Use of nano engineered approaches to overcome the stromal barrier in pancreatic cancer

https://doi.org/10.1016/j.addr.2018.06.014Get rights and content

Abstract

While chemotherapy is the only approved non-surgical option for the majority of pancreatic cancer patients, it rarely results in a cure. The failure to respond to chemotherapy is due to the presence of an abundant dysplastic stroma that interferes in drug delivery and as a result of drug resistance. It is appropriate, therefore, to consider the stromal contribution to the resistance to chemotherapy and sidestepping this barrier with nanocarriers that improve survival outcome. In this paper, we provide a short overview of the role of the stroma in chemotherapy resistance, including the use of nanocarriers to negate this barrier. We provide a perspective and guidance towards the implementation of nanotherapeutic approaches to improve therapeutic delivery and efficacy of PDAC management.

Introduction

Pancreatic cancer (PDAC) is the 4th leading cause of cancer death, with ~43,090 deaths in the US in 2017 [1]. In terms of mortality trends, Cancer Facts & Figures 2018 demonstrates that the 5-year survival rate has remained unchanged from 2006 to 2015 [2]. In the US, around 56,000 new cases of PDAC will be diagnosed in 2018 [2]. Collectively, for all stages of disease, the 5-year relative survival rate is only 8%. This number includes patients with metastatic (~52% patients) and local disease (~10% patients), with a 5-year survival rate of 3% and 32%, respectively. Due to the late diagnosis and early metastasis, for the majority patients with advanced disease, chemotherapy is considered as the only approved treatment, with the standard of care involving the use of nucleoside analog gemcitabine (GEM) or a more potent but highly toxic 4-drug regimen, FOLFIRINOX (i.e. oxaliplatin, irinotecan, 5-fluorouracil, and leucovorin). Moreover, chemotherapy is also used to treat the patients who are suitable for surgery (<20%), as neoadjuvant with a hope to lower recurrence. Unfortunately, these chemo applications seldom lead to a disease cure.

Chemotherapy failure can be partly explained by the presence of a dense desmoplastic stroma serving as a physical and biological barrier for drug delivery in PDAC and an unfavorable pharmacokinetics (PK) profile [3]. It is reasonable to consider, therefore, overcoming of the stromal interference in drug delivery and chemo-resistance to improve efficacy and patient survival [3]. A popular approach to overcoming the stromal resistance is to take advantage of the ability of nanocarriers to deliver therapeutic agents to the tumor site by mechanisms that differ from the uptake and retention of classic non-encapsulated molecular drug. A recent meta-analysis on PDAC clinical trials demonstrated that nanoparticles are promising approach to increase efficacy while reducing toxicity of multiple cancer drugs in PDAC patients [4]. Another exciting development is the use of smart design of the nanocarriers to enable them to negotiate the stroma barrier and improve drug delivery [5]. In fact, understanding the stromal contribution to the tumor access by an enhanced permeability and retention effect (EPR) in solid tumors is of particular relevance to the study of PDAC [[6], [7], [8], [9]]. The concept of “enhanced permeability” as an across-the-board explanation for nanocarrier access to solid tumor sites is over-simplified and needs to be re-interpreted [[10], [11], [12], [13], [14], [15]]. While enlarged tumor vascular fenestrations, irregular branching and abnormal angiogenesis have been reported in different cancer scenarios (many of them are xenograft models in mice) [6], the dysplastic stroma in PDAC indicate that additional consideration needs to be given to the poorly perfused, collapsed and obstructed blood vessels in this cancer as a result of tight adherence of stromal fibroblasts or pericytes to the vascular wall. In this communication, we provide a short overview to address the inhibitory effect of the stroma on PDAC treatment, including the consideration for the use of nanocarriers to potentially engineer and past this obstacle. We also provide a perspective and guidance towards the implementation of nanotherapeutic approaches in PDAC and other stroma-rich solid tumor types.

Section snippets

The pathophysiological contribution of the PDAC stroma to disease progression

A key characteristic of PDAC is the presence of tumor stroma, which contains cellular components (e.g. fibroblasts, immune cells, stellate cells, pericytes, endothelial cells), acellular components (e.g. collagens, fibronectin, growth factors and cytokines) and biophysical components (e.g. low pH, hypoxia, high tumor interstitial pressure) (Fig. 1A) [16]. These components interact in a multiplicative fashion to promote PDAC progression and tumor metastasis [17]. Previous studies have

Why overcoming tumor stroma is important to PDAC nanotherapeutics?

The high stromal volume in PDAC (up to 70% of the total tumor volume, Fig. 1B) requires disease-specific consideration to eliminate its impact on therapy [3]. Not only is the stroma poorly vascularized, but the existing vessels are relatively less leaky due to a high pericyte coverage, which blocks the extravasation of small molecule chemo agents as well as nanoparticles to the PDAC tumor site (Fig. 1C) [22, 23]. Kataoka et al., compared size-controlled polymer micelle nanoparticle access in

State-of-the-art approaches to overcome the stromal barrier in PDAC, including the use of nanocarriers

A number of stromal treatment strategies are currently being considered to improve PDAC treatment. While it is too early to evaluate the impact of PDAC stromal treatment, the field is beginning to understand the impact of multi-stage, multi-wave and combination therapy, which influence a multitude of mechanisms such as vasculature permeability, blood vessel patency, drug activation/degradation enzymes, and/or target specific biological factors, etc. These efforts involve the use of enzymatic

Future discovery to develop nanotherapy for PDAC and other stroma-rich solid tumors

Nanotechnology has contributed in a significant way in improving chemotherapy for PDAC over the last a few years. Two nanomedicines, i.e. Abraxane® and Onivyde® (irinotecan liposome injection) were approved by FDA for PDAC treatment. We have discussed further improvement in the treatment of this disease through the use of synergistic drug combinations, tumor targeting, toxicity reduction, overcoming stroma barrier, etc. These efforts can be conceptualized by a nano-enabled “engineered

Acknowledgements

This study was funded by the U.S. Public Health Service Grant U01CA198846.

Competing interests

A.E.N. and H.M. are co-founders and equity holders in Westwood Biosciences, Inc.

References (66)

  • B. Hoang et al.

    Docetaxel-carboxymethylcellulose nanoparticles target cells via a SPARC and albumin dependent mechanism

    Biomaterials

    (2015)
  • L. Mei et al.

    Increased tumor targeted delivery using a multistage liposome system functionalized with RGD, TAT and cleavable PEG

    Int. J. Pharm.

    (2014)
  • B.C. Özdemir et al.

    Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival

    Cancer Cell

    (2014)
  • C.-M.J. Hu et al.

    Nanoparticle-based combination therapy toward overcoming drug resistance in cancer

    Biochem. Pharmacol.

    (2012)
  • Cancer Facts and Figures

    (2017)
  • Cancer Facts & Figures

    (2018)
  • M. Erkan et al.

    The role of stroma in pancreatic cancer: diagnostic and therapeutic implications

    Nat. Rev. Gastroenterol. Hepatol.

    (2012)
  • M. Au et al.

    Emerging therapeutic potential of nanoparticles in pancreatic cancer: a systematic review of clinical trials

    Biomedicine

    (2016)
  • T. Ji et al.

    Using functional nanomaterials to target and regulate the tumor microenvironment: diagnostic and therapeutic applications

    Adv. Mater.

    (2013)
  • H. Zhou et al.

    IGF1 receptor targeted theranostic nanoparticles for targeted and image-guided therapy of pancreatic cancer

    ACS Nano

    (2015)
  • A. Nel et al.

    New insights into “Permeability” as in the enhanced permeability and retention effect of cancer nanotherapeutics

    ACS Nano

    (2017)
  • J. Shi et al.

    Cancer nanomedicine: progress, challenges and opportunities

    Nat. Rev. Cancer

    (2016)
  • A.Z. Wang

    EPR or no EPR? The billion-dollar question

    Sci. Transl. Med.

    (2015)
  • A. Neesse et al.

    Stromal biology and therapy in pancreatic cancer

    Gut

    (2011)
  • M. Waghray et al.

    Deciphering the role of stroma in pancreatic cancer

    Curr. Opin. Gastroenterol.

    (2013)
  • Z. Xu et al.

    Pancreatic cancer and its stroma: a conspiracy theory

    World J Gastroenterol: WJG

    (2014)
  • A. Masamune et al.

    Hypoxia stimulates pancreatic stellate cells to induce fibrosis and angiogenesis in pancreatic cancer

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2008)
  • K. Thind et al.

    Immunotherapy in pancreatic cancer treatment: a new frontier

    Ther. Adv. Gastroenterol.

    (2017)
  • A. Dimou et al.

    Overcoming the stromal barrier: technologies to optimize drug delivery in pancreatic cancer

    Ther. Adv. Med. Oncol.

    (2012)
  • H. Meng et al.

    Two-wave nanotherapy to target the stroma and optimize gemcitabine delivery to a human pancreatic cancer model in mice

    ACS Nano

    (2013)
  • H. Cabral et al.

    Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size

    Nat. Nanotechnol.

    (2011)
  • L.A. Shipley et al.

    Metabolism and disposition of gemcitabine, and oncolytic deoxycytidine analog, in mice, rats, and dogs

    Drug Metab. Dispos.

    (1992)
  • R.E. Laing et al.

    Noninvasive prediction of tumor responses to gemcitabine using positron emission tomography

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • Cited by (74)

    • Partial ligand shielding nanoparticles improve pancreatic ductal adenocarcinoma treatment via a multifunctional paradigm for tumor stroma reprogramming

      2022, Acta Biomaterialia
      Citation Excerpt :

      Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer deaths worldwide, with a five-year survival rate of only 7–9% [1].

    View all citing articles on Scopus

    This review is part of the Advanced Drug Delivery Reviews theme issue on “Perspectives and review articles on nanomedicine from NanoDDS 2017”.

    View full text