1,272
Views
1
CrossRef citations to date
0
Altmetric
Immunotherapy - Cancer

A review of the clinical experience with CMN-001, a tumor RNA loaded dendritic cell immunotherapy for the treatment of metastatic renal cell carcinoma

, , , &
Article: 2220629 | Received 07 Mar 2023, Accepted 29 May 2023, Published online: 30 Jun 2023

References

  • Mastelic-Gavillet B, Balint K, Boudousquie C, Gannon PO, Kandalaft LE. Personalized dendritic cell vaccines-recent breakthroughs and encouraging clinical results. Front Immunol. 2019;10:766. doi:10.3389/fimmu.2019.00766.
  • Gardner A, de Mingo Pulido Á, Ruffell B. Dendritic cells and their role in immunotherapy. Front Immunol. 2020;11:924. doi:10.3389/fimmu.2020.00924.
  • Chick RC, Faries MB, Hale DF, Kemp Bohan PM, Hickerson AT, Vreeland TJ, Myers JW, Cindass JL, Brown TA, Hyngstrom J, et al. Multi-institutional, prospective, randomized, double-blind, placebo-controlled phase IIb trial of the tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine to prevent recurrence in high-risk melanoma patients: a subgroup analysis. Cancer Med. 2021;10:4302–14. doi:10.1002/cam4.3969.
  • Baldin AV, Savvateeva LV, Bazhin AV, Zamyatnin AA Jr. Dendritic cells in anticancer vaccination: rationale for ex vivo loading or in vivo targeting. Cancers. 2020;12:12. doi:10.3390/cancers12030590.
  • Chung DJ, Sharma S, Rangesa M, DeWolf S, Elhanati Y, Perica K, Young JW. Langerhans dendritic cell vaccine bearing mRNA-encoded tumor antigens induces antimyeloma immunity after autotransplant. Blood Adv. 2022;6:1547–58. doi:10.1182/bloodadvances.2021005941.
  • De Keersmaecker B, Claerhout S, Carrasco J, Bar I, Corthals J, Wilgenhof S, Neyns B, Thielemans K. TriMix and tumor antigen mRNA electroporated dendritic cell vaccination plus ipilimumab: link between T-cell activation and clinical responses in advanced melanoma. J ImmunoTher Cancer. 2020;8:e000329. doi:10.1136/jitc-2019-000329.
  • Dörrie J, Schaft N, Schuler G, Schuler-Thurner B. Therapeutic cancer vaccination with ex vivo RNA-transfected dendritic cells-an update. Pharmaceutics. 2020;12:12. doi:10.3390/pharmaceutics12020092.
  • Cozzi S, Najafi M, Gomar M, Ciammella P, Iotti C, Iaccarino C, Dominici M, Pavesi G, Chiavelli C, Kazemian A, et al. Delayed effect of dendritic cells vaccination on survival in glioblastoma: a systematic review and meta-analysis. Curr Oncol. 2022;29:881–91. doi:10.3390/curroncol29020075.
  • Jansen Y, Kruse V, Corthals J, Schats K, van Dam PJ, Seremet T, Heirman C, Brochez L, Kockx M, Thielemans K, et al. A randomized controlled phase II clinical trial on mRNA electroporated autologous monocyte-derived dendritic cells (TriMixDC-MEL) as adjuvant treatment for stage III/IV melanoma patients who are disease-free following the resection of macrometastases. Cancer Immunol Immunother. 2020;69:2589–98. doi:10.1007/s00262-020-02618-4.
  • Filin IY, Kitaeva KV, Rutland CS, Rizvanov AA, Solovyeva VV, Solovyeva VV. Recent advances in experimental dendritic cell vaccines for cancer. Front Oncol. 2021;11:730824. doi:10.3389/fonc.2021.730824.
  • Batich KA, Mitchell DA, Healy P, Herndon JE 2nd, Sampson JH. Once, twice, three times a finding: reproducibility of dendritic cell vaccine trials targeting cytomegalovirus in Glioblastoma. Clin Cancer Res. 2020;26:5297–303. doi:10.1158/1078-0432.CCR-20-1082.
  • Laureano RS, Sprooten J, Vanmeerbeerk I, Borras DM, Govaerts J, Naulaerts S, Berneman ZN, Beuselinck B, Bol KF, Borst J, et al. Trial watch: dendritic cell (DC)-based immunotherapy for cancer. Oncoimmunology. 2022;11:2096363. doi:10.1080/2162402X.2022.2096363.
  • Jeng LB, Liao LY, Shih FY, Teng CF. Dendritic-cell-vaccine-based immunotherapy for hepatocellular carcinoma: clinical trials and recent preclinical studies. Cancers. 2022;14:14. doi:10.3390/cancers14184380.
  • Grenier JM, Yeung ST, Khanna KM. Combination immunotherapy: taking cancer vaccines to the next level. Front Immunol. 2018;9:610. doi:10.3389/fimmu.2018.00610.
  • Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor microenvironment. Science. 2015;348:74–80. doi:10.1126/science.aaa6204.
  • Figlin RA, Tannir NM, Uzzo RG, Tykodi SS, Chen DYT, Master V, Kapoor A, Vaena D, Lowrance W, Bratslavsky G, et al. Results of the ADAPT phase 3 study of rocapuldencel-T in combination with sunitinib as first-line therapy in patients with metastatic renal cell carcinoma. Clin Cancer Res. 2020;26:2327–36. doi:10.1158/1078-0432.CCR-19-2427.
  • Coates PT, Colvin BL, Hackstein H, Thomson AW. Manipulation of dendritic cells as an approach to improved outcomes in transplantation. Expert Rev Mol Med. 2002;4:1–21. doi:10.1017/S1462399402004283.
  • Tcherepanova IY, Adams MD, Feng X, Hinohara A, Horvatinovich J, Calderhead D, Healey D, Nicolette CA. Ectopic expression of a truncated CD40L protein from synthetic post-transcriptionally capped RNA in dendritic cells induces high levels of IL-12 secretion. BMC Mol Biol. 2008;9:90. doi:10.1186/1471-2199-9-90.
  • Calderhead DM, De Benedette MA, Ketteringham H, Gamble AH, Horvatinovich JM, Tcherepanova IY, Nicolette CA, Healey DG. Cytokine maturation followed by CD40L mRNA electroporation results in a clinically relevant dendritic cell product capable of inducing a potent proinflammatory CTL response. J Immunother. 2008;31:731–41. doi:10.1097/CJI.0b013e318183db02.
  • Cesana GC, De Raffele G, Cohen S, Moroziewicz D, Mitcham J, Stoutenburg J, Cheung K, Hesdorffer C, Kim-Schulze S, Kaufman HL. Characterization of CD4+CD25+ regulatory T cells in patients treated with high-dose interleukin-2 for metastatic melanoma or renal cell carcinoma. J Clin Oncol. 2006;24:1169–77. doi:10.1200/JCO.2005.03.6830.
  • De Benedette MA, Calderhead DM, Ketteringham H, Gamble AH, Horvatinovich JM, Tcherepanova IY, Nicolette CA, Healey DG. Priming of a novel subset of CD28+ rapidly expanding high-avidity effector memory CTL by post maturation electroporation-CD40L dendritic cells is IL-12 dependent. J Immunol. 2008;181:5296–305. doi:10.4049/jimmunol.181.8.5296.
  • De Benedette MA, Calderhead DM, Tcherepanova IY, Nicolette CA, Healey DG. Potency of mature CD40L RNA electroporated dendritic cells correlates with IL-12 secretion by tracking multifunctional CD8(+)/CD28(+) cytotoxic T-cell responses in vitro. J Immunother. 2011;34:45–57. doi:10.1097/CJI.0b013e3181fb651a.
  • Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, Master VA, Pal SK, Miller WH, Karsh LI, et al. Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): phase 2 study results. J ImmunoTher Cancer. 2015;3:14. doi:10.1186/s40425-015-0055-3.
  • Harris J, Monesmith T, Ubben A, Norris M, Freedman JH, Tcherepanova I. An improved RNA amplification procedure results in increased yield of autologous RNA transfected dendritic cell-based vaccine. Biochim Biophys Acta. 2005;1724:127–36. doi:10.1016/j.bbagen.2005.03.013.
  • Slagter-Jager JG, Raney A, Lewis WE, De Benedette MA, Nicolette CA, Tcherepanova IY. Evaluation of RNA amplification methods to improve DC immunotherapy antigen presentation and immune response. Mol Ther Nucleic Acids. 2013;2:e91. doi:10.1038/mtna.2013.18.
  • Tcherepanova I, Harris J, Starr A, Cleveland J, Ketteringham H, Calderhead D, Horvatinovich J, Healey D, Nicolette CA. Multiplex RT-PCR amplification of HIV genes to create a completely autologous DC-based immunotherapy for the treatment of HIV infection. PLos One. 2008;3:e1489. doi:10.1371/journal.pone.0001489.
  • Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–24. doi:10.1056/NEJMoa065044.
  • Escudier B. Sunitinib for the management of advanced renal cell carcinoma. Expert Rev Anticancer Ther. 2010;10:305–17. doi:10.1586/era.10.26.
  • Escudier B. Sorafenib for the management of advanced renal cell carcinoma. Expert Rev Anticancer Ther. 2011;11:825–36. doi:10.1586/era.11.55.
  • Ko JJ, Choueiri TK, Rini BI, Lee JL, Kroeger N, Srinivas S, Harshman LC, Knox JJ, Bjarnason GA, MacKenzie MJ, et al. First-, second-, third-line therapy for mRCC: benchmarks for trial design from the IMDC. Br J Cancer. 2014;110:1917–22. doi:10.1038/bjc.2014.25.
  • Escudier B, Cosaert J, Pisa P. Bevacizumab: direct anti-VEGF therapy in renal cell carcinoma. Expert Rev Anticancer Ther. 2008;8:1545–57. doi:10.1586/14737140.8.10.1545.
  • Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356:2271–81. doi:10.1056/NEJMoa066838.
  • Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008;372:449–56. doi:10.1016/S0140-6736(08)61039-9.
  • Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28:1061–8. doi:10.1200/JCO.2009.23.9764.
  • Lalani AA, McGregor BA, Albiges L, Choueiri TK, Motzer R, Powles T, Wood C, Bex A. Systemic treatment of metastatic clear cell renal cell carcinoma in 2018: current paradigms, use of immunotherapy, and future directions. Eur Urol. 2018;75:100–10. doi:10.1016/j.eururo.2018.10.010.
  • Liau LM, Ashkan K, Brem S, Campian JL, Trusheim JE, Iwamoto FM, Tran DD, Ansstas G, Cobbs CS, Heth JA, et al. Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: a phase 3 prospective externally controlled cohort trial. JAMA Oncol. 2023;9(1):112–21. doi:10.1001/jamaoncol.2022.5370.
  • Master VA, Uzzo RG, Bratlavsky G, Karam JA. Autologous dendritic vaccine therapy in metastatic kidney cancer: the ADAPT trial and beyond. Eur Urol Focus. 2022;8:651–3. doi:10.1016/j.euf.2022.04.003.
  • Heng DY, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, Eigl BJ, Ruether JD, Cheng T, North S, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor–targeted agents: results from a large, multicenter study. J Clin Oncol. 2009;27:5794–9. doi:10.1200/JCO.2008.21.4809.
  • Liotta F, Gacci M, Frosali F, Querci V, Vittori G, Lapini A, Santarlasci V, Serni S, Cosmi L, Maggi L, et al. Frequency of regulatory T cells in peripheral blood and in tumour-infiltrating lymphocytes correlates with poor prognosis in renal cell carcinoma. BJU Int. 2010;107:1500–6. doi:10.1111/j.1464-410X.2010.09555.x.
  • Raimondi G, Shufesky WJ, Tokita D, Morelli AE, Thomson AW. Regulated compartmentalization of programmed cell death-1 discriminates CD4+CD25+ resting regulatory T cells from activated T cells. J Immunol. 2006;176:2808–16. doi:10.4049/jimmunol.176.5.2808.
  • Asma G, Amal G, Raja M, Amine D, Mohammed C, Amel BA. Comparison of circulating and intratumoral regulatory T cells in patients with renal cell carcinoma. Tumour Biol. 2015;36:3727–34. doi:10.1007/s13277-014-3012-8.
  • Li J, Ridgway W, Fathman CG, Tse HY, Shaw MK. High cell surface expression of CD4 allows distinction of CD4(+)CD25(+) antigen-specific effector T cells from CD4(+)CD25(+) regulatory T cells in murine experimental autoimmune encephalomyelitis. J Neuroimmunol. 2007;192:57–67. doi:10.1016/j.jneuroim.2007.09.004.
  • Hegele A, Varga Z, von Knobloch R, Heidenreich A, Kropf J, Hofmann R. TGF-beta1 in patients with renal cell carcinoma. Urol Res. 2002;30:126–9. doi:10.1007/s00240-002-0245-6.
  • Junker U, Haufe CC, Nuske K, Rebstock K, Steiner T, Wunderlich H, Junker K, Reinhold D. Elevated plasma TGF-β1 in renal diseases: cause or consequence? Cytokine. 2000;12:1084–91. doi:10.1006/cyto.1999.0645.
  • Dedobbeleer O, Stockis J, van der Woning B, Coulie PG, Lucas S. Cutting edge: active TGF-beta1 released from GARP/TGF-beta1 complexes on the surface of stimulated human B lymphocytes increases class-switch recombination and production of IgA. J Immunol. 2017;199:391–6. doi:10.4049/jimmunol.1601882.
  • Song J, Xiao L, Du G, Gao Y, Chen W, Yang S, Fan W, Shi B. The role of regulatory B cells (Bregs) in the Tregs-amplifying effect of Sirolimus. Int Immunopharmacol. 2016;38:90–6. doi:10.1016/j.intimp.2016.05.014.
  • Taylor AW. Review of the activation of TGF-beta in immunity. J Leukoc Biol. 2009;85:29–33. doi:10.1189/jlb.0708415.
  • Nouël A, Pochard P, Simon Q, Ségalen I, Le Meur Y, Pers JO, Hillion S. B-Cells induce regulatory T cells through TGF-β/IDO production in a CTLA-4 dependent manner. J Autoimmun. 2015;59:53–60. doi:10.1016/j.jaut.2015.02.004.
  • Jin N, Malcherek G, Mani J, Zurleit R, Schmitt A, Chen B, Freund M, Ho AD, Schmitt M. Suppression of cytomegalovirus-specific CD8(+)T cells by everolimus. Leuk Lymphoma. 2014;55:1144–50. doi:10.3109/10428194.2013.822496.
  • Zeng H, Chi H. mTOR signaling in the differentiation and function of regulatory and effector T cells. Curr Opin Immunol. 2017;46:103–11. doi:10.1016/j.coi.2017.04.005.
  • Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP, Ahmed R. mTOR regulates memory CD8 T-cell differentiation. Nature. 2009;460:108–12. doi:10.1038/nature08155.
  • Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B, Engleman EG, Levy R. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med. 1996;2:52–8. doi:10.1038/nm0196-52.
  • Gilboa E, Vieweg J. Cancer immunotherapy with mRNA-transfected dendritic cells. Immunol Rev. 2004;199:251–63. doi:10.1111/j.0105-2896.2004.00139.x.
  • Cibula D, Rob L, Mallmann P, Knapp P, Klat J, Chovanec J, Minar L, Melichar B, Hein A, Kieszko D, et al. Dendritic cell-based immunotherapy (DCVAC/OvCa) combined with second-line chemotherapy in platinum-sensitive ovarian cancer (SOV02): a randomized, open-label, phase 2 trial. Gynecol Oncol. 2021;162:652–60. doi:10.1016/j.ygyno.2021.07.003.
  • Zhong R, Ling X, Cao S, Xu J, Zhang B, Zhang X, Wang H, Han B, Zhong H. Safety and efficacy of dendritic cell-based immunotherapy (DCVAC/LuCa) combined with carboplatin/pemetrexed for patients with advanced non-squamous non-small-cell lung cancer without oncogenic drivers. ESMO Open. 2022;7:100334. doi:10.1016/j.esmoop.2021.100334.
  • Rob L, Cibula D, Knapp P, Mallmann P, Klat J, Minar L, Bartos P, Chovanec J, Valha P, Pluta M, et al. Safety and efficacy of dendritic cell-based immunotherapy DCVAC/OvCa added to first-line chemotherapy (carboplatin plus paclitaxel) for epithelial ovarian cancer: a phase 2, open-label, multicenter, randomized trial. J ImmunoTher Cancer. 2022;10. doi:10.1136/jitc-2021-003190.
  • Willemen Y, Versteven M, Peeters M, Berneman ZN, Smits ELJ. Ribonucleic acid engineering of dendritic cells for therapeutic vaccination: ready ‘N able to improve clinical outcome? Cancers. 2020;12:12. doi:10.3390/cancers12020299.
  • Anguille S, Van de Velde AL, Smits EL, Van Tendeloo VF, Juliusson G, Cools N, Nijs G, Stein B, Lion E, Van Driessche A, et al. Dendritic cell vaccination as postremission treatment to prevent or delay relapse in acute myeloid leukemia. Blood. 2017;130:1713–21. doi:10.1182/blood-2017-04-780155.
  • Boudewijns S, Bloemendal M, de Haas N, Westdorp H, Bol KF, Schreibelt G, Aarntzen EHJG, Lesterhuis WJ, Gorris MAJ, Croockewit A, et al. Autologous monocyte-derived DC vaccination combined with cisplatin in stage III and IV melanoma patients: a prospective, randomized phase 2 trial. Cancer Immunol Immunother. 2020;69:477–88. doi:10.1007/s00262-019-02466-x.
  • Tryggestad AMA, Axcrona K, Axcrona U, Bigalke I, Brennhovd B, Inderberg EM, Hønnåshagen TK, Skoge LJ, Solum G, Saebøe-Larssen S, et al. Long-term first-in-man phase I/II study of an adjuvant dendritic cell vaccine in patients with high-risk prostate cancer after radical prostatectomy. Prostate. 2022;82:245–53. doi:10.1002/pros.24267.
  • Lau SP, van ‘t Land FR, van der Burg SH, Homs MYV, Lolkema MP, Aerts J, van Eijck CHJ. Safety and tumour-specific immunological responses of combined dendritic cell vaccination and anti-CD40 agonistic antibody treatment for patients with metastatic pancreatic cancer: protocol for a phase I, open-label, single-arm, dose-escalation study (REACtiVe-2 trial). BMJ Open. 2022;12:e060431. doi:10.1136/bmjopen-2021-060431.
  • Storkus WJ, Maurer D, Lin Y, Ding F, Bose A, Lowe D, Rose A, DeMark M, Karapetyan L, Taylor JL, et al. Dendritic cell vaccines targeting tumor blood vessel antigens in combination with dasatinib induce therapeutic immune responses in patients with checkpoint-refractory advanced melanoma. J ImmunoTher Cancer. 2021;9:e003675. doi:10.1136/jitc-2021-003675.
  • Adams AM, Carpenter EL, Clifton GT, Vreeland TJ, Chick RC, O’Shea AE, McCarthy PM, Kemp Bohan PM, Hickerson AT, Valdera FA, et al. Divergent clinical outcomes in a phase 2B trial of the TLPLDC vaccine in preventing melanoma recurrence and the impact of dendritic cell collection methodology: a randomized clinical trial. Cancer Immunol Immunother. 2022;72:1–9. doi:10.1007/s00262-022-03272-8.
  • Lau SP, Klaase L, Vink M, Dumas J, Bezemer K, van Krimpen A, van der Breggen R, Wismans LV, Doukas M, de Koning W, et al. Autologous dendritic cells pulsed with allogeneic tumour cell lysate induce tumour-reactive T-cell responses in patients with pancreatic cancer: a phase I study. Eur J Cancer. 2022;169:20–31. doi:10.1016/j.ejca.2022.03.015.
  • Caro AA, Deschoemaeker S, Allonsius L, Coosemans A, Laoui D. Dendritic cell vaccines: a promising approach in the fight against ovarian cancer. Cancers. 2022;14:14. doi:10.3390/cancers14164037.
  • Fucikova J, Hensler M, Kasikova L, Lanickova T, Pasulka J, Rakova J, Drozenova J, Fredriksen T, Hraska M, Hrnciarova T, et al. An autologous dendritic cell vaccine promotes anticancer immunity in patients with ovarian cancer with low mutational burden and cold tumors. Clin Cancer Res. 2022;28:3053–65. doi:10.1158/1078-0432.CCR-21-4413.
  • Guo Z, Yuan Y, Chen C, Lin J, Ma Q, Liu G, Gao Y, Huang Y, Chen L, Chen L-Z, et al. Durable complete response to neoantigen-loaded dendritic-cell vaccine following anti-PD-1 therapy in metastatic gastric cancer. NPJ Precis Oncol. 2022;6:34. doi:10.1038/s41698-022-00279-3.
  • Popovic M, Matovina-Brko G, Jovic M, Popovic LS. Immunotherapy: a new standard in the treatment of metastatic clear cell renal cell carcinoma. World J Clin Oncol. 2022;13:28–38. doi:10.5306/wjco.v13.i1.28.
  • George DJ, Lee CH, Heng D. New approaches to first-line treatment of advanced renal cell carcinoma. Ther Adv Med Oncol. 2021;13:17588359211034708. doi:10.1177/17588359211034708.
  • Heng DY, Xie W, Regan MM, Harshman LC, Bjarnason GA, Vaishampayan UN, Mackenzie M, Wood L, Donskov F, Tan M-H, et al. External validation and comparison with other models of the international metastatic renal-cell carcinoma database consortium prognostic model: a population-based study. Lancet Oncol. 2013;14:141–8. doi:10.1016/S1470-2045(12)70559-4.
  • Mekhail TM, Abou-Jawde RM, Boumerhi G, Malhi S, Wood L, Elson P, Bukowski R. Validation and extension of the memorial sloan-kettering prognostic factors model for survival in patients with previously untreated metastatic renal cell carcinoma. J Clin Oncol. 2005;23:832–41. doi:10.1200/JCO.2005.05.179.
  • George DJ, Spigel DR, Gordan LN, Kochuparambil ST, Molina AM, Yorio J, Rezazadeh Kalebasty A, McKean H, Tchekmedyian N, Tykodi SS, et al. Safety and efficacy of first-line nivolumab plus ipilimumab alternating with nivolumab monotherapy in patients with advanced renal cell carcinoma: the non-randomised, open-label, phase IIIb/IV CheckMate 920 trial. BMJ Open. 2022;12:e058396. doi:10.1136/bmjopen-2021-058396.
  • Buti S, Leonetti A, Dallatomasina A, Bersanelli M. Everolimus in the management of metastatic renal cell carcinoma: an evidence-based review of its place in therapy. Core Evid. 2016;11:23–36. doi:10.2147/CE.S98687.
  • U.S. Food and Drug Administration. FDA approves nivolumab plus ipilimumab combination for intermediate or poor-risk advanced renal cell carcinoma. 2016, Apr 16
  • Motzer RJ, Tannir NM, McDermott DF, Aren Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378:1277–90. doi:10.1056/NEJMoa1712126.
  • Shah AY, Kotecha RR, Lemke EA, Chandramohan A, Chaim JL, Msaouel P, Xiao L, Gao J, Campbell MT, Zurita AJ, et al. Outcomes of patients with metastatic clear-cell renal cell carcinoma treated with second-line VEGFR-TKI after first-line immune checkpoint inhibitors. Eur J Cancer. 2019;114:67–75. doi:10.1016/j.ejca.2019.04.003.
  • Auvray M, Auclin E, Barthelemy P, Bono P, Kellokumpu-Lehtinen P, Gross-Goupil M, De Velasco G, Powles T, Mouillet G, Vano Y-A, et al. Second-line targeted therapies after nivolumab-ipilimumab failure in metastatic renal cell carcinoma. Eur J Cancer. 2019;108:33–40. doi:10.1016/j.ejca.2018.11.031.
  • Saran U, Foti M, Dufour JF. Cellular and molecular effects of the mTOR inhibitor everolimus. Clin Sci (Lond). 2015;129:895–914. doi:10.1042/CS20150149.
  • Pantuck AJ, Seligson DB, Klatte T, Yu H, Leppert JT, Moore L, O’Toole T, Gibbons J, Belldegrun AS, Figlin RA. Prognostic relevance of the mTOR pathway in renal cell carcinoma: implications for molecular patient selection for targeted therapy. Cancer. 2007;109:2257–67. doi:10.1002/cncr.22677.
  • Porta C, Tortora G, Larkin JM, Hutson TE. Management of poor-risk metastatic renal cell carcinoma: current approaches, the role of temsirolimus and future directions. Future Oncol. 2016;12:533–49. doi:10.2217/fon.15.313.
  • Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, et al. Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors. Cancer. 2010;116:4256–65. doi:10.1002/cncr.25219.
  • Motzer RJ, Hutson TE, Glen H, Michaelson MD, Molina A, Eisen T, Jassem J, Zolnierek J, Maroto JP, Mellado B, et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. Lancet Oncol. 2015;16:1473–82. doi:10.1016/S1470-2045(15)00290-9.
  • Inman s. FDA grants priority review to lenvatinib/everolimus combo for RCC. 2016, Jan 18.
  • Viana SD, Reis F, Alves R. Therapeutic use of mTOR inhibitors in renal diseases: advances, drawbacks, and challenges. Oxid Med Cell Longev. 2018;2018:3693625. doi:10.1155/2018/3693625.
  • Ma MKM, Yung S, Chan TM. mTOR inhibition and kidney diseases. Transplantation. 2018;102:S32–s40. doi:10.1097/TP.0000000000001729.
  • Lee KM, Stott RT, Zhao G, SooHoo J, Xiong W, Lian MM, Fitzgerald L, Shi S, Akrawi E, Lei J, et al. TGF-β-producing regulatory B cells induce regulatory T cells and promote transplantation tolerance. Eur J Immunol. 2014;44:1728–36. doi:10.1002/eji.201344062.
  • Kim CS, Kim Y, Kwon T, Yoon JH, Kim KH, You D, Hong JH, Ahn H, Jeong IG. Regulatory T cells and TGF-β1 in clinically localized renal cell carcinoma: comparison with age-matched healthy controls. Urol Oncol. 2015;33:113 e19–25. doi:10.1016/j.urolonc.2014.11.004.
  • Nguyen TL, Makhlouf NT, Anthony BA, Teague RM, Di Paolo RJ. In vitro induced regulatory T cells are unique from endogenous regulatory T cells and effective at suppressing late stages of ongoing autoimmunity. PLoS One. 2014;9:e104698. doi:10.1371/journal.pone.0104698.
  • Blair PA, Noreña LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, Mauri C. Cd19(+)cd24(hi)cd38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic lupus erythematosus patients. Immunity. 2010;32:129–40. doi:10.1016/j.immuni.2009.11.009.
  • Matz M, Lehnert M, Lorkowski C, Fabritius K, Unterwalder N, Doueiri S, Weber UA, Mashreghi M-F, Neumayer H-H, Budde K. Effects of sotrastaurin, mycophenolic acid and everolimus on human B-lymphocyte function and activation. Transpl Int. 2012;25:1106–16. doi:10.1111/j.1432-2277.2012.01537.x.
  • Ye L, Lee J, Xu L, Mohammed AU, Li W, Hale JS, Tan WG, Wu T, Davis CW, Ahmed R, et al. mTOR promotes antiviral humoral immunity by differentially regulating CD4 helper T cell and B cell responses. J Virol. 2017;91:91. doi:10.1128/JVI.01653-16.
  • Gammon JM, Gosselin EA, Tostanoski LH, Chiu YC, Zeng X, Zeng Q, Jewell CM. Low-dose controlled release of mTOR inhibitors maintains T cell plasticity and promotes central memory T cells. J Control Release. 2017;263:151–61. doi:10.1016/j.jconrel.2017.02.034.