Skip to main content
Engineering Biology logoLink to Engineering Biology
. 2024 May 31;8(2-3):31–40. doi: 10.1049/enb2.12032

Programmable cancer treatments: Engineering biology approaches for living cures

Marc Biarnes Carrera 1, Alexandra Sevko 1, Nicholas Glanville 1, Livija Deban 1,
PMCID: PMC11514499  PMID: 39473536

Abstract

Living cures are cell‐based, programmable therapies that integrate the latest learnings in immunology and synthetic biology. Although Adoptive Cell Therapies (ACTs) have transformed the treatment landscape of haematological malignancies by harnessing the powerful anti‐tumour properties of immune cells, commercialisation and ensuring access is challenging. Their application in solid tumour treatment has been hindered by the immunosuppressive tumour microenvironment (TME) and its associated physical barriers. Conversely, bacterial immunotherapies offer cost‐effective solutions by utilising tumour‐colonising bacteria that trigger localised inflammatory responses within the TME. The authors briefly examine advancements in ACT and propose bacterial immunotherapies as an alternative or complementary treatment modality with potential use either as standalone therapies or in conjunction with other treatments.

Keywords: healthcare, industry, microbial engineering, synthetic biology


Living cures are cell‐based, programmable therapies that integrate the latest learnings in immunology and synthetic biology. Bacterial immunotherapies offer cost‐effective solutions by utilising tumour‐colonising bacteria that trigger localised inflammatory responses within the tumour microenvironment.

graphic file with name ENB2-8-31-g002.jpg

1. INTRODUCTION

Cancer is a devastating disease that primarily manifests as solid tumours, abnormal masses of heterogenous cells that grow uncontrollably and establish their own host‐independent microenvironment. The prevailing standard of care comprises surgical resection, followed by chemotherapy or radiotherapy, both of which are associated with debilitating side effects. Immunotherapy has emerged as an alternative therapeutic approach, focusing on targeted strategies to train the patient's immune system to identify and eradicate malignant cells. This approach has resulted in prolific development of new therapies, such as monoclonal immune checkpoint inhibitor antibodies [1, 2]. A particularly exciting area that combines the latest learnings in immunology and synthetic biology is the use of live cells, either human‐derived or bacterial, as cancer therapeutics. These living cures are programmable through engineering biology techniques to enhance their interaction with the immune system. Adoptive Cell Therapies (ACTs) have leveraged the potent anti‐tumour properties of cytotoxic immune cells to develop highly targeted therapies that revolutionised the treatment landscape of haematological malignancies. However, the use of ACT in the treatment of solid tumours, which represent 90% of all diagnosed cancers [3], has yet to yield positive outcomes, partly due to the immunosuppressive nature of the tumour microenvironment (TME) and its associated physical barriers. Moreover, industrial manufacturing of adoptive therapies remains expensive, restricting access only to a small proportion of the world's population. Bacterial immunotherapies in contrast are cost‐effective treatments that harness the solid tumour‐colonising properties of certain bacteria to induce a local, broad inflammatory immune response that stimulates the recruitment of endogenous cytotoxic immune cells into TME. Whilst the first bacterial immunotherapies are attributed to William B. Coley [4], who in 1891 performed foundational experiments by injecting different mixtures of live‐attenuated Serratia marcescens and Streptococcus pyogenes into patients' tumours, in the past decade, the bacterial immunotherapeutic space has seen a resurgence spurred by advancements in our understanding of tumour immunology and ability to read and manipulate bacterial genetic information. In this review, we discuss recent work and emerging trends in the living cures space for cancer treatment. We briefly summarise advancements and ongoing challenges in both adoptive mammalian cell and bacterial immunotherapies, contrasting the different attributes of these two alternative approaches and exploring the potential transformative impact the combination of adoptive cell and bacterial therapeutic strategies can have to maximise efficacy and patient benefit by blending the malleable and tumour‐colonising properties of bacteria with the powerful responses of adoptive cells.

2. ADOPTIVE CELL THERAPIES

In cancer immunotherapy, ACT predominantly refers to the use of autologous (from the same patient) or allogeneic (from a healthy donor) immune cells to target tumour cells [5, 6]. Whilst the use of various immune cells has been explored (e.g. lymphocytes, dendritic cells, or macrophages), adoptive T cell therapy (Figure 1) currently stands out as the most prevalent therapeutic approach [7]. T cell therapy can be categorised into three main applications: tumour‐infiltrating lymphocytes (TIL), chimaeric‐antigen receptor T cells (CAR‐T), and T cell receptor T cells (TCR‐T).

FIGURE 1.

FIGURE 1

Adoptive cell therapies. Adoptive T cell therapy relies on the targeted recognition of cancer cells by natural or engineered T cell Receptors (TCRs) or engineered Chimeric Antigen Receptors (CARs). They can be classified into three major categories: (1) Tumour‐infiltrating lymphocytes can be isolated from the patient’s tumour and re‐introduced into the patient; (2) or the TCR of tumour‐antigen reactive lymphocytes could be sequenced, its expression optimised, and transduced into T‐cells (TCR‐T) prior to infusion. The last approach (3) involves using synthetic receptors (CARs) that can recognise tumour‐associated or tumour‐specific antigens. This approach has been successful in the treatment of blood cancers and is actively being researched for its use in solid tumours. CAR, Chimeric Antigen Receptors; TCR, T cell Receptors.

The autologous treatment of TIL involves the identification and ex vivo expansion of naturally occurring tumour‐specific cytotoxic T cells, followed by their re‐infusion into the patient. The expansion, mediated by exogenous addition of IL‐2 [8, 9], results in a pool of cells with multiple T cell Receptor (TCR) clones, providing an effective response against cancer heterogeneity. However, natural infiltration of T cells inside solid tumours can be limited in many cancers [10] and recovered T cells are often dysfunctional and exhausted by the immunosuppressive nature of the tumour microenvironment [11]. Moreover, the personalised manufacturing that is required for TIL therapy presents significant logistical hurdles. This, together with the short half‐life of infused TILs [12], has resulted in slow progress of TIL therapies to the market, with the first Food and Drug Administration approval only having recently been granted [13]. Biological engineering‐based approaches have been developed to address such limitations: the TCR and Chimaeric Antigen Receptor (CAR) technologies.

TCR‐T, either autologous or allogeneic, are engineered to express a TCR specific for tumour‐specific antigens (TSA), molecular signatures only found in tumours, or tumour‐associated antigens (TAA), molecular signatures that are over‐represented in tumour tissues [14]. TCRs are heterodimeric proteins, each having a unique variable domain and a constant domain [15]. The unique variable regions can recognise specific peptides presented by mammalian cells, leading to T cell activation. The TCR‐T therapy process [16] involves isolation of lymphocytes from either healthy donors or cancer patients. The antigen‐responsive lymphocytes are isolated upon expansion and their corresponding TCR is identified through sequencing techniques [17, 18]. After selection of the most promising TCRs, the TCR molecules can undergo optimisation [19] and are loaded onto T cells for validation and therapeutic lead selection and development.

CARs are engineered synthetic receptors [5] that function to direct autologous or allogeneic T cells (CAR‐T), or in some instances also Natural Killer cells (NK, [20]) or macrophages [21], to recognise and eliminate cells expressing and displaying a specific antigen, triggering potent anti‐tumour responses. They generally rely on four main components [5, 22]. The extracellular binding domain, built of a single‐chain variable domain derived from a monoclonal antibody, is the element responsible for specific antigen recognition. It is linked to the hinge region, a flexible element that connects the binding domain with the transmembrane domain, a poorly characterised region that anchors the CAR to the T cell. Finally, the intracellular signalling domain [23] provides the activating signal(s) upon ligand engagement. The signalling domain is the most engineered element of the CAR, having undergone up to four different generations of improvement by adding up to two co‐stimulatory elements (e.g. CD28 and/or 4‐1BB) and nuclear factors that induce cytokine production upon stimulation (4th generation, T cell redirected for antigen‐unrestricted cytokine‐initiated killing (TRUCK), [24]). A fifth generation of CAR‐Ts was engineered to contain additional membrane receptors that respond to antigen stimulation [25].

Prior to administering therapeutic T cells, patients must undergo treatment aimed at reducing the number of recipients’ lymphocytes, known as lymphodepletion, which is critical for the ability of donor cells to engraft and expand in the patient. However, it leads to toxicities and immune suppression [26, 27], and there is currently no agreement on the combinations of drug and treatment regimes, with cyclophosphamide and fludarabine being the most commonly used compounds [28, 29]. Additionally, ACTs themselves have been linked to significant toxicities in the clinic. Most common adverse effects are those associated with supraphysiological cytokine production (also known as cytokine‐release syndrome) and immune effector cell‐associated neurotoxicity syndrome [30]. Some of the toxicities are thought to be mediated by IL‐6 and current management involves administering IL‐6 receptor blockade (tocilizumab and corticosteroids) [31, 32]. Further unpredictable toxicities can be caused by on‐target off‐tumour T cell activation, triggered by recognition of TAAs in healthy tissues [5]. Engineering approaches have been proposed for enhancing the safety profile by adding small molecule‐responsive switches [33] or by manipulating the affinity of the CAR‐T cell antigen binding domain, which must remain in a range that allows target binding but prevents CAR‐T disengaging from dying tumour cells, causing trogocytosis (excision of antigen from cancer cell to CAR‐T [34]) or exhausting the T cells.

Cancer is a dynamic disease that evolves as it progresses, contributing to tumour heterogeneity [35]. This leads to post‐treatment relapses caused by antigen escape, where target antigen expression is abolished or reduced to levels below the T cell threshold of activation [5, 36]. This effect has been observed to be more pronounced in CARs, with reports of 30%–70% relapse in patients with acute lymphoblastic leukaemia, where 10%–30% had lost CD19 [37]. A strategy to overcome this limitation has been explored using bi‐specific CARs [38, 39, 40]. A recent example of CD19/CD20 bi‐specific CARs [41] introduced into naive and memory T‐cells [42] resulted in safe and effective responses against non‐Hodgkin lymphoma.

While ACTs have been successfully deployed in the treatment of haematological malignancies, efficacy in the treatment of solid tumours remains elusive [43, 44]. The challenges in developing T cell therapies for solid tumours stem from the immunosuppressive nature of the tumour microenvironment that prevents T cell expansion and persistence as well as tumour‐associated physical barriers, such as cancer‐associated fibroblasts [45]. Ongoing research efforts to overcome these barriers include creating CARs that rely on the co‐administration or production of immunostimulatory cytokines [22, 23] and expression of enzymes that degrade the tumour‐associated extracellular matrix, such as heparinase [46].

Finally, manufacturing challenges remain a barrier to widespread adoption of these therapies. Processes are costly, time‐consuming, laborious, and can affect the functionality of the final therapeutic cellular product [47, 48, 49], with high batch‐to‐batch variability that is unavoidable due to the use of cells from different donors to initiate manufacturing. For autologous treatments in particular, patients generally possess low numbers of lymphocytes due to cancer‐induced immunosuppression and previous treatments (e.g. with chemotherapy) and experience high vein‐to‐vein treatment times between T cell harvest and therapy injection.

3. BACTERIAL IMMUNOTHERAPIES

Bacterial immunotherapies (Figure 2) leverage the latest advancements in synthetic biology to develop cost‐effective approaches that take advantage of the tumour‐homing and immunostimulatory capabilities of some bacterial strains to reprogramme the TME from immunosuppressive ‘cold’ to triggering a ‘hot’ immune response. These therapies build upon the observation that bacteria administered to hosts bearing tumours can accumulate preferentially in the TME in ratios of up to 10,000:1 when compared to healthy tissues such as the liver or spleen [50, 51, 52]. It is thought that such refined targeting mechanisms derive from the leaky vasculature in the TME and the ability of bacteria to exploit the unique features of the tumour, such as low pH [53], enhanced nutrient availability [54], or a hypoxic core [55]. When established in the TME, shedding of microbe‐associated molecular patterns (MAMPs), such as flagella or lipopolysaccharide (LPS) [56, 57, 58], can trigger a broad stimulation of the immune system, resulting in the infiltration of immune effector cells such as NK cells or T cells (e.g. CD8+) [58]. These findings have led to the development of several bacterial strains as cancer therapeutics, including Clostridium [55], Listeria [59], Escherichia coli (e.g. Nissle strain) [60] and Salmonella [61]. Pathogenicity and bacteraemia are prevented by introducing attenuating mutations that limit the bacterial ability to replicate in healthy tissue, restricting growth to the tumour. Selection of appropriate attenuating mutations is critical as over‐attenuated microorganisms will fail to elicit an appropriate immune response and under‐attenuated bacteria can lead to toxicity. Obligate anaerobes, such as Clostridium, which can only survive in the necrotic regions of the tumour, require attenuation by the removal of any potentially produced bacterial toxins, such as the α‐toxin in C. novyi‐NT [62], the most advanced Clostridium tested in humans, or nine gene clusters with high sequence similarity to the Streptolysin S operon from S. pyogenes in Clostridium sporogenes‐NT [63]. Facultative bacteria, such as Escherichia or Salmonella, colonise the tumour through more complex mechanisms and thus require additional attenuating mutations. The Nissle strain, a naturally occurring probiotic isolated in 1917, is the most researched E. coli strain for cancer treatment, including clinically tested strain variants [64]. Salmonella enterica is perhaps the most researched species overall for cancer treatment, with the auxotrophic strains S. Typhimurium VNP20009 (deletion mutant of purI, involved in purine biosynthesis, and msbB, involved in LPS biosynthesis) [51] and S. enterica Typhi ZH9 (deletion mutant of aroC, involved in aromatic acid biosynthesis, and ssaV, involved in bacterial replication) [65] having been safely administered to humans. An alternative approach to ensuring lack of pathogenicity and viable bacterial replication is complete inactive bacteria, known as bacterial ghosts, which have been constructed through the inducible expression of lysin E from φX174 [66], yielding the bacterial cell envelope which retains all the inflammatory elements of the bacterium but no intracellular material (e.g. genomic information). Decoration of the membrane with selected cargo proteins fused to outer membrane proteins prior to bacteriolysis can be used to refine the immunostimulatory response [67]. In some instances, bacterial ghosts have been loaded with chemotherapies (e.g. doxycycline [68]) or plasmid DNA for transfer into macrophages [69].

FIGURE 2.

FIGURE 2

Bacterial immunotherapies. Bacterial therapies are cost‐effective treatments that leverage the tumour‐colonising properties of certain bacterial species, which can result in up to 10,000:1 ratios of enrichment in the malignant tissue. In the tumour, the established microbe can shed a series of immunostimulatory molecules that reprogramme the properties of the tumour microenvironment and promote the infiltration of anti‐tumour immune cells. Ease of engineering and high loading capacity of these bacteria allow the production of synthetic payloads, either on a plasmid or integrated into the chromosome, that can be delivered precisely into the tumour after sensing of tumour‐associated metabolic or physicochemical signatures. Risk of adverse events is minimised by introduction of attenuating mutations or the construction of bacterial ghosts. Bacterial therapies could provide the required immunostimulatory components, either through shedding of inflammatory molecular signatures or recombinant payloads, that promote infiltration and sustain the activity of ACT. ACT, Adoptive Cell Therapies.

The new generation of bacterial immunotherapies builds upon the foundational technology of attenuated bacteria by creating improved synthetic strains with enhanced tumour‐colonising capabilities and safety profile. This can be achieved by controlling the expression of essential genes [70] or payload production [71] through specific promoters activated by the differential attributes of the tumour versus healthy tissue or after bacteria reach a certain population threshold (i.e. quorum sensing, [72]). Such approaches can suffer on‐target off‐tumour misfiring, necessitating the exploration of external triggers, such as the arabinose‐inducible promoter (PBAD, [57, 73]), the lac promoter [74], light‐inducible promoters [75] or heat‐inducible systems [76]. These complex circuit control strategies are in their infancy and have yet to demonstrate utility in the human context but hold great promise for enhanced precision and specificity of bacterial immunotherapies. While individual promoter‐regulated circuits can be affected by leaky expression, combination of one or more gene circuits into Boolean genetic logic gates is poised to overcome such limitations [77]. Another mitigating approach has been to increase the bacterial tumour tropism by overexpressing bacterial motility components [78], encapsulating the microbes in polysaccharide capsules [74], or displaying tumour‐targeting peptides or antibody‐like molecules on the bacterial surface [79, 80, 81, 82].

Such precise bacterial therapeutics can contain engineered payloads to selectively deliver therapeutic molecules directly into the tumour, pairing cargo with target cell biology. The host invasive properties of intracellular bacteria, such as Salmonella or Listeria, have been leveraged to deliver nucleic acids, plasmid DNA or RNA, in a process known as bactofection [83, 84]. Therapeutic DNA is loaded into strains as plasmids that are released into the host cytosol upon bacteriolysis and transported to the cell nucleus. These plasmids, which typically cannot replicate in the host cell, encode TSA [83, 85] or silencing RNA [86, 87, 88, 89] downstream of strong viral promoters. However, the delivery of such molecules can be inefficient, mainly due to the rate‐limiting step of nuclear entry. RNA bactofection overcomes this obstacle [90]. A recent publication by Singer et al. demonstrates that bactofection can effectively deliver not just small silencing or epitope‐encoding RNAs but also large functional sequences, and in this case, an oncolytic virus under bacterial control, showcasing the enormous potential of bactofection as a targeted delivery vehicle for a range of RNA therapeutics [91].

Approaches whereby therapeutic proteins are expressed directly by live‐attenuated bacterial carriers have also garnered increasing attention in the recent years, with most examples using S. enterica and E. coli Nissle as bacterial chassis. Salmonella has been highlighted [54, 73] as a unique chassis due to its tendency to accumulate in tumours, its programmability, and its cell invasive nature. The initial steps of the Salmonella invasion cycle rely on the use of a needle‐like structure known as SPI‐1 [92], a type III secretion system that injects effector proteins into the host cell and can be repurposed for the delivery of heterologous payloads [93, 94, 95, 96, 97]. Internalised Salmonella establishes inside a Salmonella‐containing vacuole (SCV) that protects itself from intracellular defence mechanisms [98, 99] and enables replication [100]. Induction of bacterial lysis after Salmonella's establishment in the SCV has been shown to allow payload delivery in murine breast cancer models [73]. Escape from the vacuole can be forced by expression of Listeriolysin O [101], a pore‐forming cytolysin from Listeria monocytogenes, or by using the sifA mutant strain [102], which causes disruption of the vacuole during its maturation. Examples of intracellular payloads which have been explored include intrabodies [103] and bacterial toxins, including chimeric anticancer toxins [104]. Non‐invasive strains have also been engineered to inject protein payloads into eukaryotic cells through heterologous type III secretion systems [105] or, more recently, contractile injection systems [106] and can enable payload deployment throughout the TME by fusing targets with secretion signal peptides [107] or through synchronous release triggered by quorum sensing [60, 108, 109, 110]. Examples of extracellular payloads include cytokines [107, 110, 111, 112, 113], antibody‐like molecules [108, 114] and immunostimulatory proteins (e.g. FlaB [57]).

Engineered bacteria overcome the manufacturing and logistical drawbacks of engineered mammalian cell therapies, offering a relatively straightforward fermentation manufacturing process [115, 116] with the potential for lyophilisation to produce off‐the‐shelf therapies stable at standard refrigerator (2°C–8°C) or even room temperatures [117], as is well established for probiotic and bacterial vaccine products, or to produce bespoke therapies in as little as 3–4 days compared to the 3–5 weeks required for autologous ACT [118]. However, most examples of bacterial therapies in the literature do rely on the use of bacterial plasmids to incorporate engineered functions, which can lead to sub‐optimal final product performance derived from inherent cell‐to‐cell variability [119] and increased metabolic burden [120, 121, 122]. Moreover, a large portion of these plasmids rely on the use of antibiotic resistance cassettes [123] or addiction strategies [124, 125, 126] that can be incompatible with use in the clinical setting. Integration of synthetic cassettes into the chromosome is a strategy deployed to circumvent these limitations, although insulation from the surrounding genomic context and minimisation of polar effects are critical to achieve robust strain performance [127]; and positional effects (i.e. gene location in the genome) can impact the total payload yield [127, 128, 129, 130].

4. FUTURE PROSPECTS

Both ACTs and bacterial immunotherapies have harnessed recent advancements in immunology and engineering biology to bring new treatment opportunities to cancer patients. Adoptive cell therapies have achieved success in the treatment of haematological malignancies but to date yielded disappointing results in solid tumours which represent the vast majority of cancers. Bacterial immunotherapies are ideally positioned to address this area of unmet need due to their immense engineering potential and intrinsic tumour targeting properties, offering alternative approaches.

The recent advances in engineering biology capabilities open up opportunities not just for new monotherapies but also for the integration of engineered bacterial therapies with current standards of care and other classes of immunotherapies in development. Such bacteria‐coupled regimes could, for example, restore immune fitness in immune‐compromised patients ahead of other treatments, potentially addressing the major issue of insufficient responders or refractory patients common to many therapies. Bacteria‐dependent immune reprogramming could also directly synergise with therapies like ACT in various ways. For example, colonisation of live‐attenuated bacteria in tumours before adoptive cell infusion could induce profound changes in the TME immune landscape due to exposure and shedding of MAMPs, or through expression of engineered cytokines and chemokines, to promote and sustain engineered immune cell infiltration and activity. Additional bacterial payloads could also degrade tumour‐associated physical barriers to improve immune cell penetration. Safety profiles of ACT could be enhanced by coupling their activity to bacterial therapies that either produce an activating payload, ensuring synthetic immune cells are only triggered in appropriate tissues and thus reducing on‐target off‐tumour effects or that produce a safety switch to prevent the development of serious adverse effects. Ease of engineering and high circuit loading capacity of bacterial strains can also provide opportunities for the introduction of both such circuits on a single strain, providing a highly dynamic control of living cures therapies. Finally, the ability of bacterial therapies to colonise tumours independently of the need for specific targeting molecules can be leveraged to develop ACT treatment regimens that respond to the bacteria instead of the tumour. These beacon approaches, whose initial activity is independent from specific TAA or TSA antigens, have the potential to overcome the tumour heterogeneity hurdle and offer the opportunity to develop off‐the‐shelf bacterial and adoptive cell combination therapies that can be manufactured and distributed ahead of time, thereby reducing overall treatment costs and timelines.

Bacterial cancer therapies have thus far achieved clinical translation only in the form of Bacillus Calmette‐Guérin treatment for bladder cancer, but other live attenuated strains such as Prokarium's Salmonella Typhi ZH9 (NCT06181266) or T3 Pharmaceuticals' Yersinia enterocolitica (NCT05120596) in clinical development promise to establish regulatory pathways that will facilitate adoption of engineered bacterial therapies in the future. Due to the simple, scalable and inexpensive fermentation manufacturing processes of bacterial therapies, this transformative approach promises to democratise access to life‐altering treatments to patients worldwide, making significant strides towards equitable healthcare access on a global scale.

AUTHOR CONTRIBUTIONS

Marc Biarnes Carrera: Writing – original draft. Alexandra Sevko: Writing – review & editing. Nicholas Glanville: Writing – review & editing. Livija Deban: Writing – review & editing.

CONFLICT OF INTEREST STATEMENT

At the time of submission, all authors were full time employees of Prokarium Ltd and received funding from Prokarium Ltd in the form of salary. This does not alter adherence to journal policies.

Biarnes Carrera, B. , et al.: Programmable cancer treatments: engineering biology approaches for living cures. Eng. Biol. 8(2-3), 31–40 (2024). 10.1049/enb2.12032

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article as no new data were created or analysed in this study.

REFERENCES

  • 1. Han, Y. , Liu, D. , Li, L. : PD‐1/PD‐L1 pathway: current researches in cancer. Am. J. Cancer Res. 10(3), 727–742 (2020) [PMC free article] [PubMed] [Google Scholar]
  • 2. Lisi, L. , et al.: Clinical experience with CTLA‐4 blockade for cancer immunotherapy: from the monospecific monoclonal antibody ipilimumab to probodies and bispecific molecules targeting the tumor microenvironment. Pharmacol. Res. 175, 105997 (2022). 10.1016/j.phrs.2021.105997 [DOI] [PubMed] [Google Scholar]
  • 3. Sung, H. , et al.: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 71(3), 209–249 (2021). 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
  • 4. Dobosz, P. , Dzieciątkowski, T. : The intriguing history of cancer immunotherapy. Front. Immunol. 10 (2019). 10.3389/fimmu.2019.02965 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Sterner, R.C. , Sterner, R.M. : CAR‐T cell therapy: current limitations and potential strategies. Blood Cancer J. 11(4), 69 (2021). 10.1038/s41408-021-00459-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Kawamoto, H. , Masuda, K. : Trends in cell medicine: from autologous cells to allogeneic universal‐use cells for adoptive T‐cell therapies. Int. Immunol. 36(2), 65–73 (2024). 10.1093/intimm/dxad051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Ascierto, P.A. , et al.: The “great debate” at immunotherapy bridge 2022, Naples, November 30th–December 1st, 2022. J. Transl. Med. 21(1), 275 (2023). 10.1186/s12967-023-04117-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Rosenberg, S.A. , et al.: Use of tumor‐infiltrating lymphocytes and interleukin‐2 in the immunotherapy of patients with metastatic melanoma. N. Engl. J. Med. 319(25), 1676–1680 (1988). 10.1056/nejm198812223192527 [DOI] [PubMed] [Google Scholar]
  • 9. Kongkaew, T. , et al.: TIL expansion with high dose IL‐2 or low dose IL‐2 with anti‐CD3/anti‐CD28 stimulation provides different quality of TIL‐expanded T cell clones. J. Immunol. Methods 503, 113229 (2022). 10.1016/j.jim.2022.113229 [DOI] [PubMed] [Google Scholar]
  • 10. Manfredi, F. , et al.: TCR redirected T cells for cancer treatment: achievements, hurdles, and goals. Front. Immunol. 11 (2020). 10.3389/fimmu.2020.01689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Quail, D.F. , Joyce, J.A. : Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19(11), 1423–1437 (2013). 10.1038/nm.3394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Hsu, E.J. , et al.: A cytokine receptor‐masked IL2 prodrug selectively activates tumor‐infiltrating lymphocytes for potent antitumor therapy. Nat. Commun. 12(1), 2768 (2021). 10.1038/s41467-021-22980-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. FDA . FDA grants accelerated approval to lifileucel for unresectable or metastatic melanoma. [cited 2024 12th March 2024]. https://www.fda.gov/drugs/resources‐information‐approved‐drugs/fda‐grants‐accelerated‐approval‐lifileucel‐unresectable‐or‐metastatic‐melanoma (2024)
  • 14. Haen, S.P. , et al.: Towards new horizons: characterization, classification and implications of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 17(10), 595–610 (2020). 10.1038/s41571-020-0387-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Shafer, P. , Kelly, L.M. , Hoyos, V. : Cancer therapy with TCR‐engineered T cells: current strategies, challenges, and prospects. Front. Immunol. 13 (2022). 10.3389/fimmu.2022.835762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Baulu, E. , et al.: TCR‐engineered T cell therapy in solid tumors: state of the art and perspectives. Sci. Adv. 9(7) (2023). 10.1126/sciadv.adf3700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Augusto, D. , et al.: Dissecting efficiency of a 5’ rapid amplification of cDNA ends (5’‐RACE) approach for profiling T‐cell receptor beta repertoire. PLoS One 15(7), e0236366 (2020). 10.1371/journal.pone.0236366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Pai, J.A. , Satpathy, A.T. : High‐throughput and single‐cell T cell receptor sequencing technologies. Nat. Methods 18(8), 881–892 (2021). 10.1038/s41592-021-01201-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Froning, K. , et al.: Computational stabilization of T cell receptors allows pairing with antibodies to form bispecifics. Nat. Commun. 11(1), 2330 (2020). 10.1038/s41467-020-16231-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Wang, W. , et al.: Breakthrough of solid tumor treatment: CAR‐NK immunotherapy. Cell Death Discov. 10(1), 40 (2024). 10.1038/s41420-024-01815-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Lei, A. , et al.: A second‐generation M1‐polarized CAR macrophage with antitumor efficacy. Nat. Immunol. 25(1), 102–116 (2023). 10.1038/s41590-023-01687-8 [DOI] [PubMed] [Google Scholar]
  • 22. Zhang, C. , et al.: Engineering CAR‐T cells. Biomark. Res. 5(1), 22 (2017). 10.1186/s40364-017-0102-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Jayaraman, J. , et al.: CAR‐T design: elements and their synergistic function. EBioMedicine 58, 102931 (2020). 10.1016/j.ebiom.2020.102931 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Chmielewski, M. , Abken, H. : TRUCKs: the fourth generation of CARs. Expet Opin. Biol. Ther. 15(8), 1145–1154 (2015). 10.1517/14712598.2015.1046430 [DOI] [PubMed] [Google Scholar]
  • 25. Kagoya, Y. , et al.: A novel chimeric antigen receptor containing a JAK–STAT signaling domain mediates superior antitumor effects. Nat. Med. 24(3), 352–359 (2018). 10.1038/nm.4478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Muranski, P. , et al.: Increased intensity lymphodepletion and adoptive immunotherapy—how far can we go? Nat. Clin. Pract. Oncol. 3(12), 668–681 (2006). 10.1038/ncponc0666 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Nissani, A. , et al.: Comparison of non‐myeloablative lymphodepleting preconditioning regimens in patients undergoing adoptive T cell therapy. J. Immunother. Cancer 9(5), e001743 (2021). 10.1136/jitc-2020-001743 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Lickefett, B. , et al.: Lymphodepletion – an essential but undervalued part of the chimeric antigen receptor T‐cell therapy cycle. Front. Immunol. 14, 1303935 (2023). 10.3389/fimmu.2023.1303935 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. de Alencar Paton, E.J. , Duarte Barroso, F. : Lymphodepletion in cell therapy. J. Bone Marrow Transplant. Cell. Therapy 3(2), 170 (2022). 10.46765/2675-374x.2022v3n2p170 [DOI] [Google Scholar]
  • 30. Xiao, X. , et al.: Mechanisms of cytokine release syndrome and neurotoxicity of CAR T‐cell therapy and associated prevention and management strategies. J. Exp. Clin. Cancer Res. 40(1), 367 (2021). 10.1186/s13046-021-02148-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Le, R.Q. , et al.: FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell‐induced severe or life‐threatening cytokine release syndrome. Oncol. 23(8), 943–947 (2018). 10.1634/theoncologist.2018-0028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Luan, C. , et al.: Improved efficacy with early tocilizumab in the prophylaxis and treatment of cytokine release syndrome of chimeric antigen receptor T cell (CAR‐T) therapy for diffuse large B‐cell lymphoma than acute lymphoblastic leukemia. Biomed. Technol. 3, 59–65 (2023). 10.1016/j.bmt.2023.01.002 [DOI] [Google Scholar]
  • 33. Giordano‐Attianese, G. , et al.: A computationally designed chimeric antigen receptor provides a small‐molecule safety switch for T‐cell therapy. Nat. Biotechnol. 38(4), 426–432 (2020). 10.1038/s41587-019-0403-9 [DOI] [PubMed] [Google Scholar]
  • 34. Hamieh, M. , et al.: CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature 568(7750), 112–116 (2019). 10.1038/s41586-019-1054-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. El‐Sayes, N. , Vito, A. , Mossman, K. : Tumor heterogeneity: a great barrier in the age of cancer immunotherapy. Cancers 13(4), 806 (2021). 10.3390/cancers13040806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Majzner, R.G. , Mackall, C.L. : Tumor antigen escape from CAR T‐cell therapy. Cancer Discov. 8(10), 1219–1226 (2018). 10.1158/2159-8290.cd-18-0442 [DOI] [PubMed] [Google Scholar]
  • 37. Aparicio‐Pérez, C. , et al.: Failure of ALL recognition by CAR T cells: a review of CD 19‐negative relapses after anti‐CD 19 CAR‐T treatment in B‐ALL. Front. Immunol. 14 (2023). 10.3389/fimmu.2023.1165870 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Dai, H. , et al.: Bispecific CAR‐T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J. Hematol. Oncol. 13(1), 30 (2020). 10.1186/s13045-020-00856-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Pan, J. , et al.: Sequential CD19‐22 CAR T therapy induces sustained remission in children with r/r B‐ALL. Blood 135(5), 387–391 (2020). 10.1182/blood.2019003293 [DOI] [PubMed] [Google Scholar]
  • 40. Liu, S. , et al.: Combination of CD19 and CD22 CAR‐T cell therapy in relapsed B‐cell acute lymphoblastic leukemia after allogeneic transplantation. Am. J. Hematol. 96(6), 671–679 (2021). 10.1002/ajh.26160 [DOI] [PubMed] [Google Scholar]
  • 41. Larson, S.M. , et al.: CD19/CD20 bispecific chimeric antigen receptor (CAR) in naive/memory T cells for the treatment of relapsed or refractory non‐hodgkin lymphoma. Cancer Discov. 13(3), 580–597 (2023). 10.1158/2159-8290.cd-22-0964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Zah, E. , et al.: T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol. Res. 4(6), 498–508 (2016). 10.1158/2326-6066.cir-15-0231 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Morotti, M. , et al.: Promises and challenges of adoptive T‐cell therapies for solid tumours. Br. J. Cancer 124(11), 1759–1776 (2021). 10.1038/s41416-021-01353-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Albelda, S.M. : CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat. Rev. Clin. Oncol. 21(1), 47–66 (2023). 10.1038/s41571-023-00832-4 [DOI] [PubMed] [Google Scholar]
  • 45. Alkasalias, T. , et al.: Fibroblasts in the tumor microenvironment: shield or spear? Int. J. Mol. Sci. 19(5), 1532 (2018). 10.3390/ijms19051532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Caruana, I. , et al.: Heparanase promotes tumor infiltration and antitumor activity of CAR‐redirected T lymphocytes. Nat. Med. 21(5), 524–529 (2015). 10.1038/nm.3833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Blach, U. , et al.: Potential solutions for manufacture of CAR T cells in cancer immunotherapy. Nat. Commun. 13(1), 5225 (2022). 10.1038/s41467-022-32866-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Watanabe, N. , Mo, F. , McKenna, M.K. : Impact of manufacturing procedures on CAR T cell functionality. Front. Immunol. 13, 876339 (2022). 10.3389/fimmu.2022.876339 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Jagannath, S. , et al.: Component costs of CAR‐T therapy in addition to treatment acquisition costs in patients with multiple myeloma. Oncology and Therapy 11(2), 263–275 (2023). 10.1007/s40487-023-00228-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Pawelek, J.M. , Low, K.B. , Bermudes, D. : Tumor‐targeted Salmonella as a novel anticancer vector. Cancer Res. 57(20), 4537–4544 (1997) [PubMed] [Google Scholar]
  • 51. Clairmont, C. , et al.: Biodistribution and genetic stability of the novel antitumor agent VNP20009, a genetically modified strain of Salmonella typhimurium. J. Infect. Dis. 181(6), 1996–2002 (2000). 10.1086/315497 [DOI] [PubMed] [Google Scholar]
  • 52. Forbes, N.S. , et al.: Sparse initial entrapment of systemically injected Salmonella typhimurium leads to heterogeneous accumulation within tumors. Cancer Res. 63(17), 5188–5193 (2003) [PubMed] [Google Scholar]
  • 53. Zhang, X. , Lin, Y. , Gillies, R.J. : Tumor pH and its measurement. J. Nucl. Med. 51(8), 1167–1170 (2010). 10.2967/jnumed.109.068981 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Aganja, R.P. , et al.: Salmonella as a promising curative tool against cancer. Pharmaceutics 14(10), 2100 (2022). 10.3390/pharmaceutics14102100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Heap, J.T. , et al.: Spores of Clostridium engineered for clinical efficacy and safety cause regression and cure of tumors in vivo. Oncotarget 5(7), 1761–1769 (2014). 10.18632/oncotarget.1761 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Kocijancic, D. , et al.: Therapeutic benefit of Salmonella attributed to LPS and TNF‐α is exhaustible and dictated by tumor susceptibility. Oncotarget 8(22), 36492–36508 (2017). 10.18632/oncotarget.16906 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Zheng, J.H. , et al.: Two‐step enhanced cancer immunotherapy with engineered Salmonella typhimurium secreting heterologous flagellin. Sci. Transl. Med. 9(376) (2017). 10.1126/scitranslmed.aak9537 [DOI] [PubMed] [Google Scholar]
  • 58. Chen, J. , et al.: Salmonella flagella confer anti‐tumor immunological effect via activating Flagellin/TLR5 signalling within tumor microenvironment. Acta Pharm. Sin. B 11(10), 3165–3177 (2021). 10.1016/j.apsb.2021.04.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Flickinger, J. , Rodeck, U. , Snook, A. : Listeria monocytogenes as a vector for cancer immunotherapy: current understanding and progress. Vaccines 6(3), 48 (2018). 10.3390/vaccines6030048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Gurbatri, C.R. , et al.: Engineering tumor‐colonizing E. coli Nissle 1917 for detection and treatment of colorectal neoplasia. Nat. Commun. 15(1), 646 (2024). 10.1038/s41467-024-44776-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Glanville, N. , et al.: 878 Novel microbial immunotherapy approach for the treatment of bladder cancer. J. Immuno. Therapy Cancer 9(Suppl 2), A920 (2021) [Google Scholar]
  • 62. Dang, L.H. , et al.: Combination bacteriolytic therapy for the treatment of experimental tumors. In: Proceedings of the National Academy of Sciences 98(26), 15155–15160 (2001). 10.1073/pnas.251543698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Kubiak, A.M. , et al.: Efficient secretion of murine IL‐2 from an attenuated strain of Clostridium sporogenes, a novel delivery vehicle for cancer immunotherapy. Front. Microbiol. 12 (2021). 10.3389/fmicb.2021.669488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Luke, J.J. , et al.: Phase I study of SYNB1891, an engineered E. coli Nissle strain expressing STING agonist, with and without atezolizumab in advanced malignancies. Clin. Cancer Res. 29(13), 2435–2444 (2023). 10.1158/1078-0432.ccr-23-0118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Miranda, J.J. , et al.: A randomised trial evaluating the safety and immunogenicity of the novel single oral dose typhoid vaccine M01ZH09 in healthy Vietnamese children. PLoS One 5(7), e11778 (2010). 10.1371/journal.pone.0011778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Won, G. , Hajam, I.A. , Lee, J.H. : Improved lysis efficiency and immunogenicity of Salmonella ghosts mediated by co‐expression of λ phage holin‐endolysin and ɸX174 gene E. Sci. Rep. 7(1), 45139 (2017). 10.1038/srep45139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Lubitz, W. , et al.: Extended recombinant bacterial ghost system. J. Biotechnol. 73(2‐3), 261–273 (1999). 10.1016/s0168-1656(99)00144-3 [DOI] [PubMed] [Google Scholar]
  • 68. Rabea, S. , et al.: Salmonella‐innovative targeting carrier: loading with doxorubicin for cancer treatment. Saudi Pharmaceut. J. 28(10), 1253–1262 (2020). 10.1016/j.jsps.2020.08.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Paukner, S. , et al.: DNA‐loaded bacterial ghosts efficiently mediate reporter gene transfer and expression in macrophages. Mol. Ther. 11(2), 215–223 (2005). 10.1016/j.ymthe.2004.09.024 [DOI] [PubMed] [Google Scholar]
  • 70. Yu, B. , et al.: Explicit hypoxia targeting with tumor suppression by creating an “obligate” anaerobic Salmonella Typhimurium strain. Sci. Rep. 2(1), 436 (2012). 10.1038/srep00436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Mengesha, A. , et al.: Development of a flexible and potent hypoxia‐inducible promoter for tumor‐targeted gene expression in attenuated salmonella. Cancer Biol. Ther. 5(9), 1120–1128 (2014). 10.4161/cbt.5.9.2951 [DOI] [PubMed] [Google Scholar]
  • 72. Din, M.O. , et al.: Synchronized cycles of bacterial lysis for in vivo delivery. Nature 536(7614), 81–85 (2016). 10.1038/nature18930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Raman, V. , et al.: Intracellular delivery of protein drugs with an autonomously lysing bacterial system reduces tumor growth and metastases. Nat. Commun. 12(1), 6116 (2021). 10.1038/s41467-021-26367-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Harimoto, T. , et al.: A programmable encapsulation system improves delivery of therapeutic bacteria in mice. Nat. Biotechnol. 40(8), 1259–1269 (2022). 10.1038/s41587-022-01244-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Zhang, Y. , et al.: Upconversion optogenetic engineered bacteria system for time‐resolved imaging diagnosis and light‐controlled cancer therapy. ACS Appl. Mater. Interfaces 14(41), 46351–46361 (2022). 10.1021/acsami.2c14633 [DOI] [PubMed] [Google Scholar]
  • 76. Li, L. , et al.: Precise thermal regulation of engineered bacteria secretion for breast cancer treatment in vivo. ACS Synth. Biol. 11(3), 1167–1177 (2022). 10.1021/acssynbio.1c00452 [DOI] [PubMed] [Google Scholar]
  • 77. Nguyen, D.‐H. , et al.: Bioengineering of bacteria for cancer immunotherapy. Nat. Commun. 14(1), 3553 (2023). 10.1038/s41467-023-39224-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Raman, V. , et al.: The motility regulator flhDC drives intracellular accumulation and tumor colonization of Salmonella. Journal for ImmunoTherapy of Cancer 7(1), 44 (2019). 10.1186/s40425-018-0490-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Bereta, M. , et al.: Improving tumor targeting and therapeutic potential of Salmonella VNP20009 by displaying cell surface CEA‐specific antibodies. Vaccine 25(21), 4183–4192 (2007). 10.1016/j.vaccine.2007.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Massa, P.E. , et al.: Salmonella engineered to express CD20‐targeting antibodies and a drug‐converting enzyme can eradicate human lymphomas. Blood 122(5), 705–714 (2013). 10.1182/blood-2012-12-474098 [DOI] [PubMed] [Google Scholar]
  • 81. Park, S.‐H. , et al.: RGD peptide cell‐surface display enhances the targeting and therapeutic efficacy of attenuated salmonella‐mediated cancer therapy. Theranostics 6(10), 1672–1682 (2016). 10.7150/thno.16135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Liang, K. , et al.: Attenuated Salmonella Typhimurium with truncated LPS and outer membrane‐displayed RGD peptide for cancer therapy. Biomed. Pharmacother., 155 (2022) [DOI] [PubMed] [Google Scholar]
  • 83. Lee, C.‐H. , Wu, C.‐L. , Shiau, A.‐L. : Systemic administration of attenuated Salmonella choleraesuis carrying thrombospondin‐1 gene leads to tumor‐specific transgene expression, delayed tumor growth and prolonged survival in the murine melanoma model. Cancer Gene Ther. 12(2), 175–184 (2004). 10.1038/sj.cgt.7700777 [DOI] [PubMed] [Google Scholar]
  • 84. Jawalagatti, V. , et al.: Highly feasible immunoprotective multicistronic SARS‐CoV‐2 vaccine candidate blending novel eukaryotic expression and Salmonella bactofection. J. Adv. Res. 36, 211–222 (2022). 10.1016/j.jare.2021.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Stegantseva, M.V. , et al.: Multi‐antigen DNA vaccine delivered by polyethylenimine and Salmonella enterica in neuroblastoma mouse model. Cancer Immunol. Immunother. 69(12), 2613–2622 (2020). 10.1007/s00262-020-02652-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Blache, C.A. , et al.: Systemic delivery ofSalmonella typhimuriumTransformed with IDO shRNA enhances intratumoral vector colonization and suppresses tumor growth. Cancer Res. 72(24), 6447–6456 (2012). 10.1158/0008-5472.can-12-0193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Jiang, Z. , et al.: Using attenuated salmonella typhi as tumor targeting vector for MDR1 siRNA delivery: an experimental study. Cancer Biol. Ther. 6(4), 555–560 (2014). 10.4161/cbt.6.4.3850 [DOI] [PubMed] [Google Scholar]
  • 88. Yang, N. , et al.: Oral administration of attenuatedS. typhimuriumcarrying shRNA‐expressing vectors as a cancer therapeutic. Cancer Biol. Ther. 7(1), 145–151 (2014). 10.4161/cbt.7.1.5195 [DOI] [PubMed] [Google Scholar]
  • 89. Zhao, C. , et al.: Enhanced therapeutic effect of an antiangiogenesis peptide on lung cancer in vivo combined with salmonella VNP20009 carrying a Sox2 shRNA construct. J. Exp. Clin. Cancer Res. 35(1), 107 (2016). 10.1186/s13046-016-0381-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Senevirathne, A. , et al.: Eukaryotic expression system complemented with expressivity of Semliki Forest Virus's RdRp and invasiveness of engineered Salmonella demonstrate promising potential for bacteria mediated gene therapy. Biomaterials, 279 (2021) [DOI] [PubMed] [Google Scholar]
  • 91. Singer, Z.S. , et al.: Engineered bacteria launch and control an oncolytic virus. bioRxiv (2023) [Google Scholar]
  • 92. Lou, L. , et al.: Salmonella pathogenicity island 1 (SPI‐1) and its complex regulatory network. Front. Cell. Infect. Microbiol. 9 (2019). 10.3389/fcimb.2019.00270 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Russmann, H. , et al.: Protection against murine listeriosis by oral vaccination with recombinant Salmonella expressing hybrid Yersinia type III proteins. J. Immunol. 167(1), 357–365 (2001). 10.4049/jimmunol.167.1.357 [DOI] [PubMed] [Google Scholar]
  • 94. Igwe, E.I. , Geginat, G. , Russmann, H. : Concomitant cytosolic delivery of two immunodominant listerial antigens bySalmonella entericaSerovar typhimurium confers superior protection against murine listeriosis. Infect. Immun. 70(12), 7114–7119 (2002). 10.1128/iai.70.12.7114-7119.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Nishikawa, H. : In vivo antigen delivery by aSalmonella typhimurium type III secretion system for therapeutic cancer vaccines. J. Clin. Invest. 116(7), 1946–1954 (2006). 10.1172/jci28045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Berthoin, L. , et al.: Targeted release of transcription factors for cell reprogramming by a natural micro‐syringe. Int. J. Pharm. 513(1‐2), 678–687 (2016). 10.1016/j.ijpharm.2016.09.081 [DOI] [PubMed] [Google Scholar]
  • 97. Chabloz, A. , et al.: Salmonella‐based platform for efficient delivery of functional binding proteins to the cytosol. Commun. Biol. 3(1), 342 (2020). 10.1038/s42003-020-1072-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Eswarappa, S.M. , et al.: Division of the salmonella‐containing vacuole and depletion of acidic lysosomes in salmonella‐infected host cells are novel strategies of Salmonella enterica to avoid lysosomes. Infect. Immun. 78(1), 68–79 (2010). 10.1128/iai.00668-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Schulte, M. , Olschewski, K. , Hensel, M. : The protected physiological state of intracellular Salmonella enterica persisters reduces host cell‐imposed stress. Commun. Biol. 4(1), 520 (2021). 10.1038/s42003-021-02049-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Figueira, R. , Holden, D.W. : Functions of the Salmonella pathogenicity island 2 (SPI‐2) type III secretion system effectors. Microbiology 158(5), 1147–1161 (2012). 10.1099/mic.0.058115-0 [DOI] [PubMed] [Google Scholar]
  • 101. Gentschev, I. , et al.: Salmonella strain secreting active listeriolysin changes its intracellular localization. Infect. Immun. 63(10), 4202–4205 (1995). 10.1128/iai.63.10.4202-4205.1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Beuzon, C.R. : Salmonella maintains the integrity of its intracellular vacuole through the action of SifA. EMBO J. 19(13), 3235–3249 (2000). 10.1093/emboj/19.13.3235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Fugmann, S.D. , et al.: Direct injection of functional single‐domain antibodies from E. coli into human cells. PLoS One 5(12), e15227 (2010). 10.1371/journal.pone.0015227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Khoshnood, S. , et al.: Bacteria‐derived chimeric toxins as potential anticancer agents. Front. Oncol. 12 (2022). 10.3389/fonc.2022.953678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Reeves, A.Z. , et al.: Engineering Escherichia coli into a protein delivery system for mammalian cells. ACS Synth. Biol. 4(5), 644–654 (2015). 10.1021/acssynbio.5b00002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Kreitz, J. , et al.: Programmable protein delivery with a bacterial contractile injection system. Nature 616(7956), 357–364 (2023). 10.1038/s41586-023-05870-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Tumas, S. , et al.: Engineered E. coli Nissle 1917 for delivery of bioactive IL‐2 for cancer immunotherapy. Sci. Rep. 13(1), 12506 (2023). 10.1038/s41598-023-39365-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Gurbatri, C.R. , et al.: Engineered probiotics for local tumor delivery of checkpoint blockade nanobodies. Sci. Transl. Med. 12(530) (2020). 10.1126/scitranslmed.aax0876 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Deb, D. , et al.: Design of combination therapy for engineered bacterial therapeutics in non‐small cell lung cancer. Sci. Rep. 12(1), 21551 (2022). 10.1038/s41598-022-26105-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Savage, T.M. , et al.: Chemokines expressed by engineered bacteria recruit and orchestrate antitumor immunity. Sci. Adv. 9(10) (2023). 10.1126/sciadv.adc9436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Loeffler, M. , et al.: Salmonella typhimurium engineered to produce CCL21 inhibit tumor growth. Cancer Immunol. Immunother. 58(5), 769–775 (2008). 10.1007/s00262-008-0555-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Saltzman, D.A. , et al.: AttenuatedSalmonella typhimuriumContaining interleukin‐2 decreases MC‐38 hepatic metastases: a novel anti‐tumor agent. Cancer Biother. Rad. 11(2), 145–153 (1996). 10.1089/cbr.1996.11.145 [DOI] [PubMed] [Google Scholar]
  • 113. Loeffler, M. , et al.: IL‐18‐producing Salmonella inhibit tumor growth. Cancer Gene Ther. 15(12), 787–794 (2008). 10.1038/cgt.2008.48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Ruano‐Gallego, D. , et al.: Screening and purification of nanobodies from E. coli culture supernatants using the hemolysin secretion system. Microb. Cell Factories 18(1), 47 (2019). 10.1186/s12934-019-1094-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. FDA . Vivotif Product Information (2018). [cited 2024 25th April 2024]. https://www.fda.gov/vaccines‐blood‐biologics/vaccines/vivotif
  • 116. FDA . TICE BCG Product Information (2018). [cited 2024 25th April 2024]. https://www.fda.gov/vaccines‐blood‐biologics/vaccines/tice‐bcg
  • 117. Langemann, T. , et al.: The bacterial ghost platform system. Bioengineered Bugs 1(5), 326–336 (2014). 10.4161/bbug.1.5.12540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Geethakumari, P.R. , et al.: Balancing quality, cost, and access during delivery of newer cellular and immunotherapy treatments. Curr. Hematol. Malignancy Rep. 16(4), 345–356 (2021). 10.1007/s11899-021-00635-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Shao, B. , et al.: Single‐cell measurement of plasmid copy number and promoter activity. Nat. Commun. 12(1), 1475 (2021). 10.1038/s41467-021-21734-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Gyorgy, A. , et al.: Isocost lines describe the cellular economy of genetic circuits. Biophys. J. 109(3), 639–646 (2015). 10.1016/j.bpj.2015.06.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Ceroni, F. , et al.: Burden‐driven feedback control of gene expression. Nat. Methods 15(5), 387–393 (2018). 10.1038/nmeth.4635 [DOI] [PubMed] [Google Scholar]
  • 122. Liu, Q. , et al.: Orthogonality and burdens of heterologous and gate gene circuits in E. coli. ACS Synth. Biol. 7(2), 553–564 (2018). 10.1021/acssynbio.7b00328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Isabella, V.M. , et al.: Development of a synthetic live bacterial therapeutic for the human metabolic disease phenylketonuria. Nat. Biotechnol. 36(9), 857–864 (2018). 10.1038/nbt.4222 [DOI] [PubMed] [Google Scholar]
  • 124. Galán, J.E. , Nakayama, K. , Curtiss, R. : Cloning and characterization of the asd gene of Salmonella typhimurium: use in stable maintenance of recombinant plasmids in Salmonella vaccine strains. Gene 94(1), 29–35 (1990). 10.1016/0378-1119(90)90464-3 [DOI] [PubMed] [Google Scholar]
  • 125. Fedorec, A.J.H. , et al.: Two new plasmid post‐segregational killing mechanisms for the implementation of synthetic gene networks in Escherichia coli. iScience 14, 323–334 (2019). 10.1016/j.isci.2019.03.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Gruzdev, N. , et al.: Development of toxin‐antitoxin self‐destructive bacteria, aimed for salmonella vaccination. Vaccine 41(33), 4918–4925 (2023). 10.1016/j.vaccine.2023.06.074 [DOI] [PubMed] [Google Scholar]
  • 127. Park, Y. , et al.: Precision design of stable genetic circuits carried in highly‐insulated E. coli genomic landing pads. Mol. Syst. Biol. 16(8) (2020). 10.15252/msb.20209584 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Englaender, J.A. , et al.: Effect of genomic integration location on heterologous protein expression and metabolic engineering in E. coli. ACS Synth. Biol. 6(4), 710–720 (2017). 10.1021/acssynbio.6b00350 [DOI] [PubMed] [Google Scholar]
  • 129. Garmendia, E. , Brandis, G. , Hughes, D. : Transcriptional regulation buffers gene dosage effects on a highly expressed operon in Salmonella. mBio 9(5) (2018). 10.1128/mbio.01446-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Lato, D.F. , Golding, G.B. : Spatial patterns of gene expression in bacterial genomes. J. Mol. Evol. 88(6), 510–520 (2020). 10.1007/s00239-020-09951-3 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analysed in this study.


Articles from Engineering Biology are provided here courtesy of Wiley

RESOURCES