Theranostics 2020; 10(17):7921-7924. doi:10.7150/thno.49577 This issue Cite

Editorial

The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy

Yang Shi1 Corresponding address, Roy van der Meel2 Corresponding address, Xiaoyuan Chen3 Corresponding address, Twan Lammers1,4,5 Corresponding address

1. Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Aachen, Germany.
2. Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
3. Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Maryland, USA.
4. Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands.
5. Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands.

Citation:
Shi Y, van der Meel R, Chen X, Lammers T. The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Theranostics 2020; 10(17):7921-7924. doi:10.7150/thno.49577. https://www.thno.org/v10p7921.htm
Other styles

File import instruction

Abstract

Following its discovery more than 30 years ago, the enhanced permeability and retention (EPR) effect has become the guiding principle for cancer nanomedicine development. Over the years, the tumor-targeted drug delivery field has made significant progress, as evidenced by the approval of several nanomedicinal anticancer drugs. Recently, however, the existence and the extent of the EPR effect - particularly in patients - have become the focus of intense debate. This is partially due to the disbalance between the huge number of preclinical cancer nanomedicine papers and relatively small number of cancer nanomedicine drug products reaching the market. To move the field forward, we have to improve our understanding of the EPR effect, of its cancer type-specific pathophysiology, of nanomedicine interactions with the heterogeneous tumor microenvironment, of nanomedicine behavior in the body, and of translational aspects that specifically complicate nanomedicinal drug development. In this virtual special issue, 24 research articles and reviews discussing different aspects of the EPR effect and cancer nanomedicine are collected, together providing a comprehensive and complete overview of the current state-of-the-art and future directions in tumor-targeted drug delivery.

Keywords: EPR effect, enhanced permeability and retention (EPR), cancer nanomedicine, tumor targeting, active targeting, cancer immunotherapy, extracellular vesicles, imaging

 

Formulating therapeutic molecules in nanocarriers to yield nanomedicines is an attractive approach to improve the therapeutic index of oncology drugs. Over the last three decades, the development of cancer nanomedicines has resulted in thousands of publications and several approved drug products for the treatment of solid and hematological malignancies. In the context of solid tumors, the enhanced permeability and retention (EPR) effect has become an important driver of cancer nanomedicine design and it has served as a key cornerstone of tumor-targeted drug delivery [1-3].

Recently, however, the importance and the existence of the EPR effect in human patients have been heavily debated [4-6]. It has been demonstrated that the mechanism by which nanoparticles enter solid tumors is more complex than previously thought (potentially going beyond simple extravasation through gaps in the endothelial lining) [7], and that immune cells in the tumor microenvironment play important roles in nanomedicines' accumulation, retention and intratumoral distribution [8,9]. In addition, it is clear that the EPR effect is significantly more pronounced in the small animal xenograft tumor models which are typically used to evaluate cancer nanomedicines in preclinical settings as compared to tumor growing in humans [10]. Accumulation of nanocarriers in human tumors definitely does occur [11], but the extent varies heavily between patients and tumor types. Accordingly, quantifying the degree of EPR effect in tumors using non-invasive imaging is a promising approach to stratify patients for cancer nanomedicine treatment [12,13]. Moreover, strategies are needed to improve the effectiveness of nanomedicine therapy. This can be done via pharmacological and physical co-treatments to prime tumors for improved delivery and efficacy, via active targeting, via the use of multi-stage and/or stimuli-responsive nanocarrier materials, and via the combination of nanotherapeutics with immunotherapy [14], which has already shown initial clinical success [15]. In this virtual special issue of Theranostics, 24 research and review articles are compiled which discuss approaches aimed at improving the therapeutic efficacy of cancer nanomedicine. These strategies by themselves, and especially when combined with others, will improve cancer nanomedicine's clinical translation and ultimately improve patient outcomes [16-18].

Traditionally, EPR-mediated tumor accumulation is proposed to result from long-circulating nanoparticles with a hydrodynamic diameter size exceeding the renal clearance threshold, which can extravasate from leaky tumor vessels. However, recent studies have investigated approaches to extend the conventional concept of EPR-based tumor targeting. For example, Liu et al. describe the potential of exploring transcytosis for tumor targeting, which is a potential additional mechanism to mediate tumor targeting by nanomedicines, especially in highly stromal solid tumors such as pancreatic ductal adenocarcinoma with weak EPR effect [19]. Bort and colleagues discuss studies on the use of ultrasmall nanoparticles for tumor targeting. These include polysiloxane-based nanoparticles with a hydrodynamic diameter of approximately 4 nm, which have been successfully tested in animal models and have recently entered a clinical trial for treating patients with brain metastases [20]. In a comparative study, Xu et al. investigate the tumor targeting efficiency of ligand-modified nanoparticles of 3 and 30 nm, respectively. Their results show that functionalizing 3 nm nanoparticles with a targeting ligand increased tumor targeting efficiency and tumor penetration while this was not the case for 30 nm nanoparticles [21].

To improve the EPR effect and nanomedicine effectiveness, pharmacological and physical co-treatments have been employed to prime the tumor microenvironment. Kwon and colleagues summarize features of the tumor microenvironment that impair EPR-based tumor targeting by nanomedicines. In addition, several priming strategies to improve EPR effect are discussed, including physical and physiological measures to remodel the tumor microenvironment [22]. Dhaliwal and Zheng focus on the applications of physical strategies to improve EPR effect of tumors including ultrasound and hyperthermia. The authors also summarize assessment methods and proper use of animal models to study EPR-mediated nanomedicine targeting [23]. Among the physical strategies, Duan et al. discuss the applications of micro/nanobubbles to augment the thermal effect, acoustic streaming and cavitation mechanisms of ultrasound to enhance the EPR effect [24]. Recognizing the importance of the vasculature in tumor development, Tsioumpekou et al. demonstrate that specific suppression of PDGFRβ kinase activity by 1-NaPP1 effectively modulates the tumor microenvironment by inhibiting angiogenesis [25].

Active targeting can be used as a complementary strategy to EPR-based passive targeting to improve nanomedicine tumor accumulation and retention. Tumor targeting ligands include antibodies, fragments of antibodies (e.g. nanobodies) and peptides. Dammes and Peer summarize the applications of monoclonal antibodies in molecular imaging of cancer, autoimmune disorders and cardiovascular diseases [26]. In addition, the potential of using monoclonal antibody-based molecular imaging strategies in theranostics and precision medicine is highlighted. Oliveira and co-workers utilize epidermal growth factor receptor (EGFR)-targeted nanobodies to deliver photosensitizers to tumors for photodynamic therapy. Both monovalent nanobodies and biparatopic nanobodies are conjugated with photosensitizers. Although these two types of conjugates exhibit different biodistribution profiles, they result in similar levels of necrosis after photodynamic therapy, resulting in tumor reduction [27]. Minko and colleagues report on the use of a synthetic luteinizing hormone-releasing hormone (LHRH) decapeptide for targeting lung cancer to deliver paclitaxel and siRNAs via nanostructured lipid nanoparticles. The nanomedicine was administered via inhalation which also showed efficient homing to target cells [28]. Zhong and colleagues utilize cyclic RGD as a targeting ligand to improve the delivery of disulfide-crosslinked iodine-rich polymersomes to B16 melanoma. The actively targeted polymersomes exhibit an in vivo elimination half-life of 6.5 h in the blood circulation, thus achieving efficient tumor targeting (6.7 %ID/g) and displaying promising therapeutic efficacy [29].

Another strategy to improve cancer therapy is to employ nanomedicine-based combination treatments. Zhao et al. discuss the potential of this approach for the treatment of glioblastoma, benefiting from synergistic combinations of different therapeutic agents. The authors discuss the rationale of nanomedicine-based drug combinations and recent clinical progress in nanocarrier-based combination therapies [30]. Yu et al. discuss a special class of nanomedicines which induce cancer starvation by anti-angiogenesis and vascular blockade [31]. Such nano-interventions have been combined with other modalities such as chemotherapy, gene therapy and photodynamic therapy to achieve synergistic effects for cancer treatment. Zhu et al. report on a pH-sensitive nanomedicine formulation combining an enzyme, focused ultrasound-based tumor ablation and hypoxia alleviation to potentiate doxorubicin-based chemotherapy [32]. Their catalase-loaded nanoparticles were able to increase oxygen levels in tumors by converting H2O2 to O2, which improved the effect of ultrasound ablation and reduced tumor hypoxia, and these effects together improved doxorubicin efficacy.

In addition to conventional nanocarriers used for EPR-based tumor targeting, new carriers based on bio-inspired design and materials allowing for tumor-selective drug release have been exploited. Wolfram and co-workers review the use of extracellular vesicle-based drug delivery systems [33]. The intrinsic tissue tropism of extracellular vesicles is highly promising for tumor targeting and the authors summarize methods to load therapeutic agents in extracellular vesicle, and modification strategies to improve their tumor targeting ability. Mi summarizes nanomedicines with stimuli responsiveness for tumor targeted imaging, therapy and theranostics [34]. Nanomedicines sensitive to endogenous and exogenous stimuli as well as their potential to improve therapeutic efficacy are discussed.

Cancer nanomedicines have been extensively combined with immunotherapy to improve treatment outcomes. Yu and colleagues summarize the recent progress of combination nano-immunotherapy, with a special focus on nanomedicines modulating the tumor immune microenvironment (TIME) to improve immunotherapeutic efficacy [35]. An experimental report by Panagi et al. describes an immunomodulatory nanomedicine based on liposomes co-loaded with a transforming growth factor beta inhibitor and an immunogenic cell death inducer [36]. The liposome-based combination treatment improves the immunogenicity of triple-negative breast tumors and potentiates the efficacy of checkpoint blockade antibodies.

Imaging is instrumental in tumor targeting and translational cancer nanomedicine, as it can help capture tumor targeting efficiency and the heterogeneity of the EPR effect in tumors. Miller, Weissleder and colleagues comprehensively review the advances in image-guided systems pharmacology of cancer nanomedicines [37]. Recent developments of quantitative imaging technologies and their applications in systems pharmacology of nanomedicine are discussed, with a focus on utilizing computational modeling to understand and guide the manipulation of the EPR effect and tumor microenvironment for improving nanomedicine therapy. Dasgupta, Lammers et al. summarize the value of imaging-assistance in determining nanomedicine biodistribution, target site accumulation and drug release [38]. Imaging techniques to eventually enable patient stratification via companion nanodiagnostics, via nanotheranostics, via conventional imaging techniques and via immunohistochemistry are discussed. A comprehensive review by De Maar, Deckers and colleagues addresses multiscale imaging techniques for analyzing the heterogeneity of nanomedicines' spatial distribution in tumors, which is an important - and often overlooked - reason for inefficient nanotherapy [39]. The authors summarize the applications as well as the strengths and weaknesses of 3 classes of imaging techniques for assessing the intratumoral distribution of nanomedicines, i.e. non-invasive clinical imaging modalities (nuclear imaging, magnetic resonance imaging, computed tomography and ultrasound), optical imaging and mass spectrometry imaging. Moss and co-workers provide novel insight on the use of high-resolution ex vivo micro-computed tomography for studying the spatial distribution of liposomes in 4 different tumor models [40]. Their work identifies vessel distribution and vessel support as crucial determinants of efficient liposome accumulation and distribution in tumors. Qi et al. report on the use of hyaluronic acid conjugated with fluorescent dyes for molecular imaging of pancreatic cancer in settings allowing for intraoperative imaging [41]. Their results demonstrate that the molecular weight of hyaluronic acid and the physicochemical properties of conjugated dyes affect the efficiency of tumor-specific imaging. Finally, Goos et al. report on star polymers chelated with MRI contrast agents and radioisotopes for molecular imaging and endoradiotherapy of cancerous lesions via exploiting EPR-based tumor accumulation. In CT26 tumor-bearing mice, the star polymer-based nanoparticles demonstrated a very high tumor targeting efficiency (15-22 %ID/g), which contributed to the improved survival of mice upon endoradiotherapy intervention [42].

Altogether, this Theranostics special issue presents a timely and comprehensive collection of research and review articles focusing on the EPR effect and beyond. These articles summarize from various different angles our current understanding of nanomedicine-based tumor targeting, and they provide valuable expert perspectives on how to improve the use and the efficacy of (EPR-based) nanomedicine formulations for cancer therapy.

Competing Interests

The authors have declared that no competing interest exists.

References

1. Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46:6387-92

2. Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release. 2000;65:271-84

3. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv Drug Deliv Rev. 2015;91:3-6

4. Park K. The beginning of the end of the nanomedicine hype. J Control Release. 2019;305:221-2

5. Wilhelm S, Tavares AJ, Dai Q, Ohta S, Audet J, Dvorak HF. et al. Analysis of nanoparticle delivery to tumours. Nat Rev Mater. 2016;1:1-12

6. Lammers T. Macro-nanomedicine: targeting the big picture. J Control Release. 2019;294:372-5

7. Sindhwani S, Syed AM, Ngai J, Kingston BR, Maiorino L, Rothschild J. et al. The entry of nanoparticles into solid tumours. Nat Mater. 2020;19:566-75

8. Miller MA, Zheng YR, Gadde S, Pfirschke C, Zope H, Engblom C. et al. Tumour-associated macrophages act as a slow-release reservoir of nano-therapeutic Pt(IV) pro-drug. Nat Commun. 2015;6:8692

9. Miller MA, Gadde S, Pfirschke C, Engblom C, Sprachman MM, Kohler RH. et al. Predicting therapeutic nanomedicine efficacy using a companion magnetic resonance imaging nanoparticle. Sci Transl Med. 2015;7:314ra183

10. Petersen GH, Alzghari SK, Chee W, Sankari SS, La-Beck NM. Meta-analysis of clinical and preclinical studies comparing the anticancer efficacy of liposomal versus conventional non-liposomal doxorubicin. J Control Release. 2016;232:255-64

11. Harrington KJ, Mohammadtaghi S, Uster PS, Glass D, Peters AM, Vile RG. et al. Effective targeting of solid tumors in patients with locally advanced cancers by radiolabeled pegylated liposomes. Clin Cancer Res. 2001;7:243-54

12. Lee H, Shields AF, Siegel BA, Miller KD, Krop I, Ma CX. et al. 64Cu-MM-302 positron emission tomography quantifies variability of enhanced permeability and retention of nanoparticles in relation to treatment response in patients with metastatic breast cancer. Clin Cancer Res. 2017;23:4190-202

13. Ramanathan RK, Korn RL, Raghunand N, Sachdev JC, Newbold RG, Jameson G. et al. Correlation between ferumoxytol uptake in tumor lesions by MRI and response to nanoliposomal irinotecan in patients with advanced solid tumors: a pilot study. Clin Cancer Res. 2017;23:3638-48

14. Meel RVD, Sulheim E, Shi Y, Kiessling F, Mulder WJM, Lammers T. Smart cancer nanomedicine. Nat Nanotechnol. 2019;14:1007-17

15. Shi Y. Clinical translation of nanomedicine and biomaterials for cancer immunotherapy: progress and perspectives. Adv Ther, in press. doi: 10.1002/adtp.201900215

16. Martins JP, das Neves J, de la Fuente M, Celia C, Florindo H, Günday-Türeli N. et al. The solid progress of nanomedicine. Drug Deliv Transl Res. 2020;10:726-9

17. Min Y, Caster JM, Eblan MJ, Wang AZ. Clinical translation of nanomedicine. Chem Rev. 2015;115:11147-90

18. Anselmo AC, Mitragotri S. Nanoparticles in the clinic: an update. Bioeng Transl Med. 2019;4:e10143

19. Liu X, Jiang J, Meng H. Transcytosis - an effective targeting strategy that is complementary to “EPR effect” for pancreatic cancer nano drug delivery. Theranostics. 2019;9:8018-25

20. Bort G, Lux F, Dufort S, Crémillieux Y, Verry C, Tillement O. EPR-mediated tumor targeting using ultrasmall-hybrid nanoparticles: from animal to human with theranostic AGuIX nanoparticles. Theranostics. 2020;10:1319-31

21. Xu Y, Wu H, Huang J, Qian W, Martinson DE, Ji B. et al. Probing and enhancing ligand-mediated active targeting of tumors using sub-5 nm ultrafine iron oxide nanoparticles. Theranostics. 2020;10:2479-94

22. Park J, Choi Y, Chang H, Um W, Ryu JH, Kwon IC. Alliance with EPR effect: combined strategies to improve the EPR effect in the tumor microenvironment. Theranostics. 2019;9:8073-90

23. Dhaliwal A, Zheng G. Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: designing a new perspective in nanomedicine delivery. Theranostics. 2019;9:8091-108

24. Duan L, Yang L, Jin J, Yang F, Liu D, Hu K. et al. Micro/nano-bubble-assisted ultrasound to enhance the EPR effect and potential theranostic applications. Theranostics. 2020;10:462-83

25. Tsioumpekou M, Cunha SI, Ma H, Åhgren A, Cedervall J, Olsson AK. et al. Specific targeting of PDGFRβ in the stroma inhibits growth and angiogenesis in tumors with high PDGF-BB expression. Theranostics. 2020;10:1122-35

26. Dammes N, Peer D. Monoclonal antibody-based molecular imaging strategies and theranostic opportunities. Theranostics. 2020;10:938-55

27. de Bruijn HS, Mashayekhi V, Schreurs TJL, van Driel PBAA, Strijkers GJ, van Diest PJ. et al. Acute cellular and vascular responses to photodynamic therapy using EGFR-targeted nanobody-photosensitizer conjugates studied with intravital optical imaging and magnetic resonance imaging. Theranostics. 2020;10:2436-52

28. Garbuzenko OB, Kuzmov A, Taratula O, Pine SR, Minko T. Strategy to enhance lung cancer treatment by five essential elements: inhalation delivery, nanotechnology, tumor-receptor targeting, chemo- and gene therapy. Theranostics. 2019;9:8362-76

29. Zou Y, Wei Y, Sun Y, Bao J, Yao F, Li Z. et al. Cyclic RGD-functionalized and disulfide-crosslinked iodine-rich polymersomes as a robust and smart theranostic agent for targeted CT imaging and chemotherapy of tumor. Theranostics. 2019;9:8061-72

30. Zhao M, van Straten D, Broekman MLD, Préat V, Schiffelers RM. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics. 2020;10:1355-72

31. Yu S, Chen Z, Zeng X, Chen X, Gu Z. Advances in nanomedicine for cancer starvation therapy. Theranostics. 2019;9:8026-47

32. Zhu J, Li Z, Zhang C, Lin L, Cao S, Che H. et al. Single enzyme loaded nanoparticles for combinational ultrasound-guided focused ultrasound ablation and hypoxia-relieved chemotherapy. Theranostics. 2019;9:8048-60

33. Walker S, Busatto S, Pham A, Tian M, Suh A, Carson K. et al. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics. 2019;9:8001-17

34. Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics. 2020;10:4557-88

35. Saeed M, Gao J, Shi Y, Lammers T, Yu H. Engineering nanoparticles to reprogram the tumor immune microenvironment for improved cancer immunotherapy. Theranostics. 2019;9:7981-8000

36. Panagi M, Voutouri C, Mpekris F, Papageorgis P, Martin MR, Martin JD. et al. TGF-β inhibition combined with cytotoxic nanomedicine normalizes triple negative breast cancer microenvironment towards anti-tumor immunity. Theranostics. 2020;10:1910-22

37. Ng TSC, Garlin MA, Weissleder R, Miller MA. Improving nanotherapy delivery and action through image-guided systems pharmacology. Theranostics. 2020;10:968-97

38. Dasgupta A, Biancacci I, Kiessling F, Lammers T. Imaging-assisted anticancer nanotherapy. Theranostics. 2020;10:956-67

39. de Maar JS, Sofias AM, Siegel TP, Vreeken RJ, Moonen C, Bos C. et al. Spatial heterogeneity of nanomedicine investigated by multiscale imaging of the drug, the nanoparticle and the tumour environment. Theranostics. 2020;10:1884-909

40. Moss JI, Barjat H, Emmas SA, Strittmatter N, Maynard J, Goodwin RJA. et al. High-resolution 3D visualization of nanomedicine distribution in tumors. Theranostics. 2020;10:880-97

41. Qi B, Crawford AJ, Wojtynek NE, Talmon GA, Hollingsworth MA, Ly QP. et al. Tuned near infrared fluorescent hyaluronic acid conjugates for delivery to pancreatic cancer for intraoperative imaging. Theranostics. 2020;10:3413-29

42. Goos JACM, Cho A, Carter LM, Dilling TR, Davydova M, Mandleywala K. et al. Delivery of polymeric nanostars for molecular imaging and endoradiotherapy through the enhanced permeability and retention (EPR) effect. Theranostics. 2020;10:567-84

Author contact

Corresponding address Corresponding authors: yshide, r.v.d.meelnl, shawn.chengov, tlammersde


Published 2020-6-25


Citation styles

APA
Shi, Y., van der Meel, R., Chen, X., Lammers, T. (2020). The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Theranostics, 10(17), 7921-7924. https://doi.org/10.7150/thno.49577.

ACS
Shi, Y.; van der Meel, R.; Chen, X.; Lammers, T. The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Theranostics 2020, 10 (17), 7921-7924. DOI: 10.7150/thno.49577.

NLM
Shi Y, van der Meel R, Chen X, Lammers T. The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Theranostics 2020; 10(17):7921-7924. doi:10.7150/thno.49577. https://www.thno.org/v10p7921.htm

CSE
Shi Y, van der Meel R, Chen X, Lammers T. 2020. The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Theranostics. 10(17):7921-7924.

This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
Popup Image