Pan-TGFβ inhibition by SAR439459 relieves immunosuppression and improves antitumor efficacy of PD-1 blockade

Rita Greco, Hongjing Qu, Hui Qu, Joachim Theilhaber, Gary Shapiro, Richard Gregory, Christopher Winter, Natalia Malkova, Frank Sun, Julie Jaworski, Annie Best, Lily Pao, Andrew Hebert, Mikhail Levit, Alexei Protopopov, Jack Pollard, Keith Bahjat, Dmitri Wiederschain, Sharad Sharma, Rita Greco, Hongjing Qu, Hui Qu, Joachim Theilhaber, Gary Shapiro, Richard Gregory, Christopher Winter, Natalia Malkova, Frank Sun, Julie Jaworski, Annie Best, Lily Pao, Andrew Hebert, Mikhail Levit, Alexei Protopopov, Jack Pollard, Keith Bahjat, Dmitri Wiederschain, Sharad Sharma

Abstract

TGFβ is a pleiotropic cytokine that may have both tumor inhibiting and tumor promoting properties, depending on tissue and cellular context. Emerging data support a role for TGFβ in suppression of antitumor immunity. Here we show that SAR439459, a pan-TGFβ neutralizing antibody, inhibits all active isoforms of human and murine TGFβ, blocks TGFβ-mediated pSMAD signaling, and TGFβ-mediated suppression of T cells and NK cells. In vitro, SAR439459 synergized with anti-PD1 to enhance T cell responsiveness. In syngeneic tumor models, SAR439459 treatment impaired tumor growth, while the combination of SAR439459 with anti-PD-1 resulted in complete tumor regression and a prolonged antitumor immunity. Mechanistically, we found that TGFβ inhibition with PD-1 blockade augmented intratumoral CD8+ T cell proliferation, reduced exhaustion, evoked proinflammatory cytokines, and promoted tumor-specific CD8+ T cell responses. Together, these data support the hypothesis that TGFβ neutralization using SAR439459 synergizes with PD-1 blockade to promote antitumor immunity and formed the basis for the ongoing clinical investigation of SAR439459 in patients with cancer (NCT03192345).

Keywords: PD1 blockade; T cell response; TGFβ; Tumor microenvironment; combination immunotherapy.

© 2020 The Author(s). Published with license by Taylor & Francis Group, LLC.

Figures

Figure 1.
Figure 1.
Higher TGFβ pathway activation is associated with worse objective response and lower overall survival under anti–PD-1/PD-L1 therapies. a–c. High TGFβ pathway activation correlates with resistance to PD-1/PD-L1 therapies and with reduced overall survival. Box plots show TGFβ activation signatures in nonresponders (NR) and responders (R) in (a), melanoma data from Hugo et al.; (b), melanoma data from Sade-Feldman; and (c), urothelial cancer data. Enrichment scores are obtained via a regulated Kolmogorov-Smirnov analysis using the RNA sequencing whole-transcriptome profiles available in each study. P values are from Wilcoxon tests. (d), Overall survival for the groups of patients with urothelial cancer who had TGFβ-low pathway activation (n = 243; enrichment score ≥1) and TGFβ-high pathway activation (n = 55; enrichment score <1). TGFβ-low and TGFβ-high data from urothelial cancer was further subdivided on the basis of the respective cytotoxic T lymphocyte (CTL) scores, and patient survival was compared in each case. (e), Survival curves are shown for the TGFβ-low patients, and (f), for the TGFβ-high patients. Box plots show CTL scores in TGFβ-high and TGFβ-low groups (high versus low TGFβ pathway activation), which were not different (g).
Figure 2.
Figure 2.
SAR439459 binds to various TGFβ isoforms and inhibits TGFβ-mediated signaling similar to fresolimumab. SPR Biacore data shows similar binding properties of SAR439459 and fresolimumab against human TGFβ1, 2, and 3 proteins, (a). Murine colon carcinoma, MC38; mouse breast cancer cells, EMT6 and human colorectal carcinoma cells HCT116-overexpressing TGFβRII were cultured with human TGFβ1 (1 ng/mL) in presence of SAR439459, fresolimumab or isotype control. Total and phosphorylated SMAD2/3 protein levels were assessed by ELISA. Graph shows ratio of phospho-/total SMAD2/3 for SAR439459 and fresolimumab and isotype control, (b). P value <.0001 using two-way ANOVA test. SBE reporter cells were cultured in the presence of human TGFβ isoforms −1, −2, and −3 with various concentrations of SAR439459, fresolimumab or isotype control and luminescence activity was measured. Graphs show ability of SAR439459 and fresolimumab to prevent TGFβ1, 2 and 3-mediated SMAD activation in the reporter cell line, (c). IC50 values for SAR439459 were 0.008, 1.22, 0.45, and for fresolimumab were 0.02, 1.86, 0.61 nM for TGFβ1, TGFβ2, and TGFβ3 respectively. Each graph shows mean ± SEM and represents one of three independent experiments.
Figure 3.
Figure 3.
SAR439459 restores TGFβ-mediated suppression of primary immune cell function. Enriched CTV-labeled human CD8+ T cells were co-cultured with B-LCL cells in the presence of TGFβ1 (1 ng/mL) and, to analyze effect on CD8+ T cell proliferation and IFNγ, measured by FACS. Cells were gated on live CD8+ T cell population followed by CTV-low and IFNγ-positive cells. FACS plots show proliferation (CTV-low) of CD8+ T cells producing IFNγ under various treatment conditions, (a). Bar graphs show mean ± SEM percentage of CD8+ T cells that were functional CTV-low–IFNγ+, (b). Data are representative of 3 independent experiments with CD8+ T cells from 2 human donors. P values: ***<0.005, **<0.05, ***<0.001, ****<0.0001 for SAR439459 versus isotype at doses (µg/mL) 12.5, 25, 50, and 100, respectively. Enriched NK cells from human donors were cultured in the presence of IL-2 for 2–3 days and with various concentrations of human TGFβ1 (0.1, 1.0, and 10 ng/mL) as shown in the FACS plots. NK proliferation was examined by Ki-67 staining. FACS plots show percentages of live Ki-67-positive NK cells in the presence of varying TGFβ1 doses, (c). FACS plots show NK cell proliferation in presence of TGFβ1 (1 ng/mL) and its blockade by SAR439459, (d). Bar graph shows IL-2–induced NK cell proliferation in presence of various doses of TGFβ (0.1, 1.0, and 10 ng/mL) and treated with SAR439459 (50 nM), human IgG4, or untreated control, (e). P values: ***<0.01, ****<0.0001, and ***<0.05 at 0.1, 1.0, and 10 ng/mL TGFβ1, respectively. FACS plots show percentages of K562 cells positive for cleaved granzyme B (GZB) peptides delivered by NK activity under various conditions, (f). Bar graph shows calculated percentage of cleaved granzyme B peptides in K562 cells (co-cultured with NK cells) in the presence of 1 ng/mL TGFβ1 under various treatment conditions, with or without SAR439459 (50 nM) or human IgG4, (g). P values: **<0.005, ****<0.0001 at 0.1 and 1.0 ng/mL TGFβ1, respectively. One of three experiments is shown. APC, antigen-presenting cell; FITC, fluorescein isothiocyanate. NS, not significant. Graphs show dose dependent induction of GZB, perforin, TNFa and MIP1a in the culture supernatants of K562: NK cell co-culture experiments, (h). Data represents one of two experiments. Bar graphs show IFNγ produced in a human T cell: DC co-culture MLR assay with SAR439459 or fresolimumab treatments or isotype control, (i). P values: SAR439459 vs. untreated = 0.02; fresolimumab vs. untreated = 0.007. SAR439459 vs fresolimumab, NS.
Figure 4.
Figure 4.
PD-1 blockade-mediated T cell response is further enhanced by SAR439459. Total T cells and monocyte-derived dendritic cells from healthy human donors were co-cultured in a ratio of 10:1 with anti–PD-1 (10 nM) alone, SAR439459 (50 nM) alone, their combination, or isotype controls. Bar graph shows mean ± SEM level of IFNγ under various treatments, (a). Assay was set up with or without addition of exogenous TGFβ1 to the cultures. P values are shown between isotype control vs SAR439459 for all treatment conditions: *<0.01, **<0.008, a = 0.08, b = 0.03. Experiment was performed in 2 different donors and repeated at least four times. Graph shows mean ± SEM luciferase activity from NFAT reporter Jurkat assay under similar treatment conditions as detailed in Materials and Methods section, (b). Data represent at least 3 independent experiments. P value: ****<0.0001. RLU, relative light units. In MLR assay data demonstrates production of IL-2, GZB, Perforin, IL-6, IL-10 and TNF by TGFβ and PD1 co-inhibition, (c). P values: Combination vs. anti-PD1 or SAR439459 alone, IL2 < 0.0004 and <0.0001, GZB 0.0003 and 0.0001, Perforin, 0.01 and 0.08, IL-6 0.001 and 0.01, IL-10 < 0.0001 both and TNFα 0.03 both.
Figure 5.
Figure 5.
SAR439459 improves antitumor efficacy of PD-1 blockade in mouse tumor models and combination establishes long-lasting immunity. Antitumor efficacy of SAR439459 in combination with PD-1 antibody was examined in mouse colon and breast carcinomas, MC38 and EMT-6, respectively. Graph shows time dependent MC38 tumor volumes over time after receiving various treatments as detailed in Materials and Methods (a). Survival curves of mice from MC38 study over several days. P values: *<0.05, **<0.005 (b). Graph depicts EMT-6 breast cancer tumor volumes over time in mice after receiving various treatments (c). Survival data from EMT-6 tumor model at various days (d). Mice surviving EMT-6 experiment in combination and SAR439459 monotherapy group were re-challenged with EMT-6 tumors, (e). Curves show that tumors were immediately rejected in these mice. Tumor growth inhibition in Lovo model by SAR439459 and fresolimumab as a monotherapy at day 31 and 35 post tumor implantation, (f). Data was compared with two-way ANOVA and no difference was observed between SAR439459 vs. fresolimumab.
Figure 6.
Figure 6.
SAR439459 in combination with PD-1 blockade elicits proinflammatory cytokine response, tumor-specific CD8+ T cell response. Mice were implanted with various types of tumors and treated with three doses of SAR439459 alone, PD-1 antibody alone, or their combination. Control groups received either vehicle (PBS) or similar doses of isotype antibodies in combination. Intratumoral cytokines were evaluated. Bar graphs show mean ± SEM intratumoral levels of A, IFNγ, combo versus PBS, *P = .01; combo versus isotype, P = .07; B, TNFα, combo versus PBS, *P = .06; C, IL-6, combo versus PBS, *P = .04; and D, TGFβ, combo versus PBS, ***P = .0004; combo versus isotype, **P = .001; combo vs anti–PD-1, ***P = .0003; SAR439459 versus PBS, **P = .001; SAR439459 versus isotype, **P = .005; SAR439459 versus anti–PD-1, **P = .003. n = 5 mice/each group, and data are representative of two or more experiments. Mice implanted with EMT6 tumors were treated with three doses of anti-TGFβ (1D11), anti-PD1, their combination or vehicle (n = 5 each group) and intratumoral levels of various cytokines IFNγ, TNFα, IL-6 and TGFβ are shown, (e-h). P values; PBS vs. combination, 0.06 for IFNγ, 0.3 for TNFα, 0.1 IL-6. For TGFβ, PBS vs. combination = 0.0001 and anti-TGFβ = 0.0004, and anti-PD1 vs. combination = 0.0004 and anti-TGFβ = 0.002. In addition, time course evaluation of IFNγ was performed after various treatments in EMT6 tumor model, (i). Two-way ANOVA analysis show interaction P value 0.0009 and column factor 0.0013. Intratumoral levels of TGFβ were analyzed in Lovo model after treatment with different doses of SAR439459 or fresolimumab, (j). Mice were implanted with MC38OVA tumors and divided into various treatment groups. Tumors and draining lymph nodes were examined for presence of total and OVA-specific CD8+ T cells using FACS. Dot plots show dextramer staining on tumor samples among various groups. Graph shows counts of OVA-specific CD8+ T cells normalized to tumor weight, (k). P value combination versus isotype: *<0.03. Graph shows mean percentages of total and OVA-specific CD8+ T cells in tumor-draining lymph nodes among various treatment and control groups (l) & (m) respectively. P value, **0.009 combination versus isotype control. TDLN, tumor-draining lymph node.
Figure 7.
Figure 7.
Single-cell RNA sequencing profiling in MC38 tumor model indicates that SAR439459 induces proliferative phenotype in intratumoral CD8+ T cells. Dimensional reduction and tree inference by a graphical clustering method on a set of 3,476 CD8+ T cells generated from MC38 tumor models in 12 mice assigned to 4 treatment groups (all 4 treatment groups shown together) clusters the T cells into 5 distinct “states” (see Methods). Cells are displayed in accordance to their DDRTree coordinates; numeric membership per state is indicated next to each label, (a). Bar plots showing fractional membership in states 1 and 5 for CD8+ T cells as a function of treatment. ANOVA P values (n = 12) are indicated in the plot headers. The frequency of cells in state 1 increases under the sequence of treatments (isotype-control < anti-PD-1 < SAR439459 < SAR439459+ anti-PD-1), from about 35% to 45%, while the frequency of cells in state 5 markedly decreases in the same order, from about 45% to 15%. No such trends are seen for the states 2, 3 and 4. States 1 and 5 accounted for 75% of the CD8+ T cells, while state 4 accounted for about 15% and state 2 and 3 for approximately 10% of the total. P values are shown (b). Heat map of genes selected for specificity to each of the 5 CD8+ T cell states. The top 50 genes ranked by specificity were selected for each state (some overlap occurs between gene sets, resulting in only 227 genes selected overall). Colors represent Z-transformed log2 (transcripts per 100,000 + 1) values. While state 1 shows marked expression of genes for cell cycle and cellular proliferation (inset shows representative genes in this group), state 5 shows marked expression of genes for cytotoxic or checkpoint proteins (see inset). State 4 is enriched in genes induced by type I interferons, but its membership did not significantly change with treatment (c). Gene set enrichment analysis confirms engagement of cellular proliferation pathways in many of the state 1 CD8+ T cells, and in none of the state 5 CD8+ T cells; thus, a signature consisting of G2/M transition genes scores positively for many cells in state 1, and for none in state 5, (d). Comparison of 2 signature based CD8+ T cell categories, “primed/activated” versus “exhausted,” shows that state 1 has about twice as many primed/activated cells than state 5, with respective proportions 40% and 20%, (e).

References

    1. Wei SC, Duffy CR, Allison JP.. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8:1069–18. doi:10.1158/-18-0367.
    1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61. doi:10.1126/science.aaa8172.
    1. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 1627-39;2015:373.
    1. Herbst RS, Baas P, Kim D-W, Felip E, Perez-Gracia JL, Han J-Y, Molina J, Kim J-H, Arvis CD, Ahn M-J, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–1550. doi:10.1016/S0140-6736(15)01281-7.
    1. Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol. 2016;27:1492–1504. doi:10.1093/annonc/mdw217.
    1. Miao Y, Yang H, Levorse J, Yuan S, Polak L, Sribour M, Singh B, Rosenblum MD, Fuchs E. Adaptive Immune Resistance Emerges from Tumor-Initiating Stem Cells. Cell. 2019;177(5):1172–86 e14. doi:10.1016/j.cell.2019.03.025.
    1. Pitt JM, Vetizou M, Daillere R, Roberti MP, Yamazaki T, Routy B, Lepage P, Boneca I, Chamaillard M, Kroemer G. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity. 2016;44(6):1255–1269. doi:10.1016/j.immuni.2016.06.001.
    1. Principe DR, Doll JA, Bauer J, Jung B, Munshi HG, Bartholin L, Pasche B, Lee C, Grippo PJ. TGF- : duality of function between tumor prevention and carcinogenesis. J Natl Cancer Inst. 2014;106(2):djt369. doi:10.1093/jnci/djt369.
    1. Wrzesinski SH, Wan YY, Flavell RA. Transforming growth Factor- and the immune response: implications for anticancer therapy. Clin Cancer Res. 2007;13(18):5262–5270. doi:10.1158/1078-0432.CCR-07-1157.
    1. Travis MA, Sheppard D. TGF-beta activation and function in immunity. Annu Rev Immunol. 2014;32:51–82. doi:10.1146/annurev-immunol-032713-120257.
    1. Batlle E, Massague J. Transforming growth factor-beta signaling in immunity and cancer. Immunity. 2019;50:924–940. doi:10.1016/j.immuni.2019.03.024.
    1. Seystahl K, Papachristodoulou A, Burghardt I, Schneider H, Hasenbach K, Janicot M, Roth P, Weller M. Biological Role and Therapeutic Targeting of TGF-β3 in Glioblastoma. Mol Cancer Ther. 2017;16:1177–1186. doi:10.1158/1535-7163.MCT-16-0465.
    1. Shah S, Qiao L. Resting B cells expand a CD4+CD25+Foxp3+ Treg population via TGF-beta3. Eur J Immunol. 2008;38:2488–2498. doi:10.1002/eji.200838201.
    1. Ma GF, Miao Q, Zeng XQ, Luo TC, Ma LL, Liu YM, Lian -J-J, Gao H, Chen S-Y. Transforming growth factor-beta1 and -beta2 in gastric precancer and cancer and roles in tumor-cell interactions with peripheral blood mononuclear cells in vitro. PLoS One. 2013;8:e54249. doi:10.1371/journal.pone.0054249.
    1. Teicher BA. Transforming growth factor-beta and the immune response to malignant disease. Clin Cancer Res. 2007;13:6247–6251. doi:10.1158/1078-0432.CCR-07-1654.
    1. Marie JC, Letterio JJ, Gavin M, Rudensky AY. TGF-beta1 maintains suppressor function and Foxp3 expression in CD4+CD25+ regulatory T cells. J Exp Med. 2005;201:1061–1067. doi:10.1084/jem.20042276.
    1. Chen CH, Seguin-Devaux C, Burke NA, Oriss TB, Watkins SC, Clipstone N, et al. Transforming growth factor beta blocks Tec kinase phosphorylation, Ca2+ influx, and NFATc translocation causing inhibition of T cell differentiation. J Exp Med. 1689-99;2003:197.
    1. McKarns SC, Schwartz RH, Kaminski NE. Smad3 is essential for TGF-beta 1 to suppress IL-2 production and TCR-induced proliferation, but not IL-2-induced proliferation. J Immunol. 2004;172:4275–4284. doi:10.4049/jimmunol.172.7.4275.
    1. Kasid A, Bell GI, Director EP. Effects of transforming growth factor-beta on human lymphokine-activated killer cell precursors. Autocrine inhibition of cellular proliferation and differentiation to immune killer cells. J Immunol. 1988;141:690–698.
    1. Kuppner MC, Hamou MF, Bodmer S, Fontana A, de Tribolet N. The glioblastoma-derived T-cell suppressor factor/transforming growth factor beta 2 inhibits the generation of lymphokine-activated killer (LAK) cells. Int J Cancer. 1988;42:562–567. doi:10.1002/ijc.2910420416.
    1. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–548. doi:10.1038/nature25501.
    1. Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, Sevillano M, Ibiza S, Cañellas A, Hernando-Momblona X, et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. 2018;554:538–543. doi:10.1038/nature25492.
    1. Colak S, Ten Dijke P, Targeting TG. F-beta Signaling in Cancer. Trends Cancer. 2017;3:56–71. doi:10.1016/j.trecan.2016.11.008.
    1. Park BV, Freeman ZT, Ghasemzadeh A, Chattergoon MA, Rutebemberwa A, Steigner J, Winter ME, Huynh TV, Sebald SM, Lee S-J, et al. TGFbeta1-mediated SMAD3 Enhances PD-1 expression on antigen-specific t cells in cancer. Cancer Discov. 2016;6:1366–1381. doi:10.1158/-15-1347.
    1. Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity. 2018;48:434–452. doi:10.1016/j.immuni.2018.03.014.
    1. Formenti SC, Hawtin RE, Dixit N, Evensen E, Lee P, Goldberg JD, Li X, Vanpouille-Box C, Schaue D, McBride WH, et al. Baseline T cell dysfunction by single cell network profiling in metastatic breast cancer patients. J Immunother Cancer. 2019;7(1):177. doi:10.1186/s40425-019-0633-x.
    1. Formenti SC, Lee P, Adams S, Goldberg JD, Li X, Xie MW, Ratikan JA, Felix C, Hwang L, Faull KF, et al. Focal Irradiation and systemic TGFβ blockade in metastatic breast cancer. Clin Cancer Res. 2018;24(11):2493–2504. doi:10.1158/1078-0432.CCR-17-3322.
    1. Morris JC, Tan AR, Olencki TE, Shapiro GI, Dezube BJ, Reiss M, Hsu FJ, Berzofsky JA, Lawrence DP, et al. Phase I study of GC1008 (Fresolimumab): a human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One. 2014;9(3):e90353. doi:10.1371/journal.pone.0090353.
    1. Stevenson JP, Kindler HL, Papasavvas E, Sun J, Jacobs-Small M, Hull J, Schwed D, Ranganathan A, Newick K, Heitjan DF, et al. Immunological effects of the TGFβ-blocking antibody GC1008 in malignant pleural mesothelioma patients. Oncoimmunology. 2013;2(8):e26218. doi:10.4161/onci.26218.
    1. Theilhaber JCJ, Madden SL, Manning C, Cao S, Mankoo P, Pomponio R, Qu H, Malkova N, Shapiro G, Winter C, et al. . Abstract 5550: translational biomarkers for SAR439459, an anti-TGFβ antibody for cancer immunotherapy. . Proceedings: AACR Annual Meeting 2018. Chicago, IL 2018.
    1. Theilhaber J, Rakhade SN, Sudhalter J, Kothari N, Klein P, Pollard J, Jensen FE. Gene expression profiling of a hypoxic seizure model of epilepsy suggests a role for mTOR and Wnt signaling in epileptogenesis. PLoS One. 2013;8(9):e74428. doi:10.1371/journal.pone.0074428.
    1. Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, Selves J, Laurent-Puig P, Sautès-Fridman C, Fridman WH. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17(1):218. doi:10.1186/s13059-016-1070-5.
    1. Bellone G, Aste-Amezaga M, Trinchieri G, Rodeck U. Regulation of NK cell functions by TGF-beta 1. J Immunol. 1995;155:1066–1073.
    1. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, Rinn JL. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014;32:381–386. doi:10.1038/nbt.2859.
    1. Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas A, Kamitaki N, Martersteck E. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell. 2015;161:1202–1214. doi:10.1016/j.cell.2015.05.002.
    1. Tirosh I, Izar B, Prakadan SM, Wadsworth MH 2nd, Treacy D, Trombetta JJ, Rotem A, Rodman C, Lian C, Murphy G. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016;352(6282):189–196. doi:10.1126/science.aad0501.
    1. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44. doi:10.1016/j.cell.2016.02.065.
    1. Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, Jenkins RW, Lieb DJ, Chen JH, Frederick DT, Barzily-Rokni M. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018;175(4):998–1013 e20. doi:10.1016/j.cell.2018.10.038.
    1. Anti-TGF-Beta antibodies and their uses. WO 2018/134681 A1. 2018
    1. Moulin A, Mathieu M, Lawrence C, Bigelow R, Levine M, Hamel C, Marquette J-P, Le Parc J, Loux C, Ferrari P. Structures of a pan-specific antagonist antibody complexed to different isoforms of TGFβ reveal structural plasticity of antibody-antigen interactions. Protein Sci. 2014;23(12):1698–1707. doi:10.1002/pro.2548.
    1. Bai X, Yi M, Jiao Y, Chu Q, Wu WK. Blocking TGF-β signaling to enhance the efficacy of immune checkpoint inhibitor. Onco Targets Ther. 2019;12:9527–9538. doi:10.2147/OTT.S224013.
    1. Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018;48:812–30 e14.
    1. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348(3):203–213. doi:10.1056/NEJMoa020177.
    1. Fransen MF, Schoonderwoerd M, Knopf P, Camps MG, Hawinkels LJ, Kneilling M, van Hall T, Ossendorp F. Tumor-draining lymph nodes are pivotal in PD-1/PD-L1 checkpoint therapy. JCI Insight. 2018;3(23). doi:10.1172/jci.insight.124507.
    1. Lan Y, Zhang D, Xu C, Hance KW, Marelli B, Qi J, Yu H, Qin G, Sircar A, Hernández VM. Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-L1 and TGF-β. Sci Transl Med. 2018;10(424):eaan5488. doi:10.1126/scitranslmed.aan5488.
    1. Dodagatta-Marri E, Meyer DS, Reeves MQ, Paniagua R, To MD, Binnewies M, Broz ML, Mori H, Wu D, Adoumie M. α-PD-1 therapy elevates Treg/Th balance and increases tumor cell pSmad3 that are both targeted by α-TGFβ antibody to promote durable rejection and immunity in squamous cell carcinomas. J Immunother Cancer. 2019;7(1):62. doi:10.1186/s40425-018-0493-9.
    1. Thomas DA, Massague J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8(5):369–380. doi:10.1016/j.ccr.2005.10.012.
    1. Terabe M, Robertson FC, Clark K, De Ravin E, Bloom A, Venzon DJ, Kato S, Mirza A, Berzofsky JA. Blockade of only TGF-β 1 and 2 is sufficient to enhance the efficacy of vaccine and PD-1 checkpoint blockade immunotherapy. Oncoimmunology. 2017;6(5):e1308616. doi:10.1080/2162402X.2017.1308616.
    1. Aqbi HF, Wallace M, Sappal S, Payne KK, Manjili MH. IFN-gamma orchestrates tumor elimination, tumor dormancy, tumor escape, and progression. J Leukoc Biol. 2018. doi:10.1002/JLB.5MIR0917-351R.
    1. Ni L, Lu J. Interferon gamma in cancer immunotherapy. Cancer Med. 2018;7:4509–4516. doi:10.1002/cam4.1700.
    1. Calzascia T, Pellegrini M, Hall H, Sabbagh L, Ono N, Elford AR, Mak TW, Ohashi PS. TNF-alpha is critical for antitumor but not antiviral T cell immunity in mice. J Clin Invest. 2007;117:3833–3845. doi:10.1172/JCI32567.
    1. Fisher DT, Appenheimer MM, Evans SS. The two faces of IL-6 in the tumor microenvironment. Semin Immunol. 2014;26(1):38–47. doi:10.1016/j.smim.2014.01.008.
    1. Jiao S, Subudhi SK, Aparicio A, Ge Z, Guan B, Miura Y, Sharma P. Differences in tumor microenvironment dictate T helper lineage polarization and response to immune checkpoint therapy. Cell. 2019;179(5):1177–90 e13. doi:10.1016/j.cell.2019.10.029.
    1. Holmgaard RB, Schaer DA, Li Y, Castaneda SP, Murphy MY, Xu X, Inigo I, Dobkin J, Manro JR, Iversen PW. Targeting the TGFbeta pathway with galunisertib, a TGFbetaRI small molecule inhibitor, promotes anti-tumor immunity leading to durable, complete responses, as monotherapy and in combination with checkpoint blockade. J Immunother Cancer. 2018;6:47. doi:10.1186/s40425-018-0356-4.
    1. Anderton MJ, Mellor HR, Bell A, Sadler C, Pass M, Powell S, Steele SJ, Roberts RRA, Heier A. Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol Pathol. 2011;39(6):916–924. doi:10.1177/0192623311416259.
    1. Lacouture ME, Morris JC, Lawrence DP, Tan AR, Olencki TE, Shapiro GI, Dezube BJ, Berzofsky JA, Hsu FJ, Guitart J. Cutaneous keratoacanthomas/squamous cell carcinomas associated with neutralization of transforming growth factor β by the monoclonal antibody fresolimumab (GC1008). Cancer Immunol Immunother. 2015;64(4):437–446. doi:10.1007/s00262-015-1653-0.
    1. Yang Y, Yang HH, Tang B, Wu AML, Flanders KC, Moshkovich N, Weinberg DS, Welsh MA, Weng J, Ochoa HJ. The outcome of TGFβ antagonism in metastatic breast cancer models in vivo reflects a complex balance between tumor-suppressive and proprogression activities of TGFβ. Clin Cancer Res. 2020;26(3):643–656. doi:10.1158/1078-0432.CCR-19-2370.
    1. Strauss J, Heery CR, Schlom J, Madan RA, Cao L, Kang Z, Lamping E, Marté JL, Donahue RN, Grenga I. Phase I trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGFβ, in advanced solid tumors. Clin Cancer Res. 2018;24(6):1287–1295. doi:10.1158/1078-0432.CCR-17-2653.
    1. Tolcher AW, Berlin JD, Cosaert J, Kauh J, Chan E, Piha-Paul SA, Amaya A, Tang S, Driscoll K, Kimbung R. A phase 1 study of anti-TGFβ receptor type-II monoclonal antibody LY3022859 in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2017;79(4):673–680. doi:10.1007/s00280-017-3245-5.

Source: PubMed

3
订阅