Intratumoral delivery of TransCon™ TLR7/8 Agonist promotes sustained anti-tumor activity and local immune cell activation while minimizing systemic cytokine induction

Luis Alejandro Zúñiga, Torben Leßmann, Karan Uppal, Nicola Bisek, Enping Hong, Caroline E Rasmussen, Jens-Jakob Karlsson, Joachim Zettler, Lars Holten-Andersen, Kathy Bang, Dhruv Thakar, Yu-Chi Lee, Salomon Martinez, Simran Singh Sabharwal, Sebastian Stark, Frank Faltinger, Oliver Kracker, Samuel Weisbrod, Robin Müller, Tobias Voigt, Kornelia Bigott, Mohammad Tabrizifard, Vibeke Miller Breinholt, Amer M Mirza, David B Rosen, Kennett Sprogøe, Juha Punnonen, Luis Alejandro Zúñiga, Torben Leßmann, Karan Uppal, Nicola Bisek, Enping Hong, Caroline E Rasmussen, Jens-Jakob Karlsson, Joachim Zettler, Lars Holten-Andersen, Kathy Bang, Dhruv Thakar, Yu-Chi Lee, Salomon Martinez, Simran Singh Sabharwal, Sebastian Stark, Frank Faltinger, Oliver Kracker, Samuel Weisbrod, Robin Müller, Tobias Voigt, Kornelia Bigott, Mohammad Tabrizifard, Vibeke Miller Breinholt, Amer M Mirza, David B Rosen, Kennett Sprogøe, Juha Punnonen

Abstract

Background: Intratumoral (IT) delivery of toll-like receptor (TLR) agonists has shown encouraging anti-tumor benefit in preclinical and early clinical studies. However, IT delivery of TLR agonists may lead to rapid effusion from the tumor microenvironment (TME), potentially limiting the duration of local inflammation and increasing the risk of systemic adverse events.

Methods: To address these limitations, TransCon™ TLR7/8 Agonist-an investigational sustained-release prodrug of resiquimod that uses a TransCon linker and hydrogel technology to achieve sustained and predictable IT release of resiquimod-was developed. TransCon TLR7/8 Agonist was characterized for resiquimod release in vitro and in vivo, in mice and rats, and was assessed for anti-tumor efficacy and pharmacodynamic activity in mice.

Results: Following a single IT dose, TransCon TLR7/8 Agonist mediated potent tumor growth inhibition which was associated with sustained resiquimod release over several weeks with minimal induction of systemic cytokines. TransCon TLR7/8 Agonist monotherapy promoted activation of antigen-presenting cells in the TME and tumor-draining lymph nodes, with evidence of activation and expansion of CD8+ T cells in the tumor-draining lymph node and TME. Combination of TransCon TLR7/8 Agonist with systemic immunotherapy further promoted anti-tumor activity in TransCon TLR7/8 Agonist-treated tumors. In a bilateral tumor setting, combination of TransCon TLR7/8 Agonist with systemic IL-2 potentiated tumor growth inhibition in both injected and non-injected tumors and conferred protection against tumor rechallenge following complete regressions.

Conclusions: Our findings show that a single dose of TransCon TLR7/8 Agonist can mediate sustained local release of resiquimod in the TME and promote potent anti-tumor effects as monotherapy and in combination with systemic immunotherapy, supporting TransCon TLR7/8 Agonist as a novel intratumoral TLR agonist for cancer therapy. A clinical trial to evaluate the safety and efficacy of TransCon TLR7/8 Agonist, as monotherapy and in combination with pembrolizumab, in cancer patients is currently ongoing (transcendIT-101; NCT04799054).

Keywords: Adaptive immunity; Innate immunity; Intratumoral injection; Intratumoral treatment; Toll-like receptor; Tumor microenvironment.

Conflict of interest statement

All authors are or were employees of Ascendis Pharma and declare competing financial interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
In vivo pharmacokinetic characteristics of TransCon TLR7/8 Agonist. A TransCon TLR7/8 Agonist was designed as a sustained-release prodrug of resiquimod consisting of resiquimod bound, at multiple conjugation sites, to a suspension of polymeric PEG hydrogel microspheres via a covalent and cleavable linker (R = resiquimod; L = linker). B Male Wistar rats (n = 3/group) received a single subcutaneous injection of either soluble resiquimod (25 μg) or TransCon TLR7/8 Agonist [25 μg equivalent (eq.) of resiquimod]. Blood was withdrawn and used for plasma generation over 28 days. The resiquimod concentration in the plasma samples was quantified by LC–MS/MS. Values are represented as mean resiquimod concentration (pg/ml) per group ± SEM. C Female BALB/c mice were SC implanted with 3 × 105 CT26 tumor cells in their flank. When tumors were grown to a mean tumor volume of ~ 115 mm3, mice were randomized into treatment cohorts (Day 0). The day following randomization, animals received 5 or 20 μg (eq. of resiquimod) of TransCon TLR7/8 Agonist as a single intratumoral dose. Blood was withdrawn (n = 3/time point) and used for plasma generation at 6 h, 3 days and 7 days. The resiquimod concentration in the plasma samples was quantified by LC–MS/MS. Values are represented as mean resiquimod concentration (pg/ml) per dose group ± SEM
Fig. 2
Fig. 2
TransCon TLR7/8 Agonist promoted TGI, minimal systemic cytokine release, and sustained intratumoral cytokine expression. A Female BALB/c mice were SC implanted (flank) with 3 × 105 CT26 tumor cells. At a mean tumor volume of ~ 90 mm3, mice were randomized (Day 0; n = 10–15/group). The following day, animals received either TransCon Vehicle, 20 μg (eq. of resiquimod) of TransCon TLR7/8 Agonist, or 20 μg of soluble resiquimod (single IT dose, arrow). Values are represented as mean tumor volume ± SEM. On Day 14, *p < 0.005 vs all other groups, n = 6–11/group. B Female BALB/c mice were SC implanted (flank) with 3 × 105 CT26 tumor cells. At a mean tumor volume of ~ 120 mm3, mice were randomized (Day 0). The following day, animals received either TransCon Vehicle, 20 μg of TransCon TLR7/8 Agonist, or 20 μg of soluble resiquimod as a single IT dose. Plasma samples were (n = 3 independent mice/group/timepoint) assessed for cytokines by ProcartaPlex Multiplex Immunoassay. Values are represented as median plasma analyte concentration (pg/ml) with whiskers representing minimum and maximum replicate values. Points are connected for visualization purposes. *p < 0.05 Soluble Resiquimod vs TransCon Vehicle (same day), †p < 0.05 TransCon TLR7/8 Agonist vs TransCon Vehicle (same day), ‡p < 0.05 TransCon TLR7/8 Agonist vs Soluble Resiquimod (same day). C Female BALB/c mice were SC implanted (flank) with 3 × 105 CT26 tumor cells. At a mean tumor volume of ~ 115 mm3, mice were randomized (Day 0). The following day, animals received either TransCon Vehicle or 20 or 80 μg of TransCon TLR7/8 Agonist. Tumors were harvested and assessed for cytokine levels. Values are represented as the minimum, maximum, and median (black line) analyte concentrations normalized to protein input for each treatment group (n = 3/treatment/timepoint, except for the TransCon TLR7/8 Agonist treated group at the 168-h timepoint, where n = 2). *p < 0.05 vs TransCon Vehicle (same day)
Fig. 3
Fig. 3
TransCon TLR7/8 Agonist associated with low systemic cytokines and sustained activation of peripheral immune cells. In two independent experiments, Female BALB/c mice were SC implanted (flank) with 2 × 106 CT26 tumor cells. At a mean tumor volume of ~ 185–200 mm3, mice were randomized into treatment cohorts (Day 0; n = 18–20/group). On the same day of randomization, animals received either buffer control or TransCon TLR7/8 Agonist as a single IT dose (arrow). For each experiment, samples from 5 to 6 animals were collected for processing and analysis at each timepoint. Data from the two experiments was concatenated prior to analysis. A Mean tumor volume (mm3) ± SEM over time is shown (n = 13–38/group/timepoint). On Day 14, *p < 0.05 vs Buffer, n = 13–16/group. B Mean body weight (g) ± SEM is shown (n = 13–38/group/timepoint). C Plasma cytokine/chemokine concentrations over time are shown. D Frequency of indicated immune cell sub-type within the CD45+ gate assessed in PBMCs one day after treatment is shown. E PBMCs were isolated at various times following treatment and assessed for markers of adaptive immune-cell subsets and activation via flow cytometry. For CF *p < 0.05 vs Buffer on the same day, n = 5–11/ group/timepoint
Fig. 4
Fig. 4
IT TransCon TLR7/8 Agonist potentiated activation of APCs and T cells in tDLNs. Mice were treated as described in Fig. 3. Lymphocytes from tDLNs were isolated at various times following treatment and assessed for markers of immune-cell subsets and activation via flow cytometry. A Antigen presenting cell immunophenotyping. B Adaptive and cytolytic immune cell immunophenotyping. For A and B *p < 0.05 vs Buffer on the same day, n = 9–11/group/timepoint
Fig. 5
Fig. 5
IT TransCon TLR7/8 Agonist potentiated activation of APCs and antigen-specific CD8 T cells in tumors. Mice were treated as described in Fig. 3. Dissociated tumor cells were isolated at various times following treatment and assessed for markers of immune-cell subsets and activation via flow cytometry. A Antigen presenting cell immunophenotyping following treatment initiation. *p < 0.05 vs Buffer on the same day, n = 5–11 per group per timepoint. B CD8 T cell granzyme B and PD-1 immunophenotyping on day 14 following treatment initiation. *p < 0.05 vs Buffer, n = 9–11/group. C %AH1-tetramer+ CD8 T cells on Day 14 following treatment initiation. n = 9–11/group
Fig. 6
Fig. 6
TransCon TLR7/8 Agonist with immunotherapy promoted TGI in injected and non-injected tumors and immunological memory. A Female BALB/c mice were SC implanted (flank) with 2 × 106 CT26 tumor cells. At a mean tumor volume of ~ 80 mm3, mice were randomized into treatment cohorts (Day 0; n = 10/group). On the same day of randomization, animals received either anti-PD1, 100 μg (eq. of resiquimod) TransCon TLR7/8 Agonist as a single IT dose (arrow) or were left untreated (Control). On Day 18, *p < 0.05 vs Control, and **p < 0.05 vs control, n = 9–10/group. B Female BALB/c mice were SC implanted with 5 × 105 CT26 tumor cells into the left and right flanks. At a mean tumor volume of ~ 100 mm3, mice were randomized into treatment cohorts (Day 0). On the day of randomization, animals received either TransCon Vehicle or 216 μg (eq. of resiquimod) of TransCon TLR7/8 Agonist as a single intratumoral dose (arrow). Some cohorts were further treated with 20 μg human IL-2 dosed twice daily on Days 0–4 and once daily on Days 8–12. In this experiment, 3 out of 7 mice treated with TransCon TLR7/8 Agonist + IL-2 experienced complete regressions in injected and non-injected tumors. On Day 16, *p < 0.05 vs TransCon Vehicle and **p < 0.05 vs all other groups, n = 7/group. For non-injected tumors on Day 18, †p < 0.05 vs Control and ‡p < 0.05 vs Control and TransCon TLR7/8 Agonist, n = 7/group. C Mice from B that were treated with TransCon TLR7/8 Agonist and IL-2, and that experienced complete regressions in both treated and untreated tumors for at least 10 days (n = 3), were SC rechallenged with CT26 tumor cells in their front right dorsal region and observed for tumor growth. Naïve mice (n = 10) were used as controls for tumor growth. Values are represented as mean tumor volume ± SEM

References

    1. Zhang G, Ghosh S. Toll-like receptor-mediated NF-kappaB activation: a phylogenetically conserved paradigm in innate immunity. J Clin Invest. 2001;107:13–19. doi: 10.1172/JCI11837.
    1. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5:987–995. doi: 10.1038/ni1112.
    1. Yang Y, Huang CT, Huang X, Pardoll DM. Persistent Toll-like receptor signals are required for reversal of regulatory T cell-mediated CD8 tolerance. Nat Immunol. 2004;5:508–515. doi: 10.1038/ni1059.
    1. Urban-Wojciuk Z, et al. The role of TLRs in anti-cancer immunity and tumor rejection. Front Immunol. 2019;10:2388. doi: 10.3389/fimmu.2019.02388.
    1. Mangsbo SM, Sandin LC, Anger K, Korman AJ, Loskog A, Totterman TH. Enhanced tumor eradication by combining CTLA-4 or PD-1 blockade with CpG therapy. J Immunother. 2010;33:225–235. doi: 10.1097/CJI.0b013e3181c01fcb.
    1. Del Rosso JQ. The use of topical imiquimod for the treatment of actinic keratosis: a status report. Cutis. 2005;76:241–248.
    1. Rook AH, et al. Topical resiquimod can induce disease regression and enhance T-cell effector functions in cutaneous T-cell lymphoma. Blood. 2015;126:1452–1461. doi: 10.1182/blood-2015-02-630335.
    1. Michaelis KA, et al. The TLR7/8 agonist R848 remodels tumor and host responses to promote survival in pancreatic cancer. Nat Commun. 2019;10:4682. doi: 10.1038/s41467-019-12657-w.
    1. Spinetti T, et al. TLR7-based cancer immunotherapy decreases intratumoral myeloid-derived suppressor cells and blocks their immunosuppressive function. Oncoimmunology. 2016;5:e1230578. doi: 10.1080/2162402X.2016.1230578.
    1. Sato-Kaneko F, et al. Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight. 2017 doi: 10.1172/jci.insight.93397.
    1. Salazar LG, et al. Topical imiquimod plus nab-paclitaxel for breast cancer cutaneous metastases: a phase 2 clinical trial. JAMA Oncol. 2017;3:969–973. doi: 10.1001/jamaoncol.2016.6007.
    1. Diab A, et al. Phase Ib: preliminary clinical activity and immune activation for NKTR-262 [TLR 7/8 agonist] plus NKTR-214 [CD122-biased agonist] in patients (pts) with locally advanced or metastatic solid tumors (REVEAL Phase Ib/II Trial) J Clin Oncol. 2019;37:26–26. doi: 10.1200/JCO.2019.37.8_suppl.26.
    1. Milhem MM, et al. Phase 1b/2, open label, multicenter, study of the combination of SD-101 and pembrolizumab in patients with advanced melanoma who are naïve to anti-PD-1 therapy. J Clin Oncol. 2019;37:9534–9534. doi: 10.1200/JCO.2019.37.15_suppl.9534.
    1. Ribas A, et al. SD-101 in combination with pembrolizumab in advanced melanoma: results of a phase Ib multicenter study. Cancer Discov. 2018;8:1250–1257. doi: 10.1158/-18-0280.
    1. Kirkwood J, et al. 950 Final analysis: phase 1b study investigating intratumoral injection of toll-like receptor 9 agonist vidutolimod ± pembrolizumab in patients with PD-1 blockade–refractory melanoma. J Immunother Cancer. 2021;9:A999–A999. doi: 10.1136/jitc-2021-SITC2021.950.
    1. Babiker HM, et al. Abstract 4062: activation of innate and adaptive immunity using intratumoral tilsotolimod (IMO-2125) as monotherapy in patients with refractory solid tumors: a phase Ib study (ILLUMINATE-101) Can Res. 2019;79:4062–4062. doi: 10.1158/1538-7445.AM2019-4062.
    1. Dowling DJ, et al. TLR7/8 adjuvant overcomes newborn hyporesponsiveness to pneumococcal conjugate vaccine at birth. JCI Insight. 2017;2:e91020. doi: 10.1172/jci.insight.91020.
    1. Engel AL, Holt GE, Lu H. The pharmacokinetics of Toll-like receptor agonists and the impact on the immune system. Expert Rev Clin Pharmacol. 2011;4:275–289. doi: 10.1586/ecp.11.5.
    1. Lee M. Abstract 2755: NKTR-262: Prodrug pharmacokinetics in mice, rats, and dogs. Can Res. 2018;78:2755–2755. doi: 10.1158/1538-7445.AM2018-2755.
    1. FDA SKYTROFA. . Accessed 2 Aug 2022
    1. EMA Lonapegsomatropin. . Accessed 2 Aug 2022
    1. Hackstein H, et al. The TLR7/8 ligand resiquimod targets monocyte-derived dendritic cell differentiation via TLR8 and augments functional dendritic cell generation. Cell Immunol. 2011;271:401–412. doi: 10.1016/j.cellimm.2011.08.008.
    1. Brugnolo F, et al. The novel synthetic immune response modifier R-848 (Resiquimod) shifts human allergen-specific CD4+ TH2 lymphocytes into IFN-gamma-producing cells. J Allergy Clin Immunol. 2003;111:380–388. doi: 10.1067/mai.2003.102.
    1. van Aalst S, Jansen MAA, Ludwig IS, van der Zee R, van Eden W, Broere F. Routing dependent immune responses after experimental R848-adjuvated vaccination. Vaccine. 2018;36:1405–1413. doi: 10.1016/j.vaccine.2018.01.077.
    1. Paster EV, Villines KA, Hickman DL. Endpoints for mouse abdominal tumor models: refinement of current criteria. Comp Med. 2009;59:234–241.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Northfelt DW, et al. A phase I dose-finding study of the novel Toll-like receptor 8 agonist VTX-2337 in adult subjects with advanced solid tumors or lymphoma. Clin Cancer Res. 2014;20:3683–3691. doi: 10.1158/1078-0432.CCR-14-0392.
    1. Haymaker C, et al. Tilsotolimod with ipilimumab drives tumor responses in anti-PD-1 refractory melanoma. Cancer Discov. 2021;11:1996–2013. doi: 10.1158/-20-1546.
    1. Chandorkar Y, Ravikumar K, Basu B. The foreign body response demystified. ACS Biomater Sci Eng. 2019;5:19–44. doi: 10.1021/acsbiomaterials.8b00252.
    1. Tsou CL, et al. Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow and recruitment to inflammatory sites. J Clin Invest. 2007;117:902–909. doi: 10.1172/JCI29919.
    1. Rahman K, et al. Inflammatory Ly6Chi monocytes and their conversion to M2 macrophages drive atherosclerosis regression. J Clin Invest. 2017;127:2904–2915. doi: 10.1172/JCI75005.
    1. Dinter J, et al. Variable processing and cross-presentation of HIV by dendritic cells and macrophages shapes CTL immunodominance and immune escape. PLoS Pathog. 2015;11:e1004725. doi: 10.1371/journal.ppat.1004725.
    1. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10. doi: 10.1016/j.immuni.2013.07.012.
    1. Sheikh NA, Jones LA. CD54 is a surrogate marker of antigen presenting cell activation. Cancer Immunol Immunother. 2008;57:1381–1390. doi: 10.1007/s00262-008-0474-9.
    1. Wrammert J, Kallberg E, Agace WW, Leanderson T. Ly6C expression differentiates plasma cells from other B cell subsets in mice. Eur J Immunol. 2002;32:97–103. doi: 10.1002/1521-4141(200201)32:1<97::AID-IMMU97>;2-Y.
    1. Zhong W, et al. Comparison of the molecular and cellular phenotypes of common mouse syngeneic models with human tumors. BMC Genomics. 2020;21:2. doi: 10.1186/s12864-019-6344-3.
    1. Harrison LI, Astry C, Kumar S, Yunis C. Pharmacokinetics of 852A, an imidazoquinoline Toll-like receptor 7-specific agonist, following intravenous, subcutaneous, and oral administrations in humans. J Clin Pharmacol. 2007;47:962–969. doi: 10.1177/0091270007303766.
    1. Pockros PJ, et al. Oral resiquimod in chronic HCV infection: safety and efficacy in 2 placebo-controlled, double-blind phase IIa studies. J Hepatol. 2007;47:174–182. doi: 10.1016/j.jhep.2007.02.025.
    1. Smith M, et al. Trial Watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunology. 2018;7:e1526250. doi: 10.1080/2162402X.2018.1526250.
    1. Cohen EEW, et al. Phase 1b/2, open label, multicenter study of intratumoral SD-101 in combination with pembrolizumab in anti-PD-1 treatment naïve patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) J Clin Oncol. 2019;37:6039–6039. doi: 10.1200/JCO.2019.37.15_suppl.6039.
    1. Garon EB, et al. A Phase ib open-label, multicenter study of inhaled dv281, a tlr9 agonist, in combination with nivolumab in patients with advanced or metastatic non-small cell lung cancer. Clin Cancer Res. 2021;27:4566–4573. doi: 10.1158/1078-0432.CCR-21-0263.
    1. Zalevsky J. NKTR-214 plus NKTR-262, a scientifically-guided rational combination approach for immune oncology. World Preclinical Congress. 2017.
    1. Park CG, Hartl CA, Schmid D, Carmona EM, Kim HJ, Goldberg MS. Extended release of perioperative immunotherapy prevents tumor recurrence and eliminates metastases. Sci Transl Med. 2018 doi: 10.1126/scitranslmed.aar1916.
    1. Widmer J, et al. Polymer-based nanoparticles loaded with a TLR7 ligand to target the lymph node for immunostimulation. Int J Pharm. 2018;535:444–451. doi: 10.1016/j.ijpharm.2017.11.031.
    1. Thauvin C, et al. Development of resiquimod-loaded modified PLA-based nanoparticles for cancer immunotherapy: a kinetic study. Eur J Pharm Biopharm. 2019;139:253–261. doi: 10.1016/j.ejpb.2019.04.007.
    1. Rodell CB, et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat Biomed Eng. 2018;2:578–588. doi: 10.1038/s41551-018-0236-8.
    1. Wagner J, et al. Mesoporous silica nanoparticles as pH-responsive carrier for the immune-activating drug resiquimod enhance the local immune response in mice. ACS Nano. 2021;15:4450–4466. doi: 10.1021/acsnano.0c08384.
    1. Wang S, et al. Amelioration of systemic antitumor immune responses in cocktail therapy by immunomodulatory nanozymes. Sci Adv. 2022 doi: 10.1126/sciadv.abn3883.
    1. Bourquin C, et al. Systemic cancer therapy with a small molecule agonist of toll-like receptor 7 can be improved by circumventing TLR tolerance. Cancer Res. 2011;71:5123–5133. doi: 10.1158/0008-5472.CAN-10-3903.
    1. Asselin-Paturel C, et al. Type I interferon dependence of plasmacytoid dendritic cell activation and migration. J Exp Med. 2005;201:1157–1167. doi: 10.1084/jem.20041930.
    1. Torvinen M, Campwala H, Kilty I. The role of IFN-gamma in regulation of IFN-gamma-inducible protein 10 (IP-10) expression in lung epithelial cell and peripheral blood mononuclear cell co-cultures. Respir Res. 2007;8:80. doi: 10.1186/1465-9921-8-80.
    1. Kuo PT, Zeng Z, Salim N, Mattarollo S, Wells JW, Leggatt GR. The role of CXCR3 and its chemokine ligands in skin disease and cancer. Front Med. 2018 doi: 10.3389/fmed.2018.00271.
    1. Sharonov GV, Serebrovskaya EO, Yuzhakova DV, Britanova OV, Chudakov DM. B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat Rev Immunol. 2020;20:294–307. doi: 10.1038/s41577-019-0257-x.
    1. Kroeger DR, Milne K, Nelson BH. Tumor-infiltrating plasma cells are associated with tertiary lymphoid structures, cytolytic t-cell responses, and superior prognosis in ovarian cancer. Clin Cancer Res. 2016;22:3005–3015. doi: 10.1158/1078-0432.CCR-15-2762.
    1. Werner F, et al. A standardized analysis of tertiary lymphoid structures in human melanoma: disease progression- and tumor site-associated changes with germinal center alteration. Front Immunol. 2021;12:675146. doi: 10.3389/fimmu.2021.675146.
    1. Wong RM, et al. TLR-9 signaling and TCR stimulation co-regulate CD8(+) T cell-associated PD-1 expression. Immunol Lett. 2009;127:60–67. doi: 10.1016/j.imlet.2009.09.002.
    1. Zahm CD, Colluru VT, McIlwain SJ, Ong IM, McNeel DG. TLR Stimulation during T-cell Activation Lowers PD-1 Expression on CD8(+) T Cells. Cancer Immunol Res. 2018;6:1364–1374. doi: 10.1158/2326-6066.CIR-18-0243.
    1. Maleki VS. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer. 2018;6:157. doi: 10.1186/s40425-018-0479-7.

Source: PubMed

3
Abonner