Next Article in Journal
Evaluation of Angiogenesis Assays
Previous Article in Journal
Recent Advances in Non-Conventional Antimicrobial Approaches for Chronic Wound Biofilms: Have We Found the ‘Chink in the Armor’?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Salmonella-Based Targeted Cancer Therapy: Updates on A Promising and Innovative Tumor Immunotherapeutic Strategy

1
Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
2
Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
*
Author to whom correspondence should be addressed.
Biomedicines 2019, 7(2), 36; https://doi.org/10.3390/biomedicines7020036
Submission received: 29 March 2019 / Revised: 25 April 2019 / Accepted: 29 April 2019 / Published: 2 May 2019
(This article belongs to the Special Issue Bacterial Cancer Therapy)

Abstract

:
Presently, cancer is one of the leading causes of death in the world, primarily due to tumor heterogeneity associated with high-grade malignancy. Tumor heterogeneity poses a tremendous challenge, especially with the emergence of resistance not only to chemo- and radiation- therapies, but also to immunotherapy using monoclonal antibodies. The use of Salmonella, as a highly selective and penetrative antitumor agent, has shown convincing results, thus meriting further investigation. In this review, the mechanisms used by Salmonella in combating cancer are carefully explained. In essence, Salmonella overcomes the suppressive nature of the tumor microenvironment and coaxes the activation of tumor-specific immune cells to induce cell death by apoptosis and autophagy. Furthermore, Salmonella treatment suppresses tumor aggressive behavior via inhibition of angiogenesis and delay of metastatic activity. Thus, harnessing the natural potential of Salmonella in eliminating tumors will provide an avenue for the development of a promising micro-based therapeutic agent that could be further enhanced to address a wide range of tumor types.

Graphical Abstract

1. Introduction

Bacteria-mediated tumor therapy has become one of the focal points in cancer therapeutic research over the past decades. The use of bacteria in cancer treatment may be favored over conventional treatment modalities such as surgery, chemo- and radiation therapies for several reasons [1]. First, surgery is only efficient in removing non-metastatic solid tumors, while bacteria harbor the potential to prevent metastasis. Second, tumor heterogeneity provides an avenue for the emergence of drug resistant and aggressive phenotypes whereas certain strains of bacteria used in tumor therapy can re-sensitize chemo-resistant tumor cells [1,2,3]. Lastly, the tumor microenvironment, such as the oxygen-depleted region of tumor, limits the effect of radiation therapy, whereas bacteria are able to disperse even at the hypoxic core [1,4].
One of the species used in bacteria-mediated tumor therapy is the gram negative, facultatively anaerobic Salmonella [5]. Salmonella sp. are able to penetrate and favorably invade tumor tissue by being attracted to the compounds produced by cancer cells as well as preferentially grow within tumor tissues [6]. Attenuated and engineered Salmonella have been demonstrated, both in in-vitro and murine models, to eliminate virulence while prompting antitumor activity via different mechanisms, largely due to the activation of tumor-specific humoral and cellular immune responses [7]. Despite being highly selective in targeting tumorigenic sites, the use of Salmonella in inhibiting tumor progression has encountered drawbacks such as host immunity against Salmonella antigens [8]. Interestingly, this problem has been addressed by modifications and alterations of the surface antigens or immunogenic components of Salmonella [9] and by coating the bacteria itself with certain materials like polymer to prevent the binding of Salmonella-specific antibodies, thus bypassing immune surveillance [10]. This review carefully explains the mechanisms underlying the tumor-targeting ability of Salmonella and its strategic activity in controlling tumor-specific responses that ultimately leads to inhibition of tumor growth and progression.

2. Salmonella Efficiently Targets and Invades Tumor

An interesting fact regarding Salmonella is their efficiency in selectively colonizing tumor tissue by being able to sense tumor-specific microenvironment. Solid tumors are characterized by depleted oxygen concentration ranging between 10 to 30 mmHg—otherwise known as hypoxia within the tumor tissue [11,12]—that is beneficial for Salmonella due to their facultative nature. To leverage this capability in targeting tumor tissue, an obligate anaerobic S. typhimurium strain (YB1) was generated by placing asd (aspartate-semialdehyde dehydrogenase) gene under the control of a hypoxia-conditioned promoter, that will be essential for survival only in a hypoxic condition [13]. Moreover, certain chemicals present in the tumor microenvironment, mostly in the necrotic region, could act as chemotactic agents that attract Salmonella to invade tumor tissue [6]. Salmonella have the ability to exploit ethanolamine as a signal to target and recognize niches including tumors [14,15]. The ability of Salmonella to efficiently reach and colonize tumor is facilitated by its motility, which is essential for effective dispersal in tumor [16]. Furthermore, Salmonella migration in the tumor is directed towards the core of the tumor and away from the vasculature [17]. Most Salmonella strains used for tumor therapy experiments and in clinical trials were attenuated, such as in the case of VNP20009 strain having partial deletion of the msbB gene for lipid A modification; in addition to that, the strain used bears CheY gene non-synonymous single nucleotide polymorphism, resulting in a reduced chemotactic ability [18,19]. Lipid A modification by msbB gene mutation reduced the lipopolysaccharide-associated toxicity or septic shock induction [20]. CheY gene is part of the two-component system CheA/CheY that is necessary or chemotactic response or motility and bacterial distribution in tumor tissue [6,18]; however, some reports showed that CheY protein deficiency does not compromise colonization in tumor [15]. In VNP20009, the efficiency of chemotactic response was only recovered after the replacement of the mutated CheY with the wild-type copy and restoring msbB gene [18,20] and further developed to enhance its hypoxia-targeting capacity [21]. Although several avirulent Salmonella strains generated via mutation demonstrated a reduced targeting and fitness ability, a variety of mutants categorized as class 1 such as htrA, SPI-2, and STM3120 mutations did not exhibit significant change in fitness and exclusive targeting of tumors [22]. In vitro targeting using tumor-on-a-chip device and in vivo targeting in tumor-bearing mice was achieved by Trg-deficient Salmonella through penetration of, and dispersal in, the quiescent tumor region [23].

3. Modified Salmonella Bypasses Antibacterial Immune Response

One of the drawbacks of using bacteria as an antitumor agent is the host’s immune response, triggered upon an increase in bacterial concentration, and ultimately leading to clearance of the introduced bacteria [24]. In fact, preexposure to Salmonella may hinder the therapeutic potential of bacteria-mediated tumor therapy [8,25]. This limiting effect of preexposure has been addressed by engineering Salmonella strain SF200 resulting to a modified Lipid A structure via ∆lpxR9 ∆pagL7 and ∆pagP8 deletions, and ∆ydiV and ∆fliF mutations to modify flagella synthesis [9,25]. The optimized SF200 strain showed significantly higher tumor regression in naïve and immunized tumor-bearing mice as compared to non-optimized variant, SL7207 that showed relatively low tumor regression [25]. Moreover, SF200 induced cytokine levels in immunized mice comparable to naïve mice, while SL7207 showed significantly lower cytokine levels in tumors of immunized mice compared to naïve mice [25]. In another experiment, Salmonella was encapsulated with poly(allylamine hydrochloride) or PAH to prevent binding of antibodies specific for Salmonella [10]. PAH-coated Salmonella (PAH-S.C.) did not elicit significant change in its tumor targeting ability and effectively cleared tumor in naïve and immunized tumor-bearing mice involving the recruitment of infiltrating immune cells such as neutrophils and macrophages [10].

4. Salmonella Promotes Activation of Antitumor Immunity

The development of various immune evading strategies by tumor cells enabled the generation of more aggressive phenotypes [26]. These highly aggressive phenotypes have led to the establishment of resistance to tumor immune therapy [26,27]. Systemic administration of Salmonella, shown in Figure 1, triggers immune cell infiltration and induction of proinflammatory cytokine expressions such as IL-1β and TNF-α [5,28]. Also, macrophages in Salmonella-colonized tumor express and activate the inflammasome pathway involving NLRP3, IPAF and caspase-1 p10 [29]. When Salmonella gained access to the tumor microenvironment, intratumoral CD11b+ myeloid cells accumulate and are coaxed to undergo phenotypic and functional maturation, impairing their suppressive activity [30,31]. The decrease in suppressive activity after Salmonella treatment involves downregulation of immunosuppressive factors, particularly arginase-1, IL-4, TGF-β, and VEGF, and an enhanced expression of inducible nitric oxide synthase (iNOS) and IFN-γ [31]. The enhanced expressions of iNOS, IFN-γ and other IFN-inducible chemokines may play a crucial role in the recruitment of neutrophils, activated CD8+ T cells and an increase in intratumoral activated NK cells [31,32,33]. It was previously reported that TLR4 signaling is involved in the Salmonella-induced cytokine expression [32] and that the lipopolysaccharide (LPS) of Salmonella might be crucial in the activation and recruitment of immune cells and the subsequent production primarily of TNF-α [34]. In a recent study, an engineered S. typhimurium strain secreting heterologous flagellin B (FlaB) in tumor tissues first induced the recruitment of infiltrating immune cells via TLR4 signaling followed by the activation of the recruited intratumoral macrophages via TLR5 signaling which appears to have caused the increase in the synthesis of cytotoxic mediators and cytokines [35]. In another study, the flagellin of S. typhimurium fused with peptide P10 of the gp43 protein from Paracoccidioides brasiliensis activated TLR5 signaling that helped impair the metastatic activity of melanoma in vivo [36].
Bacterial LPS and the enhanced synthesis of TNF-α induced the activation of CD8+ T cell that plays a major role in tumor regression [34], consistent with the findings that the antitumor activity of host immune system involves both CD4+ and CD8+ T cells rather than solely relying on innate mechanisms [37,38]. Lysis of tumor cells by anti-Salmonella-specific T cells further recruits infiltration of CD8+ T in Salmonella-colonized tumor that might eventually result in the uptake of tumor debris by antigen-presenting cells leading to presentation to naïve T cells and activation of tumor-specific T cells [7,37,39]. Previous reports have already pointed out that bacteria-induced gap junctions, such as connexin-43 (Cx43), can be formed between tumor and dendritic cells and promote tumor antigen cross-presentation [40]. The upregulation of Cx43 in tumor after Salmonella treatment explains tumor antigen cross-presentation leading to immune cell-mediated antitumor activity [41].
The activation of antitumor immunity mediated by Salmonella treatment does not rely only on the recruitment of a variety of infiltrating immune cells as mentioned previously. Interestingly, Salmonella invasion of tumor tissue interferes with the immunosuppressive nature of tumor microenvironment in a variety of mechanisms [5,7]. The immunosuppressive factors arginase-1, IL-4, TGF-β, and VEGF were known to be upregulated in many solid tumors but were found to be downregulated following Salmonella treatment [31]. Another immunosuppressive factor, the indoleamine 2, 3-dioxygenase 1 (IDO), is involved in mediating activation of regulatory T cells by increasing kynurenine concentration [42]. In a recent study, Salmonella downregulated IDO expression in B16F10 and 4T1 tumor cells via inhibition of AKT /mTOR/p70S6K signaling pathway, and thus resulted in a decrease in kynurenine concentration [43]. Since kynurenine is involved in competent T cell apoptosis [44] and Salmonella can reduce kynurenine, T cell survival increased as seen in Jurkat cells (T cells) cell viability cultured in a medium of tumor cells previously treated with highest dose of Salmonella [43]. In another study, S. typhimurium was transformed with shRNA targeting IDO to further enhance intratumoral cell death by inducing an increase in polymorphonuclear neutrophils (PMN) activity [45].

5. Salmonella Mediates Tumor Cell Self-Destruction

Programmed cell death, and apoptosis in particular, is crucial in tumor clearance involving the activation of caspase cascade in response to cancer therapy; however, in a state of high-grade malignancy, apoptosis remains attenuated [2]. The efficiency of Salmonella in combating tumor malignancy primarily involves the activation of cell death pathways (Figure 1 and Table 1) by nutrient competition and an enhanced stimulation of tumor-specific immune responses [5,17]. To highlight the mechanism, Salmonella treatment increased the expression of cleaved caspase-3 required in the activation of apoptosis via caspase cascade system in two melanoma models, K1735 and B16F10 [46]. Apoptotic response of tumor was only reversed in pan-caspase inhibitor Z-VAD-FMK-treated tumor cells which verified the theory that apoptosis is involved in Salmonella-mediated tumor cell death [46]. Furthermore, autophagy adds up to the layer of responses following Salmonella treatment as indicated by the upregulation of Beclin-1 along with an enhanced conversion of LC3-I to LC3-II, an autophagosomal marker [46]. The activation of autophagic signaling pathway is mediated by downregulation of AKT/mTOR/p70S6K being a crucial target of Salmonella in tumor cells [46]. In another study, Salmonella was used as a vector to carry second mitochondria-derived activator of caspases (Smac) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) genes which enhance apoptotic cell death and resulted to almost 90% regression of tumor growth in murine melanoma, lung carcinoma, and mammary carcinoma models [47,48].

6. Salmonella Diminishes Tumor Metastasis

The prevention of metastatic activity by Salmonella through various mechanisms prevent tumor cells from migrating (Figure 1 and Table 1). Failure to detect the advancement of tumor into metastatic state where cancer cells gain access to different sites of the body leads to a significantly poor prognosis which, in turn, dramatically affects patients’ survival. This is primarily due to the changes in the behavior of cells in the primary tumor causing migration and aggressive invasion of the proximal and even distal organs forming secondary lesions [52,53].
The initiation of angiogenic sprouting in tumor marks the dramatic shift from dormancy to tumor progression [2,54]. Angiogenesis is not only critical to tumor growth per se, but also to tumor migration because the new blood vessels formed provide the major route used by disintegrating tumor cells to escape from the primary tumor and start migration [55]. Many studies have already demonstrated the role of this angiogenesis in tumor metastasis and, thus, that the inhibition of angiogenic sprouting may help contain the tumor and stop its metastatic activity. The regulation of angiogenic sprouting is orchestrated by the angiogenic factors vascular endothelial growth factor-A (VEGF-A) and thrombospondin-1 (TSP-1) that either stimulates or inhibits cell-surface receptors, respectively [56]. In the tumor microenvironment, the presence of oncogenic signals and hypoxic condition activate certain transcription regulators such as hypoxia-inducible factor 1-alpha (HIF-1α) to induce upregulation of VEGF [57,58,59] that largely influences tumor vascularization. In a study by Tu et al. [49], Salmonella inhibited angiogenesis by negatively regulating VEGF expressions in two tumor cells namely, B16F10 and 4T1. The downregulation of VEGF is attributed to the downregulation of its gene regulator HIF-1α, suggesting that the anti-angiogenic activity of Salmonella targets the HIF-1α dependent pathway. The activation of HIF-1α is mediated by AKT/mTOR/p70S6K cascade [49,59,60,61] which was found to be significantly reduced after Salmonella treatment as indicated by decline in AKT, mTOR and p70S6K phosphorylation. Interestingly, the frequency of viable human microvascular endothelial cells (HMEC-1) declined significantly post-treatment with conditioned medium of either Salmonella-treated B16F10 or 4T1 cells [49]. Consistent with the findings already mentioned, in vivo study using tumor-bearing mice showed reduced VEGF levels in tumor tissue and reduced tumor microvessel density that is necessary to confine tumor in the primary site [49].
For metastatic tumor to escape primary site and eventually migrate to other organs to seed secondary tumors, metastatic tumor cells rely on an important process involving proteolytic degradation of extracellular matrix (ECM) by proteinases such as matrix metalloproteinase [62]. Matrix metalloproteinase, a member of the zinc-dependent endopeptidases family, is known to modulate ECM remodeling in normal development, inflammation, and wound healing [63,64,65,66]. In various types of tumors, MMP-9 expression is primarily implicated in metastatic phenotypes by acting upon the ECM components thereby altering adhesive capability and in promoting tumor vascularization [67,68,69]. MMP-9 expression contributes to the epithelium to mesenchymal transition (EMT), serving as one of the markers of cancer stem cells [70]. It is therefore advantageous to utilize therapeutic strategies targeting MMP-9 expression. Negative regulation of MMP-9 expression has also been demonstrated to suppress metastatic activity of prostate cancer [71,72] malignant glioma [73] and renal cell carcinoma [74]. When Salmonella is used as an antitumor agent, it inhibits not only tumor vascularization [49], but also prevents EMT by keeping an intact ECM via suppression of MMP-9 expressions [50]. The inhibition of tumor cell migration observed in wound-healing and transwell assay was found to be due to MMP-9 suppression that is controlled by AKT/mTOR axis. The downregulation of phospho-AKT/phospho-mTOR after Salmonella treatment in both B16F10 and LL2 cells resulted in a decrease of MMP-9 expression which was reversed in tumor cells transfected with constitutively active AKT [50].

7. Salmonella Enhances Chemosensitivity of Tumor

An alarming increase of chemoresistance fueled by tumor heterogeneity among genetically-distinct sub-populations of tumor cells [3] demanded the exploration of alternative efficient therapeutic strategies that could combat drug resistance or a way to sensitize an already resistant phenotype. Mechanism of multi-drug resistance among various types of cancer primarily includes alteration of membrane permeability via transporter proteins, i.e. P-glycoprotein (P-gp), involved in drug efflux [75,76]. Many other resistance mechanisms causing poor responses to various drugs include alteration of target enzymes, alteration of drug metabolism, suppression of apoptosis, and enhancement of DNA repair mechanisms, among others [76,77]. Surprisingly, as shown in Figure 2 and Table 1, Salmonella can sensitize drug resistant-tumor cells (K1735 melanoma cells) to cisplatin—a cytotoxic drug—by enhancing gap intercellular communication (GJIC) mediated by the upregulation of connexin 43 (Cx43) [41]. Furthermore, the study showed that Cx43 was mediated by p38 signaling cascade as determined by using inhibitor of p38 that blocked the Salmonella-mediated Cx43 expression. Furthermore, overexpression Cx43 increased the expression of p38, while Cx43-silenced cells showed no significant changes on p38 expressions [41]. A recent study revealed that the same Salmonella mediated Cx43 upregulation and also contributed to the downregulation of P-gp, further confirming the ability of Salmonella to sensitize multidrug-resistant tumor cells [51]. Previous findings have also demonstrated the potential of S. enterica serovar Typhimurium in modulating the expression of P-gp in the epithelial lining of the intestine [78]. Poor prognosis in many patients with solid tumors and blood malignancies often have unfavorably enhanced P-gp efflux potential that drives cancer drug out of tumor cells [76]. The expression of P-gp, which is normally controlled by AKT/mTOR signaling via p70s6K phosphorylation [79,80], was significantly reduced in B16F10 and 4T1 cells after Salmonella treatment in a dose-dependent manner [51]. The downregulation of P-gp mediated by Salmonella showed a significant increase in Rho-123 intracellular accumulation suggesting suppressed P-gp transport activity, and an increased susceptibility of tumor cells to 5-FU [51]. Previously, Mercado-Lubo et al. [81], reported that SipA, a type III secretion effector in S. enterica, controls P-gp levels involving caspase-3-mediated protein degradation rather than transcriptional control.

8. Combination Therapy with Salmonella Further Improves Tumor-Regression

In recent years, bacteria-mediated tumor therapy is no longer centered on using Salmonella alone as a means of combating cancer. An increasing number of studies have now been reported to maximally exploit the potential of Salmonella in combination with other therapeutic strategies to combat cancer [1,7].
The ability of Salmonella to sensitize multi-drug resistant tumors have encouraged researchers to use various cancer drugs such as cisplatin or 5- Fluorouracil combined with Salmonella treatment [41,51]. A combined Salmonella therapy and cyclophosphamide drug treatment improves tumor regression and significantly decreased tumor micro-vascularization in melanoma model [82]. Recently, Bascuas et al. [83] demonstrated that Salmonella treatment in B-cell non-Hodgkin lymphoma (B-NHL)-bearing mice enhanced the effect of chemotherapy using CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone). CHOP treatment in mice model prior to Salmonella treatment revealed enhanced NK cell cytotoxic activity and a significantly higher lymphoma-specific humoral and cellular immune responses compared to Salmonella alone nor CHOP alone treatment [83].
In another study, adoptive T cell therapy was combined with Salmonella treatment, in which intravenous injection of either viable or heat killed (HK) Salmonella enhanced the proliferation of adoptively transferred OT-1 T cells and significantly improved tumor regression compared to OT-1 T cells alone [84]. Despite the successful regression of tumor by HK Salmonella combined with adoptive T cell therapy, one mouse died after 16 days post-treatment indicative of potential increase in toxicity. The observed mortality may be due to elevated levels of IL-6 and other pro-inflammatory cytokines which can be addressed by applying neutralizing antibodies [84].
The hypoxia-targeting ability of Salmonella was used in tandem with photothermal therapy using the carbon-based nanomaterial, polydopamine [85]. In the study, Salmonella strain VNP20009 was coated with polydopamine, designated as pDA-VNP, to act as photothermal agent. The efficacy of photothermal therapy is mediated by photothermal agents, such as pDA, to induce elevation of local temperature by converting incident light into heat [86]. The findings suggest that the targeting ability of Salmonella successfully delivered polydopamine to the tumor site and that near-infrared irradiation induced significant increase in temperature in the tumor site enough to cause tumor cell decay. In vitro cytotoxicity revealed significantly lower viable B16F10 cells in pDA-VNP post-irradiation group compared with VNP20009 alone [85]. Similar findings were observed in melanoma mouse model as indicated by apoptotic and necrotic cells and confirmed by infrared thermal imaging of mice showing significant increase of temperature in pDA-VNP with irradiation [85].

9. Clinical Trials, Challenges and Future Perspectives

The first attempt of using Salmonella in preclinical studies and clinical trials for the treatment of advanced or metastatic tumor can be dated back in the early 2000 where, in particular, the strain VNP20009 described earlier in this paper, was developed by Vion Pharmaceutics Inc [87]. Unfortunately, the phase 1 clinical trials of VNP20009 was discontinued due to low tumor regression and incidence of side effects at high dosage [85,87]. Despite the outcome, VNP20009 has been safely administered via intravenous infusion resulting in increased circulation of proinflammatory cytokines and marked colonization in the tumor biopsies of three out of 25 patients in the trial [87,88]; this means, it is safe to assume that Salmonella can be further developed to improve the outcomes of future clinical trials. One hindrance affecting the tumor targeting ability of VNP20009, as reflected in the results of phase-1 clinical trials, is the point mutation in CheY gene that forms part of the two-component system CheA/CheY associated with motility and bacterial distribution of Salmonella in tumor tissue [6,18]. The chemotactic ability of VNP20009 was enhanced, by 69% efficiency, with respect to the parental strain, after replacing the CheY mutated copy with the wild-type sequence [18] and then optimized by restoring msbB gene in VNP20009 CheY+ that further increased chemotactic mobility [20]. Another strategery that may better equip Salmonella in targeting tumor sites is surface modification. Recently, Park et al. [89] modified the S. typhimurium to display arginine-glycine-aspartate (RGD) peptide sequence on the external loop of outer membrane protein A (OmpA). RGD peptide binds efficiently to αvβ3 which is overexpressed in most tumor cells; therefore, RGD-displaying Salmonella can target tumor overexpressing αvβ3 at high efficiency while weakly binding to αvβ3-negative cells [89].
Majority of Salmonella strains used as anticancer agents were developed by disabling genes associated with virulence which may be required for immune activation associated with antitumor response, an essential aspect that makes cancer immunotherapy efficient. Several reports have already provided solutions for the improvement of the antitumor potential of Salmonella, mostly by surface modification and/or engineering of various strains, such as VNP20009, A1-R and the avirulent ∆ppGpp, among others, but not directly addressing over-attenuation [5,90]. Low tumor regression possibly due to over-attenuation can be addressed by altering a virulence factor to be expressed under an inducible promoter such as PBAD requiring arabinose as an inducer in an in vitro system [91]. The concentration of arabinose becomes heavily diluted after administration, which will eventually lead to a halted virulence factor expression. In turn, Salmonella will become attenuated after a few rounds of replication without the inducer arabinose. For instance, the transcriptional regulator phoP that is responsible for Salmonella virulence and a regulator of LPS structural modifications, has been engineered to be expressed under PBAD promoter [92]. By so doing, a more vigorous anti-tumor activity may be prompted compared to an over-attenuated strain. This opens another area of investigation in cancer therapeutics that is focused on using anticancer Salmonella.
In another phase 1 clinical trial, S. typhi Ty21a was used as a vector to deliver the oral DNA vaccine VXM01 targeting the vascular endothelial growth factor receptor 2 (VEGFR-2) as an antiangiogenic intervention for advanced pancreatic cancer patients [93,94]. Preliminary findings suggest that Ty21a can be safely administered with only minimal adverse effect, relatively substantial tumor regression and a significant reduction of tumor perfusion along with elevated levels of serum anti-angiogenic biomarkers [94]. In the phase 1 trial extension, treatment-associated adverse reactions were significantly decreased along with increased vaccine specific T cell responses [95]. Another vaccine strain of Salmonella called TXSVN is set for a phase-1 clinical trial for multiple myeloma [96]. This genetically altered Salmonella produces tumor-associated antigens (TAAs) known as Survivin and is expected to induce tumor-specific immune response.
Ultimately, with the right attenuated strain that is safe for systemic administration harboring the essential elements involved in tumor targeting, as well as efficiently prompting oncolysis, Salmonella-based targeted cancer therapy will play a significant part in cancer immunotherapy. In addition, investigating further on the innate mechanisms of Salmonella in disrupting tumor growth and progression may prove helpful in maximizing the potential of this bacteria for use in monotherapy, vaccine delivery vector or in tandem with other useful therapeutic interventions.

10. Conclusions

The increasing number of relevant findings from cell-based to preclinical researches clearly suggest that Salmonella-based targeted therapy is a promising therapeutic candidate that could potentially become a mainstream therapeutic strategy in cancer treatment. Because Salmonella can efficiently work in a multifaceted interplay between upregulation of immunomodulatory molecules and downregulation of aggressive phenotype-related proteins to counteract various protumor cellular processes, it can be utilized to improve survival of cancer patients or can be enhanced to improve outcomes of existing treatment strategies. Most of the obstacles in using Salmonella have already been addressed or at least, lessened, such as potential toxicity and host immune response against the bacterial agent itself. Salmonella may not be the holy grail, in terms of cancer therapeutics, but further improvement of the treatment, through bioengineering and/or combinatorial treatments, may prove to be significantly effective in combating high-grade cancer malignancies.

Author Contributions

Writing—original draft preparation, C.R.P.; writing—review and editing, C.-H.L.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AKTprotein kinase B
asdaspartate-semialdehyde dehydrogenase gene
B-NHLB-cell non-Hodgkin lymphoma
CHOPcyclophosphamide, doxorubicin, vincristine, and prednisone
Cx43connexin-43
ECMextracellular matrix
EMTepithelium to mesenchymal transition
GJICgap junction intercellular communication
HIF-1hypoxia inducible factor-1
HKheat killed
IDOindoleamine 2, 3-dioxygenase 1
IFN-γinterferon-γ
ILinterleukins
iNOSinducible nitric oxide synthase
LPSlipopolysaccharide
MMPmatrix metalloproteinase
mTORmammalian target of rapamycin
OmpAouter membrane protein A
p70S6Kribosomal protein S6 kinase beta-1
P-gpP glycoprotein
PAHpoly(allylamine hydrochloride)
pDApolydopamine
PMNpolymorphonuclear neutrophils
RGDarginine-glycine-aspartate
shRNAshort hairpin ribonucleic acid
TGF-βtransforming growth factor-β
TLRtoll like receptor
TNF-αtumor necrosis factor-α
TRAILtumor necrosis factor-related apoptosis-inducing ligand
TSP-1thrombospondin-1
VEGFvascular endothelial growth factor.

References

  1. Nguyen, V.H.; Min, J.J. Salmonella-mediated cancer therapy: Roles and potentials. Nucl. Med. Mol. Imaging 2017, 51, 118–126. [Google Scholar] [CrossRef]
  2. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  3. Dagogo-Jack, I.; Shaw, A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2018, 15, 81–94. [Google Scholar] [CrossRef]
  4. Brown, J.M.; Wilson, W.R. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 2004, 4, 437–447. [Google Scholar] [CrossRef]
  5. Lee, C.H. Engineering bacteria toward tumor targeting for cancer treatment: Current state and perspectives. Appl. Microbiol. Biotechnol. 2012, 93, 517–523. [Google Scholar] [CrossRef] [PubMed]
  6. Kasinskas, R.W.; Forbes, N.S. Salmonella typhimurium specifically chemotax and proliferate in heterogeneous tumor tissue in vitro. Biotechnol. Bioeng. 2006, 94, 710–721. [Google Scholar] [CrossRef]
  7. Chang, W.W.; Lee, C.H. Salmonella as an innovative therapeutic antitumor agent. Int. J. Mol. Sci. 2014, 15, 14546–14554. [Google Scholar] [CrossRef] [PubMed]
  8. Lee, C.H.; Wu, C.L.; Chen, S.H.; Shiau, A.L. Humoral immune responses inhibit the antitumor activities mediated by Salmonella enterica serovar choleraesuis. J. Immunother. 2009, 32, 376–388. [Google Scholar] [CrossRef]
  9. Needham, B.D.; Carroll, S.M.; Giles, D.K.; Georgiou, G.; Whiteley, M.; Trent, M.S. Modulating the innate immune response by combinatorial engineering of endotoxin. Proc. Natl. Acad. Sci. USA 2013, 110, 1469–1499. [Google Scholar] [CrossRef]
  10. Lee, C.H.; Lin, Y.H.; Hsieh, J.L.; Chen, M.C.; Kuo, W.L. A polymer coating applied to Salmonella prevents the binding of Salmonella-specific antibodies. Int. J. Cancer 2013, 132, 717–725. [Google Scholar] [CrossRef] [PubMed]
  11. Wei, M.Q.; Ellem, K.A.O.; Dunn, P.; West, M.J.; Bai, C.X.; Vogelstein, B. Facultative or obligate anaerobic bacteria have the potential for multimodality therapy of solid tumours. Eur. J. Cancer 2007, 43, 490–496. [Google Scholar] [CrossRef]
  12. St Jean, A.T.; Zhang, M.; Forbes, N.S. Bacterial therapies: Completing the cancer treatment toolbox. Curr. Opin. Biotechnol. 2008, 19, 511–517. [Google Scholar] [CrossRef]
  13. Yu, B.; Yang, M.; Shi, L.; Yao, Y.; Jiang, Q.; Li, X.; Tang, L.H.; Zheng, B.J.; Yuen, K.Y.; Smith, D.K.; et al. Explicit hypoxia targeting with tumor suppression by creating an “obligate” anaerobic Salmonella typhimurium strain. Sci. Rep. 2012, 2, 436. [Google Scholar] [CrossRef]
  14. Anderson, C.J.; Clark, D.E.; Adli, M.; Kendall, M.M. Ethanolamine signaling promotes Salmonella niche recognition and adaptation during infection. PLoS Pathog. 2015, 11, e1005278. [Google Scholar]
  15. Silva-Valenzuela, C.A.; Desai, P.T.; Molina-Quiroz, R.C.; Pezoa, D.; Zhang, Y.; Porwollik, S.; Zhao, M.; Hoffman, R.M.; Contreras, I.; Santiviago, C.A.; et al. Solid tumors provide niche-specific conditions that lead to preferential growth of Salmonella. Oncotarget 2016, 7, 35169–35180. [Google Scholar] [CrossRef] [PubMed]
  16. Toley, B.J.; Forbes, N.S. Motility is critical for effective distribution and accumulation of bacteria in tumor tissue. Integr. Biol. 2012, 4, 165–176. [Google Scholar] [CrossRef]
  17. Ganai, S.; Arenas, R.B.; Sauer, J.P.; Bentley, B.; Forbes, N.S. In tumors Salmonella migrate away from vasculature toward the transition zone and induce apoptosis. Cancer Gene Ther. 2011, 18, 457–466. [Google Scholar] [CrossRef]
  18. Broadway, K.M.; Denson, E.A.P.; Jensen, R.V.; Scharf, B.E. Rescuing chemotaxis of the anticancer agent Salmonella enterica serovar Typhimurium VNP20009. J. Biotechnol. 2015, 211, 117–120. [Google Scholar] [CrossRef]
  19. Clairmont, C.; Lee, K.C.; Pike, J.; Ittensohn, M.; Low, K.B.; Pawelek, J.; Bermudes, D.; Brecher, S.M.; Margitich, D.; Turnier, J.; et al. Biodistribution and Genetic Stability of the Novel Antitumor Agent VNP20009, a Genetically Modified Strain of Salmonella Typhimurium. J. Infect. Dis. 2000, 181, 1996–2002. [Google Scholar] [CrossRef]
  20. Broadway, K.M.; Suh, S.; Behkam, B.; Scharf, B.E. Optimizing the restored chemotactic behavior of anticancer agent Salmonella enterica serovar Typhimurium VNP20009. J. Biotechnol. 2017, 251, 76–83. [Google Scholar] [CrossRef] [PubMed]
  21. Coutermarsh-Ott, S.L.; Broadway, K.M.; Scharf, B.E.; Allen, I.C. Effect of Salmonella enterica serovar Typhimurium VNP20009 and VNP20009 with restored chemotaxis on 4T1 mouse mammary carcinoma progression. Oncotarget 2017, 8, 33601–33613. [Google Scholar] [CrossRef]
  22. Arrach, N.; Cheng, P.; Zhao, M.; Santiviago, C.A.; Hoffman, R.M.; McClelland, M. High-throughput screening for Salmonella avirulent mutants that retain targeting of solid tumors. Cancer Res. 2010, 70, 2165–2170. [Google Scholar] [CrossRef] [Green Version]
  23. Zhang, M.; Forbes, N.S. Trg-deficient Salmonella colonize quiescent tumor regions by exclusively penetrating or proliferating. J. Control Release 2015, 199, 180–189. [Google Scholar] [CrossRef]
  24. Palffy, R.; Gardlik, R.; Hodosy, J.; Behuliak, M.; Resko, P.; Radvansky, J.; Celec, P. Bacteria in gene therapy: Bactofection versus alternative gene therapy. Gene Ther. 2006, 13, 101–105. [Google Scholar] [CrossRef]
  25. Felgner, S.; Kocijancic, D.; Frahm, M.; Heise, U.; Rohde, M.; Zimmermann, K.; Falk, C.; Erhardt, M.; Weiss, S. Engineered Salmonella enterica serovar Typhimurium overcomes limitationsof anti-bacterial immunity in bacteria-mediated tumor therapy. Oncoimmunology 2018, 7, e1382791. [Google Scholar] [CrossRef]
  26. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumarai, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [Green Version]
  27. Spranger, S.; Gajewski, T.F. Mechanisms of tumor cell–intrinsic immune evasion. Ann. Rev. Cancer Biol. 2018, 2, 213–228. [Google Scholar] [CrossRef]
  28. Kim, J.E.; Phan, T.X.; Nguyen, V.H.; Dinh-Vu, H.V.; Zheng, J.H.; Yun, M.; Park, S.G.; Hong, Y.; Choy, H.E.; Szardenings, M.; et al. Salmonella typhimurium suppresses tumor growth via the pro-inflammatory cytokine interleukin-1β. Theranostics 2015, 5, 1328–1342. [Google Scholar] [CrossRef] [PubMed]
  29. Phan, T.X.; Nguyen, V.H.; Duong, M.T.; Hong, Y.; Choy, H.E.; Min, J.J. Activation of inflammasome by attenuated Salmonella typhimurium in bacteria-mediated cancer therapy. Microbiol. Immunol. 2015, 59, 664–675. [Google Scholar] [CrossRef] [PubMed]
  30. Tam, J.W.; Kullas, A.L.; Mena, P.; Bliska, J.B.; van der Velden, A.W.M. CD11b+ Ly6Chi Ly6G immature myeloid cells recruited in response to Salmonella enterica serovar Typhimurium infection exhibit protective and immunosuppressive properties. Infect. Immunity 2014, 82, 2606–2614. [Google Scholar] [CrossRef]
  31. Kaimala, S.; Mohamed, Y.A.; Nader, N.; Issac, J.; Elkord, E.; Chouaib, S.; Fernandez-Cabezudo, M.J.; al-Ramadi, B.K. Salmonella-mediated tumor regression involves targeting of tumor myeloid suppressor cells causing a shift to M1-like phenotype and reduction in suppressive capacity. Cancer Immunol. Immunother. 2014, 63, 587–599. [Google Scholar] [CrossRef]
  32. Lee, C.H.; Wu, C.L.; Shiau, A.L. Toll-like receptor 4 mediates an antitumor host response induced by Salmonella choleraesuis. Clin. Cancer Res. 2008, 14, 1905–1912. [Google Scholar] [CrossRef]
  33. Grille, S.; Moreno, M.; Bascua, T.; Marqués, J.M.; Muñoz, N.; Lens, D.; Chabalgoity, J.A. Salmonella enterica serovar Typhimurium immunotherapy for B-cell lymphoma induces broad anti-tumour immunity with therapeutic effect. Immunology 2014, 143, 428–437. [Google Scholar] [CrossRef]
  34. Kocijancic, D.; Leschner, S.; Felgner, S.; Komoll, R.M.; Frahm, M.; Pawar, V.; Weiss, S. Therapeutic benefit of Salmonella attributed to LPS and TNF-α is exhaustible and dictated by tumor susceptibility. Oncotarget 2017, 8, 36492–36508. [Google Scholar] [CrossRef]
  35. Zheng, J.H.; Nguyen, V.H.; Jiang, S.N.; Park, S.H.; Tan, W.; Hong, S.H.; Shin, M.G.; Chung, I.J.; Hong, Y.; Bom, H.S.; et al. Two-step enhanced cancer immunotherapy with engineered Salmonella typhimurium secreting heterologous flagellin. Sci. Transl. Med. 2017, 9, eaak9537. [Google Scholar] [CrossRef]
  36. De Melo, F.M.; Braga, C.J.; Pereira, F.V.; Maricato, J.T.; Origassa, C.S.; Souza, M.F.; Melo, A.C.; Silva, P.; Tomaz, S.L.; Gimenes, K.P.; et al. Anti-metastatic immunotherapy based on mucosal administration of flagellin and immunomodulatory P10. Immunol. Cell Biol. 2015, 93, 86–98. [Google Scholar] [CrossRef]
  37. Lee, C.H.; Hsieh, J.L.; Wu, C.L.; Hsu, P.Y.; Shiau, A.L. T cell augments the antitumor activity of tumor-targeting Salmonella. Appl. Microbiol. Biotechnol. 2011, 90, 1381–1388. [Google Scholar] [CrossRef]
  38. Stern, C.; Kasnitz, N.; Kocijancic, D.; Trittel, S.; Riese, P.; Guzman, C.A.; Leschner, S.; Weiss, S. Induction of CD4(+) and CD8(+) anti-tumor effector T cell responses by bacteria mediated tumor therapy. Int. J. Cancer 2015, 137, 2019–2028. [Google Scholar] [CrossRef]
  39. Avogadri, F.; Martinoli, C.; Petrovska, L.; Chiodoni, C.; Transidico, P.; Bronte, V.; Longhi, R.; Colombo, M.P.; Dougan, G.; Rescigno, M. Cancer immunotherapy based on killing of Salmonella-infected tumor cells. Cancer Res. 2005, 65, 3920–3927. [Google Scholar] [CrossRef]
  40. Saccheri, F.; Pozzi, C.; Avogadri, F.; Barozzi, S.; Faretta, M.; Fusi, P.; Rescigno, M. Bacteria-induced gap junctions in tumors favor antigen cross-presentation and antitumor immunity. Sci. Transl. Med. 2010, 2, 44–57. [Google Scholar] [CrossRef]
  41. Chang, W.W.; Lai, C.H.; Chen, M.C.; Liu, C.F.; Kuan, Y.D.; Lin, S.T.; Lee, C.H. Salmonella enhance chemosensitivity in tumor through connexin 43 upregulation. Int. J. Cancer 2013, 133, 1926–1935. [Google Scholar] [CrossRef] [Green Version]
  42. Brandacher, G.; Perathoner, A.; Ladurner, R.; Schneeberger, S.; Obrist, P.; Winkler, C.; Werner, E.R.; Werner-Felmayer, G.; Weiss, H.G.; Gobel, G.; et al. Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: Effect on tumor-infiltrating T cells. Clin. Cancer Res. 2006, 12, 1144–1151. [Google Scholar] [CrossRef]
  43. Kuan, Y.D.; Lee, C.H. Salmonella overcomes tumor immune tolerance by inhibition of tumor indoleamine 2, 3-dioxygenase 1 expression. Oncotarget 2015, 7, 374–385. [Google Scholar] [CrossRef]
  44. Sun, J.; Yu, J.; Li, H.; Yang, L.; Wei, F.; Yu, W.; Liu, J.; Ren, X. Upregulated expression of indoleamine 2, 3-dioxygenase in CHO cells induces apoptosis of competent T cells and increases proportion of Treg cells. J. Exp. Clin. Cancer Res. 2011, 30, 82. [Google Scholar] [CrossRef]
  45. Blache, C.A.; Manuel, E.R.; Kaltcheva, T.I.; Wong, A.N.; Ellenhorn, J.D.I.; Blazar, B.R.; Diamond, D.J. Systemic delivery of Salmonella typhimurium transformed with IDO shRNA enhances intratumoral vector colonization and suppresses tumor growth. Cancer Res. 2012, 72, 6447–6456. [Google Scholar] [CrossRef]
  46. Lee, C.H.; Lin, S.T.; Liu, J.J.; Chang, W.W.; Hsieh, J.L.; Wang, W.K. Salmonella induce autophagy in melanoma by the downregulation of AKT/mTOR pathway. Gene Ther. 2014, 21, 309–316. [Google Scholar] [CrossRef]
  47. Fu, W.; Chu, L.; Han, X.; Liu, X.; Ren, D. Synergistic antitumoral effects of human telomerase reverse transcriptase-mediated dual-apoptosis-related gene vector delivered by orally attenuated Salmonella enterica Serovar Typhimurium in murine tumor models. J. Gene Med. 2008, 10, 690–701. [Google Scholar] [CrossRef]
  48. Chen, J.; Yang, B.; Cheng, X.; Qiao, Y.; Tang, B.; Chen, G.; Wei, J.; Liu, X.; Cheng, W.; Du, P.; et al. Salmonella-mediated tumor-targeting TRAIL gene therapy significantly suppresses melanoma growth in mouse model. Cancer Sci. 2012, 103, 325–333. [Google Scholar] [CrossRef]
  49. Tu, D.G.; Chang, W.W.; Lin, S.T.; Kuo, C.Y.; Tsao, Y.T.; Lee, C.H. Salmonella inhibits tumor angiogenesis by downregulation of vascular endothelial growth factor. Oncotarget 2016, 7, 37513–37523. [Google Scholar] [CrossRef]
  50. Tsao, Y.T.; Kuo, C.Y.; Cheng, S.P.; Lee, C.H. Downregulations of AKT/mTOR Signaling Pathway for Salmonella-Mediated Suppression of Matrix Metalloproteinases-9 Expression in Mouse Tumor Models. Int. J. Mol. Sci. 2018, 19, 1630. [Google Scholar] [CrossRef]
  51. Yang, C.J.; Chang, W.W.; Lin, S.T.; Chen, M.C.; Lee, C.H. Salmonella Overcomes Drug resistance in tumor through P-glycoprotein downregulation. Int. J. Med. Sci. 2018, 15, 574–579. [Google Scholar] [CrossRef]
  52. Gupta, G.P.; Massagué, J. Cancer metastasis: Building a framework. Cell 2006, 127, 679–695. [Google Scholar] [CrossRef]
  53. Nguyen, D.X.; Bos, P.D.; Massagué, J. Metastasis: From dissemination to organ-specific colonization. Nat. Rev. Cancer 2009, 9, 274–284. [Google Scholar] [CrossRef]
  54. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  55. Zetter, B.R. Angiogenesis and tumor metastasis. Annu. Rev. Med. 1998, 49, 407–424. [Google Scholar] [CrossRef]
  56. Baeriswyl, V.; Christofori, G. The angiogenic switch in carcinogenesis. Semin. Cancer Biol. 2009, 19, 329–337. [Google Scholar] [CrossRef]
  57. Ferrara, N. Vascular endothelial growth factor. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 789–791. [Google Scholar] [CrossRef]
  58. Carmeliet, P. VEGF as a key mediator of angiogenesis in cancer. Oncology 2005, 69, 4–10. [Google Scholar] [CrossRef]
  59. Semenza, G.L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 2003, 3, 721–732. [Google Scholar] [CrossRef]
  60. Chen, M.C.; Hsu, W.L.; Hwang, P.A.; Chou, T.C. Low molecular weight fucoidan inhibits tumor angiogenesis through downregulation of HIF-1/VEGF signaling under Hypoxia. Mar. Drugs 2015, 13, 4436–4451. [Google Scholar] [CrossRef]
  61. Ataie-Kachoie, P.; Pourgholami, M.H.; Bahrami, B.F.; Badar, S.; Morris, D.L. Minocycline attenuates hypoxia-inducible factor-1α expression correlated with modulation of p53 andAKT/mTOR/p70S6K/4E-BP1 pathway in ovarian cancer: In vitro and in vivo studies. Am. J. Cancer Res. 2015, 5, 575–588. [Google Scholar] [PubMed]
  62. Yilmaz, M.; Christofori, G.; Lehembre, F. Distinct mechanisms of tumor invasion and metastasis. Trends Mol. Med. 2007, 13, 535–541. [Google Scholar] [CrossRef]
  63. Stamenkovic, I. Extracellular matrix remodelling: The role of matrix metalloproteinases. J. Pathol. 2003, 200, 448–464. [Google Scholar] [CrossRef]
  64. Parks, W.C.; Wilson, C.L.; Lopez-Boado, Y.S. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat. Rev. Immunol. 2004, 4, 617–629. [Google Scholar] [CrossRef]
  65. Page-McCaw, A.; Ewald, A.J.; Werb, Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat. Rev. Mol. Cell Biol. 2007, 8, 221. [Google Scholar] [CrossRef]
  66. Chen, P.; Parks, W.C. Role of matrix metalloproteinases in epithelial migration. J. Cell. Biochem. 2009, 108, 1233–1243. [Google Scholar] [CrossRef]
  67. Klein, G.; Vellenga, E.; Fraaije, M.; Kamps, W.; De Bont, E. The possible role of matrix metalloproteinase (MMP)-2 and MMP-9 in cancer, e.g., acute leukemia. Crit. Rev. Oncol. Hematol. 2004, 50, 87–100. [Google Scholar] [CrossRef]
  68. Cotignola, J.; Reva, B.; Mitra, N.; Ishill, N.; Chuai, S.; Patel, A.; Shah, S.; Vanderbeek, G.; Coit, D.; Busam, K.; et al. Matrix Metalloproteinase-9 (MMP-9) polymorphisms in patients with cutaneous malignant melanoma. BMC Med. Genet. 2007, 8, 10. [Google Scholar] [CrossRef]
  69. Mehner, C.; Hockla, A.; Miller, E.; Ran, S.; Radisky, D.C.; Radisky, E.S. Tumor cell-produced matrix metalloproteinase 9 (MMP-9) drives malignant progression and metastasis of basal-like triple negative breast cancer. Oncotarget 2014, 5, 2736–2749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Wang, S.; Yan, Y.; Cheng, Z.; Hu, Y.; Liu, T. Sotetsuflavone suppresses invasion and metastasis in non-small-cell lung cancer A549 cells by reversing EMT via the TNF-α/NF-κB and PI3K/AKT signaling pathway. Cell Death Discov. 2018, 4, 26. [Google Scholar] [CrossRef] [Green Version]
  71. Chen, Y.; Zheng, L.; Liu, J.; Zhou, Z.; Cao, X.; Lv, X.; Chen, F. Shikonin inhibits prostate cancer cells metastasis by reducing matrix metalloproteinase-2/-9 expression via AKT/mTOR and ROS/ERK1/2 pathways. Int. Immunopharmacol. 2014, 21, 447–455. [Google Scholar] [CrossRef] [PubMed]
  72. Kou, B.; Liu, W.; He, W.; Zhang, Y.Y.; Zheng, J.J.; Yan, Y.; Zhang, Y.J.; Xu, S.C.; Wang, H. Tetrandrine suppresses metastatic phenotype of prostate cancer cells by regulating Akt/mTOR/MMP-9 signaling pathway. Oncol. Rep. 2016, 35, 2880–2886. [Google Scholar] [CrossRef]
  73. Jiang, Y.; Zhang, Q.; Bao, J.; Du, C.; Wang, J.; Tong, Q.; Liu, C. Schisandrin B suppresses glioma cell metastasis mediated by inhibition of mTOR/MMP-9 signal pathway. Biomed. Pharmacother. 2015, 74, 77–82. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, S.; Liu, W.; Wang, K.; Fan, Y.; Chen, J.; Ma, J.; Wang, X.; He, D.; Zeng, J.; Li, L. Tetrandrine inhibits migration and invasion of human renal cell carcinoma by regulating Akt/NF-κB/MMP-9 signaling. PLoS ONE 2017, 12, e0173725. [Google Scholar] [CrossRef] [PubMed]
  75. Di Pietro, A.; Dayan, G.; Conseil, G.; Steinfels, E.; Krell, T.; Trompier, D.; Baubichon-Cortay, H.; Jault, J.M. P-glycoprotein-mediated resistance to chemotherapy in cancer cells: Using recombinant cytosolic domains to establish structure-function relationships. Braz. J. Med. Biol. Res. 1999, 32, 925–939. [Google Scholar] [CrossRef] [PubMed]
  76. Nikolaou, M.; Pavlopoulou, A.; Georgakilas, A.G.; Kyrodimos, E. The challenge of drug resistance in cancer treatment: A current overview. Clin. Exp. Metastasis 2018, 35, 309–318. [Google Scholar] [CrossRef] [PubMed]
  77. Mansoori, B.; Mohammadi, A.; Davudian, S.; Shirjang, S.; Baradaran, B. The different mechanisms of cancer drug resistance: A brief review. Adv. Pharm. Bull. 2017, 7, 339–348. [Google Scholar] [CrossRef]
  78. Siccardi, D.; Mumy, K.L.; Wall, D.M.; Bien, J.D.; McCormick, B.A. Salmonella enterica serovar Typhimurium modulates P-glycoprotein in the intestinal epithelium. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 294, G1392–G1400. [Google Scholar] [CrossRef] [PubMed]
  79. Chiu, L.Y.; Hu, M.E.; Yang, T.Y.; Hsin, I.L.; Ko, J.L.; Tsai, K.J.; Sheu, G.T. Immunomodulatory protein from Ganoderma microsporum induces pro-death autophagy through Akt-mTOR-p70S6K pathway inhibition in multidrug resistant lung cancer cells. PLoS ONE 2015, 10, e0125774. [Google Scholar] [CrossRef] [PubMed]
  80. Wang, L.; Wang, C.; Jia, Y.; Liu, Z.; Shu, X.; Liu, K. Resveratrol increases anti-proliferative activity of Bestatin through downregulating P-Glycoprotein expression via inhibiting PI3K/Akt/mTOR pathway in K562/ADR cells. J. Cell Biochem. 2016, 117, 1233–1239. [Google Scholar] [CrossRef]
  81. Mercado-Lubo, R.; Zhang, Y.; Zhao, L.; Rossi, K.; Wu, X.; Zou, Y.; Castillo, A.; Leonard, J.; Bortell, R.; Greiner, D.L.; et al. A Salmonella nanoparticle mimic overcomes multidrug resistance in tumours. Nat. Commun. 2016, 7, 12225. [Google Scholar] [CrossRef] [PubMed]
  82. Jia, L.J.; Wei, D.P.; Sun, Q.M.; Jin, G.H.; Li, S.F.; Huang, Y.; Hua, Z.C. Tumor-targeting Salmonella typhimurium improves cyclophosphamide chemotherapy at maximum tolerated dose and low-dose metronomic regimens in a murine melanoma model. Int. J. Cancer 2007, 121, 666–674. [Google Scholar] [CrossRef]
  83. Bascuas, T.; Moreno, M.; Grille, S.; Chabalgoity, J.A. Salmonella immunotherapy improves the outcome of CHOP chemotherapy in non-Hodgkin lymphoma-bearing mice. Front. Immunol. 2018, 9, 7. [Google Scholar] [CrossRef] [PubMed]
  84. Binder, D.C.; Arina, A.; Wen, F.; Tu, T.; Zhao, M.; Hoffman, R.M.; Wainwright, D.A.; Schreiber, H. Tumor relapse prevented by combining adoptive T cell therapy with Salmonella typhimurium. Oncoimmunology 2016, 5, e1130207. [Google Scholar] [CrossRef] [PubMed]
  85. Chen, W.; Wang, Y.; Qin, M.; Zhang, X.; Zhang, Z.; Sun, X.; Gu, Z. Bacteria-driven hypoxia targeting for combined biotherapy and photothermal therapy. ACS Nano 2018, 12, 5995–6005. [Google Scholar] [CrossRef] [PubMed]
  86. Zhu, H.; Cheng, P.; Chen, P.; Pu, K. Recent progress in the development of near-infrared organic photothermal and photodynamic nanotherapeutics. Biomater. Sci. 2018, 6, 746–765. [Google Scholar] [CrossRef] [PubMed]
  87. Toso, J.F.; Gill, V.J.; Hwu, P.; Marincola, F.M.; Restifo, N.P.; Schwartzentruber, D.J.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Stock, F.; et al. Phase I study of the intravenous administration of attenuated Salmonella typhimurium to patients with metastatic melanoma. J. Clin. Oncol. 2002, 20, 142–152. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, C.Z.; Kazmierczak, R.A.; Eisenstark, A. Strains, Mechanism, and Perspective: Salmonella-Based Cancer Therapy. Int. J. Microbiol. 2016, 5678702. [Google Scholar] [CrossRef]
  89. Park, S.H.; Zheng, J.H.; Nguyen, V.H.; Jiang, S.N.; Kim, D.Y.; Szardenings, M.; Min, J.H.; Hong, Y.; Choy, H.E.; Min, J.J. RGD Peptide cell-surface display enhances the targeting and therapeutic efficacy of attenuated salmonella-mediated cancer therapy. Theranostics 2016, 6, 1672–1682. [Google Scholar] [CrossRef] [PubMed]
  90. Zheng, J.H.; Min, J.J. Targeted Cancer Therapy Using Engineered Salmonella typhimurium. Chonnam Med. J. 2016, 52, 173–184. [Google Scholar] [CrossRef] [PubMed]
  91. Dai, Y.; Toley, B.J.; Swofford, C.A.; Forbes, N.S. construction of an inducible cell-communication system that amplifies Salmonella gene expression in tumor tissue. Biotechnol. Bioeng. 2013, 110, 1769–1781. [Google Scholar] [CrossRef] [PubMed]
  92. Gunn, J.S.; Ernst, R.K.; McCoy, A.J.; Miller, S.I. Constitutive Mutations of the Salmonella enterica Serovar Typhimurium Transcriptional Virulence Regulator phoP. Infect. Immun. 2000, 68, 3758–3762. [Google Scholar] [CrossRef]
  93. Niethammer, A.G.; Lubenau, H.; Mikus, G.; Knebel, P.; Hohmann, N.; Leowardi, C.; Beckhove, P.; Akhisaroglu, M.; Ge, Y.; Springer, M.; et al. Double-blind, placebo-controlled first in human study to investigate an oral vaccine aimed to elicit an immune reaction against the VEGF-Receptor 2 in patients with stage IV and locally advanced pancreatic cancer. BMC Cancer 2012, 12, 361. [Google Scholar] [CrossRef] [PubMed]
  94. Schmitz-Winnenthal, F.H.; Hohmann, N.; Niethammer, A.G.; Friedrich, T.; Lubenau, H.; Springer, M.; Breiner, K.M.; Mikus, G.; Weitz, J.; Ulrich, A.; et al. Anti-angiogenic activity of VXM01, an oral T-cell vaccine against VEGF receptor 2, in patients with advanced pancreatic cancer: A randomized, placebo-controlled, phase 1 trial. Oncoimmunology 2015, 4, e1001217. [Google Scholar] [CrossRef] [Green Version]
  95. Schmitz-Winnenthal, F.H.; Hohmann, N.; Schmidt, T.; Podola, L.; Friedrich, T.; Lubenau, H.; Springer, M.; Wieckowski, S.; Breiner, K.M.; Mikus, G.; et al. A phase 1 trial extension to assess immunologic efficacy and safety of prime-boost vaccination with VXM01, an oral T cell vaccine against VEGFR2, in patients with advanced pancreatic cancer. Oncoimmunology 2018, 7, e1303584. [Google Scholar] [CrossRef] [PubMed]
  96. U.S. National Institutes of Health; Clinicaltrials.gov. Multiple Myeloma Trial of Orally Administered Salmonella Based Survivin Vaccine (MAPSS). Identifier: NCT03762291. Available online: https://clinicaltrials.gov/ct2/show/NCT03762291 (accessed on 23 April 2019).
Figure 1. Salmonella-mediated tumor immunotherapy. Salmonella triggers immune cells infiltration into tumor tissue and coax tumor cells self-destruction while preventing tumor microvascularization and delaying tumor cell migration.
Figure 1. Salmonella-mediated tumor immunotherapy. Salmonella triggers immune cells infiltration into tumor tissue and coax tumor cells self-destruction while preventing tumor microvascularization and delaying tumor cell migration.
Biomedicines 07 00036 g001
Figure 2. Salmonella enhances gap intercellular communication (GJIC) via gap junctions and alters membrane permeability to re-sensitize drug-resistant tumor cells.
Figure 2. Salmonella enhances gap intercellular communication (GJIC) via gap junctions and alters membrane permeability to re-sensitize drug-resistant tumor cells.
Biomedicines 07 00036 g002
Table 1. A summary of the tumor cell signaling targets of Salmonella-mediated tumor immunotherapy.
Table 1. A summary of the tumor cell signaling targets of Salmonella-mediated tumor immunotherapy.
Protein TargetsControlMechanismsRef.
Connexin 43 Increases tumor cell chemosensitivity [41]
IDOSuppresses tumor immune tolerance[43,45]
Beclin-1 and LC3Induce autophagy [46]
Cleaved Caspase-3Activates apoptosis[46]
HIF-1/VEGFInhibits angiogenesis [49]
Matrix MMP-9Delays cell migration and metastasis[50]
P-glycoprotein Increases tumor cell chemosensitivity[51]
Note: ▲-Upregulation; ▼-downregulation.

Share and Cite

MDPI and ACS Style

Pangilinan, C.R.; Lee, C.-H. Salmonella-Based Targeted Cancer Therapy: Updates on A Promising and Innovative Tumor Immunotherapeutic Strategy. Biomedicines 2019, 7, 36. https://doi.org/10.3390/biomedicines7020036

AMA Style

Pangilinan CR, Lee C-H. Salmonella-Based Targeted Cancer Therapy: Updates on A Promising and Innovative Tumor Immunotherapeutic Strategy. Biomedicines. 2019; 7(2):36. https://doi.org/10.3390/biomedicines7020036

Chicago/Turabian Style

Pangilinan, Christian Ronquillo, and Che-Hsin Lee. 2019. "Salmonella-Based Targeted Cancer Therapy: Updates on A Promising and Innovative Tumor Immunotherapeutic Strategy" Biomedicines 7, no. 2: 36. https://doi.org/10.3390/biomedicines7020036

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

Article Metrics

Back to TopTop