Significance
Nanotechnology is a promising approach for improving cancer diagnosis and treatment with reduced side effects. A key question that has emerged is: What is the ideal nanoparticle size, shape, or surface chemistry for targeting tumors? Here, we show that tumor pathophysiology and volume can significantly impact nanoparticle targeting. This finding presents a paradigm shift in nanomedicine away from identifying and using a universal nanoparticle design for cancer detection and treatment. Rather, our results suggest that future clinicians will be capable of tailoring nanoparticle designs according to the patient's tumor characteristics. This concept of “personalized nanomedicine” was tested for detection of prostate tumors and was successfully demonstrated to improve nanoparticle targeting by over 50%.
Keywords: cancer, nanoparticles, targeting, nano–bio interactions, tumor
Abstract
Nanoparticles can provide significant improvements in the diagnosis and treatment of cancer. How nanoparticle size, shape, and surface chemistry can affect their accumulation, retention, and penetration in tumors remains heavily investigated, because such findings provide guiding principles for engineering optimal nanosystems for tumor targeting. Currently, the experimental focus has been on particle design and not the biological system. Here, we varied tumor volume to determine whether cancer pathophysiology can influence tumor accumulation and penetration of different sized nanoparticles. Monte Carlo simulations were also used to model the process of nanoparticle accumulation. We discovered that changes in pathophysiology associated with tumor volume can selectively change tumor uptake of nanoparticles of varying size. We further determine that nanoparticle retention within tumors depends on the frequency of interaction of particles with the perivascular extracellular matrix for smaller nanoparticles, whereas transport of larger nanomaterials is dominated by Brownian motion. These results reveal that nanoparticles can potentially be personalized according to a patient’s disease state to achieve optimal diagnostic and therapeutic outcomes.
Nanotechnology remains an emerging and important research discipline for detecting and treating cancer. Nanomaterials can be engineered with different sizes, shapes, and surface chemistries, as well as assembled into hierarchical nanosystems (1). Nanomaterials can also be engineered with unique properties such as emission of light for fluorescence detection (2), magnetism for magnetic resonance imaging (3, 4), and thermal emission for ablation of tumor cells (5). Despite the potential of nanomaterials, typically less than 5% of an administered dose reaches the tumor compartment (6) because of poor retention within the tumor space and uptake by the skin (7), spleen, and liver (8–10). Refinements to the size, shape, and surface chemistry of nanomaterials have improved their blood half-lives (11, 12) and interactions with cancer cells (13–15). Unfortunately, clinical translation of cancer nanomedicine remains stagnated by adherence to the ideology that nanoparticles and other agents can be designed to “universally” detect and treat tumors independent of type or stage of cancer progression. Tumor growth leads to physiological changes in their tissue composition (cell density, vascularity, necrosis, and stroma). If nanoparticles could be tailored according to the physiological state of each tumor, cancer detection and treatment may be drastically improved. However, investigations into the effect of tumor pathophysiology on nanoparticle accumulation and kinetics have been limited.
Fundamental analysis of tumor pathophysiology has identified unique cellular and structural properties associated with various stages of cancer progression. We currently understand that the increasing vascular tortuosity, inhomogeneity, and restricted blood flow (and subsequent low blood pressure) associated with tumor growth prevents chemotherapeutic agents from reaching their target. This impairment of drug delivery may lead to poor therapeutic efficacy and cancer recurrence (16, 17). As we learn more about the cellular, vascular, and compositional characteristics of tumors, it is increasingly evident that tailoring drug delivery vehicles to the physiological state of a tumor may be instrumental to improving treatment of this disease (18, 19). However, enabling clinicians to personalize patient care will require a deeper understanding of the implications of tumor anatomy and pathophysiology on the delivery and function of medicinal agents.
Here, we determine whether the delivery of spherical gold nanoparticles (AuNPs) can be affected by changes in tumor volume—a surrogate of cancer progression. Specifically, we (i) characterize the changes in the physiological structures and microenvironment of tumors as they grow in a tumor xenograft mouse model, and (ii) explore how such changes impact uptake, permeation, and retention of polyethylene glycol (PEG)-coated AuNPs. Understanding these variations will enable clinicians to personalize cancer therapy by catering nanotherapeutic regimens according to tumor characteristics. As a proof of concept, we successfully demonstrate that observable changes in tumor pathophysiology can be used in a decision matrix to rationally select AuNP designs according to desired function.
Results
Characterization of Tumors.
Pathophysiological changes associated with tumor volume were studied to identify biological parameters that might impact AuNP targeting. The degree of vascularization, cell density, and extracellular matrix (ECM) content of different-sized orthotopic human breast melanoma xenograft tumors derived from MDA-MB-435 cells in CD1 nude athymic mouse models were characterized. These parameters were selected because they have been shown to individually impact nanoparticle uptake rate, accumulation, and retention (20–22). Histological sections stained with CD31 antibodies were used to colorimetrically visualize tumor blood vessels, whereas Movat’s Pentachrome staining was performed to highlight nuclei and ECM components such as proteoglycans, mucopolysaccharides, and collagen. Vascular density was calculated by counting the number of vessels per tumor cross-section. We observed that the concentration of blood vessels increased with tumor volume but plateaued at 44 ± 3 blood vessels/mm2 for tumor volumes exceeding 1.0 cm3 (Fig. 1A). Interestingly, the tumor vasculature was only uniformly distributed in small tumors. Tumor blood vessels became increasingly concentrated near necrotic regions and at the tumor perimeter as tumors enlarged (SI Appendix, Fig. S1).
Fig. 1.
Summary of the pathophysiological changes in tumors during growth. The graphs depict the changes in vascular density (A), the proportion of tumors occupied by ECM components (B), acellular space (C), and cellular density (D) associated with tumor volume. All values were normalized to tumor cross-sectional area.
Beyond tumor vascularization, the fraction of the tumor composed of proteoglycans and mucopolysaccharides increased at a rate of 4.2 arbitrary units/cm3 (Fig. 1B), whereas tumor cell density increased at a rate of 1.70 cells/cm3 (Fig. 1C) with tumor volume. Unstained acellular space also proportionally decreased with tumor growth (Fig. 1D). These factors coincided with heightened ECM production at regions surrounding tumor blood vessels and necrotic tissue, whereas ECM content in regions of dense tumor tissue around the core became reduced (SI Appendix, Fig. S2). A closer examination of ECM composition by Picrosirius red staining (Fig. 2A) and second harmonic generation (SHG) imaging (Fig. 2B) identified that these regions contained type I collagen with a density and structure that evolved with tumor growth. Picrosirius red-stained samples spectrally shifted from deep red to pale pink (Fig. 2C), whereas SHG microscopy images decreased in intensity (Fig. 2D) as tumors enlarged. There is a decrease of 9% per cm3 in Picrosirius red intensity and the spectral shift in SHG peak intensity was characteristic of a loss in structural ECM via reduction in collagen fiber thickness and length (23–25).
Fig. 2.
Structural changes to type I collagen associated with tumor size. (A) Representative bright-field images of Picrosirius red-stained sections that depict the evolution of collagen with tumor size. (B) Representative SHG microscopy images of collagen (green) overlaid with DRAQ5-stained nuclei (blue). (C) Graph delineating how Picrosirius red intensity fades with rising tumor volume as collagen fibrils convert to lesser-organized constructs. (D) Histograms of SHG intensity in collagen-enriched zones validates that type I collagen becomes increasingly amorphous with tumor enlargement. (Scale bars, 50 µm.) DRAQ5 is 1,5-bis{[2-(di-methylamino) ethyl]amino}-4, 8-dihydroxyanthracene-9,10-dione.
Together, these results indicate that as tumors mature through growth, their tissue and vasculature become denser and more chaotic. In particular, the ECM appears to remodel during tumor enlargement, thus leading to a more amorphous phenotype. Given that ECM components were observed to encapsulate tumor blood vessels (SI Appendix, Fig. S3) and are known to biologically function as a basal support for blood vessels that interfaces with the stroma, changes in ECM may be a primary mediator of nanoparticle entry into the tumor compartment.
Gold Nanoparticle Model System.
Having characterized the evolution of tumor tissues during growth, we sought to determine whether these physiological changes could be used to tune the tumor targeting efficacy of nanoparticles. Because tumor uptake is dependent on nanoparticle diameter (12, 26, 27), a library of methoxy-PEG–coated AuNPs of varying diameter were designed to examine the effect of tumor growth on particle delivery. Although clinical trials for AuNPs are limited, AuNPs were selected over more clinically appropriate polymeric nanomaterials because AuNPs can be reproducibly and precisely synthesized in a broad range of sub-100-nm sizes. Furthermore, AuNPs provide a nondeformable formulation for testing the effect of core diameter on tumor uptake, are easily surface modified, and can be quantified in tissues with high sensitivity. A schematic illustrating the AuNP design used in this study is depicted in SI Appendix, Fig. S4A.
Spherical AuNPs with core diameters of 15, 30, 45, 60, and 100 nm (SI Appendix, Fig. S4B) were synthesized using standard citrate and hydroquinone reduction techniques (28). These sizes were selected to systematically characterize how the tumor microenvironment impacts a broad range of particle diameters. AuNP surfaces were modified with hetero-bifunctional 5-kDa PEG with methoxy and sulfhydryl termini as well as Alexa Fluor 750-labeled 10-kDa sulfhydryl-PEG to respectively stabilize particles for blood transport and to fluorescently track particles in vivo. Although it is difficult to use fluorescence as an absolute quantification technique, we have shown previously that fluorescence is an accurate modality for monitoring relative changes in nanoparticle biodistribution (26, 29). Surface modifications resulted in AuNPs with a PEG packing density of 0.3–1.5 ligands/nm2. At these densities, surface-bound PEG moieties were calculated according to their Flory diameter to be in the brush layer conformation, ensuring that the tested nanoparticles were sufficiently passivated (SI Appendix, Table S1). Surface modifications were also found to increase nanoparticle hydrodynamic diameters by 20–40 nm (SI Appendix, Fig. S4C) and positively shift nanoparticle zeta potentials by 20–30 mV (SI Appendix, Fig. S4D). Particle fluorescence was confirmed by the migration of distinct fluorescent bands during agarose gel electrophoresis (SI Appendix, Fig. S4E). AuNP fluorescence was shown to increase proportionally with particle diameter (SI Appendix, Fig. S5). Fluorescent PEG groups were also confirmed to be stably bound to particle surfaces because the rate of desorption in the presence of serum remained below 0.2 PEG molecules/h (SI Appendix, Fig. S5D). In vivo pharmacokinetics of our functionalized AuNPs was also characterized by analysis of blood plasma at 0, 2, 4, 8, and 24 h posttail vein injection (HPI) in non–tumor-bearing CD1 nude athymic mice. Inductively coupled plasma atomic emission spectroscopy (ICP-AES) analysis of blood samples revealed that the blood half-lives of our AuNPs ranged from 2 to 10 h. A complete characterization of our formulations is presented in SI Appendix, Table S2.
Analysis of Nanoparticle Accumulation in Tumors.
AuNP accumulation was evaluated via tail vein injection of formulations into CD1 nude athymic mice bearing orthotopic MDA-MB-435 human breast melanoma tumors. Tumors volumes evaluated in this study ranged from 0.05–3.00 cm3. AuNP delivery to the different sized tumors was fluorescently profiled in mice to assess tumor accumulation kinetics and to measure total AuNP exposure. Fluorescent tracking was achieved by whole-animal imaging using a Carestream In Vivo Imaging System at time points ranging from 0–24 HPI.
Total area under the curve (AUC) was calculated from the kinetic curves in SI Appendix, Fig. S6 as a metric for AuNP accumulation within the tumor. Overall, AUC values increased with tumor volume (Fig. 3A). Accumulation for 15-, 30-, and 45-nm AuNPs steadily increased with tumor volume from 490 ± 70% to 720 ± 30% ID⋅h, 280 ± 50% to 750 ± 10% ID⋅h, and 480 ± 70% to 960 ± 100% ID⋅h, respectively. Changes in accumulation of larger formulations occurred as step increases at discrete tumor volumes. There was an ∼1.5 times higher accumulation for 60-nm formulations once tumors exceeded 2.2 cm3, whereas 100-nm particles exhibited a ∼4.6 times increase in accumulation for volumes 0.5 cm3 and larger in comparison to smaller tumors. These trends were confirmed by ICP-AES measurements of gold content in tumors at 24 HPI (Fig. 3B). The ICP-AES results indicated that by 24 HPI tumor uptake of 15- and 30-nm particles were consistently higher than all other formulations and steadily increased from 0.39 ± 0.04% to 0.99 ± 0.18% ID and 0.28 ± 0.03% to 0.90 ± 0.18% ID, respectively (two-way ANOVA, P = 0.05), whereas larger particles such as 60 nm trended higher (although statistically not significant) from 0.18 ± 0.02% to 0.26 ± 0.12% ID as tumor volumes increased.
Fig. 3.
Results delineating how tumor uptake of AuNPs varies with tumor volume. (A) Bar graph of calculated AUC measurements for AuNP uptake by tumors. The yellow dotted line denotes our defined successful accumulation threshold. Overall, AuNP accumulation increases with tumor volume. (B) Total AuNP content in tumors of different volumes as measured by ICP-AES at 24 HPI. Results were normalized to injection dose per gram of tumor. (C) Bar graph summarizing how the speed of AuNP uptake varies with increasing tumor volume and particle diameter. Uptake rates remain constant for tumor volumes above 0.5 cm3 apart for 45-nm AuNPs. Error bars denote SE of mean values (n > 3). Asterisks denote statistically significant data (two-way ANOVA, P = 0.05).
In combination with our histological observations, these results suggest that the higher porosity of the ECM increasingly accommodates the entry of larger nanoparticles at later stages of tumor growth. This implies that a minimum tumor size must be reached to support entry of each AuNP diameter. An AuNP accumulation threshold of 500% was selected to illustrate this point (Fig. 3A). This threshold was defined as the mean AUC of 15-nm AuNPs in sub-0.5-cm3 tumors as particles in this size range would experience the least steric hindrance. AUC values for each AuNP diameter were statistically compared with the threshold (two-way ANOVA, P = 0.05); 15-nm AuNPs have this accumulation threshold at tumor volumes of 0.5 cm3 and larger, whereas 30-nm nanoparticles achieved a similar trend at a threshold of 0.5–1.0 cm3 and above. Similarly, 45-nm formulations attained statistically higher accumulation at tumor volumes above 1.0 cm3, and 60-nm AuNPs exceeded this threshold (although statistically insignificant) when tumor volumes were beyond 2.2 cm3; 100-nm particles never reached the defined threshold accumulation at any of the tumor volumes tested. It has been shown that AuNPs greater than 100 nm in diameter sequester near tumor blood vessels and do not penetrate into MDA-MB-435 tumors (26, 27). Hence, the difference in the accumulation pattern of 100-nm AuNPs over the other tested formulations was attributed to the inability of these AuNPs to diffuse through pores that are smaller than the particle size.
Nanoparticle Kinetics Within the Different-Sized Tumors.
Kinetics of AuNP delivery to tumors were analyzed in an effort to explain the dependence between accumulation and tumor volume. Tumor uptake rates were calculated by taking the instantaneous slope at 3 HPI of the AuNP accumulation profiles presented in SI Appendix, Fig. S6. We observed that the speed of AuNP accumulation (Fig. 3C) was largely insensitive to changes in tumor volume (two-way ANOVA, P = 0.05); 15-, 60-, and 100-nm AuNPs maintained tumor entry rates of 4.2 ± 0.6%, 3.2 ± 0.9%, and 2.9 ± 0.8% ID⋅h−1 as tumors grew to 1.0 cm3. Particles with 30- and 45-nm diameters were the exception as their rate of uptake steadily rose from 2.3 ± 0.3% to 5.7 ± 0.9% ID⋅h−1 and 2.8 ± 0.9 to 7.0 ± 1.0% ID⋅h−1, respectively, as tumors grew beyond 0.5 cm3. Although the rate of delivery did not statistically vary with growth, AuNP entry into the tumor compartment trended higher as tumors increased in size. The 15-, 30-, and 45-nm AuNPs also consistently accumulated in tumors ∼1.2–1.7 times faster than the 60- and 100-nm formulations. However, these differences became less apparent as tumor volumes increased. These results further reinforce the relationship between ECM porosity and particle size whereby smaller pores restrict larger nanoparticles from deep tumor infiltration and conversely become washed out of the tumor at a faster rate than smaller nanomaterials.
Because it is difficult to probe nanoparticle transport through ECM in animal models, we developed an in vitro system to measure diffusion of AuNPs into a hydrogel to mimic the effects of collagen structure on the transport of nanoparticles into the tumor (Fig. 4A). Although this in vitro model only evaluates diffusion through a collagen matrix independent of fluid flow or cellular interactions, the model provides a means to determine how the velocity of transport and quantity of AuNPs within tumors are dictated by the perivascular stroma upon initial AuNP entry. Self-assembled hydrogels composed of either 2.5 or 4.0 mg/mL of type I collagen were used to mimic stromal changes caused by tumor growth. Type I collagen was selected as a stromal phantom because type I collagen is the primary component of the tumor–blood vessel interface (30, 31). Entry of AuNPs from a fluid reservoir into the hydrogel was kinetically monitored by AuNP fluorescence using scanning confocal microscopy at different time points over 900 min. Overall, AuNP transport into the collagen gel occurred in two phases: (i) rapid concentration at the periphery of the hydrogel; and (ii) gradual movement from the concentrated zone to deeper regions of the matrix (Fig. 4B).
Fig. 4.
In vitro collagen hydrogel model of AuNP transport through tumor ECM. (A) Schematic depicting the in vitro setup used to profile AuNP infiltration into type I collagen hydrogels. (B) Illustration of the observed AuNP (red) infiltration process for the collagen hydrogels (green). AuNPs first concentrate at the gel–reservoir interface dependent on particle size and collagen density. Once an equilibrium is reached between AuNPs in the matrix and interface, the AuNP front gradually diffuses deeper into the hydrogel. (C) Bar graph depicting the permeation of AuNPs within the collagen hydrogels at 900 min postexposure. (D) Whisker plot depicting the cumulative results of AuNP penetration from blood vessels into tumor tissues at 24 HPI. No differences were found between tumor sizes. Bar graphs (E and F) summarize the differences in AuNP entry and exit from hydrogels based on collagen density and AuNP diameter. Error bars denote SEM for n = 3. Asterisks denote statistically significant data (two-way ANOVA, P = 0.05).
The amount of AuNPs infiltrating the hydrogel plateaued within 120–240 min postexposure for all formulations greater than 45 and 15 nm for 2.5 and 4.0 mg/mL collagen hydrogels, respectively (SI Appendix, Fig. S7A). The AuNP diffusion front also plateaued by 480 min postexposure for all particle diameters independent of collagen density (SI Appendix, Fig. S7B). Rather, AuNP penetration into the hydrogel at later time points occurred by diffusing away from the concentrated zone into the surrounding gel (seen as a broadening of the diffusion front in SI Appendix, Fig. S8). This penetration was dictated by particle diameter; 45-nm AuNPs achieved the highest permeation at 17.0 ± 2.0 and 13.2 ± 0.4 µm, whereas 100-nm AuNPs exhibited the poorest penetration at 8.0 ± 1.0 and 5.4 ± 0.6 µm for collagen densities of 2.5 and 4.0 mg/mL, respectively (Fig. 4C). Although AuNP permeation appeared to decrease with collagen concentration, differences were not statistically significant (two-way ANOVA, P > 0.05). These trends were consistent with the tumor-permeation results at 24 HPI, where AuNP infiltration did not vary with tumor volume (Fig. 4D). Particle permeation also did not change statistically between the tumor periphery, regions neighboring necrotic zones, or within the core of the tumor tissue. Despite the lower collagen density, diffusion of 15-nm AuNPs into the 2.5 mg/mL collagen gels was unexpectedly 2.0 and 1.3 times lower than our 45-nm formulation in vitro and in vivo, respectively. These differences in diffusion were similar to previous studies (26, 27) and were attributed to the speed of AuNP uptake by and expulsion from the collagen matrix (Fig. 4 E and F). In comparison to 4.0 mg/mL collagen gels, 15-nm formulations were taken up by the 2.5 mg/mL gels 14% slower and expelled 51% faster. This result leads to a lower overall AuNP concentration within the 2.5 mg/mL collagen gel and, accordingly, slower particle diffusion. Alternatively, because the uptake rate of AuNPs exceeding 45 nm does not vary with collagen density (two-way ANOVA, P = 0.05), their slower depletion from the collagen matrix allows for greater nanoparticle retention and consequently greater infiltration distances.
Computational Modeling of Nanoparticle Diffusion Through Porous Matrices.
To help understand how nanoparticles interact with the collagen matrix, Monte Carlo numerical simulations of AuNP diffusion through collagen matrices were conducted; 2D models were used to examine the frequency of AuNP collisions with collagen fibers within pores of the hydrogel matrix. The frequency of such collisions can influence the retention and path of the AuNPs in the tumor; 3D simulations were also conducted to compare AuNP permeation capacity through stroma with different collagen densities. These computational models were conducted in Matlab using custom algorithms to simulate AuNP interaction and diffusion within collagen matrices. These simulations followed similar strategies used by Stylianopoulos et al. (32). AuNP motility was modeled as step-wise random walk obeying Einstein–Stokes diffusion (Eq. 2), whereas particle–fiber interactions were modeled as elastic collisions. Fig. 5A provides an illustration delineating the path of AuNP motion within a collagen pore. Obeying Brownian motion, AuNPs move randomly and can collide with collagen fibers. For our 3D simulations, collagen fibers were approximated as cylinders with radii between 0.05 and 0.50 µm. Representative images of the collagen matrices of varying collagen density simulated in Matlab are presented in Fig. 5B. The modeled radii were chosen according to measured thicknesses from scanning electron microscopy images of our 2.5 and 4.0 mg/mL collagen hydrogels (SI Appendix, Fig. S9). A detailed summary of our model and its underlying assumptions can be found in Methods.
Fig. 5.
Monte Carlo models simulating the dynamics of AuNP transport through and interactions with collagen matrices. (A) Pictorial representation of AuNP random walk in two dimensions within collagen pores. Number of collisions with the pore was tracked as a measure of AuNP interactions with collagen matrices. (B) Representative images of simulated hydrogels of varying collagen densities in three dimensions. Images were rendered in Matlab using the same algorithms used for assessment of AuNP diffusion through collagen matrices in three dimensions. (C) Bar graph comparing the rate of AuNP collisions with collagen matrices of varying pore size obtained from 2D simulations. Collision frequency decreases with increasing pore and AuNP size. Asterisks denotes the scenario whereby AuNP size exceeded the dimensions of the pore. (D) Line graph depicting the simulated changes to AuNP-diffusion rate in collagen gels as collagen density increases. Collagen density did not appear to impact AuNP diffusivity but was instead dictated by AuNP size.
AuNP movement in our 2D models for 1,000-particle replicates was simulated in 0.005-, 0.020-, 0.108-, and 0.640-µm2 square stromal pores for 10,000 steps at 0.1-s intervals. Our simulations determined that AuNP–fiber collision rates increased with reducing pore size and decreasing AuNP diameter (Fig. 5C); 15-nm AuNPs achieved the highest frequency of interaction with collagen fibers at rates between 0.038–0.023 collisions/s (cps), whereas 100-nm formulations ranged from 0.016–0.001 cps for pore sizes between 0.005–0.640 µm2. Interestingly, collision rates for 15-, 45-, and 60-nm AuNPs were statistically similar for 0.005-µm2 pores (ANOVA, P = 0.05) but became increasingly dissimilar as pores enlarged. These simulations suggest that impact of particle size on Brownian motion is a primary mediator of AuNP motility within the hydrogel over its frequency of collision with the ECM. This result suggests that the greater the nanoparticles interact with collagen, the longer they will be retained within the tumor.
Expanding on these results, AuNP diffusion was also modeled in three dimensions to compare how AuNP diameter and collagen density might impact stromal accumulation and infiltration. Stromal–ECM of increasing collagen density was modeled computationally as 27,000-µm3 cubes containing anisotropically oriented collagen fibers. The number of fibers were chosen to achieve collagen volume fractions (8.72–87.20%), reflective of conditions found within tumors (33, 34). Diffusion distance for 500 AuNP replicates was tracked for 5,000 discrete steps at 1-s intervals. Our simulations indicate that diffusion rates changed with AuNP diameter but did not change with collagen density (Fig. 5D); 15-nm AuNPs exhibited the greatest mobility at 1.95 ± 0.03 nm/s in the simulated hydrogels, whereas 45-, 60-, and 100-nm particles diffused at rates of 0.78 ± 0.01, 0.60 ± 0.01, and 0.36 ± 0.01 nm/s, respectively. These findings support our AuNP-permeation observations from histological tumor sections whereby AuNP diffusion away from blood vessels (SI Appendix, Fig. S10) did not vary with tumor volume (Fig. 5D).
Together, these 2D and 3D models elucidate how the stromal matrix is implicated in particle permeation. Although these simplified 2D and 3D models ignore the effect of fluid flow, oncotic pressure, and inelastic collagen–AuNP interactions, the models provide a mechanism for our in vitro and in vivo permeation observations. They suggest that AuNP permeation is the balance between the effects of particle size on Brownian motion and the frequency of particle collision with the ECM. The increased mobility of smaller AuNPs afforded greater diffusion but was also inhibitory because of the higher frequency of collision with the ECM. Conversely, AuNPs of larger diameters exhibited slower motion but also a lower propensity to interact with the stroma. The volume fractions tested and simulated in this study equate to ECM pore sizes ranging from 0.45 to 1.74 µm. Because these pores exceed the size of our AuNPs, differences in diffusivity associated with collagen density would be negligible for all tested particle diameters. Extended further, these computational findings demonstrate that AuNP transport within the tumor can be distorted through collisions with ECM fibers. These collisions can limit retention within the tumor compartment if AuNP volume approaches the porosity of the stromal ECM.
Nanoparticle Selection According to Tumor Maturity.
Given the complex dependence of tumor AuNP uptake on both particle size and tumor pathophysiology, we asked whether there was a means to rationally select AuNP formulations according to tumor volume. In our proof-of-concept work, we evaluated whether a decision matrix could be used to select nanomaterials for either tumor detection (diagnostic) or drug delivery (treatment). AuNP formulations with rapid delivery and high tumor contrast were defined as effective probes for delineating tumors, whereas AuNPs capable of high tumor retention and homogeneous tissue distribution were anticipated to fare well as drug delivery vehicles.
Relative measurements of AuNP fluorescence in vitro and in vivo were used as an estimate of tumor contrast achievable by each formulation. Surface area-to-volume ratios were also calculated to approximate the drug-loading capacity of each AuNP size (SI Appendix, Fig. S5C). These parameters in conjunction with tumor accumulation, uptake rate, and penetration capacity were ranked from best (4) to worst (1) for each of the four AuNP sizes used in our experiments. Each parameter was also given a multiplier according to the parameter’s importance to a given AuNP function. The weighted sum of these rankings was then calculated for each AuNP design for the different tumor size ranges. Eq. 1 is a summary of the scoring scheme, where µ is the importance multiplier, β represents the ranking factor, and i denotes the ranked AuNP parameters. Fig. 6B highlights these parameters and the associated values used to calculate the scores found in our decision matrices (Fig. 6 A and B).
| [1] | 
Overall, smaller (<45-nm) AuNPs were favored for both diagnostic and therapeutic applications across all tumor sizes. Diameters in the 100-nm range were consistently predicted as poor candidates for either application, whereas 15- and 45-nm particles were both expected to be useful for detection and treatment of large (>1.0-cm3) tumors. AuNPs in the 60-nm range were the exception to these trends because these AuNPs were predicted to be better for detection of small, early-stage tumors (<0.5 cm3). This exception for 60-nm nanoparticles was empirically attributed to the statistical similarity in AUC values for particles with diameters between 15 and 60 nm (Fig. 3A) as well as the higher tumor contrast seen for 60-nm particles (SI Appendix, Fig. S5) in the 0.0- to 0.5-cm3 range. Because macrophage uptake of nanoparticles increases with particle diameter (35), the enhanced utility of 60-nm AuNPs may also be related to changes in phagocytic capacity of tumor-associated macrophages (36) because the macrophages’ phenotypes evolve during tumor progression (37).
Fig. 6.
Proposed method of selecting AuNPs according to tumor maturity. (A) Pseudocolored heat maps qualitatively depict the utility of each AuNP diameter for therapeutic (Left) and diagnostic (Right) applications predicted by our proposed decision matrices. Rankings for particle utility for a given tumor volume have been rated from high (red) to low (green). Tabular values were calculated by taking the weighted sum of empirically ranked tumor accumulation potential, uptake rate, contrast, and permeation data according to AuNP diameter and tumor size. (B) Weighted importance (µ) of decision matrix parameters for application of nanoparticles to tumor diagnosis and treatment. (C) Flow diagram illustrating a proposed method of personalizing AuNP selection in the clinic for cancer detection and treatment.
These results imply that passively targeted AuNPs with smaller diameters would be more applicable for detection and drug delivery when tumor size is unknown. However, 45-nm AuNPs may be the more effective vehicle for later-staged tumors because their larger surface area-to-volume ratio (SI Appendix, Fig. S5C) theoretically allows for 900% greater drug loading than 15-nm particles with merely a drop to tumor accumulation by less than 57.3%. Although these findings are specific to passively targeted AuNPs, the proposed decision matrix schema can be generalized to provide a systematic method for assessing other particle types. A flowchart detailing a potential means of implementing this strategy is outlined in Fig. 6C.
Validation of the Decision Matrix for Personalized Targeting of Prostate Tumors.
Toward validating our results, we evaluated whether our formulated decision matrices could be used to predict the ideal AuNP design for other tumor models. A blinded study was conducted in CD1 nude athymic mice bearing orthotopic human tumors with PC3 prostate cancer cells to verify whether our tumor-size dependent predictions were accurate; 15- and 100-nm AuNPs were tail vein-injected into tumor-bearing mice to evaluate AuNP efficacy for tumor detection and accumulation. Both particle designs were effective at delineating the location of the tumor (Fig. 7A) but at varying efficacies. Tumor detection speed and contrast for 15-nm AuNPs were, respectively, 53.7% and 50.8% higher than 100-nm particles; 15 nm achieved greater tumor accumulation than 100-nm designs and trended higher with increasing tumor size (Fig. 7 C–E). These findings were consistent with our decision matrix, alluding to the potential of our system for use on other particle formulations and tumor types.
Fig. 7.
Blinded study assessing passive AuNP targeting of prostate tumors. (A) Whole-animal fluorescent images of mice bearing orthotopic prostate tumors. Bright regions highlight areas of AuNP accumulation. (B) Magnetic resonance images used to confirm the presence and size of prostate tumors in mice. Dotted circles demarcate the location of the tumor. Graphs C, D, and E, respectively, compare the ICP-AES measured accumulation, tumor uptake rates, and tumor contrast of 15- and 100-nm AuNPs in small and large tumors. Error bars in all graphs denote SE mean values for n = 3. Asterisks denote statistically significant data (two-way ANOVA, P = 0.05).
Discussion
Given the observed limitations of AuNP accumulation in tumors, it is clear that careful design of nanomaterials is necessary to achieve optimal tumor delivery. Currently, interaction of nanoparticles with the hepatic and renal systems can be manipulated by the nanoparticle's size, shape, and surface chemistry (38). However, to engineer an optimal nanoparticle delivery system for cancer targeting is more complicated and one needs to balance the particular function (i.e., payload and signal intensity), tumor interaction, and the tumor-competing organs for nanoparticle sequestration. Unfortunately, optimization of only the physicochemical properties of nanomaterials has reached an impasse whereby tumor targeting efficiency remains stagnated at 5% (6, 39). In our work, we have alternatively approached tumor delivery from the biological perspective by characterizing the unique physiological changes that occur during tumor growth to tailor nanoparticles.
We determined that for MDA-MB-435 orthotopic human tumor xenografts, malignant tissues become more disordered as the tumors increase in volume. Starting from homogeneously vascularized tissues with minimal necrotic space, tumors transition toward higher cell densities with vasculature that concentrates at sparsely distributed regions. This disproportionate vascularization coincides with an increase in necrotic tissue and expression of collagen and other ECM components that surround tumor blood vessels. Type I collagen in the tumor ECM was found to convert from long filamentous fibers to shorter and more amorphous structures as tumors increase in volume. Through use of an in vitro collagen hydrogel model, we rationalized that these structural changes in the ECM are a primary mediator of passive tumor delivery of spherical AuNPs. This collagenous basal membrane acts as a “sponge” for extravasating AuNPs but can delay particle infiltration. The densely packed ECM of early-stage tumors appears to sterically restrict AuNP entry based on particle diameter. In larger tumors, the more porous and less rigid structure of type I collagen facilitates entry of larger AuNPs and enhances accumulation of smaller particles by the stroma. Because AuNP infiltration depth did not change with tumor size in vivo nor with variations to collagen density in vitro, bulk tumor accumulation of particles likely depends (i) on the capacity of ECM to interact with AuNPs and (ii) on the number of blood vessels available for AuNP entry for a given tumor volume.
These tumor growth-associated changes highlight physiological parameters that can be exploited for selecting and designing AuNPs for tumor targeting. The reduction of available interstitial volume and enhanced porosity of stroma caused by tumor growth hinder permeation of AuNPs but allow for higher AuNP extravasation into the tumor space. This finding suggests that large AuNPs become more effective when tumors mature but this improvement in accumulation comes at the expense of deep tissue permeation. While small AuNPs may have deeper penetration they may not be ideal in many applications. For example, early diagnosis and treatment of cancer is associated with increased patient survival (40–42). Unfortunately, smaller AuNPs, which are best suited to target low-volume tumors, may be less effective drug-delivery vehicles because the payloads of these AuNPs may be smaller than larger particles. This possibility illustrates the dichotomy of AuNP design for cancer targeting application, because a tradeoff must be made between the intended function of a nanomaterial and optimal tumor delivery.
Toward personalized medicine, a simplified decision matrix was developed to illustrate a means of personalizing the selection of AuNPs according to tumor stage and desired AuNP function. Our proof-of-concept decision matrix facilitates the personalization of a nanomaterial according to the patient by providing an unbiased score of how well a formulation might fare based on tumor volume and the AuNP’s design parameters: tumor signal (fluorescence), accumulation, uptake rate, and permeation. Fig. 6C presents a flowchart illustrating how such a decision matrix might be used clinically to select nanotherapeutic regimens.
Simulations established that for MDA-MB-435 tumors, passively targeted AuNPs with 60-nm diameters provide the best contrast for detecting early stages of tumor growth and sites of metastasis. Alternatively, particles in the 15- to 45-nm range appear to be more effective for diagnostics because tumors increase in size or in situations where tumor maturity and phenotype are unknown. For therapeutic regimens, our work also identifies that AuNPs with diameters between 15 and 45 nm are best used for tumors exceeding 1.0 cm3, because these AuNPs’ permeation distances exceed 60- to 100-nm AuNPs despite having lower loading capacities. These results imply that AuNPs must be rationally designed according to the intended function. Formulations optimized for diagnostic applications may not necessarily be effective designs for drug delivery or vice versa. Although we have shown that our AuNP tumor size trends were also valid for a PC3 prostate tumor model, our results may not necessarily be generalizable to all tumor types because the decision matrix presented here was constructed from a single tumor type and nanoparticle design. However, because our proposed decision matrix uses phenotypic parameters that are common to malignant tissues, the proposed strategy could be easily adopted by pathologists and researchers. With a concerted effort among researchers to elucidate how different nanoparticle physicochemical properties and microarchitectures of different tumor models and host species impact nanoparticle entry and retention within tumors, a generalized decision matrix may be realized. Production of this large database may allow future clinicians to use standard magnetic resonance, computer tomographic, and histological imaging techniques to landmark and approximate the size of a tumor, which will guide the specifications for designing the nanoparticles. This approach would cater the design of nanomedicine to the patient.
Conclusions
To improve cancer detection and therapy, researchers are now investigating how the physicochemical properties of a nanomaterial mediate nanoparticle transport and function. Although it is clear that the synthetic properties of the nanoparticle are critical to the nanoparticles’ biological interactions, how the physiological characteristics of the tumor impact nanoparticle fate remains largely unexplored. Here, we show that tumor biology is equally important as nanoparticle size in dictating nanoparticle targeting efficacy. We further show that a thorough assessment of tumor composition can be used to develop a simple algorithm for rational selection of AuNPs according to cancer stage. Implementation of nanomaterials in tandem with radiological imaging and tissue biopsies may be clinically useful to optimally detect nascent tumors and personalize therapeutic regimens. However, realization of this personalized approach to cancer nanomedicine will require a greater understanding of the physical changes in the tumor microenvironment associated with cancer progression and the microenvironment’s implications on nanoparticle function.
The conclusions presented here have been formulated with passively targeted AuNPs using an orthotopic MDA-MB-435 tumor model. Although we successfully demonstrate that our proposed decision matrix can predict AuNP targeting efficacy for orthotopic prostate tumors, ascertaining how tumor growth can affect malignant tissues in other tumor models remains critical to ensure that animal and nano-based research can be translated to humans. It would also be prudent to study how other nanoparticle types and targeting schemes may change nanoparticle interactions with the host and tumor microenvironment. For example, analysis of how tumor pathophysiology influences active targeting may help to explain why the decoration of biorecognition molecules on nanoparticle surfaces appear to only enhance tumor targeting for nanoparticles within the 60-nm range (26). Further investigation on such topics will broaden our understanding of nano–bio interactions and allow for the development of a fundamental framework for design of cancer-centric nanomaterials. Nevertheless, our results illustrate that tumor maturity is a critical parameter that both impacts the fate of a nanomaterial and can be exploited to rationally design better diagnostic probes and therapeutic vehicles in the future.
Methods
Animal-handling protocols were approved by the Faculty of Medicine and Pharmacy Animal Care Committee, University of Toronto.
Tumor Accumulation Measurements.
Efficiency of AuNP delivery to tumors was measured by ICP-AES. Tumors were harvested at 24 HPI and digested in 1 mL of aqua regia (1:3 vol/vol nitric acid to hydrochloric acid) supplemented with 1 µg/mL yttrium for 2 h at 70 °C. Yttrium was used as an internal reference to account for sample loss during the digestion and purification process. Postdigestion, acidic solutions were diluted with 2 mL of double-distilled water and filtered through 0.22-µm PVDF membranes to remove undigested tissue. Volumes of the digested samples were then adjusted to achieve a final volume of 4 mL via addition of double-distilled water. Gold and yttrium contents in each sample were measured using a Perkin-Elmer Optima 3000. AuNP accumulation in tumors at 24 HPI was determined by normalizing measured gold concentrations to yttrium content and tumor mass.
Analysis of Nanoparticle Infiltration into Collagen Matrices.
Synthesized nanoparticles were tested in vitro for their permeation capacity through type I collagen hydrogels. Self-assembled hydrogels were first prepared by mixing presolubilized rat tail type I collagen on ice with 10× PBS and 1 M sodium bicarbonate at an 8:1:1 volumetric ratio, followed by dilution with double-distilled water to achieve final collagen concentrations of 2.5 and 4.0 mg/mL collagen solutions that were then placed into gel molds and allowed to self-assemble at 37 °C for 3 h. Postpolymerization, hydrogels were equilibrated in double-distilled water for 2 h, followed by immediate water exchange and introduction of AuNPs. AuNP infiltration into hydrogels was monitored every 30 min for 15 h via laser-scanning confocal microscopy using an Olympus Fluoview FV1000. A transillumination lamp was used to determine the collagen edge, whereas differential interference contrast (DIC) and fluorescence were invoked to profile AuNP distribution within the hydrogel. AuNP permeation was profiled along the length of the collagen hydrogel by analyzing confocal images of AuNP fluorescence in ImageJ. Fluorescent-intensity profiles were then placed into GraphPad Prism to calculate total AuNP uptake and track the mean AuNP-infiltration distances. Calculated values were used to determine AuNP-accumulation rates for the different hydrogel densities by taking the slope of the linear regression curves seen in SI Appendix, Fig. S11A.
Analysis of Nanoparticle Expulsion from Collagen Matrices.
Collagen hydrogels were constructed using a similar pH-based self-assembly process as mentioned in Analysis of Nanoparticle Infiltration into Collagen Matrices. Before gelation, AuNPs equivalent to a total surface area of 30 cm2 were thoroughly mixed with hydrogel solutions on ice. AuNP–collagen mixtures were then allowed to set overnight at 37 °C, rinsed with PBS, and suspended in 1 mL of PBS. At 0, 1, 2, 3, 5, 6, 8, and 24 h, the PBS suspension solution was sampled (90 µL) to track AuNP expulsion from the hydrogels. AuNP quantity in sample solutions was approximated by measurement of sample fluorescence in 384 fluorescent well plates (Nunc 384-well optical well plates) using a Carestream Multispectral MS Fx Pro in vivo imager (excitation/emission: 750/830 nm) at an exposure time of 10 min. Fluorescent images were analyzed by densitometry in ImageJ. AuNP expulsion rates were obtained by taking the slope of the linear regression curves seen in SI Appendix, Fig. S11B.
Simulation of Nanoparticle Diffusion in Collagen Matrices.
Two-dimensional and 3D stochastic models of AuNP movement in collagen matrices were programmed and simulated in Matlab; 2D models were used to study how AuNP diameter and differences in the available area fraction of collagen matrices would affect the frequency of AuNP–collagen collisions; 3D simulations were conducted to investigate the impact of collagen density on AuNP diffusion distance. For both models, AuNP movement was taken as discrete random walk steps obeying Einstein–Stokes Brownian motion, as dictated by Eq. 2, where KB, η, T, and r denote the Boltzmann constant, solvent viscosity, temperature in kelvins, and AuNP radii, respectively:
| [2] | 
Particle movement was approximated as the mean square displacement according to Fick’s second law (Eq. 3), where δ and dt were taken as the discrete distance and time interval between steps:
| [3] | 
AuNPs were approximated as circles and spheres for two and three dimensions, respectively. AuNP–collagen fiber collisions were assumed to be elastic with collagen fibers approximated as immobile cylinders. Simulations were also conducted in the limit of dilute AuNP concentrations where AuNP–AuNP collisions could be neglected and particles could be independently tracked.
For the 2D simulations, collagen matrices of differing available area fractions were approximated as square pores of varying size. Pore sizes were selected based on empirically measured spaces between collagen fibers seen in scanning electron microscopy images of collagen hydrogels of varying concentration (SI Appendix, Fig. S9). Images were imported into ImageJ and thresholded to differentiate collagen fibers from pores. The size of each pore was measured by calculating the rolling ball radius of each pore using ImageJ’s built-in algorithm. Initial AuNP positions were randomized within the collagen pores and were permitted to move stochastically within the square. Upon AuNP movement beyond the dimensions of the pore, collision events were counted and AuNP trajectories were elastically reflected. Particles were tracked for 10,000 steps at 0.1-s intervals. AuNP–collagen collisions were tallied for each condition.
To simulate 3D collagen, matrices composed of 100, 300, 600, and 1,000 cylinders ranging in length from 0 to 30 µm and radii ranging from 0.05 to 0.50 µm were randomly distributed and oriented within 30 × 30 × 30 µm cubes to mimic hydrogels with collagen volume fractions between 8.72% and 87.20%. Collagen volume fractions were determined by calculating the ratio of volume occupied by collagen fibers (approximated as cylinders) versus the total region of interest (30 × 30 × 30 µm cube). Volume fractions were equated to empirical collagen pore sizes by taking a 2D projection of the generated tissue, followed by calculation using the same ImageJ process as mentioned for our 2D simulations. For particle-motility simulations, AuNPs were randomly placed in our 3D matrices, allowed to move freely within the confines of the cube, and reflect off collagen fibers. The direction of AuNP motion was randomized with each step. AuNPs were tracked for 5,000 steps at 0.5-s intervals. Displacement between start and end points were measured to determine AuNP diffusion distances.
Statistical Analysis.
All statistical analysis comparing between groups were performed using one-way ANOVA (one variable per group) and two-way ANOVA (two variables per group) in GraphPad Prism.
Supporting Information.
See the SI Appendix for details regarding materials, gold nanoparticle synthesis and characterization, tumor induction, nanoparticle administration, tumor histology analysis, and whole-animal imaging.
Supplementary Material
Acknowledgments
This work was supported by Natural Sciences and Engineering Research Council Grant RGPIN288231, Canadian Institute of Health Research Grants MOP130143 and COP126588, and Canadian Research Chair Grant 950-223824. E.A.S. and C.D.S. acknowledge fellowships from the Natural Sciences and Engineering Research Council. C.D.S. acknowledges a scholarship from the Alberta Innovates Technology Futures.
Footnotes
The authors declare no conflict of interest.
This article is a PNAS Direct Submission.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1521265113/-/DCSupplemental.
References
- 1.Chou LYT, Zagorovsky K, Chan WCW. DNA assembly of nanoparticle superstructures for controlled biological delivery and elimination. Nat Nanotechnol. 2014;9(2):148–155. doi: 10.1038/nnano.2013.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.He X, Wang K, Cheng Z. In vivo near-infrared fluorescence imaging of cancer with nanoparticle-based probes. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2010;2(4):349–366. doi: 10.1002/wnan.85. [DOI] [PubMed] [Google Scholar]
- 3.Nam T, et al. Tumor targeting chitosan nanoparticles for dual-modality optical/MR cancer imaging. Bioconjug Chem. 2010;21(4):578–582. doi: 10.1021/bc900408z. [DOI] [PubMed] [Google Scholar]
- 4.Yang J, et al. Multifunctional magneto-polymeric nanohybrids for targeted detection and synergistic therapeutic effects on breast cancer. Angew Chem Int Ed Engl. 2007;46(46):8836–8839. doi: 10.1002/anie.200703554. [DOI] [PubMed] [Google Scholar]
- 5.Cherukuri P, Glazer ES, Curley SA. Targeted hyperthermia using metal nanoparticles. Adv Drug Deliv Rev. 2010;62(3):339–345. doi: 10.1016/j.addr.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bae YH, Park K. Targeted drug delivery to tumors: Myths, reality and possibility. J Control Release. 2011;153(3):198–205. doi: 10.1016/j.jconrel.2011.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sykes EA, Dai Q, Tsoi KM, Hwang DM, Chan WCW. Nanoparticle exposure in animals can be visualized in the skin and analysed via skin biopsy. Nat Commun. 2014;5:3796. doi: 10.1038/ncomms4796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.De Jong WH, et al. Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials. 2008;29(12):1912–1919. doi: 10.1016/j.biomaterials.2007.12.037. [DOI] [PubMed] [Google Scholar]
- 9.Cole AJ, David AE, Wang J, Galbán CJ, Yang VC. Magnetic brain tumor targeting and biodistribution of long-circulating PEG-modified, cross-linked starch-coated iron oxide nanoparticles. Biomaterials. 2011;32(26):6291–6301. doi: 10.1016/j.biomaterials.2011.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kumar R, et al. In vivo biodistribution and clearance studies using multimodal organically modified silica nanoparticles. ACS Nano. 2010;4(2):699–708. doi: 10.1021/nn901146y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Moghimi SM, Hunter AC, Murray JC. Long-circulating and target-specific nanoparticles: Theory to practice. Pharmacol Rev. 2001;53(2):283–318. [PubMed] [Google Scholar]
- 12.Yaehne K, et al. Nanoparticle accumulation in angiogenic tissues: Towards predictable pharmacokinetics. Small. 2013;9(18):3118–3127. doi: 10.1002/smll.201201848. [DOI] [PubMed] [Google Scholar]
- 13.Huang X, Teng X, Chen D, Tang F, He J. The effect of the shape of mesoporous silica nanoparticles on cellular uptake and cell function. Biomaterials. 2010;31(3):438–448. doi: 10.1016/j.biomaterials.2009.09.060. [DOI] [PubMed] [Google Scholar]
- 14.Hauck TS, Ghazani AA, Chan WCW. Assessing the effect of surface chemistry on gold nanorod uptake, toxicity, and gene expression in mammalian cells. Small. 2008;4(1):153–159. doi: 10.1002/smll.200700217. [DOI] [PubMed] [Google Scholar]
- 15.Treuel L, Jiang X, Nienhaus GU. New views on cellular uptake and trafficking of manufactured nanoparticles. J R Soc Interface. 2013;10(82):20120939. doi: 10.1098/rsif.2012.0939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Brown JM, Giaccia AJ. The unique physiology of solid tumors: Opportunities (and problems) for cancer therapy. Cancer Res. 1998;58(7):1408–1416. [PubMed] [Google Scholar]
- 17.Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501(7467):346–354. doi: 10.1038/nature12626. [DOI] [PubMed] [Google Scholar]
- 18.Rosenblum D, Peer D. Omics-based nanomedicine: The future of personalized oncology. Cancer Lett. 2014;352(1):126–136. doi: 10.1016/j.canlet.2013.07.029. [DOI] [PubMed] [Google Scholar]
- 19.McMillin DW, Negri JM, Mitsiades CS. The role of tumour-stromal interactions in modifying drug response: Challenges and opportunities. Nat Rev Drug Discov. 2013;12(3):217–228. doi: 10.1038/nrd3870. [DOI] [PubMed] [Google Scholar]
- 20.Thurber GM, Schmidt MM, Wittrup KD. Factors determining antibody distribution in tumors. Trends Pharmacol Sci. 2008;29(2):57–61. doi: 10.1016/j.tips.2007.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Stylianopoulos T, et al. Diffusion of particles in the extracellular matrix: The effect of repulsive electrostatic interactions. Biophys J. 2010;99(5):1342–1349. doi: 10.1016/j.bpj.2010.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lu P, Weaver VM, Werb Z. The extracellular matrix: A dynamic niche in cancer progression. J Cell Biol. 2012;196(4):395–406. doi: 10.1083/jcb.201102147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rich L, Whittaker P. Collagen and picrosirius red staining: A polarized light assessment of fibrillar hue and spatial distribution. Braz J Morphol Sci. 2005;22:97–104. [Google Scholar]
- 24.Chen X, Nadiarynkh O, Plotnikov S, Campagnola PJ. Second harmonic generation microscopy for quantitative analysis of collagen fibrillar structure. Nat Protoc. 2012;7(4):654–669. doi: 10.1038/nprot.2012.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Brown E, et al. Dynamic imaging of collagen and its modulation in tumors in vivo using second-harmonic generation. Nat Med. 2003;9(6):796–800. doi: 10.1038/nm879. [DOI] [PubMed] [Google Scholar]
- 26.Sykes EA, Chen J, Zheng G, Chan WCW. Investigating the impact of nanoparticle size on active and passive tumor targeting efficiency. ACS Nano. 2014;8(6):5696–5706. doi: 10.1021/nn500299p. [DOI] [PubMed] [Google Scholar]
- 27.Perrault SD, Walkey C, Jennings T, Fischer HC, Chan WCW. Mediating tumor targeting efficiency of nanoparticles through design. Nano Lett. 2009;9(5):1909–1915. doi: 10.1021/nl900031y. [DOI] [PubMed] [Google Scholar]
- 28.Perrault SD, Chan WCW. Synthesis and surface modification of highly monodispersed, spherical gold nanoparticles of 50-200 nm. J Am Chem Soc. 2009;131(47):17042–17043. doi: 10.1021/ja907069u. [DOI] [PubMed] [Google Scholar]
- 29.Chou LYT, Chan WCW. Fluorescence-tagged gold nanoparticles for rapidly characterizing the size-dependent biodistribution in tumor models. Adv Healthc Mater. 2012;1(6):714–721. doi: 10.1002/adhm.201200084. [DOI] [PubMed] [Google Scholar]
- 30.Provenzano PP, et al. Collagen density promotes mammary tumor initiation and progression. BMC Med. 2008;6(1):11. doi: 10.1186/1741-7015-6-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Dudley DT, et al. A 3D matrix platform for the rapid generation of therapeutic anti-human carcinoma monoclonal antibodies. Proc Natl Acad Sci USA. 2014;111(41):14882–14887. doi: 10.1073/pnas.1410996111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Stylianopoulos T, Diop-Frimpong B, Munn LL, Jain RK. Diffusion anisotropy in collagen gels and tumors: The effect of fiber network orientation. Biophys J. 2010;99(10):3119–3128. doi: 10.1016/j.bpj.2010.08.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Jakobsen I, Lyng H, Kaalhus O, Rofstad EK. MRI of human tumor xenografts in vivo: Proton relaxation times and extracellular tumor volume. Magn Reson Imaging. 1995;13(5):693–700. doi: 10.1016/0730-725x(95)00019-d. [DOI] [PubMed] [Google Scholar]
- 34.Krol A, Maresca J, Dewhirst MW, Yuan F. Available volume fraction of macromolecules in the extravascular space of a fibrosarcoma: Implications for drug delivery. Cancer Res. 1999;59(16):4136–4141. [PubMed] [Google Scholar]
- 35.Walkey CD, Olsen JB, Guo H, Emili A, Chan WCW. Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake. J Am Chem Soc. 2012;134(4):2139–2147. doi: 10.1021/ja2084338. [DOI] [PubMed] [Google Scholar]
- 36.Varin A, Mukhopadhyay S, Herbein G, Gordon S. Alternative activation of macrophages by IL-4 impairs phagocytosis of pathogens but potentiates microbial-induced signalling and cytokine secretion. Blood. 2010;115(2):353–362. doi: 10.1182/blood-2009-08-236711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang B, et al. Transition of tumor-associated macrophages from MHC class II(hi) to MHC class II(low) mediates tumor progression in mice. BMC Immunol. 2011;12(1):43. doi: 10.1186/1471-2172-12-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Albanese A, Tang PS, Chan WCW. The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu Rev Biomed Eng. 2012;14:1–16. doi: 10.1146/annurev-bioeng-071811-150124. [DOI] [PubMed] [Google Scholar]
- 39.Dawidczyk CM, Russell LM, Searson PC. Nanomedicines for cancer therapy: State-of-the-art and limitations to pre-clinical studies that hinder future developments. Front Chem. 2014;2:69. doi: 10.3389/fchem.2014.00069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Josefsson A, et al. Tumor size, vascular density and proliferation as prognostic markers in GS 6 and GS 7 prostate tumors in patients with long follow-up and non-curative treatment. Eur Urol. 2005;48(4):577–583. doi: 10.1016/j.eururo.2005.05.016. [DOI] [PubMed] [Google Scholar]
- 41.Wisnivesky JP, Yankelevitz D, Henschke CI. The effect of tumor size on curability of stage I non-small cell lung cancers. Chest. 2004;126(3):761–765. doi: 10.1378/chest.126.3.761. [DOI] [PubMed] [Google Scholar]
- 42.Yu K-D, et al. Effect of large tumor size on cancer-specific mortality in node-negative breast cancer. Mayo Clin Proc. 2012;87(12):1171–1180. doi: 10.1016/j.mayocp.2012.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.







