banner

News

Jan 09, 2024

Targeted modulation of immune cells and tissues using engineered biomaterials

Nature Reviews Bioengineering volume 1, pages 107–124 (2023)Cite this article

12k Accesses

9 Citations

41 Altmetric

Metrics details

Therapies modulating the immune system offer the prospect of treating a wide range of conditions including infectious diseases, cancer and autoimmunity. Biomaterials can promote specific targeting of immune cell subsets in peripheral or lymphoid tissues and modulate the dosage, timing and location of stimulation, thereby improving the safety and efficacy of vaccines and immunotherapies. Here, we review recent advances in biomaterials-based strategies, focusing on targeting of lymphoid tissues, circulating leukocytes, tissue-resident immune cells and immune cells at disease sites. These approaches can improve the potency and efficacy of immunotherapies by promoting immunity or tolerance against different diseases.

In immunotherapy, choosing the right target cell, tissue and treatment duration is essential to ensure effective immunomodulation while avoiding toxicity.

Biomaterial-mediated targeting of immune cells in lymph nodes improves the potency and efficacy of vaccines by promoting immunity or tolerance.

Circulating migratory immune cells can be targeted to perform as living chaperones to carry therapeutics into tissues.

Systemic administration or intratumoral injection of nanomaterials and therapeutic depots can selectively accumulate and target immune cells in tumours.

Reducing biomaterial complexity is essential to facilitate clinical translation.

Modulating the immune system is a pivotal treatment strategy of modern medicine. Of the top ten drugs (by sales in 2021), seven acted on the immune system, including, notably, the messenger RNA (mRNA)-based vaccines for COVID-19 (ref. 1). Beyond its established role in vaccine development, immunomodulation has therapeutic potential for various conditions including autoimmunity and cancer, as well as inflammatory, fibrotic and infectious diseases. However, immunomodulation is a two-edged sword, as is most clearly demonstrated in the field of cancer immunotherapy, where despite inducing substantial anti-tumour immunity, systemic administration of many immunotherapy drugs has resulted in immune-related adverse events distal to tumour sites2. Therefore, delivering precise dosages of immunomodulatory drugs with spatiotemporal control to specific cells and tissues, while avoiding unwanted off-target stimulation, is essential to ensure a safe and effective immune response.

Targeting of immune cells can be achieved using traditional protein-engineering strategies, employing monoclonal antibodies, engineered binding proteins or recombinant native ligands for immune cell surface receptors. These approaches are at various stages of clinical development, with mono- and multi-specific antibodies being the most advanced3,4,5,6,7. However, the only cell surface protein that truly defines a lymphocyte as relevant to a particular disease is its antigen receptor, making targeting of disease-specific immune cells challenging. In addition, simply linking an antibody domain to an immunomodulatory drug is not always sufficient to achieve effective targeting, because the therapeutic payload can dominate the biodistribution of such fusions8. Engineered biomaterials can introduce additional functionality beyond simple binding to target cells by concentrating immunotherapy agents in specific tissue sites, controlling their release kinetics, and/or controlling their intracellular localization.

The immune system consists of well-defined regional control centres (lymphoid organs), important tissue-resident cell populations (especially at barrier tissues such as mucosal surfaces) and mobile cell populations that constantly recirculate through blood and tissues, providing challenges and opportunities as a therapeutic target (Fig. 1a). Here, we review the recent advances of biomaterials-enabled therapeutic strategies for in vivo targeted modulation of the immune system. We focus on approaches targeting immune cells and lymphoid organs, excluding direct targeting of pathogens such as viruses or bacteria, followed by a discussion on prospects and challenges for developments in this area.

a, Anatomical distribution of primary and secondary lymphoid organs. b, General organization of secondary lymphoid organs and sites of key interactions leading to adaptive immunity, using the lymph node as an example. Shown are orchestrated steps in the early activation of adaptive immune response in response to an antigen (orange). (1a) Dendritic cells (grey) acquire antigens trafficked into the lymph node or migrate to the lymph node from peripheral tissues carrying antigens, which they then present to naive T cells (blue) to drive T cell activation and proliferation. (1b) B cells (yellow) bind to antigens arriving in follicles, triggering initial B cell activation and proliferation. (2) Early-activated B cells receive help signals from activated CD4+ T cells at the T zone–follicle border, providing signals to drive entry into germinal centres. (3) Activated B cells enter germinal centres where they undergo proliferation and somatic hypermutation to affinity mature their antibody receptor through interactions with follicular helper T cells and the antigens captured on the dendrites of follicular dendritic cells (FDCs). GALT, gut-associated lymphoid tissue; NALT, nasal-associated lymphoid tissue.

Lymphoid organs coordinate the maturation and migration of immune cells while organizing and regulating immune responses (Fig. 1a). Primary lymphoid organs in adults include the bone marrow and thymus, which serve as niches for lymphocyte development. Secondary lymphoid organs — which include 600–800 lymph nodes distributed across the body, the spleen and the mucosa-associated lymphoid tissue — house and organize T cells, B cells and antigen-presenting cells (APCs). These organs serve as command centres of adaptive immunity where activation of naive B and T lymphocytes occurs (Fig. 1a,b) and are thus natural targets for vaccines and immunotherapies.

Lymph nodes interface with peripheral tissues through lymphatic vessels, which drain lymph fluid from all tissues and provide a conduit for immune cell trafficking. Drugs and vaccines can be targeted to lymph nodes through lymphatic uptake following parenteral injection. A major factor influencing lymphatic targeting is the physical size of the injected agents: following injection into tissue (including tumours), particles larger than 50–100 nm in diameter become trapped in the extracellular matrix (ECM), whereas particles in the size range of 5–50 nm convect with lymph into the lymphatic vessels and flow to draining lymph nodes (dLNs). By contrast, particles smaller than 5 nm partition preferentially into the blood rather than the lymph9,10 (Fig. 2a). These approximate size ranges depend on the tissue site of injection (which can vary in ECM and lymphatic composition) and factors such as injection volume and rate (which can cause mechanical expansion of the flaps mediating entry into lymphatic vessels), especially in small-animal models, where tissue volume relative to injection volume is much smaller. Furthermore, particle shape, charge and surface chemistry can also play a part by promoting or hindering convection through the tissue and lymphatic entry11. Notably, capture of particles at the downstream dLN is a less studied but equally important prerequisite for lymph node modulation, as evidenced by data showing that proteins12 and small hydrophilic nanoparticles13 can pass through the entire lymphatic chain, reach the thoracic duct and enter the systemic circulation following a parenteral injection. In this regard, using adjuvants that promote compound entry into lymph nodes14, or designing carriers that stimulate recognition by macrophages lining the subcapsular and medullary sinuses, can prove useful15.

a, Passive targeting of lymphatics through particle size. Particles smaller than 5 nm are preferentially cleared into the blood vasculature, whereas particles larger than 50 nm tend to become trapped in the tissue. Intermediate-sized particles (5–50 nm) exhibit preferential trafficking into lymphatic vessels. b, Targeting migratory leukocytes to traffic vaccines and therapeutics to draining lymph nodes. Shown are examples of injectable or implantable biomaterials that attract monocytes or dendritic cells from the local tissue and peripheral blood. The cells are then stimulated by the implanted material, triggering activation and differentiation into migratory antigen-presenting cells that carry antigens or other compounds to the draining lymph nodes. ECM, extracellular matrix.

Protein nanoparticles with surface-arrayed antigens and a size optimized for efficient lymphatic uptake enhance the immunogenicity of vaccines16,17,18,19,20. Protein nanoparticle vaccines are currently in clinical trials for human immunodeficiency virus (HIV), SARS-CoV-2 and influenza, and have been shown to be safe and elicit potent neutralizing antibody responses in humans21,22 (Table 1). Lipid nanodiscs and block copolymer micelles smaller than 50 nm also efficiently traffic and deliver compounds into lymphatics23,24. Cage-like nanoparticles formed by the self-assembly of saponin and lipids (termed ISCOMs) are potent vaccine adjuvants of an ideal size (about 40 nm) for lymph node targeting, which received emergency use approval in the USA against SARS-CoV-2 (clinical trial EudraCT 2020-004123-16)25. Another strategy to promote lymphatic uptake is to exploit albumin and lipoproteins that naturally traffic from blood to lymph. Amphiphilic conjugates — consisting of peptides, proteins or small molecules linked to an albumin-binding lipid tail through a hydrophilic poly(ethylene glycol) (PEG) spacer (called amph-vaccines) — bind to endogenous albumin in the tissue following parenteral injection, improving lymph node targeting26,27,28,29. For example, conjugation of an albumin-binding lipid tail to a CpG oligonucleotide adjuvant resulted in a 12-fold increase in lymph node exposure following subcutaneous immunization in mice, compared with unmodified CpG26. This approach is currently being tested in the clinic for lymph node targeting of a cancer vaccine (NCT04853017; Table 1)30. Lymph node-targeting nanoparticles are also being used to deliver latency-reverting drugs in HIV cure strategies31 and to induce immunological tolerance32,33,34,35. Nanoparticles composed of self-assembled proteins, synthetic bioresorbable polymers or lipid assemblies have different in vivo lifetimes and stabilities that can affect their functionality. For example, proteolysis of protein nanoparticle vaccines could lead to particle disassembly and loss of antigen multimerization. However, such effects have received limited attention to date.

Complementary to the strategy of direct lymphatic targeting, this approach leverages the natural process by which immune cells, including dendritic cells (DCs), monocytes and neutrophils, internalize antigens in peripheral tissues and transport them to lymph nodes as part of their constitutive immune surveillance (Fig. 2b). For example, subcutaneously implanted porous polymer scaffolds releasing the cytokine granulocyte–macrophage colony-stimulating factor (GM-CSF) attract and differentiate monocytes into the DC lineage. Molecular adjuvants and tumour antigens carried by these scaffolds are taken up by the recruited cells and transported to dLNs36,37 (Fig. 2b). This approach improved anti-tumour immunity, eliciting approximately 50% complete responses in an aggressive mouse model of melanoma and leading to a first-in-humans clinical trial of this technology (NCT01753089; Table 1). Data from this trial are not yet published, but should provide important insights into the safety of generating a strong localized inflammatory reaction at the implant site, an approach that could be extended to applications beyond cancer. To avoid implantation, injectable polymeric hydrogels38 or suspensions of biodegradable silica particles39 have been employed, resulting in both cellular and humoral immune responses against cancer or microbial antigens40. Similarly, microparticles loaded with tumour lysates as antigen and adenosine triphosphate (ATP) as a chemotaxis-inducing ‘find-me’ signal promoted recruitment of DCs and increased levels of mature DCs migrating to tumour dLNs, resulting in reduced tumour growth41.

DC-attracting biomaterials are also being developed to programme tolerance rather than immunity. Dual-sized poly(lactide-co-glycolide) (PLGA) particles consisting of small (0.5–2.5 μm) phagocytosable microparticles loaded with the tolerogenic metabolite vitamin D3 and autoantigens, combined with large (10–65 μm) non-phagocytosable microparticles designed to release extracellular transforming growth factor-β (TGFβ) and GM-CSF, prevented hyperglycaemia in a mouse model of type 1 diabetes42 and induced antigen-specific tolerance in mouse models of multiple sclerosis and collagen-induced arthritis43,44. This example highlights the potential of DC-attracting approaches not only to modulate immunity, but also to programme tolerance.

Lymph nodes are highly organized organs with defined compartments enriched in different immune cell subsets. Afferent lymph enters the subcapsular sinus (SCS) of the lymph node, a fluid space between the lymph node capsule and the lymph node parenchyma, lined by lymphatic endothelial cells and macrophages (Fig. 1b). Within the parenchyma, many of the key cell populations regulating adaptive immunity have specific niches; B cells and follicular dendritic cells (FDCs) reside in follicles arrayed around the exterior of the node, whereas T cells are localized deeper in the lymph node paracortex45,46 (Fig. 1b). This physical segregation has an important role in orchestrating sequential steps in the immune response, so targeting distinct niches in the lymph node could offer valuable therapeutic opportunities.

Under physiological conditions, substances in the lymph can pass into the parenchyma through narrow collagen conduits, or be captured and transferred into the lymph node interior by SCS macrophages47,48,49 (Fig. 1b). The conduits exclude globular proteins larger than approximately 70 kDa (refs. 49,50), although evidence suggests that this size limit can change in the presence of a live infection51. Vaccine adjuvants such as oil-in-water nanoemulsions and saponin nanoparticles have been used to boost entry of compounds into lymph nodes by inducing rapid death of SCS macrophages14,52,53 (Fig. 3a). Similarly, liposomal formulations of clodronate, which is widely used to deplete SCS macrophages, improve antigen entry into lymph nodes54. In a two-stage delivery strategy, CpG oligonucleotides were coupled to approximately 30-nm poly(propylene sulfide) (PPS) nanoparticles through cleavable oxanorbornadiene linkers and then injected intradermally in a murine model of lymphoma55. The nanoparticles trafficked from the injection site to dLNs, but were trapped in the sinuses after 24 hours. By tuning the chemistry of the oxanorbornadiene linker, the CpG adjuvant payload remained bound to the nanoparticles during trafficking to the dLN, and were later released deep into the lymph node parenchyma (Fig. 3b).

a, Promoting therapeutic entry into lymph nodes through depletion of subcapsular sinus (SCS) macrophages. Vaccine adjuvants (red) induce rapid death of SCS macrophages, leading to enhanced entry of antigens and other compounds (gold) into lymph nodes. b, Polymer nanoparticles (green) efficiently traffic into lymphatic vessels (20–30 nm) but are too large to efficiently penetrate the lymph node paracortex. They chemically release small-molecule payloads (red) that rapidly permeate throughout the node. c, Soluble polymers and polymer nanoparticles conjugated with ligands for receptors expressed by specific target cell types (such as dendritic cells) are taken up by antigen-presenting cells in lymph nodes and co-deliver vaccine antigens or dendritic cell-activating compounds (for example, TLR agonists). d, (1) Nanoparticles activate complement, (2) are captured by SCS macrophages (3) and transferred to non-antigen-specific B cells through complement receptors. (4) These nanoparticles are then transferred to follicular dendritic cells (FDCs), which express high levels of complement and fragment crystallizable (Fc) receptors. e, (1) Ligands for chimeric antigen receptor (CAR) T cells linked to albumin-binding poly(ethylene glycol) (PEG)-lipid moieties traffic from an injection site and bind to endogenous albumin. These compounds are then transferred to lymph nodes (2) where they are inserted into the plasma membrane of macrophages and dendritic cells. Subsequent encounter of CAR T cells with the ligand displayed on the surface of dendritic cells (3) will lead to CAR T cell tandem stimulation by natural costimulatory receptors and cytokine signals provided by the ligand-decorated dendritic cells.

To facilitate intra-nodal delivery, specific cell populations can be targeted within the lymph node parenchyma, in particular DCs and macrophages. For example, oral or systemic administration of rapamycin promotes tolerance by acting on DCs and myeloid cells (for example, in organ transplants), though with considerable systemic side effects56. Subcutaneous injection of rapamycin encapsulated in PEG-b-PPS polymer vesicles (about 100 nm) targeted the lymph nodes in a diabetic mouse model, where it was preferentially phagocytosed by DCs and other myeloid cells and induced a regulatory DC phenotype enabling allogeneic pancreatic islet transplants to be maintained over 100 days, without side effects57. To drive antigen-specific tolerance, lymph node DCs were targeted using liposomes co-delivering a tolerance-driving aryl hydrocarbon receptor agonist drug together with a multiple sclerosis-related peptide in a murine multiple sclerosis model58. Cancer neoantigens linked to hydrophobic peptides bearing Toll-like receptor 7 (TLR7) agonists were developed to self-assemble into approximately 20-nm micelles, which efficiently trafficked from subcutaneous injection sites to dLNs, achieving uptake in 80% of lymph node DCs, and leading to anti-tumour immunity in multiple murine tumour models59. DC targeting has also been achieved by targeting specific receptors expressed by these cells. Following intradermal vaccination, polymer chains functionalized with antigens, TLR7 agonists and mannose moieties to target mannose receptor-positive DCs improved uptake of antigens in lymph nodes (Fig. 3c), generating antigen-specific CD8+ T cell responses and increasing serum antibody concentrations (1.7- and 3.6-fold compared with the potent alternative adjuvant formulations AS01EL and poly(I:C), respectively)60. These studies highlight the potential of co-delivering antigens and adjuvant compounds to the same cell through physical conjugation or encapsulation within the polymer carrier, a widely adopted strategy59,61,62. Moreover, the physical chemistry of antigen and adjuvant compounds, together with selective design of nanoparticle carriers to enable release into DCs upon arrival in the lymph nodes, are important to consider to ensure optimal T cell priming and functional polarization62,63,64. For example, autoimmune-related peptide antigens coupled to 10-nm quantum dots trafficked from subcutaneous injection sites to dLNs, where they accumulated in macrophage receptor with collagenous structure (MARCO)-positive macrophages65. MARCO+ cells have been associated with immune tolerance and expanded regulatory T (Treg) cells, contributing to disease control in a mouse model of multiple sclerosis65. Therefore, targeting these lymph node APC populations seems promising for promoting both immunity and tolerance. Of note, in this work, quantum dots were used owing to their bright fluorescent properties, but are known to release toxic metals over time66. In general, an important question to address with any polymer-based or nanoparticle-based DC-targeting strategy is whether the material is effectively captured in the dLN chain following injection, or whether it passes through the thoracic duct and enters the systemic circulation13, raising potential toxicity concerns, in particular for potent immunostimulants.

B cell follicles are another important niche regulating humoral immune responses. Nanoparticles engineered to activate the complement system can target antigens or therapeutics to B cell follicles. For example, nanoparticles bearing a high density of mannose-containing glycans are recognized by the innate immune protein mannose-binding lectin, which triggers complement deposition on the particles. Upon arrival to the lymph nodes, SCS macrophages transfer complement-decorated particles to migratory B cells in the lymph node parenchyma, which in turn pass the particle to FDCs53,67 (Fig. 3d). For vaccines, this strategy is glycan density dependent, amplifies the germinal centre and serum antibody responses and can be engineered into synthetic particles to promote follicle localization67,68. A protein nanoparticle vaccine that triggers this FDC targeting process completed a phase I trial (NCT03547245) and demonstrated 97% efficacy in initiating broadly neutralizing antibody B cell lineages in humans69. In principle, particles triggering complement activation through natural immunoglobulin M (IgM) or the alternative pathway should also be capable of such FDC localization. However, this trafficking is size-dependent; complement-decorated particles smaller than 15 nm were internalized and cleared by FDCs, whereas particles 50 nm or larger were retained on FDC dendrites for several weeks, resulting in improved antibody responses70. An important parameter to consider when designing carriers targeting B cells is that rigid particles carrying multivalent copies of an antigen are more effective than flexible polymers in triggering B cell activation71.

T cells are another key cell type in lymph nodes, located in the paracortex and interfollicular regions (Fig. 1b). Polymer nanoparticles coated with DC-derived plasma membrane fragments and conjugated with anti-CD3 antibodies showed efficient accumulation in dLNs, activated and expanded CD8+ T cells, and, in combination with anti-PD1 therapy elicited anti-tumour immunity72. To provide vaccine-like stimulation of chimeric antigen receptor (CAR) T cells in lymph nodes, albumin-binding ‘amph-vaccine’ molecules were designed carrying CAR ligands. The lipid tails of these conjugates inserted into cell membranes of DCs and macrophages in the dLN, enabling activation of CAR T cells in the lymph node and improving anti-tumour activity73 (Fig. 3e).

Unlike live infections, current vaccines fail to elicit sufficient CD8+ T cell responses in humans, which has motivated the field to explore new ways to prime cellular immunity74. Vaccines usually act locally on a chain of lymph nodes draining the injection site, such as a muscle75. However, this means that reacting immune cells are confined to a few lymph nodes, resulting in competition for limited resources (for example, cytokines or antigens), which, theoretically, could restrain the eventual systemic immune response. One hypothesis is that systemic administration of antigens to many lymph nodes could alleviate such resource constraints and enable more potent immune priming. A key consideration is that adjuvant and antigen signals need to be delivered together to ensure that immunity, rather than tolerance, is elicited. Moreover, intravenously administered materials are rapidly opsonized and captured by macrophages in the liver, spleen and bone marrow, in particular particles larger than 100 nm. By contrast, particles smaller than 5 nm are rapidly cleared through the kidneys76. Therefore, to systemically target lymphoid organs, opsonization-resistant (for example, PEGylated) particles in the range of 5–100 nm would be ideal, although this size dependency could be altered if circulating immune cells take up the particles and actively transport them into systemic lymph nodes. For example, intravenous administration of negatively charged interferon-inducing lipid nanoparticles carrying antigen-encoding mRNA efficiently targeted DCs in the spleen and peripheral lymph nodes as well as myeloid cells in the liver and bone marrow, resulting in a substantial antigen-specific T cell response (30–60% of the total CD8+ T cell compartment), and consequent tumour rejection in aggressive murine tumour models77 (Fig. 4). These findings spurred ongoing clinical trials of mRNA lipid nanoparticles in multiple cancers, with promising interim results including objective response rates in 6 out of 17 patients with advanced melanoma78,79 (NCT04534205, NCT03418480 and NCT02410733; Table 1). Similarly, intravenous vaccination with self-assembled polymer nanoparticles carrying cancer neoantigen peptides and small-molecule TLR7/8 agonists primed a larger TCF1+ stem-like antigen-specific T cell population compared with parenteral vaccination with the same material, leading to improved anti-tumour immunity80. These examples highlight the potential beneficial effects of systemic immunization on the immune response. Following these principles, a clinical trial in the Netherlands is currently evaluating intravenously administered PLGA-based nanoparticles, delivering a tumour antigen together with a small molecule as innate immune activator designed to promote DC activation through invariant natural killer (NK) T cells (NCT04751786, Table 1)81.

Nanoparticle carriers can target lymphoid organs systemically, including lymph nodes, spleen, bone marrow and tolerogenic antigen-presenting cells in the liver. For example, apolipoprotein A-based lipid nanodiscs are preferentially taken up by myeloid cells in the liver, spleen and bone marrow. Stabilin-functionalized nanoparticles are efficiently scavenged by endothelial cells in the liver. Anionic lipid nanoparticles target antigen-presenting cells in the liver, spleen, lymph nodes and bone marrow.

APC populations in the liver and spleen are known to have important roles in systemic tolerance, and are therefore being targeted for tolerance and autoimmunity. For example, intravenously administered biodegradable PLGA nanoparticles carrying rapamycin were efficiently captured by DCs in the spleen, and elicit a Treg cell-generating, tolerizing phenotype in these cells that blocked generation of cellular and humoral responses against co-administered antigens82. Following this approach, a clinical trial aimed at blocking anti-drug antibody responses against recombinant uricase in gout patients demonstrated promising efficacy and safety data in humans (NCT02648269)83. Similarly, intravenous administration of biodegradable polymer nanoparticles coupled to autoantigens for tolerance induction led to efficient uptake in tolerance-promoting MARCO+ APCs in the spleen and liver, fostering protection in mouse models of diabetes and other autoimmune conditions84,85. Subcutaneously injected diabetes autoantigens coupled to hyaluronic acid polymers were delivered to both disseminated lymph nodes and spleen, promoting systemic expansion of CD4+ T cells with an immunosuppressive phenotype86. Intravenous injection of biodegradable PLGA nanoparticles loaded with a model antigen and functionalized with stabilin to target scavenger receptors expressed by liver sinusoidal endothelial cells resulted in an almost exclusive accumulation in the liver, providing protection from airway-induced allergic inflammation87 (Fig. 4). As a final example, liposomes were administered intraperitoneally to deliver α-galactosylceramide to marginal zone B cells in the spleen. Presentation of the ligand to invariant NK T cells promoted the development of Treg cells and tolerogenic DCs in the spleen of mice88. This treatment suppressed graft-versus-host disease following bone marrow transplant89 and is now moving into early-stage clinical trials (NCT04014790 and NCT01379209; Table 1)90. In these examples, different materials were used to target antigens and stimuli to relevant immune cells. An important but understudied parameter is how the duration of cargo release in recipient target cells affects their tolerizing effect. For example, loaded PLGA particles could be engineered to release antigens over weeks, whereas fast-degrading surface-conjugated particles or liposomes could provide rapid release following endocytosis. Whether this kinetics will have a substantial impact remains unknown.

The bone marrow is a primary lymphoid organ for haematopoiesis and myelopoiesis, as well as a site housing a large pool of memory T cells91 (Fig. 1a). Myeloid precursors, in particular, have received attention because they drive a process known as ‘trained immunity’. This phenomenon occurs when innate immune cells or their progenitors receive certain microbial stimuli (including Bacillus Calmette–Guérin bacteria, Candida albicans yeast, β-glucan or peptidoglycan92) that drive epigenetic changes rewiring their metabolic and functional state93,94. When induced in progenitor cells, these epigenetic modifications are passed on to their progeny and confer a prolonged and elevated state of responsiveness to microbial stimuli in monocytes, macrophages and NK cells. Therapeutic induction (or suppression) of trained immunity is now being considered as a promising approach to treat diverse diseases92. For example, intravenous injection of apolipoprotein-A1-based lipid nanodiscs carrying the minimal peptidoglycan muramyl dipeptide preferentially accumulated in myeloid cells and their precursors in blood, spleen and tumours in a mouse model of melanoma, resulting in enhanced cytokine production and increased efficacy of checkpoint blockade therapy95 (Fig. 4). Such innate immune modulation could be a powerful complement to therapeutic strategies focused on boosting adaptive immunity.

Many immune cells continuously recirculate between secondary lymphoid organs, blood and tissues, patrolling for foreign antigens. The ability of immune cells in blood to home in on disease sites forms the basis of adoptive cell therapies, where engineered immune cells are infused intravenously into patients and subsequently arrive at targeted distal sites. These cells can be targeted while in the blood to serve as chaperones to transport drugs into disease sites.

Adoptive cell therapies based on the injection of autologous cells engineered to have disease-specific functions is a steadily expanding approach for immunotherapy96,97. To date, the most clinically successful example is CAR T cell therapy, where lymphocytes isolated from cancer patients are transduced to express a CAR that enables recognition and selective killing of tumour cells. CAR T cells are then expanded ex vivo and re-infused into the patient98. So far, six different CAR T cell products have been approved for haematological malignancies, with several ongoing clinical trials to extend CAR T therapy to solid tumours and infectious diseases99,100,101. Adoptive cell therapies are also being developed using natural or transgenic T cell receptor (TCR)-expressing T cells, NK cells, macrophages, Treg cells (for autoimmune disease and transplant tolerance) and other immune cell populations96. Adoptively transferred cells benefit from supporting stimulation to maintain their function or promote their expansion and survival; however, as with other immunomodulators, systemic co-administration of supporting drugs can be toxic owing to non-specific effects on endogenous immune cells.

To provide sustained and localized stimulation, therapeutic carriers (‘backpacks’) have been directly attached to the surface of donor cells prior to infusion (Fig. 5a). This strategy has been used to load tumour-specific T cells and Treg cells with cytokines, small-molecule drugs or chemotherapeutic agents102,103,104,105. Early studies showed that lipid-nanoparticle backpacks carrying cytokines such as IL-15 remained on the cell surface for extended periods, inducing potent autocrine stimulation and proliferation of adopted antigen-specific T cells in vivo102,105. Backpacking protein nanogels onto T cells enabled a more controlled release of supporting cytokines in response to altered cell surface reducing activity after TCR or CAR stimulation in mice106. This strategy restricted donor cell stimulation to tumours and tumour-dLNs (where the antigen is encountered), improving therapeutic efficacy and safety106. Interim results from a phase I clinical trial of this backpacking approach in cancer patients (NCT03815682; Table 1) showed that only one-tenth the amount of administered IL-15 was detectable in the blood despite backpacking three times the maximum tolerated dose of free systemic fragment crystallizable (Fc)-fused IL-15 (ref. 107) onto T cells, leading to stable disease in 10 out of 17 patients108. A limitation of this approach is that the therapeutic payload is, by definition, finite, and will be diluted over time as T cells proliferate in vivo. However, it could still provide immediate autocrine stimulation following adoptive transfer, enabling cells to successfully engraft and initiate tumour rejection. Owing to the cytokine carrier being directly conjugated to the cell membrane, the concentration of newly released cytokines at the cell surface is substantial, and it has been estimated that the IL-15 nanogels will continue to stimulate T cells through at least seven cell divisions before stimulatory capacity is lost106.

a, ‘Backpacking’ cells by attaching drug-releasing materials to cells ex vivo prior to adoptive transfer. b, Nanoparticles functionalized with targeting moieties can bind to circulating immune cells in blood, which then traffic into different tissue sites with the drug carrier. c, T cell-targeted nanoparticles deliver nucleic acid payloads (DNA or RNA) to circulating lymphocytes, resulting in the expression of chimeric antigen receptors (CARs) and other immunomodulatory gene payloads in situ. cRGD, cyclic arginine–glycine–aspartic acid; IFN, interferon; IL-15 SA, interleukin-15 super agonist; PLGA, poly(lactide-co-glycolide); TCR, T cell receptor.

Backpacking approaches have also been developed for innate immune cells; discoid polymer particles carrying interferon-γ (IFNγ) adhered to the surface of macrophages without inducing phagocytosis, resulting in persistent polarization into an ‘M1’ phenotype that promoted tumour rejection in a murine breast cancer model109. Importantly, the use of a discoid morphology was crucial to avoiding phagocytosis of the backpack110 (Fig. 5a).

Targeting immune cells in the blood is an attractive strategy to circumvent physical barriers in tissues. For example, lymphocytes populating Peyer’s patches and mesenteric lymph nodes express well-defined adhesion and chemokine receptors (α4β7 integrin and CCR9, respectively111,112) that direct their homing to these sites (Fig. 5b). Intravenous administration of lipid-coated polymer nanoparticles loaded with an HIV antiretroviral drug and surface-functionalized with antibodies against α4β7 promoted uptake in gut lamina propria cells, which transported the particles to the gut of mice113. Functionalizing small interfering RNA (siRNA)-loaded lipid nanoparticles with recombinant mucosal vascular addressin cell adhesion molecule 1 (MAdCAM1), the ligand recognized by the high-affinity conformation of α4β7, increased the particles’ specificity to gut-homing lymphocytes and thus resulted in silencing of IFNγ in a murine model of colitis114.

Nanoparticles have also been used to target circulating immune cells that home into tumours or sites of inflammation. Intraperitoneally injected hyaluronic acid-polyethyleneimine hybrid nanoparticles carrying miR-125b (a microRNA known to induce an anti-tumour ‘M1’ phenotype in macrophages115), were taken up by these cells, which then migrated to lung tumours and repolarized tumour-associated macrophages towards an M1 phenotype. We note that choosing the correct administration route is important to avoid unexpected transduction of macrophages in the liver or bone marrow. Intravenous administration of liposomes or lipid nanoemulsions conjugated with arginine–glycine–aspartic acid (RGD) peptides (known to bind to αV integrins, which are highly expressed by monocytes and neutrophils) led to rapid uptake by these cells in the blood, which subsequently carried the particles to tumours in mice116 (Fig. 5b). The same approach enabled the delivery of neuroprotective drugs across the blood–brain barrier to sites of ischaemia in mice117. Thus, targeting of immune cells in the blood is a promising strategy to hitchhike drugs into disease sites.

Targeting of antigen-specific T cells in the blood has been pursued to enhance adoptive cell therapy and promote protective T cell responses. For example, in mouse models of autoimmunity, intravenous injection of iron oxide nanoparticles coated with disease-relevant peptide–major histocompatibility complexes (pMHC) induced expansion of CD4+ or CD8+ T cells with a regulatory phenotype118,119. Intravenously injected class II pMHC-displaying nanoparticles can further expand Treg cells to control liver autoimmune diseases without systemically suppressing immunity in other organs120. For cancer applications, artificial APCs were generated by conjugating biodegradable PLGA-poly(β-amino-ester) (PBAE) microparticles with pMHC and anti-CD28 antibodies121. Intravenous administration of these particles expanded antigen-specific cytotoxic CD8+ T cells in vivo, and, in combination with checkpoint blockade therapy, increased the median survival time by 31% compared with single checkpoint blockade therapy in a mouse model of melanoma121. The same strategy was used to develop tolerogenic particles that promoted the expansion of forkhead box P3 (FOXP3+) Treg cells, which, after a single intravenous injection, resulted in a 20% increase of these cells in lymph nodes compared with untreated mice122.

CAR T cell therapy is having a substantial clinical impact against haematological malignancies, but its patient-specific nature and the complex manufacturing process of transducing T cells ex vivo limits its widespread application123. A potential alternative is to genetically modify T cells in vivo. Engineered viral vectors are being developed for this purpose124,125; however, viral vectors raise concerns of strong immune reactions and toxicity126,127,128. Another option is to use synthetic nanoparticle gene-delivery vectors. Intravenously injected poly(β-amino ester) nanoparticles carrying transposon DNA encoding a CAR, which targets T cells through anti-CD3 antibody fragments, proved safe and effective in mouse models of leukaemia129 (Fig. 5c). Similarly, in vivo mRNA delivery to T cells generated transient CAR T cells capable of eliminating fibrosis-promoting fibroblasts in models of heart failure130. We note that immune cells (including T cells) are notoriously resistant to conventional transfection methods, so it is important to identify more effective lipid–polymer compositions for nucleic acid delivery131,132,133. CAR T cells have also been produced in vivo using an implantable macroporous alginate scaffold, which provides a contact interface for T cells and retroviruses and facilitates vector-mediated CAR gene transfer134. Subcutaneous implantation of these scaffolds seeded with human peripheral blood mononuclear cells and CD19-encoding retroviral particles reduced ex vivo cell-processing times to a single day, compared to 2–4 weeks for conventional CAR T cells, while maintaining similar anti-tumour efficacy134.

Some innate and adaptive immune cell populations permanently reside in tissues and provide functions such as immediate immune defence at barrier tissues, local production of protective antibodies and tissue-resident immune memory. In addition, these cells infiltrate sites of tissue damage, inflammation and tumours, making them important therapeutic targets for disease treatment.

Several immune cell populations reside at mucosal barriers, such as the skin, the airways, and the gastrointestinal and reproductive tracts. DCs serve as sentinels of pathogen entry by sampling foreign antigens, whereas tissue-resident memory lymphocytes and plasma cells provide frontline adaptive immune protection135,136, and, meanwhile, mucosa-associated lymphoid tissues, such as the Peyer’s patches and nasal-associated lymphoid tissue (NALT), serve as secondary lymphoid organs that rapidly respond to antigens entering through the mucosae137. These tissues are important targets for vaccination, because antigens taken up in mucosa draining lymphoid tissues prime lymphocytes that home back to mucosal tissue sites135. Moreover, mucosa-resident immune cells can be preferential targets of infection (for example, gut-resident memory CD4+ T cells in HIV infection), making them interesting therapeutic targets for viral latency-reverting drugs against HIV31,138. A key challenge in targeting immune cells at these sites is that mucosal barriers have evolved to block incoming pathogens and foreign materials efficiently139,140.

One promising strategy for effective drug delivery across mucosal surfaces is to exploit a natural bidirectional transport pathway for crossing the epithelial barrier. Following endocytosis in epithelial cells, immunoglobulin G (IgG) and albumin bind to the neonatal fragment crystallizable receptor (FcRn) in endosomes, triggering transcytosis and release at the apical surface141. Fusing therapeutic proteins with Fc domains or albumin promotes uptake at the mucosal surface142 (Fig. 6a). Similar to the backpacking approach, protein or peptide antigens linked to PEG-lipids (amph-vaccines) associate with albumin in the airway fluid and ‘hitchhike’ through the mucus across the epithelial barrier in an FcRn-dependent manner28,143. Peptide amph-vaccines that were administered into the lungs primed robust lung tissue-resident memory T cells that enhanced vaccine protection against respiratory viral or tumour challenge28. Alternatively, intranasal administration of protein amph-vaccines amplified germinal centre responses in the NALT, promoting higher mucosal antibody titres in both mice (100–1,000-fold increase) and non-human primates (10-fold increase), compared with unmodified proteins143.

a, Albumin is used as a chaperone to transport vaccines across the epithelial barrier in the nasal and respiratory pathways through the neonatal fragment crystallizable (FcRn) receptor. Nanoparticles functionalized with hyaluronic acid are phagocytosed by macrophages and other myeloid cells at sites of intestinal inflammation in the gut. b, Systemically injected nanoparticles passively accumulate in tumours through the enhanced permeation and retention effect, or functionalized nanoparticles actively target cells such as PD1+ lymphocytes. Alternatively, biomaterials can be directly injected into tumours to locally present or release immunostimulatory agents in the tumour microenvironment or promote gene delivery to tumour cells for localized expression of immunomodulatory factors. c, Nanoparticles can be modified with targeting moieties or opsonization-triggering components to bind to the endothelial cells of high endothelial venules (HEVs) of the inflamed draining lymph nodes or target different innate immune cells that home to sites of inflammation. cRGD, cyclic arginine–glycine–aspartic acid; HDL, high-density lipoprotein; NALT, nasal-associated lymphoid tissue; PNAd, peripheral node addressin.

Biomaterials are also being developed to target immune cells at sites of mucosal inflammation. For example, conjugation of the hydrophilic ECM polymer hyaluronic acid with the natural gut anti-oxidant bilirubin led to the formation of nanoparticles 80–400 nm in size. After oral administration in mouse models of colitis, these nanoparticles were efficiently taken up at the inflamed epithelium by macrophages expressing the hyaluronic acid receptor CD44, leading to polarization of these cells toward an anti-inflammatory phenotype144 (Fig. 6a). Interestingly, this treatment caused an increase in microbial diversity in the microbiome, which promoted epithelial healing and inflammation reduction.

Instead of targeting circulating immune cells destined to migrate into tumours, there is also interest in delivering drugs directly to tumour-resident immune cells. For many years, biomaterials scientists have sought to target tumours by exploiting the enhanced permeation and retention (EPR) effect, a phenomenon first described in animal models in the late 1980s in which large proteins or nanoparticles accumulate in tumours owing to their hyperpermeable vasculature and reduced lymphatic clearance76 (Fig. 6b). However, the efficiency of EPR-based tumour targeting remains low and penetration of nanoparticles deep into tumours is often poor145,146. Therefore, new approaches are being pursued to facilitate targeting of immune cells in tumours. For example, the stimulator of interferon genes (STING) pathway, a cytosolic danger sensor in host immune cells147, has gained particular interest. However, its natural ligands, cyclic dinucleotides (CDNs), are labile and are poorly taken up by cells in vivo. Conjugating PEGylated lipid nanodiscs with CDN prodrugs enabled penetration into solid tumours, activating DCs to drive T cell-mediated tumour rejection148. Passive nanoparticle uptake in tumours can be further exploited to localize immune cell activation to tumours. For example, intravenously injected PEGylated gold nanorods accumulated in tumours and were then irradiated by a near-infrared laser to induce photothermal heating of the tumour-resident nanorods149. This heating activated thermally responsive gene cassettes in co-administered engineered T cells and induced localized CAR or cytokine expression in mice149. However, the clearing mechanism of gold nanoparticles remains unclear, because these materials are not resorbable in vivo150 (Fig. 6b). Nanoparticles have also been designed to target T cells in tumours through conjugation with antibody fragments specific to T cell receptors. For example, PD1-targeted PLGA–PEG-based nanoparticles were detected on intratumoral T cells within 1 hour of intravenous injection and enabled pronounced therapeutic modulation of tumours through delivery of a small-molecule TLR7/8 agonist151.

One factor limiting nanoparticle dissemination deep into the tumour parenchyma is the propensity of nanoparticles to be captured by tumour-associated macrophages residing near blood vessels152,153,154. This affinity is now being exploited to reprogram tumour myeloid cells into a pro-immune phenotype using polymeric or lipid carriers loaded with immunomodulators such as TLR7/8 agonists and colony-stimulating factor 1 receptor inhibitors155,156. As another example, polymer nanoparticles carrying mRNA targeted to myeloid cells through di-mannose moieties reprogrammed tumour-associated macrophages towards an M1 phenotype157. Although promising, these approaches will need to assess the potential side effects of non-specific uptake by macrophages in the liver, spleen and bone marrow, given the tendency of nanoparticles to be captured by macrophages in these organs158,159.

In addition to capture by macrophages, abnormal tumour vasculature limits dissemination of nanoparticles and immune cells in the tumour microenvironment (TME). Inhibiting proangiogenic factors, such as vascular endothelial growth factor (VEGF) and angiopoietin 2 (ANG2) (at low to intermediate doses) normalizes tumour vasculature and perfusion, thereby improving the efficacy of anticancer treatments, including immunotherapy160,161,162,163,164,165. For example, tumour biopsies from breast cancer patients treated with an anti-VEGF antibody in a phase III clinical trial (NCT00546156) showed increased infiltration of CD4+ T cells, CD8+ T cells and mature DCs166. Moreover, combining antiangiogenic agents with immunotherapies, including cancer vaccines and immune-checkpoint inhibitors, enhanced tumour infiltration of effector immune cells and improved therapeutic efficacy in murine cancer models167,168,169,170,171,172,173,174.

Biomaterials can also be directly injected into the tumour. Importantly, to ensure efficacy, the delivered payload needs to remain in the tumour (or tumour-dLNs) after injection. Conjugation of immunomodulatory payloads to antibodies or engineered proteins that bind to tumour cell surface antigens175 or ECM components such as collagen176,177,178 improves safety and efficacy compared with intratumoral or peritumoral administration of free immunotherapy drugs. However, these approaches suffer from short-lived tumour stimulation (typically only a few days or less) because these agents are internalized by tumour cells or released out of the TME176. Biomaterial-based treatments can extend the window of drug exposure and improve efficacy from single injections, which is important for treating deep visceral lesions that require surgical access. For example, IL-12 bearing a phosphoserine peptide tag enables stable high-avidity binding to aluminium hydroxide (alum), a common clinical vaccine adjuvant179. Intratumoral injection of cytokine-loaded alum particles allowed drug retention in tumours for more than 2 weeks, resulting in pronounced regressions and complete responses in several tumour models in mice (Fig. 6b). Similarly, intratumoral injection of spherical nucleic acids, liposomal nanoparticles with surface-conjugated oligonucleotides as ligands for TLR9, led to strong immunomodulatory activity180. In a phase Ib/II clinical trial, as of the data cut-off date (1 July 2021), out of 14 patients, 2 had a complete response and 1 had a partial response, and side effects were mostly limited to injection site reactions and flu-like symptoms181,182 (NCT03684785; Table 1). Another promising approach consists of in situ vaccination using viral nanoparticles derived from the virus cowpea mosaic virus, which shifts the immune composition of the TME towards a pro-inflammatory profile183,184.

Biomaterials are also being considered as depots for the local release of immunotherapy payloads to the TME (Fig. 6b). For example, chitosan microparticles have been developed that, following intratumoral injection, released IL-12 over 1–2 weeks and elicited complete tumour regression in 80–100% of animals in murine cancer models185,186. Highlighting its synergistic potential, in pancreatic cancer models in mice, intratumoral injection of IL-12-loaded polymer microspheres at 24 hours following local stereotactic body radiation therapy resulted in increased TME reprogramming, systemic T cell responses and anti-tumour efficacy compared with IL-12-loaded microspheres or radiation-only treatments187. Hydrogels releasing innate immune-stimulating small-molecule drugs, such as TLR7/8 or STING agonists, decreased tumour recurrence when implanted at surgical resection sites in mouse models of breast and lung cancer188. Sprayable fibrin hydrogels releasing an anti-CD47 antibody at tumour resection sites stimulated macrophage phagocytic activity while neutralizing the acidic pH at the tumour site, triggering myeloid cells to eliminate residual tumour cells189.

Biomaterials have also been used to deliver and locally stimulate adoptively transferred T cells in tumours. For example, intratumoral implantation of alginate hydrogels loaded with polyclonal tumour-specific T cells or CAR T cells, cytokines and immunostimulatory antibodies, led to enhanced tumour elimination in mouse models of breast and ovarian cancer190. In the adjuvant setting, hyaluronic acid hydrogels encapsulating CAR T cells together with stimulatory factors for localized immunostimulation implanted into the tumour bed following surgical resection protected against tumour recurrence and inhibited distant tumour growth191. In another report, scaffolds made of nitinol metal, a material routinely used as cardiovascular stents, were functionalized with T cell-stimulatory antibodies and loaded with CAR T cells for peritumoral implantation. In a mouse model of non-resectable ovarian cancer, these scaffolds eradicated tumours in 70% of animals and extended the average tumour survival time by 2.7-fold compared with untreated controls192. These described materials have different in vivo lifetimes and might require surgical retrieval. Clinical implications are still unclear because none of these approaches have entered clinical testing yet.

Engineered materials can also be used to increase uptake and promote cytosolic delivery of therapeutics in immune cells following localized delivery to tumour sites. For example, intratumoral administration of endosome-disrupting polymersomes delivering CDNs into phagocytes improved STING activation, indicated by an 11-fold decrease in melanoma growth rate and complete tumour rejection in one-third of the mice193. Similarly, intratumoral injection of CDN-loaded lipid-calcium phosphate nanoparticles surface-treated with phosphatidyl serine promoted CDN uptake by myeloid cells and DCs in the pleural space in a mouse model of malignant pleural effusion194. mRNA also requires cytosolic delivery to immune cells (Fig. 6b). Intratumoral injection of mixtures of lipid nanoparticles delivering mRNA encoding cytokines and membrane-bound T cell costimulatory receptor OX40L led to pronounced immune activation and complete tumour regression in 50% of animals bearing subcutaneous hepatoma tumours195. Similarly, intratumoral injection of saline formulations or polymer nanoparticles delivering mRNAs encoding selected potent cytokine combinations (for example IL-12, GM-CSF, IL-15 and IFNα) led to robust tumour regressions and induction of systemic T cell responses that regressed even distal untreated lesions196,197. Interim data from a follow-up phase I clinical trial showed that lipid nanoparticles encapsulating mRNAs encoding OX40L, IL-23 and IL-36γ, in combination with a PD1 immune checkpoint inhibitor, resulted in increased levels of proinflammatory cytokines in both plasma and tumour biopsies (NCT03739931; Table 1)198. These lipid nanoparticle formulations were shown to primarily transduce cancer cells and myeloid cells. Another approach is to deliver mRNA directly into tumour-infiltrating T cells; lipid nanoparticles incorporating ionizable lipids transfected around 5% and 10% of tumour-infiltrating CD4+ and CD8+ primary T cells, respectively, following intratumorual injection in a murine melanoma model, and, when used to deliver mRNA encoding OX40 in combination with systemic administration of anti-OX40 agonist antibodies, resulted in 60% tumour rejection in a murine lymphoma model131.

Immune cells are central to the pathology of inflammatory diseases, therefore, targeting them at sites of inflammation is a potentially effective strategy for disease treatment. Phagocytes are involved in generating persistent inflammation, thereby providing a first-line target choice for drug development199. For example, polymersomes, vesicles formed by the self-assembly of amphiphilic block copolymers, were efficiently taken up by neutrophils through scavenger receptors200. Using pH-responsive polymersomes that disrupt endosomes in response to acidification of these compartments, cytosolic delivery of cyclin-dependent kinase inhibitor R-roscovitine induced neutrophil apoptosis and suppressed inflammation in a sterile injury zebrafish model. Immune cells can also serve as chaperones to concentrate nanocarriers at sites of inflammation and angiogenesis; for example, intravenous administration of nanoemulsions functionalized with cyclic RGD peptides that bind to αVβ3 integrins expressed on inflamed blood vessels rapidly adhered to circulating neutrophils and monoyctes in a mouse model of acute wound-derived inflammation within minutes. These nanoparticles also directly bound to the endothelium of inflamed vessels at later times, suggesting transfer from leukocytes to endothelial cells201. Neutrophils are known to effectively capture opsonized particles in the blood, a process exploited to target these cells at sites of lung inflammation using nanoparticles of different size, charge and composition, provided they are rapidly opsonized with complement upon injection199 (Fig. 6c). Intravenously injected synthetic high-density lipoprotein nanoparticles carrying the mTOR inhibitor efficiently targeted myeloid cells and blocked the expression of inflammatory cytokines by macrophages infiltrating heart allografts, resulting in long-term allograft survival202. Interestingly, intravenously injected empty PLGA nanoparticles were reported to be taken up by monocytes and neutrophils in the blood, leading to their reprogramming into an anti-inflammatory phenotype and localization to sites of spinal cord injury, promoting a pro-regenerative milieu that enabled axon recovery203.

Biomaterials are also being explored to modulate adaptive immune responses at tissue transplant sites. For example, biodegradable polymer microspheres releasing TGFβ promoted Treg cell development in vitro, and promoted Treg cell infiltration into allogeneic pancreatic β-cell transplants for treatment of an in vivo diabetes model204. In a rat model of vascularized hindlimb allogeneic tissue grafting, local administration of microspheres releasing TGFβ, rapamycin and IL-2 promoted induction and expansion of Treg cells at the implant site, leading to long-term tissue engraftment205.

Lymph node draining sites of disease are also modulated by upstream inflammation. For example, high endothelial venules (HEVs) are specialized blood vessels of the lymph nodes that express the lymphocyte homing protein peripheral node addressin (PNAd), which supports naive lymphocyte entry into lymph nodes. Notably, PNAd expression is upregulated in lymph node draining sites of chronic inflammation206. Exploiting this process, intravenously injected PLGA microparticles carrying the immunosuppressive drug tacrolimus and functionalized with a PNAd-targeting antibody enabled accumulation of the drug at lymph nodes draining the transplanted tissues207. As microparticles are too large to passively transport across the endothelium, this finding suggests particle accumulation on the luminal surfaces of the HEVs, followed by drug diffusion into the tissue across the endothelial barrier. Building on these findings, PNAd-targeted PLGA nanoparticles were found not only to bind to the HEVs of inflamed dLNs, but were also further transported into the lymph node parenchyma, leading to substantially prolonged allograft survival in a model of MHC-mismatched heart tissue transplantation208 (Fig. 6c).

Biomaterial-mediated targeting of immune cells and tissues is starting to have clinical impact, with great promise for the future of vaccines and immunotherapies. However, there remain important fundamental aspects of immunology to be understood, and a number of technological and translational challenges must be solved if these technologies are to reach their full potential.

For example, there is still much to learn about the dynamics of immune cells during disease and treatment. The reciprocal trafficking of lymphocytes from lymph nodes to blood and tumours, as well as between tumours, has received limited attention209,210. However, this information is essential for the design of targeted immune-oncology therapeutics that will ‘hit’ cells at a desired location or time point in their life cycle. The role of stem-like CD8+ T cells in diseases, such as autoimmunity211, infections212 and cancer213,214, is gaining particular attention, because these cells appear to play an important part as responders to immunotherapies such as checkpoint blockade212,215, by producing progenies that are important effector cells (either in lymph nodes or directly at peripheral tissue sites). These cells are promising targets for immunotherapy, but how they can be generated and expanded in vivo remains poorly understood (Box 1).

Interactions of biomaterials with the immune system also need further assessment. The SARS-CoV-2 pandemic revealed that lipid nanoparticles used for mRNA delivery have direct adjuvant activity in vaccines216,217,218 by triggering inflammatory cytokine production in lymph nodes; however, the exact mechanisms remain to be elucidated. Other biomaterials, for example biodegradable polymers such as PLGA, are generally considered passive scaffolds or drug-release matrices; however, lactic acid produced by PLGA hydrolysis is known to be immunomodulatory219,220,221, and PLGA particles promote a pro-regenerative phenotypic state when phagocytosed by innate immune cells203. These effects can promote tolerance in conditions such as autoimmune disease222, but would be undesirable in other settings, for example in cancer immunotherapy.

One reality that the field of biomaterials-based immune therapies must face is that complex multicomponent formulations requiring the production of multiple recombinant proteins and exotic multi-step chemistries using different clinical-grade materials are unlikely to be clinically translated owing to the prohibitive cost and complexities involved in the good manufacturing practice (GMP) production of such systems. To ensure clinical translation, ‘less is more’ should be a motivating mantra. This reality is evident by looking at the technologies undergoing clinical trials (Table 1). Furthermore, technologies are often confined to the expertise of individual laboratories. Sharing methods in the field will enable broader experimental validation and testing in different disease models.

The success of lipid-nanoparticle-delivered SARS-CoV-2 mRNA vaccines provides an important proof of concept in humans, but there is much room for improvement to realize their full potential. Despite important advances over the past 20 years, lipid nanoparticles remain substantially worse than biological vectors, such as viruses, at transfecting cells. Systemic or intratumoral injection of lipid nanoparticles to deliver mRNA to T cells transfects only 2% or less of T cells129,196. In addition, what makes lipid nanoparticles suitable for packaging nucleic acids (for example, their charge) also makes them prone to opsonization and non-specific binding to cells and matrices. Moreover, allergic reactions elicited in a small proportion of individuals with current PEG-stabilized lipid nanoparticle formulations represent a potential safety issue223,224. Despite the clinical success of lipid nanoparticles as delivery materials for mRNA and siRNA, other materials, such as endosome-responsive polymers225,226,227, can outperform lipid nanoparticles for nucleic acid delivery in some cell types. Furthermore, combinatorial library studies of lipid nanoparticle compositions have revealed that lipid nanoparticle formulations can be tuned to target specific tissues and immune cell types without even the need for specific antibody or other binder-based targeting199,228,229. Materials with improved delivery efficiency will increase the utility of mRNA, as well as DNA and CRISPR-based gene-editing systems.

Despite existing knowledge gaps and challenges, the use of biomaterials to enable cell-specific and organ-specific targeted immune modulation is an important and exciting area of research and clinical development, which could unlock the full potential of immunotherapies for different diseases.

Modulation of the immune system can only be effective if the targeted cells and their responses are well understood. For example, different phenotypic and functional states exist for antigen-specific T cells. Inactivated precursors, early activated cells (including short-lived and memory precursor effector cells), activated effector cells, memory cells and memory stem cells and ‘exhausted’ effector cells231 have been defined, all with different functionalities and ability to respond to immunostimulation. ‘Stem-like’ CD8+ T cells, in particular, have characteristics of both memory cells and more activated cells213,214,215,232,233, and express specific transcriptional and protein signatures (for example, T cell factor 1 (TCF1+), PD1+) and self-ligand receptor of the signalling lymphocytic activation molecule 6 (SLAM6+)), which are important modulators of anti-tumour immune response. Fundamental studies identifying these important, but possibly rare, cell types will be required to develop targeted immunotherapies and vaccines.

Urquhart, L. Top companies and drugs by sales in 2021. Nat. Rev. Drug Discov. 21, 251 (2022).

Article Google Scholar

Dougan, M., Luoma, A. M., Dougan, S. K. & Wucherpfennig, K. W. Understanding and treating the inflammatory adverse events of cancer immunotherapy. Cell 184, 1575–1588 (2021).

Article Google Scholar

Seung, E. et al. A trispecific antibody targeting HER2 and T cells inhibits breast cancer growth via CD4 cells. Nature 603, 328–334 (2022).

Article Google Scholar

Muik, A. et al. Preclinical characterization and phase I trial results of a bispecific antibody targeting PD-L1 and 4-1BB (GEN1046) in patients with advanced refractory solid tumors. Cancer Discov. 12, 1248–1265 (2022).

Article Google Scholar

Neri, D. Antibody–cytokine fusions: versatile products for the modulation of anticancer immunity. Cancer Immunol. Res. 7, 348–354 (2019).

Article Google Scholar

Jones, D. S. II et al. Cell surface-tethered IL-12 repolarizes the tumor immune microenvironment to enhance the efficacy of adoptive T cell therapy. Sci. Adv. 8, eabi8075 (2022).

Ren, Z. et al. Selective delivery of low-affinity IL-2 to PD-1+ T cells rejuvenates antitumor immunity with reduced toxicity. J. Clin. Invest. 132, e153604 (2022).

Article Google Scholar

Tzeng, A., Kwan, B. H., Opel, C. F., Navaratna, T. & Wittrup, K. D. Antigen specificity can be irrelevant to immunocytokine efficacy and biodistribution. Proc. Natl Acad. Sci. USA 112, 3320–3325 (2015).

Article Google Scholar

Reddy, S. T. et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat. Biotechnol. 25, 1159–1164 (2007).

Article Google Scholar

McLennan, D. N., Porter, C. J. H. & Charman, S. A. Subcutaneous drug delivery and the role of the lymphatics. Drug. Discov. Today Technol. 2, 89–96 (2005).

Article Google Scholar

Schudel, A., Francis, D. M. & Thomas, S. N. Material design for lymph node drug delivery. Nat. Rev. Mater. 4, 415–428 (2019).

Article Google Scholar

Mehta, N. K. et al. Pharmacokinetic tuning of protein–antigen fusions enhances the immunogenicity of T-cell vaccines. Nat. Biomed. Eng. 4, 636–648 (2020).

Article Google Scholar

Kourtis, I. C. et al. Peripherally administered nanoparticles target monocytic myeloid cells, secondary lymphoid organs and tumors in mice. PLoS ONE 8, e61646 (2013).

Article Google Scholar

Silva, M. et al. A particulate saponin/TLR agonist vaccine adjuvant alters lymph flow and modulates adaptive immunity. Sci. Immunol. 6, eabf1152 (2021).

Article Google Scholar

Moon, J. J. et al. Enhancing humoral responses to a malaria antigen with nanoparticle vaccines that expand Tfh cells and promote germinal center induction. Proc. Natl Acad. Sci. USA 109, 1080–1085 (2012).

Article Google Scholar

Boyoglu-Barnum, S. et al. Quadrivalent influenza nanoparticle vaccines induce broad protection. Nature 592, 623–628 (2021).

Article Google Scholar

Jardine, J. G. et al. HIV-1 broadly neutralizing antibody precursor B cells revealed by germline-targeting immunogen. Science 351, 1458–1463 (2016).

Article Google Scholar

Walls, A. C. et al. Elicitation of potent neutralizing antibody responses by designed protein nanoparticle vaccines for SARS-CoV-2. Cell 183, 1367–1382 (2020).

Article Google Scholar

Marcandalli, J. et al. Induction of potent neutralizing antibody responses by a designed protein nanoparticle vaccine for respiratory syncytial virus. Cell 176, 1420–1431 (2019).

Article Google Scholar

Walls, A. C. et al. Elicitation of broadly protective sarbecovirus immunity by receptor-binding domain nanoparticle vaccines. Cell 184, 5432–5447.e16 (2021).

Article Google Scholar

Houser, K. V. et al. Safety and immunogenicity of a ferritin nanoparticle H2 influenza vaccine in healthy adults: a phase 1 trial. Nat. Med. 28, 383–391 (2022).

Article Google Scholar

Song, J. Y. et al. Safety and immunogenicity of a SARS-CoV-2 recombinant protein nanoparticle vaccine (GBP510) adjuvanted with AS03: a randomized, placebo-controlled, observer-blinded phase 1/2 trial. EClinicalMedicine 51, 101569 (2022).

Article Google Scholar

Kuai, R., Ochyl, L. J., Bahjat, K. S., Schwendeman, A. & Moon, J. J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 16, 489–496 (2016).

Article Google Scholar

Karabin, N. B. et al. Sustained micellar delivery via inducible transitions in nanostructure morphology. Nat. Commun. 9, 624 (2018).

Article Google Scholar

Heath, P. T. et al. Safety and efficacy of NVX-CoV2373 Covid-19 vaccine. N. Engl. J. Med. 385, 1172–1183 (2021).

Article Google Scholar

Liu, H. et al. Structure-based programming of lymph-node targeting in molecular vaccines. Nature 507, 519–522 (2014).

Article Google Scholar

Moynihan, K. D. et al. Enhancement of peptide vaccine immunogenicity by increasing lymphatic drainage and boosting serum stability. Cancer Immunol. Res. 6, 1025–1038 (2018).

Article Google Scholar

Rakhra, K. et al. Exploiting albumin as a mucosal vaccine chaperone for robust generation of lung-resident memory T cells. Sci. Immunol. 6, eabd8003 (2021).

Article Google Scholar

Vrieze, J. D. et al. Potent lymphatic translocation and spatial control over innate immune activation by polymer-lipid amphiphile conjugates of small-molecule TLR7/8 agonists. Angew. Chem. Int. Edn 58, 15390–15395 (2019).

Article Google Scholar

Pant, S. et al. First-in-human phase 1 trial of ELI-002 immunotherapy as treatment for subjects with Kirsten rat sarcoma (KRAS)-mutated pancreatic ductal adenocarcinoma and other solid tumors. J. Clin. Oncol. 40, TPS2701–TPS2701 (2022).

Article Google Scholar

Cao, S. et al. Hybrid nanocarriers incorporating mechanistically distinct drugs for lymphatic CD4+ T cell activation and HIV-1 latency reversal. Sci. Adv. 5, eaav6322 (2019).

Article Google Scholar

Capini, C. et al. Antigen-specific suppression of inflammatory arthritis using liposomes. J. Immunol. 182, 3556–3565 (2009).

Article Google Scholar

Zhou, K. et al. Targeting peripheral immune organs with self‐assembling prodrug nanoparticles ameliorates allogeneic heart transplant rejection. Am. J. Transpl. 21, 3871–3882 (2021).

Article Google Scholar

Kishimoto, T. K. & Maldonado, R. A. Nanoparticles for the induction of antigen-specific immunological tolerance. Front. Immunol. 9, 230 (2018).

Article Google Scholar

Maldonado, R. A. et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc. Natl Acad. Sci. USA 112, E156–E165 (2015).

Article Google Scholar

Ali, O. A., Huebsch, N., Cao, L., Dranoff, G. & Mooney, D. J. Infection-mimicking materials to program dendritic cells in situ. Nat. Mater. 8, 151–158 (2009).

Article Google Scholar

Ali, O. A., Emerich, D., Dranoff, G. & Mooney, D. J. In situ regulation of DC subsets and T cells mediates tumor regression in mice. Sci. Transl Med. 1, 8ra19 (2009).

Article Google Scholar

Shah, N. J. et al. A biomaterial-based vaccine eliciting durable tumour-specific responses against acute myeloid leukaemia. Nat. Biomed. Eng. 4, 40–51 (2020).

Article Google Scholar

Kim, J. et al. Injectable, spontaneously assembling inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy. Nat. Biotechnol. 33, 64–72 (2014).

Article Google Scholar

Super, M. et al. Biomaterial vaccines capturing pathogen-associated molecular patterns protect against bacterial infections and septic shock. Nat. Biomed. Eng. 6, 8–18 (2022).

Article Google Scholar

Lee, J. A. et al. Recruitment of dendritic cells using ‘find-me’ signaling microparticles for personalized cancer immunotherapy. Biomaterials 282, 121412 (2022).

Article Google Scholar

Lewis, J. S. et al. A combination dual-sized microparticle system modulates dendritic cells and prevents type 1 diabetes in prediabetic NOD mice. Clin. Immunol. 160, 90–102 (2015).

Article Google Scholar

Allen, R., Chizari, S., Ma, J. A., Raychaudhuri, S. & Lewis, J. S. Combinatorial, microparticle-based delivery of immune modulators reprograms the dendritic cell phenotype and promotes remission of collagen-induced arthritis in mice. ACS Appl. Bio Mater. 2, 2388–2404 (2019).

Article Google Scholar

Cho, J. J. et al. An antigen-specific semi-therapeutic treatment with local delivery of tolerogenic factors through a dual-sized microparticle system blocks experimental autoimmune encephalomyelitis. Biomaterials 143, 79–92 (2017).

Article Google Scholar

Gerner, M. Y., Casey, K. A., Kastenmuller, W. & Germain, R. N. Dendritic cell and antigen dispersal landscapes regulate T cell immunity. J. Exp. Med. 214, 3105–3122 (2017).

Article Google Scholar

Eickhoff, S. et al. Robust anti-viral immunity requires multiple distinct T cell-dendritic cell interactions. Cell 162, 1322–1337 (2015).

Article Google Scholar

Junt, T. et al. Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells. Nature 450, 110–114 (2007).

Article Google Scholar

Gretz, J. E., Norbury, C. C., Anderson, A. O., Proudfoot, A. E. I. & Shaw, S. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J. Exp. Med. 192, 1425–1440 (2000).

Article Google Scholar

Sixt, M. et al. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 22, 19–29 (2005).

Article Google Scholar

Roozendaal, R. et al. Conduits mediate transport of low-molecular-weight antigen to lymph node follicles. Immunity 30, 264–276 (2009).

Article Google Scholar

Reynoso, G. V. et al. Lymph node conduits transport virions for rapid T cell activation. Nat. Immunol. 20, 602–612 (2019).

Article Google Scholar

Kim, E. H. et al. Squalene emulsion-based vaccine adjuvants stimulate CD8 T cell, but not antibody responses, through a RIPK3-dependent pathway. eLife 9, e52687 (2020).

Article Google Scholar

Detienne, S. et al. Central role of CD169+ lymph node resident macrophages in the adjuvanticity of the QS-21 component of AS01. Sci. Rep. 6, 39475 (2016).

Article Google Scholar

Zhang, Y.-N., Poon, W., Sefton, E. & Chan, W. C. W. Suppressing subcapsular sinus macrophages enhances transport of nanovaccines to lymph node follicles for robust humoral immunity. ACS Nano 14, 9478–9490 (2020).

Article Google Scholar

Schudel, A. et al. Programmable multistage drug delivery to lymph nodes. Nat. Nanotechnol. 15, 491–499 (2020).

Article Google Scholar

Hafiz, M. M. et al. Immunosuppression and procedure-related complications in 26 patients with type 1 diabetes mellitus receiving allogeneic islet cell transplantation. Transplantation 80, 1718–1728 (2005).

Article Google Scholar

Burke, J. A. et al. Subcutaneous nanotherapy repurposes the immunosuppressive mechanism of rapamycin to enhance allogeneic islet graft viability. Nat. Nanotechnol. 17, 319–330 (2022).

Article Google Scholar

Kenison, J. E. et al. Tolerogenic nanoparticles suppress central nervous system inflammation. Proc. Natl Acad. Sci. USA 117, 32017–32028 (2020).

Article Google Scholar

Lynn, G. M. et al. Peptide–TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigens. Nat. Biotechnol. 38, 320–332 (2020).

Article Google Scholar

Wilson, D. S. et al. Antigens reversibly conjugated to a polymeric glyco-adjuvant induce protective humoral and cellular immunity. Nat. Mater. 18, 175–185 (2019).

Article Google Scholar

Shae, D. et al. Co-delivery of peptide neoantigens and stimulator of interferon genes (STING) agonists enhances response to cancer vaccines. ACS Nano 14, 9904–9916 (2020).

Article Google Scholar

Wang, S. et al. Rational vaccinology with spherical nucleic acids. Proc. Natl Acad. Sci. USA. 116, 10473–10481 (2019).

Article Google Scholar

Callmann, C. E. et al. Tumor cell lysate-loaded immunostimulatory spherical nucleic acids as therapeutics for triple-negative breast cancer. Proc. Natl Acad. Sci. USA 117, 17543–17550 (2020).

Article Google Scholar

Froimchuk, E., Oakes, R. S., Kapnick, S. M., Yanes, A. A. & Jewell, C. M. Biophysical properties of self-assembled immune signals impact signal processing and the nature of regulatory immune function. Nano Lett. 21, 3762–3771 (2021).

Article Google Scholar

Hess, K. L. et al. Engineering immunological tolerance using quantum dots to tune the density of self-antigen display. Adv. Funct. Mater. 27, 1700290 (2017).

Article MathSciNet Google Scholar

Liang, Y., Zhang, T. & Tang, M. Toxicity of quantum dots on target organs and immune system. J. Appl. Toxicol. 42, 17–40 (2022).

Article Google Scholar

Tokatlian, T. et al. Innate immune recognition of glycans targets HIV nanoparticle immunogens to germinal centers. Science 363, 649–654 (2019).

Article Google Scholar

Read, B. J. et al. Mannose-binding lectin and complement mediate follicular localization and enhanced immunogenicity of diverse protein nanoparticle immunogens. Cell Rep. 38, 110217 (2022).

Article Google Scholar

Venkatesan, P. Preliminary phase 1 results from an HIV vaccine candidate trial. Lancet Microbe 2, e95 (2021).

Article Google Scholar

Zhang, Y.-N. et al. Nanoparticle size influences antigen retention and presentation in lymph node follicles for humoral immunity. Nano Lett. 19, 7226–7235 (2019).

Article Google Scholar

Veneziano, R. et al. Role of nanoscale antigen organization on B-cell activation probed using DNA origami. Nat. Nanotechnol. 15, 716–723 (2020).

Article Google Scholar

Xiao, P. et al. Engineering nanoscale artificial antigen-presenting cells by metabolic dendritic cell labeling to potentiate cancer immunotherapy. Nano Lett. 21, 2094–2103 (2021).

Article Google Scholar

Ma, L. et al. Enhanced CAR–T cell activity against solid tumors by vaccine boosting through the chimeric receptor. Science 365, 162–168 (2019).

Article Google Scholar

Pennock, N. D., Kedl, J. D. & Kedl, R. M. T cell vaccinology: beyond the reflection of infectious responses. TRENDS Immunol. 37, 170–180 (2016).

Article Google Scholar

Ols, S. et al. Route of vaccine administration alters antigen trafficking but not innate or adaptive immunity. Cell Rep. 30, 3964–3971 (2020).

Article Google Scholar

Golombek, S. K. et al. Tumor targeting via EPR: strategies to enhance patient responses. Adv. Drug. Deliv. Rev. 130, 17–38 (2018).

Article Google Scholar

Kranz, L. M. et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534, 396–401 (2016).

Article Google Scholar

Beyaert, S., Machiels, J.-P. & Schmitz, S. Vaccine-based immunotherapy for head and neck cancers. Cancers 13, 6041 (2021).

Article Google Scholar

Sahin, U. et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 585, 107–112 (2020).

Article Google Scholar

Baharom, F. et al. Intravenous nanoparticle vaccination generates stem-like TCF1+ neoantigen-specific CD8+ T cells. Nat. Immunol. 52, 41–52 (2020).

Google Scholar

Creemers, J. H. A. et al. Assessing the safety, tolerability and efficacy of PLGA-based immunomodulatory nanoparticles in patients with advanced NY-ESO-1-positive cancers: a first-in-human phase I open-label dose-escalation study protocol. BMJ Open 11, e050725 (2021).

Article Google Scholar

Kishimoto, T. K. et al. Improving the efficacy and safety of biologic drugs with tolerogenic nanoparticles. Nat. Nanotechnol. 11, 890–899 (2016).

Article Google Scholar

Sands, E. et al. Tolerogenic nanoparticles mitigate the formation of anti-drug antibodies against pegylated uricase in patients with hyperuricemia. Nat. Commun. 13, 272 (2022).

Article Google Scholar

Prasad, S. et al. Tolerogenic Ag-PLG nanoparticles induce tregs to suppress activated diabetogenic CD4 and CD8 T cells. J. Autoimmun. 89, 112–124 (2018).

Article Google Scholar

Luo, X., Miller, S. D. & Shea, L. D. Immune tolerance for autoimmune disease and cell transplantation. Annu. Rev. Biomed. Eng. 18, 181–205 (2016).

Article Google Scholar

Leon, M. A. et al. Soluble antigen arrays displaying mimotopes direct the response of diabetogenic T cells. ACS Chem. Biol. 14, 1436–1448 (2019).

Article Google Scholar

Liu, Q. et al. Use of polymeric nanoparticle platform targeting the liver to induce Treg-mediated antigen-specific immune tolerance in a pulmonary allergen sensitization model. ACS Nano 13, 4778–4794 (2019).

Article Google Scholar

Ishii, Y. et al. Alpha-galactosylceramide-driven immunotherapy for allergy. Front. Biosci. 13, 6214–6228 (2008).

Article Google Scholar

Duramad, O., Laysang, A., Li, J., Ishii, Y. & Namikawa, R. Pharmacologic expansion of donor-derived, naturally occurring CD4+ Foxp3+ regulatory T cells reduces acute graft-versus-host disease lethality without abrogating the graft-versus-leukemia effect in murine models. Biol. Blood Marrow Transplant. 17, 1154–1168 (2011).

Article Google Scholar

Chen, Y.-B. et al. Pharmacokinetics and safety of KRN7000 following intravenous infusion of RGI-2001 in allogeneic transplant patients. Blood 136, 15–16 (2020).

Google Scholar

Okhrimenko, A. et al. Human memory T cells from the bone marrow are resting and maintain long-lasting systemic memory. Proc. Natl Acad. Sci. USA 111, 9229–9234 (2014).

Article Google Scholar

Mulder, W. J. M., Ochando, J., Joosten, L. A. B., Fayad, Z. A. & Netea, M. G. Therapeutic targeting of trained immunity. Nat. Rev. Drug Discov. 18, 553–566 (2019).

Article Google Scholar

Netea, M. G. et al. Trained immunity: a program of innate immune memory in health and disease. Science 352, aaf1098 (2016).

Article Google Scholar

Netea, M. G. & van der Meer, J. W. M. Trained immunity: an ancient way of remembering. Cell Host Microbe 21, 297–300 (2017).

Article Google Scholar

Priem, B. et al. Trained immunity-promoting nanobiologic therapy suppresses tumor growth and potentiates checkpoint inhibition. Cell 183, 786–801 (2020).

Article Google Scholar

Finck, A. V., Blanchard, T., Roselle, C. P., Golinelli, G. & June, C. H. Engineered cellular immunotherapies in cancer and beyond. Nat. Med. 28, 678–689 (2022).

Article Google Scholar

Ribas, A. T cells as the future of cancer therapy. Cancer Discov. 11, 798–800 (2021).

Article Google Scholar

Hong, M., Clubb, J. D. & Chen, Y. Y. Engineering CAR-T cells for next-generation cancer therapy. Cancer Cell 38, 473–488 (2020).

Article Google Scholar

Barros, L. R. C. et al. Systematic review of available CAR-T cell trials around the world. Cancers 14, 2667 (2022).

Article Google Scholar

National Cancer Institute. CAR T cells: engineering patients’ immune cells to treat their cancers. NCI https://www.cancer.gov/about-cancer/treatment/research/car-t-cells (2022).

Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021).

Article Google Scholar

Siriwon, N. et al. CAR–T cells surface-engineered with drug-encapsulated nanoparticles can ameliorate intratumoral T-cell hypofunction. Cancer Immunol. Res. 6, 812–824 (2018).

Article Google Scholar

Huang, B. et al. Active targeting of chemotherapy to disseminated tumors using nanoparticle-carrying T cells. Sci. Transl Med. 7, 291ra94–291ra94 (2015).

Article Google Scholar

Eskandari, S. K. et al. Regulatory T cells engineered with TCR signaling–responsive IL-2 nanogels suppress alloimmunity in sites of antigen encounter. Sci. Transl Med. 12, eaaw4744 (2020).

Article Google Scholar

Stephan, M. T., Moon, J. J., Um, S. H., Bershteyn, A. & Irvine, D. J. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat. Med. 16, 1035–1041 (2010).

Article Google Scholar

Tang, L. et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 36, 707–716 (2018).

Article Google Scholar

Romee, R. et al. First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood 131, 2515–2527 (2018).

Article Google Scholar

Hamilton, E. et al. 801 PRIMETM IL-15 (RPTR-147): preliminary clinical results and biomarker analysis from a first-in-human phase 1 study of IL-15 loaded peripherally-derived autologous T cell therapy in solid tumor patients. J. Immunother. Cancer 8, A850–A850 (2020).

Google Scholar

ShieldsIV, C. W. et al. Cellular backpacks for macrophage immunotherapy. Sci. Adv. 6, eaaz6579 (2020).

Article Google Scholar

Champion, J. A. & Mitragotri, S. Role of target geometry in phagocytosis. Proc. Natl Acad. Sci. USA. 103, 4930–4934 (2006).

Article Google Scholar

Berlin, C. et al. α4β7 integrin mediates lymphocyte binding to the mucosal vascular addressin MAdCAM-1. Cell 74, 185–195 (1993).

Article Google Scholar

Mora, J. R. et al. Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature 424, 88–93 (2003).

Article Google Scholar

Cao, S., Jiang, Y., Zhang, H., Kondza, N. & Woodrow, K. A. Core–shell nanoparticles for targeted and combination antiretroviral activity in gut-homing T cells. Nanomed. Nanotechnol. Biol. Med. 14, 2143–2153 (2018).

Article Google Scholar

Dammes, N. et al. Conformation-sensitive targeting of lipid nanoparticles for RNA therapeutics. Nat. Nanotechnol. 16, 1030–1038 (2021).

Article Google Scholar

Parayath, N. N., Parikh, A. & Amiji, M. M. Repolarization of tumor-associated macrophages in a genetically engineered nonsmall cell lung cancer model by intraperitoneal administration of hyaluronic acid-based nanoparticles encapsulating microRNA-125b. Nano Lett. 18, 3571–3579 (2018).

Article Google Scholar

Sofias, A. M. et al. Tumor targeting by αvβ3-integrin-specific lipid nanoparticles occurs via phagocyte hitchhiking. ACS Nano 14, 7832–7846 (2020).

Article Google Scholar

Hou, J. et al. Accessing neuroinflammation sites: monocyte/neutrophil-mediated drug delivery for cerebral ischemia. Sci. Adv. 5, eaau8301 (2019).

Article Google Scholar

Tsai, S. et al. Reversal of autoimmunity by boosting memory-like autoregulatory T cells. Immunity 32, 568–580 (2010).

Article Google Scholar

Singha, S. et al. Peptide–MHC-based nanomedicines for autoimmunity function as T-cell receptor microclustering devices. Nat. Nanotechnol. 12, 701–710 (2017).

Article Google Scholar

Umeshappa, C. S. et al. Suppression of a broad spectrum of liver autoimmune pathologies by single peptide-MHC-based nanomedicines. Nat. Commun. 10, 2150 (2019).

Article Google Scholar

Rhodes, K. R. et al. Biodegradable cationic polymer blends for fabrication of enhanced artificial antigen presenting cells to treat melanoma. ACS Appl. Mater. Interf. 13, 7913–7923 (2021).

Article Google Scholar

Rhodes, K. R., Meyer, R. A., Wang, J., Tzeng, S. Y. & Green, J. J. Biomimetic tolerogenic artificial antigen presenting cells for regulatory T cell induction. Acta Biomater. 112, 136–148 (2020).

Article Google Scholar

Yip, A. & Webster, R. M. The market for chimeric antigen receptor T cell therapies. Nat. Rev. Drug Discov. 17, 161–162 (2018).

Article Google Scholar

Agarwal, S., Weidner, T., Thalheimer, F. B. & Buchholz, C. J. In vivo generated human CAR T cells eradicate tumor cells. Oncoimmunology 8, e1671761 (2019).

Article Google Scholar

Parayath, N. N. & Stephan, M. T. In situ programming of CAR T cells. Annu. Rev. Biomed. Eng. 23, 385–405 (2021).

Article Google Scholar

Nayak, S. & Herzog, R. W. Progress and prospects: immune responses to viral vectors. Gene Ther. 17, 295–304 (2010).

Article Google Scholar

Barnes, C., Scheideler, O. & Schaffer, D. Engineering the AAV capsid to evade immune responses. Curr. Opin. Biotech. 60, 99–103 (2019).

Article Google Scholar

Hinderer, C. et al. Severe toxicity in nonhuman primates and piglets following high-dose intravenous administration of an adeno-associated virus vector expressing human SMN. Hum. Gene Ther. 29, 285–298 (2018).

Article Google Scholar

Smith, T. T. et al. In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. Nat. Nanotechnol. 12, 813–820 (2017).

Article Google Scholar

Rurik, J. G. et al. CAR T cells produced in vivo to treat cardiac injury. Science 375, 91–96 (2022).

Article Google Scholar

Li, W. et al. Biomimetic nanoparticles deliver mRNAs encoding costimulatory receptors and enhance T cell mediated cancer immunotherapy. Nat. Commun. 12, 7264 (2021).

Article Google Scholar

Billingsley, M. M. et al. Ionizable lipid nanoparticle-mediated mRNA delivery for human CAR T cell engineering. Nano Lett. 20, 1578–1589 (2020).

Article Google Scholar

McKinlay, C. J., Benner, N. L., Haabeth, O. A., Waymouth, R. M. & Wender, P. A. Enhanced mRNA delivery into lymphocytes enabled by lipid-varied libraries of charge-altering releasable transporters. Proc. Natl Acad. Sci. USA 115, 201805358 (2018).

Article Google Scholar

Agarwalla, P. et al. Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells. Nat. Biotechnol. 40, 1250–1258 (2022).

Article Google Scholar

Mora, J. R. et al. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science 314, 1157–1160 (2006).

Article Google Scholar

Allie, S. R. et al. The establishment of resident memory B cells in the lung requires local antigen encounter. Nat. Immunol. 20, 97–108 (2018).

Article Google Scholar

Kiyono, H. & Fukuyama, S. NALT- versus PEYER’S-patch-mediated mucosal immunity. Nat. Rev. Immunol. 4, 699–710 (2004).

Article Google Scholar

Bowen, A., Sweeney, E. E. & Fernandes, R. Nanoparticle-based immunoengineered approaches for combating HIV. Front. Immunol. 11, 789 (2020).

Article Google Scholar

Li, M. et al. Mucosal vaccines: strategies and challenges. Immunol. Lett. 217, 116–125 (2019).

Article Google Scholar

Iwasaki, A. Exploiting mucosal immunity for antiviral vaccines. Annu. Rev. Immunol. 34, 575–608 (2016).

Article Google Scholar

Sockolosky, J. T. & Szoka, F. C. The neonatal Fc receptor, FcRn, as a target for drug delivery and therapy. Adv. Drug Deliv. Rev. 91, 109–124 (2015).

Article Google Scholar

Fieux, M. et al. FcRn as a transporter for nasal delivery of biologics: a systematic review. Int. J. Mol. Sci. 22, 6475 (2021).

Article Google Scholar

Hartwell, B. L. et al. Intranasal vaccination via lipid-conjugated immunogens promotes antigen persistence and transmucosal uptake to drive mucosal and systemic immunity. Sci. Transl Med. 14, eabn1413 (2022).

Article Google Scholar

Lee, Y. et al. Hyaluronic acid–bilirubin nanomedicine for targeted modulation of dysregulated intestinal barrier, microbiome and immune responses in colitis. Nat. Mater. 19, 118–126 (2019).

Article Google Scholar

Lázaro, Ide & Mooney, D. J. Obstacles and opportunities in a forward vision for cancer nanomedicine. Nat. Mater. 20, 1469–1479 (2021).

Article Google Scholar

Wilhelm, S. et al. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 1, 16014 (2016).

Article Google Scholar

Barber, G. N. STING: infection, inflammation and cancer. Nat. Rev. Immunol. 15, 760–770 (2015).

Article Google Scholar

Dane, E. L. et al. STING agonist delivery by tumour-penetrating PEG-lipid nanodiscs primes robust anticancer immunity. Nat. Mater. 21, 710–720 (2022).

Article Google Scholar

Miller, I. C. et al. Enhanced intratumoural activity of CAR T cells engineered to produce immunomodulators under photothermal control. Nat. Biomed. Eng. 5, 1348–1359 (2021).

Article Google Scholar

Longmire, M., Choyke, P. L. & Kobayashi, H. Clearance properties of nano-sized particles and molecules as imaging agents: considerations and caveats. Nanomedicine 3, 703–717 (2008).

Article Google Scholar

Schmid, D. et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 8, 1747 (2017).

Article Google Scholar

Turk, M. J., Waters, D. J. & Low, P. S. Folate-conjugated liposomes preferentially target macrophages associated with ovarian carcinoma. Cancer Lett. 213, 165–172 (2004).

Article Google Scholar

Miller, M. A. et al. Tumour-associated macrophages act as a slow-release reservoir of nano-therapeutic Pt(IV) pro-drug. Nat. Commun. 6, 8692 (2015).

Article Google Scholar

Dai, Q. et al. Quantifying the ligand-coated nanoparticle delivery to cancer cells in solid tumors. ACS Nano 12, 8423–8435 (2018).

Article Google Scholar

Rodell, C. B. et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat. Biomed. Eng. 2, 578–588 (2018).

Article Google Scholar

Kulkarni, A. et al. A designer self-assembled supramolecule amplifies macrophage immune responses against aggressive cancer. Nat. Biomed. Eng. 2, 589–599 (2018).

Article Google Scholar

Zhang, F. et al. Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat. Commun. 10, 3974 (2019).

Article Google Scholar

Gustafson, H. H., Holt-Casper, D., Grainger, D. W. & Ghandehari, H. Nanoparticle uptake: the phagocyte problem. Nano Today 10, 487–510 (2015).

Article Google Scholar

Chen, D. et al. NIR-II fluorescence imaging reveals bone marrow retention of small polymer nanoparticles. Nano Lett. 21, 798–805 (2021).

Article Google Scholar

Sorensen, A. G. et al. A “vascular normalization index” as potential mechanistic biomarker to predict survival after a single dose of cediranib in recurrent glioblastoma patients. Cancer Res. 69, 5296–5300 (2009).

Article Google Scholar

Dickson, P. V. et al. Bevacizumab-induced transient remodeling of the vasculature in neuroblastoma xenografts results in improved delivery and efficacy of systemically administered chemotherapy. Clin. Cancer Res. 13, 3942–3950 (2007).

Article Google Scholar

Willett, C. G. et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat. Med. 10, 145–147 (2004).

Article Google Scholar

Inai, T. et al. Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am. J. Pathol. 165, 35–52 (2004).

Article Google Scholar

Goel, S., Wong, A. H.-K. & Jain, R. K. Vascular normalization as a therapeutic strategy for malignant and nonmalignant disease. Cold Spring Harb. Perspect. Med. 2, a006486 (2012).

Article Google Scholar

Tong, R. T. 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. 11, 3731–3736 (2004).

Article Google Scholar

Boucher, Y. et al. Bevacizumab improves tumor infiltration of mature dendritic cells and effector T-cells in triple-negative breast cancer patients. npj Precis. Oncol. 5, 62 (2021).

Article Google Scholar

Fukumura, D., Kloepper, J., Amoozgar, Z., Duda, D. G. & Jain, R. K. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 15, 325–340 (2018).

Article Google Scholar

Shigeta, K. et al. Regorafenib combined with PD1 blockade increases CD8 T-cell infiltration by inducing CXCL10 expression in hepatocellular carcinoma. J. Immunother. Cancer 8, e001435 (2020).

Article Google Scholar

Manning, E. A. et al. A vascular endothelial growth factor receptor-2 inhibitor enhances antitumor immunity through an immune-based mechanism. Clin. Cancer Res. 13, 3951–3959 (2007).

Article Google Scholar

Yasuda, S. et al. Simultaneous blockade of programmed death 1 and vascular endothelial growth factor receptor 2 (VEGFR2) induces synergistic anti‐tumour effect in vivo. Clin. Exp. Immunol. 172, 500–506 (2013).

Article Google Scholar

Li, B. et al. Vascular endothelial growth factor blockade reduces intratumoral regulatory T cells and enhances the efficacy of a GM-CSF-secreting cancer immunotherapy. Clin. Cancer Res. 12, 6808–6816 (2006).

Article Google Scholar

Voron, T. et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J. Exp. Med. 212, 139–148 (2015).

Article Google Scholar

Allen, E. et al. Combined antiangiogenic and anti–PD-L1 therapy stimulates tumor immunity through HEV formation. Sci. Transl Med. 9, eaak9679 (2017).

Article Google Scholar

Huang, Y. et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl Acad. Sci. USA 109, 17561–17566 (2012).

Article Google Scholar

Yang, R. K. et al. Intratumoral hu14.18-IL-2 (IC) induces local and systemic antitumor effects that involve both activated T and NK cells as well as enhanced IC retention. J. Immunol. https://doi.org/10.4049/jimmunol.1200934 (2012).

Momin, N. et al. Maximizing response to intratumoral immunotherapy in mice by tuning local retention. Nat. Commun. 13, 109 (2022).

Article Google Scholar

Momin, N. et al. Anchoring of intratumorally administered cytokines to collagen safely potentiates systemic cancer immunotherapy. Sci. Transl Med. 11, eaaw2614 (2019).

Article Google Scholar

Ishihara, J. et al. Matrix-binding checkpoint immunotherapies enhance antitumor efficacy and reduce adverse events. Sci. Transl Med. 9, eaan0401 (2017).

Article Google Scholar

Agarwal, Y. et al. Intratumourally injected alum-tethered cytokines elicit potent and safer local and systemic anticancer immunity. Nat. Biomed. Eng. 6, 129–143 (2022).

Article Google Scholar

Huang, Z. N., Callmann, C. E., Cole, L. E., Wang, S. & Mirkin, C. A. Synergistic immunostimulation through the dual activation of toll-like receptor 3/9 with spherical nucleic acids. ACS Nano 15, 13329–13338 (2021).

Article Google Scholar

O’Day, S. et al. 423 Safety and preliminary efficacy of intratumoral cavrotolimod (AST-008), a spherical nucleic acid TLR9 agonist, in combination with pembrolizumab in patients with advanced solid tumors. Regul. Young Invest. Award. Abstr. https://doi.org/10.1136/jitc-2020-sitc2020.0423 (2020).

Exicure provides interim results from ongoing phase 1b/2 clinical trial of cavrotolimod (AST-008). sec.report https://sec.report/Document/0001698530-21-000079/exhibit992pressrelease0805.htm (2021).

Wang, C., Fiering, S. N. & Steinmetz, N. F. Cowpea mosaic virus promotes anti‐tumor activity and immune memory in a mouse ovarian tumor model. Adv. Ther. 2, 1900003 (2019).

Article Google Scholar

Lizotte, P. H. et al. In situ vaccination with cowpea mosaic virus nanoparticles suppresses metastatic cancer. Nat. Nanotechnol. 11, 295–303 (2016).

Article Google Scholar

Nguyen, K. G. et al. Localized interleukin-12 for cancer immunotherapy. Front. Immunol. 11, 575597 (2020).

Article Google Scholar

Zaharoff, D. A., Hance, K. W., Rogers, C. J., Schlom, J. & Greiner, J. W. Intratumoral immunotherapy of established solid tumors with chitosan/IL-12. J. Immunother. 33, 697–705 (2010).

Article Google Scholar

Mills, B. N. et al. Stereotactic body radiation and interleukin-12 combination therapy eradicates pancreatic tumors by repolarizing the immune microenvironment. Cell Rep. 29, 406–421 (2019).

Article Google Scholar

Park, C. G. et al. Extended release of perioperative immunotherapy prevents tumor recurrence and eliminates metastases. Sci. Transl Med. 10, eaar1916 (2018).

Article Google Scholar

Chen, Q. et al. In situ sprayed bioresponsive immunotherapeutic gel for post-surgical cancer treatment. Nat. Nanotechnol. 14, 89–97 (2018).

Article Google Scholar

Stephan, S. B. et al. Biopolymer implants enhance the efficacy of adoptive T cell therapy. Nat. Biotechnol. 33, 97–101 (2014).

Article Google Scholar

Hu, Q. et al. Inhibition of post-surgery tumour recurrence via a hydrogel releasing CAR-T cells and anti-PDL1-conjugated platelets. Nat. Biomed. Eng. 5, 1038–1047 (2021).

Article Google Scholar

Coon, M. E., Stephan, S. B., Gupta, V., Kealey, C. P. & Stephan, M. T. Nitinol thin films functionalized with CAR-T cells for the treatment of solid tumours. Nat. Biomed. Eng. 4, 195–206 (2020).

Article Google Scholar

Shae, D. et al. Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy. Nat. Nanotechnol. 14, 269–278 (2019).

Article Google Scholar

Liu, Y. et al. Intrapleural nano-immunotherapy promotes innate and adaptive immune responses to enhance anti-PD-L1 therapy for malignant pleural effusion. Nat. Nanotechnol. 17, 206–216 (2022).

Article Google Scholar

Hewitt, S. L. et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36γ, and OX40L mRNAs. Sci. Transl Med. 11, eaat9143 (2019).

Article Google Scholar

Hotz, C. et al. Local delivery of mRNA-encoding cytokines promotes antitumor immunity and tumor eradication across multiple preclinical tumor models. Sci. Transl Med. 13, eabc7804 (2021).

Article Google Scholar

Tzeng, S. Y. et al. In situ genetic engineering of tumors for long-lasting and systemic immunotherapy. Proc. Natl Acad. Sci. USA 117, 4043–4052 (2020).

Article Google Scholar

Patel, M. et al. 539 Phase 1 study of mRNA-2752, a lipid nanoparticle encapsulating mRNAs encoding human OX40L/IL-23/IL-36γ, for intratumoral (ITu) injection +/− durvalumab in advanced solid tumors and lymphoma. J Immunother. Cancer 9, A569 (2021).

Article Google Scholar

Myerson, J. W. et al. Supramolecular arrangement of protein in nanoparticle structures predicts nanoparticle tropism for neutrophils in acute lung inflammation. Nat. Nanotechnol. 17, 86–97 (2022).

Article Google Scholar

Robertson, J. D., Ward, J. R., Avila-Olias, M., Battaglia, G. & Renshaw, S. A. Targeting neutrophilic inflammation using polymersome-mediated cellular delivery. J. Immunol. 198, 3596–3604 (2017).

Article Google Scholar

Sofias, A. M. et al. Cyclic arginine–glycine–aspartate‐decorated lipid nanoparticle targeting toward inflammatory lesions involves hitchhiking with phagocytes. Adv. Sci. 8, 2100370 (2021).

Article Google Scholar

Braza, M. S. et al. Inhibiting inflammation with myeloid cell-specific nanobiologics promotes organ transplant acceptance. Immunity 49, 819–828 (2018).

Article Google Scholar

Park, J. et al. Intravascular innate immune cells reprogrammed via intravenous nanoparticles to promote functional recovery after spinal cord injury. Proc. Natl Acad. Sci. USA 116, 14947–14954 (2019).

Article Google Scholar

Li, Y. et al. Immunosuppressive PLGA TGF-β1 microparticles induce polyclonal and antigen-specific regulatory T cells for local immunomodulation of allogeneic islet transplants. Front. Immunol. 12, 653088 (2021).

Article Google Scholar

Fisher, J. D. et al. Treg-inducing microparticles promote donor-specific tolerance in experimental vascularized composite allotransplantation. Proc. Natl Acad. Sci. USA 116, 25784–25789 (2019).

Article Google Scholar

Hautz, T. et al. Lymphoid neogenesis in skin of human hand, nonhuman primate, and rat vascularized composite allografts. Transpl. Int. 27, 966–976 (2014).

Article Google Scholar

Azzi, J. et al. Targeted delivery of immunomodulators to lymph nodes. Cell Rep. 15, 1202–1213 (2016).

Article Google Scholar

Bahmani, B. et al. Targeted delivery of immune therapeutics to lymph nodes prolongs cardiac allograft survival. J. Clin. Invest. 128, 4770–4786 (2018).

Article Google Scholar

Li, Z. et al. In vivo labeling reveals continuous trafficking of TCF-1+ T cells between tumor and lymphoid tissue. J. Exp. Med. 219, e20210749 (2022).

Article Google Scholar

Torcellan, T. et al. In vivo photolabeling of tumor-infiltrating cells reveals highly regulated egress of T-cell subsets from tumors. Proc. Natl Acad. Sci. USA 114, 5677–5682 (2017).

Article Google Scholar

Gearty, S. V. et al. An autoimmune stem-like CD8 T cell population drives type 1 diabetes. Nature 602, 156–161 (2022).

Article Google Scholar

Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

Article Google Scholar

Connolly, K. A. et al. A reservoir of stem-like CD8+ T cells in the tumor-draining lymph node preserves the ongoing anti-tumor immune response. Sci. Immunol. 6, eabg7836 (2021).

Article Google Scholar

Schenkel, J. M. et al. Conventional type I dendric cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity 54, 2338–2353 (2021).

Article Google Scholar

Siddiqui, I. et al. Intratumoral Tcf1+ PD-1+ D8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211 (2018).

Article Google Scholar

Tahtinen, S. et al. IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines. Nat. Immunol. 23, 532–542 (2022).

Article Google Scholar

Alameh, M.-G. et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity 54, 2877–2892 (2021).

Article Google Scholar

Li, C. et al. Mechanisms of innate and adaptive immunity to the Pfizer-BioNTech BNT162b2 vaccine. Nat. Immunol. 23, 543–555 (2022).

Article Google Scholar

Yang, X. et al. Lactate-modulated immunosuppression of myeloid-derived suppressor cells contributes to the radioresistance of pancreatic cancer. Cancer Immunol. Res. 8, 1440–1451 (2020).

Article Google Scholar

Certo, M., Tsai, C.-H., Pucino, V., Ho, P.-C. & Mauro, C. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat. Rev. Immunol. 21, 151–161 (2021).

Article Google Scholar

Sangsuwan, R. et al. Lactate exposure promotes immunosuppressive phenotypes in innate immune cells. Cell Mol. Bioeng. 13, 541–557 (2020).

Article MathSciNet Google Scholar

Allen, R. P., Bolandparvaz, A., Ma, J. A., Manickam, V. A. & Lewis, J. S. Latent, immunosuppressive nature of poly(lactic-co-glycolic acid) microparticles. ACS Biomater. Sci. Eng. 4, 900–918 (2018).

Article Google Scholar

Shimabukuro, T. T., Cole, M. & Su, J. R. Reports of anaphylaxis after receipt of mRNA COVID-19 vaccines in the US—December 14, 2020-January 18, 2021. JAMA 325, 1101–1102 (2021).

Article Google Scholar

Szebeni, J. et al. Applying lessons learned from nanomedicines to understand rare hypersensitivity reactions to mRNA-based SARS-CoV-2 vaccines. Nat. Nanotechnol. 17, 337–346 (2022).

Article Google Scholar

McKinlay, C. J. et al. Charge-altering releasable transporters (CARTs) for the delivery and release of mRNA in living animals. Proc. Natl Acad. Sci. USA. 114, E448–E456 (2017).

Article Google Scholar

Kowalski, P. S., Rudra, A., Miao, L. & Anderson, D. G. Delivering the messenger: advances in technologies for therapeutic mRNA delivery. Mol. Ther. 27, 710–728 (2019).

Article Google Scholar

Kong, N. et al. Synthetic mRNA nanoparticle-mediated restoration of p53 tumor suppressor sensitizes p53-deficient cancers to mTOR inhibition. Sci. Transl Med. 11, eaaw1565 (2019).

Article Google Scholar

Leuschner, F. et al. Therapeutic siRNA silencing in inflammatory monocytes in mice. Nat. Biotechnol. 29, 1005–1010 (2011).

Article Google Scholar

Cheng, Q. et al. Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing. Nat. Nanotechnol. 15, 313–320 (2020).

Article Google Scholar

Hewitt, S. L. et al. Intratumoral IL12 mRNA therapy promotes TH1 transformation of the tumor microenvironment. Clin. Cancer Res. 26, 6284–6298 (2020).

Article Google Scholar

Youngblood, B., Hale, J. S. & Ahmed, R. T-cell memory differentiation: insights from transcriptional signatures and epigenetics. Immunology 139, 277–284 (2013).

Article Google Scholar

Gounari, F. & Khazaie, K. TCF-1: a maverick in T cell development and function. Nat. Immunol. 23, 671–678 (2022).

Article Google Scholar

Zhao, X., Shan, Q. & Xue, H.-H. TCF1 in T cell immunity: a broadened frontier. Nat. Rev. Immunol. 22, 147–157 (2022).

Article Google Scholar

Download references

This work was supported in part by the Marble Center for Nanomedicine, the Ragon Institute of MGH, MIT and Harvard, the NIH (awards CA247632, EB031082, U01-CA265706, AI147845, AI162307, AI161297 and CA235375 to D.J.I.) and the Mark Foundation for Cancer Research. This material is based upon work supported in part by the US Army Research Office through the Institute for Soldier Nanotechnologies at MIT, under Cooperative Agreement Number W911NF-18-2-0048. D.J.I. is an investigator of the Howard Hughes Medical Institute.

Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA

Parisa Yousefpour, Kaiyuan Ni & Darrell J. Irvine

Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA

Darrell J. Irvine

Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA

Darrell J. Irvine

Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA

Darrell J. Irvine

Howard Hughes Medical Institute, Chevy Chase, MD, USA

Darrell J. Irvine

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

The manuscript was drafted and revised by P.Y., K.N. and D.J.I.

Correspondence to Darrell J. Irvine.

D.J.I. is an inventor on patents related to albumin hitchhiking (discussed under ‘Principles of lymph node targeting’), nanoparticle modification of T cells (discussed under ‘Backpacking’ cells’) and alum-binding cytokines (discussed under ‘Intratumoral delivery of biomaterials for immune cell targeting’). These patents have been licensed to Elicio Therapeutics, Repertoire Immune Medicines and Ankyra Therapeutics, respectively, and D.J.I. holds equity in these companies. The remaining authors declare no competing interests.

Nature Reviews Bioengineering thanks the anonymous reviewers for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and Permissions

Yousefpour, P., Ni, K. & Irvine, D.J. Targeted modulation of immune cells and tissues using engineered biomaterials. Nat Rev Bioeng 1, 107–124 (2023). https://doi.org/10.1038/s44222-022-00016-2

Download citation

Accepted: 28 November 2022

Published: 30 January 2023

Issue Date: February 2023

DOI: https://doi.org/10.1038/s44222-022-00016-2

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Cellular & Molecular Immunology (2023)

SHARE