Fact-checked by Grok 2 weeks ago

Pharmaceutical engineering

Pharmaceutical engineering is a multidisciplinary branch of engineering that integrates principles from chemical, , , and biomedical engineering to design, develop, optimize, and manufacture pharmaceutical products, processes, and facilities. It focuses on the research, , , testing, and large-scale production of medicinal drugs, ensuring they are safe, effective, and compliant with regulatory standards such as (GMP). This field plays a critical role in the pharmaceutical industry by bridging scientific discovery with practical manufacturing, enabling the efficient production of therapies ranging from traditional small-molecule drugs to advanced biopharmaceuticals like vaccines, gene therapies, and personalized medicines. Key aspects include process design using (QbD) principles, cleanroom technology for contamination control, for efficiency, recent advances incorporating for process optimization, and adherence to guidelines from bodies like the FDA and to minimize risks and accelerate time-to-market. The pharmaceutical industry has roots in early drug formulation efforts dating back to the 16th century, while pharmaceutical engineering emerged as a distinct discipline in the mid-20th century with the adoption of GMP standards by the World Health Organization in 1969. Today, it addresses modern challenges such as sustainability, digitalization, and innovations like 3D-printed medications and nanoparticle formulations, supporting an industry where approximately 80% of active pharmaceutical ingredients are sourced from regions including India, China, and the European Union as of 2023. Professionals in this field typically hold advanced degrees in engineering or related sciences and work in roles involving facility design, equipment validation, and process optimization to ensure reliable supply of life-saving treatments.

Overview

Definition and scope

Pharmaceutical engineering is an interdisciplinary field that applies principles from chemical, , and to the , , and of pharmaceutical products, including small-molecule drugs, biologics, and systems. This discipline focuses on creating processes that ensure the production of safe, effective, and high-quality medications while addressing challenges such as formulation stability and therapeutic efficacy. At its core, pharmaceutical engineering integrates scientific and methodologies to translate laboratory discoveries into scalable industrial processes. The scope of pharmaceutical engineering encompasses process design, optimization, and scale-up from laboratory to commercial production levels, ensuring efficient and reproducible manufacturing. A key aspect is the integration of Quality by Design (QbD) principles, which involve systematic risk assessment and predefined quality targets to build robustness into products and processes from the outset. This approach minimizes variability and enhances control over critical quality attributes during scale-up, facilitating seamless transitions from pilot to full-scale operations. Pharmaceutical engineering is inherently interdisciplinary, combining expertise in , , , and various domains to achieve safe, effective, and efficient production outcomes. Professionals in this field collaborate across disciplines to address complex interactions between drug formulation, manufacturing equipment, and biological responses. The primary objectives of pharmaceutical engineering include attaining product stability under various storage conditions, optimizing to ensure adequate absorption and therapeutic effect, and maintaining compliance with stringent regulatory standards for safety and efficacy. These goals are pursued through engineered solutions that enhance solubility, protect against , and align with guidelines from bodies like the FDA and .

Importance in healthcare

Pharmaceutical engineering significantly contributes to the global economy through its pivotal role in the , which is projected to reach a market size of USD 1.77 trillion in 2025. This sector supports millions of jobs worldwide; for example, in , direct exceeds 950,000 individuals and generates approximately three times more indirect jobs in upstream and downstream activities, while globally it supports millions of direct and indirect jobs, fostering and stability. Furthermore, the industry's activities contribute substantially to global GDP, adding around USD 2,295 billion through direct, indirect, and induced effects in 2022, highlighting its influence on national and international economic landscapes. In terms of health benefits, pharmaceutical engineering enables faster by optimizing processes, such as through practices and , which can reduce timelines by 25-50% by streamlining scale-up and phases. This is exemplified in the adoption of model-based approaches and digital twins that cut experimental requirements by over 50%, allowing quicker progression from to commercialization. Additionally, engineered processes ensure consistent product quality via methodologies like (QbD), which embeds quality controls to minimize variability, thereby improving outcomes through reliable and profiles that reduce adverse events and enhance therapeutic adherence. Pharmaceutical engineering plays a crucial societal role in addressing global health crises, particularly by facilitating rapid vaccine production during pandemics like COVID-19, where process engineering innovations enabled unprecedented scale-up of manufacturing capacities to deliver billions of doses within months. For instance, technology transfer and just-in-time manufacturing techniques were instrumental in accelerating production timelines and ensuring supply chain resilience amid surging demand. Beyond emergencies, it supports the management of chronic diseases through advanced engineered delivery systems, such as long-acting injectables and targeted nanoparticles, which provide sustained release and precise dosing to improve long-term disease control and quality of life for patients with conditions like diabetes and cancer. As an driver, pharmaceutical engineering facilitates the of affordable generics and biosimilars by refining and technologies, which enable cost-effective replication of complex molecules and biologics. This has led to substantial healthcare reductions, with oral generics often priced 80% lower than branded counterparts, making essential medications accessible in various markets. For biologics, biosimilars typically cost 50% less than originators, though savings can reach up to 80% in competitive environments, thereby alleviating financial burdens on healthcare systems and patients while promoting equitable access to treatments.

History

Early synthesis and development

The origins of pharmaceutical engineering trace back to the early 19th century, when systematic efforts to isolate and synthesize active pharmaceutical compounds began to shift from empirical herbalism to more structured chemical processes. A pivotal milestone occurred in 1804, when German pharmacist Friedrich Sertürner successfully isolated morphine from opium derived from the Papaver somniferum plant, marking the first isolation of a pure alkaloid and the inception of systematic drug synthesis. This achievement enabled precise dosing of the analgesic, mitigating the risks of variable potency in crude opium preparations, and laid the groundwork for alkaloid chemistry as a cornerstone of pharmaceutical development. By the late 19th century, chemists increasingly applied proto-engineering techniques to synthesize novel drugs, exemplified by Felix Hoffmann's work at . In 1897, Hoffmann synthesized acetylsalicylic acid (aspirin) by acetylating , producing a more tolerable alternative to sodium salicylate for pain relief, and purified the compound to achieve a stable, high-purity form suitable for therapeutic use. This process involved recrystallization to separate the product from impurities, representing an early integration of purification methods that foreshadowed engineering principles in drug production. Early efforts were hampered by manual techniques and small-scale reactions, which often resulted in inconsistent purity and low yields due to rudimentary equipment and variable raw material quality. In the , pharmaceutical relied on handicraft methods, with operations limited to workshops employing 20–30 workers, leading to variability that prompted efforts like the 1820 U.S. Pharmacopeia to address purity concerns. Limited availability of commercial reagents further compounded these issues, requiring chemists to prepare starting materials and yielding compounds of unpredictable potency. The transition toward pharmaceutical engineering gained momentum in the , as and emerged as foundational unit operations for refining synthetic intermediates and isolating pure compounds. These techniques, adapted from broader chemical practices, allowed for scalable purification of alkaloids and other drugs, bridging artisanal with industrialized processes by improving and in small-batch production.

Mass production era

The mass production era of pharmaceutical engineering emerged in the mid-20th century, propelled by the urgent demands of , which necessitated scaling production from laboratory quantities to industrial volumes. A pivotal event was the rapid commercialization of penicillin, initially discovered in 1928 but impractical for widespread use until wartime efforts. In 1941, the U.S. coordinated a collaborative project involving 21 companies, academic institutions, and government agencies to achieve , transforming penicillin from a curiosity into a strategic resource. By 1944, production had surged over 250-fold from mid-1943 levels, reaching millions of doses monthly through standardized processes. Pfizer played a central role by pioneering deep-tank , a submerged aerobic method adapted from production, which enabled efficient scaling in large stainless-steel vessels up to 10,000 gallons. Engineers Jasper L. Kane and John C. McKeen led this innovation at , proposing the technique in 1943 and opening the first commercial facility in , , on March 1, 1944, which quickly made the company the world's largest penicillin producer. This approach shifted output from surface fermentation's limited grams per batch to tons annually, supplying Allied forces and civilians while reducing costs from $25,000 per pound in 1943 to under $300 by 1945. Other firms, including Merck and Squibb, joined early in 1942 via information-sharing pacts, followed by , , , and , establishing dedicated facilities like 's fermentation plants in to meet rising demands. Post-war, the 1950s marked a technological shift toward mechanized and semi-continuous processes, extending wartime gains to broader drug manufacturing. Introduction of rotary tablet compression machines and early instrumented presses, developed by figures like T. Higuchi, allowed for higher throughput and precise control, increasing tablet output from manual presses' hundreds per hour to thousands, facilitating production scales from grams to tons for solid like antibiotics. Continuous flow elements, inspired by , began integrating into drying and extraction steps, standardizing operations across facilities like those of Merck and to handle surging post-war demand for antibiotics such as . Engineering contributions focused on optimizing unit operations to ensure product uniformity and minimize contamination, critical for efficacy and safety. In penicillin processing, engineers refined mixing protocols for extraction solvents and techniques—such as vacuum or —to achieve consistent and potency while preventing microbial ingress in sterile environments. These optimizations, informed by wartime data-sharing, reduced batch variability from over 20% to under 5% in key parameters like yield and purity, laying foundations for Good Manufacturing Practices. Merck's facilities, for instance, standardized to stabilize penicillin salts, supporting global distribution. These post-war developments culminated in the formal adoption of Good Manufacturing Practices (GMP) by the in 1969, which established international standards for pharmaceutical production quality and safety, solidifying pharmaceutical engineering's role as a distinct .

Advances in drug delivery

Advances in drug delivery during the mid-to-late marked a shift toward controlled and targeted release mechanisms, enabling more precise therapeutic outcomes in pharmaceutical engineering. A pivotal early was the of the Spansule® sustained-release capsule in by Laboratories, which encapsulated dexedrine sulfate within coated beads of varying thicknesses to achieve prolonged drug release over approximately 12 hours. This system represented one of the first commercial oral controlled-release formulations, relying on timed dissolution of multiple layers to sustain therapeutic levels and minimize peak-trough fluctuations in drug concentration. Subsequent milestones expanded these principles to alternative routes and carriers. In the , patches emerged as a noninvasive option, with the first FDA-approved product being Transderm Scop® () in 1979, developed by CIBA-Geigy for prevention through steady diffusion over 72 hours. By the 1980s, liposomes gained prominence for targeted delivery, with early applications demonstrating enhanced ; for instance, liposomal formulations of compounds were investigated for treating by exploiting uptake to localize drug action and reduce systemic exposure. These systems were engineered to encapsulate hydrophilic or lipophilic drugs within phospholipid bilayers, facilitating site-specific release and overcoming barriers like enzymatic degradation. Pharmaceutical engineers played a crucial role by applying principles, governed by Fick's laws, alongside coatings such as ethylcellulose or hydroxypropyl methylcellulose, to modulate over extended periods—ranging from hours in oral matrices to days in reservoirs. This engineering approach allowed for zero-order release profiles in ideal cases, where elution rates remained constant independent of remaining . The resulting innovations significantly reduced dosing frequency, from multiple daily administrations to once-daily or less, thereby enhancing patient compliance in managing chronic conditions like , where adherence rates improved due to simplified regimens and steadier plasma levels.

Establishment of professional bodies

The formalization of pharmaceutical engineering as a distinct discipline gained momentum in the late through the creation of dedicated professional organizations that emphasized standardized practices, knowledge sharing, and regulatory alignment in drug manufacturing and facility design. The International Society for Pharmaceutical Engineering (ISPE) was established in 1980 by a group of pioneering pharmaceutical engineers who recognized the need for a dedicated to advance , networking, and the exchange of best practices in pharmaceutical design and manufacturing processes. This quickly became a cornerstone for professionals, focusing on improving efficiency and compliance in the sector without overlapping into broader domains. Complementing ISPE's efforts, the (AIChE) chartered its Food, Pharmaceutical & Bioengineering Division in 1966, providing a specialized platform within the broader community to address process challenges unique to pharmaceutical production, such as and . A pivotal development occurred in the when ISPE initiated its Baseline Guides series, with the first guide on "Bulk Pharmaceutical Chemicals" approved in November 1994 and published in June 1996; these guides established industry benchmarks for facility design, validation, and operational standards, influencing global regulatory expectations. ISPE's global expansion accelerated thereafter, reaching over 25,000 members in more than 120 countries as of 2025 and exerting substantial influence on pharmaceutical standards in —through annual conferences and regulatory collaborations—and in , via affiliates and harmonization efforts like those supporting PIC/S in ASEAN regions.

Fundamental principles

Unit operations in pharmaceutical processes

Unit operations in pharmaceutical processes encompass the fundamental engineering steps adapted from to handle the , , purification, and of active pharmaceutical ingredients () and , with a strong emphasis on separation techniques to ensure purity and transformation processes to achieve desired physical properties. These operations are critical for maintaining product quality while addressing the sensitivity of pharmaceutical materials to temperature, shear, and contamination. Unlike general chemical processing, pharmaceutical unit operations prioritize sterility, from to production, and compliance with good manufacturing practices, often involving batch modes for precision control. Key separation operations such as , , and are tailored for isolation to remove impurities and recover solvents efficiently. , for instance, employs depth or filters to separate solids from liquids in , as seen in removal during steps where waste cake achieves high purity. purifies volatile components and facilitates solvent recovery, commonly used in batch processes to isolate by evaporating solvents like , with continuous variants emerging for efficiency in flow . leverages differences to partition into preferred phases, exemplified by in-line liquid-liquid in production under high pressure, enabling continuous isolation while minimizing waste. These operations collectively support high efficiency and sustainable manufacturing. Applications of in pharmaceutical processes focus on mixing and milling to promote homogeneity in powders, suspensions, and granulates, preventing segregation that could lead to inconsistent dosing. Mixing involves convective and diffusive mechanisms to blend with excipients, using tumblers or high-shear mixers to achieve relative standard deviations below 5% in blend uniformity for solid . Milling reduces particle size to enhance and flowability, with techniques like jet milling applied to to target mean diameters of 1-10 μm, improving without excessive heat generation that might degrade compounds. These operations ensure uniform distribution in formulations, critical for therapeutic efficacy.30010-1/abstract) Heat and principles underpin drying processes, which remove moisture to stabilize APIs against and microbial growth. , a prominent , atomizes feeds into hot gas streams to produce microparticles with rapid , achieving residual moisture contents below 2% essential for long-term stability in dispersions. This operation is particularly suited for biologics and inhalable powders, where inlet temperatures of 100-200°C yield particles with controlled and surface area, enhancing drug release profiles. Proper control prevents and maintains API potency during scale-up. Scale considerations highlight differences between batch and continuous operations, especially for sensitive biologics where batch modes allow precise monitoring of residence times to minimize , while continuous flows offer higher throughput and reduced variability in downstream purification. dominates API isolation due to flexibility in handling variable yields, but in unit operations like and reduces production times by up to 50% for monoclonal antibodies, though it requires advanced controls to manage shear-sensitive proteins. This shift enhances efficiency without compromising quality in biopharmaceutical manufacturing.

Thermodynamics and kinetics

In pharmaceutical engineering, provides the foundational principles for predicting the feasibility and direction of chemical reactions involved in . The change, \Delta G = \Delta H - T\Delta S, where \Delta H is the change, T is the absolute , and \Delta S is the change, determines the spontaneity of reactions; a negative \Delta G indicates a , guiding the selection of conditions for efficient of active pharmaceutical ingredients (). This principle is particularly applied in evaluating binding affinities during , where favorable contributions enhance ligand-receptor interactions in synthetic compared to natural ones. diagrams, incorporating \Delta G of mixing, further aid in screening drug-polymer combinations for dispersions, ensuring stability and by assessing energy changes in mixtures.00086-1/fulltext) Phase equilibria play a crucial role in crystallization processes, where vapor-liquid equilibrium influences the formation and control of polymorphs—different crystal structures of the same API that can drastically affect solubility and dissolution rates. In pharmaceutical crystallization, understanding these equilibria allows engineers to manipulate conditions like temperature and solvent composition to selectively produce desired polymorphs, as the stable form minimizes the under given conditions. For instance, vapor-liquid equilibria data help predict phase boundaries in binary systems, preventing unwanted polymorphic transformations that could lead to variations in drug efficacy or issues. This control is essential, as polymorphs exhibit distinct thermodynamic stabilities, with the most stable form having the lowest at equilibrium. Kinetics governs the rate at which reactions proceed in pharmaceutical synthesis, enabling optimization of reactor conditions to maximize yields and minimize byproducts. laws, expressed as rate = k [A]^n, where k is the rate constant, [A] is the reactant concentration, and n is the reaction order, describe how concentrations influence reaction speed, allowing engineers to model and scale processes in continuous reactors. In complex API networks, such as those for antineoplastic agents, kinetic modeling using these laws identifies rate-limiting steps, facilitating adjustments in or catalyst use to achieve high-purity outputs efficiently. These principles integrate with unit operations like mixing and to ensure precise control over reaction progression. Stability analysis in pharmaceuticals relies on kinetic models to forecast degradation under storage conditions, directly impacting shelf-life predictions. The , k = A e^{-E_a / RT}, where A is the , E_a is the , R is the , and T is temperature, quantifies how degradation rate constants vary with temperature, enabling extrapolation from accelerated stability tests to real-time conditions.00059-X/fulltext) For solid-state drugs, this equation supports early shelf-life estimation by modeling moisture and temperature effects, with activation energies typically ranging from 20-100 kJ/mol for common degradation pathways. Advanced applications, such as the Accelerated (), refine Arrhenius-based predictions by incorporating humidity factors, ensuring for product expiration dating.

Bioprocessing fundamentals

Bioprocessing in pharmaceutical engineering involves the use of living organisms or biological systems to produce therapeutic proteins, such as insulin and monoclonal antibodies, through controlled microbial or mammalian cell cultures. basics distinguish between and processes, with being predominant for recombinant protein production in bacteria like . In processes, oxygen is essential for and high-yield expression of target proteins, whereas conditions limit growth and productivity due to reliance on less efficient metabolic pathways. For instance, recombinant human insulin production via E. coli employs fed-batch at 37°C, where oxygen transfer is optimized through the volumetric (kLa), typically ranging from 100 to 500 h-1 in stirred-tank bioreactors to prevent oxygen limitation and maximize accumulation up to 100 g/L dry cell weight. Cell culture engineering extends these principles to mammalian systems for complex biologics like monoclonal antibodies, focusing on bioreactor designs that support prolonged viability and secretion. Stirred-tank bioreactors, equipped with impellers for mixing and spargers for gas distribution, are widely used for batch or fed-batch operations, maintaining critical parameters such as pH between 6.8 and 7.4 using CO2 and base additions, and temperature at 37°C to mimic physiological conditions. In contrast, perfusion bioreactors enable continuous media exchange and cell retention via alternating tangential flow or centrifugation, achieving higher cell densities (up to 108 cells/mL) and extended production phases beyond 30 days compared to the 10-14 day cycles in stirred-tank systems. These designs optimize nutrient delivery and waste removal, enhancing antibody glycosylation and folding for therapeutic efficacy. Downstream processing recovers and purifies the biologics from complex culture broths, ensuring high purity for clinical use. Initial clarification employs at 5,000-10,000 × g to separate cells and debris, followed by depth filtration to achieve clear supernatants with minimal turbidity. Subsequent purification relies on steps, including affinity for capture (yielding 95% purity in one step), ion-exchange for charge-based separation, and size-exclusion for final polishing, collectively attaining >99% purity while removing host cell proteins and aggregates to below 100 ppm. These unit operations are integrated in a approach, minimizing steps to reduce costs and maintain product stability. Yield metrics in quantify process efficiency, with specific measuring protein output per per day and volumetric assessing overall performance. For monoclonal antibodies in CHO cultures, specific typically ranges from 20-70 pg//day, while volumetric has advanced from 0.05-0.1 g/L/day in traditional batch modes to 0.5-2.0 g/L/day in systems, enabling cumulative titers exceeding 20 g/L. Recovery rates in average 80-90%, influenced by resin capacity and process robustness, underscoring the need for integrated upstream-downstream optimization to achieve economic viability in large-scale .

Key manufacturing processes

Drug formulation and design

Drug formulation and design in pharmaceutical engineering involves the systematic development of that ensure the (API) is delivered effectively, stably, and safely to the patient. This process integrates principles of chemistry, , and to select appropriate excipients, optimize drug release profiles, and create prototypes suitable for further testing. Key considerations include the physicochemical properties of the API, such as and , which directly influence and therapeutic efficacy. Formulation types are categorized based on their physical state and intended . Solid , such as tablets and capsules, are the most common for oral delivery due to their , ease of , and precise dosing. In these forms, excipients play critical roles: binders like (PVP) promote cohesion by agglomerating powder particles into granules, enhancing tablet integrity during compression and preventing . Liquid , including syrups and injections, facilitate rapid absorption and are ideal for pediatric or intravenous use; here, excipients such as (e.g., Tween 80) improve and reduce interfacial tension for lipophilic . Semi-solid forms like creams and gels are designed for topical application, where polymers such as hydroxypropyl methylcellulose (HPMC) provide and controlled release while ensuring compatibility. Excipients across all types must be inert, biocompatible, and selected to avoid interactions that could alter or efficacy. The design process begins with preformulation studies to characterize the API's fundamental properties and ensure compatibility with excipients. assessments are pivotal, often employing the Noyes-Whitney equation to model rates: \frac{dM}{dt} = \frac{D \cdot S \cdot (C_s - C_b)}{h} where \frac{dM}{dt} is the rate, D is the coefficient, S is the surface area, C_s and C_b are the and bulk concentrations, respectively, and h is the layer thickness. This equation highlights how particle size reduction increases S, accelerating for poorly soluble drugs. testing involves stressing the API with excipients under varied conditions (e.g., , , ) to detect products via techniques like or , thereby guiding stable formulation selection. Dosage form engineering refines these studies into viable prototypes, particularly for solid forms where mechanical properties are optimized. For tablets, forces typically range from 5 to 20 to achieve adequate (e.g., 4-10 ) without excessive , balancing structural against rapid release. Higher forces reduce , increasing but prolonging disintegration time; thus, targets are set for disintegration under 15 minutes in pharmacopeial media (e.g., at 37°C) to ensure >80% within physiological ranges. Disintegrants like croscarmellose sodium are incorporated to facilitate wicking and swelling, countering compression-induced delays. Quality by Design (QbD) principles are applied throughout to build quality into the formulation via systematic risk assessment. Using tools like (FMEA), engineers identify critical quality attributes (CQAs) such as (drug content uniformity) and impurities ( products <1%), prioritizing variables like excipient ratios that impact them. This risk-based approach defines a design space where formulations meet quality targets, enhancing robustness and reducing variability in prototype development.

Scale-up and production

Scale-up in pharmaceutical engineering involves translating laboratory-scale formulations to commercial production while maintaining product quality and process efficiency. Strategies typically progress from pilot-scale batches (1-100 L) to full production scales (1000 L or larger), focusing on geometric, kinematic, and dynamic similarities to avoid deviations in critical quality attributes such as particle size and density. Dimensionless numbers, including the for viscous forces in mixing and the for centrifugal effects, guide this process by ensuring proportional scaling of impeller speeds and power inputs across equipment sizes. For instance, maintaining a constant impeller tip speed of 5-10 m/s helps replicate mixing conditions from lab to production, as demonstrated in model-based approaches for . Key equipment in scale-up includes high-shear mixers and fluidized bed granulators, which facilitate granulation by promoting uniform binder distribution and agglomeration. High-shear mixers, equipped with impellers and choppers operating at 150-200 rpm, are widely used for wet granulation in batch processes, enabling rapid nucleation and growth while controlling granule size through adjustable wet massing times (30 seconds to 5 minutes). Fluidized bed granulators, by contrast, integrate granulation and drying in a single step via air suspension, producing porous granules with improved flowability, though they require careful management of fill levels (10-80%) to optimize energy efficiency. These systems aim to achieve granule uniformity indices exceeding 95%, ensuring consistent drug content distribution that meets pharmacopeial standards for downstream tableting. Process validation during scale-up relies on in-process controls to monitor and adjust parameters in real time, minimizing variability. Near-infrared (NIR) spectroscopy, for example, enables non-destructive, in-line assessment of moisture content during fluid bed drying, with partial least squares models calibrated against Karl Fischer titration achieving root-mean-square errors as low as 0.1%. This technique supports validation in continuous lines like the ConsiGma™ 25, where multipoint probes track drying across segments, ensuring compliance with predefined quality thresholds. The industry is shifting from traditional batch manufacturing to continuous processes, with the FDA approving several facilities and products in the 2020s, including expansions at Thermo Fisher's Greenville site in 2024 and drugs like (2017) and (2019). Continuous manufacturing eliminates hold times between unit operations, offering efficiency gains of 20-30% in productivity through 24/7 operation and reduced equipment footprint, while enhancing quality consistency via integrated process analytical technology. This transition, supported by FDA guidance like , addresses scalability challenges in high-volume production.

Sterilization and packaging

Sterilization in pharmaceutical engineering ensures the elimination of viable microorganisms from final drug products, preventing contamination and extending shelf life, particularly for injectable, ophthalmic, and other sterile formulations. For heat-stable products, such as certain aqueous solutions and equipment, autoclaving remains a primary method, involving saturated steam under pressure at 121°C and 15 psi for at least 15 minutes to achieve microbial inactivation through protein denaturation and cell lysis. This process is effective against a broad spectrum of bacteria, spores, and fungi but is unsuitable for thermolabile substances like proteins or plastics that degrade under high temperatures. For such heat-sensitive materials, including many drug delivery systems and biologics, gamma irradiation using cobalt-60 sources is preferred, delivering a dose of 25-40 kGy to penetrate packaging and disrupt microbial DNA without generating residues or requiring post-process aeration. Validation of these sterilization processes is critical to confirm efficacy, typically targeting a sterility assurance level (SAL) of $10^{-6}, meaning the probability of a single viable microorganism surviving is less than one in a million units processed. This standard is achieved through half-cycle testing, overkill methods, or bioburden-based approaches, where biological indicators such as spores of are used for steam validation due to their high resistance to heat, while serves for radiation confirmation. Physical parameters like temperature, pressure, and dose are monitored via sensors and dosimeters, with fractional cycle runs ensuring the process meets pharmacopeial requirements without compromising product integrity. Packaging engineering in pharmaceuticals focuses on engineered barriers to protect against environmental factors like moisture, oxygen, and light, while maintaining sterility post-sterilization. Common formats include packs for solid oral dosage forms, which combine thermoformed PVC or PVDC cavities with aluminum foil lidding to provide unit-dose protection, and glass or high-density polyethylene (HDPE) vials for injectables, where HDPE offers robust moisture barrier properties limiting weight gain to less than 0.5% under controlled humidity conditions. These materials are selected for their compatibility with sterilization methods—gamma-irradiated packaging retains integrity without delamination—and tested per USP <671> for rates, ensuring minimal transmission of (e.g., Class A packs with average rates below 0.5 mg/day). Child-resistant and tamper-evident designs are integral to , enhancing and product security in compliance with regulatory standards. Child-resistant closures, such as push-and-turn caps on HDPE bottles, prevent accidental access by young children while allowing adult usability, often validated through sequential testing protocols. Tamper-evident features, like breakable seals or perforated bands on vials and blisters, provide visible evidence of unauthorized access, with container integrity assessed under USP <671> for and seal strength to safeguard against . These elements ensure the packaged product remains sterile and unaltered from the point of filling through distribution.

Advanced technologies

Controlled release mechanisms

Controlled release mechanisms in pharmaceutical engineering enable the precise modulation of to maintain therapeutic concentrations over extended periods, reducing dosing frequency and improving patient compliance. These systems are designed to overcome limitations of immediate-release formulations by incorporating physical and chemical principles that govern diffusion, behavior, and environmental interactions. Key mechanisms include diffusion-controlled, swelling-controlled, and erosion-based release, each tailored to specific properties and therapeutic needs. Diffusion-controlled systems rely on the concentration gradient-driven movement of molecules through a polymeric barrier or , as described by Fick's first law of diffusion: the J = -D \frac{dc}{dx}, where D is the diffusion coefficient, c is the concentration, and x is the within the . This mechanism predominates in both and reservoir configurations, where the rate-limiting step is the 's permeation through the polymer network. In reservoir systems, a -loaded core is encased in a semi-permeable that controls release, often achieving zero-order for constant delivery independent of external factors like . systems, in contrast, embed the homogeneously within an inert polymer, resulting in release primarily following the Higuchi model for monolithic matrices: the cumulative amount released Q = k \sqrt{t}, where k is a constant incorporating , , and , and t is time; this square-root-of-time dependence reflects gradual depletion from the surface. Swelling-controlled mechanisms involve hydrophilic polymers that absorb water upon exposure to physiological fluids, leading to hydration, chain relaxation, and matrix expansion that facilitates drug diffusion out of the gel layer. This process creates a dynamic front where polymer swelling pushes the drug toward the surface, with release rates influenced by the polymer's crosslinking density and hydrophilicity; for instance, high-swelling polymers like hydroxypropyl methylcellulose can sustain release over hours by balancing osmotic uptake and elastic retraction forces. Erosion-based systems complement these by degrading the polymer matrix itself—either via surface erosion, where hydration and hydrolysis progressively dissolve the outer layers, or bulk erosion, involving uniform internal breakdown—to liberate entrapped drug molecules. These mechanisms are particularly suited for hydrophilic polymers like polyanhydrides, where the erosion rate directly correlates with drug release, enabling predictable profiles in gastrointestinal environments. A prominent example of advanced controlled release is the Osmotic Release Oral System (OROS) technology, developed by Alza Corporation in the 1970s, which uses a semi-permeable membrane around an osmotic core to deliver drugs at a constant rate via water influx and zero-order pumping, unaffected by gastrointestinal pH variations. This reservoir-based design has been applied in products like Glucotrol XL for sustained antidiabetic therapy. To ensure reliability, these mechanisms are modeled and validated through dissolution testing, as outlined in the <711> general chapter, which employs apparatus like baskets or paddles to simulate release under controlled conditions and correlate with in vivo performance via metrics such as f2 similarity factors.

Nanotechnology applications

Nanotechnology in pharmaceutical engineering leverages nanoscale materials, typically in the range of 1-100 nm, to enhance systems by improving , , and specificity in therapeutic applications. Liposomes, a prominent class of nanoparticles with sizes often between 100-200 nm, serve as versatile carriers due to their amphiphilic structure, consisting of bilayers that mimic cell membranes. These vesicles can encapsulate both hydrophilic drugs in their aqueous core and hydrophobic drugs within the , thereby facilitating the solubilization and transport of poorly water-soluble compounds that constitute a significant portion of modern pharmaceuticals. For hydrophobic drugs, liposomes integrate the active pharmaceutical ingredient into the bilayer, effectively increasing their apparent aqueous solubility by factors of 10-100 fold depending on the drug-lipid interaction and formulation parameters, which overcomes limitations in oral or intravenous administration. This encapsulation protects the drug from degradation and enzymatic attack, while the nanoscale size enables better compared to larger particulates. In cancer , such systems exploit the enhanced permeability and retention () effect, where leaky tumor vasculature allows preferential accumulation of nanoparticles at the tumor site, thereby minimizing systemic exposure. Targeted delivery further refines this approach through surface conjugation of liposomes with ligands, such as antibodies or peptides, that bind specifically to overexpressed receptors on diseased cells, like HER2 in . This active targeting reduces off-target effects by promoting , enhancing intracellular drug release while sparing healthy tissues, and has shown up to 10-fold improvement in in preclinical models. The EPR effect complements this by passively directing unmodified liposomes to solid tumors, but ligand conjugation amplifies specificity, particularly in heterogeneous tumor environments. Fabrication of liposomes with controlled properties is critical for and in pharmaceutical production. Emulsion-based methods, such as the thin-film hydration followed by or the ethanol injection technique, involve dissolving in organic solvents, hydrating with aqueous media, and applying shear forces to form vesicles, yielding uniform particles with polydispersity index (PDI) values below 0.2 for narrow size distributions essential for consistent . processing, using (scCO2) as a , enables solvent-free assembly under high pressure, producing liposomes with PDI <0.2 and high encapsulation efficiency (>90%) while avoiding residual solvents that could compromise safety. These scalable techniques support industrial manufacturing, ensuring batch-to-batch uniformity. Recent advancements include lipid nanoparticles (LNPs) for mRNA delivery, building on liposomal technology, with optimized ionizable lipids improving stability and targeted delivery, as seen in vaccines and ongoing applications as of 2025. A landmark clinical example is Doxil, the first FDA-approved nanodrug in 1995, comprising pegylated liposomes encapsulating for treatment. By remote loading into the aqueous core and coating with (PEG) to evade clearance, Doxil extends the drug's circulation from approximately 1 hour for free to 55 hours, enabling sustained tumor exposure and reduced cardiotoxicity. This formulation exemplifies how integrates with controlled release principles to optimize dosing regimens in .

Process analytical technology

Process Analytical Technology (PAT) is a regulatory framework established by the U.S. (FDA) in to promote innovative through enhanced process understanding and control. This initiative integrates , process analyzers, and multivariate to enable monitoring of critical process parameters and quality attributes, shifting from traditional end-product testing to proactive quality assurance during production. By emphasizing risk-based approaches, PAT facilitates the design, analysis, and control of manufacturing processes to ensure consistent product quality while minimizing variability. Key tools within the PAT framework include spectroscopic and chromatographic methods for in-line or on-line analysis. serves as a non-destructive process analyzer for quantifying active pharmaceutical ingredient () content in solid , achieving prediction accuracies typically within ±1-2% for amorphous content in low-drug-load products. Online (HPLC) enables real-time impurity detection during chemical reactions, allowing precise measurement of product purity and supporting decisions on process collection or adjustments. These analyzers, combined with chemometric models such as , process spectral data to extract actionable insights on composition and process state. Implementation of often involves loops in continuous setups, where sensors provide data for automated systems. For instance, pH monitoring via PAT tools can trigger immediate adjustments in reaction conditions through closed-loop controllers, maintaining optimal process parameters and preventing deviations. This integration supports seamless scale-up from lab to production, as referenced in broader contexts, by enabling adaptive responses that align with defined quality targets. Recent developments as of 2025 include integration of (AI) and for in PAT, enabling proactive deviation detection and optimization in manufacturing processes. The adoption of PAT yields significant benefits, including reductions in batch failure rates through early detection and correction of process excursions, as demonstrated in specific (QbD) case studies, alongside accelerated product release via real-time release testing (RTRT) that bypasses lengthy offline assays. These outcomes enhance overall efficiency, reduce waste, and align with FDA goals for flexible, science-based regulation.

Regulatory framework

Good manufacturing practices

Good manufacturing practices (GMP) form the cornerstone of pharmaceutical engineering, establishing regulatory guidelines to ensure the hygiene, consistency, and quality of drug production processes. Developed by organizations such as the (WHO) and harmonized through the International Council for Harmonisation (ICH), GMP emphasizes a comprehensive that integrates to mitigate contamination risks and maintain product integrity throughout manufacturing. Core principles include meticulous of all procedures, from master production formulas to batch records, to enable and reproducibility; personnel training programs that ensure staff qualifications and hygiene practices; and facility design that minimizes environmental risks, such as the use of cleanrooms classified up to Class 100,000 (equivalent to ISO 8) for non-sterile operations to control . These elements collectively safeguard against deviations that could compromise . In key operational areas, GMP mandates stringent controls over raw materials and equipment to prevent cross-contamination and ensure reliability. Raw materials must be sourced from qualified vendors through rigorous qualification processes, including audits of supplier quality systems and testing of incoming lots for identity, purity, and compliance with specifications before release for use. Equipment requires regular calibration, validation, and maintenance as per standard operating procedures (SOPs), with records demonstrating traceability to international standards, thereby reducing the risk of mechanical failures or residue carryover between batches. These practices are particularly critical in multi-product facilities, where dedicated production lines or validated cleaning protocols are employed to isolate potent or hazardous substances. GMP guidelines have evolved to incorporate technological advancements, notably in the 2022 revision of the (EU) GMP Annex 1 for sterile medicinal products, which introduces a contamination control strategy (CCS) and emphasizes barrier technologies like restricted access barrier systems (RABS). RABS, featuring rigid enclosures with integrated gloves and HEPA-filtered airflows, provide Grade A conditions while restricting personnel intervention, marking a shift from earlier versions that focused primarily on unidirectional airflow alone. This update, effective from August 2023, aligns with ICH Q9 principles of quality risk management to enhance sterility assurance in . Compliance with GMP is maintained through systematic audits and corrective mechanisms, including annual self-inspections conducted by multidisciplinary teams to evaluate adherence across personnel, , and processes, culminating in detailed reports with actionable recommendations. Deviations trigger corrective and preventive actions (CAPA), involving , implementation of fixes, and verification of effectiveness to prevent recurrence, with all steps documented for regulatory review. These audits extend to manufacturers and suppliers, ensuring a holistic oversight.

Quality assurance and control

Quality assurance (QA) and quality control (QC) in pharmaceutical engineering encompass systematic processes to ensure that drug products meet predefined standards for safety, efficacy, and consistency throughout their lifecycle. focuses on establishing and maintaining organizational structures and procedures to prevent defects, while involves the actual testing and verification of materials, processes, and products. These systems are integral to pharmaceutical quality systems, as outlined in the International Council for Harmonisation (ICH) guidelines Q8, Q9, and Q10, which provide a framework for pharmaceutical development, , and overall quality systems. ICH Q8 emphasizes the use of principles to define critical quality attributes and design space during development. ICH Q9 introduces structured risk management tools, such as , to identify and mitigate potential quality risks. ICH Q10 establishes a model for a pharmaceutical quality system that integrates these elements across the , including processes to manage modifications while ensuring ongoing product quality. QC methods are essential for verifying product attributes at various stages, with (HPLC) serving as a primary for assessing potency. HPLC separates and quantifies active pharmaceutical ingredients based on their with a stationary phase, typically achieving acceptance criteria of 95-105% of the label claim to confirm potency within therapeutic limits. This method ensures accurate measurement of content, supporting batch release decisions. Complementing potency testing, evaluates the release profile of the from its , simulating gastrointestinal conditions to predict . Apparatus such as the paddle or basket method is used under standardized conditions (e.g., 37°C in simulated gastric fluid), with acceptance criteria often requiring at least 80% dissolution within 30 minutes for immediate-release formulations to ensure consistent performance. These QC tests are validated per ICH Q2(R1) to demonstrate accuracy, precision, and specificity. Stability testing is a cornerstone of , assessing how drug products degrade over time under various environmental conditions to determine shelf-life. The ICH Q1A(R2) guideline mandates protocols including long-term testing at 25°C/60% relative and accelerated testing at 40°C/75% relative for at least six months to predict and establish expiration dates. Data from a minimum of three batches are analyzed for trends in potency, impurities, and physical characteristics, with shelf-life assigned based on when quality attributes fall below acceptance criteria. This testing informs labeling and storage recommendations, ensuring product integrity until the end of shelf-life. Process validation underpins QA by confirming that manufacturing processes consistently produce products meeting quality specifications. As per U.S. (FDA) regulations in 21 CFR Part 211, validation requires documented evidence from three consecutive production-scale batches demonstrating reproducibility in critical process parameters and quality attributes. This includes prospective validation for new processes and concurrent validation where appropriate, with statistical to verify process capability. Validation protocols must include predefined criteria, ensuring compliance with current good manufacturing practices (cGMP) to mitigate variability risks.

International standards

International standards in pharmaceutical engineering ensure the safety, quality, and efficacy of medicinal products across global markets by harmonizing regulatory requirements and facilitating cross-border compliance. These standards address critical aspects of drug development, manufacturing, and supply chains, minimizing variations in practices among major economies while promoting equivalence in product quality. Key frameworks include those developed by the International Council for Harmonisation (ICH), the World Health Organization (WHO), and the International Organization for Standardization (ISO), alongside mutual recognition agreements that reduce redundant oversight. The ICH, established as a tripartite initiative involving regulatory authorities and industry associations from the European Union (EU), the United States (US), and Japan, develops harmonized guidelines to streamline technical requirements for pharmaceuticals. Notable examples include the Q1A(R2) guideline on stability testing of new drug substances and products, which outlines protocols for assessing degradation under various conditions to support registration applications. Similarly, the Q3A(R2) and Q3B(R2) guidelines set thresholds and qualification procedures for impurities in new drug substances and products, respectively, ensuring control of potentially hazardous contaminants. For biotechnological products, the Q5C guideline specifies stability testing tailored to biological entities, accounting for their unique sensitivities. These guidelines, adopted by ICH member regions, reduce the need for region-specific studies and enhance global consistency in pharmaceutical engineering practices. The WHO Prequalification Programme evaluates finished pharmaceutical products (FPPs) and active pharmaceutical ingredients () against unified standards of quality, safety, and efficacy, particularly for used in low- and middle-income countries. This programme assesses manufacturing sites for compliance with WHO Good Manufacturing Practices (GMP), ensuring bioequivalence and therapeutic interchangeability with originator products, which supports procurement by international agencies like and the Global Fund. By focusing on priority diseases such as , , and , it facilitates access to affordable, reliable medicines in developing regions without compromising engineering standards. ISO standards integrate quality management principles with pharmaceutical-specific requirements, notably ISO 15378:2017 for primary materials used in medicinal products, which combines ISO 9001 elements with GMP to cover , , and supply processes. This ensures packaging integrity against and issues during and . Complementing this, :2016 provides a framework for medical devices, with overlaps in pharmaceutical engineering for combination products or systems where device components interact with pharmaceuticals, emphasizing risk-based controls and regulatory compliance. Mutual recognition agreements further streamline international oversight, exemplified by the EU-US Mutual Recognition Agreement (MRA) on GMP for pharmaceuticals, which entered into force on November 1, 2017. Under this sectoral annex, the US Food and Drug Administration (FDA) and EU authorities recognize each other's GMP inspections of manufacturing sites, accepting official certificates to avoid duplicate audits and expedite for exported products. This agreement covers human and veterinary medicines, fostering efficiency in global pharmaceutical engineering while maintaining high safety standards.

Education and professional practice

Academic programs and curricula

Pharmaceutical engineering academic programs typically span multiple degree levels, beginning with bachelor's degrees that provide foundational training in engineering principles applied to drug development and manufacturing. A Bachelor of Science (B.S.) in Pharmaceutical Engineering or related fields, such as Biological Engineering with a Pharmaceutical Process Engineering concentration, is commonly a four-year program requiring 120-140 credits. For instance, Purdue University's BSBE in Pharmaceutical Process Engineering integrates biological systems design for pharmaceutical applications, encompassing core engineering and biology coursework. Graduate programs include master's degrees, often one to two years in duration, such as the Master of Engineering (M.E.) in Pharmaceutical Engineering at Rutgers University, which focuses on advanced unit operations and materials science for pharmaceutical production. Doctoral programs, like the Ph.D. in Pharmaceutical Sciences at institutions such as Northeastern University, emphasizing research in bioprocesses and regulatory compliance, typically requiring 30-60 additional credits beyond the master's level. Core curricula in pharmaceutical engineering degrees emphasize interdisciplinary knowledge blending , , and pharmaceutical sciences to address drug formulation, process optimization, and . Common required courses include to model drug behavior in the body, reactor design for scalable manufacturing, and for biological therapeutics production. At the (NJIT), the M.S. program mandates foundational classes in reaction kinetics, , and principles of pharmaceutical engineering, totaling 30 credits with a focus on practical applications like systems. Programs often total 120-140 credits at the bachelor's level, incorporating , physics, and specialized electives such as crystallization systems and principles, as seen in Purdue's . These courses prioritize conceptual understanding of scale-up challenges in pharmaceutical production while ensuring compliance with industry standards. Hands-on training forms a critical component of pharmaceutical engineering education, bridging theoretical knowledge with practical skills through laboratory modules, simulations, and industry placements. Students engage in formulation labs to develop drug prototypes, (GMP) simulations for sterile processing, and co-operative education (co-op) experiences lasting several months in pharmaceutical facilities. For example, Mellon University's M.S. in and Pharmaceutical Engineering includes 51 core units with lab-based biology and components, emphasizing experimental design for optimization. Such training equips graduates to handle real-world tasks like aseptic manipulations and , often integrated into projects or internships required for degree completion. Global variations in pharmaceutical engineering curricula reflect regional priorities, with European programs like Zurich's Bachelor and Master in Pharmaceutical Sciences stressing and scientific research alongside industry-specific training in and . In contrast, Asian programs, such as the Indian (BHU) Varanasi's four-year B.Tech. in Pharmaceutical Engineering and Technology, emphasize processes, drug testing, and cost-effective production techniques tailored to large-scale industries. These differences arise from local economic focuses, with European curricula allocating more time to advanced biotech (e.g., 7-week practical methods labs at ) versus Asia's integration of with for rapid market entry.

Certifications and career paths

Professional certifications play a crucial role in validating expertise in pharmaceutical engineering, particularly in areas like good manufacturing practices (GMP) and . The (CPGP), offered by the (ASQ), requires candidates to demonstrate five years of on-the-job experience in GMP-related areas, with at least three years in decision-making positions involving authority over projects or processes, followed by passing a comprehensive open-book consisting of 150 scored multiple-choice questions. Similarly, the ASQ (CQE) certification, relevant for pharmaceutical , demands eight years of experience in principles, reducible through educational waivers (e.g., four years waived for a ), and successful completion of a 160-question focused on product evaluation and control. These credentials enhance by confirming adherence to regulatory standards and are often pursued after initial academic preparation in disciplines. Entry-level career paths in pharmaceutical engineering typically begin with roles such as process engineers, who design, optimize, and scale processes for drug production in (R&D) or environments. Starting salaries for these positions average around $80,000 USD annually in 2025, reflecting the demand for technical skills in process optimization and compliance within the U.S. pharmaceutical sector. With hands-on experience in areas like operations or development, professionals gain foundational knowledge essential for advancing in the field. Career advancement often involves progression to senior roles such as project managers, who oversee multidisciplinary teams in timelines and budgets, or specialists, responsible for ensuring compliance with agencies like the FDA. This typically requires 5-10 years of experience, building on initial engineering roles to develop leadership and regulatory expertise. Such trajectories emphasize continuous , including certifications, to navigate the industry's emphasis on and . Pharmaceutical engineers find employment across key industry sectors, with significant opportunities in large pharmaceutical companies like , which dominate drug manufacturing and R&D; biotechnology firms focused on novel therapies; and the expanding contract manufacturing organizations () that provide outsourced production services. These sectors collectively drive the field's growth, projected at 3% through 2034, supported by ongoing advancements in production.

Challenges and future directions

Sustainability and green engineering

Pharmaceutical manufacturing processes present notable environmental challenges, including high water consumption, often tens of cubic meters per kg of active pharmaceutical ingredient (), driven by needs for purification, cooling, and cleaning in and stages. Solvent emissions from these operations further exacerbate impacts, with the healthcare sector, including pharmaceuticals, contributing approximately 4-5% of global . These factors highlight the sector's resource intensity, where and gaseous releases can strain local ecosystems if not managed effectively. To address these issues, principles emphasize waste minimization through advanced and strategies. Enzymatic , for instance, enables reactions under milder conditions, reducing demands by up to 50% compared to traditional chemical methods, as seen in biocatalytic routes for antibiotics and analgesics. Solvent complements this by recovering and reusing organic solvents like or , cutting waste generation and associated emissions while aligning with the 12 principles of . These approaches prioritize and benign auxiliaries to lower the overall of drug production. A key metric for evaluating sustainability is the E-factor, defined as kilograms of waste per kilogram of product, with the typically ranging from 25 to over 100 due to multi-step syntheses and purification losses. The ACS Green Chemistry Institute advocates targeting E-factors below 25 through process optimization, promoting innovations like continuous flow reactors that enhance efficiency and reduce byproducts. Illustrative case studies demonstrate practical implementation, such as Pfizer's green chemistry program launched in the 2000s, which has integrated biocatalysis and solvent recovery across its operations, yielding cumulative CO₂ savings of approximately 814,000 tons since 2000 through energy-efficient redesigns and waste avoidance. This initiative underscores how targeted engineering can scale sustainability without compromising product quality or regulatory compliance. As of 2025, EU regulations under the Green Deal are increasingly mandating reduced environmental impacts in pharmaceutical manufacturing.

Personalized and precision medicine

Pharmaceutical engineering plays a pivotal role in enabling personalized and precision medicine by developing technologies that tailor drug formulations and delivery systems to individual patient profiles, often based on genomic, proteomic, or phenotypic data. Microfluidic devices facilitate on-demand drug formulation by enabling precise control over mixing, reaction times, and reagent volumes at the microscale, which reduces waste and allows for rapid customization of dosages and release profiles for specific patients. For instance, droplet-based microfluidics have been applied in synthesizing personalized drug delivery systems, offering advantages such as shorter synthesis times and improved reproducibility compared to traditional batch methods. Complementing this, 3D printing technologies, such as binder jetting, enable the fabrication of personalized tablets with complex geometries and controlled porosity to optimize disintegration and absorption. A landmark example is Spritam (levetiracetam), the first FDA-approved 3D-printed pharmaceutical product in 2015, which features a highly porous structure for rapid dissolution in epilepsy treatment, demonstrating how additive manufacturing can produce patient-specific dosage forms with enhanced precision. In gene therapies, such as chimeric antigen receptor T-cell (CAR-T) therapies, pharmaceutical engineering integrates artificial intelligence () for process control to handle variable batch sizes inherent to autologous production, where each patient's cells are uniquely modified. -driven platforms predict T-cell quality metrics during manufacturing, using to optimize parameters like expansion rates and efficiency in bioreactors, thereby adapting to inter-patient variability and ensuring consistent therapeutic outcomes. For CAR-T processes, digital twins or shadows of cell expansion in perfusion systems employ to monitor real-time growth dynamics, enabling that scales from single-patient lots to semi-automated workflows while maintaining GMP compliance. These integrations address the bespoke nature of precision therapies by automating and reducing manual interventions, as seen in automated multi-step systems for CAR-T , , and . Despite these advances, significant challenges persist in scaling single-patient production for personalized therapies, particularly due to the labor-intensive, autologous processes required for treatments like CAR-T cells. Manufacturing timelines often exceed weeks, compounded by logistical complexities in sourcing and processing patient-specific materials, which limits throughput and increases variability. Costs for CAR-T therapies frequently surpass $400,000 per treatment, driven by specialized facilities, production, and extensive quality controls, posing barriers to broader accessibility despite their efficacy in cancers. Looking ahead, FDA initiatives in medicine, including enhanced regulatory pathways for companion diagnostics and cell/ therapies, are poised to expand personalized adoption. Personalized medicines already represent approximately 38% of newly approved therapeutic molecular entities in , a trend that underscores the shift toward individualized treatments. Projections indicate that by 2030, a substantial portion of the pharmaceutical market—potentially over 50% of patent-protected therapies—will incorporate elements, driven by ongoing innovations in and . As of November 2025, the FDA has continued this momentum with additional approvals in oncology and editing technologies.

References

  1. [1]
    Pharmaceutical Engineering - Zamann Pharma Support GmbH
    Pharmaceutical engineering is a multidisciplinary field that applies principles of chemical, mechanical, and industrial engineering to the design, development, ...
  2. [2]
    Pharmaceutical Engineering Explained | JD Pharma Consultants
    May 20, 2025 · Pharmaceutical engineering is a field that combines science and engineering to design, develop, and manage systems and processes in the pharmaceutical industry.
  3. [3]
    What Is Pharmaceutical Engineering? | UC Riverside
    Pharmaceutical engineering involves the research, development, creation, and manufacturing of medicinal drugs.
  4. [4]
    What is pharmaceutical engineering - NNE
    Pharmaceutical engineering focuses on designing, building, and improving manufacturing facilities that produce drugs.
  5. [5]
    Pharmaceutical Engineering - an overview | ScienceDirect Topics
    Pharmaceutical engineering is an interdisciplinary science that involves the development and manufacturing of products, process, and components as drugs in the ...
  6. [6]
    [PDF] Pharmaceutical Engineering and Process Design
    What is Pharmaceutical Engineering? Definition. • Pharmaceutical engineering is an interdisciplinary field that applies engineering principles, techniques ...
  7. [7]
    Drug Substance Manufacturing Process Optimization & Scale-Up
    Enhance efficiency and scalability with expert process optimization and scale-up solutions for small-molecule drug substances during late-stage development.
  8. [8]
    Pharmaceutical Quality by Design | ISPE - ISPE
    Pharmaceutical Quality by Design (QbD) is a systematic approach to development that begins with predefined objectives and emphasizes product and process ...
  9. [9]
    Model-Based Scale-up Methodologies for Pharmaceutical Granulation
    In this study, model-based scale-up methodologies for pharmaceutical granulation are presented, along with data from various related reports.
  10. [10]
    Pharmaceutical Engineering: Applications & Explained - StudySmarter
    Sep 5, 2024 · Pharmaceutical engineering is an interdisciplinary field that combines principles of engineering, pharmaceutical sciences, and biotechnology ...
  11. [11]
    Pharmaceutical Engineering, MS, Master's - Northeastern University
    Students have the opportunity to gain advanced interdisciplinary training in chemical engineering and pharmaceutical sciences with core and elective classes in ...
  12. [12]
    [PDF] Formulation Engineering and Bioavailability Analysis
    Jan 29, 2024 · Formulation engineering involves the art and science of designing a drug product that ensures stability, efficacy and patient compliance. It ...Missing: objectives | Show results with:objectives
  13. [13]
    Quality by design | European Medicines Agency (EMA)
    Quality by design is an approach that aims to ensure the quality of medicines by employing statistical, analytical and risk-management methodology.
  14. [14]
    Pharmaceutical Market Size Expected to Hit USD 3.03 Trillion by 2034
    Sep 29, 2025 · The global pharmaceutical market size is calculated at USD 1.77 trillion in 2025 and is predicted to exceed USD 3.03 trillion by 2034, ...
  15. [15]
    [PDF] The Pharmaceutical Industry in Figures - EFPIA
    Nov 28, 2024 · The world pharmaceutical (prescription) market was worth an estimated € 1,413,609 million ($ 1,528,535 million) at ex-factory prices in 2024.
  16. [16]
    The economic impact of the global pharmaceutical industry | IFPMA
    Nov 20, 2024 · 22.4 million jobs supported through induced effects in the supply chain. The industry contributed USD 2,295 billion to the global GDP, including ...
  17. [17]
    When Speed to Market Counts | Pharmaceutical Engineering - ISPE
    Apr 28, 2023 · Lean manufacturing practices and automation can optimize a company's internal processes and thereby reduce production times and costs.
  18. [18]
    How to reduce time to market by >50% - Körber Pharma
    In this article we would like to introduce a self-learning digital twin that enables the reduction of required experiments in PCS by more than 50% and hence ...Missing: engineering | Show results with:engineering
  19. [19]
    Understanding Pharmaceutical Quality by Design - PMC - NIH
    This paper is intended to describe the objectives of pharmaceutical QbD, detail its concept and elements, and explain implementation tools and studies.
  20. [20]
    Why tech transfer may be critical to beating COVID-19 - McKinsey
    Jul 23, 2020 · How quickly COVID-19 vaccine production ramps up will depend on technology transfer—the capabilities and processes that can speed vaccines ...
  21. [21]
    Accelerating drug production with “just-in-time" manufacturing
    Oct 7, 2024 · “Just-in-time” manufacturing makes drug production faster, flexible and less wasteful, getting vital medicines to patients quicker.
  22. [22]
    Long-acting parenteral drug delivery systems for the treatment of ...
    Long-acting parenteral drug delivery systems have the potential to improve the treatment of chronic conditions. These systems use various technologies, such as ...
  23. [23]
    [PDF] Price Declines after Branded Medicines Lose Exclusivity in the US
    Generic drugs greatly reduce the price of medicines. Generics enter ... price reductions for oral medicines. • Oral generics cost 80% less than the brands.
  24. [24]
    Biosimilars can reduce employer Rx costs | Kaiser Permanente
    and are driving down biologic costs. On average, biosimilars cost 50% less than their biologic reference products.
  25. [25]
    The Chemical History of Morphine: An 8000-year Journey, from ...
    Aug 6, 2025 · The isolation of morphine by Friedrich Sertürner in 1804 added an essential pharmacological tool in the treatment of pain and spawned the ...
  26. [26]
    The isolation of morphine by Serturner - PMC - NIH
    Through diligent research, Serturner was the first to successfully isolate and extract morphine crystals from the tarry poppy seed juice.
  27. [27]
    The Discovery of Aspirin's Antithrombotic Effects - PMC - NIH
    In 1897, Felix Hoffman, a German chemist working for the Bayer company, was able to modify salicylic acid to create acetylsalicylic acid, which was named ...
  28. [28]
    The discovery of aspirin: a reappraisal - PMC - NIH
    The page on which Hoffmann reported his synthesis of pure acetylsalicylic acid in 1897 is retained in the archives of Bayer AG in Leverkusen. The significance ...Missing: purification recrystallization
  29. [29]
    [PDF] Pharmaceutical Manufacturing in America: A Brief History
    Abstract: This article reviews the history of drug manufacturing and changes in the compounding of drugs by pharmacies in the United States and outlines ...
  30. [30]
    Analysis of Past and Present Synthetic Methodologies on Medicinal ...
    Nov 16, 2015 · The first observation was the lack of new synthetic reactions that became firmly integrated into modern medicinal chemistry practices, with ...
  31. [31]
    The First Century of Chemical Engineering | Science History Institute
    Nov 11, 2008 · As developed by the two men, “unit operations” referred to the idea that all chemical processes can be analyzed by dividing them into distinct ...Missing: pharmaceutical | Show results with:pharmaceutical
  32. [32]
    Rethinking Antibiotic Research and Development: World War II and ...
    The US pharmaceutical industry's involvement in antibiotics originated in a government-sponsored project during World War II. A historical analysis of the broad ...
  33. [33]
    Pfizer's Penicillin Pioneers – Jasper Kane and John McKeen
    Dec 1, 2017 · Before deep-tank fermentation was developed, penicillin was produced through surface fermentation. Chemicals production via fermentation was new ...
  34. [34]
    Penicillin | National Museum of American History
    On March 1, 1944, Pfizer opened the first commercial large-scale penicillin production plant which used deep tank (submerged) fermentation methods. The ...
  35. [35]
    [PDF] Fact Sheet: The Challenge of Mass Production
    Penicillin production at pharmaceutical company, Eli Lilly. Medics using ... Penicillin quickly became known as the war's. “miracle drug,” curing ...
  36. [36]
    4 historical designations highlight groundbreaking scientific ...
    Mar 2, 2023 · 4 historical designations highlight groundbreaking scientific achievements - Merck.com.
  37. [37]
    The physics of tablet compression. III. Design and construction of an ...
    Aug 7, 2025 · The knowledge of the process of tableting changed radically in the early 1950s, when Higuchi introduced the instrumented compression machines ...Missing: continuous | Show results with:continuous
  38. [38]
    History of Tablet Compression | PDF - Scribd
    Eccentric tablet presses were commonly used through the 1950s when instrumented presses enabled better understanding of the compression process.
  39. [39]
    [PDF] How the Mass Production of Penicillin Became Possible in the Early ...
    With WWII in full swing across Europe, the British pharmaceutical companies could not produce penicillin on a large scale due to the war-related constraints on.
  40. [40]
    Merck's legacy of antimicrobial innovation and action
    Oct 12, 2023 · 1930s. Merck Research Laboratories played a central role in the development of sulfas, the first synthetic antimicrobial.Merck's Legacy Of... · 1930s · 1940s
  41. [41]
    The Controlled Drug Delivery Systems: Past Forward and Future Back
    Apr 30, 2014 · It began in 1952 with the introduction of the first sustained release formulation. The 1st generation (1950-1980) of drug delivery was focused
  42. [42]
    Evolution of drug delivery systems: From 1950 to 2020 and beyond
    Dec 29, 2021 · Modern drug delivery technology began in 1952 with the advent of the Spansule® sustained-release capsule technology, which can deliver a drug for 12 h after ...
  43. [43]
    Transdermal patches: history, development and pharmacology - PMC
    ... Transdermal System (Barr Pharm Labs Div Teva). The corresponding transdermal patches for Japan were first developed by the Nitto Denko Corporation in the 1970s ...
  44. [44]
    Liposomes in Drug Delivery: How It All Happened - PubMed Central
    The use of liposomes in drug delivery and targeting is often discussed in the context of decades marked by significant milestones. Thus, the 1970s are noted ...
  45. [45]
  46. [46]
    Controlled Drug Delivery Systems: Current Status and Future ...
    The membrane is a thin film that controls the diffusion rate of the drug from the patch to the skin. The adhesive layer helps the patch adhere to the skin [18].
  47. [47]
    Novel drug-delivery systems for hypertension - PubMed
    Potential advantages include reduced dosing frequency, enhanced compliance and convenience, reduced toxicity, stable drug levels, uniform drug effect, and ...
  48. [48]
    Memorable Milestones: 40 Years of ISPE
    1980. In August six men met to discuss the formation of an engineering society that would primarily focus on education, networking, and the exchange of ...
  49. [49]
    Founding Members | ISPE | International Society for Pharmaceutical ...
    44 years ago, a small group of innovative pharmaceutical engineers recognized the need for better manufacturing facility development guidelines and created ISPE ...
  50. [50]
    About - AIChE
    Charted: 1966 Dues: $10 Mission The FP&BE division ("Division 15") provides engineers and scientists interested in the field of food, pharmaceuticals, ...
  51. [51]
    ISPE Baseline® Guide: Volume 1 – Active Pharmaceutical ...
    In November 1994, the ISPE sanctioned the beginning of this important projectand the first, “Bulk Pharmaceutical Chemicals,” was published in June 1996. The ...
  52. [52]
    Homepage | ISPE | International Society for Pharmaceutical ...
    The International Society for Pharmaceutical Engineering (ISPE) is a global nonprofit association serving its members through leading scientific, technical ...About ISPE · Conferences · Pharmaceutical Engineering · Membership
  53. [53]
    ISPE Affiliates and Chapters Span the Globe
    ISPE has more than 21000 members in more than 120 countries worldwide. As an ISPE member, you have access to this network, which can be exciting and ...
  54. [54]
    ASEAN Effort Progressing on CMC/GMP Harmonization, But ...
    Dec 20, 2010 · At the ISPE session, Stoltz and Komas both pointed to the positive impact of the ASEAN country participation in PIC/S on industry. Noting that ...<|control11|><|separator|>
  55. [55]
    [PDF] PHEN 603 - Pharmaceutical Unit Operations
    Sep 4, 2018 · Course Description: This course covers state-of-the-art pharmaceutical processing involving primarily liquid and dispersed-phase systems, ...
  56. [56]
  57. [57]
    In-Line Purification: A Key Component to Facilitate Drug Synthesis ...
    We discussed four techniques: scavenger resins, distillation, nanofiltration, and extraction. They are potential tools for drug synthesis. They can be ...
  58. [58]
    MILLING OPERATIONS IN THE PHARMACEUTICAL INDUSTRY
    Mar 29, 2019 · Milling is a powerful unit operation to aid in the control of particle size for a variety of processing, bioavailability, reactivity, and safety-related ...
  59. [59]
    The Pharmaceutical Drying Unit Operation: An Industry Perspective on Advancing the Science and Development Approach for Scale-Up and Technology Transfer
    **Summary of Drying Processes in Pharmaceuticals from "The Pharmaceutical Drying Unit Operation: An Industry Perspective on Advancing the Science and Development Approach for Scale-Up and Technology Transfer" by Various Authors**
  60. [60]
    Comparative Studies of Different Preservation Methods and Relative ...
    Tg′, Teu, and Tc of the Formulations Measured with DSC and FDM, Residual Moisture Content ... below 2%, avoiding the action plasticizer, which can lower ...
  61. [61]
    Economic Analysis of Batch and Continuous Biopharmaceutical ...
    Unlike batch processing, continuous processing is not widely used and is not well defined, even in many research studies on continuous biopharmaceutical ...
  62. [62]
    SBE Special Section: Biopharmaceuticals - Continuous Processing ...
    The transition from batch to continuous downstream processing of biologics has been gradual, with some processes integrating continuous and batch operations.
  63. [63]
    On Free Energy Calculations in Drug Discovery - ACS Publications
    Oct 10, 2025 · Binding free energy is a crucial metric in drug discovery, as it measures the affinity of a ligand for its target receptor. Free energy and ...
  64. [64]
    Thermodynamic Studies for Drug Design and Screening - PMC - NIH
    Synthetic, rationally designed drugs have a proportionately greater favorable entropy contribution to binding free energy than natural, biological ligands [13] ...
  65. [65]
    [PDF] Thermodynamic stability and transformation of pharmaceutical ...
    Thermodynamic stability of polymorphs. The equilibrium conditions between the solid and liquid phases can be expressed by eq. 1 using the chemical potential ...
  66. [66]
    Phase equilibrium and mechanisms of crystallization in liquid–liquid ...
    Jan 15, 2015 · Introduction. Crystallization is a key step in production of a wide range of chemical products, e.g. organic fine chemicals and pharmaceuticals.Phase Equilibrium And... · Graphical Abstract · IntroductionMissing: vapor- | Show results with:vapor-
  67. [67]
    A practical guide to pharmaceutical polymorph screening & selection
    This review introduces the basic concepts of “What is polymorphism?”, addresses a fundamental question of “Why do polymorphs form?”, and provides practical ...
  68. [68]
    A Brief Introduction to Chemical Reaction Optimization
    (98,99) The determination of reaction orders and rate laws within a model is used to optimize outputs but also to increase overall chemical understanding.
  69. [69]
    Flow synthesis kinetics for lomustine, an anti-cancer active ...
    Jul 19, 2021 · In this study, we postulate kinetic rate laws based on hereby proposed reaction mechanisms presented for the first time in the literature for this API ...Missing: optimization | Show results with:optimization
  70. [70]
    Modelling chemical kinetics of a complex reaction network of active ...
    The characterization and understanding of reaction kinetics is therefore an important part of the chemical development in the pharmaceutical industry [1].
  71. [71]
    Algorithm for the early prediction of drug stability using bayesian ...
    Sep 5, 2023 · In such studies, the Arrhenius equation has been used to predict the stability of the solid state of a pharmaceutical product under the ...
  72. [72]
    An Introduction to the Accelerated Stability Assessment Program
    Aug 15, 2017 · Based on the Arrhenius equation the chemical degradation increases with the temperature and therefore it should be possible to project the ...
  73. [73]
    Cell factories for insulin production
    Oct 2, 2014 · Recombinant human insulin was first produced in E. coli by Genentech in 1978, using a approach that required the expression of chemically ...Missing: aerobic fermentation
  74. [74]
    Cell culture processes for monoclonal antibody production - PMC
    A typical stirred tank bioreactor is equipped with temperature, pressure, agitation, pH and dissolved oxygen controls. Figure 4 illustrates the affects of cell ...
  75. [75]
    Trends in Monoclonal Antibody Production Using Various Bioreactor ...
    This review describes recent trends in high-density cell culture systems established for monoclonal antibody production that are excellent methods to scale up.
  76. [76]
    Recovery and purification process development for monoclonal ...
    The first unit operation in a downstream process is the removal of cells and cell debris from the culture broth and clarification of the cell culture ...Missing: biomanufacturing | Show results with:biomanufacturing
  77. [77]
    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 ...
  78. [78]
    Advancing Monoclonal Antibody Manufacturing - PubMed Central
    Apr 9, 2025 · This review examines recent advances in monoclonal antibody (mAb) manufacturing, focusing on process optimization, cost reduction strategies, and emerging ...
  79. [79]
    Pharmaceutical Formulation - StatPearls - NCBI Bookshelf - NIH
    Pharmaceutical formulation is the multistep process where the active drug is mixed with all other components by considering the factors of particle size, ...Missing: solid liquid semi-
  80. [80]
    Pharmaceutical assessment of polyvinylpyrrolidone (PVP)
    Sep 2, 2020 · It can produce non-friable tablets with optimum hardness and a reliable rate of dissolution. PVP can also be used as a dry binder on mixing ...
  81. [81]
    PVP in Pharmaceuticals: The Role of Povidone as a Binder
    Oct 27, 2025 · It helps to agglomerate powder particles into granules, providing the necessary cohesion for the tablet to withstand the stresses of compression ...
  82. [82]
    Pharmaceutical Excipients and Drug Metabolism: A Mini-Review - NIH
    Nov 3, 2020 · The functional roles of pharmaceutical excipients include modulating bioavailability and solubility of APIs, increasing the stability of APIs ...
  83. [83]
    CHAPTER 1: Preformulation Studies - Books
    Jun 25, 2018 · The chapter covers the measurement of solubility and dissolution rate, molecular dissociation, pKa, diffusion, partition and permeability; and ...
  84. [84]
    High-Speed Tableting of High Drug-Loaded Tablets Prepared ... - NIH
    Apr 13, 2023 · While the maximum recommended compression force for this punch type was 19 kN, the compaction force was varied between 4.8 and 15.5 kN. The ...
  85. [85]
    A Review of Disintegration Mechanisms and Measurement ...
    Pharmaceutical solid dosage forms (tablets or capsules) are the predominant form to administer active pharmaceutical ingredients (APIs) to the patient.
  86. [86]
    Disintegration of Highly Soluble Immediate Release Tablets - NIH
    One of the ICH Q6A Decision Trees #7(1) criterion for using disintegration test for drug release is >80% dissolution in 15 min across the physiological pH range ...
  87. [87]
    [PDF] Q8(R2) Pharmaceutical Development - FDA
    • Identifying potential critical quality attributes (CQAs) of the drug product, so that those product characteristics having an impact on product quality can be ...
  88. [88]
    A review on the scale-up of high-shear wet granulation processes ...
    Mechanical mixing or air introduction helps to uniform granulation liquid distribution in high/low-shear granulators and fluidized-bed processes, respectively ( ...
  89. [89]
    [PDF] 〈905〉 UNIFORMITY OF DOSAGE UNITS - US Pharmacopeia (USP)
    Aug 1, 2023 · The term “uniformity of dosage unit” is defined as the degree of uniformity in the amount of the drug substance among dosage units. Therefore, ...
  90. [90]
    NIR spectroscopic method for the in-line moisture assessment ...
    This study focuses on the thorough validation of an in-line NIR based moisture quantification method in the six-segmented fluid bed dryer of a continuous ...
  91. [91]
    Continuous Manufacturing: Recent Trends and Analysis
    Mar 27, 2024 · Over the past few years, the process of continuous manufacturing is continuously gaining momentum in the pharmaceutical and biotech industry.
  92. [92]
    Q13 Continuous Manufacturing of Drug Substances and Drug ... - FDA
    Mar 1, 2023 · This guidance describes scientific and regulatory considerations for the development, implementation, operation, and lifecycle management of continuous ...Missing: 2020s | Show results with:2020s
  93. [93]
    Sterilization for Medical Devices - FDA
    Medical devices are sterilized in a variety of ways including using moist heat (steam), dry heat, radiation, ethylene oxide (EtO) gas, vaporized hydrogen ...
  94. [94]
    Treatment of Clinical Solid Waste Using a Steam Autoclave as ... - NIH
    Log reduction of bacteria in sterilized waste. Sterilization conditions: temperature 121 °C, time 60 minutes and pressure 15 psi. Bacteria, Initial ...
  95. [95]
    Radiation sterilization of new drug delivery systems - PMC - NIH
    Sterilization by γ radiation is used for the sterilization of active substances as well as drug delivery systems. The new studies on radiation sterilization of ...
  96. [96]
    Gamma irradiation of animal sera for inactivation of viruses and ...
    The available data indicate that inactivation of mollicutes in serum is essentially complete at the gamma radiation doses normally employed (25-40 kGy), while ...
  97. [97]
    Which parameters must be validated during a steam sterilization ...
    Dec 7, 2021 · For example, an SAL of ≤ 10-6 means that there is a probability of one in a million devices being non-sterile after exposure to a sterilization ...
  98. [98]
    The limits of sterility assurance - PMC - NIH
    A sterility assurance level (SAL) of 10 –6 is generally accepted for pharmacopoeial sterilization procedures, ie, a probability of not more than one viable ...Missing: stearothermophilus | Show results with:stearothermophilus
  99. [99]
    [PDF] generalChapter671RevisionBull...
    The packs as tested in Method II are designated Class A if no pack tested exceeds 0.5 mg per day in average blister moisture per- meation rate; they are ...Missing: engineering | Show results with:engineering
  100. [100]
    [PDF] Guidance for Industry on Container Closure Systems for ... - FDA
    The most common forms of flexible packaging are the blister package and the pouch. ... Because LDPE is not a particularly good moisture barrier, a drug.
  101. [101]
    USP <671> - Smithers
    USP General Chapter <671> provides a series of test methods specific to the functionality and performance of containers and package systems.Missing: HDPE | Show results with:HDPE
  102. [102]
    How Are Controlled-Release Products Administered?
    Two types of diffusion based controlled release formulations – matrix and reservoir are most commonly used for extended or sustained release applications.Missing: engineering | Show results with:engineering
  103. [103]
    Higuchi's equation and beyond: Overview of the formulation and ...
    Nov 1, 2012 · The Higuchi model describes the rate of release of drug from a matrix where the drug loading exceeds it solubility in the matrix into a ...
  104. [104]
    Swelling-Controlled Release | SpringerLink
    Swelling-controlled release occurs when a polymer's interaction with the elution medium causes a glass-to-gel transition, controlling the release of the active ...
  105. [105]
    Quantification of Swelling and Erosion in the Controlled Release of ...
    In swellable matrices loaded with sparingly or slightly soluble drugs, solid drug particles can be transported within the hydration layer by polymer swelling.
  106. [106]
    Polymer particle erosion controlling drug release. I. Factors ...
    Erosion controlled systems are prepared by simply incorporating the drug in water soluble polymeric carriers (hydrocolloids) which are available in a broad ...
  107. [107]
    Osmotic Pump Drug Delivery Systems—A Comprehensive Review
    Nov 18, 2022 · A patent was granted to Alza Corporation in 1976 for an oral osmotic pump. The osmotic bursting drug delivery system was then developed in 1979 ...
  108. [108]
    [PDF] 〈711〉 DISSOLUTION - US Pharmacopeia (USP)
    〈711〉 DISSOLUTION basket. The vessel is partially immersed in a suitable water bath of any convenient size or heated by a suitable device.
  109. [109]
    A Review of Liposomes as a Drug Delivery System - PubMed Central
    Feb 17, 2022 · Liposomes have been considered promising and versatile drug vesicles. Compared with traditional drug delivery systems, liposomes exhibit better properties.Missing: engineering | Show results with:engineering
  110. [110]
    The suitability of liposomes for the delivery of hydrophobic drugs
    In this study, curcumin was chosen as a model drug to investigate the suitability of liposomes for the delivery of hydrophobic drugs.Research Paper · Introduction · Liposome Characterization...
  111. [111]
    Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role ...
    Passive targeting of NPs is mainly achieved by the enhanced permeability and retention (EPR) effect, which exploits the increased vascular permeability and ...
  112. [112]
    Antibody-conjugated nanoparticles for target-specific drug delivery ...
    Sep 4, 2023 · Antibody-conjugated nanoparticles (ACNPs) are an effective targeted therapy for the efficient delivery of chemotherapeutics specifically to the targeted cancer ...
  113. [113]
    Smart nanoparticles for cancer therapy - Nature
    Nov 3, 2023 · Smart nanoparticles, which can respond to biological cues or be guided by them, are emerging as a promising drug delivery platform for precise cancer treatment.
  114. [114]
    Methods of Liposomes Preparation: Formation and Control Factors ...
    This article analyzes the main features of the formation and fabrication techniques of liposome nanocarriers, with a special focus on the structure, parameters,
  115. [115]
    Liposome Formation in the Tube Process Using Supercritical CO2
    A novel flow process for liposome production in a microfluidic tube using supercritical CO 2 (scCO 2 ) named as LipTube has been developed in this work.
  116. [116]
    Doxil®--the first FDA-approved nano-drug: lessons learned - PubMed
    Jun 10, 2012 · Doxil, the first FDA-approved nano-drug (1995), is based on three unrelated principles: (i) prolonged drug circulation time and avoidance of the RES due to the ...Missing: half- life
  117. [117]
    [PDF] DOXIL Label - accessdata.fda.gov
    the elimination half-life of DOXIL is longer and the clearance lower compared ... liposomal doxorubicin suggests that DOXIL is largely confined to vascular.
  118. [118]
    [PDF] FDA Guidance for Industry PAT – A Framework for Innovative ...
    The scientific, risk-based framework outlined in this guidance, Process Analytical Technology or. PAT, is intended to support innovation and efficiency in ...
  119. [119]
    PAT — A Framework for Innovative Pharmaceutical Development ...
    Aug 24, 2018 · This guidance is intended to describe a regulatory framework (Process Analytical Technology, PAT) that will encourage the voluntary development and ...
  120. [120]
    Measurement of an Amorphous Form in a Pharmaceutical Drug ...
    Aug 1, 2010 · The prediction accuracy of amorphous content with respect to API, in a drug product of low drug load, was less than 2.00% at both sites. The ...
  121. [121]
    [PDF] On-Line HPLC as a Process Analytical Technology (PAT) for ...
    This poster discusses how on-line HPLC is a tool that can be utilized to measure the product purity in near real time which can reduce product variability, ...
  122. [122]
    Raman spectroscopy as a process analytical technology for ...
    Aug 4, 2016 · Raman spectroscopy has made a positive impact as a process analytical and control tool for pharmaceutical manufacturing and bioprocessing.Raman Spectroscopy As A... · Instrumentation And Data... · Bioprocessing<|control11|><|separator|>
  123. [123]
    Advantages of Integrated Continuous Manufacturing Throughout the ...
    Advanced process analytical technology (PAT) tools, including real-time pH, density, Raman, and infrared monitoring, feed into closed-loop control systems ...
  124. [124]
    Model-based real-time optimization in continuous pharmaceutical ...
    In this work, real-time optimization (RTO) schemes are proposed and applied on a continuous pharmaceutical manufacturing process which consists of three units.Missing: adjustment | Show results with:adjustment
  125. [125]
    Aspects and Implementation of Pharmaceutical Quality by Design ...
    May 8, 2025 · Rooted in ICH Q8–Q11 guidelines, QbD emphasizes defining Critical Quality Attributes (CQAs), establishing design spaces, and integrating risk ...
  126. [126]
    [PDF] Annex 2
    The first WHO draft text on good manufacturing practices (GMP) was prepared in 1967 by a group of consultants at the request of the Twentieth World Health.
  127. [127]
    [PDF] Annex 6 WHO good manufacturing practices for sterile ...
    Clean room and clean-air device classification. 4.6 Clean rooms and clean-air devices should be classified in accordance with ISO 14644 (2–3, 5–7). 4.6.1 ...
  128. [128]
    [PDF] 20220825_gmp-an1_en_0.pdf
    Aug 22, 2022 · This document provides technical guidance on the principles and guidelines of good manufacturing practice (GMP) for medicinal products as laid ...
  129. [129]
    [PDF] Q8(R2) - ICH
    This guideline is intended to provide guidance on the contents of Section 3.2.P.2. (Pharmaceutical Development) for drug products as defined in the scope of ...
  130. [130]
    [PDF] quality risk management q9(r1) - ICH
    Jan 18, 2023 · Risk management principles are effectively utilized in many areas of business and government including finance, insurance, occupational safety, ...
  131. [131]
    [PDF] Pharmaceutical Quality System Q10 - ICH
    Jun 4, 2008 · ICH. Q10 is a model for a pharmaceutical quality system that can be implemented throughout the different stages of a product lifecycle. Much of ...
  132. [132]
    [PDF] Analytical Procedures and Methods Validation for Drugs and Biologics
    quality, purity, and potency of drug substances and drug products. 6. It will help you assemble. 21 information and present data to support your analytical ...
  133. [133]
    Analytical Methods: A Statistical Perspective on the ICH Q2A and ...
    Depending on the method being validated, acceptance criteria should be established. For the following example, one can use a percent recovery of 95–105%. A ...
  134. [134]
    [PDF] Dissolution Testing of Immediate Release Solid Oral Dosage Forms
    This guidance is developed for immediate release (IR) dosage forms and is intended to provide. (1) general recommendations for dissolution testing; (2) ...
  135. [135]
    [PDF] Q1A(R2) Guideline - ICH
    The purpose of the stability study is to establish, based on testing a minimum of three batches of the drug substance and evaluating the stability information.
  136. [136]
    21 CFR Part 211 -- Current Good Manufacturing Practice for ... - eCFR
    The regulations in this part contain the minimum current good manufacturing practice for preparation of drug products (excluding positron emission tomography ...Title 21 · Subpart I —Laboratory Controls · Subpart D —Equipment · 211.1 – 211.3
  137. [137]
    [PDF] Process Validation: General Principles and Practices | FDA
    pharmaceuticals are provided in 21 CFR parts 210 and 211. The CGMP regulations require that manufacturing processes be designed and controlled to assure ...
  138. [138]
    Quality Guidelines - ICH
    A more flexible approach to pharmaceutical quality based on Good Manufacturing Practice (GMP) risk management.
  139. [139]
    Home | WHO - Prequalification of Medical Products (IVDs, Medicines ...
    WHO Essential Medicines and Health Products · WHO Global Malaria Programme · WHO Medical Devices · WHO Medicines Quality Assurance · WHO Regulation and ...Medicines · About Medicines Prequalification · Prequalification Reports · Vaccines
  140. [140]
    ISO 15378:2017 - Primary packaging materials for medicinal products
    In stock 2–5 day deliveryISO 15378:2017 is an application standard for the design, manufacture and supply of primary packaging materials for medicinal products.
  141. [141]
    [PDF] Impurities in new drug substances Q3A (R2) - ICH
    Oct 25, 2006 · Guideline Q1A on Stability) used to identify potential impurities arising during storage. The impurity profile of the drug substance batches ...
  142. [142]
    [PDF] Guidance for Industry Q3B(R2) Impurities in New Drug Products - FDA
    This guidance provides recommendations for registration applications on the content and qualification of impurities in new drug products produced from ...Missing: harmonized Q1A
  143. [143]
    Prequalification of medicines by WHO
    Jan 31, 2013 · WHO prequalification of medicines is a service provided by WHO to assess the quality, safety and efficacy of medicinal products.
  144. [144]
    Medicines | WHO - Prequalification of Medical Products (IVDs ...
    The standards used to evaluate FPPs and APIs, and their manufacturing sites, are based on the principles and practices agreed by the world's leading regulatory ...WHO Medicines... · Prequalification Reports · Prequalification Pipeline
  145. [145]
    ISO 13485:2016 - Medical devices — Quality management systems
    In stock 2–5 day deliveryWhat is ISO 13485? ISO 13485 is the internationally recognized standard for quality management systems in the design and manufacture of medical devices.Missing: pharmaceutical overlap
  146. [146]
    European Union (EU) Mutual Recognition Agreement - FDA
    The U.S.-EU Mutual Recognition Agreement (MRA) Sectoral Annex for Pharmaceutical Good Manufacturing Practices (GMP) MRA entered into force on November 1, 2017.
  147. [147]
    [PDF] EU-US agreement on mutual recognition of pharmaceutical ...
    Mar 1, 2017 · 1. A Party shall recognize pharmaceutical inspections and accept official GMPs documents issued by a recognized authority of the other Party for ...
  148. [148]
    Pharmaceutical GMP Professional Certification (CPGP) | GMP Certification ASQ
    ### Certified Pharmaceutical GMP Professional (CPGP) Summary
  149. [149]
    Certified Quality Engineer (CQE) Certification | ASQ
    ### Certified Quality Engineer (CQE) Summary
  150. [150]
  151. [151]
    Pharmaceutical Project Manager | George Washington University
    Apr 20, 2020 · The path to becoming a pharmaceutical project manager typically begins with earning a four-year undergraduate degree. The fields we mentioned ...
  152. [152]
    Chemical Engineers : Occupational Outlook Handbook
    The median annual wage for chemical engineers was $121,860 in May 2024. Job Outlook. Employment of chemical engineers is projected to grow 3 percent from 2024 ...
  153. [153]
    Guidelines For Optimum Water Consumption in Bulk Drugs ...
    Guidelines For Optimum Water Consumption in Bulk Drugs Manufacturing Industry PDF. Central Pollution Control Board has taken up various projects covering ...
  154. [154]
    Pharmaceutical Company Targets and Strategies to Address ... - NIH
    Feb 11, 2023 · Health systems produce 4–5% of national greenhouse gas (GHG) emissions [1], contributing to climate change, with its subsequent negative impact ...
  155. [155]
    API manufacturing: The cost of decarbonization | McKinsey
    Jul 26, 2024 · In 2022, approximately 35 percent of solvent waste generated in the US pharmaceutical industry was recycled, while the remaining 65 percent was ...
  156. [156]
    [PDF] Green Synthesis in Drug Development: Advances in Enzyme
    Sep 15, 2024 · Enzymatic reactions often operate under mild conditions, such as ambient temperatures and neutral. pH, reducing energy consumption by up to 50%.
  157. [157]
    Green Chemistry for Sustainable API development - Patheon
    Jul 16, 2024 · To mitigate their impact, we focus on reducing solvent usage and improving recycling processes. Evaluating solvents carefully, considering their ...
  158. [158]
    Twelve Principles of Green Chemistry - Open Textbook Publishing
    Ideally, the e-factor would be zero. Oil refining industries report e-factors <0.1 whereas pharmaceutical industries report a range from 25-100. Process ...
  159. [159]
    12 Principles of Green Chemistry - American Chemical Society
    Solvents and separation agents provide for mass and energy transfer and without this, many reactions will not proceed. It has also been shown that solvents ...
  160. [160]
    Case Study - Pfizer - Science Based Targets Initiative
    For example, since 2000, approximately 3300 energy projects have been implemented resulting in roughly $150 million in annualized savings and a reduction of ...Missing: chemistry 1.5