View Article

Abstract

Pharmaceutical technology is crucial for the advancement of innovative drug delivery systems that significantly enhance the efficacy of treatments and improve patient outcomes. With rapid technological progress, the pharmaceutical landscape has witnessed transformative changes, particularly with the emergence of personalized medicines and targeted therapies that cater to the individual needs of patients. These innovations have been pivotal in addressing the complexities of various diseases by allowing for tailored therapeutic approaches that optimize treatment efficacy and minimize side effects. One of the most exciting developments in this field is the application of nanotechnology. Nanoparticles and nanocarriers facilitate the precise delivery of therapeutics, ensuring that drugs reach their intended targets with improved bioavailability and reduced systemic toxicity. Additionally, the advent of 3D printing technology has opened new avenues for the customization of drug formulations, enabling the production of complex dosage forms that can be tailored to patient-specific requirements, including varying dosages and release profiles. Artificial intelligence (AI) is also reshaping pharmaceutical technology, enhancing drug discovery processes, optimizing clinical trials, and personalizing treatment plans through data analysis and predictive modeling. Furthermore, continuous manufacturing represents a significant shift from traditional batch production methods, offering advantages such as increased efficiency, reduced costs, and faster response times to market demands. This review consolidates these key advancements in pharmaceutical technology,illustrating their collective impact on the development of novel drug delivery systems and therapeutic innovations. By exploring these trends, we can better understand the future direction of pharmaceutical technology and its potential to revolutionize healthcare, ultimately leading to improved patient care and outcomes.

Keywords

Pharmaceutical technology, nanotechnology, Nanoparticles, 3D printing, Artificial intelligence, novel drug delivery systems

Introduction

Molecular diagnosis encompasses a range of advanced technologies designed to identify, elucidate, and monitor human diseases at a molecular level. This field extends beyond traditional DNA diagnostics to include the analysis of RNA, genes, proteins, and other biomolecules, providing a comprehensive approach to understanding complex diseases. Utilizing techniques such as polymerase chain reaction (PCR), sequencing, and microarrays, molecular diagnosis enables the detection of genetic mutations, pathogen identification, and assessment of gene expression profiles, which can inform treatment decisions and predict disease outcomes. By providing precise molecular information, it enhances early detection and risk stratification of conditions such as cancer, infectious diseases, and genetic disorders. Additionally, molecular diagnostics facilitates personalized medicine, allowing healthcare providers to tailor therapies according to individual genetic and molecular profiles. The integration of bioinformatics and computational tools further strengthens the ability to interpret complex data, highlight relevant biomarkers, and support clinical decision-making. As technology advances, the scope of molecular diagnosis continues to expand, promising to improve patient outcomes through earlier intervention and more effective treatment strategies. Ultimately, the clinical application of molecular technologies represents a transformative shift in the approach to diagnosing and managing diseases, paving the way for innovations in healthcare that are grounded in a deeper understanding of individual molecular characteristics.Molecular diagnostics extends far beyond traditional approaches, encompassing innovative techniques such as in vivo imaging and the examination of biological processes at the single-molecule level. This advanced scope allows for a deeper understanding of disease mechanisms and enhances diagnostic accuracy. Techniques such as fluorescence microscopy, magnetic resonance imaging (MRI), and positron emission tomography (PET) are pivotal in visualizing molecular interactions in real-time, facilitating early diagnosis and treatment monitoring. The ability to track the behavior of single molecules within living organisms has revealed invaluable insights into biochemical pathways and disease progression, notably in cancers and neurological disorders. Simultaneously, industries involved in molecular diagnostics, including biotechnology, pharmaceuticals, and clinical laboratories, are continuously striving for innovation to exceed customer expectations. They employ various methodologies, such as high-throughput screening, next-generation sequencing, and biochip technology, to develop and deliver precise diagnostic tools. This aligns with a growing demand for personalized healthcare solutions, where diagnostic insights directly inform treatment strategies tailored to individual patient profiles. Furthermore, regulatory agencies and standardization bodies are increasingly emphasizing the importance of quality control and validation in diagnostic products, driving industries to invest in enhanced research and development frameworks. Collaborations between academia, industry, and healthcare providers are essential for fostering innovation, ensuring the translational potential of molecular diagnostic advancements. By leveraging cross-disciplinary expertise, these collaborations aim to develop rapid, cost-effective diagnostic solutions that can be integrated into clinical settings, ultimately improving patient care and healthcare outcomes. The evolution of molecular diagnostics, enriched by cutting-edge technologies and robust industry efforts, positions it as a cornerstone of modern medical practice and research. The pharmaceutical industry is instrumental in advancing healthcare and saving lives through the development of effective medications and therapies. Continuous research and innovation focused on small molecules are essential for creating better drugs that can address diverse health challenges. Small molecules offer numerous advantages, including the ability to efficiently target specific biochemical pathways, facilitating rapid absorption and improved bioavailability. By enhancing product efficacy and safety, pharmaceutical companies aim to meet the growing demands of healthcare practitioners and patients alike, contributing to an overall increase in customer satisfaction. In parallel, the field of nanorobotics is emerging as a groundbreaking area with the potential to revolutionize drug delivery and medical procedures. Nanorobots, designed to operate at the nanoscale (typically measuring between 1 and 100 nanometers), can navigate physiological environments with remarkable precision. These tiny robots are capable of performing complex tasks, such as targeted drug delivery, where they can transport therapeutic agents directly to diseased cells, minimizing systemic side effects and maximizing therapeutic effects. Furthermore, advancements in nanorobotics enable precise manipulation of nanoscale objects which can lead to innovations in diagnostics, including real-time imaging and biomarker detection. The intersection of nanotechnology and pharmaceuticals opens up exciting opportunities for novel therapeutics, particularly in oncology and chronic disease management. By combining the targeted capabilities of nanorobots with the therapeutic potential of small molecules, researchers can develop more effective treatment modalities. As the pharmaceutical industry continues to explore the benefits of nanorobotics, there is considerable potential not only for enhanced drug efficacy and delivery but also for improving patient outcomes and overall healthcare experiences. The integration of these technologies promises to propel the industry toward a future characterized by more personalized and effective therapeutic interventions. A nanorobot is an advanced, active nanostructure that possesses the capability to perform multiple functions essential for various applications at the nanoscale, including actuation, sensing, manipulation, propulsion, signaling, and information processing. These functions enable nanorobots to operate in complex environments, particularly within biological systems, where they can facilitate a range of tasks with unprecedented precision. One of the most intriguing prospects of nanorobots is their potential involvement in molecular biology, particularly in manipulating DNA.  Through techniques like DNA hacking, nanorobots can be engineered to modify genetic material to correct mutations, deliver gene therapies, or even edit genes using technologies such as CRISPR-Cas9. This ability opens the door to transformative applications in medicine, including personalized treatments for genetic disorders, cancer therapies that target specific genetic mutations, and the potential to combat infectious diseases through rapid response mechanisms. By precisely targeting and altering specific sequences within the genome, nanorobots have the promise to usher in a new era of precision medicine that tailors therapeutic interventions to the individual patient’s genetic profile. However, with these capabilities come significant ethical and safety considerations. The notion of "DNA hacking" raises critical questions about the consequences of altering genetic material, both at an individual and societal level. Ensuring responsible use, developing regulatory frameworks, and addressing potential misuse are essential components as researchers navigate this cutting-edge field. Ultimately, while nanorobots present tremendous opportunities to advance biotechnology and medicine, they also necessitate ongoing discussions about their impact on humanity, emphasizing the importance of ethical governance in the pursuit of scientific revolution. Nanomolecular diagnostics, or "nanodiagnostics," represents a cutting-edge application of nanobiotechnology in the realm of molecular diagnostics, as first articulated by Jain in 2003. This innovative field leverages the unique properties of nanomaterials to enhance diagnostic processes, enabling the detection and analysis of diseases at unprecedented sensitivity and specificity levels. Nanodevices and nanosystems, including those designed for sequencing single molecules of DNA, exemplify the transformative potential of nanotechnology in diagnostics. These systems can identify genetic variations, pathogen presence, or disease states with remarkable accuracy, leading to quicker and more reliable diagnoses. The integration of inorganic nanostructures into biological systems as biomarkers amplifies the potential of nanodiagnostics. These nanostructures, such as nanoparticles, quantum dots, and nanoshells, can be engineered to bind to specific biological targets, allowing for the early detection of diseases including cancer and infectious diseases. Their small size, coupled with high surface-area-to-volume ratios, contributes to their ability to interact with biomolecules, facilitating signal amplification and enhancing detection capabilities. Furthermore, due to their tunable optical, electrical, and magnetic properties, these nanomaterials can provide real-time imaging and monitoring, enabling clinicians to assess disease progression and treatment responses more effectively. As research expands in this domain, the prospects for nanodiagnostics are promising, with potential applications spanning personalized medicine, point-of-care testing, and biosensing. However, alongside these advances, it is crucial to address safety, regulatory, and ethical considerations to ensure that nanomaterials are used responsibly and effectively in clinical settings. Ultimately, nanomolecular diagnostics holds the key to revolutionizing the landscape of healthcare by enhancing diagnostic accuracy, enabling timely intervention, and paving the way for personalized therapeutic strategies. The development of a new class of nanoscale probes is poised to revolutionize the detailed monitoring and analysis of receptors, pores, and other functional components of living cells, which operate at the nanoscale. These nanoscale probes are engineered to interact with biological components on a molecular level, providing unprecedented insights into cellular activities and processes. By utilizing materials such as nanoparticles, nanowires, and carbon nanotubes, researchers can create highly sensitive tools that enhance the detection and quantification of biomolecules, allowing for real-time observation of cellular functions. One of the most significant advancements lies in the use of nanoscale particles as tags or labels in biological tests. These nanoparticles can be designed to attach to specific biomolecules, enabling the visualization of their presence and activity within complex biological systems. The small size of these particles means they can penetrate tissues and cells more effectively than conventional markers, thus improving the sensitivity and specificity of assays. Moreover, the unique optical, electrical, and magnetic properties of nanoscale particles facilitate various detection methods, such as fluorescence, surface plasmon resonance, and magnetic resonance imaging, enabling diverse applications across diagnostics and research. The flexibility offered by nanoscale probes also enhances the potential for multiplexing, where multiple assays can be conducted simultaneously on a single sample, significantly reducing time and costs associated with testing. This capability allows for comprehensive profiling and analysis, essential in areas such as personalized medicine and drug discovery, where understanding the interactions of multiple biomolecules is critical. As these nanoscale probes continue to evolve, they will not only improve the efficiency of biological tests but also deepen our understanding of cellular mechanisms, leading to breakthroughs in medical research and therapeutic development. The exciting potential of nanoscale probes thus represents a significant leap forward in the field of molecular diagnostics and cellular biology. Nanotechnology holds significant promise in enhancing the sensitivity and integration of analytical methods, allowing for a more coherent evaluation of life processes. In particular, it can address the challenges associated with the therapeutic application of nucleic acids, which are large, negatively charged macromolecules. Due to their inherent physicochemical properties, nucleic acids face several biological barriers that hinder their effectiveness as therapeutic agents. These limitations include nuclease degradation, which results in the rapid breakdown of nucleic acids in biological environments; unfavorable pharmacokinetics and pharmacodynamics, which impair their distribution and efficacy in target tissues; and clearance via the reticuloendothelial system (RES), which leads to premature elimination from the bloodstream. Additionally, the ability of nucleic acids to permeate cellular membranes is compromised due to their size and charge, making it difficult for them to enter target cells effectively. Once inside, they must navigate intracellular trafficking to reach their designated sites for action, which further complicates their therapeutic application. Nanotechnology provides solutions to these challenges by enabling the design of nanocarriers that can encapsulate nucleic acids, protecting them from degradation and facilitating their transport within the body. These nanocarriers can be engineered to improve pharmacokinetics and pharmacodynamics, promoting longer circulation times and enhanced accumulation in target tissues. Moreover, nanocarriers can be modified with targeting ligands that enable them to bind specifically to receptors on target cells, thereby enhancing cellular uptake through endocytosis. This specificity not only increases the efficacy of nucleic acid delivery but also minimizes off-target effects. By overcoming these barriers, nanotechnology not only opens new avenues for the therapeutic use of nucleic acids but also enhances our ability to comprehensively analyze and manipulate complex biological processes at the molecular level, ultimately leading to innovative treatments for various diseases. Upon administration, nucleic acids, particularly RNA, face significant challenges due to the abundant presence of nucleases, such as 3' exonuclease and endonucleases, in biological compartments including plasma and tissues. These nucleases rapidly degrade RNA molecules, limiting their therapeutic potential and making them susceptible to swift inactivation before they can reach their intended targets. This degradation not only diminishes the efficacy of RNA-based therapies but also complicates their use in clinical settings, where maintaining RNA stability is crucial for therapeutic outcomes. Additionally, the pharmacokinetics and pharmacodynamics of RNA are further impaired by a rapid clearance mechanism through the kidneys, which quickly filters and excretes free RNA molecules from circulation. When RNA is associated with larger carriers, such as liposomes or polymeric nanoparticles, the liver becomes a primary site for clearance, exacerbating the problem of systemic distribution. This clearance occurs within minutes, leading to an acute systemic distribution phase that restricts the time available for the RNA to exert its therapeutic effect in target tissues. To overcome these hurdles, researchers are exploring various strategies to enhance the stability and bioavailability of RNA therapies. These strategies include the use of chemical modifications to confer nuclease resistance, the design of nanocarriers that shield RNA from degradation, and the incorporation of targeting moieties that facilitate selective uptake by target cells. Improving the pharmacokinetic and pharmacodynamic profiles of RNA is essential for achieving longer circulation times in the bloodstream, increasing tissue accumulation, and ultimately enhancing the therapeutic efficacy of RNA-based treatments. As advancements in drug delivery systems and nanotechnology continue to evolve, they hold great promise for maximizing the potential of RNA therapies in treating a variety of diseases, including cancer and genetic disorders. The reticuloendothelial system (RES) poses a significant barrier to the in vivo delivery of nucleic acids, primarily due to the highly efficient phagocytic activity of immune cells such as monocytes and macrophages. These cells are adept at recognizing and engulfing foreign particles, including nucleic acid formulations encapsulated in nanocarriers or liposomes. As a result, once nucleic acids are administered, they can be rapidly cleared from circulation, impeding their therapeutic efficacy and distribution to target tissues. This rapid clearance through the RES not only diminishes the half-life of nucleic acid-based therapies but also disrupts the intended pharmacokinetics needed for effective treatment. Consequently, overcoming this challenge necessitates innovative strategies that enhance the stealth properties of nucleic acid formulations, such as PEGylation or the use of biodegradable materials that can evade immune detection. Furthermore, tailoring the size, surface charge, and composition of nanocarriers can significantly influence their interaction with RES components, potentially improving the systemic availability of nucleic acids and extending their therapeutic window. As research in drug delivery systems advances, finding ways to effectively navigate the barriers presented by the RES will be crucial for the successful implementation of nucleic acid therapies in clinical practice. The COVID-19 crisis has unveiled the fundamental interdependence between public contributions and the advancement of medical innovations, particularly in the context of vaccine development. The global response to the pandemic showcased how public funding, collaboration, and expertise were pivotal in accelerating research and development, enabling the rapid deployment of vaccines in an unprecedented timeframe. However, the ongoing inequities in vaccine access have starkly revealed the pitfalls of placing control over such crucial innovations in the hands of a limited number of private corporations. This situation underscores a significant imbalance between the public health interests of populations and the profit-driven motives of these few companies. The disproportionate control exerted by private entities over vaccine technologies and distribution has led to alarming disparities in access, even within wealthier nations. While affluent countries hoarded supplies, many lower-income nations struggled to secure vaccines, exacerbating health inequities on a global scale. This scenario illustrates how valuable public resources, which funded much of the research and development efforts, are being utilized without sufficient accountability measures to ensure that public health needs are prioritized. The lack of equitable access can have devastating consequences, prolonging the pandemic and undermining the collective effort to achieve herd immunity. The deployment of public health interventions necessitates robust checks and balances that align private sector activities with the public interest. This can include mechanisms such as equitable licensing agreements, transparent pricing strategies, and the promotion of open-source approaches to vaccine technology that allow broader access. Furthermore, governments and international organizations must emphasize the importance of global partnerships and solidarity in overcoming the pandemic, ensuring that innovations developed with public funding are accessible to all, particularly those in marginalized communities. The COVID-19 pandemic serves as a critical lesson in the need for a paradigm shift in how medical innovations are governed. It calls for an urgent reevaluation of the current models that prioritize profit over public health. Moving forward, it is essential to establish frameworks that foster collaboration between public and private sectors, ensuring that health innovations are developed and distributed equitably. Ultimately, the protection and promotion of public health must be paramount, requiring a commitment to dismantling existing barriers and redefining the terms of engagement in medical research and innovation. Through these changes, we can work toward a more just and equitable healthcare landscape, particularly in times of crisis. The ongoing challenges within the health and pharmaceutical sectors underscore that tinkering at the margins of the status quo will not yield the transformative solutions needed to address pressing public health issues. Current market-based policies, predominantly shaped by the interests of private corporations, have failed to generate essential health technologies that are universally accessible and affordable. The global response to crises such as the COVID-19 pandemic has highlighted the systemic flaws in these policies, showcasing significant gaps in the development and distribution of vaccines and therapeutics. Therefore, it is imperative to rethink and redesign research and development (R&D) and access policies that are no longer captive to a Western-dominated global health order.  A shift toward needs-driven research and the production of pharmaceuticals is critical. This means prioritizing public health needs over market profits, emphasizing the principle of health as a commons rather than a commodity. Such an approach requires recognizing that healthcare technologies—including vaccines, medications, and medical devices—should be designed and developed with public health goals as their foremost priority. By organizing R&D around public needs, it becomes possible to ensure that innovations are directly relevant to the populations that require them most, rather than being dictated by market forces that predominantly serve wealthier nations or consumers.  To realize this vision, we must establish transparent and equitable governance structures that facilitate collaboration between the public sector, private companies, and civil society. Ensuring transparency in funding, decision-making, and outcomes can foster greater accountability and trust among stakeholders. These collaborative arrangements should be designed to align the interests of private innovators with public health goals, incentivizing companies to prioritize accessibility and affordability in their R&D processes. For instance, public-private partnerships can be structured to require that the results of publicly funded research, whether in terms of patents, licensing, or distribution, are made available to all without exorbitant costs. Additionally, enhancing public capacities for R&D and manufacturing is essential. Governments and international organizations could invest in public sector research institutions and innovation hubs that prioritize health equity. By bolstering local production capabilities in low- and middle-income countries, it becomes possible to decentralize pharmaceutical production, reducing dependency on multinational corporations and ensuring that life-saving technologies are produced closer to the communities that need them. A holistic approach that includes diverse stakeholders will also help expand the dialogue around intellectual property rights, advocating for flexible frameworks that prioritize widespread access over strict commercialization. Models such as open-source pharmaceuticals could enable a more collaborative approach to drug development, allowing multiple entities to contribute to and benefit from innovations without the constraints imposed by proprietary rights. Ultimately, the transition from a market-driven model to one that actively invests in health as a shared global resource is crucial for sustainable change. By prioritizing the public good in health innovation, we have the potential to create a more equitable system that values human life over profit and ensures that essential health technologies are made available to all who need them. Embracing this fundamental shift is not just an ethical imperative but a pathway to a healthier, more just world, equipping us to better address the health challenges of today and tomorrow.

Ai Importance In Pharmacy:

Artificial Intelligence (AI) is revolutionizing drug discovery and development by harnessing its computational power to analyze various data types, including genetic, proteomic, genomic, and clinical information. This capability allows researchers to identify potential therapeutic targets with a level of efficiency and precision previously unattainable. Through sophisticated algorithms and machine learning techniques, AI systems can uncover disease-associated biomarkers and elucidate complex molecular pathways critical to disease progression. This process enables the design of medications that can effectively modulate biological processes, targeting the root causes of diseases rather than merely alleviating symptoms. One of AI's most significant contributions to drug discovery is its ability to efficiently screen vast chemical libraries for potential drug candidates. Traditional approaches to drug discovery often relied on high-throughput screening methods that, while effective, can be time-consuming and resource-intensive. In contrast, AI can rapidly analyze thousands of compounds virtually by simulating chemical interactions and predicting binding affinities to specific biological targets. This predictive capacity is pivotal in prioritizing and selecting lead compounds for experimental testing, effectively streamlining the drug development process. AI employs a variety of techniques, including deep learning, reinforcement learning, and natural language processing. These methodologies allow the system to learn from previously established data sets, refining its predictions and improving accuracy based on the outcomes of earlier studies. For instance, by training on large-scale clinical data sets, AI can identify patterns and correlations that may inform the discovery of novel therapeutic targets. Additionally, AI’s capacity to process unstructured data, such as scientific literature and clinical notes, further enriches the information landscape, yielding insights that enhance the drug discovery pipeline. Moreover, AI's ability to simulate and predict chemical interactions is essential in the early phases of drug development. By modeling how potential drug candidates will interact with biological targets, researchers can assess factors such as specificity and efficacy, facilitating the identification of compounds with the highest probability of success in subsequent preclinical and clinical trials. This predictive modeling significantly reduces the attrition rate of drug candidates, which has historically plagued the pharmaceutical industry and contributed to lengthy and costly development timelines. In addition to identifying and screening potential drug candidates, AI can also optimize lead compounds through iterative design processes. By employing techniques such as generative adversarial networks (GANs), researchers can generate new molecular structures that fit desired properties. These AI-guided approaches can lead to the discovery of innovative compounds that might not have been considered using traditional methodologies. Furthermore, AI can aid in understanding drug metabolism and toxicity, anticipating adverse effects before clinical trials begin, thereby enhancing the overall safety profile of potential drugs. The integration of AI into pharmaceutical research is not without challenges. Issues such as data variability, the need for high-quality datasets, and the complexity of biological systems can hinder the effectiveness of AI models. However, ongoing advancements in AI technologies, coupled with collaborative efforts between researchers, clinicians, and data scientists, are paving the way for the continued elevation of AI's role in drug discovery. In conclusion, AI is transforming the landscape of pharmaceutical research by streamlining the identification of therapeutic targets, enabling efficient screening of drug candidates, and facilitating the design of innovative therapeutic agents. By optimizing these processes, AI holds the promise of accelerating the pace of drug discovery and improving therapeutic outcomes in various diseases, ultimately contributing to more effective healthcare solutions.AI models are increasingly pivotal in linking the chemical structure of compounds to their biological activity, a critical step in the drug discovery process. By employing machine learning algorithms and deep learning techniques, these models can analyze vast datasets that encompass diverse chemical entities and their corresponding biological effects. This capability enables researchers to understand the relationships between molecular features and therapeutic potentials, leading to the identification of structural characteristics that contribute to high potency, selectivity, and optimal pharmacokinetic profiles. Once these links are established, researchers can utilize AI to optimize lead compounds. For example, AI can suggest modifications to existing molecules to enhance their activity against specific biological targets while minimizing off-target effects. This rational design approach allows for the creation of drug candidates tailored to meet specific criteria, such as improved absorption, distribution, metabolism, and excretion (ADME) properties, which are critical for ensuring that drugs perform effectively in clinical settings. Moreover, by predicting how structural changes impact biological activity, AI can significantly reduce the time and costs associated with trial-and-error methods in drug design. The ability to rapidly iterate and refine compounds based on data-driven insights empowers researchers to expand their creativity and innovation in developing new therapeutics. Ultimately, AI-driven optimization of drug candidates promises to enhance the efficacy and safety profiles of new medications, accelerating their journey from the lab bench to the clinic.AI algorithms, particularly those utilizing reinforcement learning and generative models, are revolutionizing the discovery of novel drug-like chemical structures. By leveraging extensive chemical libraries and experimental data, these algorithms can explore and expand the chemical space more efficiently than traditional methods. Reinforcement learning allows the AI to navigate this vast space by optimizing for specific objectives, such as maximizing biological activity or minimizing toxicity, through iterative feedback loops. Generative models, such as variational autoencoders and generative adversarial networks, can synthesize new chemical structures while adhering to predefined pharmacological properties. These models learn to recognize patterns and relationships in existing compounds, enabling them to create innovative drug candidates that may not have been previously considered.  The integration of these AI techniques can significantly accelerate the drug discovery process by producing diverse, actionable design proposals, thereby enhancing the chances of identifying effective therapeutics. This capability not only streamlines R&D but also encourages creativity in drug design, ultimately leading to the development of more effective and targeted medications customized for specific diseases. As a result, AI's role in expanding the chemical space holds immense potential for transforming pharmaceutical innovation and improving patient outcomes. AI algorithms have become indispensable in the analysis and optimization of drug candidates, significantly improving the drug development process by taking into account critical factors such as efficacy, safety, and pharmacokinetics. By mining large datasets that include previous experimental results and clinical trial data, these algorithms can uncover complex relationships between a drug's chemical properties and its biological effects.  Through advanced modeling techniques, AI can predict how modifications to a molecule might impact its therapeutic potential. For instance, machine learning techniques can model dose-response relationships, helping researchers identify optimal dosage levels that maximize efficacy while minimizing adverse side effects. Additionally, AI algorithms can assess the pharmacokinetic profiles of drug candidates, analyzing parameters such as absorption, distribution, metabolism, and excretion (ADME) to ensure that a compound has the desired bioavailability and half-life.Furthermore, AI can facilitate virtual screening processes to prioritize candidates with the best potential profiles, thereby reducing the time and cost associated with experimental testing. By simulating how different compounds behave in biological systems, researchers can focus their efforts on the most promising candidates. Ultimately, the use of AI in drug candidate optimization leads to a streamlined and more effective development pipeline, enabling the design of therapeutic molecules that are not only highly effective but also carry lower risks in terms of safety, which is essential for successful clinical outcomes.According to a U.S. News survey conducted among 150 professionals, pharmacists rank as the 13th best-paid professionals, with an average salary of approximately $120,950. This attractive compensation reflects the level of expertise, responsibility, and the critical role pharmacists play in the healthcare system. Moreover, the profession boasts a relatively low unemployment rate of just 1.6%, indicating a strong demand for skilled pharmacists in various settings, from community pharmacies to hospitals and research institutions. The core function of a pharmacist revolves around ensuring the safe and effective use of medications. With the healthcare landscape continually evolving, pharmacists have adapted to take on more significant responsibilities beyond merely dispensing medications. Their expertise is crucial in interpreting medication prescriptions, ensuring the correct drugs are dispensed in the proper amounts, and providing patients with essential counseling on how to use their medications effectively. One of the vital aspects of a pharmacist's role is to prevent adverse drug-drug interactions, especially for patients taking multiple medications. Given that pharmaceutical treatments often require a combination of various drugs, pharmacists meticulously analyze each patient’s medication profile to identify potential interactions that could compromise safety or treatment efficacy. This process helps in averting negative outcomes and is particularly important for elderly patients or individuals with complex health conditions who are often prescribed multiple medications. Pharmacists also serve as a vital link between patients and healthcare providers. They collaborate closely with physicians and other healthcare professionals to ensure a holistic approach to patient care. This collaboration helps optimize therapeutic outcomes, and pharmacists are often consulted for their medication expertise, contributing valuable insights about drug selection, dosing adjustments, and alternative therapies. This collaborative environment enhances patient care by leveraging the unique skills of pharmacists alongside those of other healthcare professionals. Education typically plays a significant role in the preparation of pharmacists for these responsibilities. Most pharmacists hold a Doctor of Pharmacy (PharmD) degree, which typically involves four years of professional study after at least two years of undergraduate education. The coursework includes pharmacology, medicinal chemistry, and clinical pharmacy, along with practical experiences in various healthcare settings. Continuous education is also essential for pharmacists to stay updated with the latest medication therapies, advancements in pharmaceuticals, and changes in regulations, ensuring they provide accurate and up-to-date information to their patients. Moreover, the advent of technology has revolutionized the pharmacy profession, allowing pharmacists to utilize software systems that enhance their ability to track prescriptions, alert them to potential issues, and manage inventory efficiently. This integration of technology not only streamlines workflows but also maximizes patient safety, as digital systems often include databases that flag potential drug interactions automatically.The role of pharmacists has evolved over time, and they are increasingly recognized as integral members of healthcare teams. They play a significant part in preventative care through immunization programs and have been crucial during public health crises, such as the COVID-19 pandemic, where they provided vaccines to millions of individuals. Additionally, pharmacists are often at the forefront of health education, helping patients understand their conditions and the importance of adhering to prescribed medication regimens. Despite the rewarding aspects of the profession, pharmacists often face challenges, including workload pressures and the need to manage complex health conditions in patients. The aforementioned low unemployment rate reflects a robust job market that demands highly qualified professionals, but it also suggests that pharmacists must continually adapt to meet evolving healthcare needs. In summary, the pharmacist profession is characterized by its competitive salary, low unemployment rates, and an essential role within the healthcare system. Pharmacists ensure the safe use of medications while mitigating risks through vigilant monitoring of drug interactions. Their extensive training, ability to communicate effectively with patients and other healthcare providers, and commitment to ongoing education all contribute to their invaluable position in promoting public health and patient safety. As healthcare continues to evolve, pharmacists will undoubtedly take on even more significant roles in improving health outcomes and providing quality care to a diverse patient population.The landscape of the pharmaceutical profession has undergone significant transformation over the past five years, primarily driven by advancements in big data, artificial intelligence (AI), and robotics. As healthcare increasingly embraces these technologies, robots are gaining trust from medical professionals, allowing them to assist in various tasks traditionally performed by pharmacists and other healthcare workers. Automation, enabled by AI algorithms, can handle tasks ranging from medication dispensing to inventory management with heightened accuracy and efficiency, thereby enhancing operational workflows in pharmacies and hospitals. Incorporating robotics into pharmacy operations offers several advantages: robots can work tirelessly, reducing the risk of human error in medication dispensing, thereby increasing patient safety. Additionally, these automated systems can efficiently manage high volumes of prescriptions, particularly in busy environments, significantly reducing wait times for patients. As institutions begin to lean more on robotic systems, pharmacists can reallocate their time to more complex responsibilities, such as patient counseling and medication therapy management, which require human empathy and clinical judgement. Moreover, AI-powered analytics can sift through vast datasets to identify patterns, such as optimal drug combinations or emerging drug interactions, enabling pharmacists to make more informed decisions. This synergy of human expertise with technological capabilities creates a hybrid model of care that enhances the overall effectiveness of pharmaceutical services. Yet, while automation offers numerous benefits, it also raises important questions about the future role of pharmacists in a technology-driven environment. As certain tasks become automated, pharmacists will need to shift their focus towards areas where human interaction and clinical assessment remain irreplaceable, making the profession more centered on strategic care, patient education, and personalized medicine. This evolution underscores the importance of adaptability in the profession, highlighting that pharmacists will continue to play a crucial role in the healthcare continuum, albeit in a transformed capacity empowered by technology. Klopman’s introduction of a pioneering computer-automated program for the analysis of structure-activity relationships (SAR) marks a significant advancement in the field of computational chemistry and toxicology. This innovative program leverages the KLN code, a linear coding routine that facilitates the identification and evaluation of organic molecular structures. The core objective of SAR studies is to elucidate the relationship between the chemical structure of a molecule and its biological activity, a pursuit that is critical for pharmaceutical development, environmental chemistry, and the assessment of chemicals' safety. The KLN code plays a fundamental role in the automation of structure recognition, which vastly improves the efficiency of molecular analysis. By employing this linear coding system, the program can discern the features and configurations of complex organic molecules with remarkable precision. This capability not only streamlines the identification of molecular structures but also sets the stage for a deeper understanding of the underlying biophysical interactions that determine a compound’s biological activity.One of the standout features of Klopman’s program is its ability to identify, tabulate, and statistically analyze biophores, which are the essential substructures within a molecule that directly contribute to its biological effects. The biological activity of a compound often hinges on these specific functional groups or structural motifs, making the identification of biophores crucial for predicting the behavior of new compounds. By systematically cataloging these biophores, researchers can develop predictive models that inform the design of new molecules with desired biological properties, thereby accelerating the drug discovery process and enhancing the efficacy of environmental assessments. The application of Klopman’s method to study the carcinogenicity of various compounds showcases the practical utility of the program. Polycyclic aromatic hydrocarbons (PAHs) are complex organic molecules that have been extensively studied due to their presence in fossil fuel combustion products and their recognized potential as carcinogens. Using the SAR program, researchers were able to analyze the structural features of different PAHs and correlate these features with their observed carcinogenic effects. Through the statistical analysis of biophores, insights were gained regarding the specific components of PAHs that contribute to their toxicity, potentially guiding future regulatory measures and public health initiatives. In another compelling application, Klopman’s method was employed to investigate the activity of ketoxime carbamate pesticides. These pesticides are widely used in agriculture, but their safety and environmental impact remain a concern. The SAR program enabled a comprehensive analysis of the molecular structures of various ketoxime carbamates, allowing researchers to determine which structural elements were associated with effective pest control as well as toxicological outcomes. By identifying the biophores responsible for both the beneficial and harmful effects of these pesticides, the program paves the way for the development of safer alternatives that maintain efficacy while minimizing risks to human health and the ecosystem. Additionally, the program’s efficacy was demonstrated in studying the carcinogenicity of N-nitrosamines in rats. N-nitrosamines are a group of potent carcinogens resulting from the reaction of amines with nitrosating agents. Understanding their molecular structure and activity relationship can provide vital information for assessing their risks. Klopman’s structure-activity program facilitated the identification of key structural features of various N-nitrosamines, allowing researchers to correlate these features with their carcinogenic potential. This analysis is crucial in toxicology, as it helps regulatory agencies and researchers prioritize which compounds require further study or risk assessment based on their structural characteristics. The implications of Klopman’s program extend far beyond the immediate case studies. As scientists aim to develop a more predictive understanding of chemical behavior, the analytical capabilities imbued within this program contribute to a growing body of computational methods that facilitate research across diverse fields, including medicinal chemistry, environmental science, and regulatory toxicology. Furthermore, the evolution of computational techniques represented by Klopman’s work highlights a broader trend in science towards data-driven research, where large datasets and quantitative analyses provide insights that were once impossible to obtain through traditional experimental approaches alone. The automation of structure evaluation not only accelerates the pace of research but also reduces costs and enhances reproducibility, enabling scientists to focus on interpreting results and devising novel solutions. Despite its many strengths, the integration of automated programs like Klopman’s into scientific research also calls for careful consideration of the limitations of computational methods. While the program offers powerful tools for SAR analysis, the inherent complexity of biological systems necessitates a balanced approach that also includes experimental validation. Combining computational predictions with laboratory experiments can provide a more comprehensive understanding of how molecular structures translate into biological activity, ultimately enhancing the reliability of predictions made through SAR analyses. In conclusion, Klopman’s automated program for studying structure-activity relationships represents a transformative development in the analysis of organic molecules. Through the KLN coding system, the program facilitates the identification and characterization of biophores, allowing for in-depth statistical analyses that enhance our understanding of molecular biology and toxicology. Its successful applications in evaluating the carcinogenicity of various compounds underscore its potential to inform chemical safety assessments and drug discovery efforts. Moving forward, the continued integration of computational methods with experimental research will be key to unlocking new insights in the fields of chemistry and toxicology, guiding the development of safer pharmaceuticals and chemicals for public health and environmental protection.

NDDS ( Noval Darug Delivary System ) :

The method by which a drug isdelivered can have a significant effect on its efficacy. Some drugs have an optimum concentration range within which maximum benefit is derived, and concentrations above or below this range can be toxic or produce no therapeutic benefit at all 1. On the other hand, the very slow progress in the efficacy of the treatment of severe diseases, has suggested a growing need for a multidisciplinary approach to the delivery of therapeutics to targets in tissues. From this, new ideas on controlling the pharmacokinetics, pharmacodynamics, non-specific toxicity, immunogenicity, biorecognition, and efficacy of drugs were generated. These new strategies, often called drug delivery systems (DDS), which are based on interdisciplinary approaches that combine polymer science, pharmaceutics, bioconjugate chemistry, and molecular biology. To minimize drug degradation and loss, to prevent harmful side-effects and to increase drug bioavailability and the fraction of the drug accumulated in the required zone, various drug delivery and drug targeting systems are currently under development Controlled and Novel Drug Delivery which was only a dream or at best a possibility is now a reality. During the last decade and half pharmaceutical and other scientists have carried out extensive and intensive investigations in this field of drug research. Among drug carriers one can name soluble polymers, microparticles made of insoluble or biodegradable, natural and synthetic polymers, microcapsules, cells, cell ghosts, lipoproteins, liposomes, and micelles. The carriers can be made slowly degradable, stimuli-reactive (e.g., pH- or temperature-sensitive), and even targeted (e.g., by conjugating them with specific antibodies against certain characteristic components of the area of interest). Targeting is the ability to direct the drug-loaded system to the site of interest. Two major mechanisms can be distinguished for addressing the desired sites for drug release.Active targeting is an advanced strategy used to ensure selectivity and specificity of SMART Nanosystem to the targeted site/organ/cells. Commonly, targeting of antitumor drugs to the cancerous tissue has become the main strategy to reduce the effects the drugs might have on healthy tissues. The technique of active targeting should be considered during preparation of the SMART noncolloidal system via functionalization of the surface of the carrier with ligands, which specially bind with its corresponding receptor on the surface of the targeted cell. Ligand receptor attachment can guarantee that the Nano system will be optimally delivered to the dis- eased cells rather than the surrounding healthy tissue . The attached ligand on the surface of the Nano system can be classified as several subtypes including antibodies or parts of their fragments, nucleic acids (aptamers), and various classes of peptides. These ligands bind with their specific receptors that are densely localized on the surface of tumour cells. This approach also ensures higher cellular uptake through the endocentric pathway. The affinity of binding between the ligand and the receptor which is overexpressed on the surface of the targeted cell is the most important factor affecting the delivery of the drug. After ligand—receptor interaction, two possible mechanisms might occur, the Nano system might start to release part of its encapsulated drug in the close proximity of the tar- get cells and act as sustained release drug reservoir or the intact Nano system is engulfed via endocytosis and the release begins inside the cell. The second mechanism is desirable to ensure efficient delivery of drug inside the cells. Recently, SMART NPs have been widely used as a Nano carrier drug delivery system for cancer therapy. The surface of SMART NPs is functionalized with specific ligands for active targeting These systems take advantage of the fact that tumorous cells

3D Printing:

3D printing, or additive manufacturing, has revolutionized manufacturing technology by significantly reducing product development cycles and enabling rapid prototyping. Originally devised to expedite the design process, these machines have found diverse applications across various fields, including engineering, medicine, design, science, education, market research, and art, showcasing their versatility and transformative impact on innovation and production.3D printing relies on complementary technologies such as Computer Aided Design (CAD), Computer Aided Manufacturing (CAM), and Computer Numerical Control (CNC) to create three-dimensional objects through a layered approach, starting from a digital file. Invented by Charles W. Hull in 1983, the initial process, known as stereolithography, utilized UV rays to cure photopolymers. The first step involves designing the product in CAD software or scanning an existing object to create a 3D digital model, which is then sliced into numerous horizontal layers for the printing process.3D printing commonly utilizes fine plastic or metal powder, with particle sizes as small as 20 micrometers, stored in cartridges or beds and spread in thin layers using a roller or blade. An array of nozzles then applies a binding agent according to the design specified in CAD, after which a new layer of powder is added, and the process is repeated with the build platform lowered accordingly. Upon completion, the finished part is removed from the surrounding unconsolidated powder, then cleaned and potentially finished. This technology excels at producing highly complex and customizable products, highlighting its versatility in manufacturing.

Types Of 3d Printing:

A 3D printer is a type of industrial robot. It can be classified on the basis of form of materials used to make model. The form of material can be solid, liquid and powder (1). The classification is given below:

  1. Stereolithography (SL)
  2. Fused Deposition Modelling (FDM)
  3. PolyJet or Multi jet Printing
  4. Selective Laser Sintering (SLS)
  1. Stereolithography (SL)

SL was developed by 3D system, Inc. It is widely used process of rapid prototyping. In this process, firstly, STL file of the part/object is created in CAD software. The STL file contains details of each layer. Then the layering of liquid photopolymer is done on support material in the machine. Support material is required for the cavity or overhung parts in the object. The machine automatically generates support structure for the model under the construction. After layering, curing process is done with UV laser at specific locations. During curing process, solidification of polymer takes place. After that the platform is lowered by one layer thickness. The process of layering and curing continue till the part/ object is completed. Then, the object is taken out and the support material is removed manually. The object is then finished. Here UV light acts as catalyst for polymerization. SL process is selected when it is required to make visual prototypes for photoshoot, show fine details, market testing, checking 3D drawings, low volume production of complex geometries etc.

  1. Fused Deposition Modelling (FDM):

The technology was developed by S. Scott Crump in the late 1980s and was commercialized in 1990 by a company named Stratasys. In this process the object is built layer –by – layer from the bottom up by heating and extruding thermoplastic filament. The first step is to create 2D slice of the object with the help of AutoCad or any design software. The software also determines path to extrude the thermoplastic material and support material if needed. Then the material is fed into the temperature controlled extruder head, where it is converted into semi liquid state. The thermoplastic material is heated just beyond its Glass Transition temperature (Tg). The head extrudes fine filament of thickness in the range 0.254 mm and deposits in layers on a fixtureless base. Where a support or buffering is needed, the 3D printer deposits a removable material that acts as scaffolding. When layer is finished, it moves in the Z direction to the next layer. The layering process is done vey precisely and bonded and solidified. Once the process is complete, the support material is removed and the object is finished. Mainly polymeric materials such as Acrylonitrile Butadiene Styrene, Polylactic acid, Polycarbonate, Polyamide, Polystyrene etc. are used in FDM 3D printers. This technology is widely used in various industries such as aerospace, automotive, medical etc. apart from that it attracts a large number of professionals such as engineers, designers, educators and they can make any prototype or product for their requirements.

  1. PolyJet or Multi jet Printing:

In this technology, two or more ink jets are used for spraying liquid polymers. For making object, photopolymer is used which is cured instantly in presence of UV light. Another jet can be used for support spraying support material, which is not cured by UV light. Support material is used for overhung parts. It is then removed after completion of the layering process by hand and water jetting. PlyJet 3D printing machine is used to create objects or parts which have very high details. The thickness of the horizontal layers can be as low as 16 micron and ultra-thin walls down to 0.6mm (0.024") depending on geometry. The limitation of this process is the objects or parts produced are weaker compared to the other process such as stereolithography and selective laser sintering. Here it is also possible to produce multi colored objects (1, 4, 5, 6). Companies have developed their own wide range of different materials in terms of properties to suit various needs. Materialse, a popular company provides 3D printing Solutions, offers two primary materials. They are VeroWhite Plus and TangoWhite Plus. VeroWhite Plus is a general purpose resin, available in white color and has very good mechanical properties. TangoWhite Plus is flexible rubber like resin, which has exceptional elongation at break and can be used as rubber like products. This company also offers a number of composite materials (5). Another company named Stratasys, also offers a vast array of materials which can work on Polyjet 3D printing machines. They vary in properties and colors. Stratasys also offers two Polyjet copolymers which simulate the functionality and appearance of Polypropylene.

  1. Selective Laser Sintering (SLS)

SLS was developed and patented by Dr. Carl Deckard and academic adviser, Dr. Joe Beaman at the University of Texas at Austin in the mid-1980s. In this technology, the basic material is powdered form of metal, combinations of metals, polymer, combinations of polymers, combinations of metals and polymers, combinations of metals and ceramics are used. The particle sizes are in the order of 50 µm. The carbon di oxide laser beam fuses metal or polymeric power at specific locations as per the design. After completion of every layer, the powder bed is lowered by one layer thickness and a new layer of the material is applied on the top. The process is repeated until the part or object is completed. The process is carried out in an inert atmosphere to avoid oxidation. For metals, binders are used. The binder is removed after the process. (1, 3). Unlike FDM, SLS does not require any support material as the part which is being constructed, always remains surrounded by unsintered powder. A wide range of materials can be processed on SLS. These include polymers such as nylon (neat, glass-filled, or with other fillers) or polystyrene, metals including steel, titanium, alloy mixtures, and composites and green sand. The physical process can be full melting, partial melting, or liquid-phase sintering. Depending on the material, up to 100?nsity can be achieved with material properties comparable to those from conventional manufacturing methods (13). Other techniques of additive manufacturing are also available. These are 3DP, Prometal, Electron beam melting (EBM), Laminated Object Manufacturing LOM), Laser Engineering Net Shaping

BENEFITS:

Technology plays a crucial role in enhancing the pharmaceutical industry across multiple dimensions, including providing better healthcare solutions, improving regulatory compliance, and boosting operational and financial performance. By streamlining processes, technology helps reduce costs while ensuring that healthcare services remain effective and accessible. Additionally, it contributes to improved patient outcomes by facilitating personalized treatment and enhancing the overall quality of care. Technology significantly enhances the pharmaceutical industry by improving confidentiality standards, enabling easier sharing of information, and facilitating real-time decision-making for healthcare professionals. Additionally, advancements such as RFID technology help reduce the threat of counterfeit drugs, ensuring that patients receive safe and authentic medications. Furthermore, the use of artificial intelligence in clinical trials enhances efficiency and safety, ultimately contributing to more reliable research outcomes. Personalized medicine, also known as precision medicine, enables the tailoring of treatments to align with an individual's unique response to medications, enhancing therapeutic effectiveness. Meanwhile, automation plays a pivotal role in digitally transforming the pharmaceutical supply chain, improving efficiency and accuracy in the delivery of products. Additionally, reliable packaging machines ensure that pharmaceutical products are packaged safely and efficiently, further contributing to the overall quality and safety of medications available to patients.

LIMITATIONS:

Pharmaceutical technology faces several challenges that can impede treatment efficacy, including inadequate drug concentrations reaching the target site, resulting in suboptimal therapeutic effects. Additionally, nonspecific drug distribution can lead to off-target effects and increased side effects, compromising patient safety. Furthermore, the development of acquired resistance during chemotherapy poses a significant hurdle, as cancer cells may adapt to various drugs and dosages, decreasing treatment effectiveness over time. These limitations highlight the need for continued advancements in pharmaceutical technology to address these critical issues.

  • Technical Limitations:

Pharmaceutical technology faces significant limitations, including scalability issues, where scaling up production processes can compromise product quality. Stability is another concern, particularly for sensitive formulations that require specific conditions to remain effective. Maintaining sterility throughout manufacturing processes is crucial to ensure patient safety, yet it poses considerable challenges. Additionally, ensuring material compatibility between drugs and packaging is essential to prevent interactions that could alter efficacy or safety. Finally, analytical challenges arise in accurately analyzing complex formulations, which can hinder development and quality control efforts. Addressing these limitations is critical for the advancement of effective pharmaceutical solutions.

  • Regulatory Limitations:

Pharmaceutical technology faces several limitations, including complex regulatory frameworks that delay product approvals, stringent quality control standards necessitating extensive testing, challenges in protecting intellectual property, and difficulties in conducting clinical trials due to high costs and recruitment issues. These factors hinder the timely development and evaluation of new therapies, emphasizing the need for improvements in the pharmaceutical development process.

  • Economic Limitations:

The pharmaceutical industry is challenged by high development costs that encompass research, development, and manufacturing, making it financially burdensome to bring new medications to market. This is exacerbated by the limited accessibility of high-priced medications, which many patients cannot afford. Additionally, securing reimbursement from payers poses significant hurdles, further complicating the financial viability of new drugs. Finally, the threat of generic competition diminishes profitability for brand-name products, underscoring the need for strategies to navigate these obstacles effectively.

  • Ethical Limitations:

The pharmaceutical industry faces significant ethical and safety challenges, including the need to balance innovation with patient safety. There are ongoing debates regarding the necessity of animal testing, alongside concerns about protecting patient data during clinical trials to maintain privacy and trust. Additionally, managing conflicts of interest between the industry and academia is critical to preserve research integrity and ensure unbiased results. These factors reflect the complexities of ethics in pharmaceutical development.

  • Other Limitations:

The pharmaceutical industry is grappling with critical challenges in waste management, aiming to handle hazardous waste from manufacturing while minimizing environmental impact and developing sustainable practices amid resource constraints such as water and energy. Social issues like disparities in access to healthcare, medication affordability, cultural sensitivity in packaging, and reliance on adequate healthcare infrastructure further complicate the landscape. Future directions involve addressing these technical challenges through innovative solutions, streamlining regulatory processes, enhancing accessibility and affordability, embracing sustainability, and fostering collaboration among industry, academia, and regulatory bodies to ensure a responsible and equitable healthcare approach.

REFERENCES

  1. A Bodhani, ‘From computer to catwalk', Engineering & Technology, vol.
  2. A. Kapczynski, “How to vaccinate the world, part 1,” LPE Project Blog (April 30, 2021). Available at https://lpeproject.org/blog/how-to-vaccinate-the-world-part-1/.
  3. Alam SR, Shah AS, Richards J, et  al. Ultrasmall superparamagnetic particles of iron oxide in patients with acute myocardial infarction: early clinical experience. Circ Cardiovasc Imaging. 2012;5:559–65.
  4. Almutairi A, Rossin R, Shokeen M, et al. Biodegradable dendritic positron-emitting nanoprobes for the noninvasive imaging of angiogenesis. Proc Natl Acad Sci U S A. 2009;106:685–90.
  5. Avci O, Ünlü NL, Özkumur AY, Ünlü MS. Interferometric reflectance imaging sensor (IRIS) – a platform technology for multiplexed diagnostics and digital detection. Sensors (Basel). 2015;15:17649–65.
  6. Bernstein E, Caudy AA, Hammond SM, Hannon GJ. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001;409:363-6
  7. Biggest Challenges Facing the Pharmaceutical Industry in 2023. Available online: https://www.pssindia.com/2023/01/23/bigg est-challenges-facing-the-pharmaceutical-industry-in-2023/ (accessed on 5 May 2023).
  8. Breitbart. Your Pharmacist will Soon be a Robot. Available from: http://www.breitbart.com/california/2016/05/02/ pharmacist-will-soon-app-robot/. [Last accessed on 2017 Jun 24].
  9. C. C. Mu¨ller-Goymann, Physicochemical characterization of colloidal drug delivery sys- tems such as reverse micelles, vesicles, liquid crystals and nanoparticles for topical administration, Eur. J. Pharm. Biopharm. 58 (2004) 343—356. Available from: https:// doi. org/10. 1016/j. ejpb. 2004. 03. 028.
  10. C. Chaudhuri, “Making Covid-19 medical products affordable: Voluntary patent pool and TRIPS flexibilities,” South Centre Views 200 (June 2020).
  11. C. Pathak, F. U. Vaidya, S. M. Pandey, Mechanism for development of nanobased drug delivery system, Appl. Target. Nano Drugs Deliv. Syst. , Elsevier Inc, 2019, pp. 35—67. Available from: http://doi. org/10. 1016/b978-0-12-814029-1. 00003-x.
  12. Cellan-Jones R. Stephen Hawking Warns Artificial Intelligence could End Mankind. Available from: http:// www.bbc.com/news/technology-30290540. [Last accessed on 2017 Jun 24].
  13. Chavda, V.; Valu, D.; Parikh, P.; Tiwari, N.; Chhipa, A.; Shukla, S.; Patel, S.; Balar, P.; Paiva-Santos, A.; Patravale, V. Conventional and Novel Diagnostic Tools for the Diagnosis of Emerging SARS-CoV-2 Variants. Vaccines 2023, 11, 374. [CrossRef].
  14. Chavda, V.P.; Vihol, D.; Patel, A.; Redwan, E.M.; Uversky, V.N. Introduction to Bioinformatics, AI, and ML for Pharmaceuticals. In Bioinformatics Tools for Pharmaceutical Drug Product Development; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2023; pp. 1–18.
  15. Colombo, S.; Beck-Broichsitter, M.; Bøtker, J.P.; Malmsten, M.; Rantanen, J.; Bohr, A. Transforming Nanomedicine Manufacturing toward Quality by Design and Microfluidics. Adv. Drug Deliv. Rev. 2018, 128, 115–131. [CrossRef].
  16. Colombo, S.; Zeng, X.; Ragelle, H.; Foged, C. Complexity in the Therapeutic Delivery of RNAi Medicines: An Analytical Challenge. Expert Opin. Drug Deliv. 2014, 11, 1481–1495. [CrossRef].
  17. Daka, A.; Peer, D. RNAi-Based Nanomedicines for Targeted Personalized Therapy. Adv. Drug Deliv. Rev. 2012, 64, 1508–1521. [CrossRef].
  18. Dasta JF. Application of artificial intelligence to pharmacy and medicine. Hosp Pharm 1992;27:312-5, 319-22.
  19. Dickherber, A.; Morris, S.A.; Grodzinski, P. NCI Investment in Nanotechnology: Achievements and Challenges for the Future. Wiley Interdiscip. Rev. Nanomed. Nanobio Technol. 2015, 7, 251–265. [CrossRef] [PubMed]
  20. E. Torreele, M. Kazatchkine, and M. Mazzucato, “Preparing for the next pandemic requires public health focused industrial policy,” BMJ Blog (April 1, 2021). Available at https:// blogs.bmj.com/bmj/2021/04/01/preparing-for-the-next-pandemic-requires-public-health-focused-industrial-policy.
  21. E. Torreele, M. Mazzucato, and H. Lishi Li, “Delivering the people’s vaccine: Challenges and proposals for the biopharmaceutical innovation system,” UCL Institute for Innovation and Public Purpose, Policy Brief 12. Available at https://www.ucl.ac.uk/bartlett/public-purpose/ sites/public-purpose/files/iipp-pb12_delivering-the-peoples-vaccine_final.pdf; G. Krikorian, “Refusing state funding of vaccine research in a corporate ‘double-blind’,” Remix the Commons Blog (2021). Available at https://www. remixthecommons.org/?p=5022.
  22. Flasi?ski M. Introduction to Artificial Intelligence. 1st ed. Switzerland: Springer International Publishing; 2016. p. 4.
  23. Gaëlle Krikorian And Els Torreele, We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis, JUNE 2021 VOLUME 23 NUMBER 1.
  24. gaëlle krikorian and els torreele, We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis, JUNE 2021 VOLUME 23 NUMBER 1.
  25. Global Health Centre, Graduate Institute Geneva. Covid-19 vaccine R&D investment. Available at https://www. knowledgeportalia.org/covid19-r-d-funding.
  26. Grilo, A.L.; Mantalaris, A. The Increasingly Human and Profitable Monoclonal Antibody Market. Trends Biotechnol. 2019, 37, 9–16. [CrossRef] [PubMed].
  27. H. Schellekens, M. Aldosari, H. Talsma, and E. Mastrobattista, “Making individualized drugs a reality,” Nature Biotechnology 35 (2017), pp. 507–513.
  28. Hammond SM. Dicing and slicing: the core machinery of the RNA interference pathway. FEBS Lett 2005;579:5822-9
  29. Hariom Rajput, Saurabh raj Pandey, A Review Article on Novel Drug Delivery System, Volume 10 ~ Issue 4 (2024) pp: 21-31 ISSN(Online) : 2347-2995.
  30. http://manufacturing.materialise.com/cases/wearable-tech-just-got-smarter-anouk-wipprechtsintel-edison-powered-3d-printed-synapse-dress viewed on 1 June 2015
  31. Jain, K.K.; Jain, K.K. Nanomolecular Diagnostics. In The Handbook of Nanomedicine; Springer: Berlin/Heidelberg, Germany, 2017; pp. 133–200. [CrossRef].
  32. K. Bajwa, B. Qorri, A. Decarlo, M. R. Szewczuk, Recent advances in “smart” delivery systems for extended drug release in cancer therapy, Int. J. Nanomed. 13 (2018) 4727—4745.
  33. K. Basu, L. Gostin, and N. Hassoun, “Pandemic preparedness and response: Beyond the WHO’s Access to COVID-19 Tools Accelerator,” Brookings Global Economy and Development Working Paper (2021). Available at https:// www.brookings.edu/wp-content/uploads/2021/04/Pandemic-preparedness-and-response_final.pdf. 31. A. M. Büyüm, C. Kenney, A. Koris, et al., “Decolonising global health: If not now, when?,” BMJ Global Health 5 (2020).
  34. K. Kupferschmidt, “‘Vaccine nationalism’ threatens global plan to distribute COVID-19 shots fairly,” Science Magazine (July 28, 2020). Available at https://www. sciencemag.org/news/2020/07/vaccine-nationalism-threatens-global-plan-distribute-covid-19-shots-fairly.
  35. K.K. Jain, The Handbook of Nanomedicine, DOI 10.1007/978-1-4939-6966-1_4.
  36. Kalepu, S.; Nekkanti, V. Insoluble Drug Delivery Strategies: Review of Recent Advances and Business Prospects. Acta Pharm. Sin. B 2015, 5, 442–453. [CrossRef].
  37. Kgmg. Available online: https://kpmg.com/xx/en/home/insights/2022/12/the-supply-chain-trends-shaking-up-2023.html (accessed on 2 May 2023).
  38. Kovacs, M. TECHNOLOGY: 3D printing gears up for fashion industry change. Aroq - JustStyle.Com (Global News), 1 (2015)
  39. Krikorian, G.; Torreele, E. We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis. Health Hum. Rights 2021, 23, 119–127.
  40. Lau JR, Geho WB, Snedekar GH, Inventors; SDG INC, An Ohio Corporation, Assignee; Targeted Liposomal Drug Delivery System. US Patent 20100209492. 2010 Aug 19.
  41. Lau JR,Geho WB, Snedekar GH, Inventors; Targeted Liposomal Drug Delivery System. US Patent 20070104777. 2007 May 10.
  42. Mahesh Shaw, 3D Printing Technology: Its application and scope in Fashion Industry, 27 August 2018.
  43. Mak, K.-K.; Pichika, M.R. Artificial Intelligence in Drug Development: Present Status and Future Prospects. Drug Discov. Today 2019, 24, 773–780. [CrossRef].
  44. Manish Vyas, Sourav Thakur, Bushra Riyaz, Kuldeep K Bansal, Bhupendra Tomar, Vijay Mishra, Artificial Intelligence: The Beginning of a New Era in Pharmacy Profession, Received: 28-07-2017 Revised: 12-04-2018 Accepted: 27-04-2018.
  45. Müller, R. Junghanns Nanocrystal Technology, Drug Delivery and Clinical Applications. Int. J. Nanomed. 2008, 3, 295–310. [CrossRef] [PubMed].
  46. Munos, B. Lessons from 60 Years of Pharmaceutical Innovation. Nat. Rev. Drug Discov. 2009, 8, 959–968. [CrossRef].
  47. National Nanotechnology Initiative. NNI Budget Supplements and Strategic Plans. Available online: https://www.nano.gov/N NIBudgetSupplementsandStrategicPlans (accessed on 1 May 2023).
  48. no. 12, (2015) p. 68. 9. L Adendorff, '3D technology: 3D printing the next frontier?’ Aroq - Just-Style.Com (Global News), (2014) p. 12, Entrepreneurial Studies Source, EBSCOhost, viewed 1 June 2015.)
  49.  Parker JS, Roe SM, Barford D. Structural insights into mRNA recognition from a PIWI domain-siRNA guide complex. Nature 2005;434:663-6 16. Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev 2001;15:188-200.
  50. R. R. Bhagwat, I. S. Vaidhya, Novel drug delivery systems: an overview, Int. J. Pharm. Sci. Res. 4 (2013) 970—982.
  51. R.R. Bhagwat and I.S. Vaidhya, Novel Drug Delivery Systems: An Overview, IJPSR (2013), Vol. 4, Issue 3.
  52. Sarpatwari, A.; Barenie, R.; Curfman, G.; Darrow, J.J.; Kesselheim, A.S. The US Biosimilar Market: Stunted Growth and Possible Reforms. Clin. Pharmacol. Ther. 2019, 105, 92–100. [CrossRef].
  53. Scannell, J.W.; Blanckley, A.; Boldon, H.; Warrington, B. Diagnosing the Decline in Pharmaceutical R&D Efficiency. Nat. Rev. Drug Discov. 2012, 11, 191–200. [CrossRef] [PubMed].
  54. Schmidt FR. About the nature of RNA interference. Appl Microbiol Biotechnol 2005;67:429-35
  55. Sharma, R.; Shishodia, A.; Gunasekaran, A.; Min, H.; Munim, Z.H. The Role of Artificial Intelligence in Supply Chain Management: Mapping the Territory. Int. J. Prod. Res. 2022, 60, 7527–7550. [CrossRef].
  56. Statistica. Artificial Intelligence (AI). Available from: https://www.statista.com/study/38609/artificialintelligence-ai-statista-dossier/. [Last accessed on 2017 Jun 24].
  57. Stefano Colombo, Xianghui Zeng, He´lo?¨se Ragelle & Camilla Foged, Complexity in the therapeutic delivery of RNAi medicines: an analytical challenge, 2014 Informa UK, Ltd. ISSN 1742-5247, e-ISSN 1744-7593.
  58. Takagi A, Yamashita N, Sonobe T,Inventors; Astellas Pharma INC Tokyo, Assignee; Intracellular Drug Delivery Improving Liposomes. US Patent 20070286898. 2007 Dec 13.
  59. Times of India Pharmaceutical Supply Chain Management: Crises and Innovations. Available online: https://timesofindia.indiat imes.com/blogs/voices/pharmaceutical-supply-chain-management-crises-and-innovations/ (accessed on 15 May 2023).
  60. Troiano, G.; Nolan, J.; Parsons, D.; Van Geen Hoven, C.; Zale, S. A Quality by Design Approach to Developing and Manufacturing Polymeric Nanoparticle Drug Products. AAPS J. 2016, 18, 1354–1365. [CrossRef] [PubMed].
  61. Whitehead KA, Langer R, Anderson DG. Knocking down barriers: advances in siRNA delivery. Nat Rev Drug Discov 2009;8:129-38
  62. World Health Assembly, Improving the Transparency of Markets for Medicines, Vaccines, and Other Health Products, WHA72.8 (2019). 38. Ahmed v. Union of India, para. 1., as cited in A. Kapcyzinski, “The right to medicines in an age of neoliberalism,” Humanity Journal 10/1 (2019).
  63. Y. H. Bae, K. Park, Targeted drug delivery to tumors: myths, reality and possibility, J. Control. Release. 153 (2011) 198—205. Available from: https://doi. org/10. 1016/j. jconrel. 2011. 06. 001.
  64. Yamauchi H, Morita H, Kikuchi H, Inventors; Daiichi pharmaceuticals Co.LTD, Assignee; Liposomes And Liposomal Dispersion. US Patent 20020182248. 2002 Dec 5.
  65. Zhang H, Kolb FA, Jaskiewicz L, et al. Single processing center models for human Dicer and bacterial RNase III. Cell 2004;118:57-68
  66. Zhang Y, Luo B, Iyer L, Inventors; Liposomal Delivery Vehicle For Hydrophobic Drugs. US Patent 20070014845. 2007 Jan 18.
  67. Zignify. Available online: https://zignify.net/what-is-supply-chain-disruption-and-what-are-its-causes/#:~:text=LogisticsDe laysandFailures,resultingincongestionanddelays (accessed on 2 May 2023).

Reference

  1. A Bodhani, ‘From computer to catwalk', Engineering & Technology, vol.
  2. A. Kapczynski, “How to vaccinate the world, part 1,” LPE Project Blog (April 30, 2021). Available at https://lpeproject.org/blog/how-to-vaccinate-the-world-part-1/.
  3. Alam SR, Shah AS, Richards J, et  al. Ultrasmall superparamagnetic particles of iron oxide in patients with acute myocardial infarction: early clinical experience. Circ Cardiovasc Imaging. 2012;5:559–65.
  4. Almutairi A, Rossin R, Shokeen M, et al. Biodegradable dendritic positron-emitting nanoprobes for the noninvasive imaging of angiogenesis. Proc Natl Acad Sci U S A. 2009;106:685–90.
  5. Avci O, Ünlü NL, Özkumur AY, Ünlü MS. Interferometric reflectance imaging sensor (IRIS) – a platform technology for multiplexed diagnostics and digital detection. Sensors (Basel). 2015;15:17649–65.
  6. Bernstein E, Caudy AA, Hammond SM, Hannon GJ. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001;409:363-6
  7. Biggest Challenges Facing the Pharmaceutical Industry in 2023. Available online: https://www.pssindia.com/2023/01/23/bigg est-challenges-facing-the-pharmaceutical-industry-in-2023/ (accessed on 5 May 2023).
  8. Breitbart. Your Pharmacist will Soon be a Robot. Available from: http://www.breitbart.com/california/2016/05/02/ pharmacist-will-soon-app-robot/. [Last accessed on 2017 Jun 24].
  9. C. C. Mu¨ller-Goymann, Physicochemical characterization of colloidal drug delivery sys- tems such as reverse micelles, vesicles, liquid crystals and nanoparticles for topical administration, Eur. J. Pharm. Biopharm. 58 (2004) 343—356. Available from: https:// doi. org/10. 1016/j. ejpb. 2004. 03. 028.
  10. C. Chaudhuri, “Making Covid-19 medical products affordable: Voluntary patent pool and TRIPS flexibilities,” South Centre Views 200 (June 2020).
  11. C. Pathak, F. U. Vaidya, S. M. Pandey, Mechanism for development of nanobased drug delivery system, Appl. Target. Nano Drugs Deliv. Syst. , Elsevier Inc, 2019, pp. 35—67. Available from: http://doi. org/10. 1016/b978-0-12-814029-1. 00003-x.
  12. Cellan-Jones R. Stephen Hawking Warns Artificial Intelligence could End Mankind. Available from: http:// www.bbc.com/news/technology-30290540. [Last accessed on 2017 Jun 24].
  13. Chavda, V.; Valu, D.; Parikh, P.; Tiwari, N.; Chhipa, A.; Shukla, S.; Patel, S.; Balar, P.; Paiva-Santos, A.; Patravale, V. Conventional and Novel Diagnostic Tools for the Diagnosis of Emerging SARS-CoV-2 Variants. Vaccines 2023, 11, 374. [CrossRef].
  14. Chavda, V.P.; Vihol, D.; Patel, A.; Redwan, E.M.; Uversky, V.N. Introduction to Bioinformatics, AI, and ML for Pharmaceuticals. In Bioinformatics Tools for Pharmaceutical Drug Product Development; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2023; pp. 1–18.
  15. Colombo, S.; Beck-Broichsitter, M.; Bøtker, J.P.; Malmsten, M.; Rantanen, J.; Bohr, A. Transforming Nanomedicine Manufacturing toward Quality by Design and Microfluidics. Adv. Drug Deliv. Rev. 2018, 128, 115–131. [CrossRef].
  16. Colombo, S.; Zeng, X.; Ragelle, H.; Foged, C. Complexity in the Therapeutic Delivery of RNAi Medicines: An Analytical Challenge. Expert Opin. Drug Deliv. 2014, 11, 1481–1495. [CrossRef].
  17. Daka, A.; Peer, D. RNAi-Based Nanomedicines for Targeted Personalized Therapy. Adv. Drug Deliv. Rev. 2012, 64, 1508–1521. [CrossRef].
  18. Dasta JF. Application of artificial intelligence to pharmacy and medicine. Hosp Pharm 1992;27:312-5, 319-22.
  19. Dickherber, A.; Morris, S.A.; Grodzinski, P. NCI Investment in Nanotechnology: Achievements and Challenges for the Future. Wiley Interdiscip. Rev. Nanomed. Nanobio Technol. 2015, 7, 251–265. [CrossRef] [PubMed]
  20. E. Torreele, M. Kazatchkine, and M. Mazzucato, “Preparing for the next pandemic requires public health focused industrial policy,” BMJ Blog (April 1, 2021). Available at https:// blogs.bmj.com/bmj/2021/04/01/preparing-for-the-next-pandemic-requires-public-health-focused-industrial-policy.
  21. E. Torreele, M. Mazzucato, and H. Lishi Li, “Delivering the people’s vaccine: Challenges and proposals for the biopharmaceutical innovation system,” UCL Institute for Innovation and Public Purpose, Policy Brief 12. Available at https://www.ucl.ac.uk/bartlett/public-purpose/ sites/public-purpose/files/iipp-pb12_delivering-the-peoples-vaccine_final.pdf; G. Krikorian, “Refusing state funding of vaccine research in a corporate ‘double-blind’,” Remix the Commons Blog (2021). Available at https://www. remixthecommons.org/?p=5022.
  22. Flasi?ski M. Introduction to Artificial Intelligence. 1st ed. Switzerland: Springer International Publishing; 2016. p. 4.
  23. Gaëlle Krikorian And Els Torreele, We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis, JUNE 2021 VOLUME 23 NUMBER 1.
  24. gaëlle krikorian and els torreele, We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis, JUNE 2021 VOLUME 23 NUMBER 1.
  25. Global Health Centre, Graduate Institute Geneva. Covid-19 vaccine R&D investment. Available at https://www. knowledgeportalia.org/covid19-r-d-funding.
  26. Grilo, A.L.; Mantalaris, A. The Increasingly Human and Profitable Monoclonal Antibody Market. Trends Biotechnol. 2019, 37, 9–16. [CrossRef] [PubMed].
  27. H. Schellekens, M. Aldosari, H. Talsma, and E. Mastrobattista, “Making individualized drugs a reality,” Nature Biotechnology 35 (2017), pp. 507–513.
  28. Hammond SM. Dicing and slicing: the core machinery of the RNA interference pathway. FEBS Lett 2005;579:5822-9
  29. Hariom Rajput, Saurabh raj Pandey, A Review Article on Novel Drug Delivery System, Volume 10 ~ Issue 4 (2024) pp: 21-31 ISSN(Online) : 2347-2995.
  30. http://manufacturing.materialise.com/cases/wearable-tech-just-got-smarter-anouk-wipprechtsintel-edison-powered-3d-printed-synapse-dress viewed on 1 June 2015
  31. Jain, K.K.; Jain, K.K. Nanomolecular Diagnostics. In The Handbook of Nanomedicine; Springer: Berlin/Heidelberg, Germany, 2017; pp. 133–200. [CrossRef].
  32. K. Bajwa, B. Qorri, A. Decarlo, M. R. Szewczuk, Recent advances in “smart” delivery systems for extended drug release in cancer therapy, Int. J. Nanomed. 13 (2018) 4727—4745.
  33. K. Basu, L. Gostin, and N. Hassoun, “Pandemic preparedness and response: Beyond the WHO’s Access to COVID-19 Tools Accelerator,” Brookings Global Economy and Development Working Paper (2021). Available at https:// www.brookings.edu/wp-content/uploads/2021/04/Pandemic-preparedness-and-response_final.pdf. 31. A. M. Büyüm, C. Kenney, A. Koris, et al., “Decolonising global health: If not now, when?,” BMJ Global Health 5 (2020).
  34. K. Kupferschmidt, “‘Vaccine nationalism’ threatens global plan to distribute COVID-19 shots fairly,” Science Magazine (July 28, 2020). Available at https://www. sciencemag.org/news/2020/07/vaccine-nationalism-threatens-global-plan-distribute-covid-19-shots-fairly.
  35. K.K. Jain, The Handbook of Nanomedicine, DOI 10.1007/978-1-4939-6966-1_4.
  36. Kalepu, S.; Nekkanti, V. Insoluble Drug Delivery Strategies: Review of Recent Advances and Business Prospects. Acta Pharm. Sin. B 2015, 5, 442–453. [CrossRef].
  37. Kgmg. Available online: https://kpmg.com/xx/en/home/insights/2022/12/the-supply-chain-trends-shaking-up-2023.html (accessed on 2 May 2023).
  38. Kovacs, M. TECHNOLOGY: 3D printing gears up for fashion industry change. Aroq - JustStyle.Com (Global News), 1 (2015)
  39. Krikorian, G.; Torreele, E. We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis. Health Hum. Rights 2021, 23, 119–127.
  40. Lau JR, Geho WB, Snedekar GH, Inventors; SDG INC, An Ohio Corporation, Assignee; Targeted Liposomal Drug Delivery System. US Patent 20100209492. 2010 Aug 19.
  41. Lau JR,Geho WB, Snedekar GH, Inventors; Targeted Liposomal Drug Delivery System. US Patent 20070104777. 2007 May 10.
  42. Mahesh Shaw, 3D Printing Technology: Its application and scope in Fashion Industry, 27 August 2018.
  43. Mak, K.-K.; Pichika, M.R. Artificial Intelligence in Drug Development: Present Status and Future Prospects. Drug Discov. Today 2019, 24, 773–780. [CrossRef].
  44. Manish Vyas, Sourav Thakur, Bushra Riyaz, Kuldeep K Bansal, Bhupendra Tomar, Vijay Mishra, Artificial Intelligence: The Beginning of a New Era in Pharmacy Profession, Received: 28-07-2017 Revised: 12-04-2018 Accepted: 27-04-2018.
  45. Müller, R. Junghanns Nanocrystal Technology, Drug Delivery and Clinical Applications. Int. J. Nanomed. 2008, 3, 295–310. [CrossRef] [PubMed].
  46. Munos, B. Lessons from 60 Years of Pharmaceutical Innovation. Nat. Rev. Drug Discov. 2009, 8, 959–968. [CrossRef].
  47. National Nanotechnology Initiative. NNI Budget Supplements and Strategic Plans. Available online: https://www.nano.gov/N NIBudgetSupplementsandStrategicPlans (accessed on 1 May 2023).
  48. no. 12, (2015) p. 68. 9. L Adendorff, '3D technology: 3D printing the next frontier?’ Aroq - Just-Style.Com (Global News), (2014) p. 12, Entrepreneurial Studies Source, EBSCOhost, viewed 1 June 2015.)
  49.  Parker JS, Roe SM, Barford D. Structural insights into mRNA recognition from a PIWI domain-siRNA guide complex. Nature 2005;434:663-6 16. Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev 2001;15:188-200.
  50. R. R. Bhagwat, I. S. Vaidhya, Novel drug delivery systems: an overview, Int. J. Pharm. Sci. Res. 4 (2013) 970—982.
  51. R.R. Bhagwat and I.S. Vaidhya, Novel Drug Delivery Systems: An Overview, IJPSR (2013), Vol. 4, Issue 3.
  52. Sarpatwari, A.; Barenie, R.; Curfman, G.; Darrow, J.J.; Kesselheim, A.S. The US Biosimilar Market: Stunted Growth and Possible Reforms. Clin. Pharmacol. Ther. 2019, 105, 92–100. [CrossRef].
  53. Scannell, J.W.; Blanckley, A.; Boldon, H.; Warrington, B. Diagnosing the Decline in Pharmaceutical R&D Efficiency. Nat. Rev. Drug Discov. 2012, 11, 191–200. [CrossRef] [PubMed].
  54. Schmidt FR. About the nature of RNA interference. Appl Microbiol Biotechnol 2005;67:429-35
  55. Sharma, R.; Shishodia, A.; Gunasekaran, A.; Min, H.; Munim, Z.H. The Role of Artificial Intelligence in Supply Chain Management: Mapping the Territory. Int. J. Prod. Res. 2022, 60, 7527–7550. [CrossRef].
  56. Statistica. Artificial Intelligence (AI). Available from: https://www.statista.com/study/38609/artificialintelligence-ai-statista-dossier/. [Last accessed on 2017 Jun 24].
  57. Stefano Colombo, Xianghui Zeng, He´lo?¨se Ragelle & Camilla Foged, Complexity in the therapeutic delivery of RNAi medicines: an analytical challenge, 2014 Informa UK, Ltd. ISSN 1742-5247, e-ISSN 1744-7593.
  58. Takagi A, Yamashita N, Sonobe T,Inventors; Astellas Pharma INC Tokyo, Assignee; Intracellular Drug Delivery Improving Liposomes. US Patent 20070286898. 2007 Dec 13.
  59. Times of India Pharmaceutical Supply Chain Management: Crises and Innovations. Available online: https://timesofindia.indiat imes.com/blogs/voices/pharmaceutical-supply-chain-management-crises-and-innovations/ (accessed on 15 May 2023).
  60. Troiano, G.; Nolan, J.; Parsons, D.; Van Geen Hoven, C.; Zale, S. A Quality by Design Approach to Developing and Manufacturing Polymeric Nanoparticle Drug Products. AAPS J. 2016, 18, 1354–1365. [CrossRef] [PubMed].
  61. Whitehead KA, Langer R, Anderson DG. Knocking down barriers: advances in siRNA delivery. Nat Rev Drug Discov 2009;8:129-38
  62. World Health Assembly, Improving the Transparency of Markets for Medicines, Vaccines, and Other Health Products, WHA72.8 (2019). 38. Ahmed v. Union of India, para. 1., as cited in A. Kapcyzinski, “The right to medicines in an age of neoliberalism,” Humanity Journal 10/1 (2019).
  63. Y. H. Bae, K. Park, Targeted drug delivery to tumors: myths, reality and possibility, J. Control. Release. 153 (2011) 198—205. Available from: https://doi. org/10. 1016/j. jconrel. 2011. 06. 001.
  64. Yamauchi H, Morita H, Kikuchi H, Inventors; Daiichi pharmaceuticals Co.LTD, Assignee; Liposomes And Liposomal Dispersion. US Patent 20020182248. 2002 Dec 5.
  65. Zhang H, Kolb FA, Jaskiewicz L, et al. Single processing center models for human Dicer and bacterial RNase III. Cell 2004;118:57-68
  66. Zhang Y, Luo B, Iyer L, Inventors; Liposomal Delivery Vehicle For Hydrophobic Drugs. US Patent 20070014845. 2007 Jan 18.
  67. Zignify. Available online: https://zignify.net/what-is-supply-chain-disruption-and-what-are-its-causes/#:~:text=LogisticsDe laysandFailures,resultingincongestionanddelays (accessed on 2 May 2023).

Photo
Mr. Kuna Akash
Corresponding author

Bhaskara Institute of Pharmacy, Komatipalli(V), Bobbili, Vizianagaram, Andhara Pradesh, India

Photo
Damu Uma Sankar
Co-author

Assistent professor, Bhaskara Institute of Pharmacy, Komatipalli(V), Bobbili, Vizianagaram, Andhara Pradesh, India

Photo
Gudisi Rama Krishna
Co-author

Assistent professor, Bhaskara Institute of Pharmacy, Komatipalli(V), Bobbili, Vizianagaram, Andhara Pradesh, India

Photo
Dr. B. Kiran Kumar
Co-author

Professor of Sri Venkateswara college of pharmacy Etcherlla, Srikakulam, Andhra Pradesh, India

Photo
Mylapalli Ajay Kumar
Co-author

Bhaskara Institute of Pharmacy, Komatipalli(V), Bobbili, Vizianagaram, Andhara Pradesh, India

Photo
Dr. Konathala Rajesh
Co-author

Principal, Bhaskara Institute of Pharmacy Komatipalli(V), Bobbili, Vizianagaram, Andhara Pradesh, India

Gudisi Rama Krishna, Dr. Konathala Rajesh, Damu Uma Sankar, Kuna Akash*, Mylapalli Ajay Kumar, Dr. B. Kiran Kumar, Present Scenario in Pharmaceutical Technology Development: An Over View, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 2, 2656-2677. https://doi.org/10.5281/zenodo.14782383

More related articles
Formulation And Evaluation of Sustained Release Ma...
Muni Raja Lakshmi K, Naseema Shaik, Lavanya E, Venkata Rao Gadde,...
Investigating an Out of Specifications results –...
Abhinav Sawant, Amey Bhosale, Tejashri Kamble, Dr. Nilesh Chougul...
Overview Of Nanotechnology ...
Rajvardhan Desai , Seema Shinde, Dr. Nilesh Chougule, ...
A Comprehensive Review Of Phytochemical Profile And Pharmacological Attributes O...
R Ghana Shyam , Ananda V, D. Visagaperumal, Vineeth Chandy, ...
Drug Resistance In Various Disorders...
T. V. Sowjanya, B. Akhila, N. Ahalya, T. Sai Ram, T. Maneesha, V. Pavithra, ...
Related Articles
Effect Of Leech Application In Prolapsed Thrombosed Haemorrhoid A Case Study ...
Anjana H. Yadav , Rajendra Sonekar , Amol padole , Aruna sonekar, ...
In-Vitro Evaluation Of Anti-Inflammatory Property Of Sesamum Indicum Seeds And W...
Vishakha Shelke , Prathamesh Choudhari, S. N. Belhekar, ...
Review On Micro-Gas Chromatography System For Analysis Of Multiple Low-Concentra...
Mrunal Shashikant Pagare, Varun Sunil Dev, Rajashree Gulshan Jagtap, Shraddha Prakashdas Vaishnav, ...
Formulation And Evaluation of Sustained Release Matrix Tablets A Review...
Muni Raja Lakshmi K, Naseema Shaik, Lavanya E, Venkata Rao Gadde, ...
More related articles
Formulation And Evaluation of Sustained Release Matrix Tablets A Review...
Muni Raja Lakshmi K, Naseema Shaik, Lavanya E, Venkata Rao Gadde, ...
Investigating an Out of Specifications results – The most challenging function...
Abhinav Sawant, Amey Bhosale, Tejashri Kamble, Dr. Nilesh Chougule, ...
Overview Of Nanotechnology ...
Rajvardhan Desai , Seema Shinde, Dr. Nilesh Chougule, ...
Formulation And Evaluation of Sustained Release Matrix Tablets A Review...
Muni Raja Lakshmi K, Naseema Shaik, Lavanya E, Venkata Rao Gadde, ...
Investigating an Out of Specifications results – The most challenging function...
Abhinav Sawant, Amey Bhosale, Tejashri Kamble, Dr. Nilesh Chougule, ...
Overview Of Nanotechnology ...
Rajvardhan Desai , Seema Shinde, Dr. Nilesh Chougule, ...