Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Aug 12;16(1):94.
doi: 10.1186/s13045-023-01487-5.

Synergistic efficacy of simultaneous anti-TGF-β/VEGF bispecific antibody and PD-1 blockade in cancer therapy

Affiliations

Synergistic efficacy of simultaneous anti-TGF-β/VEGF bispecific antibody and PD-1 blockade in cancer therapy

Mengke Niu et al. J Hematol Oncol. .

Abstract

Background: Recently, therapeutic antibodies against programmed cell death 1 (PD-1) and its ligand (PD-L1) have exerted potent anticancer effect in a variety of tumors. However, blocking the PD-1/PD-L1 axis alone is not sufficient to restore normal immune response. Other negative regulators of antitumor immunity, like TGF-β and VEGFA, are also involved in immune escape of tumor cells and induce immunotherapy resistance.

Methods: We developed a novel anti-TGF-β/VEGF bispecific antibody Y332D based on the Nano-YBODY™ technology platform. The CCK-8, flow cytometry, SBE4 luciferase reporter assay, western blotting and transwell assays were used to measure the biological activities of the anti-TGF-β moiety. The NFAT luciferase reporter assay, luminescent cell viability assay and tube formation assay were used to measure the biological activities of the anti-VEGF moiety. The in vivo anticancer efficacy of Y332D alone or in combination with PD-1 blockade was evaluated in H22, EMT-6, 4T1, and AKT/Ras-driven murine hepatocellular carcinoma tumor models. Immunofluorescent staining, flow cytometry, RNA-seq and quantitative RT-PCR were adopted to analyze the alterations in the tumor microenvironment.

Results: Y332D could maintain specific binding affinities for TGF-β and VEGFA. Y332D almost entirely counteracted the in vitro biological functions of TGF-β and VEGFA, including immunosuppression, activated TGF-β signaling, epithelial-mesenchymal transition (EMT), activated VEGF/VEGFR signaling, HUVEC proliferation and tube formation. The in vivo experiment data demonstrated that Y332D was more effective in inhibiting tumor growth and metastasis than anti-TGF-β and anti-VEGF monotherapies. In combination therapies, Y332D plus PD-1 blockade exhibited the most potent and durable anticancer effect. Mechanistically, Y332D plus PD-1 blockade upregulated the density and function of tumor-infiltrating lymphocytes and exerted reinvigorated antitumor immunity.

Conclusion: Y332D could simultaneously block TGF-β and VEGF signalings. In comparison with the monotherapies, Y332D combined with PD-1 blockade exerts superior antitumor effect through improving immune microenvironment.

Keywords: Bispecific antibody; Cancer immunotherapy; PD-1; TGF-β; The tumor microenvironment; VEGF.

PubMed Disclaimer

Conflict of interest statement

QH, RY, LZ, JS, JZ and PZ were employees of Wuhan YZY Biopharma Co., Ltd.

Figures

Fig. 1
Fig. 1
The structure characteristics of Y332D and the binding affinity of Y332D to TGF-β and VEGF. a Schematic representation of Y332D. Y332D is designed as a tetravalent and symmetric bispecific antibody that contains two anti-VEGF regions and two anti-TGF-β regions. The long chain of Y332D consists of five domains: VHb, CH1, CH2, CH3, VHH. The short chain of Y332D contains two domains: VLb, CL. The Fc region of Y332D is an engineered hybrid fragment: the CH2 domain is from IgG2, and the CH3 domain is from IgG1. b The non-reduced and reduced SDS-PAGE analysis of Y332D. Under nonreducing conditions, a single band was displayed. Two bands were observed under reducing conditions, representing the heavy and light chains. c, d The non-reduced and reduced CE-SDS analysis of Y332D. One peak was observed in the non-reduced CE-SDS, and two peaks were detected in the reduced CE-SDS. The purity of Y332D was more than 97%. e The results of SPR assay to detect the binding kinetics of Y332D to TGF-β1. f The results of SPR assay to detect the binding kinetics of Y332D to VEGFA. g The ELISA binding affinity of serially diluted Y332D or controls to plate-coated TGF-β1. h The ELISA binding affinity of serially diluted Y332D or controls to plate-coated VEGFA. i The simultaneous binding activity of Y332D to TGF-β1 and VEGFA by ELISA. Serially diluted Y332D or controls were incubated with plate-coated TGF-β1. Then, VEGFA-Biotin and peroxidase-conjugated streptavidin were added sequentially. Bars, SDs
Fig. 2
Fig. 2
Y332D counteracted TGF-β1-induced inhibition of T cell proliferation and activation as well as epithelial-mesenchymal transition (EMT). a, b CCK-8 assays were performed to show the antagonistic effect of Y332D on TGF-β1-hampered proliferation in T cells. 1 × 103 CTLL-2 and HT-2 cells were seeded in 96-well plates. Then, 5 ng/ml TGF-β1 plus 106 pM antibodies or control were added. Cell viability was continuously monitored by CCK-8 reagent. cg Multi-cytokine assay was performed to analyze the effect of Y332D on the alteration of TGF-β1-caused cytokine secretion during T cell activation. Murine T cells were obtained from the isolation of splenocytes from C57BL/6 mice. T cells (1 × 106/ml) supplemented with anti-CD28 (3 μg/ml), TGF-β1 (5 ng/ml) and 106 pM antibodies or control were cultured in 96 well flat-bottom plates precoated with anti-CD3 (3 μg/ml). After 4 days, the cellular supernatants were harvested to measure cytokines concentration. h, i Transwell migration/invasion assays were performed to demonstrate the antagonistic effect of Y332D on TGF-β-enhanced tumor cell motility. 4T1 and EMT-6 mammary tumor cells were cultured in RPIM-1640 with 1% FBS and treated with 5 ng/ml TGF-β1 plus 106 pM antibodies or untreated for 96 h. Then, about 5 × 104 cells in 100 µl RPIM-1640 supplemented with 1%FBS were seeded in the upper chambers. The lower chambers were added with 600 µl of RPIM-1640 containing 10% FBS. After incubation for 24 h, the migratory and invasive cells were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet. Bars, SDs; **p < 0.01, ***p < 0.001, and ****p < 0.0001 denote the significant difference relative to Y332D treatment. α-TGF-β: anti-TGF-β, α-VEGF: anti-VEGF
Fig. 3
Fig. 3
The antagonistic effect of Y332D on the activation of VEGF/VEGFR pathway, VEGFA-induced proliferation and tube formation in HUVECs. a NFAT-luciferase reporter assay was performed to show the blockade effect of Y332D on VEGF/VEGFR pathway. HEK-293 cells overexpressing VEGFR2 were transfected with the lentiviral vectors carrying the NFAT and luciferase gene (NFAT-RE-Luci) to construct stable transfected cell lines 293-NFAT. 293-NFAT cells were cultured in 2% FBS-DMEM with VEGFA (20 ng/ml) and serially diluted Y332D or controls for 6 h. Then, the luminescence was detected. b Luminescent cell viability assay was performed to measure the inhibitory effect of Y332D on VEGFA-induced HUVEC proliferation. 5 × 103 HUVECs were seeded in 96-well plates overnight at 37 °C. Then, the medium was replaced with endothelial cell basal medium mixed with VEGFA (50 ng/ml) and serially diluted Y332D or controls. Cell viability was detected after incubation at 37 °C for 72 h. c Tube formation assay was performed to show the inhibitory effect of Y332D on VEGFA-induced vessel-like tube formation. 2 × 104 HUVECs were seeded in 96 well flat-bottom plates after plates were precoated with 50 μl Matrigel for 30 min at 37 °C. The cells were incubated in endothelial cell complete medium mixing with 100 ng/ml VEGFA and 106 pM antibodies or control for 12 h at 37 °C. Then, HUVECs were fixed with 4% paraformaldehyde for 15 min. The images of tube-like structures were captured with inverted microscope. Bars, SDs; α-TGF-β: anti-TGF-β, α-VEGF: anti-VEGF
Fig. 4
Fig. 4
Y332D inhibited tumor growth in murine tumor models, and the combination of Y332D and PD-1 blockade demonstrated synergistic antitumor effects. Tumor volume (TV) of tumor-bearing mice was measured every other day or every two days. Mice were euthanatized when TV exceeded 2500 mm3 or the study ended. a Model establishment and treatment schedule of H22 and EMT-6 tumor models. 8.7 mg/kg α-PD-1 were administrated every two days by intraperitoneal injection for four times. Equivalent mole hIgG (8.7 mg/kg), α-VEGF (8.7 mg/kg), α-TGF-β (6 mg/kg), Y332D (10 mg/kg) were administrated on alternate days by intraperitoneal injection for six times. bd 5 × 105 H22 cells were inoculated subcutaneously in the right groin of BALB/c mice on day 0. Start of treatment on day 6. The representative tumors image, tumor growth curve, tumor weight of H22-bearing mice receiving α-PD-1 plus Y332D or controls treatment were shown. e The overall survival curves of H22-bearing mice receiving α-PD-1 plus Y332D or controls treatment were shown. fh 5 × 105 EMT-6 cells were inoculated in the right mammary fat pad of BALB/c mice on day 0. Start of treatment on day 10. The representative tumors image, tumor growth curve and tumor weight of EMT-6-bearing mice receiving α-PD-1 plus Y332D or controls treatment were shown. i, j The representative image and tumor growth curve of EMT-6 tumors in the rechallenge assay were shown. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001 denote the significant difference relative to Y332D or Y332D plus anti-PD-1 treatment. CR: complete regression. Bars, SDs; α-PD-1: anti-PD-1, α-TGF-β: anti-TGF-β, α-VEGF: anti-VEGF
Fig. 5
Fig. 5
The combination of Y332D and PD-1 blockade demonstrated synergistic antitumor efficacy in AKT/Ras-driven murine hepatocellular carcinoma tumor model. a Model establishment and treatment schedule of AKT/Ras-driven murine hepatocellular carcinoma tumor model. The plasmid encoding myr-AKT1 and/or NRasV12 along with sleeping beauty transposase were injected into the lateral tail vein of C57BL/6 mice by hydrodynamic injection on day 0. Treatment was started on day 26 (5 mice for each group). All tumor-bearing mice were randomly divided into six groups (Vehicle, Y332D, α-PD-1, α-PD-1 plus α-VEGF, α-PD-1 plus α-TGF-β, α-PD-1 plus Y332D). 8.7 mg/kg α-PD-1 were administrated every two days by intraperitoneal injection for four times. Equivalent mole hIgG (8.7 mg/kg), α-VEGF (8.7 mg/kg), α-TGF-β (6 mg/kg), Y332D (10 mg/kg) were administrated on alternate days by intraperitoneal injection for six times. 40 days after injection, the mice were euthanized, and the liver tissues were collected. b, c The representative liver tumor images and liver weight of AKT/Ras-driven murine hepatocellular carcinoma mice receiving α-PD-1 plus Y332D or controls treatment were shown. d The overall survival curves of AKT/Ras-driven murine hepatocellular carcinoma mice receiving α-PD-1 plus Y332D or controls treatment were shown. e The representative H&E staining images of liver tissues of mice receiving the treatment of combination therapies or controls. The size of the scale bar in the immunofluorescence images refer to 5 mm or 250 μm. *p < 0.05, **p < 0.01, and ****p < 0.0001 denote the significant difference relative to Y332D plus anti-PD-1 treatment. Bars, SDs. α-PD-1: anti-PD-1, α-TGF-β: anti-TGF-β, α-VEGF: anti-VEGF
Fig. 6
Fig. 6
Immunofluorescence staining to measure the status of epithelial-mesenchymal transition (EMT) of cancer cells, carcinoma-associated fibroblast (CAF) and tumor angiogenesis in H22 tumor model. The representative images and quantitative analysis of ae EMT-related markers, including anti-E-cadherin staining, anti-Vimentin staining, anti-N-cadherin staining, f CAF marker, anti-α-SMA staining, g Anti-CD31 staining. The size of the scale bar in the immunofluorescence images refer to 100 μm. Bars, SDs; ***p < 0.001, and ****p < 0.0001 denote the significant difference relative to Y332D treatment. ns: not significant, α-TGF-β: anti-TGF-β, α-VEGF: anti-VEGF
Fig. 7
Fig. 7
Immunofluorescence staining and flow cytometry assay to analyze tumor-infiltrating lymphocytes in H22 tumor model. Mice were euthanatized when the study ended. The harvested tumor tissues were subjected to immunofluorescence and flow cytometry. a The representative immunofluorescence images and quantitative analysis of tumor-infiltrating CD8+ T cells. Harvested tumor tissues were dissociated with Collagenase B and hyaluronidase to prepare single-cell suspensions. Then, the cells were fluorescently stained with the detection antibodies. The representative images and quantitative analysis of tumor-infiltrating b, h CD8+ T cells, c, i Ki67+CD8+ T cells, d, j CD25+CD8+ T cells, e, k CD69+CD8+ T cells, f, l Granzyme B+CD8+ T cells, g, m IFN-γ+CD8+ T cells. The proportion of tumor-infiltrating lymphocytes in the total live cells was calculated. The size of the scale bar in the immunofluorescence images refer to 100 μm. Bars, SDs; *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001 denote the significant difference relative to combination treatment. ns: not significant, α-PD-1: anti-PD-1
Fig. 8
Fig. 8
RNA-seq assay to explore the immune profile of H22 tumors after different treatments. a Heat map to represent the differentially expressed genes among four groups. b Heat map to represent the expression levels of cytotoxicity-related genes (Gzma, Gzmb, Prf1, Ifng, Tnf, etc.). c Heat map to represent the expression levels of chemokines (Ccl11, Cxcl9, Cxcl11, Cxcl12, Cxcl16). d, e Heat map to represent the expression levels of signature genes in T cells and NK cells, and signature scores were calculated to quantify. f–h The top 10 significantly enriched immune-related Gene Ontology (GO) terms (α-PD-1 + Y332D vs. Vehicle; α-PD-1 + Y332D vs. α-PD-1; α-PD-1 + Y332D vs. Y332D). i, j Quantitative RT-PCR validation of selected genes identified by RNA-seq. Bars, SDs; *p < 0.05 and **p < 0.01 denote the significant difference relative to combination treatment. α-PD-1: anti-PD-1
Fig. 9
Fig. 9
Schematic diagram demonstrating the synergistic antitumor immune effect of Y332D plus PD-1 blockade. a Y332D restored the cytotoxic effects of TGF-β-suppressed CD8+ T cells and inhibited TGF-β-mediated collagen production in cancer-associated fibroblasts (CAF) cells. b Y332D promoted T cells infiltration through vascular normalization. c Y332D in combination with PD-1 blockade synergistically enhanced antitumor immunity

References

    1. Boussiotis VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med. 2016;375:1767–1778. doi: 10.1056/NEJMra1514296. - DOI - PMC - PubMed
    1. Liu Z, Yu X, Xu L, Li Y, Zeng C. Current insight into the regulation of PD-L1 in cancer. Exp Hematol Oncol. 2022;11:44. doi: 10.1186/s40164-022-00297-8. - DOI - PMC - PubMed
    1. Yang X, Ma L, Zhang X, Huang L, Wei J. Targeting PD-1/PD-L1 pathway in myelodysplastic syndromes and acute myeloid leukemia. Exp Hematol Oncol. 2022;11:11. doi: 10.1186/s40164-022-00263-4. - DOI - PMC - PubMed
    1. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–1034. doi: 10.1084/jem.192.7.1027. - DOI - PMC - PubMed
    1. Niu M, Liu Y, Yi M, Jiao D, Wu K. Biological characteristics and clinical significance of soluble PD-1/PD-L1 and exosomal PD-L1 in cancer. Front Immunol. 2022;13:827921. doi: 10.3389/fimmu.2022.827921. - DOI - PMC - PubMed

Publication types

Substances