1 s2.0 S0304383524002106 Main

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Cancer Letters 589 (2024) 216817

Contents lists available at ScienceDirect

Cancer Letters
journal homepage: www.elsevier.com/locate/canlet

Engineered bacteria in tumor immunotherapy


Hua Chen 1, Yinrui Zhu 1, Chonghai Zhang , Lin Hu **, Kai Yang *
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of
Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, 199 Renai Road, Suzhou, 215123, China

A R T I C L E I N F O A B S T R A C T

Keywords: As the limitations of cancer immunotherapy become increasingly apparent, there is considerable anticipation
Engineered bacteria regarding the utilization of biological tools to enhance treatment efficacy, particularly bacteria and their de­
Immunotherapy rivatives. Leveraging advances in genetic and synthetic biology technologies, engineered bacteria now play
Combined therapy
important roles far beyond those of conventional immunoregulatory agents, and they could function as tumor-
Gene editing
Synthetic biology
targeting vehicles and in situ pharmaceutical factories. In recent years, these engineered bacteria play a role in
Controlled release almost every aspect of immunotherapy. It is nothing short of impressive to keep seeing different strain of bacteria
modified in diverse ways for unique immunological enhancement. In this review, we have scrutinized the
intricate interplay between the immune system and these engineered bacteria. These interactions generate
strategies that can directly or indirectly optimize immunotherapy and even modulate the effects of combination
therapies. Collectively, these engineered bacteria present a promising novel therapeutic strategy that promises to
change the current landscape of immunotherapy.

1. Introduction attempts by bacteria to fight cancer [8]. In 1893, Dr. Coley treated
cancer patients with "Coley’s toxins” made from inactivated Strepto­
As one crucial approaches in clinical cancer treatment, immuno­ coccus pyogenes and Serratia marcescens. People then could hardly un­
therapy activates patients’ inherent immune system, eradicating tumors derstand how these dead bacteria benefited the medical treatment until
while reducing damage to normal tissues [1]. When combined with 1970s, molecular cloning techniques revealed the interaction between
chemotherapy or radiotherapy, immunotherapy even significantly ex­ immunity and bacteria [9–11]. In the 21st century, Beutler [12]
tends an individual’s median survival time [1,2]. However, this kind of explained the activation of the Toll-like receptor family. Soon after,
therapy shows poor performance in general population, wherein only researchers drew inspiration from new anti-tumor strategies that acti­
20–40% of patients respond to immunotherapy in clinical samples [3]. vate or enhance the immune system, marking the beginning of tumor
The majority of deaths reported to occur during treatment are associated immunotherapy. This article has arranged the key advances during the
with serious immune-related adverse events (irAEs), which remain past hundred years and concluded them into three parts (Fig. 1). Prior to
difficult to avoid [4]. Resistance to related drugs also complicates the 1970s, attempts were made to treat tumors without exploring the
treatments [5]. Furthermore, the high cost of medication deters many mechanisms involved. It was nearly 30 years later that we discovered
patients from receiving treatment [6,7]. Given the heterogeneity, the relationship between bacteria and immunity, and bacteria were
complexity, and diversity of tumors, traditional immunotherapy stra­ shown to be engineered and applied to tumor therapy. Since the 21st
tegies no longer meet clinical needs. Therefore, it is highly significant to century, advanced strategies for engineered bacteria to be used for
identify a new type of efficient, convenient, and safe immunomodula­ tumor immunity have emerged, ushering in a new era of
tory agent to better assist tumor immunotherapy. immunotherapy.
Bacteria have an innate ability to stimulate the host immune system With the widespread use of genetic engineering and synthetic
and may be suitable agents for novel immunotherapy. A funny fact is biology, living bacteria have for the first time broken the natural limi­
that the acknowledged modern cancer immunotherapy began with tations of pathogen and developed some new properties [13]. Some

* Corresponding author.
** Corresponding author.
E-mail addresses: hulin@suda.edu.cn (L. Hu), kyang@suda.edu.cn (K. Yang).
1
Contributed equally to this work.

https://doi.org/10.1016/j.canlet.2024.216817
Received 14 December 2023; Received in revised form 12 March 2024; Accepted 12 March 2024
Available online 15 March 2024
0304-3835/© 2024 Elsevier B.V. All rights reserved.
H. Chen et al. Cancer Letters 589 (2024) 216817

types of bacteria, such as Gram-negative Salmonella (e.g., S. typhimurium (Fig. 2), to explore the role of engineering bacteria in immunotherapy.
YS8211 and YS1629 [14]), non-pathogenic bacterial strain E. coli Nissle
1917 (EcN) [15], and Gram-positive Clostridium (e.g., Clostridium 2. Engineered bacteria generate immune killing effect
butyricum M − 55 [16]), Lactococcus lactis [17], Bifidobacterium [18]
have a tendency to accumulate at tumor sites after intravenous admin­ 2.1. Engineered bacteria release tumor necrosis factor α (TNFα)
istration. Thus, they are also called ‘Oncolytic Bacteria’ [19]. Among
them, an attenuated S. typhimurium strain VNP20009 had completed Cytokines are among the most direct immune effector molecules in
Phase I clinical trials as an anti-tumor agent [20]. Their potential in cancer immunotherapy. As the first cytokine used in tumor immuno­
tumor therapy has been recognized, and advances in synthetic biology therapy, tumor necrosis factor α (TNFα) has also been found to be
have further enhanced their medical applications [21]. With the help of associated with bacteria [37]. After binding to the TNFR-1 receptor,
advanced biochemical tools, scientists have attempted to artificially TNFα sends a cell apoptosis signal, thereby initiating a proteinase
modify these bacteria with extra biological behavior, which are also cascade reaction with Fas-related death domain protein recruitment
known as engineered bacteria [22,23]. Through genetic modification, (FADD) that eventually triggers complete tumor cell apoptosis [38].
biological modules could be “inserted” into these living bacteria on the However, direct systemic injection of TNFα will induce severe toxic side
basis of prokaryotic expression systems. As a result, a number of new effects and has the potential to promote tumor cell metastasis [39].
biological functions (e.g., biosensing, control, and synthesis) could be Therefore, targeting bacteria to secrete TNFα at the tumor site is an ideal
added to the host bacteria [24–28]. Some oncolytic bacteria could alternative.
multiply in vivo after intravenous administration, which means that a S. Nuyts et al. were the first to successfully modify Bacillus subtilis
single injection may cause large and lasting bacterial effect at a low [40], whereas S.L. Young et al. employed non-pathogenic Bacteroides
dose. As for engineered oncolytic bacteria, their proliferation ensures fragilis for recombinant secretion modification of TNFα in the treatment
continuous production or secretion, and their tumor-targeting property of primary and metastatic tumors in mice [41]. The secreted TNFα
allow them to produce a superior target/non-target ratio, thereby activated macrophages and dendritic cells, while Bacteroides functioned
making engineered bacteria excellent drug delivery vehicles for as a rich source of immunogens and antigenic substances that acted as
anti-tumor agent. Some types of engineered bacteria successfully survive effective adjuvants and together produce efficient antigen presentation.
inside tumor cells and even immune cells [29,30], thus facilitating Their study found that the antitumor effect of modified bacteria was
intracellular drug-delivery [31–33]. To realize more precise drug mainly related to natural killer (NK) cells, further emphasizing the po­
release, researchers can introduce specific stimulus-response element tential efficacy of NK cells in immunotherapy. W.S. Yoon et al. also
upstream of the concerned gene. Certain bacteria strains also acquire emphasized the importance of NK cells in their work [42]. They inge­
additional biological behaviors through the integration of artificial niously fused recombinant TNFα downstream to SipB, an outer mem­
materials (which are usually nano-scaled), without extensive genetic brane protein sequence that was actively expressed by Salmonella when
modification of the bacteria themselves [34,35]. In this way live bacteria it invaded tumor cells, so that TNFα only appeared inside tumor cells
could as well perform new biological behavior due to the artificially rather than normal cells or interstitial tissue (Fig. 3a). The results ulti­
additional materials (usually are nano-scaled), so we classify them as mately verified that the antitumor effect produced by such engineered
part of engineered bacteria as well in this article. To sum up, genetic bacteria mainly was mainly from NK cells and bacterial toxicity itself,
engineering and synthetic biology have enabled live bacteria-based with almost no immune memory. These studies suggested that the use of
immunotherapy with the advantages of high efficacy, low cost and engineered bacterial in conjunction with TNFα therapy might tempo­
few side effects [36] compared with other intravenous immune agents. rarily cause tumor regression, but lack significance in inhibiting tumor
While many engineered bacteria have shown their immunothera­ recurrence. Therefore, further combinations with immunomodulators
peutic potential, there is no comprehensive classification or summary of targeting T cells or alternative products with more widely acting cyto­
how they participate in immunotherapy so far. In this review, we will kines are necessary. Nonetheless, these studies confirmed the conve­
focus on the interactions between engineered bacteria, responsive nience, low cost, and safety of engineered bacterial preparations,
external stimuli, engineer contents, and the host immune system highlighting the important role of synthetic biology in modifying

Fig. 1. Timeline of major advances in the application of engineered bacteria in immunotherapy. The use of engineered bacteria in immunotherapy has gone through
three phases. In the first phase until the 1970s, researchers were still exploring the possibilities of bacteria as therapeutic agents, in the second phase from 1970 to
2000, interactions between bacteria and immunity were discovered, while engineered bacteria showed great potential in cancer therapy, and in the third phase in the
21st century, engineered bacteria began to treat tumors from an immunological perspective. Produced with permission from Biorender.

2
H. Chen et al. Cancer Letters 589 (2024) 216817

Fig. 2. Bacteria are engineered using gene editing technology and synthetic biology. Gene editing reduces the virulence of wild hosts, and synthetic biology installs
non-intrinsic functions such as sensing, control, tumor targeting, and biosynthesis. These engineered bacteria participate in the immunotherapy in three main ways:
engineered bacteria assisting immune effect, generating immune killing effect and participating in combination immunotherapy. 1) When assisting immune effect,
the role of engineered bacteria is either to enhance innate immune, regulate intestinal flora or nourish immune cells, 2) while engineered bacteria generate immune
effect through releasing cytokines, blocking immune checkpoint or presenting antigen, 3) they also participate in combination therapy with ICIs, PTT or radio­
therapy. Produced with permission from Biorender.

bacteria as immunomodulators. 2.3. Engineered bacteria release interleukins

2.2. Engineered bacteria release interferon γ In addition to TNFα and IFN-γ, interleukins also play a crucial role in
transmitting information between immune cells and are typically
Interferon γ (IFN-γ) is essential for inducing immunogenic death of participate in anti-tumor immune responses by activating and regulating
tumor cells and participates in both innate and adaptive immunity. specific immune cells. Although bacteria can strongly stimulate the body
However, due to its toxicity, a systemic dosing strategy for IFN-γ is not to produce various interleukins such as IL-6, IL-12 [49], IL-17 [50], and
feasible. Additionally, IFN-γ was thought difficult to be directly secreted IL-18 [51], the effect of modifying bacteria to secrete interleukins is not
by bacterial carriers for its anti-bacterial effects [43]. With the advent of obvious.
synthetic biology technology, the sensing control function of bacteria According to the literatures, IL-2 secreted by S. typhimurium signifi­
can be altered, making it possible for tumor-targeting bacteria to release cantly induced NK cell-mediated innate immunity [52–55], with much
IFN-γ directly, killing tumor cells without damaging themselves until less systemic toxicity than that of IL-2 injection alone [53]. However,
specific conditions are met. W.S. Yoon first fused the SipB sequence with researchers differed on whether this bacterium is effective in activating
IFN-γ and modified it into S. typhimurium BRD509 [44] (Fig. 3b). In this specific immunity. Some studies have provided direct evidence that
study, it was found that the powerful anti-tumor effect came from NK there was no significant activation or proliferation of T cells at the tumor
cells. However, although no immunological memory was detected, site or in circulation [52,53], while other studies have shown that
macrophages as antigen-presenting cells were significantly activated, engineered SalpIL2 bacteria inhibited tumor metastasis [54] and acti­
suggesting that this modified strain might have some ability to inhibit vated proliferation of CD8+T cells in the spleen. Researchers have also
tumor metastasis. attempted to modify bacteria to secrete other interleukins. Y. Wu inte­
In a study by Y. Chen et al. [45], an ultrasonic-responsive VNP20009 grated the function of secreting IL-lβ to S. typhimurium substrain
strain was developed, which secreted IFN-γ induced by ultrasound to ΔpGlux/pT-ClyA [56], which resulted in an upregulation of expression
promote cancer cell apoptosis and induce macrophage polarization from levels of NLRP3, Caspase 1, MyD88, NF-κB, and IL-lβ-related signaling
M0 towards M1 phenotype (Fig. 3b). CD4+ T cells and CD8+ T cells were pathway molecules in tumors, and ultimately enhanced the inhibitory
also found to be significantly activated. Interestingly, the IFN-γ secretion effect of bacteria on colon cancer without observing tumor recurrence.
by bacteria into cytoplasm polarized splenic macrophages and enhanced L. Wang et al. used Bifidobacterium as a carrier to deliver the IL-24 gene
their antigen presentation function, resulting in increased activation of into tumor cells [57], inducing the apoptosis of tumor cells. IL-18 could
CD4+ and CD8+ T-cells, and ultimately inhibiting distant tumor stimulate T cell proliferation and enhance NK cells activity, thus some
metastasis and growth. In C. Wu’s study [46], the recombinant studies conferred on S. typhimurium the ability to express IL-18 in mice
VNP20009 strain secreted IFN-γ under the control of light. Combining directly [58]. Intratumorally injection of these bacteria could increase T
this with photodynamic therapy (PDT) treatment could be more effec­ cells and NK cells at the tumor site, along with significant infiltration by
tive in suppressing tumor at distant sites. IFN-γ has also been reported to neutrophils, thereby inhibiting tumor growth and metastasis.
upregulate PD-L1 expression in tumors [47]. When combined with
PD-L1 antibodies, IFN-γ secreted by non-pathogenic extracellular bac­
teria could significantly inhibited tumor growth [48]. 2.4. Controlled release of immune checkpoint inhibitors by engineered
bacteria

The most classic immune checkpoint inhibitors, αPD-L1 and αCTLA-

3
H. Chen et al. Cancer Letters 589 (2024) 216817

Fig. 3. Engineered bacteria generate immune killing effect, with or without specific control. a. Bacteria acquire the ability to release TNFα after engineering. TNFα
can be secreted directly or during bacterial invasion by fusing with invasion protein SipB. b. Similarly, bacteria can be engineered to release IFN-γ, both directly or
with the help of SipB. The release of IFN-γ can be responsive to external stimuli like ultrasound, etc. c. Optimized antibodies (scFv and nanobodies) can also be
therapeutic payloads. These products could be regulated by light irradiation (photoswitch) or bacterial density (synchronized lysis circuit, SLC). d. Engineered
bacteria can suppress immune checkpoint expression genetically. This strategy mainly relies on outer membrane vesicles (OMV), inclusion bodies, plasmid or other
nucleic acid that released once bacteria are lysed.

4, have been desired to be loaded on special carriers to reduce their Interestingly, studies have reported that chronic infections with some
extra-tumoral concentrations and thus reduce irAEs events. To reduce bacteria could upregulate PD-L1 expression in cancer cells [63–65]. In
the cost of drug, researchers have developed single-chain antibodies addition, even dead bacteria could also achieve this effect, further
(ScFv, ≈30 kDa) and smaller nanobodies (≈15 kDa) using recombinant reducing T cell activation [65]. Therefore, whether or not these bacteria
DNA technology [59,60] (Fig. 3c). Although these molecules retain are used as immune checkpoint inhibitor vectors, they promote the ef­
certain antigen affinity [61,62], their small molecular weight may cause ficacy of αPD-L1.
rapid clearance and poor distribution within tumor tissues. The engi­ Although the target/non-target ratio of tumor-targeted bacteria is
neered tumor-targeting bacteria may hold promise for solving these high, it is important to note that bacteria may still temporarily accu­
challenges. These bacteria have an unparalleled target/non-target ratio mulate in other organs such as the liver and spleen. Antibodies released
and unique synthetic capabilities, which means that they can continu­ directly from bacteria may temporarily increase in these tissues, causing
ously release antibodies into tumor sites, maintain longer intratumoral local side effects. Fortunately, researchers were able to control the
drug concentrations and reduce instances of tumor resistance events. timing and location of antibodies release through synthetic biology

4
H. Chen et al. Cancer Letters 589 (2024) 216817

programs. Additionally, by controlling the rate of release, they were able directly attack tumor cells and promote apoptosis. Conversely, the
to breach the maximum threshold of antibody concentration within the tumor inflammatory microenvironment promotes the proliferation and
tumor, resulting in stronger immune checkpoint inhibition. Subse­ metastasis of malignant tumor cells [78]. To avoid counterproductive
quently, various external stimulus including focused ultrasound and effect, the innate immune system needs to be appropriately modulated
photothermal effect were applied to transfer antibody for enhancing in immunotherapy to generate a balanced inflammatory cell phenotype
tumor immunotherapy [66,67]. In a special research, C.R. Gurbatri et al. and quantity [79,80]. As a unicellular microorganism, bacterial infec­
engineered the expression of PD-L1/CTLA-4 dual-nanobody by incor­ tion could activate the innate immune system, particularly macrophages
porating SLC in E. coli DE3 [68]. Since none of the nanobodies used in and natural killing (NK) cells [81]. Taking advantage of this properties,
this engineered bacterium exceeded 15 kDa, this bacterium could ex­ some researchers have used engineered bacteria to modulate immune
press two nanobodies at the same time, resulting in stress-free dual-­ cells within tumors and enhance the effectiveness of immunotherapy.
targeted inhibition and higher efficacy than monoclonal antibody The efficacy of immunotherapy is limited in a tumor immunosup­
therapies. pression microenvironment in which tumor-associated macrophages
Additionally, blocking CD47 not only increases cancer cell phago­ (TAMs) exhibit antigenic and immunosuppressive properties. In general,
cytosis but also promotes cross-presentation of tumor antigens, which in the M2 phenotype of TAMs promote tumor growth while the M1-like
turn activates the anti-tumor response of effector T cells. However, CD47 phenotype transition can inhibit tumor progression [31]. A novel ther­
is also highly expressed on red blood cells and platelets, and systemic apy strategy combining nanoparticle-loaded E. coli with immunogenic
CD47 blockade can cause anemia and thrombocytopenia, respectively. cell death (ICD) had been developed to regulate TAMs polarization and
To confine αCD47 to tumor tissue as much as possible, S. Chowdhury activate innate immune responses within the body. By combining with
et al. loaded engineered E. coli producing αCD47nb into an SLC (syn­ black phosphorus nanoparticles (NPs), E. coli strain MG1655 gained
chronized lysis circuit) [69] (Fig. 3c). This technology allows extra power to polarize TAMs towards the M1 phenotype, thereby
tumor-targeting bacteria to sense bacterial density and lyse them when a improving therapeutic efficacy [34]. Besides E. coli, Salmonella typhi­
critical density is reached. Since antibodies are not produced outside the murium strain YB1 had also been engineered to redirect M2 macrophages
tumor site, the delivery of therapeutic agent is limited to the tumor site. towards the M1 phenotype. These M2 macrophages thus exhibited a
The bacteria lysed about 20 h after growth initiation, followed by pe­ decreased expression of mannose receptor (an M2 phenotypic marker)
riodic growth-lysis oscillations during the log-phase growth period and and increased the expression of human leukocyte antigen D (an M1
beyond. Consequently, a larger number of actual antibody-releasing phenotypic marker) [82]. The resetting of macrophage to M1 phenotype
bacteria were present. Q. Feng et al. also attempted to block could also be attributed to the secretion of flagellin B (flaB) from the
CD47-SIRPα axis to repolarize TAMs towards M1, but they immobilized strain (Fig. 4a), as well as the TLR5-mediated host reactions in tumor
αCD47nb on bacterial-produced outer membrane vesicles (OMVs), and microenvironment [83]. The aforementioned findings showed that
facilitated the delivery of αCD47nb to macrophage [70,71]. Besides, the macrophages in the tumor microenvironments are a new target for
modification of PEG/selenium on the surface of OMVs further gave them tumor treatment.
radiation-response capability so that CD47 blockade scope could be Additionally, NK cells are a crucial element of the innate immune
easily controlled by adjusting irradiated area. system, lysing a variety of target cells and acting as early regulators of
cytokines [84]. In view of this, as one of the earliest bacterial vaccines,
2.5. Engineered bacteria inhibit tumor immune checkpoint expression Bacillus Calmette-Guérin (BCG) vaccine had demonstrated superior tumor
through gene delivery therapeutic efficacy through innate immune response mediated by NK
cells [85]. Now through the production of interferon-gamma (IFN-γ),
The anti-tumor efficacy of blocking the PD-1/PD-L1 axis has been engineered Salmonella YB1 could also activate NK cells and promote NK
repeatedly demonstrated. However, targeted antibodies are not the only cells accumulation, activation (Fig. 4a), and cytotoxicity through a
means to achieve this goal. For instance, PD-L1 genes in tumors could be self-regulating feedback loop, thereby significantly inhibiting metastasis
specifically silenced by using small interfering RNAs (siRNAs), thereby of multiple cancers [86].
preventing possible autoimmune side effects at the genetic level. Yet, the
application of siRNAs remains challenging due to its easy degradation in 3.2. Presentation of antigens by engineered bacteria
vivo and difficulty in effectively penetrating tumors. As a potential so­
lution, engineered bacteria have been explored as vehicles for siRNA Bacteria, as pathogens, have a complex chemical composition.
delivery due to their efficacy for intratumoral delivery and capacity for Moreover, each bacterium is a complex entity composed of multiple
cancer cell remodeling [72] (Fig. 3d). Even if the mutated progeny cells antigenic components that can be recognized by the host’s immune
counteract the effects of the gene therapy, the proliferating bacteria system, thereby eliciting an immune response. Therefore, if bacteria
ensure that new siRNA enters these cells. In view of this, T. Zhao et al. could transport and deliver tumor-associated antigens (TAAs) recog­
successfully developed attenuated S. typhimurium carrying a PD-L1 nized by the immune system, it has the potential to induce a specific and
siRNA sequence, increasing in the numbers of CD4+, CD8+ T cells and enduring anti-tumor adaptive immune response. Bacteria could be
NK cells in the spleen [73,74]. Besides, Salmonella was used to deliver genetically modified to present tumor-targeting components, including
PD-L1/PD-1 siRNA with similar effect in the similar effect in eliminating adhesive peptides and tumor-associated antigens (TAAs), on their cell
immunosuppression [75,76]. membrane [87]. Importantly, bacterial vector-based delivery of TAAs
As mentioned above, engineered bacteria generated immune killing could ingeniously bypass immune tolerance to self-tumor antigens and
effect through different therapeutic payloads (chemokines, cytokines elicit a potent targeting effect by inducing MHC-1 presentation and
and optimized antibodies). To perform their functions, these payloads activating specific CD8+ T cells [88]. Currently, bacteria have the ability
were also released in diverse ways, indicating greater combination may to deliver and promote immune system recognition of various types of
be applied in future studies. TAAs, such as tyrosinase-related protein-2 (TPR-2) [89], melanoma
antigen gene-b (Mage-B) [90,91], AH1 [92,93], and others. Engineered
3. Engineered bacteria assist immune effect Salmonella released the tumor antigens by a variety of methods, mainly
including direct release of TAAs, intracellular release of antigens fused
3.1. Engineered bacteria enhance innate cellular anti-tumor activity to type 3 secretion system (T3SS) proteins, or display of antigens on the
bacterial flagellum [94]. In addition, the use of engineered Salmonella
The innate immune system plays a critical role in regulating tumor for anti-tumor vaccine administration has also achieved encouraging
immunotherapy [77]. Certain cytokines and innate immune cells can results. For example, engineered Salmonella had been developed to

5
H. Chen et al. Cancer Letters 589 (2024) 216817

Fig. 4. Engineered bacteria assist immune effect in the immunotherapy. a. The additional expression help engineered bacteria increase specific activation of immune
cells. Expressed flagella protein flaB induced polarization of M1 to M2 and activation of NK cells. The IFN-γ secreted by engineered bacteria induced similar effects.
Engineered EcN with mutant N-acetylglutamate synthase (ArgAfgr) accumulated more L-arginine to nourish cytotoxic T cell inside tumor. b. Engineered bacteria
released endobacterial DNA to activate STING pathway, which could be also activated by secreted agonist cyclic di-AMP (c-di-AMP) from engineered bacteria. c.
Engineered Gram-positive bacteria like Lactobacillus plantarum regulated intestinal flora, to inhibit p53-p21 signaling pathway, and to suppress overexpression of
TLR1/NF-κB along with other proinflammatory factor, hence strengthen gut barrier to prevent colorectal cancer. d. Bacteria engineered to secreted chemokines
intratumorally like CXCL1, CXCL9 and CCL21. As a result, macrophages (Mφ), T cells, dendritic cells (DC), NK cells are effectively recruited into tumor microen­
vironment, eventually leads to death of tumor cells. Produced with permission from Biorender.

deliver virus-like particles of human papillomavirus type 16 (HPV16), Moreover, delivery of PDAC-associated antigen Annexin A2 (ANXA2)
thereby effectively inhibiting the growth of HPV16-expressing tumors using L. monocytogenes could activate specific CD8+ T cells and improve
[95]. In another case, engineered Salmonella decorated with survival rate [98]. Combination therapy of one HCC vaccine called
antigen-coated cationic polymer nanoparticles could enhance the Lmdd-MPFG based on the L. monocytogenes and anti-PD-1 immune
cross-talk between antigens and dendritic cells after intratumoral in­ checkpoint blockade antibody promoted the expression of PD-L1 in HCC
jection, leading to a significant increase in activation of cells while re-sensitizing the tumor-localized T cells in a response to
ovalbumin-specific dendritic cells and systemic anti-tumor effects [96]. PD-1 immunotherapy [99]. A recent research made another way, the
Except for engineered Salmonella, Listeria monocytogenes, another antigen released by tumor-colonizing probiotics labeled tumor tissue for
engineered bacteria, is widely used as a carrier for cancer vaccines. CAR-mediated lysis in situ [100]. After the probiotic was administered,
L. monocytogenes could invade antigen-presenting cells (APCs) and ex­ the CAR-T cells could be directed to the solid tumors, thus remarkably
press listeriolysin O (LLO) for antigen delivery. Currently, various enhance the CAR-T immunotherapy.
engineered L. monocytogenes expressing different antigens and targeting In addition to the two types of the abovementioned bacteria, many
different types of tumors have been developed. For example, a other bacteria have been genetically modified as antigen delivery vec­
microbial-based immunotherapeutic approach had been developed to tors. Pseudomonas aeruginosa could deliver TAAs into cancer cells
selectively deliver immunogenic tetanus toxin protein (TT856-1313) into through its T3SS system [101]. After three months of vaccination with
pancreatic ductal adenocarcinoma (PDAC) tumor cells through attenu­ engineered Pseudomonas aeruginosa vaccine in mice, significant rejection
ated L. monocytogenes [97]. This treatment reactivated pre-existing reactions were observed in melanomas expressing ovalbumin, indicating
TT-specific memory T cells to kill the infected tumor cells in mice. activation of memory T cells [102]. Furthermore, enhanced vaccination

6
H. Chen et al. Cancer Letters 589 (2024) 216817

significantly increased the activation of memory T cells and improved response to alterations in the microbiome [115]. Currently, numerous
the survival rate of mice, suggesting that engineered Pseudomonas aer­ types of probiotics have been developed and shown to be beneficial in a
uginosa could provide effective and long-lasting immunity in mice. E. coli mouse models of colitis-associated colorectal cancer (CAC) induced by
was also developed as an antigen delivery vector, which was genetically azoxymethane (AOM)/sodium dextran sulfate (DSS) [116–119].
modified to co-express antigen with LLO [103]. A recent study used Another article had revealed the antitumor immunity enhancement in
genetically modified Staphylococcus epidermidis to express melanoma of bacteria by elucidating a critical microbial-host crosstalk
tumor-specific antigens. This modified commensal bacterium induced between probiotic-product and CD8 T cells within the tumor microen­
specific CD8+ and CD4+ T cell responses after topical application to vironment [120].
mouse skin. This novel commensal microorganism did not cause infec­ Recently, a study reported a dual-gene box-equipped Lactobacillus
tion and did not require internal transport to enter the tumor. Moreover, plantarum encoded therapeutic protein, P8, called PP*-P8, which
it did not induce high inflammatory signals in vivo. In summary, sig­ inhibited the p53-p21 signaling pathway and induced G1 cell cycle ar­
nificant progress has been made in the introduction of tumor-associated rest in human colorectal adenocarcinoma epithelial cells (DLD-2 cells).
antigens using engineered bacteria, providing a safer and more direct The administration of PP*-P8 probiotics alleviated the ecological
approach to cancer therapy [104]. imbalance induced by AOM/DSS through the regulation of α- and β-di­
versity of intestinal microbial communities and the proportions of
3.3. Activation of STING pathway in antigen-presenting cells (APC) potentially beneficial taxa [116]. Lactobacillus rhamnosus LS8 (LRL)
could also alleviate colorectal cancer (CRC) by modulating the gut mi­
Research on innate immune agonists has gained momentum in the crobial community structure and preventing intestinal barrier damage
biomedical field due to the limited effectiveness of immune checkpoint and inflammation. Specifically, by suppressing overexpression of
inhibitors in cancer patients [105]. Among these agonists, stimulator of TLR1/NF-κB, pro-inflammatory cytokines (TNF-α, IL-6β, IL-17, IL-γ, and
interferon genes (STING) agonists [106–108] have garnered significant IL-1a), chemokines (CXCL1, CXCL2, CXCL3, CXCL5, and CXCL7), LRL
interest. By activating the STING pathway in immune cells, these ago­ ameliorated colonic inflammation [117] (Fig. 4c). Moreover, other
nists stimulate the production of interferons, thereby eliciting probiotics, such as Bifidobacterium [118] and butyrate-producing Clos­
anti-tumor immune responses. Compared to conventional small mole­ tridia [119], have the potential to enhance gut microbiota regulation of
cule STING (smSTING) agonists that may cause “off-target” effects on immunity, thereby improving the effectiveness of immunotherapy.
effector T cells, engineered bacteria, such as modified E. coli (EcN) and
S. typhimurium [109], have emerged as more desirable carriers for STING 3.5. Release of chemokines to enhance immune cell penetration
agonists [110]. Immune cells could present or phagocytose these bac­
teria, thereby triggering complementary immune pathways via pattern Engineered bacteria can produce and release chemokines within the
recognition receptors (PRRs) like Toll-like receptors (TLRs) and estab­ tumor to attract adaptive immune cells into the environment. Chemo­
lishing immune memory. kines are mediators of transporting immune cells and play a crucial role
An engineered strain of S. typhimurium expressing cGAMP was also in the tumor microenvironment, both promoting and inhibiting tumor
designed to activate the STING pathway in THP-1 macrophages and growth [121]. Many chemokines are engineered for expression in bac­
human primary dendritic cells (DCs) [111]. Likewise, a previous study teria and for recruitment or maturation of immune cells within tumors.
found that the modified E. coli strain SYNB1891 producing the Some previous studies have shown that the interactions of therapeutic
STING-agonist cyclic di-AMP (CDA) could specifically target Listeria monocytogenes with TAM could increase the production of CXCL1
antigen-presenting cells (APCs) within tumors [110] (Fig. 4b). This [122] and other chemokines [123]. On the contrary, CXCL9 and CXCL10
activation of the STING pathway in APCs leaded to the production of protein synthesis were upregulated by TLR-4 after infection with Sal­
type I interferons (IFNs). Importantly, the modified SYNB1891 could monella choleraesuis [124].
further activate complementary innate immune pathways via its bac­ A study successfully engineered two bacterial strains expressing
terial chassis, thereby inducing immune memory. The work of Waanders CXCL16 and CCL20, respectively, which recruited T cells and dendritic
L on S. typhimurium expressing cGAMP showed that an 87-fold stronger cells to synergistically activate anti-tumor immune responses [125]. The
IFN-β response in THP-I macrophages [111]. Apart from DC activation first bacterial strain was designed to recruit activated T cells within
and type I IFN response, they firstly found T3S system necessary for tumors by intravenous or intratumoral injection of the activated mutant
IFN-β induction in epithelial cells. A nother study also confirmed that human chemokine CXCL16 (hCXCL16K42A) and had shown therapeutic
two modified E. coli strains, CIBT4523 and CIBT4712, had potent tumor effect in a variety of mouse tumor models. In parallel, the second
suppressive effects and activated the STING pathway in a mouse model engineered bacterial strain expressing CCL20 was designed to recruit
[112]. Encouragingly, SYNB1891 was evaluated in an ongoing Phase I dendritic cells presenting tumor antigens, thereby triggering an immune
clinical trial, which demonstrated a favorable safety and tolerability activation cascades [125]. This combined strategy resulted in the
profile as monotherapy or in combination with atezolizumab via recruitment of type I conventional dendritic cells, which effectively
repeated intratumoral injection [113]. In one recent research, W. Chen synergized with hCXCL16K42A-induced T-cell recruitment, thereby
coated STING agonist MSA-2 with biocompatible polydopamine on the providing additional therapeutic benefits. Such engineered bacterial
bacterial surface of ECN [114]. Such engineered bacterial delivery co-recruitment strategy activated innate and adaptive anti-tumor im­
platform holds potential in elevating levels of type I interferon (IFN) and mune responses, and was considered as a novel strategy for cancer
proinflammatory cytokines, serving as a potential personalized cancer immunotherapy [125]. The chemokine CCL21 expressed by engineered
vaccine with enhanced biosafety and immune activation profile for S. typhimurium also enhanced antitumor activity [126]. In a multi-drug
diverse applications. resistant preclinical mouse cancer model, it was demonstrated that
attenuated S. typhimurium expressing CCL21 significantly inhibited the
3.4. Probiotics regulate intestinal flora to influence immune system growth of primary tumors and lung metastases. Histological analysis of
the tumors showed a significant increase in the infiltration of inflam­
The intestinal flora plays a vital role in mediating various immune matory cells (e.g., CXCL9, CXCL10, and INF-γ) in mice treated with
activities, including the differentiation of naive T cells, production of CCL21-expressing bacteria, whereas no significant difference was
cytokines, and activation of bone marrow cells. Additionally, the rela­ observed in mice treated with control bacteria (Fig. 4d).
tive abundance, and immune characteristics of the microbiome are
associated with the incidence of tumors. Due to its high abundance and
robust characteristics, the colon displays a more delicate immune

7
H. Chen et al. Cancer Letters 589 (2024) 216817

3.6. Nourish immune cells immunotherapy.


Besides, another study surprisingly discovered that a strain of L-
In addition to their immunoregulatory function, tumor-infiltrating arginine producing bacteria had better antitumor efficacy. In this strain,
bacteria can directly nourish immune cells, remodel the tumor micro­ the arginine repressor gene ArgR was deleted, and a feedback-resistant
environment (TME), and indirectly promote adaptive immune re­ dominant mutant gene ArgAfbr not inhibited by high levels of L-argi­
sponses. A well-known example is the genetic engineering of a non- nine was integrated into the bacterial genome. When such bacteria
pathogenic E. coli strain, ECN, to convert the tumor metabolic waste infected MC38 tumor in mice models, higher concentrations of arginine
product ammonia into L-arginine, thereby increasing the accumulation converted from immunomodulatory metabolites was observed
of tumor-infiltrating lymphocytes (TILs). Through transamination ac­ compared to wild-type EcN, thus resulting in significant TIL infiltration
tivity, ECN increased the concentration of L-arginine in the tumor and [128] (Fig. 4a). Meanwhile, the study also found that mice whose tu­
nourished CD4+ and CD8+ T cells while decreasing the number of mors had completely regressed no longer grew when they were
FOXP3+ T regulatory cells [15]. The results further showed that ECN re-infected three months later, suggesting the formation of a long-term
had a significant synergistic effect in tumor clearance when used immune anti-tumor memory, demonstrating the great potential of this
together with PD-L1 blocking antibodiess [15,127]. The antitumor ef­ bacterium [128]. Interestingly, clinical trials with L-arginine-producing
fects of these bacteria are mediated by L-arginine and are dependent on T bacteria had yielded positive results in the treatment of hyper­
cells. These findings suggest the potential of engineered microbial ammonemia and hepatic encephalopathy [129–131].
therapies in modulating tumor metabolism to enhance the efficacy of In addition to its ability to metabolize ammonia, the gene-edited ECN

Fig. 5. Engineered bacteria participate in combination therapy. a. Engineered bacteria combined with ICIs treatments. The combination treatment with anti-4-1BB,
anti-PD-1, anti-CTLA-4 achieved more adaptive immune effect. b. Engineered bacteria combined with phototherapy. Near infrared ray (NIR) produces heat in the
presence of photosensitizer. Tumor-invading bacteria attached photosensitizer through linker, thus produced intratumoral heat. The upconversion nanoparticles on
bacteria transformed NIR into blue light, which activated bacterial photoswitch to regulate downstream expression. c. Fe3O4 nanoparticles attached to engineered
bacteria transform magnetic power into heat, exposure to magnetic field activates thermos-lysis circuit inside such bacteria to express CD47 nanobody downstream.
d. Ionic radiation activates endobacterial DNA repair response. Based on that, the downstream TNF-α of repairing-element RecA elements expresses response to
external irradiation. Produced with permission from Biorender.

8
H. Chen et al. Cancer Letters 589 (2024) 216817

was also able to utilize phenylalanine (Phe) to treat phenylketonuria. 4.2. Combined with photothermal therapy
Under hypoxic conditions in the mammalian gut, the engineered strain
SYNB1618 encoding a phenylalanine metabolizing enzyme could Photothermal therapy (PTT) has enormous potential in the field of
convert phenylalanine to trans-cinnamic acid salts. The host then cancer treatment. Under near-infrared irradiation, light-absorbing ma­
quantitatively synthesized these salts into hippuric acid salts, which terial with high photothermal conversion efficiency can convert light
were excreted in the urine, thereby reducing neurotoxicity [132]. energy into heat and induce immunogenic cell death (ICD) in tumor
Through regulating the nutrient metabolism in tumor microenvi­ cells. However, the limited depth of penetration by near-infrared light
ronment, we can see new ways for engineered bacteria to participate in and the inability of most light-absorbing material to reach deep-seated
cancer immunotherapy. This is also a new strategy for cancer immu­ tumors often leads to insufficient tumor killing, resulting in tumor
notherapy, as localized precise trophic remodeling is considered an recurrence and metastasis [144–146]. This limitation can be overcome
effective way to treat tumor sites, but was previously difficult to achieve by using bacteria as carriers to deliver light-absorbing materials deep
[133,134]. Therefore, engineered bacteria show us great potential not into tumor tissue. Additionally, the design of photoactivatable expres­
only as carriers for drug delivery, but also to address new therapeutic sion circuits in bacteria allows for the targeted release of drugs at the
theories. appropriate time, enhancing ICD and improving the efficacy of immu­
notherapy (Fig. 5b).
4. Engineered bacteria participating in combination X. Sun et al. utilized the biocompatible hybrid “N–V-J″ agent in his
immunotherapy study [147]. Under NIR irradiation, the resulting local high temperature
released the BET inhibitor JQ-1, and then significantly reduced the
4.1. Combination therapy with immune checkpoint inhibitors (ICIs) expression of PD-L1 on tumor cells and tumor-associated dendritic cells.
In W. Lin’s latest research, an iridium (III) photosensitizer was cova­
Bacteria have limited ability to activate specific antitumor immunity lently labeled to the surface of genetically engineered EcN [148]. This
through innate immunity. To achieve more effective and stable specific strain is the first metal complex decorated bacteria capable of inducing
antitumor immunity, researchers have combined bacterial therapies pyroptosis and immunogenic cell death of tumor cells under irradiation,
with immune checkpoint inhibitors (ICIs) (Fig. 5a). In a study conducted remarkably evoking anti-tumor innate and adaptive immune responses
by B.H. Al-Saafeen [135], treatment of tumors with attenuated in vivo. Superparamagnetic Fe3O4 nanoparticles convert magnetic en­
S. typhimurium induced an increase in CD11b + Gr-1+ myeloid cells and ergy into heat, too [149]. With the help of that, engineered E. coli could
M1 polarization of Tumor-associated macrophages (TAMs), as well as release the pre-expressed CD47 nanobodies inside the bacteria by a
significant increases in the expression of various activation markers such heat-sensitive cell-lysing process, to deliver precise tumor immuno­
as MHC II and costimulatory molecules. Notably, the combination of therapy [150] (Fig. 5c).
αPD-L1 significantly enhanced these effects and inhibited tumor growth Optogenetic modules have become important tools in the field of
more effectively than αPD-L1 alone [136]. For example, D.C. Binder synthetic biology. Blue-light control systems, which are led by CRY2/
[90] created attenuated S. typhimurium A1-R with tumor antigens. When CIB, VVD, and EL222, can regulate gene transcription, cell metabolism,
combined with αPD-L1, effector CD8+ T cells showed enhanced killing and other processes rapidly by activating or deactivating photosensitive
of antigenically homologous tumors. The continuous antigen secretion proteins. For instance, using EL222 as a template, X. Zhu designed EcN
by engineered bacteria also established long-term specific responses in T that perceived the release of blue light-encoded flaB [137]. By trans­
cells. Y. Li used attenuated S. typhimurium SL3261 to co-stimulate the forming nanoparticles, near-infrared light is converted into blue light,
activation and proliferation of tumor antigen-recognized T cells through which initiates synchronous cancer immunotherapy. Furthermore, the
4-1BB/4-1BBL. This was achieved by restoring 4-1BBL expression on secretion of flab which activated by light started the Myd88-dependent
colon cancer tumors with a eukaryotic plasmid and then binding to immune response, which combined with PDT to provide a combined
αPD-L1 to eliminate CD8+ T cell suppression [137]. Besides, a tumor immunotherapy effect of inhibiting distant tumor growth. C. Wu also
vaccine VXM01 that delivered VEGFR2 via Salmonella also showed used upconversion particles to transform NIR into blue-violet light to
complementary effects with αPD-L1 in tumor treatment. Both vaccines activate bacterial production of interferon-gamma (INF-γ) in the intes­
are currently being used in phase II clinical trials for the combined tine [46] (Fig. 5b). Y. Xiao controlled the high expression of tyrosinase
treatment of glioblastoma [138]. in EcN [151], generating in-situ synthesis of melanin within tumors,
Besides, Bifidobacterium has also shown potential to enhance αPD-L1 providing stronger photothermal performance that assisted in treating
efficacy. In mouse lymphoid tissue, infection with bifidobacterium cancer. By encoding luciferase [152] or fluorophore activating protein
induced T cell proliferation at lower antigen concentrations and acti­ (FAP) [153], engineered bacteria could interact with fluorescent sub­
vated CD8+ T cells more efficiently [139]. Subsequently, the same strates for photothermal therapy (PTT) or photodynamic treatment
research group engineered Bifidobacterium to upregulate PD-L1 expres­ (PDT). Additionally, Zhu, Z. found Cerenkov radiation-induced photo­
sion in tumors with IFN-γ secretion, followed by combination with dynamic therapy (CR-PDT) a feasible scheme for the PDT excited by the
αPD-L1 for remolding the tumor matrix and promoting cancer cell death. internal light [154]. Their EcN loaded with aggregation-induced emis­
Additionally, K. Shioya developed Bifidobacterium longum APS001 sion photosensitizer was co-enriched with 18F-FDG in the tumor site to
[140], expressing uracil deaminase, which could convert fluorouracil trigger CR-PDT. The presence of Cerenkov radiation seemed able to
into 5-fluorouracil. When combined with αPD-L1, the animal survival induce more ICD than traditional PDT.
rates were significantly improved compared to single treatments.
Recently, Akkermansia muciniphila, a naturally occurring bacterium 4.3. Combined with radiotherapy
extracted from the gastrointestinal tract of healthy human, was reported
to predict clinical response to PD-1 blockade in patients with advanced Radiotherapy is a non-invasive method with high direct killing effi­
non-small-cell lung cancer [141]. This kind of bacterium quickly ciency, allowing high precision and penetrating treatment of deep-
attracted a great deal of attention because it was the first gut microbe to seated tumors. Importantly, radiotherapy could significantly induce
reverse epithelial tumor resistance to PD-1 blockers [142,143]. A bio­ immunogenic cell death (ICD) [155]. However, the efficacy of conven­
logical agents based on this bacterium called Oncobax®-AK has already tional radiotherapy is limited by the “oxygen effect,” which reduces the
been under phase I clinical trial. With the aid of that, NSCLC patients are lethality of the hypoxic regions of the tumor. Fortunately, most
expected to safely take oral administration for better immunotherapy parthenogenetic anaerobic bacteria are able to colonize and proliferate
effect. in the hypoxic regions of the tumor, playing a complementary role. The
combination of radiotherapy with bacterial therapy could enhance the

9
H. Chen et al. Cancer Letters 589 (2024) 216817

immune response to metastatic or distant tumors through the abscopal tumor cells by radiation (Fig. 5d). The discovery of radio-responsive
effect stimulated by immune formation [156,157]. These findings sug­ elements is exciting. This modification could produce alternative stra­
gested that radiation and engineered bacteria could work together to tegies for weakly penetrating near-infrared light used earlier in combi­
achieve better therapeutic results. nation therapy and enable these strategies to be more effective. In 2020,
Platt verified that attenuated S. typhimurium significantly enhanced Gao and his team modified this strategy with promoter recN, another
the anti-tumor effect of radiotherapy [158]. This effect was observed in radiation-inducible promoter that was more sensitive than recA. The
the dose ranging of 5–15 Gy, regardless of whether radiation was new engineered bacteria showed a synergistic anti-tumor effect in mu­
segmented or not. In a separate study, Bettegowda explored the effec­ rine tumor model with radiotherapy.
tiveness of combining anaerobic bacteria C. novyi-NT with external As mentioned above, this article has already summarized 3 popular
beam radiation therapy (EBRT), brachytherapy (BT), and stereotactic combination therapy in which engineered bacteria participate. Among
body radiation therapy (SBRT) [159]. It was found that anaerobic bac­ them, the combination with ICIs shows the greatest potential to be
teria could selectively destroy tumor hypoxia regions, thus improving translated into clinical practice, but the shortcoming of ICIs listed at the
the efficacy of all forms of radiotherapy. This was the first instance of beginning of this article still remain unsolved. The combination with
bacterial-induced radiosensitization explained by the oxygen effect in PTT therapy and radiotherapy may get rid of this problem, but associ­
radiotherapy, which proved inspiring to future research. Later, Yoon ated modules are now not sensitive enough to meet clinical needs.
et al. [160] observed similar radiosensitization results on Several light-control systems of bacteria has shown good efficiency,
irradiation-resistant B16F10 melanoma treated with S. typhimurium. since themselves are product of evolution in prokaryotic biota, but the
Further experiments indicated that S. typhimurium amplified the actual performance seem limited by penetrating power of visible light.
γ-ray-induced increase of reactive oxygen species (ROS) and H2AX After all, the combination with radiotherapy could carry out under
phosphorylation, while upregulating the Caspase-3 mediated apoptosis realistic condition, so it shall become one novel promising chance for
pathway of tumor cells. The induced apoptosis suggested that it might engineered bacteria to come into clinical applications, but only if the
activate the body’s immune system to further eliminate tumors through radiation-responsive elements are correctly upgraded.
ICD. Moreover, Burdelya et al. used the purified flagellin CBLB502 to
activate TLR5 receptors for enhanced radiotherapy [161]. In parallel, 5. Discussion
CBLB502 also showed direct anti-proliferative effects on certain tumors
without irradiation. These results suggested that bacterial components The concept of engineered bacteria offers a new strategy for single or
could enhance radio-immunotherapy by activating the immune system. combined immunotherapy utilizing their targeted immunologic regu­
Engineered bacteria with specific secretion functions were used to lation, demonstrating their unique influence on immunotherapy directly
coordinate re-establishing radiotherapy almost simultaneously when or indirectly. The molecular modification of bacteria not only enhances
engineered bacteria began to be used in tumor treatment experiments. In their interaction with the immune system, but also achieve spatial and
2010, Jiang et al. [162] utilized a sub-strain of E. coli K-12 expressing the temporal drug release profiles that differ from those of conventional
cytotoxin ClyA to significantly improve the efficacy of radiotherapy and treatment [23], facilitating the ability of tumors to tolerate bacterial or
significantly inhibit lung metastasis. Similarly, Liu et al. [163] observed immunological payloads. For the first time, immunotherapeutic agents
improvements in radiotherapy results using a S. typhimurium ΔppGpp have overcome the limitations of traditional pharmacokinetics in cancer
strain expressing ClyA, which could enter cells. This effect may also be treatment by virtue of live bacteria secretion. Now, bacteria are not only
related to intracellular induction of TNF-α by bacteria or activated immunogenic vectors but also producers of cytokines [40–42,44,45] and
antitumor T cells within bacterial colonization areas. Pan et al. [164] antibodies [66–68], converters of tumor metabolic waste into nutrients
built on Jiang’s work by attaching Bi2S3 nanoparticles to the surface of [128–131], and initiators of bacterial production via external signals
ClyA-expressing E. coli to increase DNA damage to tumor cells. Yoon [40,45,46,137,147–149]. Participation of engineered bacteria even
et al. [164] successfully delivered INHA miRNA into B16F10 melanoma creates innovative opportunities for immunotherapy. In such combina­
cells of mice through the genetic information transfer function of engi­ tion immunotherapy, engineered bacteria can flexibly switch functions
neered bacteria, achieving radiosensitization at the gene level. to provide either immunoregulatory enhancement, greater ICDs, or
Some engineered bacterial derivative have also showed great po­ more importantly, immunotherapeutic agents itself. In recent years, we
tential in radio-immunotherapy. A recent research utilized radionuclide have witnessed the gradual entry of engineered bacteria for immuno­
labeled inactivated salmonella to further activation of cGAS-STING therapy into clinical trials, as summarized in Table 1.
pathway [165]. Feng and his team successfully extracted outer mem­ Despite the potential for better outcomes, the application of engi­
brane vesicles (OMVs) enriched with CD47 nanobodies from E. coli and neered bacteria in immunotherapy has been restricted. The primary
coated them with a layer of poly (ethylene glycol) containing diselenide concern is safety, which is necessary for such live bacteria preparation
bonds (PEG/Se) [70]. This coating not only reduced the immunogenicity [167]. During treatment, intravenous injection of engineered bacteria
of OMVs before irradiation but also added radiation-responsive may create a transient bacteremia hazard, which will lead to a higher
controlled release ability to OMVs. In their experiments, OMVs were risk of mortality and risk factors [168,169]. Moreover, the proliferation
induced by radiation to expose CD47nb on their surface, thereby of live bacteria would complicate the assessment of equivalent doses of
inducing M1 polarization while blocking the SIRPα pathway, and acti­ immunological agent, making applications susceptible to overdose. The
vating the phagocytosis of TAM, effectively reshaping the tumor second limitation is efficiency. The interactions between different bac­
microenvironment (TME) at the irradiated tumor sites. Qi et al. [166] terial within the tumor may lead to the change of their phenotypes such
constructed intratumoral bacterial cellulose nanofibers framework for as invasiveness and virulence, thus hindering therapeutic effects
long-term retention of 131I-labeled αPD-L1. Besides from the immobili­ [170–172]. Synthetic-biologically engineered Bacteria may not be effi­
zation of radionuclide, bacterial cellulose also enhanced the immuno­ ciently modified due to plasmid loss. Therefore, despite the great
genic cell death (ICD) of cancer cells, activating the maturation of achievements of synthetic biology today, there is still a need for more
multiple immune cells to induce a systemic anti-tumor immune effect. advanced engineering approaches, such as cleaner deletion of wild
Platt et al. had already achieved the modification of bacterial radio- virulence genes and tighter control of responses to external stimuli.
response elements as early as 2001 [40]. They found that recA compo­
nents in the SOS repair system could upregulate downstream tran­ 6. Future outlook
scription when bacteria were irradiated. By adding this element
upstream of TNFα-cDNA transferred into S. typhimurium, bacteria After years of development and practice, bioengineering techniques
secreted 44% more TNF-α after radiation, which greatly aided in killing have become quite sophisticated. However, both immunization and the

10
H. Chen et al. Cancer Letters 589 (2024) 216817

Table 1
Clinical trials applying engineered bacteria for immunotherapy.
Clinical trial Species Strain Engineered agents Combination Applications Start Status
identifiersa year

NCT01099631 Salmonella — IL-2 — Hepatoma 2010 phase I


NCT04589234 typhimurium Saltikva IL-2 — Metastatic Pancreatic 2020 phase II
Cancer
NCT00004216 VNP20009 — — Solid tumors 1999 phase I
NCT00006254 Solid tumors 2000 phase I
NCT00004988 metastatic cancer 2000 phase I
NCT03762291 CVD908ssb-TXSVN Antigen: survivin — Multiple Myeloma 2021 phase I
NCT04167137 Escherichia coli SYNB1891 STING Agonist atezolizumab Solid Tumors and 2019 phase I
Lymphoma
NCT01266460 Listeria ADXS11-001 Antigen: Human Papilloma — Cervical Cancer 2011 phase II
NCT01598792 monocytogenes Virus HPV+ Oropharyngeal 2012 phase I
NCT02002182 Genotype 16 Target Antigens Carcinoma 2013 phase II
NCT01967758 ADU-623 Antigen: EGFRvIII-NY-ESO-1 — Astrocytomas 2014 phase I
NCT03847519 ADXS-503 Antigen: 22 NSCLC-associated Pembrolizumab Lung Cancer 2019 phase II
tumor antigens
NCT02004262 CRS 207 Antigen: mesothelin GVAX Pancreas Pancreatic 2014 phase II
Vaccine Adenocarcinoma
NCT02575807 Epacadostat Ovarian, Fallopian, 2016 phase II
Peritoneal Cancer
NCT01675765 — Malignant Pleural 2014 phase I
Mesothelioma
NCT02592967 JNJ-64041757 NSCLC 2015 phase I
NCT03371381 Nivolumab Lung Cancer 2018 phase II
NCT02625857 JNJ-64041809 Antigen: 4 antigens relevant — Metastatic Prostate Cancer 2015 phase I
NCT02906605 to prostate cancer Apalutamide 2016 phase II
NCT03189030 pLADD personalized antigen — Metastatic Colorectal 2017 phase I
Cancer
NCT04025307 Bifidobacterium bacTRL-IL-12 pro-inflammatory IL-12 — Solid tumors 2019 phase I
longum transgene
NCT00358397 Clostridium novy-NT C. novyi-NT spores — — Solid tumors 2006 phase I
NCT01118819 2011 phase I
NCT03435952 Pembrolizumab 2018 phase I
NCT04857697 gut microbiome engineered gut — — Breast and Lung Cancer 2021 early
microbiome phase I
NCT05865730 Akkermansia Oncobax®-AK — anti PD-1 NSCLC or RCC 2022 phase II
muciniphila
a
Clinical trial identifiers currently listed on ClinicalTrials.gov.

Fig. 6. Outlook for immunotherapeutic future of engineered bacteria. a. Commensal bacteria show more affinity for relevant tumor. With help of single-bacteria
sequencing, we can filtrate better engineering hosts. b. Researchers could find advanced control circuit for more sensitivity and more specificity. c. Genetic code
expansion (GCE) enables bacteria express unusual amino acids, indicating advanced engineered bacteria could present special chemical group for wider application.
To click to drug using click-chemistry, for example. d. As a recent big hit, artificial intelligence could assist engineered bacteria development. With the aid of AI
biocomputing technology, more protein could be predicted correctly and guide further discovery. Produced with permission from Biorender.

11
H. Chen et al. Cancer Letters 589 (2024) 216817

microbiome are fraught with complexity and finesse, and there are still study, Zhang’s team used AI characterized and engineered the extra­
more ingenious targets and more efficient immunomodulatory mecha­ cellular contractile injection systems (eCISs) called Photorhabdus viru­
nisms to be discovered in the content of bacterial engineering. At the lence cassette (PVC) [175]. This brave attempt reminds us that some
same time, the diversity of engineered bacteria in the future means a field difficult to study in past time shall hand over to AI. We will soon
wider range of clinical application scenarios, hence the dilemma of understand how facultative anaerobe can aggregate inside tumor, we
engineered bacteria’s poor performance in clinical trials may alter will soon know on what those rare bacteria rely to survive extreme
radically. environment, and we will soon facilitate inverse translation of newfound
In the near future, the list of bacteria to engineer is firstly expected to structure and thus engineer them into different cell (Fig. 6d). AI means
update. Although diverse payloads of engineered bacteria have shown productivity, with that we will be able to challenge the boundary of our
therapeutic potential for clinical patients, those bacteria host remain too modern cognition. We believe that in the near future, a large number of
virulent for them. Michael A. Fischbach’s work on commensal bacteria special biological structures will be fully studied and transferred into our
had enlightened us [104]. Knowing that bacteria often play different tool bacteria.
roles according to different somatotopic sites, scientists may dig out
more commensal bacteria from patient’s tumors as new personalized CRediT authorship contribution statement
tools in future works (Fig. 6a). The unique ways to safely apply
commensal bacteria (for example, surface coating for skin bacteria) are Hua Chen: Writing – original draft. Yinrui Zhu: Writing – original
also hopeful to replace intravenous administration. Inspired by CAR-T, draft. Chonghai Zhang: Software. Lin Hu: Writing – review & editing.
we can further optimize such strategy: extract bacteria from tumor tis­ Kai Yang: Writing – review & editing.
sues, engineer them according to individual need in the lab and then
apply back to the patients. Bacteria are usually easier to culture and
require less time for genetic editing comparing to CAR-T, so this strategy Declaration of competing interest
will certainly lead to lower costs. Besides bacteria, commensal micro­
organism are expected to be modified as well, such as fungi in the skin The authors declare that they have no known competing financial
and viruses in the genitals, which may exert better immune effects than interests or personal relationships that could have appeared to influence
bacteria. We believe, for greater therapeutic effects on related tumors or the work reported in this paper.
other diseases, great efforts shall be soon devoted on analyzing and
engineering those commensal microorganisms.
Acknowledgments
Secondly, the strategy of engineered bacteria shall be redesigned.
Engineered bacteria have created an opportunity for in situ protein
This work was partially supported by the National Natural Science
production, promising greater controllability in clinical drug delivery
Foundation of China (U1932208, 32171382), Key Research and Devel­
strategies. Nevertheless, the control circuits within engineered bacteria
opment Program of Social Development of Jiangsu Province
still have room for improvement, as the present modules cannot totally
(BE2022725), Gusu Leading Talent Program for Innovation and Entre­
prevent leakage while ensuring sensitivity [40,155,173]. Nature is full
preneurship (No. ZXL2021441), and the Project Funded by the Priority
of wonders, variety kinds of bacteria have evolved to survive extreme
Academic Program Development of Jiangsu Higher Education In­
environment, so there must exist advanced response mechanism among
stitutions (PAPD).
them waiting to be discovered, optimized, and applied for our engi­
neering strategy (Fig. 6b). On the other hand, therapeutic payloads as
well need optimizing. In clinic, most drugs cannot easily synthesized by References
bacteria due to the limits of prokaryotic system. Recently, an advanced
[1] M. Abbott, Y. Ustoyev, Cancer and the immune system: the history and
gene editing technique called genetic code expansion (GCE) have background of immunotherapy, Semin. Oncol. Nurs. 35 (2019) 150923, https://
inspired us [174]. Through inserting unnatural amino acid (UAA) into doi.org/10.1016/j.soncn.2019.08.002.
[2] H.A. Tawbi, et al., Relatlimab and nivolumab versus nivolumab in untreated
target protein molecular, we can induce abundant chemical groups that
advanced melanoma, N. Engl. J. Med. 386 (2022) 24–34, https://doi.org/
never exist in nature (Fig. 6c). Bacteria can even be engineered to utilize 10.1056/NEJMoa2109970.
microelement of host body to assemble specific chemical group if con­ [3] P. Sharma, S. Hu-Lieskovan, J.A. Wargo, A. Ribas, Primary, adaptive, and
dition permits. These chemical groups may have therapeutic effects. acquired resistance to cancer immunotherapy, Cell 168 (2017) 707–723, https://
doi.org/10.1016/j.cell.2017.01.017.
Once produced steadily from engineered bacteria, the total effect de­ [4] L.B. Kennedy, A.K.S. Salama, A review of cancer immunotherapy toxicity, CA A
pends on the real-time amount of the bacteria inside concerning tissue. Cancer J. Clin. 70 (2020) 86–104, https://doi.org/10.3322/caac.21596.
However, direct therapeutic group displayed on bacteria may do same [5] S. Bagchi, R. Yuan, E.G. Engleman, Immune checkpoint inhibitors for the
treatment of cancer: clinical impact and mechanisms of response and resistance,
harm to themselves to influence their viability, so those chemical groups Annual review of pathology 16 (2021) 223–249, https://doi.org/10.1146/
may also act as linkers of external medicine. For example, displaying annurev-pathol-042020-042741.
alkyne groups on bacteria surface, covalently attaching antibodies using [6] Z. Chen, Y. Cheng, D. DeRemer, V. Diaby, Cost-effectiveness and drug wastage of
immunotherapeutic agents for hematologic malignancies: a systematic review,
click chemistry, and making the bacterial surface more attuned to tu­ Expert Rev. Pharmacoecon. Outcomes Res. 21 (2021) 923–941, https://doi.org/
mors or immune cells, resulting in more immune interactions. In this 10.1080/14737167.2021.1913056.
way, although effector (external medicine) cannot automatically pro­ [7] S. Mucherino, et al., Cost-effectiveness of treatment optimisation with biomarkers
for immunotherapy in solid tumours: a systematic review protocol, BMJ Open 11
duce, the precise targeting is insured. Therefore, the clinical doctors only
(2021) e048141, https://doi.org/10.1136/bmjopen-2020-048141.
need to inject bacteria once, wait for a period of time for targeting, and [8] W.B. Coley, The treatment of malignant tumors by repeated inoculations of
periodically replenish these topical drugs until the patient is cured. erysipelas. With a report of ten original cases, Clin. Orthop. Relat. Res. 3–11
(1991), 1893.
Either way, therapeutic effect ends with the removal of engineered
[9] A.C. Allison, J. Ferluga, Editorial: how lymphocytes kill tumor cells, N. Engl. J.
bacteria, so the doctor can quit the therapy any time only if antibiotics Med. 295 (1976) 165–167, https://doi.org/10.1056/nejm197607152950311.
are used. [10] J. Wybran, Cancer immunotherapy, Dermatol. 157 (1978) 415–417, https://doi.
Artificial intelligence (AI) technology is altering the academic world. org/10.1159/000250868.
[11] R. Mitra, S. Singh, A. Khar, Antitumour immune responses, Expet Rev. Mol. Med.
We are now facing the liberated computing power automatically to 5 (2003) 1–19, https://doi.org/10.1017/s1462399403005623.
collect and analyze vast amounts of data, which is revolutionary for [12] B.A. Beutler, TLRs and innate immunity, Blood 113 (2009) 1399–1407, https://
molecular biology and immunology. With the help of AI program, we are doi.org/10.1182/blood-2008-07-019307.
[13] L. Rong, Q. Lei, X.Z. Zhang, Engineering living bacteria for cancer therapy, ACS
able to predict protein structure and function precisely. As to the simple Appl. Bio Mater. 3 (2020) 8136–8145, https://doi.org/10.1021/
prokaryotes, AI may predict even the subcellular structures. In the latest acsabm.0c01286.

12
H. Chen et al. Cancer Letters 589 (2024) 216817

[14] J.R. Moese, G. Moese, Oncolysis by clostridia. I. Activity of Clostridium butyricum [43] G. Kak, M. Raza, B.K. Tiwari, Interferon-gamma (IFN-γ): exploring its
(M-55) and other nonpathogenic clostridia against the ehrlich carcinoma, Cancer implications in infectious diseases, Biomol. Concepts 9 (2018) 64–79, https://doi.
Res. 24 (1964) 212–216. org/10.1515/bmc-2018-0007.
[15] Q. Liu, Y. Gai, Y. Chen, X. Lan, D. Jiang, Escherichiacoli Nissle 1917 as a novel [44] W. Yoon, et al., Application of genetically engineered Salmonella typhimurium
microrobot for tumor-targeted imaging and therapy, Pharmaceutics 13 (2021), for interferon-gamma-induced therapy against melanoma, Eur. J. Cancer 70
https://doi.org/10.3390/pharmaceutics13081226. (2017) 48–61, https://doi.org/10.1016/j.ejca.2016.10.010.
[16] K.B. Low, et al., Lipid A mutant Salmonella with suppressed virulence and TNFα [45] Y. Chen, M. Du, Z. Yuan, Z. Chen, F. Yan, Spatiotemporal control of engineered
induction retain tumor-targeting in vivo, Nat. Biotechnol. 17 (1999) 37–41, bacteria to express interferon-gamma by focused ultrasound for tumor
https://doi.org/10.1038/5205. immunotherapy, Nat. Commun. 13 (2022) 4468, https://doi.org/10.1038/
[17] X. Zhang, et al., Heterologous expression of carcinoembryonic antigen in s41467-022-31932-x.
Lactococcus lactis via LcsB-mediated surface displaying system for oral vaccine [46] C. Wu, et al., NIR-responsive photodynamic nanosystem combined with
development, Journal of microbiology, immunology, and infection = Wei mian antitumor immune optogenetics bacteria for precise synergetic therapy, ACS
yu gan ran za zhi 49 (2016) 851–858, https://doi.org/10.1016/j. Appl. Mater. Interfaces 14 (2022) 13094–13106, https://doi.org/10.1021/
jmii.2014.11.009. acsami.2c01138.
[18] T. Sasaki, et al., Genetically engineered Bifidobacterium longum for tumor- [47] Y. Wu, et al., Disrupting the phase separation of KAT8-IRF1 diminishes PD-L1
targeting enzyme-prodrug therapy of autochthonous mammary tumors in rats, expression and promotes antitumor immunity, Nature cancer 4 (2023) 382–400,
Cancer Sci. 97 (2006) 649–657, https://doi.org/10.1111/j.1349- https://doi.org/10.1038/s43018-023-00522-1.
7006.2006.00221.x. [48] Y. Seki, et al., Abstract 2631: enhanced anti-tumor effects by a combination
[19] L.K. Hawkins, N.R. Lemoine, D. Kirn, Oncolytic biotherapy: a novel therapeutic approach of interferon-γ producing recombinant Bifidobacterium and anti-mPD-1
platform, Lancet Oncol. 3 (2002) 17–26, https://doi.org/10.1016/s1470-2045 antibody in syngeneic mouse model, Cancer Res. 77 (2017) 2631, https://doi.
(01)00618-0. org/10.1158/1538-7445.
[20] J.F. Toso, et al., Phase I study of the intravenous administration of attenuated [49] D.M. Klinman, A.K. Yi, S.L. Beaucage, J. Conover, A.M. Krieg, CpG motifs present
Salmonella typhimurium to patients with metastatic melanoma, J. Clin. Oncol. 20 in bacteria DNA rapidly induce lymphocytes to secrete interleukin 6, interleukin
(2002) 142–152, https://doi.org/10.1200/jco.2002.20.1.142. 12, and interferon gamma, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 2879–2883,
[21] A.S. Khalil, J.J. Collins, Synthetic biology: applications come of age, Nat. Rev. https://doi.org/10.1073/pnas.93.7.2879.
Genet. 11 (2010) 367–379, https://doi.org/10.1038/nrg2775. [50] M.M. Curtis, S.S. Way, Interleukin-17 in host defence against bacterial,
[22] J.X. Fan, M.T. Niu, Y.T. Qin, Y.X. Sun, X.Z. Zhang, Progress of engineered bacteria mycobacterial and fungal pathogens, Immunology 126 (2009) 177–185, https://
for tumor therapy, Adv. Drug Deliv. Rev. 185 (2022) 114296, https://doi.org/ doi.org/10.1111/j.1365-2567.2008.03017.x.
10.1016/j.addr.2022.114296. [51] F. Biet, C. Locht, L. Kremer, Immunoregulatory functions of interleukin 18 and its
[23] C.R. Gurbatri, N. Arpaia, T. Danino, Engineering bacteria as interactive cancer role in defense against bacterial pathogens, J. Mol. Med. (Berl.) 80 (2002)
therapies, Science 378 (2022) 858–864, https://doi.org/10.1126/science. 147–162, https://doi.org/10.1007/s00109-001-0307-1.
add9667. [52] al, B.K. Ramadi, M.J. Fernandez-Cabezudo, N. Mustafa, D. Xu, Activation of
[24] T. Keren-Kaplan, et al., Synthetic biology approach to reconstituting the innate immune responses by IL-2-expressing Salmonella typhimurium is
ubiquitylation cascade in bacteria, EMBO J. 31 (2012) 378–390, https://doi.org/ independent of Toll-like receptor 4, Mol. Immunol. 40 (2004) 671–679, https://
10.1038/emboj.2011.397. doi.org/10.1016/j.molimm.2003.09.005.
[25] C.A. Voigt, Bacteria collaborate to sense arsenic, Nature 481 (2012) 33–34, [53] B. Sorenson, et al., Safety and immunogenicity of Salmonella typhimurium
https://doi.org/10.1038/481033a. expressing C-terminal truncated human IL-2 in a murine model, Biologics 4
[26] C.A. Voigt, Synthetic biology: bacteria collaborate to sense arsenic, Nature 481 (2010) 61–73, https://doi.org/10.2147/btt.s9121.
(2012) 33–34, https://doi.org/10.1038/481033a. [54] B.S. Sorenson, K.L. Banton, N.L. Frykman, A.S. Leonard, D.A. Saltzman,
[27] M. Fussenegger, Synchronized bacterial clocks, Nature 463 (2010) 301–302, Attenuated Salmonella typhimurium with interleukin 2 gene prevents the
https://doi.org/10.1038/463301a. establishment of pulmonary metastases in a model of osteosarcoma, J. Pediatr.
[28] J. Liang, Y. Luo, H. Zhao, Synthetic biology: putting synthesis into biology, in: Surg. 43 (2008) 1153–1158, https://doi.org/10.1016/j.jpedsurg.2008.02.048.
Wiley Interdiscip Rev Syst Biol Med, vol. 3, 2011, pp. 7–20, https://doi.org/ 55 B.K. al-Ramadi, et al., Potent anti-tumor activity of systemically-administered IL2-
10.1002/wsbm.104. expressing Salmonella correlates with decreased angiogenesis and enhanced tumor
[29] B.B. Finlay, S. Falkow, Salmonella as an intracellular parasite, Mol. Microbiol. 3 apoptosis, Clin Immunol 130 (2009) 89–97, https://doi.org/10.1016/j.
(1989) 1833–1841, https://doi.org/10.1111/j.1365-2958.1989.tb00170.x. clim.2008.08.021.
[30] Y. Zhang, et al., Listeria hijacks host mitophagy through a novel mitophagy [56] Y. Wu, et al., Secreting-lux/pT-ClyA engineered bacteria suppresses tumor growth
receptor to evade killing, Nat. Immunol. 20 (2019) 433–446, https://doi.org/ via interleukin-1 beta in two pathways, Amb. Express 9 (2019) 189, https://doi.
10.1038/s41590-019-0324-2. org/10.1186/s13568-019-0910-6.
[31] A. Lukic, et al., GM-CSF- and M-CSF-primed macrophages present similar [57] L. Wang, et al., Bifidobacterium breve as a delivery vector of IL-24 gene therapy
resolving but distinct inflammatory lipid mediator signatures, Faseb. J. 31 (2017) for head and neck squamous cell carcinoma in vivo, Gene Ther. 24 (2017)
4370–4381, https://doi.org/10.1096/fj.201700319R. 699–705, https://doi.org/10.1038/gt.2017.74.
[32] X. Cheng, X. Zhang, Y. Zhou, C. Zhang, Z.C. Hua, A Salmonella Typhimurium [58] M. Loeffler, G. Le’Negrate, M. Krajewska, J.C. Reed, IL-18-producing Salmonella
mutant strain capable of RNAi delivery: higher tumor-targeting and lower inhibit tumor growth, Cancer Gene Ther. 15 (2008) 787–794, https://doi.org/
toxicity, Cancer Biol. Ther. 15 (2014) 1068–1076, https://doi.org/10.4161/ 10.1038/cgt.2008.48.
cbt.29185. [59] Z.A. Ahmad, et al., scFv antibody: principles and clinical application, Clin. Dev.
[33] H. Li, et al., Nanozyme-coated bacteria hitchhike on CD11b(+) immune cells to Immunol. (2012) 980250, https://doi.org/10.1155/2012/980250, 2012.
boost tumor radio-immunotherapy, Adv. Mater. (2023) e2309332, https://doi. [60] T. Chanier, P. Chames, Nanobody engineering: toward next generation
org/10.1002/adma.202309332. immunotherapies and immunoimaging of cancer, Antibodies 8 (2019), https://
[34] B. Wei, et al., Polarization of tumor-associated macrophages by nanoparticle- doi.org/10.3390/antib8010013.
loaded Escherichia coli combined with immunogenic cell death for cancer [61] H. Lou, X. Cao, Antibody variable region engineering for improving cancer
immunotherapy, Nano Lett. 21 (2021) 4231–4240, https://doi.org/10.1021/acs. immunotherapy, Cancer Commun. 42 (2022) 804–827, https://doi.org/10.1002/
nanolett.1c00209. cac2.12330.
[35] C. Yin, et al., Light-triggered photosynthetic engineered bacteria for enhanced- [62] S. Ahangarzadeh, et al., An update on antiviral antibody-based
photodynamic therapy by relieving tumor hypoxic microenvironment, biopharmaceuticals, Int. Immunopharm. 86 (2020) 106760, https://doi.org/
Theranostics 13 (2023) 1632–1648, https://doi.org/10.7150/thno.81718. 10.1016/j.intimp.2020.106760.
[36] S. Zhou, C. Gravekamp, D. Bermudes, K. Liu, Tumour-targeting bacteria [63] M. Lopez-Medina, A. Perez-Lopez, C. Alpuche-Aranda, V. Ortiz-Navarrete,
engineered to fight cancer, Nat. Rev. Cancer 18 (2018) 727–743, https://doi.org/ Salmonella induces PD-L1 expression in B cells, Immunol. Lett. 167 (2015)
10.1038/s41568-018-0070-z. 131–140, https://doi.org/10.1016/j.imlet.2015.08.004.
[37] E.A. Carswell, et al., An endotoxin-induced serum factor that causes necrosis of [64] C. Michikawa, et al., Fusobacterium is enriched in oral cancer and promotes
tumors, Proc. Natl. Acad. Sci. U. S. A. 72 (1975) 3666–3670, https://doi.org/ induction of programmed death-ligand 1 (PD-L1), Neoplasia 31 (2022) 100813,
10.1073/pnas.72.9.3666. https://doi.org/10.1016/j.neo.2022.100813.
[38] A. Degterev, M. Boyce, J. Yuan, A decade of caspases, Oncogene 22 (2003) [65] S.A. Lee, et al., Escherichia coli K12 upregulates programmed cell death ligand 1
8543–8567, https://doi.org/10.1038/sj.onc.1207107. (PD-L1) expression in gamma interferon-sensitized intestinal epithelial cells via
[39] F. Balkwill, Tumour necrosis factor and cancer, Nat. Rev. Cancer 9 (2009) the NF-kappaB pathway, Infect. Immun. 89 (2020), https://doi.org/10.1128/
361–371, https://doi.org/10.1038/nrc2628. IAI.00618-20.
[40] S. Nuyts, et al., Radio-responsive recA promoter significantly increases TNFα [66] M.H. Abedi, et al., Ultrasound-controllable engineered bacteria for cancer
production in recombinant clostridia after 2 Gy irradiation, Gene Ther. 8 (2001) immunotherapy, Nat. Commun. 13 (2022) 1585, https://doi.org/10.1038/
1197–1201, https://doi.org/10.1038/sj.gt.3301499. s41467-022-29065-2.
[41] S.L. Young, et al., Cytokine-modified Mycobacterium smegmatis as a novel [67] W. Xu, et al., Bacteria-mediated tumor immunotherapy via photothermally-
anticancer immunotherapy, Int. J. Cancer 112 (2004) 653–660, https://doi.org/ programmed PD1 expression, Nanoscale Adv. 4 (2022) 1577–1586, https://doi.
10.1002/ijc.20442. org/10.1039/d1na00857a.
[42] W.S. Yoon, Y.S. Chae, J. Hong, Y.K. Park, Antitumor therapeutic effects of a [68] C.R. Gurbatri, et al., Engineered probiotics for local tumor delivery of checkpoint
genetically engineered Salmonella typhimurium harboring TNF-alpha in mice, blockade nanobodies, Sci. Transl. Med. 12 (2020), https://doi.org/10.1126/
Appl. Microbiol. Biotechnol. 89 (2011) 1807–1819, https://doi.org/10.1007/ scitranslmed.aax0876.
s00253-010-3006-4.

13
H. Chen et al. Cancer Letters 589 (2024) 216817

[69] S. Chowdhury, et al., Programmable bacteria induce durable tumor regression [97] B.C. Selvanesan, et al., Listeria delivers tetanus toxoid protein to pancreatic
and systemic antitumor immunity, Nat. Med. 25 (2019) 1057–1063, https://doi. tumors and induces cancer cell death in mice, Sci. Transl. Med. 14 (2022)
org/10.1038/s41591-019-0498-z. eabc1600, https://doi.org/10.1126/scitranslmed.abc1600.
[70] Q. Feng, et al., Engineered bacterial outer membrane vesicles as controllable two- [98] V.M. Kim, et al., Anti-pancreatic tumor efficacy of a Listeria-based, Annexin A2-
way adaptors to activate macrophage phagocytosis for improved tumor targeting immunotherapy in combination with anti-PD-1 antibodies,
immunotherapy, Adv. Mater. 34 (2022) e2206200, https://doi.org/10.1002/ J Immunother Cancer 7 (2019) 132, https://doi.org/10.1186/s40425-019-0601-
adma.202206200. 5.
[71] M. Kaparakis-Liaskos, R.L. Ferrero, Immune modulation by bacterial outer [99] G. Xu, et al., Listeria-based hepatocellular carcinoma vaccine facilitates anti-PD-1
membrane vesicles, Nat. Rev. Immunol. 15 (2015) 375–387, https://doi.org/ therapy by regulating macrophage polarization, Oncogene 39 (2020) 1429–1444,
10.1038/nri3837. https://doi.org/10.1038/s41388-019-1072-3.
[72] C.K. Baban, M. Cronin, D. O’Hanlon, G.C. O’Sullivan, M. Tangney, Bacteria as [100] R.L. Vincent, et al., Probiotic-guided CAR-T cells for solid tumor targeting 382
vectors for gene therapy of cancer, Bioengineered Bugs 1 (2010) 385–394, (2023) 211–218, https://doi.org/10.1126/science.add7034.
https://doi.org/10.4161/bbug.1.6.13146. [101] O. Epaulard, et al., Anti-tumor immunotherapy via antigen delivery from a live
[73] T. Zhao, et al., PD-1-siRNA delivered by attenuated Salmonella enhances the attenuated genetically engineered Pseudomonas aeruginosa type III secretion
antimelanoma effect of pimozide, Cell Death Dis. 10 (2019) 164, https://doi.org/ system-based vector, Mol. Ther. 14 (2006) 656–661, https://doi.org/10.1016/j.
10.1038/s41419-019-1418-3. ymthe.2006.06.011.
[74] T. Zhao, et al., Combination of attenuated Salmonella carrying PD-1 siRNA with [102] Y. Wang, et al., Optimization of antitumor immunotherapy mediated by type III
nifuroxazide for colon cancer therapy, J. Cell. Biochem. 121 (2020) 1973–1985, secretion system-based live attenuated bacterial vectors, J. Immunother. 35
https://doi.org/10.1002/jcb.29432. (2012) 223–234, https://doi.org/10.1097/CJI.0b013e31824747e5.
[75] X. Jia, et al., Antitumor effects and mechanisms of CpG ODN combined with [103] K.J. Radford, et al., A recombinant E. coli vaccine to promote MHC class I-
attenuated Salmonella-delivered siRNAs against PD-1, Int. Immunopharm. 90 dependent antigen presentation: application to cancer immunotherapy, Gene
(2021) 107052, https://doi.org/10.1016/j.intimp.2020.107052. Ther. 9 (2002) 1455–1463, https://doi.org/10.1038/sj.gt.3301812.
[76] S. Lu, et al., PD-1-siRNA delivered by attenuated Salmonella enhances the [104] Y.E. Chen, et al., Engineered skin bacteria induce antitumor T cell responses
antitumor effect of chloroquine in colon cancer, Front. Immunol. 12 (2021) against melanoma, Science 380 (2023) 203–210, https://doi.org/10.1126/
707991, https://doi.org/10.3389/fimmu.2021.707991. science.abp9563.
[77] S.M. Crusz, F.R. Balkwill, Inflammation and cancer: advances and new agents, [105] P. Darvin, S.M. Toor, V. Sasidharan Nair, E. Elkord, Immune checkpoint
Nat. Rev. Clin. Oncol. 12 (2015) 584–596, https://doi.org/10.1038/ inhibitors: recent progress and potential biomarkers, Exp. Mol. Med. 50 (2018)
nrclinonc.2015.105. 1–11, https://doi.org/10.1038/s12276-018-0191-1.
[78] A. Mantovani, P. Allavena, A. Sica, F. Balkwill, Cancer-related inflammation, [106] J. Fu, et al., STING agonist formulated cancer vaccines can cure established
Nature 454 (2008) 436–444, https://doi.org/10.1038/nature07205. tumors resistant to PD-1 blockade, Sci. Transl. Med. 7 (2015), https://doi.org/
[79] S. Yan, G. Wan, Tumor-associated macrophages in immunotherapy, FEBS J. 288 10.1126/scitranslmed.aaa4306, 283ra252.
(2021) 6174–6186, https://doi.org/10.1111/febs.15726. [107] L. Corrales, et al., Direct activation of STING in the tumor microenvironment
[80] O. Demaria, et al., Harnessing innate immunity in cancer therapy, Nature 574 leads to potent and systemic tumor regression and immunity, Cell Rep. 11 (2015)
(2019) 45–56, https://doi.org/10.1038/s41586-019-1593-5. 1018–1030, https://doi.org/10.1016/j.celrep.2015.04.031.
[81] C.H. Liu, H. Liu, B. Ge, Innate immunity in tuberculosis: host defense vs pathogen [108] S.R. Woo, et al., STING-dependent cytosolic DNA sensing mediates innate
evasion, Cell. Mol. Immunol. 14 (2017) 963–975, https://doi.org/10.1038/ immune recognition of immunogenic tumors, Immunity 41 (2014) 830–842,
cmi.2017.88. https://doi.org/10.1016/j.immuni.2014.10.017.
[82] M. Yang, J. Xu, Q. Wang, A.-Q. Zhang, K. Wang, An obligatory anaerobic [109] B. Larkin, et al., Cutting edge: activation of STING in T cells induces type I IFN
Salmonella typhimurium strain redirects M2 macrophages to the M1 phenotype, responses and cell death, J. Immunol. 199 (2017) 397–402, https://doi.org/
Oncol. Lett. 15 (2018) 3918–3922, https://doi.org/10.3892/ol.2018.7742. 10.4049/jimmunol.1601999.
[83] J.H. Zheng, et al., Two-step enhanced cancer immunotherapy with engineered [110] D.S. Leventhal, et al., Immunotherapy with engineered bacteria by targeting the
Salmonella typhimurium secreting heterologous flagellin, Sci. Transl. Med. 9 STING pathway for anti-tumor immunity, Nat. Commun. 11 (2020) 2739, https://
(2017), https://doi.org/10.1126/scitranslmed.aak9537. doi.org/10.1038/s41467-020-16602-0.
[84] M.A. Cooper, T.A. Fehniger, M.A. Caligiuri, The biology of human natural killer- [111] L. Waanders, et al., Ectopic expression of cGAS in Salmonella typhimurium
cell subsets, Trends Immunol. 22 (2001) 633–640. enhances STING-mediated IFN-β response in human macrophages and dendritic
[85] H. Suttmann, et al., Mechanisms of bacillus Calmette-Guerin mediated natural cells, J Immunother Cancer 11 (2023), https://doi.org/10.1136/jitc-2022-
killer cell activation, J. Urol. 172 (2004) 1490–1495. 005839.
[86] Q. Lin, et al., IFN-γ-dependent NK cell activation is essential to metastasis [112] Y. Jiang, et al., Fine-tuning bacterial cyclic di-AMP production for durable
suppression by engineered Salmonella, Nat. Commun. 12 (2021) 2537, https:// antitumor effects through the activation of the STING pathway, Research 6
doi.org/10.1038/s41467-021-22755-3. (2023) 0102, https://doi.org/10.34133/research.0102.
[87] B.F. Sieow, K.S. Wun, W.P. Yong, I.Y. Hwang, M.W. Chang, Tweak to treat: [113] J.J. Luke, et al., Phase I study of SYNB1891, an engineered E. coli Nissle strain
reprograming bacteria for cancer treatment, Trends Cancer 7 (2021) 447–464, expressing STING agonist, with and without atezolizumab in advanced
https://doi.org/10.1016/j.trecan.2020.11.004. malignancies, Clin. Cancer Res. 29 (2023) 2435–2444, https://doi.org/10.1158/
[88] S. Chouaib, C. Asselin-Paturel, F. Mami-Chouaib, A. Caignard, J.Y. Blay, The host- 1078-0432.CCR-23-0118.
tumor immune conflict: from immunosuppression to resistance and destruction, [114] W. Chen, et al., in: Potentiating the Systemic Immunity by Bacteria-Delivered
Immunol. Today 18 (1997) 493–497, https://doi.org/10.1016/s0167-5699(97) Sting Activation in a Tumor Microenvironment, vol. 33, 2023 2307001, https://
01115-8. doi.org/10.1002/adfm.202307001.
[89] X. Zhu, et al., Tumor antigen delivered by Salmonella III secretion protein fused [115] K. Martinez-Guryn, V. Leone, E.B. Chang, Regional diversity of the
with heat shock protein 70 induces protection and eradication against murine gastrointestinal microbiome, Cell Host Microbe 26 (2019) 314–324, https://doi.
melanoma, Cancer Sci. 101 (2010) 2621–2628, https://doi.org/10.1111/j.1349- org/10.1016/j.chom.2019.08.011.
7006.2010.01722.x. [116] Y. Chung, et al., A synthetic probiotic engineered for colorectal cancer therapy
[90] D.C. Binder, et al., Antigen-specific bacterial vaccine combined with anti-PD-L1 modulates gut microbiota, Microbiome 9 (2021) 122, https://doi.org/10.1186/
rescues dysfunctional endogenous T cells to reject long-established cancer, Cancer s40168-021-01071-4.
Immunol. Res. 1 (2013) 123–133, https://doi.org/10.1158/2326-6066.CIR-13- [117] T. Wang, et al., Lacticaseibacillus rhamnosus LS8 ameliorates azoxymethane/
0058. dextran sulfate sodium-induced colitis-associated tumorigenesis in mice via
[91] A. Jahangir, et al., Immunotherapy with Listeria reduces metastatic breast cancer regulating gut microbiota and inhibiting inflammation, Probiotics Antimicrob
in young and old mice through different mechanisms, OncoImmunology 6 (2017) Proteins 14 (2022) 947–959, https://doi.org/10.1007/s12602-022-09967-9.
e1342025, https://doi.org/10.1080/2162402X.2017.1342025. [118] Q. Wang, et al., Administration of Bifidobacterium bifidum CGMCC 15068
[92] W. Deng, et al., Recombinant Listeria promotes tumor rejection by CD8(+) T cell- modulates gut microbiota and metabolome in azoxymethane (AOM)/dextran
dependent remodeling of the tumor microenvironment, Proc. Natl. Acad. Sci. U. sulphate sodium (DSS)-induced colitis-associated colon cancer (CAC) in mice,
S. A. 115 (2018) 8179–8184, https://doi.org/10.1073/pnas.1801910115. Appl. Microbiol. Biotechnol. 104 (2020) 5915–5928, https://doi.org/10.1007/
[93] D.G. Brockstedt, et al., Listeria-based cancer vaccines that segregate s00253-020-10621-z.
immunogenicity from toxicity, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) [119] M. Liu, W. Xie, X. Wan, T. Deng, Clostridium butyricum modulates gut microbiota
13832–13837, https://doi.org/10.1073/pnas.0406035101. and reduces colitis associated colon cancer in mice, Int. Immunopharm. 88 (2020)
[94] T.D. Doan, H.Y. Wang, G.M. Ke, L.T. Cheng, N-Terminus of flagellin fused to an 106862, https://doi.org/10.1016/j.intimp.2020.106862.
antigen improves vaccine efficacy against pasteurella multocida infection in [120] M.J. Bender, et al., Dietary tryptophan metabolite released by intratumoral
chickens, Vaccines 8 (2020), https://doi.org/10.3390/vaccines8020283. Lactobacillus reuteri facilitates immune checkpoint inhibitor treatment, Cell 186
[95] V. Revaz, et al., Mucosal vaccination with a recombinant Salmonella (2023) 1846–1862.e1826, https://doi.org/10.1016/j.cell.2023.03.011.
typhimurium expressing human papillomavirus type 16 (HPV16) L1 virus-like [121] F. Markl, D. Huynh, S. Endres, S. Kobold, Utilizing chemokines in cancer
particles (VLPs) or HPV16 VLPs purified from insect cells inhibits the growth of immunotherapy, Trends Cancer 8 (2022) 670–682, https://doi.org/10.1016/j.
HPV16-expressing tumor cells in mice, Virology 279 (2001) 354–360, https:// trecan.2022.04.001.
doi.org/10.1006/viro.2000.0717. [122] P.H. Lizotte, et al., Attenuated Listeria monocytogenes reprograms M2-polarized
[96] W. Wang, et al., Systemic immune responses to irradiated tumours via the tumor-associated macrophages in ovarian cancer leading to iNOS-mediated tumor
transport of antigens to the tumour periphery by injected flagellate bacteria, Nat. cell lysis, OncoImmunology 3 (2014) e28926.
Biomed. Eng. 6 (2022) 44–53, https://doi.org/10.1038/s41551-021-00834-6.

14
H. Chen et al. Cancer Letters 589 (2024) 216817

[123] W. Deng, et al., Recombinant Listeria promotes tumor rejection by CD8+ T cell- [150] X. Ma, et al., Modular-designed engineered bacteria for precision tumor
dependent remodeling of the tumor microenvironment, Proc. Natl. Acad. Sci. U. immunotherapy via spatiotemporal manipulation by magnetic field, Nat.
S. A. 115 (2018) 8179–8184, https://doi.org/10.1073/pnas.1801910115. Commun. 14 (2023) 1606, https://doi.org/10.1038/s41467-023-37225-1.
[124] C.H. Lee, C.L. Wu, A.L. Shiau, Toll-like receptor 4 mediates an antitumor host [151] Y. Xiao, et al., In-situ synthesis of melanin in tumor with engineered probiotics for
response induced by Salmonella choleraesuis, Clin. Cancer Res. 14 (2008) hyperbaric oxygen-synergized photothermal immunotherapy, Nano Today 47
1905–1912, https://doi.org/10.1158/1078-0432.CCR-07-2050. (2022), https://doi.org/10.1016/j.nantod.2022.101632.
[125] T.M. Savage, et al., Chemokines expressed by engineered bacteria recruit and [152] X. Zhu, et al., Near-infrared nano-optogenetic activation of cancer
orchestrate antitumor immunity, Sci. Adv. (2022), https://doi.org/10.1126/ immunotherapy via engineered bacteria, Adv. Mater. (2022) e2207198, https://
sciadv.adc9436. doi.org/10.1002/adma.202207198.
[126] M. Loeffler, G. Le’Negrate, M. Krajewska, J.C. Reed, Salmonella typhimurium [153] Y. Guo, et al., Photodynamic therapy-improved oncolytic bacterial
engineered to produce CCL21 inhibit tumor growth, Cancer Immunol. immunotherapy with FAP-encoding S. typhimurium, J. Contr. Release 351 (2022)
Immunother. 58 (2009) 769–775, https://doi.org/10.1007/s00262-008-0555-9. 860–871, https://doi.org/10.1016/j.jconrel.2022.09.050.
CII. [154] Z. Zhu, et al., Aggregation-induced emission photosensitizer/bacteria biohybrids
[127] J.R. Cubillos-Ruiz, A. Cubillos-Ruiz, Engineered bacteria recycle tumor metabolic enhance Cerenkov radiation-induced photodynamic therapy by activating anti-
waste to boost immunotherapy, Cell Host Microbe 29 (2021) 1725–1727, https:// tumor immunity for synergistic tumor treatment, Acta Biomater. (2023), https://
doi.org/10.1016/j.chom.2021.11.008. doi.org/10.1016/j.actbio.2023.06.009.
[128] F.P. Canale, et al., Metabolic modulation of tumours with engineered bacteria for [155] T. Liu, P. Pei, W. Shen, L. Hu, K. Yang, Radiation-induced immunogenic cell death
immunotherapy, Nature 598 (2021) 662–666, https://doi.org/10.1038/s41586- for cancer radioimmunotherapy, Small Methods 7 (2023) e2201401, https://doi.
021-04003-2. org/10.1002/smtd.202201401.
[129] C.B. Kurtz, et al., An engineered E. coli Nissle improves hyperammonemia and [156] E.B. Golden, et al., Local radiotherapy and granulocyte-macrophage colony-
survival in mice and shows dose-dependent exposure in healthy humans, Sci. stimulating factor to generate abscopal responses in patients with metastatic solid
Transl. Med. 11 (2019), https://doi.org/10.1126/scitranslmed.aau7975. tumours: a proof-of-principle trial, Lancet Oncol. 16 (2015) 795–803, https://doi.
[130] C. Nicaise, et al., Control of acute, chronic, and constitutive hyperammonemia by org/10.1016/s1470-2045(15)00054-6.
wild-type and genetically engineered Lactobacillus plantarum in rodents, [157] Y. Zhang, et al., The role of bacteria and its derived biomaterials in cancer
Hepatology 48 (2008) 1184–1192, https://doi.org/10.1002/hep.22445. radiotherapy, Acta Pharm. Sin. B (2022), https://doi.org/10.1016/j.
[131] R. Ochoa-Sanchez, et al., Genetically engineered E. coli Nissle attenuates apsb.2022.10.013.
hyperammonemia and prevents memory impairment in bile-duct ligated rats, [158] J. Platt, et al., in: Antitumour Effects of Genetically Engineered Salmonella in
Liver Int. 41 (2021) 1020–1032, https://doi.org/10.1111/liv.14815. Combination with Radiation, vol. 36, 2000, pp. 2397–2402, https://doi.org/
[132] V.M. Isabella, et al., Development of a synthetic live bacterial therapeutic for the 10.1016/s0959-8049(00)00336-1. European journal of cancer (Oxford, England :
human metabolic disease phenylketonuria, Nat. Biotechnol. 36 (2018) 857–864, 1990).
https://doi.org/10.1038/nbt.4222. [159] C. Bettegowda, et al., Overcoming the hypoxic barrier to radiation therapy with
[133] F. Peyraud, et al., Circulating L-arginine predicts the survival of cancer patients anaerobic bacteria, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 15083–15088,
treated with immune checkpoint inhibitors, Ann. Oncol. 33 (2022) 1041–1051, https://doi.org/10.1073/pnas.2036598100.
https://doi.org/10.1016/j.annonc.2022.07.001. [160] W.S. Yoon, S. Kim, S. Seo, Y. Park, Salmonella typhimurium with gamma-
[134] C.L. Chen, et al., Arginine is an epigenetic regulator targeting TEAD4 to modulate radiation induced H2AX phosphorylation and apoptosis in melanoma, Biosci.
OXPHOS in prostate cancer cells, Nat. Commun. 12 (2021) 2398, https://doi.org/ Biotechnol. Biochem. 78 (2014) 1082–1085, https://doi.org/10.1080/
10.1038/s41467-021-22652-9. 09168451.2014.905173.
[135] S. Kaimala, et al., Salmonella-mediated tumor regression involves targeting of [161] L.G. Burdelya, et al., Toll-like receptor 5 agonist protects mice from dermatitis
tumor myeloid suppressor cells causing a shift to M1-like phenotype and and oral mucositis caused by local radiation: implications for head-and-neck
reduction in suppressive capacity, Cancer Immunol. Immunother. 63 (2014) cancer radiotherapy, Int. J. Radiat. Oncol. Biol. Phys. 83 (2012) 228–234,
587–599, https://doi.org/10.1007/s00262-014-1543-x. https://doi.org/10.1016/j.ijrobp.2011.05.055.
[136] B.H. Al-Saafeen, et al., Attenuated Salmonella potentiate PD-L1 blockade [162] X. Liu, S. Jiang, L. Piao, F. Yuan, Radiotherapy combined with an engineered
immunotherapy in a preclinical model of colorectal cancer, Front. Immunol. 13 Salmonella typhimurium inhibits tumor growth in a mouse model of colon
(2022) 1017780, https://doi.org/10.3389/fimmu.2022.1017780. cancer, Exp. Anim. 65 (2016) 413–418, https://doi.org/10.1538/expanim.16-
[137] Y. Li, et al., Efficacy of bivalent CEACAM6/4-1BBL genetic vaccine combined with 0033.
anti-PD1 antibody in MC38 tumor model of mice, Heliyon 8 (2022) e10775, [163] P. Pan, X. Dong, Y. Chen, X. Zeng, X.Z. Zhang, Engineered bacteria for enhanced
https://doi.org/10.1016/j.heliyon.2022.e10775. radiotherapy against breast carcinoma, ACS Nano 16 (2022) 801–812, https://
[138] W. Wick, et al., in: Oral DNA Vaccination Targeting VEGFR-2 Combined with doi.org/10.1021/acsnano.1c08350.
Anti-PD-L1 Avelumab in Patients with Progressive Glioblastoma, a Phase I/II [164] W. Yoon, Y. Park, S. Kim, Y. Park, C.Y. Kim, Combined therapy with microRNA-
Study: NCT03750071, vol. 37, 2019, p. TPS2076, https://doi.org/10.1200/ expressing Salmonella and irradiation in melanoma, Microorganisms 9 (2021),
JCO.2019.37.15_suppl.TPS2076. https://doi.org/10.3390/microorganisms9112408.
[139] A. Sivan, et al., Commensal Bifidobacterium promotes antitumor immunity and [165] P. Pei, et al., Pleiotropic immunomodulatory functions of radioactive inactivated
facilitates anti–PD-L1 efficacy, Science 350 (2015) 1084–1089, https://doi.org/ bacterial vectors for enhanced cancer radio-immunotherapy, ACS Nano 16 (2022)
10.1126/science.aac4255. 11325–11337, https://doi.org/10.1021/acsnano.2c04982.
[140] K. Shioya, et al., Potentiated antitumor effects of APS001F/5-FC combined with [166] Z. Qi, et al., 131)I-alphaPD-L1 immobilized by bacterial cellulose for enhanced
anti-PD-1 antibody in a CT26 syngeneic mouse model, Biosci. Biotechnol. radio-immunotherapy of cancer, J. Contr. Release 346 (2022) 240–249, https://
Biochem. 85 (2021) 324–331, https://doi.org/10.1093/bbb/zbaa057. doi.org/10.1016/j.jconrel.2022.04.029.
[141] L. Derosa, et al., Intestinal Akkermansia muciniphila predicts clinical response to [167] Q. Tang, et al., Current status and future directions of bacteria-based
PD-1 blockade in patients with advanced non-small-cell lung cancer, Nat. Med. 28 immunotherapy, Front. Immunol. 13 (2022) 911783, https://doi.org/10.3389/
(2022) 315–324, https://doi.org/10.1038/s41591-021-01655-5. fimmu.2022.911783.
[142] B. Routy, et al., Gut microbiome influences efficacy of PD-1-based [168] R. Kullar, E.J.C. Goldstein, S. Johnson, L.V. McFarland, Lactobacillus bacteremia
immunotherapy against epithelial tumors, Science (New York, N.Y.) 359 (2018) and probiotics: a review, Microorganisms 11 (2023), https://doi.org/10.3390/
91–97, https://doi.org/10.1126/science.aan3706. microorganisms11040896.
[143] M. Bae, et al., Akkermansia muciniphila phospholipid induces homeostatic [169] M. Shimura, et al., Probiotic-related bacteremia after major hepatectomy for
immune responses, Nature 608 (2022) 168–173, https://doi.org/10.1038/ biliary cancer: a report of two cases, Surg Case Rep 7 (2021) 133, https://doi.org/
s41586-022-04985-7. 10.1186/s40792-021-01216-5.
[144] D. Zhi, T. Yang, J. O’Hagan, S. Zhang, R.F. Donnelly, Photothermal therapy, [170] A.R. McWhorter, R. Tearle, T.S. Moyle, K.K. Chousalkar, In vivo passage of
J. Contr. Release 325 (2020) 52–71, https://doi.org/10.1016/j. Salmonella Typhimurium results in minor mutations in the bacterial genome and
jconrel.2020.06.032. increases in vitro invasiveness, Vet. Res. 50 (2019) 71, https://doi.org/10.1186/
[145] X. Huang, Y. Lu, M. Guo, S. Du, N. Han, Recent strategies for nano-based PTT s13567-019-0688-1.
combined with immunotherapy: from a biomaterial point of view, Theranostics [171] K.E. Kram, et al., Adaptation of Escherichia coli to long-term serial passage in
11 (2021) 7546–7569, https://doi.org/10.7150/thno.56482. complex medium: evidence of parallel evolution, mSystems 2 (2017), https://doi.
[146] Y. Liu, P. Bhattarai, Z. Dai, X. Chen, Photothermal therapy and photoacoustic org/10.1128/mSystems.00192-16.
imaging via nanotheranostics in fighting cancer, Chem. Soc. Rev. 48 (2019) [172] G.G. Sgro, et al., Bacteria-killing type IV secretion systems, Front. Microbiol. 10
2053–2108, https://doi.org/10.1039/c8cs00618k. (2019) 1078, https://doi.org/10.3389/fmicb.2019.01078.
[147] W. Chen, et al., Dual drugs decorated bacteria irradiate deep hypoxic tumor and [173] X. Yi, et al., Bacteria-triggered tumor-specific thrombosis to enable potent
arouse strong immune responses, Biomaterials 286 (2022) 121582, https://doi. photothermal immunotherapy of cancer, Sci. Adv. 6 (2020) eaba3546, https://
org/10.1016/j.biomaterials.2022.121582. doi.org/10.1126/sciadv.aba3546.
[148] W. Lin, et al., Engineered bacteria labeled with iridium(III) photosensitizers for [174] K. Sakamoto, Genetic code expansion: another solution to codon assignments, Int.
enhanced photodynamic immunotherapy of solid tumors, Angew Chem. Int. Ed. J. Mol. Sci. 24 (2022), https://doi.org/10.3390/ijms24010361.
Engl. 62 (2023) e202310158, https://doi.org/10.1002/anie.202310158. [175] J. Kreitz, et al., Programmable protein delivery with a bacterial contractile
[149] B. Xie, et al., Supramolecularly engineered conjugate of bacteria and cell injection system, Nature 616 (2023) 357–364, https://doi.org/10.1038/s41586-
membrane-coated magnetic nanoparticles for enhanced ferroptosis and 023-05870-7.
immunotherapy of tumors, Adv. Sci. (2023) e2304407, https://doi.org/10.1002/
advs.202304407.

15

You might also like