Yonsei Med J.  2018 Jul;59(5):595-601. 10.3349/ymj.2018.59.5.595.

A Tumor-Specific Tissue-Penetrating Peptide Enhances the Efficacy of Chemotherapy Drugs in Gastric Cancer

Affiliations
  • 1The Second Outpatient Department of Chengdu Army Region Authority, Chengdu, China.
  • 2Department of Gastroenterology, the 520th Hospital of People's Liberation Army, Mianyang, China.
  • 3Sichuan Province Administration of Traditional Chinese Medicine, Chengdu, China.
  • 4Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China. zhoulin1105@163.com

Abstract

PURPOSE
C-end rule (CendR) peptides are found to enhance the penetration of chemotherapeutic agents into tumor cells, while GX1 is a peptide that homes to gastric cancer (GC) vasculature. This study aimed to synthesize a novel peptide GX1-RPAKPAR (GXC) and to explore the effect of GXC on sensitizing GC cells to chemotherapeutic agents.
MATERIALS AND METHODS
Intracellular Adriamycin concentration analysis was applied to conform whether GXC peptide increases the penetration of chemotherapeutic agents into GC cells in vitro. The effect of GXC peptide on sensitizing GC cells to chemotherapeutics was validated by apoptosis assay and in vitro/vivo drug sensitivity assay. The specificity of GXC to GC tissue was validated by ex vivo fluorescence imaging.
RESULTS
In vitro, administration of GXC significantly increased Adriamycin concentrations inside SGC-7901 cells, and enhanced the efficacy of chemotherapeutic agents by decreasing the IC50 value. In vivo, FITC-GXC specifically accumulated in GC tissue. Moreover, systemic co-injection with GXC peptide and Adriamycin statistically improved the therapeutic efficacy in SGC-7901 xenograft models, surprisingly, without obviously increasing side effects.
CONCLUSION
These results demonstrated that co-administration of the novel peptide GXC with chemotherapeutic agents may be a potential way to enhance the efficacy of anticancer drugs in GC treatment.

Keyword

GXC; GX1; C-end rule peptide; gastric cancer; penetration

MeSH Terms

Apoptosis
Doxorubicin
Drug Therapy*
Heterografts
In Vitro Techniques
Inhibitory Concentration 50
Optical Imaging
Peptides
Sensitivity and Specificity
Stomach Neoplasms*
Doxorubicin
Peptides

Figure

  • Fig. 1 GXC peptide enhanced the drug sensitivity of SGC-7901 cells in vitro. (A) Cytotoxicity of RPAKPAR, GX1, and GXC to SGC-7901 cells was acceptable. (B–D) RPAKPAR and GXC peptides sensitize gastric cancer cells to chemotherapeutics in vitro. (B) The effect of co-administration of three kinds of peptides on IC50 values of SGC-7901 cells to ADR, VCR, 5FU, and CDDP. (C) The apoptotic rates of SGC-7901 cells treated with ADR (0.4 µg/mL) or 5FU (10 µg/mL), co-administrated with GX1 (1×10−6 µmol/mL) or RPAKPAR (1×10−6 µmol/mL), respectively, for 24 h, were measured by flow cytometry. (D) The statistical analyses of cell apoptosis of SGC-7901 to ADR and 5FU. *p<0.05. GXC, GX1-RPAKPAR.

  • Fig. 2 GXC increased intracellular ADR concentrations in vitro. (A) SGC-7901 cells were treated with ADR (10 µg/mL) combined with GX1, RPAKPAR, and GXC (1×10−6 µmol/mL) respectively for 1 h. Green fluorescence indicates the cytoplasmic membrane stained with DIO, and red fluorescence indicates ADR. (B) The average red fluorescence intensity at different times. GXC, GX1-RPAKPAR.

  • Fig. 3 GXC specifically accumulated in gastric cancer in nude mice bearing tumor xenografts derived from SGC-7901 cells. (A) Ex vivo imaging of tumor and normal tissues of FITC-GXC (40 µmol/kg) uptake at 2 h after euthanizing the mice. (B) Fluorescence intensity ratio of tumor and normal tissues were quantified using regions of interest analysis (p<0.01). GXC, GX1-RPAKPAR; GFP, green fluorescent protein.

  • Fig. 4 Co-administration with GXC enhanced the anti-tumor effect of free ADR in nude mice bearing tumor xenografts. (A) Fluorescence imaging of GFP-SGC-7901 tumor models every 4 days. (B) Fluorescence intensity ratio of tumors was quantified by region of interest analysis. (C) The statistical analyses of tumor weight collected after treating for 24 days. (D) The statistical analyses of mice body weight shift. *p<0.05; **p<0.01. GXC, GX1-RPAKPAR; GFP, green fluorescent protein.


Reference

1. Bray F, Jemal A, Grey N, Ferlay J, Forman D. Global cancer transitions according to the Human Development Index (2008–2030): a population-based study. Lancet Oncol. 2012; 13:790–801.
Article
2. Sha H, Zou Z, Xin K, Bian X, Cai X, Lu W, et al. Tumor-penetrating peptide fused EGFR single-domain antibody enhances cancer drug penetration into 3D multicellular spheroids and facilitates effective gastric cancer therapy. J Control Release. 2015; 200:188–200.
Article
3. Teesalu T, Sugahara KN, Kotamraju VR, Ruoslahti E. C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetration. Proc Natl Acad Sci U S A. 2009; 106:16157–16162.
Article
4. Sugahara KN, Teesalu T, Karmali PP, Kotamraju VR, Agemy L, Greenwald DR, et al. Coadministration of a tumor-penetrating peptide enhances the efficacy of cancer drugs. Science. 2010; 328:1031–1035.
Article
5. Schmithals C, Köberle V, Korkusuz H, Pleli T, Kakoschky B, Augusto EA, et al. Improving drug penetrability with iRGD leverages the therapeutic response to sorafenib and doxorubicin in hepatocellular carcinoma. Cancer Res. 2015; 75:3147–3154.
Article
6. Hui X, Han Y, Liang S, Liu Z, Liu J, Hong L, et al. Specific targeting of the vasculature of gastric cancer by a new tumor-homing peptide CGNSNPKSC. J Control Release. 2008; 131:86–93.
Article
7. Chen B, Cao S, Zhang Y, Wang X, Liu J, Hui X, et al. A novel peptide (GX1) homing to gastric cancer vasculature inhibits angiogenesis and cooperates with TNF alpha in anti-tumor therapy. BMC Cell Biol. 2009; 10:63.
Article
8. Chen K, Sun X, Niu G, Ma Y, Yap LP, Hui X, et al. Evaluation of 64Cu labeled GX1: a phage display peptide probe for PET imaging of tumor vasculature. Mol Imaging Biol. 2012; 14:96–105.
Article
9. Xin J, Zhang X, Liang J, Xia L, Yin J, Nie Y, et al. In vivo gastric cancer targeting and imaging using novel symmetric cyanine dye-conjugated GX1 peptide probes. Bioconjug Chem. 2013; 24:1134–1143.
Article
10. Green M, Loewenstein PM. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat transactivator protein. Cell. 1988; 55:1179–1188.
Article
11. Oliveira EA, Faintuch BL. Radiolabeling and biological evaluation of the GX1 and RGD-GX1 peptide sequence for angiogenesis targeting. Nucl Med Biol. 2015; 42:123–130.
Article
12. Xiong D, Liu Z, Bian T, Li J, Huang W, Jing P, et al. GX1-mediated anionic liposomes carrying adenoviral vectors for enhanced inhibition of gastric cancer vascular endothelial cells. Int J Pharm. 2015; 496:699–708.
Article
13. Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell. 1988; 55:1189–1193.
Article
14. Elliott G, O'Hare P. Intercellular trafficking and protein delivery by a herpesvirus structural protein. Cell. 1997; 88:223–233.
Article
15. Morris MC, Deshayes S, Heitz F, Divita G. Cell-penetrating peptides: from molecular mechanisms to therapeutics. Biol Cell. 2008; 100:201–217.
Article
16. Arap W, Pasqualini R, Ruoslahti E. Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science. 1998; 279:377–380.
Article
17. Aird WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ Res. 2007; 100:158–173.
18. Arap W, Haedicke W, Bernasconi M, Kain R, Rajotte D, Krajewski S, et al. Targeting the prostate for destruction through a vascular address. Proc Natl Acad Sci U S A. 2002; 99:1527–1531.
Article
19. Svensen N, Walton JG, Bradley M. Peptides for cell-selective drug delivery. Trends Pharmacol Sci. 2012; 33:186–192.
Article
20. Sugahara KN, Teesalu T, Karmali PP, Kotamraju VR, Agemy L, Girard OM, et al. Tissue-penetrating delivery of compounds and nanoparticles into tumors. Cancer Cell. 2009; 16:510–520.
Article
21. Du Y, Zhang Q, Jing L, Liang X, Chi C, Li Y, et al. GX1-conjugated poly(lactic acid) nanoparticles encapsulating Endostar for improved in vivo anticolorectal cancer treatment. Int J Nanomedicine. 2015; 10:3791–3802.
Article
22. Mantis C, Kandela I, Aird F. Reproducibility Project: Cancer Biology. Replication study: coadministration of a tumor-penetrating peptide enhances the efficacy of cancer drugs. Elife. 2017; 6:e17584.
Article
23. Baker M, Dolgin E. Cancer reproducibility project releases first results. Nature. 2017; 541:269–270.
Article
24. Staton CA, Kumar I, Reed MW, Brown NJ. Neuropilins in physiological and pathological angiogenesis. J Pathol. 2007; 212:237–248.
Article
25. Jia H, Bagherzadeh A, Hartzoulakis B, Jarvis A, Löhr M, Shaikh S, et al. Characterization of a bicyclic peptide neuropilin-1 (NP-1) antagonist (EG3287) reveals importance of vascular endothelial growth factor exon 8 for NP-1 binding and role of NP-1 in KDR signaling. J Biol Chem. 2006; 281:13493–13502.
Article
26. Pellet-Many C, Frankel P, Jia H, Zachary I. Neuropilins: structure, function and role in disease. Biochem J. 2008; 411:211–226.
Article
27. Akagi M, Kawaguchi M, Liu W, McCarty MF, Takeda A, Fan F, et al. Induction of neuropilin-1 and vascular endothelial growth factor by epidermal growth factor in human gastric cancer cells. Br J Cancer. 2003; 88:796–802.
Article
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr