Microbiota and Extracellular Vesicles in Anti-PD-1/PD-L1 Therapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Methods
3. Microbiota in Cancer and Anti-PD-1/PD-L-1 Therapy
3.1. PD-1/PD-L-1 Axis and Microbiota
3.2. Microbiota in Anti-PD-1/PD-L1 Therapy
- (i)
- through the activation of immune cell responses due to presence of microbial antigens: bacterial antigens such as peptide or lipid structures can activate a large range of T cell receptors. These microbial antigens either help tumor-specific immune responses and facilitate anti-tumor activity, or in some cases may cross-react against tumor-specific antigens and in turn become responsible for anti-tumor drug resistance [25,39].
- (ii)
- through the involvement of pattern recognition receptors: immune cells when exposed to microbes such as Bacteroides fragilis or Akkermansia muciniphila activate systemic interleukin dependent immune responses, which facilitate tumor control [13,40]. Zitvogel et al., (2018) suggested that ligands of toll-like receptors or Nod-like receptors may cause these microorganisms to produce such immune responses [38].
- (iii)
3.3. Effect of Fecal Microbiota Transplant (FMT) on Anti-PD-1 Therapy
4. Extracellular Vesicles in Cancer and Anti-PD-1/PD-L1 Therapy
4.1. Mammalian Extracellular Vesicles (MEVs)
4.2. Bacterial Extracellular Vesicles (bEVs)
4.2.1. bEVs as Diagnostic Markers
4.2.2. bEVs as Cancer Immunotherapy Agents
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Global Health Estimates: Leading Causes of Death. Available online: https://www.who.int/data/gho/data/themes/mortality-and-global-health-estimates/ghe-leading-causes-of-death (accessed on 21 July 2022).
- Comparative Oncology-PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/20806453/ (accessed on 21 July 2022).
- Rommasi, F. Bacterial-based methods for cancer treatment: What we know and where we are. Oncol. Ther. 2022, 10, 23–54. [Google Scholar] [CrossRef] [PubMed]
- Chaudhri, A.; Xiao, Y.; Klee, A.N.; Wang, X.; Zhu, B.; Freeman, G.J. PD-L1 binds to B7-1 only in cis on the same cell surface. Cancer Immunol. Res. 2018, 6, 921–929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Satelli, A.; Batth, I.S.; Brownlee, Z.; Rojas, C.; Meng, Q.H.; Kopetz, S.; Li, S. Potential role of nuclear PD-L1 expression in cell-surface vimentin positive circulating tumor cells as a prognostic marker in cancer patients. Sci. Rep. 2016, 6, 28910. [Google Scholar] [CrossRef] [Green Version]
- Jia, L.; Gao, Y.; Zhou, T.; Zhao, X.L.; Hu, H.Y.; Chen, D.W.; Qiao, M.X. Enhanced response to PD-L1 silencing by modulation of TME via balancing glucose metabolism and robust co-delivery of siRNA/Resveratrol with dual-responsive polyplexes. Biomaterials 2021, 271, 120711. [Google Scholar] [CrossRef] [PubMed]
- Burcelin, R.; Serino, M.; Chabo, C.; Garidou, L.; Pomié, C.; Courtney, M.; Amar, J.; Bouloumié, A. Metagenome and metabolism: The tissue microbiota hypothesis. Diabetes Obes. Metab. 2013, 15 (Suppl. 3), 61–70. [Google Scholar] [CrossRef] [PubMed]
- Peng, H.; He, X.; Wang, Q. Immune checkpoint blockades in gynecological cancers: A review of clinical trials. Acta Obstet. Gynecol. Scand. 2022, 101, 941–951. [Google Scholar] [CrossRef]
- Zhu, Q.; Gao, R.; Wu, W.; Qin, H. The role of gut microbiota in the pathogenesis of colorectal cancer. Tumor Biol. 2013, 34, 1285–1300. [Google Scholar] [CrossRef] [PubMed]
- Sonnenberg, G.F.; Artis, D. Innate lymphoid cell interactions with microbiota: Implications for intestinal health and disease. Immunity 2012, 37, 601–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; San Lucas, A.; et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 2019, 178, 795–806.e12. [Google Scholar] [CrossRef]
- Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef]
- Routy, B.; le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
- Chronopoulos, A.; Kalluri, R. Emerging role of bacterial extracellular vesicles in cancer. Oncogene 2020, 39, 6951–6960. [Google Scholar] [CrossRef] [PubMed]
- Amatya, S.B.; Salmi, S.; Kainulainen, V.; Karihtala, P.; Reunanen, J. Bacterial extracellular vesicles in gastrointestinal tract cancer: An unexplored territory. Cancers 2021, 13, 5450. [Google Scholar] [CrossRef] [PubMed]
- Botticelli, A.; Zizzari, I.; Mazzuca, F.; Ascierto, P.A.; Putignani, L.; Marchetti, L.; Napoletano, C.; Nuti, M.; Marchetti, P.; Botticelli, A.; et al. Cross-talk between microbiota and immune fitness to steer and control response to anti PD-1/PDL-1 treatment. Oncotarget 2016, 8, 8890–8899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahmadzadeh, M.; Johnson, L.A.; Heemskerk, B.; Wunderlich, J.R.; Dudley, M.E.; White, D.E.; Rosenberg, S.A. Tumor antigen–specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009, 114, 1537. [Google Scholar] [CrossRef] [PubMed]
- Ohaegbulam, K.C.; Assal, A.; Lazar-Molnar, E.; Yao, Y.; Zang, X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol. Med. 2015, 21, 24. [Google Scholar] [CrossRef] [Green Version]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science 2015, 350, 1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zitvogel, L.; Daillère, R.; Roberti, M.P.; Routy, B.; Kroemer, G. Anticancer effects of the microbiome and its products. Nat. Rev. Microbiol. 2017, 15, 465–478. [Google Scholar] [CrossRef]
- Asmar, N.; Ibrahim, T.; Rey, J.F. Checkpoint inhibitors: Conquering cancer with a little (T)-help from our microbial friends. Dig. Dis. Sci. 2018, 63, 2177–2179. [Google Scholar] [CrossRef] [PubMed]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
- Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 2017, 28, 1368–1379. [Google Scholar] [CrossRef] [PubMed]
- Zitvogel, L.; Ayyoub, M.; Routy, B.; Kroemer, G. Microbiome and anticancer immunosurveillance. Cell 2016, 165, 276–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, Z.; Cheng, S.; Kou, Y.; Wang, Z.; Jin, R.; Hu, H.; Zhang, X.; Gong, J.F.; Li, J.; Lu, M.; et al. The gut microbiome is associated with clinical response to anti-PD-1/PD-L1 immunotherapy in gastrointestinal cancer. Cancer Immunol. Res. 2020, 8, 1251–1261. [Google Scholar] [CrossRef] [PubMed]
- Thaiss, C.A.; Zmora, N.; Levy, M.; Elinav, E. The microbiome and innate immunity. Nature 2016, 535, 65–74. [Google Scholar] [CrossRef] [PubMed]
- Frankel, A.E.; Coughlin, L.A.; Kim, J.; Froehlich, T.W.; Xie, Y.; Frenkel, E.P.; Koh, A.Y. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia 2017, 19, 848–855. [Google Scholar] [CrossRef] [PubMed]
- Chu, S.; Cheng, Z.; Yin, Z.; Xu, J.; Wu, F.; Jin, Y.; Yang, G. Airway fusobacterium is associated with poor response to immunotherapy in lung cancer. Onco Targets Ther. 2022, 15, 201–213. [Google Scholar] [CrossRef]
- Zheng, Y.; Wang, T.; Tu, X.; Huang, Y.; Zhang, H.; Tan, D.; Jiang, W.; Cai, S.; Zhao, P.; Song, R.; et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J. Immunother. Cancer 2019, 7, 193. [Google Scholar] [CrossRef] [Green Version]
- Szczyrek, M.; Bitkowska, P.; Chunowski, P.; Czuchryta, P.; Krawczyk, P.; Milanowski, J. Diet, Microbiome, and cancer immunotherapy—A comprehensive review. Nutrients 2021, 13, 2217. [Google Scholar] [CrossRef]
- Ouaknine Krief, J.; Helly De Tauriers, P.; Dumenil, C.; Neveux, N.; Dumoulin, J.; Giraud, V.; Labrune, S.; Tisserand, J.; Julie, C.; Emile, J.F.; et al. Role of antibiotic use, plasma citrulline and blood microbiome in advanced non-small cell lung cancer patients treated with nivolumab. J. Immunother. Cancer 2019, 7, 176. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.; Lee, J.E.; Hong, S.H.; Lee, M.A.; Kang, J.H.; Kim, I.H. The effect of antibiotics on the clinical outcomes of patients with solid cancers undergoing immune checkpoint inhibitor treatment: A retrospective study. BMC Cancer 2019, 19, 1100. [Google Scholar] [CrossRef] [PubMed]
- Reed, J.P.; Devkota, S.; Figlin, R.A.; Li, X. Gut microbiome, antibiotic use, and immunotherapy responsiveness in cancer. Ann. Transl Med. 2019, 7, S309. [Google Scholar] [CrossRef] [PubMed]
- Hakozaki, T.; Okuma, Y.; Omori, M.; Hosomi, Y. Impact of prior antibiotic use on the efficacy of nivolumab for non-small cell lung cancer. Oncol. Lett. 2019, 17, 2946–2952. [Google Scholar] [CrossRef] [Green Version]
- Bertrand, A.; Kostine, M.; Barnetche, T.; Truchetet, M.E.; Schaeverbeke, T. Immune related adverse events associated with anti-CTLA-4 antibodies: Systematic review and meta-analysis. BMC Med. 2015, 13, 211. [Google Scholar] [CrossRef] [Green Version]
- Bernicker, E.H.; Quigley, E.M.M. The gut microbiome influences responses to programmed death 1 therapy in chinese lung cancer patients—The benefits of diversity. J. Thorac. Oncol. 2019, 14, 1319–1322. [Google Scholar] [CrossRef]
- Yan, X.; Zhang, S.; Deng, Y.; Wang, P.; Hou, Q.; Xu, H. Prognostic factors for checkpoint inhibitor based immunotherapy: An update with new evidences. Front. Pharmacol. 2018, 9, 1050. [Google Scholar] [CrossRef] [Green Version]
- Zitvogel, L.; Ma, Y.; Raoult, D.; Kroemer, G.; Gajewski, T.F. The microbiome in cancer immunotherapy: Diagnostic tools and therapeutic strategies. Science 2018, 359, 1366–1370. [Google Scholar] [CrossRef] [Green Version]
- Kalaora, S.; Nagler, A.; Nejman, D.; Alon, M.; Barbolin, C.; Barnea, E.; Ketelaars, S.L.C.; Cheng, K.; Vervier, K.; Shental, N.; et al. Identification of bacteria-derived HLA-bound peptides in melanoma. Nature 2021, 592, 138–143. [Google Scholar] [CrossRef]
- Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.M.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [Green Version]
- Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021, 371, 602–609. [Google Scholar] [CrossRef]
- Davar, D.; Dzutsev, A.K.; McCulloch, J.A.; Rodrigues, R.R.; Chauvin, J.M.; Morrison, R.M.; Deblasio, R.N.; Menna, C.; Ding, Q.; Pagliano, O.; et al. Fecal microbiota transplant overcomes resistance to anti–PD-1 therapy in melanoma patients. Science 2021, 371, 595. [Google Scholar] [CrossRef]
- Yin, Z.; Yu, M.; Ma, T.; Zhang, C.; Huang, S.; MR, K.; AA, M.-B.; Chen, S. Mechanisms underlying low-clinical responses to PD-1/PD-L1 blocking antibodies in immunotherapy of cancer: A key role of exosomal PD-L1. J. Immunother. Cancer 2021, 9, e001698. [Google Scholar] [CrossRef]
- Ren, D.; Hua, Y.; Yu, B.; Ye, X.; He, Z.; Li, C.; Wang, J.; Mo, Y.; Wei, X.; Chen, Y.; et al. Predictive biomarkers and mechanisms underlying resistance to PD1/PD-L1 blockade cancer immunotherapy. Mol. Cancer 2020, 19, 1–19. [Google Scholar] [CrossRef] [Green Version]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
- Xu, Z.; Zeng, S.; Gong, Z.; Yan, Y. Exosome-based immunotherapy: A promising approach for cancer treatment. Mol. Cancer 2020, 19, 160. [Google Scholar] [CrossRef]
- Xie, F.; Xu, M.; Lu, J.; Mao, L.; Wang, S. The role of exosomal PD-L1 in tumor progression and immunotherapy. Mol. Cancer 2019, 18, 146. [Google Scholar] [CrossRef] [Green Version]
- Tkach, M.; Théry, C. Communication by extracellular vesicles: Where we are and where we need to go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef] [Green Version]
- Bobrie, A.; Colombo, M.; Raposo, G.; Théry, C. Exosome secretion: Molecular mechanisms and roles in immune responses. Traffic 2011, 12, 1659–1668. [Google Scholar] [CrossRef]
- Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
- McAndrews, K.M.; Kalluri, R. Mechanisms associated with biogenesis of exosomes in cancer. Mol. Cancer 2019, 18, 52. [Google Scholar] [CrossRef]
- Chetty, V.K.; Ghanam, J.; Anchan, S.; Reinhardt, K.; Brenzel, A.; Gelléri, M.; Cremer, C.; Grueso-navarro, E.; Schneider, M.; von Neuhoff, N.; et al. Efficient small extracellular vesicles (EV) isolation method and evaluation of EV-associated DNA role in cell-cell communication in cancer. Cancers 2022, 14, 2068. [Google Scholar] [CrossRef]
- Theodoraki, M.N.; Yerneni, S.S.; Hoffmann, T.K.; Gooding, W.E.; Whiteside, T.L. Clinical significance of PD-L1 + exosomes in plasma of head and neck cancer patients. Clin. Cancer Res. 2018, 24, 896–905. [Google Scholar] [CrossRef] [Green Version]
- Ludwig, S.; Floros, T.; Theodoraki, M.N.; Hong, C.S.; Jackson, E.K.; Lang, S.; Whiteside, T.L. Suppression of lymphocyte functions by plasma exosomes correlates with disease activity in patients with head and neck cancer. Clin. Cancer Res. 2017, 23, 4843–4854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ricklefs, F.L.; Alayo, Q.; Krenzlin, H.; Mahmoud, A.B.; Speranza, M.C.; Nakashima, H.; Hayes, J.L.; Lee, K.; Balaj, L.; Passaro, C.; et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci. Adv. 2018, 4, eaar2766. [Google Scholar] [CrossRef] [Green Version]
- Zhou, J.; Mahoney, K.M.; Giobbie-Hurder, A.; Zhao, F.; Lee, S.; Liao, X.; Rodig, S.; Li, J.; Wu, X.; Butterfield, L.H.; et al. Soluble PD-L1 as a biomarker in malignant melanoma treated with checkpoint blockade. Cancer Immunol. Res. 2017, 5, 480–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carretero-González, A.; Hergueta-Redondo, M.; Sánchez-Redondo, S.; Ximénez-Embún, P.; Manso Sánchez, L.; Gil, E.C.; Castellano, D.; de Velasco, G.; Peinado, H. Characterization of plasma circulating small extracellular vesicles in patients with metastatic solid tumors and newly diagnosed brain metastasis. Oncoimmunology 2022, 11, 2067944. [Google Scholar] [CrossRef]
- Cordonnier, M.; Nardin, C.; Chanteloup, G.; Derangere, V.; Algros, M.P.; Arnould, L.; Garrido, C.; Aubin, F.; Gobbo, J. Tracking the evolution of circulating exosomal-PD-L1 to monitor melanoma patients. J. Extracell. Vesicles 2020, 9, 1710899. [Google Scholar] [CrossRef] [Green Version]
- Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [Green Version]
- Yin, Y.; Liu, B.; Cao, Y.; Yao, S.; Liu, Y.; Jin, G.; Qin, Y.; Chen, Y.; Cui, K.; Zhou, L.; et al. Colorectal cancer-derived small extracellular vesicles promote tumor immune evasion by upregulating PD-L1 expression in tumor-associated macrophages. Adv. Sci. 2022, 9, 2102620. [Google Scholar] [CrossRef]
- Cappellesso, R.; Tinazzi, A.; Giurici, T.; Simonato, F.; Guzzardo, V.; Ventura, L.; Crescenzi, M.; Chiarelli, S.; Fassina, A. Programmed cell death 4 and MicroRNA 21 inverse expression is maintained in cells and exosomes from ovarian serous carcinoma effusions. Cancer Cytopathol. 2014, 122, 685–693. [Google Scholar] [CrossRef]
- Chan, J.A.; Krichevsky, A.M.; Kosik, K.S. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005, 65, 6029–6033. [Google Scholar] [CrossRef]
- Meng, F.; Henson, R.; Wehbe-Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Venturutti, L.; Romero, L.V.; Urtreger, A.J.; Chervo, M.F.; Cordo Russo, R.I.; Mercogliano, M.F.; Inurrigarro, G.; Pereyra, M.G.; Proietti, C.J.; Izzo, F.; et al. Stat3 regulates ErbB-2 expression and co-opts ErbB-2 nuclear function to induce MiR-21 expression, PDCD4 downregulation and breast cancer metastasis. Oncogene 2016, 35, 2208–2222. [Google Scholar] [CrossRef] [PubMed]
- MiR-200a, MiR-200b and MiR-429 Are Onco-MiRs That Target the PTEN Gene in Endometrioid Endometrial Carcinoma-PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/25750291/ (accessed on 25 July 2022).
- Yang, X.; Wang, J.; Qu, S.; Zhang, H.; Ruan, B.; Gao, Y.; Ma, B.; Wang, X.; Wu, N.; Li, X.; et al. MicroRNA-200a suppresses metastatic potential of side population cells in human hepatocellular carcinoma by decreasing ZEB2. Oncotarget 2015, 6, 7918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, S.J.; Hu, J.Y.; Kuang, X.Y.; Luo, J.M.; Hou, Y.F.; Di, G.H.; Wu, J.; Shen, Z.Z.; Song, H.Y.; Shao, Z.M. MicroRNA-200a promotes anoikis resistance and metastasis by targeting YAP1 in human breast cancer. Clin. Cancer Res. 2013, 19, 1389–1399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, T.; Zhang, Y.; Qu, X.; Che, X.; Li, C.; Fan, Y.; Wan, X.; Ma, R.; Hou, K.; Zhou, H.; et al. MiR-200a enhances TRAIL-induced apoptosis in gastric cancer cells by targeting A20. Cell Biol. Int. 2018, 42, 506–514. [Google Scholar] [CrossRef] [PubMed]
- Zang, Y.; Tai, Y.; Wan, B.; Jia, X. MiR-200a-3p promotes the proliferation of human esophageal cancer cells by post-transcriptionally regulating cytoplasmic collapsin response mediator protein-1. Int. J. Mol. Med. 2016, 38, 1558–1564. [Google Scholar] [CrossRef] [Green Version]
- Peng, F.; Jiang, J.; Yu, Y.; Tian, R.; Guo, X.; Li, X.; Shen, M.; Xu, M.; Zhu, F.; Shi, C.; et al. Direct targeting of SUZ12/ROCK2 by MiR-200b/c inhibits cholangiocarcinoma tumourigenesis and metastasis. Br. J. Cancer 2013, 109, 3092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tixeira, R.; Caruso, S.; Paone, S.; Baxter, A.A.; Atkin-Smith, G.K.; Hulett, M.D.; Poon, I.K.H. Defining the morphologic features and products of cell disassembly during apoptosis. Apoptosis 2017, 22, 475–477. [Google Scholar] [CrossRef] [PubMed]
- Surman, M.; Hoja-łukowicz, D.; Szwed, S.; Kędracka-Krok, S.; Jankowska, U.; Kurtyka, M.; Drożdż, A.; Lityńska, A.; Stępień, E.; Przybyło, M. An insight into the proteome of uveal melanoma-derived ectosomes reveals the presence of potentially useful biomarkers. Int. J. Mol. Sci. 2019, 20, 3789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciardiello, C.; Leone, A.; Lanuti, P.; Roca, M.S.; Moccia, T.; Minciacchi, V.R.; Minopoli, M.; Gigantino, V.; de Cecio, R.; Rippa, M.; et al. Large oncosomes overexpressing integrin alpha-V promote prostate cancer adhesion and invasion via AKT activation. J. Exp. Clin. Cancer Res. 2019, 38, 317. [Google Scholar] [CrossRef] [PubMed]
- Wright, P.K.; Jones, S.B.; Ardern, N.; Ward, R.; Clarke, R.B.; Sotgia, F.; Lisanti, M.P.; Landberg, G.; Lamb, R. 17β-estradiol regulates giant vesicle formation via estrogen receptor-alpha in human breast cancer cells. Oncotarget 2014, 5, 3055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patton, M.C.; Zubair, H.; Khan, M.A.; Singh, S.; Singh, A.P. Hypoxia alters the release and size distribution of extracellular vesicles in pancreatic cancer cells to support their adaptive survival. J. Cell Biochem. 2020, 121, 828–839. [Google Scholar] [CrossRef] [PubMed]
- Kang, M.; Kim, S.; Ko, J. Roles of CD133 in microvesicle formation and oncoprotein trafficking in colon cancer. FASEB J. 2019, 33, 4248–4260. [Google Scholar] [CrossRef] [PubMed]
- Surman, M.; Hoja-Łukowicz, D.; Szwed, S.; Drożdż, A.; Stępień, E.; Przybyło, M. Human melanoma-derived ectosomes are enriched with specific glycan epitopes. Life Sci. 2018, 207, 395–411. [Google Scholar] [CrossRef] [PubMed]
- Ciardiello, C.; Cavallini, L.; Spinelli, C.; Yang, J.; Reis-Sobreiro, M.; Candia, P.D.; Minciacchi, V.R.; di Vizio, D. Focus on extracellular vesicles: New frontiers of cell-to-cell communication in cancer. Int. J. Mol. Sci. 2016, 17, 175. [Google Scholar] [CrossRef] [Green Version]
- Minciacchi, V.R.; Freeman, M.R.; di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vagner, T.; Spinelli, C.; Minciacchi, V.R.; Balaj, L.; Zandian, M.; Conley, A.; Zijlstra, A.; Freeman, M.R.; Demichelis, F.; De, S.; et al. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J. Extracell. Vesicles 2018, 7, 1505403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morello, M.; Minciacchi, V.R.; de Candia, P.; Yang, J.; Posadas, E.; Kim, H.; Griffiths, D.; Bhowmick, N.; Chung, L.W.K.; Gandellini, P.; et al. Large oncosomes mediate intercellular transfer of functional MicroRNA. Cell Cycle 2013, 12, 3526–3536. [Google Scholar] [CrossRef] [Green Version]
- Bertolini, I.; Terrasi, A.; Martelli, C.; Gaudioso, G.; di Cristofori, A.; Storaci, A.M.; Formica, M.; Braidotti, P.; Todoerti, K.; Ferrero, S.; et al. A GBM-like V-ATPase signature directs cell-cell tumor signaling and reprogramming via large oncosomes. eBioMedicine 2019, 41, 225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Minciacchi, V.R.; Spinelli, C.; Reis-Sobreiro, M.; Cavallini, L.; You, S.; Zandian, M.; Li, X.; Mishra, R.; Chiarugi, P.; Adam, R.M.; et al. MYC mediates large oncosome-induced fibroblast reprogramming in prostate cancer. Cancer Res. 2017, 77, 2306–2317. [Google Scholar] [CrossRef] [PubMed]
- di Vizio, D.; Morello, M.; Dudley, A.C.; Schow, P.W.; Adam, R.M.; Morley, S.; Mulholland, D.; Rotinen, M.; Hager, M.H.; Insabato, L.; et al. Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. Am. J. Pathol. 2012, 181, 1573–1584. [Google Scholar] [CrossRef] [PubMed]
- Hager, M.H.; Morley, S.; Bielenberg, D.R.; Gao, S.; Morello, M.; Holcomb, I.N.; Liu, W.; Mouneimne, G.; Demichelis, F.; Kim, J.; et al. DIAPH3 governs the cellular transition to the amoeboid tumour phenotype. EMBO Mol. Med. 2012, 4, 743. [Google Scholar] [CrossRef] [PubMed]
- di Vizio, D.; Kim, J.; Hager, M.H.; Morello, M.; Yang, W.; Lafargue, C.J.; True, L.D.; Rubin, M.A.; Adam, R.M.; Beroukhim, R.; et al. Oncosome formation in prostate cancer: Association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res. 2009, 69, 5601–5609. [Google Scholar] [CrossRef] [Green Version]
- Muralidharan-Chari, V.; Clancy, J.; Plou, C.; Romao, M.; Chavrier, P.; Raposo, G.; D’Souza-Schorey, C. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr. Biol. 2009, 19, 1875–1885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaparakis-Liaskos, M.; Ferrero, R.L. Immune modulation by bacterial outer membrane vesicles. Nat. Rev. Immunol. 2015, 15, 375–387. [Google Scholar] [CrossRef] [PubMed]
- Toyofuku, M.; Nomura, N.; Eberl, L. Types and origins of bacterial membrane vesicles. Nat. Rev. Microbiol. 2019, 17, 13–24. [Google Scholar] [CrossRef] [PubMed]
- Tulkens, J.; Vergauwen, G.; van Deun, J.; Geeurickx, E.; Dhondt, B.; Lippens, L.; de Scheerder, M.A.; Miinalainen, I.; Rappu, P.; de Geest, B.G.; et al. Increased levels of systemic LPS-positive bacterial extracellular vesicles in patients with intestinal barrier dysfunction. Gut 2020, 69, 191–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, S.I.; Kang, N.; Leem, S.; Yang, J.; Jo, H.; Lee, M.; Kim, H.S.; Dhanasekaran, D.N.; Kim, Y.K.; Park, T.; et al. Metagenomic analysis of serum microbe-derived extracellular vesicles and diagnostic models to differentiate ovarian cancer and benign ovarian tumor. Cancers 2020, 12, 1309. [Google Scholar] [CrossRef]
- Lee, Y.S.; Kim, J.H.; Lim, D.H. Urine microbe-derived extracellular vesicles in children with asthma. Allergy Asthma Immunol. Res. 2021, 13, 75–87. [Google Scholar] [CrossRef]
- Yoo, J.Y.; Rho, M.; You, Y.A.; Kwon, E.J.; Kim, M.H.; Kym, S.; Jee, Y.K.; Kim, Y.K.; Kim, Y.J. 16S RRNA gene-based metagenomic analysis reveals differences in bacteria-derived extracellular vesicles in the urine of pregnant and non-pregnant women. Exp. Mol. Med. 2016, 48, e208. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.J.; Yang, J.; Seo, H.; Lee, W.H.; Ho Lee, D.; Kym, S.; Park, Y.S.; Kim, J.G.; Jang, I.J.; Kim, Y.K.; et al. Colorectal cancer diagnostic model utilizing metagenomic and metabolomic data of stool microbial extracellular vesicles. Sci. Rep. 2020, 10, 2860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, J.; Li, Q.; Haesebrouck, F.; van Hoecke, L.; Vandenbroucke, R.E. The tremendous biomedical potential of bacterial extracellular vesicles. Trends Biotechnol. 2022, 40, 1173–1194. [Google Scholar] [CrossRef]
- Starnes, C.O. Coley’s toxins in perspective. Nature 1992, 357, 11–12. [Google Scholar] [CrossRef]
- Kim, O.Y.; Park, H.T.; Dinh, N.T.H.; Choi, S.J.; Lee, J.; Kim, J.H.; Lee, S.W.; Gho, Y.S. Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response. Nat. Commun. 2017, 8, 626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, K.S.; Choi, K.H.; Kim, Y.S.; Hong, B.S.; Kim, O.Y.; Kim, J.H.; Yoon, C.M.; Koh, G.Y.; Kim, Y.K.; Gho, Y.S. Outer membrane vesicles derived from escherichia coli induce systemic inflammatory response syndrome. PLoS ONE 2010, 5, e11334. [Google Scholar] [CrossRef] [Green Version]
- Park, K.S.; Lee, J.; Jang, S.C.; Kim, S.R.; Jang, M.H.; Lötvall, J.; Kim, Y.K.; Gho, Y.S. Pulmonary inflammation induced by bacteria-free outer membrane vesicles from pseudomonas aeruginosa. Am. J. Respir. Cell Mol. Biol. 2013, 49, 637–645. [Google Scholar] [CrossRef]
- Svennerholm, K.; Park, K.S.; Wikström, J.; Lässer, C.; Crescitelli, R.; Shelke, G.V.; Jang, S.C.; Suzuki, S.; Bandeira, E.; Olofsson, C.S.; et al. Escherichia coli outer membrane vesicles can contribute to sepsis induced cardiac dysfunction. Sci. Rep. 2017, 7, 17434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Zhao, R.; Cheng, K.; Zhang, K.; Wang, Y.; Zhang, Y.; Li, Y.; Liu, G.; Xu, J.; Xu, J.; et al. bacterial outer membrane vesicles presenting programmed death 1 for improved cancer immunotherapy via immune activation and checkpoint inhibition. ACS Nano 2020, 14, 16698–16711. [Google Scholar] [CrossRef]
- KS, P.; Svennerholm, K.; Crescitelli, R.; Lässer, C.; Gribonika, I.; Lötvall, J. Synthetic bacterial vesicles combined with tumour extracellular vesicles as cancer immunotherapy. J. Extracell. Vesicles 2021, 10, e12120. [Google Scholar] [CrossRef]
- Khan, A.N.H.; Emmons, T.R.; Magner, W.J.; Alqassim, E.; Singel, K.L.; Ricciuti, J.; Eng, K.H.; Odunsi, K.; Tomasi, T.B.; Lee, K.; et al. VSSP abrogates murine ovarian tumor-associated myeloid cell-driven immune suppression and induces M1 polarization in tumor-associated macrophages from ovarian cancer patients. Cancer Immunol. Immunother. 2022, 71, 2355–2369. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Huang, L.; Mao, M.; Ding, J.; Wu, G.; Fan, W.; Yang, T.; Zhang, M.; Huang, Y.; Xie, H.-Y. viral protein-pseudotyped and SiRNA-electroporated extracellular vesicles for cancer immunotherapy. Adv. Funct. Mater. 2020, 30, 2006515. [Google Scholar] [CrossRef]
- Liu, H.; Geng, Z.; Su, J. Engineered mammalian and bacterial extracellular vesicles as promising nanocarriers for targeted therapy. Extracell. Vesicles Circ. Nucl. Acids 2022, 3, 63–86. [Google Scholar] [CrossRef]
- Cheng, K.; Zhao, R.; Li, Y.; Qi, Y.; Wang, Y.; Zhang, Y.; Qin, H.; Qin, Y.; Chen, L.; Li, C.; et al. Bioengineered bacteria-derived outer membrane vesicles as a versatile antigen display platform for tumor vaccination via plug-and-display technology. Nat. Commun. 2021, 12, 2041. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Bai, H.; Wu, W.; Huang, G.; Li, Y.; Wu, M.; Tang, G.; Ping, Y. Bioengineering bacterial vesicle-coated polymeric nanomedicine for enhanced cancer immunotherapy and metastasis prevention. Nano Lett. 2020, 20, 11–21. [Google Scholar] [CrossRef]
- Kuerban, K.; Gao, X.; Zhang, H.; Liu, J.; Dong, M.; Wu, L.; Ye, R.; Feng, M.; Ye, L. Doxorubicin-loaded bacterial outer-membrane vesicles exert enhanced anti-tumor efficacy in non-small-cell lung cancer. Acta Pharm. Sin. B 2020, 10, 1534–1548. [Google Scholar] [CrossRef]
- Shi, J.; Ma, Z.; Pan, H.; Liu, Y.; Chu, Y.; Wang, J.; Chen, L. Biofilm-encapsulated nano drug delivery system for the treatment of colon cancer. J. Microencapsul. 2020, 37, 481–491. [Google Scholar] [CrossRef]
- Kim, O.Y.; Dinh, N.T.H.; Park, H.T.; Choi, S.J.; Hong, K.; Gho, Y.S. Bacterial protoplast-derived nanovesicles for tumor targeted delivery of chemotherapeutics. Biomaterials 2017, 113, 68–79. [Google Scholar] [CrossRef]
Studied by | Study Group | Disease Studied | Treatment Used | Microorganism Involved | Clinical Response to Therapy |
---|---|---|---|---|---|
Peng et al. [25] | Human | Gastrointestinal cancer | Anti–PD-1/PD-L1 | Bacteroidetes, Firmicutes | High in responders |
Routy et al. [13] | Human | Non-small cell lung cancer (NSCLC) and renal cell carcinoma (RCC) | Anti–PD-1 | Akkermansia muciniphila | High in responders |
Matson et al. [12] | Human | Metastatic melanoma | Anti–PD-1 | Bifidobacterium longum, Collinsella aerofaciens, Enterococcus faecium | High in responders |
Frankel et al. [27] | Human | Metastatic melanoma | Anti-PD-1 and Anti-CTLA4 | Bacteroides caccae, Dorea formicogenerans | High in responders |
Gopalkrishnan et al. [22] | Human | Metastatic melanoma | Anti–PD-1 | Ruminococcaceae family | High in responders |
Chu et al. [28] | Human | Lung cancer | Anti–PD-1 | Fusobacterium | Produced resistance to Anti–PD-1 therapy |
Zheng et al. [29] | Human | Hepatocellular carcinoma (HCC) | Anti–PD-1 | Akkermansia muciniphila & Ruminococcaceae spp. | High in responders |
Sivan et al. [19] | Mice | Melanoma | Anti-PD-L1 | Bifidobacterium | Promotes anti-tumor immunity |
Study Done by | Study Model | Disease Studied | Clinical Response in Patients Receiving FMT |
---|---|---|---|
Gopalakrishnan et al. [22] | Germ free mice | Metastatic melanoma | Improved responses to anti–PD-L1 therapy; significant reduction in tumor size |
Matson et al. [12] | Germ free mice | Metastatic melanoma | Showed slow tumor growth |
Routy et al. [13] | Germ free mice | Non-small cell lung cancer (NSCLC) | Delay in tumor growth |
Baruch et al. [41] | Human | Metastatic melanoma | Out of 10 PD-1–refractory metastatic melanoma patients, 3 patients showed improved response to anti-PD-1 therapy |
Davar et al. [42] | Human | Metastatic melanoma | Out of 15 PD-1–refractory metastatic melanoma patients, 6 patients showed improved response to anti-PD-1 therapy |
Molecules | Functions | References | |
---|---|---|---|
Nucleic acids | MYC, AKT1, PTK2, KLF10, PTEN | Genes encoded by chromosomal DNA, their copy number variations favor cancer cell progression | [80,81] |
miR-1227 | Increases migration of cancer-associated fibroblasts (CAFs) when overexpressed | ||
GAPDH, GPI, LDHB, HSPA5, MDH, GOT, GLS | Metabolic enzymes | [82] | |
V-ATPase subunit V1G1 | Promote tumor progression by delivering oncogenic signals and reprogramming the tumor microenvironment | ||
Proteins | Urokinase-type plasminogen activator receptor (uPAR) | Promote cancer progression when released by the aggressive counterpart | [73,83] |
Eukaryotic elongation factor 1 gamma (eEF1γ) | |||
Serine-threonine protein kinase AKT1 | |||
Caveolin-1, CK18, MMP 2, MMP9 | Scaffolding protein/ cytoskeleton components and gelatinase activity | [73,84,85] | |
Small GTP-binding protein ARF6 | Coordinates the release of plasma membrane-derived microvesicles containing protease from tumor cells into the surrounding environment | [86,87] | |
αV-integrin | Imparts the adhesive and invasive properties of aggressive cancer cell line to the less aggressive equivalent | [73] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mishra, S.; Amatya, S.B.; Salmi, S.; Koivukangas, V.; Karihtala, P.; Reunanen, J. Microbiota and Extracellular Vesicles in Anti-PD-1/PD-L1 Therapy. Cancers 2022, 14, 5121. https://doi.org/10.3390/cancers14205121
Mishra S, Amatya SB, Salmi S, Koivukangas V, Karihtala P, Reunanen J. Microbiota and Extracellular Vesicles in Anti-PD-1/PD-L1 Therapy. Cancers. 2022; 14(20):5121. https://doi.org/10.3390/cancers14205121
Chicago/Turabian StyleMishra, Surbhi, Sajeen Bahadur Amatya, Sonja Salmi, Vesa Koivukangas, Peeter Karihtala, and Justus Reunanen. 2022. "Microbiota and Extracellular Vesicles in Anti-PD-1/PD-L1 Therapy" Cancers 14, no. 20: 5121. https://doi.org/10.3390/cancers14205121
APA StyleMishra, S., Amatya, S. B., Salmi, S., Koivukangas, V., Karihtala, P., & Reunanen, J. (2022). Microbiota and Extracellular Vesicles in Anti-PD-1/PD-L1 Therapy. Cancers, 14(20), 5121. https://doi.org/10.3390/cancers14205121