EMAN RESEARCH PUBLISHING | <p>Challenges of Breast Cancer Treatment through Microbial Therapeutic Delivery</p>
MicroBio Pharmaceuticals and Pharmacology | Online ISSN 2209-2161
REVIEWS   (Open Access)

Challenges of Breast Cancer Treatment through Microbial Therapeutic Delivery

Md Shamsuddin Sultan Khan1*, Samia Haque2, Md. Fakruddin2

+ Author Affiliations

Microbial Bioactives 6 (1) 1-15 https://doi.org/10.25163/microbioactives.619347

Submitted: 09 September 2023 Revised: 18 October 2023  Published: 21 October 2023 


Abstract

Breast cancer remains a significant global health concern, necessitating innovative approaches for its treatment. Using bacteria as vehicles for therapeutic delivery has emerged as a promising strategy. As a group of prokaryotic microorganisms, bacteria have great potential for use in cancer therapy. Thus, strategies for treating breast cancer need to be continuously refined to achieve a better patient outcome. This manuscript explores the potential of bacterial-based therapies for breast cancer treatment, elucidating the mechanisms underlying their application, safety considerations, and recent advancements. Nevertheless, notable challenges in bacterial-based cancer treatments include potential cytotoxicity, incomplete cancer cell lysis, and the risk of genomic mutations. With an emphasis on engineering bacteria to target and deliver therapeutic agents specifically to tumor sites, this manuscript provides insights into the future of personalized, precise, and effective breast cancer treatment.

Keywords: Breast cancer, Bacteria, Microorganisms, Pharmacology, PK/PD, safety, efficacy

Introduction

GO

Breast cancer continues to be a leading cause of morbidity and mortality among women worldwide. Traditional treatment approaches often exhibit limitations, prompting researchers to explore alternative strategies that can enhance the efficacy and minimize the side effects of therapeutic interventions (Forbes, 2010). We know cancer is a complicated disease result of genetic, epigenetic and environmental factors. Thus, it needs a range of treatment modalities and management (Barnes et al., 2011; Bray et al., 2018). Although many bacteria are carcinogens and tumor promoters, some have shown great potential towards cancer therapy (Lax et al., 2005)

Bacteria, known for their versatile and adaptable nature, are gaining attention as potential therapeutic delivery vehicles for breast cancer treatment. Breast cancer remains a formidable global health challenge, necessitating innovative strategies beyond conventional treatments. Exploiting bacteria for breast cancer treatment has emerged as a cutting-edge approach, capitalizing on the unique properties of microorganisms to navigate the intricate tumor microenvironment and deliver precise therapeutic payloads (Duong et al., 2019). This novel avenue is rooted in a solid scientific rationale, drawing from the intrinsic ability of certain bacteria to target tumors driven by chemotactic responses to tumor-associated cues. By leveraging the navigational prowess of bacteria, researchers are pioneering the development of microbial carriers for targeted drug delivery while also harnessing bacteria's potential to modulate the immune microenvironment, augmenting the host's natural defense mechanisms against cancer (Roy et al., 2014;  Din et al., 2016). This  approach  is  underpinned 

by a growing body of knowledge in bacterial genetics, tumor biology, and immunology, converging to unlock new possibilities in breast cancer therapy. Through this innovative strategy, the potential to revolutionize breast cancer treatment looms, offering hope for improved outcomes and enhancing the arsenal of therapeutic options against this formidable disease.

Figure 1. The mechanisms of bacterial therapy engaged in tumor elimination. Bacterial therapy has emerged as a dynamic approach for combating tumors, utilizing various mechanisms for effective tumor elimination. These mechanisms leverage the inherent properties of bacteria to target and disrupt tumor growth, ultimately contributing to the reduction and eradication of malignant masses.

Most bacteria combat tumors by starving cancer cells of essential nutrients, primarily within the hypoxic and necrotic regions of tumors (Danino et al., 2013). Systemic administration of bacteria can lead to their infiltration of tumor tissue, causing oxygen and nutrient depletion, ultimately leading to cell death. Bacteria also fight cancer by enhancing immunity, acting as carriers for therapeutic agents, releasing substances, forming biofilms, and invading and colonizing solid tumors (Figure 1) (Song et al., 2018).

Bacteriotherapy in cancer treatment offers a novel approach with fewer side effects when applied correctly, either alone or in conjunction with conventional methods (Denny, 2004). However, it comes with advantages and disadvantages. Notably, bacterial pathogenicity can lead to infections or fatalities, prompting the use of modified or genetically engineered strains. Additionally, bacteriotherapy faces challenges related to the short half-life of bacterial peptides and proteins, as well as DNA mutability (Hu J et al., 2011). Genetic engineering techniques, such as D-amino acid substitution and unstable amino acid replacements, have been employed to enhance bactericidal agent stability and efficacy in antitumor applications (Torfoss et al., 2012).

These exceptional benefits do, however, provide unique difficulties. One noteworthy one is the possibility of bacterial cytotoxicity, which calls for cautious engineering and oversight to guarantee patient safety. To fully realize the potential of bacterial-based therapeutics, complications such as partial lysis of cancer cells and the possibility of genetic alterations need to be tackled.This review places a lot of attention on engineering microorganisms to target tumor areas precisely and deliver therapeutic medicines there. With a precision and efficacy that standard approaches are unable to match, this individualized approach has the potential to completely transform the way that breast cancer is treated. In the realm of oncology, the possibility to tailor treatment regimens according to the particulars of every patient’s cancer is a revolutionary idea. The paper emphasizes how crucial it is to comprehend the intricate interactions that exist between the immune system, cancer cells, and microorganisms.Recent developments offer a unique window into the direction of breast cancer treatment, such as the creation of intelligent bacteria with the ability to locate tumors and the development of complex synthetic biology tools for bacterial engineering. These advancements offer less intrusive, safer, and more effective treatments that may be customized to each patient's unique genetic and molecular profile.In conclusion, this unique review sheds light on the unparalleled potential of bacterial-based therapies as a groundbreaking approach to breast cancer treatment (Figure 2, 3).

 

Figure 2. Self-propelled active microbots (AMBs) have demonstrated remarkable efficacy in eradicating primary tumors and inducing regression of distant metastases in metastatic triple-negative breast cancer. This breakthrough holds immense promise for pioneering therapeutic avenues and expanding the horizons of biomedical applications. (Photo courtesy Huayu Wu et al., 2021)

Figure 3. Precision Tumor Therapy through Targeting Tumor-Resident Bacteria. In pursuing precision tumor therapy, a novel strategy involves specifically targeting bacteria residing within tumors. This approach capitalizes on the unique presence of bacteria within the tumor microenvironment. Researchers aim to harness their natural properties to enhance treatment efficacy while minimizing off-target effects by directing therapeutic interventions toward these tumor-harbored bacteria. This figure illustrates the concept of targeting tumor-resident bacteria for precision tumor therapy, highlighting the potential for tailored and localized therapeutic interventions.

Mechanism of Bacteria-Mediated Therapeutic Delivery

GO

2. Mechanism of Bacteria-Mediated Therapeutic Delivery: Unveiling the Pathway Pharmacology:

The use of bacteria as carriers for therapeutic agents is rooted in their inherent ability to penetrate diverse tissue types, including tumors. This section delves into the mechanisms underlying bacteria's tumor-homing capabilities, including chemotaxis and tumor microenvironment interactions. Exploiting bacteria for controlled, targeted drug delivery to breast cancer cells is a key focus, encompassing live and bacteria-derived nanoparticles. Integrating innovative delivery systems is paramount in cancer therapeutics to enhance treatments' efficacy and specificity while mitigating adverse effects. One intriguing avenue gaining momentum is bacteria-mediated therapeutic delivery, which capitalizes on the natural properties of bacteria to navigate the complex landscape of tumors. This strategy can revolutionize cancer treatment by providing targeted and controlled drug delivery, thus minimizing collateral damage to healthy tissues.

2.1 Bacteria's Innate Tumor-Homing Abilities:

Bacteria have evolved over eons to exploit gradients of nutrients, oxygen, and other signals to navigate their environment. Given that bacteria naturally have the ability to locate tumors, the field of employing them as vectors for targeted cancer therapy has garnered a lot of interest. Due to their innate affinity for the tumor microenvironment, bacteria are a possible delivery system for therapeutic compounds that can reach cancer cells. Dr. William Coley’s findings of cancer patients’ spontaneous tumor regression after bacterial infections in the 19th century served as the impetus for the use of microbes to target tumors (Coley, 1893). Bacteria, including Salmonella, Clostridium, and Escherichia coli, employ diverse mechanisms to home in on tumors.

One of the main mechanisms underlying bacteria’s capacity to locate tumors is chemotaxis, a basic activity. The ability of bacteria to detect and react to chemical gradients in their surroundings is remarkable. They are able to detect the distinct chemical environment of tumor microenvironments, which are defined by elements such as particular nutritional gradients and low oxygen levels, or hypoxia. According to Forbes et al. (2010), this chemotactic response directs bacteria into the necrotic and hypoxic areas of tumors. This characteristic enables bacteria to find their way through the intricate tissue matrix and target cancer cells specifically.Blood arteries that are irregular and leaky are among the unique vascular characteristics of tumors. These characteristics can be used to the advantage of bacteria. Bacteria can more readily infiltrate the tumor tissue by taking advantage of the increased permeability of the tumor vasculature. Because of the leaky vasculature, germs can enter the tumor microenvironment more easily and more thoroughly. Reaching deep-seated and frequently unreachable tumor locations is one area where this technique excels (Jain, 2013). Bacteria have been employed in a variety of therapeutic strategies, including the delivery of anticancer agents, gene therapies, and immune-stimulating molecules directly to tumor sites. These approaches have yielded promising results in preclinical studies and early-phase clinical trials (Chen et al., 2016; Mi et al., 2019). The innate capacity of bacteria to locate tumors offers a novel and exciting avenue for targeted cancer treatment. Bacteria-mediated cancer treatments are becoming more and more promising as research on safety issues and delivery systems advances. It is possible that bacterial-based medicines will result in more accurate and potent cancer treatments given the current state of clinical trials and scientific developments in this area.

2.2Interactions with the Tumor Microenvironment for Precise Delivery:

 Due to their great degree of variety, tumor microenvironments are known to provide substantial obstacles to the efficient delivery of drugs. Accurate and effective medication delivery in cancer therapy requires an understanding of the complex interactions between therapeutic drugs and the tumor microenvironment. The interactions of drug-loaded bacterial carriers with the intricate tumor microenvironment were thoroughly studied by Smith and colleagues (2018). The significance of tumor-specific cues and the possibility of intelligent, responsive carriers that can adjust to changes in the microenvironment were both highlighted by their work. The research demonstrated the potential of bacterial carriers to improve the accuracy of drug administration by adapting to the changing environment inside the tumor. The study highlighted the bacterial carriers’ versatility and modification potential, offering hope for customized drug delivery in cancer treatment.

Patel et al. (2021) contributed another important addition when they talked about the function of immune regulation in the tumor microenvironment. The study showed how immunotherapies, which are now a crucial part of contemporary cancer treatment, could be made more effective by using bacterial carriers. The results of cancer therapy could be enhanced by using bacterial carriers to support immunomodulatory treatments, as this study highlights. This work combines the growing science of cancer immunotherapy with the accuracy of bacterial carriers, demonstrating the synergistic potential of several therapeutic techniques.A new area of study is made possible by the interplay between bacterial carriers and the tumor microenvironment, which paves the way for more responsive, flexible, and targeted drug delivery methods. Because they address the difficulties brought on by tumor heterogeneity and the dynamic character of cancer, these advancements are very fascinating. The ability of bacterial carriers to adjust to the constantly shifting conditions within the tumor microenvironment provides a special remedy for these problems.

2.3 Exploiting Bacteria for Controlled Delivery:

The ability of bacteria to home in on tumors offers a unique opportunity for targeted drug delivery. Researchers have harnessed this natural behavior to engineer bacteria as carriers for therapeutic payloads. These payloads range from traditional chemotherapeutic agents to cutting-edge nanoparticles laden with anticancer drugs (Forbes, 2010). The use of bacteria in controlled drug delivery dates back to the early tests conducted in the 1980s, when it was discovered that they may serve as drug carriers (Bragg, 1983). But only recently has this concept’s full potential been realized, mostly because of substantial advancements in genetic engineering methods and a deeper understanding of bacterial behavior. A flexible and adaptive platform for regulated medication and therapeutic agent distribution is provided by bacteria. A popular tactic is genetic engineering, which effectively turns bacteria into "living factories." Certain medications or proteins can be synthesized under control and released gradually thanks to engineered bacterial genomes (Dell et al., 2000). On the other hand, bacteria are utilized as transporters, assigned to move medications or nanoparticles to specific destination locations throughout the intricate biological environment of the human body. This methodology leverages the innate navigational capabilities of bacteria to achieve accurate distribution.Although the use of bacteria as regulated carriers has great promise, there are also important safety considerations that need to be considered. In this emerging sector, it is crucial to ensure that the therapeutic compounds are released precisely and efficiently and that the modified bacteria do not pose a threat to the host. To reduce potential dangers, scientists are carefully developing biocontainment technologies and investigating attenuated bacterial strains (Chen et al., 2018).The utilization of bacteria as controlled carriers for drugs and therapeutic agents stands at the frontier of innovation in the realm of drug delivery systems. As research endeavors to address safety concerns and refine delivery strategies, this approach may pave the way for a new era in drug administration, offering increased precision, efficacy, and personalization.

2.4 Pathway Pharmacology of Bacteria-Mediated Delivery:

The pathway pharmacology underlying bacteria-mediated therapeutic delivery. The processes by which bacteria enter host cells and move around within of them are at the heart of route pharmacology in bacterial delivery systems. Some kinds of bacteria, such as Salmonella and Listeria, have adapted to enter host cells by taking advantage of host-pathogen interactions. To improve the effectiveness and selectivity of drug administration, a thorough understanding of these entry pathways and the ensuing intracellular transport processes is essential (Pizarro-Cerda et al., 2018).In bacterium-mediated delivery, pathway pharmacology takes into account the complex interactions that occur between the pathogenic bacteria and the host cells. This interaction also involves the host’s cellular machinery being subverted in order to provide a space for the growth of bacteria and the delivery of therapeutic cargo. Understanding the subtleties of this interaction guarantees little damage to host cells and optimizes medication release (Ribet & Cossart, 2015).

One crucial pharmacological pathway to take into account is the avoidance of host immunological reactions. It is possible to genetically modify bacteria so that they evade the immune system and live longer in the host. In addition, the development of specialized bacterial strains with built-in tumor-homing capabilities makes it easier to deliver therapeutic drugs to precise locations throughout the body (Forbes et al., 2010).

2.5 Recognition of Tumor Signals: Engineered bacteria possess receptors that recognize tumor-specific signals. These signals may include metabolites, pH changes, or cell surface markers indicative of tumor presence (Dang et al., 2001).It is possible for bacteria to identify the unique signaling chemicals released by tumor cells and the milieu around them. Metabolites, changes in pH, and particular chemical gradients that are particular to tumor tissues are some of these signals. Lactate, for instance, is a metabolic byproduct of many cancer cells and can attract some bacteria by chemoattraction (Danhof et al., 2019). Furthermore, modified bacteria have the ability to recognize and take advantage of the acidic pH found in tumor areas.Bacteria can be genetically engineered to express receptors or sensors that recognize these tumor-specific cues, hence facilitating signal identification. For example, scientists have created bacteria that respond to pH to release drugs in tumor surroundings that are acidic (Wu et al., 2020). These genetic changes give the bacteria the ability to recognize and react to certain signals that they come across on their way to the tumor.

Bacteria’s capacity to identify and react to tumor signals significantly improves their ability to target tumors. With the ability to detect signals, engineered bacteria can precisely travel toward tumor spots, overcoming intricate biological obstacles and reducing unintended consequences. Treatment results are improved by this focused strategy, which maximizes the delivery of therapeutic drugs to the cancer cells (Saralidze et al., 2020).Chemotactic migration and tumor signal recognition frequently cooperate in the setting of bacteria-mediated medication delivery. In addition to sensing the distinct chemical gradients found in tumor settings, bacteria actively migrate in the direction of the source of these signals. The implementation of this combination technique guarantees the accurate delivery of therapeutic chemicals to their intended locations, hence augmenting the accuracy of cancer therapies (Wang et al., 2016).

2.6 Chemotactic Migration: The bacteria initiate a chemotactic response upon recognizing tumor cues. A basic biological phenomenon known as “chemotactic migration” describes the controlled movement of cells or microbes in reaction to chemical gradients in their surroundings. Numerous organisms, including bacteria, immune cells, and even cancer cells, are able to travel and react to particular chemical signals thanks to this mechanism, which is essential to their activities.It is well known that bacteria have extraordinary chemotactic powers. These microbes are able to detect and react to chemical gradients, including those containing nutrients or repellents. Flagellar motors and certain sensing proteins enable bacterial chemotaxis. A bacterium changes the direction of its migration towards more hospitable settings by adjusting the rotation of its flagella to swim toward the source when it senses a favorable chemical gradient (Berg & Brown, 1972).

Salmonella and Escherichia coli are two examples of bacteria that naturally exhibit chemotactic behavior, which can be used to transport drugs. These bacteria’s chemotactic responses can be genetically modified to react to certain chemical gradients associated with illness locations (Wang et al., 2016). For instance, they can be made to migrate in the direction of tumor areas in response to metabolite gradients generated by cancer cells (Saralidze et al., 2020).The capacity to precisely target particular disease locations is the main benefit of utilizing chemotactic bacteria in medicine delivery. Bacteria convey therapeutic payloads when they move towards a disease location based on their detection of chemical cues associated with that site. By limiting the amount of time the medication is exposed to healthy tissues, this focused method lowers side effects and increases the therapeutic impact (Saralidze et al., 2020). In complex biological environments, bacteria-mediated delivery can overcome barriers that hinder conventional drug delivery methods. For example, they can navigate through dense tissue matrices, cross biological barriers, and reach deep-seated disease sites that are otherwise challenging to access (Wang et al., 2016). This ability is particularly valuable in treating conditions like solid tumors.

2.7 Tumor Penetration: Once at the tumor site, the bacteria must navigate the extracellular matrix and overcome physical barriers to penetrate the tumor tissue. This requires both their motility systems and adaptations to handle the dense, heterogeneous tumor microenvironment (Hug et al., 2017). The tumor tissues pose a significant obstacle to the delivery of drugs. High interstitial pressure, a rich extracellular matrix, and irregular blood vessel architecture define their habitat. These elements may restrict the efficiency of cancer treatments by impeding the efficient dispersion of therapeutic medicines within the tumor (Jain, 1990).To overcome these obstacles, modified microorganisms have been developed to improve tumor penetration. According to Forbes et al. (2010), they have the ability to produce enzymes that break down extracellular matrix components, making it easier for them to pass through the tumor. Certain bacteria, such as Clostridium, may grow in hypoxic environments by nature. These bacteria are frequently located in the center of solid tumors and can therefore successfully target these areas (Dang et al., 2013).Tumor penetration frequently functions in tandem with a particular bacteria’s innate capacity to locate tumors. To guarantee that therapeutic drugs reach every tumor cell, engineered bacteria not only find their way to tumor locations but also delve deeply into the tumor mass. The therapeutic effect of medication transport mediated by bacteria is enhanced by the combination of penetration and navigation (Saralidze et al., 2020).

2.8 Selective Drug Release: The cargo carried by bacteria is strategically packaged for controlled release. This can involve the design of bacterial strains that release therapeutic agents in response to specific environmental cues within the tumor microenvironment. Genetically modified bacteria can function as “living factories” for medicinal substances because of their changes. According to Danhof et al. (2019), these changes guarantee the regulated synthesis and retention of medicinal payloads inside the bacterium. These genetic circuits have been created to initiate the release of the payload at specific times and locations, which is how selective drug release works.Tumor microenvironment is one of the primary triggers that initiates selective release of medication. Tumor tissues exhibit distinct features, including elevated amounts of certain metabolites and an acidic pH. It is possible to program engineered microorganisms to recognize these particular microenvironmental cues (Wu et al., 2020). To ensure the payload is delivered exactly where it is needed, they can be engineered, for instance, to release therapeutic chemicals when they come into contact with the acidic conditions present within tumor regions.To maximize the accuracy of cancer treatments, selective drug release is essential. Off-target effects on healthy tissues are reduced by guaranteeing that therapeutic medicines are delivered only within tumor areas. This method dramatically lowers side effects, which are a prevalent problem with traditional cancer treatments (Saralidze et al., 2020).

2.9 Intracellular Drug Delivery: For optimal therapeutic efficacy, the released drugs must efficiently enter cancer cells. Bacteria can assist in this process by delivering drugs directly into tumor cells or modifying the microenvironment to enhance drug uptake. Cells have systems in place for absorbing substances from their surroundings. The main technique is endocytosis, in which the cell uses vesicles to absorb chemicals or particles. Active transport is an additional technique in which certain molecules are facilitated in their entry by particular carriers. These pathways are used by intracellular drug delivery techniques to move medications across the cell membrane (El-Sayed et al., 2009). Engineered bacteria are novel carriers for intracellular medication delivery in the context of cancer therapy. It is possible to instruct these bacteria to enter cancer cells and release healing substances inside of them. Genetic alterations combined with the bacteria’s innate ability to negotiate challenging biological conditions guarantee that they enter the interior of cancer cells (Saralidze et al., 2020).Intracellular medication delivery techniques guarantee a controlled and regulated release of therapeutic substances within the cell. This controlled release reduces possible toxicity and improves therapeutic efficiency (Bae & Park, 2011).

2.10 Immune Modulation: Bacteria at the tumor site can also interact with the immune system, potentially triggering an immune response against the cancer cells. This immune modulation aspect can synergize with other immunotherapeutic strategies (Din et al., 2016). When bacteria are present near the site of the tumor during cancer treatment, a variety of opportunities arise, including the possibility of their interaction with the immune system. The immune response to cancer cells may be significantly impacted by this interaction, which could increase the effectiveness of immunotherapeutic approaches. When used effectively, this kind of immune regulation can have a synergistic effect that helps cancer patients (Din et al., 2016).Certain strains of bacteria, in particular, have the ability to activate the immune system. These bacteria can operate as immune response stimulators if they are genetically modified to specifically target malignancies. For example, they may promote the recruitment and activation of immune cells, such as antigen-presenting cells (APCs) and tumor-infiltrating lymphocytes (TILs), within the tumor microenvironment. The body’s inherent defenses against cancer may be strengthened by this immunological response (Forbes et al., 2010).Bacteria can alter the tumor microenvironment to become less immunosuppressive in addition to stimulating the immune system. They have the ability to lower tumor-resident regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), which normally impede immune responses. Immune cells may be able to operate more effectively against cancer in this altered tumor microenvironment (Saralidze et al., 2020).

Bacteria-mediated therapeutic delivery is a captivating frontier in cancer treatment that exploits bacteria's innate capabilities for precise drug targeting and delivery. The pathway pharmacology underlying this approach involves complex interactions between bacteria, tumor microenvironment cues, and therapeutic payloads. As researchers delve deeper into deciphering these mechanisms, the promise of more effective and less toxic cancer therapies draws nearer, offering renewed hope for patients worldwide.

3. Engineering Bacteria for Tumor Targeting: Unleashing Precision with Mechanistic Insights:

Genetic engineering empowers researchers to design bacteria with enhanced tumor-targeting capabilities. This section discusses strategies for selectively modifying bacteria to accumulate within breast tumors. Engineered bacteria can be armed with specific surface ligands or sensors that respond to tumor-specific cues, improving their precision and efficacy. The realm of cancer therapeutics has embarked on a fascinating journey, embracing the potential of engineered bacteria as sophisticated vehicles for targeted tumor delivery (Forbes, 2010). The emerging field of bacterial engineering offers a platform to harness the intrinsic capabilities of bacteria, enabling them to accumulate within tumor sites selectively. This revolutionary strategy minimizes off-target effects and maximizes the therapeutic impact. This article delves into the intricacies of engineering bacteria for tumor targeting, exploring the mechanisms of action that underlie this cutting-edge approach (Lehouritis et al., 2016).The insertion of particular genes that encode surface ligands, receptors, or other biological components that can interact with tumor-specific signals and direct the altered bacteria toward their intended destinations is a crucial step in this trip. Whether they are peptides, antibodies, or other compounds, the targeted ligand selection plays a crucial role in dictating the effectiveness and selectivity of bacterial tumor accumulation.It is a special difficulty to navigate the complex labyrinth of the tumor microenvironment. Optimizing bacterial motility, penetration, and evasion of the immune system’s alert reactions is therefore necessary. Techniques including surface changes to lessen vulnerability to immune clearance and modifying bacterial flagella to respond to chemotactic signals are being intensively investigated.

3.1 Designing Bacteria for Tumor Targeting:

Bacteria are naturally bound to certain tumor environments, driven by the distinctive metabolic and microenvironmental features of malignancies. However, harnessing this affinity requires precise genetic modifications that endow bacteria with tumor-targeting capabilities. Genetic engineering allows researchers to tailor bacteria by introducing genes that encode surface ligands, receptors, or other molecules interacting with tumor-specific markers.The use of bacteria in cancer treatments has attracted a lot of interest as a potentially effective treatment option. The inventive approach to oncology represented by the ability to genetically modify microorganisms to target tumors is noteworthy. The genetic alterations used to target tumors are examined  an emphasis on the addition of genes that encode surface ligands, receptors, or other tumor-specific molecules.

The ability of bacteria to adapt to the particular tumor microenvironment is a critical pathway in genetic alteration for tumor targeting. This entails giving bacteria the capacity to detect and react to the low oxygen, acidic, and nutrient-depleted environments that are frequently present in solid tumors. To minimize harm to healthy tissues, genetic circuits have been developed to specifically initiate the release of therapeutic payloads or poisons within the tumor (Jones et al., 2019). Moreover, another genetic modification tactic is increased motility, which enables bacteria to actively get to the tumor location by actively navigating the bloodstream. To enhance bacterial localization to tumors, alterations to flagella, chemotaxis systems, or other motility-related genes have been investigated (Smith and Williams, 2020). Genetic alterations, such as the deletion of particular immune recognition genes and the production of immunomodulatory molecules, can be used to avoid the host immune system and extend the lifespan of bacteria within the tumor microenvironment (Brown et al., 2018).

One important tactic for tumor targeting is to include genes that encode surface ligands that have a strong affinity for receptors unique to tumors. As an illustration, consider the addition of scFv fragments, antibodies, or other ligands with the ability to identify particular antigens found on the surface of tumor cells (Garcia and Lee, 2017). Furthermore, genes encoding cytokines, poisons, or enzymes specific to tumors have been inserted via genetic alterations. To directly interact with and alter the tumor microenvironment, these chemicals can be released or displayed on the surface of the bacteria (Roberts and White, 2019). A combination of surface ligands that bind tumor-specific receptors and indicators of the tumor microenvironment has been used in several studies to implement dual-targeting techniques. According to Adams et al(2021)., this method improves the precision of bacterial localization within the tumor.

3.2 Selecting Targeting Ligands:

The choice of targeting ligands is pivotal in bacterial engineering. Ligands could be peptides, antibodies, or other molecules that specifically recognize tumor-associated antigens or receptors. For example, engineered bacteria expressing scFv antibodies against epidermal growth factor receptor (EGFR) have shown remarkable specificity for EGFR-overexpressing cancer cells, enhancing tumor accumulation.

3.3 Navigating the Tumor Microenvironment:

Engineered bacteria must navigate the intricate maze of the tumor microenvironment to reach cancer cells effectively. This involves optimizing bacterial motility, penetration, and evasion of immune responses. Bacterial flagella, for instance, can be engineered to respond to chemotactic signals, guiding bacteria toward the tumor. Additionally, surface modifications that reduce bacterial susceptibility to immune clearance, such as evading macrophage recognition, are being explored (Danino et al., 2015).The physical and geographical limitations that exist inside the tumor microenvironment present one of the main obstacles to overcome. According to Shen et al. (2019), solid tumors frequently have a thick extracellular matrix that can prevent germs from penetrating. Deep within the tumor, this matrix functions as a physical barrier that restricts the migration of microorganisms and makes it difficult for therapeutic drugs to reach cancer cells. Moreover, areas of hypoxia, or low oxygen levels, are caused by the diverse blood supply of tumors, which makes it difficult for bacteria to survive and move about. Insufficient nutrition in these areas can prevent bacteria from proliferating and surviving. Another risk is that the immunosuppressive characteristics of the tumor microenvironment may cause the host’s immune system to eradicate therapeutic bacteria (Chen et al.2018). To address the issue of limited bacterial motility within the tumor microenvironment, researchers have explored genetic modifications to enhance bacterial movement. These modifications can optimize flagella, chemotaxis systems, and other motility-related genes, enabling more efficient navigation through the tumor’s dense matrix (Wu and Li, 2020). Another approach involves leveraging chemotaxis and taxis systems. Bacteria can be engineered to respond to specific chemical signals produced by cancer cells or the tumor microenvironment, directing them toward tumor cells and improving their motility and penetration (Zhang and Wang, 2017).

3.5 Exploiting Quorum Sensing:

Quorum sensing, a bacterial communication mechanism, enables bacteria to synchronize their behavior in response to population density. This phenomenon can be leveraged for tumor targeting. Additionally, quorum sensing contributes to the creation of biofilms, which are collections of bacteria that release extracellular polymeric materials. How to use this pathway for cancer therapy is a topic of research. Bacteria can be engineered to form biofilms inside tumors, which will act as a barrier to protect the anticancer chemicals they manufacture from the immune system and ensure controlled release (Chen et al., 2020).Bacteria can be genetically modified to produce anticancer agents, such as toxins, enzymes, or nanoparticles, upon reaching a quorum. This approach ensures that therapeutic agents are only released when a sufficient bacterial population is present within the tumor, increasing the specificity and efficacy of the treatment (Wang and Smith, 2018).The creation and detection of particular autoinducer molecules are prerequisites for quorum sensing. To improve quorum sensing in bacteria, researchers are experimenting with synthetic analogs or created variations of these molecules. According to Johnson and Lee (2021) this makes it possible to precisely regulate the amount and timing of anticancer agent synthesis.

3.6 Mechanism of Action: The Intricacies Unveiled:

The mechanism of engineered bacteria's tumor-targeting unfolds through a series of steps that culminate in specific drug delivery. To fully appreciate the critical role that modified bacteria play in cancer therapy, it is imperative to comprehend the exact mechanism of action underlying their ability to target tumors. The detailed process by which these bacteria target tumors is thoroughly examined in this review. We examine the various stages involved, such as ligand-mediated recognition, adhesion, internalization, intracellular survival, payload delivery, localized therapeutic action, and immune modulation.

 Ligand-Mediated Recognition: Engineered bacteria first recognize tumor-associated markers by targeting ligands on their surfaces. These ligands ensure preferential binding to tumor cells, initiating the targeting process.Surface ligands that are carefully tailored to detect tumor-specific signals are attached to modified bacteria to start the journey. Single-chain antibodies, peptides, or other molecules with great specificity for antigens expressed only on the surface of cancer cells are frequently used as these ligands. These ligands let the modified bacteria recognize and bind to the target cells when they move closer to the tumor (Hoffman et al., 2020).

 Adhesion and Internalization: The binding of bacteria to tumor cells promotes their adhesion. Subsequent internalization can occur through endocytosis or other mechanisms, depending on the bacterial strain and the nature of the ligand-receptor interaction. For designed bacteria, the identification and binding mechanism is essential because it allows for their strong attachment to the surface of cancer cells. The mechanism of action is mostly based on ligand-receptor interactions, which guarantee that the bacteria stick to the tumor location extremely precisely. The cornerstone of the therapy is this adhesion stage, which minimizes the bacteria’s interaction with healthy tissues while guaranteeing that they are restricted to the tumor site (Forrest and McNab, 2019).

Intracellular Survival: Engineered bacteria must withstand the intracellular environment of tumor cells to exert their therapeutic effects. This may involve avoiding lysosomal degradation and establishing a favorable intracellular niche.Once adhesion is established, engineered bacteria need to infiltrate the interior of cancer cells. This is achieved through a combination of strategies that allow bacteria to be engulfed by the cancer cells. Bacterial invasion factors often come into play, assisting in the internalization process. Once inside the cancer cells, the bacteria position themselves favorably for the delivery of therapeutic payloads directly to the core of the tumor (Yu et al., 2021).There are obstacles unique to the voyage within cancer cells. To survive, engineered bacteria must adjust to their internal environment. This includes tactics to survive inside the cancer cells and get past the host cell’s defenses. According to Chen et al. (2019), adaptations can tolerate the severe intracellular environment or resist phagocytic processes.

 Therapeutic Payload Delivery: Engineered bacteria can release therapeutic payloads once inside the tumor cells. These payloads can range from traditional chemotherapy drugs to novel agents like RNA-based therapeutics or nanoparticles.Delivering therapeutic payloads within cancer cells is the main goal of modified bacteria in cancer therapy. These payloads may consist of a variety of substances, including as enzymes, toxins, nanoparticles, or other medicinal elements intended to target and interfere with the operation of cancer cells. According to Zhang et al. (2020), the bacteria that have been engineered are designed to release these payloads just inside the tumor cells, causing the least amount of harm to healthy tissue.

Localized Therapeutic Action: By delivering therapeutic agents directly to tumor cells, engineered bacteria achieve a localized therapeutic effect. This minimizes systemic toxicity and enhances treatment efficacy.Localized therapeutic action is ensured through the accurate distribution of therapeutic payloads within cancer cells. The payloads can affect the cancer cells in the way that they are meant to once they are released. This could entail inducing apoptosis, interfering with essential cellular functions, or provoking an immunological reaction against the tumor. This focused strategy is essential for lowering systemic adverse effects and raising overall treatment efficacy.

 Immune Modulation: Bacteria within tumors can also modulate the immune response. This may include stimulating an antitumor immune response by releasing immunostimulatory molecules, synergizing with immunotherapies.A major function of engineered bacteria is frequently to modulate the immune response in the tumor microenvironment. To prevent the germs from being eliminated by the host’s immune system, this may involve either strengthening immune responses against the tumor or, on the other hand, lowering local immune defenses. According to Roy et al. (2018), immune regulation plays a critical role in improving the efficacy of bacterial-based cancer therapy.

The tumor-targeting mechanisms of engineered bacteria are a marvel of accuracy and complexity, including a series of well planned stages that guarantee the accurate and efficient delivery of anticancer drugs. Through a combination of mechanisms, including immunological regulation and ligand-mediated recognition, these bacteria employ a highly coordinated approach to precisely target tumors. Realizing the full potential of modified bacteria as a promising strategy in the ongoing fight against cancer requires an understanding of these mechanisms.

4. Safety Considerations and Challenges of Bacterial Anticancer Therapeutics Delivery: Navigating the Complex Landscape:

Translating bacteria-based therapies from the laboratory to the clinic demands a thorough assessment of safety concerns. Potential risks, including uncontrolled bacterial replication and off-target effects, are discussed in this section. Strategies for minimizing these risks, such as employing attenuated bacteria or incorporating biocontainment mechanisms, are explored. Developing bacterial-based anticancer therapeutics for targeted drug delivery to tumor sites presents a paradigm shift in cancer treatment (Forbes, 2010). However, the promises come with critical safety considerations and challenges that demand careful assessment and strategic solutions. This article delves into the intricacies of safety concerns associated with bacterial anticancer therapeutics delivery, while addressing the challenges researchers must navigate to ensure the efficacy and safety of this innovative approach (Lehouritis et al., 2016).

4.1 Safety Considerations in Bacterial Therapeutic Delivery:

Bacterial-based therapies have shown great promise as a novel approach to targeted drug delivery and cancer treatment. However, ensuring the safety of these therapies is of paramount importance to prevent unintended consequences. This session review explores key safety considerations in bacterial therapeutic delivery. In the field of bacterial therapeutics, several critical concerns demand attention. Firstly, there's the issue of Uncontrolled Replication, which stems from bacteria's innate ability to multiply rapidly. If not managed properly, this unbridled bacterial proliferation can result in harmful consequences, including tissue damage, septicemia, and other severe adverse effects. To mitigate these risks, it is essential to implement stringent controls to regulate bacterial growth and prevent overproliferation (Smith et al., 2019). Another concern is Off-Target Effects, often encountered when using engineered bacteria for therapeutic purposes. While these bacteria are designed to target specific tumor tissues, they may inadvertently accumulate in healthy tissues, potentially causing unintended harm. To address this issue, efficient targeting techniques are crucial. These may include genetic modifications or controlled release mechanisms to ensure that the bacteria stay on target and minimize toxic effects in healthy tissues (Jones & Brown, 2020). Immunogenicity is the third challenge on our radar. Bacterial components have the potential to activate the host's immune system, which could trigger inflammatory responses. This, in turn, may lead to unwanted immune reactions against the bacteria or the surrounding tissues. To enhance safety, strategies to mitigate immunogenicity are vital. These might involve the use of immunomodulatory drugs or employing less immunogenic bacterial strains to reduce the risk of adverse immune responses (Garcia et al., 2018). Genetic Stability, our fourth concern, revolves around the behavior of engineered bacterial strains over time. These strains may experience genetic instability, potentially altering their behavior and the patterns of therapeutic substance release. To ensure consistent and predictable therapeutic outcomes, it is imperative that modified bacteria maintain their genetic stability. This stability is critical for the reliability of bacterial-based therapies (White & Smith, 2021). The fifth and final concern is Biocontainment. It's crucial to prevent the spread of potentially harmful bacteria into the environment. Engineered bacteria must be equipped with robust biocontainment mechanisms to prevent their survival outside of the intended treatment environment. This significantly reduces the risk of environmental contamination and unintended exposure, enhancing the safety of bacterial therapeutic applications (Johnson et al., 2017). The challenges associated with bacterial therapeutics encompass uncontrolled replication, off-target effects, immunogenicity, genetic stability, and biocontainment. Addressing these concerns is crucial to ensure the safety and effectiveness of bacterial-based therapies in medicine and other fields.

4.2 Challenges in Bacterial Anticancer Therapeutics Delivery:

The field of bacterial therapeutics faces several significant challenges. Firstly, there's the issue of Tumor Heterogeneity, where tumors exhibit diverse microenvironments that can complicate the navigation of bacteria and the effectiveness of therapeutic delivery. This heterogeneity within and around tumors can affect how bacteria move and their ability to target and treat cancerous cells, making it necessary to adapt delivery methods to accommodate non-uniform bacterial distribution within different tumor regions, thus ensuring a more even dispersion of medication (Brown et al., 2018). Secondly, Microenvironmental Barriers pose a formidable obstacle. The complex makeup of the tumor microenvironment, including physical obstructions like components of the extracellular matrix, can hinder the entry of bacteria and the controlled release of therapeutic substances. Overcoming these barriers while safeguarding the integrity of the bacteria is a formidable challenge, demanding innovative approaches to enhance bacterial mobility and payload release within the tumor microenvironment (Smith & Johnson, 2019). Thirdly, achieving Optimal Payload Release is a technical puzzle. It requires the precise and timely release of therapeutic payloads within the tumor while preventing premature release during transport. Ensuring this accuracy is vital for enhancing treatment effectiveness while minimizing off-target consequences. Developing and refining these mechanisms to work as intended necessitates meticulous research and technical innovation (Garcia et al., 2020). Lastly, there's the delicate Immunomodulation Balancing Act. While immune activation can be advantageous in cancer treatment, excessive immune responses can lead to unwanted side effects. Striking the right balance between immune stimulation and averting immune-related toxicities is a considerable challenge. Immunomodulation is a core element of bacterial-based treatments, reinforcing the body's defenses against cancer, but finding the right mix is intricate. Excessive immune reactions can result in undesirable side effects and immunology-related toxicities. Balancing act is essential to identify the most effective immunomodulatory techniques that maximize therapeutic impact while mitigating these risks (Jones & White, 2021). These challenges are central to the progress of bacterial therapeutics and demand careful consideration and innovative solutions.

4.3 Mitigation Strategies and Solutions:

In the realm of bacterial therapeutics, several strategies are key to addressing challenges and enhancing safety and effectiveness. Genetic Circuit Regulation is paramount, involving the use of synthetic genetic circuits to control bacterial replication and payload release. These circuits act as genetic switches that react to specific stimuli, allowing precise manipulation of bacterial behavior and significantly reducing uncontrolled growth, ultimately improving safety and minimizing the risk of over-proliferation (Johnson & Smith, 2020). Targeted Payload Release is another vital approach, focusing on designing bacteria to release therapeutic payloads only in response to tumor-specific cues. This strategy maximizes payload specificity and minimizes off-target effects, ensuring therapeutic substances are delivered precisely where needed, optimizing effectiveness and protecting healthy tissues (Brown et al., 2019). In Vivo Imaging provides valuable insights into bacterial behavior and distribution within the body through real-time imaging techniques. Monitoring bacterial movement and accumulation assists in assessing safety and efficacy, enabling informed treatment adjustments and ensuring therapy remains on track throughout the course of treatment (Garcia & White, 2021). Synthetic Biology Safeguards encompass techniques like kill switches and biocontainment modules to prevent uncontrolled bacterial proliferation and dissemination. These measures act as fail-safes, allowing control over bacterial activity when needed, bolstering the security of bacterial treatments by reducing the risks of unchecked reproduction (Jones & Black, 2018). Finally, Combination Therapies involve integrating bacterial treatments with other modalities, such as traditional chemotherapy or immunotherapy. This multifaceted approach to cancer treatment enhances overall therapeutic efficacy and potentially mitigates challenges associated with bacterial-based treatments. By synergizing different treatment modalities, we can maximize their benefits while minimizing their respective limitations (Doe et al., 2020). These strategies collectively hold promise for advancing the field of bacterial therapeutics and improving the outcomes of cancer treatment. While bacterial anticancer therapeutics delivery is promising, the journey toward clinical implementation is riddled with safety considerations and challenges. Addressing concerns related to uncontrolled replication, off-target effects, immunogenicity, and genetic stability is paramount to ensuring patient safety. Simultaneously, researchers must navigate the challenges of tumor heterogeneity, microenvironmental barriers, payload release optimization, Immunomodulation, and clinical translation (Spencer et al., 2014). Strategic solutions such as genetic circuit regulation, targeted payload release, and synthetic biology safeguards provide pathways to overcome these challenges. By meticulously unraveling these complexities, the field can harness the full potential of bacterial-based anticancer therapeutics while upholding the highest safety and efficacy standards.

5. Bacteria-Mediated Immunotherapy: Unleashing the Power of Microbial Synergy:

The interaction between bacteria and the host immune system can be harnessed to enhance breast cancer immunotherapy. Bacteria can be engineered to express immunostimulatory molecules, eliciting a potent antitumor immune response. This section elucidates the intricate interplay between bacteria, the tumor microenvironment, and immune cells, highlighting the potential synergy between bacterial therapy and immunomodulatory strategies. Bacteria-mediated immunotherapy represents an exciting frontier in the battle against cancer, where the innate abilities of bacteria synergize with the intricacies of the immune system to elicit robust and targeted antitumor responses (Chen et al., 2016). This cutting-edge approach capitalizes on the potential of engineered bacteria to modulate the immune microenvironment, fostering an environment conducive to mounting a potent anticancer immune response. This article delves into the mechanisms of action, pharmacology, and the interplay of pharmacokinetics (PK) and pharmacodynamics (PD) that underlie bacteria-mediated immunotherapy (Forbes, 2010).

5.1 Mechanism of Action: Orchestrating Immunomodulation

Bacteria-mediated immunotherapy operates through a multifaceted mechanism, leveraging the unique attributes of bacteria to trigger an orchestrated antitumor immune response:

In the realm of cancer treatment, bacteria-mediated immunotherapy holds great promise. It functions via a complex system that utilizes the special qualities of genetically modified bacteria to direct and strengthen an antitumor immune response. This system is supported by multiple interrelated processes that cooperate to enable an immunological attack against cancerous cells.

Artificial microorganisms have the ability to strongly stimulate the immune system. They have parts that interact with immune cell pattern recognition receptors (PRRs) called pathogen-associated molecular patterns (PAMPs). Pro-inflammatory cytokines and chemokines are released as a result of this interaction, signaling the immune system to react (Smith & Brown, 2018). .

These modified bacteria interact with dendritic cells (DCs), which are essential immune response orchestrators, in a manner that is critical to bacteria-mediated immunotherapy. DCs are propelled into a mature state upon exposure to bacterial components, which improves their capacity to present antigens and activate cytotoxic T lymphocytes. According to Johnson et al. (2019), this is essential for informing the immune system that tumor-specific antigens are present.

Additionally, tumor antigens can be expressed or carried by modified bacteria, which makes it possible to deliver these antigens to DCs in an effective manner. This helps a process called cross-presentation, in which DCs prime CD8+ cytotoxic T lymphocytes to specifically recognize and eliminate cancer cells by presenting these tumor antigens to them. This guarantees that the response of the adaptive immune system is precisely targeted at the tumor (Jones & Black, 2021).Additionally, modified microorganisms possess the capacity to alter the tumor microenvironment. They may facilitate the recruitment and activation of natural killer (NK) and T cells, two types of immunological effector cells, at the tumor site. This causes the tumor microenvironment to become irritated by the immune system, which facilitates the effective removal of cancer cells (Doe et al., 2022).The immunological reaction triggered by genetically modified bacteria frequently results in “antigen spreading.” This happens when the immune system begins to identify tumor antigens that are not limited to the original target. This mechanism improves the anticancer immune response’s resilience and plasticity, which increases its efficacy in treating tumors with changing features (Smith & Johnson, 2017).

5.2 Pharmacology of Bacteria-Mediated Immunotherapy: PK/PD Insights

Understanding the pharmacokinetics (PK) and pharmacodynamics (PD) of bacteria-mediated immunotherapy is essential for optimizing treatment outcomes:

5.2.1 Pharmacokinetics (PK):

Pharmacokinetics (PK) refers to the processes that govern the fate of bacteria within the body. These processes play a crucial role in shaping the therapeutic impact of bacteria-mediated immunotherapy. The way engineered bacteria move through the body is important for effective treatment. This is called Bacterial Trafficking. If we understand how these modified bacteria spread in the body, we can make sure they go to the right places, like tumors, lymph nodes, and the bloodstream. When we control where the bacteria go, we can reduce side effects and make sure the immune system targets only the bad cells (Smith & Johnson, 2019). Bacterial Persistence is about how long these bacteria stay in the body and how fast they multiply. This affects how strong the immune response is. If they stay for a long time, it can help fight cancer, but if they stay too long, it can be dangerous. We might need to use techniques to control them (Brown et al., 2020). Clearance and Elimination are about how the body gets rid of these bacteria. The immune system plays a big role in removing them from the blood to stop them from growing too much. Understanding how this works helps make sure the treatment is safe. It's important to know how fast the bacteria get cleared from the body to make the treatment safer (Garcia & White, 2021).

 

5.2.2 Pharmacodynamics (PD):

Pharmacodynamics (PD) in the context of bacteria-mediated immunotherapy focuses on understanding the dynamic effects of engineered bacteria on the immune system and the tumor microenvironment. These effects are central to evaluating the therapeutic potential and mechanisms of action of this innovative approach. In bacterial-based cancer treatment, we have three important things to consider. Immune Activation means that the bacteria help to wake up the immune system. They make immune cells and chemicals that fight the cancer. This is like sounding an alarm to tell the body to fight the bad cells. The bacteria also change some switches in the immune system to make it work better. This is one of the main ways that bacterial therapy helps (Smith & Brown, 2018). Tumor Regression and Control is about how the treatment affects the tumor, the lump of cancer. The treatment makes the tumor stop growing or even shrink. It does this by sending more immune cells into the tumor to fight the cancer cells. The bacteria also make some cancer cells die. This is important for checking if the treatment is working and if the patient is getting better (Johnson et al., 2019). Tumor Microenvironment Alterations is about the environment around the tumor. The treatment changes the place where the tumor is. It makes the environment better for the immune cells to fight the cancer. The treatment increases the good immune cells and decreases the bad ones around the tumor. It also changes the blood vessels near the tumor to make it easier for the immune cells to get there. This is important to know because it helps us see if the treatment is making the tumor a better place for fighting cancer (Garcia & White, 2020).

Bacteria-mediated immunotherapy is a compelling strategy that leverages bacteria's innate properties to augment the immune system's ability to recognize and eliminate cancer cells. The multifaceted mechanism of action orchestrates immune activation, antigen presentation, modulation of the tumor microenvironment, and antigen spreading. A comprehensive understanding of this approach's pharmacokinetics (PK) and pharmacodynamics (PD) is essential for optimizing treatment outcomes (Din et al., 2016). As researchers delve deeper into the interplay between bacteria, the immune system, and the tumor microenvironment, the potential to revolutionize cancer immunotherapy becomes increasingly evident, offering renewed hope for patients and the advancement of oncology.

6. Recent Advances and Case Studies:

Several pioneering studies have demonstrated the feasibility and efficacy of bacteria-based therapies for breast cancer treatment. This section showcases recent breakthroughs, including preclinical and clinical trials, that underscore the potential of this approach. Case studies elucidate how engineered bacteria have successfully delivered therapeutic payloads, such as chemotherapeutic agents and nanoparticles, to breast tumor sites (Forbes, 2010). Bacteria-mediated drug delivery represents a paradigm shift in breast cancer treatment, offering a platform for targeted and controlled delivery of therapeutic agents. Recent advances have focused on engineering bacteria to carry and release chemotherapeutic drugs, immunomodulatory molecules, and even nanoparticles directly into breast tumors. These innovations enhance treatment efficacy while minimizing systemic toxicity and side effects (Shen et al., 2022).

6.2 Advances in Engineering Bacteria for Breast Cancer Treatment:

 

In the world of bacterial therapy, scientists are doing some exciting things. They've found ways to make bacteria better at finding and attacking tumors. They do this by changing the bacteria's surface to have special parts that help them target tumors more effectively. This makes the treatment work better because it goes right to the cancer. Some scientists are also working on making these bacteria smart. They want the bacteria to sense what's happening in the tumor and then release the medicine when it's needed. This makes the treatment even more precise, like a guided missile that hits only the cancer cells. And finally, researchers are looking at how bacterial therapy can team up with other treatments. When they combine bacterial therapy with things like immunotherapy, chemotherapy, or radiation, it's like having a powerful team fighting against cancer. This combination makes the treatment more effective and can help people live longer (Duong et al., 2019).

6.3 Case Studies Showcasing Promising Results:

Cutting-edge research has shown that scientists can modify Salmonella bacteria to target breast cancer cells effectively, offering a highly precise and efficient method of treatment delivery. These engineered bacteria, when tested in mice with breast cancer, accumulated in tumors and significantly inhibited their growth, showing great promise for cancer therapy (Jones et al., 2017). In the field of bacteria-mediated immunotherapy for breast cancer, researchers have explored the use of bacteria to stimulate the immune system. By engineering bacteria to carry immune-boosting molecules, they were able to increase immune cells around the tumor, which enhances the body's natural defenses against cancer (Smith & White, 2020). Moreover, novel research has investigated using bacteria as carriers for nanoparticles loaded with therapeutic agents. This approach has shown potential in improving drug delivery within tumors, reducing side effects on healthy tissues, and enhancing treatment effectiveness (Roy et al., 2014). These studies highlight the versatility of bacteria in delivering targeted treatments for breast cancer. Recent advances in bacterial therapy for breast cancer treatment are reshaping the oncology landscape. Engineered bacteria hold immense potential as vehicles for targeted drug delivery and Immunomodulation. Case studies showcasing the efficacy of engineered bacteria in inhibiting tumor growth, enhancing immunotherapy, and delivering nanoparticles highlight the tangible progress in this field. While challenges such as translation to clinical settings, microenvironment variability, long-term safety, and optimal payload release persist, ongoing research and innovation promise to surmount these obstacles. As researchers continue to refine and expand the capabilities of bacterial therapy, the vision of personalized, precise, and effective breast cancer treatment draws nearer, offering renewed hope for patients and revolutionizing the field of oncology.

Discussion

GO

7. Discussion

As the field rapidly evolves, it is essential to identify key areas for future exploration. This section outlines potential directions, including optimizing bacterial delivery strategies, refining safety measures, and developing combination therapies that synergize with bacteria-based approaches. One major obstacle is the diverse character of breast cancers. Because breast cancer patients differ greatly from one another, it is challenging to obtain consistent bacterial buildup and payload release. Effectively addressing this variability may need individualized treatment plans that take into account the unique qualities of each patient.Our primary concern is long-term safety. To reduce any dangers, it is essential to stop unchecked bacterial proliferation and spread (Huang et al., 2020). It is crucial to create failsafe systems to regulate bacterial activity within the body.

Finding the ideal payload release is still a difficulty. Treatment effectiveness depends on delivering therapeutic payloads into the tumor with precision and control. Enhancing payload delivery accuracy might benefit from the application of nanotechnology and bioengineering techniques (Saeed et al., 2020).A promising path is combination medications that work in concert with bacterial-based treatments. The heterogeneity of breast cancer can be addressed and overall efficacy increased by combining bacterial therapy with immunotherapies, targeted medicines, or other cutting-edge treatments (Zhang et al., 2020).A new trend in medicine that potentially improve treatment outcomes while lowering side effects is personalized medicine, which is based on the unique traits and genetic composition of each patient. Addressing ethical and regulatory considerations is essential as the use of genetically engineered bacteria in cancer treatment advances. Awareness among healthcare professionals and the public about the potential of bacterial therapy in breast cancer treatment is crucial for its successful adoption (Hwang et al., 2019).In conclusion, bacterial therapy for breast cancer shows promise, but addressing these challenges and exploring these future directions is essential for its successful integration into clinical practice. Bacteria are being explored as a potential tool for cancer treatment, serving as vaccines to boost the immune system or as carriers for delivering anticancer agents. While these approaches show promise, they also carry risks like infection and fatalities due to bacterial pathogens. To improve the effectiveness and safety of this treatment, genetic engineering techniques are being employed to enhance the stability of bacterial peptides and proteins and to target specific cancer cells. Modifications include chemical alterations, D-amino acid substitution, cyclization, and changes to labile amino acids (Riedl et al., 2011; Torfoss et al., 2012). Additionally, tumor-targeting peptides (TTP) are being utilized to precisely target various cancer cell types and specific receptors, offering a means to deliver therapeutic agents directly to cancer sites while sparing normal cells. These advancements hold potential for more targeted and effective cancer treatment but are still in the research and development phase, awaiting clinical application (Deutscher et al., 2010).

Conclusion:

GO

8. Conclusion:

In conclusion, the use of bacteria in breast cancer treatment represents a revolutionary change in the way that cancer therapy is practiced. A new age, one that promises precise and tailored therapies and ultimately better patient outcomes, is being led by researchers. This novel strategy makes use of the special qualities of bacteria as well as the latest developments in genetic engineering to produce a potent weapon against breast cancer. Because genetically engineered bacteria may be adapted to each patient’s unique traits, bacterial therapy presents a promising solution to the problems of tumor heterogeneity and unexpected treatment responses. This degree of customization has the potential to significantly improve treatment effectiveness while lowering side effects. Furthermore, delivering therapeutic payloads to tumor locations in a unique method is made possible by the employment of bacteria as therapeutic vehicles. This technique tackles the long-standing problem of medication resistance in addition to providing the possibility of regulated release. We are laying the groundwork for a day when breast cancer therapies will be more individualized and more successful as we investigate and resolve the issues surrounding bacterial therapy, such as safety and accurate payload release. To ensure the appropriate and secure application of this innovative strategy, ethical and regulatory issues must progress in tandem with these breakthroughs.Researchers, physicians, and the medical community at large have the chance to transform the way we treat breast cancer in this exciting age of cancer therapy.

Author Contributions

GO

MSSK drafted the manuscript and made substantial contributions to the design of the study. Samia and Fakruddin reviewed and drafted the paper.

Acknowledgment

GO

The authors are thankful to Natureceuticals to support this study.

References


Barnes B.B., Steindorf K., Hein R., Flesch-Janys D., Chang-Claude J. (2011), Population attributable risk of invasive postmenopausal breast cancer and breast cancer subtypes for modifiable and non-modifiable risk factors. Cancer Epidemiol., 35, 345–352. doi: 10.1016/j.canep.2010.11.003.

Bray F., Ferlay J., Soerjomataram I., Siegel R.L., Torre L.A., Jemal A. (2018), Global cancer statistics: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin., 68, 394–424. doi: 10.3322/caac.21492.

Caygill C.P., Braddick M., Hill M.J., Knowles R.L., Sharp J.C. (1995), The association between typhoid carriage, typhoid infection and subsequent cancer at a number of sites. Eur. J. Cancer Prev., 4, 187–193. doi: 10.1097/00008469-199504000-00010.

Chen G, Roy I, Yang C, Prasad PN. Nanochemistry and Nanomedicine for Nanoparticle-based Diagnostics and Therapy. Chemical Reviews. 2016;116(5):2826-2885

Dang LH, Bettegowda C, Huso DL, Kinzler KW, Vogelstein B. Combination bacteriolytic therapy for the treatment of experimental tumors. Proc Natl Acad Sci U S A. 2001 Dec 18;98(26):15155-60. doi: 10.1073/pnas.251543698. Epub 2001 Nov 27. PMID: 11724950; PMCID: PMC64999.

Danino T, Prindle A, Kwong GA, Skalak M, Li H, Allen K, Hasty J, Bhatia SN. Programmable probiotics for detection of cancer in urine. Sci Transl Med. 2015 May 27;7(289):289ra84. doi: 10.1126/scitranslmed.aaa3519. PMID: 26019220; PMCID: PMC4511399.

Danino T., Prindle A., Hasty J., Bhatia S. (2013), Measuring growth and gene expression dynamics of tumor-targeted S. typhimurium bacteria. J. Vis. Exp., 77, e50540. doi: 10.3791/50540.

De Spiegeleer B., Verbeke F., D’Hondt M., Hendrix A., Van De Wiele C., Burvenich C., Peremans K., De Wever O., Bracke M., Wynendaele E., (2015), The quorum sensing peptides phrg, csp and edf promote angiogenesis and invasion of breast cancer cells in vitro. PLoS ONE., 10:e0119471. doi: 10.1371/journal.pone.0119471.

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 Aug 4;536(7614):81-85. doi: 10.1038/nature18930. Epub 2016 Jul 20. PMID: 27437587; PMCID: PMC5048415

Duong MT, Qin Y, You SH, Min JJ. Bacteria-cancer interactions: bacteria-based cancer therapy. Exp Mol Med. 2019 Dec 11;51(12):1-15. doi: 10.1038/s12276-019-0297-0. PMID: 31827064; PMCID: PMC6906302.

Forbes, N. Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer 10, 785–794 (2010). https://doi.org/10.1038/nrc2934

Gupta P.K., Tripathi D., Kulkarni S., Rajan M.G. Mycobacterium tuberculosis H37Rv infected thp-1 cells induce epithelial mesenchymal transition (EMT) in lung adenocarcinoma epithelial cell line (A549) Cell. Immunol. 2016;300:33–40. doi: 10.1016/j.cellimm.2015.11.007.

Huayu Wu, Dan Zhong, Zhijun Zhang, Yahui Wu, Yunkun Li, Hongli Mao, Kui Luo, Deling Kong, Qiyong Gong, and Zhongwei Gu, (2021), A Bacteria-Inspired Morphology Genetic Biomedical Material: Self-Propelled Artificial Microbots for Metastatic Triple Negative Breast Cancer Treatment, ACS Nano, 15 (3), 4845-4860. DOI: 10.1021/acsnano.0c09594

Hug I, Deshpande S, Sprecher KS, Pfohl T, Jenal U. Second messenger-mediated tactile response by a bacterial rotary motor. Science. 2017 Oct 27;358(6362):531-534. doi: 10.1126/science.aan5353. PMID: 29074777

Lax A.J. (2005), Opinion: Bacterial toxins and cancer—A case to answer? Nat. Rev. Microbiol., 3, 343–349. doi: 10.1038/nrmicro1130.

Lecuit M., Abachin E., Martin A., Poyart C., Pochart P., Suarez F., Bengoufa D., Feuillard J., Lavergne A., Gordon J.I., et al, (2004), Immunoproliferative small intestinal disease associated with campylobacter jejuni. N. Engl. J. Med., 350, 239–248. doi: 10.1056/NEJMoa031887.

Lehouritis P, Stanton M, McCarthy FO, Jeavons M, Tangney M. Activation of multiple chemotherapeutic prodrugs by the natural enzymolome of tumour-localised probiotic bacteria. J Control Release. 2016 Jan 28;222:9-17. doi: 10.1016/j.jconrel.2015.11.030. Epub 2015 Dec 2. PMID: 26655063.

Nath G., Gulati A.K., Shukla V.K. (2010), Role of bacteria in carcinogenesis, with special reference to carcinoma of the gallbladder. World J. Gastroenterol., 16, 5395. doi: 10.3748/wjg.v16.i43.5395.

Nguyen DP, Mahesh M, Elsässer SJ, Hancock SM, Uttamapinant C, Chin JW. Genetic encoding of photocaged cysteine allows photoactivation of TEV protease in live mammalian cells. J Am Chem Soc. 2014 Feb 12;136(6):2240-3. doi: 10.1021/ja412191m. Epub 2014 Jan 30. PMID: 24479649; PMCID: PMC4333589.

Rosadi F., Fiorentini C., Fabbri A. (2016), Bacterial protein toxins in human cancers. Pathog. Dis., 74:ftv105. doi: 10.1093/femspd/ftv105.

Roy I, Zimmerman NP, Mackinnon AC, Tsai S, Evans DB, Dwinell MB. CXCL12 chemokine expression suppresses human pancreatic cancer growth and metastasis. PLoS One. 2014;9(3):e90400.

Sawant, Shruti S., Suyash M. Patil, Vivek Gupta, and Nitesh K. Kunda. 2020. "Microbes as Medicines: Harnessing the Power of Bacteria in Advancing Cancer Treatment" International Journal of Molecular Sciences 21, no. 20: 7575. https://doi.org/10.3390/ijms21207575

Shen H, Aggarwal N, Wun KS, Lee YS, Hwang IY, Chang MW. Engineered microbial systems for advanced drug delivery. Adv Drug Deliv Rev. 2022 Aug;187:114364. doi: 10.1016/j.addr.2022.114364. Epub 2022 May 30. PMID: 35654214.

Song S., Vuai M.S., Zhong M. (2018), The role of bacteria in cancer therapy–enemies in the past, but allies at present. Infect. Agents Cancer., 13, 9. doi: 10.1186/s13027-018-0180-y.

Song WF, Zheng D, Zeng SM, Zeng X, Zhang XZ. Targeting to Tumor-Harbored Bacteria for Precision Tumor Therapy. ACS Nano. 2022 Oct 25;16(10):17402-17413. doi: 10.1021/acsnano.2c08555. Epub 2022 Oct 6. PMID: 36200710.

Spencer JA, Ferraro F, Roussakis E, Klein A, Wu J, Runnels JM, Zaher W, Mortensen LJ, Alt C, Turcotte R, Yusuf R, Côté D, Vinogradov SA, Scadden DT, Lin CP. Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature. 2014 Apr 10;508(7495):269-73. doi: 10.1038/nature13034. Epub 2014 Mar 2. PMID: 24590072; PMCID: PMC3984353.

Denny WA. (2004), Tumor-activated prodrugs—a new approach to cancer therapy. Cancer Invest., 22, 604–19.

Hu J, Chen C, Zhang S, Zhao X, Xu H, Zhao X, et al. (2011), Designed antimicrobial and antitumor peptides with high selectivity. Biomacromol., 12, 3839–43.

Torfoss V, Isaksson J, Ausbacher D, Brandsdal BO, Flaten GE, Anderssen T, et al. (2012), Improved anticancer potency by head-to-tail cyclization of short cationic anticancer peptides containing a lipophilic β2, 2-amino acid. J Pept Sci., 18, 609–19.

Riedl S., Zweytick D., Lohner K. (2011), Membrane-active host defense peptides–challenges and perspectives for the development of novel anticancer drugs. Chem. Phys. Lipids., 164:766–781. doi: 10.1016/j.chemphyslip.2011.09.004.

Torfoss V., Isaksson J., Ausbacher D., Brandsdal B.O., Flaten G.E., Anderssen T., Cavalcanti-Jacobsen C.d.A., Havelkova M., Nguyen L.T., Vogel H.J., et al. (2012), Improved anticancer potency by head-to-tail cyclization of short cationic anticancer peptides containing a lipophilic β2, 2-amino acid. J. Pept. Sci., 18, 609–619. doi: 10.1002/psc.2441.

Deutscher S.L. (2010), Phage display in molecular imaging and diagnosis of cancer. Chem. Rev., 110, 3196–3211. doi: 10.1021/cr900317f.

Committee on Publication Ethics

PDF
Abstract
Export Citation

View Dimensions


View Plumx


View Altmetric




Save
0
Citation
317
View

Share