Mechanisms underlying low-clinical responses to PD-1/PD-L1 blocking antibodies in immunotherapy of cancer: a key role of exosomal PD-L1

Zi Yin, Min Yu, Tingting Ma, Chuanzhao Zhang, Shanzhou Huang, Mohammad Reza Karimzadeh, Amir Abaas Momtazi-Borojeni, Sheng Chen, Zi Yin, Min Yu, Tingting Ma, Chuanzhao Zhang, Shanzhou Huang, Mohammad Reza Karimzadeh, Amir Abaas Momtazi-Borojeni, Sheng Chen

Abstract

Exosomes, as the main group of extracellular vesicles, are biologically active lipid-bilayer vesicles that are naturally released from different types of normal or tumor cells. These vesicles play an important role in intercellular communication and influence the extracellular environment and the immune system. Emerging evidence demonstrates that cancer-derived exosomes are enriched in immunosuppressive proteins, such as the programmed death-ligand 1 (PD-L1). PD-L1 and its receptor programmed cell death protein 1 (PD-1) are the key immune checkpoint molecules that promote tumor progression via negative regulation of immune responses. PDL-1 is highly expressed on the surface of tumor cells and binds to PD-1 on the surface of activated T cells, leading to suppression of T cells, which consequently enables cancer cells to escape antitumor immunity. Currently, there are several Food and Drug Administration-approved monoclonal antibodies blocking PD-1/PD-L1 interaction, which are clinically used for cancer treatment. However, despite impressive treatment outcomes, some patients show poor response to PD-1/PD-L1 blockade. Of note, tumor-derived exosomes containing PD-L1 can recapitulate the effect of cell-surface PD-L1. There is evidence that reveals a significant association between levels of circulating exosomal PD-L1 and rate of response to anti-PD-1/PD-L1 antibody therapy. The present article reviews the role of exosomal PDL-1 in the therapeutic resistance to anti-PD-1/PD-L1 treatment. Importantly, it is suggested that the removal of exosomal PDL-1 could serve as a therapeutic adjuvant for enhancing the efficacy of anti-PD-1/PD-L1 therapy in patients with cancer.

Keywords: immunotherapy; programmed cell death 1 receptor; tumor escape.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
The role of exosomal PD-L1 in cancer progression. Tumor cells secrete exosomes containing PD-L1 leading to suppression of immunity by reducing T-cell activity and inhibition of interferon-gamma and interleukin 2 production as well as reducing total number of CD8+ T cells by inducing apoptosis through PD-1/PD-L1 pathway. PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1.
Figure 2
Figure 2
Exosomal PD-L1 induces acquired resistance to anti-PD-1/PD-L1 therapy. Exosomes carrying PD-L1 limit effectiveness of anti-PD-1/PD-L1 therapy through binding to antibodies and suppression of T-cell activity. However, elimination of PD-L1 exosomes can improve anti-PD-1/PD-L1 therapy. IFN-γ, interferon-gamma; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1.

References

    1. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov 2018;8:1069–86. 10.1158/-18-0367
    1. Agata Y, Kawasaki A, Nishimura H, et al. . Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 1996;8:765–72. 10.1093/intimm/8.5.765
    1. Keir ME, Liang SC, Guleria I, et al. . Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med 2006;203:883–95. 10.1084/jem.20051776
    1. Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, et al. . Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med 2012;209:1201–17. 10.1084/jem.20112741
    1. Garcia-Diaz A, Shin DS, Moreno BH, et al. . Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep 2017;19:1189–201. 10.1016/j.celrep.2017.04.031
    1. Wu Y, Chen W, Xu ZP, et al. . Pd-L1 distribution and perspective for cancer Immunotherapy-Blockade, knockdown, or inhibition. Front Immunol 2019;10:2022. 10.3389/fimmu.2019.02022
    1. Sfanos KS, Bruno TC, Meeker AK, et al. . Human prostate-infiltrating CD8+ T lymphocytes are oligoclonal and PD-1+. Prostate 2009;69:1694–703. 10.1002/pros.21020
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, et al. . Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114:1537–44. 10.1182/blood-2008-12-195792
    1. Dong H, Strome SE, Salomao DR, et al. . Tumor-Associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 2002;8:793–800. 10.1038/nm730
    1. Blank C, Brown I, Peterson AC, et al. . PD-L1/B7H-1 Inhibits the Effector Phase of Tumor Rejection by T Cell Receptor (TCR) Transgenic CD8 + T Cells. Cancer Res 2004;64:1140–5. 10.1158/0008-5472.CAN-03-3259
    1. Iwai Y, Ishida M, Tanaka Y, et al. . Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A 2002;99:12293–7. 10.1073/pnas.192461099
    1. Rittmeyer A, Barlesi F, Waterkamp D, et al. . Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (oak): a phase 3, open-label, multicentre randomised controlled trial. Lancet 2017;389:255–65. 10.1016/S0140-6736(16)32517-X
    1. Baxi S, Yang A, Gennarelli RL, et al. . Immune-Related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis. BMJ 2018;360:k793. 10.1136/bmj.k793
    1. Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors. JAMA Oncology 2016;2:1346–53. 10.1001/jamaoncol.2016.1051
    1. Kwok G, Yau TCC, Chiu JW, et al. . Pembrolizumab (Keytruda). Hum Vaccin Immunother 2016;12:2777–89. 10.1080/21645515.2016.1199310
    1. Peters S, Kerr KM, Stahel R. Pd-1 blockade in advanced NSCLC: a focus on pembrolizumab. Cancer Treat Rev 2018;62:39–49. 10.1016/j.ctrv.2017.10.002
    1. Sim F, Leidner R, Bell RB. Immunotherapy for head and neck cancer. Oral Maxillofac Surg Clin North Am 2019;31:85–100. 10.1016/j.coms.2018.09.002
    1. Bellmunt J, de Wit R, Vaughn DJ, et al. . Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med Overseas Ed 2017;376:1015–26. 10.1056/NEJMoa1613683
    1. Herbst RS, Soria J-C, Kowanetz M, et al. . Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563–7. 10.1038/nature14011
    1. Tumeh PC, Harview CL, Yearley JH, et al. . Pd-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71. 10.1038/nature13954
    1. Im SJ, Hashimoto M, Gerner MY, et al. . Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016;537:417–21. 10.1038/nature19330
    1. Huang AC, Postow MA, Orlowski RJ, et al. . T-Cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017;545:60–5. 10.1038/nature22079
    1. Forde PM, Chaft JE, Smith KN, et al. . Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med Overseas Ed 2018;378:1976–86. 10.1056/NEJMoa1716078
    1. Bengsch B, Johnson AL, Kurachi M, et al. . Bioenergetic Insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8 + T cell exhaustion. Immunity 2016;45:358–73. 10.1016/j.immuni.2016.07.008
    1. Gubin MM, Zhang X, Schuster H, et al. . Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014;515:577–81. 10.1038/nature13988
    1. Loke P'ng, Allison JP. Pd-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Proc Natl Acad Sci U S A 2003;100:5336–41. 10.1073/pnas.0931259100
    1. Dahan R, Sega E, Engelhardt J, et al. . FcγRs modulate the anti-tumor activity of antibodies targeting the PD-1/PD-L1 axis. Cancer Cell 2015;28:285–95. 10.1016/j.ccell.2015.08.004
    1. Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer 2018;118:9–16. 10.1038/bjc.2017.434
    1. Polk A, Svane I-M, Andersson M, et al. . Checkpoint inhibitors in breast cancer – current status. Cancer Treat Rev 2018;63:122–34. 10.1016/j.ctrv.2017.12.008
    1. Sheng Z, Zhu X, Sun Y, et al. . The efficacy of anti-PD-1/PD-L1 therapy and its comparison with EGFR-TKIs for advanced non-small-cell lung cancer. Oncotarget 2017;8:57826–35. 10.18632/oncotarget.18406
    1. Ribas A. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov 2015;5:915–9. 10.1158/-15-0563
    1. Reck M, Rodríguez-Abreu D, Robinson AG, et al. . Pembrolizumab versus chemotherapy for PD-L1–positive non–small-cell lung cancer. N Engl J Med Overseas Ed 2016;375:1823–33. 10.1056/NEJMoa1606774
    1. Ding W, LaPlant BR, Call TG, et al. . Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 2017;129:3419–27. 10.1182/blood-2017-02-765685
    1. Patel SP, Kurzrock R. Pd-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther 2015;14:847–56. 10.1158/1535-7163.MCT-14-0983
    1. Yagi T, Baba Y, Ishimoto T, et al. . Pd-L1 expression, tumor-infiltrating lymphocytes, and clinical outcome in patients with surgically resected esophageal cancer. Ann Surg 2019;269:471–8. 10.1097/SLA.0000000000002616
    1. Teng MWL, Ngiow SF, Ribas A, et al. . Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 2015;75:2139–45. 10.1158/0008-5472.CAN-15-0255
    1. Thommen DS, Koelzer VH, Herzig P, et al. . A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 2018;24:994–1004. 10.1038/s41591-018-0057-z
    1. Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. . Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 2018;175:e1020:998–1013. 10.1016/j.cell.2018.10.038
    1. Tran E, Ahmadzadeh M, Lu Y-C, et al. . Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 2015;350:1387–90. 10.1126/science.aad1253
    1. Tran E, Turcotte S, Gros A, et al. . Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641–5. 10.1126/science.1251102
    1. Monach PA, Meredith SC, T.Siegel C, et al. . A unique tumor antigen produced by a single amino acid substitution. Immunity 1995;2:45–59. 10.1016/1074-7613(95)90078-0
    1. Robbins PF, El-Gamil M, Li YF, et al. . A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med 1996;183:1185–92. 10.1084/jem.183.3.1185
    1. Dubey P, Hendrickson RC, Meredith SC, et al. . The immunodominant antigen of an ultraviolet-induced regressor tumor is generated by a somatic point mutation in the DEAD box helicase p68. J Exp Med 1997;185:695–706. 10.1084/jem.185.4.695
    1. Lennerz V, Fatho M, Gentilini C, et al. . The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc Natl Acad Sci U S A 2005;102:16013–8. 10.1073/pnas.0500090102
    1. Rizvi NA, Hellmann MD, Snyder A, et al. . Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 2015;348:124–8. 10.1126/science.aaa1348
    1. Chen Y, Liu Q, Chen Z, et al. . Pd-L1 expression and tumor mutational burden status for prediction of response to chemotherapy and targeted therapy in non-small cell lung cancer. J Exp Clin Cancer Res 2019;38:193. 10.1186/s13046-019-1192-1
    1. Hellmann MD, Nathanson T, Rizvi H, et al. . Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell 2018;33:e844:843–52. 10.1016/j.ccell.2018.03.018
    1. Hellmann MD, Ciuleanu T-E, Pluzanski A, et al. . Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med 2018;378:2093–104. 10.1056/NEJMoa1801946
    1. Rosenberg JE, Hoffman-Censits J, Powles T, et al. . Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. The Lancet 2016;387:1909–20. 10.1016/S0140-6736(16)00561-4
    1. Riaz N, Havel JJ, Makarov V, et al. . Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 2017;171:e916:934–49. 10.1016/j.cell.2017.09.028
    1. Hugo W, Zaretsky JM, Sun L, et al. . Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016;165:35–44. 10.1016/j.cell.2016.02.065
    1. Goodman AM, Kato S, Bazhenova L, et al. . Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther 2017;16:2598–608. 10.1158/1535-7163.MCT-17-0386
    1. Legrand FA, Gandara DR, Mariathasan S, et al. . Association of high tissue TMB and atezolizumab efficacy across multiple tumor types. JCO 2018;36:12000. 10.1200/JCO.2018.36.15_suppl.12000
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med Overseas Ed 2017;377:2500–1. 10.1056/NEJMc1713444
    1. Le DT, Durham JN, Smith KN, et al. . Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–13. 10.1126/science.aan6733
    1. Le DT, Uram JN, Wang H, et al. . Pd-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. 10.1056/NEJMoa1500596
    1. Germano G, Lamba S, Rospo G, et al. . Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 2017;552:116–20. 10.1038/nature24673
    1. Yi M, Jiao D, Xu H, et al. . Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer 2018;17:1–14. 10.1186/s12943-018-0864-3
    1. Kalluri R. The biology and function of exosomes in cancer. J Clin Invest 2016;126:1208–15. 10.1172/JCI81135
    1. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer 2019;1871:455–68. 10.1016/j.bbcan.2019.04.004
    1. Tavasolian F, Moghaddam AS, Rohani F. Exosomes: Effectual players in rheumatoid arthritis, 2020.
    1. Moghaddam AS, Afshari JT, Esmaeili S-A, et al. . Cardioprotective microRNAs: lessons from stem cell-derived exosomal microRNAs to treat cardiovascular disease. Atherosclerosis 2019;285:1–9. 10.1016/j.atherosclerosis.2019.03.016
    1. Moghiman T, Barghchi B, Esmaeili S-A, et al. . Therapeutic angiogenesis with exosomal microRNAs: an effectual approach for the treatment of myocardial ischemia, 2020: 1–9.
    1. Zhou R, Wang L, Zhao G. Circulating exosomal microRNAs as emerging non‐invasive clinical biomarkers in heart failure: Mega bio‐roles of a nano bio‐particle;72. 10.1002/iub.2396
    1. Greening DW, Gopal SK, Xu R, et al. . Exosomes and their roles in immune regulation and cancer. Semin Cell Dev Biol 2015;40:72–81. 10.1016/j.semcdb.2015.02.009
    1. Yi M, Xu L, Jiao Y, et al. . The role of cancer-derived microRNAs in cancer immune escape. J Hematol Oncol 2020;13:1–14. 10.1186/s13045-020-00848-8
    1. Sun Z, Shi K, Yang S, et al. . Effect of exosomal miRNA on cancer biology and clinical applications. Mol Cancer 2018;17:147. 10.1186/s12943-018-0897-7
    1. McAndrews KM, Kalluri R. Mechanisms associated with biogenesis of exosomes in cancer. Mol Cancer 2019;18:52. 10.1186/s12943-019-0963-9
    1. Chen G, Huang AC, Zhang W, et al. . Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018;560:382–6. 10.1038/s41586-018-0392-8
    1. Yang Y, Li C-W, Chan L-C, et al. . Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res 2018;28:862–4. 10.1038/s41422-018-0060-4
    1. Theodoraki M-N, Yerneni SS, Hoffmann TK, et al. . Clinical Significance of PD-L1+ Exosomes in Plasma of Head and Neck Cancer Patients. Clin Cancer Res 2018;24:896–905. 10.1158/1078-0432.CCR-17-2664
    1. Li C, Li C, Zhi C, et al. . Clinical significance of PD-L1 expression in serum-derived exosomes in NSCLC patients. J Transl Med 2019;17. 10.1186/s12967-019-2101-2
    1. Poggio M, Hu T, Pai C-C, et al. . Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 2019;177:e413:414–27. 10.1016/j.cell.2019.02.016
    1. Ricklefs FL, Alayo Q, Krenzlin H, et al. . Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci Adv 2018;4:eaar2766. 10.1126/sciadv.aar2766
    1. Kim DH, Kim H, Choi YJ, et al. . Exosomal PD-L1 promotes tumor growth through immune escape in non-small cell lung cancer. Exp Mol Med 2019;51:1–13. 10.1038/s12276-019-0295-2
    1. Razzo BM, Ludwig N, Hong C-S, et al. . Tumor-Derived exosomes promote carcinogenesis of murine oral squamous cell carcinoma. Carcinogenesis 2020;41:625–33. 10.1093/carcin/bgz124
    1. Theodoraki M-N, Yerneni S, Gooding WE, et al. . Circulating exosomes measure responses to therapy in head and neck cancer patients treated with cetuximab, ipilimumab, and IMRT. Oncoimmunology 2019;8:e1593805. 10.1080/2162402X.2019.1593805
    1. Fan Y, Liu Y, Qu X. ASO author reflections: the prognostic role of exosomal PD-L1 in patients with gastric cancer. Ann Surg Oncol 2019;26:851–2. 10.1245/s10434-019-07864-0
    1. Fan Y, Che X, Qu J, et al. . Exosomal PD-L1 retains immunosuppressive activity and is associated with gastric cancer prognosis. Ann Surg Oncol 2019;26:3745–55. 10.1245/s10434-019-07431-7
    1. Lux A, Kahlert C, Grützmann R, et al. . C-Met and PD-L1 on circulating exosomes as diagnostic and prognostic markers for pancreatic cancer. Int J Mol Sci 2019;20:3305. 10.3390/ijms20133305
    1. Foster BA, Gingrich JR, Kwon ED, et al. . Characterization of prostatic epithelial cell lines derived from transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Cancer Res 1997;57:3325–30.
    1. Yu P, Steel JC, Zhang M, et al. . Simultaneous inhibition of two regulatory T-cell subsets enhanced interleukin-15 efficacy in a prostate tumor model. Proc Natl Acad Sci U S A 2012;109:6187–92. 10.1073/pnas.1203479109
    1. EL Andaloussi S, Mäger I, Breakefield XO, et al. . Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov 2013;12:347–57. 10.1038/nrd3978
    1. Datta A, Kim H, Lal M, et al. . Manumycin a suppresses exosome biogenesis and secretion via targeted inhibition of Ras/Raf/ERK1/2 signaling and hnRNP H1 in castration-resistant prostate cancer cells. Cancer Lett 2017;408:73–81. 10.1016/j.canlet.2017.08.020
    1. Datta A, Kim H, McGee L, et al. . High-Throughput screening identified selective inhibitors of exosome biogenesis and secretion: a drug repurposing strategy for advanced cancer. Sci Rep 2018;8:8161. 10.1038/s41598-018-26411-7
    1. Bobrie A, Krumeich S, Reyal F, et al. . Rab27A supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res 2012;72:4920–30. 10.1158/0008-5472.CAN-12-0925
    1. Nishida-Aoki N, Tominaga N, Takeshita F, et al. . Disruption of circulating extracellular vesicles as a novel therapeutic strategy against cancer metastasis. Mol Ther 2017;25:181–91. 10.1016/j.ymthe.2016.10.009
    1. Im E-J, Lee C-H, Moon P-G, et al. . Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor a. Nat Commun 2019;10:1387. 10.1038/s41467-019-09387-4
    1. Chalmin F, Ladoire S, Mignot G, et al. . Membrane-Associated HSP72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest 2010;67:457–71. 10.1172/JCI40483
    1. Savina A, Furlán M, Vidal M, et al. . Exosome release is regulated by a calcium-dependent mechanism in K562 cells. J Biol Chem 2003;278:20083–90. 10.1074/jbc.M301642200
    1. Spugnini EP, Citro G, Fais S. Proton pump inhibitors as anti vacuolar-ATPases drugs: a novel anticancer strategy. J Exp Clin Cancer Res 2010;29:44. 10.1186/1756-9966-29-44
    1. Luciani F, Spada M, De Milito A, et al. . Effect of proton pump inhibitor pretreatment on resistance of solid tumors to cytotoxic drugs. J Natl Cancer Inst 2004;96:1702–13. 10.1093/jnci/djh305
    1. Trajkovic K, Hsu C, Chiantia S, et al. . Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008;319:1244–7. 10.1126/science.1153124
    1. Tullis RH, Duffin RP, Handley HH, et al. . Reduction of hepatitis C virus using lectin affinity plasmapheresis in dialysis patients. Blood Purif 2009;27:64–9. 10.1159/000167011
    1. Marleau AM, Chen C-S, Joyce JA, et al. . Exosome removal as a therapeutic adjuvant in cancer. J Transl Med 2012;10:134. 10.1186/1479-5876-10-134

Source: PubMed

3
購読する