Description

The increasing amounts of diagnosed breast cancer have attracted public attention globally.1-3 There are millions of people diagnosed with different subtypes of breast cancers, which represents the abnormal growth of epithelium cells of the ducts or lobules in the glandular tissue of the breast.4-6 Some of them are non-invasive that are confined to the duct or lobule with no symptoms. Triple negative breast cancer (TNBC) is the most aggressive subtype with poor prognosis and high risk of recurrence. It can continuously progress and invade the surrounding breast tissues or even local metastasis to the nearby lymph nodes or distant metastasis to other organs.7-9 Because of the lack of specific targets, the treatment of TNBC is highly challenging. We are aimed to develop an immune therapeutic approach that can convert the acidic and hypoxia microenvironment caused by the high glycolytic activity of breast tumor. Figure 1 represents the overview of our project below.

Figure 1. The overview of our project.


We are looking for an approach that can tackle with the acidic and hypoxia tumor microenvironment for efficient immunotherapy of triple negative breast cancer. Our effort is focused on the genetic engineering and chemical modification of E coli Nissle 1917 that can specifically attack on tumor cells and live on lactic acid. The consumption of lactic acid is expected to invert the acidic pH and re-boost macrophage-driven anti-tumor immune responses. Manganese dioxide nanoparticles are used as nano-immunomodulators that can catalyze hydrogen peroxide (H2O2) to produce oxygen (O2) under acidic conditions and promote innate and adaptive immunity.

Glycolytic activity of TNBC cells and macrophage polarization

Reprogramming of cellular metabolism is the emerging hallmark of TNBC that exacerbates proliferation, metastasis and angiogenesis of tumor cells.10The high glycolytic activity of TNBC cells usually causes the accumulation of metabolic byproduct lactic acid and hypoxia tumor microenvironment (TME).11As shown in Figure 2, the acidification of tumor microenvironment can be sensed by tumor associated macrophages (TAMs) through proton-sensing G protein-coupled receptors (GPCRs).12 The tumor acidosis subsequently induces the expression of a transcriptional repressor called ICER in tumor-associated macrophages, which are then converted from pro-inflammatory M1 phenotype into a non-inflammatory M2 phenotype.13 Classical M1 and alternative M2 activation of macrophages represent two dynamic changing states of macrophage activation.14In contrast to M1-type, cytokines released from M2-type macrophages promote but not inhibit the proliferation of contiguous cells.15 M1–M2 polarization of macrophage is usually tightly balanced by transcriptional and posttranscriptional regulatory networks.16 However, an imbalance of macrophage M1–M2 polarization often leads to immunoevasion including not only local metastasis to the nearby lymph nodes but also distant metastasis to other organs.17

Figure 2. Macrophage M1-M2 polarization induced by acidic tumor microenvironment (TME).


Technique challenges and current solution

Triple negative breast cancer (TNBC) is the most dangerous subtype with least favorable outcomes.18 It is characterized by profound clinical features such as high invasiveness, high metastasis, high risk of recurrence, and poor prognosis.19 Because TNBC tumor does not express estrogen receptor (ER), progesterone receptor (PR), or human epidermal growth factor receptor 2 (HER-2), it lacks actionable targets for regular endocrine therapy or HER2 treatment.20 Currently, the treatment of TNBC is still challenging. There is limited responses to chemotherapy. And surgical removal or radiation therapy are also not able to prevent the recurrence. Advances in omics technologies have revealed the dynamics of TNBC microenvironment heterogeneity.21 These new knowledges facilitate the development of novel poly (ADP-ribose) polymerase (PARP) inhibitors22 and antibody–drug conjugates.23 In particular, immune-checkpoint inhibitors (ICI) have been revolutionizing the therapeutic opportunities for early-stage and advanced-stage TNBC patients.24 Recent clinical trials have coupled paclitaxel with immune checkpoint inhibitors in TNBC patients.25 Increased stromal tumor-infiltrating lymphocytes and micronucleation over baseline have been found.26 It shows paclitaxel can induce chromosome missegregation on multipolar spindles during mitosis. Then post-mitotic cells containing micronuclei may activate cyclic GMP-AMP synthase (cGAS) and induce type I IFN responses in the stimulator of IFN genes (STING) pathway.26 As shown in Figure 3, the combination of ICI with paclitaxel promotes the M1 polarization of TAMs and provides a new immunotherapy to treat metastatic TNBC.27 However, while such microtubule-targeting agent potentiates an immune response in TNBC, the acquired resistance to ICI-based therapies has clinically emerged, which demands further development for the immunotherapy of TNBC.28

Figure 3. Macrophage M1 polarization facilitated by the combination of immune checkpoint inhibitors with microtubule-targeting agent paclitaxel.


Our solution: remodeling the acidic and hypoxia tumor microenvironment

Our solution is aimed to rescue tumor associated macrophages (TAMs) and re-boost macrophage driven anti-tumor immune responses through the remodeling of the acidic and hypoxia tumor microenvironment. It is expected to convert TAMs from non-inflammatory back to inflammatory phenotype by adjusting the acidic pH and relieving hypoxia of TME. Because bacteria infection can induce innate and adaptive immune responses,29 we hope combine the advantage of intrinsic antitumor activities of bacteria with nano-immunomodulators and enhanced capability to specifically attack on TNBC cells. E coli Nissle 1917 cells are genetically engineered with SIRPα gene that can recognize CD47 present on surfaces of TNBC cells.30 Engineered bacteria then undergo surface thiolation through a simple one-step imidoester reaction by which primary amino groups on bacterial surface can be converted to free thiols under cytocompatible conditions.31 Gold nanoparticles are deposited on surfaces of hollow manganese dioxide nanospheres that are subsequently bound with thiolated bacteria because of the high affinity of Au nanoparticles with the thiol groups.32 The genetically and chemically modified live tumor-targeting bacteria are named as TuTaBa (兔大巴). As shown in Figure 4, TuTaBa is expected to live on lactic acid that is transported with monocarboxylate transporter 1 (MCT1) and not disturb the normal glucose metabolism. There are great advantages to use manganese dioxide nanospheres as nano-immunomodulators because they have shown lots of biological functions that can directly regulate tumor growth.33 Cancer cells often produce large amounts of H2O2 and GSH for their metabolism and resistance to immunological killings.34 With the peroxidase-like activity, MnO2 nanospheres can effectively catalyze the in situ production of O2 and relieve hypoxia by reacting with endogenous H2O2 to release Mn2+.35 They can also react with GSH in tumor cells to generate GSSH and Mn2+.36 The ability of MnO2 nanospheres to modulate TME allows them to enhance the antitumor immunotherapy. When the mission is completed and the tumor microenvironment (TME) goes back to normal pH value, engineered bacteria undergo clearance program.

Figure 4. The sketch of genetically and chemically modified TuTaBa (tumor targeted bacteria).


Feasibility for the engineering of bacteria carrying with nano-immunomodulators

Genetically or chemically modified bacteria have been recognized as new therapeutic tools that are able to grow in tumors and prevent their proliferation. The feasibility of bacteria-based treatment can be summarized as follows for the immunotherapy of triple negative breast cancer.

✓ Bacteria can initiate the antitumor effects by activating innate and adaptive immune responses or directly killing tumors with secreted toxins.29

✓ With the aid of flagella, live bacteria with the facultative anaerobic property can naturally move toward solid tumors for survival and reproduction in which there are usually insufficient oxygen and low pH. High levels of local colonization of bacteria can be formed there.37

✓ Bacteria can compete with tumors for nutrients that are required for cell metabolism. The colonization of bacteria in the surrounding areas of tumors reduces local nutrient supply. Tumor cells may then die from starvation and suffocation in resultant necrotic regions.38

✓ Bacteria can live on lactic acid that is transported into cells by ubiquitously expressed MCT1. It is responsible for the transport of circulating lactate, pyruvate, and acetoacetic acid and also facilitates the unidirectional proton-linked transport of monocarboxylates across the plasma membrane. The research group supervised by Professor Bernhard Ø. Palsson has systematically investigated the adaptive evolution of the metabolic network of E. Coli strains on lactic acid.39 They have experimentally demonstrated the convergence and reproducible growth of E. coli on lactic acid.40

✓ Overexpression of cluster of differentiation 47 (CD47) has been found in triple negative breast cancer.41 Then it can be used as a marker for engineered bacteria to look for tumor cells. Meanwhile CD47 can also functions as the engulfment signal and dominant macrophage immune checkpoint that represent a signal “do not eat me” to the immune system.42 It has been demonstrated that the interaction of CD47 with its receptor signal-regulatory protein alpha (SIRPα) cause the suppression of phagocytic activities. The expression of SIRPα on bacteria should compete with the SIRPα on macrophage to interact with CD47 on TNBC tumor cells. Then the phagocytic and cytotoxic activities of macrophages against TNBC tumor cells would be enhanced.

✓There are increasing evidences that manganese can regulate immunological responses in different conditions.43 Its role as an alarm protein of innate immunity and the regulation of adaptive immunity have been experimentally proved.44 Under acidic conditions, MnO2 nanospheres can efficiently catalyze the production of O2 by interact with H2O2 to release Mn2+, or interact with the intracellular GSH to release Mn2+.45

Perspectives

Compared with other types of invasive cancer, TNBC has fewer treatment options due to the absence of actionable targets. The proposed immunotherapy approach combines genetically engineered bacteria with hollow manganese dioxide nanospheres that function as chemical nano-immunomodulators. It not only remodels the acidic and hypoxia tumor microenvironment and re-boost macrophage-driven anti-tumor responses but also provide hollow nanospheres as potential carriers for the delivery of therapeutic payloads of clinical needs in the future. In particular, because the technique strategy is based on bacteria depletion of lactic acid instead of blocking or knocking out the generation pathway of lactic acid, it does not disturb normal glucose metabolism while the acidification of TME is stopped. This immunotherapeutic approach is helpful for those people suffering from chemotherapy and radiation that cannot prevent the recurrence of TNBC but may damage normal tissues. It is shown self-propelled bacteria can be developed as perfect robot therapies with genetic and chemical manipulation. The distinguished advantage is the capability to penetrate into solid tumor regions that are usually inaccessible to passive therapies. With the aid of the expression of genes that can recognize molecular markers on surfaces of tumor cells, differentiation of tumor cells with normal cells and specific target is possible. In the meanwhile, the chemical nano-immunomodulators relieve hypoxia and oxidative stress. Those unique features of genetically engineered and chemically modified bacteria make them well-suited as perspective anticancer agents.

References

1. Woolston, C., Breast cancer. Nature 2015, 527, S101.
2. Polyak, K. Breast cancer: origins and evolution. J. Clin. Invest. 2007, 117, 3155-3163.
3. Fan, L., Strasser-Weippl, K., Li, J. J., St Louis, J., Finkelstein, D. M., Yu, K. D., Chen, W. Q., Shao, Z. M., Goss, P. E., Breast cancer in China. Lancet Oncol. 2014, 15, e279-e289.
4. Gray, J., Druker, B., The breast cancer landscape. Nature 2012, 486, 328-329..
5. Lei, S., Zheng, R., Zhang, S., Chen, R., Wang, S., Sun, K., Zeng, H., Wei, W., He, J., Breast cancer incidence and mortality in women in China: temporal trends and projections to 2030. Cancer biol. Med. 2021, 18, 900.
6. Nolan, E., Lindeman, G. J., Visvader, J. E., Deciphering breast cancer: from biology to the clinic. Cell 2023, 186, 1708-1728.
7. Gray, J., & Druker, B., The breast cancer landscape. Nature 2012, 486, 328-329.
8. Bianchini, G., Balko, J. M., Mayer, I. A., Sanders, M. E., Gianni, L., Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat. Rev. Clin. Oncol. 2016, 13, 674-690.
9. Li, L., Zhang, F., Liu, Z., Fan, Z., Immunotherapy for triple-negative breast cancer: combination strategies to improve outcome. Cancers 2023, 15, 321.
10. Hay, N., Reprogramming glucose metabolism in cancer: can it be exploited for cancer therapy?. Nat. Rev. Cancer 2016, 16, 635-649.
11. Deepak, K. G. K., Vempati, R., Nagaraju, G. P., Dasari, V. R., Nagini, S., Rao, D. N., Malla, R. R., Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol. Res. 2020, 153, 104683.
12. Sisignano, M.; Fischer, M.J.M.; Geisslinger, G., Proton-sensing GPCRs in health and disease. Cells 2021, 10, 2050.
13. Lv, S., Li, J., Qiu, X., Li, W., Zhang, C., Zhang, Z. N., Luan, B., A negative feedback loop of ICER and NF-κB regulates TLR signaling in innate immune responses. Cell Death Differ. 2017, 24, 492-499.
14. Sekine, H.; Yamamoto, M.; Motohashi, H., Tumors sweeten macrophages with acids. Nat. Immunol. 2018, 19, 1281–1283.
15. Tariq, M., Zhang, J., Liang, G., Ding, L., He, Q., & Yang, B., Macrophage polarization: anti‐cancer strategies to target tumor‐associated macrophage in breast cancer. J. Cell. Biochem. 2017, 118, 2484-2501.
16. Mehta, A. K,; Kadel, S.; Townsend, M. G,; Oliwa. M,; Guerriero, J. L., Macrophage biology and mechanisms of immune suppression in breast cancer. Front. Immunol. 2021, 12, 643771.
17. Bohn, T., Rapp, S., Luther, N., Klein, M., Bruehl, T. J., Kojima, N., Lopez, P. A., Hahlbrock, J., Muth, S., Endo, S., Pektor, S., Brand, A., Renner, K., Popp, V., Gerlach, K., Vogel, D., Lueckel, C., Arnold-Schild, D., Pouyssegur, J., Kreutz, M., Huber, M., Koenig, J., Weigmann, B., Probst, H. C., Bopp, T., Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat. Immunol. 2018, 19, 1319–1329.
18. William, D. Foulkes, W. D.; Smith, I. E.; Reis-Filho, J. S. Triple negative breast cancer. N. Engl. J. Med. 2010, 363, 1938-1948.
19. de Ruijter, T. C., Veeck, J., de Hoon, J. P., van Engeland, M., Tjan-Heijnen, V. C., Characteristics of triple-negative breast cancer. J. Cancer Res. Clin. Oncol. 2011, 137, 183-192.
20. Cleator, S.; Heller, W.; Coombes, R. C. Triple-negative breast cancer: therapeutic options. Lancet Oncol. 2007, 8, 235-244.
21. Kashyap, D., Bal, A., Irinike, S., Khare, S., Bhattacharya, S., Das, A., Singh, G., Heterogeneity of the tumor microenvironment across molecular subtypes of breast cancer. Appl. Immunohistochem. Mol. Morphol. 2023, 31, 533-543.
22. Barchiesi, G., Roberto, M., Verrico, M., Vici, P., Tomao, S., Tomao, F., Emerging role of PARP inhibitors in metastatic triple negative breast cancer. Current scenario and future perspectives. Fron. Oncol. 2021, 11, 769280.
23. Seligson, J. M., Patron, A. M., Berger, M. J., Harvey, R. D., Seligson, N. D., Sacituzumab govitecan-hziy: an antibody-drug conjugate for the treatment of refractory, metastatic, triple-negative breast cancer. Annu. Pharmacother. 2021, 55(7), 921-931.
24. Thomas, R., Al-Khadairi, G., Decock, J., Immune checkpoint inhibitors in triple negative breast cancer treatment: promising future prospects. Front. Oncol. 2021, 10, 600573.
25. Napier, T.S.; Hunter, C.L.; Song, P.N.; Larimer, B. M.; Sorace, A. G. Preclinical PET imaging of granzyme B shows promotion of immunological response following combination paclitaxel and immune checkpoint inhibition in triple negative breast cancer. Pharmaceutics 2022, 14, 440.
26. Hu, Y., Manasrah, B. K., McGregor, S. M., Lera, R. F., Norman, R. X., Tucker, J. B., Scribano, C. M., Yan, R. E., Humayun, M., Wisinski, K. B., Tevaarwerk, A. J., O'Regan, R. M., Wilke, L. G., Weaver, B. A., Beebe, D. J., Jin, N., Burkard, M. E., Paclitaxel induces micronucleation and activates pro-inflammatory cGAS–STING signaling in triple-negative breast cancer. Mol. Cancer Ther. 2021, 20, 2553-2567.
27. Sharmni Vishnu, K., Win, T. T., Aye, S. N., Basavaraj, A. K., Combined atezolizumab and nab-paclitaxel in the treatment of triple negative breast cancer: a meta-analysis on their efficacy and safety. BMC cancer 2022, 22, 1-13.
28. Kim, H., Choi, J. M., Lee, K. M., Immune checkpoint blockades in triple-negative breast cancer: current state and molecular mechanisms of resistance. Biomed. 2022, 10, 1130.
29. Gurbatri, C. R., Arpaia, N., Danino, T., Engineering bacteria as interactive cancer therapies. Science 2022, 378, 858-864.
30. Willingham, S. B., Volkmer, J. P., Gentles, A. J., Sahoo, D., Dalerba, P., Mitra, S. S., Wang, J., Contreras-Trujillo, H., Martin, R., Cohen, J. D., Lovelace, P., Scheeren, F. A., Chao, M. P., Weiskopf, K., Tang, C., Volkmer, A. K., Naik, T. J., Storm, T. A., Mosley, A. R., Edris, B., Schmid, S. M., Sun, C. K., Chua, M. S., Murillo, O., Rajendran, P., Cha, A. C., Chin, R. K., Adorno, D. M., Raveh, T., Tseng, D., Jaiswal, S., Øyvind Enger, P., Steinberg, G. K., Li, G., So, S. K., Majeti, R., Harsh, G. R., van de Rijn, M., Teng, N. N. ., Sunwoo, J. B., Alizadeh, A. A., Clarke, M. F., Weissman, I. L., The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. 2012, 109, 6662-6667.
31. Luo, H., Chen, Y., Kuang, X., Wang, X., Yang, F., Cao, Z., Wang, L., Lin, S., Wu, F., Liu, J., Chemical reaction-mediated covalent localization of bacteria. Nat. Commun. 2022, 13, 7808.
32. Karyakin, A. A., Presnova, G. V., Rubtsova, M. Y., Egorov, A. M., Oriented immobilization of antibodies onto the gold surfaces via their native thiol groups. Anal. Chem. 2000, 72, 3805-3811.
33. Haase H. Innate immune cells speak manganese. Immun. 2018, 48, 616–618.
34. Liu, X., Zhou, Y., Xie, W., Liu, S., Zhao, Q., Huang, W., Construction of smart manganese dioxide‐based all‐in‐one nanoplatform for cancer diagnosis and therapy. Small Methods 2020, 4, 2000566.
35. Huang, Y., Ruan, Y., Ma, Y., Chen, D., Zhang, T., Fan, S., Lin, W., Huang, Y., Lu, H., Xu, J. F., Pi, J., Zheng, B., Immunomodulatory activity of manganese dioxide nanoparticles: Promising for novel vaccines and immunotherapeutics. Front. Immunol. 2023, 14, 1128840.
36. Zhang, Z., Ji, Y., Nanostructured manganese dioxide for anticancer applications: preparation, diagnosis, and therapy. Nanoscale 2020, 12, 17982-18003.
37. Forbes, N. S., Engineering the perfect (bacterial) cancer therapy. Nat. Rev. Cancer 2010, 10, 785-794.
38. Yu, S., Chen, Z., Zeng, X., Chen, X., Gu, Z., Advances in nanomedicine for cancer starvation therapy. Theranostics 2019, 9, 8026.
39. Hua, Q.; Joyce, A. R.; Palsson, B. Ø.; Fong, S. S. Metabolic characterization of Escherichia coli strains adapted to growth on lactate. Appl. Environ. Microbiol. 2007, 74, 4639–4647.
40. Fong, S. S., A. R. Joyce, and B. O. Palsson. Parallel adaptive evolution cultures of Escherichia coli lead to convergent growth phenotypes with different gene expression states. Genome Res. 2005, 15, 1365–1372.
41. Hayat, S. M. G., Bianconi, V., Pirro, M., Jaafari, M. R., Hatamipour, M., Sahebkar, A., CD47: role in the immune system and application to cancer therapy. Cell. Oncol. 2020, 43, 19-30.
42. Takimoto, C. H., Chao, M. P., Gibbs, C., McCamish, M. A., Liu, J., Chen, J. Y., Majeti, I.L., Weissman, I. L., The Macrophage ‘Do not eat me’signal, CD47, is a clinically validated cancer immunotherapy target. Ann. Oncol. 30, 486-489.
43. Horning, K. J., Caito, S. W., Tipps, K. G., Bowman, A. B,, Aschner, M., Manganese is essential for neuronal health. Annu. Rev. Nutr. 2015, 35, 71–108.
44. Sun, Y., Wang, Y., Liu, Y., Wang, H., Yang, C., Liu, X., Wang, F., Integration of manganese dioxide‐based nanomaterials for biomedical applications. Adv. Nanobiomed. Res. 2023, 3, 2200093.
45. Yang, G., Ji, J., Liu, Z., Multifunctional MnO2 nanoparticles for tumor microenvironment modulation and cancer therapy. WIRES Nanomed. Nanobi. 2021, 13, e1720.