Treatment options for triple negative breast cancer (TNBC) are generally limited to cytotoxic chemotherapy. Recently, anti-epidermal growth factor receptor (EGFR) therapy has been introduced for TNBC patients. We engineered a novel nanobioconjugate based on a poly(β-L-malic acid) (PMLA) nanoplatform for TNBC treatment. The nanobioconjugate carries anti-tumor nucleosome-specific monoclonal antibody (mAb) 2C5 to target breast cancer cells, anti-mouse transferrin receptor (TfR) antibody for drug delivery through the host endothelial system, and Morpholino antisense oligonucleotide (AON) to inhibit EGFR synthesis. The nanobioconjugates variants were: (1) P (BioPolymer) with AON, 2C5 and anti-TfR for tumor endothelial and cancer cell targeting, and EGFR suppression (P/AON/2C5/TfR), and (2) P with AON and 2C5 (P/AON/2C5). Controls included (3) P with 2C5 but without AON (P/2C5), (4) PBS, and (5) P with PEG and leucine ester (LOEt) for endosomal escape (P/mPEG/LOEt). Drugs were injected intravenously to MDA-MB-468 TNBC bearing mice. Tissue accumulation of injected nanobioconjugates labeled with Alexa Fluor 680 was examined by Xenogen IVIS 200 (live imaging) and confocal microscopy of tissue sections. Levels of EGFR, phosphorylated and total Akt in tumor samples were detected by western blotting. In vitro western blot showed that the leading nanobioconjugate P/AON/2C5/TfR inhibited EGFR synthesis significantly better than naked AON. In vivo imaging revealed that 2C5 increased drug-tumor accumulation. Significant tumor growth inhibition was observed in mice treated with the lead nanobioconjugate (1) [P = 0.03 vs. controls; P<0.05 vs. nanobioconjugate variant (2)]. Lead nanobioconjugate (1) also showed stronger inhibition of EGFR expression and Akt phosphorylation than other treatments. Treatment of TNBC with the new nanobioconjugate results in tumor growth arrest by inhibiting EGFR and its downstream signaling intermediate, phosphorylated Akt. The nanobioconjugate represents a new generation of nanodrugs for treatment of TNBC.
References
[1]
Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, et al. (2007) Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 13: 4429–4434.
[2]
Bevers TB, Anderson BO, Bonaccio E, Buys S, Daly MB, et al. (2009) NCCN clinical practice guidelines in oncology: breast cancer screening and diagnosis. J Natl Compr Canc Netw 7: 1060–1096.
[3]
Pal SK, Childs BH, Pegram M (2011) Triple negative breast cancer: unmet medical needs. Breast Cancer Res Treat 125: 627–636.
[4]
Agrawal A, Gutteridge E, Gee JM, Nicholson RI, Robertson JF (2005) Overview of tyrosine kinase inhibitors in clinical breast cancer. Endocr Relat Cancer 12: Suppl 1S135–144.
[5]
Flynn JF, Wong C, Wu JM (2009) Anti-EGFR Therapy: Mechanism andAdvances in Clinical Efficacy in Breast Cancer. J Oncol 2009: 526963.
[6]
Baselga J (2002) Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 7: Suppl 42–8.
[7]
Huang S, Armstrong EA, Benavente S, Chinnaiyan P, Harari PM (2004) Dual-agent molecular targeting of the epidermal growth factor receptor (EGFR): combining anti-EGFR antibody with tyrosine kinase inhibitor. Cancer Res 64: 5355–5362.
[8]
Liu D, He J, Yuan Z, Wang S, Peng R, et al. (2010) EGFR expression correlates with decreased disease-free survival in triple-negative breast cancer: a retrospective analysis based on a tissue microarray. Med Oncol.
[9]
Atkinson RL, Zhang M, Diagaradjane P, Peddibhotla S, Contreras A, et al. (2010) Thermal enhancement with optically activated gold nanoshells sensitizes breast cancer stem cells to radiation therapy. Sci Transl Med 2: 55ra79.
[10]
Chandna P, Khandare JJ, Ber E, Rodriguez-Rodriguez L, Minko T (2010) Multifunctional tumor-targeted polymer-peptide-drug delivery system for treatment of primary and metastatic cancers. Pharm Res 27: 2296–2306.
[11]
van Renswoude J, Bridges KR, Harford JB, Klausner RD (1982) Receptor-mediated endocytosis of transferrin and the uptake of fe in K562 cells: identification of a nonlysosomal acidic compartment. Proc Natl Acad Sci U S A 79: 6186–6190.
[12]
Xia CQ, Shen WC (2001) Tyrphostin-8 enhances transferrin receptor-mediated transcytosis in Caco-2- cells and inreases hypoglycemic effect of orally administered insulin-transferrin conjugate in diabetic rats. Pharm Res 18: 191–195.
[13]
Lee BS, Fujita M, Khazenzon NM, Wawrowsky KA, Wachsmann-Hogiu S, et al. (2006) Polycefin, a new prototype of a multifunctional nanoconjugate based on poly(beta-L-malic acid) for drug delivery. Bioconjug Chem 17: 317–326.
[14]
Lee B-S, Vert M, Holler E (2002) Water-soluble aliphatic polyesters: poly(malic acid)s. In: Doi YSA, editor. Biopolymers. Weinheim: Wiley–VCH. pp. 75–103.
[15]
Ding H, Inoue S, Ljubimov AV, Patil R, Portilla-Arias J, et al. (2010) Inhibition of brain tumor growth by intravenous poly (beta-L-malic acid) nanobioconjugate with pH-dependent drug release [corrected]. Proc Natl Acad Sci U S A 107: 18143–18148.
[16]
Inoue S, Ding H, Portilla-Arias J, Hu J, Konda B, et al. (2011) Polymalic Acid-Based Nanobiopolymer Provides Efficient Systemic Breast Cancer Treatment by Inhibiting both HER2/neu Receptor Synthesis and Activity. Cancer Res 71: 1454–1464.
[17]
Ljubimova JY, Fujita M, Ljubimov AV, Torchilin VP, Black KL, et al. (2008) Poly(malic acid) nanoconjugates containing various antibodies and oligonucleotides for multitargeting drug delivery. Nanomedicine (Lond) 3: 247–265.
[18]
Braud C, Vert M (1992) Degradation of poly(p-malic acid) - monitoring of oligomers formation by aqueous SEC and HPCE. Polym Bull 29: 177–183.
[19]
Gasslmaier B, Holler E (1997) Specificity and direction of depolymerization of beta-poly(L-malate) catalysed by polymalatase from Physarum polycephalum–fluorescence labeling at the carboxy-terminus of beta-poly(L-malate). Eur J Biochem 250: 308–314.
[20]
Gasslmaier B, Krell CM, Seebach D, Holler E (2000) Synthetic substrates and inhibitors of beta-poly(L-malate)-hydrolase (polymalatase). Eur J Biochem 267: 5101–5105.
[21]
Fujita M, Lee BS, Khazenzon NM, Penichet ML, Wawrowsky KA, et al. (2007) Brain tumor tandem targeting using a combination of monoclonal antibodies attached to biopoly(beta-L-malic acid). J Control Release 122: 356–363.
[22]
Busch RK, Perlaky L, Valdez BC, Henning D, Busch H (1994) Apoptosis in human tumor cells following treatment with p120 antisense oligodeoxynucleotide ISIS 3466. Cancer Lett 86: 151–157.
[23]
Sekhon HS, London CA, Sekhon M, Iversen PL, Devi GR (2008) c-MYC antisense phosphosphorodiamidate morpholino oligomer inhibits lung metastasis in a murine tumor model. Lung Cancer 60: 347–354.
[24]
Wu B, Lu P, Benrashid E, Malik S, Ashar J, et al. (2010) Dose-dependent restoration of dystrophin expression in cardiac muscle of dystrophic mice by systemically delivered morpholino. Gene Ther 17: 132–140.
[25]
Kinali M, Arechavala-Gomeza V, Feng L, Cirak S, Hunt D, et al. (2009) Local restoration of dystrophin expression with the morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: a single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurol 8: 918–928.
[26]
Elbayoumi TA, Pabba S, Roby A, Torchilin VP (2007) Antinucleosome antibody-modified liposomes and lipid-core micelles for tumor-targeted delivery of therapeutic and diagnostic agents. J Liposome Res 17: 1–14.
[27]
Elbayoumi TA, Torchilin VP (2008) Tumor-specific antibody-mediated targeted delivery of Doxil reduces the manifestation of auricular erythema side effect in mice. Int J Pharm 357: 272–279.
[28]
ElBayoumi TA, Torchilin VP (2009) Tumor-targeted nanomedicines: enhanced antitumor efficacy in vivo of doxorubicin-loaded, long-circulating liposomes modified with cancer-specific monoclonal antibody. Clin Cancer Res 15: 1973–1980.
[29]
Lorenz MR, Holzapfel V, Musyanovych A, Nothelfer K, Walther P, et al. (2006) Uptake of functionalized, fluorescent-labeled polymeric particles in different cell lines and stem cells. Biomaterials 27: 2820–2828.
[30]
Wilhelm C, Billotey C, Roger J, Pons JN, Bacri JC, et al. (2003) Intracellular uptake of anionic superparamagnetic nanoparticles as a function of their surface coating. Biomaterials 24: 1001–1011.
[31]
Gupta B, Levchenko TS, Mongayt DA, Torchilin VP (2005) Monoclonal antibody 2C5-mediated binding of liposomes to brain tumor cells in vitro and in subcutaneous tumor model in vivo. J Drug Target 13: 337–343.
[32]
Fujita M, Khazenzon NM, Ljubimov AV, Lee BS, Virtanen I, et al. (2006) Inhibition of laminin-8 in vivo using a novel poly(malic acid)-based carrier reduces glioma angiogenesis. Angiogenesis 9: 183–191.
[33]
Ljubimova JY, Fujita M, Khazenzon NM, Lee BS, Wachsmann-Hogiu S, et al. (2008) Nanoconjugate based on polymalic acid for tumor targeting. Chem Biol Interact 171: 195–203.
[34]
Herbst RS, Bajorin DF, Bleiberg H, Blum D, Hao D, et al. (2006) Clinical Cancer Advances 2005: major research advances in cancer treatment, prevention, and screening–a report from the American Society of Clinical Oncology. J Clin Oncol 24: 190–205.
[35]
Jemal A, Siegel R, Ward E, Hao Y, Xu J, et al. (2009) Cancer statistics, 2009. CA Cancer J Clin 59: 225–249.
[36]
Kallergi G, Agelaki S, Kalykaki A, Stournaras C, Mavroudis D, et al. (2008) Phosphorylated EGFR and PI3K/Akt signaling kinases are expressed in circulating tumor cells of breast cancer patients. Breast Cancer Res 10: R80.
[37]
Sibilia M, Fleischmann A, Behrens A, Stingl L, Carroll J, et al. (2000) The EGF receptor provides an essential survival signal for SOS-dependent skin tumor development. Cell 102: 211–220.
[38]
Lurje G, Lenz HJ (2009) EGFR signaling and drug discovery. Oncology 77: 400–410.
[39]
Baselga J, Albanell J, Ruiz A, Lluch A, Gascon P, et al. (2005) Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J Clin Oncol 23: 5323–5333.
Modi S, D'Andrea G, Norton L, Yao TJ, Caravelli J, et al. (2006) A phase I study of cetuximab/paclitaxel in patients with advanced-stage breast cancer. Clin Breast Cancer 7: 270–277.
[42]
Nahta R, Yuan LX, Du Y, Esteva FJ (2007) Lapatinib induces apoptosis in trastuzumab-resistant breast cancer cells: effects on insulin-like growth factor I signaling. Mol Cancer Ther 6: 667–674.
[43]
Liu X, Wang Y, Nakamura K, Kubo A, Hnatowich DJ (2008) Cell studies of a three-component antisense MORF/tat/Herceptin nanoparticle designed for improved tumor delivery. Cancer Gene Ther 15: 126–132.
[44]
Maeda H, Bharate GY, Daruwalla J (2009) Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect. Eur J Pharm Biopharm 71: 409–419.
[45]
Maeda H, Fang J, Inutsuka T, Kitamoto Y (2003) Vascular permeability enhancement in solid tumor: various factors, mechanisms involved and its implications. Int Immunopharmacol 3: 319–328.
[46]
Kang CS, Zhang ZY, Jia ZF, Wang GX, Qiu MZ, et al. (2006) Suppression of EGFR expression by antisense or small interference RNA inhibits U251 glioma cell growth in vitro and in vivo. Cancer Gene Ther 13: 530–538.
[47]
Inoue S, Branch CD, Gallick GE, Chada S, Ramesh R (2005) Inhibition of Src kinase activity by Ad-mda7 suppresses vascular endothelial growth factor expression in prostate carcinoma cells. Mol Ther 12: 707–715.