key: cord-0843137-biyhr1l8 authors: Papi, Massimiliano; Pozzi, Daniela; Palmieri, Valentina; Caracciolo, Giulio title: Principles for optimization and validation of mRNA lipid nanoparticle vaccines against COVID-19 using 3D bioprinting date: 2022-01-21 journal: Nano Today DOI: 10.1016/j.nantod.2022.101403 sha: ffc8f4d74a28cb720402f3273b860c67150020a9 doc_id: 843137 cord_uid: biyhr1l8 BioNTech/Pfizer’s Comirnaty and Moderna’s SpikeVax vaccines consist in mRNA encapsulated in lipid nanoparticles (LNPs). The modularity of the delivery platform and the manufacturing possibilities provided by microfluidics let them look like an instant success, but they are the product of decades of intense research. There is a multitude of considerations to be made when designing an optimal mRNA-LNP vaccine. Herein, we provide a brief overview of what is presently known and what still requires investigation to optimize mRNA LNP vaccines. Lastly, we give our perspective on the engineering of 3D bioprinted validation systems that will allow faster, cheaper, and more predictive vaccine testing in the future compared with animal models. for the SARS-CoV-2 receptor binding domain (RBD) that is the target of the virus neutralizing antibodies (nAb). Both the vaccines were granted ‗Emergency Use Authorization' by the U.S. Food and Drug Administration (FDA) and, in December 2020, ‗conditional approval' by the European Medicinal Agency (EMA). These mRNA-loaded nanomedicines are formulated through microfluidic mixing and made of four main lipid components: a neutral lipid (e.g., DSPC), cholesterol, a PEG-lipid and an ionizable cationic lipid. Frontrunner drugs (e.g., Doxil®) showed that the neutral lipid and cholesterol provide LNPs with bilayer stability and fusogenic properties [7, 8] ; while PEG lipids control particle size and prevent aggregation and fusion [9] . Ionizable lipids -the most important component of mRNA-LNPs [10] -are designed with a pKa below 7 to allow high encapsulation efficiencies of negatively charged mRNA during the mixing step (pH < 6), while preserving a neutral charge at physiological pH, reducing toxicity associated with the cationic charge and extending the circulation lifetime of the LNP system. At a cellular level, protonation occurring in acidic endosomal compartments favors interaction and mixing with anionic cellular lipids leading to intracellular release of mRNA. In one of the fundamental steps in the process, libraries of different ionizable lipids are screened to identify the optimal lipid species to boost protein expression and induce better immune responses while maintaining low toxicity profiles [11] [12] [13] . In order to speed up the development process, this optimization step was skipped and Moderna and BioNTech/Pfizer turned to a lipid composition inspired by Patirisan (trade name Onpattro® by Alnylam Pharmaceuticals), an orphan drug with a short interfering RNA (siRNA)-LNP system, that was already approved by the FDA for the treatment of hereditary transthyretin (TTR) amyloidosis [14] . The main difference with respect to Onpattro® was the usage of new ionizable cationic lipids (ALC-0315 and SM-102 in BNT162b2/Comirnaty and mRNA-1273/SpikeVax respectively) [15] . However, a persistent theme J o u r n a l P r e -p r o o f in gene delivery has been the design of transport technologies according to biomedical demand. Design considerations are made about the payload to be delivered, the therapeutic target and the route of administration, i.e., typically intramuscular (IM) or intravenous (IV). It is well known that the encapsulated genetic payload affects size, shape and, most importantly, nanoscale arrangement of lipid systems. The exact arrangement of mRNA encapsulated in LNPs has been poorly addressed so far, while most information has been obtained with siRNA-LNP systems. Depending on the siRNA/lipid molar ratio (i.e., phosphate over ionizable cationic lipid molar ratios) and on the lipid ingredients, siRNA-LNPs may exhibit a multilamellar structure, a nanostructure core a homogeneous core shell [16, 17] . Some authors also reported the existence of cubic structures with Ia3d and Pm3n as crystallographic space groups where siRNA is confined within bicontinuous aqueous channels [18, 19] . According to previous findings [20] the superior efficient silencing of these structures was attributed to enhanced fusion with endosomal membranes promoted by the positive Gaussian modulus of the cubic phases K G >0. However, whether and to what extent the lessons learned from the structure of siRNA-LNPs can apply to the mRNA-LNPs is under debate [17, 21] . mRNA is at least 100-fold larger than siRNA, and this may affect the inner structure of the LNP. Using cryo-TEM, small-angle X-ray scattering (SAXS) and small-angle neutron scattering (SANS) Arteta et al. [22] suggested that mRNA is located inside water cylinders, which are coated by cationic lipids and proposed that this peculiar arrangement can determine instability under non-frozen storage conditions. While mRNA is certainly positioned within the particle interior, the nanoscale organization of lipids and mRNA remains ambiguous, with more research needed to confirm the morphology of mRNA-LNPs. Nanoscale arrangement is a critical determinant for the functional delivery of nucleic acids and it is a central dogma that distinct morphologies and nanostructures are associated with totally different transfection performances [23] . This shows how using the same standards (research results, rules, procedures, and protocols) of an approved drug (e.g., Onpattro®) can generate safe and well-functioning vaccines in a short J o u r n a l P r e -p r o o f time, but better understanding of particle attributes (e.g., structure-activity relationship) should be prioritized to optimize mRNA-LNPs and create effective immunization delivery systems. Most of the time, the therapeutic target dictates the choice of the administration route as it influences organ distribution, expression kinetics and therapeutic outcomes [24] . The lipid composition of Onpattro® (a ionizable cationic lipid, a neutral lipid, cholesterol and a PEGylated lipid in proper proportions) was specifically designed for hepatocyte targeting following IV administration. A major challenge often linked with IV administration of LNPs is their short circulation time. Ionizable amino-lipids help maintain a more biocompatible neutral charge, while PEG-lipids also give particles a ‗stealth' effect. This shields the LNP surface and limits the adsorption of serum proteins, ensuring protection against mononuclear phagocyte systemic uptake and thus contributing to increased circulation time [25] . However, both lipid components have their own drawbacks. For instance, ionizable cationic lipids are susceptible to temperature-and pHdependent hydrolysis, which has a detrimental effect on particle stability, while high concentrations of PEG lipids prevent the delivery of RNA into cells (the -PEG dilemma‖) [26] . Immune responses to PEG molecules have also been reported to result in the accelerated blood clearance (‗ABC') phenomenon [27] . A major pitfall of IV administration in the context of immunomodulatory agents is the risk of systemic impact and cytokine storm [28] , making this route less desirable for LNP-mRNA vaccine candidate development. Injection by IV also requires administration in a medical setting by a trained professional. [29] . This means that the use of ionizable and PEGylate lipids that are key components of LNPs given to patients by IV administration may not be necessary upon IM injection. Future research will be aimed at understanding whether cationic lipids and alternative materials to PEG-lipids such as zwitterionic materials [30] and artificial protein coatings [31] may even provide a superior efficacy of COVID-19 vaccine [32] . So far, the synthetic identity of LNP-based vaccines in terms of proportions of lipids or lipid/mRNA ratio has been developed through phenomenological approaches. Recent research has clarified the wide gap existing between benchtop discoveries and clinical success of LNP-based vaccines is partly due to partly a result of our poor understanding of the relationship between their synthetic identity and cellular activity. Decrypting this code is a priority to maximize the intracellular release of mRNA, the synthesis of the S protein and the stimulation of the immune system to produce nAb. In this regard, quantitative structure-activity relationship (QSAR) methods can be used to predict the relationships between the synthetic identity of mRNA-LNPs and their mechanism of action at a cellular level. mRNA-LNPs can be internalized by multiple mechanisms, including macropinocytosis, clathrin-mediated, caveolae-mediated endocytosis and temperature-independent fusion processes [33, 34] . Once inside the cell, the ability of mRNA-LNPs to escape from the autophagy-lysosomal pathway is still unclear. As endosomes mature and their pH drops to 6 or less ionizable lipids become protonated, changing from being neutral to positively charged. This protonation is likely accompanied by a change in the lipid molecular shape that, ultimately, promotes the formation of inverted highly-fusogenic structure [35, 36] . Recently, Maugeri et al. [37] also demonstrated that some internalized mRNA-LNPs are secreted into the extracellular matrix as extracellular vesicles (EVs). QSAR approaches assume that LNPs with comparable synthetic identities will interact similarly with cells thus eliciting similar physiological responses (e.g., protein expression and production of antibodies). We envision that J o u r n a l P r e -p r o o f QSAR investigations will enable in vitro prediction of bioactivity and accelerate the development of optimized mRNA-LNP vaccines against COVID-19 [38, 39] . Validation of delivery systems requires the establishment of proper in vitro models for formulation screening. Although 2D cell culture systems play a key role in advancing our understanding of molecular signaling, cellular morphology, and drug discovery, not all results are translatable to physiological in vivo systems [40] . Indeed, 2D cellular models lack the complex tissue structure consisting of neighboring cells, molecules gradients, and extracellular matrix (ECM). It has been demonstrated that nanoparticle uptake in 2D systems reaches a plateau in a few hours while continues to increase over time if the same cells are grown in 3D matrices [41] . These differences are likely the result of unique diffusive behavior, interaction with ECM components [42] [43] [44] , and limited exposure of the cell surface to nanoparticle uptake in 2D cultures. It also seems that endocytic pathways are enhanced when cells are in a 3D arrangement [41] . 3D cell cultures, as well as microfluidic and organ-on-chip systems, represent the current frontline in the recreation of in vivo cell arrangement, mechanical environment, and molecular turnover [45] . In vivo testing brings several disadvantages, including cost, lack of similarity with human response, for example for species-specific pathogens [46] , and ethical concerns [47] . In 1959, the establishment of the 3R principles (replacement, reduction, and refinement) set the basis for experimental research, with the replacement of animal models top of the priority list. Even if complete substitution of animal testing is still unimaginable, the creation of suitable 3D models would allow a reduction of the number of animals used in trials and speed up high throughput testing necessary in fast vaccine development. Several 3D models of lung tissue have been developed for the study of host response towards coronaviruses as well as multiorgan failure, as recently reviewed [48, 49] . 3D cell culture is extremely technically and economically challenging. Recently, 3D additive manufacturing has been applied to biology, where it is referred to as bioprinting. It consists of a layer-by-layer positioning of cells and biochemicals with a high throughput workflow and customizable biological material, including patient-derived cells and body fluids [50] . Even if fabrication efficiency and resolution still do not match the same performance of robotics-based high-throughput screening platforms (10,000-100,000 tests per day), 3D bioprinting is continuously being improved [51] . Recently, Hwang et al. created 9can integrated 3D bioprinter based on microscale continuous optical printing capable of the large-scale production of biological samples production within multi-wells [51, 52] . Administration route dramatically impacts the performance of a vaccine formulation and consequently, 3D models should be chosen accordingly, e.g., for IM, subcutaneous (SC), or intradermal (ID) delivery. 3D bioprinting of muscular tissue has been recently reviewed [53, 54] . Ghobolova and colleagues demonstrated the feasibility of a human bio-artificial muscle model formed by mixing muscle cells, obtained from biopsies, in a fibrin hydrogel to observe the effect of micro-injections. Measured parameters included the release of the injected compounds and their metabolites over time [55] . This kind of model could be further improved by adding immune cells to study responses to the vaccine. Indeed, in only a few models have immune cells been inserted into muscular tissues [56] . Most interestingly, bone marrow-derived macrophages injected into adult-derived muscle 3D cultures enable near-complete tissue repair post-injury, limiting myofiber apoptosis and attenuating inflammatory environment [57] . It should also be noted that considerable progress has been made towards vascularization of 3D muscle tissues and that a proper blood supply could also help mimic cell recruitment during IM vaccination [58] . COVID-19 vaccines are usually IM administered but could be switched to SC or ID routes [59] . For example, the accidental SC injection of the BNT162b2/Comirnaty vaccine, resulted in high immunogenicity (98%) after the first dose in 790 patients [60] . In recent experiments in vivo, J o u r n a l P r e -p r o o f administration of fluorescently labeled HIV-1 envelope glycoprotein on liposomes was used to demonstrate that both SC and IM routes induce efficient immune cell activation, though antigen is delivered to different lymph nodes depending on the route [61, 62] . A few skin-targeted SARS-CoV-2 vaccines are being studied, based on DNA, mRNA, or viral subunits [59] . Indeed, the cutaneous microenvironment is an ideal target for vaccination since the skin contains a high density of APC and generates a high sustained innate immune response, even at distant organs. The use of 3D cell cultures also is advantageous to follow dynamic cell-cell interactions via multiple fluorescent labeling or other techniques designed to track interactions between immune cells [63, 64] . However, proper training is necessary for ID administration unless specific technologies like microneedle patches are adopted [65] . Certain adjuvants, which may be toxic with IM injection, could be feasible via SC or ID delivery [59] . The identification of route-specific adjuvants, as well as LNP specific formulations, will contribute significantly to next-generation vaccine development. 3D tissue models will enable a fundamental understanding of immune activation and overall efficiency to be obtained rapidly using different administration routes as is necessary in a pandemic situation (Figure 1 ). Skin bioprinting, including the reconstruction of skin appendages, is a research field that has mainly focused on wound healing to replace autologous skin transplantation [66, 67] . Natural skin contains several types of cells, including Langerhans cells, dendritic cells, macrophages, T cells and pluripotent stem cells. Importantly, there are several differences between mouse and human skin immune systems in terms of the amount and position of T cells, and chemokines expressed. These differences may contribute to the failure of translating results obtained in animal models to humans [67] . Since a specialized medium is required to maintain different cell types and can influence marker expression, the combination of many cell types in one model is technically complicated. Concerning immune response, it has been demonstrated that skin differentiation cell media influence response to stimuli and migration of macrophages [68, 69] . Furthermore, spatial organization and the physical constraints provided by 3D culture influence gene expression and cell behavior therefore a controlled bioprinting technology is indispensable for skin reconstruction. [72] . Moreover, a 3D-printed functional human skin has been implanted in vivo in immunodeficient mouse and became indistinguishable from mouse skin after transplantation [73] . Whether these systems will effectively recapitulate human immunological response and substitute in vivo testing in the future is still an open question, however immunocompetent skin is currently available from companies such as Genoskin® for the testing of allergens, psoriasis and drug efficacy after subcutaneous injections, with the InflammaSkin® model able to establish the interplay between activated skin-resident T cells (Th17) and keratinocytes [74] . Finally, it should be mentioned that bioprinting technology can be also used to assess the effect of vaccination on newborns, in terms of safety and efficacy for protection by the placental passage of antibodies against SARS-CoV-2. Several case reports of 2021, reported antibodies in the cord blood after maternal vaccination [75] [76] [77] . In vivo study of blood-placenta barrier (BPB) passage is clearly impossible in human and difficult and time-consuming in animal models. Furthermore BPB physiology is complicated to reproduce with classical 2D systems [78] . [48, 79] . This model was further improved by using primary placental fibroblasts within the biological membrane and primary human placental endothelial cells [80] . In the coming years, currently available 3D bioprinting techniques of tissues for vaccine testing will likely improve. In summary, there is ample room to improve mRNA-based candidates for vaccines or therapeutics that are under clinical trials and at the preclinical stage [81] . We foresee that the knowledge gained from the development and clinical use of LNPs in SARS-CoV-2 vaccines gives new impetus to development of other LNP-based therapeutics. Of equal importance is the development of ever more accurate biomimetic 3D tissue models capable of simulating human immune response. 3D bioprinted models allow to optimization and choice of administration route. We are convinced that the enormous efforts that we are observing in the technological development of new methods such as 3D bioprinting will allow faster and more predictive vaccine testing than animal models in the future. Nanomaterial delivery systems for mRNA vaccines Engineering of the current nucleoside-modified mRNA-LNP vaccines against SARS-CoV-2 Middle East respiratory syndrome: obstacles and prospects for vaccine development The effect of different lipid components on the in vitro stability and release kinetics of liposome formulations Influence of cholesterol on liposome stability and on in vitro drug release Liposome longevity and stability in circulation: effects on the in vivo delivery to tumors and therapeutic efficacy of encapsulated anthracyclines Next-generation lipids in RNA interference therapeutics Ionizable lipid nanoparticle-mediated mRNA delivery for human CAR T cell engineering A combinatorial library of lipid nanoparticles for RNA delivery to leukocytes Ionizable lipid nanoparticles encapsulating barcoded mRNA for accelerated in vivo delivery screening The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs Lipid-Based Nanoparticles in the Clinic and Clinical Trials: From Cancer Nanomedicine Structure of lipid nanoparticles containing siRNA or mRNA by dynamic nuclear polarization-enhanced NMR spectroscopy Crommelin, mRNA-lipid nanoparticle COVID-19 vaccines: structure and stability Biocompatible Nanovector of siRNA Consisting of Arginine-Based Cationic Lipid for Gene Knockdown in Cancer Cells Efficient cellular knockdown mediated by siRNA nanovectors of gemini cationic lipids having delocalizable headgroups and oligo-oxyethylene spacers Highly efficient gene silencing activity of siRNA embedded in a nanostructured gyroid cubic lipid matrix mRNA-based therapeutics-developing a new class of drugs Successful reprogramming of cellular protein production through mRNA delivered by functionalized lipid nanoparticles Naturally-occurring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA mRNA vaccine: a potential therapeutic strategy PEGylation as a strategy for improving nanoparticle-based drug and gene delivery The polyethyleneglycol dilemma: advantage and disadvantage of PEGylation of liposomes for systemic genes and nucleic acids delivery to tumors The accelerated blood clearance (ABC) phenomenon: clinical challenge and approaches to manage Current concepts in the diagnosis and management of cytokine release syndrome Optimization of lipid nanoparticles for intramuscular administration of mRNA vaccines A decade of the protein corona Interplay of protein corona and immune cells controls blood residency of liposomes The importance of poly (Ethylene glycol) and lipid structure in targeted gene delivery to lymph nodes by lipid nanoparticles Mechanistic understanding of gene delivery mediated by highly efficient multicomponent envelope-type nanoparticle systems Cytosolic delivery of nucleic acids: The case of ionizable lipid nanoparticles Overcoming delivery barriers with LNPs Tailoring lipoplex composition to the lipid composition of plasma membrane: a Trojan horse for cell entry? Linkage between endosomal escape of LNP-mRNA and loading into EVs for transport to other cells Biological identity of nanoparticles in vivo: clinical implications of the protein corona Nano-engineered tools in the diagnosis, therapeutics, prevention, and mitigation of SARS-CoV-2 Advances in the Biofabrication of 3D Skin in vitro: Healthy and Pathological Models Dynamics of nanoparticle diffusion and uptake in three-dimensional cell cultures New perspectives on the roles of nanoscale surface topography in modulating intracellular signaling How do surface properties of nanoparticles influence their diffusion in the extracellular matrix? A model study in Matrigel using polymer-grafted nanoparticles Preventing obstructions of nanosized drug delivery systems by the extracellular matrix Deconstructing the third dimension-how 3D culture microenvironments alter cellular cues Three-dimensional cell culture models for investigating human viruses Innovative human three-dimensional tissue-engineered models as an alternative to animal testing 3D tissue models as an effective tool for studying viruses and vaccine development Bioengineered in vitro tissue models to study SARS-CoV-2 pathogenesis and therapeutic validation An insight on advances and applications of 3d bioprinting: A review 3D Printing of Pharmaceutical Application: Drug Screening and Drug Delivery High throughput direct 3D bioprinting in multiwell plates 3D graphene scaffolds for skeletal muscle regeneration: future perspectives Bioprinting of 3D in vitro skeletal muscle models: A review Human tissue-engineered skeletal muscle: a novel 3D in vitro model for drug disposition and toxicity after intramuscular injection Engineering skeletal muscle: Building complexity to achieve functionality, in: Semin Incorporation of macrophages into engineered skeletal muscle enables enhanced muscle regeneration Vascularization of tissueengineered skeletal muscle constructs Microarray patches enable the development of skin-targeted vaccines against COVID-19 Sub-cutaneous Pfizer/BioNTech COVID-19 vaccine administration results in seroconversion among young adults Route of vaccine administration alters antigen trafficking but not innate or adaptive immunity Vaccine delivery systems toward lymph nodes Monitoring T cell-dendritic cell interactions in vivo by intercellular enzymatic labelling A novel sample holder for 4D live cell imaging to study cellular dynamics in complex 3D tissue cultures Transdermal vaccination via 3D-printed microneedles induces potent humoral and cellular immunity 3D bioprinting for skin tissue engineering: Current status and perspectives Toward immunocompetent 3D skin models In Vitro Models Mimicking Immune Response in the Skin In vitro skin culture media influence the viability and inflammatory response of primary macrophages Construction of three-dimensional dermo-epidermal skin equivalents using cell coating technology and their utilization as alternative skin for permeation studies and skin irritation tests Development of full-thickness human skin equivalents with blood and lymph-like capillary networks by cell coating technology Human skin 3D bioprinting using scaffold-free approach 3D bioprinting of functional human skin: production and in vivo analysis Development and characterization of a human Th17-driven ex vivo skin inflammation model Newborn antibodies to SARS-CoV-2 detected in cord blood after maternal vaccination-a case report Maternal and neonatal SARS-CoV-2 antibodies assessment after mRNA maternal vaccination in the third trimester of pregnancy Cord blood antibody following maternal SARS-CoV-2 inactive vaccine (CoronaVac) administration during the pregnancy Bioengineered Microphysiological Placental Models: Towards Improving Understanding of Pregnancy Health and Disease Biomimetic placenta-fetus model demonstrating maternal-fetal transmission and fetal neural toxicity of zika virus Inspired by the human placenta: a novel 3D bioprinted membrane system to create barrier models COVID-19 vaccines: where we stand and challenges ahead During his post-doctoral formation, he carried out research in the area of biophysics, from single molecules to cells and tissues. Current main research interests are the development of delivery systems for macromolecular drugs, the cytotoxicity of nanomaterials and their interactions with biological systems and 3D printing She is co-founder and co-Principal investigator of the NanoDelivery Lab whose mission is to generate and disseminate knowledge in nanomedicine by combining physics, chemistry, biology, pharmacology and nanotechnology and their translation into innovative, clinically applicable therapeutics and diagnostics. She has been a recipient of grants from the Italian Ministry of Health and the Italian Foundation for Cancer Research VP is a medical biotechnologist and obtained a PhD in Oncobiology and Medical Oncology in 2014. She worked in collaboration with several national and international groups and obtained National awards and International Grants such as Fondazione Umberto Veronesi Post-Doctoral Grant He is a recipient of multiple grants from the Italian Foundation for Cancer Research (AIRC), other public agencies and private companies. HIGHLIGHTS -Lipid nanoparticles allow high encapsulation and protection of mRNA in vaccines -New methods predict the LNPs composition-cellular response relationship -3D bioprinting recapitulate human tissues and could expedite vaccine availability ☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.