Elsevier

Biomaterials

Volume 32, Issue 15, May 2011, Pages 3862-3874
Biomaterials

Integrin-assisted drug delivery of nano-scaled polymer therapeutics bearing paclitaxel

https://doi.org/10.1016/j.biomaterials.2011.01.073Get rights and content

Abstract

Angiogenesis plays a prominent role in cancer progression. Anti-angiogenic therapy therefore, either alone or in combination with conventional cytotoxic therapy, offers a promising therapeutic approach. Paclitaxel (PTX) is a widely-used potent cytotoxic drug that also exhibits anti-angiogenic effects at low doses. However, its use, at its full potential, is limited by severe side effects. Here we designed and synthesized a targeted conjugate of PTX, a polymer and an integrin-targeted moiety resulting in a polyglutamic acid (PGA)-PTX-E-[c(RGDfK)2] nano-scaled conjugate. Polymer conjugation converted PTX to a macromolecule, which passively targets the tumor tissue exploiting the enhanced permeability and retention effect, while extravasating via the leaky tumor neovasculature. The cyclic RGD peptidomimetic enhanced the effects previously seen for PGA–PTX alone, utilizing the additional active targeting to the αvβ3 integrin overexpressed on tumor endothelial and epithelial cells. This strategy is particularly valuable when tumors are well-vascularized, but they present poor vascular permeability. We show that PGA is enzymatically-degradable leading to PTX release under lysosomal acidic pH. PGA-PTX-E-[c(RGDfK)2] inhibited the growth of proliferating αvβ3-expressing endothelial cells and several cancer cells. We also showed that PGA-PTX-E-[c(RGDfK)2] blocked endothelial cells migration towards vascular endothelial growth factor; blocked capillary-like tube formation; and inhibited endothelial cells attachment to fibrinogen. Orthotopic studies in mice demonstrated preferential tumor accumulation of the RGD-bearing conjugate, leading to enhanced anti-tumor efficacy and a marked decrease in toxicity as compared with free PTX-treated mice.

Introduction

Pharmacological, molecular and genetic evidence demonstrate that tumor progression is angiogenesis-dependent [1], [2]. Cell adhesion mechanisms facilitating migration and invasion through the extracellular matrix are critical to the growth of new blood vessels [3]. Integrins, a class of receptors involved in cell adhesion, play a key role in cell matrix interactions and thus in angiogenesis. Integrin alterations are responsible for a number of pathological manifestations, such as defective embryogenesis, blood coagulation, osteoporosis, acute renal failure, retinopathy and cancer [4], [5].

αvβ3 integrin receptors are involved in angiogenesis and tumor invasiveness, essential in cell adhesion, motility, growth and differentiation [6]. αvβ3 integrin binds the Arg-Gly-Asp (RGD) sequence, which constitutes the recognition domain of adhesion proteins including laminin, fibronectin and vitronectin [7]. RGD peptidomimetics compete with extracellular matrix proteins to bind to integrin receptors [8]. αvβ3 receptors are expressed mainly on the luminal surface of the endothelial cell predominantly during angiogenesis, making it a compelling target for agents within the vascular space. In addition, αvβ3 integrin is overexpressed on proliferating tumor endothelial cells, as well as on various tumor cells, such as glioblastoma [9]. It is a marker of poor prognosis for some tumor types [10], [11]. The bis-cyclic peptide E-[c(RGDfK)2] is a vascular-targeting ligand used in this study to actively and selectively target the chemotherapeutic drug paclitaxel (PTX) to tumor cells and their surrounding tumor endothelial cells via binding to αvβ3 integrin receptor.

The microtubule-interfering agent PTX is a clinically well-established and highly-effective anti-neoplastic drug used for the treatment of many carcinomas including prostate, breast, ovarian, and non-small cell lung cancer. It is also the drug of choice for the treatment of metastatic breast carcinoma. In addition to its anti-neoplastic activity, PTX exhibits anti-angiogenic and pro-apoptotic properties [11]. Due to its hydrophobic nature, PTX is solubilized in CremophorEL or ethanol, when used clinically, causing hypersensitivity reactions in addition to the severe side effects associated with PTX itself (such as neurotoxicity). Moreover, PTX’s poor pharmacokinetics (short half-life, low selectivity) lead to the fact that only a small amount of the drug localizes in the tumor. An additional limitation of PTX is that it is a substrate of efflux pumps [12], resulting in multiple drug resistance.

One of the most successful approaches to deliver PTX, has been the development of an albumin-based PTX nanoparticle named Abraxane® (ABI-007, Celgene Corporation). It was approved by the FDA in 2004 for the treatment of breast cancer after the failure of combination chemotherapy for metastatic disease or relapse within 6 months of adjuvant chemotherapy. PTX is physically entrapped in the Abraxane® nanoparticle, leading to enhanced solubility of PTX and thus avoids harmful solubilizing agent [13].

A chemical conjugation of PTX to a carrier could offer pharmacological advantages. Conjugation of PTX with the non-degradable polymer, N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer showed improved pharmacokinetics and promising advantages [14]. The use of this strategy failed clinically due to premature release of PTX in the circulation causing a similar toxicity profile as free PTX. Cell Therapeutics Inc. (Seattle) took a different approach conjugating PTX with the biodegradable polyglutamic acid (PGA), OPAXIO™. It showed clinical benefits, such as safety superiority compared to free PTX, in paclitaxel-based cancer treatment alone or in combination with radiotherapy or other small drugs such as cisplatin [15], [16], [17].

PGA is a water-soluble multivalent polymer, which allows the conjugation of more than one compound or targeting residue within the polymer backbone. It is non-immunogenic, non-toxic, and biodegradable by cathepsin B [18], [19], [20], an enzyme that is highly expressed in most tumor tissues [21], [22], [23], [24]. In this manuscript, we evaluated the anti-angiogenic effect on top of the known anti-tumor effect of polymer conjugate of PTX. In addition, we improved PGA–PTX conjugate’s anti-tumor efficiency and widened its clinical applications. We designed and synthesized a PGA-PTX-E-[c(RGDfK)2] conjugate that targets the tumor both (i) passively due to the enhanced permeability and retention (EPR) effect by virtue of its size and structure as PGA–PTX conjugate [25], and (ii) actively due to integrin binding.

We have characterized an original and superior targeted therapeutic synthetic nanoconjugate named PGA-PTX-E-[c(RGDfK)2] with the potential to target angiogenesis [26], [27] detected by in vitro assays on endothelial cells. In addition, we evaluated its anti-tumor effect on cancer cell lines both in vitro and in vivo in tumor-bearing mice.

Section snippets

Materials

All chemicals and solvents were A.R. grade or purified by standard techniques. Chemical reagents were purchased from Sigma–Aldrich (Madrid, Spain). HPLC grade solvents were from Merck (Barcelona, Spain). PTX was from Petrus Chemicals and Materials Ltd. (Israel) and Shaanxi Sciphar Hi-Tech Industry Co. (Xi’an, China). E-[c(RGDfK)2] and c(RADfK) were from Peptides International (KY, USA). Fluorescence dye Oregon Green-cadaverine (OG) was from Invitrogen (Barcelona, Spain). Cy5.5 dye was kindly

Molecular weight, drug loading and polydispersity

The general two step synthesis of a PGA-PTX-E-[c(RGDfK)2] is depicted in Fig. 1A. The ester linker is hydrolytically labile. A series of PGA-based conjugates with general chemical structure (Fig. 1A,C) has been synthesized and fully characterized. Average molecular weight (Mw) and polydispersity (Mw/Mn) were determined by size exclusion chromatography (SEC) as 17700 Da (Mw/Mn = 1.3) for PGA, 48600 Da (Mw/Mn = 1.3) for PGA-PTX-E-[c(RGDfK)2] conjugate and 35700 Da (Mw/Mn = 1.3) for PGA-E-[c(RGDfK)

Discussion

Two successful strategies in cancer therapy that are currently being evaluated in clinical trials consist of targeting αvβ3 integrin receptors and conjugating PTX to a polymer. However, the thought of combining these powerful approaches by developing an anti-αvβ3 and PTX bi-specific macromolecule targeting the tumor cells and their endothelial microenvironment had not been given previous consideration. The synergism found here, results in a therapy that constitutes a significant step within the

Conclusions

The targeted polymer-based PTX delivery conjugate, PGA-PTX-E-[c(RGDfK)2], significantly augments the anti-tumor activity of both the free drug and the conjugated PGA–PTX. Inclusion of an active targeting moiety to integrin expressing-cells, leads to a selective anti-angiogenic mechanism of action and an alternative manipulation to overcome acquired multi-drug resistance.

Acknowledgements

This study was supported (in part) by grant no. 5145-300000 from the Chief Scientist Office of the Ministry of Health, Israel, by THE ISRAEL SCIENCE FOUNDATION (Grant No. 1300/06), by the Israel Cancer Research Fund, by the Recanati Foundation (RSF) and by the United States-Israel Binational Science Foundation (Grant No. 2007347, RSF and RL). We thank the Spanish Ministry of Science and Education (MICINN, CTQ2007-060601), Generalitat Valenciana (ACOMP/2009/086), European Commission FP7-Health

References (42)

  • J. Folkman

    Angiogenesis: an organizing principle for drug discovery?

    Nat Rev Drug Discov

    (2007)
  • S.A. Alavi et al.

    Integrins in angiogenesis

  • D. Cox et al.

    The pharmacology of the integrins

    Med Res Rev

    (1994)
  • C.J. Avraamides et al.

    Integrins in angiogenesis and lymphangiogenesis

    Nat Rev Cancer

    (2008)
  • E. Lohof et al.

    Carbohydrate derivatives for use in drug design:cyclic alpha(v)-selective RGD peptides

    Angew Chem Int Ed Engl

    (2000)
  • S. Monferran et al.

    Alphavbeta3 and alphavbeta5 integrins control glioma cell response to ionising radiation through ILK and RhoB

    Int J Cancer

    (2008)
  • B. Felding-Habermann et al.

    Involvement of integrin alpha V gene expression in human melanoma tumorigenicity

    J Clin Invest

    (1992)
  • J. Wang et al.

    Paclitaxel at ultra low concentrations inhibits angiogenesis without affecting cellular microtubule assembly

    Anticancer Drugs

    (2003)
  • E. Miele et al.

    Albumin-bound formulation of paclitaxel (Abraxane ABI-007) in the treatment of breast cancer

    Int J Nanomedicine

    (2009)
  • W.J. Gradishar

    Albumin-bound paclitaxel: a next-generation taxane

    Expert Opin Pharmacother

    (2006)
  • K. Miller et al.

    Targeting bone metastases with a bispecific anticancer and antiangiogenic polymer-alendronate-taxane conjugate

    Angew Chem Int Ed Engl

    (2009)
  • Cited by (115)

    • Drug–polymer conjugates: Challenges, opportunities, and future prospects in clinical trials

      2023, Polymer-Drug Conjugates: Linker Chemistry, Protocols and Applications
    • Smart Polymeric Nanocarriers for Drug Delivery

      2019, Smart Polymers and Their Applications
    • Molecular platforms for targeted drug delivery

      2019, International Review of Cell and Molecular Biology
    View all citing articles on Scopus
    View full text