Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma
Abstract
:1. Introduction
2. Recent Cancer Therapies Possibly Applied for the Treatment of HCC
3. A New Concept for the Effective Cancer Therapy
4. Epigenetics with Gut Microbiota Alteration Involved in the Superior Cancer Therapy
5. Association Between Gut Microbiota and Useful Epigenetics for the Development of Effective Cancer Therapy Against HCC
6. Future Perspectives
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
ALD | alcohol-associated liver disease |
CAR | chimeric antigen receptor |
CTLA4 | cytotoxic T-lymphocyte-associated protein 4 |
FMT | fecal microbiota transplantation |
HBV | hepatitis B virus |
HCV | hepatitis C virus |
HCC | hepatocellular carcinoma |
LPS | lipopolysaccharide |
MASH | metabolic dysfunction associated steatohepatitis |
MASLD | metabolic dysfunction associated steatotic liver disease |
MHC | major histocompatibility complex |
PD-1 | programmed cell death protein-1 |
PD-L1 | programmed cell death protein ligand-1 |
PDT | photodynamic therapy |
PTT | photothermal therapy |
ROS | reactive oxygen species |
SCFAs | short-chain fatty acids |
STAT | signal transducer and activator of transcription |
Th17 | T helper 17 cell |
TLRs | toll-like receptors |
Treg | regulatory T cell |
VEGF | vascular endothelial growth factor A |
References
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [PubMed]
- Canale, M.; Ulivi, P.; Foschi, F.G.; Scarpi, E.; De Matteis, S.; Donati, G.; Ercolani, G.; Scartozzi, M.; Faloppi, L.; Passardi, A.; et al. Clinical and circulating biomarkers of survival and recurrence after radiofrequency ablation in patients with hepatocellular carcinoma. Crit. Rev. Oncol. Hematol. 2018, 129, 44–53. [Google Scholar] [PubMed]
- Choi, J.W.; Lee, J.M.; Lee, D.H.; Yoon, J.H.; Kim, Y.J.; Lee, J.H.; Yu, S.J.; Cho, E.J. Radiofrequency ablation using internally cooled wet electrodes in bipolar mode for the treatment of recurrent hepatocellular carcinoma after locoregional treatment: A randomized prospective comparative study. PLoS ONE 2020, 15, e0239733. [Google Scholar]
- Ilagan, C.H.; Goldman, D.A.; Gönen, M.; Aveson, V.G.; Babicky, M.; Balachandran, V.P.; Drebin, J.A.; Jarnagin, W.R.; Wei, A.C.; Kingham, T.P.; et al. Recurrence of Hepatocellular Carcinoma After Complete Radiologic Response to Trans-Arterial Embolization: A Retrospective Study on Patterns, Treatments, and Prognoses. Ann. Surg. Oncol. 2022, 29, 6815–6826. [Google Scholar]
- Bruix, J.; Qin, S.; Merle, P.; Granito, A.; Huang, Y.H.; Bodoky, G.; Pracht, M.; Yokosuka, O.; Rosmorduc, O.; Breder, V.; et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 389, 56–66. [Google Scholar]
- Hu, W.Y.; Wei, H.Y.; Li, K.M.; Wang, R.B.; Xu, X.Q.; Feng, R. LINC00511 as a ceRNA promotes cell malignant behaviors and correlates with prognosis of hepatocellular carcinoma patients by modulating miR-195/EYA1 axis. Biomed. Pharmacother. 2020, 121, 109642. [Google Scholar]
- Ma, C.; Kesarwala, A.H.; Eggert, T.; Medina-Echeverz, J.; Kleiner, D.E.; Jin, P.; Stroncek, D.F.; Terabe, M.; Kapoor, V.; ElGindi, M.; et al. NAFLD causes selective CD4(+) T lymphocyte loss and promotes hepatocarcinogenesis. Nature 2016, 531, 253–257. [Google Scholar]
- Mehal, W.Z. The Gordian Knot of dysbiosis, obesity and NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 637–644. [Google Scholar]
- Villanueva, A. Hepatocellular Carcinoma. N. Engl. J. Med. 2019, 380, 1450–1462. [Google Scholar]
- Chen, W.; Desert, R.; Ge, X.; Han, H.; Song, Z.; Das, S.; Athavale, D.; You, H.; Nieto, N. The Matrisome Genes From Hepatitis B-Related Hepatocellular Carcinoma Unveiled. Hepatol. Commun. 2021, 5, 1571–1585. [Google Scholar]
- Quesada-Vázquez, S.; Bone, C.; Saha, S.; Triguero, I.; Colom-Pellicer, M.; Aragonès, G.; Hildebrand, F.; Del Bas, J.M.; Caimari, A.; Beraza, N.; et al. Microbiota Dysbiosis and Gut Barrier Dysfunction Associated with Non-Alcoholic Fatty Liver Disease Are Modulated by a Specific Metabolic Cofactors’ Combination. Int. J. Mol. Sci. 2022, 23, 13675. [Google Scholar] [CrossRef] [PubMed]
- Guo, X.; Okpara, E.S.; Hu, W.; Yan, C.; Wang, Y.; Liang, Q.; Chiang, J.Y.L.; Han, S. Interactive Relationships between Intestinal Flora and Bile Acids. Int. J. Mol. Sci. 2022, 23, 8343. [Google Scholar] [CrossRef] [PubMed]
- Tohme, S.; Simmons, R.L.; Tsung, A. Surgery for Cancer: A Trigger for Metastases. Cancer Res. 2017, 77, 1548–1552. [Google Scholar]
- Goldstein, M.R.; Mascitelli, L. Surgery and cancer promotion: Are we trading beauty for cancer? QJM 2011, 104, 811–815. [Google Scholar]
- Shaverdian, N.; Lisberg, A.E.; Bornazyan, K.; Veruttipong, D.; Goldman, J.W.; Formenti, S.C.; Garon, E.B.; Lee, P. Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: A secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol. 2017, 18, 895–903. [Google Scholar]
- Seiwert, T.Y.; Kiess, A.P. Time to Debunk an Urban Myth? The “Abscopal Effect” with Radiation and Anti-PD-1. J. Clin. Oncol. 2021, 39, 1–3. [Google Scholar]
- Chen, D.; Verma, V.; Patel, R.R.; Barsoumian, H.B.; Cortez, M.A.; Welsh, J.W. Absolute Lymphocyte Count Predicts Abscopal Responses and Outcomes in Patients Receiving Combined Immunotherapy and Radiation Therapy: Analysis of 3 Phase 1/2 Trials. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 196–203. [Google Scholar]
- Zhai, D.; An, D.; Wan, C.; Yang, K. Radiotherapy: Brightness and darkness in the era of immunotherapy. Transl. Oncol. 2022, 19, 101366. [Google Scholar]
- Terasaki, Y.; Ohsawa, I.; Terasaki, M.; Takahashi, M.; Kunugi, S.; Dedong, K.; Urushiyama, H.; Amenomori, S.; Kaneko-Togashi, M.; Kuwahara, N.; et al. Hydrogen therapy attenuates irradiation-induced lung damage by reducing oxidative stress. Am. J. Physiol. Lung Cell Mol. Physiol. 2011, 301, L415–L426. [Google Scholar]
- Lundgren, S.; Karnevi, E.; Elebro, J.; Nodin, B.; Karlsson, M.C.I.; Eberhard, J.; Leandersson, K.; Jirström, K. The clinical importance of tumour-infiltrating macrophages and dendritic cells in periampullary adenocarcinoma differs by morphological subtype. J. Transl. Med. 2017, 15, 152. [Google Scholar]
- Ryter, S.W.; Kim, H.P.; Hoetzel, A.; Park, J.W.; Nakahira, K.; Wang, X.; Choi, A.M. Mechanisms of cell death in oxidative stress. Antioxid. Redox Signal. 2007, 9, 49–89. [Google Scholar] [PubMed]
- Siegel, R.J.; Singh, A.K.; Panipinto, P.M.; Shaikh, F.S.; Vinh, J.; Han, S.U.; Kenney, H.M.; Schwarz, E.M.; Crowson, C.S.; Khuder, S.A.; et al. Extracellular sulfatase-2 is overexpressed in rheumatoid arthritis and mediates the TNF-α-induced inflammatory activation of synovial fibroblasts. Cell Mol. Immunol. 2022, 19, 1185–1195. [Google Scholar] [PubMed]
- Raguz, S.; Yagüe, E. Resistance to chemotherapy: New treatments and novel insights into an old problem. Br. J. Cancer 2008, 99, 387–391. [Google Scholar] [PubMed]
- Choy, H.; Kim, D.W. Chemotherapy and irradiation interaction. Semin. Oncol. 2003, 30 (Suppl. S9), 3–10. [Google Scholar]
- McGuigan, A.; Kelly, P.; Turkington, R.C.; Jones, C.; Coleman, H.G.; McCain, R.S. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J. Gastroenterol. 2018, 24, 4846–4861. [Google Scholar]
- Jung, H.S.; Verwilst, P.; Sharma, A.; Shin, J.; Sessler, J.L.; Kim, J.S. Organic molecule-based photothermal agents: An expanding photothermal therapy universe. Chem. Soc. Rev. 2018, 47, 2280–2297. [Google Scholar]
- Fan, Z.; Zhuang, C.; Wang, S.; Zhang, Y. Photodynamic and Photothermal Therapy of Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 787780. [Google Scholar]
- Li, X.; Lovell, J.F.; Yoon, J.; Chen, X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol. 2020, 17, 657–674. [Google Scholar]
- Dun, X.; Liu, S.; Ge, N.; Liu, M.; Li, M.; Zhang, J.; Bao, H.; Li, B.; Zhang, H.; Cui, L. Photothermal effects of CuS-BSA nanoparticles on H22 hepatoma-bearing mice. Front. Pharmacol. 2022, 13, 1029986. [Google Scholar]
- Casini, A.; Leone, S.; Vaccaro, R.; Vivacqua, G.; Ceci, L.; Pannarale, L.; Franchitto, A.; Onori, P.; Gaudio, E.; Mancinelli, R. The Emerging Role of Ferroptosis in Liver Cancers. Life 2022, 12, 2128. [Google Scholar] [CrossRef]
- Kaufmann, S.H.E.; Dorhoi, A.; Hotchkiss, R.S.; Bartenschlager, R. Host-directed therapies for bacterial and viral infections. Nat. Rev. Drug Discov. 2018, 17, 35–56. [Google Scholar] [PubMed]
- Dyck, L.; Mills, K.H.G. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur. J. Immunol. 2017, 47, 765–779. [Google Scholar] [PubMed]
- Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar]
- Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar]
- Korman, A.J.; Peggs, K.S.; Allison, J.P. Checkpoint blockade in cancer immunotherapy. Adv. Immunol. 2006, 90, 297–339. [Google Scholar]
- Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar]
- Andrews, L.P.; Yano, H.; Vignali, D.A.A. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: Breakthroughs or backups. Nat. Immunol. 2019, 20, 1425–1434. [Google Scholar]
- Donisi, C.; Puzzoni, M.; Ziranu, P.; Lai, E.; Mariani, S.; Saba, G.; Impera, V.; Dubois, M.; Persano, M.; Migliari, M.; et al. Immune Checkpoint Inhibitors in the Treatment of HCC. Front. Oncol. 2021, 10, 601240. [Google Scholar]
- Lai, E.; Astara, G.; Ziranu, P.; Pretta, A.; Migliari, M.; Dubois, M.; Donisi, C.; Mariani, S.; Liscia, N.; Impera, V.; et al. Introducing immunotherapy for advanced hepatocellular carcinoma patients: Too early or too fast? Crit. Rev. Oncol. Hematol. 2021, 157, 103167. [Google Scholar]
- Greten, T.F.; Lai, C.W.; Li, G.; Staveley-O’Carroll, K.F. Targeted and Immune-Based Therapies for Hepatocellular Carcinoma. Gastroenterology 2019, 156, 510–524. [Google Scholar]
- Katariya, N.N.; Lizaola-Mayo, B.C.; Chascsa, D.M.; Giorgakis, E.; Aqel, B.A.; Moss, A.A.; Uson Junior, P.L.S.; Borad, M.J.; Mathur, A.K. Immune Checkpoint Inhibitors as Therapy to Down-Stage Hepatocellular Carcinoma Prior to Liver Transplantation. Cancers 2022, 14, 2056. [Google Scholar] [CrossRef] [PubMed]
- Abdelrahim, M.; Esmail, A.; Saharia, A.; Abudayyeh, A.; Abdel-Wahab, N.; Diab, A.; Murakami, N.; Kaseb, A.O.; Chang, J.C.; Gaber, A.O.; et al. Utilization of Immunotherapy for the Treatment of Hepatocellular Carcinoma in the Peri-Transplant Setting: Transplant Oncology View. Cancers 2022, 14, 1760. [Google Scholar] [CrossRef] [PubMed]
- Hewitt, D.B.; Rahnemai-Azar, A.A.; Pawlik, T.M. Potential experimental immune checkpoint inhibitors for the treatment of cancer of the liver. Expert. Opin. Investig. Drugs 2021, 30, 827–835. [Google Scholar]
- Zhang, Y.; Liu, Z.; Tian, M.; Hu, X.; Wang, L.; Ji, J.; Liao, A. The altered PD-1/PD-L1 pathway delivers the ’one-two punch’ effects to promote the Treg/Th17 imbalance in pre-eclampsia. Cell Mol. Immunol. 2018, 15, 710–723. [Google Scholar]
- Kim, S.T.; Chu, Y.; Misoi, M.; Suarez-Almazor, M.E.; Tayar, J.H.; Lu, H.; Buni, M.; Kramer, J.; Rodriguez, E.; Hussain, Z.; et al. Distinct molecular and immune hallmarks of inflammatory arthritis induced by immune checkpoint inhibitors for cancer therapy. Nat. Commun. 2022, 13, 1970. [Google Scholar]
- Okiyama, N.; Tanaka, R. Immune-related adverse events in various organs caused by immune checkpoint inhibitors. Allergol. Int. 2022, 71, 169–178. [Google Scholar]
- Li, S.; Na, R.; Li, X.; Zhang, Y.; Zheng, T. Targeting interleukin-17 enhances tumor response to immune checkpoint inhibitors in colorectal cancer. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188758. [Google Scholar]
- Ramesh, R.; Kozhaya, L.; McKevitt, K.; Djuretic, I.M.; Carlson, T.J.; Quintero, M.A.; McCauley, J.L.; Abreu, M.T.; Unutmaz, D.; Sundrud, M.S. Pro-inflammatory human Th17 cells selectively express P-glycoprotein and are refractory to glucocorticoids. J. Exp. Med. 2014, 211, 89–104. [Google Scholar]
- Melin, A.; Routier, É.; Roy, S.; Pradere, P.; Le Pavec, J.; Pierre, T.; Chanson, N.; Scoazec, J.Y.; Lambotte, O.; Robert, C. Sarcoid-like Granulomatosis Associated with Immune Checkpoint Inhibitors in Melanoma. Cancers 2022, 14, 2937. [Google Scholar] [CrossRef]
- Downs-Canner, S.; Berkey, S.; Delgoffe, G.M.; Edwards, R.P.; Curiel, T.; Odunsi, K.; Bartlett, D.L.; Obermajer, N. Suppressive IL-17A+Foxp3+ and ex-Th17 IL-17AnegFoxp3+ Treg cells are a source of tumour-associated Treg cells. Nat. Commun. 2017, 8, 14649. [Google Scholar]
- Saenz, S.A.; Local, A.; Carr, T.; Shakya, A.; Koul, S.; Hu, H.; Chourb, L.; Stedman, J.; Malley, J.; D’Agostino, L.A.; et al. Small molecule allosteric inhibitors of RORγt block Th17-dependent inflammation and associated gene expression in vivo. PLoS ONE 2021, 16, e0248034. [Google Scholar]
- Kendall, T.; Verheij, J.; Gaudio, E.; Evert, M.; Guido, M.; Goeppert, B.; Carpino, G. Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int. 2019, 39 (Suppl. S1), 7–18. [Google Scholar]
- Yoo, H.J.; Harapan, B.N. Chimeric antigen receptor (CAR) immunotherapy: Basic principles, current advances, and future prospects in neuro-oncology. Immunol. Res. 2021, 69, 471–486. [Google Scholar]
- Huang, R.; Wang, X.; Zhang, X. Unity brings strength: Combination of CAR-T cell therapy and HSCT. Cancer Lett. 2022, 549, 215721. [Google Scholar]
- Chung, H.; Jung, H.; Noh, J.Y. Emerging Approaches for Solid Tumor Treatment Using CAR-T Cell Therapy. Int. J. Mol. Sci. 2021, 22, 12126. [Google Scholar] [CrossRef]
- Sun, B.; Yang, D.; Dai, H.; Liu, X.; Jia, R.; Cui, X.; Li, W.; Cai, C.; Xu, J.; Zhao, X. Eradication of Hepatocellular Carcinoma by NKG2D-Based CAR-T Cells. Cancer Immunol. Res. 2019, 7, 1813–1823. [Google Scholar]
- Jose, A.; Bavetta, M.G.; Martinelli, E.; Bronte, F.; Giunta, E.F.; Manu, K.A. Hepatocellular Carcinoma: Current Therapeutic Algorithm for Localized and Advanced Disease. J. Oncol. 2022, 2022, 3817724. [Google Scholar]
- Howells, A.; Marelli, G.; Lemoine, N.R.; Wang, Y. Oncolytic Viruses-Interaction of Virus and Tumor Cells in the Battle to Eliminate Cancer. Front. Oncol. 2017, 7, 195. [Google Scholar]
- Luo, C.; Wang, P.; He, S.; Zhu, J.; Shi, Y.; Wang, J. Progress and Prospect of Immunotherapy for Triple-Negative Breast Cancer. Front. Oncol. 2022, 12, 919072. [Google Scholar]
- Zhou, Y.; Wang, Q.; Ying, Q.; Zhang, X.; Chen, K.; Ye, T.; Li, G. Effects of Oncolytic Vaccinia Viruses Harboring Different Marine Lectins on Hepatocellular Carcinoma Cells. Int. J. Mol. Sci. 2023, 24, 3823. [Google Scholar] [CrossRef]
- Zheng, X.; Xu, W.; Ying, Q.; Ni, J.; Jia, X.; Zhou, Y.; Ye, T.; Li, G.; Chen, K. Oncolytic Vaccinia Virus Carrying Aphrocallistes vastus Lectin (oncoVV-AVL) Enhances Inflammatory Response in Hepatocellular Carcinoma Cells. Mar. Drugs 2022, 20, 667. [Google Scholar] [CrossRef] [PubMed]
- Qi, X.; Liu, Y.; Hussein, S.; Choi, G.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Li, G. The Species of Gut Bacteria Associated with Antitumor Immunity in Cancer Therapy. Cells 2022, 11, 3684. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Li, L.; Wang, S.; Wei, J.; Qu, L.; Pan, L.; Xu, K. The role of the gut microbiota and probiotics associated with microbial metabolisms in cancer prevention and therapy. Front. Pharmacol. 2022, 13, 1025860. [Google Scholar]
- Luo, M.; Chen, X.; Gao, H.; Yang, F.; Chen, J.; Qiao, Y. Bacteria-mediated cancer therapy: A versatile bio-sapper with translational potential. Front. Oncol. 2022, 12, 980111. [Google Scholar]
- Nejman, D.; Livyatan, I.; Fuks, G.; Gavert, N.; Zwang, Y.; Geller, L.T.; Rotter-Maskowitz, A.; Weiser, R.; Mallel, G.; Gigi, E.; et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Scinence 2020, 368, 973–980. [Google Scholar]
- Kaźmierczak-Siedlecka, K.; Daca, A.; Fic, M.; van de Wetering, T.; Folwarski, M.; Makarewicz, W. Therapeutic methods of gut microbiota modification in colorectal cancer management—Fecal microbiota transplantation, prebiotics, probiotics, and synbiotics. Gut Microbes 2020, 11, 1518–1530. [Google Scholar]
- Ikeda, Y.; Taniguchi, K.; Yoshikawa, S.; Sawamura, H.; Tsuji, A.; Matsuda, S. A budding concept with certain microbiota, anti-proliferative family proteins, and engram theory for the innovative treatment of colon cancer. Explor. Med. 2022, 3, 468–478. [Google Scholar]
- Wang, C.; Li, W.; Wang, H.; Ma, Y.; Zhao, X.; Zhang, X.; Yang, H.; Qian, J.; Li, J. Saccharomyces boulardii alleviates ulcerative colitis carcinogenesis in mice by reducing TNF-α and IL-6 levels and functions and by rebalancing intestinal microbiota. BMC Microbiol. 2019, 19, 246. [Google Scholar]
- Shi, Y.; Xu, L.Z.; Peng, K.; Wu, W.; Wu, R.; Liu, Z.Q.; Yang, G.; Geng, X.R.; Liu, J.; Liu, Z.G.; et al. Specific immunotherapy in combination with Clostridium butyricum inhibits allergic inflammation in the mouse intestine. Sci. Rep. 2015, 5, 17651. [Google Scholar]
- Uchugonova, A.; Zhang, Y.; Salz, R.; Liu, F.; Suetsugu, A.; Zhang, L.; Koenig, K.; Hoffman, R.M.; Zhao, M. Imaging the Different Mechanisms of Prostate Cancer Cell-killing by Tumor-targeting Salmonella typhimurium A1-R. Anticancer Res. 2015, 35, 5225–5229. [Google Scholar]
- Li, L.; You, L.S.; Mao, L.P.; Jin, S.H.; Chen, X.H.; Qian, W.B. Combing oncolytic adenovirus expressing Beclin-1 with chemotherapy agent doxorubicin synergistically enhances cytotoxicity in human CML cells in vitro. Acta Pharmacol. Sin. 2018, 39, 251–260. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Liu, Z.; Sui, X.; Wu, Q.; Wang, J.; Xu, C. Elemene injection as adjunctive treatment to platinum-based chemotherapy in patients with stage III/IV non-small cell lung cancer: A meta-analysis following the PRISMA guidelines. Phytomedicine 2019, 59, 152787. [Google Scholar] [PubMed]
- Din, M.O.; Danino, T.; Prindle, A.; Skalak, M.; Selimkhanov, J.; Allen, K.; Julio, E.; Atolia, E.; Tsimring, L.S.; Bhatia, S.N.; et al. Synchronized cycles of bacterial lysis for in vivo delivery. Nature 2016, 536, 81–85. [Google Scholar] [CrossRef] [PubMed]
- McNerney, M.P.; Doiron, K.E.; Ng, T.L.; Chang, T.Z.; Silver, P.A. Theranostic cells: Emerging clinical applications of synthetic biology. Nat. Rev. Genet. 2021, 22, 730–746. [Google Scholar]
- Lu, Y.; Luo, X.; Yang, D.; Li, Y.; Gong, T.; Li, B.; Cheng, J.; Chen, R.; Guo, X.; Yuan, W. Effects of probiotic supplementation on related side effects after chemoradiotherapy in cancer patients. Front. Oncol. 2022, 12, 1032145. [Google Scholar] [CrossRef]
- Burdelya, L.G.; Krivokrysenko, V.I.; Tallant, T.C.; Strom, E.; Gleiberman, A.S.; Gupta, D.; Kurnasov, O.V.; Fort, F.L.; Osterman, A.L.; Didonato, J.A.; et al. An agonist of toll-like receptor 5 has radioprotective activity in mouse and primate models. Science 2008, 320, 226–230. [Google Scholar]
- Abdollahi, H. Beneficial effects of cellular autofluorescence following ionization radiation: Hypothetical approaches for radiation protection and enhancing radiotherapy effectiveness. Med. Hypotheses 2015, 84, 194–198. [Google Scholar]
- Bettegowda, C.; Dang, L.H.; Abrams, R.; Huso, D.L.; Dillehay, L.; Cheong, I.; Agrawal, N.; Borzillary, S.; McCaffery, J.M.; Watson, E.L.; et al. Overcoming the hypoxic barrier to radiation therapy with anaerobic bacteria. Proc. Natl. Acad. Sci. USA 2003, 100, 15083–15088. [Google Scholar]
- Poonacha, K.N.T.; Villa, T.G.; Notario, V. The Interplay among Radiation Therapy, Antibiotics and the Microbiota: Impact on Cancer Treatment Outcomes. Antibiotics 2022, 11, 331. [Google Scholar] [CrossRef]
- Lee, S.H.; Cho, S.Y.; Yoon, Y.; Park, C.; Sohn, J.; Jeong, J.J.; Jeon, B.N.; Jang, M.; An, C.; Lee, S.; et al. Bifidobacterium bifidum strains synergize with immune checkpoint inhibitors to reduce tumour burden in mice. Nat. Microbiol. 2021, 6, 277–288. [Google Scholar]
- Zhuo, Q.; Yu, B.; Zhou, J.; Zhang, J.; Zhang, R.; Xie, J.; Wang, Q.; Zhao, S. Lysates of Lactobacillus acidophilus combined with CTLA-4-blocking antibodies enhance antitumor immunity in a mouse colon cancer model. Sci. Rep. 2019, 9, 20128. [Google Scholar]
- Ahmadi Badi, S.; Moshiri, A.; Ettehad Marvasti, F.; Mojtahedzadeh, M.; Kazemi, V.; Siadat, S.D. Extraction and Evaluation of Outer Membrane Vesicles from Two Important Gut Microbiota Members, Bacteroides fragilis and Bacteroides thetaiotaomicron. Cell J. 2020, 22, 344–349. [Google Scholar] [PubMed]
- Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [PubMed]
- Zhang, M.; Qiu, X.; Zhang, H.; Yang, X.; Hong, N.; Yang, Y.; Chen, H.; Yu, C. Faecalibacterium prausnitzii inhibits interleukin-17 to ameliorate colorectal colitis in rats. PLoS ONE 2014, 9, e109146. [Google Scholar]
- Zhou, L.; Zhang, M.; Wang, Y.; Dorfman, R.G.; Liu, H.; Yu, T.; Chen, X.; Tang, D.; Xu, L.; Yin, Y.; et al. Faecalibacterium prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase. Inflamm. Bowel Dis. 2018, 24, 1926–1940. [Google Scholar] [CrossRef]
- Kaźmierczak-Siedlecka, K.; Skonieczna-Żydecka, K.; Hupp, T.; Duchnowska, R.; Marek-Trzonkowska, N.; Połom, K. Next-generation probiotics—Do they open new therapeutic strategies for cancer patients? Gut Microbes 2022, 14, 2035659. [Google Scholar]
- Behary, J.; Raposo, A.E.; Amorim, N.M.L.; Zheng, H.; Gong, L.; McGovern, E.; Chen, J.; Liu, K.; Beretov, J.; Theocharous, C.; et al. Defining the temporal evolution of gut dysbiosis and inflammatory responses leading to hepatocellular carcinoma in Mdr2 −/− mouse model. BMC Microbiol. 2021, 21, 113. [Google Scholar]
- Ponziani, F.R.; Bhoori, S.; Castelli, C.; Putignani, L.; Rivoltini, L.; Del Chierico, F.; Sanguinetti, M.; Morelli, D.; Paroni Sterbini, F.; Petito, V.; et al. Hepatocellular Carcinoma Is Associated With Gut Microbiota Profile and Inflammation in Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 107–120. [Google Scholar] [CrossRef]
- Zhang, X.; Coker, O.O.; Chu, E.S.; Fu, K.; Lau, H.C.H.; Wang, Y.X.; Chan, A.W.H.; Wei, H.; Yang, X.; Sung, J.J.Y.; et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut 2021, 70, 761–774. [Google Scholar]
- Rezasoltani, S.; Yadegar, A.; Asadzadeh Aghdaei, H.; Reza Zali, M. Modulatory effects of gut microbiome in cancer immunotherapy: A novel paradigm for blockade of immune checkpoint inhibitors. Cancer Med. 2021, 10, 1141–1154. [Google Scholar]
- Wu, Y.; Zheng, L. Dynamic education of macrophages in different areas of human tumors. Cancer Microenviron. 2012, 5, 195–201. [Google Scholar] [PubMed]
- Ahmed, F.; Steele, J.C.; Herbert, J.M.; Steven, N.M.; Bicknell, R. Tumor stroma as a target in cancer. Curr. Cancer Drug Targets 2008, 8, 447–453. [Google Scholar] [PubMed]
- Zhang, J.P.; Yan, J.; Xu, J.; Pang, X.H.; Chen, M.S.; Li, L.; Wu, C.; Li, S.P.; Zheng, L. Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients. J. Hepatol. 2009, 50, 980–989. [Google Scholar] [CrossRef]
- Zhao, F.; Hoechst, B.; Gamrekelashvili, J.; Ormandy, L.A.; Voigtländer, T.; Wedemeyer, H.; Ylaya, K.; Wang, X.W.; Hewitt, S.M.; Manns, M.P.; et al. Human CCR4+ CCR6+ Th17 cells suppress autologous CD8+ T cell responses. J. Immunol. 2012, 188, 6055–6062. [Google Scholar]
- Liao, R.; Sun, J.; Wu, H.; Yi, Y.; Wang, J.X.; He, H.W.; Cai, X.Y.; Zhou, J.; Cheng, Y.F.; Fan, J.; et al. High expression of IL-17 and IL-17RE associate with poor prognosis of hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2013, 32, 3. [Google Scholar]
- Kuang, D.M.; Peng, C.; Zhao, Q.; Wu, Y.; Chen, M.S.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma promote expansion of memory T helper 17 cells. Hepatology 2010, 51, 154–164. [Google Scholar] [CrossRef]
- Michaelis, L.; Treß, M.; Löw, H.C.; Klees, J.; Klameth, C.; Lange, A.; Grießhammer, A.; Schäfer, A.; Menz, S.; Steimle, A.; et al. Gut Commensal-Induced IκBζ Expression in Dendritic Cells Influences the Th17 Response. Front. Immunol. 2021, 11, 612336. [Google Scholar]
- Sung, C.Y.; Lee, N.P.; El-Nezami, H. Regulation of T helper 17 by bacteria: An approach for the treatment of hepatocellular carcinoma. Int. J. Hepatol. 2012, 2012, 439024. [Google Scholar]
- Gu, F.M.; Li, Q.L.; Gao, Q.; Jiang, J.H.; Zhu, K.; Huang, X.Y.; Pan, J.F.; Yan, J.; Hu, J.H.; Wang, Z.; et al. IL-17 induces AKT-dependent IL-6/JAK2/STAT3 activation and tumor progression in hepatocellular carcinoma. Mol. Cancer 2011, 10, 150. [Google Scholar]
- Qin, H.; Yuan, B.; Huang, W.; Wang, Y. Utilizing Gut Microbiota to Improve Hepatobiliary Tumor Treatments: Recent Advances. Front. Oncol. 2022, 12, 924696. [Google Scholar]
- Mikó, E.; Vida, A.; Bai, P. Translational aspects of the microbiome-to be exploited. Cell Biol. Toxicol. 2016, 32, 153–156. [Google Scholar] [PubMed]
- Kovács, T.; Mikó, E.; Vida, A.; Sebő, É.; Toth, J.; Csonka, T.; Boratkó, A.; Ujlaki, G.; Lente, G.; Kovács, P.; et al. Cadaverine, a metabolite of the microbiome, reduces breast cancer aggressiveness through trace amino acid receptors. Sci. Rep. 2019, 9, 1300. [Google Scholar]
- Yu, L.X.; Schwabe, R.F. The gut microbiome and liver cancer: Mechanisms and clinical translation. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 527–539. [Google Scholar] [PubMed]
- Yuan, J.; Chen, C.; Cui, J.; Lu, J.; Yan, C.; Wei, X.; Zhao, X.; Li, N.; Li, S.; Xue, G.; et al. Fatty Liver Disease Caused by High-Alcohol-Producing Klebsiella pneumoniae. Cell Metab. 2019, 30, 675–688.e7. [Google Scholar]
- Lapidot, Y.; Amir, A.; Nosenko, R.; Uzan-Yulzari, A.; Veitsman, E.; Cohen-Ezra, O.; Davidov, Y.; Weiss, P.; Bradichevski, T.; Segev, S.; et al. Alterations in the Gut Microbiome in the Progression of Cirrhosis to Hepatocellular Carcinoma. mSystems 2020, 5, e00153-20. [Google Scholar]
- Ma, C.; Han, M.; Heinrich, B.; Fu, Q.; Zhang, Q.; Sandhu, M.; Agdashian, D.; Terabe, M.; Berzofsky, J.A.; Fako, V.; et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018, 360, eaan5931. [Google Scholar]
- 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]
- 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]
- 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]
- Yoshimoto, S.; Loo, T.M.; Atarashi, K.; Kanda, H.; Sato, S.; Oyadomari, S.; Iwakura, Y.; Oshima, K.; Morita, H.; Hattori, M.; et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013, 499, 97–101. [Google Scholar] [CrossRef]
- Dapito, D.H.; Mencin, A.; Gwak, G.Y.; Pradere, J.P.; Jang, M.K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Fernandes, M.R.; Aggarwal, P.; Costa, R.G.F.; Cole, A.M.; Trinchieri, G. Targeting the gut microbiota for cancer therapy. Nat. Rev. Cancer 2022, 22, 703–722. [Google Scholar] [CrossRef]
- Dai, E.; Zhu, Z.; Wahed, S.; Qu, Z.; Storkus, W.J.; Guo, Z.S. Epigenetic modulation of antitumor immunity for improved cancer immunotherapy. Mol. Cancer 2021, 20, 171. [Google Scholar] [CrossRef]
- Burr, M.L.; Sparbier, C.E.; Chan, K.L.; Chan, Y.C.; Kersbergen, A.; Lam, E.Y.N.; Azidis-Yates, E.; Vassiliadis, D.; Bell, C.C.; Gilan, O.; et al. An evolutionarily conserved function of polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell 2019, 36, 385–401. e8. [Google Scholar] [CrossRef]
- Greger, V.; Passarge, E.; Höpping, W.; Messmer, E.; Horsthemke, B. Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma. Hum. Genet. 1989, 83, 155–158. [Google Scholar] [CrossRef]
- Saito, Y.; Liang, G.; Egger, G.; Friedman, J.M.; Chuang, J.C.; Coetzee, G.A.; Jones, P.A. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell 2006, 9, 435–443. [Google Scholar] [CrossRef]
- Senga, S.S.; Grose, R.P. Hallmarks of cancer-the new testament. Open Biol. 2021, 11, 200358. [Google Scholar] [CrossRef]
- Wang, G.; He, X.; Wang, Q. Intratumoral bacteria are an important “accomplice” in tumor development and metastasis. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188846. [Google Scholar] [CrossRef]
- Yang, F.; Yang, Y.; Chen, L.; Zhang, Z.; Liu, L.; Zhang, C.; Mai, Q.; Chen, Y.; Chen, Z.; Lin, T.; et al. The gut microbiota mediates protective immunity against tuberculosis via modulation of lncRNA. Gut Microbes 2022, 14, 2029997. [Google Scholar] [CrossRef]
- Niller, H.H.; Minarovits, J. Patho-epigenetics of Infectious Diseases Caused by Intracellular Bacteria. Adv. Exp. Med. Biol. 2016, 879, 107–130. [Google Scholar] [PubMed]
- Luu, M.; Schütz, B.; Lauth, M.; Visekruna, A. The Impact of Gut Microbiota-Derived Metabolites on the Tumor Immune Microenvironment. Cancers 2023, 15, 1588. [Google Scholar] [CrossRef] [PubMed]
- Cerf-Bensussan, N.; Gaboriau-Routhiau, V. The immune system and the gut microbiota: Friends or foes? Nat. Rev. Immunol. 2010, 10, 735–744. [Google Scholar]
- Ledezma, D.K.; Balakrishnan, P.B.; Cano-Mejia, J.; Sweeney, E.E.; Hadley, M.; Bollard, C.M.; Villagra, A.; Fernandes, R. Indocyanine Green-Nexturastat A-PLGA Nanoparticles Combine Photothermal and Epigenetic Therapy for Melanoma. Nanomaterials 2020, 10, 161. [Google Scholar] [CrossRef]
- Wachowska, M.; Gabrysiak, M.; Muchowicz, A.; Bednarek, W.; Barankiewicz, J.; Rygiel, T.; Boon, L.; Mroz, P.; Hamblin, M.R.; Golab, J. 5-Aza-2′-deoxycytidine potentiates antitumour immune response induced by photodynamic therapy. Eur. J. Cancer 2014, 50, 1370–1381. [Google Scholar] [CrossRef]
- Wachowska, M.; Muchowicz, A.; Golab, J. Targeting Epigenetic Processes in Photodynamic Therapy-Induced Anticancer Immunity. Front. Oncol. 2015, 5, 176. [Google Scholar]
- Trifylli, E.M.; Koustas, E.; Papadopoulos, N.; Sarantis, P.; Aloizos, G.; Damaskos, C.; Garmpis, N.; Garmpi, A.; Karamouzis, M.V. An Insight into the Novel Immunotherapy and Targeted Therapeutic Strategies for Hepatocellular Carcinoma and Cholangiocarcinoma. Life 2022, 12, 665. [Google Scholar] [CrossRef]
- Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar]
- Pace, F.; Macchini, F.; Castagna, V.M. Safety of probiotics in humans: A dark side revealed? Dig. Liver Dis. 2020, 52, 981–985. [Google Scholar]
- Wallace, C.; Sinopoulou, V.; Gordon, M.; Akobeng, A.K.; Llanos-Chea, A.; Hungria, G.; Febo-Rodriguez, L.; Fifi, A.; Fernandez Valdes, L.; Langshaw, A.; et al. Probiotics for treatment of chronic constipation in children. Cochrane Database Syst. Rev. 2022, 3, CD014257. [Google Scholar]
- Sun, F.; Zhang, Q.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. A potential species of next-generation probiotics? The dark and light sides of Bacteroides fragilis in health. Food Res. Int. 2019, 126, 108590. [Google Scholar] [PubMed]
- Bayer, A.S.; Chow, A.W.; Betts, D.; Guze, L.B. Lactobacillemia--report of nine cases. Important clinical and therapeutic considerations. Am. J. Med. 1978, 64, 808–813. [Google Scholar] [PubMed]
- Dickgiesser, U.; Weiss, N.; Fritsche, D. Lactobacillus gasseri as the cause of septic urinary infection. Infection 1984, 12, 14–16. [Google Scholar] [PubMed]
- Tan, K.P.; Yang, M.; Ito, S. Activation of nuclear factor (erythroid-2 like) factor 2 by toxic bile acids provokes adaptive defense responses to enhance cell survival at the emergence of oxidative stress. Mol. Pharmacol. 2007, 72, 1380–1390. [Google Scholar]
- Hiam-Galvez, K.J.; Allen, B.M.; Spitzer, M.H. Systemic immunity in cancer. Nat. Rev. Cancer 2021, 21, 345–359. [Google Scholar]
- Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC strategy for prognosis prediction and treatment recommendation: The 2022 update. J. Hepatol. 2022, 76, 681–693. [Google Scholar]
- Jiang, X.; Zheng, J.; Zhang, S.; Wang, B.; Wu, C.; Guo, X. Advances in the Involvement of Gut Microbiota in Pathophysiology of NAFLD. Front. Med. 2020, 7, 361. [Google Scholar]
- Alexander, J.L.; Wilson, I.D.; Teare, J.; Marchesi, J.R.; Nicholson, J.K.; Kinross, J.M. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 356–365. [Google Scholar]
- Mukaida, N. Intestinal microbiota: Unexpected alliance with tumor therapy. Immunotherapy 2014, 6, 231–233. [Google Scholar]
- Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar]
- Pagadala, M.; Sears, T.J.; Wu, V.H.; Pérez-Guijarro, E.; Kim, H.; Castro, A.; Talwar, J.V.; Gonzalez-Colin, C.; Cao, S.; Schmiedel, B.J.; et al. Germline modifiers of the tumor immune microenvironment implicate drivers of cancer risk and immunotherapy response. Nat. Commun. 2023, 14, 2744. [Google Scholar] [PubMed]
- Yoshikawa, S.; Taniguchi, K.; Sawamura, H.; Ikeda, Y.; Tsuji, A.; Matsuda, S. Encouraging probiotics for the prevention and treatment of immune-related adverse events in novel immunotherapies against malignant glioma. Explor. Target. Antitumor Ther. 2022, 3, 817–827. [Google Scholar] [CrossRef] [PubMed]
- Ayob, A.Z.; Ramasamy, T.S. Cancer stem cells as key drivers of tumour progression. J. Biomed. Sci. 2018, 25, 20. [Google Scholar]
- Garza Treviño, E.N.; González, P.D.; Valencia Salgado, C.I.; Martinez Garza, A. Effects of pericytes and colon cancer stem cells in the tumor microenvironment. Cancer Cell Int. 2019, 19, 173. [Google Scholar]
- Yoshikawa, S.; Taniguchi, K.; Sawamura, H.; Ikeda, Y.; Tsuji, A.; Matsuda, S. A New Concept of Associations between Gut Microbiota, Immunity and Central Nervous System for the Innovative Treatment of Neurodegenerative Disorders. Metabolites 2022, 12, 1052. [Google Scholar] [CrossRef]
- Abenavoli, L.; Maurizi, V.; Rinninella, E.; Tack, J.; Di Berardino, A.; Santori, P.; Rasetti, C.; Procopio, A.C.; Boccuto, L.; Scarpellini, E. Fecal Microbiota Transplantation in NAFLD Treatment. Medicina 2022, 58, 1559. [Google Scholar] [CrossRef]
- Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The Severity of Nonalcoholic Fatty Liver Disease Is Associated with Gut Dysbiosis and Shift in the Metabolic Function of the Gut Microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 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
Nakashima, M.; Fukumoto, A.; Matsuda, S. Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma. Diseases 2025, 13, 111. https://doi.org/10.3390/diseases13040111
Nakashima M, Fukumoto A, Matsuda S. Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma. Diseases. 2025; 13(4):111. https://doi.org/10.3390/diseases13040111
Chicago/Turabian StyleNakashima, Moeka, Akari Fukumoto, and Satoru Matsuda. 2025. "Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma" Diseases 13, no. 4: 111. https://doi.org/10.3390/diseases13040111
APA StyleNakashima, M., Fukumoto, A., & Matsuda, S. (2025). Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma. Diseases, 13(4), 111. https://doi.org/10.3390/diseases13040111