Anna Sokolovska’s research while affiliated with Synlogic, Inc. and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (14)


Activation of HIF-1α by lactate inhibits the pro-inflammatory activities of DCs
a–c, Ingenuity pathway analysis (IPA) (a), hypoxia score (b) and Hif1a expression (c) in total DCs from the CNS of EAE mice. d, Expression of HIF-1α in DCs isolated from the CNS, draining lymph nodes (dLNs) and spleen of wild-type mice before (‘naive’, n = 3), 12 days (‘onset’) or 17 days (‘peak’) after EAE induction (n = 4 for CNS peak and spleen onset, n = 5 otherwise). e, EAE in wild-type (WT) (n = 15) and HIF-1αItgax (n = 9) mice. Experiment repeated three times. f, IFNγ⁺, IL-17⁺ and IFNγ⁺IL-17⁺GM-CSF⁺ CD4⁺ T cells in the CNS of wild-type and HIF-1αItgax mice 15 days after EAE induction (n = 5 mice per group). g, RNA sequencing (RNA-seq) analysis of splenic DCs at peak EAE (n = 5 mice per group). h,i, IPA (h) and GSEA for the human phenotype ontology term ‘abnormal activity of mitochondrial respiratory chain’ (i) in wild-type and HIF-1αItgax splenic DCs at peak EAE. NES, normalized enrichment score. j, Mean fluorescence intensity (MFI) of HIF-1α expression in splenic DCs treated with metabolites and LPS for 6 h, normalized to LPS stimulation. DMF, dimethyl fumarate; Pyr, pyruvate; Mal, malonate; Succ, succinate; 4-OI, 4-octyl itaconate; α-KG, α-ketoglutarate; L-LA, l-sodium lactate (n = 4 per condition). k, HIF-1α⁺ DCs after treatment with L-LA (0.1, 1 and 10 mM) and LPS for 6 h (n = 3 for medium, L-LA 0.1 mM, L-LA 10 mM, LPS + L-LA 10 mM, n = 4 otherwise). l, mRNA expression in wild-type BMDCs after treatment with LPS, L-LA, GPR81 antagonist or MCT1 inhibitor (n = 3 per group). m, Ifng, Il17a and Csf2 expression in 2D2⁺CD4⁺ T cells co-cultured with wild-type BMDCs pre-stimulated with L-LA and LPS (n = 4 for Il17a LPS + L-LA 0.1 mM, n = 3 otherwise). n, IFNG and IL17A expression in human T cells co-cultured with DCs (n = 3 per group). siNT, non-targeting siRNA. Statistical analysis was performed using one-way ANOVA with Dunnett’s or Šídák’s post-hoc test for selected multiple comparisons for d,k,l–n, unpaired Student’s t-test for f and two-way ANOVA for e. Data are mean ± s.e.m.
Source data
HIF-1α-induced NDUFA4L2 limits the production of mtROS
a, OCR in wild-type and HIF-1αItgax BMDCs after stimulation with L-LA (n = 7 for WT, n = 3 for WT + L-LA, n = 4 otherwise). b, ATP levels in BMDCs as a result of 1 mM L-LA for 6 h (n = 3 per group). c, Expression of HIF-1α target genes in BMDCs stimulated with LPS under normoxic or hypoxic conditions (n = 3 per group). d, Ndufa4l2 expression in wild-type and HIF-1αItgax splenic DCs at EAE peak stimulation (n = 4 for WT, n = 5 for HIF-1αItgax). e, Ndufa4l2 expression after 6 h of treatment with LPS + L-LA, LPS + D-LA or LPS (n = 8 for vehicle, n = 6 for LPS, n = 3 otherwise). f, Transactivation of the Ndufa4l2 promoter after overexpression of HIF-1α (n = 7 per group). g, Effect of Ndufa4l2 overexpression in LPS-stimulated BMDCs (n = 3). h–k, Effect of Ndufal2 knockdown on OCR (h,i), mtROS production (j) and gene expression (k) in BMDCs treated with LPS, LPS + L-LA or LPS + D-LA (n = 3 per group). l, mRNA expression in BMDCs treated with LPS, mitoPQ or mitoTempo for 6 h (n = 3 per group). Statistical analysis was performed using one-way ANOVA with Tukey’s, Dunnett’s or Šídák’s post-hoc test for selected multiple comparisons for a,e,i–l, two-way ANOVA for g and unpaired Student’s t-test for b,d,f. Data are mean ± s.e.m.
Source data
L-LA limits the mtROS-driven activation of XBP1 in DCs
a, Spliced Xbp1 mRNA normalized to total Xbp1 mRNA (sXbp1/Xbp1 ratio) in BMDCs isolated from wild-type mice and treated with LPS (n = 6), LPS + L-LA (n = 5) or LPS + L-LA + mitoPQ (n = 3) for 6 h. b, sXPB1/XBP1 mRNA ratio in human DCs treated with LPS, LPS + D-LA or LPS + L-LA for 6 h (n = 5 per group). c–e, XBP1-binding sites (top) and recruitment of XBP1 to Il1b (c), Il6 (d) and Il23a (e) promoters in BMDCs isolated from wild-type mice determined by chromatin immunoprecipitation (ChIP) after treatment with LPS, LPS + D-LA or LPS + L-LA for 6 h (n = 4 for Il1b LPS + D-LA, n = 3 otherwise). f, sXbp1/Xbp1 mRNA ratio in wild-type (n = 5) and HIF-1αItgax (n = 5) splenic DCs isolated at peak EAE. g, sXbp1/Xbp1 mRNA ratio in wild-type BMDCs transfected with either an empty plasmid (n = 5) or an Ndufa4l2-overexpression plasmid (n = 5). h, EAE development in wild-type (n = 15) and XBP1Itgax (n = 15) mice. Experiment repeated three times. i, IFNγ⁺, IFNγ⁺IL-17⁺ and IL-17⁺ CD4⁺ T cells isolated from the CNS of wild-type (n = 5–6) and XBP1Itgax (n = 5) EAE mice. j,k, Cytokine production (j) and proliferative recall response to ex vivo MOG35–55 restimulation for 72 h (k) of splenocytes from EAE wild-type and Xbp1Itgax mice (n = 6 for IFNγ and IL-10 WT, n = 5 mice otherwise). CPM, counts per million. l,m, IPA (l) and heat map (m) of RNA-seq analysis of wild-type (n = 5) and Xbp1Itgax (n = 5) splenic DCs isolated 30 days after EAE induction. Statistical analysis was performed using one-way ANOVA with Dunnett’s or Šídák’s post-hoc test for selected multiple comparisons for a–e, unpaired Student’s t-test for f,g,i,j and two-way ANOVA for h,k. Data are mean ± s.e.m.
Source data
Activating HIF-1α–NDUFA4L2 with engineered probiotics ameliorates EAE
a, Engineered EcNLac strain. b, Concentrations of D-LA and L-LA in culture supernatants after EcNLac or EcN incubation at 37 °C (n = 3 per group). c, Concentration of D-LA in small-intestinal tissue after daily administration of EcN (n = 3 at 1 h and 24 h, n = 5 at 4 h) or EcNLac (n = 4 at 1 h and 24 h, n = 5 at 4 h) for a week compared to untreated controls (n = 5 mice). d,e, Immunostaining analysis of HIF-1α, NDUFA4L2 and spliced XBP1 (d) and quantification (e) in CD11c⁺ cells from the small intestine after daily administration of EcNLac or EcN for a week (n = 5 mice per group). Data quantified from tile scan images of full tissue sections from each mouse. Experiment repeated twice. AU, arbitrary units. f, EAE in wild-type and HIF-1αItgax mice treated with EcN (n = 5 WT, 5 HIF-1αItgax mice) or EcNLac (n = 19 WT, 4 HIF-1αItgax mice). Experiment repeated three times. g, Quantification of IFNγ⁺, IL-17⁺ and IFNγ⁺IL-17⁺ CD4⁺ T cells in the CNS of mice from f (n = 5 for IL-17 WT EcN, n = 4 for WT EcNLac, n = 3 otherwise). h, Quantification of IFNγ⁺ and IL-17⁺ CD4⁺ T cells in the small intestine of wild-type mice treated with EcN (n = 8) or EcNLac (n = 9) after EAE induction. i,j, sXbp1/Xbp1 mRNA ratio (i) and Il1b, Il6, Il23a and Tnf mRNA expression (j) in small-intestinal DCs from EcN- or EcNLac-treated mice (n = 4 for Il23a and Tnf, n = 3 otherwise). k, Representative dot plot of Kaede photoconversion in CD4⁺ T cells isolated from the spleen. l, Total, IFNγ⁺ and IL-17⁺ CD4⁺ T cells photoconverted in the spleen of wild-type mice treated with EcN (n = 5) or EcNLac (n = 4). Statistical analysis was performed using two-way ANOVA followed by Šídák’s multiple comparisons test for b,c,f, unpaired Student’s t-test for e,h–j,l and one-way ANOVA with Šídák’s post-hoc test for selected multiple comparisons for g. Data are mean ± s.e.m.
Source data
Lactate limits CNS autoimmunity by stabilizing HIF-1α in dendritic cells
  • Article
  • Publisher preview available

August 2023

·

404 Reads

·

74 Citations

Nature

·

Joseph M. Rone

·

·

[...]

·

Francisco J. Quintana

Dendritic cells (DCs) have a role in the development and activation of self-reactive pathogenic T cells1,2. Genetic variants that are associated with the function of DCs have been linked to autoimmune disorders3,4, and DCs are therefore attractive therapeutic targets for such diseases. However, developing DC-targeted therapies for autoimmunity requires identification of the mechanisms that regulate DC function. Here, using single-cell and bulk transcriptional and metabolic analyses in combination with cell-specific gene perturbation studies, we identify a regulatory loop of negative feedback that operates in DCs to limit immunopathology. Specifically, we find that lactate, produced by activated DCs and other immune cells, boosts the expression of NDUFA4L2 through a mechanism mediated by hypoxia-inducible factor 1α (HIF-1α). NDUFA4L2 limits the production of mitochondrial reactive oxygen species that activate XBP1-driven transcriptional modules in DCs that are involved in the control of pathogenic autoimmune T cells. We also engineer a probiotic that produces lactate and suppresses T cell autoimmunity through the activation of HIF-1α–NDUFA4L2 signalling in DCs. In summary, we identify an immunometabolic pathway that regulates DC function, and develop a synthetic probiotic for its therapeutic activation.

View access options

Phase I Study of SYNB1891, an Engineered E. coli Nissle Strain Expressing STING Agonist, with and without Atezolizumab in Advanced Malignancies

May 2023

·

82 Reads

·

71 Citations

Clinical Cancer Research

Purpose: SYNB1891 is a live, modified strain of the probiotic E. coli Nissle 1917 (EcN) engineered to produce cyclic dinucleotides under hypoxia leading to stimulator of interferon genes (STING)-activation in phagocytic antigen-presenting cells in tumors and activating complementary innate immune pathways. Patients and methods: This first-in-human study (NCT04167137) enrolled participants with refractory advanced cancers to receive repeat intratumoral (IT) injections of SYNB1891 either alone or in combination atezolizumab, with the primary objective of evaluating the safety and tolerability of both regimens. Results: Twenty-four participants received monotherapy across 6 cohorts, and 8 participants received combination therapy in 2 cohorts. Five cytokine release syndrome events occurred with monotherapy, including one that met the criteria for dose-limiting toxicity at the highest dose; no other SYNB1891-related serious adverse events occurred, and no SYNB1891-related infections were observed. SYNB1891 was not detected in the blood at 6 or 24h after the first IT dose or in tumor tissue 7 days following the first dose. Treatment with SYNB1891 resulted in activation of the STING pathway and target engagement as assessed by upregulation of interferon-stimulated genes, chemokines/cytokines, and T-cell response genes in core biopsies obtained pre-dose and 7 days following the third weekly dose. In addition, a dose‑related increase in serum cytokines was observed, as well as stable disease in 4 participants refractory to prior PD-1/L1 antibodies. Conclusions: Repeat IT injection of SYNB1891 as monotherapy and in combination atezolizumab was safe and well tolerated and evidence of STING pathway target engagement was observed.


Engineered probiotics limit CNS autoimmunity by stabilizing HIF-1α in dendritic cells

March 2023

·

182 Reads

·

2 Citations

Dendritic cells (DCs) control the generation of self-reactive pathogenic T cells. Thus, DCs are considered attractive therapeutic targets for autoimmune diseases. Using single-cell and bulk transcriptional and metabolic analyses in combination with cell-specific gene perturbation studies we identified a negative feedback regulatory pathway that operates in DCs to limit immunopathology. Specifically, we found that lactate, produced by activated DCs and other immune cells, boosts NDUFA4L2 expression through a mechanism mediated by HIF-1a. NDUFA4L2 limits the production of mitochondrial reactive oxygen species that activate XBP1-driven transcriptional modules in DCs involved in the control of pathogenic autoimmune T cells. Moreover, we engineered a probiotic that produces lactate and suppresses T-cell autoimmunity in the central nervous system via the activation of HIF-1α/NDUFA4L2 signaling in DCs. In summary, we identified an immunometabolic pathway that regulates DC function, and developed a synthetic probiotic for its therapeutic activation.


Engineered microbes for cancer immunotherapy

January 2022

·

41 Reads

The field of cancer immunotherapy began with the injection of pathogenic bacteria intratumorally, but the broad adoption of bacteria in cancer therapeutics was historically limited by difficulty finding a balance between efficacy and safety. Synthetic biology now provides tools to precisely engineer bacteria to address some of the limitations of currently available immunotherapies by genetically modifying bacteria to express one or more immune effectors and deliver them to the tumor microenvironment. Multiple approaches have been developed preclinically, and several have progressed to early phase clinical trials. We describe the available synthetic biology toolkit as well as the current understanding of immune effectors that can be engineered into bacteria. Finally, we describe some of the regulatory considerations for developing engineered microbes for cancer immunotherapy.


500 SYNB1891, a bacterium engineered to produce a STING agonist, demonstrates target engagement in humans following intratumoral injection

November 2021

·

82 Reads

·

5 Citations

Background SYNB1891 is a live, modified strain of probiotic E. coli Nissle engineered to produce cyclic dinucleotides under hypoxia leading to stimulator of interferon genes (STING)-activation in phagocytic antigen-presenting cells in tumors and activating complementary innate immune pathways. Methods This first-in-human study ( NCT04167137 ) enrolled patients with refractory advanced solid tumors to receive intratumoral (IT) injections of SYNB1891 monotherapy or in combination atezolizumab. Patients enrolled in the monotherapy arms received doses of 1x10 ⁶ - 3x10 ⁸ live cells on Days 1, 8 and 15 of the first 21-day cycle and then on Day 1 of each subsequent cycle. Patients enrolled in the 2 combination cohorts received doses of 1x10 ⁷ - 3x10 ⁷ live cells in combination with atezolizumab administered on a 21-day cycle. The primary objective of the study was to evaluate safety and tolerability of SYNB1891 alone and in combination with atezolizumab. Other objectives include SYNB1891 kinetics in blood and injected tumor, STING-target engagement as assessed by IT gene expression and serum cytokines, and tumor responses. Results This interim analysis includes 23 patients across 6 monotherapy cohorts dosed at 1x10 ⁶ , 3x10 ⁶ , 1x10 ⁷ , or 3x10 ⁷ , 1x10 ⁸ and 3x10 ⁸ live cells, and 7 patients dosed in 2 combination therapy cohorts (1x10 ⁷ and 3x10 ⁷ live cells). The mean (range) age was 61 (25–82); 19 patients were female. There were 4 cytokine release syndrome events in monotherapy cohorts, including one grade 3 event which met the criterion for dose limiting toxicity at 3x10 ⁸ live cells; there were no other SYNB1891-related serious adverse events. There were no SYNB1891-related infections. SYNB1891 was not detected in the blood at 6 or 24 hours after the first dose or intratumorally 7 days following the first dose. Treatment with SYNB1891 demonstrated activation of the STING pathway and target engagement as assessed by upregulation of interferon-stimulated genes (ISG15, IFIT1, IFIt2), chemokines/cytokines (CXCL9, CXCL10, TNFRS18, TNFSF10) and T-cell response genes (GZMA, CD4, PD-L2) in core biopsies obtained pre-dose and 7 days following the third weekly dose. In addition, there was a dose-response increase in serum cytokines. Durable, stable disease was observed in two patients treated with SYNB1891 monotherapy refractory to prior PD-1/L1 antibodies with vulvar melanoma (1x10 ⁶ live cells; RECIST -28%) and small cell lung cancer (1x10 ⁷ live cells; RECIST -12%). Conclusions Repeat IT injection of SYNB1891 as monotherapy and in combination atezolizumab in this ongoing study is safe and well-tolerated up to at least 1x10 ⁸ live cells, and shows evidence of STING pathway target engagement. Acknowledgements We thank Inessa Vulfova for her clinical support in conduct of this study. Trial Registration clinicaltrials.gov ( NCT04167137 ) Ethics Approval The study protocol, the informed consent form (ICF), and printed subject information materials were reviewed and approved by the institutional review board (IRB) at the investigational site before any study procedures were performed. Written informed consent to participate in the study was obtained from each subject before any study-specific procedures were performed.The Ohio State University Cancer Institutional Review Board; Approval ID: 2020C0194MD Anderson Cancer Center Institutional Review Board; Approval ID: 2019–0576Mary Crawley Medical Research Center Institutional Review Board; Approval ID: 19–31 SYNB1891-CP-001North Texas Institutional Review Board; Approval ID: 2019.040WIRB Approval ID: 20192779University of Pittsburgh Institutional Review Board Approval ID: STUDY20010116


Metabolic modulation of tumours with engineered bacteria for immunotherapy

October 2021

·

510 Reads

·

386 Citations

Nature

The availability of l-arginine in tumours is a key determinant of an efficient anti-tumour T cell response1–4. Consequently, increases of typically low l-arginine concentrations within the tumour may greatly potentiate the anti-tumour responses of immune checkpoint inhibitors, such as programmed death-ligand 1 (PD-L1)-blocking antibodies⁵. However, currently no means are available to locally increase intratumoural l-arginine levels. Here we used a synthetic biology approach to develop an engineered probiotic Escherichia coli Nissle 1917 strain that colonizes tumours and continuously converts ammonia, a metabolic waste product that accumulates in tumours⁶, to l-arginine. Colonization of tumours with these bacteria increased intratumoural l-arginine concentrations, increased the number of tumour-infiltrating T cells and had marked synergistic effects with PD-L1 blocking antibodies in the clearance of tumours. The anti-tumour effect of these bacteria was mediated by l-arginine and was dependent on T cells. These results show that engineered microbial therapies enable metabolic modulation of the tumour microenvironment leading to enhanced efficacy of immunotherapies.


Engineering living therapeutics with synthetic biology

October 2021

·

240 Reads

·

232 Citations

Nature Reviews Drug Discovery

The steadfast advance of the synthetic biology field has enabled scientists to use genetically engineered cells, instead of small molecules or biologics, as the basis for the development of novel therapeutics. Cells endowed with synthetic gene circuits can control the localization, timing and dosage of therapeutic activities in response to specific disease biomarkers and thus represent a powerful new weapon in the fight against disease. Here, we conceptualize how synthetic biology approaches can be applied to programme living cells with therapeutic functions and discuss the advantages that they offer over conventional therapies in terms of flexibility, specificity and predictability, as well as challenges for their development. We present notable advances in the creation of engineered cells that harbour synthetic gene circuits capable of biological sensing and computation of signals derived from intracellular or extracellular biomarkers. We categorize and describe these developments based on the cell scaffold (human or microbial) and the site at which the engineered cell exerts its therapeutic function within its human host. The design of cell-based therapeutics with synthetic biology is a rapidly growing strategy in medicine that holds great promise for the development of effective treatments for a wide variety of human diseases.


Abstract CT110: Intratumoral injection of SYNB1891, a synthetic biotic designed to activate the innate immune system, demonstrates target engagement in humans including intratumoral STING activation

July 2021

·

5 Reads

·

5 Citations

Cancer Research

Introduction: SYNB1891 is a live, modified strain of the probiotic E. coli Nissle engineered to produce cyclic dinucleotides under hypoxia leading to stimulator of interferon genes (STING)-activation in phagocytic antigen-presenting cells in tumors and activating complementary innate immune pathways. Methods: This first-in-human study (NCT04167137) is enrolling patients with refractory advanced solid tumors or lymphoma to receive an intratumoral (IT) injection of SYNB1891 on Days 1, 8 and 15 of the first 21-day cycle and then on Day 1 of each subsequent cycle. Dose escalation is planned across 7 cohorts (1x106 - 1x109 live cells) with Arm 1 consisting of escalating doses of SYNB1891 as monotherapy, and Arm 2 in combination with atezolizumab. The primary objective is to determine the single-agent maximum tolerated dose as monotherapy and the recommended Phase 2 dose in combination with atezolizumab. Other objectives include SYNB1891 kinetics in blood and the injected tumor, STING-target engagement as assessed by IT gene expression and serum cytokines, and tumor responses. Results: This interim analysis includes the first 11 patients across 4 cohorts dosed at 1x106, 3x106, 1x107, or 3x107 live cells, with a total of 59 doses administered. The mean (range) age was 56 (25-70); 9 patients were female. There were no dose-limiting toxicities, SYNB1891-related infections or discontinuations due to adverse events. There was one SYNB1891-related serious adverse event in a patient who experienced a grade 2 cytokine release syndrome that resolved within one day, and one patient experienced a grade 2 injection site reaction of erythema which resolved. SYNB1891 was not detected in the blood at 6 or 24 hours after the first dose or intratumorally 7 days following the first dose. Treatment with SYNB1891 demonstrated activation of the STING pathway and target engagement as assessed by upregulation of interferon-stimulated genes (ISG15, IFIT1, IFIt2), chemokines/cytokines (CXCL9, CXCL10, TNFRS18, TNFSF10) and T-cell response genes (GZMA, CD4, PD-L2) in core biopsies obtained pre-dose and 7 days following the third weekly dose. In addition, there was a dose-response increase in serum cytokines at dose levels of 1x107 and 3x107 live cells (IL-6, TNFα, IFNγ, IL-1Ra) at 6 hours post-dose. Durable, stable disease was observed in two patients refractory to prior PD-1/L1 antibodies with vulvar melanoma (1x106 live cells) and small cell lung cancer (1x107 live cells). Conclusions: Repeat IT injection of SYNB1891 as monotherapy is safe and well-tolerated up to at least 3x107 cells and shows evidence of STING pathway target engagement. These data support the continued dose escalation of SYNB1891 as monotherapy, and initiation of Arm 2 in combination with atezolizumab. Citation Format: Filip Janku, Jason J. Luke, Aoife Brennan, Richard Riese, Mary Varterasian, Michael B. Armstrong, Karen L. Kuhn, Anna Sokolovska, James F. Strauss. Intratumoral injection of SYNB1891, a synthetic biotic designed to activate the innate immune system, demonstrates target engagement in humans including intratumoral STING activation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT110.


Metabolic modulation of tumors with engineered bacteria for immunotherapy

May 2021

·

865 Reads

The availability of L-arginine in tumors is a key determinant of an efficient anti-tumor T cell response. Consequently, elevation of typically low L-arginine levels within the tumor may greatly potentiate the anti-tumor responses of immune checkpoint inhibitors, such as PD-L1 blocking antibodies. However, currently no means are available to locally increase intra-tumoral L-arginine levels. Here, we used a synthetic biology approach to develop an engineered probiotic Escherichia coli Nissle 1917 strain that colonizes tumors and continuously converts ammonia, a metabolic waste product that accumulates in tumors4, into L-arginine. Colonization of tumors with these bacteria elevated intra-tumoral L-arginine concentrations, increased the amount of tumor-infiltrating T cells, and had striking synergistic effects with PD-L1 blocking antibodies in the clearance of tumors. The anti-tumor effect of the living therapeutic was mediated by L-arginine and was dependent on T cells. These results show that engineered microbial therapies enable metabolic modulation of the tumor microenvironment leading to enhanced efficacy of immunotherapies.



Citations (8)


... Inhibition of PHD activity by lactate stabilizes HIF1α, leading to exacerbation of glycolysis [7]. A recent study found that lactate can upregulate the expression of NDUFA4L2 (NADH dehydrogenase (ubiquinone)-1α subcomplex 4-like 2) through a transcription factor HIF1α mediated mechanism [8]. The information suggested that the stabilized HIF1α/β heterodimer activates gene transcription pathways involved in angiogenesis and anaerobic glycolysis [9]. ...

Reference:

Lactate induces oxidative stress by HIF1α stabilization and circadian clock disturbance in mammary gland of dairy cows
Lactate limits CNS autoimmunity by stabilizing HIF-1α in dendritic cells

Nature

... The application of STING agonists is additionally constrained by their lack of specificity. Presently, nearly all available STING agonists do not exhibit specificity for tumors and may inadvertently activate STING in a non-specific manner across the organism, potentially resulting in "on-target off-tumor" toxicity; furthermore, the intravenous or intraperitoneal delivery of STING agonists could induce cytokine storms (Luke et al. 2023). Consequently, it will be crucial to recognize STING agonists that are specific to tumor tissues, as well as to optimize the dosages and timing in order to prevent excessive activation of the STING pathway, thereby avoiding the deterioration of clinical outcomes. ...

Phase I Study of SYNB1891, an Engineered E. coli Nissle Strain Expressing STING Agonist, with and without Atezolizumab in Advanced Malignancies
  • Citing Article
  • May 2023

Clinical Cancer Research

... F10 melanoma tumorbearing mice induced the production of type I IFNs with a concomitant significant reduction in tumor growth eight days after treatment. Phase I clinical trials of SYNB 1981 are currently ongoing in participants with advanced/metastatic solid tumors and lymphoma (NCT04167137) to identify the antitumor efficacy of intratumoral SYNB1891 as both a monotherapy and in combination with atezolizumab (Riese et al. 2021). ...

500 SYNB1891, a bacterium engineered to produce a STING agonist, demonstrates target engagement in humans following intratumoral injection

... Precise and programmable regulation of multiple gene expression is increasingly crucial for a wide range of applications in biotechnology (1). Over the past decade, the advancements in genetic engineering tools have greatly facilitated the notable progress of synthetic biology applications in mammalian cells (2)(3)(4)(5)(6). In transcriptional regulation, transcription factors can be controlled by specific molecule ligands to bind to or dissociate from target doublestranded DNA, leading to the activation or inhibition of the transcriptional process. ...

Engineering living therapeutics with synthetic biology
  • Citing Article
  • October 2021

Nature Reviews Drug Discovery

... Furthermore, cGAS/STING axis plays a pivotal role in the antitumor efficacy of LGG and the initiation of type I IFN. The cGAS-STING-TBK1-IRF7-IFN-β cascade activates a robust adaptive immune response to LGG in DCs, which is essential for the amplification of immune surveillance against neoplastic growth and establishment of an antiviral state [13] . ...

Metabolic modulation of tumours with engineered bacteria for immunotherapy

Nature

... Targeting the cytosolic DNA sensor cGMP-AMP synthase (cGAS) and its downstream effector, the stimulator of interferon (IFN) genes (STING), within antigen-presenting cells (APCs) is one therapeutic strategy for generating tumor-specific T cell immunity 5-7 . Several STING agonists are currently in clinical development with varying degrees of success [8][9][10][11] at present, no ADCs in advanced development or approved for clinical use at this time specifically target cytosolic immune sensors or are designed to enhance APC functionalities. ...

Abstract CT110: Intratumoral injection of SYNB1891, a synthetic biotic designed to activate the innate immune system, demonstrates target engagement in humans including intratumoral STING activation
  • Citing Conference Paper
  • July 2021

Cancer Research

... Our choice of EcN as a delivery vehicle is due to its genetic malleability, probiotic status, distinction from pathogenic E. coli strains, and nonpathogenic nature. 6,22,51,52 We engineered EcN to express anti-spike nanobodies using Intimin and Lpp-OmpA as surface anchors, ensuring efficient antigen presentation, direct interaction with host cells, and nanobodymediated neutralization. 53 These nanobodybearing EcN constructs successfully inhibited the interaction of spike protein-expressing pseudoviruses with the ACE2 receptor. ...

Immunotherapy with engineered bacteria by targeting the STING pathway for anti-tumor immunity

... Synthetic biology is increasingly seeking to modulate, control and even design (Alnahhas et al., 2020) the microbial consortia that form microbiomes. Synthetic organisms can be used as medicines, engineered to perform metabolic functions that once ingested could treat disease (West et al., 2018). Physiological processes are connected such that alterations to biological activity are relayed to the brain to induce behavioral changes, when necessary. ...

Abstract 2920: Metabolic modulation of the tumor microenvironment using Synthetic Biotic™ Medicines
  • Citing Article
  • July 2018

Cancer Research