New Approaches for T Cell Immunomodulation

Eman Bahattab ( National Center for Biotechnology. King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia )

Khalid Shah ( BWH Center of Excellence for Biomedicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. Center for Stem Cell Therapeutics and Imaging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA )

Mohannad Fallatah ( Center of Excellence for Biomedicine. National Center for Biotechnology. King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia )

https://doi.org/10.37155/2717-5278-0301-3

Abstract

Cancer is one of the leading causes of death worldwide. Despite the evolution in cancer treatment, the mortality rate is still high. The astonishing results of immune checkpoint inhibitors in patients bearing solid tumors have encouraged scientists to develop new strategies for cancer immunotherapy to further improve the clinical outcomes. Immunomodulation for cancer therapy is growing rapidly as a promising alternative approach for conventional cancer therapeutics, due to its specificity and efficacy in suppressing growth and metastasis. However, the complexity of the tumor microenvironment and the various mechanisms whereby tumor cells escape the immune response diminishes the efficacy of many cancer immunotherapeutic drugs, especially when the drug is administered as monotherapy. Some of these factors include poor immune recognition of tumor cells, lack of sufficient drug concentration within the tumor site, and suppression of tumor-reactive T cell proliferation and effector functions. This review describes some of the novel immunomodulation strategies that have been recently developed to overcome these limitations and to augment strong antitumor immune response against different tumor types. For example, monoclonal antibodies (mAbs), CAR- T cells, Bi-specific T-cell engagers (BiTE) antibody, oncolytic viruses (OVs), cytokine-antibody fusion protein, genetically engineered mesenchymal stem cells (MSCs), oncolytic virotherapy, gut microbiota modulation and nanoparticles mediated cancer therapy. These novel strategies have been proven to augment strong antitumor immune response against different tumor types, reduced toxicity, as well as generated long-lived memory T cell population that was able to protect the host from tumor relapse. Combination therapies either with conventional or immunotherapy further improved the efficacy of cancer treatment.

Keywords

Immunomodulation; Checkpoint inhibitors; Co-stimulatory receptors; Bispecific antibody; Cytokine/Antibody fusion; Bi-specific T-cell engagers (BiTE) antibody; Oncolytic virus (OVs); Genetically engineered mesenchymal stem cells (MSCs); Nano-based vaccine and Gut microbiota

Full Text

PDF

References

Walsh SR, Simovic B, Chen L, Bastin D, Nguyen A, Stephenson K, et al. Endogenous T cells prevent tumor immune escape following adoptive T cell therapy. J Clin Invest. 2019;129(12):5400-10.
[2]. Li D, Li X, Zhou W-L, Huang Y, Liang X, Jiang L, et al. Genetically engineered T cells for cancer immunotherapy. Signal Transduction and Targeted Therapy. 2019;4(1):35.
[3]. Durgeau A, Virk Y, Corgnac S, Mami-Chouaib F. Recent Advances in Targeting CD8 T-Cell Immunity for More Effective Cancer Immunotherapy. Frontiers in Immunology. 2018;9(14).
[4]. Dyck L, Mills KHG. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur J Immunol. 2017;47(5):765-79.
[5]. Donini C, D'Ambrosio L, Grignani G, Aglietta M, Sangiolo D. Next generation immune-checkpoints for cancer therapy. J Thorac Dis. 2018;10(Suppl 13):S1581-S601.
[6]. Velcheti V, Schalper K. Basic Overview of Current Immunotherapy Approaches in Cancer. Am Soc Clin Oncol Educ Book. 2016;35:298-308.
[7]. Gun SY, Lee SWL, Sieow JL, Wong SC. Targeting immune cells for cancer therapy. Redox Biol. 2019;25:101174-.
[8]. Wu L, Yun Z, Tagawa T, Rey-McIntyre K, de Perrot M. CTLA-4 Blockade Expands Infiltrating T Cells and Inhibits Cancer Cell Repopulation during the Intervals of Chemotherapy in Murine Mesothelioma. Molecular Cancer Therapeutics. 2012;11(8):1809-19.
[9]. Tarhini A. Immune-mediated adverse events associated with ipilimumab ctla-4 blockade therapy: the underlying mechanisms and clinical management. Scientifica (Cairo). 2013;2013:857519-.
[10]. Zhao Y, Yang W, Huang Y, Cui R, Li X, Li B. Evolving Roles for Targeting CTLA-4 in Cancer Immunotherapy. Cell Physiol Biochem. 2018;47(2):721-34.
[11]. Riley JL. PD-1 signaling in primary T cells. Immunological reviews. 2009;229(1):114-25.
[12]. Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 Blockade in Cancer Immunotherapy. Comput Struct Biotechnol J. 2019;17:661-74.
[13]. Lee HT, Lee SH, Heo YS. Molecular Interactions of Antibody Drugs Targeting PD-1, PD-L1, and CTLA-4 in Immuno-Oncology. Molecules. 2019;24(6).
[14]. Sundar R, Cho B-C, Brahmer JR, Soo RA. Nivolumab in NSCLC: latest evidence and clinical potential. Ther Adv Med Oncol. 2015;7(2):85-96.
[15]. Chae YK, Arya A, Iams W, Cruz MR, Chandra S, Choi J, et al. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). Journal for ImmunoTherapy of Cancer. 2018;6(1):39.
[16]. Rotte A. Combination of CTLA-4 and PD-1 blockers for treatment of cancer. Journal of Experimental & Clinical Cancer Research. 2019;38(1):255.
[17]. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med. 2019;381(16):1535-46.
[18]. Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am Soc Clin Oncol Educ Book. 2019;39:147-64.
[19]. Heath WR, Belz GT, Behrens GM, Smith CM, Forehan SP, Parish IA, et al. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol Rev. 2004;199:9-26.
[20]. Bousso P. T-cell activation by dendritic cells in the lymph node: lessons from the movies. Nat Rev Immunol. 2008;8(9):675-84.
[21]. Ward-Kavanagh LK, Lin WW, Sedy JR, Ware CF. The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity. 2016;44(5):1005-19.
[22]. Linsley PS, Ledbetter JA. The role of the CD28 receptor during T cell responses to antigen. Annu Rev Immunol. 1993;11:191-212.
[23]. Attanasio J, Wherry EJ. Costimulatory and Coinhibitory Receptor Pathways in Infectious Disease. Immunity. 2016;44(5):1052-68.
[24]. Buchan SL, Fallatah M, Thirdborough SM, Taraban VY, Rogel A, Thomas LJ, et al. PD-1 Blockade and CD27 Stimulation Activate Distinct Transcriptional Programs That Synergize for CD8(+) T-Cell-Driven Antitumor Immunity. Clinical cancer research : an official journal of the American Association for Cancer Research. 2018;24(10):2383-94.
[25]. Huang J, Jochems C, Anderson AM, Talaie T, Jales A, Madan RA, et al. Soluble CD27-pool in humans may contribute to T cell activation and tumor immunity. Journal of immunology (Baltimore, Md : 1950). 2013;190(12):6250-8.
[26]. Buchan SL, Dou L, Remer M, Booth SG, Dunn SN, Lai C, et al. Antibodies to Costimulatory Receptor 4-1BB Enhance Anti-tumor Immunity via T Regulatory Cell Depletion and Promotion of CD8 T Cell Effector Function. Immunity. 2018;49(5):958-70.e7.
[27]. Pan PY, Zang Y, Weber K, Meseck ML, Chen SH. OX40 ligation enhances primary and memory cytotoxic T lymphocyte responses in an immunotherapy for hepatic colon metastases. Mol Ther. 2002;6(4):528-36.
[28]. Reiss KA, Forde PM, Brahmer JR. Harnessing the power of the immune system via blockade of PD-1 and PD-L1: a promising new anticancer strategy. Immunotherapy. 2014;6(4):459-75.
[29]. Burris HA, Infante JR, Ansell SM, Nemunaitis JJ, Weiss GR, Villalobos VM, et al. Safety and Activity of Varlilumab, a Novel and First-in-Class Agonist Anti-CD27 Antibody, in Patients With Advanced Solid Tumors. J Clin Oncol. 2017;35(18):2028-36.
[30]. Hintzen RQ, de Jong R, Lens SM, Brouwer M, Baars P, van Lier RA. Regulation of CD27 expression on subsets of mature T-lymphocytes. The Journal of Immunology. 1993;151(5):2426-35.
[31]. Hayakawa Y, Smyth MJ. CD27 Dissects Mature NK Cells into Two Subsets with Distinct Responsiveness and Migratory Capacity. The Journal of Immunology. 2006;176(3):1517-24.
[32]. Prasad KV, Ao Z, Yoon Y, Wu MX, Rizk M, Jacquot S, et al. CD27, a member of the tumor necrosis factor receptor family, induces apoptosis and binds to Siva, a proapoptotic protein. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(12):6346-51.
[33]. Wu YC, Kipling D, Dunn-Walters DK. The relationship between CD27 negative and positive B cell populations in human peripheral blood. Front Immunol. 2011;2:81.
[34]. Turaj AH, Hussain K, Cox KL, Rose-Zerilli MJJ, Testa J, Dahal LN, et al. Antibody Tumor Targeting Is Enhanced by CD27 Agonists through Myeloid Recruitment. Cancer Cell. 2017;32(6):777-91.e6.
[35]. Buchan SL, Fallatah M, Thirdborough SM, Taraban VY, Rogel A, Thomas LJ, et al. PD-1 Blockade and CD27 Stimulation Activate Distinct Transcriptional Programs That Synergize for CD8(+) T-Cell-Driven Antitumor Immunity. Clin Cancer Res. 2018;24(10):2383-94.
[36]. Wasiuk A, Testa J, Weidlick J, Sisson C, Vitale L, Widger J, et al. CD27-Mediated Regulatory T Cell Depletion and Effector T Cell Costimulation Both Contribute to Antitumor Efficacy. J Immunol. 2017;199(12):4110-23.
[37]. Aspeslagh S, Postel-Vinay S, Rusakiewicz S, Soria JC, Zitvogel L, Marabelle A. Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer. 2016;52:50-66.
[38]. Voo KS, Bover L, Harline ML, Vien LT, Facchinetti V, Arima K, et al. Antibodies targeting human OX40 expand effector T cells and block inducible and natural regulatory T cell function. J Immunol. 2013;191(7):3641-50.
[39]. Polesso F, Sarker M, Weinberg AD, Murray SE, Moran AE. OX40 Agonist Tumor Immunotherapy Does Not Impact Regulatory T Cell Suppressive Function. The Journal of Immunology. 2019:ji1900696.
[40]. Linch SN, McNamara MJ, Redmond WL. OX40 Agonists and Combination Immunotherapy: Putting the Pedal to the Metal. Frontiers in Oncology. 2015;5(34).
[41]. Weinberg AD, Rivera MM, Prell R, Morris A, Ramstad T, Vetto JT, et al. Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol. 2000;164(4):2160-9.
[42]. Peng W, Williams LJ, Xu C, Melendez B, McKenzie JA, Chen Y, et al. Anti-OX40 Antibody Directly Enhances The Function of Tumor-Reactive CD8(+) T Cells and Synergizes with PI3Kbeta Inhibition in PTEN Loss Melanoma. Clin Cancer Res. 2019;25(21):6406-16.
[43]. Oberst MD, Auge C, Morris C, Kentner S, Mulgrew K, McGlinchey K, et al. Potent Immune Modulation by MEDI6383, an Engineered Human OX40 Ligand IgG4P Fc Fusion Protein. Mol Cancer Ther. 2018;17(5):1024-38.
[44]. Kvarnhammar AM, Veitonmaki N, Hagerbrand K, Dahlman A, Smith KE, Fritzell S, et al. The CTLA-4 x OX40 bispecific antibody ATOR-1015 induces anti-tumor effects through tumor-directed immune activation. J Immunother Cancer. 2019;7(1):103.
[45]. Knödler M, Körfer J, Kunzmann V, Trojan J, Daum S, Schenk M, et al. Randomised phase II trial to investigate catumaxomab (anti-EpCAM × anti-CD3) for treatment of peritoneal carcinomatosis in patients with gastric cancer. British Journal of Cancer. 2018;119(3):296-302.
[46]. Oberholzer A, Oberholzer C, Moldawer LL. Cytokine signaling--regulation of the immune response in normal and critically ill states. Crit Care Med. 2000;28(4 Suppl):N3-12.
[47]. Robinson TO, Schluns KS. The potential and promise of IL-15 in immuno-oncogenic therapies. Immunol Lett. 2017;190:159-68.
[48]. Hu Q, Ye X, Qu X, Cui D, Zhang L, Xu Z, et al. Discovery of a novel IL-15 based protein with improved developability and efficacy for cancer immunotherapy. Scientific Reports. 2018;8(1):7675.
[49]. Schwartz RN, Stover L, Dutcher JP. Managing toxicities of high-dose interleukin-2. Oncology (Williston Park). 2002;16(11 Suppl 13):11-20.
[50]. Panelli MC, White R, Foster M, Martin B, Wang E, Smith K, et al. Forecasting the cytokine storm following systemic interleukin (IL)-2 administration. J Transl Med. 2004;2(1):17-.
[51]. Fehniger TA, Suzuki K, Ponnappan A, VanDeusen JB, Cooper MA, Florea SM, et al. Fatal leukemia in interleukin 15 transgenic mice follows early expansions in natural killer and memory phenotype CD8+ T cells. J Exp Med. 2001;193(2):219-31.
[52]. Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology. 2006;117(4):433-42.
[53]. Neri D. Antibody-Cytokine Fusions: Versatile Products for the Modulation of Anticancer Immunity. Cancer Immunol Res. 2019;7(3):348-54.
[54]. Hutmacher C, Neri D. Antibody-cytokine fusion proteins: Biopharmaceuticals with immunomodulatory properties for cancer therapy. Adv Drug Deliv Rev. 2019;141:67-91.
[55]. Gillies SD, Lo KM, Burger C, Lan Y, Dahl T, Wong WK. Improved circulating half-life and efficacy of an antibody-interleukin 2 immunocytokine based on reduced intracellular proteolysis. Clin Cancer Res. 2002;8(1):210-6.
[56]. Gutbrodt KL, Schliemann C, Giovannoni L, Frey K, Pabst T, Klapper W, et al. Antibody-Based Delivery of Interleukin-2 to Neovasculature Has Potent Activity Against Acute Myeloid Leukemia. Science Translational Medicine. 2013;5(201):201ra118-201ra118.
[57]. Ziffels B, Stringhini M, Probst P, Fugmann T, Sturm T, Neri D. Antibody-Based Delivery of Cytokine Payloads to Carbonic Anhydrase IX Leads to Cancer Cures in Immunocompetent Tumor-Bearing Mice. Mol Cancer Ther. 2019;18(9):1544-54.
[58]. Chen S, Huang Q, Liu J, Xing J, Zhang N, Liu Y, et al. A targeted IL-15 fusion protein with potent anti-tumor activity. Cancer Biol Ther. 2015;16(9):1415-21.
[59]. Jochems C, Tritsch SR, Knudson KM, Gameiro SR, Rumfield CS, Pellom ST, et al. The multi-functionality of N-809, a novel fusion protein encompassing anti-PD-L1 and the IL-15 superagonist fusion complex. Oncoimmunology. 2019;8(2):e1532764.
[60]. Srivastava S, Riddell SR. Engineering CAR-T cells: Design concepts. Trends Immunol. 2015;36(8):494-502.
[61]. Guedan S, Calderon H, Posey AD, Jr., Maus MV. Engineering and Design of Chimeric Antigen Receptors. Mol Ther Methods Clin Dev. 2019;12:145-56.
[62]. Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 2019;94(S1):S3-s9.
[63]. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. New England Journal of Medicine. 2017;377(26):2531-44.
[64]. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. New England Journal of Medicine. 2018;378(5):439-48.
[65]. Frey N, Porter D. Cytokine Release Syndrome with Chimeric Antigen Receptor T Cell Therapy. Biol Blood Marrow Transplant. 2019;25(4):e123-e7.
[66]. Hunter BD, Jacobson CA. CAR T-Cell Associated Neurotoxicity: Mechanisms, Clinicopathologic Correlates, and Future Directions. J Natl Cancer Inst. 2019;111(7):646-54.
[67]. Rodríguez JA. HLA-mediated tumor escape mechanisms that may impair immunotherapy clinical outcomes via T-cell activation. Oncol Lett. 2017;14(4):4415-27.
[68]. Garrido F, Aptsiauri N, Doorduijn EM, Garcia Lora AM, van Hall T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr Opin Immunol. 2016;39:44-51.
[69]. Zhukovsky EA, Morse RJ, Maus MV. Bispecific antibodies and CARs: generalized immunotherapeutics harnessing T cell redirection. Curr Opin Immunol. 2016;40:24-35.
[70]. Smits NC, Sentman CL. Bispecific T-Cell Engagers (BiTEs) as Treatment of B-Cell Lymphoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(10):1131-3.
[71]. Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321(5891):974-7.
[72]. Stein A, Franklin JL, Chia VM, Arrindell D, Kormany W, Wright J, et al. Benefit-Risk Assessment of Blinatumomab in the Treatment of Relapsed/Refractory B-Cell Precursor Acute Lymphoblastic Leukemia. Drug Saf. 2019;42(5):587-601.
[73]. Orange M, Reuter U, Hobohm U. Coley's Lessons Remembered: Augmenting Mistletoe Therapy. Integr Cancer Ther. 2016;15(4):502-11.
[74]. Sivanandam V, LaRocca CJ, Chen NG, Fong Y, Warner SG. Oncolytic Viruses and Immune Checkpoint Inhibition: The Best of Both Worlds. Mol Ther Oncolytics. 2019;13:93-106.
[75]. Schirrmacher V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review). Int J Oncol. 2019;54(2):407-19.
[76]. Harrington KJ, Puzanov I, Hecht JR, Hodi FS, Szabo Z, Murugappan S, et al. Clinical development of talimogene laherparepvec (T-VEC): a modified herpes simplex virus type-1-derived oncolytic immunotherapy. Expert Rev Anticancer Ther. 2015;15(12):1389-403.
[77]. Russell L, Peng KW, Russell SJ, Diaz RM. Oncolytic Viruses: Priming Time for Cancer Immunotherapy. BioDrugs. 2019;33(5):485-501.
[78]. Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17(3):718-30.
[79]. D'Souza N, Burns JS, Grisendi G, Candini O, Veronesi E, Piccinno S, et al. MSC and Tumors: Homing, Differentiation, and Secretion Influence Therapeutic Potential. Adv Biochem Eng Biotechnol. 2013;130:209-66.
[80]. Reagan MR, Kaplan DL. Concise review: Mesenchymal stem cell tumor-homing: detection methods in disease model systems. Stem Cells. 2011;29(6):920-7.
[81]. Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev. 2012;64(8):739-48.
[82]. Kidd S, Caldwell L, Dietrich M, Samudio I, Spaeth EL, Watson K, et al. Mesenchymal stromal cells alone or expressing interferon-beta suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment. Cytotherapy. 2010;12(5):615-25.
[83]. Ren C, Kumar S, Chanda D, Kallman L, Chen J, Mountz JD, et al. Cancer gene therapy using mesenchymal stem cells expressing interferon-beta in a mouse prostate cancer lung metastasis model. Gene Ther. 2008;15(21):1446-53.
[84]. Choi SH, Stuckey DW, Pignatta S, Reinshagen C, Khalsa JK, Roozendaal N, et al. Tumor Resection Recruits Effector T Cells and Boosts Therapeutic Efficacy of Encapsulated Stem Cells Expressing IFNbeta in Glioblastomas. Clin Cancer Res. 2017;23(22):7047-58.
[85]. Xu C, Lin L, Cao G, Chen Q, Shou P, Huang Y, et al. Interferon-alpha-secreting mesenchymal stem cells exert potent antitumor effect in vivo. Oncogene. 2014;33(42):5047-52.
[86]. Andreeff M, Marini FC, Westin SN, Coleman RL, Thall PF, Aljahdami V, et al. Abstract 75: A phase I trial of mesenchymal stem cells transfected with a plasmid secreting interferon beta in advanced ovarian cancer. Cancer Research. 2018;78(13 Supplement):75-.
[87]. Guo Y, Lei K, Tang L. Neoantigen Vaccine Delivery for Personalized Anticancer Immunotherapy. Frontiers in Immunology. 2018;9(1499).
[88]. Bezu L, Kepp O, Cerrato G, Pol J, Fucikova J, Spisek R, et al. Trial watch: Peptide-based vaccines in anticancer therapy. Oncoimmunology. 2018;7(12):e1511506-e.
[89]. Kuai R, Ochyl LJ, Bahjat KS, Schwendeman A, Moon JJ. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat Mater. 2017;16(4):489-96.
[90]. Yoon HY, Selvan ST, Yang Y, Kim MJ, Yi DK, Kwon IC, et al. Engineering nanoparticle strategies for effective cancer immunotherapy. Biomaterials. 2018;178:597-607.
[91]. Makkouk A, Joshi VB, Wongrakpanich A, Lemke CD, Gross BP, Salem AK, et al. Biodegradable microparticles loaded with doxorubicin and CpG ODN for in situ immunization against cancer. Aaps j. 2015;17(1):184-93.
[92]. Luo M, Liu Z, Zhang X, Han C, Samandi LZ, Dong C, et al. Synergistic STING activation by PC7A nanovaccine and ionizing radiation improves cancer immunotherapy. J Control Release. 2019;300:154-60.
[93]. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nature Reviews Microbiology. 2021;19(1):55-71.
[94]. Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971-6.
[95]. Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079-84.
[96]. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084-9.

Copyright © 2021 Eman Bahattab, Khalid Shah, Mohannad Fallatah Creative Commons License Publishing time:2022-06-30
This work is licensed under a Creative Commons Attribution 4.0 International License