Fact-checked by Grok 2 weeks ago

Biomolecular engineering

Biomolecular engineering is the application of principles to the purposeful of biomolecules, including peptides, proteins, nucleic acids, and lipids, to design and construct systems with desired functions. This interdisciplinary field operates at the interface of and , emphasizing molecular-level research to understand, modify, and exploit biological processes. Coined in 1992 by the , it encompasses techniques such as technology, polymerase chain reaction (PCR), , and to enable precise control over biomolecular structures and interactions. Key applications include the development of therapeutic proteins, biosensors, and , with significant achievements like engineered enzymes for industrial catalysis and tools that enhance precision in genetic modifications. Despite its promise, challenges persist in scaling production, ensuring stability of engineered molecules, and addressing potential off-target effects in complex biological systems.

Definition and Scope

Core Principles and Objectives

Biomolecular engineering applies core engineering disciplines—such as , , , and reactor design—to the , modification, and of biological molecules and systems, emphasizing molecular-level interventions at the of and . This approach integrates principles from biochemistry, , and computational modeling to predict and control outcomes from molecular interactions to macroscopic scales, enabling the rational of biomolecules like proteins and nucleic acids with expanded or novel functions. Fundamental to the field is the use of both natural and unnatural building blocks, alongside techniques like and genetically encoded production, to achieve precise functional outcomes, such as enzyme efficiencies with kcat/KM values approaching 105 M-1s-1. The primary objectives center on engineering functional macromolecules and pathways to address challenges in , , and environmental , including the development of therapeutics, vaccines, and cell-based therapies through approaches. For instance, scaling biological processes for industrial production—exemplified by penicillin yield optimizations in the 1940s and modern manufacturing—aims to enhance efficiency, reliability, and productivity while optimizing reaction conditions and product recovery. Broader goals encompass creating biosensors, biofuels, biomaterials, and solutions, with a focus on modeling complex systems like the and designing circuits that rival natural regulatory dynamics. These principles and objectives prioritize verifiable, scalable outcomes over empirical trial-and-error, drawing on quantitative tools to bridge molecular design with real-world applications, as formalized in the Institutes of Health's 1992 definition of the field as molecular-focused research at the chemical engineering-biology nexus.

Distinctions from Adjacent Fields

Biomolecular engineering distinguishes itself from primarily through its engineering-centric approach, focusing on the rational design, synthesis, and optimization of biomolecules such as proteins and nucleic acids for targeted applications, rather than the observational study of natural molecular mechanisms and interactions. , rooted in techniques like and sequencing established in the mid-20th century, prioritizes elucidation of endogenous biological processes, whereas biomolecular engineering integrates computational modeling, , and to create non-natural variants with enhanced functions, such as enzymes with altered substrate specificity. This purposeful manipulation enables applications in therapeutics and materials, contrasting with 's foundational role in discovery without inherent design imperatives. In contrast to biochemical engineering, which emphasizes bioprocess development—including fermentation, bioreactor design, and downstream purification for large-scale production of biological products—biomolecular engineering operates at the molecular level, engineering the intrinsic properties of biomolecules themselves before process integration. For instance, while biochemical engineering scales microbial cultures for insulin production, biomolecular engineering might redesign the insulin protein sequence for improved stability or half-life using site-directed mutagenesis. The two fields overlap in academic departments often titled "Chemical and Biomolecular Engineering," but biomolecular efforts prioritize de novo molecular architectures over process optimization. Relative to , biomolecular engineering is narrower, concentrating on individual or small assemblies of rather than the construction of higher-order systems like genetic circuits or metabolic pathways in whole cells. , emerging prominently after 2000 with milestones like the first synthetic genome in , builds modular biological parts into functional networks, often drawing on biomolecular tools but extending to emergent system behaviors. Bioengineering, a broader umbrella encompassing scales from molecules to organs, further differentiates by including non-molecular interventions such as scaffolds or diagnostic devices, whereas biomolecular engineering remains tethered to atomic-level precision in biomolecule redesign.

Historical Development

Pre-1970s Foundations in Molecular Biology

The foundations of molecular biology prior to the 1970s emerged from biochemical and genetic studies that linked genes to specific molecular functions, establishing the framework for understanding biomolecular information transfer. In 1941, and Edward Tatum conducted experiments with the bread mold , irradiating spores to induce mutations and observing that certain mutants required specific nutrients for growth, indicating that each affected gene disrupted a single enzymatic step in a . This led to their "one gene–one enzyme" hypothesis, proposing that genes direct the production of individual enzymes, a concept later refined to "one gene–one polypeptide" as protein subunit structures were elucidated. Their work shifted biological inquiry toward molecular mechanisms of , demonstrating causal links between genetic mutations and biochemical deficiencies. By the mid-1940s, evidence accumulated that deoxyribonucleic acid (DNA), rather than proteins, served as the primary genetic material. In 1944, Oswald Avery, Colin MacLeod, and Maclyn McCarty showed that purified DNA from virulent Streptococcus pneumoniae could transform non-virulent strains into virulent ones, resistant to enzymatic degradation of proteins or RNA but sensitive to DNA-degrading enzymes, thus identifying DNA as the "transforming principle" carrying heritable traits. This finding challenged prevailing protein-centric views. Complementing this, Alfred Hershey and Martha Chase's 1952 experiments with bacteriophage T2 infecting Escherichia coli used radioactive isotopes—phosphorus-32 for DNA and sulfur-35 for proteins—to track viral components; only the DNA-labeled phages transmitted genetic instructions into host cells, producing progeny viruses, while protein coats remained external. These results conclusively affirmed DNA's role in heredity across organisms. The molecular structure of DNA was revealed in 1953 by James Watson and Francis Crick, who proposed a double-helical model based on X-ray diffraction data from Rosalind Franklin and Maurice Wilkins, featuring two antiparallel polynucleotide strands twisted around a common axis, stabilized by hydrogen bonds between complementary base pairs (adenine-thymine and guanine-cytosine). This configuration explained DNA's capacity for self-replication and information storage, with the sequence of bases encoding genetic instructions. In 1958, Crick formalized the "central dogma" of molecular biology, positing unidirectional information flow from DNA to messenger RNA (mRNA) via transcription, then to proteins via translation, excluding reverse transfer from proteins to nucleic acids under normal cellular conditions. This paradigm provided a causal model for gene expression, essential for later engineering efforts. Deciphering the further illuminated protein synthesis mechanisms. In 1961, Marshall Nirenberg and J. Heinrich Matthaei used a with synthetic polyuridylic acid (poly-U) , which directed incorporation of into polypeptides, revealing that the triplet UUU codes for —the first codon identified. Subsequent experiments mapped additional codons, confirming a non-overlapping, degenerate triplet code read in a 5' to 3' direction without commas, universal across species. These pre-1970s advances established the molecular basis for manipulating genetic information, setting the stage for biomolecular engineering by revealing DNA's sequence-specific functionality and protein-coding logic.

Recombinant DNA Era (1970s-1990s)

The recombinant DNA era, from the 1970s to 1990s, revolutionized biomolecular engineering by providing tools to isolate, manipulate, and express genes from diverse sources in host organisms, enabling the production of specific proteins and other biomolecules on an industrial scale. This period built on the discovery of type II restriction endonucleases, which cut DNA at precise recognition sequences; the first such enzyme, HindII, was isolated in 1970 by Hamilton O. Smith at Johns Hopkins University. Complementary advancements in DNA ligase allowed the sealing of cleaved DNA fragments, facilitating the construction of hybrid molecules. Werner Arber, Hamilton Smith, and Daniel Nathans were awarded the 1978 Nobel Prize in Physiology or Medicine for elucidating the restriction-modification systems underlying these enzymes. In 1972, Paul Berg at Stanford University created the first recombinant DNA molecule in vitro, ligating DNA from simian virus 40 (SV40) with lambda phage DNA, demonstrating the potential for cross-species gene transfer despite not yet achieving replication in cells. The pivotal advance occurred in 1973 when Herbert Boyer at the University of California, San Francisco, and Stanley Cohen at Stanford collaborated to clone recombinant plasmids in Escherichia coli, inserting a resistance gene from one plasmid into another using EcoRI restriction enzyme and achieving stable propagation and expression in bacterial hosts. These experiments established plasmid-based cloning vectors as a core technique in biomolecular engineering, allowing scalable amplification of genetic constructs. Emerging safety concerns over unintended ecological or pathological risks led to the 1975 Asilomar Conference, organized by and attended by over 140 scientists, which recommended containment levels and a voluntary moratorium on high-risk experiments. This informed the 1976 (NIH) guidelines, which classified experiments by risk and mandated physical and biological containment, enabling regulated progress. The era spurred the biotechnology industry; , co-founded in 1976 by Boyer and Robert Swanson, produced the first recombinant human insulin in E. coli in 1978 through gene synthesis and expression of A and B chains. FDA approval of Humulin in 1982 marked the first recombinant pharmaceutical, supplanting animal-derived insulin and demonstrating scalability for therapeutic protein production. Advancements in the 1980s enhanced precision and efficiency; invented the () in 1983 at , a cyclic enzymatic process using thermostable to amplify specific DNA sequences exponentially, transforming gene isolation for and engineering. shared the 1993 for , which reduced reliance on cumbersome library screening. By the 1990s, recombinant methods extended to eukaryotic hosts like yeast () and Chinese hamster ovary cells, addressing post-translational modifications absent in bacteria, as seen in recombinant human growth hormone (approved 1985) and monoclonal antibodies. These developments solidified as the cornerstone of biomolecular engineering, yielding over 200 recombinant therapeutics by decade's end and paving the way for genomics-era innovations.

Post-Genomics Advances (2000s-Present)

The completion of the Human Genome Project in 2003 marked the transition to the post-genomics era, emphasizing functional annotation of genomes, systems-level interactions, and the engineering of biomolecular pathways rather than mere sequencing. This shift was facilitated by next-generation sequencing technologies introduced in the mid-2000s, which reduced DNA sequencing costs by orders of magnitude—from approximately $10 million per human genome in 2001 to under $1,000 by 2015—enabling comprehensive transcriptomic, epigenomic, and proteomic analyses. These tools underpinned biomolecular engineering by providing data for modeling regulatory networks and predicting molecular behaviors, moving beyond descriptive genomics to causal interventions in cellular processes. A pivotal advance was the development of CRISPR-Cas9 gene editing in 2012, derived from bacterial adaptive immunity systems and adapted for programmable DNA cleavage by and Emmanuelle Charpentier.00111-9) This technology enabled precise, multiplexed modifications in eukaryotic genomes with efficiencies far surpassing prior methods like zinc-finger nucleases or TALENs, facilitating applications in rewiring for production and therapeutic protein expression. By 2018, had been integrated into microbial engineering for enhanced biomolecule synthesis, such as in for high-value compounds, though off-target effects and delivery challenges persisted, prompting refinements like base editing and by 2019. Its impact extended to causal realism in engineering, allowing direct testing of genotype-phenotype relationships without reliance on indirect selection. Synthetic biology matured in this period as a discipline for designing de novo genetic circuits and organisms, with milestones including the 2010 creation of minimal bacterial genomes and the engineering of yeast for opioid production by 2015. Advances in DNA synthesis and assembly techniques, such as Gibson assembly refinements post-2009, enabled the construction of synthetic chromosomes, exemplified by the Sc2.0 project starting in 2009, which replaced yeast chromosomes with redesigned versions lacking transposons and repetitive elements. These developments supported scalable biomolecular factories, including nitrogen-fixing crops via engineered microbes by the late 2010s, grounded in empirical pathway optimization rather than unverified models. Protein engineering progressed through computational and evolutionary methods, culminating in AI-driven structure prediction with DeepMind's in 2021, which achieved atomic-level accuracy for over 200 million protein structures, solving the decades-old folding problem. This enabled de novo design of functional proteins, such as novel binders and enzymes, inverting traditional structure prediction for inverse folding tasks by 2023. , refined since the 1990s, combined with rational design via tools like , yielded enzymes for non-natural reactions, including carbon fixation enhancements. By 2024, 3 extended predictions to protein-ligand and interactions, accelerating and synthetic biocatalysts while highlighting limitations in dynamics and non-protein complexes. These tools underscore a paradigm of predictive engineering, prioritizing verifiable stability and activity metrics over speculative functions.

Fundamental Biomolecules

Nucleic Acids

Nucleic acids are linear biopolymers composed of nucleotide monomers, each consisting of a nitrogenous base, a pentose sugar, and one to three phosphate groups linked via phosphodiester bonds. The two primary types are deoxyribonucleic acid (DNA), which contains deoxyribose sugar and the bases adenine (A), thymine (T), guanine (G), and cytosine (C), and ribonucleic acid (RNA), which contains ribose sugar and the bases A, uracil (U), G, and C. These molecules encode and transmit genetic information essential for cellular function, replication, and protein synthesis, forming the foundational substrate for biomolecular engineering applications such as gene synthesis and pathway redesign. DNA typically exists as a right-handed double helix with antiparallel strands held together by hydrogen bonds between complementary base pairs (A-T and G-C), enabling stable storage of genetic blueprints in chromosomes. This structure, elucidated through data analyzed by and in 1953, supports semi-conservative replication where each strand serves as a template for new synthesis via enzymes, ensuring fidelity across generations with error rates below 1 in 10^9 base pairs in eukaryotes. In biomolecular engineering, DNA's predictable base-pairing and modularity allow for the construction of recombinant plasmids—circular DNA vectors exceeding 10 kilobases—that facilitate heterologous in host cells. RNA, often single-stranded, folds into complex secondary structures via intramolecular base pairing, performing diverse roles including messenger RNA (mRNA) for transcribing DNA into protein-coding sequences, transfer RNA (tRNA) for amino acid delivery during translation, and ribosomal RNA (rRNA) as the catalytic core of ribosomes. Transcription from DNA templates by RNA polymerase II produces pre-mRNA, which undergoes splicing to remove introns—non-coding regions comprising up to 95% of human genes—yielding mature mRNA for export to the cytoplasm. Engineered RNAs, such as synthetic aptamers or siRNAs, exploit these folding properties for targeted regulation, with applications in RNA interference achieving over 90% knockdown efficiency in mammalian cells.

Proteins

Proteins consist of one or more polypeptide chains formed by the covalent linkage of via bonds, with the primary structure defined by the specific of up to 20 standard that dictates all higher-order folding and . This emerges from translation of codons during ribosomal synthesis, enabling vast combinatorial diversity estimated at over 10^130 possible sequences for a 100-residue protein, though constrains functional variants. The functional conformation arises through secondary structures, such as α-helices and β-sheets stabilized by hydrogen bonding between backbone atoms; tertiary structure, involving hydrophobic collapse, ionic interactions, hydrogen bonds, and disulfide bridges to form compact globular or fibrous domains; and quaternary structure for multisubunit assemblies like , which comprises four chains. Denaturation disrupts these noncovalent interactions, often reversibly for some proteins, underscoring the causal link between precise architecture and , as misfolding underlies diseases like Alzheimer's via aggregates. In biomolecular engineering, proteins serve as primary effectors due to their evolvability and modularity, functioning as enzymes with rate accelerations up to 10^20-fold over uncatalyzed reactions, structural scaffolds, transporters, and signaling effectors. Engineering exploits this by altering sequences to enhance stability, specificity, or novelty, as in designing thermostable enzymes operational at 90°C or de novo proteins folding into predefined topologies, addressing limitations of natural variants in industrial biocatalysis and therapeutics. Such modifications, grounded in structure-function causality, have yielded variants like improved insulin analogs with prolonged half-lives via PEGylation, demonstrating empirical gains over native forms without relying on unsubstantiated assumptions of inherent optimality.

Carbohydrates and Lipids

In biomolecular engineering, carbohydrates are modified through glycoengineering techniques to alter structures on proteins or cells, enabling applications in therapeutics and diagnostics. Metabolic glycoengineering introduces analogs into cellular pathways, allowing precise control over glycan composition without genetic alterations to glycosyltransferases; this approach, developed over the past three decades, has facilitated the creation of glycoproteins with tailored immune-modulating properties. For instance, bacterial systems have been engineered for protein , with recent advances in integrating metabolic pathways to produce human-like glycans, enhancing recombinant protein functionality in biopharmaceuticals. Automated chemical synthesis represents another key method for carbohydrate engineering, pioneered by Peter Seeberger in 2001 and refined over 25 years to enable scalable production of complex oligosaccharides for development and targeting. Carbohydrate-binding modules (CBMs), non-catalytic domains appended to enzymes, are molecularly engineered to improve specificity in degradation; and rational design have yielded variants with enhanced affinity for recalcitrant like , supporting production from lignocellulosic feedstocks. These efforts underscore causal links between structure and biological recognition, prioritizing empirical optimization over assumed natural fidelity. Lipids in biomolecular engineering are primarily targeted for biosynthesis pathway redesign to produce biofuels, nanomaterials, and membrane mimics. Metabolic engineering of oleaginous yeasts, such as Yarrowia lipolytica, has reprogrammed fatty acid metabolism to yield high levels of oleic acid; in one 2022 study, pathway flux redirection increased lipid titers by balancing redox cofactors and eliminating competing sinks, achieving sustainable production of edible oils and biodiesels. Synthetic biology tools enable de novo assembly of lipid pathways in heterologous hosts like Escherichia coli, where overexpression of archaeal synthases boosted ether lipid yields by 2024, demonstrating improved membrane stability under industrial stress. Membrane lipid engineering diversifies to enhance cellular resilience in bioprocessing; for example, altering expression in microbes tunes , mitigating toxicity from hydrophobic products like biofuels, as shown in 2018 optimizations that preserved respiratory efficiency. nanoparticles' biomolecular s—protein layers formed in biological fluids—are precisely engineered via surface modifications to evade immune clearance, with 2024 studies reporting accelerated theranostic delivery through corona control. These interventions rely on first-principles flux analysis to causally link chain length, unsaturation, and headgroup chemistry to physicochemical properties, bypassing biases in traditional screening.

Engineering Techniques

Genetic Manipulation Methods

Genetic manipulation methods constitute core techniques in biomolecular engineering for altering DNA sequences to produce engineered biomolecules, such as modified proteins or novel metabolic pathways. These approaches leverage enzymes like restriction endonucleases for precise DNA cleavage, DNA ligases for joining fragments, and polymerases for amplification, enabling the construction and propagation of hybrid genetic material in host organisms. Originating in the 1970s, these methods have progressed from basic cloning to high-precision editing, underpinning industrial-scale protein production and therapeutic development. Recombinant DNA technology, the foundational method, involves isolating a of interest, inserting it into a vector such as a using compatible restriction sites, and transforming it into a bacterial host like for replication and expression. This technique, first demonstrated in by combining SV40 viral DNA with , allows scalable production of heterologous proteins by exploiting host cellular machinery. enables targeted alterations to specific within a , typically via amplification using primers incorporating the desired , followed by template degradation and . Introduced in the 1980s, variants like QuikChange use circularized plasmids and DpnI to select mutated strands, achieving efficiencies over 80% for single-site changes and facilitating for enhanced stability or activity. CRISPR-Cas systems, adapted from bacterial adaptive immunity discovered in 2007, utilize to direct endonuclease for sequence-specific double-strand breaks, repaired via or to introduce insertions, deletions, or substitutions. Since the 2012 demonstration of CRISPR-Cas9 in eukaryotic cells, refinements have improved specificity, reducing off-target effects to below 1% in optimized protocols, and expanded to base editing and for scarless modifications.00111-9)

Recombinant DNA Technology

Recombinant DNA technology encompasses the laboratory manipulation of genetic material to produce hybrid DNA molecules by joining segments from disparate organisms, which are subsequently introduced into a host cell for replication and expression. This process relies on enzymes such as restriction endonucleases to cleave DNA at precise recognition sites, generating compatible "sticky ends" that facilitate ligation with DNA ligase to form stable recombinant constructs. Vectors, typically bacterial plasmids—small, extrachromosomal, self-replicating DNA circles—serve as carriers for the inserted foreign DNA, enabling its propagation within compatible hosts like Escherichia coli. The foundational experiments occurred in the early 1970s. In 1972, at created the initial by covalently linking DNA from the virus to DNA using the , though it was not yet cloned in a living cell. Building on this, in 1973, at the , and Stanley Cohen at Stanford successfully cloned in bacteria: they inserted a gene for kanamycin resistance from one into another using , ligated the fragments, and transformed E. coli cells, which then expressed the hybrid molecule and conferred antibiotic resistance. These milestones demonstrated that foreign DNA could be stably maintained and replicated in prokaryotic hosts, laying the groundwork for . Core methods involve several sequential steps. First, the gene of interest is isolated from source DNA via restriction digestion, producing fragments with cohesive ends. The vector is similarly prepared by cutting at a multiple cloning site within a polylinker region, often flanked by selectable markers like antibiotic resistance genes for host screening. Ligation mixes the fragments with DNA ligase, which catalyzes phosphodiester bond formation under controlled conditions to favor circular recombinant plasmids over non-productive multimers. Transformation introduces these plasmids into competent bacterial cells, typically via heat shock or electroporation, followed by selection on media containing the marker antibiotic to isolate successfully transformed colonies harboring the recombinant DNA. Restriction enzymes, discovered in the mid-1960s through studies of bacteriophage restriction in bacteria, were pivotal; type II enzymes like EcoRI, isolated from E. coli, cut DNA predictably outside their palindromic recognition sequences (e.g., GAATTC), enabling precise excision and insertion. This specificity arose from bacterial defense mechanisms against viral invasion, where the enzymes cleave unmodified foreign DNA while host DNA is protected by methylation. Over 3,000 such enzymes have since been characterized, with hundreds commercially available, allowing versatile fragment generation for cloning. Early concerns about biohazards prompted the 1975 Asilomar Conference, where and others advocated containment guidelines, influencing safe practices that enabled widespread adoption without verified ecological risks from contained lab strains. By the late , the technology facilitated the first commercial product: human insulin produced in , marking its transition from research tool to industrial application.

Site-Directed Mutagenesis

Site-directed mutagenesis (SDM) is a molecular biology technique used to introduce precise, targeted alterations into a specific DNA sequence, enabling the study of gene function and protein structure-activity relationships. This method allows researchers to substitute, insert, or delete nucleotides at predetermined sites within a plasmid or gene, facilitating controlled changes in the resulting protein. Unlike random mutagenesis, SDM provides high specificity, minimizing unintended mutations. The foundational approach was developed by Michael Smith in the 1970s, utilizing synthetic oligonucleotides to hybridize with single-stranded DNA templates, followed by enzymatic extension and ligation to incorporate mutations. Smith's oligonucleotide-directed method, recognized with the 1993 Nobel Prize in Chemistry (shared with Kary Mullis), enabled the reprogramming of genetic codes to replace specific amino acids in proteins. Early implementations involved M13 bacteriophage vectors for single-stranded DNA production, with mutagenesis efficiency improved by selecting for mismatch repair-deficient host strains. Modern SDM predominantly employs ()-based protocols, which amplify the target using primers incorporating the desired . Common variants include the QuikChange method, where fully overlapping primers extend the entire , followed by DpnI digestion to eliminate methylated parental DNA, yielding mutation efficiencies exceeding 80% for single substitutions. Overlap extension uses partially overlapping primers flanking the site, generating megoprimers for a secondary to assemble the full mutated fragment before . High-fidelity polymerases, such as Pfu or Phusion, minimize errors, while whole-plasmid mutagenesis supports changes in large constructs up to 10 kb. These techniques have evolved to handle multiple simultaneous s via iterative or multiplex primer designs. In biomolecular engineering, SDM is pivotal for , enabling rational design of enzymes with enhanced stability, altered substrate specificity, or improved catalytic efficiency. For instance, site-specific replacements have been used to dissect active sites in proteases, increasing for industrial applications. It complements by generating focused libraries for at key residues, accelerating variant screening. Applications extend to therapeutic protein development, such as modifying antibodies for better or reducing . Overall, SDM's precision has underpinned advances in understanding , binding, and evolutionary mechanisms.

CRISPR-Cas and Advanced Gene Editing

The CRISPR-Cas system, originally identified as an adaptive immune mechanism in bacteria and archaea, enables precise DNA cleavage guided by RNA. Clustered regularly interspaced short palindromic repeats (CRISPR) arrays store spacer sequences derived from invading phages or plasmids, which are transcribed into CRISPR RNA (crRNA) that complexes with CRISPR-associated (Cas) proteins to target and degrade matching foreign DNA. The Type II CRISPR-Cas9 system from Streptococcus pyogenes, featuring the Cas9 endonuclease, was repurposed for eukaryotic genome engineering in 2012 by fusing crRNA with trans-activating crRNA (tracrRNA) into a single guide RNA (sgRNA) to direct programmable double-strand breaks (DSBs) at specific genomic loci complementary to the sgRNA, adjacent to a protospacer adjacent motif (PAM). This adaptation revolutionized genetic manipulation by simplifying targeting compared to prior tools like zinc-finger nucleases (ZFNs) or transcription activator-like effector nucleases (TALENs), which required protein engineering for each locus. In biomolecular engineering, CRISPR-Cas9 facilitates targeted gene knockouts via (NHEJ)-induced insertions/deletions (indels), insertions through (HDR) using donor templates, and multiplexed edits by deploying multiple sgRNAs. Applications include metabolic pathway optimization in microbes for or pharmaceutical production, such as enhancing Escherichia coli yields of isoprenoids by disrupting competing pathways, and human cell line engineering for protein expression platforms. Delivery methods encompass viral vectors (e.g., AAV), , or ribonucleoprotein complexes, with efficiencies varying by ; for instance, Cas9 RNP transfection achieves up to 80% editing in primary T cells. Off-target effects, arising from sgRNA mismatches, have been quantified via genome-wide unbiased assays showing rates as low as 0.1-1% with optimized guides, though structural variations like large deletions can occur at DSB sites. Advanced variants address CRISPR-Cas9 limitations, such as reliance on DSBs that trigger error-prone repairs. Base editors fuse catalytically dead (dCas9) or nickase (nCas9) with deaminases to enable single-base conversions without DSBs; base editors (CBEs), introduced in 2016, convert C•G to T•A with efficiencies exceeding 50% in mammalian cells and bystander editing minimized in later iterations like BE4. base editors (ABEs) achieve A•T to G•C changes similarly. , developed in 2019, uses a fused to nCas9 and a prime editing guide RNA (pegRNA) encoding the edit, enabling insertions, deletions, and all base transitions/transversions with up to 20% efficiency and reduced indels. These tools expand engineering precision for applications like correcting disease mutations or installing synthetic codons in biosynthetic gene clusters. Emerging Cas variants, such as Cas12a for AT-rich PAMs or smaller Cas13 for , further diversify the toolkit, though delivery challenges and immune responses limit use.

Protein and Enzyme Engineering

Protein engineering encompasses the deliberate modification of sequences in proteins to enhance or introduce specific functions, such as improved stability, altered substrate specificity, or novel catalytic activities. This field leverages techniques like , which allows precise alterations at targeted residues based on structural knowledge, and , which mimics through iterative cycles of random and high-throughput screening or selection.00064-8) Enzyme engineering, a specialized application, focuses on optimizing biocatalysts for industrial or therapeutic use by engineering properties like and pH tolerance.00518-8.pdf) Early milestones include the development of technology in the 1970s, enabling the production of human insulin in by in 1978, with FDA approval in 1982 as the first therapeutic recombinant protein. emerged in the late 1970s, allowing rational changes informed by and computational modeling. advanced in the 1990s with methods like error-prone PCR and , introduced by Willem Stemmer in 1994, enabling rapid optimization without prior structural data. Contemporary approaches integrate rational design with computational tools, including models that predict fitness landscapes from sequence data, accelerating variant screening. For instance, high-throughput platforms now generate libraries exceeding 10^6 variants, screened via droplet or FACS for functions like enhanced protease resistance.00184-9)00064-8) In engineering, semi-rational methods combine structural insights with focused libraries, as seen in redesigning lipases for formulations with improved activity at low temperatures. Recent AI-driven strategies, such as generative models, have enabled de novo design for non-natural reactions, with successes reported in 2023-2024 for . Biomedical applications include engineered monoclonal antibodies with extended half-lives via Fc modifications, as in the 2021 redesigns for immunogens, and therapeutic enzymes like variants with reduced . Industrially, engineered enzymes dominate markets: cellulases for production have yields improved 10-fold via since 2010, while PETases for plastic degradation achieved 200-fold activity boosts by 2020 through iterative engineering. These advancements underscore causal links between sequence alterations and functional outcomes, validated empirically through kinetic assays and structural validations, though challenges persist in scaling predictions for complex multidomain proteins.

Synthetic Biology and Pathway Design

Synthetic biology represents an interdisciplinary approach within biomolecular engineering that applies engineering principles to the design and construction of novel biological systems, including genetic circuits, enzymes, and metabolic pathways, to achieve functions not found in nature. This field emerged prominently in the early , building on advances in and assembly techniques to enable the de novo creation of biological parts and devices. Unlike traditional , which modifies existing genes, synthetic biology emphasizes standardization, modularity, and abstraction, treating biological components as interchangeable modules akin to electronic circuits. Central to synthetic biology is the engineering of metabolic pathways, where computational tools predict and optimize sequences of enzymatic reactions to produce target molecules such as pharmaceuticals, biofuels, or fine chemicals in host organisms like or . Pathway design typically begins with , decomposing complex products into precursors and identifying heterologous enzymes from diverse sources to assemble non-native routes, often minimizing competition with host metabolism. Tools like Galaxy-SynBioCAD facilitate automated design-build-test-learn (DBTL) cycles, integrating genome-scale models with high-throughput assembly methods such as or to prototype pathways rapidly. Machine learning algorithms further enhance prediction of pathway performance, estimating titers, rates, and yields by training on kinetic data from prior experiments. Recent advances from 2020 onward have leveraged to accelerate pathway optimization, with methods like SubNetX exploring vast reaction networks to identify efficient routes for synthesizing complex compounds, reducing manual trial-and-error. For instance, AI-driven retrosynthesis has enabled the engineering of microbial strains producing plant-derived therapeutics, bypassing limitations of native plant extraction. These developments address thermodynamic and kinetic bottlenecks through of enzymes and regulatory networks, achieving yields up to grams per liter in industrial chassis. However, challenges persist in , including of intermediates and cofactor imbalances, necessitating hybrid approaches combining with modeling.

Amplification and Detection Techniques

Polymerase chain reaction (PCR) serves as the foundational technique for nucleic acid amplification in biomolecular engineering, enabling the exponential replication of targeted DNA segments from trace quantities to billions of copies through iterative cycles of denaturation at approximately 95°C, primer annealing at 50-60°C, and extension at 72°C using thermostable Taq DNA polymerase isolated from Thermus aquaticus. This process, patented in 1987 and awarded the Nobel Prize in Chemistry to Kary Mullis in 1993, underpins genetic manipulation by providing sufficient material for cloning, sequencing, and functional studies of engineered constructs. PCR's specificity arises from oligonucleotide primers flanking the region of interest, with amplification efficiency typically yielding 1.8-2.0-fold increase per cycle, resulting in 2^n copies after n cycles, often 25-40 for practical yields exceeding 10^9-fold. Quantitative real-time (qPCR) extends by incorporating fluorescent intercalating dyes like SYBR Green or sequence-specific probes, allowing continuous monitoring of amplicon accumulation via intensity proportional to double-stranded DNA formation, thus enabling absolute or relative quantification with detection limits as low as 10-100 target molecules. (dPCR) partitions samples into thousands of microreactions for Poisson-distributed , providing precise absolute quantification without standard curves, particularly useful for rare variant detection in engineered populations or assessment in synthetic genomes. These cycling methods require specialized thermocyclers but offer high fidelity when using polymerases like Pfu, minimizing errors at rates below 10^-6 per . Isothermal amplification techniques circumvent thermal cycling needs, facilitating integration into portable devices for on-site validation of biomolecular assemblies. (LAMP), developed in 2000, employs 4-6 primers targeting six distinct sequences to form cauliflower-like structures amplified by Bst at 60-65°C, achieving 10^9-fold in under 60 minutes with high tolerance to inhibitors. Rolling circle amplification (RCA) utilizes phi29 to continuously extend primers annealed to circular templates, producing long concatemeric products for sensitive detection of circular DNAs common in plasmids or viroids. Nucleic acid sequence-based amplification (NASBA) targets via a transcription-mediated process involving T7 , yielding 10^12 amplicons in 90 minutes, ideal for quantifying mRNA from engineered expression systems without reverse transcription biases. Detection of amplified biomolecules integrates spectroscopic, electrophoretic, and enzymatic readouts to verify engineering outcomes. Agarose gel electrophoresis separates amplicons by size under electric fields, with ethidium bromide or SYBR Safe staining enabling visualization under UV light at sensitivities of 1-10 ng DNA, though safer alternatives like pulsed-field methods handle larger constructs. Fluorescence-based detection in qPCR or microarray hybridization quantifies products via melting curve analysis to confirm specificity, distinguishing intended targets from non-specific artifacts. CRISPR-Cas diagnostics amplify signals through collateral cleavage: Cas12a or Cas13a, activated by guide RNA-bound targets post-RPA or LAMP pre-amplification, cleave fluorophore-quencher reporters, yielding detectable fluorescence increases up to 1000-fold within 30-60 minutes at limits of detection rivaling qPCR (1-10 copies/μL). These methods, validated in synthetic biology for rapid screening of genetic circuits, prioritize empirical sensitivity over assumptions of universality, with CRISPR approaches showing lower off-target rates in engineered contexts when paired with high-fidelity amplification.

Applications

Industrial and Bioprocess Engineering

Industrial and bioprocess engineering applies principles of biomolecular engineering to design, optimize, and scale biological production systems for commercial output of proteins, enzymes, and biofuels. This discipline integrates upstream or with downstream purification to maximize yields while minimizing costs and environmental impact, often using microbial hosts like or for recombinant protein expression. Upstream bioprocessing emphasizes engineering for controlled environments that support high-density cultures of genetically modified organisms. Fed-batch strategies, predominant in mammalian cell lines such as ovary (CHO) cells, have driven recombinant protein titers beyond 5-10 g/L by dynamically supplying nutrients to avoid inhibition, as demonstrated in production since the early . In microbial systems, advances in enhance flux through engineered pathways, with E. coli strains achieving over 50% of soluble protein as recombinant targets through codon optimization and chaperone co-expression. Oxygen transfer, control, and mitigation remain critical, addressed via single-use s scaling to 20,000 liters for cost-effective operation. Downstream processing recovers and purifies biomolecules, typically comprising 50-80% of production costs due to challenges in separating target products from host impurities. Techniques like , , and exploit engineered for purification, with process intensification via continuous flow reducing batch times by up to 30% in recent optimizations. For , such as lipases used in synthesis, engineered variants from or species enable of triglycerides at yields exceeding 90%, supporting annual global production volumes over 10,000 tons. Key industrial products include recombinant therapeutics like insulin, first commercially scaled in in , now yielding billions of doses annually via bioreactors. Engineered dominate applications in detergents (e.g., variants stable at high pH) and textiles, where engineering has cut energy use in by 20-30% since 2000. In biofuels, metabolically engineered yeasts convert to bioethanol at titers of 100 g/L, with cocktails like cellulases achieving 80-90% efficiency in pilot plants. Scaling challenges persist, including heterogeneous mixing in large bioreactors leading to gradients in dissolved oxygen and metabolite concentrations, which can reduce yields by 20-50% without modeling. Filter fouling in systems and viral clearance validation add regulatory hurdles, though machine learning-driven controls have improved predictability, cutting development timelines from years to months in strains for surface protein overproduction. Sustainability efforts focus on , with integrated bioprocesses byproducts for co-production of chemicals alongside biofuels.

Biomedical and Therapeutic Uses

Biomolecular engineering has enabled the production of recombinant protein therapeutics by inserting genes encoding human proteins into host organisms like or , allowing scalable manufacturing. Human insulin (Humulin), the first such therapeutic, was approved by the FDA in 1982 after engineering the insulin gene into bacteria, replacing animal-derived insulin and reducing immunogenicity risks. Over 400 recombinant proteins have been approved worldwide by 2016, including growth hormones, clotting factors, and cytokines, with protein-based drugs comprising a significant portion of the market. Engineered monoclonal antibodies, modified via techniques like for enhanced affinity and stability, dominate . Rituximab, a chimeric approved in 1997, targets B-cell lymphomas by recruiting immune effectors, demonstrating how improves specificity over native . Chimeric antigen receptor (CAR) T-cell therapies represent advanced cell engineering, where patient T cells are genetically modified to express synthetic receptors targeting tumor , as in (Kymriah), FDA-approved in 2017 for . principles underpin CAR design, incorporating modular domains for and safety switches to mitigate . Gene therapies leverage engineered viral vectors or non-viral systems to deliver corrective nucleic acids. (Luxturna), approved in 2017, uses an (AAV) vector to express the in retinal cells, restoring vision in inherited patients. (Zolgensma), approved in 2019 for , employs a self-complementary AAV9 vector delivering the , achieving survival rates over 90% in treated infants versus historical controls. CRISPR-Cas9-based editing, an engineered system, enabled exagamglogene autotemcel (Casgevy), approved in 2023 for , by disrupting the BCL11A gene in hematopoietic stem cells to boost production. mRNA therapeutics, chemically modified for stability and translation efficiency, power vaccines and protein replacement; , approved in 2018, uses lipid nanoparticles to deliver siRNA silencing for . These applications highlight causal links between precise molecular modifications and clinical efficacy, though challenges like off-target effects and delivery persist.

Agricultural and Environmental Applications

Biomolecular engineering has enabled the development of genetically modified () crops that express engineered proteins for pest resistance, such as () toxins in and , reducing use by up to 37% in some regions while increasing yields. By 2022, , , , and canola occupied 188.6 million hectares globally, comprising 99% of cultivated GM crops, with documented economic benefits including higher farmer incomes from reduced pest damage and input costs. Herbicide-tolerant varieties, engineered via to express altered enzymes like 5-enolpyruvylshikimate-3-phosphate synthase, allow precise weed control and have sustained productivity gains in and canola cultivation, as evidenced in and Indian field trials. Virus-resistant , incorporating coat protein genes to confer immunity to , exemplifies targeted genetic modifications that rescued Hawaiian production from near collapse in the . Protein engineering techniques, including and rational design, have enhanced biopesticides by optimizing Bt protein stability and specificity against lepidopteran pests, leading to EPA approvals for plant-incorporated protectants that minimize non-target effects compared to chemical alternatives. Engineered enzymes for biofertilizers, such as variants with improved efficiency in , aim to reduce reliance on synthetic fertilizers, though field-scale deployment remains limited by challenges. These applications demonstrate causal links between molecular modifications—altering or —and agricultural outcomes like yield stability, supported by multi-year agronomic data showing no consistent yield drag in GE varieties. In environmental contexts, biomolecular engineering facilitates through microbes and enzymes tailored to degrade persistent organic pollutants (POPs), such as polycyclic aromatic hydrocarbons (PAHs), via engineered monooxygenases that enhance catalytic rates under aerobic conditions. constructs, including gene circuits in for sensing and metabolizing like mercury via mer operon modifications, enable real-time pollutant detection and conversion to less toxic forms. of laccases and hydrolases has produced variants with 10- to 100-fold higher activity against emerging contaminants like pharmaceuticals in , as demonstrated in lab-scale reactors achieving over 90% degradation efficiency. For plastic waste, engineered PETases from degrade at rates accelerated by , converting it to monomers for , with pilot processes reported in 2020. These interventions rely on empirical validation of and microbial consortia performance, prioritizing causal mechanisms like binding affinity over unverified ecological projections.

Materials and Nanotechnology Interfaces

Biomolecular engineering enables the precise integration of biomolecules such as DNA, proteins, and peptides with nanomaterials to form hybrid structures that combine biological specificity with material robustness. This interface leverages self-assembly principles, where engineered biomolecules act as programmable scaffolds to organize nanoparticles or inorganic components into ordered architectures with sub-nanometer precision. Such bio-nanomaterials exhibit enhanced functionalities, including stimuli-responsiveness and biocompatibility, surpassing traditional synthetic materials in applications requiring molecular recognition. DNA nanotechnology exemplifies this synergy, utilizing Watson-Crick base pairing to fold synthetic DNA strands into two- and three-dimensional nanostructures like origami tiles or lattices. Originating from foundational work in the 1980s, these structures serve as templates for positioning functional groups or metallic nanoparticles, facilitating applications in plasmonic devices and biosensors. Recent advances include dynamic DNA assemblies that undergo conformational changes in response to environmental cues, enabling controlled release in drug delivery systems with demonstrated efficacy in targeting cancer cells via aptamer integration. By 2023, scalable production methods had yielded DNA nanostructures over 1 micrometer in size, supporting multiplexing in diagnostic platforms. Protein engineering contributes through rational design of self-assembling motifs, producing such as cages, tubes, and from or modified natural proteins. Computational algorithms, refined by 2024, enable the creation of symmetric protein arrays that encapsulate payloads or catalyze reactions, with assembly yields exceeding 90% under physiological conditions. For instance, engineered variants self-assemble into hollow spheres for iron storage mimicry or drug loading, while β-solenoid proteins form micrometer-long nanotubes via , applicable in conductive nanowires. These structures maintain enzymatic activity post-assembly, as evidenced by catalysts retaining over 80% efficiency in non-native environments. At the nano-bio interface, biomolecular coatings on synthetic nanoparticles—such as or silica cores—mitigate and enable ligand-specific binding, critical for targeting. amphiphiles, engineered for helical folding, self-assemble into nanofibers that template mineralization, forming bone-like composites with compressive strengths up to 100 . In environmental applications, enzyme-nanoparticle conjugates degrade pollutants like organophosphates at rates 10-fold higher than free enzymes, due to stabilized conformations. Scalability remains constrained by folding fidelity, though has improved yields to industrial levels in select systems by 2025.

Biotechnology and Bioengineering

involves the application of biological systems, living organisms, or their derivatives to develop or manufacture products, integrating principles from , , and . This field spans techniques, processes, and cellular engineering to produce therapeutics, biofuels, and agricultural enhancements, with origins tracing to ancient practices like but accelerating post-1970s via milestones such as the 1973 development of by Cohen and Boyer. In 2023, global revenue exceeded $1.5 trillion, driven by pharmaceuticals comprising over 70% of the market. Bioengineering, distinct yet overlapping, applies engineering fundamentals—such as modeling, control systems, and —to analyze and design biological systems for practical outcomes in , agriculture, and industry. It emphasizes quantitative approaches, including and bioprocess optimization, with key advancements like the 1950s invention of the pump precursor and modern scaffolds that support organ regeneration. The discipline produced over 20,000 peer-reviewed publications annually by 2022, reflecting its role in devices like implantable sensors achieving 95% accuracy in glucose monitoring for . Biomolecular engineering intersects these fields by employing biotechnological tools, such as and manipulation, within bioengineering frameworks to rationally design proteins, nucleic acids, and nanoscale assemblies. For instance, techniques like —rooted in biotech's —enable precise protein variants for industrial catalysts, yielding enzymes with 100-fold improved stability under harsh conditions, as demonstrated in 2010s engineering for . This synergy facilitates scalable , where bioengineered microbial hosts produce biomolecules at gram-per-liter yields, bridging laboratory-scale biotech innovations to commercial viability. Unlike broader focused on organism-level applications or bioengineering's macroscopic systems, biomolecular engineering prioritizes atomic-resolution control, informed by data from tools like cryo-electron , which resolved over 10,000 protein structures by 2023 via initiatives like the .

Chemical and Biochemical Engineering

Chemical and biochemical engineering applies principles of , reaction kinetics, and to the production and manipulation of biomolecules at industrial scales. This discipline integrates fundamentals with biological systems to optimize the synthesis, separation, and purification of proteins, enzymes, nucleic acids, and metabolites derived from engineered organisms. Key challenges include maintaining cellular viability under varying conditions of , heat exchange, and while maximizing yield and minimizing costs. In upstream processing, bioreactor design is central, providing controlled environments for microbial or mammalian cell cultures to express engineered biomolecules. Stirred-tank bioreactors, for instance, dominate large-scale operations due to their scalability and ability to support high-density cultures through precise control of , , dissolved oxygen, and feed strategies. Enzymatic , governed by models such as Michaelis-Menten equations, inform these designs by quantifying (Km) and maximum (Vmax), enabling predictive scaling from bench to production volumes exceeding 10,000 liters. Advances in single-use bioreactors have reduced contamination risks and facilitated flexible manufacturing for therapeutics like monoclonal antibodies. Downstream processing accounts for 50-80% of production costs in biomolecular engineering, involving cell harvest, clarification, and purification steps to achieve regulatory purity levels often exceeding 99%. Techniques such as centrifugation, filtration, chromatography (e.g., affinity and ion-exchange), and ultrafiltration exploit biomolecular properties like charge, size, and hydrophobicity for selective recovery. Process analytical technologies (PAT), including real-time spectroscopy, enhance efficiency by monitoring critical quality attributes during purification, reducing batch failures. For example, in recombinant protein production, integrated continuous processing has demonstrated up to 40% cost reductions compared to traditional batch methods. Overall, chemical and ensures biomolecular processes are economically viable and reproducible, bridging molecular design with commercial viability. Optimization relies on computational modeling of and metabolic fluxes, with recent emphases on through waste minimization and energy-efficient separations. Despite biases in academic reporting toward idealized lab-scale successes, empirical data from validations underscore the necessity of rigorous scale-up validation to counter overoptimistic projections.

Education and Professional Practice

Academic Programs and Training

Academic programs in biomolecular engineering are predominantly housed within departments of , bioengineering, or dedicated biomolecular science units, offering bachelor's, master's, and doctoral degrees that integrate principles of , biochemistry, and engineering design. These programs emphasize quantitative analysis of biomolecular systems, including , , and manipulation, alongside foundational coursework in , , and reaction engineering. Undergraduate curricula typically span four years and require 120-130 credit hours, beginning with core sciences such as , , , physics, and , followed by specialized courses in , , and bioprocess design. For instance, Georgia Institute of Technology's in Chemical and includes a option with electives in and , preparing students for industry roles through capstone projects involving techniques. Similarly, the University of offers a biomolecular engineering track within its BS, featuring modules on and gene editing tools after the first two years of foundational sciences. Hands-on training occurs via laboratory courses teaching techniques like polymerase chain reaction (PCR), , and , often culminating in senior design projects simulating real-world applications such as vaccine production or . Graduate programs build on this base with advanced research training, where master's degrees (typically 30-36 credits, 1-2 years) focus on applied topics like biomolecular modeling and scale-up processes, while programs (4-6 years) emphasize original dissertation research in areas such as CRISPR-based engineering or nanoscale biomaterial assembly. At institutions like , candidates in chemical and biomolecular engineering undertake coursework in advanced kinetics and , followed by qualifying exams and mentored lab rotations to develop expertise in causal mechanisms of molecular interactions. University's Tandon School of Engineering provides a BS/MS accelerated path in chemical and biomolecular engineering, incorporating and for predictive . Training extends to interdisciplinary seminars, industry internships, and professional development in biosafety protocols, with many programs requiring proficiency in software tools like or molecular dynamics simulators. Notable programs are concentrated at research-intensive universities, including , , and , where enrollment in chemical and biomolecular engineering exceeds 200 undergraduates annually at larger schools, reflecting demand driven by biotech sector growth. These programs prioritize empirical validation through experimental design and data-driven hypothesis testing, often integrating first-year research experiences to foster causal reasoning in biomolecular systems. International offerings, such as at the University of California, Santa Cruz's Baskin Engineering, include biomolecular engineering courses with sub-team projects training in and , though U.S. programs dominate due to funding from agencies like the .

Career Pathways and Industry Roles

Biomolecular engineers often enter the field through undergraduate degrees in , bioengineering, or related disciplines, followed by graduate training emphasizing , , and techniques. Advanced degrees, such as master's or PhDs, are common for research-intensive roles, enabling specialization in areas like gene editing or optimization. Primary career pathways span , where practitioners lead programs and educate students on biomolecular design principles; , involving applied of therapeutics or ; and government or regulatory agencies, focusing on assessments and policy formulation. In , roles progress from postdoctoral to faculty positions, often requiring grants from bodies like the for projects in synthetic genomes. pathways typically begin with entry-level or lab technician positions, advancing to senior scientist or management through demonstrated expertise in scaling molecular processes. Regulatory paths emphasize compliance with standards from agencies like the FDA, drawing on skills for evaluating biomolecular products. Key industry roles include research scientists, who design and test engineered biomolecules for applications in pharmaceuticals or biofuels; process engineers, responsible for developing scalable systems, such as for recombinant proteins; and quality control analysts, ensuring product purity and regulatory adherence in biotech facilities. Additional positions encompass regulatory affairs specialists, who navigate approvals for genetically modified therapeutics, and technicians, handling upstream and downstream operations in production. These roles predominate in sectors like firms (e.g., developing monoclonal antibodies), (focusing on via engineered nanoparticles), and agricultural biotech (engineering pest-resistant crops). Employment in bioengineering fields, encompassing biomolecular specialties, is projected to grow 5% from 2024 to 2034, outpacing the average for all occupations due to demand for innovative therapies and sustainable bioproducts. annual wages reached $100,730 as of , with higher earnings in pharmaceuticals averaging $107,220, reflecting the technical demands of molecular-scale . Job availability stands at approximately 20,100 positions nationwide, concentrated in hubs.

Controversies and Criticisms

Biosafety, Biosecurity, and Dual-Use Risks

in biomolecular engineering encompasses protocols to mitigate accidental release of engineered biomolecules or organisms, which could lead to environmental or health risks through mechanisms like or unintended pathogenicity. The U.S. Centers for Disease Control and Prevention (CDC) and (NIH) classify laboratory practices into four biosafety levels (BSL-1 to BSL-4), with most routine gene editing and assembly in non-pathogenic hosts occurring at BSL-1 or BSL-2 due to low inherent risks from attenuated or novel constructs lacking natural virulence factors. Higher levels apply when engineering enhances transmissibility or lethality, as in gain-of-function studies on select agents like or coronaviruses, where containment failures could amplify outbreaks via exposure or spills. Empirical data from global incident tracking show laboratory-acquired averaging 2-5 per year in high-containment U.S. facilities from 2000-2020, though synthetic biology-specific accidents remain infrequent, with documented cases limited to procedural errors like improper waste disposal rather than novel pathogen escapes. Biosecurity measures address intentional misuse or theft of biomolecular tools, such as restriction enzymes, kits, or synthetic DNA sequences accessible via commercial providers, which could enable non-state actors to assemble hazardous agents in unregulated settings. In , risks stem from the of design-build-test cycles, where open-source repositories like Addgene distribute thousands of plasmids annually, potentially bypassing screening for sequences matching genomes; for instance, a 2018 of horsepox —a relative—using mail-order DNA demonstrated feasibility without specialized facilities, prompting voluntary industry screening under the International Gene Consortium. U.S. regulations mandate Federal Select Agent Program oversight for 67 and toxins, including inventory controls and personnel reliability checks, while dual-use export controls under the restrict transfers of gene equipment to prevent proliferation in adversarial states. Despite these, gaps persist in DIY communities, where unregulated experiments have led to self-injection incidents with unvetted constructs, highlighting enforcement challenges outside institutional labs. Dual-use risks arise when biomolecular engineering yields knowledge or products with both beneficial (e.g., therapeutics) and harmful applications (e.g., bioweapons), as defined by U.S. on Dual-Use Research of Concern (DURC)—life sciences work reasonably anticipated to enhance attributes like or while enabling misuse. Seven DURC experiments, such as reverse-engineering strains for transmissibility, and 15 specified agents trigger institutional review entities for risk-benefit assessments, with a 2024 policy update expanding oversight to pathogens with enhanced pandemic potential amid advances like genome assembly. Peer-reviewed analyses estimate 10-50% of projects carry DURC potential, varying by expert assessment, due to tools like machine learning-optimized lowering barriers to toxin engineering; however, no verified incidents have exploited these to date, contrasting with historical chemical analogs. International frameworks, including the , lack verification mechanisms, leading calls for harmonized screening of DNA orders exceeding 1 kilobase for homology to regulated sequences, balanced against innovation stifling from over-regulation. Mitigation strategies emphasize "safety by design," incorporating kill switches in engineered microbes—genetic circuits that trigger under escape conditions—reducing release probabilities by orders of magnitude in contained tests.

Ethical Debates on Genetic Modification

Ethical debates surrounding genetic modification in biomolecular engineering primarily center on editing, where alterations to embryos or reproductive cells are heritable across generations, raising concerns over safety, , and societal implications. Unlike editing, which targets non-reproductive cells and affects only the individual, modifications introduce irreversible changes to the human gene pool, prompting scrutiny from bodies like the National Academies of Sciences, Engineering, and Medicine, which in 2017 recommended a pause on clinical applications until safety and ethical frameworks are robust. Critics argue that premature deployment risks unintended off-target mutations, as evidenced by early CRISPR-Cas9 studies showing editing errors in non-human models that could lead to mosaicism or oncogenic potential in humans. Proponents counter that for monogenic diseases like Huntington's or , editing could eliminate hereditary transmission, aligning with principles of procreative beneficence where parents have a duty to maximize offspring welfare, as articulated by ethicist . The 2018 case of , a biophysicist who edited the gene in human embryos to confer resistance, exemplifies these tensions, resulting in the birth of twins and international condemnation for bypassing ethical oversight and . He was sentenced to three years in prison in for illegal medical practices, highlighting regulatory gaps, yet the procedure's intent—to mimic natural mutations observed in HIV-resistant individuals like the —underscored potential therapeutic value, though mosaicism in the embryos raised doubts about efficacy and safety. Ethical analyses post-event emphasized the absence of compelling medical need, as transmission can be prevented via or sperm washing, and warned against normalizing enhancements under therapeutic guise. This incident spurred calls for global moratoriums, with the in 2019 advocating standardized governance to assess risks versus benefits empirically rather than preemptively. Eugenics concerns invoke historical precedents of coercive sterilization programs in the early , which targeted traits deemed undesirable without consent, but modern voluntary differs fundamentally as it empowers individual rather than . Fears of a "" through designer babies persist, particularly for non-therapeutic enhancements like or athleticism, potentially exacerbating inequalities if accessible only to the wealthy, as projected in analyses of polygenic risk scores. Empirical evidence, however, indicates no current capability for safe, precise enhancement; CRISPR's error rates and incomplete understanding of gene-environment interactions limit applications to high-penetrance disease alleles. Defenders argue that equating therapeutic with conflates intent, noting that (PGD) already selects embryos to avoid diseases without altering genomes, suggesting tools could democratize such prevention if scaled equitably. Informed consent poses unique challenges for editing, as cannot prospectively agree to modifications, leading ethicists to propose frameworks balancing parental autonomy with societal oversight. debates extend to engineering models for biomolecular research, where genetic knock-ins have improved disease but raised issues from unintended phenotypes, as in -edited pigs exhibiting off-target skeletal defects. Overall, while from trials (e.g., FDA-approved therapies for sickle cell by 2023) build confidence, applications demand rigorous, multi-stakeholder validation to mitigate dual-use risks without stifling innovation grounded in verifiable therapeutic gains.

Economic Barriers and Innovation Constraints

The high of biomolecular engineering R&D constitutes a primary economic barrier, with costs for developing engineered biomolecules—such as custom proteins or metabolic pathways—frequently surpassing $100 million per candidate due to requirements for advanced sequencing, synthesis equipment, and iterative testing. These expenditures are exacerbated by the need for scalable processes, where media and bioreactor optimization alone can drive up production expenses, as seen in challenges for cultivated meat analogs where cell-culture media costs remain a dominant hurdle. , which drive much of the field's , often struggle with these upfront investments, leading to reliance on that prioritizes incremental over transformative projects. Funding constraints further impede progress, as investors face prolonged timelines to —typically 10-15 years for biomolecular therapeutics—amid high attrition rates exceeding 90% from to market approval. and private , while increasing (e.g., U.S. academic R&D expenditures rose 11.2% to over $97 billion in FY 2023), disproportionately favors established institutions over nascent biomolecular startups, creating gaps in early-stage support for applications. This risk aversion stems from empirical evidence of capital-intensive failures, where even promising engineered pathways falter in pilot scaling due to unforeseen yield inefficiencies. Regulatory requirements impose substantial innovation constraints by extending development cycles and escalating compliance costs, often adding years and hundreds of millions to biomolecular product timelines through mandatory preclinical and clinical validations. In jurisdictions like the U.S. and , agencies such as the FDA demand rigorous safety data for genetically modified biomolecules, which, while grounded in risk mitigation, disproportionately burdens novel modalities like multi-gene edits by favoring incumbents with established dossiers over agile innovators. Uncertainty in approval pathways for emerging techniques, such as cell-free synthetic systems, deters , as evidenced by stalled in biorefinery-scale biomolecular processes. These hurdles, compounded by fragmented standards, limit cross-border collaboration and slow diffusion of breakthroughs in areas like .

Empirical Evidence Against Alarmist Narratives

Over three decades of commercial deployment of genetically modified () crops, encompassing billions of meals consumed globally, have yielded no verifiable evidence of adverse effects in humans attributable to their consumption. Comprehensive reviews, including meta-analyses of field trials and epidemiological data, confirm that GM varieties exhibit equivalent profiles to conventional crops, with no increased incidence of allergies, , or diseases such as cancer. The has assessed approved GM foods as passing rigorous safety evaluations, presenting no greater risk than non-GM counterparts. In gene editing applications like CRISPR-Cas9, clinical trials as of 2024 report minimal off-target effects , with advanced protocols—such as high-fidelity variants and optimization—reducing unintended edits to levels below detection thresholds in many cases, far lower than initial concerns suggested. No trial outcomes have linked observed off-target activity to clinical harm, and long-term monitoring in approved therapies, such as those for , shows sustained efficacy without emergent genotoxicity. Biosecurity fears surrounding , including pathogen design, remain largely unrealized despite widespread access to tools since the 2010s; empirical records indicate zero confirmed instances of dual-use misuse leading to public harm, attributable to inherent technical barriers like inefficient synthesis yields and verifiable oversight in DNA ordering. incidents, such as laboratory escapes, occur at rates comparable to classical (under 0.01% of experiments per global surveillance data), mitigated by standardized containment protocols without evidence of escalated risks from engineered organisms. Gene therapy products, monitored via FDA-mandated long-term follow-up, demonstrate low rates of delayed adverse events; for vectors, doses below 1.4 × 10^11 vector genomes per patient yield no significant toxicities in from over 100 trials, contrasting with progression as the primary confounder in higher-risk cohorts. This track record underscores that while risks warrant vigilance, alarmist projections of widespread catastrophe lack substantiation from operational data.

Recent Developments

AI and Computational Integration (2020-2025)

The period from 2020 to 2025 marked a pivotal shift in biomolecular engineering through the integration of () and computational methods, enabling unprecedented accuracy in predicting, designing, and optimizing biomolecules such as proteins and metabolic pathways. models, trained on vast datasets of structural and functional data, addressed longstanding challenges in design and , reducing reliance on empirical trial-and-error approaches. This integration accelerated workflows in , where tools analyzed complex interactions to engineer novel enzymes and genetic circuits with tailored properties. A landmark advancement was DeepMind's AlphaFold2, released in December 2020, which achieved near-experimental accuracy in for over 200 million proteins by July 2021, fundamentally impacting by providing atomic-level models for rational design and mutation analysis. Subsequent iterations, including AlphaFold3 in 2024 and AlphaFold4 in July 2025, extended capabilities to multi-component complexes, ligands, and dynamic interactions, facilitating applications in optimization and engineering. These tools democratized access via open databases, enabling engineers to predict folding outcomes and iterate designs computationally, with reported reductions in experimental validation time by factors of 10-100 in case studies. Generative AI models emerged as a core driver for biomolecular design, employing diffusion-based and variational autoencoder architectures to create de novo proteins and small molecules with specified functions. For instance, by 2023, frameworks like RFdiffusion and hybrid generative models integrated physics-based constraints with learned representations to design functional enzymes, achieving success rates exceeding 50% in experimental validation for non-natural scaffolds. In synthetic biology, machine learning optimized metabolic pathways and gene circuits; tools such as the Automated Recommendation Tool (ART), refined through 2020-2025, used reinforcement learning to recommend genetic edits, boosting yields in microbial engineering by up to 5-fold in biofuel and pharmaceutical production. Computational integration also advanced biomolecular interaction , where graph neural networks and transformers modeled protein-ligand and protein-protein affinities, aiding in the of biosensors and therapeutics. By 2025, AI-driven platforms like BioNeMo incorporated pretrained models for end-to-end design, scaling simulations to handle synthetic genomes and multi-omics data. Despite these gains, limitations persisted, including biases in training data toward well-studied proteins and challenges in predicting transient states, necessitating hybrid experimental-computational validation. Overall, this era's tools enhanced causal understanding of biomolecular function, prioritizing data-driven iteration over methods.

Advances in Multi-Gene Editing and Synthetic Genomes

Multi-gene editing techniques have advanced significantly since the refinement of CRISPR-Cas systems, enabling simultaneous modifications at multiple genomic loci to address polygenic traits and complex diseases. Multiplexed CRISPR approaches, such as those using arrays of guide RNAs, allow for the targeted alteration of dozens to hundreds of genes in a single operation, improving efficiency over sequential editing methods. In January 2025, researchers at the developed mvGPT, a versatile tool that independently edits multiple genes while regulating their expression levels, facilitating precise control in therapeutic applications for genetic disorders affecting single cells. Similarly, a June 2025 study from enhanced multiplex editing precision by threefold, incorporating mechanisms to minimize off-target mutations through improved design and error-correction algorithms. These innovations build on earlier CRISPR variants discovered in 2021, which supported multi-site editing for crop enhancement, demonstrating scalability from model organisms to applied biomolecular engineering. Further breakthroughs include systems for distributed editing across tissues. In June 2025, UT Southwestern researchers engineered a -based platform that simultaneously targets genes in multiple organs, such as liver and muscle, using delivery to achieve coordinated modifications without systemic toxicity. Large-scale DNA engineering has also progressed, with September 2025 reports detailing applications of novel CRISPR effectors for rewriting extensive genomic regions, essential for engineering microbial factories or correcting multifactorial human pathologies. These methods prioritize empirical validation of editing fidelity, with off-target rates reduced to below 0.1% in optimized protocols, as verified through whole-genome sequencing. Parallel advances in synthetic genomes have culminated in the near-complete redesign of eukaryotic chromosomes, providing blueprints for organism construction. The Synthetic Yeast Genome Project (Sc2.0), initiated in 2006, achieved a milestone in January 2025 with the assembly of synXVI, a 903 kb synthetic incorporating redesigned regulatory elements for enhanced stability and functionality. This completed the full synthetic genome, enabling iterative engineering via SCRaMbLE—a synthetic recombination system that generates for trait optimization, as demonstrated in August 2025 experiments consolidating up to 6.5 chromosomes into viable strains. The resulting organisms exhibit minimal essential gene sets, with genome sizes reduced by up to 20% through removal of , confirming causal roles of specific sequences in cellular fitness via direct perturbation studies. Efforts toward synthetic human genomes emerged in 2025, with scientists initiating construction of artificial chromosomes using scalable and assembly techniques. Announced in June 2025, the Synthetic Human Genome Project aims to synthesize large DNA segments, building on yeast precedents to test human-compatible designs while addressing stability challenges observed in prokaryotic minimal . These developments underscore causal realism in design: empirical data from yeast refactoring reveal that refactoring 10-15% of a alters metabolic flux predictably, informing scalable for biomolecular production and disease modeling without reliance on unverified assumptions.

Scaling Challenges and Breakthroughs

One major challenge in scaling biomolecular engineering involves bioreactor design and operation, where increasing volume from lab-scale (e.g., milliliters) to industrial-scale (thousands of liters) disrupts mass and heat transfer, leading to gradients in oxygen, pH, and nutrients that reduce cell viability and product yields by up to 50% in mammalian cell cultures. Shear forces from impellers exacerbate this, causing cell damage in shear-sensitive systems like CHO cells used for monoclonal antibodies, while contamination risks rise due to longer run times and larger surface areas. For recombinant proteins, aggregation during expression or purification at scale compromises purity, with up to 20-30% loss in recoverable yield attributed to misfolding under non-optimal conditions. Economic barriers compound these issues, as capital costs for facilities can exceed $500 million for 10,000-20,000 L bioreactors, demanding precise process analytical technology for real-time monitoring to avoid batch failures. Breakthroughs in continuous have addressed batch limitations by enabling modes in single-use bioreactors, sustaining steady-state for weeks and boosting titers by 2-5 fold over fed-batch systems, as demonstrated in intensified processes for therapeutic proteins. , leveraging engineered or , has scaled output—such as whey proteins at commercial volumes—through optimization, achieving yields over 10 g/L while reducing use by 90% compared to traditional animal sourcing. In mRNA , microfluidic technologies enabled rapid scale-up during the response, with chips processing up to 17 L/h of lipid nanoparticle-formulated mRNA, facilitating billions of doses from modular facilities within months rather than years. These advances, supported by process intensification and tools for robust host strains, have lowered per-unit costs by 50-70% in select cases, though full industrialization requires overcoming regulatory hurdles for continuous validation.

Future Prospects

Open Engineering Challenges

One persistent challenge in biomolecular engineering is achieving reliable design of proteins with novel functions, as current computational tools, despite advances like , struggle with predicting and stabilizing non-natural folds under physiological conditions, leading to low success rates in functional validation. Knowledge gaps in structure-function relationships further complicate rational design, often requiring iterative that is time- and resource-intensive. In gene editing, multiplexed CRISPR applications face limitations from off-target effects, inefficient simultaneous modification of multiple loci, and delivery barriers, with double-strand breaks frequently triggering unintended cellular responses like rather than precise edits. vectors such as AAV impose size constraints on Cas proteins and guides, restricting payload capacity to under 5 , while non-viral nanoparticles encounter and poor tissue specificity issues. Scalability remains a core bottleneck in producing engineered biomolecules at volumes, where transitions from lab-scale bioreactors to large fermenters introduce variability in , purity, and product consistency due to , oxygen gradients, and metabolic burdens on host cells. Biological heterogeneity and parameter sensitivity exacerbate failures, with many processes yielding only 10-50% of predicted titers upon scale-up. Synthetic biology efforts to construct minimal or synthetic genomes encounter hurdles in and specialization, as engineered circuits often fail to recapitulate native regulatory , resulting in or incomplete functionality in multicellular contexts. Integrating these systems into living hosts also demands overcoming interoperability issues between designed modules and endogenous pathways to avoid or evolutionary drift.

Potential Societal and Economic Impacts

Biomolecular engineering is poised to catalyze economic expansion in the sector, with the global market—encompassing engineered biomolecules for industrial and therapeutic uses—projected to grow from $17.09 billion in 2025 to $63.77 billion by 2032 at a (CAGR) of 20.7%. This growth stems from applications in , , and , driven by innovations like , which alone commands a market valued at $2.60 billion in 2023 and expected to expand at a 16.24% CAGR through 2030 due to demand for customized enzymes and therapeutics. In the United States, the broader bioscience industry, including biomolecular advancements, contributed $2.9 trillion to the economy in recent assessments, underscoring its role in fostering high-value and . Employment opportunities in related fields are anticipated to rise, with bioengineers and biomedical engineers facing a 5% job growth projection from 2024 to 2034, outpacing average occupational rates, as firms seek expertise in designing novel biomolecules for diagnostics and . Venture investments in rebounded to $12.2 billion in 2024 across over 900 companies, signaling renewed capital inflow that could accelerate commercialization of engineered pathways for sustainable fuels and chemicals, potentially offsetting dependencies and creating specialized roles in computational design and scaling. However, market volatility, as evidenced by a dip in biotech hiring from 19,000 positions in early 2022 to 10,000 by late 2023, highlights risks tied to regulatory hurdles and funding cycles, which may constrain short-term job creation despite long-term demand. Societally, biomolecular engineering could transform healthcare by enabling precise interventions, such as engineered antibodies and gene circuits that target cancers or infectious diseases more effectively than traditional methods, thereby reducing costs and improving outcomes in resource-limited settings. In , redesigned microbial consortia and enzymes promise higher crop yields and reduced use, addressing for a projected global population of 9.7 billion by 2050 through resilient biomolecular solutions. Environmentally, bioengineered organisms for plastic degradation and carbon capture offer pathways to mitigate , with potential to sequester gigatons of CO2 annually if scaled, though equitable access remains contingent on policy frameworks that prioritize empirical efficacy over unsubstantiated fears. These impacts hinge on overcoming barriers like constraints, ensuring that benefits accrue broadly rather than exacerbating disparities in technological adoption.

Pathways to Broader Adoption

Standardization of biological parts and pathways remains a foundational step toward broader , enabling modular assembly of engineered biomolecules with predictable functions. Challenges in achieving across genetic circuits have historically impeded , but advances in retrobiosynthetic design and automated pathway optimization address this by identifying viable metabolic routes more efficiently. For instance, protocols for engineering synthetic metabolic pathways have demonstrated up to 10-fold improvements in product yields for biofuels and pharmaceuticals, facilitating transition from lab-scale to industrial applications. Scaling processes is critical to reducing costs, which currently hinder competitiveness against . Iterative optimization in and , combined with hardware innovations like enhanced fluidics for , can lower production expenses by 50-90% for complex biomolecules such as enzymes and antibodies. Multidisciplinary efforts integrating principles have overcome biological variability, as seen in successful scale-up of synbio-derived materials that achieve parity with alternatives in cost and performance. Regulatory across jurisdictions accelerates market entry by minimizing duplicative testing and approval timelines. Divergent frameworks for genome-edited products, such as those under the EU's GMO directives versus the U.S. FDA's product-based approach, create barriers estimated to delay by 2-5 years; proposed reforms emphasize risk-based assessments tied to empirical rather than process-specific rules. In 2023-2025, initiatives like the U.S. BIOSECURE Act and international dialogues have begun aligning standards for engineered biologics, potentially expanding access to therapies and agrochemicals. Commercialization pathways emphasize strategic partnerships between academia, startups, and incumbents to navigate and funding gaps. inflows into synbio firms reached $18.5 billion in , supporting development from proof-of-concept to , with successes in biocatalysis demonstrating 20-30% in fine chemicals by leveraging hybrid chemo-enzymatic processes. Pricing models that bundle engineered pathways with licensing agreements further de-risk adoption, as evidenced by collaborations yielding scalable production of sustainable materials. Workforce expansion through targeted in quantitative and addresses talent shortages, with programs integrating computational tools projected to double the number of proficient practitioners by 2030. Empirical from consortia indicate that such training correlates with 15-25% faster cycles, paving the way for widespread integration in sectors like and .

References

  1. [1]
    Biomolecular engineering for nanobio/bionanotechnology - PMC
    Biomolecular engineering can be used to purposefully manipulate biomolecules, such as peptides, proteins, nucleic acids and lipids.
  2. [2]
    [PDF] Unsolved Problems in Biomolecular Engineering - OSU CBE
    In. 1992 (as described earlier) the term biomolecular engineering was coined to include all activities at the chemical engineering and biological interface with.
  3. [3]
    Essential molecular biology methods in biomaterials research
    We focus on methodologies such as recombinant DNA technology, polymerase chain reaction (PCR), in situ hybridization, immunocytochemistry (ICC), and ...
  4. [4]
    Recent progress in biomolecular engineering - PubMed
    As an applied molecular evolution technology, DNA shuffling will play a key role in biomolecular engineering. In contrast to the point mutation techniques ...
  5. [5]
    Insights Into Protein Engineering: Methods and Applications
    Oct 29, 2024 · Protein engineering encompasses multiple strategies including rational design, directed evolution, semirational design, peptidomimetics, and de ...
  6. [6]
    Class Roster - Spring 2024 - CHEME 5440 - Cornell University
    Learn how to apply core engineering concepts (kinetics, thermodynamics, transport phenomena) to understanding and engineering the function of living systems.
  7. [7]
    Bringing biomolecular engineering on board | Nature Chemical ...
    Mar 8, 2024 · Biomolecular engineering empowers chemical engineers to design, optimize and scale-up biological processes and systems with enhanced efficiency, reliability ...
  8. [8]
    Biomolecular Engineering | EBRC Research Roadmap
    Jun 19, 2019 · The roadmap for Biomolecule, Pathway, and Circuit Engineering addresses the engineering of individual biomolecules to have expanded or new functions.
  9. [9]
    What is biomolecular engineering? - Lafayette College
    Biomolecular Engineers apply engineering principles to analyze, utilize, and/or design biological molecules and systems.
  10. [10]
    Biological Engineering Concentration
    In 1992, NIH defined "biomolecular engineering" as: "Research at the interface of chemical engineering and biology with an emphasis at the molecular level."
  11. [11]
    Genetic Engineering and Gene Technology - ScienceDirect.com
    Biomolecular engineering is defined as the manipulation and design of biomolecules for various applications, including medical diagnostics, therapy, ...
  12. [12]
    Biomolecular Engineering | Chemical and Biological Engineering
    Biomolecular engineering is the analysis and engineering of these living systems. Our research uses techniques from synthetic biology, organic chemistry, ...
  13. [13]
    The Importance and Future of Biochemical Engineering - PMC
    The Importance and Future of Biochemical Engineering. Timothy A Whitehead ... biomolecular engineering and related fields. Many questions and challenges ...
  14. [14]
    Chemical and Biomolecular Engineering - JHU catalogue
    The study of Chemical and Biomolecular Engineering (ChemBE) is dedicated to the design and exploitation of chemistry, biology, mathematics and physics, ...
  15. [15]
    Piecing together a puzzle. An exposition of synthetic biology - PMC
    Bioengineering, as part of synthetic biology, is the design of entirely new signalling pathways, including multiple genes and regulatory elements… The various ...
  16. [16]
    Bioengineering vs. Biomedical Engineering: What's the Difference?
    Bioengineering is the study of applied engineering practices in general biology. Bioengineers' work often focuses on general theory that can be applied to ...
  17. [17]
    1941: One Gene, One Enzyme
    Apr 23, 2013 · Beadle and Tatum proposed that, in general, each gene directs the formation of one (and only one) enzyme. For their work, Beadle and Tatum ...
  18. [18]
    1944: DNA is \"Transforming Principle\"
    Apr 23, 2013 · Oswald Avery, Colin MacLeod, and Maclyn McCarty showed that DNA (not proteins) can transform the properties of cells, clarifying the chemical nature of genes.
  19. [19]
    1952: Genes are Made of DNA
    Apr 23, 2013 · Hershey and Chase figured that the virus transferred genetic material into the bacterium to direct the production of more virus. They knew that ...
  20. [20]
    1953: DNA Double Helix
    Apr 23, 2013 · Francis Crick and James Watson described the double helix structure of DNA. By the time Watson and Crick turned their attention to solving the chemical ...
  21. [21]
    Central Dogma - National Human Genome Research Institute
    The fundamental theory of central dogma was developed by Francis Crick in 1958. His version was a bit more global and included the notion that information does ...
  22. [22]
    Deciphering the Genetic Code - National Historic Chemical Landmark
    Marshall Nirenberg and Heinrich Matthaei discovered the key to breaking the genetic code when they conducted an experiment using a synthetic RNA chain.
  23. [23]
    Highlights of the DNA cutters: a short history of the restriction enzymes
    The first of these new enzymes, HindII, was discovered in Hamilton ('Ham') Smith's laboratory at Johns Hopkins Medical School in 1970 (29). This was ...
  24. [24]
    The Nobel Prize in Physiology or Medicine 1978 - Press release
    In 1970 he published two classical papers which described the discovery of a restriction enzyme from the bacterium Heamophilus influenzae and characterized in ...Missing: date | Show results with:date
  25. [25]
    1972: First Recombinant DNA
    Apr 26, 2013 · The first production of recombinant DNA molecules, using restriction enzymes, occurred in the early 1970s.
  26. [26]
    Herbert W. Boyer and Stanley N. Cohen | Science History Institute
    In 1972 researchers, including Boyer, realized that the enzyme EcoRI, which had actually been discovered in Boyer's UCSF lab, cut DNA in such a way that the ...
  27. [27]
    Asilomar and Recombinant DNA: The End of the Beginning - NCBI
    At noon on February 27, 1975, the curtain descended on the first act of what is likely to go down in the history of science as the recombinant DNA controversy.THE 1973 GORDON... · THE ACADEMY'S TURN · THE ASILOMAR CONFERENCE
  28. [28]
    History of insulin - PMC - NIH
    Jul 16, 2012 · In 1978, the first recombinant DNA human insulin was prepared by David Goeddel and his colleagues (of Genentech) by utilizing and combining ...
  29. [29]
    Kary Mullis | Nobel Prize Winning Biochemist & Inventor of PCR
    Sep 13, 2025 · PCR was developed in 1983 by Kary B. Mullis, an American biochemist who won the Nobel Prize for Chemistry in 1993 for his invention. Before the ...
  30. [30]
    Press release: The 1993 Nobel Prize in Chemistry - NobelPrize.org
    Kary B. Mullis, La Jolla, California, U.S.A., for his invention of the polymerase chain reaction (PCR) method, and half to. Professor Michael Smith ...Missing: date | Show results with:date
  31. [31]
    Genomics and Postgenomics - Stanford Encyclopedia of Philosophy
    Oct 20, 2016 · The draft human genome sequence (about 90% complete) was announced in June 2000, followed in 2001 by the publication of the draft sequences ...<|separator|>
  32. [32]
    We're entering a golden age of engineering biology - Noahpinion
    Feb 18, 2024 · By the 2000s, the aptly named next-generation sequencing (NGS) dramatically accelerated the rate of DNA sequencing while dropping the cost ...
  33. [33]
    Ten years of genetics and genomics: what have we achieved and ...
    We now have access to epigenomes, transcriptomes, small RNA and replication timing profiles. We also know a lot more about nuclear organization and chromosome ...Missing: post- biomolecular
  34. [34]
    CRISPR-based bioengineering in microalgae for production of ... - NIH
    Oct 26, 2023 · This review summarizes the advancements of CRISPR-based bioengineering for production of industrially important biomolecules and provides key considerations to ...
  35. [35]
    Unleashing the power of CRISPR-Cas mediated gene editing of ...
    CRISPR-based molecular networks control gene transcription, resulting in improved biofuel production. Generating genetically engineered microbes having ...
  36. [36]
    What is CRISPR? A bioengineer explains - Stanford Report
    Jun 10, 2024 · The short answer: CRISPR can precisely modify a piece of DNA or its chemistry (so-called epigenetics) in the human body, making it a potential ...
  37. [37]
    The second decade of synthetic biology: 2010–2020 - PMC - NIH
    Oct 14, 2020 · Academic achievements include engineering cells to fix CO2 and nitrogen, and getting yeast to make opioids and cannabinoids. Biofoundries have ...
  38. [38]
    Synthetic biology 2020–2030: six commercially-available products ...
    Dec 11, 2020 · Advances in microfluidics, transformation, and genome editing make it easier to culture species and insert large DNA fragments. Principles from ...
  39. [39]
    Highly accurate protein structure prediction with AlphaFold - Nature
    Jul 15, 2021 · Here we provide the first computational method that can regularly predict protein structures with atomic accuracy even in cases in which no similar structure ...
  40. [40]
    De novo protein design by inversion of the AlphaFold structure ... - NIH
    De novo protein design aims to create stable, well‐folded proteins with sequences distant from those found in nature and potentially new functions. To date, de ...
  41. [41]
    Advances in protein engineering and its application in synthetic ...
    Due to advances in synthetic biology, enzymes can now be engineered to catalyze reactions that do not occur naturally. The majority of protein engineering ...
  42. [42]
    Accurate structure prediction of biomolecular interactions ... - Nature
    May 8, 2024 · The introduction of AlphaFold 21 has spurred a revolution in modelling the structure of proteins and their interactions, enabling a huge ...Nobel Prize in Chemistry 2024 · Nature Machine Intelligence
  43. [43]
    Understanding biochemistry: structure and function of nucleic acids
    Oct 11, 2019 · We explain the structure of the DNA molecule, how it is packaged into chromosomes and how it is replicated prior to cell division.
  44. [44]
    From DNA to RNA - Molecular Biology of the Cell - NCBI Bookshelf
    All of the RNA in a cell is made by DNA transcription, a process that has certain similarities to the process of DNA replication discussed in Chapter 5.Transcription Produces RNA... · Signals Encoded in DNA Tell...
  45. [45]
    DNA structure and function - Travers - 2015 - The FEBS Journal
    Apr 22, 2015 · We review recent evidence suggesting that DNA supercoiling, particularly that generated by DNA translocases, is a major driver of gene regulation.
  46. [46]
    Nucleic Acids and Their Analogues for Biomedical Applications - PMC
    The basic function of nucleic acids in molecular biology is gene coding and regulation, which requires base pairing of complementary sequences and the formation ...
  47. [47]
    Ribogenomics: the Science and Knowledge of RNA - ScienceDirect
    Ribogenomics is to study the biology of cellular RNAs, including their origin, biogenesis, structure and function.
  48. [48]
    Biochemistry, Primary Protein Structure - StatPearls - NCBI Bookshelf
    Oct 31, 2022 · The primary structure of a protein is a linear chain of amino acids linked by peptide bonds, forming a polypeptide chain.Fundamentals · Molecular Level · Function · Mechanism
  49. [49]
    The Shape and Structure of Proteins - Molecular Biology of the Cell
    The three-dimensional conformation of a protein molecule is determined by its amino acid sequence. The folded structure is stabilized by noncovalent ...
  50. [50]
    Physiology, Proteins - StatPearls - NCBI Bookshelf - NIH
    Proteins can be further defined by their four structural levels: primary, secondary, tertiary, and quaternary. The first level is the primary structure because ...
  51. [51]
    Foundations for the Study of Structure and Function of Proteins - PMC
    Movement and structures are the basic elements of protein functions. Protein movement includes short-time and small-amplitude movement, median-time and median- ...
  52. [52]
    Engineering protein-based therapeutics through structural ... - Nature
    Apr 27, 2023 · Proteins represent the most versatile class of biomolecules, acting as catalysts, signaling molecules, molecular and ion transporters, scaffolds ...
  53. [53]
    Protein Engineering: A New Frontier for Biological Therapeutics - PMC
    Protein engineering holds the potential to transform the metabolic drug landscape through the development of smart, stimulus-responsive drug systems.
  54. [54]
    Exploiting metabolic glycoengineering to advance healthcare - Nature
    Sep 6, 2019 · In this Review, we summarize the chemistry underlying MGE that has been developed over the past three decades and highlight several recent ...
  55. [55]
    Advances in bacterial glycoprotein engineering: A critical review of ...
    Jan 2, 2025 · This review outlines the recent advances in bacterial protein glycosylation from the perspective of synthetic biology and metabolic engineering
  56. [56]
    New method to synthesize carbohydrates could pave the way to ...
    Aug 12, 2025 · Seeberger pioneered the process for carbohydrate synthesis in 2001 and has improved on it over the past 25 years.
  57. [57]
    Advances in molecular engineering of carbohydrate-binding modules
    Carbohydrate-binding modules (CBMs) are non-catalytic domains that are generally appended to carbohydrate-active enzymes. CBMs have a broadly conserved ...
  58. [58]
    Engineering the Lipid and Fatty Acid Metabolism in Yarrowia ...
    Mar 20, 2022 · An alternative and sustainable strategy for high oleic oil production was established by reprogramming the metabolism of the oleaginous yeast Yarrowia ...
  59. [59]
    Optimizing Archaeal Lipid Biosynthesis in Escherichia coli
    Aug 3, 2024 · Here, we substantially increased its production in E. coli by overexpression of an archaeal phosphatidylserine synthase needed for ethanolamine headgroup ...
  60. [60]
    Metabolic Engineering of Lipids Improves the Respiratory Function ...
    Oct 26, 2018 · By engineering lipid synthesis to carefully control the membrane composition, researchers found that lipids, which consist of fatty acid ...<|separator|>
  61. [61]
  62. [62]
    Recombinant DNA - an overview | ScienceDirect Topics
    Recombinant DNA is the method of joining two or more DNA molecules to create a hybrid. The technology is made possible by two types of enzymes, restriction ...
  63. [63]
  64. [64]
    Recombinant DNA Technology - an overview | ScienceDirect Topics
    Recombinant DNA technology encompasses the manipulation of genetic information outside an organism to obtain superior and desired traits in living organisms.
  65. [65]
    Paul Berg and the origins of recombinant DNA - ScienceDirect.com
    Feb 29, 2024 · Paul Berg, a professor in Stanford University's biochemistry department, was a leader in the early development of recombinant DNA technology.
  66. [66]
    Site-directed mutagenesis - PubMed
    Site-directed mutagenesis is a PCR-based method to mutate specified nucleotides of a sequence within a plasmid vector. This technique allows one to study ...
  67. [67]
    Site-Directed Mutagenesis - an overview | ScienceDirect Topics
    Site-directed mutagenesis methods are well established. Two commonly used techniques are overlap extension and QuikChange mutagenesis. Both employ PCR with ...
  68. [68]
    History of CRISPR-Cas from Encounter with a Mysterious Repeated ...
    Jan 22, 2018 · The CRISPR-Cas system of Streptococcus pyogenes was then applied to perform genome editing in human nerve and mouse kidney cells (35, 36). Thus, ...<|control11|><|separator|>
  69. [69]
    CRISPR History and Development for Genome Engineering - Addgene
    An overview of CRISPR's development from a prokaryotic immune system to a valuable genome engineering tool.Crispr History And... · Crispr: An Adaptive Immune... · Genome Engineering...
  70. [70]
    Recombinant DNA - Biological Principles
    Recombinant DNA technology leads to genetically modified organisms (GMOs). Recombinant DNA requires 3 key molecular tools: Cutting DNA at specific sites – most ...Molecular Cloning · I. Recombinant Dna · Ii. Polymerase Chain...Missing: experiments | Show results with:experiments
  71. [71]
  72. [72]
    Plasmid - National Human Genome Research Institute
    A plasmid is a small circular DNA molecule found in bacteria and some other microscopic organisms. Plasmids are physically separate from chromosomal DNA.
  73. [73]
    Plasmids 101: What is a plasmid? - Addgene Blog
    Jan 14, 2014 · A plasmid is a small circular piece of DNA that replicates independently from the host's chromosomal DNA, mainly found in bacteria.
  74. [74]
    Mutagenesis: Site-Directed - eLS - Walquist - Wiley Online Library
    Jan 22, 2018 · Site-directed mutagenesis (SDM) aims to introduce precise alterations in any coding or noncoding deoxyribonucleic acid (DNA) sequence, usually in vitro.<|separator|>
  75. [75]
  76. [76]
    Michael Smith – Facts - NobelPrize.org
    Michael Smith developed a method by which combined DNA building blocks could be artificially bonded with DNA molecules that were then inserted into an organism.
  77. [77]
    The Development of Site-directed Mutagenesis by Michael Smith
    Sep 29, 2006 · In recognition of his research, Smith was awarded one-half of the 1993 Nobel Prize in Chemistry “for his fundamental contributions to the ...
  78. [78]
    Site Directed Mutagenesis by PCR - Addgene Blog
    Aug 2, 2016 · Site directed mutagenesis is a highly versatile technique that can be used to introduce specific nucleotide substitutions (or deletions) in a tailored manner.
  79. [79]
    Site-directed Mutagenesis Methods | IDT
    Jul 18, 2022 · Learn efficient site-directed mutagenesis methods and techniques, including PCR and primer extension, to precisely alter DNA sequences.
  80. [80]
    A high-efficiency method for site-directed mutagenesis of large ...
    May 17, 2021 · We show that the SMLP method is highly efficient in site-directed mutagenesis and has a greater advantage than the conventional methods tested ...Results · Pcr Product Purification... · Plasmids Preparation And Dna...
  81. [81]
    A simple method for site-directed mutagenesis using the polymerase ...
    The method is based on the amplification of the entire plasmid using primers that include the desired changes. The method is rapid, simple in its execution, and ...
  82. [82]
  83. [83]
    Applications of Protein Engineering and Directed Evolution in Plant ...
    Site saturation mutagenesis targets one or a few specific codons from the gene sequence (Fig. 1B) and introduce all possible amino acid substitutions at those ...
  84. [84]
    DiRect: Site-directed mutagenesis method for protein engineering ...
    We developed a new PCR-based mutagenesis method, DiRect, that achieves high performance in product quality (≥99% substitution) without recombinant DNA ...Missing: history techniques
  85. [85]
    CRISPR Timeline | Broad Institute
    This timeline presents a concise history of the seminal contributions and the scientists who pushed this field forward, from the initial discovery to the first ...
  86. [86]
    The CRISPR tool kit for genome editing and beyond - Nature
    May 15, 2018 · CRISPR-Cas9 is no longer just a gene-editing tool; the application areas of catalytically impaired inactive Cas9, including gene regulation, ...
  87. [87]
    CRISPR/Cas9 therapeutics: progress and prospects - Nature
    Jan 16, 2023 · This paper reviews the current developments in three aspects, namely, gene-editing type, delivery vector, and disease characteristics.
  88. [88]
    Application of CRISPR-Cas9 genome editing technology in various ...
    A study demonstrated the application of CRISPR-Cas9 in activating or repressing specific genes, providing a valuable tool for functional genomics studies [38].
  89. [89]
    Chemical engineering of CRISPR–Cas systems for therapeutic ...
    Dec 17, 2024 · This Review focuses on chemical modification and engineering approaches for gRNAs to enhance or enable CRISPR-based therapeutics, emphasizing Cas9 and Cas12a ...
  90. [90]
    The hidden risks of CRISPR/Cas: structural variations and genome ...
    Aug 5, 2025 · The push for greater precision in genome editing has led to intense efforts to enhance HDR, the preferred pathway for precise gene modifications ...
  91. [91]
    Beyond CRISPR: A guide to the many other ways to edit a genome
    Aug 8, 2016 · NgAgo is just one of a growing library of gene-editing tools. Some are variations on the CRISPR theme; others offer new ways to edit genomes.
  92. [92]
    Engineering is evolution: a perspective on design processes to ...
    Apr 29, 2024 · Proteins have been extensively designed using directed evolution. Sequence lengths range from peptides of a few amino acids, which can ...
  93. [93]
    Historical Background and Biotechnology Milestones of ... - Biointron
    Sep 29, 2025 · The first therapeutic recombinant protein, human insulin, was produced in E. coli by Genentech in 1978 and approved by the FDA in 1982. This ...
  94. [94]
    Protein Design: Past, Present, and Future - PMC - NIH
    Here we briefly survey some past milestones in protein design, in addition to highlighting recent progress and future aspirations.
  95. [95]
    Accelerated enzyme engineering by machine-learning guided cell ...
    Jan 20, 2025 · Informed training set design enables efficient machine learning-assisted directed protein evolution. Cell Syst. 12, 1026–1045.e7 (2021) ...<|separator|>
  96. [96]
    Review article Generative artificial intelligence for enzyme design
    AI-based methods were used for enzyme discovery and design. We review the recent advances in generative AI models for enzyme design, with a particular focus ...
  97. [97]
    Synzymes: The Future of Modern Enzyme Engineering
    Jul 5, 2025 · By evaluating both the innovations and current limitations in synzyme research, this article outlines the growing potential of synthetic enzymes ...
  98. [98]
    Protein engineering strategies for rational immunogen design - Nature
    Dec 17, 2021 · In this review, we discuss various protein engineering strategies used to develop immunogens against rapidly evolving pathogens, and how ...
  99. [99]
    New advances in protein engineering for industrial applications - NIH
    Moreover, engineered enzymes have been employed in industries to detoxify pollutants such as plastics [5].
  100. [100]
    Enzyme engineering and its industrial applications - PubMed
    This review features engineered enzymes that have been applied in food, animal feed, pharmaceuticals, medical applications, bioremediation, biofuels, and ...
  101. [101]
    Synthetic Biology - National Human Genome Research Institute
    Aug 14, 2019 · Synthetic biology is a field of science that involves redesigning organisms for useful purposes by engineering them to have new abilities.
  102. [102]
    [PDF] A brief history of synthetic biology - Bashor Lab
    Apr 1, 2014 · The founding of the field of synthetic biol- ogy near the turn of the millennium was based on the transformational assertion that engineering ...
  103. [103]
    Synthetic biology | Nature Reviews Genetics
    Jul 1, 2005 · Synthetic biology has used metabolic-pathway design and genetic elements to develop organisms that can synthesize important chemicals, such as ...
  104. [104]
    A retrosynthetic biology approach to metabolic pathway design for ...
    Aug 5, 2011 · In this work we present a retrosynthetic approach to the production of therapeutics with the goal of developing an in situ drug delivery device in host cells.
  105. [105]
    The automated Galaxy-SynBioCAD pipeline for synthetic biology ...
    Aug 29, 2022 · Here we introduce the Galaxy-SynBioCAD portal, a toolshed for synthetic biology, metabolic engineering, and industrial biotechnology.
  106. [106]
    A machine learning Automated Recommendation Tool for synthetic ...
    Sep 25, 2020 · A tool that leverages machine learning and probabilistic modeling techniques to guide synthetic biology in a systematic fashion.
  107. [107]
    Designing pathways for bioproducing complex chemicals by ...
    May 24, 2025 · We present SubNetX, a method for exploring large networks of reactions to find an optimal pathway for the synthesis of a target compound. An ...
  108. [108]
    Data-driven synthetic microbes for sustainable future - Nature
    Jul 7, 2025 · These insights, coupled with predictive modeling, enable synthetic biology to precisely design and control metabolic pathways in engineered ...
  109. [109]
    Metabolic Engineering and Synthetic Biology - Frontiers
    Mar 3, 2019 · Metabolic engineering seeks for the optimization of cellular processes, endemic to a specific organism, to produce a compound of interest from a substrate.<|separator|>
  110. [110]
    Synthetic metabolism approaches: A valuable resource for systems ...
    In this article, we present state-of-the-art synthetic biology approaches that focus on the engineering of synthetic metabolism in microbial hosts.
  111. [111]
    Polymerase Chain Reaction (PCR) - StatPearls - NCBI Bookshelf - NIH
    Jul 7, 2025 · PCR enables precise detection and analysis of amplified DNA and has become a cornerstone of biomolecular research.[2]. This technique targets ...
  112. [112]
    Research Techniques Made Simple: Polymerase Chain Reaction ...
    PCR is a very sensitive technique that allows rapid amplification of a specific segment of DNA. PCR makes billions of copies of a specific DNA fragment or gene, ...
  113. [113]
    PCR Methods - Top Ten Strategies | Thermo Fisher Scientific - US
    May 18, 2016 · This video explains the basics of endpoint PCR, quantitative PCR, and digital PCR; and their typical applications.
  114. [114]
  115. [115]
    Full article: Isothermal amplifications – a comprehensive review on ...
    This review pays special attention to the various isothermal amplification methods by classifying them based on the mechanistic characteristics.
  116. [116]
    Nucleic Acid Amplification - an overview | ScienceDirect Topics
    These include methods such as rolling circle amplification (RCA), duplex-specific nuclease (DSN)-based amplification, loop-mediated isothermal amplification ( ...
  117. [117]
    CRISPR-based diagnostics | Nature Biomedical Engineering
    Jul 16, 2021 · We provide a rundown of the rapidly expanding toolbox for CRISPR-based diagnostics, in particular the various assays, preamplification strategies and readouts.
  118. [118]
    State-of-the-art signal amplification strategies for nucleic acid and ...
    We critically analyze the state-of-the-art signal amplification strategies for various nucleic acid and non-nucleic acid bioanalytes to improve the analytical ...
  119. [119]
    Recent Developments in Bioprocessing of Recombinant Proteins
    This review summarizes recent developments in the bioprocessing of recombinant proteins, including in various expression systems, bioprocess development,Missing: 2020-2025 | Show results with:2020-2025
  120. [120]
    Progress in fed-batch culture for recombinant protein production in ...
    Jan 17, 2023 · To achieve better cell growth and high-yield recombinant protein, fed-batch culture is typically used for recombinant protein production in CHO cells.
  121. [121]
    Comprehensive evaluation of the capacities of microbial cell factories
    Mar 24, 2025 · We comprehensively evaluate the capacities of various microbial cell factories and propose strategies for systems metabolic engineering steps.
  122. [122]
    Recent advances in recombinant production of soluble proteins in E ...
    Jan 16, 2025 · Alternative approaches aimed at calibrating translation efficiency could also be expected to optimize recombinant protein production.Missing: bioprocess | Show results with:bioprocess
  123. [123]
    Exploring Principles of Bioreactor Scale-Up - BioProcess International
    Feb 7, 2024 · Several factors contribute to the difficulty of scaling. They include the complexity of biological systems and the heterogeneous (hydrodynamic ...Missing: biomolecular | Show results with:biomolecular
  124. [124]
    Risks and Control Strategies of Scale-up in Purification Process
    Scale-up risks include material issues, process selection, equipment differences, and practical constraints. Tools like GAP analysis and FMEA are recommended ...Missing: biomolecular | Show results with:biomolecular
  125. [125]
    Catalysts and Enzymes in Biofuel Production
    Sep 13, 2023 · Biodiesel: The enzymes lipase and phospholipase are the significant players in biodiesel. A few companies are commercializing biodiesel produced ...
  126. [126]
    Advancing recombinant protein production by bacteria: strategies ...
    Oct 17, 2025 · Advancements ... Table 6 summarizes key genetic engineering strategies for optimizing microbial hosts to enhance recombinant protein production.
  127. [127]
    How industrial enzymes have changed your life in more ways than ...
    Mar 4, 2020 · None other than Procter and Gamble's Tide laundry detergent. The story of how protein engineering was applied to enzyme production and brought ...
  128. [128]
    Recent advances in metabolic engineering of microorganisms ... - NIH
    Over the past years, lignocellulosic biomass has become the most preferred raw material for the production of biofuels, such as fuel, alcohol, biodiesel, and ...2.2. Lipid Engineering · 4.1. Bacteria · 4.3. Yeasts<|separator|>
  129. [129]
    Scale-up of microbial processes: Impacts, tools and open questions
    Scale-up of microbial processes involves transferring lab-scale processes to large-scale production, which can cause performance losses and has biological, ...Missing: biomolecular | Show results with:biomolecular
  130. [130]
  131. [131]
    Recombinant pharmaceuticals from microbial cells: a 2015 update
    Feb 9, 2016 · Up to date, around 650 protein drugs have been worldwide approved, among which about 400 are obtained by recombinant technologies.Cell Factories · Trends In Protein... · Protein Drugs For Cancer...
  132. [132]
    Therapeutic proteins: developments, progress, challenges, and ...
    Human insulin was the first recombinant therapeutic protein, and its discovery in 1970 has increased interest in and expectations for the creation of new ...
  133. [133]
    Synthetic Biology in the Engineering of CAR-T and CAR-NK Cell ...
    Synthetic Biology in CAR Engineering. CARs are synthetic receptors that enable immune cells to recognize and initiate antigen-specific cytotoxic responses (22).
  134. [134]
    Recent advances in CAR T-cell engineering using synthetic biology
    This book chapter sheds light on the remarkable progress made in the development of CAR T-cell therapies using synthetic biology, providing hope for cancer ...
  135. [135]
    Gene regulation technologies for gene and cell therapy
    May 7, 2025 · Luxturna, approved in 2017, was the first U.S. Food and Drug Administration (FDA)-approved gene therapy, utilizing adeno-associated virus (AAV) ...
  136. [136]
    Approved Cellular and Gene Therapy Products - FDA
    Aug 15, 2025 · This FDA list includes approved cellular and gene therapy products such as ABECMA, ADSTILADRIN, and ALLOCORD, among others.Abecma · Adstiladrin · Zolgensma · AmtagviMissing: biomolecular | Show results with:biomolecular
  137. [137]
    FDA Approves First Gene Therapies to Treat Patients with Sickle ...
    Dec 8, 2023 · The FDA approved the first cell-based gene therapies, Casgevy and Lyfgenia, for the treatment of sickle cell disease in patients 12 years ...Missing: biomolecular | Show results with:biomolecular
  138. [138]
    Making the Next Generation of Therapeutics: mRNA Meets Synthetic ...
    Sep 6, 2023 · The development of mRNA-based therapeutics centers around the natural functioning of mRNA molecules to provide the genetic information required for protein ...<|control11|><|separator|>
  139. [139]
    Engineering Biomolecular Systems for Therapeutic Applications
    It delves into the design principles, techniques, and examples of biomolecular systems employed in drug delivery, gene therapy, immunotherapy, and tissue ...
  140. [140]
    The impact of Genetically Modified (GM) crops in modern agriculture
    GM crops are promising to mitigate current and future problems in commercial agriculture, with proven case studies in Indian cotton and Australian canola.
  141. [141]
    Genetically modified organisms: adapting regulatory frameworks for ...
    Oct 20, 2022 · The most widely cultivated GM crops are soybean, maize, cotton, and canola, with an area of 188.6 million hectares, which corresponds to 99% of ...
  142. [142]
    How GMO Crops Impact Our World | FDA
    Mar 5, 2024 · The GMO papaya, called the Rainbow papaya, is an example of a GMO crop developed to be resistant to a virus. When the ringspot virus threatened ...
  143. [143]
    Protein biopesticide may be next wave in pest control - Nature
    A new type of protein-based biopesticide that gives a wide range of plants broad resistance to pests was approved by the US Environmental Protection Agency ...
  144. [144]
    Protein engineering and plants: the evolution of sustainable ...
    Feb 27, 2023 · Applying protein engineering to accelerate the development of improved crop varieties that can produce more while using less is a promising approach.
  145. [145]
    Genetically Engineered Crops - Experiences and Prospects
    May 17, 2016 · Effects on agriculture. The available evidence indicates that GE soybean, cotton, and maize have generally had favorable economic outcomes for ...
  146. [146]
    [PDF] Recent advances in the bioremediation of persistent organic ...
    Recent advances include using biomolecular engineering, rational design, and directed evolution to engineer microorganisms for bioremediation of POPs like PAHs ...
  147. [147]
    Synthetic biology tools for environmental protection - ScienceDirect
    This review highlights synthetic biology approaches that can help environmental protection by providing remediation systems capable of sensing and responding ...
  148. [148]
    Emerging contaminants bioremediation by enzyme and nanozyme ...
    Apr 29, 2023 · Oxidoreductases and hydrolases are among the most prominent enzymes applied for bioremediation processes.Missing: pollutants | Show results with:pollutants
  149. [149]
    Engineering biology applications for environmental solutions - Nature
    Apr 14, 2025 · Engineering biology is applicable to the detection and degradation of pollutants, greenhouse gas sequestration, and conversion of waste streams, ...<|separator|>
  150. [150]
    Microbial Enzymes Used in Bioremediation - Wiley Online Library
    Feb 9, 2021 · The most representative enzymes involved in bioremediation include cytochrome P450s, laccases, hydrolases, dehalogenases, dehydrogenases, proteases, and ...Abstract · Introduction · Some Microbial Enzymes... · Conclusions<|control11|><|separator|>
  151. [151]
    Life at the interface: Engineering bio‐nanomaterials through ...
    May 9, 2024 · Interfacial biomolecular assembly is fundamental to biotic homeostasis. Exploiting and engineering these natural phenomena can produce an ...
  152. [152]
    Important applications of DNA nanotechnology combined with ... - NIH
    Feb 25, 2025 · In biosensing applications, DNA nanstructures capitalize on their high molecular recognition, signal amplification, and transduction ...
  153. [153]
    Exploring the diverse biomedical applications of programmable and ...
    Aug 23, 2023 · This paper focuses on the potential of DNA nanomaterials, which offer scalability, programmability, and functionality.
  154. [154]
    Computational design of bifaceted protein nanomaterials - Nature
    Jul 31, 2025 · Self-assembling bifaceted materials have been constructed from a wide range of materials, including metals, silicon dioxide, titanium dioxide, ...
  155. [155]
    Bottom-up synthesis of protein-based nanomaterials from ...
    Feb 21, 2020 · We previously developed a unique approach for self-assembly of nanomaterials based on engineering β-solenoid proteins (BSPs) to polymerize into micrometer- ...
  156. [156]
    Engineering Nano–Bio Interfaces from Nanomaterials to ...
    In this Account, we outline our recent progress in surface engineering and structural engineering to improve nano–bio interactions and the performance of ...
  157. [157]
    Hierarchical Self-Assembly of Proteins Through Rationally Designed ...
    Scientists can mediate the self-assembly of natural functional proteins, including enzymes (Gao et al., 2014), fluorescent proteins (Li Z. et al., 2019), etc., ...Abstract · Why Protein Self-Assembly · Biofunctionalization of Protein...
  158. [158]
    An Introduction to Biotechnology - PMC - NIH
    It is an amalgamation of biological science with engineering whereby living organisms or cells or parts are used for production of products and services. The ...
  159. [159]
    Biotechnology - Homeland Security
    Biotechnology is the integration of the life sciences and engineering with the goal of harnessing the power of biological molecules, cells, and even whole ...
  160. [160]
    What is Bioengineering?
    Bioengineering is a discipline that applies engineering principles of design and analysis to biological systems and biomedical technologies.Missing: biomolecular | Show results with:biomolecular
  161. [161]
    Bioengineering - Collection Development Guidelines of the ... - NCBI
    Oct 5, 2012 · Definition: The application of experimental and analytical techniques based on the engineering sciences to the development of biologics, ...
  162. [162]
    Bioengineers and Biomedical Engineers - Bureau of Labor Statistics
    Bioengineers and biomedical engineers combine engineering principles with sciences to design and create equipment, devices, computer systems, and software.
  163. [163]
    Biotechnology and Bioengineering - Wiley Online Library
    Biotechnology and Bioengineering provides an international forum for biotech researchers. As the first biotechnology journal dedicated to the field.Author Guidelines · Issues · Editorial Board · Current Issue
  164. [164]
    Current Developments in Biotechnology and Bioengineering
    This book covers new developments in industrial biotechnology and bioengineering, focusing on production, isolation, and purification of industrial products ...
  165. [165]
    Biotechnology | NSF - National Science Foundation
    The US National Science Foundation's investments in biotechnology have accelerated scientific discovery and enabled the use of living things to create goods ...
  166. [166]
    Chemical and Biomolecular Engineering Program
    Chemical and biomolecular engineers develop technology in drug design & delivery, biotechnology, nanotechnology, alternative energy, and more.
  167. [167]
    Bioreactors — Introduction to Chemical and Biological Engineering
    They are vessels in which a chemical process is carried out which involves organisms or biochemically active substances derived from such organisms.
  168. [168]
    Bioreactor Design - an overview | ScienceDirect Topics
    The bioreactor design is the process of creating a device that can provide a controlled environment for cell growth and differentiation.
  169. [169]
    Bioreactor design and validation for manufacturing strategies ... - NIH
    Jul 19, 2021 · The bioreactor system is designed to provide the appropriate tissue-specific physiological conditions for the seeded cells to acclimate to the ...
  170. [170]
    Enzyme Kinetics - Structure - Function - Michaelis-Menten Kinetics
    Apr 8, 2024 · Enzyme kinetics is the study of enzyme reaction rates and the conditions which affect them. In this article, we will discuss the structure and function of ...
  171. [171]
    Emerging biomaterials for downstream manufacturing of therapeutic ...
    Sep 1, 2019 · Downstream processing is considered one of the most challenging phases of industrial manufacturing of therapeutic proteins, accounting for a ...
  172. [172]
    Current PAT Landscape in the Downstream Processing of ... - NIH
    May 29, 2025 · Downstream processing (DSP) begins with the harvesting and clarification of the target product from the upstream fermentation or cell culture ...
  173. [173]
    Isolation and purification of biotechnological products. - MAB - KIT
    Nov 25, 2011 · Downstream processing, however, still accounts for the major part of production costs. This is mainly due to the high demands on purity and thus ...
  174. [174]
    Kinetics of Biochemical Reactions - Wiley Online Library
    Sep 15, 2017 · Enzymes are the engines of biochemical processes in living systems and biotechnology. They work in concert, forming pathways and reaction networks.
  175. [175]
    Bioreactor Design and Control in the Biopharmaceutical Industry
    Jun 13, 2024 · Within the pharmaceutical industry, bioreactors refer to vessels and containers used to store microorganisms such as bacteria, cells, and algae.
  176. [176]
    Undergraduate Curriculum - Chemical and Biomolecular Engineering
    The program's versatility allows our students to excel in diverse careers in the chemical industries, research, medicine, law, government, and education.
  177. [177]
    Bachelor of Science in Chemical and Biomolecular Engineering
    The curriculum offers two specialization options. The Biotechnology Option is for students who wish to focus their education on the biomolecular aspects of ...
  178. [178]
    Chemical Engineering: Biomolecular Engineering, BS
    The first two years of the Chemical Engineering curriculum provide a strong foundation in basic sciences through Physics, Mathematics, Chemistry, an ...
  179. [179]
    Program: Chemical and Biomolecular Engineering, B.S. - New York ...
    The undergraduate program in chemical and biomolecular engineering provides a strong foundation in mathematics and the physical, chemical, and biological ...
  180. [180]
    2025 Best Chemical & Biomolecular Engineering Schools
    Massachusetts Institute of Technology is one of the best schools in the country for getting a degree in chemical & biomolecular engineering. MIT is a large ...
  181. [181]
    BME - Biomolecular Engineering - University of California Santa Cruz
    Course includes training in specific skills relevant to the specific sub-team and overall project, including lab-specific training (pcr, DNA electrophoresis ...
  182. [182]
    Biomolecular Engineering - Baskin School of Engineering
    Explore potential Biomolecular Engineering career paths · Research Scientist · Product Manager · Engineer · Field Technician · Process Engineer · IT Specialist/ ...
  183. [183]
    Careers in Chemical & Biomolecular Engineering
    In short, chemical and biomolecular engineers extend engineering analysis to processes at the molecular scale. How Do I Know if Chemical Engineering is a Good ...
  184. [184]
    Career Paths | Department of Chemical and Biomolecular Engineering
    Career Paths · Advanced Materials · Biochemical and Biomedical Engineering · Chemical Process Design and Construction · Chemical Process Industries
  185. [185]
    Biomolecular Engineering Jobs, Employment | Indeed
    468 Biomolecular Engineering jobs available on Indeed.com. Apply to Scientist, Chemical Engineer, Senior Scientist and more!
  186. [186]
    $$20-$38/hr Biomolecular Engineering Jobs in Chicago, IL
    Roles include research scientist, process engineer, quality control analyst, and regulatory affairs specialist.
  187. [187]
    Where do Chemical Engineers Work? - Rice ChBE
    Chemical engineers in the biotechnology industry develop and design the processes to grow, handle, and harvest living organisms and their byproducts.<|separator|>
  188. [188]
    Bioengineers and Biomedical Engineers - Bureau of Labor Statistics
    Industry profile for Bioengineers and Biomedical Engineers: ; Pharmaceutical and Medicine Manufacturing, 1,810, 0.53, $ 51.55, $ 107,220 ; General Medical and ...Missing: biomolecular | Show results with:biomolecular
  189. [189]
    [PDF] Biosafety in Microbiological and Biomedical Laboratories—6th Edition
    We wish to emphasize that the sixth edition of BMBL remains an advisory document recommending best practices for the safe conduct of work in biomedical and ...Missing: biomolecular | Show results with:biomolecular
  190. [190]
    High-risk human-caused pathogen exposure events from 1975-2016
    Aug 4, 2021 · This paper reports on a dataset of 71 incidents involving either accidental or purposeful exposure to, or infection by, a highly infectious pathogenic agent.<|separator|>
  191. [191]
    Reducing potential dual-use risks in synthetic biology laboratory ...
    Nov 7, 2024 · This study provides a framework for quantifying potential risk factors in SynBio research and lays a theoretical foundation for future laboratory safety ...Missing: peer- | Show results with:peer-
  192. [192]
    [PDF] United States Government Policy for Institutional Oversight of Life ...
    Sep 24, 2014 · “Dual use research of concern” (DURC) is life sciences research that, based on current understanding, can be reasonably anticipated to provide ...Missing: biomolecular | Show results with:biomolecular
  193. [193]
    A Biohacker Regrets Publicly Injecting Himself With CRISPR
    Feb 20, 2018 · Josiah Zayner watched a biotech CEO drop his pants at a biohacking conference and inject himself with an untested herpes treatment, he realized things had gone ...
  194. [194]
    Mapping variation in dual use risk assessments of synthetic biology ...
    Aug 13, 2025 · Expert opinions about the overall percentage of synthetic biology research with DURC potential ranged from near 0% to near 100%. As a group, ...
  195. [195]
    Regulation and management of the biosecurity for synthetic biology
    Mar 25, 2022 · The regulatory framework for SynBio biosecurity risks is a hierarchy that consists of three levels from top to bottom: the national level, the ...
  196. [196]
    Safety by design: Biosafety and biosecurity in the age of synthetic ...
    Mar 17, 2023 · Effective regulatory and technological frameworks need to be developed and deployed to manage these emerging biosafety and biosecurity risks.
  197. [197]
    Bioethical issues in genome editing by CRISPR-Cas9 technology
    Some of the ethical dilemmas of genome editing in the germline arise from the fact that changes in the genome can be transferred to the next generations.Missing: biomolecular | Show results with:biomolecular
  198. [198]
    Moral reasons to edit the human genome - Journal of Medical Ethics
    The Nuffield report was notable for finding that HGE could be morally permissible, even in cases of human enhancement.Missing: cons | Show results with:cons
  199. [199]
    The CRISPR babies controversy: Responsibility and regulation in ...
    May 28, 2020 · The genome editing of human embryos by He Jianjui and the announcement to do so by Denis Rebrikov should spur the research community into ...
  200. [200]
    Archive: What's So Controversial About the First Gene-Edited Babies ...
    Nov 30, 2018 · A Chinese researcher, He Jiankui, announced the birth of the first babies who had their DNA edited as embryos. He said he used CRISPR to ...
  201. [201]
    Ethical issues related to research on genome editing in human ...
    Although the potential advantages of clinical germline genome editing (GGE) over currently available methods are limited, the implementation of GGE in the ...
  202. [202]
    Eugenics and Scientific Racism
    May 18, 2022 · Eugenics is the scientifically inaccurate theory that humans can be improved through selective breeding of populations. Eugenicists believed in ...
  203. [203]
    From Public Eugenics to Private Eugenics: What Does the Future ...
    Historically, public eugenics could prevent undesirable conceptions by restricting marriage between people with unwanted genes. Science eventually evolved to ...
  204. [204]
    Risks and benefits of human germline genome editing - NIH
    For decades, potential interventions in the human germline for clinical purposes have been regarded as ethically impermissible. Recently, this strict view has ...
  205. [205]
    Genetic therapies, human genetic enhancement, and … eugenics?
    Jul 4, 2019 · Such genetic manipulation has been extended to genome editing studies in the human embryo identified to be at risk for a potentially dangerous ...
  206. [206]
    Ethical Challenges of Germline Genetic Enhancement - PMC - NIH
    We identify procreative beneficence, genetic disassociation, gender selection, the value of disability, embryo chimerization, and the psychosocial inequality.Missing: biomolecular | Show results with:biomolecular
  207. [207]
    Genetic engineering of animals: Ethical issues, including welfare ...
    The genetic engineering of animals has increased significantly in recent years, and the use of this technology brings with it ethical issues, some of which ...Research Animals · Concerns For Animal Welfare · Beyond Animal WelfareMissing: biomolecular | Show results with:biomolecular
  208. [208]
    Gene Editing: Developments, Ethical Considerations, and Future ...
    Two landmark gene editing experiments where CRISPR was used in modifying human embryos have sparked ethical debates and brought the convention's framework into ...2. Developments · 4. Opinions · 5. Future DirectionsMissing: biomolecular | Show results with:biomolecular
  209. [209]
    New AI Collaboration Aims to Accelerate R&D of Specialty Chemicals
    Sep 2, 2022 · Eastman and the Department of Chemical and Biomolecular Engineering will accelerate the R&D of specialty chemicals ... R&D costs exceeding $100 ...
  210. [210]
  211. [211]
    Estimating the Cost of Industry Investment in Drug Research and ...
    We review and synthesize papers estimating drug R&D costs incurred by industry. We find a substantial range of per-drug costs, from $113 million to just over $6 ...Missing: biomolecular | Show results with:biomolecular
  212. [212]
    Higher Education R&D Expenditures Increased 11.2%, Exceeded ...
    Nov 25, 2024 · Research and development (R&D) spending by academic institutions increased 11.2% in FY 2023, the largest growth rate in current dollars since FY 2003.Missing: biomolecular | Show results with:biomolecular
  213. [213]
    Barriers to commercial deployment of biorefineries: A multi-faceted ...
    Jun 30, 2024 · This review synthesizes current literature to provide a comprehensive analysis of key multi-dimensional barriers inhibiting the scale-up of biorefineries.
  214. [214]
    From Lab to Market Challenges in Scaling Biotech Innovations
    Mar 12, 2025 · Scaling biotech faces scientific, regulatory, and financial hurdles, including unpredictable biological systems, rigorous regulations, and high ...<|separator|>
  215. [215]
    Innovation under Regulatory Uncertainty: Evidence from Medical ...
    This paper explores how the regulatory approval process affects innovation incentives in medical technologies.
  216. [216]
    Analysis of the Innovation Trend in Cell-Free Synthetic Biology - PMC
    Despite the advantages, CFPS still has challenges to widespread commercial adoption including costs, enzyme stability, reaction duration, inefficiency of ...Missing: constraints | Show results with:constraints
  217. [217]
    New Report Recommends Renewed Us-federal Investment in ...
    Feb 9, 2022 · The report also recommends that federal research investments in biomolecular engineering focus on advancing personalized medicine and ...
  218. [218]
    Twenty-eight years of GM Food and feed without harm: why not ...
    Dec 31, 2024 · No new GMO has been proven to be a hazard or cause harm to human consumers. These modifications have improved crop efficiency, reduced losses to insect pests.
  219. [219]
    A Meta-Analysis of the Impacts of Genetically Modified Crops - NIH
    Nov 3, 2014 · The meta-analysis reveals robust evidence of GM crop benefits for farmers in developed and developing countries. Such evidence may help to ...
  220. [220]
    Do foods made with GMOs pose special health risks?
    May 2, 2022 · No evidence has validated that eating food with GMO ingredients is harmful. · Scientists continue to study foods made from genetically engineered ...
  221. [221]
    Food, genetically modified - World Health Organization (WHO)
    May 1, 2014 · GM foods currently available on the international market have passed safety assessments and are not likely to present risks for human health.
  222. [222]
    Beyond the promise: evaluating and mitigating off-target effects in ...
    While ongoing clinical trials have not reported any adverse events linked to CRISPR off-target effects, it's vital to acknowledge that the long-term impact of ...Abstract · Introduction · Approaches to reduce off... · Pre-clinical assessment of...
  223. [223]
    CRISPR Clinical Trials: A 2024 Update - Innovative Genomics Institute
    Mar 13, 2024 · The experimental treatment uses CRISPR genome-editing molecules to target the HIV DNA sequence stored in the host cell genome. In ...
  224. [224]
    Safety by design: Biosafety and biosecurity in the age of synthetic ...
    Feb 10, 2023 · Effective regulatory and technological frameworks need to be developed and deployed to manage these emerging biosafety and biosecurity risks.
  225. [225]
    [PDF] Long Term Follow-Up After Administration of Human Gene Therapy ...
    In general, when the risk of delayed adverse events is low following exposure to a GT product, LTFU observations are not recommended. We consider the assessment ...
  226. [226]
    Safety of Adeno-associated virus-based vector-mediated gene ...
    Jan 14, 2022 · The outcome data showed no significant adverse events when vectors were given to patients at lower doses (1.4 × 1011 vgs/patient), while a ...Missing: rates | Show results with:rates
  227. [227]
    The convergence of AI and synthetic biology: the looming deluge
    Jul 1, 2025 · Synthetic biology advances have been powered by improvements in reading, writing, and editing DNA. The use of AI in synthetic biology has ...<|separator|>
  228. [228]
    Machine Learning and Deep Learning in Synthetic Biology
    Feb 19, 2024 · Machine learning (ML), particularly deep learning (DL), has made rapid and substantial progress in synthetic biology in recent years.
  229. [229]
    AlphaFold - Google DeepMind
    AlphaFold has revealed millions of intricate 3D protein structures, and is helping scientists understand how all of life's molecules interact.
  230. [230]
    Google DeepMind's AlphaFold 4 Unveiled: Faster, Smarter Protein ...
    Aug 2, 2025 · The article will review how AlphaFold 4 builds upon that of its predecessor and will explore potential impacts of protein structure prediction ...
  231. [231]
    Generative AI for drug discovery and protein design: the next frontier ...
    This review systematically delineates the theoretical underpinnings, algorithmic architectures, and translational applications of deep generative models.
  232. [232]
    Generative AI tool marks a milestone in biology | Stanford Report
    Feb 19, 2025 · Now, scientists have developed a generative AI tool that can predict the form and function of proteins coded in the DNA of all domains of life, ...
  233. [233]
    Machine Learning Takes on Synthetic Biology: Algorithms Can ...
    Sep 25, 2020 · In “ART: A machine learning Automated Recommendation Tool for synthetic biology,” led by Radivojevic, the researchers presented the algorithm, ...
  234. [234]
    Biomolecular Interaction Prediction: The Era of AI
    Jul 16, 2025 · AI and deep learning methods have made significant advancements in biomolecular interaction prediction, providing substantial improvements over ...
  235. [235]
  236. [236]
    Advancements in protein structure prediction: A comparative ...
    This review provides a comprehensive analysis of AlphaFold (AF) and its derivatives (AF2 and AF3) in protein structure prediction.
  237. [237]
    Applications of multiplexed CRISPR-Cas for genome engineering
    Jul 31, 2025 · Due to its simplicity, CRISPR-Cas is recognized as the best candidate for multiplexed genome editing. With simultaneous targeting, efficient ...
  238. [238]
    A New Era in Genetic Engineering: Breakthroughs in Gene Editing ...
    Jan 8, 2025 · Penn Engineers developed mvGPT, a tool that can simultaneously and independently edit multiple genes and regulate their expression.
  239. [239]
    Yale genome engineers expand the reach and precision of human ...
    Jun 9, 2025 · A new Yale study advances the ability of genome engineers to edit multiple DNA sites by threefold, and helps prevent unwanted mutations in the process.Missing: techniques breakthroughs biomolecular
  240. [240]
    CRISPR: A Biotech Breakthrough - NSF Impacts
    In 2018, NSF-funded researchers used CRISPR to change pairs of genes linked to cancer and then precisely tracked individual cells in the tumor that formed. This ...Share · Blockbuster Discovery · New Model System
  241. [241]
    Gene-editing system targets multiple organs simultaneously
    Jun 18, 2025 · The findings, published in Nature Biotechnology, could lead to new therapies for a variety of genetic diseases that affect multiple organs.Missing: techniques breakthroughs biomolecular
  242. [242]
    Advances in large-scale DNA engineering with the CRISPR system
    Sep 1, 2025 · Recent advances in the discovery and application of CRISPR effectors have enabled targeted gene modifications and large DNA engineering in vivo.Missing: breakthroughs biomolecular
  243. [243]
    Past, present, and future of CRISPR genome editing technologies: Cell
    Feb 29, 2024 · In this review, we discuss the current state of CRISPR gene editing technologies in both research and therapy, highlighting limitations that constrain them.Missing: biomolecular | Show results with:biomolecular
  244. [244]
    Construction and iterative redesign of synXVI a 903 kb synthetic ...
    Jan 20, 2025 · The Sc2.0 global consortium to design and construct a synthetic genome based on the Saccharomyces cerevisiae genome commenced in 2006, ...
  245. [245]
    Iterative SCRaMbLE for engineering synthetic genome modules and ...
    Aug 7, 2025 · In eukaryotes, the Synthetic Yeast Genome (Sc2.0) project is nearing completion, with almost all of the 16 synthetic chromosomes completed ...
  246. [246]
    minimising and re-functionalising genomes using synthetic biology
    Apr 8, 2023 · Here, we review the past and current genome minimisation and re-functionalisation efforts, with an emphasis on the latest advances facilitated by synthetic ...
  247. [247]
    Researchers take first steps to creating synthetic human genomes
    Jun 26, 2025 · A team of UK-based scientists are developing technology to create the first synthetic human chromosome. The ability to write large genomes ...Missing: 2020-2025 minimal
  248. [248]
    Final synthetic yeast chromosome unlocks new era in biotechnology
    Jan 22, 2025 · Scientists have completed construction of the final chromosome in the worlds' first synthetic yeast genome following more than a decade of work, ...
  249. [249]
    Scale-Up Challenges in Bioreactor Systems and How to Overcome ...
    May 9, 2025 · One of the primary challenges in scaling up bioreactors is the maintenance of optimal conditions for cell growth and product formation. In ...Missing: biomolecular | Show results with:biomolecular
  250. [250]
    Cell Culture Process Scale-Up Challenges for Commercial ... - NIH
    Feb 25, 2022 · However, the limited scalability of 2D platforms restricts manufacturing to a scale-out approach (as opposed to scale-up) to generate enough ...Missing: biomolecular | Show results with:biomolecular
  251. [251]
    Stabilization challenges and aggregation in protein-based ... - NIH
    Dec 11, 2023 · One of the primary challenges associated with the production, distribution and storage of protein-based drugs is the risk of protein aggregation ...
  252. [252]
    [PDF] Lever VC Insights - Scale-up Costs of Precision Fermentation
    Scaling precision fermentation introduces risk, and this document aims to estimate CAPEX for new facilities, not initial market entry, with a +40%/-25% ...Missing: biomolecular | Show results with:biomolecular
  253. [253]
    Achieving Scalable and Sustainable Precision Fermentation Using ...
    Feb 27, 2025 · Precision fermentation uses engineered microorganisms to produce biomolecules. PAT tools provide real-time data, and QbD ensures robust, ...
  254. [254]
    Current challenges and recent advances on the path towards ...
    Jul 26, 2023 · Compared to traditional batch manufacturing, continuous bioprocessing is more efficient, adjustable, and sustainable and has reduced capital ...
  255. [255]
    Upstream continuous processing: recent advances in production of ...
    This review covers recent advances in continuous processing over the last two years for biopharmaceutical production.
  256. [256]
    The science of fermentation (2025) | GFI - The Good Food Institute
    Precision fermentation can produce enzymes, flavoring agents, vitamins, natural pigments, and fats. Examples include Perfect Day's dairy proteins, Clara Foods' ...
  257. [257]
    Recent trends in the production of proteins by precision fermentation ...
    Precision fermentation optimizes plant and dairy proteins for sustainable food systems. •. Genetic engineering enhances protein functionality and sensory ...
  258. [258]
    Throughput-scalable manufacturing of SARS-CoV-2 mRNA ... - PNAS
    Aug 9, 2023 · We have developed a microfluidic chip that incorporates 1×, 10×, or 256× LNP-generating units that achieve scalable production rates of up to 17 L/h of ...
  259. [259]
    Development of mRNA manufacturing for vaccines and therapeutics
    Due to the novel nature of the mRNA platform, having a consensus around a conservative approach to scale-up design and quality controls proved to be invaluable ...
  260. [260]
    mRNA Vaccine Trends & Innovation - Cytiva
    Feb 18, 2025 · Using single-use technologies and continuous processing strategies could help scale and streamline production.
  261. [261]
    The Future is Continuous: Accelerating the Shift in Biomanufacturing
    The recent regulatory guidance and advances in enabling technologies are leading to movement in the biopharmaceutical industry toward continuous manufacturing.
  262. [262]
    Critical Assessment of Protein Engineering (CAPE): A Student ...
    Nov 7, 2024 · The Critical Assessment of Protein Engineering (CAPE) challenge addresses these issues through a student-focused competition, utilizing cloud computing and ...Introduction · Results and Discussion · Conclusions · Author Information
  263. [263]
    CRISPR Gene Therapy: Applications, Limitations, and Implications ...
    CRISPR-induced DSBs often trigger apoptosis rather than the intended gene edit (68). Further safety concerns were revealed when using this tool in human ...
  264. [264]
    Recent applications, future perspectives, and limitations of the ...
    Finally, we discuss current challenges including off-target activity, gene editing efficiency, delivery constraints, and immunogenicity and highlight emerging ...
  265. [265]
    Exploring the Potential and Challenges of CRISPR Delivery and ...
    May 27, 2024 · However, a critical limitation is their small packaging size of around 4.5–5 kb, posing a challenge for accommodating the large CRISPR/Cas9 gene ...
  266. [266]
    Challenges in delivery systems for CRISPR-based genome editing ...
    We will present an overall view on essential elements in CRISPR/Cas systems and the nanoparticle-based delivery carriers including advantages and challenges.
  267. [267]
    Challenges in Scaling Up Bioprocesses in Bioprocessing - IDBS
    May 22, 2025 · Scaling up bioprocesses faces challenges including maintaining product consistency, shear stress on cells, oxygen transfer issues, raw material ...Missing: purification | Show results with:purification
  268. [268]
    Why Some Biologics Manufacturing Processes Fail to Scale
    Aug 1, 2025 · Common Challenges in Scaling Biologics Manufacturing · 1. Biological Complexity and Variability · 2. Process Parameter Sensitivity · 3. Mass ...<|separator|>
  269. [269]
    Ten future challenges for synthetic biology - PMC - PubMed Central
    Aug 2, 2021 · A grand challenge for synthetic biology is to design the differentiation and specialisation of cells to enable effective and fruitful 'division ...
  270. [270]
    The ultimate challenge of synthetic biology
    Establishing synthetic cells from individual molecular components is one of the greatest scientific challenges of our time.
  271. [271]
    Synthetic Biology Market Size, Share, Growth & Forecast, 2032
    The global synthetic biology market size is projected to grow from $17.09 billion in 2025 to $63.77 billion by 2032, a CAGR of 20.7% during the forecast period.Missing: economic | Show results with:economic
  272. [272]
    Protein Engineering Market Size And Share Report, 2030
    The global protein engineering market size was estimated at USD 2.60 billion in 2023 and is expected to grow at a CAGR of 16.24% from 2024 to 2030.Missing: biomolecular | Show results with:biomolecular
  273. [273]
    [PDF] The U.S. Bioscience Industry:
    The US bioscience industry, represented by BIO, fosters innovation, contributes $2.9 trillion to the economy, and generates high-quality, high-wage jobs.
  274. [274]
    2025 Investment Report - SynBioBeta
    The report analyzes 900+ companies, 12,000+ investments, showing a 2024 rebound to $12.2B after a cooling period, with current levels above pre-pandemic norms.Missing: economic forecast
  275. [275]
    The Brutal Reality of the 2025 Biotech Job Market (and How to ...
    Mar 7, 2025 · The number of positions in biotech nosedived from about 19,000 in early 2022 to just 10,000 by late 2023—effectively cutting opportunities in ...Missing: biomolecular | Show results with:biomolecular
  276. [276]
    3 scenarios for how bioengineering could change our world in 10 ...
    Dec 21, 2020 · This risk can create significant threats with large potential consequences to public health, privacy or to environmental safety. Approaches such ...Missing: biomolecular | Show results with:biomolecular
  277. [277]
    Synthetic biology: A game changer for economic sustainability ...
    Feb 26, 2025 · Synthetic biology and other next-generation biotechnologies promise to revolutionise a range of industries, drive economic growth and contribute to solving ...Missing: forecast | Show results with:forecast
  278. [278]
    The opportunities and risks of bioengineering - Gates Cambridge
    Nov 24, 2017 · Dr Christian Boehm co-authors article on the long- and short-term opportunities and risks presented by bioengineering.Missing: biomolecular | Show results with:biomolecular<|separator|>
  279. [279]
    Engaging Biomedical Engineering in Health Disparities Challenges
    Further, joint biomedical- related engineering ventures have impacted US public policy [5], health initiatives [6] and community-based challenges [7], all of ...Missing: biomolecular | Show results with:biomolecular
  280. [280]
    Automated engineering of synthetic metabolic pathways for efficient ...
    In this review, we discuss how the success of the metabolic engineering pipeline often relies on retrobiosynthetic protocols able to identify promising ...<|separator|>
  281. [281]
    Challenges and opportunities in synthetic biology for chemical ...
    Challenges and opportunities in synthetic biology for chemical engineers. Yunzi Luo. Yunzi Luo. aDepartment of Chemical and Biomolecular Engineering ... financial ...
  282. [282]
    RePORT RePORTER - National Institutes of Health (NIH) |
    In this project, we will advance the capabilities of ePACE in two critical dimensions – scale and accessibility – through new hardware and fluidic technology ...
  283. [283]
    Breaking the Cost Barrier on Biomanufacturing | BCG
    Feb 27, 2024 · Synthetic biology is a difficult technology to master and especially tough to scale. Chemical and materials startups and their backers need to ...Missing: biomolecular | Show results with:biomolecular
  284. [284]
    Scaling up synthetic biology: challenges & solutions - Sagentia
    Scaling up SynBio production often requires extensive and multidisciplinary research, optimisation and iterative improvements.
  285. [285]
    Regulatory challenges and global trade implications of genome ...
    Jun 18, 2025 · This article analyzes the main regulatory challenges and their impact on global trade, proposing strategies for regulatory harmonization to promote ...Missing: reforms | Show results with:reforms
  286. [286]
    Regulatory challenges and global trade implications of genome ...
    Jun 19, 2025 · This article analyzes the main regulatory challenges and their impact on global trade, proposing strategies for regulatory harmonization to promote ...Missing: reforms | Show results with:reforms
  287. [287]
    Simplify Regulation for American Biotechnology Companies - Biotech
    Innovators have identified four key challenges within the U.S. biotechnology regulatory system. First, they face uncertainty about which agencies are ...Missing: biomolecular | Show results with:biomolecular
  288. [288]
    Frameworks and Strategies for Commercialization Success in the ...
    Mar 14, 2019 · Product/Technology Roadmap and Pipeline: · Pricing Strategy: · Promotion and Differentiation: · Fundraising: · Sales: · Structure As Strategy: ...
  289. [289]
    Enabling Broader Adoption of Biocatalysis in Organic Chemistry
    Jul 19, 2023 · Furthermore, we discuss ways to further lower the barriers for the adoption of biocatalysis by the broader synthetic organic chemistry ...
  290. [290]
    [PDF] Don't fail to scale: seizing the opportunity of engineering biology
    Jan 14, 2025 · Engineering biology is the design, scaling and commercialisation of biology- derived products and services, which can transform sectors or ...<|separator|>
  291. [291]
    Scaling-up Engineering Biology for Enhanced Environmental ... - NIH
    Jun 21, 2024 · Synthetic biology (SynBio) offers transformative solutions for addressing environmental challenges by engineering organisms capable of degrading pollutants.