Abstract
Two camps have emerged for targeting nanoparticles to specific organs and cell types: affinity moiety targeting, and physicochemical tropism. Here we directly compare and combine both, using intravenous (IV) lipid nanoparticles (LNPs) designed to target the lungs. We utilized PECAM antibodies as affinity moieties and cationic lipids for physicochemical tropism. These methods yield nearly identical lung uptake, but aPECAM LNPs show higher endothelial specificity. LNPs combining these targeting methods had >2-fold higher lung uptake than either method alone, and markedly enhanced epithelial uptake. To determine if lung uptake is because the lungs are the first organ downstream of IV injection, we compared IV vs. intra-arterial (IA) injection into the carotid artery, finding that IA combined-targeting LNPs achieve 35% of the injected dose per gram (%ID/g) in the first-pass organ, the brain, among the highest reported. Thus, combining affinity moiety and physicochemical strategies provides benefits that neither targeting method achieves alone.
Keywords: Extrahepatic delivery, physicochemical, antibody-mediated, lung targeting, cell-type expression
Graphical Abstract
Nanoparticles have held the promise of selectively targeting specific cell types and organs, though that goal has remained largely elusive for organs other than the liver, the natural clearance organ for intravenous nanoscale materials1–5. In working to achieve this targeting goal in non-liver organs, two strategies have predominated, termed here “affinity moiety targeting” and “physicochemical tropism.”
“Affinity moiety targeting,” developed first, employs “affinity moieties” which bind specifically to chosen epitopes on target cells, e.g., conjugating nanoparticles to monoclonal antibodies that have affinity for the target cell. First achieved by Torchilin and others, who showed the benefits of conjugating liposomes to monoclonal antibodies6–11. Numerous other affinity moieties have been used besides monoclonal antibodies, including small molecules that bind to receptors on target cells, aptamers, antibody fragments (Fab, F(ab’)2 , etc.)12–21. At least 10 affinity-moiety-targeted nanoparticles have reached clinical studies, though none are approved19,22,23.
“Physicochemical tropism” utilizes nanoparticle physical properties (size, shape, charge) or chemical features (binding to specific endogenous proteins because of the nanoparticles’ chemical makeup) to effect organ tropism. In general, physicochemical tropism is not easy to design from first principles, and therefore commonly achieved by screening libraries of nanoparticles with slightly varying formulations to find ones with tropism for a particular cell or organ, as exemplified by lipid nanoparticle (LNP) screens, especially Anderson’s studies of the early 2010s24–27. These optimized searches through LNP formulation space have produced numerous formulations with physicochemical tropism for particular organs or cell types, ranging from the lungs to spleen, and achieving cell-type-specificity to macrophages and T-cells28–32. The first FDA-approved RNA-loaded lipid nanoparticle (LNP), patisiran, approved in 2018, has physicochemical tropism to the plasma protein ApoE, which selectively shuttles the LNPs to hepatocytes33.
Much has been written comparing affinity moiety targeting and physicochemical tropism24–32,34–44. Advantages cited for affinity moieties include a known mechanism of action, enabling rational engineering. Physicochemical targeting usually does not have a known mechanism of action (ApoE binding is a rare exception), and therefore rational engineering is difficult. However, physicochemical targeting has a large advantage for industrialization, as affinity moieties such as antibodies are not easy to conjugate to nanoparticles and purify at scale45,46. Physicochemical targeting, for LNPs at least, usually just involves changing the lipid formulation, which is facile at industrial scale47. Thus, each approach has relative advantages and disadvantages. These two approaches have rarely been tested head-to-head, especially for LNPs. This lack of head-to-head testing might be because in general these two approaches are performed in separate labs, falling into separate “camps.”
Here, we directly compare cell and organ delivery efficiency and specificity when delivering to a single organ, the lung, using PECAM for affinity targeting vs. cationic lipids for physicochemical targeting. We do this for LNPs, since they are the leading nanoparticle format currently under development in industry, with billions invested in the last few years48,49. We chose to target the lungs because many formulations with both affinity and physicochemical targeting have been created with the goal of ameliorating alveolar diseases, especially acute respiratory distress syndrome (ARDS), the alveolar inflammatory disease that was the primary cause of death during the COVID-19 pandemic, and still causes >75,000 deaths each year from non-COVID types of ARDS50–55.
For affinity-moiety-targeting to the alveolar vasculature, we chose LNPs conjugated to antibodies against PECAM-1. Compared to untargeted LNPs, PECAM targeting has been shown to increase mRNA delivery and protein expression in the lung by ~200-fold and 25-fold respectively36. PECAM has been compared to other affinity moieties, and none perform markedly better with respect to lung uptake. For physicochemical tropism to the lungs, we chose LNPs incorporating a cationic lipid. Numerous LNPs have been formulated that have physicochemical tropism to the lungs, with the commonality that nearly all possess lipids bearing positive charge within biologically-relevant pH ranges. Some of these are ionizable lipids with tertiary amines, while others are permanently cationic with quaternary amines28,30,56. Here we chose the most cited such LNP formulation, which includes DOTAP, a permanently cationic lipid with a quaternary amine,28–30,57 shown to drive expression of mRNA-luciferase to the lungs28–30,57.
In comparing aPECAM and DOTAP LNPs, we find that they have nearly identical localization to the lungs, despite dramatically different targeting mechanisms. We then show that combining PECAM antibodies and DOTAP into a single LNP produces a synergistic benefit, achieving the highest reported delivery of an LNP to the lungs, suggesting that bringing together these two approaches can produce higher uptake than either alone.
To begin these studies, we chose our the ionizable lipid cKK-E12 for our base formulation, which others in the field have shown drives high expression from mRNA cargo58–60. We formulated affinity-moiety-targeted LNPs by conjugating aPECAM antibodies to the LNP surface (herein referred to as “aPECAM LNPs”). We fabricated LNPs with physicochemical tropism to the lungs by adding in a cationic lipid, DOTAP, as published previously (herein referred to as “DOTAP LNPs”). “Combined LNPs” were formulated by conjugating aPECAM antibody to the surface of DOTAP LNPs (Fig 1A). We added a radiotracer lipid (Indium-111-DTPA-PE) to the LNPs and intravenously (IV) injected them into healthy mice at a dose of 7.5μg mRNA per mouse. After allowing the LNPs to circulate for 30 minutes, the amount of radioactivity in each organ was measured and recorded as the percent of injected dose per gram of tissue (%ID/g). We found that aPECAM and DOTAP LNPs achieved essentially identical lung uptake, despite different targeting mechanisms. LNPs combining the two targeting techniques localize to the lungs >2-fold more than either DOTAP or aPECAM LNPs (Fig 1B). Similarly, Combined LNPs have a >2-fold higher lung-to-liver localization ratio than aPECAM or DOTAP LNPs (Fig 1C).
Figure 1: LNPs formulated with cationic lipids and targeted to PECAM show improved lung localization and expression.
A. Formulations and molar percentages of aPECAM, DOTAP, and Combined LNPs. B. Biodistributions of aPECAM, DOTAP, and Combined LNPs 30 minutes after LNP injection. Combined LNPs localize to the lungs >2-fold more than aPECAM or DOTAP LNPs. C.Lung-to-liver ratios obtained from B, calculated by dividing the %ID/g of tissue in the lung by that in the liver. Combined LNPs have a >2-fold higher lung-to-liver ratio compared to aPECAM or DOTAP LNPs. D. Luciferase expression of aPECAM, DOTAP, and Combined LNPs in the lung, liver, and spleen 4 hours after LNP injection. E. Lung-to-liver ratios calculated from D by dividing the normalized luciferase expression values in the lung by that in the liver. The lung-to-liver ratio of combined LNPs is about 10-fold and 5-fold higher than both aPECAM and DOTAP LNPs respectively. F. Ratios of lung expression to localization, calculated by dividing the normalized luciferase expression values from D to the %ID/g of tissue values from A for each LNP formulation. The expression to localization ratios are not significantly different between formulations indicating a good correlation between LNP localization and luciferase mRNA expression. Statistics: n=3 and data shown represents mean ± SEM. Comparisons between groups were made using 1-way ANOVA with Tukey’s post-hoc test. *=p<0.05, ***=p<0.001, ****=p<0.0001.
To measure protein expression, we fabricated aPECAM, DOTAP, and Combined LNPs loaded with luciferase mRNA. LNPs were IV-injected into healthy mice, then sacrificed 4 hours later for measurement of luciferase activity in each organ. Combined LNPs have a >2-fold and >1.6-fold higher luciferase expression in the lungs compared to aPECAM or DOTAP LNPs, respectively (Fig 1D). Combined LNPs also have a 10-fold and 5-fold higher lung-to-liver luciferase expression ratio than aPECAM and DOTAP LNPs, respectively (Fig 1E). Lung-expression-to-uptake ratios were obtained by dividing the normalized luciferase expression values in Fig 1E by the %ID/g values in Fig 1A (Fig 1F). We found no significant difference in the expression-to-uptake ratio between formulations, indicating that Combined LNPs’ advantage in luciferase expression is due entirely to their improved lung localization (deposition of nanoparticles in the lung), rather than an advantage in the amount of luciferase expression produced by each LNP that stays in the lungs.
To investigate the different cell types that take up our 3 different LNP formulations, we used flow cytometry to trace fluorescent aPECAM, DOTAP, and Combined LNPs in cells isolated from lungs 30 minutes after IV injection of the LNPs (Fig 2A, gating strategy shown in Supp Fig 2). We determined the percentage of endothelial cells and neutrophils that take up our LNPs (Fig 2B). All three formulations achieve high uptake in endothelial cells (>85% of endothelial cells were LNP-positive) with no significant difference between them. Neutrophils took up DOTAP and Combined LNPs in significant quantities (90% and 80% of neutrophils were positive for DOTAP and Combined, respectively), whereas only 41% of neutrophils were positive for aPECAM LNPs. Endothelial-to-neutrophil uptake ratios (Fig 2C) show that aPECAM LNPs have clear endothelial preference compared to DOTAP and Combined LNPs. aPECAM and Combined LNPs had similar mean fluorescence intensities, with a small but significant edge to aPECAM LNPs over DOTAP LNPs (Fig 2D). A large fraction of neutrophils took up DOTAP and Combined LNPs however, MFI data show that these LNPs were taken up in significantly greater concentrations by endothelial cells, compared to neutrophils. We assessed LNP uptake in alveolar mesenchymal and epithelial cells, cells further away from the alveolar capillary lumen (Fig 2E). DOTAP-containing LNPs can reach these deeper cell types28,29. aPECAM and DOTAP LNPs are taken up by <1% and 4% of epithelial cells respectively. However, epithelial cells take up a staggering 40% of Combined LNPs. Similarly, mesenchymal cells take up 35% of the Combined formulation (Fig 2E). In summary, aPECAM LNPs are more endothelial-specific than DOTAP-containing LNPs, with DOTAP-containing LNPs being taken up by a high fraction of neutrophils, but with only a moderate amount of uptake per cell, as compared to endothelial cells.
Figure 2 : Combined LNPs have enhanced uptake in epithelial and mesenchymal cells in vivo in the lung, but also high uptake by neutrophils.
A. Schematic showing the flow cytometry protocol used, where fluorescently labeled aPECAM, DOTAP, or Combined LNPs were IV injected into mice at a dose of 7.5 ug RNA, allowed to circulate for 30 min, before sacrificed and the lungs prepared as single-cell suspensions. B. Gating first by cell type e.g., endothelial cells and neutrophils, we then looked at the percentage of each cell type that was positive for aPECAM, DOTAP, or combined LNPs. Data shows enhanced neutrophil uptake of DOTAP and Combined LNPs C. Endothelial-to-neutrophil (E:N) uptake ratio of aPECAM, DOTAP, and Combined LNPs. Values greater than 1 indicate endothelial cell preference compared to neutrophils. aPECAM LNPs are shown to have higher endothelial preference. D. LNP mean fluorescence intensity (MFI) for LNP formulations show that while DOTAP and Combined LNPs have a lower E:N ratio compared to aPECAM LNPs, the endothelial cells that take up these LNPs do so highly and equally to aPECAM LNPs. E. Gating first by cell type (as in B), and then plotting LNP positivity among epithelial and mesenchymal cells. These cells are further away from the vascular lumen than endothelial cells and neutrophils, and generally harder to access via intravenous administration. DOTAP and more so Combined LNPs are able to cross and be taken up these two cell types, with Combined LNPs achieving higher epithelial uptake than both DOTAP and aPECAM LNPs. F. Schematic depicting Cre mRNA LNP-driven deletion of LoxP-flanked STOP cassette on Ai6 reporter mice to determine cell types that take up and express Cre-mRNA cargo. We inject aPECAM, DOTAP, or Combined LNPs via the intravenous route. After 24 hours, mice were sacrificed and cell types assessed for enhanced GFP (eGFP) signal driven by the ZsGreen1 gene. G. Using this method, we found that pulmonary endothelial cells had the highest rate of expressing eGFP. We observed that aPECAM and Combined LNPs outcompeted DOTAP LNPs for endothelial cell expression of eGFP. Statistics: n=3 and data shown represents mean ± SEM. For Ai6 mouse experiments a n=4 was used, data presented as mean ± SEM. Comparisons between groups were made using 1-way ANOVA or 2-way ANOVA with Tukey’s post-hoc test where appropriate. *=p<0.05, ***=p<0.001, ****=p<0.0001.
We assessed monocytes/macrophages and lymphocytes (Supp. Fig 3A and F). DOTAP and Combined LNPs were taken up in monocytes/macrophages at a significantly higher rate than aPECAM LNPs. But uptake in monocytes/macrophages was low, with only 30% of monocytes and macrophages taking up DOTAP and Combined LNPs. Uptake in other leukocytes was similarly low for all LNP formulations.
We next investigated correlation between cell type uptake and expression. To do this we generated our three LNP formulations loaded with mRNA expressing Cre-recombinase. These LNPs were then injected into Ai6 Cre reporter mice. Ai6 mice have a loxP-flanked Stop cassette preventing transcription of an enhanced green fluorescent protein (eGFP), all inserted into the ROSA-26locus as shown in Fig 2F. Using this method, we find that all three formulations drive eGFP expression in endothelial cells but aPECAM and Combined LNPs outcompete DOTAP LNPs for endothelial expression. Interestingly, we also find that these 2 formulations also express in monocytes and macrophages Fig 2G.
We next used the most common model for ARDS, nebulized LPS (neb-LPS)61,62, to investigate how acute lung inflammation affects LNP lung targeting and cell type specificity. Radiolabeled or fluorescent LNPs were administered IV, 4 hours after neb-LPS, after inflammatory processes and neutrophilic infiltration into the lung have developed (Fig 3A).
Figure 3: Combined LNPs show superior lung localization in a mouse model of acute lung inflammation.
A. Schematic of nebulized-LPS injury and resulting assays performed, briefly 4 hours after neb-LPS injury, mice are either injected with fluorescently labeled or Indium-111 (In-111) radiolabeled nanoparticles, allowed to circulate for 30 minutes before sacrifice to determine either cell type distribution or whole organ particle localization. B. Comparative biodistribution between healthy and neb-LPS challenged mice given aPECAM, DOTAP, and combined LNPs show enhancement of localization for all formulations to the lung following neb-LPS challenge. Combined LNPs localize to the lungs 3-fold and 2.25-fold more than aPECAM and DOTAP LNPs respectively following neb-LPS challenge. C. Comparison of lung-to-liver ratios between healthy and neb-LPS challenged mice, shows significant enhancement to lung localization after neb-LPS challenge for all formulations but especially for the combined formulation, getting an almost 2-fold improvement in lung targeting. Similarly, combined LNPs have a >2-fold higher lung to liver ratio compared to aPECAM or DOTAP LNPs also after neb-LPS injury. D. Quantification of the effect of pathology lung uptake and delivery aPECAM, DOTAP, and Combined LNP formulations. Dividing the %ID/g of LNP in the lung after neb-LPS by the %ID/g in healthy lung shows that aPECAM, DOTAP, and Combined LNPs increase localization between 1.5-2 fold increase in lung localization. E. Gating first by cell type (endothelial cells) then identifying the percentage that are LNP positive, shows that pathology does not influence uptake of LNPs by endothelial cells. F. Similarly, this is the case for neutrophils. Statistics: n=3 and data shown represents mean ± SEM. Comparisons between groups were made using 1-way ANOVA or 2-way ANOVA with Tukey’s post-hoc test where appropriate. *=p<0.05, ***=p<0.001, ****=p<0.0001.
In radiotracer data, Combined LNPs had a 2-fold improvement in lung uptake in neb-LPS mice vs. healthy animals (Fig 3B and Supp Fig 1). Lung uptake of Combined LNPs in neb-LPS mice was also 2-fold higher than that of aPECAM or DOTAP LNPs. Lung-to-liver uptake ratio follows a similar trend (Fig 3C), showing high lung specificity of Combined LNPs after neb-LPS injury. We define a “pathology ratio” as lung uptake in neb-LPS mice divided by lung uptake in healthy mice. aPECAM, DOTAP, and Combined LNPs had statistically similar pathology ratios, with neb-LPS causing a ~1.5 to 2-fold increase in lung uptake (Fig 3D).
Despite increased lung uptake in radiotracer data, in flow cytometry, there was no increase in the fraction of either endothelial cells or neutrophils positive for LNPs when comparing neb-LPS to healthy mice (Figs 3E–F). Epithelial and mesenchymal uptake of DOTAP and Combined LNPs was similarly conserved (Supp Fig 3G). There was an increase in monocyte and macrophage uptake of DOTAP and Combined LNPs, from 30% positive in healthy mice to 50% in neb-LPS mice (Supp Fig 3B and F). However, the mean LNP fluorescence for these cell types does not increase, suggesting they are not the source for the increase in lung uptake (Supp Fig 3D).
Since the lungs possess the first capillary bed downstream of an IV injection, we used intra-arterial (IA) injection in the right carotid artery to see if a first pass effect may: a) contribute to lung uptake of our LNPs; b) allow for increased LNP uptake in the brain after intracarotid injection (Fig 4A). After IA injection, radiolabeled aPECAM, DOTAP, and Combined LNPs were allowed to circulate for 30 min before assessing biodistributions.
Figure 4: DOTAP and Combined LNP formulations are dependent on first pass privilege for brain localization, but provide unprecedented targeting to the brain.
A test for targeted therapeutics specificity to the lung is to measure the dependence of first pass effect as the lung is the first major capillary bed targeted therapeutics face. A. To eliminate first pass effect, In-111 radiolabeled aPECAM, DOTAP, and Combined LNPs were injected intra-arterially (IA) into the right carotid artery and allowed to circulate for 30 minutes, the animals were then sacrificed and organs measured for radioactivity, this was compared to direct intravenous delivery (IV) via retro orbital injection. Segments of the brain were dissected as shown in A (right) identifying ipsilateral (same side of injection), contralateral (opposing side of injection), and the cerebellum. B. Following IA injection of aPECAM, DOTAP, and Combined LNPs we show enhanced localization by DOTAP and Combined LNPs in the brain, specifically in the ipsilateral cerebrum (same side where the LNP was injected). C. Directly comparing the %ID/g in the brain after IA injection to %ID/g after IV injection highlights that for brain localization, where IA administration gives the brain capillary bed first pass privilege, enhances delivery by DOTAP and Combined LNPs by an order of magnitude. D. We next compare this IA:IV ratio in the brain to that of the lung to better evaluate intraarterial first pass effect dependence on targeting to the brain. E. To determine the cell types in the brain that take up Combined LNPs, we injected fluorescently labeled Combined LNPs and separately prepared the ipsilateral and contralateral hemispheres of the brain. In the contralateral side of the brain, LNPs are taken up more by cerebral vascular endothelial cells whereas LNPs are taken up more by monocytes and macrophages in the ipsilateral side of the brain. Statistics. n=3 and data shown represents mean ± SEM. Comparisons between groups were made using 1-way ANOVA, where appropriate with Tukey’s post-hoc test. *=p<0.05, ***=p<0.001
DOTAP LNPs have increased uptake in the lungs after IA injection, compared to IV injection (Supp Fig 4A and B). This may be because IA injection provides the DOTAP LNPs an extra cardiac cycle before lung exposure, allowing for accumulation of additional opsonins (see Discussion for further hypotheses). aPECAM and Combined LNPs’ lung uptake was unaffected by the injection method. Our results indicate a first-pass effect is not the main reason for lung uptake of our targeted LNPs.
After IA injection in the right carotid artery, we observed high uptake of DOTAP and Combined LNPs in the brain, but no such uptake with aPECAM LNPs. Uptake of DOTAP and Combined LNPs in the right (ipsilateral to injection site) hemisphere was 35% ID/g, which is, to our knowledge, the highest reported delivery to the brain (Fig 4B)63,64. IA injection moderately increases DOTAP and Combined LNP uptake in the contralateral side and the cerebellum. Comparing ipsilateral brain uptake of LNPs after IA vs. IV delivery shows that IA delivery increases brain delivery ~50-100-fold (Fig 4C). Implying DOTAP-containing LNP brain uptake is almost entirely first-pass mediated. Comparing the IA-to-IV ratio for brain uptake vs lung uptake shows a greater first-pass effect for brain delivery than for lung delivery of DOTAP-containing LNPs, especially Combined LNPs (Fig 4D).
Last, we tested the cell type uptake of our Combined LNP following intraarterial injection (Fig 4E). Following circulation, the brains were separated into ipsilateral and contralateral hemispheres. We found that about 20-40% of the endothelial cells are positive for LNP delivery, which is impressive compared to prior delivery technologies.
Two classes of targeting methods have achieved LNP delivery to specific cell types and organs: affinity moiety targeting34,36,37,40 and physicochemical tropism24,28–30,65. Here, we directly compare these two methods. We focused on targeted delivery to the lungs, using aPECAM LNPs to represent affinity moiety targeting, and DOTAP LNPs to represent physicochemical tropism. We also incorporated aPECAM and DOTAP in the same LNPs, to achieve a “Combined” targeting LNP.
Focusing on comparing affinity moiety targeting and physicochemical tropism, both strategies produced nearly identical overall lung uptake. Indeed different targeting strategies yield similar lung uptake, nanoparticles targeted via aPECAM, aICAM66, the technique of RBC-hitchhiking67–69, and physicochemical tropism to marginated neutrophils70. Suggesting a saturation value for lung uptake focused on the alveolar capillaries. Combined LNPs overcome this saturation. Comparing cell type selectivity of affinity moiety targeting vs. physicochemical tropism, aPECAM LNPs have better endothelial specificity than DOTAP LNPs. Endothelial cells are traditionally the cells of interest in lung targeting. However, DOTAP provides delivery to cells outside of the vasculature, to epithelial and mesenchymal, promising RNA delivery to lung cell types inaccessible with IV agents. Combined LNPs had >2x-higher lung uptake than either aPECAM or DOTAP alone and provided unprecedented delivery to alveolar epithelial cells. They exhibited 2x-higher delivery to inflamed lungs than to healthy lungs. These advantages encourage investigation of Combined LNPs and their mechanisms of uptake, especially in pathology, like pulmonary inflammation.
Combined LNPs required optimization to work. Particularly, the identity and molar ratio of PEG-lipids had to be optimized. Our choice was utilizing DSPE-PEG (18 carbons) for aPECAM-LNPs, DMG-PEG (10 carbons) for DOTAP-LNPs, and a half mix of both to generate our Combined LNPs. DSPE with 18 carbons in its acyl chain, anchors the lipid to the surface layer more strongly than DMG, which only has 10 carbons9. Conversely, DMG desorbs very quickly in serum, allowing serum proteins (e.g., ApoE) to adsorb onto the LNP surface, driving localization (as shown with FDA-approved patisiran). DSPE is used to anchor lipids for affinity moieties (here anti-PECAM antibodies), to keep the lipid-affinity-moiety conjugate on the LNP surface at least long enough for the affinity moiety to reach its target. DMG is the most used lipid for anchoring PEG when physicochemical tropism is desired. For our Combined LNPs, we “thread the needle” achieving a balance of antibody targeting and protein corona guided localization. Indeed, in Supp Fig 4E and F, we see that changing the PEG ratio from 1.5% DSPE PEG to a ratio of 0.75% DMG PEG and 0.75% DSPE PEG matching the PEG ratio used in our Combined formulation, we observed ablation of aPECAM LNP lung targeting. Likely, half of the LNPs’ PEG desorbed quickly, allowing serum proteins, especially complement proteins such as C3, to adsorb onto the LNPs’ antibodies12,13. Complement C3-coated antibodies are avidly cleared by Kupffer cells, hence the high liver expression. Thus, changing the PEG-lipid ratios has a very large effect when comparing the various LNPs.
DOTAP and Combined LNPs strongly localize to the brain following intra-arterial injection. They achieve ~35% ID/g on the side of the brain ipsilateral to injection site. This level of brain uptake exceeds standard brain targeting methods, including transferrin and VCAM targeting63,64 and is largely unilateral. This can be useful for brain injuries that present unilaterally, including stroke and traumatic brain injuries.
When targeting the brain, Combined LNPs were mostly taken up by endothelial cells and innate immune cells, instead of neurons. While in some applications the goal is to target the neurons, targeting endothelial cells and leukocytes is actually quite advantageous for acute brain injuries, like ischemic stroke, hemorrhagic stroke, and traumatic brain injury34,63,71–73. Our goal is not to target LNPs past the BBB (blood-brain barrier), but rather to the endothelial cells of the BBB. In all these acute brain injuries, the BBB endothelium is leaky, allowing toxic plasma proteins and innate immune cells to cross the BBB and contact the parenchyma, injuring the neurons. We have shown that targeting LNPs to endothelial cells, delivering anti-inflammatory proteins like IL-10 or thrombomodulin, can seal up the BBB and reduce inflammation and neuronal injury63,71. Additionally, targeting LNPs to the neutrophils and monocytes that surround the BBB can ameliorate neuronal inflammation34. Thus, targeting the brain endothelial cells, neutrophils, and macrophages is a major goal, but is limited by other technologies achieving ~1% of the injected dose (%ID) to the brain, a tiny fraction going to these cells.
The intra-carotid injections of Combined LNPs provides >20x improvement in the %ID in the brain. Therefore, we hoped that it also provided good delivery to endothelial cells, neutrophils, and monocytes, and indeed, the data in Fig 4E and Supp Fig 7A shows that it does. 20-40% of endothelial cells are positive for LNP delivery, impressive compared to prior delivery technologies.
Safety and side effects are critical issues which must be investigated in future studies. Combined LNPs will have side effects, and it is important to investigate these and attempt to engineer solutions. Of greatest concern with DOTAP-containing LNPs is that IV LNPs formulated with any of various cationic lipids cause thrombosis74. It is reasonable to hypothesize that Combined LNPs may cause similar effects. However, our recent work also demonstrated techniques to mitigate clotting induced by cationic lipid LNPs, including anticoagulation and reduced LNP size. These techniques should be applied to Combined LNPs in future studies. It remains to be seen whether these or other engineered solutions will be sufficient to make the risk-benefit ratio meet the criterion for further preclinical development of what appears to be a promising targeting technology.
Additional safety concerns include LNP-associated inflammation. We have previously shown that LNPs as a class worsen pre-existing inflammation75. In this manuscript, we attempted to measure LNP-based mRNA expression after intra-carotid artery injection, however, the mice did not survive. Cannulation and injection of the carotid artery in mice is a technically difficult procedure, usually done for 2-hour short-term experiments. After many attempts, we were unable to get mice to survive the 4 hours found to be necessary to observe significant expression from mRNA LNPs. A big surgery like carotid artery exposure and cannulation induces a major inflammatory response, which is compounded by mRNA-LNPs worsening pre-existing inflammation75. Thus, we could not determine the correlation between delivery and expression in the brain.
However, we successfully correlated mRNA delivery vs expression after intravenous (IV) delivery. First, we showed in Fig 1B–F in the lungs that, for these formulations (aPECAM, DOTAP, Combined), the LNPs’ expression and localization correlate very well with one another and can be used as surrogate measures. Additionally, in Fig 2G, we examined cell-type-specific expression of LNPs in the Ai6 mouse model. Figure 2G shows that the expression data looks like the delivery data, with the exception that neutrophils do not express highly, primarily due to their 6–8-hour half-life and we measured expression at 24 hours. At the level of whole-organ biodistribution and comparative cell type level, these LNPs appear to have delivery/uptake that correlate well with expression of their cargo mRNA. While we cannot be sure of such a correlation upon intra-carotid delivery to the brain, the main point of Figure 4 shows DOTAP provides first-pass delivery, indicating localization to the first capillary bed encountered.
DOTAP-containing LNPs have greater uptake by innate immune cells, possibly leading to even greater inflammation than other LNPs. Flow cytometry data shows that DOTAP and Combined LNPs are avidly taken up by innate immune cells, especially in acute lung injury (Figs 2B, 3F, and Supp Fig 3A, B, and F). This increased immune uptake of LNPs increases the potential for activation of inflammatory pathways, possibly allowing later adaptive immunity against the LNPs. Future studies should aim to engineer solutions to uptake of DOTAP-containing LNPs by innate immune cells, such as adding on “don’t eat me” signals such as CD47-mimetic peptides76. Conversely, DOTAP-LNPs’ uptake by innate immune cells also present new targets for RNA delivery and disease-specific treatment62,77–79.
Supplementary Material
References
- (1).Wilhelm S; Tavares AJ; Dai Q; Ohta S; Audet J; Dvorak HF; Chan WCW Analysis of Nanoparticle Delivery to Tumours. Nature Reviews Materials 2016, 1 (5), 1–12. [Google Scholar]
- (2).Poon W; Zhang Y-N; Ouyang B; Kingston BR; Wu JLY; Wilhelm S; Chan WCW. Elimination Pathways of Nanoparticles. ACS Nano 2019, 13 (5), 5785–5798. [DOI] [PubMed] [Google Scholar]
- (3).Bachmaier K; Stuart A; Singh A; Mukhopadhyay A; Chakraborty S; Hong Z; Wang L; Tsukasaki Y; Maienschein-Cline M; Ganesh BB; Kanteti P; Rehman J; Malik AB Albumin Nanoparticle Endocytosing Subset of Neutrophils for Precision Therapeutic Targeting of Inflammatory Tissue Injury. ACS Nano 2022, 16 (3), 4084–4101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (4).Ouyang B; Poon W; Zhang Y-N; Lin ZP; Kingston BR; Tavares AJ; Zhang Y; Chen J; Valic MS; Syed AM; MacMillan P; Couture-Senécal J; Zheng G; Chan WCW The Dose Threshold for Nanoparticle Tumour Delivery. Nat. Mater 2020, 19 (12), 1362–1371. [DOI] [PubMed] [Google Scholar]
- (5).Chen S-J; Sanmiguel J; Lock M; McMenamin D; Draper C; Limberis MP; Kassim SH; Somanathan S; Bell P; Johnston JC; Rader DJ; Wilson JM Biodistribution of AAV8 Vectors Expressing Human Low-Density Lipoprotein Receptor in a Mouse Model of Homozygous Familial Hypercholesterolemia. Hum Gene Ther Clin Dev 2013, 24 (4), 154–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (6).Torchilin VP; Khaw BA; Smirnov VN; Haber E Preservation of Antimyosin Antibody Activity after Covalent Coupling to Liposomes. Biochem. Biophys. Res. Commun 1979, 89 (4), 1114–1119. [DOI] [PubMed] [Google Scholar]
- (7).Huang A; Kennel SJ; Huang L Interactions of Immunoliposomes with Target Cells. J. Biol. Chem 1983, 258 (22), 14034–14040. [PubMed] [Google Scholar]
- (8).Heath TD; Fraley RT; Papahdjopoulos D Antibody Targeting of Liposomes: Cell Specificity Obtained by Conjugation of F(ab’)2 to Vesicle Surface. Science 1980, 210 (4469), 539–541. [DOI] [PubMed] [Google Scholar]
- (9).Gregoriadis G Homing of Liposomes to Target Cells. Biochem. Soc. Trans 1975, 3 (5), 613–618. [DOI] [PubMed] [Google Scholar]
- (10).Gregoriadis G Targeting of Drugs. Nature 1977, 265 (5593), 407–411. [DOI] [PubMed] [Google Scholar]
- (11).Magee WE; Miller OV Liposomes Containing Antiviral Antibody Can Protect Cells from Virus Infection. Nature 1972, 235 (5337), 339–341. [DOI] [PubMed] [Google Scholar]
- (12).Kułdo JM; Ogawara KI; Werner N; Asgeirsdóttir SA; Kamps JAAM; Kok RJ; Molema G Molecular Pathways of Endothelial Cell Activation for (targeted) Pharmacological Intervention of Chronic Inflammatory Diseases. Curr. Vasc. Pharmacol 2005, 3 (1), 11–39. [DOI] [PubMed] [Google Scholar]
- (13).Ahmad I; Arsalan A; Ali SA; Sheraz MA; Ahmed S; Anwar Z; Munir I; Shah MR Formulation and Stabilization of Riboflavin in Liposomal Preparations. J. Photochem. Photobiol. B 2015, 153, 358–366. [DOI] [PubMed] [Google Scholar]
- (14).Suntres ZE; Shek PN Prophylaxis against Lipopolysaccharide-Induced Acute Lung Injury by Alpha-Tocopherol Liposomes. Crit. Care Med 1998, 26 (4), 723–729. [DOI] [PubMed] [Google Scholar]
- (15).Yamakawa T; Ohigashi H; Hashimoto D; Hayase E; Takahashi S; Miyazaki M; Minomi K; Onozawa M; Niitsu Y; Teshima T Vitamin A-Coupled Liposomes Containing siRNA against HSP47 Ameliorate Skin Fibrosis in Chronic Graft-versus-Host Disease. Blood 2018, 131 (13), 1476–1485. [DOI] [PubMed] [Google Scholar]
- (16).Khan AA; Allemailem KS; Almatroodi SA; Almatroudi A; Rahmani AH Recent Strategies towards the Surface Modification of Liposomes: An Innovative Approach for Different Clinical Applications. 3 Biotech 2020, 10 (4), 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Xu L; Huang C-C; Huang W; Tang W-H; Rait A; Yin YZ; Cruz I; Xiang L-M; Pirollo KF; Chang EH Systemic Tumor-Targeted Gene Delivery by Anti-Transferrin Receptor scFv-Immunoliposomes. Mol. Cancer Ther 2002, 1 (5), 337–346. [PubMed] [Google Scholar]
- (18).Mamot C; Drummond DC; Hong K; Kirpotin DB; Park JW Liposome-Based Approaches to Overcome Anticancer Drug Resistance. Drug Resist. Updat 2003, 6 (5), 271–279. [DOI] [PubMed] [Google Scholar]
- (19).Miller K; Cortes J; Hurvitz SA; Krop IE; Tripathy D; Verma S; Riahi K; Reynolds JG; Wickham TJ; Molnar I; Yardley DA HERMIONE: A Randomized Phase 2 Trial of MM-302 plus Trastuzumab versus Chemotherapy of Physician’s Choice plus Trastuzumab in Patients with Previously Treated, Anthracycline-Naïve, HER2-Positive, Locally Advanced/metastatic Breast Cancer. BMC Cancer 2016, 16, 352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (20).Xie F; Yao N; Qin Y; Zhang Q; Chen H; Yuan M; Tang J; Li X; Fan W; Zhang Q; Wu Y; Hai L; He Q Investigation of Glucose-Modified Liposomes Using Polyethylene Glycols with Different Chain Lengths as the Linkers for Brain Targeting. Int. J. Nanomedicine 2012, 7, 163–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Abu-Dahab R; Schäfer UF; Lehr CM Lectin-Functionalized Liposomes for Pulmonary Drug Delivery: Effect of Nebulization on Stability and Bioadhesion. Eur. J. Pharm. Sci 2001, 14 (1), 37–46. [DOI] [PubMed] [Google Scholar]
- (22).Anselmo AC; Mitragotri S Nanoparticles in the Clinic. Bioeng Transl Med 2016, 1 (1), 10–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Anselmo AC; Mitragotri S Nanoparticles in the Clinic: An Update. Bioeng Transl Med 2019, 4 (3), e10143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (24).Siegwart DJ; Whitehead KA; Nuhn L; Sahay G; Cheng H; Jiang S; Ma M; Lytton-Jean A; Vegas A; Fenton P; Levins CG; Love KT; Lee H; Cortez C; Collins SP; Li YF; Jang J; Querbes W; Zurenko C; Novobrantseva T; Langer R; Anderson DG Combinatorial Synthesis of Chemically Diverse Core-Shell Nanoparticles for Intracellular Delivery. Proc. Natl. Acad. Sci. U. S. A 2011, 108 (32), 12996–13001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (25).Lee H; Lytton-Jean AKR; Chen Y; Love KT; Park AI; Karagiannis ED; Sehgal A; Querbes W; Zurenko CS; Jayaraman M; Peng CG; Charisse K; Borodovsky A; Manoharan M; Donahoe JS; Truelove J; Nahrendorf M; Langer R; Anderson DG Molecularly Self-Assembled Nucleic Acid Nanoparticles for Targeted in Vivo siRNA Delivery. Nat. Nanotechnol 2012, 7 (6), 389–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (26).Love KT; Mahon KP; Levins CG; Whitehead KA; Querbes W; Dorkin JR; Qin J; Cantley W; Qin LL; Racie T; Frank-Kamenetsky M; Yip KN; Alvarez R; Sah DWY; de Fougerolles A; Fitzgerald K; Koteliansky V; Akinc A; Langer R; Anderson DG Lipid-like Materials for Low-Dose, in Vivo Gene Silencing. Proc. Natl. Acad. Sci. U. S. A 2010, 107 (5), 1864–1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Akinc A; Zumbuehl A; Goldberg M; Leshchiner ES; Busini V; Hossain N; Bacallado SA; Nguyen DN; Fuller J; Alvarez R; Borodovsky A; Borland T; Constien R; de Fougerolles A; Dorkin JR; Narayanannair Jayaprakash K; Jayaraman M; John M; Koteliansky V; Manoharan M; Nechev L; Qin J; Racie T; Raitcheva D; Rajeev KG; Sah DWY; Soutschek J; Toudjarska I; Vornlocher H-P; Zimmermann TS; Langer R; Anderson DG A Combinatorial Library of Lipid-like Materials for Delivery of RNAi Therapeutics. Nat. Biotechnol 2008, 26 (5), 561–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (28).Cheng Q; Wei T; Farbiak L; Johnson LT; Dilliard SA; Siegwart DJ Selective Organ Targeting (SORT) Nanoparticles for Tissue-Specific mRNA Delivery and CRISPR-Cas Gene Editing. Nat. Nanotechnol 2020, 15 (4), 313–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (29).Wang X; Liu S; Sun Y; Yu X; Lee SM; Cheng Q; Wei T; Gong J; Robinson J; Zhang D; Lian X; Basak P; Siegwart DJ Preparation of Selective Organ-Targeting (SORT) Lipid Nanoparticles (LNPs) Using Multiple Technical Methods for Tissue-Specific mRNA Delivery. Nat. Protoc 2022. 10.1038/s41596-022-00755-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (30).LoPresti ST; Arral ML; Chaudhary N; Whitehead KA The Replacement of Helper Lipids with Charged Alternatives in Lipid Nanoparticles Facilities Targeted mRNA Delivery to the Spleen and Lungs. J. Control. Release 2022. 10.1016/j.jconrel.2022.03.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (31).Billingsley MM; Singh N; Ravikumar P; Zhang R; June CH; Mitchell MJ Ionizable Lipid Nanoparticle-Mediated mRNA Delivery for Human CAR T Cell Engineering. Nano Lett. 2020, 20 (3), 1578–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (32).Billingsley MM; Hamilton AG; Mai D; Patel SK; Swingle KL; Sheppard NC; June CH; Mitchell MJ Orthogonal Design of Experiments for Optimization of Lipid Nanoparticles for mRNA Engineering of CAR T Cells. Nano Lett. 2022, 22 (1), 533–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (33).Adams D; Suhr OB; Dyck PJ; Litchy WJ; Leahy RG; Chen J; Gollob J; Coelho T Trial Design and Rationale for APOLLO, a Phase 3, Placebo-Controlled Study of Patisiran in Patients with Hereditary ATTR Amyloidosis with Polyneuropathy. BMC Neurol. 2017, 17 (1), 181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (34).Nong J; Glassman PM; Myerson JW; Zuluaga-Ramirez V; Rodriguez-Garcia A; Mukalel A; Omo-Lamai S; Walsh LR; Zamora ME; Gong X; Wang Z; Bhamidipati K; Kiseleva RY; Villa CH; Greineder CF; Kasner SE; Weissman D; Mitchell MJ; Muro S; Persidsky Y; Brenner JS; Muzykantov VR; Marcos-Contreras OA Targeted Nanocarriers Co-Opting Pulmonary Intravascular Leukocytes for Drug Delivery to the Injured Brain. ACS Nano 2023. 10.1021/acsnano.2c08275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (35).Paris AJ; Guo L; Dai N; Katzen JB; Patel PN; Worthen GS; Brenner JS Using Selective Lung Injury to Improve Murine Models of Spatially Heterogeneous Lung Diseases. PLoS One 2019, 14 (4), e0202456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (36).Parhiz H; Shuvaev VV; Pardi N; Khoshnejad M; Kiseleva RY; Brenner JS; Uhler T; Tuyishime S; Mui BL; Tam YK; Madden TD; Hope MJ; Weissman D; Muzykantov VR PECAM-1 Directed Re-Targeting of Exogenous mRNA Providing Two Orders of Magnitude Enhancement of Vascular Delivery and Expression in Lungs Independent of Apolipoprotein E-Mediated Uptake. J. Control. Release 2018, 291, 106–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (37).Glassman PM; Myerson JW; Ferguson LT; Kiseleva RY; Shuvaev VV; Brenner JS; Muzykantov VR Targeting Drug Delivery in the Vascular System: Focus on Endothelium. Adv. Drug Deliv. Rev 2020, 157, 96–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (38).Reyes-Esteves S; Nong J; Glassman PM; Omo-Lamai S; Ohashi S; Myerson JW; Zamora ME; Ma X; Kasner SE; Sansing L; Muzykantov VR; Marcos-Contreras OA; Brenner JS Targeted Drug Delivery to the Brain Endothelium Dominates over Passive Delivery via Vascular Leak in Experimental Intracerebral Hemorrhage. J. Control. Release 2023, 356, 185–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (39).Muro S; Garnacho C; Champion JA; Leferovich J; Gajewski C; Schuchman EH; Mitragotri S; Muzykantov VR Control of Endothelial Targeting and Intracellular Delivery of Therapeutic Enzymes by Modulating the Size and Shape of ICAM-1-Targeted Carriers. Mol. Ther 2008, 16 (8), 1450–1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (40).Hood ED; Greineder CF; Shuvaeva T; Walsh L; Villa CH; Muzykantov VR Vascular Targeting of Radiolabeled Liposomes with Bio-Orthogonally Conjugated Ligands: Single Chain Fragments Provide Higher Specificity than Antibodies. Bioconjug. Chem 2018, 29 (11), 3626–3637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (41).Shuvaev VV; Muro S; Arguiri E; Khoshnejad M; Tliba S; Christofidou-Solomidou M; Muzykantov VR Size and Targeting to PECAM vs ICAM Control Endothelial Delivery, Internalization and Protective Effect of Multimolecular SOD Conjugates. J. Control. Release 2016, 234, 115–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (42).Manthe RL; Muro S ICAM-1-Targeted Nanocarriers Attenuate Endothelial Release of Soluble ICAM-1, an Inflammatory Regulator. Bioeng Transl Med 2017, 2 (1), 109–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (43).Han J; Zern BJ; Shuvaev VV; Davies PF; Muro S; Muzykantov V Acute and Chronic Shear Stress Differently Regulate Endothelial Internalization of Nanocarriers Targeted to Platelet-Endothelial Cell Adhesion Molecule-1. ACS Nano 2012, 6 (10), 8824–8836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (44).McIntosh DP; Tan X-Y; Oh P; Schnitzer JE Targeting Endothelium and Its Dynamic Caveolae for Tissue-Specific Transcytosis in Vivo: A Pathway to Overcome Cell Barriers to Drug and Gene Delivery. Proc. Natl. Acad. Sci. U. S. A 2002, 99 (4), 1996–2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (45).Ireson CR; Kelland LR Discovery and Development of Anticancer Aptamers. Mol. Cancer Ther 2006, 5 (12), 2957–2962. [DOI] [PubMed] [Google Scholar]
- (46).Santos M. L. dos; Quintilio W; Manieri TM; Tsuruta LR; Moro AM Advances and Challenges in Therapeutic Monoclonal Antibodies Drug Development. Braz. J. Pharm. Sci 2018, 54 (spe), e01007. [Google Scholar]
- (47).Deng Z; Kalin GT; Shi D; Kalinichenko VV Nanoparticle Delivery Systems with Cell-Specific Targeting for Pulmonary Diseases. Am. J. Respir. Cell Mol. Biol 2021, 64 (3), 292–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (48).Arthur R CordenPharma increases xRNA-based capabilities with LNP formulation investment in Italy. biopharma-reporter.com. https://www.biopharma-reporter.com/Article/2022/06/02/CordenPharma-increases-xRNA-based-capabilities-with-LNP-formulation-investment-in-Italy (accessed 2023-11-01). [Google Scholar]
- (49).Verma M; Ozer I; Xie W; Gallagher R; Teixeira A; Choy M The Landscape for Lipid-Nanoparticle-Based Genomic Medicines. Nat. Rev. Drug Discov 2023, 22 (5), 349–350. [DOI] [PubMed] [Google Scholar]
- (50).Oud L; Garza J The Contribution of COVID-19 to Acute Respiratory Distress Syndrome-Related Mortality in the United States. J. Clin. Med. Res 2023, 15 (5), 279–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (51).Matthay MA; Ware LB; Zimmerman GA The Acute Respiratory Distress Syndrome. J. Clin. Invest 2012, 122 (8), 2731–2740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (52).Ware LB; Matthay MA The Acute Respiratory Distress Syndrome. N. Engl. J. Med 2000, 342 (18), 1334–1349. [DOI] [PubMed] [Google Scholar]
- (53).Máca J; Jor O; Holub M; Sklienka P; Burša F; Burda M; Janout V; Ševčík P Past and Present ARDS Mortality Rates: A Systematic Review. Respir. Care 2017, 62 (1), 113–122. [DOI] [PubMed] [Google Scholar]
- (54).Hasan SS; Capstick T; Ahmed R; Kow CS; Mazhar F; Merchant HA; Zaidi STR Mortality in COVID-19 Patients with Acute Respiratory Distress Syndrome and Corticosteroids Use: A Systematic Review and Meta-Analysis. Expert Rev. Respir. Med 2020, 14 (11), 1149–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (55).Silva PL; Pelosi P; Rocco PRM Personalized Pharmacological Therapy for ARDS: A Light at the End of the Tunnel. Expert Opin. Investig. Drugs 2020, 29 (1), 49–61. [DOI] [PubMed] [Google Scholar]
- (56).Qiu M; Tang Y; Chen J; Muriph R; Ye Z; Huang C; Evans J; Henske EP; Xu Q Lung-Selective mRNA Delivery of Synthetic Lipid Nanoparticles for the Treatment of Pulmonary Lymphangioleiomyomatosis. Proc. Natl. Acad. Sci. U. S. A 2022, 119 (8). 10.1073/pnas.2116271119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (57).Dilliard SA; Sun Y; Brown MO; Sung Y-C; Chatterjee S; Farbiak L; Vaidya A; Lian X; Wang X; Lemoff A; Siegwart DJ The Interplay of Quaternary Ammonium Lipid Structure and Protein Corona on Lung-Specific mRNA Delivery by Selective Organ Targeting (SORT) Nanoparticles. J. Control. Release 2023, 361, 361–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (58).Melamed JR; Hajj KA; Chaudhary N; Strelkova D; Arral ML; Pardi N; Alameh M-G; Miller JB; Farbiak L; Siegwart DJ; Weissman D; Whitehead KA Lipid Nanoparticle Chemistry Determines How Nucleoside Base Modifications Alter mRNA Delivery. J. Control. Release 2021. 10.1016/j.jconrel.2021.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (59).Hatit MZC; Lokugamage MP; Dobrowolski CN; Paunovska K; Ni H; Zhao K; Vanover D; Beyersdorf J; Peck HE; Loughrey D; Sato M; Cristian A; Santangelo PJ; Dahlman JE Species-Dependent in Vivo mRNA Delivery and Cellular Responses to Nanoparticles. Nat. Nanotechnol 2022, 17 (3), 310–318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (60).Miao L; Lin J; Huang Y; Li L; Delcassian D; Ge Y; Shi Y; Anderson DG Synergistic Lipid Compositions for Albumin Receptor Mediated Delivery of mRNA to the Liver. Nat. Commun 2020, 11 (1), 2424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (61).de Souza Xavier Costa N; Ribeiro Júnior G; Dos Santos Alemany AA; Belotti L; Zati DH; Frota Cavalcante M; Matera Veras M; Ribeiro S; Kallás EG; Nascimento Saldiva PH; Dolhnikoff M; Ferraz da Silva LF. Early and Late Pulmonary Effects of Nebulized LPS in Mice: An Acute Lung Injury Model. PLoS One 2017, 12 (9), e0185474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (62).Tsikis ST; Fligor SC; Hirsch TI; Pan A; Yu LJ; Kishikawa H; Joiner MM; Mitchell PD; Puder M Lipopolysaccharide-Induced Murine Lung Injury Results in Long-Term Pulmonary Changes and Downregulation of Angiogenic Pathways. Sci. Rep 2022, 12 (1), 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (63).Marcos-Contreras OA; Greineder CF; Kiseleva RY; Parhiz H; Walsh LR; Zuluaga-Ramirez V; Myerson JW; Hood ED; Villa CH; Tombacz I; Pardi N; Seliga A; Mui BL; Tam YK; Glassman PM; Shuvaev VV; Nong J; Brenner JS; Khoshnejad M; Madden T; Weissmann D; Persidsky Y; Muzykantov VR Selective Targeting of Nanomedicine to Inflamed Cerebral Vasculature to Enhance the Blood-Brain Barrier. Proc. Natl. Acad. Sci. U. S. A 2020, 117 (7), 3405–3414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (64).Thomsen MS; Johnsen KB; Kucharz K; Lauritzen M; Moos T Blood-Brain Barrier Transport of Transferrin Receptor-Targeted Nanoparticles. Pharmaceutics 2022, 14 (10). 10.3390/pharmaceutics14102237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (65).Sago CD; Lokugamage MP; Paunovska K; Vanover DA; Monaco CM; Shah NN; Gamboa Castro M; Anderson SE; Rudoltz TG; Lando GN; Munnilal Tiwari P; Kirschman JL; Willett N; Jang YC; Santangelo PJ; Bryksin AV; Dahlman JE High-Throughput in Vivo Screen of Functional mRNA Delivery Identifies Nanoparticles for Endothelial Cell Gene Editing. Proc. Natl. Acad. Sci. U. S. A 2018, 115 (42), E9944–E9952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (66).Brenner JS; Bhamidipati K; Glassman PM; Ramakrishnan N; Jiang D; Paris AJ; Myerson JW; Pan DC; Shuvaev VV; Villa CH; Hood ED; Kiseleva R; Greineder CF; Radhakrishnan R; Muzykantov VR Mechanisms That Determine Nanocarrier Targeting to Healthy versus Inflamed Lung Regions. Nanomedicine 2017, 13 (4), 1495–1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (67).Brenner JS; Mitragotri S; Muzykantov VR Red Blood Cell Hitchhiking: A Novel Approach for Vascular Delivery of Nanocarriers. Annu. Rev. Biomed. Eng 2021, 23, 225–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (68).Ferguson LT; Hood ED; Shuvaeva T; Shuvaev VV; Basil MC; Wang Z; Nong J; Ma X; Wu J; Myerson JW; Marcos-Contreras OA; Katzen J; Carl JM; Morrisey EE; Cantu E; Villa CH; Mitragotri S; Muzykantov VR; Brenner JS Dual Affinity to RBCs and Target Cells (DART) Enhances Both Organ-and Cell Type-Targeting of Intravascular Nanocarriers. ACS Nano 2022, 16 (3), 4666–4683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (69).Brenner JS; Pan DC; Myerson JW; Marcos-Contreras OA; Villa CH; Patel P; Hekierski H; Chatterjee S; Tao J-Q; Parhiz H; Others. Red Blood Cell-Hitchhiking Boosts Delivery of Nanocarriers to Chosen Organs by Orders of Magnitude. Nat. Commun 2018, 9 (1), 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (70).Myerson JW; Patel PN; Rubey KM; Zamora ME; Zaleski MH; Habibi N; Walsh LR; Lee Y-W; Luther DC; Ferguson LT; Marcos-Contreras OA; Glassman PM; Mazaleuskaya LL; Johnston I; Hood ED; Shuvaeva T; Wu J; Zhang H-Y; Gregory JV; Kiseleva RY; Nong J; Grosser T; Greineder CF; Mitragotri S; Worthen GS; Rotello VM; Lahann J; Muzykantov VR; Brenner JS Supramolecular Arrangement of Protein in Nanoparticle Structures Predicts Nanoparticle Tropism for Neutrophils in Acute Lung Inflammation. Nat. Nanotechnol 2022, 17 (1), 86–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (71).Nong J; Glassman PM; Shuvaev VV; Reyes-Esteves S; Descamps HC; Kiseleva RY; Papp TE; Alameh M-G; Tam YK; Mui BL; Omo-Lamai S; Zamora ME; Shuvaeva T; Arguiri E; Gong X; Brysgel TV; Tan AW; Woolfork AG; Weljie A; Thaiss CA; Myerson JW; Weissman D; Kasner SE; Parhiz H; Muzykantov VR; Brenner JS; Marcos-Contreras OA Targeting Lipid Nanoparticles to the Blood Brain Barrier to Ameliorate Acute Ischemic Stroke. Mol. Ther 2024. 10.1016/j.ymthe.2024.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (72).Nong J; Glassman PM; Muzykantov VR Targeting Vascular Inflammation through Emerging Methods and Drug Carriers. Adv. Drug Deliv. Rev 2022, 184, 114180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (73).Marcos-Contreras OA; Brenner JS; Kiseleva RY; Zuluaga-Ramirez V; Greineder CF; Villa CH; Hood ED; Myerson JW; Muro S; Persidsky Y; Muzykantov VR Combining Vascular Targeting and the Local First Pass Provides 100-Fold Higher Uptake of ICAM-1-Targeted vs Untargeted Nanocarriers in the Inflamed Brain. J. Control. Release 2019, 301, 54–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (74).Omo-Lamai S; Zamora ME; Patel MN; Wu J; Nong J; Wang Z; Peshkova A; Majumder A; Melamed JR; Chase LS; Essien E-O; Weissman D; Muzykantov VR; Marcos-Contreras OA; Myerson JW; Brenner JS Physicochemical Targeting of Lipid Nanoparticles to the Lungs Induces Clotting: Mechanisms and Solutions. Adv. Mater 2024, e2312026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (75).Parhiz H; Brenner JS; Patel PN; Papp TE; Shahnawaz H; Li Q; Shi R; Zamora ME; Yadegari A; Marcos-Contreras OA; Natesan A; Pardi N; Shuvaev VV; Kiseleva R; Myerson JW; Uhler T; Riley RS; Han X; Mitchell MJ; Lam K; Heyes J; Weissman D; Muzykantov VR Added to Pre-Existing Inflammation, mRNA-Lipid Nanoparticles Induce Inflammation Exacerbation (IE). J. Control. Release 2022, 344, 50–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (76).Rodriguez PL; Harada T; Christian DA; Pantano DA; Tsai RK; Discher DE Minimal “Self” Peptides That Inhibit Phagocytic Clearance and Enhance Delivery of Nanoparticles. Science 2013, 339 (6122), 971–975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (77).Huang X; Xiu H; Zhang S; Zhang G The Role of Macrophages in the Pathogenesis of ALI/ARDS. Mediators Inflamm. 2018, 2018, 1264913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (78).Dang W; Tao Y; Xu X; Zhao H; Zou L; Li Y The Role of Lung Macrophages in Acute Respiratory Distress Syndrome. Inflamm. Res 2022, 71 (12), 1417–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (79).Lomas-Neira JL; Heffernan DS; Ayala A; Monaghan SF BLOCKADE OF ENDOTHELIAL GROWTH FACTOR, ANGIOPOIETIN-2, REDUCES INDICES OF ARDS AND MORTALITY IN MICE RESULTING FROM THE DUAL-INSULTS OF HEMORRHAGIC SHOCK AND SEPSIS. Shock 2016, 45 (2), 157–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.