Abstract
Advances in nanoparticle synthesis and engineering have produced nanoscale agents affording both therapeutic and diagnostic functions that are often referred to by the portmanteau ‘nanotheranostics’. The field is associated with many applications in the clinic, especially in cancer management. These include patient stratification, drug-release monitoring, imaging-guided focal therapy and post-treatment response monitoring. Recent advances in nanotheranostics have expanded this notion and enabled the characterization of individual tumours, the prediction of nanoparticle–tumour interactions, and the creation of tailor-designed nanomedicines for individualized treatment. Some of these applications require breaking the dogma that a nanotheranostic must combine both therapeutic and diagnostic agents within a single, physical entity; instead, it can be a general approach in which diagnosis and therapy are interwoven to solve clinical issues and improve treatment outcomes. In this Review, we describe the evolution and state of the art of cancer nanotheranostics, with an emphasis on clinical impact and translation.
An optimized cancer therapy would deliver the right type of therapy to the right target, to achieve localized control of the disease efficiently with minimal systemic toxicity. This task is daunting, because there is considerable variation among tumours and individual patients. Increasingly, it is clear that the battle against cancer cannot be won with a single formulation. Rather, it will require the careful coordination of diagnosis and therapy, stratification of patient and tumour subpopulations, and treatments tailored to individual needs. Emerging nanotechnologies offer a promising opportunity for this new campaign: extensive efforts from the past decade have produced a large arsenal of nanoplatforms with diversified capabilities for drug loading and release, and for tumour targeting. It is possible to impart imaging functions to these nanoplatforms, such that cancer can be diagnosed for individualized therapy, and therapy can be monitored non-invasively and in real time. Such a nano-enabled amalgamation of therapy and diagnosis is often known as ‘nanotheranostics’ (BOX 1) and consolidates advances in nanomaterials with those on other fronts, including imaging, biomarkers and therapy. A timeline of advances in nanotheranostics is provided in FIG. 1.
Box 1. Conventional nanotheranostics.
A traditional nanotheranostic agent is integrated with both diagnostic and therapeutic moieties. Recently, nanotheranostics with diagnostic moieties and therapeutic moieties on separate nanoentities have demonstrated unique utility for cancer theranostics. To improve nanoparticle pharmacokinetics, nanotheranostics are often coated with a layer of anti-fouling agent and, sometimes, coupled with a ligand for active targeting.
Diagnostic agents
Positron emission tomography: 64Cu and 68Ga
Magnetic resonance imaging: Gd3+, Mn2+ and iron oxide nanoparticles
Ultrasound imaging: microbubbles
Computed tomography: I and Au
Optical imaging: quantum dots and fluorophores
Single-photon emission computed tomography: 99mTc and 123I
Photoacoustic imaging: Au nanostructures and porphyrin
Therapeutic agents
Chemotherapy: doxorubicin and paclitaxel
Radiation therapy: Au, Hf and Gd
Immunotherapy: cancer vaccines and immune checkpoint inhibitors
Photodynamic therapy: indocyanine green
Photothermal therapy: Au nanostructures
Gene therapy: small interfering RNA, plasmids and CRISPR
Figure 1. Historical timeline of key advances in cancer nanotheranostics.
EB, Evans blue; FDA, US Food and Drug Administration; HPMA, N-(2-hydroxypropyl)methacrylamide; MRI, magnetic resonance imaging; PEG, polyethylene glycol; PET, positron emission tomography; siRNA, small interfering RNA.
At present, such advances have been largely confined to academic settings. Moreover, most nanotheranostic publications imply that imaging and therapy are performed essentially independently, rather than in an integrated protocol. We need to ask how to exploit these advances to solve previously unanswerable clinical questions. Also, we need to determine to what extent and in what ways integrated nanotheranostics are advantageous over discrete steps of imaging and therapy. How can nanotheranostics promote the development of nanomedicine? How can nanotheranostics transform from ‘bench to bedside’?
One debate throughout the development of nanomedicine has been about the efficiency of the enhanced permeability and retention (EPR) effect in solid tumours. Discussions have included the advantages and disadvantages of passive and active targeting, and recent debates have been fuelled by a controversial paper in which it was concluded that the average tumour uptake of nanoparticles is only 0.7% of the injected dose1. When the EPR effect was first discovered, it was extolled by many as the Achilles’ heel of cancer2; it is clear now that this is not the case. Delivering nanoparticle drugs to cancer cells is an extremely complicated process3–5. It requires that nanoparticles evade immune surveillance and avoid adsorption of serum opsonin proteins, and selectively extravasate at a tumour site. It also requires nanoparticles to overcome cancer cell intravasation, thick tumour stroma (the supportive tissue), uptake by macrophages, high interstitial fluid pressure (IFP) and slow diffusion, and to achieve homogeneous distribution throughout the tumour. From this perspective, the conventional EPR model, at least in its classical definition, is oversimplified and inadequate to predict nanoparticle accumulation in the tumour. Moreover, nanoparticle deposition in tumours is not governed by only one effect, such as EPR, but by a group of factors. Abstraction of delivery rules without taking these variables into consideration underlies many of the contradictions and controversies (for example, passive targeting versus active targeting) in this field6. Even worse, the initial assumption was that the magnitude of the EPR effect would be of the same order among different solid tumours, or at least among those of the same origin. This led to early, unrealistic hopes for nanomedicine7. The right attitude, we now believe, is to embrace the benefits of nanotechnologies but acknowledge their limitations. We need to give up the one-for-all or all-in-one obsession; instead, nanotherapies, perhaps even more than other modalities, need to work closely with diagnosis and be given only to the right patients8. Nanotheranostics, we believe, can and should have an important role in this new campaign9–13.
In this Review, we discuss fundamental notions of nanotheranostics and review the history and state of the art of cancer nanotheranostics, including immune nanotheranostics to treat the suppressed immune system. We focus on methodologies with potential to make an impact in the clinic, such as patient stratification to identify subpopulations that are most likely to benefit from nanotherapy; tracking drug release and penetration within tumours; imaging-guided focal therapy; and the monitoring of therapeutic responses. Additionally, we lay out challenges and opportunities of cancer nanotheranostics, such as comprehensive individual tumour characterization, understanding and predicting nanoparticle–tumour interactions, and tailoring nanomedicines for optimized treatment. Many of these developments are at an early stage — we are still at the dawn of personalized medicine — but hold potential to revolutionize drug research and development, and clinical oncology.
Key concepts and the status quo
With both diagnostic and therapeutic functionalities, cancer nanotheranostics have been explored for applications beyond diagnosis or therapy alone. Specifically, some unique applications range from patient stratification to patient subpopulation screening, from monitoring intratumoural drug release to optimizing it for therapeutic efficacy, and from image-guided local therapy to therapy response monitoring. These unique applications are especially important in the context of tumour heterogeneity among patients, which demands personalized theranostic strategies.
Patient stratification
It is common to take labelled nanotherapeutic particles (FIG. 2) of similar compositions, but different sizes or shapes, and determine their optimal morphology by comparing their tumour uptake in the same animal model. However, it is rare to inject the same nanotherapeutics into tumour-bearing animals and stratify these ‘patients’ on the basis of differences in tumour uptake. This probably stems from the assumption that tumours in preclinical models often share similar background and characteristics. In particular, tumour variations among xenograft models are often considered small and their responses to nanotherapy comparable, which is not necessarily the case. For example, iodine-labelled liposome nanoprobes were injected into rats bearing breast tumour xenografts and tumour uptake was assessed by mammography14. Based on tumour uptake, the animals were divided into good- and bad-prognosis groups and treated with liposomal doxorubicin. The therapeutic efficacy correlated well with the classification, with the good-prognosis group showing slower tumour progression. This suggests that even in artificial xenograft models, tumour variation may have a considerable, underappreciated impact on treatment efficacy. For large animals bearing spontaneous tumours, the discrepancy is even greater. For example, a positron emission tomography–computed tomography (PET–CT) study of 64Cu-labelled liposomes in 11 canine patients with cancer15 found EPR-mediated tumour uptake in 6 out of 7 carcinomas but only 1 out of 4 sarcomas. Further, because tumour heterogeneity is increasingly recognized in both human patients and xenograft tumour models, it is certainly biased to choose to explore nanotheranostic applications solely in tumour models for which a nanomedicine works well. Therefore, we encourage researchers to consider tumour heterogeneity when exploring nanotheranostics, by using multiple relevant models and humanized tumour models, despite the inevitably elevated cost and effort involved.
Figure 2. Nanotheranostics for cancer diagnosis.
In nanotheranostic agents, imaging functions are imparted to nanomedicines by adding moieties that are readily detected by imaging methods. These nanotheranostic agents have been exploited for tumour diagnosis and subsequent patient stratification, for understanding the pharmacokinetics and pharmacokinetics of nanomedicines, and for monitoring therapy response. a | Liposome with radioisotope in the core. b | Liposome labelled with fluorophores on the surface. c | Polymeric conjugate labelled with radioisotopes. d | Polymeric micelle loaded with T1 MRI mediators. e | PLGA nanoparticle with T1 MRI mediators loaded inside and fluorophores labelled on the surface. f | Iron oxide nanoparticle labelled with fluorophores on the surface. g | Iron oxide nanoparticle coated with photoacoustic or photothermal material. h | Gold nanorod. MRI, magnetic resonance imaging; PLGA, poly(lactic-co-glycolic acid); T1, longitudinal relaxation time.
From a technological perspective, patient stratification with nanotheranostics may not be far away (FIG. 3). In 2001, 111In-labelled PEGylated liposomes (with no drug loaded) were injected into human patients who had different types of locally advanced tumours16. Effective tumour accumulation was found in 15 out of 17 cases, but there were large variations in tumour uptake among tumour types. Then, in a 2009 phase II clinical trial, a nanoparticle formulation consisting of N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer combined with doxorubicin was mixed with a 123I-labelled analogue and injected into patients with different cancer types17. Accumulation of nanoparticles in primary tumours and, in some cases, in metastases, was observed. Moreover, liposomal doxorubicin plus cisplatin was investigated for treatment against unresectable malignant pleural mesothelioma18, and tumour uptake, quantified by 99mTc-liposome imaging, was positively correlated with patient response and survival19. However, in the above studies, the nanotheranostic agents were not assessed for patient stratification, but to evaluate the therapeutic efficacy of the nanomedicines. As far as we know, there is currently no nanotheranostic formulation in the pipeline for translation to clinical practice. Despite the technical soundness of the methodology, much work needs to be done to bring it forward for regulatory approval20.
Figure 3. Applications of nanotheranostics in cancer therapy.
Patients go through pretreatment imaging to understand the pharmacokinetics and pharmacodynamics of the nanomedicines as well as intratumoural distributions and drug release. Based on the imaging results, prognoses can be made, along with selection of patients who are likely to benefit from the nanotherapy. Next, the select patients will receive the nanomedicine and — from the earliest stages — receive monitoring of therapy responses, the feedback from which will in turn guide the evaluation of therapeutic efficacy and, if necessary, adjust future treatment regimens for optimal therapy outcome. TME, total mesorectal exision.
A specialized probe for each nanotherapeutic formulation would be ideal but is practically challenging in the short term. For the present, it is probably more realistic to develop generic imaging probes that can adequately predict the performance of a wide range of nano-therapeutics21. Recently, ferumoxytol, a clinically used iron oxide nanoparticle formulation, was assessed as such an imaging agent22. Dye-labelled ferumoxytol (~30 nm) and nanoparticles (~90 nm) consisting of poly(ethylene glycol) methyl ether-block-poly(lactide-co-glycolide) (PEG–PLGA) block copolymer were injected into mice bearing subcutaneous human fibrosarcoma, and high-resolution intravital microscopy was used to study the tumour accumulation of both particles. Despite the relatively large difference in size and composition, ferumoxytol showed a similar tumour accumulation pattern to the PLGA–PEG nanoparticles and can well predict the distribution of PLGA–PEG nanoparticles within tumours and microvasculature. Ferumoxytol-based MRI can adequately predict accumulation and treatment response of paclitaxel-loaded therapeutic nanoparticles. This approach is now being assessed in a clinical trial, where ferumoxytol-based MRI is used to predict patient response to MM-398, an irinotecan liposomal formulation23. Note that ferumoxytol is not an ideal imaging agent, but was developed to treat iron-deficiency anaemia in adult patients with chronic kidney disease. In the next stage, it will be worthwhile to develop dedicated tumour imaging probes22,24, for example radioisotope-labelled liposomes (with no drug loaded), to screen patients for specific nanotherapeutics. In one relevant study, PET-radiolabelled nanoparticles were demonstrated to predict the efficacy of nanotherapies in animal models24. In particular, a zirconium-89 nanoreporter (89Zr-NRep) co-administered with nanomedicines including Doxil precisely quantified the biodistribution of nanomedicines. Although substantial intertumour heterogeneity in accumulation was revealed by 89Zr-NRep PET imaging, the therapeutic efficacy also positively correlated with the tumour accumulation of nanomedicine.
Monitoring intratumoural drug distribution
In many nanoparticle studies, the overall tumour uptake is considered the most important criterion in nanocarrier screening. However, this notion is potentially problematic. The intratumoural distribution of nanoparticles is never homogeneous but is affected by factors such as interstitial fluid pressure (IFP), blood flow, diffusion and stroma thickness25 (FIG. 4). In particular, it is difficult for nanoparticles to migrate into the central, often hypoxic and sometimes necrotic, regions of a tumour26–29, where the cancer cells have a high propensity to acquire stem-like phenotypes and enhanced tumorigenicity30. Hence, nanomedicines with comparable tumour uptakes may show different treatment outcomes because of discrepancies in intratumoural distribution. In addition to evaluating overall tumour uptake, it is equally important to develop tools to map intratumoural nanoparticle deposition and drug release, and then include the information in prognosis11.
Figure 4. Tumour characteristics that affect the intratumoural fates of nanotheranostics.
Slow blood flow may affect the extravasation of nanoparticles in a size-dependent manner. Leaky blood vessels affect the extravasation of nanoparticles in a size-dependent manner. This vasculature leakiness varies considerably between tumours of different types and stages. Dense blood vessels typically enhance tumour accumulation of nanotheranostics. A dense extracellular matrix (ECM), especially in the tumour periphery, may restrict the tumour penetration of nanotheranostics. An increased interstitial fluid pressure (IFP) in many tumours represents a barrier for transcapillary transport of nanotheranostics. Nonspecific uptake by stromal cells, such as tumour-associated macrophages (TAMs), may negatively affect deep tumour penetration and delivery of nanoparticles to cancer cells, but, on the other hand, may serve as an intermediate reservoir22.
In preclinical studies, this can be achieved by intravital microscopy. The high spatial and temporal resolution of this technology permits in-depth analysis of interactions between nanoparticles and tumours, and can guide therapeutic optimization. For example, this method was used to investigate tumour accumulation and drug release of doxorubicin-loaded, temperature-sensitive liposomes, whereby drug release induced by hyperthermia intravascularly was found to lead to increased free drug in the tumour interstitial space31. A phase III clinical trial of a temperature-sensitive liposomal doxorubicin, ThermoDox, with radiofrequency ablation is under way for the treatment of hepatocellular carcinoma32. Intravital microscopy was also used to study the dynamic interaction between tumours and single-walled carbon nanotubes33. Unlike in previous thoughts, the nanotubes were first taken up by Ly-6Chi monocytes and delivered to tumours by cell-mediated transportation. In another example, intravital microscopy was used to study micelle nanoparticle delivery to BxPC3 tumours. In addition to static pores in the endothelium, dynamic vascular bursts were identified that mediate nanoparticle extravasation at the tumour sites34.
In the clinic, intravital microscopy has limited use owing to its invasiveness. Instead, MRI-based imaging methods have been developed35. MRI affords high spatial and temporal resolution, which is essential for in-depth analysis of the intratumoural behaviour of a nanomedicine. MRI-based imaging can be achieved by imparting a T1 or T2 imaging probe into nanoparticles, preferably ones that are currently used in the clinic. Different nanoparticle-based systems for drug-release monitoring are depicted in FIG. 5 and detailed as follows. For example, both Gd-DTPA and (1,2-diaminocyclohexane)platinum(II) (DACHPt) were incorporated into polymeric micelles through reversible chelation. The R1 (relaxivity of T1) of the resulting micelles was 24 times higher than that of Gd chelates, making it an effective nanotheranostic agent for MRI-based tumour accumulation tracking36. However, probably a more advantageous approach to monitoring drug release is to convert nanoparticles into a switchable MRI probe that experiences a signal change following drug liberation. This approach has been used successfully in liposome-based nanotherapeutics, where Mn2+/Gd3+-based salts or chelates are loaded into the interior of nanoparticles along with drug molecules. Owing to limited access to the bulk water, Mn2+/Gd3+ induces only slight T1 shortening in an intact liposome particle; when the lipid layer is breached, for example, in the tumour extracellular environment, the payloads are released to the aqueous surroundings, causing hyperintensities on a T1-weighted image that can serve as indicators of drug release. For example, in one study, MRI was used to monitor drug release with MnSO4–doxorubicin liposome nanoparticles37,38. In another study, Gd(HPDO3A) was incorporated into doxorubicin liposomes to track drug release39. In both cases, good spatial correlation was found between T1-shortening effects and doxorubicin liberation, thereby demonstrating that MRI can be used to derive intratumoural drug-dose painting38. Such an activatable MRI imaging approach has also been seen with other types of nanotherapeutics. For example, it was shown that drug molecules can be adsorbed to the coating of ferumoxytol and released in the acidic tumour extracellular environment40. This is accompanied by a significant change of the T2 relaxation time, making ferumoxytol an interesting theranostic nanoplatform. In another study, arsenic trioxide (ATO) and Mn2+ were combined to prepare water-insoluble manganese arsenite complexes41. In the tumour microenvironment, acidic stimuli triggered the simultaneous release of manganese ions and ATO, leading to a shortened T1, which could then be monitored by MRI in real-time.
Figure 5. Nanotheranostics for drug-release monitoring.
Designer nanotheranostics have been developed to monitor the intratumoural drug release from nanomedicines by various mechanisms. a | T1 MRI mediators co-released with drug molecules from a liposomal carrier, which generates T1 hyperintensity in magnetic resonance scans. b | Mn2+ and drug molecules (HAsO3−)are released on decomposition of Mn2+-doped arsenic trioxide nanoparticles from the mesoporous silica shell, resulting in T1 hyperintensity in magnetic resonance scans. c | T1 mediators are co-released with drug molecules from the polymeric micelle, resulting in T1 hyperintensity in magnetic resonance scans. d | Drugs are released from the surface of iron oxide nanoparticles, resulting in T2 hyperintensity in magnetic resonance scans. e | Iron oxide nanoparticles, Gd-DTPA and drug molecules (5-FU) are loaded in PLGA nanoparticles. On drug release, T1 hyperintensity is generated in the magnetic resonance scans owing to deshielding. f | Chemical-shift agents (for 1H CEST detection) and highly fluorinated compounds (for 19F detection) are loaded into liposomes together with drug molecules. Before release, signal enhancement is generated in 1H CEST magnetic resonance images; after drug release, hyperintensity results in the 19F magnetic resonance images. CEST, chemical exchange-dependent saturation transfer; DTPA, diethylenetriamine pentaacetate; MRI, magnetic resonance imaging; PLGA, poly(lactic-co-glycolic acid); 5-FU, fluorouracil. T1, longitudinal relaxation time; T2, transverse relaxation time.
Some nanomedicines are designed in such a manner that the drug release is governed by an exogenous rather than endogenous stimulus, such as ultrasound, heat, photo-irradiation or X-rays42,43. Imaging guidance is important for these nanomedicines, because the timing to apply the stimulus may greatly affect intratumoural drug distribution. For example, Mn2+-doxorubicin temperature-sensitive liposomes were injected into fibro-sarcoma-bearing rats, and heat was delivered to the tumour centres by means of a catheter38. The total amount of drug released was found to be higher when the liposomes were administered during, but not after, hyperthermia. However, in this case, drugs accumulated mostly at the tumour periphery, where large arteries are located. By contrast, when nanoparticles were injected before hyperthermia, there was relatively low overall drug uptake in tumours, but the perfusion was substantially greater. This is good evidence that intratumoural drug distribution can be independent of overall tumour uptake and should be monitored separately. It is worth mentioning that recent advances in high-intensity focused ultrasound (HIFU) allow hyperthermia to be delivered to tumours in a non-invasive manner. This makes heat-regulated drug release with temperature-sensitive liposomes highly promising in the clinic44. MRI-based drug-release monitoring may play an important role in the clinical translation of this technology45. Ultrasound imaging will be another important tool to guild HIFU ablation and to monitor drug release46.
It may be possible to use one nanotheranostic agent to monitor both tumour uptake and drug release. This can be achieved by exploiting multiple imaging modalities (for example, PET for tumour uptake analysis and MRI for assessment of intratumoural drug release). Alternatively, multiplexed MRI can be used to track the two processes. For example, both iron oxide nanoparticles and Gd-DTPA were incorporated into 5-fluorouracil (5-FU)-loaded PLGA nanospheres and microspheres47. Tumour targeting was tracked by T2-weighted MRI, while the drug release was monitored by T1-weighted MRI. In another example, Tm(HPDO3A)(H2O) and NH4PF6 (used as 1H chemical exchange saturation transfer (CEST) and 19F contrast agents, respectively) were loaded into liposomes48. The CEST signal was strong for intact nanoparticles, but dropped significantly when the liposome membrane was breached during drug release. By contrast, the 19F signal was quenched in intact nanoparticles, but enhanced when Tm(HPDO3A)(H2O) was released to the surroundings. In the next stage of development, more effort should be invested in developing nanotheranostics that permit simultaneous assessment of overall tumour uptake and intratumoural drug release, and tools that can integrate the information for accurate prognosis.
Clinical imaging has its intrinsic limitations. Even with advanced nanotheranostics, it is not possible to use current clinical imaging technologies to examine nanodrug dynamics at the cellular or molecular levels, which is critical to the delivery of certain therapeutics, especially small interfering RNA (siRNA) and microRNA49–51. Companion biopsy and histological studies analysis may complement current imaging technologies to reveal information in a relatively systemic manner. However, protocols remain to be established at the clinical level.
Imaging-guided focal therapy
Nanoparticle-enabled focal therapy, such as photodynamic therapy (PDT) and photothermal therapy (PTT)52–56, has advantages including low systemic toxicity, no induced resistance and high tumour selectivity. Unlike the delivery of chemotherapeutics, the tumour selectivity in PDT and PTT is mainly governed by photo-irradiation rather than nanoparticle distribution. Still, it is important to monitor nanoparticle tumour accumulation so that the irradiation can be given at the best time interval (for example, to achieve the highest tumour-to-normal-tissue ratio of nanoparticle accumulation) for optimal treatment outcomes. However, in most nanoparticle-based PDT and PTT studies, imaging and therapy are conducted separately, rather than as an integrated protocol. Another problem is that, owing to limited tissue penetration of light, PDT and PTT are more promising in the treatment of cancers that are close to the skin, internal linings accessible by endoscopy, or organ surfaces that can be exposed during surgery. Therefore, it is important to assess nanotheranostic PDT and PTT in relevant animal models, with clinically compatible methods of light delivery and an emphasis on minimizing collateral damage to surrounding normal tissues. These assessments are rarely found in current studies.
Despite the problems outlined above, encouraging progress is being made. In one recent study, a porphyrin lipoprotein (PLP)-mimicking nanoparticle was investigated for imaging-guided surgery and PDT57. Each PLP contains multiple porphyrin molecules and can efficiently chelate with 64Cu and produce singlet oxygen (1O2) under photo-irradiation. PLPs also afford switchable near-infrared fluorescence, which is quenched in intact nanoparticles, but activated when the particles are disassembled. When tested in VX-2 buccal carcinoma rabbit models, PLPs showed selective accumulation in primary tumours and metastatic nodes. This was attributable to the EPR effect and possibly the high affinity of porphyrin against cancer cells. The tumours were visualized by both PET imaging and intraoperative fluorescence imaging, enabling precise photo-irradiation and tumour eradication. In another example, a PTT agent called porphysome was used, which features high porphyrin packing density and high light-to-heat conversion efficiency55. When tested in a rat orthotopic prostate cancer model, 64Cu-porphysome showed good tumour homing effect, manifesting a tumour-to-normal-prostate ratio of 6:1. This was followed by MRI-guided insertion of an optical fibre to the tumour region to initiate PTT, and magnetic resonance thermometry to monitor regional temperature changes. The zone of temperature increase matched the tumour boundary well, leading to selective PTT damage while minimally affecting normal prostate tissues. These systems underscore the unique capability of nanotheranostic agents for imaging-guided focal therapy.
The good light-to-heat conversion efficiency of most PTT agents also makes them capable photoacoustic imaging (PAI) agents. Therefore, it is possible to track PTT nanoparticles by PAI, saving the need for an extra imaging probe. For example, several nanoparticle-based PTT agents with multiple imaging capabilities, including gold nanovesicles58, gold nanorods and melanin-coated Fe3O4 nanoparticles59, have been reported. In an orthotopic U87MG mouse model, it was shown that PAI can be used to guide selective photothermal ablation induced by hollow gold nanospheres60. It was demonstrated that the migration of gold-plated carbon nanotubes, or golden carbon nanotubes (GCNs), can be tracked by PAI. In particular, antibody–GCN conjugates home to lymphatic endothelial walls, and the PAI results can then guide a laser to induce localized damage to the lymphatic vessels with great accuracy61.
Nanoparticle-enhanced radiation therapy is less studied than PDT and PTT, which is unfortunate because it is a mainstay in clinical oncology and nanotechnology holds great potential to enhance its efficacy. Unlike PDT and PTT, clinical radiation therapy has almost no difficulties with tissue penetration. With advanced radiation planning and delivery (for example, intensity-modulated radiation therapy and stereotactic body radiotherapy), X-rays can be delivered in a 3D conformal manner to cover tumours lying under deep tissues62. However, one main problem of radiation therapy is that some cancer cells are refractory to the treatment. To improve efficacy, chemotherapy is often administered during radiation therapy to sensitize the cancer cells (known as chemoradiation therapy)63–66. However, the trade-off of the accompanying chemotherapy is increased systemic toxicity and even increased mortality67. Nanomedicines may find wide application in this context by reducing side effects and increasing drug bioavailability. For example, promising results were obtained from a series of clinical studies conducted to investigate liposomal doxorubicin as a radiosensitizer for non-small-cell lung cancer (NSCLC), breast cancer and bladder cancer management68,69. However, nanoparticle-based chemo-radiotherapy has not yet been adopted in the clinic. In addition to the relatively slow clinical translation of nanomedicine (and thus limited options for these therapeutics), another important reason is that current clinical nanomedicines were not designed as adjuncts to radiotherapy, and hence their radiosensitizing effects are suboptimal. Fortunately, efforts are emerging to develop dedicated nanotherapeutics for chemoradiation therapy. For example, Genexol-PM, a clinically used micelle formulation, was shown to improve paclitaxel accumulation in rodent NSCLC tumours70. This led to enhanced radiation therapy efficacy, while reducing toxicity to healthy lung tissues and other major organs. Nanoparticle formulations of histone deacetylase inhibitors71 and wortmanin72,73 were also investigated as radiosensitizers with encouraging results.
In addition to chemotherapeutics, nanoparticles containing high-atomic-number elements (for example, gold, iodine and gadolinium) have also been used for radiosensitization. With high absorption cross sections, these heavy-element nanoparticles can improve energy deposition in tumour areas, leading to improved radiation therapy efficacy and reduced radiation doses74. For example, it was shown in Panc-1 xenograft models that systemic administration of Arg–Gly–Asp (RGD) peptide-conjugated gold nanoparticles and conformal-image-guided radiation therapy can lead to site-specific damage of tumour neoendothelium75. In another study, it was shown that NBTXR3, a 50 nm hafnium oxide nanosphere, can substantially enhance radiation therapy efficacy when intratumourally injected into different tumour xenograft models76. This nanoparticle formula is in phase II/III clinical trials for improving radiation therapy against advanced soft tissue sarcoma of the extremities77, and in phase I/II for head and neck cancer, liver cancers (both hepatocellular carcinoma and liver metastases), prostate cancer and rectal cancer radiation therapy. Recently, AGuIX, a ~3 nm polysiloxane and a Gd-DTPA conjugate, was investigated as a nanoparticle radiosensitizer14. With a high Gd content, these nanoparticles showed good T1 contrast and impressive radiosensitizing effects. For example, in a clonogenic assay against B16F10 cells, high sensitivity enhancement ratio and dose enhancement fractions were observed78. Owing to efficient renal clearance, these nanoparticles induced little systemic toxicity, which was confirmed in mouse and cynomolgus monkey models79. When injected into murine brain metastasis models, AGuIX efficiently accumulated in tumours, as verified by T1-weighted MRI. Whole-brain radiotherapy was applied at a time post-injection determined from the imaging results for optimal tumour selectivity. The radiosensitizing effect of AGuIX was also demonstrated in mouse pancreatic xenograft models and a good safety profile and imaging-guidance potential was shown in cynomolgus monkeys79. This nanotheranostics is now under clinical trial for treating brain metastasis80.
It is possible to combine radiation therapy with another focal treatment. A good example is the combination of radiation therapy and PDT, which can induce a synergistic effect in cancer cell killing81. However, synergy has been shown only to occur when the two modalities are applied simultaneously82, which is not practical in the clinic. New scintillator nanoparticles, such as those made of Tb2O3 (REF. 83), LaF3:Tb (REF. 84) and SrAl2O4:Eu (REFS 85,86), can be used to down-convert X-ray photons to visible photons, and in turn, activate a PDT process. Alternatively, Cherenkov radiation from radionuclides can be harnessed to activate titanium dioxide nanoparticles and produce reactive oxygen species87. Although different, the two approaches both constitute an effective PDT and radiation therapy combination, which explains the efficient killing of cancer cells, including those that are refractory to radiation therapy alone. The combination of such a radiation therapy and PDT also represents a significant advance for overcoming the shallow penetration of conventional PDT.
Nanoparticle probes can also function as intraoperative imaging agents to guide surgical procedures. This is important because tumours are often irregular in shape, and the margins are difficult to identify during surgery. Incomplete resection may cause lethal recurrence, whereas removal of normal tissues induces excessive morbidity. Previously, intraoperative MRI, often facilitated by Gd agents, has been investigated to guide surgery in the clinic. However, this method has certain restrictions, including narrow imaging time windows, high injection doses and suboptimal specificity. To address these issues, there has been an interest in developing intraoperative optical imaging probes, which potentially offer better sensitivity and specificity with regard to margin delineation. For example, plasmonic nanobubbles were investigated as intraoperative acoustic probes88. These nanobubbles are not introduced exogenously but generated in vivo from around gold nanoclusters under short laser pulses. In another study, a MRI–PAI–Raman trimodality nanoprobe was evaluated for assisting surgical removal of brain tumours. In this probe, the MRI component allows for preoperative detection and surgical planning, while the PAI component can be used to guide bulk tumour resection during surgery, and the Raman imaging enables accurate removal of residual microscopic tumour burden89. Another type of material that has been used for tumour imaging is silica nanoparticles, called Cornell dots or C dots90. Specifically, the C dots were encapsulated with the cyanine dye Cy5 and 124I, and conjugated with cyclic RGD peptide on the surface, making them compact optical-PET probes with great tumour affinity as well as good renal clearance. A recent study revealed that the ultrasmall C dots can also be used for cancer treatment by nanoparticle-induced modulation of iron and reactive oxygen species levels within cancer cells and the tumour microenvironment, to induce ferroptosis in nutrient-deprived cancer cells, and to suppress tumour growth91.
Therapy response monitoring
Post-therapy response monitoring is an appealing concept that has frequently appeared in nanomedicine papers92. The rationale is rapid prognosis after each cycle of treatment, so that timely adjustments can be made long before the conventional endpoints. In preclinical studies, tumours are often implanted subcutaneously, and their dimensions can be easily measured by calipers. Tumours that are inoculated into internal organs have often been genetically engineered to express fluorescent, bioluminescent, MRI or PET reporters, so that their tumour load can be monitored non-invasively93,94. However, these techniques are obviously not viable in the clinic. Hence, many researchers suggest introducing imaging functions into nanoparticles to permit therapy response monitoring. Although this sounds logical, it is not clear exactly how this could work in a clinical setting and what the benefits might be. The concept of early response monitoring originates from nuclear diagnosis (PET and single-photon emission computed tomography), with an emphasis on sensitive detection of alterations at the molecular or genetic level95. However, owing to the relatively large sizes and limited access to intracellular targets, nanoparticles are suboptimal or even inadequate for the task.
Perhaps nanotheranostics can find applications in sensing a change in the tumour microenvironment (TME) that is relevant to progression in therapy. Compared with a healthy microenvironment, the TME has some distinct physiological features such as lower pH and more hypoxia in some tumours, and may have altered signature of molecular biomarkers, such as matrix metalloproteinase (MMP). Therefore, it is logical that effective therapeutics would normalized these TME parameters, and thus monitoring these TME parameters would help to monitor therapy response and provide feedback to evaluate previous therapeutic efficacy and guide future therapy. For example, optical nanoprobes with a sharp pH response (ΔpH10–90% < 0.25) were used in the acidic tumour extracellular environment96. However, because of the limited tissue penetration of light, this technique is yet to find its way to the clinic. A Mn2+-doped calcium phosphate nanoprobe was reported, which disintegrates at low pH, liberating Mn2+ and mediating signal enhancement on T1 images97. However, MRI has low sensitivity, takes a relatively long acquisition time (up to 1 h) and is difficult for quantification. We and others have developed a series of optical probes that can detect changes in MMP concentration in the tumour extracellular matrix98. This means, however, that the therapeutic and imaging functionalities of the same nanoparticle target different components of a tumour, which is considered unfavourable. Moreover, there are different sets of requirements on pharmacokinetics, clearance and dosage for diagnostic and therapeutic agents, which can hardly be satisfied within a single nanoscale particle.
For the present, it is probably more realistic to use separate diagnostic methods to follow nanotherapy, rather than imposing the function of imaging on a nanomedicine. This by no means defies the concept of treatment response monitoring; by contrast, owing to the importance of the notion, it is worth breaking the stereotype that a nanotheranostic must be a physical object concurrently carrying both prognosis (or diagnosis) and therapy functionalities. In several successful studies, conventional diagnostic tools, such as 18F-fluorodeoxyglucose (18F-FDG), 2-fluoropropionyl labelled PEGylated dimeric RGD peptide (18F-FPPRGD2), diffusion-weighted MRI and blood-oxygenation-level-dependent MRI99–103, have been successfully used for early prognosis for nanotherapy with adequate prediction accuracy. Because the concept is no different from traditional prognosis, we will not expand the discussion here. Therapeutic response monitoring should provide feedback that can guide follow-up regimens. This has rarely been demonstrated in nanotherapy, probably owing to a lack of established treatment options, and needs to be explored in more depth.
Challenges and new opportunities
One of the main issues in nanomedicine development has been underestimation of tumour heterogeneity104. This is largely attributable to an oversimplified pharmacokinetic model, in which the tumour is seen as a leaky sponge into which nanoparticles can be efficiently deposited, given a sufficiently long circulation half-life. Based on this model, nanomedicine research has focused on inhibiting or minimizing opsonin adsorption, reticuloendothelial system uptake and renal clearance. It is increasingly clear that this model downplays the impacts of microvessel density, vessel leakiness, interstitial fluid pressure, blood flow, stromal thickness and macrophage abundance, as well as variations of these factors among tumours105,106. Each of these properties may strongly affect nanoparticle accumulation and penetration in tumours. For optimal nanoparticle delivery, it is important to treat each tumour as a unique and complex organ. Instead of attributing all factors to the EPR effect, which is almost impossible to quantify, it is useful to look into each individual factor and combine the information for comprehensive prognosis. Nanotheranostics can play an important role in this arena, but because of the large number of variables, it is not always feasible to derive full information from one type of nanoparticle. Similar to the case of therapeutic response monitoring, we suggest for many of the new challenges discussed below not to treat nanotheranostics as a physical entity, but as a generic approach in which diagnosis is exploited to guide or assist a nanotherapeutic procedure. Only then can we maximally harness advances on multiple fronts to identify variations among tumours and devise optimal therapeutic regimens, and thus develop innovative and effective cancer nanotheranostics. In this section, we will specifically discuss tumour heterogeneity (especially tumour vasculature variation), approaches to tailor nanoparticles and enhance the EPR effect, and emerging cancer immunotherapeutic nanomedicine.
Variation in vascular density among tumours
Although high vascular density is considered a common tumour characteristics and a key factor contributing to the EPR effect, there is great variation in vascular density among tumours. For example, tumour microvessel density may be only 14.8 vessels per microscopic field (mm2) for squamous cell carcinoma, but up to 145 vessels per field for colorectal carcinoma107. Analysing individual tumour microvessel density would provide valuable information with which to predict nanoparticle accumulation in tumours. Tumour vascular density can be assessed non-invasively by CT-based108, MRI-based109 or ultrasound-based110 angiography.
There is also considerable variation in the leakiness of tumour vasculature. In preclinical studies, vasculature leakiness is often assessed by an extravasation assay that uses Evans blue dye111. However, because this is a histological method, it is invasive and has clear limitations in the clinic. Alternatively, vasculature leakiness can be evaluated by dynamic contrast-enhanced MRI, which provides functional information on capillary blood flow and volume in tumours112,113. Vascular leakiness may also be assessed by MRI angiogram with gadofosveset, which is an albumin-binding blood tool agent that has recently obtained regulatory approval114. Recently, we developed a series of Evans-blue-derived radiotracers, which includes a 1,4,7-triazacyclononane-N,N′,N″-triacetic acid (NOTA)-conjugated truncated form of Evans blue (NEB)115. In particular, 68Ga-NEB is now under clinical trials for diagnosis of haemangiomas116 and evaluation of lymphatic disorders117.
It is difficult to differentiate the impact of microvessel density from that of the vessel leakiness; the compound effect of both is measured in all methods mentioned above. For better assessment of vessel leakiness, it is beneficial to evaluate a group of nanoprobes with similar compositions but varied sizes. This is commonly seen in nanoparticle optimization, but has rarely been pursued as a tool in tumour imaging.
In addition to vasculature leakiness and density, increasing attention is being paid to other tumour properties that may regulate nanoparticle deposition. A methodical model that takes into account spatially distributed diffusion-convection properties was developed to depict nanoparticle uptake by tumours118. The simulation suggested that the complex transport micro-environment was responsible for the large inter-subject variations in nanoparticle tumour accumulation. In particular, the IFP was identified as an important contributing factor in restricting transvascular transport of nanoparticles and their migration within tumours. In another study, multiple imaging methods were used to investigate liposome accumulation in tumours, including CT angiography to examine tumour vascularity, perfusion CT to assess blood flow and fluorescence molecular tomography to assess nanoparticle spatial-temporal distribution108. Their study suggested that liposome deposition is highly dependent on regional blood flow, which shows large intertumour and intratumour variation. Such variations in tumour blood flow and IFP, along with other heterogeneities, such as collagen density and perivascular cancer cell density, all contribute to the complexity of nanoparticle delivery to tumours.
In summary, accurate prognosis should be based on thorough characterizations of each contributing or restricting factor within a tumour, rather than on the broad, sometimes vague, EPR effect. Piecing together the information to enable precise predictions will demand extensive efforts as individual diagnostic tools become increasingly available.
Tailored nanoparticles for optimal tumour uptake
In many studies, nanoparticle extravasation at a tumour site is considered the endpoint of the delivery, and to improve tumour uptake, an extended circulation half-life is favoured. Hence, tremendous efforts have been spent on elucidating the relationship between the physicochemical properties of nanoparticles and their circulation times. However, extravasation at a tumour site is only half of the journey for nanomedicine transportation. For effective treatment, nanoparticles (or their payloads) need to distribute evenly throughout tumours, and this is not easy for many nanoparticles, whose migration within tumours is governed by the slow process of diffusion, rather than convection. Potential differences in tumour distribution constitute a source of heterogeneity for nanoparticle-based therapy. We also need to understand the impact of nanoparticle physicochemical properties on intratumoural migration and distribution, based on tumour characterizations, to be able to predict whether a patient is suitable for particular nanomedicines. Unfortunately, the required investigations are still in their infancy.
In TABLE 1, we summarize nanoparticle design rationale with an emphasis on enhancing perfusion and retention of nanomedicines within tumours. Taking size, the most widely studied nanoparticle physical property, as an example, it is now generally accepted that nanoparticles larger than 200 nm or smaller than 5 nm are inappropriate for tumour targeting, because they are rapidly cleared from the body. There is no consensus, however, on what sizes within the 5–200 nm range are optimal. For example, 90 nm Doxil was chosen on the basis of early studies suggesting that ~100 nm was a good balance between high drug loading and favourable pharmacokinetics. However, later studies found that in many solid tumours, Doxil accumulated mainly at the periphery and perfused only minimally into the centre. By contrast, smaller nanoparticles, despite having advantages in tumour penetration, are not necessarily associated with high tumour uptake. This has been explained by means of a pharmacokinetic model for quantitative analysis of the EPR effect119. According to this model, the EPR effect, although aiding nanoparticle extravasation, also permits nanoparticles to re-enter the blood circulation. This is important because the tumour interstitial fluid has a much slower flow rate than the pulsatile flow; hence, compared with nanoparticles in the vessel lumen, those in the interstitial space stay much longer at an endothelial defect, meaning a high probability of intravasation, especially for small nanoparticles. This effect was observed in a study in which 20 nm, 50 nm and 200 nm drug–silica nanoconjugates were injected into MCF-7 breast-tumour-bearing mice120. Nanoconjugates with a diameter of 200 nm were poor in tumour extravasation, whereas 20 nm nanoconjugates were quickly cleared from the tumour region. The 50 nm particles, by contrast, seemed to offer a good balance, leading to the highest tumour tissue retention.
Table 1.
Enhancing tumour uptake of nanoparticles through nanoparticle engineering and tumour microenvironment modulation
Approach | Details | |
---|---|---|
Engineering nanoparticles | ||
Tumour penetration | Small particle size | In general, small nanoparticles penetrate relatively deep into tumours |
Minimized TAM uptake | Appropriate surface engineering (for example, PEGylation) can minimize nanoparticle uptake by TAMs | |
In situ size shrinkage | Smart nanoparticles of relatively large sizes reduce their dimensions in response to a stimulus in tumours, leading to improved penetration126–128 | |
Tumour-specific nanoparticle screening | Based on imaging results and tumour characterizations, nanoparticle formulations that would afford the best penetration can be selected or designed108,120,121 | |
Tumour retention | Large nanoparticle size | Large nanoparticles prolong retention in the tumour |
In situ nanoparticle size expansion | Building-block molecules or small nanoparticles self-assemble into relatively large nanostructures within tumours, leading to prolonged tumour retention125 | |
Active targeting of nanoparticle | Through binding with a tumour biomarker, intravasation is reduced to prolong tumour retention108,119 | |
Magnetic guidance | Use of an external magnetic field can enrich magnetic nanoparticles in tumours | |
Modulating the tumour microenvironment | ||
Increase blood vessel leakiness | Use photodynamic therapy or ultrasound to enlarge gaps on endothelial walls134–138 | |
Vascular normalization | Prune tumour blood vessels to enhance drug delivery and lower tumour IFP129–133 | |
Kill perivascular cancer cells | Use radiotherapy or immunophototherapy to selectively eliminate these relatively oxygenated cells139,140 | |
Break down ECM | Melt down ECM or reduce the deposition of the related components | |
Eliminate CAFs or other stroma cells | Specifically kill, inactivate or quiesce CAFs | |
Blood pressure regulators | Modulate the constriction or tension of blood vessels141–143 |
CAF, cancer-associated fibroblasts; ECM, extracellular matrix; IFP, interstitial fluid pressure; PEG, polyethylene glycol; TAM, tumour-associated macrophages
Such rules of thumb are not strict but are largely dependent on individual tumours. For example, the tumour accumulation and treatment efficacy was compared for polymeric micelle nanoparticles of different diameters (30, 50, 70 and 100 nm) in animals bearing either highly permeable C26 colon tumours or poorly permeable BxPC3 pancreatic tumours121. With C26 tumour models, there were no significant impacts of size on either delivery or treatment efficacy. By contrast, with BxPC3 tumour models, 50, 70 and 100 nm nanoparticles showed poor tumour penetration owing to their relatively bulky sizes. However, 30 nm nanoparticles could diffuse into the central areas of tumours, leading to the best treatment outcome. In another important study, disposition of liposomes in the tumours was found to be dependent on both nanoparticle size and local blood flow, with relatively large nanoparticles (for example, 100 nm) accumulating more in the fast-flow regions and small nanoparticles (for example, 30 nm) in the slow-flow regions108.
Such complexity in nanoparticle–tumour interactions again underscores the flaws of a one-size-fits-all approach. For optimal treatment, nanoparticle size may need to be tailored to suit tumour haemodynamics and pathology. Nanoparticles with varied sizes have not been pursued as a tool in tumour imaging, and this approach may face challenges in clinical translation owing to toxicity and cost concerns. This notion can also be expanded to other nanoparticle properties, such as shape, rigidity and surface charge. Whereas there have been tremendous efforts to elucidate the impact of these physicochemical properties on nanoparticle blood circulation half-lives and uptake in the reticuloendothelial system, their influences on intratumoural distribution and penetration have not been adequately studied. In addition, recent investigation of the protein corona effect of nanoparticles has identified key factors that influence nanoparticle–tumour interactions, such as the physicochemical properties of the nanoparticles, exposure time, protein type, and nanoparticle concentration122,123.
Tumour-tailored targeting approaches may also include active targeting. The results of a recent simulation suggest that tumour-targeting ligands may improve nanoparticle retention after extravasation and minimize intravasation119. The approach would probably work better for relatively small nanoparticles, which are more susceptible to washout from the interstitial space. For example, conjugating a ligand that targeted epidermal growth factor receptor (EGFR) to a 30 nm liposome nanoparticle was found to increase deposition in tumours108. By contrast, adding the same EGFR-targeting ligand to 100 nm liposomes had minimal impact. One should also keep in mind that other factors are relevant in tumour deposition. For example, in the study mentioned above, increased tumour uptake for 30 nm active targeting particles was only seen in regions of slow flow rate. Overlooking these impacts of tumour specifics is probably behind many of the controversies regarding active versus passive targeting.
Another approach explored to improve tumour delivery is ‘size expansion’. Briefly, molecules or small nanoparticles are injected systemically and undergo self-assembly within tumours. This approach enables enhanced tumour uptake through favourable extravasation and minimized intravasation. An example was the use of pH-sensitive PDPA-b-PAMA/SA (succinic anhydride (SA)-modified poly(2-diisopropylaminoethyl methacrylate)-block-poly(2-aminoethyl methacrylate hydrochloride)) micelles, which agglomerated in the acidic tumour microenvironment, forming aggregates that were less susceptible to intravasation124. In another study, a gelatinase-responsive molecule, P18-PLGVRGRGD, which can self-assemble into fibrous nanostructures within tumours, led to increased retention125. Interestingly, an opposite, ‘size reduction’ approach has also been explored to aid nanoparticle delivery. The idea is to design relatively large nanoparticles that can ‘smartly’ reduce their dimensions in tumour areas, thereby migrating deeper into tumours. For example, a composite nanosystem was developed that contained a 100 nm gelatin core and multiple ~10 nm quantum dot satellites. These nanoparticles were degraded by MMPs within tumours, releasing quantum dots to the surroundings126. In another example, ~100 nm polymeric clustered nanoparticles were used, which were disassembled in the acidic tumour extracellular environment, releasing ~5 nm platinum prodrug-conjugated poly(amidoamine) dendrimers127. Other examples include porphyrin microbubbles that can burst into nanoparticles in tumour areas upon in situ ultrasound stimuli and an injectable nanoparticle generator consisting of micrometre-sized porous silicon particles encapsulated with doxorubicin–polymer conjugates, which, once released within tumours, self- assemble into nanoparticles that can be internalized by cancer cells to induce cell death128.
Increasing the EPR effect for optimal tumour uptake
As mentioned above, many factors, including low micro-vascular permeability, high IFP and thick stroma, may prevent efficient delivery of nanoparticles to and perfusion within a tumour. Choosing the right nanoparticle formulation and targeting strategy may improve the delivery, but opposing factors in some tumours may simply be too strong. However, in such cases it may be possible to artificially modify the tumour microenvironment and tip the balance in favour of nanoparticle extravasation and penetration3, as summarized in TABLE 1.
One such example is IFP management. IFP is determined by several factors, including hyperpermeable and tortuous tumour vasculature, increased transvascular fluid flow, high concentrations of plasma proteins, compromised lymphatic drainage, and compression of interstitial space by fast-proliferating tumour and stroma cells. Instead of addressing these individual factors, a more sophisticated approach is to ‘normalize’ the abnormal tumour vasculature that underlies the high IFP. For example, treating tumours with an anti-angiogenesis agent (an anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibody) combined with radiation was shown to lower the IFP in tumours by up to 74%129. In a different study, treatment with an anti-VEGF was found to markedly increase the penetration distance of bovine serum albumin from tumour vessels130. This strategy was also tested in the clinic with bevacizumab, which is a humanized anti-VEGF monoclonal antibody. In patients with rectal carcinoma, the treatment led to a decrease of IFP by 73%131. In patients with NSCLC, bevacizumab treatment was found to enhance tumour vasculature and blood perfusion in some patients, improving therapy efficacy with carboplatin and abraxane132. Such a normalization treatment can be used in conjunction with nanomedicines to enhance intratumour delivery and therapeutic efficacy133.
Another barrier to efficient nanoparticle delivery is insufficient tumour vascular leakiness. Tumours often possess discontinuous endothelial layers, but fenestration sizes range widely, probably from 50 to 4,700 nm (REF. 134). Clearly, the dimensions of the fenestra may greatly affect the extravasation of nanoparticle drugs, especially relatively large ones, and it is hoped that an external stimulus can enlarge the endothelial gaps to augment the EPR effect and promote nanoparticle accumulation. This can be achieved by vasculature-targeting photodynamic therapy, which causes cell rounding and contraction, leading to enlarged endothelial gaps. The approach was previously demonstrated with small-molecule photosensitizer-mediated PDT135 and with ferritin-mediated vascular targeting PDT136. Moreover, ultrasound has been explored as a weapon against poorly penetrable endothelium. This often involves the use of microbubbles, which, in response to an external ultrasound stimulus, collapse within blood vessels, generating mechanical stress that can disrupt cell membranes and enhance capillary permeability. This strategy has been used in preclinical and clinical studies to temporarily loosen the blood–brain barrier, improving extravasation of macromolecules and nanoparticle drugs137,138.
Owing to a relative abundance of nutrients and oxygen, tumour perivascular regions often have a dense layer of cancer cells that may restrict penetration of nanoparticles and, by compressing interstitial space, cause increased IFP. These perivascular cancer cells have also been investigated as a target for enhanced EPR. Radiotherapy, which preferentially kills oxygenated cells, can be applied to eliminate perivascular cells, leading to increased nanoparticle accumulation. For example, a single radiation dose led to a transient increase of tumour uptake of polyamidoamine dendrimers139, and immunophototherapy as an EPR-enhancing method was demonstrated to enhance tumour accumulation of nanoparticles by an order of magnitude140. Unlike vasculature-targeting methods, which need to be carefully gauged to avoid thrombus formulation and vessel collapse, perivascular targeting is potentially associated with a lower risk of occluding circulation.
Poor blood pressure regulation in tumours represents another challenge. This is attributable to several factors, including the lack of a smooth muscle layer around tumour blood vessels and a high level of nitric oxide126. In the 1980s, the vasoconstrictor angiotensin II (AT-II) was explored as an agent to increase EPR141. AT-II causes vasoconstriction and a systemic increase in blood pressure. However, owing to a reduced expression of AT-II receptors, tumour blood vessels barely respond to AT-II treatment. This leads to an increased blood flow and enlarged microvascular lumen, which favours deposition of macromolecules and nanoparticles into tumours. It was found that AT-II can increase the tumour uptake of poly(styrene-co-maleic-acid)–neocarzinostatin conjugate several-fold, in contrast to healthy tissues, the uptake into which was unchanged, or even reduced142. Recently, losartan, an angiotensin inhibitor, was studied to modulate the tumour microenvironment143. Losartan reduced solid stress in tumours, leading to increased vascular perfusion, as well as improved delivery of drug and oxygen.
Several other methods to increase the EPR effect have been investigated, including the use of hyperthermia, PTT, radiofrequency energy and convection-enhanced delivery144–147. Despite great promise, more studies are needed to assess the side effects of these treatments, including toxicity to normal tissue and circulation occlusion. Just as the EPR effect is affected by intertumour and intratumour heterogeneities, the EPR increase is expected to differ among tumours, which again underscores the importance of imaging guidance. The impact of EPR enhancement is often transient, and therefore it is important to understand the time window of the effect by prognostic imaging, and to inject nanomedicines within the optimal interval. The combination of diagnosis and therapy is crucial.
Therapeutic efficacy versus systemic toxicity
The premise of nanotherapy is to enhance treatment efficacy against tumours and reduce toxicity to normal tissues. Although most of the research focus has been directed toward the former, the benefits of the latter are just as important. A survey of the nanomedicine literature from the past decade highlighted the overall dismal delivery of injected nanoparticle dose to solid tumours1. But the tumour delivery efficiency is neither the sole factor that contributes to therapeutic index nor a parameter that governs regulatory approval of nanoformulations148. Reducing drug toxicity and side effects is as important as enhancing tumour uptake efficiency, if not more so. Indeed, with the goal to benefit patients, nanomedicine is an excellent platform to shift from off-target to on-target drug accumulation149.
Thus, instead of just looking at the absolute tumour accumulation numbers, we should examine the relative increase of drug accumulation in tumours relative to normal tissues and the relative decrease of accumulation in healthy tissues, which is equally important. For example, the benefits of Doxil, relative to free doxorubicin, in tumour uptake are often marginal. However, Doxil can universally reduce cardiotoxicity, which is a dose-limiting side effect of doxorubicin (the altered pharmacokinetics may, however, lead to increased incidence of palmar–plantar erythrodysaesthesia). This allows for better tolerance and prolonged dosing, which are important factors in Doxil’s clinical success. A similar story applies to Abraxane. It is now clear that Abraxane nanoparticles quickly disassemble in the blood and thus are not necessarily delivered efficiently to tumours. Nevertheless, it is a blockbuster drug in the clinic, and a key reason is, again, improved patient tolerance. Chemotherapy-induced toxicities, including those to the pulmonary system, cardiovascular system, liver, bowel and pancreas, are well-documented and may be detected by conventional imaging methods. Future nanotherapy should take treatment-induced side effects into consideration for prognosis, regimen selection and dosage escalation.
In view of vast inter-patient heterogeneity, individualized cancer nanotheranostics are desirable for optimal therapeutic efficacy and minimal toxicity. In this regard, nanotheranostics holds great potential in ‘N-of-1’ trials (in which a single patient constitutes the whole trial)150 and in interim imaging-guided treatment (to decide between standard chemotherapy and a more intensive, but more toxic, regimen). Under the concept of N-of-1 trials, one patient obtains a series of prognoses using nanotheranostics for regimen selection. After a period of treatment using the selected regimen, another prognosis is performed for regimen adjustment. In this pattern, therapeutic outcomes can be constantly monitored. Although there are some N-of-1 clinical trials to improve patient management for osteoarthritis151, chronic pain152 and cancers153, there have been no attempts to use this strategy to study nanoparticle drugs.
Nanotheranostics for cancer immunotherapy
Immune nanotheranostics are an important class of cancer nanotheranostics154–156. Cancer immunotherapy treats cancer by means of the immune system157, and the companion diagnostics and prognostics allow cancer diagnosis, patient stratification, and therapy response monitoring. Unlike chemotherapeutic nanotheranostics, in which the diagnostics/prognostics and therapeutics both target tumour, immune nanotheranostics may target peripheral lymphocytes, tumour-infiltrating lymphocytes or tumour cells. Therefore, immune nanotheranostics is an example in which diagnostics/prognostics and therapeutics are separately incorporated. Because the diagnostics/prognostics of immune nanotheranostics are similar to nanotheranostics discussed above, we focus our discussion on nanotherapeutics, specifically on cancer immunotherapeutic nanovaccines.
The development of subunit vaccines is partially hampered by their limited half-life. Depot-forming water-in-oil emulsions, such as incomplete Freund’s adjuvant, can prolong the half-life of vaccines and potentiate the immunogenicity of antigen, but they failed to elicit a robust T cell response upon boosting vaccination and yielded limited clinical outcome158. Nanovaccines155,159–165 can penetrate tissue barriers, co-deliver antigen and adjuvant, and efficiently deliver vaccine into antigen-presenting cells (APCs) for antigen cross-presentation. For example, for a nanovaccine made of a gold nanoparticle core and a shell of CpG oligonucleotides (CpG ODN), a Toll-like receptor 9 (TLR9) agonist was used for delivering CpG to inhibit tumour growth166. Liposome was also studied to improve lymph-node-targeted delivery of cyclic di-GMP (cdGMP), a potential robust immunostimulatory adjuvant167,168. In addition to synthetic nanocarriers, naturally derived nanovaccines have been explored because of their good biocompatibility. For example, lipid-modified molecular vaccine can hitchhike endogenous albumin to deliver vaccine to lymph nodes, thus eliciting a robust anti-tumour T cell response. Compared with exogenous nanocarriers, nanovaccines assembled in vivo from exogenous and chemically defined molecular vaccines are attractive for potential good manufacturing practices169. In another example, high-density-lipoprotein (HDL)-mimicking nanodisks co-delivered adjuvant and cancer-specific neoantigen for personalized cancer immunotherapy162. Use of cancer-cell or erythrocyte membranes170–172 to cloak nanoparticles can also transplant the biomolecular signature of the membrane onto the nanoparticle and impart the biocompatibility of the host cell membrane. Nanovaccines also confers optimal immunization by efficient intracellular delivery of many molecular adjuvants162,166,173 and antigens. Specifically, intracellular delivery of antigen is pivotal to elicit robust antigen-specific T cell responses, because antigen needs to be internalized and processed by intracellular protease machinery in APCs, and transported into specialized intracellular compartments to bind with newly synthesized major histocompatibility complex (MHC) molecules for antigen cross-presentation. In this regard, antigen delivered by exogenous nanoparticles is internalized by APCs, processed, loaded onto MHC class I and cross-presented to CD8+ T cells174,175.
Nanovaccines are also expected to aid the development of neoantigens, which are derived from somatic mutation in tumours, and are thus expressed exclusively in tumour cells. The signature of tumour mutations (termed the mutatome) of each patient is unique, making neoantigens ideal for personalized cancer immunotherapy. However, the natural frequency of neoantigen-specific T cells is often small. Thus, it is desirable to deliver exogenous neoantigen — which nanovaccines can do efficiently, as discussed above162 — to increase the frequency of tumour-specific T cells. Despite the hope of neoantigen-based nanovaccines, several challenges remain. First, the load of naturally occurring neoantigen is extremely low in some tumour types, such as glioblastoma, and pancreatic and breast cancer, which hinders the identification of effective neoantigen176. Second, current technology takes ~3 months to identify and manufacture synthetic neoantigen peptides for vaccination. Nanotechnology can also improve the delivery of mRNA vaccines. Antigen-encoding, in vitro transcribed modified mRNA vaccine generally has low risk of latent viral infection and potent T cell response177–180. Delivery of mRNA to APCs is prerequisite for optimal therapeutic efficacy but is challenged by nuclease susceptibility, inefficient intracellular delivery, and endosome mRNA trapping. Nanovaccines can be explored to address these challenges181,182. For example, liposome was used to deliver neoantigen-encoding mRNA, resulting in efficient mRNA delivery to lymphoid dendritic cells in the spleen and potent T cell responses in mice and in human181,182. Worth noting, one mRNA can be engineered to encode multi-epitope neoantigens, thus conferring a broad spectrum of T cell responses for optimal cancer therapy. Several ongoing clinical trials are likely to unveil detailed potential and challenges of nanovaccines.
Conclusion
Cancer nanomedicine has evolved considerably in the past two decades, beginning with a great expansion of the materials repertoire, characterized by diversity and multifunctionality. Some of these nanomaterials were then further explored for scaled-up manufacturing and clinical translation to address unmet oncological needs. Tumour targeting has been a central theme to nanomedicine that has also gradually evolved. This started with the community concentrating on pure EPR-based tumour uptake, focusing almost entirely on extending nanoparticle circulation half-lives, and largely ignoring intertumour variations. The concept of active targeting was then introduced, which to a certain degree touches upon the issue of tumour variation. Now, entering the era of personalized medicine, modern nanomedicine is focusing more than ever on tumour heterogeneity and tailoring of regimens for individual patients. Nanotheranostics, which had been considered an interesting but not necessarily mainstream concept in nanomedicine, may find many opportunities in this campaign. Its utility is highly application-oriented, so it is important to break the dogma and embrace all possibilities that could make cancer treatment more efficient. A physically integrated diagnostic-plus-therapeutic nanoparticle may be advantageous in some applications but cumbersome in others. Hence, we suggest extending the concept of nanotheranostics to a broad approach that uses diagnosis to aid or guide nanoparticle therapy. The take-home messages are summarized in BOX 2.
Box 2. Take-home messages for the future developments of nanotheranostics.
Nanotheranostics
We suggest extending the concept of nanotheranostics beyond its current meaning of a nanomedicine that affords both diagnostic and therapeutic functions, to a broad approach that uses diagnosis to aid or guide nanoparticle therapy procedures.
Tumour heterogeneity and patient stratification
Conventional nanomedicine is founded on the enhanced permeability and retention (EPR) effect. This effect is, however, an oversimplified model because it largely neglects heterogeneity within and among tumours. Future nanomedicines should treat each tumour as a unique and complex organ. Instead of putting all factors under the EPR effect, it is worthwhile to consider each individual factor and combine the information for comprehensive prognosis, and to stratify patients based on prediction of nanotherapeutic efficacy. Nanotheranostics can have an important role in this new campaign.
Nanoparticle accumulation in target tissues
For tumour-targeting nanotheranostics (for example, chemotherapy) based on comprehensive tumour characterizations, it is possible to choose or even tailor nanoparticles for optimal tumour targeting; it is also possible to artificially modulate the tumour microenvironment to favour accumulation of nanoparticles within tumours. For nanotheranostics (for example, nanovaccines) that target secondary lymphoid organs, nanoparticle accumulation in these organs can be optimized.
Nanoparticle–body interactions
It is crucial to understand the interactions between nanoparticles and the human body (for example, tumour, healthy tissues, blood cells and proteins), to ultimately enhance nanoparticle delivery to tumour cells, prevent opsonization (chemical modification that makes the nanoparticle more readily identified by phagocytes) and reduce distribution in healthy tissues.
High efficacy and low toxicity
Although tumour uptake is a very important parameter when evaluating a nanomedicine, it is not the sole criterion. Reducing systemic toxicity and improving patient tolerance, for example, are crucial benefits of nanoparticle delivery and critical benchmarks in the clinical translation and implementation of nanotechnology.
Clinical translation
Clinical translation of cancer nanotheranostics requires commitment in scaled-up synthesis, detailed understanding of the interactions between cancer nanotheranostics and the human body, long-term assessment of toxicity, and establishment of regulatory protocols for cancer nanotheranostics.
In addition to the delivery of traditional chemotherapeutics, there is great promise in the delivery of immunotherapeutics using nanocarriers. Immunotherapy induces durable and systemic antitumour immunity, which is especially beneficial for the treatment of metastatic cancer. Many types of immunotherapeutics, including cancer therapeutic vaccine, can be efficiently delivered to targeted tissues for optimal therapeutic efficacy with reduced side effects. Moreover, companion diagnostics is critical for patient stratification and for the design of neoantigen-based personalized cancer immunotherapy. In this sense, almost all future nanomedicine may be considered nanotheranostics, given the increasing emphasis on the combination.
Despite the promise of nanotheranostics, there are multiple barriers to successful clinical translation. The community should seek in-depth understanding of nanoparticle–tumour interactions and cooperation between diagnosis and therapy. More efforts should also be directed at scaled-up synthesis, long-term assessment of toxicity and establishment of regulatory protocols for nanotheranostics183. Only then can we deliver the technology at the patient’s bedside for effective and personalized therapy.
Acknowledgments
We thank H. S. Eden for critically reading the manuscript. This work was supported in part by the Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, US National Institutes of Health (NIH); and by the Department of Defense (CDMRP grant CA140666), National Science Foundation (CAREER grant NSF1552617), University of Georgia–Georgia Regents University (seed grant) and NIH (R01 grants R01EB022596 and R01NS093314).
Footnotes
Competing interests statement
The authors declare no competing interests.
References
- 1.Wilhelm S, et al. Analysis of nanoparticle delivery to tumours. Nat Rev Mater. 2016;1:16014. This perspective stimulates an interesting discussion on the efficiency of nanoparticle delivery to tumours. [Google Scholar]
- 2.Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46:6387–6392. [PubMed] [Google Scholar]
- 3.Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33:941–951. doi: 10.1038/nbt.3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kobayashi H, Watanabe R, Choyke PL. Improving conventional enhanced permeability and retention (EPR) effects; what is the appropriate target? Theranostics. 2013;4:81–89. doi: 10.7150/thno.7193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Prabhakar U, et al. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. Cancer Res. 2013;73:2412–2417. doi: 10.1158/0008-5472.CAN-12-4561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev. 2014;66:2–25. doi: 10.1016/j.addr.2013.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Rajora AK, Ravishankar D, Osborn HMI, Greco F. Impact of the enhanced permeability and retention (EPR) effect and cathepsins levels on the activity of polymer–drug conjugates. Polymers. 2014;6:2186–2220. [Google Scholar]
- 8.Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17:20–37. doi: 10.1038/nrc.2016.108. A noteworthy and comprehensive review on cancer nanomedicine development. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kim TH, Lee S, Chen X. Nanotheranostics for personalized medicine. Expert Rev Mol Diagn. 2013;13:257–269. doi: 10.1586/erm.13.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mura S, Couvreur P. Nanotheranostics for personalized medicine. Adv Drug Deliv Rev. 2012;64:1394–1416. doi: 10.1016/j.addr.2012.06.006. [DOI] [PubMed] [Google Scholar]
- 11.Kunjachan S, Ehling J, Storm G, Kiessling F, Lammers T. Noninvasive imaging of nanomedicines and nanotheranostics: principles, progress, and prospects. Chem Rev. 2015;115:10907–10937. doi: 10.1021/cr500314d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Melancon MP, Stafford RJ, Li C. Challenges to effective cancer nanotheranostics. J Control Release. 2012;164:177–182. doi: 10.1016/j.jconrel.2012.07.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Mura S, Couvreur P. Nanotheranostics for Personalized Medicine. World Scientific; 2016. [DOI] [PubMed] [Google Scholar]
- 14.Karathanasis E, et al. Imaging nanoprobe for prediction of outcome of nanoparticle chemotherapy by using mammography. Radiology. 2009;250:398–406. doi: 10.1148/radiol.2502080801. [DOI] [PubMed] [Google Scholar]
- 15.Hansen AE, et al. Positron emission tomography based elucidation of the enhanced permeability and retention effect in dogs with cancer using copper-64 liposomes. ACS Nano. 2015;9:6985–6995. doi: 10.1021/acsnano.5b01324. [DOI] [PubMed] [Google Scholar]
- 16.Harrington KJ, et al. Effective targeting of solid tumors in patients with locally advanced cancers by radiolabeled pegylated liposomes. Clin Cancer Res. 2001;7:243–254. [PubMed] [Google Scholar]
- 17.Seymour LW, et al. Phase II studies of polymer–doxorubicin (PK1, FCE28068) in the treatment of breast, lung and colorectal cancer. Int J Oncol. 2009;34:1629–1636. doi: 10.3892/ijo_00000293. A clinical study showing that a radiolabelled polymer–drug formulation, which is essentially a nanotheranostic, can be used to predict patient responses to nanoparticle therapy. [DOI] [PubMed] [Google Scholar]
- 18.Arrieta O, et al. First-line chemotherapy with liposomal doxorubicin plus cisplatin for patients with advanced malignant pleural mesothelioma: phase II trial. Br J Cancer. 2012;106:1027–1032. doi: 10.1038/bjc.2012.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Arrieta O, et al. High liposomal doxorubicin tumour tissue distribution, as determined by radiopharmaceutical labelling with (99m)Tc-LD, is associated with the response and survival of patients with unresectable pleural mesothelioma treated with a combination of liposomal doxorubicin and cisplatin. Cancer Chemother Pharmacol. 2014;74:211–215. doi: 10.1007/s00280-014-2477-x. [DOI] [PubMed] [Google Scholar]
- 20.Hamburg MA, Collins FS. The path to personalized medicine. N Engl J Med. 2010;363:301–304. doi: 10.1056/NEJMp1006304. [DOI] [PubMed] [Google Scholar]
- 21.Weissleder R, Schwaiger MC, Gambhir SS, Hricak H. Imaging approaches to optimize molecular therapies. Sci Transl Med. 2016;8:355ps16. doi: 10.1126/scitranslmed.aaf3936. [DOI] [PubMed] [Google Scholar]
- 22.Miller MA, et al. Predicting therapeutic nanomedicine efficacy using a companion magnetic resonance imaging nanoparticle. Sci Transl Med. 2015;7:314ra183. doi: 10.1126/scitranslmed.aac6522. This study demonstrates the use of a clinically used nanoparticle formulation as a probe to accurately predict tumour accumulation and treatment efficacy of a range of nanotherapeutics. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.US National Library of Medicine. ClinicalTrials.gov. 2017 https://clinicaltrials.gov/ct2/show/NCT01770353.
- 24.Pérez-Medina C, et al. Nanoreporter PET predicts the efficacy of anti-cancer nanotherapy. Nat Commun. 2016;7:11838. doi: 10.1038/ncomms11838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Miao L, Huang L. Exploring the tumor microenvironment with nanoparticles. Cancer Treat Res. 2015;166:193–226. doi: 10.1007/978-3-319-16555-4_9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nat Rev Clin Oncol. 2010;7:653–664. doi: 10.1038/nrclinonc.2010.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Barua S, Mitragotri S. Challenges associated with penetration of nanoparticles across cell and tissue barriers: a review of current status and future prospects. Nano Today. 2014;9:223–243. doi: 10.1016/j.nantod.2014.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev Cancer. 2006;6:583–592. doi: 10.1038/nrc1893. [DOI] [PubMed] [Google Scholar]
- 29.Nakamura Y, Mochida A, Choyke PL, Kobayashi H. Nanodrug delivery: is the enhanced permeability and retention effect sufficient for curing cancer? Bioconjug Chem. 2016;27:2225–2238. doi: 10.1021/acs.bioconjchem.6b00437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lee J, Abdeen AA, Wycislo KL, Fan TM, Kilian KA. Interfacial geometry dictates cancer cell tumorigenicity. Nat Mater. 2016;15:856–862. doi: 10.1038/nmat4610. [DOI] [PubMed] [Google Scholar]
- 31.Manzoor AA, et al. Overcoming limitations in nanoparticle drug delivery: triggered, intravascular release to improve drug penetration into tumors. Cancer Res. 2012;72:5566–5575. doi: 10.1158/0008-5472.CAN-12-1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.US National Library of Medicine. ClinicalTrials.gov. 2017 https://clinicaltrials.gov/ct2/show/NCT00617981.
- 33.Smith BR, et al. Selective uptake of single-walled carbon nanotubes by circulating monocytes for enhanced tumour delivery. Nat Nanotechnol. 2014;9:481–487. doi: 10.1038/nnano.2014.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Matsumoto Y, et al. Vascular bursts enhance permeability of tumour blood vessels and improve nanoparticle delivery. Nat Nanotechnol. 2016;11:533–538. doi: 10.1038/nnano.2015.342. [DOI] [PubMed] [Google Scholar]
- 35.Huang J, et al. Magnetic nanoparticle facilitated drug delivery for cancer therapy with targeted and image-guided approaches. Adv Funct Mater. 2016;26:3818–3836. doi: 10.1002/adfm.201504185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kaida S, et al. Visible drug delivery by supramolecular nanocarriers directing to single-platformed diagnosis and therapy of pancreatic tumor model. Cancer Res. 2010;70:7031–7041. doi: 10.1158/0008-5472.CAN-10-0303. [DOI] [PubMed] [Google Scholar]
- 37.Viglianti BL, et al. Chemodosimetry of in vivo tumor liposomal drug concentration using MRI. Magn Reson Med. 2006;56:1011–1018. doi: 10.1002/mrm.21032. [DOI] [PubMed] [Google Scholar]
- 38.Ponce AM, et al. Magnetic resonance imaging of temperature-sensitive liposome release: drug dose painting and antitumor effects. J Natl Cancer Inst. 2007;99:53–63. doi: 10.1093/jnci/djk005. [DOI] [PubMed] [Google Scholar]
- 39.Wang D, et al. Novel dexamethasone–HPMA copolymer conjugate and its potential application in treatment of rheumatoid arthritis. Arthritis Res Ther. 2007;9:R2. doi: 10.1186/ar2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kaittanis C, et al. Environment-responsive nanophores for therapy and treatment monitoring via molecular MRI quenching. Nat Commun. 2014;5:3384. doi: 10.1038/ncomms4384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhao Z, et al. Real-time monitoring of arsenic trioxide release and delivery by activatable T1 imaging. ACS Nano. 2015;9:2749–2759. doi: 10.1021/nn506640h. [DOI] [PubMed] [Google Scholar]
- 42.Torchilin VP. Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat Rev Drug Discov. 2014;13:813–827. doi: 10.1038/nrd4333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat Mater. 2013;12:991–1003. doi: 10.1038/nmat3776. [DOI] [PubMed] [Google Scholar]
- 44.Gasselhuber A, et al. Targeted drug delivery by high intensity focused ultrasound mediated hyperthermia combined with temperature-sensitive liposomes: computational modelling and preliminary in vivo validation. Int J Hyperthermia. 2012;28:337–348. doi: 10.3109/02656736.2012.677930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hsiao YH, Kuo SJ, Tsai HD, Chou MC, Yeh GP. Clinical application of high-intensity focused ultrasound in cancer therapy. J Cancer. 2016;7:225–231. doi: 10.7150/jca.13906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.You Y, et al. Nanoparticle-enhanced synergistic HIFU ablation and transarterial chemoembolization for efficient cancer therapy. Nanoscale. 2016;8:4324–4339. doi: 10.1039/c5nr08292g. [DOI] [PubMed] [Google Scholar]
- 47.Onuki Y, Jacobs I, Artemov D, Kato Y. Noninvasive visualization of in vivo release and intratumoral distribution of surrogate MR contrast agent using the dual MR contrast technique. Biomaterials. 2010;31:7132–7138. doi: 10.1016/j.biomaterials.2010.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Langereis S, et al. A temperature-sensitive liposomal 1H CEST and 19F contrast agent for MR image-guided drug delivery. J Am Chem Soc. 2009;131:1380–1381. doi: 10.1021/ja8087532. [DOI] [PubMed] [Google Scholar]
- 49.Zhu X, et al. Long-circulating siRNA nanoparticles for validating Prohibitin1-targeted non-small cell lung cancer treatment. Proc Natl Acad Sci USA. 2015;112:7779–7784. doi: 10.1073/pnas.1505629112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sahin U, Kariko K, Tureci O. mRNA-based therapeutics — developing a new class of drugs. Nat Rev Drug Discov. 2014;13:759–780. doi: 10.1038/nrd4278. [DOI] [PubMed] [Google Scholar]
- 51.Kormann MSD, et al. Expression of therapeutic proteins after delivery of chemically modified mRNA in mice. Nat Biotechnol. 2011;29:154–157. doi: 10.1038/nbt.1733. [DOI] [PubMed] [Google Scholar]
- 52.Idris NM, et al. In vivo photodynamic therapy using upconversion nanoparticles as remote-controlled nanotransducers. Nat Med. 2012;18:1580–1585. doi: 10.1038/nm.2933. [DOI] [PubMed] [Google Scholar]
- 53.Lecaros RL, Huang L, Lee TC, Hsu YC. Nanoparticle delivered VEGF-A siRNA enhances photodynamic therapy for head and neck cancer treatment. Mol Ther. 2016;24:106–116. doi: 10.1038/mt.2015.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Muhanna N, et al. Phototheranostic porphyrin nanoparticles enable visualization and targeted treatment of head and neck cancer in clinically relevant models. Theranostics. 2015;5:1428–1443. doi: 10.7150/thno.13451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jin CS, et al. Nanoparticle-enabled selective destruction of prostate tumor using MRI-guided focal photothermal therapy. Prostate. 2016;76:1169–1181. doi: 10.1002/pros.23203. [DOI] [PubMed] [Google Scholar]
- 56.US National Library of Medicine. ClinicalTrials.gov. 2016 https://clinicaltrials.gov/ct2/show/NCT02680535.
- 57.Muhanna N, et al. Multimodal image-guided surgical and photodynamic interventions in head and neck cancer: from primary tumor to metastatic drainage. Clin Cancer Res. 2016;22:961–970. doi: 10.1158/1078-0432.CCR-15-1235. [DOI] [PubMed] [Google Scholar]
- 58.Lin J, et al. Photosensitizer-loaded gold vesicles with strong plasmonic coupling effect for imaging-guided photothermal/photodynamic therapy. ACS Nano. 2013;7:5320–5329. doi: 10.1021/nn4011686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lin J, et al. Multimodal-imaging-guided cancer phototherapy by versatile biomimetic theranostics with UV and γ-irradiation protection. Adv Mater. 2016;28:3273–3279. doi: 10.1002/adma.201505700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lu W, et al. Effects of photoacoustic imaging and photothermal ablation therapy mediated by targeted hollow gold nanospheres in an orthotopic mouse xenograft model of glioma. Cancer Res. 2011;71:6116–6121. doi: 10.1158/0008-5472.CAN-10-4557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kim JW, Galanzha EI, Shashkov EV, Moon HM, Zharov VP. Golden carbon nanotubes as multimodal photoacoustic and photothermal high-contrast molecular agents. Nat Nanotechnol. 2009;4:688–694. doi: 10.1038/nnano.2009.231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jaffray DA. Image-guided radiotherapy: from current concept to future perspectives. Nat Rev Clin Oncol. 2012;9:688–699. doi: 10.1038/nrclinonc.2012.194. [DOI] [PubMed] [Google Scholar]
- 63.Strom HH, et al. Concurrent palliative chemoradiation leads to survival and quality of life benefits in poor prognosis stage III non-small-cell lung cancer: a randomised trial by the Norwegian lung cancer study group. Br J Cancer. 2013;109:1467–1475. doi: 10.1038/bjc.2013.466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lukianova-Hleb EY, et al. On-demand intracellular amplification of chemoradiation with cancer-specific plasmonic nanobubbles. Nat Med. 2014;20:778–784. doi: 10.1038/nm.3484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Begg AC, Stewart FA, Vens C. Strategies to improve radiotherapy with targeted drugs. Nat Rev Cancer. 2011;11:239–253. doi: 10.1038/nrc3007. [DOI] [PubMed] [Google Scholar]
- 66.Adams SR, et al. Anti-tubulin drugs conjugated to anti-ErbB antibodies selectively radiosensitize. Nat Commun. 2016;7:13019. doi: 10.1038/ncomms13019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Strom TJ, et al. Increased acute mortality with chemoradiotherapy for locally advanced head and neck cancer in patients ≥70 years. J Geriatr Oncol. 2017;8:50–55. doi: 10.1016/j.jgo.2016.09.003. [DOI] [PubMed] [Google Scholar]
- 68.Koukourakis MI, et al. Liposomal doxorubicin and conventionally fractionated radiotherapy in the treatment of locally advanced non-small-cell lung cancer and head and neck cancer. J Clin Oncol. 1999;17:3512–3521. doi: 10.1200/JCO.1999.17.11.3512. [DOI] [PubMed] [Google Scholar]
- 69.Eblan MJ, Wang AZ. Improving chemoradiotherapy with nanoparticle therapeutics. Transl Cancer Res. 2013;2:320–329. doi: 10.3978/j.issn.2218-676X.2013.08.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Werner ME, et al. Preclinical evaluation of Genexol-PM, a nanoparticle formulation of paclitaxel, as a novel radiosensitizer for the treatment of non-small cell lung cancer. Int J Radiat Oncol Biol Phys. 2013;86:463–468. doi: 10.1016/j.ijrobp.2013.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wang EC, et al. Nanoparticle formulations of histone deacetylase inhibitors for effective chemoradiotherapy in solid tumors. Biomaterials. 2015;51:208–215. doi: 10.1016/j.biomaterials.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Caster JM, et al. Nanoparticle delivery of chemosensitizers improve chemotherapy efficacy without incurring additional toxicity. Nanoscale. 2015;7:2805–2811. doi: 10.1039/c4nr07102f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Karve S, et al. Revival of the abandoned therapeutic wortmannin by nanoparticle drug delivery. Proc Natl Acad Sci USA. 2012;109:8230–8235. doi: 10.1073/pnas.1120508109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Baumann M, et al. Radiation oncology in the era of precision medicine. Nat Rev Cancer. 2016;16:234–249. doi: 10.1038/nrc.2016.18. [DOI] [PubMed] [Google Scholar]
- 75.Kunjachan S, et al. Nanoparticle mediated tumor vascular disruption: a novel strategy in radiation therapy. Nano Lett. 2015;15:7488–7496. doi: 10.1021/acs.nanolett.5b03073. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 76.US National Library of Medicine. ClinicalTrials.gov. 2017 https://clinicaltrials.gov/ct2/show/NCT02379845?term.
- 77.Bonvalot S, et al. First-in-human study testing a new radioenhancer using nanoparticles (NBTXR3) activated by radiation therapy in patients with locally advanced soft tissue sarcomas. Clin Cancer Res. 2017;23:908–917. doi: 10.1158/1078-0432.CCR-16-1297. A first-in-human study which uses hafnium oxide nanoparticles (NBTXR3) as radiosensitizers to aid cancer therapy. [DOI] [PubMed] [Google Scholar]
- 78.Kotb S, et al. Gadolinium-based nanoparticles and radiation therapy for multiple brain melanoma metastases: proof of concept before phase I trial. Theranostics. 2016;6:418–427. doi: 10.7150/thno.14018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Detappe A, et al. Advanced multimodal nanoparticles delay tumor progression with clinical radiation therapy. J Control Release. 2016;238:103–113. doi: 10.1016/j.jconrel.2016.07.021. [DOI] [PubMed] [Google Scholar]
- 80.US National Library of Medicine. ClinicalTrials.gov. 2016 https://clinicaltrials.gov/ct2/show/NCT02820454.
- 81.Chen W, Zhang J. Using nanoparticles to enable simultaneous radiation and photodynamic therapies for cancer treatment. J Nanosci Nanotechnol. 2006;6:1159–1166. doi: 10.1166/jnn.2006.327. [DOI] [PubMed] [Google Scholar]
- 82.Pogue BW, et al. Photodynamic therapy with verteporfin in the radiation-induced fibrosarcoma-1 tumor causes enhanced radiation sensitivity. Cancer Res. 2003;63:1025–1033. [PubMed] [Google Scholar]
- 83.Bulin AL, et al. X-Ray-induced singlet oxygen activation with nanoscintillator-coupled porphyrins. J Phys Chem C. 2013;117:21583–21589. [Google Scholar]
- 84.Tang YG, Hu J, Elmenoufy AH, Yang XL. Highly efficient FRET system capable of deep photodynamic therapy established on X-ray excited mesoporous LaF3:Tb scintillating nanoparticles. ACS Appl Mater Interfaces. 2015;7:12261–12269. doi: 10.1021/acsami.5b03067. [DOI] [PubMed] [Google Scholar]
- 85.Chen H, et al. Nanoscintillator-mediated X-ray inducible photodynamic therapy for in vivo cancer treatment. Nano Lett. 2015;15:2249–2256. doi: 10.1021/nl504044p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wang GD, et al. X-Ray induced photodynamic therapy: a combination of radiotherapy and photodynamic therapy. Theranostics. 2016;6:2295–2305. doi: 10.7150/thno.16141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kotagiri N, Sudlow GP, Akers WJ, Achilefu S. Breaking the depth dependency of phototherapy with Cerenkov radiation and low-radiance-responsive nanophotosensitizers. Nat Nanotechnol. 2015;10:370–379. doi: 10.1038/nnano.2015.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lukianova-Hleb EY, et al. Intraoperative diagnostics and elimination of residual microtumours with plasmonic nanobubbles. Nat Nanotechnol. 2016;11:525–532. doi: 10.1038/nnano.2015.343. [DOI] [PubMed] [Google Scholar]
- 89.Kircher MF, et al. A brain tumor molecular imaging strategy using a new triple-modality MRI–photoacoustic–Raman nanoparticle. Nat Med. 2012;18:829–834. doi: 10.1038/nm.2721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Phillips E, et al. Clinical translation of an ultrasmall inorganic optical-PET imaging nanoparticle probe. Sci Transl Med. 2014;6:260ra149. doi: 10.1126/scitranslmed.3009524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kim SE, et al. Ultrasmall nanoparticles induce ferroptosis in nutrient-deprived cancer cells and suppress tumour growth. Nat Nanotechnol. 2016;11:977–985. doi: 10.1038/nnano.2016.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Brindle K. New approaches for imaging tumour responses to treatment. Nat Rev Cancer. 2008;8:94–107. doi: 10.1038/nrc2289. [DOI] [PubMed] [Google Scholar]
- 93.Shuhendler AJ, et al. Molecular magnetic resonance imaging of tumor response to therapy. Sci Rep. 2015;5:14759. doi: 10.1038/srep14759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Bussink J, Kaanders JH, van der Graaf WT, Oyen WJ. PET–CT for radiotherapy treatment planning and response monitoring in solid tumors. Nat Rev Clin Oncol. 2011;8:233–242. doi: 10.1038/nrclinonc.2010.218. [DOI] [PubMed] [Google Scholar]
- 95.Jorgensen JT, et al. Single particle and PET-based platform for identifying optimal plasmonic mano-heaters for photothermal cancer therapy. Sci Rep. 2016;6:30076. doi: 10.1038/srep30076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wang Y, et al. A nanoparticle-based strategy for the imaging of a broad range of tumours by nonlinear amplification of microenvironment signals. Nat Mater. 2014;13:204–212. doi: 10.1038/nmat3819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Mi P, et al. A pH-activatable nanoparticle with signal-amplification capabilities for non-invasive imaging of tumour malignancy. Nat Nanotechnol. 2016;11:724–730. doi: 10.1038/nnano.2016.72. [DOI] [PubMed] [Google Scholar]
- 98.Lee S, Xie J, Chen X. Activatable molecular probes for cancer imaging. Curr Top Med Chem. 2010;10:1135–1144. doi: 10.2174/156802610791384270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Guo J, et al. 18F-alfatide II and 18F-FDG dual-tracer dynamic PET for parametric, early prediction of tumor response to therapy. J Nucl Med. 2014;55:154–160. doi: 10.2967/jnumed.113.122069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sun X, et al. 18F-FPPRGD2 and 18F-FDG PET of response to abraxane therapy. J Nucl Med. 2011;52:140–146. doi: 10.2967/jnumed.110.080606. [DOI] [PubMed] [Google Scholar]
- 101.Notohamiprodjo M, et al. Combined diffusion-weighted, blood oxygen level-dependent, and dynamic contrast-enhanced MRI for characterization and differentiation of renal cell carcinoma. Acad Radiol. 2013;20:685–693. doi: 10.1016/j.acra.2013.01.015. [DOI] [PubMed] [Google Scholar]
- 102.Mittra ES, et al. Pilot pharmacokinetic and dosimetric studies of 18F-FPPRGD2: a PET radiopharmaceutical agent for imaging αv β3 integrin levels. Radiology. 2011;260:182–191. doi: 10.1148/radiol.11101139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Park SY, et al. Assessment of early renal allograft dysfunction with blood oxygenation level-dependent MRI and diffusion-weighted imaging. Eur J Radiol. 2014;83:2114–2121. doi: 10.1016/j.ejrad.2014.09.017. [DOI] [PubMed] [Google Scholar]
- 104.Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity in the clinic. Nature. 2013;501:355–364. doi: 10.1038/nature12627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Alizadeh AA, et al. Toward understanding and exploiting tumor heterogeneity. Nat Med. 2015;21:846–853. doi: 10.1038/nm.3915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501:346–354. doi: 10.1038/nature12626. [DOI] [PubMed] [Google Scholar]
- 107.Taurin S, Nehoff H, Greish K. Anticancer nanomedicine and tumor vascular permeability; where is the missing link? J Control Release. 2012;164:265–275. doi: 10.1016/j.jconrel.2012.07.013. [DOI] [PubMed] [Google Scholar]
- 108.Toy R, et al. Multimodal in vivo imaging exposes the voyage of nanoparticles in tumor microcirculation. ACS Nano. 2013;7:3118–3129. doi: 10.1021/nn3053439. A good example of using multiple imaging methods to comprehensively characterize tumours and understand the impact of individual factors on nanoparticle–tumour interactions. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ullrich RT, et al. In-vivo visualization of tumor microvessel density and response to anti-angiogenic treatment by high resolution MRI in mice. PLoS ONE. 2011;6:e19592. doi: 10.1371/journal.pone.0019592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Theek B, et al. Characterizing EPR-mediated passive drug targeting using contrast-enhanced functional ultrasound imaging. J Control Release. 2014;182:83–89. doi: 10.1016/j.jconrel.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Chen B, et al. Tumor vascular permeabilization by vascular-targeting photosensitization: effects, mechanism, and therapeutic implications. Clin Cancer Res. 2006;12:917–923. doi: 10.1158/1078-0432.CCR-05-1673. [DOI] [PubMed] [Google Scholar]
- 112.Hamzah J, et al. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature. 2008;453:410–414. doi: 10.1038/nature06868. [DOI] [PubMed] [Google Scholar]
- 113.Jain R. Measurements of tumor vascular leakiness using DCE in brain tumors: clinical applications. NMR Biomed. 2013;26:1042–1049. doi: 10.1002/nbm.2994. [DOI] [PubMed] [Google Scholar]
- 114.Goyen M. Gadofosveset-enhanced magnetic resonance angiography. Vasc Health Risk Manag. 2008;4:1–9. doi: 10.2147/vhrm.2008.04.01.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Niu G, et al. In vivo labeling of serum albumin for PET. J Nucl Med. 2014;55:1150–1156. doi: 10.2967/jnumed.114.139642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhang J, et al. Clinical translation of an albumin-binding PET radiotracer 68Ga-NEB. J Nucl Med. 2015;56:1609–1614. doi: 10.2967/jnumed.115.159640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Zhang W, et al. Potential applications of using 68Ga-Evans blue PET/CT in the evaluation of lymphatic disorder: preliminary observations. Clin Nucl Med. 2016;41:302–308. doi: 10.1097/RLU.0000000000001171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Stapleton S, et al. A mathematical model of the enhanced permeability and retention effect for liposome transport in solid tumors. PLoS ONE. 2013;8:e81157. doi: 10.1371/journal.pone.0081157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wong AD, Ye M, Ulmschneider MB, Searson PC. Quantitative analysis of the enhanced permeation and retention (EPR) effect. PLoS ONE. 2015;10:e0123461. doi: 10.1371/journal.pone.0123461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Tang L, et al. Investigating the optimal size of anticancer nanomedicine. Proc Natl Acad Sci USA. 2014;111:15344–15349. doi: 10.1073/pnas.1411499111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Cabral H, et al. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nat Nanotechnol. 2011;6:815–823. doi: 10.1038/nnano.2011.166. A systematic study that compared the accumulation and therapeutic efficacy of different-sized, long-circulating, drug-loaded polymeric micelles. The study was conducted in animals bearing tumours of either high or low permeability. [DOI] [PubMed] [Google Scholar]
- 122.Salvati A, et al. Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. Nat Nanotechnol. 2013;8:137–143. doi: 10.1038/nnano.2012.237. [DOI] [PubMed] [Google Scholar]
- 123.Liu R, Jiang W, Walkey CD, Chan WCW, Cohen Y. Prediction of nanoparticles–cell association based on corona proteins and physicochemical properties. Nanoscale. 2015;7:9664–9675. doi: 10.1039/c5nr01537e. [DOI] [PubMed] [Google Scholar]
- 124.Wu W, et al. Tumor-targeted aggregation of pH-sensitive nanocarriers for enhanced retention and rapid intracellular drug release. Polymer Chem. 2014;5:5668–5679. [Google Scholar]
- 125.Zhang D, et al. In situ formation of nanofibers from purpurin18–peptide conjugates and the assembly induced retention effect in tumor sites. Adv Mater. 2015;27:6125–6130. doi: 10.1002/adma.201502598. [DOI] [PubMed] [Google Scholar]
- 126.Wong C, et al. Multistage nanoparticle delivery system for deep penetration into tumor tissue. Proc Natl Acad Sci USA. 2011;108:2426–2431. doi: 10.1073/pnas.1018382108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Li HJ, et al. Stimuli-responsive clustered nanoparticles for improved tumor penetration and therapeutic efficacy. Proc Natl Acad Sci USA. 2016;113:4164–4169. doi: 10.1073/pnas.1522080113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Xu R, et al. An injectable nanoparticle generator enhances delivery of cancer therapeutics. Nat Biotechnol. 2016;34:414–418. doi: 10.1038/nbt.3506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Lee CG, et al. Anti-vascular endothelial growth factor treatment augments tumor radiation response under normoxic or hypoxic conditions. Cancer Res. 2000;60:5565–5570. [PubMed] [Google Scholar]
- 130.Tong RT, et al. Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors. Cancer Res. 2004;64:3731–3736. doi: 10.1158/0008-5472.CAN-04-0074. [DOI] [PubMed] [Google Scholar]
- 131.Willett CG, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med. 2004;10:145–147. doi: 10.1038/nm988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Heist RS, et al. Improved tumor vascularization after anti-VEGF therapy with carboplatin and nab-paclitaxel associates with survival in lung cancer. Proc Natl Acad Sci USA. 2015;112:1547–1552. doi: 10.1073/pnas.1424024112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Pham E, et al. Preclinical efficacy of bevacizumab with CRLX101, an investigational nanoparticle-drug conjugate, in treatment of metastatic triple-negative breast cancer. Cancer Res. 2016;76:4493–4503. doi: 10.1158/0008-5472.CAN-15-3435. [DOI] [PubMed] [Google Scholar]
- 134.Nehoff H, Parayath NN, Domanovitch L, Taurin S, Greish K. Nanomedicine for drug targeting: strategies beyond the enhanced permeability and retention effect. Int J Nanomed. 2014;9:2539–2555. doi: 10.2147/IJN.S47129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Snyder JW, Greco WR, Bellnier DA, Vaughan L, Henderson BW. Photodynamic therapy: a means to enhanced drug delivery to tumors. Cancer Res. 2003;63:8126–8131. [PubMed] [Google Scholar]
- 136.Zhen Z, et al. Tumor vasculature targeted photodynamic therapy for enhanced delivery of nanoparticles. ACS Nano. 2014;8:6004–6013. doi: 10.1021/nn501134q. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Carpentier A, et al. Clinical trial of blood–brain barrier disruption by pulsed ultrasound. Sci Transl Med. 2016;8:343re2. doi: 10.1126/scitranslmed.aaf6086. A first-in-human trial showing initial successes on disrupting the blood–brain barrier with ultrasound. [DOI] [PubMed] [Google Scholar]
- 138.Leinenga G, Gotz J. Scanning ultrasound removes amyloid-β and restores memory in an Alzheimer’s disease mouse model. Sci Transl Med. 2015;7:278ra33. doi: 10.1126/scitranslmed.aaa2512. [DOI] [PubMed] [Google Scholar]
- 139.Kobayashi H, et al. Application of a macromolecular contrast agent for detection of alterations of tumor vessel permeability induced by radiation. Clin Cancer Res. 2004;10:7712–7720. doi: 10.1158/1078-0432.CCR-04-1175. [DOI] [PubMed] [Google Scholar]
- 140.Sano K, Nakajima T, Choyke PL, Kobayashi H. Markedly enhanced permeability and retention effects induced by photo-immunotherapy of tumors. ACS Nano. 2013;7:717–724. doi: 10.1021/nn305011p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Suzuki M, Hori K, Abe I, Saito S, Sato H. A new approach to cancer chemotherapy: selective enhancement of tumor blood flow with angiotensin II. J Natl Cancer Inst. 1981;67:663–669. [PubMed] [Google Scholar]
- 142.Li CJ, Miyamoto Y, Kojima Y, Maeda H. Augmentation of tumour delivery of macromolecular drugs with reduced bone marrow delivery by elevating blood pressure. Br J Cancer. 1993;67:975–980. doi: 10.1038/bjc.1993.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y, Jain RK. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci USA. 2011;108:2909–2914. doi: 10.1073/pnas.1018892108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Buckway B, Frazier N, Gormley AJ, Ray A, Ghandehari H. Gold nanorod-mediated hyperthermia enhances the efficacy of HPMA copolymer–90Y conjugates in treatment of prostate tumors. Nucl Med Biol. 2014;41:282–289. doi: 10.1016/j.nucmedbio.2013.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Tamarov KP, et al. Radio frequency radiation-induced hyperthermia using Si nanoparticle-based sensitizers for mild cancer therapy. Sci Rep. 2014;4:7034. doi: 10.1038/srep07034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Yin D, et al. Convection-enhanced delivery improves distribution and efficacy of tumor-selective retroviral replicating vectors in a rodent brain tumor model. Cancer Gene Ther. 2013;20:336–341. doi: 10.1038/cgt.2013.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Perlstein B, et al. Convection-enhanced delivery of maghemite nanoparticles: Increased efficacy and MRI monitoring. Neuro Oncol. 2008;10:153–161. doi: 10.1215/15228517-2008-002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.McNeil SE. Evaluation of nanomedicines: stick to the basics. Nat Rev Mater. 2016;1:16073. [Google Scholar]
- 149.Lammers T, et al. Cancer nanomedicine: is targeting our target? Nat Rev Mater. 2016;1:16069. doi: 10.1038/natrevmats.2016.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Schork NJ. Personalized medicine: time for one-person trials. Nature. 2015;520:609–611. doi: 10.1038/520609a. [DOI] [PubMed] [Google Scholar]
- 151.Scuffham PA, et al. Using N-of-1 trials to improve patient management and save costs. J Gen Intern Med. 2010;25:906–913. doi: 10.1007/s11606-010-1352-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.US National Library of Medicine. ClinicalTrials.gov. 2015 https://clinicaltrials.gov/ct2/show/NCT02116621.
- 153.US National Library of Medicine. ClinicalTrials.gov. 2017 https://clinicaltrials.gov/ct2/show/NCT02142036.
- 154.Irvine DJ. Materializing the future of vaccines and immunotherapy. Nat Rev Mater. 2016;1:15008. [Google Scholar]
- 155.Hubbell JA, Thomas SN, Swartz MA. Materials engineering for immunomodulation. Nature. 2009;462:449–460. doi: 10.1038/nature08604. [DOI] [PubMed] [Google Scholar]
- 156.Stephan MT, Moon JJ, Um SH, Bershteyn A, Irvine DJ. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat Med. 2010;16:1035–1041. doi: 10.1038/nm.2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Rosenberg SA. Raising the bar: the curative potential of human cancer immunotherapy. Sci Transl Med. 2012;4:127ps8. doi: 10.1126/scitranslmed.3003634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hailemichael Y, et al. Persistent antigen at vaccination sites induces tumor-specific CD8+ T cell sequestration, dysfunction and deletion. Nat Med. 2013;19:465–472. doi: 10.1038/nm.3105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Moon JJ, Huang B, Irvine DJ. Engineering nano- and microparticles to tune immunity. Adv Mater. 2012;24:3724–3746. doi: 10.1002/adma.201200446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Irvine DJ, Hanson MC, Rakhra K, Tokatlian T. Synthetic nanoparticles for vaccines and immunotherapy. Chem Rev. 2015;115:11109–11146. doi: 10.1021/acs.chemrev.5b00109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Zhu G, et al. DNA–inorganic hybrid nanovaccine for cancer immunotherapy. Nanoscale. 2016;8:6684–6692. doi: 10.1039/c5nr08821f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Kuai R, Ochyl LJ, Bahjat KS, Schwendeman A, Moon JJ. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat Mater. 2017;16:489–496. doi: 10.1038/nmat4822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Nguyen DN, et al. Lipid-derived nanoparticles for immunostimulatory RNA adjuvant delivery. Proc Natl Acad Sci USA. 2012;109:E797–E803. doi: 10.1073/pnas.1121423109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Jeanbart L, Swartz MA. Engineering opportunities in cancer immunotherapy. Proc Natl Acad Sci USA. 2015;112:14467–14472. doi: 10.1073/pnas.1508516112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Shao K, et al. Nanoparticle-based immunotherapy for cancer. ACS Nano. 2015;9:16–30. doi: 10.1021/nn5062029. [DOI] [PubMed] [Google Scholar]
- 166.Radovic-Moreno AF, et al. Immunomodulatory spherical nucleic acids. Proc Natl Acad Sci USA. 2015;112:3892–3897. doi: 10.1073/pnas.1502850112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Koshy ST, Cheung AS, Gu L, Graveline AR, Mooney DJ. Liposomal delivery enhances immune activation by STING agonists for cancer immunotherapy. Adv Biosystems. 2017;1:1600013. doi: 10.1002/adbi.201600013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Hanson MC, et al. Nanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvants. J Clin Invest. 2015;125:2532–2546. doi: 10.1172/JCI79915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Liu H, et al. Structure-based programming of lymph-node targeting in molecular vaccines. Nature. 2014;507:519–522. doi: 10.1038/nature12978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Guo Y, et al. Erythrocyte membrane-enveloped polymeric nanoparticles as nanovaccine for induction of antitumor immunity against melanoma. ACS Nano. 2015;9:6918–6933. doi: 10.1021/acsnano.5b01042. [DOI] [PubMed] [Google Scholar]
- 171.Fang RH, et al. Cancer cell membrane-coated nanoparticles for anticancer vaccination and drug delivery. Nano Lett. 2014;14:2181–2188. doi: 10.1021/nl500618u. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Cheung AS, Koshy ST, Stafford AG, Bastings MMC, Mooney DJ. Adjuvant-loaded subcellular vesicles derived from disrupted cancer cells for cancer vaccination. Small. 2016;12:2321–2333. doi: 10.1002/smll.201600061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15:760–770. doi: 10.1038/nri3921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Guermonprez P, et al. ER–phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells. Nature. 2003;425:397–402. doi: 10.1038/nature01911. [DOI] [PubMed] [Google Scholar]
- 175.Burgdorf S, Schölz C, Kautz A, Tampé R, Kurts C. Spatial and mechanistic separation of cross-presentation and endogenous antigen presentation. Nat Immunol. 2008;9:558–566. [Google Scholar]
- 176.Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–420. doi: 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Schlake T, Thess A, Fotin-Mleczek M, Kallen KJ. Developing mRNA-vaccine technologies. RNA Biol. 2012;9:1319–1330. doi: 10.4161/rna.22269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Benteyn D, Heirman C, Bonehill A, Thielemans K, Breckpot K. mRNA-based dendritic cell vaccines. Expert Rev Vaccines. 2014;14:161–176. doi: 10.1586/14760584.2014.957684. [DOI] [PubMed] [Google Scholar]
- 179.Sahin U, Karikó K, Türeci Ö. mRNA-based therapeutics — developing a new class of drugs. Nat Rev Drug Discov. 2014;13:759–780. doi: 10.1038/nrd4278. [DOI] [PubMed] [Google Scholar]
- 180.Li J, et al. Structurally programmed assembly of translation initiation nanoplex for superior mRNA delivery. ACS Nano. 2017;11:2531–2544. doi: 10.1021/acsnano.6b08447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Kranz LM, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534:396–401. doi: 10.1038/nature18300. [DOI] [PubMed] [Google Scholar]
- 182.Kreiter S, et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature. 2015;520:692–696. doi: 10.1038/nature14426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Mitragotri S, et al. Drug delivery research for the future: expanding the nano horizons and beyond. J Control Release. 2017;246:183–184. doi: 10.1016/j.jconrel.2017.01.011. [DOI] [PubMed] [Google Scholar]