Targeting intracellular cancer proteins with tumor-microenvironment-responsive bispecific nanobody-PROTACs for enhanced therapeutic efficacy

Changping Deng , Jiacheng Ma , Yuping Liu , Xikui Tong , Lei Wang , Jiayi Dong , Ping Shi , Meiyan Wang , Wenyun Zheng , Xingyuan Ma

MedComm ›› 2025, Vol. 6 ›› Issue (2) : e70068

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (2) : e70068 DOI: 10.1002/mco2.70068
ORIGINAL ARTICLE

Targeting intracellular cancer proteins with tumor-microenvironment-responsive bispecific nanobody-PROTACs for enhanced therapeutic efficacy

Author information +
History +
PDF

Abstract

Proteolysis targeting chimeras (PROTACs) are pivotal in cancer therapy for their ability to degrade specific proteins. However, their non-specificity can lead to systemic toxicity due to protein degradation in normal cells. To address this, we have integrated a nanobody into the PROTACs framework and leveraged the tumor microenvironment to enhance drug specificity. In this study, we engineered BumPeD, a novel bispecific nanobody-targeted PROTACs-like platform, by fusing two nanobodies with a Furin protease cleavage site (RVRR) and a degron sequence (ALAPYIP or KIGLGRQKPPKATK), enabling the tumor microenvironment to direct the degradation of intracellular proteins. We utilized KN035 and Nb4A to target PD-L1 (programmed death ligand 1) on the cell surface and intracellular Survivin, respectively. In vitro experiments showed that BumPeD triggers Survivin degradation via the ubiquitin-proteasome pathway, inducing tumor apoptosis and suppressing bladder tumor cell proliferation and migration. In vivo experiments further confirmed BumPeD’s robust anti-tumor efficacy, underscoring its potential as a precise protein degradation strategy for cancer therapy. Our platform provides a systematic approach to developing effective and practical protein degraders, offering a targeted theoretical basis and experimental support for the development of novel degradative drugs, as well as new directions for cancer therapy.

Keywords

nanobody / PD-L1 and Survivin / proteolysis targeting chimeras (PROTACs) / targeted degradation / tumor microenvironment

Cite this article

Download citation ▾
Changping Deng, Jiacheng Ma, Yuping Liu, Xikui Tong, Lei Wang, Jiayi Dong, Ping Shi, Meiyan Wang, Wenyun Zheng, Xingyuan Ma. Targeting intracellular cancer proteins with tumor-microenvironment-responsive bispecific nanobody-PROTACs for enhanced therapeutic efficacy. MedComm, 2025, 6(2): e70068 DOI:10.1002/mco2.70068

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Li X, Pu W, Zheng Q, Ai M, Chen S, Peng Y. Proteolysis-targeting chimeras (PROTACs) in cancer therapy. Mol Cancer. 2022; 21(1): 99.

[2]

He S, Dong G, Cheng J, Wu Y, Sheng C. Strategies for designing proteolysis targeting chimaeras (PROTACs). Med Res Rev. 2022; 42(3): 1280-1342.

[3]

Martin-Acosta P, Xiao X. PROTACs to address the challenges facing small molecule inhibitors. Eur J Med Chem. 2021; 210: 112993.

[4]

Wang Y, Jiang X, Feng F, Liu W, Sun H. Degradation of proteins by PROTACs and other strategies. Acta Pharm Sin B. 2020; 10(2): 207-238.

[5]

Han X, Sun Y. PROTACs: a novel strategy for cancer drug discovery and development. MedComm. 2023; 4(3): e290.

[6]

Bekes M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022; 21(3): 181-200.

[7]

Raina K, Lu J, Qian Y, et al. PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci U S A. 2016; 113(26): 7124-7129.

[8]

Yang C, Yang Y, Li Y, Ni Q, Li J. Radiotherapy-triggered proteolysis targeting chimera prodrug activation in tumors. J Am Chem Soc. 2023; 145(1): 385-391.

[9]

Nalawansha DA, Crews CM. PROTACs: an emerging therapeutic modality in precision medicine. Cell Chem Biol. 2020; 27(8): 998-1014.

[10]

Paiva SL, Crews CM. Targeted protein degradation: elements of PROTAC design. Curr Opin Chem Biol. 2019; 50: 111-119.

[11]

Verma S, Manna D. Controlling PROTACs with light. ChemMedChem. 2020; 15(14): 1258-1261.

[12]

Zinn S, Vazquez-Lombardi R, Zimmermann C, Sapra P, Jermutus L, Christ D. Advances in antibody-based therapy in oncology. Nature Cancer. 2023; 4(2): 165-180.

[13]

Marei H, Tsai WK, Kee YS, et al. Antibody targeting of E3 ubiquitin ligases for receptor degradation. Nature. 2022; 610(7930): 182-189.

[14]

Wang T, Zhao J, Ren JL, et al. Recombinant immunoproapoptotic proteins with furin site can translocate and kill HER2-positive cancer cells. Cancer Res. 2007; 67(24): 11830-11839.

[15]

Cao Y, Marks JD, Marks JW, Cheung LH, Kim S, Rosenblum MG. Construction and characterization of novel, recombinant immunotoxins targeting the Her2/neu oncogene product: in vitro and in vivo studies. Cancer Res. 2009; 69(23): 8987-8995.

[16]

Wang F, Ren J, Qiu XC, et al. Selective cytotoxicity to HER2-positive tumor cells by a recombinant e23sFv-TD-tBID protein containing a furin cleavage sequence. Clin Cancer Res. 2010; 16(8): 2284-2294.

[17]

Muyldermans S. Applications of nanobodies. Annu Rev Anim Biosci. 2021; 9: 401-421.

[18]

Muyldermans S. Nanobodies: natural single-domain antibodies. Annu Rev Biochem. 2013; 82: 775-797.

[19]

Jovcevska I, Muyldermans S. The therapeutic potential of nanobodies. BioDrugs. 2020; 34(1): 11-26.

[20]

Salvador JP, Vilaplana L, Marco MP. Nanobody: outstanding features for diagnostic and therapeutic applications. Anal Bioanal Chem. 2019; 411(9): 1703-1713.

[21]

Shen F, Zheng G, Setegne M, et al. A cell-permeant nanobody-based degrader that induces fetal hemoglobin. ACS Cent Sci. 2022; 8(12): 1695-1703.

[22]

Roth S, Fulcher LJ, Sapkota GP. Advances in targeted degradation of endogenous proteins. Cell Mol Life Sci. 2019; 76(14): 2761-2777.

[23]

Simpson LM, Macartney TJ, Nardin A, et al. Inducible degradation of target proteins through a tractable affinity-directed protein missile system. Cell Chem Biol. 2020; 27(9): 1164-1180.e5.

[24]

Ibrahim AFM, Shen L, Tatham MH, et al. Antibody RING-mediated destruction of endogenous proteins. Mol Cell. 2020; 79(1): 155-166.e9.

[25]

Fulcher LJ, Macartney T, Bozatzi P, Hornberger A, Rojas-Fernandez A, Sapkota GP. An affinity-directed protein missile system for targeted proteolysis. Open Biol. 2016; 6(10): 160255.

[26]

Zhang H, Han Y, Yang Y, et al. Covalently engineered nanobody chimeras for targeted membrane protein degradation. J Am Chem Soc. 2021; 143(40): 16377-16382.

[27]

Schneekloth JS Jr, Fonseca FN, Koldobskiy M, et al. Chemical genetic control of protein levels: selective in vivo targeted degradation. J Am Chem Soc. 2004; 126(12): 3748-3754.

[28]

Si L, Shen Q, Li J, et al. Generation of a live attenuated influenza A vaccine by proteolysis targeting. Nat Biotechnol. 2022; 40(9): 1370-1377.

[29]

Ryan A, Liu J, Deiters A. Targeted protein degradation through fast optogenetic activation and its application to the control of cell signaling. J Am Chem Soc. 2021; 143(24): 9222-9229.

[30]

He Z, Khatib A-M, Creemers JWM. The proprotein convertase furin in cancer: more than an oncogene. Oncogene. 2022; 41(9): 1252-1262.

[31]

Kiba Y, Oyama R, Misawa S, Tanikawa T, Kitamura M, Suzuki R. Screening for inhibitory effects of crude drugs on furin-like enzymatic activities. J Nat Med. 2021; 75(4): 1080-1085.

[32]

Hu X, Hai Z, Wu C, Zhan W, Liang G. A golgi-targeting and dual-color “turn-on” probe for spatially precise imaging of furin. Anal Chem. 2021; 93(3): 1636-1642.

[33]

Chu TT, Gao N, Li QQ, et al. Specific knockdown of endogenous Tau protein by peptide-directed ubiquitin-proteasome degradation. Cell Chem Biol. 2016; 23(4): 453-461.

[34]

Zhang Z, Wang Y, Ding Y, Hattori M. Structure-based engineering of anti-GFP nanobody tandems as ultra-high-affinity reagents for purification. Sci Rep. 2020; 10(1): 6239.

[35]

Zhang F, Wei H, Wang X, et al. Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov. 2017; 3: 17004.

[36]

Zhang N, Guo H, Zheng W, Wang T, Ma X. Design and screening of a chimeric survivin-specific nanobody and its anticancer activities in vitro. Anti-Cancer Drugs. 2016; 27(9): 839-847.

[37]

Cha JH, Chan LC, Li CW, Hsu JL, Hung MC. Mechanisms controlling PD-L1 expression in cancer. Mol Cell. 2019; 76(3): 359-370.

[38]

Liu J, Hong H, Shi J, et al. Dinitrophenol-mediated modulation of an anti-PD-L1 VHH for Fc-dependent effector functions and prolonged serum half-life. Eur J Pharm Sci. 2021; 165: 105941.

[39]

Wheatley SP, Altieri DC. Survivin at a glance. J Cell Sci. 2019; 132(7): jcs223826.

[40]

Qi Y, Liu Y, Yu B, et al. A lactose-derived CRISPR/Cas9 delivery system for efficient genome editing in vivo to treat orthotopic hepatocellular carcinoma. Adv Sci. 2020; 7(17): 2001424.

[41]

Zhao L, Zhao J, Zhong K, Tong A, Jia D. Targeted protein degradation: mechanisms, strategies and application. Signal Transduct Target Ther. 2022; 7(1): 113.

[42]

Guo N, Peng Z. MG132, a proteasome inhibitor, induces apoptosis in tumor cells. Asia Pac J Clin Oncol. 2013; 9(1): 6-11.

[43]

Mauthe M, Orhon I, Rocchi C, et al. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy. 2018; 14(8): 1435-1455.

[44]

Zhang Q, Liu S, Wang H, et al. ETV4 mediated tumor-associated neutrophil infiltration facilitates lymphangiogenesis and lymphatic metastasis of bladder cancer. Adv Sci (Weinh). 2023; 10(11): e2205613.

[45]

Xiao K, Peng S, Lu J, et al. UBE2S interacting with TRIM21 mediates the K11-linked ubiquitination of LPP to promote the lymphatic metastasis of bladder cancer. Cell Death & Disease. 2023; 14(7): 408.

[46]

Schapira M, Calabrese MF, Bullock AN, Crews CM. Targeted protein degradation: expanding the toolbox. Nature Reviews Drug Discovery. 2019; 18(12): 949-963.

[47]

Cheng X, Zhou X, Xu J, et al. Furin enzyme and pH synergistically triggered aggregation of gold nanoparticles for activated photoacoustic imaging and photothermal therapy of tumors. Anal Chem. 2021; 93(26): 9277-9285.

[48]

Deng C, Hu F, Zhao Z, et al. The establishment of quantitatively regulating expression cassette with sgRNA targeting BIRC5 to elucidate the synergistic pathway of Survivin with P-glycoprotein in cancer multi-drug resistance. Front Cell Dev Biol. 2021; 9: 797005.

[49]

Hu F, Deng C, Zhou Y, et al. Multistage targeting and dual inhibiting strategies based on bioengineered tumor matrix microenvironment-mediated protein nanocages for enhancing cancer biotherapy. Bioeng Transl Med. 2022; 7(2): e10290.

[50]

Sun X, Zhou C, Xia S, Chen X. Small molecule-nanobody conjugate induced proximity controls intracellular processes and modulates endogenous unligandable targets. Nat Commun. 2023; 14(1): 1635.

[51]

Deng CP, Li SH, Liu YP, et al. Split-Cas9-based targeted gene editing and nanobody-mediated proteolysis-targeting chimeras optogenetically coordinated regulation of Survivin to control the fate of cancer cells. Clin Transl Med. 2023; 13(8): e1382.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

171

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/