293
Views
0
CrossRef citations to date
0
Altmetric
Review

Next-generation immunotherapy for solid tumors: combination immunotherapy with crosstalk blockade of TGFβ and PD-1/PD-L1

, , , , , ORCID Icon & show all
Pages 1187-1202 | Received 30 Jun 2022, Accepted 23 Nov 2022, Published online: 05 Dec 2022

References

  • Massagué J. TGFβ in Cancer. Cell. 2008;134(2):215–230.
  • Akhurst RJ, Hata A. Targeting the TGFß signalling pathway in disease. Nat Rev Drug Discov. 2012;11(10):790–811.
  • Jakowlew SB. Transforming growth factor-ß in cancer and metastasis. Cancer and Metastasis Rev. 2006;25(3):435.
  • Colak S, ten Dijke P. Targeting TGF-ß signaling in cancer. Trends Cancer. 2017;3(1):56–71.
  • Tian M, Schiemann WP. The TGF-beta paradox in human cancer: an update. Future Oncol. 2009;5(2):259–271.
  • Batlle E, Massagué J. Transforming growth factor-ß signaling in immunity and cancer. Immunity. 2019;50(4):924–940.
  • Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19(7):813–824.
  • Nishimura H, Nose M, Hiai H, et al. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11(2):141–151.
  • Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.
  • Latchman Y, Wood CR, Chernova T, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2(3):261–268.
  • Ishida Y, Agata Y, Shibahara K, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11(11):3887–3895.
  • Sheppard KA, Fitz LJ, Lee JM, et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett. 2004 Sep 10;574(1–3):37–41.
  • Patsoukis N, Brown J, Petkova V, et al. Selective effects of PD-1 on akt and ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal. 2012;5(230):ra46–ra46.
  • Chemnitz JM, Parry RV, Nichols KE, et al. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173(2):945–954.
  • Chang C-H, Qiu J, O’Sullivan D, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162(6):1229–1241.
  • Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015;36(4):265–276.
  • Oestreich KJ, Yoon H, Ahmed R, et al. NFATc1 regulates PD-1 expression upon T cell activation. J Immunol. 2008;181(7):4832–4839.
  • Macian F, Lopez-Rodriguez C, Rao A. Partners in transcription: NFAT and AP-1. Oncogene. 2001;20(19):2476–2489.
  • Mathieu M, Cotta-Grand N, Daudelin JF, et al. Notch signaling regulates PD-1 expression during CD8+ T-cell activation. Immunol Cell Biol. 2012;91(1):82–88.
  • Kao C, Oestreich KJ, Paley MA, et al. Transcription factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8+ T cell responses during chronic infection. Nat Immunol. 2011;12(7):663–671.
  • Kallies A, Xin A, Belz GT, et al. Blimp-1 transcription factor is required for the differentiation of effector CD8(+) T cells and memory responses. Immunity. 2009;31(2):283–295.
  • Bally APR, Lu P, Tang Y, et al. NF-?B regulates PD-1 expression in macrophages. J Immunol. 2015;194(9):4545–4554.
  • Shaffer AL, Yu X, He Y, et al. BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control. Immunity. 2000;13(2):199–212.
  • Staron MM, Gray SM, Marshall HD, et al. The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8(+) T cells during chronic infection. Immunity. 2014;41(5):802–814.
  • Horvath CM, Stark GR, Kerr IM, et al. Interactions between STAT and non-STAT proteins in the interferon-stimulated gene factor 3 transcription complex. Mol Cell Biol. 1996;16(12):6957–6964.
  • Terawaki S, Chikuma S, Shibayama S, et al. IFN-alpha directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J Immunol. 2011;186(5):2772–2779.
  • Hirahara K, Ghoreschi K, Yang XP, et al. Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1. Immunity. 2012;36(6):1017–1030.
  • Huynh LK, Hipolito CJ, Ten Dijke P. A perspective on the development of TGF-beta inhibitors for cancer treatment. Biomolecules. 2019;9(11):743.
  • Uckun FM, Qazi S, Hwang L, et al. Recurrent or refractory high-grade gliomas treated by convection-enhanced delivery of a TGFbeta2-targeting RNA therapeutic: a post-hoc analysis with long-term follow-up. Cancers (Basel). 2019;11(12):1892.
  • Hwang L, Ng K, Wang W, et al. OT-101: an anti-TGF-beta-2 antisense- primed tumors to subsequent chemotherapies. J Clin Oncol. 2016;34(15_suppl):e15727–e15727.
  • Smith AL, Robin TP, Ford HL. Molecular pathways: targeting the TGF-ß pathway for cancer therapy. Clin Cancer Res. 2012;18(17):4514–4521.
  • Hau P, Jachimczak P, Bogdahn U. Treatment of malignant gliomas with TGF-beta2 antisense oligonucleotides. Expert Rev Anticancer Ther. 2009;9(11):1663–1674.
  • Connolly EC, Freimuth J, Akhurst RJ. Complexities of TGF-beta targeted cancer therapy. Int J Biol Sci. 2012;8(7):964–978.
  • Rijavec E, Biello F, Genova C, et al. Belagenpumatucel-L for the treatment of non-small cell lung cancer. Expert Opin Biol Ther. 2015;15(9):1371–1379.
  • Fakhrai H, Mantil JC, Liu L, et al. Phase I clinical trial of a TGF-beta antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Ther. 2006;13(12):1052–1060.
  • Nemunaitis J, Dillman RO, Schwarzenberger PO, et al. Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer. J Clin Oncol. 2006;24(29):4721–4730.
  • Nemunaitis J, Nemunaitis M, Senzer N, et al. Phase II trial of belagenpumatucel-L, a TGF-beta2 antisense gene modified allogeneic tumor vaccine in advanced non small cell lung cancer (NSCLC) patients. Cancer Gene Ther. 2009;16(8):620–624.
  • Giaccone G, Bazhenova LA, Nemunaitis J, et al. A phase III study of belagenpumatucel-L, an allogeneic tumour cell vaccine, as maintenance therapy for non-small cell lung cancer. Eur J Cancer. 2015;51(16):2321–2329.
  • Katz LH, Li Y, Chen J-S, et al. Targeting TGF-ß signaling in cancer. Expert Opin Ther Targets. 2013;17(7):743–760.
  • Neuzillet C, Tijeras-Raballand A, Cohen R, et al. Targeting the TGFß pathway for cancer therapy. Pharmacol Ther. 2015;147:22–31.
  • Morris JC, Tan AR, Olencki TE, et al. Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-beta (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PloS one. 2014;9(3):e90353.
  • Formenti SC, Lee P, Adams S, et al. Focal irradiation and systemic TGFß blockade in metastatic breast cancer. Clin Cancer Res. 2018;24(11):2493–2504.
  • Thwaites M, Koropatnick J, Tremblay G, et al. AVID200: a novel TGF-ß inhibitor for the treatment of anemia associated with myelodysplastic syndromes. Blood. 2017;130(Supplement 1):2532–2532.
  • O’Connor-McCourt MD, Tremblay G, Lenferink A, et al. Abstract 1759: AVID200, a highly potent TGF-beta trap, exhibits optimal isoform selectivity for enhancing anti-tumor T-cell activity, without promoting metastasis or cardiotoxicity. Cancer Res. 2018;78(13 Supplement):1759–1759.
  • Zhong Z, Carroll KD, Policarpio D, et al. Anti-transforming growth factor beta receptor II antibody has therapeutic efficacy against primary tumor growth and metastasis through multieffects on cancer, stroma, and immune cells. Clin Cancer Res. 2010;16(4):1191–1205.
  • Tolcher AW, Berlin JD, Cosaert J, et al. A phase 1 study of anti-TGFbeta receptor type-II monoclonal antibody LY3022859 in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2017;79(4):673–680.
  • Foster AE, Dotti G, Lu A, et al. Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-ß receptor. J Immunother. 2008;31(5):500–505.
  • Kloss CC, Lee J, Zhang A, et al. Dominant-negative TGF-beta receptor enhances PSMA-targeted human car t cell proliferation and augments prostate cancer eradication. Mol Ther. 2018;26(7):1855–1866.
  • Vong Q, Nye C, Hause R, et al. Inhibiting TGFß signaling in CAR T-cells may significantly enhance efficacy of tumor immunotherapy. Blood. 2017;130(Supplement 1):1791–1791.
  • Bardhan K, Anagnostou T, Boussiotis VA. The PD1:PD-L1/2 pathway from discovery to clinical implementation. Front Immunol. 2016;7:550.
  • Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8(9):1069–1086.
  • Lin X, Lu X, Luo G, et al. Progress in PD-1/PD-L1 pathway inhibitors: from biomacromolecules to small molecules. Eur J Med Chem. 2020;186:111876.
  • Boyerinas B, Jochems C, Fantini M, et al. Antibody-dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody avelumab (MSB0010718C) on human tumor cells. Cancer Immunol Res. 2015;3(10):1148–1157.
  • Marciscano AE, Gulley JL, Kaufman HL. Avelumab: is it time to get excited? Expert Rev Anticancer Ther. 2018;18(9):815–821.
  • Collins JM, Gulley JL. Product review: avelumab, an anti-PD-L1 antibody. Hum Vaccin Immunother. 2019;15(4):891–908.
  • Zhang F, Wei H, Wang X, et al. Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov. 2017;3(1):17004.
  • J-M X, Qin S, Zhang Y, et al. Phase I study of KN035, the first subcutaneously administered, novel fusion anti-PD-L1 antibody in patients with advanced solid tumors in China. J Clin Oncol. 2019;37(15_suppl):2608–2608.
  • Shimizu T, Nakajima TE, Lu N, et al. Phase I safety and pharmacokinetic study of KN035, the first subcutaneously administered, novel fusion anti-PD-L1 antibody in Japanese patients with advanced solid tumors. J Clin Oncol. 2019;37(15_suppl):2609–2609.
  • Papadopoulos KP, Harb W, Lu N, et al. Phase I study of KN035, a novel fusion Anti-PD-L1 antibody administered subcutaneously in patients with advanced solid tumors in the USA. Ann Oncol. 2018;29(8):viii405.
  • Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 2017;377(20):1919–1929.
  • Ferris RL, Haddad R, Even C, et al. Durvalumab with or without tremelimumab in patients with recurrent or metastatic head and neck squamous cell carcinoma: EAGLE, a randomized, open-label phase III study. Ann Oncol. 2020;31(7):942–950.
  • Nowak AK, Lesterhuis WJ, Kok PS, et al. Durvalumab with first-line chemotherapy in previously untreated malignant pleural mesothelioma (DREAM): a multicentre, single-arm, phase 2 trial with a safety run-in. Lancet Oncol. 2020;21(9):1213–1223.
  • Maestri M, Pallozzi M, Santopaolo F, et al. Durvalumab: an investigational agent for unresectable hepatocellular carcinoma. Expert Opin Investig Drugs. 2022;31(4):347–360.
  • Powles T, van der Heijden MS, Castellano D, et al. Durvalumab alone and durvalumab plus tremelimumab versus chemotherapy in previously untreated patients with unresectable, locally advanced or metastatic urothelial carcinoma (Danube): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol. 2020;21(12):1574–1588.
  • Goldman JW, Dvorkin M, Chen Y, et al. Durvalumab, with or without tremelimumab, plus platinum-etoposide versus platinum-etoposide alone in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): updated results from a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2021;22(1):51–65.
  • Duan J, Cui L, Zhao X, et al. Use of immunotherapy with programmed cell death 1 vs programmed cell death ligand 1 inhibitors in patients with cancer: a systematic review and meta-analysis. JAMA Oncol. 2020;6(3):375–384.
  • Lee A, Keam SJ. Tislelizumab: first approval. Drugs. 2020;80(6):617–624.
  • Zhou CC, Huang DZ, Yu XM, et al. Results from RATIONALE 303: a global phase 3 study of tislelizumab (TIS) vs docetaxel (TAX) as second- or third-line therapy for patients with locally advanced or metastatic NSCLC. Cancer Res. 2021;81(13_Supplement):CT039.
  • Folliguet TA, Malergue MC, Temkine J, et al. Calcified embolus of the left coronary ostia after aortic valve replacement. Ann Thorac Surg. 1997;63(4):1162–1163.
  • Costa R, Carneiro BA, Agulnik M, et al. Toxicity profile of approved anti-PD-1 monoclonal antibodies in solid tumors: a systematic review and meta-analysis of randomized clinical trials. Oncotarget. 2017;8(5):8910–8920.
  • Johnson DB, Nebhan CA, Moslehi JJ, et al. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol. 2022;19(4):254–267.
  • Ingles Garces AH, Au L, Mason R, et al. Building on the anti-PD1/PD-L1 backbone: combination immunotherapy for cancer. Expert Opin Investig Drugs. 2019;28(8):695–708.
  • Chang A, Schlafer D, Flowers CR, et al. Investigational PD-1 inhibitors in HL and NHL and biomarkers for predictors of response and outcome. Expert Opin Investig Drugs. 2018;27(1):55–70.
  • Ghanem S, Valecha GK, Hossri S, et al. Investigational PD-1 inhibitors for advanced non-small lung cancer: new players in a shifting paradigm. Expert Opin Investig Drugs. 2017;26(12):1317–1319.
  • Vick E, Mahadevan D. Programming the immune checkpoint to treat hematologic malignancies. Expert Opin Investig Drugs. 2016;25(7):755–770.
  • Gunturi A, McDermott DF. Nivolumab for the treatment of cancer. Expert Opin Investig Drugs. 2015;24(2):253–260.
  • Cherkassky L, Hou Z, Amador-Molina A, et al. Regional CAR T cell therapy: an ignition key for systemic immunity in solid tumors. Cancer Cell. 2022;40(6):569–574.
  • Grosser R, Cherkassky L, Chintala N, et al. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell. 2019;36(5):471–482.
  • Kiesgen S, Chicaybam L, Chintala NK, et al. Chimeric antigen receptor (CAR) T-cell therapy for thoracic malignancies. J Thorac Oncol. 2018;13(1):16–26.
  • Morello A, Sadelain M, Adusumilli PS. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 2016;6(2):133–146.
  • Tang N, Cheng C, Zhang X, et al. TGF-beta inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight. 2020;5(4):e133977.
  • Narayan V, Gladney W, Plesa G, et al. A phase I clinical trial of PSMA-directed/TGFß-insensitive CAR-T cells in metastatic castration-resistant prostate cancer. J Clin Oncol. 2019;37(7_suppl): TPS347–TPS347.
  • Cherkassky L, Morello A, Villena-Vargas J, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016;126(8):3130–3144.
  • Adusumilli PS, Zauderer MG, Riviere I, et al. A phase I trial of regional mesothelin-targeted CAR t-cell therapy in patients with malignant pleural disease, in combination with the anti-PD-1 agent pembrolizumab. Cancer Discov. 2021;11(11):2748–2763.
  • Fang J, Ding N, Guo X, et al. alphaPD-1-mesoCAR-T cells partially inhibit the growth of advanced/refractory ovarian cancer in a patient along with daily apatinib. J Immunother Cancer. 2021;9(2):e001162.
  • Wang Z, Li N, Feng K, et al. Phase I study of CAR-T cells with PD-1 and TCR disruption in mesothelin-positive solid tumors. Cell Mol Immunol. 2021;18(9):2188–2198.
  • Odorizzi PM, Pauken KE, Paley MA, et al. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J Exp Med. 2015;212(7):1125–1137.
  • Priceman SJ, Cheema W, Adusumilli PS. Advancing together and moving forward: combination gene and cellular immunotherapies. Mol Ther Oncolytics. 2022;25:330–334.
  • Rekik R, Belhadj Hmida N, Ben Hmid A, et al. PD-1 induction through TCR activation is partially regulated by endogenous TGF-beta. Cell Mol Immunol. 2015;12(5):648–649.• This publication proposed the role of TGFß in increasing PD-1 level through TCR activation
  • Baas M, Besancon A, Goncalves T, et al. TGFbeta-dependent expression of PD-1 and PD-L1 controls CD8(+) T cell anergy in transplant tolerance. Elife. 2016;5:e08133.
  • Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009;19(1):128–139.
  • Dhillon AS, Hagan S, Rath O, et al. MAP kinase signalling pathways in cancer. Oncogene. 2007;26(22):3279–3290.
  • Sorrentino A, Thakur N, Grimsby S, et al. The type I TGF-beta receptor engages TRAF6 to activate TAK1 in a receptor kinase-independent manner. Nat Cell Biol. 2008;10(10):1199–1207.
  • Yamashita M, Fatyol K, Jin C, et al. TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-beta. Mol Cell. 2008;31(6):918–924.
  • Stuber T, Monjezi R, Wallstabe L, et al. Inhibition of TGF-beta-receptor signaling augments the antitumor function of ROR1-specific CAR T-cells against triple-negative breast cancer. J Immunother Cancer. 2020;8(1):e000676.
  • 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(1):35–44.
  • Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol. 2018;18(3):153–167.
  • Vanpouille-Box C, Formenti SC. Dual transforming growth factor-beta and programmed death-1 blockade: a strategy for immune-excluded tumors? Trends Immunol. 2018;39(6):435–437.
  • Mariathasan S, Turley SJ, Nickles D, et al. TGFß attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554(7693):544–548. • This publication highlighted that the lack of response in patients who were treated with checkpoint therapy was associated with TGFß genes in tumor fibroblast
  • Tauriello DVF, Palomo-Ponce S, Stork D, et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. 2018;554(7693):538–543.
  • Chen X, Wang L, Li P, et al. Dual TGF-ß and PD-1 blockade synergistically enhances MAGE-A3-specific CD8+ T cell response in esophageal squamous cell carcinoma. Int J Cancer. 2018;143(10):2561–2574.
  • Feun LG, Y-Y L, Wu C, et al. Phase 2 study of pembrolizumab and circulating biomarkers to predict anticancer response in advanced, unresectable hepatocellular carcinoma. Cancer. 2019;125(20):3603–3614.
  • Lind H, Gameiro SR, Jochems C, et al. Dual targeting of TGF-ß and PD-L1 via a bifunctional anti-PD-L1/TGF-ßRII agent: status of preclinical and clinical advances. J Immunother Cancer. 2020;8(1):e000433.
  • Strauss J, Heery CR, Schlom J, et al. Phase I trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGFbeta, in advanced solid tumors. Clin Cancer Res. 2018;24(6):1287–1295.•• This publication was one of the first clinical trials that discussed the safety of bifunctional fusion protein M7824 in patients with advanced solid tumor
  • Knudson KM, Hicks KC, Luo X, et al. M7824, a novel bifunctional anti-PD-L1/TGFbeta Trap fusion protein, promotes anti-tumor efficacy as monotherapy and in combination with vaccine. Oncoimmunol. 2018;7(5):e1426519.
  • Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29(34):4741–4751.
  • David JM, Dominguez C, McCampbell KK, et al. A novel bifunctional anti-PD-L1/TGF-beta Trap fusion protein (M7824) efficiently reverts mesenchymalization of human lung cancer cells. Oncoimmunol. 2017;6(10):e1349589.
  • Jochems C, Tritsch SR, Wang P, et al. Analyses of functions of an anti-PD-L1/TGFßR2 bispecific fusion protein (M7824). Oncotarget. 2017;8(43):75217–75231.
  • Lan Y, Zhang D, Xu C, et al. Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-L1 and TGF-beta. Sci Transl Med. 2018;10(424):eaan5488.
  • Grenga I, Donahue RN, Gargulak ML, et al. Anti-PD-L1/TGFbetaR2 (M7824) fusion protein induces immunogenic modulation of human urothelial carcinoma cell lines, rendering them more susceptible to immune-mediated recognition and lysis. Urol Oncol. 2018;36(3):93.e1–93.e11.
  • Gulley JL, Heery CR, Schlom J, et al. Preliminary results from a phase 1 trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGF-ß, in advanced solid tumors. J Clin Oncol. 2017;35(15_suppl):3006–3006.
  • Paz-Ares LG, Kim TM, Baz DV, et al. Results from a second-line (2L) NSCLC cohort treated with M7824 (MSB0011359C), a bifunctional fusion protein targeting TGF-ß and PD-L1. J Clin Oncol. 2018;36(15_suppl):9017–9017.
  • Lin CC, Doi T, Muro K, et al. Bintrafusp alfa, a bifunctional fusion protein targeting TGFbeta and PD-l1, in patients with esophageal squamous cell carcinoma: results from a phase 1 cohort in Asia. Target Oncol. 2021;16(4):447–459.
  • Redman JM, Steinberg SM, Gulley JL. Quick efficacy seeking trial (QuEST1): a novel combination immunotherapy study designed for rapid clinical signal assessment metastatic castration-resistant prostate cancer. J Immunother Cancer. 2018;6(1):91.
  • Cheng B, Ding K, Chen P, et al. Anti-PD-L1/TGF-betaR fusion protein (SHR-1701) overcomes disrupted lymphocyte recovery-induced resistance to PD-1/PD-L1 inhibitors in lung cancer. Cancer Commun (Lond). 2022;42(1):17–36.
  • Liu D, Gong JF, Liu TS, et al. Phase 1 study of SHR-1701, a bifunctional fusion protein targeting PD-L1 and TGF-beta, in patients with advanced solid tumors. J Clin Oncol. 2021;39(15_suppl):2503–2503.
  • Shi MQ, Chen JH, Li KY, et al. SHR-1701, a bifunctional fusion protein targeting PD-L1 and TGF-beta, for advanced NSCLC with EGFR mutations: data from a multicenter phase 1 study. J Clin Oncol. 2021;39(15_suppl):9055–9055.
  • Feng J, Wang J, Tang D, et al. SHR-1701, a bifunctional fusion protein targeting PD-L1 and TGF-beta, for pretreated advanced cervical cancer: data from a clinical expansion cohort of a phase I study. Ann Oncol. 2021;32:S753–S753.
  • Huang X, Sun JM, Qin YR, et al. LBL-015, a novel anti-PD-1 fused with TGF-beta RII, shows a great anti-tumor activity in a mouse MC38 model. Cancer Res. 2021;81(13_Supplement):1791.
  • Li HJ, Wang CM, Guo HH, et al. The preclinical characterization of TST005, a bi-functional anti-PD-L1 and TGF-beta trap fusion protein. Cancer Res. 2021;81(13_Supplement):917.
  • Gao ZZ, Li C, Chen G, et al. Optimization strategies for expression of a novel bifunctional anti-PD-L1/TGFBR2-ECD fusion protein. Protein Expr Purif. 2022;189:105973.
  • Ravi R, Noonan KA, Pham V, et al. Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFbeta enhance the efficacy of cancer immunotherapy. Nat Commun. 2018;9(1):741.
  • Terabe M, Robertson FC, Clark K, et al. Blockade of only TGF-beta 1 and 2 is sufficient to enhance the efficacy of vaccine and PD-1 checkpoint blockade immunotherapy. Oncoimmunol. 2017;6(5):e1308616.
  • Vanpouille-Box C, Diamond JM, Pilones KA, et al. TGFbeta is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res. 2015;75(11):2232–2242.
  • Lim YW, Coles GL, Sandhu SK, et al. Single-cell transcriptomics reveals the effect of PD-L1/TGF-beta blockade on the tumor microenvironment. BMC Biol. 2021;19(1):107.
  • Grell P, Lin CC, Milella M, et al. Phase II study of the anti-TGF-beta monoclonal antibody (mAb) NIS793 with and without the PD-1 inhibitor spartalizumab in combination with nabpaclitaxel/gemcitabine (NG) versus NG alone in patients (pts) with first-line metastatic pancreatic ductal adenocarcinoma (mPDAC). J Clin Oncol. 2021;39(15_suppl): TPS4173–TPS4173.
  • Robbrecht D, Jean-Jacques G, Bechter O, et al. 520 preliminary biomarker and pharmacodynamic (PD) activity of the TGFß inhibitor SAR439459, alone or in combination with cemiplimab, in a phase 1 clinical study in patients with advanced solid tumors. J Immunother Cancer. 2021;9:A550–A550.
  • Bertrand C, Van Meerbeeck P, de Streel G, et al. Combined blockade of GARP:TGF-beta1 and PD-1 increases infiltration of T cells and density of pericyte-covered GARP(+) blood vessels in mouse MC38 tumors. Front Immunol. 2021;12:704050.
  • Principe DR, Park A, Dorman MJ, et al. TGFbeta blockade augments PD-1 inhibition to promote T-cell-mediated regression of pancreatic cancer. Mol Cancer Ther. 2019;18(3):613–620.
  • Zhao F, Evans K, Xiao C, et al. Stromal fibroblasts mediate anti-PD-1 resistance via MMP-9 and dictate TGFbeta inhibitor sequencing in melanoma. Cancer Immunol Res. 2018;6(12):1459–1471.
  • Sow HS, Ren J, Camps M, et al. Combined inhibition of TGF-beta signaling and the PD-l1 immune checkpoint is differentially effective in tumor models. Cells. 2019;8(4):320.
  • Panagi M, Voutouri C, Mpekris F, et al. TGF-beta inhibition combined with cytotoxic nanomedicine normalizes triple negative breast cancer microenvironment towards anti-tumor immunity. Theranostics. 2020;10(4):1910–1922.
  • Melisi D, Hollebecque A, Oh DY, et al. A phase Ib dose-escalation and cohort-expansion study of safety and activity of the transforming growth factor (TGF) beta receptor I kinase inhibitor galunisertib plus the anti-PD-L1 antibody durvalumab in metastatic pancreatic cancer. J Clin Oncol. 2019;37(15_suppl):4124–4124.
  • Yang Y, Xu W, Peng D, et al. An oncolytic adenovirus targeting transforming growth factor beta inhibits protumorigenic signals and produces immune activation: a novel approach to enhance anti-PD-1 and anti-CTLA-4 therapy. Hum Gene Ther. 2019;30(9):1117–1132.
  • Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2019;7(1):306.
  • Weinmann SC, Pisetsky DS. Mechanisms of immune-related adverse events during the treatment of cancer with immune checkpoint inhibitors. Rheumatology (Oxford). 2019;58(Suppl 7):vii59–vii67.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.