Fact-checked by Grok 2 weeks ago

3D bioprinting

3D bioprinting is an additive that precisely deposits bioinks—viscous formulations containing living cells, components, factors, and biomaterials—in a layer-by-layer manner to engineer functional biological structures mimicking native tissues. This adapts principles from conventional , originating in the biomedical field with early cell-encapsulation demonstrations in hydrogels as far back as and formalized bioprinting of liver tissue models by , evolving through iterative advancements in and . Key bioprinting modalities include extrusion-based systems for high-viscosity bioinks yielding macro-scale constructs, inkjet methods for droplet-based with viable ejection, and laser-induced forwarding for non-contact deposition minimizing on cells, though each contends with trade-offs in structural , throughput, and post-print maturation. Notable achievements the of perfusable vascular , stratified equivalents for repair, and cartilage scaffolds exhibiting akin to native , alongside organoid models for high-throughput screening that reduce reliance on . These developments hold causal promise for addressing organ shortages via patient-specific grafts, grounded in empirical validations of and within printed matrices. Persistent challenges encompass inadequate neovascularization beyond millimeter-scale volumes, heterogeneous nutrient leading to central in thicker constructs, and regulatory hurdles for standardization and long-term implant safety, limiting translation from proofs to clinical deployment despite accelerating in multi-material and sacrificial templating strategies. Empirical from recent studies underscore that while viability often exceeds 80% post-, functional integration—such as electrically responsive myocardium or hormone-secreting endocrine tissues—remains empirically constrained by microenvironmental cues and degradation kinetics.

Definition and Principles

Core Concepts and Mechanisms

3D bioprinting represents an additive paradigm adapted for biological applications, the precise, layer-by-layer deposition of cellularized biomaterials to engineer tissue constructs that mimic native and cellular . This approach departs from traditional by providing spatiotemporal over multiple cell types, biochemical gradients, and structural heterogeneity, potentially addressing limitations in manual fabrication such as uniformity and . At its , the integrates living cells within supportive matrices to form viable, functional structures, with core viability hinging on maintaining cellular integrity during mechanical stresses of printing, typically achieving post-print survival rates above 85% in optimized systems. Central to the is the , a shear-thinning composite encapsulating cells at densities of million per milliliter, which must extrudability—facilitated by non-Newtonian under —for deposition with rapid upon from the , often quantified by metrics like circularity exceeding 0.8. Post-deposition stabilization occurs via crosslinking , such as photoinitiated or ionic gelation, which solidify layers within seconds to minutes, preventing collapse under gravitational or forces while permitting nutrient and . These processes emulate embryonic by enabling programmed assembly, where sequential layering builds vascularized or anisotropic constructs, though resolution is constrained to 20–400 micrometers depending on diameter and rheology. Causal drivers of success include rheological optimization to minimize shear-induced apoptosis, with bioinks exhibiting storage moduli of 100–1000 Pa in oscillatory tests to support self-supporting structures, and incorporation of growth factors like VEGF at nanogram scales to induce angiogenesis post-printing. Empirical validation from scaffold-free spheroid fusion models demonstrates that printed aggregates coalesce via cadherin-mediated adhesion within 24–72 hours, forming contiguous tissues with mechanical properties approaching 1–10 kPa modulus akin to soft organs. However, persistent challenges arise from diffusive limitations in avascular cores exceeding 200 micrometers, underscoring the need for hybrid mechanisms integrating sacrificial inks for perfusable channels.

First-Principles Foundations

3D bioprinting derives from the biological reality that functional tissues consist of cells organized in hierarchical structures within an extracellular matrix (ECM), which supplies mechanical support, biochemical gradients, and cues for adhesion, migration, proliferation, and differentiation. This matrix, composed of proteins like collagen and glycosaminoglycans, imposes topological and stiffness gradients that cells sense via integrins and mechanotransduction pathways, influencing phenotype and tissue homeostasis. Conventional 2D cultures or random 3D seeding fail to replicate these anisotropies, resulting in dedifferentiation or necrosis beyond diffusion-limited depths, underscoring the need for precise spatial engineering. The foundation adapts additive to biological constraints, depositing bioinks—viscoelastic mixtures of cells, polymers, and factors—layer by layer under computer control to mimic native architectures. Bioinks must balance printability with cytocompatibility: shear-thinning enables through micron-scale nozzles without clogging, while post-extrusion gelation via ionic (e.g., calcium crosslinking of alginate), , or UV-induced preserves against and . Cell densities typically from 1-10 million per milliliter to viability while avoiding agglomeration that disrupts . A core physical limitation arises from mass transport: oxygen and nutrients diffuse only 100-200 micrometers in acellular hydrogels before hypoxic gradients cause central cell death, per Fick's laws and empirical measurements in spheroids. Bioprinting circumvents this by enabling vascular channel integration during fabrication, though fusion of printed layers demands interfacial wetting and dehydration to eliminate voids, with mechanical integrity hinging on matched viscosities (often 10^2-10^6 Pa·s) and printing speeds (1-100 mm/s). During extrusion, the predominant modality, pneumatic or piston-driven pressure generates Poiseuille flow, imposing shear (τ ≈ μ · γ, where μ is and γ ) and extensional stresses on cells; viability drops above 10^3-10^5 s⁻¹ rates for sensitive types like stem cells, necessitating rheological optimization to limit τ below 5-50 . Hydrostatic (up to 10^5 Pa in long nozzles) add compressive forces, but causal damage primarily stems from rupture or cytoskeletal disruption, mitigated by larger diameters (200-500 μm) at the cost of . These enforce trade-offs: higher demands finer nozzles, amplifying stresses and reducing throughput, while empirical thresholds bioink formulation for scalable, viable constructs.

Historical Development

Origins in Additive Manufacturing

Additive manufacturing (AM), the foundational technology underlying 3D bioprinting, emerged in the early as a layer-by-layer fabrication contrasting traditional subtractive methods, precise of complex geometries from digital designs using materials like photopolymers and thermoplastics. The core principle of controlled material deposition—whether via , jetting, or photopolymerization—directly translates to bioprinting, where or filaments are replaced by biocompatible hydrogels laden with living cells to mimic extracellular matrices. This leverages AM's to achieve micrometer-scale , for replicating architectures, though biological constraints like cell required modifications to designs and printing parameters. Key early AM innovations established the mechanical and optical frameworks later adapted for bioprinting. In 1981, Hideo Kodama proposed the first layer-by-layer photopolymerization technique, using ultraviolet light to solidify liquid resin into solid prototypes, laying groundwork for light-mediated bioink crosslinking. Hull advanced this in with (), employing a UV to selectively photopolymer layers in a vat, a process patented in 1986 that founded 3D Systems and influenced laser-assisted bioprinting for high-resolution cell patterning. By 1989, S. Scott Crump patented fused deposition modeling (FDM), which extrudes heated thermoplastic filaments through a nozzle to build parts, providing the basis for extrusion bioprinting systems capable of handling shear-thinning bioinks at physiological temperatures. Inkjet-based AM techniques further bridged to biological applications by enabling non-contact droplet ejection. Early inkjet methods, rooted in printing but adapted for AM in the —such as Emanuel Sachs' binder jetting at , which deposits binder droplets onto powder beds—facilitated precise, low-force material placement suitable for fragile cellular payloads. The first explicit connection to biology occurred in , when Klebe modified a inkjet printer to deposit living cells onto substrates in a process called cytoscribing, demonstrating AM's potential for patterning viable mammalian cells without scaffolds. These origins highlight how AM's emphasis on automated, scalable deposition overcame limitations of manual tissue assembly, though initial efforts faced viability issues due to droplet shear and lack of optimized bioinks.

Early Bioprinting Experiments

In 1988, J. Klebe at the of Center introduced cytoscribing, an early for micropositioning cells using a modified inkjet printer to deposit proteins and living cells onto a substratum in precise patterns. This enabled the of two-dimensional cell arrays and rudimentary three-dimensional synthetic tissues by leveraging the printer's ability to eject droplets containing extracellular matrix components like fibronectin, followed by cell seeding, demonstrating cell viability and adhesion post-deposition without significant damage from the printing process. Klebe's experiments highlighted the feasibility of automated cell patterning but were limited to surface-bound structures and did not involve layer-by-layer buildup of hydrogels with embedded cells, marking it as a foundational precursor rather than full 3D bioprinting. Advancing toward true three-dimensional constructs, in 2003, Thomas Boland and W. Cris at developed the first inkjet-based bioprinter by retrofitting a Hewlett-Packard with reservoirs containing mammalian cells suspended in biocompatible hydrogels, such as alginate cross-linked with . Their experiments demonstrated the printing of viable cells and other types in layered patterns, achieving cell densities comparable to native tissues and showing subsequent and within the printed gels over several days, with viability rates exceeding 80-90% immediately post-printing. This work established key principles of formulation—balancing suspension viscosity for nozzle ejection, minimization to preserve integrity, and post-print gelation for —paving the way for organ-like architectures, though early constructs remained due to limits (droplet sizes around 50-100 μm) and lack of vascularization. These efforts underscored bioprinting's potential for while revealing challenges like nozzle from cellular aggregates and the need for thermosensitive or photo-crosslinkable materials to enhance , influencing subsequent refinements in the mid-2000s. Boland's innovations, patented shortly thereafter, shifted from mere patterning to functional 3D cellular , distinguishing bioprinting from scaffold-based methods.

Key Milestones and Commercialization

The foundational concept of bioprinting emerged in 1988 when Dr. Robert J. Klebe proposed using a modified inkjet printer to pattern living cells onto a substrate, laying the groundwork for precise cellular deposition. Early experimental systems followed in the early 2000s; by 2004, researchers developed a bioprinting setup employing 12 piezoelectric ejectors to deposit biological materials via droplet ejection, enabling initial demonstrations of printed cell structures. A pivotal advancement occurred in 2007 with the founding of Organovo Holdings, Inc., which licensed bioprinting technology from the to engineer functional human tissues, marking the transition from academic prototypes to structured development efforts. In 2010, Organovo unveiled the first commercial 3D bioprinter designed for producing multilayered human tissue constructs, such as basic blood vessels, making the technology accessible beyond research labs. Progress accelerated in 2013 when Organovo achieved the world's first fully cellular 3D bioprinted liver tissue, capable of performing basic metabolic functions, which demonstrated potential for drug testing models. Subsequent years saw refinements in printing vascular networks and multicellular tissues, though persistent challenges like achieving sufficient vascularization for larger constructs limited . Commercialization has focused on research tools and select clinical applications rather than whole organs. Companies such as CELLINK (now part of BICO Group) and have commercialized bioprinters, bioinks, and tissue models for pharmaceutical screening and R&D, with the global for bioprinting exceeding research prototypes by the mid-2010s. A landmark regulatory milestone came in June 2025, when secured FDA approval for a bioprinted for peripheral repair, the first such clearance for a bioprinted therapeutic device, enabling clinical use in damage treatment. Despite these strides, full commercialization of transplantable organs remains preclinical, constrained by issues including immune rejection, long-term functionality, and regulatory hurdles for complex biologics.

Bioprinting Process

Pre-Bioprinting Preparation

Pre-bioprinting preparation constitutes the foundational stage of 3D bioprinting, encompassing the digital modeling of architectures and the of bioinks that integrate viable s with supportive biomaterials. This phase ensures construct fidelity, cell viability, and compatibility with downstream parameters, drawing from and principles to replicate native geometries and microenvironments. Digital scaffold design begins with acquiring high-resolution 3D structural data via medical imaging modalities such as magnetic resonance imaging (MRI) or computed tomography (CT) scans, which provide anatomical blueprints for patient-specific constructs. These datasets are processed using computer-aided design (CAD) and computer-aided manufacturing (CAM) software to generate printable models, often sliced into sequential 2D layers with thicknesses ranging from 20–100 μm for laser-based systems to 100–500 μm for extrusion or inkjet methods. The resulting stereolithography (STL) or other bioprinter-compatible files dictate pore sizes, filament orientations, and overall porosity to optimize nutrient diffusion and mechanical integrity, with resolutions limited by imaging quality and software algorithms. Bioink formulation integrates cells—typically sourced from stem cells, primary isolates, or derivatives—with extracellular matrix-mimicking hydrogels to form printable suspensions. Cells are cultured for expansion, harvested via enzymatic dissociation, and suspended at densities such as 1.5 × 10⁶ cells/mL to promote uniform distribution and post-print viability exceeding 90% in compatible matrices. Biomaterials, including natural polymers like alginate (2–6% w/v, cross-linked ionically with Ca²⁺) or methacryloyl (GelMA, ~10% for UV cross-linking), are selected for shear-thinning ( ~30 mPa·s), , and gelation kinetics that minimize during . Hybrid formulations, blending synthetic elements like (PEG) with natural components, address trade-offs in printability and , though challenges persist in mitigating viability losses from high-pressure dispensing (up to 40% reduction) and ensuring long-term structural fidelity. Preparatory considerations extend to sterilization of components under aseptic conditions and via additives to and cell encapsulation , with empirical testing validating parameters like nozzle shear rates against tissue-specific demands such as vascular or load-bearing . This iterative optimization, informed by material-cell interactions, underpins causal between pre-print variables and emergent tissue functionality, prioritizing empirical metrics over idealized assumptions.

Core Printing Techniques

Extrusion-based bioprinting is one of the most widely used core techniques, involving the continuous extrusion of viscoelastic bioinks through a computer-controlled via pneumatic, , or screw-driven , depositing in a layer-by-layer to build three-dimensional structures. This supports high densities approaching physiological levels and accommodates a broad range of bioink viscosities, enabling the creation of complex, multi- constructs with tunable biodegradability. Typical resolutions range from 100 to 200 μm, though lower precision compared to other can limit intricate feature fabrication, and shear stress from extrusion may reduce viability to 85-95%. Inkjet or droplet-based bioprinting operates by ejecting micro-scale droplets (1-100 picoliters) of bioink onto a substrate using thermal, piezoelectric, or electrostatic actuation, allowing for precise, non-contact deposition suitable for low-viscosity hydrogels like alginate or collagen. It achieves resolutions below 100 μm and high-speed printing, with cell viability often exceeding 90% due to minimal mechanical stress, making it ideal for vascular or tissue patterning applications. However, requirements for low viscosity restrict bioink options, potentially limiting structural integrity and cell density in thicker constructs. Laser-assisted bioprinting employs a to vaporize a thin layer on a donor , propelling cell-laden droplets toward a receiving surface in a nozzle-free, non-contact process that minimizes shear forces and supports high cell densities. Resolutions can reach 10-50 μm, with viability rates of 90-95%, enabling precise patterning of sensitive cells like stem cells. Drawbacks include high equipment costs, slower processing times, and challenges in scaling to large constructs due to laser scanning limitations. These techniques form the foundation of the bioprinting process, often selected based on bioink properties, required resolution, and target tissue complexity, with extrusion dominating due to versatility despite trade-offs in precision. Variations like coaxial extrusion for core-shell structures or hybrid systems combining methods are emerging to address limitations in vascularization and mechanical fidelity.

Post-Processing and Tissue Maturation

Following the deposition of cellular bioinks, post-processing stabilizes the nascent construct through crosslinking mechanisms tailored to the bioink composition. Photocrosslinking via (UV) light exposure, often applied in stereolithography-based systems, polymerizes acrylic or methacrylated hydrogels like methacryloyl (GelMA), enhancing mechanical integrity while preserving cell viability above 80% in many protocols. Chemical crosslinking with agents such as or calcium ions solidifies alginate-based structures, whereas thermal or ionic gelation provides milder alternatives for temperature-sensitive cells. Support baths, composed of yield-stress materials like granular hydrogels or Pluronic-nanoclay composites, prevent structural collapse during printing and are subsequently removed via dissolution, enabling complex geometries. Sacrificial bioprinting introduces transient templates—such as or lattices—to form perfusable microchannels, which are dissolved post-printing and lined with endothelial cells to foster vascular . These steps address immediate challenges like poor and cell , with viability post-crosslinking typically exceeding 85% in optimized processes. Tissue maturation ensues in controlled environments to recapitulate native , involving in nutrient-rich supplemented with factors like transforming growth factor-beta (TGF-β) to drive , , and (ECM) remodeling. Static yields initial cell-ECM integration, but dynamic bioreactors applying mimic hemodynamic forces, improving oxygen and reducing in constructs thicker than 200–500 μm; for instance, mesenchymal stem cell-seeded norbornene-modified (NorHA) hydrogels exhibit compressive moduli increases from 50 kPa to over 500 kPa after 6 weeks of , approximating cartilage mechanics. Electrical or mechanical stimuli, such as cyclic strain at 5–10% elongation, further promote alignment and functionality in muscle or cardiac tissues. Maturation timelines vary by tissue type—days for simple epithelial layers to months for stratified organs—with endpoints assessed via metrics like metabolic activity (e.g., MTT assays), gene expression for maturation markers, and functional assays such as contractility in bioprinted myocardium. Persistent hurdles include incomplete vascularization beyond millimeter scales and heterogeneous maturation due to diffusion gradients, necessitating hybrid approaches like co-culture with angiogenic factors. Peer-reviewed studies emphasize empirical validation over theoretical models, revealing that bioreactor-conditioned constructs outperform static ones in viability (up to 95% vs. 70%) and tissue-specific outputs.

Bioinks and Biomaterials

Classification of Bioinks

Bioinks are classified primarily by their compositional into , synthetic, and categories, as this determines attributes like biocompatibility, , and suitability for specific modalities. bioinks, derived from biological sources, excel in mimicking extracellular matrix () environments to promote , proliferation, and differentiation, though they often suffer from inconsistent strength and batch variability. Synthetic bioinks, engineered from non-biological polymers, provide tunable rheological and for high-fidelity but typically lack inherent bioactivity, necessitating additives for . bioinks integrate and synthetic components to these trade-offs, enhancing overall printability and functionality for constructs. Additional classification criteria include gelation mechanisms—such as ionic (e.g., alginate with Ca²⁺), (e.g., ), photochemical (e.g., GelMA under UV), or enzymatic (e.g., via )—which affect post-printing stability and viability, often exceeding 80-90% in optimized formulations. Bioinks may also be categorized by incorporation: cell-laden (with suspended live cells like mesenchymal stem cells at densities of 1-10 × 10⁶ cells/mL) versus acellular scaffolds for later seeding, or by form as hydrogels, microgels, or scaffold-free aggregates like spheroids, where viability reaches ≥95% due to self-supporting structures. Decellularized ECM (dECM) bioinks represent a specialized natural subclass, processed from tissues like porcine heart or to retain organ-specific biochemical cues while removing risks.
  • Natural Bioinks: Comprise proteins (e.g., collagen type I, providing RGD motifs for adhesion and >80% cell viability in skin constructs), polysaccharides (e.g., alginate for shear-thinning gelation at 2-5% w/v, hyaluronic acid for chondrogenesis support with >90% viability), and others like fibrin or silk fibroin, which degrade enzymatically to aid remodeling but yield low stiffness (0.1-10 kPa).
  • Synthetic Bioinks: Include polyethylene glycol (PEG) derivatives like PEGDA for photocrosslinking and moduli up to 100 kPa, polycaprolactone (PCL) for thermoplastic extrusion with melting points around 60°C, and Pluronic for temperature-responsive behavior; these ensure shape fidelity but require functionalization (e.g., RGD grafting) to achieve >85% viability over weeks.
  • Hybrid Bioinks: Blend materials like alginate-GelMA or collagen-PEG to yield composites with enhanced viscosity (10-100 Pa·s) and bioactivity, as in chitosan-PCL for vascular applications or nanomaterial-doped variants (e.g., hydroxyapatite in gelatin for osteogenesis).
CategoryExamplesAdvantagesLimitations
NaturalAlginate, , , dECMHigh ; native Weak ; sourcing variability
Synthetic, PCL, PluronicPrecise tunability; high Poor bioactivity; potential
HybridGelMA-alginate, Silk-dECMBalanced ; improved vascularization formulation; optimization challenges
This compositional framework guides selection, with natural and hybrid types dominating applications in soft tissues due to superior cellular outcomes, while synthetics suit supportive roles in load-bearing constructs.

Physicochemical Properties and Selection

Bioinks require tailored physicochemical properties to balance printability, structural fidelity, and biocompatibility during 3D bioprinting. Rheological attributes, such as viscosity and shear-thinning behavior, govern flow through nozzles under applied pressure, while mechanical properties like storage modulus (G') and yield stress ensure post-extrusion shape retention against gravitational and surface tension forces. Chemical crosslinking mechanisms enable rapid gelation to stabilize constructs, and overall selection prioritizes properties matching the target printing modality and tissue mechanics. Rheological are paramount for extrusion-based systems, where bioinks must exhibit pseudoplastic (shear-thinning) characterized by a power-law n < 1, allowing viscosity to decrease under high shear rates (up to 2000 s⁻¹ during printing) for smooth deposition, followed by rapid recovery to a solid-like state. Viscosity typically ranges from 10³ to 10⁷ mPa·s for pneumatic or piston-driven extrusion, with higher values (up to 10⁴ Pa·s) feasible in screw extruders; inadequate ranges lead to filament breakage or spreading. Yield stress, often exceeding 72 Pa (e.g., 166–227 Pa in alginate-poloxamer blends), prevents slumping by providing resistance to deformation below a critical shear threshold. Mechanical properties post-crosslinking, including viscoelasticity where G' surpasses loss modulus G'' for elastic dominance, must approximate native tissue stiffness—soft hydrogels (e.g., <10 kPa for neural tissues) versus stiffer matrices (>100 kPa for ). Swelling ratios and degradation rates influence long-term stability, with controlled enzymatic or hydrolytic breakdown aligning to tissue remodeling timelines. , modulated by (decreasing with higher concentrations due to interfacial adsorption), affects droplet formation in inkjet methods but is secondary in . Selection of bioinks hinges on quantitative printability assessments, such as filament circularity (ideal P_r ≈ 1 for uniform pores) and critical stacking height, alongside biological compatibility ensuring >85–95% cell viability post-printing. For extrusion, shear-thinning hydrogels with high yield stress (e.g., gellan gum additives) are favored over low-viscosity inks suited to lithography (0.25–10 Pa·s, photo-crosslinkable). Natural bioinks like alginate offer tunable viscosity and ionic crosslinking (e.g., Ca²⁺ gelation), but hybrids address weaknesses in mechanical strength; criteria also encompass low immunogenicity and biodegradability matched to repair processes, evaluated via rheometry and empirical trials.

Recent Innovations in Bioink Formulations

Recent developments in bioink formulations emphasize materials that shear-thinning for printability with biomechanical and bioactivity to encapsulation and post-printing maturation. Innovations incorporate decellularized extracellular matrices (dECM), nanoparticles, and stimuli-responsive polymers to mimic native microenvironments, addressing limitations in traditional hydrogels like alginate or methacryloyl (GelMA) that often compromise or viability under extrusion stresses. A notable advancement is the gellan gum/dECM bioink (GG/dECMb), formulated by combining with urea-extracted decellularized cartilage ECM and dual crosslinking via Ca²⁺ ions and , which exhibits shear-thinning behavior, high printability (area fidelity near 100%), and viscoelastic dominance (G' > G''). This achieves 97.41% cell viability after 5 days with chondrocytes, promotes proliferation and chondrogenic (evidenced by Alcian staining at 21 days), and supports cartilage scaffolds with enhanced swelling (974% at 24 hours) and controlled degradation (~95% over 14 days). For applications in cellular agriculture, κ-CAM bioinks—comprising κ-carrageenan, alginic acid, and methylcellulose in Leibovitz-15 medium—enable extrusion printing of sea bass cell-laden structures at 20–30°C with viabilities of 87–95% over 8 days, while mFAT inks integrate microalgae (e.g., Nannochloropsis oceanica), plant oils, soy protein, and κ-carrageenan to yield fat mimics with up to 97.44% viability and improved sensory profiles for hybrid seafood products. Nanoengineered granular hydrogels, incorporating nanoparticles into microporous matrices, enhance strength and reduce during , preserving attachment and architectures, as demonstrated in formulations from 2022 onward. Biphasic colloidal hydrogels, featuring particles in phases, offer tunable for improved in 2024 constructs. photocurable systems, such as / blends, with for vascularized tissues.

Bioprinter Technologies

Extrusion-Based Bioprinters

Extrusion-based bioprinters deposit through a under controlled to form continuous filaments, building constructs layer by layer in a precise spatial . The process relies on shear-thinning of the , which temporarily reduces during to enable , followed by rapid to maintain structural post-deposition. Common actuation mechanisms include pneumatic systems using air , mechanical piston or screw-driven extruders for precise control, and hybrid variants combining these for multi-material printing. This technique supports high cell densities exceeding 10^6 cells/mL, accommodating spheroids and heterogeneous cell populations suitable for complex tissue mimics. It excels in fabricating large-scale constructs due to compatibility with viscous hydrogels and the to print in supportive for soft materials, enhancing . Key advantages encompass versatility with biomaterials like alginate, , and synthetic polymers, as well as scalability for clinical translation in . However, limitations include reduced typically ranging from 100 to 400 μm, constrained by and , which hinders fine vascular . Cell viability post-extrusion often exceeds 85-90%, though from high pressures can induce mechanotransduction effects, potentially altering if not mitigated by low-viscosity formulations or optimized geometries. parameters such as (10-100 kPa), speed (1-20 /s), and critically uniformity and viability; for instance, excessive rates above 10^3 s^-1 correlate with in sensitive . Recent advancements incorporate nozzles for core-shell structures to protect and improve , alongside in yield-stress gels to achieve resolutions below 50 μm without scaffolds. Commercial systems from companies like CELLINK and BioBots feature modular extrusion heads for multi-bioink deposition, enabling applications in cartilage and skin models with demonstrated maturation in vitro. Milestones include the 2010s shift to multi-arm robotic arms for freeform printing and 2020s integration of real-time imaging for adaptive deposition, addressing inconsistencies in heterogeneous tissues. Despite hype around whole-organ printing, empirical data emphasize incremental progress in vascularized patches, with clinical trials for extrusion-printed implants reported as of 2024.

Inkjet and Droplet-Based Systems

Inkjet bioprinting, a subset of droplet-based systems, employs modified commercial inkjet printer heads to deposit precise picoliter-scale droplets of bioink containing cells and biomaterials onto a substrate, enabling non-contact layer-by-layer fabrication of tissue constructs. This technique originated in 2003 when Wilson and Boland adapted a standard Hewlett-Packard inkjet printer for biological applications, followed by the first demonstration of cell viability printing in 2004 using mammalian cells. Droplet formation relies on either thermal (bubble-jet) or piezoelectric actuation: thermal methods heat the bioink to generate vapor bubbles that eject droplets, while piezoelectric systems use mechanical deformation of a crystal to pressurize and dispense fluid without heat. These systems achieve resolutions down to 20-50 micrometers, suitable for cellular patterning, due to precise control over droplet volume (typically 10-300 picoliters) and placement via digital rasterization. Cell viability post-printing often exceeds 85-95%, attributed to minimal shear stress compared to extrusion methods, though thermal inkjet variants risk temporary heat-induced damage mitigated by low energy pulses. Bioinks must exhibit low viscosity (3-20 mPa·s) for reliable jetting, limiting structural support and necessitating post-deposition crosslinking, such as via calcium ions for alginate or UV for gelatin methacryloyl. Advantages include high throughput (up to thousands of droplets per second), cost-effectiveness from off-the-shelf components, and with multi-material for heterogeneous tissues, as demonstrated in 2016 studies fabricating vascular-like with endothelial cells. However, drawbacks encompass restricted z-axis to droplet stacking challenges, potential clogging from cellular aggregates, and suboptimal mechanical integrity in thicker constructs without supportive hydrogels. Recent innovations, such as hybrid piezoelectric systems integrated with reported in 2023, enhance shape fidelity for and models by combining droplets with fibrous scaffolds. Applications span regenerative medicine, including 2022 demonstrations of inkjet-printed skin equivalents with dermis and epidermis layers achieving 90% cell survival, and drug screening platforms modeling tumor microenvironments via precise cell gradients. In droplet-based variants beyond inkjet, like solenoid microvalve systems, larger droplets (microliter scale) enable co-printing of multiple cell types for organoids, though with reduced resolution. Ongoing challenges involve scaling to vascularized organs, addressed in 2024 reviews emphasizing stimuli-responsive bioinks for improved post-print perfusion.

Laser and Light-Assisted Methods

Laser-assisted bioprinting, exemplified by laser-induced forward (), utilizes a focused to propel microdroplets of from a transparent donor to a receiving surface, enabling precise, nozzle-free deposition. The process involves a laser beam passing through a quartz slide coated with a thin absorbing layer and bioink film; upon absorption, rapid vaporization generates a high-speed jet that transfers the material without direct contact, achieving resolutions down to 10-20 μm and single-cell accuracy. This method accommodates high-viscosity bioinks up to 300 mPa·s and yields cell viabilities often exceeding 95%, particularly with picosecond or femtosecond pulses that limit thermal damage to localized regions. Light-assisted bioprinting techniques, including () and (), rely on photopolymerization where or visible light crosslinks photocurable layer by layer within a . In , a scanning selectively cures the surface, while DLP employs systems like digital micromirror devices to solidify entire layers simultaneously, enabling faster throughput for structures up to centimeters in scale. These approaches deliver high resolutions as fine as 10 μm, superior surface quality, and compatibility with complex geometries such as vascular channels, though they necessitate low-concentration photoinitiators like Irgacure 2959 to preserve cell viability above 90% during times typically under 1 second per layer. Compared to extrusion-based systems, laser and light-assisted methods minimize shear forces on encapsulated cells, enhancing post-print functionality in applications like neural tissue models and organ-on-chip devices; however, LIFT requires precise pulse energy control (e.g., 0.1-1 μJ) to avoid bubble formation or incomplete transfer, while light methods are constrained to transparent, light-sensitive bioinks prone to oxygen inhibition without inert atmospheres. Hybrid variants, such as laser-assisted projection stereolithography, combine transfer precision with volumetric curing for improved depth penetration up to 500 μm using multi-photon processes. Ongoing refinements, including absorbent-free LIFT variants reported in 2023, aim to reduce substrate contamination and expand to in vivo printing.

Emerging Hybrid and Advanced Systems

Hybrid systems in 3D bioprinting integrate multiple deposition techniques to address limitations of individual methods, such as resolution constraints in or scalability issues in laser-assisted printing, enabling the fabrication of scaffolds with enhanced mechanical strength, biomimicry, and cellular compatibility. For instance, -electrospinning hybrids combine robotic for macro-scale structural deposition with electrospinning for nanoscale fibrous , producing dual-scale scaffolds that promote osteogenesis in bone , as demonstrated in polycaprolactone-based constructs exhibiting improved compressive and cell proliferation compared to single-technique outputs. Similarly, -laser hybrids, such as those employing laser-guided direct writing alongside pneumatic , achieve sub-micrometer for vascular patterns while maintaining bulk deposition, with applications in cartilage regeneration using collagen-polycaprolactone composites that support chondrocyte viability over 28 days. These systems often incorporate additional modalities like UV curing or for surface functionalization, as in zonal -assisted bio, which enhances scaffold bioactivity without compromising print . Embedded bioprinting represents an advanced variant, where bioinks are extruded directly into a viscoelastic support bath—such as granular microgels or yield-stress fluids—allowing for freeform printing of overhangs and complex geometries without external supports, thus facilitating large-scale vascularized constructs like branched vessel networks or biomimetic organs. This approach overcomes in traditional extrusion, yielding resolutions down to 20-50 μm and enabling perfusable channels mimicking native vasculature, with demonstrated efficacy in heart and bone models post-2023. platforms like RegenHU's BioFactory exemplify fully integrated hybrids, merging screw-assisted , inkjet droplet ejection, and crosslinking for multi-material organ-on-chip models, achieving heterogeneous tissue layers with gradients in stiffness and composition. Further advancements incorporate into bioinks for multi-material systems, such as graphene oxide-reinforced hydrogels printed via combined and light-assisted methods, which bolster (up to 10-fold increase in ) and electrical for neural interfaces, while in these setups supports of cellular spheroids into functional mini-tissues. Despite these gains, challenges persist in for human-scale organs and long-term vascular , with ongoing prioritizing of support and automated multi-nozzle .

Design and Fabrication Approaches

Biomimicry and Structural Replication

Biomimicry in 3D bioprinting entails engineering constructs that replicate the , compositional gradients, and biomechanical properties of native tissues to foster functional recapitulation. This draws from biological precedents, such as the extracellular matrix's nanofibrillar and multicellular , to guide bioink and deposition strategies that promote cell-ECM interactions and tissue maturation. Techniques emphasize layer-by-layer fabrication to emulate spatial heterogeneity, including stiffness variations and porosity akin to those in cartilage or bone, where compressive moduli range from 0.1 to 1 MPa in engineered analogs matching native values. Structural replication advances through multi-material printing, enabling precise positioning of cells and matrices to mimic native architectures like aligned collagen fibers in tendons or vascular bifurcations. For example, sacrificial alginate inks have been used to create perfusable channels with diameters of 100-500 μm, replicating capillary networks and sustaining cell viability beyond the diffusion limit of 200 μm in avascular constructs. In cardiac tissue engineering, bioinks derived from decellularized myocardial ECM preserve anisotropic fiber orientations, achieving conduction velocities of 20-30 cm/s comparable to native myocardium when printed with induced pluripotent stem cell-derived cardiomyocytes. Recent developments integrate computational modeling of native blueprints to optimize parameters, such as diameter and speed, for high- replication. A platform using magnetized bioprinting enabled spatially controlled assembly of organoids into assembloids, mimicking embryonic fusion with over 90% alignment accuracy in neural structures. Similarly, high-throughput spheroid bioprinting in fused cellular aggregates into dense s, replicating stromal interfaces with densities exceeding 10^8 cells/, as validated by and metabolic assays. These methods underscore causal between structural and emergent functions, such as induction via VEGF gradients in printed vessels. Challenges persist in scaling biomimetic resolution to microscale features like podocyte arrangements in kidneys, limited by current printer resolutions of 10-50 μm versus native 1-5 μm scales, necessitating hybrid approaches combining printing with self-assembly. Empirical validation through histological comparisons and in vivo implantation confirms that biomimetic designs enhance integration, with printed skin grafts showing 80% re-epithelialization rates in porcine models versus 50% for non-mimetic controls.

Autonomous Self-Assembly Techniques

Autonomous self-assembly techniques in 3D bioprinting replicate embryonic morphogenesis by enabling cells to spontaneously form complex tissues through inherent biological processes, such as cell-cell adhesion, migration, and extracellular matrix (ECM) production, with minimal external structural constraints. This bottom-up strategy contrasts with scaffold-dependent methods by emphasizing cellular autonomy, where printed bioinks—typically high-density cell suspensions or aggregates—rely on biochemical cues like cadherins and integrins to drive organization, often resulting in scaffold-free constructs that better mimic native tissue composition. Such approaches prioritize histogenesis, allowing cells to modulate tissue properties endogenously, though they require precise control of initial deposition to initiate fusion. Key mechanisms involve the deposition of viable cellular units that fuse post-printing: for instance, multicellular spheroids (typically 300–500 μm in diameter) or linear tissue strands are extruded or assembled, leveraging natural fusion kinetics driven by ECM secretion and cytoskeletal remodeling. In a 2009 study, Norotte et al. utilized extrusion-based bioprinting with spheroids composed of human smooth muscle cells and fibroblasts, temporarily stabilized by agarose rods (dissolved post-assembly), to create branched tubular vascular structures up to several centimeters in length; these conduits exhibited patent lumens and ECM deposition akin to native vessels after 21–60 days in culture. Cell viability remained above 90% throughout, highlighting the technique's biocompatibility, though fusion times extended to weeks for larger scales. More recent advancements incorporate robotic-assisted systems for scalable strand . Domínguez-Rubio et al. (2016) fabricated scaffold-free tissue strands from bovine articular chondrocytes by microinjecting approximately 200 million cells into temporary alginate capsules (outer ~1,250 μm), dissolving the capsules to strands that self-assembled via coaxial extrusion. Printed into 3 mm × 3 mm lattices using a Multi-Arm BioPrinter, these strands initiated within 12 hours, achieving near-complete by day 7, with 87 ± 3% cell viability and a of 1,094 ± 26 kPa—comparable to cartilage—alongside sulfated glycosaminoglycan content of 290 μg/ng DNA. In vivo implantation into rabbit osteochondral defects demonstrated partial remodeling but limited host , underscoring potential for cartilage repair while revealing needs for vascularization enhancements. These techniques excel in producing biologically faithful tissues by avoiding synthetic scaffold degradation products, which can elicit immune responses or hinder remodeling, but face limitations in spatiotemporal precision for heterogeneous structures and extended maturation periods (often days to weeks). Integration with factors or co-cultures can accelerate , as seen in endothelial spheroids forming perfusable vascular networks via angiogenic self-organization, yet scalability remains constrained by diffusion-limited delivery in avascular cores exceeding 200–500 μm. Ongoing focuses on bioinks to balance with guided cues, aiming for clinically viable organs.

Mini-Tissue and Modular Assembly

Mini-tissue and modular assembly in 3D bioprinting represents a bottom-up fabrication paradigm, where discrete cellular building blocks—termed mini-tissues, such as spheroids, strands, or microgels—are engineered individually and then combined to construct larger, hierarchically organized tissues. This contrasts with top-down extrusion or layering by enabling modular scalability, precise cellular positioning, and integration of functional elements like vasculature within subunits, thereby mitigating diffusion limitations in avascular constructs exceeding 200–500 μm in thickness. Fabrication of mini-tissues typically employs cell aggregation methods: spheroids form via low-adherence cultures or hanging-drop techniques, achieving diameters of 100–500 μm with high viability (>90%) for organoid-like structures; cylindrical strands are generated through coaxial extrusion bioprinting of cell suspensions in sacrificial hydrogels, yielding perfusable filaments 300–600 μm in diameter; microgels arise from emulsification or microfluidic droplet generation, encapsulating cells in 50–200 μm crosslinked matrices. A 2016 study demonstrated scaffold-free strand production by microinjecting chondrocytes into 130 μm alginate capsules, followed by dissolution to yield self-supporting bioinks printable without molds, resulting in strands that fused into 3 mm patches with 87% viability by day 7 and sGAG content of 290 μg/ng DNA after 21 days. Modular assembly integrates these units via directed techniques including robotic bioprinting for layer-by-layer stacking, with superparamagnetic nanoparticles for non-contact positioning (e.g., aligning modules into tubular vasculatures), acoustic wave manipulation for levitation and fusion, or passive relying on cell-mediated adhesion and shape complementarity. In 2011, sequential assembly of cell-laden microgels produced vascularized networks with endothelial-lined channels, demonstrating fusion times of hours to days and improved over monolithic gels. This approach has yielded functional prototypes, such as 2019 modular cardiac patches from fused strands exhibiting synchronous , and pre-vascularized organoids assembled from endothelialized spheroids supporting blood in murine models. Such strategies enhance biological fidelity by preserving module-specific microenvironments—e.g., zonal gradients in cartilage mini-tissues mimicking native ECM deposition—while allowing customization for applications like cartilage regeneration, where assembled modules achieved 1.5-fold higher mechanical stiffness than uniform prints. Limitations include inter-module interface integrity, with fusion requiring optimized cadherin expression or fibrin glues to prevent delamination under shear, though yields exceed 80% in optimized protocols.02891-8)

Applications

Regenerative Medicine and Organ Engineering

3D bioprinting facilitates regenerative medicine by enabling the precise fabrication of patient-specific tissue constructs, incorporating living cells, biomaterials, and growth factors to mimic native extracellular matrices and promote tissue regeneration. This technology supports the engineering of scaffolds that guide cellular behavior, enhancing repair processes for damaged tissues and potentially alleviating organ donor shortages through personalized implants. Advances since 2020 have emphasized vascular integration and multi-cellular layering to improve construct functionality and integration with host tissues. In tissue regeneration, 3D bioprinted skin equivalents have shown promise for treating burns and chronic wounds, utilizing keratinocytes, fibroblasts, and dermal matrices to form stratified structures that promote re-epithelialization and vascularization in preclinical models. Cartilage constructs, printed with chondrocytes in bioinks, have been developed for repair and auricular , demonstrating mechanical properties akin to native and successful implantation in . scaffolds incorporating osteoblasts and composites support osteogenesis, with reports of enhanced bone regeneration in defect models. Clinical remains , but as of 2025, interventional trials have autologous bioprinted vessels, tracheal segments, and external implants, focusing on and long-term viability. Organ engineering leverages bioprinting to assemble complex architectures, such as vascularized myocardial patches that have repaired damaged heart tissue in rodent models by restoring contractile function post-infarction. Perfusable scaffolds, including collagen-based internally vascularized structures printed in 2025, enable nutrient delivery to cells in volumes approaching human-scale tissues, addressing necrosis in avascular cores. Progress toward full organs includes bioprinted airway components using patient-derived cells, achieving patency and mucociliary clearance in vitro, though full organ transplantation remains preclinical due to scalability and innervation challenges. In situ bioprinting, applied directly to injury sites, enhances adhesion and reduces surgical invasiveness, with demonstrations in skin and cartilage repair showing superior integration over ex vivo constructs.

Drug Discovery and Disease Modeling

3D bioprinting facilitates the fabrication of tissue constructs that emulate physiological environments, more accurate preclinical screening compared to traditional two-dimensional () cultures, which often fail to replicate extracellular matrix interactions and three-dimensional . These bioprinted models support high-throughput testing of , , and by incorporating patient-derived cells and biomimetic scaffolds, thereby bridging gaps between results and clinical outcomes. For instance, bioprinted liver tissues have been used to model first-pass and , demonstrating superior over monolayers in detecting compound-induced . In cardiac drug screening, bioprinted spheroids derived from AC16 cardiomyocytes have assessed doxorubicin-induced , revealing dose-dependent viability and functional impairments not fully captured in assays. models, such as glioblastoma-on-a-chip printed with patient-derived cells and cancer-associated fibroblasts, evaluate chemotherapeutic responses like temozolomide resistance, where matrix stiffness influences penetration and efficacy. models printed with osteoblasts have tested osteogenic s like icariin, showing enhanced mineralization and markers such as and ALP, which correlate with in formation. with organ-on-a-chip systems further allows multi-organ simulations for pharmacokinetic studies, reducing reliance on models that exhibit interspecies variability. For disease modeling, bioprinted neural constructs replicate neurodegenerative pathologies, such as , by arranging dopaminergic neurons in stiffness-controlled hydrogels (2-4 kPa) that mimic brain tissue, enabling observation of spontaneous action potentials and aggregation. These models incorporate blood-brain barrier to study barriers, improving screening for therapeutics targeting amyloid-beta in Alzheimer's or motor neuron degeneration in . Post-2020 advancements include microfluidic-assisted bioprinting for gradient-patterned, patient-specific neural models using induced pluripotent stem cells, which enhance resolution in replicating progression and therapeutic responses. Overall, these applications have supported planned clinical validations, such as comparing bioprinted tumor responses to against , aiming to refine lead selection and de-risk pipelines.

Cultured Meat and Food Production

3D bioprinting facilitates the production of structured cultured meat by enabling the precise deposition of animal-derived cells, such as bovine muscle, fat, and endothelial cells, alongside edible bioinks to replicate the hierarchical architecture of whole cuts like steaks. This approach addresses limitations in traditional cell culturing methods, which often yield unstructured products resembling ground meat, by allowing control over tissue composition, marbling, and vascular networks essential for texture and flavor development. In February 2021, Aleph Farms, in collaboration with the Technion – Israel Institute of Technology, unveiled the world's first 3D bioprinted cultivated ribeye steak using non-genetically modified bovine cells differentiated into muscle, fat, and connective tissues, printed layer-by-layer to form a marbled structure approximately 1.5 cm thick. Research demonstrates that bioprinted constructs can incorporate antioxidative protein hydrolysates in alginate/gelatin-based bioinks to enhance cell viability and reduce during printing and maturation, yielding meat-like tissues with viable myoblasts up to post-printing. A 2021 study reported the assembly of engineered steak-like tissue from bovine fibers—muscle fibers aligned for texture, fat strips for marbling, and vascular fibers for nutrient —achieving a structured of 0.8 cm³ with histological features mimicking beef. These advancements extend to , where bioprinting modalities like extrusion and inkjet systems have been explored to fabricate muscle tissues from fish cells, though scalability remains constrained by cell sourcing and media costs. Challenges in 3D bioprinting for cultured meat include ensuring food-grade biomaterials that support long-term cell maturation without cytotoxicity, as many hydrogels degrade too rapidly or fail to mimic extracellular matrices adequately. Print resolution limits the replication of fine marbling patterns, often requiring post-printing bioreactor conditioning to promote hypertrophy and fiber alignment, which extends production timelines to weeks or months. Economic barriers persist, with current processes yielding small-scale prototypes at costs exceeding $10,000 per kilogram, far from competitive with conventional meat, necessitating innovations in high-throughput printers and serum-free media. Regulatory hurdles involve verifying nutritional equivalence and safety, as bioprinted tissues must demonstrate absence of contaminants and comparable protein digestibility to animal-derived products. Despite these, bioprinting holds potential for sustainable food production by reducing land use and emissions associated with livestock, provided technical viability improves.

Bioremediation and Environmental Engineering

3D bioprinting facilitates the creation of engineered living materials incorporating microorganisms, such as bacteria and microalgae, to degrade environmental pollutants through structured biofilms and scaffolds. These constructs enable precise spatial arrangement of microbial consortia, promoting synergistic metabolic pathways that enhance degradation efficiency over unstructured cultures. For example, extrusion-based bioprinting of bacteria-laden hydrogels has been employed to target organic pollutants and heavy metals, with viability rates exceeding 80% post-printing in optimized bioinks. In bioremediation applications, 3D-printed microbial structures have demonstrated superior performance in wastewater treatment by immobilizing degraders like Pseudomonas species within porous matrices, achieving up to 95% removal of phenols in batch tests conducted in 2023. Similarly, microalgae-based bioprinting constructs 3D lattices that improve nutrient uptake and pollutant sequestration, with studies reporting 2-3 fold higher bioremediation rates for nitrogen compounds compared to suspended cells. For soil remediation, printed fungal-bacterial hybrids form mycorrhizal-like networks that accelerate hydrocarbon breakdown, as evidenced by field trials reducing total petroleum hydrocarbons by 70% within 60 days. Environmental engineering benefits from bioprinted biosensors, where living materials detect contaminants via metabolic responses; for instance, E. coli-embedded prints signal heavy metal presence through colorimetric changes, enabling real-time monitoring with detection limits below 1 ppm for lead and cadmium. Recent developments include 3D-printed synthetic consortia in microbial fuel cells, boosting electricity generation and dye decolorization by 150% through optimized electron transfer gradients. In marine settings, bioprinted Ideonella sakaiensis variants degrade polyethylene terephthalate at rates tunable from 0.1 to 1.0 mg/cm²/day via bio-sticker formats tested in 2025 simulations. These approaches limitations of traditional , such as microbial washout in systems, by providing mechanically , customizable architectures that sustain activity under varying and conditions. However, remains constrained by shear-thinning and long-term microbial , with ongoing focusing on hybrid polymer-microbe formulations for industrial deployment.

Challenges and Limitations

Technical and Engineering Constraints

One primary constraint in 3D bioprinting is the limited resolution of printing techniques, which restricts the fabrication of structures at the cellular or subcellular scale required for functional tissues. , the most common method, typically achieves resolutions of 100–400 μm due to nozzle diameters and bioink extrusion dynamics, insufficient for replicating microvasculature or precise cell arrangements. offers higher precision but is constrained to approximately 300 μm resolution with current bioinks, limiting its applicability to finer vascular networks. Bioink formulation poses significant engineering challenges, as materials must balance printability—requiring suitable , shear-thinning behavior, and rapid crosslinking—with to support cell viability and proliferation. High-viscosity bioinks enable structural integrity but increase during , reducing post-print cell survival rates to 70–90% in many cases, with further declines due to inadequate in avascular constructs thicker than 200–500 μm. clogging from heterogeneous cell-laden inks or inconsistent crosslinking further hampers reliability, necessitating precise control of temperature, pressure, and humidity in printing environments. Vascularization remains a critical , as perfusable capillary-scale (5–10 μm ) exceeds multi-material capabilities, leading to necrotic cores in larger constructs and limiting tissue volumes to millimeters rather than organ-scale dimensions. Layer-by-layer deposition often results in anisotropic mechanical and poor interlayer , compromising long-term structural under physiological stresses, while slow speeds (e.g., 10–20 mm/s for ) exacerbate cell to damaging conditions and for clinical applications.

Biological and Viability Issues

One primary biological in 3D bioprinting is maintaining viability during the , where high from through nozzles damages membranes and induces . Studies indicate that viability often drops below 85% at shear rates exceeding 10-30 s⁻¹, depending on type and , with non-spherical cells experiencing up to 50% higher deformation. Efforts to mitigate this include shear-thinning and preconditioning cells to moderate , which can post-print by enhancing cytoskeletal . Post-printing, avascular constructs larger than 200-500 μm thick face necrosis due to diffusion-limited oxygen and nutrient delivery, restricting viable tissue scale to thin sheets or small modules. Vascularization remains a core viability barrier, as bioprinted endothelium-lined channels struggle to form hierarchical, perfusable networks mimicking native capillary densities (10⁴-10⁵ vessels/cm²), leading to thrombosis or incomplete anastomosis with host vessels upon implantation. Recent co-printing of angiogenic factors like VEGF has improved sprout formation but fails to sustain long-term patency in complex geometries. Long-term tissue functionality is compromised by incomplete cellular maturation and extracellular matrix (ECM) remodeling, where printed stem cells often arrest in partial differentiation states, yielding constructs with 20-50% of native mechanical strength and metabolic output. Bioprinted cardiomyocytes, for instance, exhibit asynchronous beating and limited contractility beyond 4-6 weeks in vitro, attributable to disrupted cell-cell junctions and absent native ECM cues. Host integration poses further risks, including immune-mediated rejection of allogeneic cells unless autologous iPSCs are used, though these carry tumorigenicity hazards from incomplete reprogramming. Overall, these issues underscore that while short-term viability exceeds 70-90% in optimized simple tissues, clinically viable organs demand unresolved advances in dynamic culture and biomimicry.

Scalability and Economic Barriers

One primary scalability barrier in 3D bioprinting stems from the protracted fabrication times required for clinically relevant constructs, as extrusion-based systems—the dominant —typically operate at speeds of 10-20 /s, rendering the assembly of centimeter- to organ-scale tissues a process spanning hours to days. This duration exceeds the viability window for many cell types, which often succumb to forces, deprivation, and , limiting printed structures to millimeter-scale modules rather than functional organs. Achieving vascularization for larger volumes compounds these issues, as resolutions below 100 μm are needed for capillary networks (approximately 5-10 μm in diameter), yet finer nozzles drastically reduce deposition rates and amplify from increased gradients. Techniques like laser-assisted or inkjet bioprinting offer higher but throughput, with overall inefficiencies preventing mass production akin to traditional . Efforts to parallelize printing heads or integrate sacrificial inks for post-print perfusion have shown promise in lab settings but fail to scale without custom , underscoring the trade-off between , speed, and biological . Economic impediments further deployment, with bioprinters priced between and exceeding million, depending on features like multi-material and sterility controls, which deter beyond well-funded institutions. Bioinks, comprising hydrogels or decellularized matrices laden with cells, incur high costs—often $200-500 per milliliter for specialized, cell-compatible variants—driven by sourcing of components and rigorous to minimize . Operational expenses, including cell under GMP conditions and extended maturation (weeks to months), amplify per-unit costs to levels uncompetitive with synthetic implants or donor organs, with analyses indicating that full organ bioprinting could exceed million per construct without optimization. These factors contribute to the field's nascent , valued at approximately billion in 2024 but projected to grow slowly amid unresolved cost reductions.

Bioethical Debates on Enhancement and Dignity

3D bioprinting raises bioethical concerns regarding , where the technology could extend beyond therapeutic applications to augment physical or cognitive capacities, such as printing enhanced tissues for superior strength or longevity. Critics argue that such enhancements risk exacerbating social inequalities, as access would likely favor the wealthy, potentially creating a divide between "enhanced" and "" humans. Proponents, however, contend that enhancements align with , akin to historical adaptations like or prosthetics, provided they are regulated to and . Empirical evidence remains limited, as clinical enhancements via bioprinting are not yet realized, but analogies to debates highlight risks of , including loss of genetic diversity. Debates on human dignity center on whether bioprinting commodifies the body by treating tissues as manufacturable products, potentially eroding the intrinsic derived from origins. Philosophers invoking warn that fabricating organs or enhancements instrumentalizes , reducing persons to editable components and challenging the of embodiment. In contrast, utilitarian perspectives emphasize dignity through improved , arguing that bioprinting restores wholeness for the diseased rather than diminishing it. Specific worries include the of printed structures; if vascularized tissues develop rudimentary , ethical dilemmas akin to arise, necessitating guidelines on and disposal. These concerns are amplified by the technology's potential for "designer" body parts, which some view as hubristic interference with limits, echoing critiques in literature since the . Regulatory proposals balancing with protections, such as prohibiting non-therapeutic enhancements until societal forms, informed by rather than unchecked . Source analyses reveal biases; papers often prioritize precautionary principles, potentially underplaying benefits to institutional , while industry reports may overstate enhancement feasibility without rigorous trials. As of 2023, no international specifically addresses bioprinting enhancements, leaving debates unresolved amid advancing prototypes like printed vascular networks.

Regulatory and Intellectual Property Concerns

Regulatory frameworks for bioprinted constructs remain underdeveloped as of , primarily adapting existing classifications for devices, biologics, and products rather than establishing bioprinting-specific guidelines. The U.S. () bioprinted tissues as products involving cells, scaffolds, and printing processes, subjecting them to oversight under the for Biologics and or and Radiological pathways, which necessitates demonstrating , , and manufacturing through preclinical and . No full bioprinted organs have received FDA approval for clinical use, though point-of-care bioprinting initiatives, such as on-site dressings or patches, face evolving for decentralized risks like variability in cell viability and sterility. In Europe, the European Medicines Agency (EMA) lacks dedicated regulations for bioprinted tissues, paralleling tissue-engineered products under advanced therapy medicinal product rules, which demand rigorous quality controls but have not authorized any bioprinted constructs beyond simple scaffolds. Key regulatory challenges include of bioinks, printers, and post-printing maturation processes, as variability in extrusion-based or laser-assisted methods can lead to inconsistent tissue functionality and risks not fully captured by current good practices (GMP). Risk classification debates persist, with bioprinted vascularized tissues potentially qualifying as high-risk class III devices requiring extensive trials, while simpler acellular scaffolds might fall under class II with 510(k) clearance; however, the integration of living cells complicates predictability of long-term integration and rejection. Internationally, harmonization efforts via bodies like the International Council for Harmonisation lag, exacerbating barriers for global commercialization, as evidenced by the absence of approved bioprinted implants in major markets despite preclinical successes in and analogs. Intellectual property (IP) in 3D bioprinting centers on patents for bioprinters, bioinks, scaffold designs, and algorithmic controls, with over 1,000 filings by 2023 dominated by companies like Organovo Holdings and Cellink AB, focusing on proprietary hydrogel formulations and multi-material extrusion techniques to enable vascularization. Patent eligibility remains contentious, particularly under U.S. law post-Alice Corp. v. CLS Bank, where bioprinting methods risk invalidation as abstract ideas unless tied to specific, non-obvious hardware innovations like novel nozzle geometries or cell encapsulation protocols. Disputes have arisen, including Organovo's 2022 counterclaims against Cellink for infringing patents on tissue printing platforms, highlighting tensions over method claims that could encompass broad applications in organoid fabrication. Ownership concerns extend to digital blueprints for patient-specific prints and derived cell lines, raising questions of inventorship when autologous cells are combined with patented scaffolds, potentially invoking ethical limits on patenting life forms under precedents like Association for Molecular Pathology v. Myriad Genetics. Bioink IP, critical for print fidelity, involves trade secrets for crosslinking chemistries, but enforcement challenges emerge from reverse-engineering risks in open-source printer adaptations, as seen in trademark suits like Advanced Solutions Life Sciences v. BioBots Inc. over branding overlaps. Licensing models are evolving to balance innovation, with cross-licensing agreements among firms addressing overlapping claims on decellularized matrices, though commercialization hurdles persist due to fragmented portfolios that deter investment without clear freedom-to-operate analyses.

Access, Equity, and Societal Impact

The high costs associated with 3D bioprinting and materials currently its primarily to well-funded institutions in developed . bioprinters from USD 13,000 to 300,000, while bioinks can USD 3.85 to 100,000 per gram, creating significant financial barriers that restrict widespread beyond specialized labs. These expenses, combined with the need for advanced such as sterile facilities and skilled personnel, further confine the to regions with robust biomedical ecosystems, such as , which held a 36.7% in 2025. In developing , analogous challenges with general 3D printing— including inadequate and expertise—suggest that bioprinting faces even greater hurdles, potentially perpetuating disparities in medical . Equity concerns arise from the risk that bioprinted tissues or organs could initially serve as luxuries for the affluent, exacerbating existing healthcare inequalities. As noted in bioethical analyses, the technology's commercialization may stratify access along socioeconomic lines, with high costs rendering it unavailable to lower-income populations and widening health gaps rather than bridging them. “Access to care… is challenged by the possibility of 3D bioprinting becoming a luxury available only to the privileged few,” highlighting how resource allocation could favor wealthier patients, potentially leading to social stratification in organ replacement therapies. While efforts to develop low-cost prototypes—such as those using recycled materials or off-the-shelf components—aim to democratize the field, these remain experimental and have not yet scaled to clinical use. Societally, 3D bioprinting holds potential to alleviate organ shortages and reduce reliance on donors, which could mitigate issues like illegal trafficking by addressing supply-demand imbalances. However, without deliberate policies for equitable , it risks amplifying of vulnerable groups through uneven , as barriers and market-driven may prioritize over . Projections indicate market from USD 2.95 billion in 2025 to USD 8.53 billion by 2032, driven by applications in , but this expansion could entrench inequalities if affordability improvements lag behind technological advances. Overall, realizing equitable societal impacts requires addressing these barriers through innovation in cost reduction and international regulatory to prevent the technology from becoming an .

Recent Advances and Future Directions

Breakthroughs Since 2020

In 2021, researchers developed a full-thickness skin model using an acellular dermal matrix combined with methacrylamide , enabling the fabrication of stratified epidermal and dermal layers that supported viability and maturation, advancing applications. Concurrently, a biomimetic bilayered for osteochondral repair was bioprinted using decellularized and fibroin, demonstrating enhanced integration with native and subchondral in preclinical models. Vascularization emerged as a critical focus, with 2022 innovations including tough hydrogel-based conduits that maintained structural integrity under physiological pressures and supported endothelial cell lining for functional blood vessel formation. That same year, multimaterial bioprinting produced liver tissue with branched vascular networks using human hepatocytes and endothelial cells, exhibiting bile acid secretion and albumin production indicative of metabolic functionality. Large-scale aortic models were also achieved via decellularized porcine cardiac tissue, providing perfusable vascular structures for cardiovascular research. Clinical translation advanced in 2022 when 3DBio Therapeutics launched the first for AuriNovo, a 3D bioprinted autologous auricular cartilage implant for microtia reconstruction, involving patient-derived chondrocytes embedded in a collagen-based bioink to form anatomically precise ears with over 90% cell viability post-printing. In reproductive medicine, 2022 bioprinting of ovary-derived structures with primary ovarian cells restored folliculogenesis and fertility in mouse models of ovarian failure, highlighting potential for endocrine tissue regeneration. By 2023, acoustic bioprinting enabled precise deposition of patient-derived organoids, improving predictions of cancer therapy responses through tumor microenvironment mimicry. Bone reconstruction saw a 2024 milestone with patient-specific 3D-printed mesh scaffolds optimized via CAD for critical defects, achieving and functional ambulation in a clinical case after 1.5 years, though cellular bioprinting remains preclinical. These developments underscore progress in formulation and , yet functional maturity and persist as barriers to broader organoid viability. 4D bioprinting extends 3D bioprinting by incorporating a temporal , enabling printed constructs to undergo programmable shape changes or functional adaptations in response to external stimuli such as , , or . This approach leverages biomaterials, including shape-memory polymers and hydrogels, to mimic dynamic biological processes like tissue morphogenesis or kinetics. For instance, in a 2024 study, researchers demonstrated 4D-printed biomaterials that self-fold into complex geometries post-printing, facilitating applications in adaptive scaffolds for tissue engineering. Recent advances emphasize integrating cellular contractile forces with biofabrication to drive spatiotemporal tissue , as evidenced by experiments in 2025 where engineered constructs exhibited controlled mimicking embryonic folding. Polymeric 4D bioprinting has also progressed toward regenerative applications, with bioinks designed for on-demand degradation or expansion to support vascularization and organoid growth. These developments, building on foundational work from 2019 onward, address limitations in static prints by enabling constructs that evolve post-fabrication, though challenges persist in achieving precise temporal control and under physiological conditions. Multi-material bioprinting involves the simultaneous deposition of diverse bioinks—such as cell-laden hydrogels, supportive matrices, and vascular components—to fabricate heterogeneous tissues that replicate native complexity. Advances since 2020 have focused on extrusion-based systems capable of switching between materials mid-print, structures like tri-layered models or composite bone grafts with integrated osteogenic and endothelial cells. A 2025 review highlighted multidimensional extensions, including 5D printing (adding ) and 6D (incorporating electrical stimuli), which enhance mechanical through embedded high-strength supports within porous bioinks. These trends converge in hybrid 4D multi-material systems, where stimuli-responsive inks allow sequential assembly and reconfiguration, as seen in prototypes for dynamic cardiac patches that contract and perfuse over time. Clinical remains nascent, with projections for 2025-2030 emphasizing improved below 100 micrometers and integration with for design optimization, though scalability and regulatory validation of long-term functionality are ongoing hurdles.

Projections for Clinical Translation and Market Growth

Clinical translation of 3D bioprinting remains limited to early-stage applications, with projections indicating incremental progress toward routine use for non-vascularized tissues by the early , while faces extended timelines to persistent challenges in vascularization, immune , and long-term functionality. As of 2025, eleven clinical trials incorporating bioprinted constructs have been globally since 2016, predominantly involving models, patient-specific implants for or , or testing rather than function restoration. The inaugural trial utilizing a 3D bioprinted tissue began in 2022, led by 3DBio Therapeutics for autologous in microtia patients, demonstrating feasibility for personalized, avascular implants but highlighting limitations for broader . Regulatory agencies, including the FDA and EMA, initiated framework development in 2024-2025 to address bespoke manufacturing and cellular variability, yet experts anticipate approvals for advanced constructs like grafts or bone scaffolds within 5-10 years, with full vascularized organs unlikely before 2040 owing to unresolved bioengineering hurdles. Market growth forecasts reflect optimism from R&D investments and ancillary applications in drug screening and tissue modeling, though tempered by translational delays and high costs, projecting expansion from current valuations around USD 2 billion to USD 3-5 billion by 2030.
SourceBase Year Value (USD Billion)Projected Value by 2030 (USD Billion)CAGR (%)
2.0 (2022)5.312.5
1.67 (2025)3.4915.89
These estimates from consulting firms, while consistent in , incorporate assumptions of accelerated maturation and regulatory streamlining that may prove overly given historical in regenerative therapies; nonetheless, sustained venture —exceeding USD million annually in recent years—underpins realistic mid-term in preclinical and diagnostic segments.

References

  1. [1]
    3D Bioprinting Methods and Techniques: Applications on Artificial ...
    Bioprinting is defined as the positioning of biochemicals, biological materials, and living cells for the generation of bioengineered structures (i.e. additive ...
  2. [2]
    3D Bioprinting:principles, fantasies and prospects - ScienceDirect
    3D bioprinting is the process of creating biomedical structures using living cells, biological chemicals, and biomaterials [1]. Cell culture techniques are ...
  3. [3]
    Evolution of bioprinting and current applications - PMC
    The first print associated with the encapsulation of cells in hydrogels was produced in 2006 by bioprinting liver cells for an in vitro model for ...Missing: peer- | Show results with:peer-
  4. [4]
    Applications, advancements, and challenges of 3D bioprinting in ...
    Feb 20, 2024 · This review respectively introduces the development of 3D bioprinting as well as its working principles and main applications in the medical field.
  5. [5]
  6. [6]
    Printability and Shape Fidelity of Bioinks in 3D Bioprinting
    Aug 28, 2020 · This review discusses qualitative and quantitative methodologies to evaluate printability of bioinks for extrusion- and lithography-based bioprinting.
  7. [7]
    Advancements in tissue and organ 3D bioprinting - ScienceDirect.com
    The present review is focused on tissue- and organ-based 3D bioprinting to achieve regenerative alternatives. We briefly summarized and evaluated various ...
  8. [8]
    3D Bioprinting in Skin Related Research: Recent Achievements and ...
    The objective of this paper was to review the latest advances in 3D bioprinting dedicated to skin applications.3. Bioprinting · 3.1. 3d Bioprinting Methods · 3.2. Bioink<|separator|>
  9. [9]
    Advancements of 3D bioprinting in regenerative medicine
    Feb 15, 2024 · In this review, we explore the impact of different 3D bioprinting technologies and bioink materials on seed cells, providing a comprehensive overview of the ...4.1. Cell Lines · 4.2. Stem Cells · 4.4. Primary Cells
  10. [10]
    Full article: Recent advances in 3D bioprinting of tissues and organs ...
    In the present review, we summarized recent advances in the field of 3D bioprinting tissues and organs for transplantation and drug screening. This review first ...2. 3d Bioprinting Techniques · 3. Bioinks · 3.2. Biomaterials Of Bioink
  11. [11]
    Advances, Challenges, and the Promise of 3D Bioprinting
    Aug 20, 2025 · This comprehensive review article aims to provide an in-depth analysis of current 3D bioprinting technologies, emphasizing advanced printing ...<|separator|>
  12. [12]
    3D Bioprinting in Tissue Engineering: Advancements, Challenges ...
    Jan 10, 2025 · This review covers the most common 3D bioprinting technologies. Post Bioprinting stage. Most bioprinted structures require stabilization ...3d Bioprinting Process · Recent Advances In... · Applications In Tissue...Missing: achievements | Show results with:achievements
  13. [13]
    Applications, advancements, and challenges of 3D bioprinting in ...
    This review respectively introduces the development of 3D bioprinting as well as its working principles and main applications in the medical field.
  14. [14]
    3D Bioprinting at the Frontier of Regenerative Medicine ...
    3D printing technology has emerged as a key driver behind an on-going paradigm shift in the production process of various industrial domains.Missing: milestones | Show results with:milestones
  15. [15]
    3D bioprinting and its innovative approach for biomedical applications
    Dec 24, 2022 · 3D bioprinting or additive manufacturing is an emerging innovative technology revolutionizing the field of biomedical applications.Missing: milestones | Show results with:milestones
  16. [16]
    Printability and Shape Fidelity of Bioinks in 3D Bioprinting - PMC
    This review discusses qualitative and quantitative methodologies to evaluate printability of bioinks for extrusion- and lithography-based bioprinting.
  17. [17]
    Development of 3D bioprinting: From printing methods to biomedical ...
    We systematically review the evolution, process and classification of 3D bioprinting with an emphasis on the fundamental printing principles and commercialized ...
  18. [18]
    3-D bioprinting technologies in tissue engineering and regenerative ...
    Known as 3D bioprinting, this technology involves the precise layering of cells, biologic scaffolds, and growth factors with the goal of creating bioidentical ...
  19. [19]
    Quantifying Oxygen Levels in 3D Bioprinted Cell-Laden Thick ...
    May 30, 2020 · Diffusion limit of oxygen in vivo is approximately 200 µm, and cells ... The body of work on 3D printing of vascularized tissues and organs is ...
  20. [20]
    Biomechanical factors in three-dimensional tissue bioprinting - PMC
    The mechanical forces exerted on cells during 3D bioprinting generally include hydrostatic pressure, shear stress, and extensional stress. Hydrostatic pressure ...
  21. [21]
    3D Printing History: A Complete Timeline of Additive Manufacturing ...
    Jun 25, 2024 · 3D printing's roots began with the inventive efforts of Dr. Hideo Kodama. In 1981, he laid the foundation for additive manufacturing.
  22. [22]
    The History of 3D Printing: From the 80s to Today - Sculpteo
    The first 3D printing attempts are granted to Dr Kodama for his development of a rapid prototyping technique. He was the first to describe a layer by layer ...
  23. [23]
    Additive Manufacturing History: From the 1980's to Now - Markforged
    In 1984, Charles Hull developed the material Stereolithography Apparatus known as SLA. He went on to establish the first 3D printing company in 1986, that then ...White Paper: Why Additive... · History Of Additive... · Today's Additive...<|separator|>
  24. [24]
    The complete history of 3D printing - UltiMaker
    Shortly after, in 1989, Scott Crump patented fused deposition modeling (FDM) – also known as fused filament fabrication (FFF) – and founded Stratasys, one ...
  25. [25]
    Additive Manufacture of Ceramics Components by Inkjet Printing
    Inkjet printing was one of the first technologies to be developed for additive manufacture. In 1992, Sachs et al. at MIT described a method for manufacturing ...
  26. [26]
    Cytoscribing: a method for micropositioning cells and the ... - PubMed
    Cells may be positioned in precise, predetermined patterns by a technique, termed cytoscribing, in which cell adhesion proteins are deposited on a substratum ...Missing: bioprinting | Show results with:bioprinting
  27. [27]
    3D Bioprinting Technology and Hydrogels Used in the Process - NIH
    Nov 3, 2022 · In 1988, the researcher Robert J. Klebe used cytoscribing technology to demonstrate the potential of positioning biological products, using ...
  28. [28]
    Cell and organ printing 2: Fusion of cell aggregates in three ...
    Apr 15, 2003 · We recently developed a cell printer (Wilson and Boland, 2003) that enables us to place cells in positions that mimic their positions in an ...Missing: bioprinting | Show results with:bioprinting
  29. [29]
    fusion of cell aggregates in three-dimensional gels - PubMed
    We recently developed a cell printer (Wilson and Boland, 2003) that enables us to place cells in positions that mimic their respective positions in organs.Missing: experiment | Show results with:experiment
  30. [30]
    Is 3D Bioprinting the Future of Tailor-Made Medicine? - 3Dnatives
    Oct 10, 2024 · Klebe of the University of Texas presented his cytoscribing process, a method of micropositioning cells to construct two and three-dimensional ...
  31. [31]
    Printing cures: Organovo advances with 3D-printed liver tissue
    Feb 17, 2017 · Founded in late 2007, the company licensed their bioprinting technology from the University of Missouri, where it was developed in the lab of ...
  32. [32]
    Organovo Develops First Commercial 3D Bio-Printer for ...
    Jun 8, 2010 · Organovo Develops First Commercial 3D Bio-Printer for Manufacturing Human Tissue and Organs. SAN DIEGO, Calif.
  33. [33]
    The history of 3D printing in healthcare - RCSEng
    In April 2013 US company Organovo created the world's first fully cellular 3D bioprinted liver tissue. Commenting at the time, Keith Murphy, Chairman and Chief ...
  34. [34]
    3D Bioprinting: These Are The Top 15 Bioprinting Companies
    Aug 22, 2024 · The company recently received FDA approval for their 3D-printed resorbable surgical mesh designed for use in the reinforcement of soft ...Missing: commercialization | Show results with:commercialization
  35. [35]
    3D Systems Advances Regenerative Medical Solutions for First-of ...
    Jun 26, 2025 · 3D Systems (NYSE: DDD) announced that its 3D bioprinting technologies have enabled FDA approval for the regenerative repair of peripheral nerve damage.Missing: commercialization | Show results with:commercialization
  36. [36]
    3D Systems receives FDA approval for 3D printed first in peripheral ...
    Jun 26, 2025 · 3D Systems receives FDA approval for 3D printed first in peripheral nerve repair ... "I truly believe this will redefine treatment paradigms.".3d Scanning And Metrology... · Post-Processing News · Supply Chain NewsMissing: commercialization | Show results with:commercialization
  37. [37]
    3D Bioprinting in 2025: What Medical Device Makers Need to Know ...
    The FDA oversees 3D bioprinting through three primary centers: CDRH for medical devices, CBER for biological applications, and CDER for drug-related aspects.
  38. [38]
    Current Developments in 3D Bioprinting for Tissue and Organ ...
    Briefly, the process follows a general outline in the form of three basic steps: Preparatory phase, Printing phase and Post-handling. Preparatory phase is the ...
  39. [39]
    Narrative review of 3D bioprinting for the construction of in vitro ...
    Preparation for bioprinting. Bioprinting preparation involves two steps: readying the print material and acquiring 3D structural data of the target object.Bioprinting Stages · 3d Bioprinting Technologies · Tumor Models<|separator|>
  40. [40]
    3D Printing of Scaffolds for Tissue Regeneration Applications - PMC
    Evolving from conventional techniques, 3D printing provides tissue engineers with a way to design scaffolds capable of mimicking the complex structures of the ...
  41. [41]
    Bioinks for 3D bioprinting: an overview - PMC - NIH
    Bioinks are solutions of biomaterials or mixtures in hydrogel form, used in bioprinting to create tissue constructs, and can be natural, synthetic, or hybrid.
  42. [42]
    Advances in 3D Bioprinting: Techniques, Applications, and Future ...
    Jul 16, 2023 · The four main bioprinting techniques are droplet-based, laser-assisted, stereolithography, and extrusion-based, and are used with a range of ...2.3. 3d Bioprinting · 3. Bioprinting Techniques · 3.4. Extrusion-Based...
  43. [43]
    3D Bioprinting Technologies for Tissue Engineering: A Mini Review
    Aug 19, 2020 · 3D bioprinting uses layer-by-layer deposition of biological materials to create 3D organ constructs, with primary techniques including inkjet, ...
  44. [44]
    3D bioprinting of cells, tissues and organs | Scientific Reports - Nature
    Aug 18, 2020 · The discovery of a 3D printer dates back to early 1980s when Charles Hull, an American engineer, built the 1st 3D printer, capable of ...Missing: first | Show results with:first<|separator|>
  45. [45]
    Three-Dimensional Bioprinting Strategies for Tissue Engineering
    In this article, we review the application of 3D bioprinting to tissue engineering. We first discuss the general strategies for printing functional tissue ...
  46. [46]
    [PDF] 3-D bioprinting technologies in tissue engineering and regenerative ...
    accelerated tissue maturation when using a microtissue-based approach.8,16 ... 3D bioprinting of tissues and organs. Nat Biotechnol. 2014;32(8):773-785 ...
  47. [47]
    Innovative bioinks for 3D bioprinting - PubMed Central - NIH
    Jan 20, 2025 · The various types of bioinks discussed are decellularized extracellular matrix, protein based, cell based and hybrid based bioink (Table 2) and ...
  48. [48]
    Proposal to assess printability of bioinks for extrusion-based ...
    Although extrusion-based bioprinting offers a flexible fabrication platform, the rheological demands placed on the bioinks are stringent.
  49. [49]
    Bioinks of Natural Biomaterials for Printing Tissues - PMC - NIH
    Jun 10, 2023 · Bioinks are inks—in other words, hydrogels—prepared from biomaterials with certain physiochemical properties together with cells to establish ...
  50. [50]
    Advances in Polymeric Nanoparticles and Hydrogels in 3D Bioprinting
    Sep 13, 2025 · Hydrogels and polymeric nanoparticles are used in 3D bioprinting to produce bioinks, which enhances the fabrication of tissues and organs ...
  51. [51]
    The introduction and evaluation of novel decellularized extracellular ...
    Sep 30, 2025 · The bioink composed of gellan gum and decellularized extracellular matrix was abbreviated as GG/dECMb for convenience throughout the manuscript.
  52. [52]
    Microalgae-enriched (bio)inks for 3D bioprinting of cultured seafood
    Feb 12, 2025 · This study introduces novel (bio)inks: (i) κ-CAM bioinks (κ-carrageenan, alginate, and methylcellulose) compatible with seabass cells; and (ii) mFAT inks.
  53. [53]
  54. [54]
    Extrusion bioprinting: meeting the promise of human tissue ... - NIH
    Mar 11, 2025 · Extrusion is the most popular bioprinting platform. Predictions of human tissue and whole-organ printing have been made for the technology.
  55. [55]
    Biomaterials / bioinks and extrusion bioprinting - ScienceDirect.com
    Important properties of bioinks include their physical, rheological, crosslinking, mechanical, and biological ones.
  56. [56]
    Embedded 3D bioprinting – An emerging strategy to fabricate ...
    Embedded bioprinting is a gel–in–gel approach developed to overcome the gravitational and overhanging limits of bioprinting to print large–sized constructs.
  57. [57]
    Recent advances in extrusion-based 3D printing for biomedical ...
    One major advantage for extrusion based-printing, is the ability to fabricate designs with high cell densities (e.g. >1×106 cells/mL or even spheroids).Missing: principles | Show results with:principles
  58. [58]
    Extrusion bioprinting of soft materials: An emerging technique for ...
    Mar 19, 2019 · In extrusion-based bioprinting, soft materials are loaded into the cartridges and extruded from the nozzle via pneumatic or mechanical actuation.I. Introduction · 2. Stereolithography · 4. Extrusion
  59. [59]
    Biomaterials for extrusion-based bioprinting and biomedical ...
    Jun 21, 2024 · New applications of extrusion-based bioprinting are systematically emerging in the biomedical field in relation to tissue and organ fabrication.
  60. [60]
    Biomaterials for extrusion-based bioprinting and biomedical ...
    Jun 21, 2024 · Extrusion-based bioprinting presents a series of specific challenges in relation to achievable resolutions, accuracy and speed. Resolution and ...
  61. [61]
    An overview of extrusion-based bioprinting with a focus on induced ...
    During the extrusion-based bioprinting process, cells are subjected to mechanical forces of different kinds. Among these mechanical forces, shear stress is of a ...Review Article · Introduction · Extrusion-Based Bioprinting...
  62. [62]
    Persuasive factors on the bioink printability and cell viability in the ...
    The bioink must be tuned for biocompatibility to accommodate live cells, mechanical stability after printing, and good resolution [34]. It is perceived from the ...
  63. [63]
    CELLINK's 2022 Year in Review
    Here is a round up of your favorite resources and key milestones of 2022 that have brought us one step closer to reducing our dependence on animal testing.
  64. [64]
    Recent advances in bioprinting techniques: approaches ...
    Sep 20, 2016 · Scientists have made great progress in patterning molecules, cells and organs by inkjet printing. Molecules such as DNA have been successfully ...Inkjet-Based Bioprinting · Bioprinting Cells · Bioprinting Stem Cells
  65. [65]
    Droplet-based bioprinting | Springer Nature Experiments
    Droplet-based bioprinting (DBB) is unique among the existing bioprinting modalities in its ability to handle and manipulate bioprinting at the cellular level.
  66. [66]
    Recent Advances in Additive Manufacturing and 3D Bioprinting for ...
    Similar to the inkjet technology, extrusion bioprinters employ either mechanical or pneumatic forces to extrude bioink in a continuous cylindrical stream ...
  67. [67]
    The promising rise of bioprinting in revolutionalizing medical science
    Droplet-based bioprinting offers excellent spatial resolution which makes it desirable for application in tissue engineering and regenerative medicine. These ...
  68. [68]
    3D Bioprinting of Biomaterials: A Review of Advances in Techniques ...
    Aug 28, 2025 · 3D bioprinting is an innovative and revolutionary technological model that fuses additive manufacturing concepts with bioengineering approaches ...
  69. [69]
    Laser-induced forward transfer based laser bioprinting in biomedical ...
    Aug 21, 2023 · The working principle of laser-assisted bioprinting is to concentrate laser energy on an absorbent layer on a glass plate, which pushes the ...
  70. [70]
    A Review of Bioprinting Techniques, Scaffolds, and Bioinks - NIH
    The purpose of this review is to provide a background on the leading 3D bioprinting methods and bioink selections for tissue engineering applications.
  71. [71]
    A review on cell damage, viability, and functionality during 3D ...
    Dec 16, 2022 · Benefiting from its nozzle-free mechanism, a large range of bioink viscosity up to 300 mPa·s can be applicable to laser-assisted bioprinting, ...
  72. [72]
    Laser-induced forward transfer based laser bioprinting in biomedical ...
    Aug 21, 2023 · This review briefly introduces the experimental setup, bioink ejection mechanisms, and parameters relevant to LIFT bioprinting. Furthermore, it ...
  73. [73]
    Stereolithography 3D Bioprinting - PubMed
    Stereolithography (SLA) 3D bioprinting has emerged as a prominent bioprinting method addressing the requirements of complex tissue fabrication.
  74. [74]
    Stereolithography apparatus and digital light processing-based 3D ...
    Feb 17, 2023 · Our review aims to provide an overview of SLA- and DLP-based 3D bioprinting strategies, starting from factors that affect these bioprinting processes.
  75. [75]
    Recent advances in the development of stereolithography-based ...
    Compared to other additive manufacturing (AM) techniques, SLA provides superior resolution (as low as 10 μm), enhanced surface quality and higher printing speed ...
  76. [76]
    Light-based 3D bioprinting techniques for illuminating the advances ...
    Light-assisted strategies leverage light-induced polymerization to modify the rheological properties and mechanical strengths of bioinks on-demand, thereby ...
  77. [77]
    An Overview of Laser-assisted Bioprinting (LAB) in Tissue ...
    Jun 30, 2021 · This review tackles the basic principles and key aspects of Laser-Assisted Bioprinting. Recent advances, limitations, and future perspectives ...
  78. [78]
    Laser-Induced Forward Transfer in Organ-on-Chip Devices - MDPI
    Laser-induced forward transfer (LIFT) bioprinting uses a laser to propel a droplet of bioink, enabling precise, non-contact deposition of biological materials.
  79. [79]
    Bioprinting the future using light: A review on photocrosslinking ...
    Light-based bioprinting uses light to control biomaterial formation, using photocrosslinking reactions and photoinitiators to create bonds between molecules.
  80. [80]
  81. [81]
  82. [82]
  83. [83]
  84. [84]
  85. [85]
    Biomimetic structural design in 3D-printed scaffolds for bone tissue ...
    By employing 3D bioprinting techniques, we aim to more precisely mimic the natural process of bone growth and support tissue regeneration. This approach not ...
  86. [86]
    Advances in 3D bioprinting for urethral tissue reconstruction
    Dec 5, 2023 · Moreover, diverse 3D bioprinting methods can replicate the native tissue microenvironment by integrating the patient cells and providing ...
  87. [87]
    Harnessing native blueprints for designing bioinks to bioprint ...
    Jan 23, 2025 · Engineered cardiac tissues developed through three-dimensional bioprinting (3DBP) offer a promising alternative. However, reproducing the native ...
  88. [88]
    Spatially controlled construction of assembloids using bioprinting
    Jul 19, 2023 · We develop a platform, termed Spatially Patterned Organoid Transfer (SPOT), consisting of an iron-oxide nanoparticle laden hydrogel and magnetized 3D printer.<|control11|><|separator|>
  89. [89]
    High-throughput bioprinting of spheroids for scalable tissue fabrication
    Nov 21, 2024 · This study presents HITS-Bio (High-throughput Integrated Tissue Fabrication System for Bioprinting), a multiarray bioprinting technique for rapidly positioning ...
  90. [90]
    Development and deployment of a functional 3D-bioprinted blood ...
    Apr 5, 2025 · When bioprinting tissue, the aim is to accurately replicate the three-dimensional architecture, along with the physical and biochemical ...
  91. [91]
    Advanced bioprinting strategies for fabrication of biomimetic tissues ...
    This review paper systematically explores the challenges and design requirements in the fabrication of 3D biomimetic tissue constructs, emphasizing the need for ...
  92. [92]
    Application of 3D bioprinting in the prevention and the therapy for ...
    May 14, 2021 · Light-assisted printing is a bioprinting technique that uses light to solidify a photocurable bioink. Light-assisted printing can be divided ...
  93. [93]
    Three-dimensional bioprinting using self-assembling scalable ...
    Jun 27, 2016 · Here, we fabricate and engineer scaffold-free scalable tissue strands as a novel bioink material for robotic-assisted bioprinting technologies.
  94. [94]
  95. [95]
    3-D bioprinting technologies in tissue engineering and regenerative ...
    Three central approaches to bioprinting are biomimicry, autonomous self-assembly, and a microtissue-based method. These general strategies are not exclusive to ...
  96. [96]
    Utilizing bioprinting to engineer spatially organized tissues from the ...
    Apr 8, 2024 · In this review, we summarize the techniques applied in fabrication and assembly of building blocks. We elaborate mechanisms and applications of bioprinting.Inkjet Bioprinting Assembles... · Extrusion-Based Bioprinting... · Bioprinting Tissues...Missing: autonomous | Show results with:autonomous
  97. [97]
    Engineering transferrable microvascular meshes for subcutaneous ...
    Oct 10, 2019 · Vascularized organoid engineered by modular assembly ... 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs.
  98. [98]
    In situ 3D bioprinting: The future of regenerative medicine
    Jun 9, 2025 · In situ 3D bioprinting offers enhanced tissue integration and surface construction capabilities, demonstrating greater clinical potential.
  99. [99]
    Assessing the landscape of clinical and observational trials ...
    Feb 17, 2025 · Four interventional trials aimed to implant bioprinted tissues made with autologous cells, including blood vessels, trachea, external ear, and ...
  100. [100]
    [PDF] ICMRA Innovation Project 3D Bio-Printing Case Study
    Multiple studies have demonstrated that 3D bio-printed myocardial tissue patches can be successfully transplanted to repair damaged myocardium in animals.
  101. [101]
    3D bioprinting of collagen-based high-resolution internally ... - Science
    Apr 23, 2025 · We three-dimensionally (3D) bioprint extracellular matrix (ECM) and cells into collagen-based high-resolution internally perfusable scaffolds (CHIPS)
  102. [102]
    A Milestone in 3D Printed Organs
    Lund University researchers have designed a new bioink which allows small human-sized airways to be 3D-bioprinted with the help of patient cells.
  103. [103]
    3D bioprinting for the construction of drug testing models ... - Frontiers
    Feb 13, 2025 · 3D models will provide a valuable tool in screening drugs at different stages as they are one step closer to bio-mimic human tissues.
  104. [104]
    Recent applications of three-dimensional bioprinting in drug ...
    This review provides a comprehensive summary of the latest applications of 3D bioprinting in drug discovery and development.
  105. [105]
    The rise of 3D bioprinting advancements in modeling ...
    Today, 3D bioprinting is gaining increasing attention as a powerful platform for disease modeling, moving beyond dated 2D culture systems. 3D bioprinting ...
  106. [106]
    Engineered whole cut meat-like tissue by the assembly of cell fibers ...
    Aug 24, 2021 · We demonstrate in vitro construction of engineered steak-like tissue assembled of three types of bovine cell fibers (muscle, fat, and vessel).<|separator|>
  107. [107]
    An overview of cultured meat and stem cell bioprinting: How to make ...
    3D printing has the potential to provide distinct solutions for the crucial challenges in cultured meat production, specifically in controlling the protein, fat ...
  108. [108]
    Aleph Farms & The Technion Reveal First Cultivated Ribeye Steak
    We have successfully cultivated the world's first slaughter-free ribeye steak, using three-dimensional (3D) bioprinting technology and natural building blocks ...
  109. [109]
    World's First 3D Bioprinted And Cultivated Ribeye Steak Is Revealed
    Feb 12, 2021 · The world's first 3D bioprinted and cultivated ribeye steak was made without genetic engineering. Created by Aleph Farms Ltd. and the Faculty of Biomedical ...
  110. [110]
    Bioengineered Lab-Grown Meat-like Constructs through 3D ...
    Jul 18, 2022 · We developed a 3D bioprinted and bioactive meat culture platform using Alg/Gel/PHs as a printable and edible component for the mass production of cultured meat.
  111. [111]
    A narrative review: 3D bioprinting of cultured muscle meat and ... - NIH
    Aug 14, 2024 · This review article covers these bioprinting modalities and the-state-of-art in relevant cells, biomaterials, printability, and nutritional ...
  112. [112]
    3D bioprinting of cultured muscle meat and seafood products and its ...
    Overall, several challenges, including overcoming consumer skepticism, improving scalability and cost-effectiveness, and ensuring the safety and quality of 3D ...
  113. [113]
    Trends and Technological Challenges of 3D Bioprinting in Cultured ...
    The purpose of this article is to review and analyze published studies related to the biofabrication of cultured meat using 3D bioprinting techniques.
  114. [114]
    (PDF) Trends and Technological Challenges of 3D Bioprinting in ...
    Nov 9, 2023 · However, like any scientific advance, 3D bioprinting of cultured meat faces challenges involving regulation, acceptance, the selection of ideal ...
  115. [115]
    A Decade of Progress in Cultured and Bioprinted Meat - 3DPrint.com
    Oct 2, 2023 · Diving into the world of 3D bioprinted cultivated meat reveals a dynamic landscape where biotechnology, food science, and engineering converge.
  116. [116]
    Material extrusion-based 3D printing for the fabrication of bacteria ...
    This article posits 3D bioprinting as a cornerstone of next-generation bioremediation practices, offering scalable, customizable, and potentially greener ...
  117. [117]
    3D bioprinting of microorganisms: principles and applications
    Jan 31, 2024 · This review covers current developments in 3D bioprinting methods for microorganisms. We go over the bioink compositions designed to promote microbial ...
  118. [118]
    3D bioprinting in bioremediation: a comprehensive review of ...
    Feb 8, 2024 · 3D bioprinting is an additive manufacturing technology that involves the precise layer-by-layer positioning of biological materials, ...
  119. [119]
    Microalgae-Based 3D Bioprinting: Recent Advances, Applications ...
    Current studies demonstrate that 3D bioprinting technology primarily enhances bioremediation efficacy through two practical strategies: constructing ...
  120. [120]
    Advances in 3D bioprinting for environmental remediation and ...
    Sep 9, 2024 · This review highlights the applications of 3D bioprinting in the environmental monitoring and bioremediation. 3D printing in solid waste ...
  121. [121]
    3D Bioprinting of Microbial-based Living Materials for Advanced ...
    3D bioprinting provides a powerful tool for engineering microorganisms and polymers into living materials with delicate structures.
  122. [122]
    3D printing of synthetic microbial consortium for boosting ...
    Apr 12, 2025 · A 3D printing approach for construction of starch MFC with rationally designed synthetic bacterial consortium was developed, which dramatically improved the ...
  123. [123]
    3D-bioprinted marine bacteria for the degradation of bioplastics
    Feb 5, 2025 · 3D-bioprinted marine bacteria, integrated into bio-stickers, degrade bioplastics in marine environments. The rate of degradation can be tuned.<|control11|><|separator|>
  124. [124]
    Controllable and biocompatible 3D bioprinting technology for ...
    This article posits 3D bioprinting as a cornerstone of next-generation bioremediation practices, offering scalable, customizable, and potentially greener ...
  125. [125]
    3D Bioprinting for Vascularization - PMC - NIH
    May 18, 2023 · With current technology, SLA bioprinting is limited to a print resolution of about 300 μ m, thus preventing it from printing even smaller ...
  126. [126]
    3D Bioprinting of Vascularized Tissues for in vitro and in vivo ...
    When examining the printed tissue's physiological needs, we see that the limit of diffusion for O2 in tissue is generally accepted to be limited to 100–200 ...
  127. [127]
    A review on cell damage, viability, and functionality during 3D ...
    Dec 16, 2022 · For example, the cells mainly endure strong shear stress during microextrusion-based bioprinting, resulting in loss of cell viability to various ...Fig. 1 · Cell Damage Modelling · Abbreviations
  128. [128]
    Harnessing shear stress preconditioning to improve cell viability in ...
    We hypothesized that moderate shear stress preconditioning increases the ability of cells to tolerate bioprinting induced stress, thus improving their viability ...2. Materials And Methods · 2.2. Shear Stress... · 3. Results
  129. [129]
    3D Bioprinting for Vascularization - MDPI
    May 18, 2023 · The difficulty in creating a sufficient blood vessel system generally stems from a difficulty in mimicking natural vascular architecture and ...3d Bioprinting For... · 5.1. Hydrogels · 8. Feasibility, Limitations...
  130. [130]
    Synergistic coupling between 3D bioprinting and vascularization ...
    Nov 20, 2023 · 3D bioprinting can enable the creation of personalized, tissue-specific vascular network while the vascularization enhances tissue viability and function.
  131. [131]
    Tissue Bioprinting: Promise and Challenges - PMC - NIH
    Dec 7, 2023 · Some of these challenges include engineering tissue complexity, post-print tissue maturation and maintenance, standardized and scalable ...Missing: constraints | Show results with:constraints
  132. [132]
    Advances In Three-Dimensional Bioprinted Stem Cell-Based Tissue ...
    A major challenge of 3D bioprinting strategies is the inability of matrix molecules to reconstitute the complexity of the extracellular matrix and the intrinsic ...
  133. [133]
    Three-Dimensional Bioprinting: A Comprehensive Review for ...
    This review highlights the integration of 3D printing, specifically bioprinting, across several surgical disciplines over the last five years.
  134. [134]
    Progress and challenges on extrusion based three dimensional (3D ...
    The present paper reviews the bioprinting process in general together with challenges associated with that to overcome various limitations.<|separator|>
  135. [135]
    3D Bioprinting: Challenges in Commercialization and Clinical ...
    One challenge related to scalability (printing human scale tissues and organs) will be to maintain the cells without damage during the long printing process.
  136. [136]
    Breaking the resolution limits of 3D bioprinting: future opportunities ...
    Dec 10, 2022 · This is especially relevant, as high resolution is often associated with long fabrication timescales, and cell viability could thus be impaired ...
  137. [137]
    3D Bioprinting: Advantages, Challenges, and Applications
    Jan 24, 2025 · Economic factors also play a significant role. A bioprinter can cost anywhere from $100,000 to over $500,000, depending on the sophistication ...
  138. [138]
    Development of a high-performance open-source 3D bioprinter
    Dec 31, 2022 · Chief among these barriers is the high cost of commercial, research-grade 3D bioprinting platforms, which range from $5000 to over $1,000,000.
  139. [139]
    [PDF] A Qualitative Assessment of Current Challenges Facing the ...
    Jul 1, 2025 · Main challenges include low cell viability, high costs of biomaterials/printers, and high customer expectations of bioprinting.
  140. [140]
    3D Bioprinting Market Worth US$2.4 billion | MarketsandMarkets
    Apr 8, 2025 · High cost of 3D bioprinters and bioinks. Opportunities: Rising demand for organ transplants. Challenge: Biocompatibility issues and rigorous ...
  141. [141]
    3D Bioprinting Market valued at USD 1.41 billion in 2024 |
    Sep 16, 2024 · The global 3D bioprinting market, valued at USD 1.41 billion in 2024, is expected to reach USD 2.86 billion by 2030, growing at a CAGR of 11.5%.
  142. [142]
    Bioethical and Legal Issues in 3D Bioprinting - PMC - NIH
    ... human enhancement”[19], the issues of “technological design”[20] youth ... 3D bioprinting taking into account the human rights and dignity. Furthermore ...
  143. [143]
    [PDF] 3D bio-printing for medical and enhancement purposes
    for medical recovery and human enhancement'. The project was ... (7), 863–883. Li, P. H., '3D bioprinting technologies: patents, innovation and access'.
  144. [144]
    Bioethical and Legal Issues in 3D Bioprinting - AccScience Publishing
    The scope of 3D bioprinting includes not only the issues of the advanced ... Yudin B, 2016, Technoscience and “Human Enhancement”. Epistemol Philos Sci ...
  145. [145]
    [PDF] Ethical guidelines for using bioprinting for humans
    In this paper, bioprinting will be unpacked, analysed, and interpreted in the context of bioethics focusing on human beings. 2. Conceptualisation and benefit.
  146. [146]
    a socioethical view of bioprinting human organs and tissues
    In this article, we review the extant social science and ethical literature on three-dimensional (3D) bioprinting.
  147. [147]
    Personalized 3D printed scaffolds: The ethical aspects - ScienceDirect
    Dec 25, 2023 · ... human enhancement which violates the intrinsic limit of human nature ... 3D bioprinting – an ethical, legal and social aspects (ELSA) framework.
  148. [148]
    [PDF] Bioethical and Legal Issues in 3D Bioprinting - Semantic Scholar
    Apr 28, 2020 · This article addresses the ethical and legal issues of bioprinting of artificial human organs. Keywords: Three-dimensional printing, Bioethics, ...
  149. [149]
    [PDF] Human Fabrication: Ethical and Legal Aspects of 3D Bioprinting
    Feb 19, 2024 · 3D bioprinting of human tissues and organs for the ... However, they see unethical the development of projects of human enhancement [15].
  150. [150]
    [PDF] Brain organoids and organoid intelligence from ethical, legal, and ...
    Jan 5, 2024 · This includes considerations of human dignity and the rights of both donors and organoids. An interdisciplinary collaboration is crucial to ...
  151. [151]
  152. [152]
    The Regulatory Challenge of 3D Bioprinting - Taylor & Francis Online
    We consider a range of specific challenges pertaining to 3D bioprinting in regenerative medicine, including classification, risk, standardization and quality ...
  153. [153]
    the role of critical feedstock materials and process parameters
    Sep 16, 2025 · The EMA has not established a regulatory framework for 3D-printed pharmaceuticals, nor has it authorized any 3D-printed product.<|separator|>
  154. [154]
    Bioprinted Constructs in the Regulatory Landscape: Current State ...
    Oct 18, 2025 · The current regulatory status of bioprinting is traced by exploring parallels with existing categories such as 3D printed medical devices, ...
  155. [155]
    Expanding Quality by Design Principles to Support 3D Printed ...
    The main obstacles to bringing 3DP into the mainstream healthcare system are regulatory, technological, and technology-related investment challenges [77,153,154] ...
  156. [156]
    [PDF] Regulatory Hurdles in the Commercialization of 3D Printed ...
    Regulators are challenged with the task of ensuring quality, safety, and efficacy while accommodating the flexible, decentralized, and digital nature of 3D ...
  157. [157]
    The 3D Bioprinting Patent Landscape Takes Shape as IP Leaders ...
    Jul 7, 2016 · 3D bioprinting inventions are originating from all corners of the world. The majority of the inventors of the 950 patents and applications ...Missing: key connecting
  158. [158]
    The Patent Eligibility of 3D Bioprinting: Towards a New Version of ...
    Jan 12, 2022 · It is argued that the patentability of inventions involving 3D bioprinting processes and pre-printing materials, such as bio-ink and scaffold, ...
  159. [159]
    Organovo Files Counterclaims In Patent Lawsuit Brought Against It ...
    Jan 10, 2022 · Organovo is accusing Cellink of infringing several of Organovo's patents, and thus Organovo filed counterclaims to the Delaware suit on Friday.Missing: disputes | Show results with:disputes
  160. [160]
    Bioprinting and Intellectual Property: Challenges, Opportunities, and ...
    Jan 15, 2025 · The study explores three critical areas of concern: IP protection for bioprinting hardware and bioinks, ownership and ethical management of digital files.
  161. [161]
    3D printing, the Maker Movement, IP litigation and legal reform - WIPO
    In the field of bioprinting, Advanced Solutions Life Sciences has sued Biobots Inc. for trademark infringement (Advanced Solutions Life Sciences, LLC v BioBiots ...
  162. [162]
    3D Bioprinting Intellectual Property Issues - Attorney Aaron Hall
    Common disputes in bioprinting intellectual property often revolve around patent infringement and licensing agreements. Conflicts may arise when one entity ...
  163. [163]
    Three-Dimensional Bioprinting of Human Organs and Tissues - NIH
    Sep 7, 2023 · The fundamental technical–biological basis of 3D bioprinting lies in the combination of computer-aided design (CAD) and biological materials to ...
  164. [164]
    3d Bioprinting Market Size, Trends & Forecast, 2025-2032
    Jun 19, 2025 · By Regional Insight: North America is projected to maintain its lead in the global 3D bioprinting market with a 36.7% share in 2025.
  165. [165]
    Three-dimensional Printing in Developing Countries - PMC - NIH
    The advent of 3-dimensional (3D) printing technology has facilitated the creation of customized objects. The lack of regulation in developing countries ...
  166. [166]
    Three-Dimensional Bioprinting of Human Organs and Tissues - MDPI
    Sep 7, 2023 · Access to care, an essential principle in healthcare ethics, is challenged by the possibility of 3D bioprinting becoming a luxury available ...
  167. [167]
  168. [168]
  169. [169]
  170. [170]
  171. [171]
  172. [172]
    Advancements of 3D bioprinting in regenerative medicine
    3D bioprinting has unlocked new possibilities for generating complex and functional tissues and organs. However, one of the greatest challenges lies in ...4.1. Cell Lines · 4.2. Stem Cells · 4.4. Primary Cells
  173. [173]
    Recent Advances in 4D Bioprinting - PubMed
    4D bioprinting has emerged as a powerful technique where the fourth dimension time is incorporated with 3D bioprinting.
  174. [174]
    Advances and future perspectives in 4D bioprinting - PMC - NIH
    Advances in 4D bioprinting will allow the development of smart nanomaterial-based constructs with capability to release biomolecules from them [115,116]. Smart ...
  175. [175]
    4D Printing for Biomedical Applications - 2024 - Wiley Online Library
    Apr 5, 2024 · 4D (bio-)printing endows 3D printed (bio-)materials with multiple functionalities and dynamic properties. 4D printed materials have been ...
  176. [176]
    Cell contractile forces drive spatiotemporal morphogenesis in 4D ...
    Sep 11, 2025 · Recent advances in 4D biofabrication indicate that manipulating CCFs in conjunction with biomaterials holds significant potential for generating ...
  177. [177]
    4D printing polymeric biomaterials for adaptive tissue regeneration
    This review aims to comprehensively summarize the principles and research progress of 4D printing polymeric biomaterials for adaptive tissue regeneration.
  178. [178]
    4D bioprinting of programmed dynamic tissues - ScienceDirect
    The goal of this review is to offer useful insights into the latest development of the 4D bioprinting technology and its wider future biomedical application in ...
  179. [179]
    Multimaterial and Multidimensional Bioprinting in Regenerative ...
    Jun 4, 2025 · This review outlines the principles and progress of multimaterial and multidimensional bioprinting, from 3D to 4D, 5D, and emerging 6D ...Introduction · Characteristics of Multimaterial... · Progresses and Challenges in...
  180. [180]
    Applications and Recent Advances in 3D Bioprinting Sustainable ...
    This article reviews the characteristics and applications of different biomaterials printed by 3D technology, such as gelatin, sodium alginate, and starch, ...
  181. [181]
    Recent Advances in Polymer 4D Printing: 3D Printing Techniques ...
    Sep 9, 2025 · This review provides a comprehensive overview of recent advancements in the 4D printing of polymer composites, emphasizing three pivotal areas: ...
  182. [182]
    3D Bioprinting Market Size & Share Analysis Report, 2030
    The global 3D bioprinting market size was valued at USD 2.0 billion in 2022 and is projected to reach USD 5.3 billion by 2030, growing at a CAGR of 12.5% ...
  183. [183]
    3D Bioprinting Market Growth, Size and Industry Trends 2030
    Jul 1, 2025 · The 3D Bioprinting Market is expected to reach USD 1.67 billion in 2025 and grow at a CAGR of 15.89% to reach USD 3.49 billion by 2030.