Fact-checked by Grok 2 weeks ago

Enhanced permeability and retention effect

The enhanced permeability and retention (EPR) effect is a pathophysiological phenomenon in solid tumors whereby macromolecules and nanoparticles (typically 10–200 nm in size) selectively accumulate in tumor tissues due to the abnormal leakiness of tumor vasculature and deficient lymphatic drainage, enabling passive targeting for anticancer drug delivery. This effect was first described in 1986 by researchers Y. Matsumura and H. Maeda, who observed the tumoritropic accumulation of a polymer-conjugated antitumor agent in animal models, marking a foundational concept in nanomedicine. The mechanism of the EPR effect arises from the unique , where rapid leads to irregularly structured blood vessels with wide endothelial gaps (vascular pore cutoff sizes of 380–780 nm in many tumors), allowing of circulating nanoparticles that would otherwise be confined in normal tissues. Once in the tumor , these agents are retained due to suppressed lymphatic clearance and high pressure, resulting in concentrations 10–100 times higher than in healthy organs. Key mediators include (VEGF) and , which further enhance in response to tumor-induced . In clinical applications, the EPR effect underpins the design of nanoparticle-based therapeutics, such as liposomal (Doxil) and albumin-bound (Abraxane), which exploit passive targeting to improve drug efficacy while reducing systemic toxicity. However, the effect's heterogeneity across tumor types, patient physiologies, and locations—such as lower expression in pancreatic or cancers—with growing recognition of its variable clinical expression (as of 2024 reviews)—limits its universality, with studies showing only modest 1.5–2-fold delivery enhancements over normal tissues in some cases. Emerging strategies, including EPR enhancers like donors or tumor-priming agents, aim to standardize and amplify this phenomenon for broader therapeutic impact.

History and Discovery

Initial Observations

The initial documentation of the enhanced permeability and retention (EPR) effect emerged from studies on passive targeting of macromolecules to tumors in rodent models during the 1980s. In a seminal 1986 investigation, researchers observed that a polymer-conjugated anticancer agent, styrene maleic anhydride-neocarzinostatin (SMANCS), exhibited significantly higher accumulation in tumor tissues compared to the unconjugated protein neocarzinostatin alone. This selective tumoritropic behavior was attributed to the unique physiological properties of tumor vasculature, marking the conceptual origin of the EPR effect as a mechanism for macromolecular drug delivery. The experimental setup involved tumor-bearing mice injected intravenously with a range of 51Cr-labeled proteins varying in molecular weight from 12,000 to 160,000 , including representatives like (Mr 69,000) and . These proteins progressively accumulated in tumor tissues, achieving a tumor-to-blood of up to 5 within 19 to 72 hours for most macromolecules, with larger proteins such as IgG requiring longer times to reach this level. In contrast, smaller proteins like neocarzinostatin (Mr 12,000) failed to achieve even a ratio of 1. Retention was further demonstrated using an -dye complex, which visualized prolonged accumulation exclusively in tumor tissue due to minimal lymphatic drainage, with little recovery observed over extended periods. Early observations highlighted the leaky nature of tumor vasculature in animal models, particularly in tumors exceeding 150-200 μm in diameter, where cells beyond the oxygen diffusion limit induce to sustain growth. This contrasted sharply with normal tissues, where endothelial barriers restrict macromolecular to maintain vascular integrity. The defective architecture in tumors allowed passive leakage of even large molecules into the , a phenomenon absent in healthy vasculature. The initial posited a direct link between tumor-induced and enhanced , supported by electron microscopy evidence revealing fenestrations and gaps up to 600-800 in tumor vessel endothelia. These structural abnormalities, arising from rapid and disorganized angiogenic sprouting, facilitated the of macromolecules while impairing their clearance, laying the groundwork for the EPR effect as a tumor-specific targeting .

Key Milestones and Researchers

The enhanced permeability and retention (EPR) effect was first described in 1986 by Hiroshi Maeda and colleagues at University School of Medicine, based on observations from experiments showing selective accumulation of macromolecular proteins in tumor tissues. Maeda's pioneering work extended to the development of styrene-maleic acid-conjugated neocarzinostatin (SMANCS), a polymer-drug conjugate that exploited the EPR effect for targeted delivery to solid tumors, leading to its approval in in 1993 for the treatment of . In the 1990s, the EPR concept gained traction through clinical applications of liposomal formulations, notably pegylated liposomal (Doxil), which was approved by the U.S. in 1995 for and later for other cancers. Doxil's design leveraged the EPR effect to achieve passive tumor targeting, resulting in reduced compared to free by limiting exposure to healthy tissues while enhancing accumulation in permeable tumor vasculature. The 2000s marked the integration of the effect with advancements, exemplified by the work of Halas and Jennifer West at , who developed gold nanoshells for near-infrared photothermal therapy between 2003 and 2005. These silica-core gold-shell nanoparticles were engineered to accumulate in tumors via the EPR effect and convert light to heat for localized ablation, demonstrating efficacy in preclinical mouse models of cancer. A pivotal 2016 review by Stefan Wilhelm and colleagues in Nature Reviews Materials analyzed over 100 studies and quantified the typical delivery efficiency to tumors at a of 0.7% of the injected dose, highlighting limitations in -based accumulation and spurring refinements in design. From 2023 to 2025, the effect has received renewed recognition through commemorative publications honoring Maeda's legacy following his passing in 2021, alongside updated models that incorporate tumor heterogeneity to better predict variable across patient tumors. These models emphasize microenvironmental factors like and vascular to enhance reliability, as detailed in recent reviews on precision strategies.

Mechanism of the EPR Effect

Tumor Angiogenesis and Vascular Permeability

Tumors exceeding 1-2 mm in diameter induce angiogenesis to sustain growth, a process primarily driven by intratumoral hypoxia and the release of angiogenic growth factors such as vascular endothelial growth factor (VEGF). This hypoxic environment activates hypoxia-inducible factor-1α (HIF-1α), which upregulates VEGF expression in tumor cells, promoting endothelial cell proliferation and migration from nearby normal vessels. The resulting neovasculature is structurally immature, characterized by the absence or sparse coverage of pericytes and smooth muscle cells, which normally stabilize mature vessels and regulate permeability.00528-1) These immature vessels exhibit tortuous architecture, uneven diameters, and increased branching, contributing to heterogeneous blood flow and elevated interstitial fluid pressure within the tumor microenvironment. The endothelial lining of these tumor vessels displays pronounced abnormalities that enhance permeability, including wide interendothelial gaps ranging from 200 to 2000 nm, an irregular and discontinuous , and elevated vascular density. These gaps, often observed via electron microscopy in various tumor models, arise from defective endothelial junctions and cytoskeletal disorganization, allowing components to leak into the tumor . In contrast to normal vessels, where tight junctions limit , the high vascular density—often exceeding that of surrounding normal tissue—facilitates rapid of leaky conduits, particularly in solid tumors like carcinomas and sarcomas. VEGF serves as the primary mediator of this leakiness, though its detailed role is further explored in molecular contexts. These vascular defects enable selective extravasation of macromolecules and nanoparticles, forming the basis of enhanced permeability in the EPR effect. Molecules larger than 40 or nanoparticles sized 10-200 readily pass through the enlarged endothelial gaps, accumulating in the tumor , whereas normal vessels restrict passage with an effective cutoff of approximately 5-10 due to tighter junctions. This size-dependent threshold ensures that low-molecular-weight compounds (<40 ) circulate freely without significant tumor retention, while larger entities exploit the leaky vasculature for targeted delivery. Quantitatively, the permeability coefficient (P) for —a key protein— in tumor vessels is 10-100 times higher than in normal tissues, reflecting the profound impact of . This elevated permeability can be modeled using an adaptation of Starling's principle for fluid flux across the , accounting for the leaky nature of tumor vessels: \text{Flux} = K_f \left[ (P_c - P_i) - \sigma (\pi_c - \pi_i) \right] Here, K_f is the filtration coefficient, P_c and P_i are capillary and interstitial hydrostatic pressures, \pi_c and \pi_i are oncotic pressures, and \sigma is the , which is markedly reduced (approaching 0) in tumors due to protein leakage through gaps, diminishing the oncotic barrier. In normal , \sigma is closer to 1, effectively opposing filtration, but tumor-specific reductions amplify net outward flux of macromolecules like .

Impaired Lymphatic Drainage and Retention

In solid tumors, lymphatic vessels are often absent or functionally impaired, particularly in the tumor core, due to mechanical compression caused by rapid cellular proliferation and elevated interstitial fluid pressure (IFP). This compression collapses lymphatic structures, preventing effective drainage of extravasated fluid and macromolecules from the interstitial space. Tumor IFP typically ranges from 10 to 40 mmHg, compared to less than 5 mmHg in normal tissues, further exacerbating lymphatic dysfunction by counteracting the pressure gradient needed for lymph flow. This elevated IFP arises in part from the leaky tumor vasculature, which allows excessive fluid influx into the interstitium. The impaired lymphatic drainage results in prolonged retention of macromolecules and nanoparticles within the tumor , a key component of the EPR effect. In tumors, the of these agents is typically 2-10 times longer than in normal —often extending to days rather than hours—due to the lack of efficient clearance pathways. This extended retention enables accumulation levels that can be 10- to 100-fold higher in tumor compared to or normal organs, facilitating selective . The tumor's dense interstitial matrix, rich in and hyaluronan, further contributes to retention by physically trapping extravasated nanoparticles and reducing their back-diffusion to the bloodstream. Collagen fibers form a rigid scaffold that limits molecular mobility, while hyaluronan creates a hydrated gel-like barrier that hinders convective and diffusive clearance. These components, overexpressed in many tumors, effectively prolong the local residence time of therapeutic agents, enhancing their . Experimental evidence from animal models underscores the extent of lymphatic impairment in tumors. Lymphoscintigraphy studies in have demonstrated that lymphatic clearance of radiolabeled tracers from tumor sites is markedly reduced, often less than 1% within hours, compared to approximately 50% clearance in normal tissues over the same period. These findings highlight how dysfunctional lymphatics promote sustained intratumoral accumulation, distinguishing tumor microenvironments from healthy ones.

Molecular and Physiological Mediators

The enhanced permeability and retention (EPR) effect in tumors is significantly influenced by specific molecular mediators that disrupt endothelial barriers, allowing macromolecular . Vascular endothelial growth factor (VEGF) is a primary mediator, acting through activation to phosphorylate and disrupt proteins such as , thereby increasing endothelial gaps and . VEGF binds to VEGFR2 on endothelial cells, triggering Src-dependent signaling that loosens intercellular junctions and promotes fluid leakage into the tumor interstitium. Bradykinin, a generated from kininogen by enzymes in the , further amplifies permeability by engaging B2 receptors on endothelial cells, which induce cytoskeletal rearrangements and gap formation between cells. This receptor-mediated action elevates intracellular calcium and activates pathways that transiently open paracellular routes, facilitating the passage of solutes and nanoparticles. (NO), produced by endothelial (eNOS) and inducible (iNOS) overexpressed in tumor endothelium and infiltrating immune cells, relaxes vascular and disrupts tight junctions by S-nitrosylation of junctional proteins. Elevated NO levels in hypoxic tumors maintain dilated vessels and enhance leakiness, contributing directly to the EPR phenomenon. Prostaglandins, such as (PGI2) and (PGE2), are lipid mediators released by tumor-associated inflammatory cells and , binding to IP and EP receptors respectively to modulate junctional integrity and increase permeability. These eicosanoids activate cyclic AMP-dependent pathways that weaken adherens junctions, promoting in inflamed tumor tissues. Physiological triggers in the further drive these mediators. Inflammation, characterized by and infiltration, upregulates production of VEGF, , and NO through signaling, sustaining a permissive vascular state. Hypoxia-inducible factor-1α (HIF-1α), stabilized under low oxygen conditions prevalent in solid tumors, transcriptionally induces VEGF and other permeability factors, linking metabolic stress to barrier disruption. Matrix metalloproteinases (MMPs), secreted by tumor and stromal cells, degrade components like and claudins, creating persistent endothelial gaps that amplify mediator effects. Mediator interactions often exhibit , enhancing permeability beyond individual contributions. For instance, VEGF upregulates eNOS and iNOS expression, boosting NO production and doubling vascular leakiness in endothelial cell models compared to VEGF alone. Such cooperative signaling, observed with human umbilical vein endothelial cells, underscores how interconnected pathways in tumors intensify the EPR effect. Mediator-induced permeability can be conceptually modeled as a change in , \Delta P = f([\text{mediator}]), where f represents a dose-response derived from like the Miles assay, which quantifies via dye leakage after intradermal mediator injection (e.g., at 10^{-7} M elicits measurable increases in ). This simplified relation highlights the nonlinear amplification in tumor contexts, informed by dose-response data.

Factors Influencing the EPR Effect

Tumor Heterogeneity and Microenvironment

Tumor heterogeneity significantly influences the enhanced permeability and retention (EPR) effect, as variations in vascular architecture and tissue composition across different tumor types lead to inconsistent nanoparticle accumulation. Sarcomas often exhibit a pronounced EPR effect due to their leaky vessels, facilitating greater extravasation of macromolecules compared to normal tissues. In contrast, prostate and pancreatic cancers typically display a diminished EPR effect, attributed to dense stroma and high interstitial pressure that restrict permeability. This inter-tumor variability underscores the need for type-specific considerations in EPR-dependent therapies. The stage and size of tumors further modulate the EPR effect, with optimal accumulation observed in mid-stage nodules measuring 1-5 mm, where active promotes leaky vasculature without extensive . As tumors progress to larger sizes, the development of necrotic cores impairs and reduces EPR efficiency by creating avascular regions that limit . Similarly, metastatic sites often show attenuated EPR compared to primary tumors, due to altered microenvironmental pressures and heterogeneous vascularization in distant organs. Key microenvironmental factors, including elevated interstitial fluid pressure (IFP), , and , profoundly impact by hindering and penetration. High IFP, often reaching 20-40 mmHg in solid tumors versus near 0 mmHg in normal tissues, compresses vessels and opposes convective , thereby reducing EPR-mediated accumulation. Tumor , with extracellular typically ranging from 6.5 to 7.0, further exacerbates this by promoting vessel collapse and limiting diffusion. , characterized by dense stromal collagen deposition, restricts interstitial flow and , particularly in fibrotic tumors like pancreatic adenocarcinomas. Conversely, immune cell infiltration, such as by tumor-associated macrophages, can dynamically alter , potentially enhancing local through cytokine-mediated vessel dilation. Patient-specific physiological factors, such as and cardiovascular health, also influence the EPR effect by modulating systemic blood flow and pressure, with preclinical studies showing enhanced tumor accumulation under hypertensive conditions (as of 2025). Quantitative assessments reveal substantial variability in the EPR effect, with the EPR index—defined as the accumulation ratio of nanoparticles in tumor versus normal tissue—ranging from 2- to 20-fold across preclinical models. Recent intravital studies in 2024 have highlighted this heterogeneity, showing that while some tumor regions achieve high ratios due to focal leaky vessels, others exhibit minimal enhancement owing to microenvironmental barriers.

Nanocarrier Properties and Design

The design of nanocarriers is critical for maximizing exploitation of the enhanced permeability and retention (EPR) effect, with physical properties such as size, shape, and surface characteristics directly influencing into tumor and prolonged circulation. Optimal nanocarrier diameters typically range from 20 to 100 nm, allowing passage through leaky tumor vasculature while facilitating within the dense . This size range balances accumulation via EPR with sufficient mobility, as larger particles exceeding 200 nm are predominantly sequestered in systemic circulation by splenic or phagocytic clearance. The diffusion coefficient D of nanocarriers follows the Stokes-Einstein relation, where D \propto 1/r (with r as the ), underscoring how smaller sizes enhance interstitial transport despite reduced vascular penetration for particles below 20 nm. Nanoparticle shape profoundly affects tumor penetration and retention, with non-spherical morphologies often outperforming spheres. Linear polymers or filomicelles, characterized by high aspect ratios greater than 3:1, demonstrate improved tumor accumulation compared to spherical counterparts, achieving up to 2- to 3-fold enhancements in efficiency due to better alignment with blood flow and reduced margination to vessel walls. These elongated structures prolong circulation times—up to 10-fold longer than spheres in preclinical models—and facilitate deeper tissue infiltration post-extravasation. Surface modifications further optimize nanocarrier performance by mitigating immune recognition and modulating interactions with biological barriers. Poly(ethylene glycol) (PEG)ylation imparts a stealth coating that evades opsonization and uptake, extending plasma to 24-48 hours in formulations like PEGylated liposomes. Optimal surface charge, characterized by near-neutral zeta potentials between -10 and +10 mV, promotes longevity in circulation by minimizing protein adsorption and nonspecific binding, thereby enhancing EPR-mediated tumor delivery. Slightly negative or neutral charges are preferred over highly positive ones, which accelerate clearance despite favoring cellular uptake. Recent advancements as of 2025 emphasize responsive and hybrid designs to refine exploitation. pH-sensitive linkers, such as or Schiff-base bonds in chitosan-based or mesoporous silica nanocarriers, enable triggered disassembly in the acidic (pH ~6.5), promoting on-site drug release while maintaining stability during circulation. Hybrid nanoparticles, exemplified by liposomes conjugated with targeting ligands like anti-EGFR antibodies post- accumulation, combine passive retention with active receptor engagement, achieving 1.5- to 3-fold increases in tumor-specific uptake in preclinical cancer models.

Applications in Medicine

Therapeutic Drug Delivery

The enhanced permeability and retention (EPR) effect enables passive targeting of nanocarriers to solid tumors, facilitating the delivery of therapeutic agents by exploiting leaky tumor vasculature and impaired lymphatic clearance. This approach has revolutionized anticancer drug formulations, allowing for higher intratumoral drug concentrations while minimizing exposure to healthy tissues. Liposomal and polymeric systems, in particular, have leveraged EPR to encapsulate cytotoxic drugs, improving efficacy and safety profiles in clinical settings. One of the earliest and most successful applications is Doxil, a pegylated liposomal formulation of approved by the FDA in , which achieves higher drug levels in tumors compared to free through EPR-mediated accumulation. This selective retention reduces systemic toxicity, particularly cardiotoxicity, enabling safer administration in patients with and . Polymeric conjugates represent another key advancement, with PK1 (an N-(2-hydroxypropyl)methacrylamide copolymer conjugated to doxorubicin) demonstrating EPR-dependent efficacy in Phase II trials for breast and ovarian cancers. In these studies, PK1 showed partial responses in 21% of breast cancer patients (3/14 evaluable) and 5% of ovarian cancer patients (1/19), attributed to enhanced tumor extravasation and reduced off-target effects compared to conventional doxorubicin. Inorganic nanoparticles, such as gold nanoshells developed by the Halas group, have been employed for photothermal ablation, where near-infrared laser irradiation induces localized heating to destroy tumor cells following EPR-based accumulation. These nanoshells, often combined with chemotherapeutic agents like , enhance synergistic effects by improving drug release and penetration in solid tumors, as shown in preclinical models around 2005. Recent innovations include phthalocyanine-nanoparticle conjugates for (PDT), which utilize to deliver s selectively to solid tumors for light-activated generation. From 2023 to 2025, these conjugates have shown improved EPR-mediated tumor accumulation, enabling enhanced PDT outcomes in preclinical cancer models by overcoming limitations in solubility and .

Diagnostic Imaging and Theranostics

The enhanced permeability and retention (EPR) effect has been leveraged to develop nanoparticle-based contrast agents for non-invasive tumor visualization, enabling improved detection and characterization of solid tumors through modalities such as (MRI) and (PET). These agents exploit leaky tumor vasculature for preferential accumulation, providing enhanced signal intensity at the tumor site compared to normal tissues. Gadolinium-loaded liposomes serve as a prominent example of EPR-targeted , where the nanoparticles accumulate in tumors via passive , resulting in significant signal enhancement for better delineation of tumor margins. For instance, PEGylated gadolinium liposomes demonstrate prolonged circulation and tumor-specific uptake, achieving up to several-fold increase in relaxivity and contrast compared to free agents, thereby facilitating high-resolution of tumor heterogeneity. Co-loading with therapeutic agents in such liposomes further supports image-guided interventions, though the primary diagnostic benefit stems from EPR-mediated accumulation. Radiolabeled nanoparticles, such as those incorporating (64Cu), have enabled quantitative assessment of the effect using imaging, particularly in clinical settings during the . 64Cu-labeled liposomes, like MM-302, accumulate in metastatic tumors through EPR, allowing scans to measure nanoparticle deposition variability across patients and tumors, with uptake ratios often exceeding 2-5 times background in responsive lesions. These studies highlight PET's role in stratifying patients for EPR-dependent therapies by visualizing and quantifying tumor permeability non-invasively. In theranostics, nanoparticles (IONPs) combined with imaging and therapeutic payloads exemplify integrated platforms that utilize for and treatment. Doxorubicin-loaded IONPs, for example, provide T2-weighted MRI upon EPR-driven tumor accumulation, enabling real-time monitoring of nanoparticle delivery and drug release in models of and . This approach allows for MRI-guided assessment of intratumoral distribution, with signal changes correlating to therapeutic efficacy and retention due to impaired lymphatic clearance. Recent advances as of 2025 incorporate image-guided techniques to transiently amplify the effect during diagnostic procedures, enhancing extravasation and accuracy. Ultrasound-mediated strategies, such as with microbubble contrast agents, temporarily increase in targeted tumor regions, boosting EPR accumulation of nanoparticles and improving (CEUS) visualization of tumor by up to 2-3 times. This method supports precise, on-demand enhancement for better tumor border definition and EPR heterogeneity mapping without systemic side effects.

Clinical Evidence and Translation

Preclinical Models and Findings

Preclinical investigations of the enhanced permeability and retention () effect have primarily utilized models to demonstrate and quantify accumulation in tumors. In subcutaneous xenograft models, commonly employed in mice and rats, nanoparticles of approximately 50 nm in size have shown tumor accumulation ranging from 2% to 7% of the injected dose per gram of (% ID/g), significantly higher than the less than 1% ID/g observed in major organs like the liver under similar conditions. These models, involving implantation of human or murine tumor cells under , provide accessible sites for monitoring but often exhibit more uniform vascularization compared to natural tumor settings. Early observations in xenografts from the 1980s confirmed the foundational EPR mechanism through elevated macromolecular retention in tumors relative to normal tissues. Orthotopic models, where tumors are implanted at their native organ sites, offer improved representation of human tumor heterogeneity and microenvironmental factors, leading to variations in EPR efficiency. For instance, in orthotopic pancreatic tumor models, nanoparticle uptake via EPR is typically 2- to 5-fold lower than in corresponding subcutaneous xenografts due to denser stromal barriers and reduced . These models better recapitulate clinical scenarios, such as hypovascularity in pancreatic ductal adenocarcinoma, where EPR-mediated accumulation may reach only 0.5-2% ID/g, highlighting the influence of tumor location on . In vitro assays, including Transwell permeability models using endothelial cell monolayers derived from tumor vasculature, have corroborated the size-dependent extravasation central to the EPR effect. These systems demonstrate that nanoparticles smaller than 200 nm exhibit significantly higher permeability across leaky barriers mimicking tumor , with flux rates increasing up to 3-fold for 50 nm particles compared to larger ones exceeding 100 nm. Such assays isolate from systemic factors, confirming that pore sizes in tumor-like (around 200-800 nm) favor selective nanoparticle escape while retaining them due to absent lymphatics. Key findings from meta-analyses and advanced imaging underscore the variability and modest efficiency of EPR in preclinical settings. A comprehensive meta-analysis of 117 studies encompassing 5331 tumors reported a median nanoparticle delivery efficiency of 0.7% ID/g to solid tumors, with only 0.64% of the administered dose reaching the target site across various rodent models. Recent intravital microscopy studies have further revealed the dynamic nature of the EPR effect, showing temporal fluctuations in vascular permeability and nanoparticle extravasation over hours to days, influenced by tumor growth stages and immune interactions in murine xenografts. These insights emphasize that while EPR enables preferential accumulation, its magnitude is often limited by inter- and intra-tumor heterogeneity in preclinical models. A 2023 meta-analysis of nanoparticle distribution confirmed median tumor delivery of approximately 0.76% ID/g, highlighting ongoing challenges in translation.

Human Trials and Outcomes

Early clinical investigations into the enhanced permeability and retention (EPR) effect focused on pegylated liposomal (Doxil), approved by the FDA in 1995 for AIDS-related . In a pivotal phase III randomized involving 258 patients with advanced , Doxil administered at 20 mg/m² every 2-3 weeks achieved an overall response rate of 45.9% (95% CI: 37%-54%), significantly outperforming the standard regimen of , , and , which yielded 24.8% (95% CI: 17%-32%; P < .001). This superior efficacy was linked to Doxil's exploitation of the EPR effect, with biopsy analyses from phase I/II studies revealing concentrations in lesions 21 times higher than in adjacent normal , facilitating 5- to 11-fold greater intralesional drug levels compared to free . More recent trials have continued to evaluate EPR-mediated nanocarrier delivery, though with mixed outcomes highlighting variability in clinical translation. The phase II/III trial (NCT02379845) of hafnium oxide s (NBTXR3) in locally advanced , completed with results reported in 2020 and follow-up analyses through 2023, demonstrated improved pathological complete response rates of 16.7% when NBTXR3 was intratumorally injected and activated by radiotherapy, compared to 7.7% with radiotherapy alone, underscoring radiosensitization benefits potentially augmented by nanoparticle retention in permeable tumor vasculature. However, the U.S. approval of polymeric (Cynviloq) failed in 2018 due to manufacturing issues following its phase III bioequivalence trial against albumin-bound . General studies indicate EPR-mediated accumulation is often below 1% of the injected dose per gram of tumor tissue in xenografts and patient-derived models. Across these trials, EPR-targeted nanotherapeutics have generally shown 20-40% higher overall response rates compared to free-drug counterparts in responsive cohorts, such as in recurrent where liposomal doxorubicin yielded 19.7% vs. 16.1% for . Patient heterogeneity remains a key limiter, with imaging and data indicating variability in across tumor types and individuals, contributing to variable therapeutic outcomes and underscoring the need for patient selection strategies such as EPR imaging biomarkers.

Limitations and Controversies

Efficiency and Accumulation Challenges

Despite the promise of the enhanced permeability and retention (EPR) effect for targeted to tumors, quantitative analyses reveal persistently low efficiency in achieving therapeutic accumulation at the tumor site. A comprehensive of preclinical studies from 2005 to 2015 found that the median tumor was only 0.7% of the injected dose per gram of tumor tissue (%ID/g), with most nanoparticles failing to exceed 5% ID/g even under optimized conditions. In human applications, this translates to less than a 2-fold increase in to tumors compared to normal tissues, as seen in clinical data on nanomedicines like Doxil. A major challenge stems from substantial off-target accumulation in non-tumor organs, primarily driven by uptake from the (RES). A large fraction of systemically administered can accumulate in the liver, with up to 90-99% clearance observed, severely curtailing the fraction available for tumor targeting. This RES-mediated clearance not only reduces overall nanoparticle availability but also imposes risks in hepatic tissues, thereby limiting safe dose escalation and therapeutic indexing in clinical settings. Pharmacokinetic barriers further exacerbate inefficient accumulation by promoting rapid systemic clearance and impeding intratumoral transport. Non-PEGylated nanoparticles often exhibit half-lives shorter than due to opsonization and , drastically shortening the circulation time needed for EPR-mediated . Additionally, elevated interstitial fluid pressure (IFP) within tumors, often substantially higher than in normal tissues, compresses vessels and hinders from perivascular spaces into the tumor , resulting in heterogeneous distribution primarily near vessels. Recent meta-analyses, including those up to , underscore low performance overall, with delivery efficiencies around 0.7% /g, highlighting quantitative shortfalls in advanced disease settings. Tumor heterogeneity, as detailed elsewhere, contributes to these variations but does not fully account for the quantitative shortfalls observed.

Scientific Debates and Criticisms

Early critiques of the enhanced permeability and retention () effect emerged in the mid-2010s, questioning its reliability and overestimation in preclinical settings. In a 2014 review, Nichols and Bae argued that animal tumor models often exaggerate EPR due to differences in vascular distribution and blood flow compared to tumors, leading to inconsistent translation where drug carriers fail to outperform free s in clinical trials. They highlighted challenges such as high fluid pressure and irregular that undermine EPR-dependent , suggesting its application should be limited to susceptible tumor types. Similarly, Danhier's 2016 analysis emphasized that despite thousands of preclinical successes, the EPR effect largely fails in clinical contexts, prompting a reevaluation of strategies beyond passive tumor targeting. By 2024, controversies intensified with analyses of failed clinical trials attributing poor outcomes to the inconsistent presence of EPR in human tumors. For instance, the BIND-014 nanoparticle, designed for prostate-specific membrane antigen targeting, showed no superior efficacy over free in phase II trials, largely due to EPR variability where only about 50% of human tumors exhibit sufficient permeability for accumulation, unlike more uniform preclinical models. This sparked broader debates framing the EPR effect as a "myth" of universal passive targeting versus a "heterogeneous reality" influenced by tumor type, stage, and patient factors, with low nanocarrier accumulation (often <1% injected dose reaching tumors) underscoring these limitations. Such discussions highlighted how overreliance on EPR has contributed to nanomedicine's translational challenges. Alternative perspectives have proposed mechanisms beyond the classic EPR model, emphasizing active transvascular transport processes. A 2024 review in detailed how —via caveolae-mediated and —dominates delivery in many tumors, particularly those with low endothelial gaps, rather than passive leakage through vessel walls as traditionally described in EPR. This non-passive pathway, enhanced by ligand-receptor interactions or specific endothelial cell subsets, accounts for much of the observed tumor accumulation and explains EPR heterogeneity across species and tumor types. These views challenge the EPR paradigm by shifting focus to cellular machinery and barriers like the that limit . In response to these critiques, originator Hiroshi Maeda defended the EPR effect's foundational role while advocating for refined applications, particularly through patient selection for tumors with high EPR potential. In discussions published in 2022, Maeda stressed that EPR remains viable for but requires targeting tumors with elevated and reduced lymphatic drainage, achievable via or histopathological to improve clinical outcomes (noting Maeda's passing in 2021). This approach aims to address heterogeneity without dismissing EPR, emphasizing its utility in select solid tumors when combined with stromal barrier modulation. As of 2025, recent reviews continue to affirm the heterogeneous nature of the EPR effect and ongoing translational challenges in .

Strategies for Enhancement

Physiological Interventions

Physiological interventions aim to modulate the to enhance and reduce barriers to nanoparticle , thereby amplifying the enhanced permeability and retention () effect. These approaches leverage biological and thermal stimuli to temporarily alter tumor physiology, improving without relying on nanoparticle modifications. Key strategies include , paradoxical use of inhibitors, and specific pharmacological agents that target vascular and stromal components. Mild hyperthermia, typically applied at 40-43°C for 30-60 minutes, increases tumor vascular permeability by inducing vasodilation, enlarging endothelial gaps up to 10 μm, and upregulating nitric oxide (NO) and vascular endothelial growth factor (VEGF), which downregulate VE-cadherin junctions. This also lowers interstitial fluid pressure (IFP) immediately and for 24-48 hours post-treatment while disrupting the extracellular matrix to facilitate deeper nanoparticle penetration. Studies in animal models demonstrate a 2-4-fold increase in nanoparticle accumulation, such as a threefold enhancement in doxorubicin-loaded thermosensitive liposomes in sarcoma-bearing rats. Tumor blood flow improves by 15-250% in human cancers under these conditions, supporting broader clinical translation. Paradoxically, low-dose angiogenesis inhibitors like anti-VEGF agents (e.g., bevacizumab or cediranib) can normalize aberrant tumor vessels by pruning inefficient ones, reducing hypoxia, and decreasing IFP, which enhances perfusion and nanoparticle delivery via the EPR effect. Unlike high doses that exacerbate vessel collapse, low doses promote more uniform vessel structure, improving accumulation of nanoparticles sized 12-40 nm, as shown in preclinical breast cancer models where enzyme-responsive gold nanoparticles exhibited greater retention post-treatment. This normalization window, lasting days to weeks, balances permeability with improved blood flow for better therapeutic outcomes. Pharmacological agents further target tumor barriers to boost . analogs, such as RMP-7 (labradimil), activate B2 receptors on endothelial cells to widen intercellular clefts and increase pinocytotic activity, enhancing permeability for molecules up to 2 MDa and amplifying the effect in tumor models. For instance, RMP-7 improved delivery across the blood-tumor barrier in trials by up to twofold. Similarly, losartan, an , reduces IFP by approximately 50% through and hyaluronan degradation in the tumor stroma, decompressing vessels and enhancing drug in desmoplastic tumors like pancreatic and ovarian cancers. This leads to broader intratumoral distribution of agents like and , increasing efficacy in preclinical settings. Clinical examples illustrate these interventions' potential. In a phase I/II trial of neoadjuvant liposomal doxorubicin (Doxil) combined with and in locally advanced , the regimen achieved a 72% clinical response rate and 60% pathological response rate, enabling breast-conserving surgery in 37% of patients and correlating thermal dose with improved outcomes. These results suggest enhances Doxil accumulation, contributing to superior response compared to alone.

Technological and Formulation Advances

Recent advancements in from 2023 to 2025 have focused on stimuli-responsive nanoparticles (NPs) designed to address limitations in the enhanced permeability and retention (EPR) effect by enabling tumor-specific drug release. These NPs respond to endogenous cues, such as elevated levels, to trigger payload liberation precisely at the site of action, thereby improving therapeutic efficacy while minimizing off-target effects. For instance, enzyme-cleavable NPs, which degrade in response to matrix metalloproteinases overexpressed in tumors, have demonstrated enhanced drug penetration and release in preclinical models of solid tumors. A 2025 review highlights how these systems, including those responsive to and gradients, augment EPR-mediated accumulation by promoting deeper tissue distribution post-extravasation. Hybrid organic-inorganic NPs have emerged as a promising class of formulations between 2023 and 2025, combining the of organic components with the structural stability and multifunctionality of inorganic materials to enhance tumor beyond traditional reliance. These hybrids, such as those integrating polymeric matrices with silica or cores, exhibit improved mechanical properties that facilitate navigation through dense tumor , leading to superior intracellular delivery compared to purely or inorganic counterparts. In glioblastoma models, hybrid NPs have shown up to twofold greater depth due to their tunable surface charge and size, as reported in a 2024 study. Image-guided delivery techniques, particularly those employing ultrasound and focused beams, have advanced in 2025 reviews as methods to transiently enhance vascular permeability and EPR efficiency without permanent tissue disruption. Focused ultrasound (FUS) combined with microbubbles induces cavitation that temporarily opens the blood-brain barrier (BBB) for brain tumors or amplifies vascular leaks in peripheral solid tumors, allowing greater NP extravasation. For example, FUS-mediated BBB opening has been shown to increase NP delivery to glioma sites by facilitating EPR-like accumulation in otherwise impermeable regions. Surface engineering of NPs has seen significant progress in 2023-2025, incorporating active targeting ligands post-EPR accumulation to boost specificity and cell-mimetic coatings to evade (RES) clearance. Folate ligands conjugated to NP surfaces exploit overexpressed receptors on cancer cells, enhancing cellular uptake after initial EPR-driven tumor homing and improving therapeutic outcomes in ovarian and models. Complementarily, cell-mimetic coatings, such as erythrocyte or membranes, mimic host cells to reduce RES uptake, prolong circulation, and amplify EPR-mediated tumor delivery by up to 14-fold in blood retention studies. A notable example from involves Intralipid co-administration, a clinically approved lipid emulsion that modulates NP clearance by saturating RES macrophages, thereby increasing tumor delivery approximately threefold in preclinical tumor models. This approach enhances EPR utilization by improving NP circulation time and reducing hepatic sequestration, as demonstrated in studies on solid tumor xenografts.

References

  1. [1]
    A new concept for macromolecular therapeutics in cancer ... - PubMed
    Cancer Res. 1986 Dec;46(12 Pt 1):6387-92. Authors. Y Matsumura, H Maeda. PMID: 2946403. Abstract. We previously found that a polymer conjugated to the ...
  2. [2]
    The 35th Anniversary of the Discovery of EPR Effect - NIH
    Mar 22, 2021 · This Special Issue on the enhanced permeability and retention (EPR) effect commemorates the 35th anniversary of its discovery, the original 1986 ...
  3. [3]
    The Enhanced Permeability and Retention (EPR) Effect - PMC
    Aug 6, 2021 · The EPR effect occurs primarily in the peritumoral area [28]. Interstitial fluid pressure and solid stress provide the mechanism for the ...
  4. [4]
    Tumor vascular permeability and the EPR effect in macromolecular ...
    The phenomenon now known as the enhanced permeability and retention (EPR) effect for lipid and macromolecular agents has been observed to be universal in solid ...
  5. [5]
  6. [6]
    Nanodrug Delivery: Is the Enhanced Permeability and Retention ...
    The EPR effect improves nanosized drug accumulation in the extravascular space of tumors. In normal organs, circulating nanosized drugs are cleared ...
  7. [7]
    Beginning of angiogenesis research - Holland-Frei Cancer Medicine
    Because the limit of oxygen diffusion is 100 to 200 μm, tumor cells that ... The size limits of experimental tumors when angiogenesis is blocked or ...
  8. [8]
    MAGNETIC NANOPARTICLE HYPERTHERMIA IN CANCER ... - NIH
    This type of vascularization results in large (600–800 nm) fenestrations (gaps) between adjacent tumor endothelial cells. These fenestrations permit ...
  9. [9]
    The Enhanced Permeability and Retention (EPR) Effect - MDPI
    Aug 6, 2021 · This review highlights the significance of the concept and mechanism of the EPR effect and discusses methods for better utilizing the EPR effect.<|control11|><|separator|>
  10. [10]
    Challenges and Key Considerations of the Enhanced Permeability ...
    Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology.
  11. [11]
    Doxil®--the first FDA-approved nano-drug: lessons learned - PubMed
    Jun 10, 2012 · Due to the EPR effect, Doxil is "passively targeted" to tumors and its doxorubicin is released and becomes available to tumor cells by as yet ...
  12. [12]
    Photo-thermal tumor ablation in mice using NIR nanoparticles
    This technique takes advantage of the strong near infrared (NIR) absorption of nanoshells, a new class of gold nanoparticles with tunable optical absorptivities ...
  13. [13]
    Nanoshell-Enabled Photothermal Cancer Therapy - ACS Publications
    This property is referred to as the “enhanced permeability and retention (EPR) effect”. (12, 13) Without an appropriate surface coating, nanoparticles in the ...
  14. [14]
    Commemorative Issue in Honor of Professor Hiroshi Maeda - MDPI
    This momentous result led to discovery of the enhanced permeability and retention effect (EPR effect) in 1986. Later, the EPR effect became known worldwide ...Missing: approval | Show results with:approval<|separator|>
  15. [15]
    Perspectives for Improving the Tumor Targeting of Nanomedicine ...
    Jun 13, 2023 · Unlike the xenograft model, the EPR effect in existing human cancers is highly affected by tumor heterogeneity and the complexity of the tumor ...<|control11|><|separator|>
  16. [16]
    Physiologic upper limit of pore size in the blood-tumor barrier of ...
    Jun 23, 2009 · The physiologic upper limit of pore size in the BTB of malignant solid tumor microvasculature is approximately 12 nanometers.
  17. [17]
    Tumor Targeting via EPR: Strategies to Enhance Patient Responses
    By means of improved circulation times, nanomedicines can accumulate in tumors via the so called Enhanced Permeability and Retention (EPR) effect, which was ...
  18. [18]
    Tumor Interstitial Fluid Formation, Characterization, and ... - Frontiers
    Other factors contributing to the high tumor Pif would be intratumoral blood vessel compression due to solid stress due to growth (54), and direct effects of ...<|control11|><|separator|>
  19. [19]
    Overview of Methods for Overcoming Hindrance to Drug Delivery to ...
    ... tumor interstitial fluid pressure (TIFP), and a multifactorial increase in ... 10–40 mmHg) (60–62). As outlined by Jain, TIFP decreases from the center ...
  20. [20]
    Tumor vascular permeability and the EPR effect in macromolecular ...
    The phenomenon now known as the enhanced permeability and retention (EPR) effect for lipid and macromolecular agents has been observed to be universal in solid ...<|control11|><|separator|>
  21. [21]
    A Phase I and Pharmacological Study of the Platinum Polymer ...
    As a result of these characteristics, concentrations of polymer-drug conjugates in tumor tissue can reach 10–100 times higher levels than those produced after ...<|control11|><|separator|>
  22. [22]
    The Use of Alternative Strategies for Enhanced Nanoparticle ...
    Jan 14, 2021 · In the case of solid tumors, various factors such as leaky vasculature, angiogenesis, interstitial fluid pressure and lymphatic drainage are ...
  23. [23]
    Extracellular matrix remodeling in vivo for enhancing tumor-targeting ...
    Oct 10, 2017 · It has been reported that dense collagen and hyaluronan matrix might act as physical barrier hindering the access and deep penetration of ...Missing: trap | Show results with:trap
  24. [24]
    Impaired lymphatic function accelerates cancer growth - PMC
    An impaired lymphatic washout was observed from the peritumoral area in Chy mice with C3HBA tumors, and the number of macrophages was significantly reduced.
  25. [25]
    A Mathematical Model of the Enhanced Permeability and Retention ...
    The EPR effect describes the preferential accumulation of nanoparticles at tumor sites due to leaky vasculature (i.e. enhanced permeation) and impaired ...
  26. [26]
    Endothelial barrier disruption by VEGF-mediated Src activity ... - NIH
    Oct 25, 2004 · We demonstrate that VEGF-mediated Src kinase activity leads to a breakdown in endothelial barrier function which, in turn, hastens tumor cell extravasation and ...
  27. [27]
    VEGFR2 induces c-Src signaling and vascular permeability in vivo ...
    Jun 11, 2012 · Regulation of vascular endothelial (VE) growth factor (VEGF)–induced permeability is critical in physiological and pathological processes.
  28. [28]
    Roles of bradykinin in vascular permeability and angiogenesis in ...
    These results suggest that BK would promote angiogenesis by increasing vascular permeability in the early phase via B2 receptor in the endothelial cells.
  29. [29]
    Passive targeting of nanoparticles to cancer - Spandidos Publications
    Jul 23, 2014 · Therefore, bradykinin, a key factor controlling vascular permeability, is an important mediator controlling the EPR effect in neoplastic tissues ...
  30. [30]
    Enhancement of EPR Effect for Passive Tumor Targeting - MDPI
    The Enhanced Permeability and Retention (EPR) effect is a key mechanism for passive tumor targeting, which involves the selective accumulation of therapeutic ...
  31. [31]
    Prostaglandins and Inflammation | Arteriosclerosis, Thrombosis, and ...
    It has also been suggested that bradykinin induces PGI2 formation leading to enhancement of microvascular permeability and edema. The IP receptor has been shown ...
  32. [32]
    Vascular permeability enhancement in solid tumor: various factors ...
    Common denominators affecting both cancer and inflammatory lesions are now well known: bradykinin (BK), nitric oxide (NO), peroxynitrite (ONOO−), prostaglandins ...
  33. [33]
    Hypoxia-inducible factors: a central link between inflammation ... - JCI
    Aug 15, 2016 · Hypoxia and expression of HIF-1α and HIF-2α are characteristic features of all solid tumors. HIF signaling serves as a major adaptive mechanism ...
  34. [34]
    Matrix metalloproteinases in the brain and blood–brain barrier - PMC
    Inhibiting MMPs prevented the loss of tight junction proteins, indicating that MMPs disturb barrier integrity by degrading tight junction proteins. Thus, while ...
  35. [35]
    Targeting VEGF signaling for tumor microenvironment remodeling ...
    VEGF-A increases permeability mostly by upregulating eNOS and iNOS [29]. This increases NO production and relaxes vascular smooth muscle, which leads to blood ...
  36. [36]
    Analysis of Vascular Permeability by a Modified Miles Assay - PMC
    Factors increasing permeability include histamine, bradykinin, thrombin, and platelet-activating factor (PAF), among many others. For example, histamine and ...
  37. [37]
    Vascular Endothelial Growth Factor/Vascular Permeability Factor ...
    Accordingly, we used an in vivo assay described originally by Miles and Miles14 to study the putative mediators of VP induced by VEGF/VPF. This in vivo assay, ...
  38. [38]
  39. [39]
    Nanomedicine in cancer therapy | Signal Transduction and Targeted ...
    Aug 7, 2023 · The EPR effect has been found to vary substantially not only among patients, but also among tumor types, and even within the same patient or ...
  40. [40]
  41. [41]
  42. [42]
  43. [43]
    Phase II studies of polymer-doxorubicin (PK1, FCE28068 ... - PubMed
    These results show 6/62 PR with limited side effects, supporting the concept that polymer-bound therapeutics can have modified and improved anticancer ...Missing: EPR ovarian
  44. [44]
    Photo-thermal tumor ablation in mice using NIR nanoparticles
    Jun 25, 2004 · This study introduces a new laser-induced thermal therapy employing systemically delivered, NIR absorbing nanoparticles called nanoshells.
  45. [45]
    Phthalocyanine-nanoparticle conjugates for enhanced cancer ...
    It provides a comprehensive overview of key advancements from 2023 to 2025 in nanoparticle-mediated delivery strategies for phthalocyanine-based PSs in cancer ...Missing: effect | Show results with:effect
  46. [46]
    Gadolinium-loaded polychelating polymer-containing tumor ...
    A single nanoparticle of long-circulating (PEGylated) liposomes could provide a new contrast agent for highly specific and efficient tumor MRI.
  47. [47]
    Sorafenib and gadolinium co-loaded liposomes for drug delivery ...
    May 1, 2016 · Longer imaging time and higher signal enhancement was observed at the tumor tissue by SF/Gd-liposome. •. SF/Gd-liposomes allowed the MRI-guided ...
  48. [48]
    Theranostic liposomes as a bimodal carrier for magnetic resonance ...
    The present study is focused on the development of liposomes bearing gadolinium chelate (GdLip) providing two functionalities for magnetic resonance imaging ( ...
  49. [49]
    64Cu-MM-302 Positron Emission Tomography Quantifies Variability ...
    These findings provide important evidence and quantification of the EPR effect in human metastatic tumors, and support imaging nanoparticle deposition in tumors ...
  50. [50]
    Doxorubicin-loaded iron oxide nanoparticles for glioblastoma therapy
    Jul 9, 2020 · Tailored functionalization of iron oxide nanoparticles for MRI, drug delivery, magnetic separation and immobilization of biosubstances.
  51. [51]
    Theranostic MRI liposomes for magnetic targeting and ultrasound ...
    Jun 10, 2020 · A recent study reported the encapsulation of iron oxide nanoparticles and doxorubicin into a redox-responsive liposome structure for breast ...<|separator|>
  52. [52]
    Efficient uptake and retention of iron oxide-based nanoparticles in ...
    Jun 29, 2020 · Efficient uptake and retention of iron oxide-based nanoparticles in HeLa cells leads to an effective intracellular delivery of doxorubicin.
  53. [53]
    New strategies for tumor diagnosis and therapy with ultrasound ...
    Sep 5, 2025 · Ultrasound-guided delivery facilitated tumor accumulation via the EPR effect, with CEUS imaging confirming enhanced intratumoral contrast.
  54. [54]
    Image-guided strategies to improve drug delivery to tumors beyond ...
    Jul 14, 2025 · This review discusses strategies to refine and complement EPR-based drug delivery. One possibility is to better stratify patients for therapy ...
  55. [55]
    Magnitude of Enhanced Permeability and Retention Effect in Tumors ...
    This study investigated the influence of different tumor phenotypes on the EPR effect, hypothesizing that a baseline level of uptake must be exceeded.
  56. [56]
    A Comparative Analysis of Orthotopic and Subcutaneous Pancreatic ...
    The PE was more than 2.5 times higher for the subcutaneous tumours compared to orthotopic ones, indicating a greater vascular volume (Figure 7b, Table 2). In ...
  57. [57]
    Development of an Orthotopic Model of Invasive Pancreatic Cancer ...
    Using tumor cells obtained from mice with targeted Kras and p53 mutations, we have developed a rapid and predictable orthotopic model of invasive pancreatic ...Abstract · Materials and Methods · Results · Discussion
  58. [58]
    A Biomimetic Microfluidic Tumor Microenvironment Platform ... - Nature
    Aug 24, 2017 · EPR effect is one of the most widely used modalities for passive targeting of macromolecules to solid tumor, although the significance of the ...
  59. [59]
    Size-Dependent EPR Effect of Polymeric Nanoparticles on Tumor ...
    Passive targeting of large nanoparticles by the enhanced permeability and retention (EPR) effect is a crucial concept for solid tumor targeting in cancer ...2. Results · Figure 2 · Figure 3<|separator|>
  60. [60]
    Analysis of nanoparticle delivery to tumours - Nature
    Apr 26, 2016 · In this article, we explore the influence of delivery on the efficacy of nanoparticles for cancer-targeting applications. Delivery is important ...
  61. [61]
    Beyond the EPR effect: Intravital microscopy analysis of nanoparticle ...
    The EPR effect, first observed by Matsumura and Maeda in 1986, refers to the enhanced accumulation of NPs via the abnormal leaky vasculature of tumors and the ...Missing: initial | Show results with:initial
  62. [62]
    results of a randomized phase III clinical trial - PubMed
    We conducted a randomized phase III clinical trial to compare the efficacy and toxicities of a new form of therapy, pegylated-liposomal doxorubicin, with ...
  63. [63]
    Pegylated Liposomal Doxorubicin: Scientific Rationale and ...
    Oct 1, 1997 · Biopsies of Kaposi's sarcoma (KS) lesion tissue and adjacent normal skin were obtained in 22 patients (Table 2).[37] Doxorubicin levels in KS ...Missing: EPR | Show results with:EPR
  64. [64]
    Clinical Oncology Original Article
    In patients with advanced Kaposi's sarcoma, a single-dose study has shown intralesional doxorubicin levels are 5-11 times greater with Doxil ® than with ...<|control11|><|separator|>
  65. [65]
    NBTXR3, a first-in-class radioenhancer hafnium oxide ... - PubMed
    This phase 2-3 trial evaluated the safety and efficacy of the hafnium oxide (HfO 2 ) nanoparticle NBTXR3 activated by radiotherapy versus radiotherapy alone as ...Missing: 2023 EPR
  66. [66]
    Image-guided strategies to improve drug delivery to tumors beyond ...
    Passive drug targeting relies on the Enhanced Permeability and Retention (EPR) effect, which enables nanomedicines to passively accumulate in solid tumors due ...
  67. [67]
    Thirty years from FDA approval of pegylated liposomal doxorubicin ...
    Jan 9, 2025 · The enhancement of tumour growth and immunosuppressive effect related to drug-free liposomes have not been observed with PLD.
  68. [68]
    Investigating the EPR effect of nanomedicines in human renal ...
    Considerable EPR effect was observed in more than 87% of human renal tumors, which showed significant diversity and heterogeneity. Abstract. The enhanced ...Missing: strong | Show results with:strong
  69. [69]
    Nano-drug delivery: Is the enhanced permeability and retention ...
    While the EPR effect is widely held to improve delivery of nano-drugs to tumors, it in fact offers less than a 2-fold increase in nano-drug delivery.
  70. [70]
    Directing Nanoparticle Biodistribution through Evasion and ...
    Jan 10, 2018 · Up to 99% of systemically administered nanoparticles are cleared through the liver. Within the liver, most nanoparticles are thought to be ...A Zebrafish Model For... · Figure 1 · Materials And Methods
  71. [71]
    Tissue Distribution and Pharmacokinetics of Stable Polyacrylamide ...
    (2001) reported a very rapid elimination from the blood, with the half life ranging from 13 to 35 s, whereas pegylated PLGA (PLGA-mPEG) nanoparticles of 113 nm ...
  72. [72]
    Nanoparticle uptake in tumors is mediated by the interplay of ... - NIH
    Nanoparticle delivery into solid tumors is affected by vessel density, interstitial fluid pressure (IFP) and collagen, as shown here.
  73. [73]
    Meta-Analysis of Nanoparticle Distribution in Tumors and Major ...
    Oct 9, 2023 · (5,9) Wilhelm et al. (2016) (5) summarized the studies between 2005 and 2015 and calculated the median NP delivery efficiency of 0.7%ID. Cheng ...
  74. [74]
    Review EPR: Evidence and fallacy - ScienceDirect.com
    The enhanced permeability and retention (EPR) of nanoparticles in tumors has long stood as one of the fundamental principles of cancer drug delivery.Missing: effect critique
  75. [75]
    To exploit the tumor microenvironment: Since the EPR effect fails in ...
    The aim of this review is to provide a general overview on (i) the current state of the EPR effect, (ii) the future of nanomedicine and (iii) the strategies of ...Missing: irrelevance | Show results with:irrelevance
  76. [76]
    Clinical cancer nanomedicines - ScienceDirect.com
    Sep 10, 2025 · In this study, BIND-014 did not show enhanced therapeutic performance compared to free docetaxel. When discussing the failed trial, the ...
  77. [77]
    Transvascular transport of nanocarriers for tumor delivery - Nature
    Sep 17, 2024 · The heterogeneity between tumors and vessels affects the pathway of vascular extravasation of NCs. In 2023, a research work demonstrated that ...
  78. [78]
    35 years of discussions with Prof. Maeda on the EPR effect and ...
    Oct 13, 2025 · The phenomenon of enhanced permeation and retention (EPR) effect further boosted the case for clinical translation of cancer nanomedicine.
  79. [79]
    Augmentation of the EPR effect by mild hyperthermia to improve ...
    The HT-induced enhanced permeability is attributed to increasing the size of the endothelial gaps (up to 10 μm) after HT [166]. In addition, HT downregulates VE ...
  80. [80]
    Nanoparticle Delivery and Tumor Vascular Normalization - NIH
    Here we propose an overview of the nanodelivery approaches that target tumor vasculature, aiming to achieve vascular normalization.
  81. [81]
    Pharmacological Modulation of Blood–Brain Barrier Permeability by ...
    They are also potent enhancers of the desirable enhanced permeability and retention (EPR) effect in tumors [10,11,12]. Kinin effects are mediated through ...
  82. [82]
    Losartan treatment enhances chemotherapy efficacy and reduces ...
    Jan 18, 2019 · Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma.
  83. [83]
    A phase I/II study of neoadjuvant liposomal doxorubicin, paclitaxel ...
    Our study shows for the first time that neoadjuvant therapy with paclitaxel, liposomal doxorubicin and hyperthermia is a feasible and well tolerated treatment ...Missing: Doxil | Show results with:Doxil
  84. [84]
  85. [85]
    Advances in Nano-Drug Delivery for Tumor Microenvironment and ...
    Jul 21, 2025 · Among these, endogenous stimulus-responsive nanocarriers, designed to react to specific tumor-associated stimuli such as pH, redox gradients, ...
  86. [86]
    New insights into targeted therapy of glioblastoma using smart ...
    May 7, 2024 · Diverse inorganic, organic, and hybrid organic–inorganic nanoparticles, each possessing unique properties, have been introduced into this domain ...Routine Theranostic... · Drug Delivery Using Targeted... · Gold Nanoparticles (gnps Or...
  87. [87]
    Focused Ultrasound-Mediated Blood-Brain Barrier Opening and ...
    Sep 3, 2025 · We introduce a two-pronged strategy comprising focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening and long-circulating ...Missing: EPR | Show results with:EPR
  88. [88]
    Folate-engineered chitosan nanoparticles: next-generation ...
    Oct 31, 2024 · Chitosan nanoparticles (NPs) are well-recognized as promising vehicles for delivering anticancer drugs due to their distinctive characteristics.
  89. [89]
    Cell membrane biomimetic nanoplatforms: a new strategy for ...
    Sep 23, 2025 · Enhanced EPR effect; reduced uptake by liver Kupffer cells; decreased plasma protein adsorption, 24h blood retention rate up to 14.2 ...Missing: mimetic | Show results with:mimetic
  90. [90]
    Rediscovery of mononuclear phagocyte system blockade for ...
    May 22, 2024 · Rapid uptake of nanoparticles by mononuclear phagocyte system (MPS) significantly hampers their therapeutic efficacy.