to provide a safer and more potent cancer treatment. Examples of obligatory anaerobes, such as Bifidobacterium and Clostridium, demonstrate modest invasion of healthy tissues and relatively strong tumor selectivity. In contrast, facultative anaerobes like Listeria and Salmonella could multiply in oxygen-rich conditions, posing a risk to healthy tissues [159]. However, genetic modification of these facultative anaerobes can mitigate their detrimental effects on healthy tissues and enhance their ability to target tumors. For improved safety and precision in targeting, "obligate" anaerobic Salmonella strains, like YB1 and ST4, exclusively colonize the necrotic portions of tumor tissue. Nevertheless, these strains may face challenges efficiently targeting early metastatic tumor cells that typically have sufficient oxygen.

To address this issue, auxotroph mutant strains can be engineered for tumor targeting. For example, mutant Salmonella strains lacking specific nutrients may be designed to flourish only in tumor microenvironments rich in amino acids, such as Leu- and Arg-rich regions. The use of bacteria-driven microswimmers for drug delivery, responsive to external stimuli, has gained popularity. Bacteria exhibit various taxis mechanisms, including pH taxis, magnetotaxis, and chemotaxis. Notably, Serratia marcescens bacteria, displaying efficient chemotaxis, can be observed in microswimmers moving towards l-serine gradients. Fe3O4 nanoparticle-modified natural Spirulina platensis can be employed for magnetically targeted accumulation in malignancies. Further enhancing these bacteria's on-site adherence to affected tissues may further improve targeting efficiency. Despite advancements in exploiting bacterial taxis capabilities, various processes, such as thermotaxis, phototaxis, and galvanotaxis, remain undiscovered [160].

According to Zhang et al. A number of bacteria are suitable towards cognitive bio-hybrid robots because they have built-in motility and sensing capabilities enabling taxis-based autonomy. Bacteria-based robots that incorporate active nano-hybrids may act as cognitive drug delivery vehicles, reacting to a variety of simulated signals like magnetotaxis or chemotaxis to reach desired locations. The creation, propulsion, imaging, and treatment of bacteria-based bio-hybrid magnetic robots for a range of illnesses have advanced significantly in the past few decades. Therapeutic genes and gene reporters for tumor treatment and in vivo imaging are additionally expressed by genetic alteration [161]. Several peritrichous flagellad bacteria such as Escherichia coli might be bioengineered into microrobots for targeted administration that is noninvasive in physiological conditions (Fig. 11).

Fig. 11
figure 11

Reproduced from ref. [161] MDPI, Copyright 2024

Diagram illustrating the creation of bio-hybrid magnetic robots using bacteria and its uses in specific therapy. a A TEM picture of MC-1 and MC-1 that has been liposome-decorated, allowing for the loading of medicines. b Transverse MC-1 tumor segments following targeted and liposome attachment. Utilizing a fluorescent optical microscope, pictures of every slice were taken. The photographs demonstrate an excellent dispersion among the injected MC-1 cells within the tumor. c An illustration of the AMB-1-based microrobots in successive transmission beneath magnetic and optical fields, together with a SEM picture of a typical microrobot. 5 µm is the scale bar. d Setting up and characterizing robots according to AMB-1 that can perform consecutive magneto/optics. e Diagram of the bio-hybrid robots created using E. coli coupled with SPION and nickel nanoparticles (NLs). Doxorubicin (DOX) and indocyanine green (ICG) are put into the NLs. f Intellectual drawings showing how magnetic guidance might direct bacterial bio-hybrid robots across permeable microenvironments and into target tissues, such tumors. g Diagram showing how to build hybrid magnetically robots utilizing E. coli that can sense three different environments: temperature, hypoxic, and magnetic.

Enhancing therapeutic outcomes

Since William Coley’s pioneering use of live infectious agents, specifically Streptococcus pyogenes, for cancer treatment in 1891, extensive research has been conducted on various bacterial strains in clinical settings [162]. Despite these efforts, the outcomes were not consistently associated with significant tumor reduction; instead, they often resulted in mild side effects and successful bacterial colonization in patients. The collective experience from clinical trials indicates that although modified bacteria may demonstrate reduced virulence, they frequently fall short of delivering the intended therapeutic effects.

Despite these challenges, a substantial number of bacterial strains with significant potential for synergistic therapy remain untested in human subjects [22]. For instance, studies conducted by Xing et al. showed that AMB-1 combined with laser irradiation successfully reduced tumor cells at temperatures as high as 58 °C [153]. NPs gathered in the hypoxic area of the tumor in a research by Chen et al. When the ICG payload was exposed to near-infrared (NIR) light, the oxygenated tumor tissue around it was killed. Significant tumor shrinking was the outcome of this photothermal tumor lysis mechanism, which both provided nutrients and promoted enhanced bacterial penetration into the tumor tissue. Significantly, the mouse survival rate was 100% after 28 days, and the primary organs' hematoxylin and eosin (H&E) staining examination showed no harm [163].

Furthermore, in a study conducted by Chen et al., OMV-coated polymeric NPs exhibited remarkable efficacy, achieving a 70% tumor suppression in a 4T1 xenograft breast cancer model [164]. Additionally, Chowdhury et al. employed genetically modified E. coli to produce an encoded nanobody antagonist of CD47. This innovative approach enhanced tumor cell phagocytosis by macrophages, ultimately resulting in complete tumor regression. The method demonstrated promising therapeutic success across various mouse tumor models, including melanomas and triple-negative breast cancers [21].

The microbes inherent traits, such as having non-specific metabolic sites, hazardous contaminants, and unchecked growth, prevent them from being used in therapeutic settings like tumor treatment. Li et al. describe a biohybrid that has been developed to precisely ablate tumors by effectively targeting malignant regions using a pre-established metabolic route. With this approach, lactate oxidase genes and hypoxia-inducible promoters are added to DH5α Escherichia coli, which is then extensively surface-armored using iron-doped ZIF-8 nanoparticles. As response to a hypoxic tumor microenvironment, this bioengineered E. coli is able to manufacture and release lactate oxidase, which lowers the quantity of lactate also activates the immune system. Because the nanoparticles have peroxidase-like properties, they may convert hydrogen peroxide (H2O2) into very harmful hydroxyl radicals, which is the final result of lactate metabolism. Significant cancer cell iron deficiency being a consequence of this as well as the conversion of tirapazamine-loaded nanoparticles to poisonous benzotriazinyl [165]. This biohybrid when injected intravenously dramatically reduces tumor growth and metastasis (Fig. 12).

Fig. 12
figure 12

Reproduced from ref. [165] Wiley, Copyright 2024

Features and architecture of PP3244@Fe-ZT. a Pictorial representation of PP3244@Fe-ZT production and functions. b PP3244 and c PP3244@Fe-ZT scanning electron microscopy (SEM) pictures. d At pH = 6.5 (n = 3), growth curves for PP3244 and PP3244@Fe-ZT assuming the identical starting concentration. e PP3244 and PP3244@Fe-ZT lactate absorption in various oxygen environments (n = 3).

Conclusion and future perspectives

The use of bacterial derivative-mediated drug delivery in cancer therapy displays promising potential for targeted and efficient treatment. By harnessing the unique properties of bacterial derivatives, such as their ability to target specific cells and tissues, researchers can develop innovative strategies to enhance drug delivery and improve therapeutic outcomes for cancer patients. Further research and clinical trials are needed to fully explore the benefits and challenges of this approach, but the initial results suggest that bacterial derivative-mediated drug delivery could be a valuable tool in the fight against cancer. However, there haven't been many clinical trials conducted yet, and the majority of research on bacteria-mediated cancer treatment is still in the pre-clinical stages. Further clinical trials on such microbes are required in the future. Prior research has previously demonstrated that single therapy is not a particularly effective cancer treatment (Table 4) [9, 10, 16, 200,201,202,203,204,205,206,207].

Table 4 Clinical trials with engineered tumor-targeting bacteria

In the future, bacterial derivative-mediated drug delivery for cancer holds great promise for revolutionizing cancer therapy. With ongoing advancements in biotechnology and nanomedicine, researchers can further optimize the design and delivery of bacterial derivatives to enhance their efficacy and specificity in targeting cancer cells. Additionally, the development of personalized medicine approaches, such as utilizing patient-specific bacterial derivatives, could lead to more tailored and effective treatments for individual cancer patients. Collaborations between multidisciplinary teams of scientists, clinicians, and industry partners will be crucial in translating these innovative strategies from the lab to the clinic, ultimately improving outcomes for cancer patients and potentially transforming the landscape of cancer treatment.

Combination therapy leverages the strengths of different therapeutic approaches to provide a more robust, effective, and personalized treatment for cancer. It addresses the complexities and challenges of treating a heterogeneous and adaptable disease, ultimately aiming to improve patient outcomes and quality of life. The strategic use of combination therapy represents a sophisticated and evolving approach in the ongoing battle against cancer.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  2. Clancy E. ACS report shows prostate cancer on the rise, cervical cancer on the decline. Renal Urol News. 2023. https://doi.org/10.3322/caac.21763.

    Article  Google Scholar 

  3. Hasan Mujahid M, Upadhyay TK, Upadhye V, Sharangi AB, Saeed M. Phytocompound identification of aqueous Zingiber officinale rhizome (ZOME) extract reveals antiproliferative and reactive oxygen species mediated apoptotic induction within cervical cancer cells: an in vitro and in silico approach. J Biomol Struct Dyn. 2023;12:1–28.

    Article  Google Scholar 

  4. Cao Z, Liu J. Bacteria and bacterial derivatives as drug carriers for cancer therapy. J Control Release. 2020;10(326):396–407.

    Article  Google Scholar 

  5. Wu L, Bao F, Li L, Yin X, Hua Z. Bacterially mediated drug delivery and therapeutics: strategies and advancements. Adv Drug Deliv Rev. 2022;1(187):114363.

    Article  Google Scholar 

  6. Fan JY, Huang Y, Li Y, Muluh TA, Fu SZ, Wu JB. Bacteria in cancer therapy: a new generation of weapons. Cancer Med. 2022;11(23):4457–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hossen S, Hossain MK, Basher MK, Mia MN, Rahman MT, Uddin MJ. Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: a review. J Adv Res. 2019;1(15):1–8.

    Article  Google Scholar 

  8. Chen J, Ning C, Zhou Z, Yu P, Zhu Y, Tan G, Mao C. Nanomaterials as photothermal therapeutic agents. Prog Mater Sci. 2019;1(99):1–26.

    Article  CAS  Google Scholar 

  9. Zhao X, Xie N, Zhang H, Zhou W, Ding J. Bacterial drug delivery systems for cancer therapy: “Why” and “How”. Pharmaceutics. 2023;15(9):2214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Krick EL, Sorenmo KU, Rankin SC, Cheong I, Kobrin B, Thornton K, Kinzler KW, Vogelstein B, Zhou S, Diaz LA. Evaluation of Clostridium novyi–NT spores in dogs with naturally occurring tumors. Am J Vet Res. 2012;73(1):112–8.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Wang L, Wang Q, Tian X, Shi X. Learning from Clostridium novyi-NT: how to defeat cancer. J Cancer Res Ther. 2018;14(Suppl 1):S1-6.

    Article  CAS  PubMed  Google Scholar 

  12. Mi Z, Feng ZC, Li C, Yang X, Ma MT, Rong PF. Salmonella-mediated cancer therapy: an innovative therapeutic strategy. J Cancer. 2019;10(20):4765.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kasinskas RW, Forbes NS. Salmonella typhimurium specifically chemotax and proliferate in heterogeneous tumor tissue in vitro. Biotechnol Bioeng. 2006;94(4):710–21.

    Article  CAS  PubMed  Google Scholar 

  14. Kucerova P, Cervinkova M. Spontaneous regression of tumour and the role of microbial infection–possibilities for cancer treatment. Anticancer Drugs. 2016;27(4):269.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chen W, Wang Y, Qin M, Zhang X, Zhang Z, Sun X, Gu Z. Bacteria-driven hypoxia targeting for combined biotherapy and photothermal therapy. ACS Nano. 2018;12(6):5995–6005.

    Article  CAS  PubMed  Google Scholar 

  16. Toso JF, Gill VJ, Hwu P, Marincola FM, Restifo NP, Schwartzentruber DJ, Sherry RM, Topalian SL, Yang JC, Stock F, Freezer LJ. Phase I study of the intravenous administration of attenuated Salmonella typhimurium to patients with metastatic melanoma. J Clin Oncol. 2002;20(1):142–52.

    Article  PubMed  Google Scholar 

  17. Mercado-Lubo R, Zhang Y, Zhao L, Rossi K, Wu X, Zou Y, Castillo A, Leonard J, Bortell R, Greiner DL, Shultz LD. A Salmonella nanoparticle mimic overcomes multidrug resistance in tumours. Nat Commun. 2016;7(1):12225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Felgner S, Kocijancic D, Frahm M, Heise U, Rohde M, Zimmermann K, Falk C, Erhardt M, Weiss S. Engineered Salmonella enterica serovar Typhimurium overcomes limitations of anti-bacterial immunity in bacteria-mediated tumor therapy. Oncoimmunology. 2018;7(2): e1382791.

    Article  PubMed  Google Scholar 

  19. Fritz SE, Henson MS, Greengard E, Winter AL, Stuebner KM, Yoon U, Wilk VL, Borgatti A, Augustin LB, Modiano JF, Saltzman DA. A phase I clinical study to evaluate safety of orally administered, genetically engineered Salmonella enterica serovar Typhimurium for canine osteosarcoma. Vet Med Sci. 2016;2(3):179–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dang LH, Bettegowda C, Huso DL, Kinzler KW, Vogelstein B. Combination bacteriolytic therapy for the treatment of experimental tumors. Proc Natl Acad Sci. 2001;98(26):15155–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chowdhury S, Castro S, Coker C, Hinchliffe TE, Arpaia N, Danino T. Programmable bacteria induce durable tumor regression and systemic antitumor immunity. Nat Med. 2019;25(7):1057–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhou S, Gravekamp C, Bermudes D, Liu K. Tumour-targeting bacteria engineered to fight cancer. Nat Rev Cancer. 2018;18(12):727–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Forbes NS. Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer. 2010;10(11):785–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Katuri J, Ma X, Stanton MM, Sánchez S. Designing micro-and nanoswimmers for specific applications. Acc Chem Res. 2017;50(1):2–11.

    Article  CAS  PubMed  Google Scholar 

  25. Zhuang J, Sitti M. Chemotaxis of bio-hybrid multiple bacteria-driven microswimmers. Sci Rep. 2016;6(1):32135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhuang J, Wright Carlsen R, Sitti M. pH-taxis of biohybrid microsystems. Sci Rep. 2015;5(1):11403.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Kefayat A, Ghahremani F, Motaghi H, Rostami S, Mehrgardi MA. Alive attenuated Salmonella as a cargo shuttle for smart carrying of gold nanoparticles to tumour hypoxic regions. J Drug Target. 2019;27(3):315–24.

    Article  CAS  PubMed  Google Scholar 

  28. Felfoul O, Mohammadi M, Taherkhani S, De Lanauze D, Zhong XuY, Loghin D, Essa S, Jancik S, Houle D, Lafleur M, Gaboury L. Magneto-aerotactic bacteria deliver drug-containing nanoliposomes to tumour hypoxic regions. Nat Nanotechnol. 2016;11(11):941–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Takahashi M, Sukowati EW, Nomura S, Kato A, Mizuseki K, Watanabe Y, Mukai H. Impact of tumoral structure and bacterial species on growth and biodistribution of live bacterial therapeutics in xenografted tumours. J Drug Target. 2023;31(2):194–205.

    Article  CAS  PubMed  Google Scholar 

  30. Fu L, He Q, Lu X, Hu L, Qiang H, Pei P. Surface engineering on bacteria for tumor immunotherapy: strategies and perspectives. Adv Funct Mater. 2024. https://doi.org/10.1002/adfm.202405304.

    Article  Google Scholar 

  31. Montanaro J, Inic-Kanada A, Ladurner A, Stein E, Belij S, Bintner N, Schlacher S, Schuerer N, Mayr UB, Lubitz W, Leisch N. Escherichia coli Nissle 1917 bacterial ghosts retain crucial surface properties and express chlamydial antigen: an imaging study of a delivery system for the ocular surface. Drug Des Dev Ther. 2015;21:3741–54.

    Google Scholar 

  32. Langemann T, Koller VJ, Muhammad A, Kudela P, Mayr UB, Lubitz W. The bacterial ghost platform system: production and applications. Bioeng Bugs. 2010;1(5):326–36.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Henrich B, Lubitz W, Plapp R. Lysis of Escherichia coli by induction of cloned ϕX174 genes. Mol Gen Genet MGG. 1982;185:493–7.

    Article  CAS  PubMed  Google Scholar 

  34. Bläsi U, Linke RP, Lubitz W. Evidence for membrane-bound oligomerization of bacteriophage ϕ X174 lysis protein-E. J Biol Chem. 1989;264(8):4552–8.

    Article  PubMed  Google Scholar 

  35. Witte A, Bläsi U, Halfmann G, Szostak M, Wanner G, Lubitz W. PhiX174 protein E-mediated lysis of Escherichia coli. Biochimie. 1990;72(2–3):191–200.

    Article  CAS  PubMed  Google Scholar 

  36. Witte A, Wanner G, Sulzner M, Lubitz W. Dynamics of PhiX174 protein E-mediated lysis of Escherichia coli. Arch Microbiol. 1992;157:381–8.

    Article  CAS  PubMed  Google Scholar 

  37. Tabrizi CA, Walcher P, Mayr UB, Stiedl T, Binder M, McGrath J, Lubitz W. Bacterial ghosts–biological particles as delivery systems for antigens, nucleic acids and drugs. Curr Opin Biotechnol. 2004;15(6):530–7.

    Article  CAS  PubMed  Google Scholar 

  38. Huter V, Szostak MP, Gampfer J, Prethaler S, Wanner G, Gabor F, Lubitz W. Bacterial ghosts as drug carrier and targeting vehicles. J Control Release. 1999;61(1–2):51–63.

    Article  CAS  PubMed  Google Scholar 

  39. Haslberger AG, Kohl G, Felnerova D, Mayr UB, Fürst-Ladani S, Lubitz W. Activation, stimulation and uptake of bacterial ghosts in antigen presenting cells. J Biotechnol. 2000;83(1–2):57–66.

    Article  CAS  PubMed  Google Scholar 

  40. Chen Z, Liu Y, Yu Y, Yang S, Feng J, Zhu Y, Huang W, Qin B, Guan X, He Z, Sun M. Micro-to-nano oncolytic microbial system shifts from tumor killing to tumor draining lymph nodes remolding for enhanced immunotherapy. Adv Mater. 2023;16:2306488.

    Google Scholar 

  41. Paukner S, Kohl G, Lubitz W. Bacterial ghosts as novel advanced drug delivery systems: antiproliferative activity of loaded doxorubicin in human Caco-2 cells. J Control Release. 2004;94(1):63–74.

    Article  CAS  PubMed  Google Scholar 

  42. Stein E, Inic-Kanada A, Belij S, Montanaro J, Bintner N, Schlacher S, Mayr UB, Lubitz W, Stojanovic M, Najdenski H, Barisani-Asenbauer T. In vitro and in vivo uptake study of Escherichia coli Nissle 1917 bacterial ghosts: cell-based delivery system to target ocular surface diseases. Invest Ophthalmol Vis Sci. 2013;54(9):6326–33.

    Article  CAS  PubMed  Google Scholar 

  43. Koller VJ, Dirsch VM, Beres H, Donath O, Reznicek G, Lubitz W, Kudela P. Modulation of bacterial ghosts–induced nitric oxide production in macrophages by bacterial ghost-delivered resveratrol. FEBS J. 2013;280(5):1214–25.

    Article  CAS  PubMed  Google Scholar 

  44. Wang C, Zhong L, Xu J, Zhuang Q, Gong F, Chen X, Tao H, Hu C, Huang F, Yang N, Li J. Oncolytic mineralized bacteria as potent locally administered immunotherapeutics. Nat Biomed Eng. 2024;21:1–8.

    Google Scholar 

  45. Adler HI, Fisher WD, Cohen A, Hardigree AA. Miniature Escherichia coli cells deficient in DNA. Proc Natl Acad Sci. 1967;57(2):321–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Di Ventura B, Sourjik V. Self-organized partitioning of dynamically localized proteins in bacterial cell division. Mol Syst Biol. 2011;7(1):457.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Khachatourians GG, Clark DJ, Adler HI, Hardigree AA. Cell growth and division in Escherichia coli: a common genetic control involved in cell division and minicell formation. J Bacteriol. 1973;116(1):226–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Inselburg J. Replication of colicin E1 plasmid DNA in minicells from a unique replication initiation site. Proc Natl Acad Sci. 1974;71(6):2256–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. MacDiarmid JA, Amaro-Mugridge NB, Madrid-Weiss J, Sedliarou I, Wetzel S, Kochar K, Brahmbhatt VN, Phillips L, Pattison ST, Petti C, Stillman B. Sequential treatment of drug-resistant tumors with targeted minicells containing siRNA or a cytotoxic drug. Nat Biotechnol. 2009;27(7):643–51.

    Article  CAS  PubMed  Google Scholar 

  50. Mujahid MH, Upadhyay TK, Upadhye VJ. Targeted identification of antibacterial phytocompounds from plant extracts against multidrug-resistant bacteria: a systematic review.

  51. Kudela P, Koller VJ, Lubitz W. Bacterial ghosts (BGs)—advanced antigen and drug delivery system. Vaccine. 2010;28(36):5760–7.

    Article  CAS  PubMed  Google Scholar 

  52. Gao J, Chu D, Wang Z. Cell membrane-formed nanovesicles for disease-targeted delivery. J Control Release. 2016;28(224):208–16.

    Article  Google Scholar 

  53. Gao J, Dong X, Su Y, Wang Z. Human neutrophil membrane-derived nanovesicles as a drug delivery platform for improved therapy of infectious diseases. Acta Biomater. 2021;15(123):354–63.

    Article  Google Scholar 

  54. Gao J, Dong X, Wang Z. Generation, purification and engineering of extracellular vesicles and their biomedical applications. Methods. 2020;1(177):114–25.

    Article  Google Scholar 

  55. Wang S, Gao J, Li M, Wang L, Wang Z. A facile approach for development of a vaccine made of bacterial double-layered membrane vesicles (DMVs). Biomaterials. 2018;1(187):28–38.

    Article  Google Scholar 

  56. Wang S, Gao J, Wang Z. Outer membrane vesicles for vaccination and targeted drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11(2): e1523.

    Article  PubMed  Google Scholar 

  57. Toyofuku M, Nomura N, Eberl L. Types and origins of bacterial membrane vesicles. Nat Rev Microbiol. 2019;17(1):13–24.

    Article  CAS  PubMed  Google Scholar 

  58. Gerritzen MJ, Martens DE, Wijffels RH, van der Pol L, Stork M. Bioengineering bacterial outer membrane vesicles as vaccine platform. Biotechnol Adv. 2017;35(5):565–74.

    Article  CAS  PubMed  Google Scholar 

  59. Hu R, Liu H, Wang M, Li J, Lin H, Liang M, Gao Y, Yang M. An OMV-based nanovaccine confers safety and protection against pathogenic Escherichia coli via both humoral and predominantly Th1 immune responses in poultry. Nanomaterials. 2020;10(11):2293.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Kulp A, Kuehn MJ. Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu Rev Microbiol. 2010;13(64):163–84.

    Article  Google Scholar 

  61. Mujahid MH, Upadhyay TK, Khan F, Pandey P, Park MN, Sharangi AB, Saeed M, Upadhye VJ, Kim B. Metallic and metal oxide-derived nanohybrid as a tool for biomedical applications. Biomed Pharmacother. 2022;1(155):113791.

    Article  Google Scholar 

  62. Lee EY, Choi DS, Kim KP, Gho YS. Proteomics in gram-negative bacterial outer membrane vesicles. Mass Spectrom Rev. 2008;27(6):535–55.

    Article  CAS  PubMed  Google Scholar 

  63. Biller SJ, Schubotz F, Roggensack SE, Thompson AW, Summons RE, Chisholm SW. Bacterial vesicles in marine ecosystems. Science. 2014;343(6167):183–6.

    Article  CAS  PubMed  Google Scholar 

  64. Schertzer JW, Whiteley M. A bilayer-couple model of bacterial outer membrane vesicle biogenesis. MBio. 2012;3(2):10–128.

    Article  Google Scholar 

  65. Kohl P, Zingl FG, Eichmann TO, Schild S. Isolation of outer membrane vesicles including their quantitative and qualitative analyses. In: Sikora A, editor. Vibrio cholerae: methods and protocols. New York: Humana Press; 2018. p. 117–34.

    Chapter  Google Scholar 

  66. Alzahrani H, Winter J, Boocock D, De Girolamo L, Forsythe SJ. Characterization of outer membrane vesicles from a neonatal meningitic strain of Cronobacter sakazakii. FEMS Microbiol Lett. 2015;362(12): fnv085.

    Article  PubMed  Google Scholar 

  67. Badamchi A, Bahrami F, Tasbiti AH, Yari S, Shafiei M, Shahcheraghi F, Siadat SD. Immuno-proteomics analysis between OMV of vaccine and dominant wild type strains of Bordetella pertussis in Iran. Iran J Microbiol. 2020;12(2):77.

    PubMed  PubMed Central  Google Scholar 

  68. Schmitt S, Prokisch H, Schlunck T, Camp DG, Ahting U, Waizenegger T, Scharfe C, Meitinger T, Imhof A, Neupert W, Oefner PJ. Proteome analysis of mitochondrial outer membrane from Neurospora crassa. Proteomics. 2006;6(1):72–80.

    Article  CAS  PubMed  Google Scholar 

  69. McBroom AJ, Kuehn MJ. Release of outer membrane vesicles by Gram-negative bacteria is a novel envelope stress response. Mol Microbiol. 2007;63(2):545–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Eddy JL, Gielda LM, Caulfield AJ, Rangel SM, Lathem WW. Production of outer membrane vesicles by the plague pathogen Yersinia pestis. PLoS ONE. 2014;9(9): e107002.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Kim OY, Choi SJ, Jang SC, Park KS, Kim SR, Choi JP, Lim JH, Lee SW, Park J, Di Vizio D, Lötvall J. Bacterial protoplast-derived nanovesicles as vaccine delivery system against bacterial infection. Nano Lett. 2015;15(1):266–74.

    Article  CAS  PubMed  Google Scholar 

  72. Kim OY, Dinh NT, Park HT, Choi SJ, Hong K, Gho YS. Bacterial protoplast-derived nanovesicles for tumor targeted delivery of chemotherapeutics. Biomaterials. 2017;1(113):68–79.

    Article  Google Scholar 

  73. Kuerban K, Gao X, Zhang H, Liu J, Dong M, Wu L, Ye R, Feng M, Ye L. Doxorubicin-loaded bacterial outer-membrane vesicles exert enhanced anti-tumor efficacy in non-small-cell lung cancer. Acta Pharm Sin B. 2020;10(8):1534–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhang D, Qin X, Wu T, Qiao Q, Song Q, Zhang Z. Extracellular vesicles based self-grown gold nanopopcorn for combinatorial chemo-photothermal therapy. Biomaterials. 2019;1(197):220–8.

    Article  Google Scholar 

  75. Gao J, Wang S, Dong X, Wang Z. RGD-expressed bacterial membrane-derived nanovesicles enhance cancer therapy via multiple tumorous targeting. Theranostics. 2021;11(7):3301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Brown L, Wolf JM, Prados-Rosales R, Casadevall A. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol. 2015;13(10):620–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Liu Y, Defourny KA, Smid EJ, Abee T. Gram-positive bacterial extracellular vesicles and their impact on health and disease. Front Microbiol. 2018;9(9):1502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kim JH, Lee J, Park J, Gho YS. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin Cell Dev Biol. 2015;40:97–104.

    Article  CAS  PubMed  Google Scholar 

  79. Toyofuku M, Cárcamo-Oyarce G, Yamamoto T, Eisenstein F, Hsiao CC, Kurosawa M, Gademann K, Pilhofer M, Nomura N, Eberl L. Prophage-triggered membrane vesicle formation through peptidoglycan damage in Bacillus subtilis. Nat Commun. 2017;8(1):481.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Marsollier L, Brodin P, Jackson M, Korduláková J, Tafelmeyer P, Carbonnelle E, Aubry J, Milon G, Legras P, André JP, Leroy C. Impact of Mycobacterium ulcerans biofilm on transmissibility to ecological niches and Buruli ulcer pathogenesis. PLoS Pathog. 2007;3(5): e62.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Klieve AV, Yokoyama MT, Forster RJ, Ouwerkerk D, Bain PA, Mawhinney EL. Naturally occurring DNA transfer system associated with membrane vesicles in cellulolytic Ruminococcus spp. of ruminal origin. Appl Environ Microbiol. 2005;71(8):4248–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Tzipilevich E, Habusha M, Ben-Yehuda S. Acquisition of phage sensitivity by bacteria through exchange of phage receptors. Cell. 2017;168(1):186–99.

    Article  CAS  PubMed  Google Scholar 

  83. Rodriguez GM, Prados-Rosales R. Functions and importance of mycobacterial extracellular vesicles. Appl Microbiol Biotechnol. 2016;100:3887–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lee J, Lee EY, Kim SH, Kim DK, Park KS, Kim KP, Kim YK, Roh TY, Gho YS. Staphylococcus aureus extracellular vesicles carry biologically active β-lactamase. Antimicrob Agents Chemother. 2013;57(6):2589–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Olaya-Abril A, Prados-Rosales R, McConnell MJ, Martín-Peña R, González-Reyes JA, Jiménez-Munguía I, Gómez-Gascón L, Fernández J, Luque-García JL, García-Lidón C, Estévez H. Characterization of protective extracellular membrane-derived vesicles produced by Streptococcus pneumoniae. J Proteomics. 2014;25(106):46–60.

    Article  Google Scholar 

  86. Lee WH, Choi HI, Hong SW, Kim KS, Gho YS, Jeon SG. Vaccination with Klebsiella pneumoniae-derived extracellular vesicles protects against bacteria-induced lethality via both humoral and cellular immunity. Exp Mol Med. 2015;47(9): e183.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Eisenbach M. Bacterial chemotaxis. eLS. 2001. https://doi.org/10.1038/npg.els.0001251.

    Article  Google Scholar 

  88. Sitti M. Voyage of the microrobots. Nature. 2009;458(7242):1121–2.

    Article  CAS  PubMed  Google Scholar 

  89. Sitti M. Microscale and nanoscale robotics systems [grand challenges of robotics]. IEEE Robot Autom Mag. 2007;14(1):53–60.

    Article  Google Scholar 

  90. Diller E, Sitti M. Micro-scale mobile robotics. Found Trends® Robot. 2013;2(3):143–259.

    Article  Google Scholar 

  91. Vikram Singh A, Sitti M. Targeted drug delivery and imaging using mobile milli/microrobots: a promising future towards theranostic pharmaceutical design. Curr Pharm Des. 2016;22(11):1418–28.

    Article  Google Scholar 

  92. Carlsen RW, Sitti M. Bio-hybrid cell-based actuators for microsystems. Small. 2014;10(19):3831–51.

    Article  CAS  PubMed  Google Scholar 

  93. Kim D, Liu A, Diller E, Sitti M. Chemotactic steering of bacteria propelled microbeads. Biomed Microdevice. 2012;14:1009–17.

    Article  CAS  Google Scholar 

  94. Jo SD, Ku SH, Won YY, Kim SH, Kwon IC. Targeted nanotheranostics for future personalized medicine: recent progress in cancer therapy. Theranostics. 2016;6(9):1362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Au JL, Abbiati RA, Wientjes MG, Lu Z. Target site delivery and residence of nanomedicines: application of quantitative systems pharmacology. Pharmacol Rev. 2019;71(2):157–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mujahid MH, Upadhyay TK, Upadhye VJ, Mathad PS. Antioxidant, antimicrobial, antidiabetic, antiglycation, and biocompatibility potential of Aqueous Zingiber officinale Rhizome (AZOME) Extract.

  97. Stauber JM, Qian EA, Han Y, Rheingold AL, Král P, Fujita D, Spokoyny AM. An organometallic strategy for assembling atomically precise hybrid nanomaterials. J Am Chem Soc. 2019;142(1):327–34.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Ling D, Lee N, Hyeon T. Chemical synthesis and assembly of uniformly sized iron oxide nanoparticles for medical applications. Acc Chem Res. 2015;48(5):1276–85.

    Article  CAS  PubMed  Google Scholar 

  99. Das P, Fatehbasharzad P, Colombo M, Fiandra L, Prosperi D. Multifunctional magnetic gold nanomaterials for cancer. Trends Biotechnol. 2019;37(9):995–1010.

    Article  CAS  PubMed  Google Scholar 

  100. Zhu D, Roy S, Liu Z, Weller H, Parak WJ, Feliu N. Remotely controlled opening of delivery vehicles and release of cargo by external triggers. Adv Drug Deliv Rev. 2019;1(138):117–32.

    Article  Google Scholar 

  101. Chen L, Zhou L, Wang C, Han Y, Lu Y, Liu J, Hu X, Yao T, Lin Y, Liang S, Shi S. Tumor-targeted drug and CpG delivery system for phototherapy and docetaxel-enhanced immunotherapy with polarization toward M1-type macrophages on triple negative breast cancers. Adv Mater. 2019;31(52):1904997.

    Article  CAS  Google Scholar 

  102. Cun X, Chen J, Li M, He X, Tang X, Guo R, Deng M, Li M, Zhang Z, He Q. Tumor-associated fibroblast-targeted regulation and deep tumor delivery of chemotherapeutic drugs with a multifunctional size-switchable nanoparticle. ACS Appl Mater Interfaces. 2019;11(43):39545–59.

    Article  CAS  PubMed  Google Scholar 

  103. Cartmell T, Mitchell D, Lamond FJ, Laburn HP. Route of administration differentially affects fevers induced by Gram-negative and Gram-positive pyrogens in rabbits. Exp Physiol. 2002;87(3):391–9.

    Article  CAS  PubMed  Google Scholar 

  104. Cortes-Perez NG, Lefèvre F, Corthier G, Adel-Patient K, Langella P, Bermúdez-Humarán LG. Influence of the route of immunization and the nature of the bacterial vector on immunogenicity of mucosal vaccines based on lactic acid bacteria. Vaccine. 2007;25(36):6581–8.

    Article  CAS  PubMed  Google Scholar 

  105. Crull K, Bumann D, Weiss S. Influence of infection route and virulence factors on colonization of solid tumors by Salmonella enterica serovar Typhimurium. FEMS Immunol Med Microbiol. 2011;62(1):75–83.

    Article  CAS  PubMed  Google Scholar 

  106. Zhao M, Yang M, Li XM, Jiang P, Baranov E, Li S, Xu M, Penman S, Hoffman RM. Tumor-targeting bacterial therapy with amino acid auxotrophs of GFP-expressing Salmonella typhimurium. Proc Natl Acad Sci. 2005;102(3):755–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Rosenberg GA. Neurological diseases in relation to the blood–brain barrier. J Cereb Blood Flow Metab. 2012;32(7):1139–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Van Sorge NM, Doran KS. Defense at the border: the blood–brain barrier versus bacterial foreigners. Future Microbiol. 2012;7(3):383–94.

    Article  PubMed  Google Scholar 

  109. Zwagerman NT, Friedlander RM, Monaco EA III. Intratumoral Clostridium novyi as a potential treatment for solid necrotic brain tumors. Neurosurgery. 2014;75(6):N17–8.

    Article  PubMed  Google Scholar 

  110. Lee CH. Engineering bacteria toward tumor targeting for cancer treatment: current state and perspectives. Appl Microbiol Biotechnol. 2012;93:517–23.

    Article  CAS  PubMed  Google Scholar 

  111. Liu Y, Zhou M, Luo D, Wang L, Hong Y, Yang Y, Sha Y. Bacteria-mediated in vivo delivery of quantum dots into solid tumor. Biochem Biophys Res Commun. 2012;425(4):769–74.

    Article  CAS  PubMed  Google Scholar 

  112. Ganai S, Arenas RB, Sauer JP, Bentley B, Forbes NS. In tumors Salmonella migrate away from vasculature toward the transition zone and induce apoptosis. Cancer Gene Ther. 2011;18(7):457–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Loeffler M, Le’Negrate G, Krajewska M, Reed JC. Inhibition of tumor growth using Salmonella expressing Fas ligand. J Natl Cancer Inst. 2008;100(15):1113–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Yam C, Zhao M, Hayashi K, Ma H, Kishimoto H, McElroy M, Bouvet M, Hoffman RM. Monotherapy with a tumor-targeting mutant of S. typhimurium inhibits liver metastasis in a mouse model of pancreatic cancer. J Surg Res. 2010;164(2):248–55.

    Article  PubMed  Google Scholar 

  115. Sant S, Tao SL, Fisher OZ, Xu Q, Peppas NA, Khademhosseini A. Microfabrication technologies for oral drug delivery. Adv Drug Deliv Rev. 2012;64(6):496–507.

    Article  CAS  PubMed  Google Scholar 

  116. Sastry SV, Nyshadham JR, Fix JA. Recent technological advances in oral drug delivery—a review. Pharm Sci Technol Today. 2000;3(4):138–45.

    Article  CAS  PubMed  Google Scholar 

  117. Devriendt B, De Geest BG, Goddeeris BM, Cox E. Crossing the barrier: targeting epithelial receptors for enhanced oral vaccine delivery. J Control Release. 2012;160(3):431–9.

    Article  CAS  PubMed  Google Scholar 

  118. Correia-Pinto JF, Csaba N, Alonso MJ. Vaccine delivery carriers: insights and future perspectives. Int J Pharm. 2013;440(1):27–38.

    Article  CAS  PubMed  Google Scholar 

  119. Urbanska AM, Karagiannis ED, Au AS, Dai SY, Mozafari M, Prakash S. What’s next for gastrointestinal disorders: no needles? J Control Release. 2016;10(221):48–61.

    Article  Google Scholar 

  120. Berlec A, Ravnikar M, Štrukelj B. Lactic acid bacteria as oral delivery systems for biomolecules. Die Pharm Int J Pharm Sci. 2012;67(11):891–8.

    CAS  Google Scholar 

  121. Hanson ML, Hixon JA, Li W, Felber BK, Anver MR, Stewart CA, Janelsins BM, Datta SK, Shen W, McLean MH, Durum SK. Oral delivery of IL-27 recombinant bacteria attenuates immune colitis in mice. Gastroenterology. 2014;146(1):210–21.

    Article  CAS  PubMed  Google Scholar 

  122. Robert S, Gysemans C, Takiishi T, Korf H, Spagnuolo I, Sebastiani G, Van Huynegem K, Steidler L, Caluwaerts S, Demetter P, Wasserfall CH. Oral delivery of glutamic acid decarboxylase (GAD)-65 and IL10 by Lactococcus lactis reverses diabetes in recent-onset NOD mice. Diabetes. 2014;63(8):2876–87.

    Article  CAS  PubMed  Google Scholar 

  123. Guo S, Yan W, McDonough SP, Lin N, Wu KJ, He H, Xiang H, Yang M, Moreira MA, Chang YF. The recombinant Lactococcus lactis oral vaccine induces protection against C. difficile spore challenge in a mouse model. Vaccine. 2015;33(13):1586–95.

    Article  CAS  PubMed  Google Scholar 

  124. Ahmed B, Loos M, Vanrompay D, Cox E. Oral immunization with Lactococcus lactis-expressing EspB induces protective immune responses against Escherichia coli O157: H7 in a murine model of colonization. Vaccine. 2014;32(31):3909–16.

    Article  CAS  PubMed  Google Scholar 

  125. Wei P, Yang Y, Liu Z, Huang J, Gong Y, Sun H. Oral Bifidobacterium longum expressing alpha-melanocyte-stimulating hormone to fight experimental colitis. Drug Deliv. 2016;23(6):2058–64.

    Article  CAS  PubMed  Google Scholar 

  126. Takei S, Omoto C, Kitagawa K, Morishita N, Katayama T, Shigemura K, Fujisawa M, Kawabata M, Hotta H, Shirakawa T. Oral administration of genetically modified Bifidobacterium displaying HCV-NS3 multi-epitope fusion protein could induce an HCV-NS3-specific systemic immune response in mice. Vaccine. 2014;32(25):3066–74.

    Article  CAS  PubMed  Google Scholar 

  127. Yu Z, Huang Z, Sao C, Huang Y, Zhang F, Yang J, Lian J, Zeng Z, Luo W, Zeng W, Deng Q. Bifidobacterium as an oral delivery carrier of interleukin-12 for the treatment of Coxsackie virus B3-induced myocarditis in the Balb/c mice. Int Immunopharmacol. 2012;12(1):125–30.

    Article  CAS  PubMed  Google Scholar 

  128. Ning JF, Zhu W, Xu JP, Zheng CY, Meng XL. Oral delivery of DNA vaccine encoding VP28 against white spot syndrome virus in crayfish by attenuated Salmonella typhimurium. Vaccine. 2009;27(7):1127–35.

    Article  CAS  PubMed  Google Scholar 

  129. Chen G, Wei DP, Jia LJ, Tang B, Shu L, Zhang K, Xu Y, Gao J, Huang XF, Jiang WH, Hu QG. Oral delivery of tumor-targeting Salmonella exhibits promising therapeutic efficacy and low toxicity. Cancer Sci. 2009;100(12):2437–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Grillot-Courvalin C, Goussard S, Courvalin P, Silva A, Bodyak N, Vaze M, Sauer N, Laroux FS, Harborth J, Xiang S, Li CJ. Development of a therapeutic RNAi delivery system using nonpathogenic bacteria expressing inv and hly: transkingdom RNA interference (tkRNAi). Inhuman Gene Therapy. 2009;20(6):670–670.

    Google Scholar 

  131. Ivory K, Chambers SJ, Pin C, Prieto E, Arques JL, Nicoletti C. Oral delivery of Lactobacillus casei Shirota modifies allergen-induced immune responses in allergic rhinitis. Clin Exp Allergy. 2008;38(8):1282–9.

    Article  CAS  PubMed  Google Scholar 

  132. Huq T, Khan A, Khan RA, Riedl B, Lacroix M. Encapsulation of probiotic bacteria in biopolymeric system. Crit Rev Food Sci Nutr. 2013;53(9):909–16.

    Article  CAS  PubMed  Google Scholar 

  133. Sohail A, Turner MS, Coombes A, Bostrom T, Bhandari B. Survivability of probiotics encapsulated in alginate gel microbeads using a novel impinging aerosols method. Int J Food Microbiol. 2011;145(1):162–8.

    Article  CAS  PubMed  Google Scholar 

  134. Urbanska AM, Bhathena J, Prakash S. Live encapsulated Lactobacillus acidophilus cells in yogurt for therapeutic oral delivery: preparation and in vitro analysis of alginate–chitosan microcapsules. Can J Physiol Pharmacol. 2007;85(9):884–93.

    Article  CAS  PubMed  Google Scholar 

  135. Jiang T, Singh B, Maharjan S, Li HS, Kang SK, Bok JD, Cho CS, Choi YJ. Oral delivery of probiotic expressing M cell homing peptide conjugated BmpB vaccine encapsulated into alginate/chitosan/alginate microcapsules. Eur J Pharm Biopharm. 2014;88(3):768–77.

    Article  CAS  PubMed  Google Scholar 

  136. Mei L, He F, Zhou RQ, Wu CD, Liang R, Xie R, Ju XJ, Wang W, Chu LY. Novel intestinal-targeted Ca-alginate-based carrier for pH-responsive protection and release of lactic acid bacteria. ACS Appl Mater Interfaces. 2014;6(8):5962–70.

    Article  CAS  PubMed  Google Scholar 

  137. Cook MT, Tzortzis G, Khutoryanskiy VV, Charalampopoulos D. Layer-by-layer coating of alginate matrices with chitosan–alginate for the improved survival and targeted delivery of probiotic bacteria after oral administration. J Mater Chem B. 2013;1(1):52–60.

    Article  CAS  PubMed  Google Scholar 

  138. Lin J, Yu W, Liu X, Xie H, Wang W, Ma X. In vitro and in vivo characterization of alginate-chitosan-alginate artificial microcapsules for therapeutic oral delivery of live bacterial cells. J Biosci Bioeng. 2008;105(6):660–5.

    Article  CAS  PubMed  Google Scholar 

  139. Danino T, Prindle A, Kwong GA, Skalak M, Li H, Allen K, Hasty J, Bhatia SN. Programmable probiotics for detection of cancer in urine. Scie Transl Med. 2015;7(289):289ra84.

    Google Scholar 

  140. Bermudes D, Low B, Pawelek J. Tumor-targeted Salmonella: highly selective delivery vectors. In: Habib NA, editor. Cancer gene therapy past achievements and future challenges. New York: Springer; 2002. p. 57–63.

    Chapter  Google Scholar 

  141. Paton AW, Morona R, Paton JC. Bioengineered microbes in disease therapy. Trends Mol Med. 2012;18(7):417–25.

    Article  CAS  PubMed  Google Scholar 

  142. Ciabattini A, Pettini E, Andersen P, Pozzi G, Medaglini D. Primary activation of antigen-specific naive CD4+ and CD8+ T cells following intranasal vaccination with recombinant bacteria. Infect Immun. 2008;76(12):5817–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Ciabattini A, Giomarelli B, Parigi R, Chiavolini D, Pettini E, Aricò B, Giuliani MM, Santini L, Medaglini D, Pozzi G. Intranasal immunization of mice with recombinant Streptococcus gordonii expressing NadA of Neisseria meningitidis induces systemic bactericidal antibodies and local IgA. Vaccine. 2008;26(33):4244–50.

    Article  CAS  PubMed  Google Scholar 

  144. Izumo T, Maekawa T, Ida M, Noguchi A, Kitagawa Y, Shibata H, Yasui H, Kiso Y. Effect of intranasal administration of Lactobacillus pentosus S-PT84 on influenza virus infection in mice. Int Immunopharmacol. 2010;10(9):1101–6.

    Article  CAS  PubMed  Google Scholar 

  145. Kotnik T, Rems L, Tarek M, Miklavčič D. Membrane electroporation and electropermeabilization: mechanisms and models. Annu Rev Biophys. 2019;6(48):63–91.

    Article  Google Scholar 

  146. Zoaby N, Shainsky-Roitman J, Badarneh S, Abumanhal H, Leshansky A, Yaron S, Schroeder A. Autonomous bacterial nanoswimmers target cancer. J Control Release. 2017;10(257):68–75.

    Article  Google Scholar 

  147. Xie S, Zhang P, Zhang Z, Liu Y, Chen M, Li S, Li X. Bacterial navigation for tumor targeting and photothermally-triggered bacterial ghost transformation for spatiotemporal drug release. Acta Biomater. 2021;1(131):172–84.

    Article  Google Scholar 

  148. Nguyen VH, Kim HS, Ha JM, Hong Y, Choy HE, Min JJ. Genetically engineered Salmonella typhimurium as an imageable therapeutic probe for cancer. Can Res. 2010;70(1):18–23.

    Article  CAS  Google Scholar 

  149. Chou CK, Hung JY, Liu JC, Chen CT, Hung MC. An attenuated Salmonella oral DNA vaccine prevents the growth of hepatocellular carcinoma and colon cancer that express α-fetoprotein. Cancer Gene Ther. 2006;13(8):746–52.

    Article  CAS  PubMed  Google Scholar 

  150. Yoon W, Park YC, Kim J, Chae YS, Byeon JH, Min SH, Park S, Yoo Y, Park YK, Kim BM. Application of genetically engineered Salmonella typhimurium for interferon-gamma–induced therapy against melanoma. Eur J Cancer. 2017;1(70):48–61.

    Article  Google Scholar 

  151. Din MO, Danino T, Prindle A, Skalak M, Selimkhanov J, Allen K, Julio E, Atolia E, Tsimring LS, Bhatia SN, Hasty J. Synchronized cycles of bacterial lysis for in vivo delivery. Nature. 2016;536(7614):81–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Nguyen DH, Chong A, Hong Y, Min JJ. Bioengineering of bacteria for cancer immunotherapy. Nat Commun. 2023;14(1):3553.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Xing J, Yin T, Li S, Xu T, Ma A, Chen Z, Luo Y, Lai Z, Lv Y, Pan H, Liang R. Targeted cancer therapy: sequential magneto-actuated and optics-triggered biomicrorobots for targeted cancer therapy. Adv Funct Mater. 2021;31(11):2170074.

    Article  Google Scholar 

  154. Deng X, Yang W, Shao Z, Zhao Y. Genetically modified bacteria for targeted phototherapy of tumor. Biomaterials. 2021;1(272):120809.

    Article  Google Scholar 

  155. Min JJ, Nguyen VH, Kim HJ, Hong Y, Choy HE. Quantitative bioluminescence imaging of tumor-targeting bacteria in living animals. Nat Protoc. 2008;3(4):629–36.

    Article  CAS  PubMed  Google Scholar 

  156. Uebe R, Schüler D. Magnetosome biogenesis in magnetotactic bacteria. Nat Rev Microbiol. 2016;14(10):621–37.

    Article  CAS  PubMed  Google Scholar 

  157. Mowday AM, Copp JN, Syddall SP, Dubois LJ, Wang J, Lieuwes NG, Biemans R, Ashoorzadeh A, Abbattista MR, Williams EM, Guise CP. E. coli nitroreductase NfsA is a reporter gene for non-invasive PET imaging in cancer gene therapy applications. Theranostics. 2020;10(23):10548.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Wang W, Song J, Yu W, Chen M, Li G, Chen J, Chen L, Yu L, Chen Y. Engineering photothermal and H2S-producing living nanomedicine by bacteria-enabled self-mineralization. Adv Funct Mater. 2024. https://doi.org/10.1002/adfm.202400929.

    Article  PubMed  Google Scholar 

  159. Felgner S, Frahm M, Kocijancic D, Rohde M, Eckweiler D, Bielecka A, Bueno E, Cava F, Abraham WR, Curtiss R III, Häussler S. aroA-deficient Salmonella enterica serovar Typhimurium is more than a metabolically attenuated mutant. MBio. 2016;7(5):10–128.

    Article  Google Scholar 

  160. Paster E, Ryu WS. The thermal impulse response of Escherichia coli. Proc Natl Acad Sci. 2008;105(14):5373–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Zhang Q, Zeng Y, Zhao Y, Peng X, Ren E, Liu G. Bio-hybrid magnetic robots: from bioengineering to targeted therapy. Bioengineering. 2024;11(4):311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Nauts HC, Swift WE, Coley BL. The treatment of malignant tumors by bacterial toxins as developed by the late William B. Coley, MD, reviewed in the light of modern research. Cancer Res. 1946;6(4):205–16.

    CAS  PubMed  Google Scholar 

  163. Chen F, Zang Z, Chen Z, Cui L, Chang Z, Ma A, Yin T, Liang R, Han Y, Wu Z, Zheng M. Nanophotosensitizer-engineered Salmonella bacteria with hypoxia targeting and photothermal-assisted mutual bioaccumulation for solid tumor therapy. Biomaterials. 2019;1(214):119226.

    Article  Google Scholar 

  164. Chen Q, Bai H, Wu W, Huang G, Li Y, Wu M, Tang G, Ping Y. Bioengineering bacterial vesicle-coated polymeric nanomedicine for enhanced cancer immunotherapy and metastasis prevention. Nano Lett. 2019;20(1):11–21.

    Article  PubMed  Google Scholar 

  165. Li F, Zhu P, Zheng B, Lu Z, Fang C, Fu Y, Li X. A customized biohybrid presenting cascade responses to tumor microenvironment. Adv Mater. 2024;9:2404901.

    Article  Google Scholar 

  166. Chen W, Wang Y, Qin M, Zhang X, Zhang Z, Sun X, Gu Z. Bacteria-driven hypoxia targeting for combined biotherapy and photothermal therapy. ACS Nano. 2018;12(6):5995–6005.

    Article  CAS  PubMed  Google Scholar 

  167. Gao J, Su Y, Wang Z. Engineering bacterial membrane nanovesicles for improved therapies in infectious diseases and cancer. Adv Drug Deliv Rev. 2022;1(186):114340.

    Article  Google Scholar 

  168. Han JW, Choi YJ, Cho S, Zheng S, Ko SY, Park JO, Park S. Active tumor-therapeutic liposomal bacteriobot combining a drug (paclitaxel)-encapsulated liposome with targeting bacteria (Salmonella Typhimurium). Sens Actuators B Chem. 2016;1(224):217–24.

    Google Scholar 

  169. Ektate K, Munteanu MC, Ashar H, Malayer J, Ranjan A. Chemo-immunotherapy of colon cancer with focused ultrasound and Salmonella-laden temperature sensitive liposomes (thermobots). Sci Rep. 2018;8(1):13062.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Park BW, Zhuang J, Yasa O, Sitti M. Multifunctional bacteria-driven microswimmers for targeted active drug delivery. ACS Nano. 2017;11(9):8910–23.

    Article  CAS  PubMed  Google Scholar 

  171. Alapan Y, Yasa O, Schauer O, Giltinan J, Tabak AF, Sourjik V, Sitti M. Soft erythrocyte-based bacterial microswimmers for cargo delivery. Sci Robot. 2018;3(17): eaar4423.

    Article  PubMed  Google Scholar 

  172. Quispe-Tintaya W, Chandra D, Jahangir A, Harris M, Casadevall A, Dadachova E, Gravekamp C. Nontoxic radioactive Listeriaat is a highly effective therapy against metastatic pancreatic cancer. Proc Natl Acad Sci. 2013;110(21):8668–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Chandra D, Selvanesan BC, Yuan Z, Libutti SK, Koba W, Beck A, Zhu K, Casadevall A, Dadachova E, Gravekamp C. 32-Phosphorus selectively delivered by listeria to pancreatic cancer demonstrates a strong therapeutic effect. Oncotarget. 2017;8(13):20729.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Fan JX, Peng MY, Wang H, Zheng HR, Liu ZL, Li CX, Wang XN, Liu XH, Cheng SX, Zhang XZ. Engineered bacterial bioreactor for tumor therapy via Fenton-like reaction with localized H2O2 generation. Adv Mater. 2019;31(16):1808278.

    Article  Google Scholar 

  175. Zheng JH, Nguyen VH, Jiang SN, Park SH, Tan W, Hong SH, Shin MG, Chung IJ, Hong Y, Bom HS, Choy HE. Two-step enhanced cancer immunotherapy with engineered Salmonella typhimurium secreting heterologous flagellin. Sci Transl Med. 2017;9(376): eaak9537.

    Article  PubMed  Google Scholar 

  176. Nishikawa H, Sato E, Briones G, Chen LM, Matsuo M, Nagata Y, Ritter G, Jäger E, Nomura H, Kondo S, Tawara I. In vivo antigen delivery by a Salmonella typhimurium type III secretion system for therapeutic cancer vaccines. J Clin Investig. 2006;116(7):1946–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Jiang SN, Park SH, Lee HJ, Zheng JH, Kim HS, Bom HS, Hong Y, Szardenings M, Shin MG, Kim SC, Ntziachristos V. Engineering of bacteria for the visualization of targeted delivery of a cytolytic anticancer agent. Mol Ther. 2013;21(11):1985–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. MacDiarmid JA, Langova V, Bailey D, Pattison ST, Pattison SL, Christensen N, Armstrong LR, Brahmbhatt VN, Smolarczyk K, Harrison MT, Costa M. Targeted doxorubicin delivery to brain tumors via minicells: proof of principle using dogs with spontaneously occurring tumors as a model. PLoS ONE. 2016;11(4): e0151832.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Solomon BJ, Desai J, Rosenthal M, McArthur GA, Pattison ST, Pattison SL, MacDiarmid J, Brahmbhatt H, Scott AM. A first-time-in-human phase I clinical trial of bispecific antibody-targeted, paclitaxel-packaged bacterial minicells. PLoS ONE. 2015;10(12): e0144559.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Sagnella SM, Yang L, Stubbs GE, Boslem E, Martino-Echarri E, Smolarczyk K, Pattison SL, Vanegas N, Clair ES, Clarke S, Boockvar J. Cyto-immuno-therapy for cancer: a pathway elicited by tumor-targeted, cytotoxic drug-packaged bacterially derived nanocells. Cancer Cell. 2020;37(3):354–70.

    Article  CAS  PubMed  Google Scholar 

  181. Sagnella SM, Trieu J, Brahmbhatt H, MacDiarmid JA, MacMillan A, Whan RM, Fife CM, McCarroll JA, Gifford AJ, Ziegler DS, Kavallaris M. Targeted doxorubicin-loaded bacterially derived nano-cells for the treatment of neuroblastoma. Mol Cancer Ther. 2018;17(5):1012–23.

    Article  CAS  PubMed  Google Scholar 

  182. Whittle JR, Lickliter JD, Gan HK, Scott AM, Simes J, Solomon BJ, MacDiarmid JA, Brahmbhatt H, Rosenthal MA. First in human nanotechnology doxorubicin delivery system to target epidermal growth factor receptors in recurrent glioblastoma. J Clin Neurosci. 2015;22(12):1889–94.

    Article  CAS  PubMed  Google Scholar 

  183. Gujrati V, Prakash J, Malekzadeh-Najafabadi J, Stiel A, Klemm U, Mettenleiter G, Aichler M, Walch A, Ntziachristos V. Bioengineered bacterial vesicles as biological nano-heaters for optoacoustic imaging. Nat Commun. 2019;10(1):1114.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Carleton HA, Lara-Tejero M, Liu X, Galán JE. Engineering the type III secretion system in non-replicating bacterial minicells for antigen delivery. Nat Commun. 2013;4(1):1590.

    Article  PubMed  Google Scholar 

  185. Schetters ST, Jong WS, Horrevorts SK, Kruijssen LJ, Engels S, Stolk D, Daleke-Schermerhorn MH, Garcia-Vallejo J, Houben D, Unger WW, den Haan JM. Outer membrane vesicles engineered to express membrane-bound antigen program dendritic cells for cross-presentation to CD8+ T cells. Acta Biomater. 2019;1(91):248–57.

    Article  Google Scholar 

  186. Huang W, Shu C, Hua L, Zhao Y, Xie H, Qi J, Gao F, Gao R, Chen Y, Zhang Q, Li W. Modified bacterial outer membrane vesicles induce autoantibodies for tumor therapy. Acta Biomater. 2020;1(108):300–12.

    Article  Google Scholar 

  187. Chen Q, Huang G, Wu W, Wang J, Hu J, Mao J, Chu PK, Bai H, Tang G. A hybrid eukaryotic–prokaryotic nanoplatform with photothermal modality for enhanced antitumor vaccination. Adv Mater. 2020;32(16):1908185.

    Article  CAS  Google Scholar 

  188. Akin D, Sturgis J, Ragheb K, Sherman D, Burkholder K, Robinson JP, Bhunia AK, Mohammed S, Bashir R. Bacteria-mediated delivery of nanoparticles and cargo into cells. Nat Nanotechnol. 2007;2(7):441–9.

    Article  CAS  PubMed  Google Scholar 

  189. Xie S, Zhao L, Song X, Tang M, Mo C, Li X. Doxorubicin-conjugated Escherichia coli Nissle 1917 swimmers to achieve tumor targeting and responsive drug release. J Control Release. 2017;28(268):390–9.

    Article  Google Scholar 

  190. Hu Q, Wu M, Fang C, Cheng C, Zhao M, Fang W, Chu PK, Ping Y, Tang G. Engineering nanoparticle-coated bacteria as oral DNA vaccines for cancer immunotherapy. Nano Lett. 2015;15(4):2732–9.

    Article  CAS  PubMed  Google Scholar 

  191. Xie S, Chen M, Song X, Zhang Z, Zhang Z, Chen Z, Li X. Bacterial microbots for acid-labile release of hybrid micelles to promote the synergistic antitumor efficacy. Acta Biomater. 2018;15(78):198–210.

    Article  Google Scholar 

  192. Suh S, Jo A, Traore MA, Zhan Y, Coutermarsh-Ott SL, Ringel-Scaia VM, Allen IC, Davis RM, Behkam B. Nanoscale bacteria-enabled autonomous drug delivery system (NanoBEADS) enhances intratumoral transport of nanomedicine. Adv Sci. 2019;6(3):1801309.

    Article  Google Scholar 

  193. Li Q, Chen H, Feng X, Yu C, Feng F, Chai Y, Lu P, Song T, Wang X, Yao L. Nanoparticle-regulated semiartificial magnetotactic bacteria with tunable magnetic moment and magnetic sensitivity. Small. 2019;15(15):1900427.

    Article  Google Scholar 

  194. Moreno VM, Álvarez E, Izquierdo-Barba I, Baeza A, Serrano-López J, Vallet-Regí M. Bacteria as nanoparticles carrier for enhancing penetration in a tumoral matrix model. Adv Mater Interfaces. 2020;7(11):1901942.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Zheng DW, Chen Y, Li ZH, Xu L, Li CX, Li B, Fan JX, Cheng SX, Zhang XZ. Optically-controlled bacterial metabolite for cancer therapy. Nat Commun. 2018;9(1):1680.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Park W, Cho S, Huang X, Larson AC, Kim DH. Branched gold nanoparticle coating of clostridium novyi-NT spores for CT-guided intratumoral injection. Small. 2017;13(5):1602722.

    Article  Google Scholar 

  197. Luo CH, Huang CT, Su CH, Yeh CS. Bacteria-mediated hypoxia-specific delivery of nanoparticles for tumors imaging and therapy. Nano Lett. 2016;16(6):3493–9.

    Article  CAS  PubMed  Google Scholar 

  198. Dong H, Sarkes DA, Rice JJ, Hurley MM, Fu AJ, Stratis-Cullum DN. Living bacteria–nanoparticle hybrids mediated through surface-displayed peptides. Langmuir. 2018;34(20):5837–48.

    Article  CAS  PubMed  Google Scholar 

  199. Dogra N, Izadi H, Vanderlick TK. Micro-motors: a motile bacteria based system for liposome cargo transport. Sci Rep. 2016;6(1):29369.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Thamm DH, Kurzman ID, King I, Li Z, Sznol M, Dubielzig RR, Vail DM, MacEwen EG. Systemic administration of an attenuated, tumor-targeting Salmonella typhimurium to dogs with spontaneous neoplasia: phase I evaluation. Clin Cancer Res. 2005;11(13):4827–34.

    Article  CAS  PubMed  Google Scholar 

  201. Heimann DM, Rosenberg SA. Continuous intravenous administration of live genetically modified salmonella typhimurium in patients with metastatic melanoma. J Immunother. 2003;26(2):179.

    Article  PubMed  PubMed Central  Google Scholar 

  202. NCT00004216. ClinicalTrials.gov. http://www.clinicaltrials.gov/ct2/show/NCT00004216.

  203. NCT00358397. ClinicalTrials.gov. http://www.clinicaltrials.gov/ct2/show/NCT00358397.

  204. NCT01099631. ClinicalTrials.gov. http://www.clinicaltrials.gov/ct2/show/NCT01099631.

  205. NCT01118819. ClinicalTrials.gov. http://www.clinicaltrials.gov/ct2/show/NCT01118819.

  206. NCT01562626. ClinicalTrials.gov. http://www.clinicaltrials.gov/ct2/show/NCT01562626.

  207. NCT01924689. ClinicalTrials.gov. http://www.clinicaltrials.gov/ct2/show/NCT01924689.

Download references

Acknowledgements

Muhammad Ijaz and Ikram Hasan contributed equally to this manuscript. The authors thank for the support from the “Chunhui Plan” cooperative scientific research project of the Ministry of Education, China (HZKY20220312), the General Project of Guangdong Natural Science Foundation (2022A1515011781), the Science and Technology Innovation Commission of Shenzhen (JCYJ20210324132816039), and Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application (ZDSYS20220527171407017).

Funding

This work was supported by grants from the “Chunhui Plan” cooperative scientific research project of the Ministry of Education, China (HZKY20220312), the General Project of Guangdong Natural Science Foundation (2022A1515011781), the Science and Technology Innovation Commission of Shenzhen (JCYJ20210324132816039), and Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application (ZDSYS20220527171407017).

Author information

Authors and Affiliations

Authors

Contributions

Muhammad Ijaz and Ikram Hasan: conceptualization, writing—original draft, methodology, data curation. Tamoor Hamid Chaudhry: visualization, data curation. Rui Huang and Lan Zhang: writing—review and editing. Ziwei Hu and Qingqin Tan: writing—review and editing, supervision, resources. Bing Guo: investigation, writing—review and editing, Project administration, funding acquisition.

Corresponding authors

Correspondence to Ziwei Hu, Qingqin Tan or Bing Guo.

Ethics declarations

Consent for publication

All authors contributed to the article and approved the final manuscript.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ijaz, M., Hasan, I., Chaudhry, T.H. et al. Bacterial derivatives mediated drug delivery in cancer therapy: a new generation strategy. J Nanobiotechnol 22, 510 (2024). https://doi.org/10.1186/s12951-024-02786-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-024-02786-w

Keywords