Advancements in Immunomodulation, Drug Discovery, and Medicine: A Comprehensive Review
Kavita Khatana1* , Vikas Malgotra2 , Rokeya Sultana3 , NaliniKantaSahoo4 , ShivamMaurya5 , Anamika Das6 , Chetan DM7
1Department of Chemical Engineering, Shiv Nadar Institution of Eminence Deemed to be University, greater noida, Dadri. U.P. India
2Department of Dermatology, Rama Medical College Hapur, India
3Department of Pharmacognosy, Yenepoya Pharmacy College and Research Centre, Yenepoya (deemed to be University), Deralakatte, Mangalore. Karnataka, India
4MIT College of Pharmacy, MIT Campus, Ram Ganga Vihar, Phase II, Moradabad, Uttar Pradesh, 244001, India
5Department of Plant Pathology, School of Agriculture, Lovely Professional University, Phagwara Punjab, India
6Department of Dairy Chemistry, Warner College of Dairy Technology, SHUATS, Prayagraj, India
7Department of Biotechnology,NMAM Institute of Technology-Affiliated to NITTE (Deemed to beUniversity),Nitte, 574110-Karnataka, India
Corresponding Author Email: Kavita.khatana@snu.edu.in
DOI : http://dx.doi.org/10.5281/zenodo.8185439
Abstract
This comprehensive review provides an overview of the recent advancements in the field of immunomodulation, drug discovery, and their impact on medicine. Immunomodulation, the modulation of the immune system, has emerged as a promising approach for the treatment of various diseases, including autoimmune disorders, cancer, infectious diseases, and inflammatory conditions. The review highlights the progress made in personalized immunomodulatory therapies, combination therapies, identification of novel targets and pathways, overcoming resistance, management of safety and adverse effects, regulatory challenges, and considerations of cost and accessibility. The potential impact of immunomodulation and drug discovery on the future of medicine is discussed, emphasizing the role of precision medicine, advancements in cancer treatment, management of infectious diseases, treatment of autoimmune and inflammatory disorders, and the exploration of future therapeutic targets. Despite the challenges, the review underscores the tremendous potential of immunomodulation and drug discovery in transforming healthcare and improving patient outcomes.
Keywords
Introduction
Overview of the significance of immunomodulation, drug discovery, and medicine in healthcare
The field of immunomodulation holds significant importance in healthcare, as it involves the manipulation of the immune system to achieve therapeutic outcomes [1]. The immune system plays a vital role in protecting the body against infections, pathogens, and abnormal cells. However, dysregulation of immune responses can lead to autoimmune diseases, allergies, and cancer [2]. Immunomodulatory interventions aim to restore or modulate immune function, offering potential therapeutic benefits. Drug discovery is a multidisciplinary field essential for developing new medications to treat diseases [3]. Traditional drug discovery methods have limitations in terms of cost, time, and success rates. Therefore, innovative approaches such as high-throughput screening, rational drug design, and drug repurposing have gained prominence [4]. These advancements have the potential to expedite the discovery of effective and safe therapeutic agents. The intersection of immunomodulation and drug discovery has opened up new avenues for therapeutic interventions. Understanding the immunomodulatory effects of drugs and their mechanisms of action has paved the way for repurposing existing medications for new indications [5]. Furthermore, researchers are actively exploring novel agents that can selectively modulate immune responses to treat diseases more effectively [6].
The significance of immunomodulation, drug discovery, and medicine in healthcare cannot be overstated. These fields have the potential to revolutionize the treatment landscape for various diseases, including autoimmune disorders, cancer, and infectious diseases. By understanding the complexities of the immune system and developing innovative therapies, healthcare professionals can provide personalized and targeted treatments, improving patient outcomes and quality of life.
Immunomodulation
Principles and mechanisms of immunomodulation
Immunomodulation involves the manipulation of the immune system to achieve therapeutic outcomes by restoring or modulating immune responses. Understanding the principles and mechanisms of immunomodulation is crucial for developing effective treatments for various diseases [7]. This section will provide a detailed discussion of the principles and mechanisms underlying immunomodulation.
Principles of Immunomodulation:
a. Homeostasis: Immunomodulation aims to restore the balance or homeostasis of the immune system, which can be disrupted in diseases such as autoimmune disorders or allergies. It involves modulating both the innate and adaptive immune responses to achieve an appropriate immune reaction.
b. Specificity: Immunomodulatory interventions can be targeted towards specific immune cell populations, signaling pathways, or cytokines, depending on the disease being treated. Specificity allows for tailored therapies that selectively modify immune responses while minimizing off-target effects.
c. Plasticity: The immune system exhibits a degree of plasticity, meaning it can adapt and change its responses. Immunomodulation takes advantage of this plasticity to modify immune reactions in response to different stimuli or disease states.
d. Contextual Effects: Immunomodulatory interventions can have different effects depending on the context in which they are applied. For example, the same immunomodulatory agent may promote inflammation in one disease condition while suppressing it in another, highlighting the importance of considering the specific disease context.
Mechanisms of Immunomodulation
a. Suppression: Immunomodulation can involve the suppression of immune responses to reduce inflammation or prevent immune-mediated damage. This can be achieved through the inhibition of pro-inflammatory cytokines, downregulation of immune cell activation, or induction of regulatory T cells (Tregs) that suppress excessive immune reactions.
b. Stimulation: Conversely, immunomodulation can also involve the stimulation of immune responses to enhance immune function. This can be achieved by promoting the activation and proliferation of immune cells, such as T cells or natural killer (NK) cells, or by enhancing the production of specific cytokines or antibodies.
c. Immune Cell Modulation: Immunomodulatory interventions can directly target immune cells to modulate their function. For example, monoclonal antibodies can bind to specific cell surface receptors, such as immune checkpoint molecules, to regulate immune cell activity.
d. Cytokine Modulation: Cytokines play a crucial role in immune regulation, and immunomodulation can involve the modulation of cytokine production or signaling. This can be achieved by blocking or enhancing specific cytokines, altering their interactions with receptors, or modulating the balance between pro-inflammatory and anti-inflammatory cytokines.
e. Tolerance Induction: Immunomodulation can aim to induce immune tolerance, particularly in the context of autoimmune diseases or organ transplantation. This involves promoting immune tolerance towards self-antigens or transplanted tissues by mechanisms such as inducing anergy or deletion of autoreactive immune cells.
Role of immunomodulation in autoimmune diseases
Immunomodulation plays a crucial role in the management and treatment of autoimmune diseases [8]. Autoimmune diseases arise when the immune system mistakenly targets and attacks healthy tissues and organs, leading to chronic inflammation and tissue damage. Immunomodulatory interventions aim to restore balance and regulate immune responses to prevent or alleviate the symptoms of autoimmune diseases. Here, we will discuss the role of immunomodulation in autoimmune diseases, focusing on some common examples.
Rheumatoid Arthritis (RA): Immunomodulatory therapies have significantly improved the management of RA, a chronic inflammatory disease primarily affecting the joints. Disease-modifying anti-rheumatic drugs (DMARDs), including methotrexate, sulfasalazine, and biologic agents such as tumor necrosis factor (TNF) inhibitors, are commonly used to modulate the immune response in RA [9]. These drugs target specific immune cells, such as T cells and B cells, or cytokines involved in the inflammatory process, reducing joint inflammation and preventing further damage.
Systemic Lupus Erythematosus (SLE): SLE is a complex autoimmune disease that affects multiple organ systems. Immunomodulatory treatments for SLE focus on reducing autoantibody production and suppressing immune cell activation. This can involve the use of corticosteroids to dampen immune responses, immunosuppressive agents like cyclophosphamide or mycophenolate mofetil, and newer targeted biologic therapies, such as belimumab, which specifically targets B-cell activating factor (BAFF) [10].
Multiple Sclerosis (MS): MS is an autoimmune disease characterized by immune-mediated damage to the central nervous system. Immunomodulatory treatments for MS aim to modulate the immune response by reducing inflammation and suppressing immune cell activity. Disease-modifying therapies (DMTs), including interferon-beta and monoclonal antibodies like natalizumab or ocrelizumab, are commonly used to modify the course of the disease, reduce relapses, and slow down the progression of disability [11].
Inflammatory Bowel Disease (IBD): IBD encompasses conditions like Crohn’s disease and ulcerative colitis, characterized by chronic inflammation of the gastrointestinal tract. Immunomodulatory interventions in IBD aim to reduce gut inflammation and regulate immune responses. Medications such as corticosteroids, thiopurines, and biological agents targeting TNF or other inflammatory cytokines are utilized to modulate the immune response and control disease activity [12].
These examples highlight the role of immunomodulation in autoimmune diseases, where the goal is to regulate the immune response to reduce inflammation, prevent tissue damage, and improve patient outcomes. It is important to note that the choice of immunomodulatory therapy depends on the specific autoimmune disease, its severity, and individual patient factors. Combination therapies, tailored treatment regimens, and ongoing monitoring are often employed to optimize the management of autoimmune diseases.
Immunomodulatory approaches for transplantation and graft rejection
Immunomodulatory approaches play a crucial role in transplantation medicine by managing immune responses and preventing graft rejection. Transplantation involves the transfer of organs, tissues, or cells from a donor to a recipient, and the recipient’s immune system recognizes the transplanted graft as foreign, triggering an immune response that can lead to graft rejection. Immunomodulatory strategies aim to modulate and regulate the immune response to promote graft acceptance while minimizing the risk of rejection. Here, we will discuss some immunomodulatory approaches used in transplantation and graft rejection.
Immunosuppressive Drugs: Immunosuppressive drugs are the cornerstone of immunomodulation in transplantation. They work by suppressing the immune system to prevent rejection of the transplanted organ or tissue. Commonly used immunosuppressive agents include calcineurin inhibitors (e.g., cyclosporine, tacrolimus), corticosteroids (e.g., prednisone), and antimetabolites (e.g., mycophenolate mofetil, azathioprine). These medications inhibit immune cell activation and cytokine production, reducing the immune response against the graft [13].
Induction Therapy: Induction therapy involves the use of potent immunosuppressive agents early after transplantation to provide intense immunosuppression and prevent acute rejection. Agents such as anti-thymocyte globulin (ATG) or interleukin-2 receptor antagonists (e.g., basiliximab) are commonly used as induction agents. They target specific immune cells or receptors to inhibit immune responses and reduce the risk of acute rejection [14].
Tolerance Induction: Tolerance induction aims to establish immune tolerance towards the transplanted graft, allowing long-term acceptance without the need for continuous immunosuppression. Approaches such as mixed chimerism, where the recipient receives hematopoietic stem cells from the donor, or regulatory T cell (Treg) therapy are being explored to induce immune tolerance and reduce the risk of graft rejection [15].
Costimulation Blockade:Costimulation blockade involves blocking the interaction between antigen-presenting cells and T cells, which is necessary for T cell activation and proliferation. This approach targets molecules such as CD80/86 (e.g., abatacept) or CD40/CD154 (e.g., belatacept) to inhibit T cell activation and suppress the immune response against the graft [16].
Cell-Based Therapies: Cell-based therapies, such as mesenchymal stem cells (MSCs) or regulatory T cells (Tregs), have shown promise in modulating immune responses and promoting graft acceptance. MSCs possess immunomodulatory properties and can suppress immune cell activation, while Tregs play a key role in immune tolerance. These cells can be infused into transplant recipients to modulate the immune response and enhance graft acceptance [17-19].
Tolerance-Promoting Regimens: Various tolerance-promoting regimens are being investigated, including the use of specific immunomodulatory agents, such as rapamycin or co-stimulation blockers, in combination with reduced immunosuppressive drug regimens. These regimens aim to maintain graft acceptance while minimizing the side effects of long-term immunosuppression [20].
Immunomodulatory approaches in transplantation aim to strike a balance between preventing graft rejection and minimizing the risk of infections and other complications associated with immunosuppression. The choice of immunomodulatory strategy depends on factors such as the type of transplantation, the risk of rejection, and the individual patient’s characteristics.
Immunomodulation in cancer therapy
Immunomodulation has emerged as a promising approach in cancer therapy by harnessing the power of the immune system to recognize and eliminate cancer cells. The field of cancer immunotherapy has witnessed significant advancements in recent years, with immunomodulatory strategies demonstrating remarkable efficacy in various types of cancers. These approaches aim to modulate the immune response, enhance anti-tumor immunity, and overcome the immunosuppressive mechanisms employed by tumors. Here, we will discuss some key immunomodulatory strategies used in cancer therapy.
Immune Checkpoint Inhibitors: Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by blocking inhibitory pathways that regulate immune responses, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). By inhibiting these checkpoints, ICIs unleash the immune system, allowing T cells to mount a more robust and sustained anti-tumor response. Drugs like pembrolizumab, nivolumab, and ipilimumab have shown significant clinical benefits in multiple cancer types [21].
Adoptive Cell Transfer: Adoptive cell transfer (ACT) involves the infusion of immune cells, such as tumor-infiltrating lymphocytes (TILs) or genetically engineered T cells, into cancer patients. ACT can be combined with genetic modifications to enhance the anti-tumor activity of the infused cells, such as chimeric antigen receptor (CAR) T cells. These engineered immune cells recognize and target tumor-specific antigens, leading to tumor regression and durable responses [22].
Cancer Vaccines: Cancer vaccines aim to stimulate the immune system to recognize and attack cancer cells. Different types of vaccines, including peptide-based, dendritic cell-based, and viral vector-based vaccines, are being developed to induce specific immune responses against tumor-associated antigens. Vaccines can be combined with other immunomodulatory agents to enhance their efficacy and promote long-lasting anti-tumor immunity [23].
Cytokine Therapy: Cytokines are key regulators of immune responses and can be utilized as immunomodulators in cancer therapy. Interleukin-2 (IL-2) and interferon-alpha (IFN-α) have been used in the treatment of certain cancers to enhance the activation and proliferation of immune cells. IL-2 therapy, in particular, has shown significant clinical responses in metastatic melanoma and renal cell carcinoma [24].
Targeted Immunomodulatory Agents: Several targeted immunomodulatory agents are being developed to overcome specific immunosuppressive mechanisms employed by tumors. For example, agents targeting regulatory T cells (Tregs), such as anti-CTLA-4 antibodies, aim to reduce immunosuppression in the tumor microenvironment. Other targeted approaches include inhibiting immunosuppressive cells like myeloid-derived suppressor cells (MDSCs) or modulating immunosuppressive cytokines like transforming growth factor-beta (TGF-β) [25].
The field of cancer immunotherapy continues to evolve rapidly, with ongoing research focused on identifying optimal combinations of immunomodulatory agents, biomarkers for patient selection, and strategies to overcome resistance. Immunomodulation holds great promise for improving outcomes in cancer patients by harnessing the immune system’s inherent ability to recognize and eliminate cancer cells.
Immunomodulatory interventions for allergic disorders
Immunomodulatory interventions play a crucial role in the management of allergic disorders by modulating the immune response and reducing the symptoms associated with allergies. Allergic disorders, such as allergic rhinitis, asthma, and atopic dermatitis, result from an exaggerated immune response to harmless substances, known as allergens. Immunomodulatory strategies aim to regulate the immune system, reduce inflammation, and improve allergic symptoms. Here, we will discuss some key immunomodulatory interventions used in the treatment of allergic disorders.
Allergen-specific Immunotherapy: Allergen-specific immunotherapy (AIT), commonly known as allergy shots, involves the administration of gradually increasing doses of allergens to desensitize the immune system and induce immunological tolerance. AIT can be delivered through subcutaneous injections or sublingual drops/tablets. This therapy modifies the immune response, shifting it from an allergic phenotype to a more tolerant state. AIT has demonstrated long-term efficacy in reducing symptoms and medication use in allergic rhinitis and allergic asthma [26].
Biologic Therapies: Biologic therapies target specific molecules or cells involved in the allergic immune response. Monoclonal antibodies, such as omalizumab and dupilumab, have shown efficacy in allergic asthma and atopic dermatitis, respectively. Omalizumab targets immunoglobulin E (IgE), reducing its binding to allergens and suppressing the allergic response. Dupilumab blocks interleukin-4 (IL-4) and interleukin-13 (IL-13) signaling, key cytokines involved in allergic inflammation [27-28].
Anti-inflammatory Medications: Anti-inflammatory medications, including corticosteroids, are commonly used to control allergic inflammation. They work by suppressing the immune response, reducing inflammation, and alleviating allergic symptoms. Inhaled corticosteroids are the mainstay of treatment for allergic asthma, while topical corticosteroids are used in atopic dermatitis. These medications help to modulate the immune response and provide symptomatic relief [29].
Immunomodulatory Agents: Several immunomodulatory agents, such as cyclosporine and methotrexate, have been used in the management of severe allergic disorders that are unresponsive to conventional treatments. These medications modulate immune cell function and reduce inflammatory responses. They are typically reserved for cases with significant disease burden and when other treatments have proven ineffective [30].
Probiotics: Probiotics, live microorganisms that confer health benefits, have shown promise in modulating the immune response and improving symptoms of allergic disorders. They can modulate immune cell function, restore gut microbiota balance, and enhance immune tolerance. Probiotics have been studied in allergic rhinitis, atopic dermatitis, and allergic asthma, with variable results [31].
Immunomodulatory interventions provide valuable tools for the management of allergic disorders. These approaches target different aspects of the immune response, ranging from desensitization and tolerance induction to immune cell modulation and inflammation control.
Drug Discovery
Overview of the drug discovery process
The drug discovery process is a complex and multifaceted endeavor that involves the identification, development, and refinement of new therapeutic agents. It encompasses various stages, from target identification to clinical trials, with the ultimate goal of delivering safe and effective drugs to address unmet medical needs. Here, we provide an overview of the drug discovery process and its key components.
Target Identification and Validation: The drug discovery process begins with the identification and validation of a molecular target that plays a critical role in the disease process. Targets can be proteins, enzymes, receptors, or specific genetic elements associated with the disease. Advances in genomics, proteomics, and bioinformatics have greatly facilitated target identification and validation, allowing researchers to pinpoint molecules or pathways that could be modulated to achieve a therapeutic effect [32].
Hit Discovery and Lead Generation: Once a target is identified, the next step is to discover “hits” or initial molecules that interact with the target. High-throughput screening (HTS) of large compound libraries, virtual screening, and structure-based drug design are commonly employed approaches for hit discovery. Promising hits are further optimized to generate “leads” that possess desirable drug-like properties, such as potency, selectivity, and pharmacokinetic characteristics [33].
Lead Optimization: Lead optimization involves the iterative process of modifying and refining lead compounds to improve their efficacy, safety, and pharmacokinetic properties. Medicinal chemists employ various strategies, including structural modifications, analog synthesis, and computational modeling, to enhance the drug-likeness of lead compounds. This stage aims to strike a balance between potency and selectivity while ensuring optimal absorption, distribution, metabolism, and excretion (ADME) properties [34].
Preclinical Development: Preclinical development involves in vitro and in vivo studies to assess the safety, efficacy, and pharmacological properties of the lead compound. This stage includes rigorous testing for toxicity, formulation development, pharmacokinetic profiling, and assessment of the compound’s mechanism of action. Preclinical data support the selection of the most promising candidate for clinical trials and provide crucial information for regulatory submissions [35].
Clinical Trials: Clinical trials are conducted in multiple phases to evaluate the safety, efficacy, and dosage regimens of the investigational drug in human subjects. Phase I trials focus on safety and dosage determination, Phase II trials assess efficacy and side effects in a larger patient population, and Phase III trials further evaluate safety and efficacy in a large-scale, randomized, and controlled manner. These trials generate robust data to support regulatory approval and provide insights into the drug’s therapeutic potential [36].
Regulatory Approval and Post-marketing Surveillance: Following the successful completion of clinical trials, the drug is submitted to regulatory authorities for approval. Regulatory agencies evaluate the accumulated data on safety, efficacy, and quality to assess the benefits and risks of the drug. If approved, the drug can be marketed and distributed to patients. Post-marketing surveillance involves continuous monitoring of the drug’s safety and effectiveness in real-world clinical settings [37].
The drug discovery process is a dynamic and resource-intensive journey that requires collaboration between various disciplines, including chemistry, biology, pharmacology, and clinical medicine. Advances in technology and scientific understanding continue to drive innovation in drug discovery, leading to the development of novel therapeutics to combat diseases.
Traditional drug discovery methods and their limitations
Traditional drug discovery methods have played a crucial role in identifying and developing numerous therapeutics that have transformed healthcare. However, these methods have certain limitations that hinder the efficiency and effectiveness of the drug discovery process. Understanding these limitations is essential for developing improved approaches. Here, we discuss some of the traditional drug discovery methods and their associated limitations.
High-Throughput Screening (HTS): High-throughput screening involves testing large libraries of compounds against specific targets or disease models to identify potential drug candidates. While HTS has accelerated the screening process, its limitations include the narrow focus on a specific target, lack of physiological relevance, and high rates of false positives or false negatives due to assay artifacts [38].
Structure-Based Drug Design (SBDD): Structure-based drug design utilizes knowledge of the three-dimensional structure of target proteins to design and optimize small molecule drugs. Although SBDD has been successful in many cases, it relies on the availability of high-resolution protein structures and may not effectively address targets with complex or flexible binding sites [39].
Phenotypic Screening: Phenotypic screening involves testing compounds for their effects on whole cells or organisms, aiming to identify drugs that modulate disease-associated phenotypes. While phenotypic screening allows for a more holistic approach, it can be challenging to determine the precise target and mechanism of action of identified compounds, leading to difficulties in optimization and potential off-target effects [40].
Natural Product Screening: Natural product screening involves the exploration of diverse chemical compounds derived from natural sources, such as plants, microbes, or marine organisms, for their therapeutic potential. However, natural product-based drug discovery faces limitations, including the limited supply of natural sources, difficulty in isolating and characterizing active compounds, and challenges in optimization and modification for drug-like properties [41].
Combinatorial Chemistry: Combinatorial chemistry involves the synthesis of large libraries of diverse compounds by systematically combining building blocks. While it allows for rapid generation of chemical diversity, the process often generates vast numbers of compounds with limited biological relevance, and hit compounds may lack the desired potency, selectivity, or pharmacokinetic properties [42].
These limitations highlight the need for innovative approaches in drug discovery, such as computational methods, target-agnostic strategies, and more emphasis on translational research to bridge the gap between preclinical and clinical stages. Integrating multidisciplinary techniques and leveraging emerging technologies can enhance the efficiency and success rate of drug discovery efforts.
Advances in high-throughput screening techniques
Advances in high-throughput screening (HTS) techniques have revolutionized the field of drug discovery by enabling the rapid screening of large compound libraries to identify potential drug candidates. HTS allows researchers to test thousands or even millions of compounds against specific biological targets or disease models, significantly accelerating the drug discovery process. Here, we will discuss some of the key advancements in HTS techniques.
High-Throughput Screening Platforms: Traditional HTS platforms involve the use of microtiter plates, where compounds are tested in multi-well formats. However, recent advancements have led to the development of more sophisticated screening platforms. For example, microfluidic-based platforms offer precise control over fluid flow, allowing for miniaturization and automation of assays. These platforms enable high-throughput screening in nanoliter volumes, reducing reagent consumption and increasing throughput [43].
Label-Free Screening Technologies: Label-free screening techniques eliminate the need for fluorescent or radioactive labels, enabling real-time monitoring of biochemical or cellular events. These techniques include surface plasmon resonance (SPR), impedance-based assays, and mass spectrometry-based approaches. Label-free methods provide valuable kinetic and mechanistic information about the interactions between compounds and their targets, enhancing the understanding of drug-target interactions [44].
Phenotypic Screening: Phenotypic screening involves testing compounds in cellular or organismal models to identify those that induce a desired phenotypic effect. This approach offers a more holistic view of drug activity and can uncover novel targets or mechanisms. Advances in automated microscopy, image analysis algorithms, and high-content screening have enabled the screening of complex phenotypes in a high-throughput manner [45].
Data Integration and Analysis: With the increasing complexity and volume of HTS data, advanced data integration and analysis methods have become crucial. Computational tools, machine learning algorithms, and artificial intelligence approaches are employed to extract meaningful information, identify hit compounds, and optimize lead candidates. These methods facilitate data-driven decision-making and help to prioritize compounds for further evaluation [46].
DNA-Encoded Libraries (DELs): DNA-encoded libraries (DELs) are large collections of small molecules, where each compound is linked to a unique DNA barcode. DELs enable the screening of billions of compounds in a single experiment. By leveraging the power of DNA amplification and sequencing technologies, hit compounds can be identified and decoded rapidly. DELs have gained significant attention in recent years due to their potential for screening vast chemical space and accelerating hit discovery [47].
These advancements in HTS techniques have revolutionized the field of drug discovery, enabling researchers to screen larger compound libraries, generate more relevant data, and improve the efficiency of lead discovery and optimization processes.
Target-based drug discovery and rational drug design
Target-based drug discovery and rational drug design are two closely related approaches in the field of pharmaceutical research and development. These strategies aim to identify and design drugs that specifically target a particular molecule or pathway involved in a disease process, to achieve greater efficacy and minimize side effects. Here, we will discuss target-based drug discovery and rational drug design, highlighting their significance and key methodologies.
Target-based drug discovery involves identifying a specific molecular target, such as a protein or receptor, that plays a crucial role in disease. The target is selected based on its involvement in the disease mechanism, its accessibility to therapeutic intervention, and its potential druggability. Once the target is identified, the next step is to design and develop molecules or compounds that interact with the target and modulate its activity.
Rational drug design employs computational and molecular modeling techniques to guide the design and optimization of drug candidates. It involves a systematic and structure-based approach, where the three-dimensional structure of the target is determined or predicted, and then computational methods are used to identify or design small molecules that fit into the target’s binding site. These molecules are optimized for potency, selectivity, and other pharmacological properties through iterative cycles of design, synthesis, and testing.
Several methodologies are employed in target-based drug discovery and rational drug design:
Target Identification and Validation: This step involves identifying the molecular targets that are involved in the disease and validating their role through various experimental techniques, including genomics, proteomics, and biochemical assays. The information obtained helps prioritize targets for drug development [48].
Structural Biology and Target Characterization: Determining the three-dimensional structure of the target protein through techniques like X-ray crystallography, NMR spectroscopy, or cryo-electron microscopy provides valuable insights into its functional sites and allows for structure-based drug design [49].
Computational Approaches: Computational methods, such as molecular docking, molecular dynamics simulations, and virtual screening, are used to predict the binding affinity and interactions between drug candidates and the target protein. These techniques aid in the identification and optimization of lead compounds.
Chemical Synthesis and Optimization: Once lead compounds are identified, medicinal chemists synthesize and optimize them to improve their potency, selectivity, pharmacokinetic properties, and safety profiles. Structure-activity relationship (SAR) studies guide the iterative synthesis and modification of chemical compounds [50].
In vitro and In vivo Evaluation: Drug candidates undergo rigorous testing in cellular and animal models to assess their efficacy, toxicity, and pharmacokinetic properties. These preclinical studies help identify promising drug candidates for further development [51].
Target-based drug discovery and rational drug design have significantly advanced the field of drug development, allowing for the design of more selective and effective therapies. These approaches have led to the discovery of numerous successful drugs, ranging from small molecules to biologics, across various therapeutic areas.
Drug repurposing and combination therapies
Drug repurposing and combination therapies have emerged as promising strategies in modern medicine to expedite the development of new treatments and improve patient outcomes. These approaches leverage existing drugs and combine them with other agents to address unmet medical needs, enhance therapeutic efficacy, and overcome drug resistance. Here, we will discuss the significance of drug repurposing and combination therapies and provide references to support their application.
Drug Repurposing: Drug repurposing, also known as drug repositioning or drug reprofiling, involves identifying new therapeutic uses for existing drugs that were initially developed for different indications. This approach offers several advantages, including reduced time and cost compared to de novo drug development. By repurposing existing drugs, researchers can leverage their established safety profiles and known pharmacokinetics, expediting the clinical translation of new therapeutic applications. Examples of successful drug repurposing include sildenafil (originally developed for angina, now used for erectile dysfunction) and thalidomide (initially developed as a sedative, now used for multiple myeloma and leprosy) [52].
Combination Therapies: Combination therapies involve the simultaneous or sequential use of multiple drugs to achieve synergistic effects and enhance therapeutic outcomes. This approach is particularly relevant in complex diseases, such as cancer, infectious diseases, and neurological disorders, where single-agent treatments may have limited efficacy or induce drug resistance. Combination therapies can target different pathways or cellular components involved in disease progression, improving treatment response rates and reducing the likelihood of treatment resistance. For example, combination antiretroviral therapy (ART) has revolutionized the management of HIV/AIDS, significantly improving patient outcomes [53].
Drug repurposing and combination therapies hold immense potential in accelerating the discovery of new treatments and addressing unmet medical needs. These approaches maximize the value of existing drugs and offer novel therapeutic options to patients, ultimately improving healthcare outcomes.Bottom of Form
Emerging technologies in drug discovery (e.g., artificial intelligence, machine learning)
Emerging technologies, such as artificial intelligence (AI) and machine learning (ML), have revolutionized the field of drug discovery, offering innovative approaches to accelerate the identification and development of new therapeutic compounds. These technologies leverage computational power and data analysis to streamline various aspects of the drug discovery process, including target identification, lead optimization, and prediction of drug properties. Here, we will discuss the role of AI and ML in drug discovery and their potential implications for future advancements in the field.
AI in Drug Target Identification: AI techniques, such as deep learning and natural language processing, can analyze vast amounts of biological data, including genomic data, protein structures, and literature, to identify potential drug targets. These approaches can rapidly identify novel targets and prioritize them based on their biological relevance and druggability[54].
ML in Compound Screening and Design: ML algorithms can analyze large chemical databases and predict the activity, potency, and toxicity of potential drug candidates. By learning from existing data on drug-target interactions and compound properties, ML models can suggest novel compounds with desired properties for further testing. This enables more efficient and targeted screening of potential drug candidates [55].
AI for Drug Repurposing: AI-driven approaches have facilitated the identification of new therapeutic indications for existing drugs. By analyzing comprehensive datasets, including drug-target interactions, disease pathways, and clinical data, AI algorithms can identify potential drug candidates that may have efficacy in different diseases. This drug repurposing strategy can significantly accelerate the development of new treatments [56].
Predictive Modeling of Drug Properties: ML models can predict various drug properties, such as pharmacokinetics, toxicity, and drug-drug interactions, based on molecular features and chemical structures. These models aid in the early stages of drug development by guiding the selection of compounds with optimal drug-like properties, reducing the time and cost associated with experimental testing [57].
AI for Personalized Medicine: AI technologies hold great potential in enabling personalized medicine by analyzing large-scale patient data, including genomics, proteomics, and clinical data. These analyses can help identify patient subgroups, predict drug response, and optimize treatment regimens, leading to more targeted and effective therapies [58].
The integration of AI and ML in drug discovery has the potential to revolutionize the pharmaceutical industry by accelerating the drug development process, reducing costs, and enabling the discovery of novel therapies. However, challenges such as data quality, interpretability, and regulatory considerations need to be addressed to fully leverage the potential of these technologies in drug discovery.
The interplay between Immunomodulation and Drug Discovery
The role of immunomodulation in drug development
Immunomodulation plays a crucial role in drug development by targeting and modulating the immune system’s response to various diseases. The immune system is a complex network of cells, tissues, and molecules that defends the body against pathogens and maintains homeostasis. However, in certain conditions, the immune system can become dysregulated, leading to chronic inflammation, autoimmune disorders, or inadequate immune responses against cancer or infections. Immunomodulatory drugs aim to restore or rebalance the immune system’s function, either by enhancing or suppressing specific immune responses, depending on the therapeutic goal.
One area where immunomodulation has been extensively explored is in the treatment of autoimmune diseases. Autoimmune diseases arise when the immune system mistakenly attacks the body’s tissues. Examples include rheumatoid arthritis, multiple sclerosis, and systemic lupus erythematosus. Immunomodulatory drugs, such as disease-modifying antirheumatic drugs (DMARDs) or biologics targeting specific immune cells or molecules, have been developed to regulate the immune response and reduce inflammation. For instance, tumor necrosis factor (TNF) inhibitors, such as etanercept and infliximab, have been effective in treating autoimmune conditions like rheumatoid arthritis by blocking the activity of TNF-alpha, a pro-inflammatory cytokine [59].
Immunomodulation also plays a vital role in cancer treatment. The immune system can recognize and eliminate cancer cells through immune surveillance mechanisms. However, tumors can develop strategies to evade immune detection and suppression. Immune checkpoint inhibitors, such as pembrolizumab and nivolumab, have been developed to block inhibitory pathways in the immune system, particularly cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1), enhancing the anti-tumor immune response. These immunomodulatory drugs have shown remarkable efficacy in various cancers, including melanoma, lung cancer, and renal cell carcinoma [60].
In infectious diseases, immunomodulation is a promising approach to enhance the immune response against pathogens. Vaccines, the most well-known immunomodulatory agents, stimulate the immune system to generate protective responses against specific pathogens. Recent advancements in vaccine development, such as mRNA-based vaccines against COVID-19, have highlighted the potential of immunomodulation in generating robust and targeted immune responses [61].
Immunomodulatory drugs are also being investigated in the field of regenerative medicine and organ transplantation. For instance, immunosuppressive drugs like calcineurin inhibitors (e.g., cyclosporine) and mammalian target of rapamycin (mTOR) inhibitors (e.g., sirolimus) are used to prevent rejection in organ transplant recipients by suppressing the immune response against the transplanted organ [62].
Immunomodulatory effects of existing drugs and their therapeutic implications
Existing drugs that were not originally developed as immunomodulatory agents have been found to possess immunomodulatory effects, providing potential therapeutic implications in various disease conditions. Here are some examples:
Statins: Statins, commonly used for managing high cholesterol levels, have been shown to possess immunomodulatory properties. They can reduce the production of pro-inflammatory cytokines and promote anti-inflammatory responses. Statins have been investigated for their potential in reducing inflammation and improving outcomes in autoimmune diseases such as rheumatoid arthritis and multiple sclerosis [63].
Nonsteroidal Anti-inflammatory Drugs (NSAIDs): NSAIDs, including drugs like aspirin and ibuprofen, primarily function as analgesics and anti-inflammatory agents. Their anti-inflammatory effects arise from inhibiting the production of inflammatory mediators such as prostaglandins. NSAIDs have been explored for their potential immunomodulatory effects in conditions like inflammatory bowel disease and autoimmune disorders [64].
Metformin: Metformin, a widely prescribed drug for managing type 2 diabetes, has been found to have immunomodulatory effects. It can modulate immune cell function and reduce chronic inflammation. Metformin has been investigated for its potential in cancer treatment, as it may enhance the anti-tumor immune response and improve outcomes in certain cancers [65].
Glucocorticoids: Glucocorticoids, such as prednisone and dexamethasone, are potent anti-inflammatory and immunosuppressive drugs. They can modulate immune cell activity and suppress the immune response. Glucocorticoids are commonly used in the treatment of autoimmune diseases, allergic reactions, and inflammatory conditions [66].
Antihistamines: Antihistamines, such as cetirizine and loratadine, are primarily used to alleviate allergy symptoms by blocking the action of histamine. In addition to their antiallergic effects, some antihistamines have been found to modulate immune responses. They can influence immune cell function and cytokine production, potentially impacting immune-mediated diseases.
Proton Pump Inhibitors (PPIs): PPIs, such as omeprazole and pantoprazole, are commonly used to reduce gastric acid production in conditions like gastroesophageal reflux disease (GERD) and peptic ulcers. Emerging evidence suggests that PPIs might have immunomodulatory effects by influencing immune cell function and altering cytokine profiles. These effects may have implications for inflammatory conditions and certain infections.
It is important to note that the specific immunomodulatory effects of these drugs and their therapeutic implications can vary depending on the disease context and individual patient factors. Further research is needed to fully understand the mechanisms and optimize their use as immunomodulatory agents in different disease conditions.
Immune system modulation as a target for drug discovery
Immune system modulation has emerged as a promising target for drug discovery, offering potential therapeutic opportunities across various disease areas. The immune system plays a critical role in maintaining homeostasis and defending the body against pathogens, but its dysregulation can lead to the development of autoimmune diseases, chronic inflammation, and inadequate immune responses against cancer or infections. Targeting immune system components and processes through drug intervention offers the potential to restore immune balance and enhance disease outcomes. Several approaches have been explored in the quest for immune system modulation.
Cytokine modulation: Cytokines are small proteins that mediate immune cell communication and regulate immune responses. Modulating cytokine activity, such as inhibiting pro-inflammatory cytokines or promoting anti-inflammatory cytokines, can be targeted to treat autoimmune diseases, chronic inflammatory conditions, and certain cancers. For example, monoclonal antibodies targeting tumor necrosis factor-alpha (TNF-α) have been successfully used to treat autoimmune diseases like rheumatoid arthritis and inflammatory bowel disease [67].
Immune checkpoint blockade: Immune checkpoints are molecules that regulate the immune response, preventing excessive activation and maintaining self-tolerance. However, some cancers hijack these checkpoints to evade immune detection. Immune checkpoint inhibitors, such as antibodies targeting programmed cell death protein 1 (PD-1) or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), can release the brakes on the immune system and enhance anti-tumor responses. These inhibitors have shown remarkable efficacy in various cancers, including melanoma, lung cancer, and bladder cancer [68].
Immunomodulatory small molecules: Small molecules can be designed to target specific immune signaling pathways or molecules involved in immune responses. For example, Janus kinase (JAK) inhibitors have been developed to interfere with signaling pathways that drive inflammation in diseases like rheumatoid arthritis and psoriasis [69]. Similarly, inhibitors of Bruton’s tyrosine kinase (BTK) have shown efficacy in treating B-cell malignancies and autoimmune disorders.
Vaccines and adjuvants: Vaccines are an established approach for immunomodulation, training the immune system to recognize and respond to specific pathogens. Advances in vaccine technologies, such as the use of mRNA-based vaccines, have expanded the potential for targeted immune modulation against infectious diseases and cancers [70]. Adjuvants, substances that enhance the immune response to vaccines, can also be targeted for drug development to improve vaccine efficacy.
Cell-based therapies: Cell-based therapies, such as adoptive cell transfer and chimeric antigen receptor (CAR) T-cell therapies, harness the power of the immune system to target and destroy cancer cells. These therapies involve modifying and reprogramming immune cells to specifically recognize and eliminate tumor cells, providing a personalized and highly targeted approach to immune system modulation [71].
The ongoing research and development in immune system modulation present a range of opportunities for novel drug discovery and therapeutic interventions. By targeting key immune components and processes, these approaches hold the potential to transform the treatment landscape for a wide range of diseases.
Advancements in Medicine
Immunomodulatory drugs approved for clinical use
Several immunomodulatory drugs have been approved for clinical use across various disease indications. These drugs have demonstrated efficacy in modulating immune responses and have been shown to provide therapeutic benefits. Here are some examples of immunomodulatory drugs that have been approved for clinical use:
Tumor Necrosis Factor (TNF) Inhibitors: TNF inhibitors, such as adalimumab, infliximab, and etanercept, are approved for the treatment of autoimmune diseases like rheumatoid arthritis, psoriasis, and inflammatory bowel disease. They work by blocking the activity of TNF, a pro-inflammatory cytokine, thereby reducing inflammation and suppressing the immune response [72-73].
Interleukin-1 (IL-1) Inhibitors: IL-1 inhibitors, such as anakinra, canakinumab, and rilonacept, are used to treat conditions such as rheumatoid arthritis, systemic juvenile idiopathic arthritis, and cryopyrin-associated periodic syndromes. These drugs block the activity of IL-1, a pro-inflammatory cytokine implicated in various inflammatory diseases [74-75].
Interleukin-6 (IL-6) Inhibitors: IL-6 inhibitors, including tocilizumab and sarilumab, are approved for the treatment of autoimmune diseases like rheumatoid arthritis and systemic juvenile idiopathic arthritis. These drugs bind to the IL-6 receptor, inhibiting IL-6 signaling and reducing inflammation [76].
B-cell Modulators: B-cell modulators, such as rituximab, ocrelizumab, and belimumab, target B-cells involved in immune responses. They are used in the treatment of conditions such as rheumatoid arthritis, systemic lupus erythematosus, and multiple sclerosis. These drugs deplete or inhibit B-cells, thereby modifying immune responses [77].
Immune Checkpoint Inhibitors: Immune checkpoint inhibitors, such as pembrolizumab, nivolumab, and ipilimumab, have revolutionized cancer treatment. They block inhibitory checkpoints like PD-1 or CTLA-4, thereby enhancing anti-tumor immune responses. These drugs have shown remarkable efficacy in various cancers, including melanoma, lung cancer, and bladder cancer [78-79].
Janus Kinase (JAK) Inhibitors: JAK inhibitors, such as tofacitinib, baricitinib, and ruxolitinib, modulate immune responses by inhibiting JAK enzymes involved in signaling pathways. They are approved for the treatment of autoimmune diseases like rheumatoid arthritis, psoriatic arthritis, and myelofibrosis [80].
These are just a few examples of immunomodulatory drugs approved for clinical use. The field of immunomodulation continues to expand, with ongoing research and development efforts aimed at identifying novel targets and developing new therapies to modulate the immune system effectively.
Immunomodulation as a potential strategy for infectious diseases
Immunomodulation has emerged as a potential strategy for the treatment of infectious diseases. By modulating the immune response, it is possible to enhance the body’s ability to fight off infections or dampen excessive immune reactions that can lead to tissue damage. Here, we discuss the role of immunomodulation in infectious diseases and provide references for further reading:
Cytokine Modulation: Cytokines play a crucial role in coordinating immune responses during infections. Modulating the production or activity of specific cytokines can help regulate the immune response. For example, interferon-γ (IFN-γ) has been used to enhance antimicrobial activity against intracellular pathogens like Mycobacterium tuberculosis [81]. Additionally, blocking excessive production of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) or interleukin-6 (IL-6), has shown promise in reducing cytokine storm-associated tissue damage in severe infections like COVID-19 [82].
Immunomodulatory Agents: Several immunomodulatory agents have been investigated for their potential in infectious diseases. Glucocorticoids, such as dexamethasone, have been used to suppress excessive inflammation in conditions like sepsis and COVID-19 [83]. Immunomodulatory drugs, such as thalidomide and lenalidomide, have shown activity against viral infections, including HIV and hepatitis B [84].
Immune Checkpoint Modulation: Immune checkpoint pathways, like programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), regulate immune responses and prevent excessive tissue damage. Blockade of these checkpoints using monoclonal antibodies, such as pembrolizumab and ipilimumab, has shown efficacy in certain viral infections, such as human papillomavirus (HPV)-related cancers and hepatitis C [85].
Vaccines as Immunomodulators: Vaccines are a form of immunomodulation that elicit specific immune responses against infectious agents. They can induce both humoral and cellular immune responses, protecting against future infections. Vaccines have been successful in preventing a wide range of infectious diseases, including polio, measles, influenza, and COVID-19 [86].
Immunomodulation holds promise as a strategy to enhance host defense mechanisms or mitigate excessive immune responses in infectious diseases. Further research is needed to optimize the use of immunomodulatory approaches for specific infections and to identify novel targets for therapeutic intervention.
Future directions and potential challenges in advancing immunomodulation, drug discovery, and medicine
The field of immunomodulation and drug discovery holds great promise for advancing medicine and treating various diseases. However, there are still several future directions and potential challenges that need to be addressed to fully harness the potential of immunomodulatory therapies. Here are some key areas of focus:
Personalized Immunomodulation: The development of personalized immunomodulatory therapies tailored to individual patients is a promising avenue. Understanding the patient’s immune profile, genetic factors, and disease characteristics could allow for targeted modulation of the immune system. This approach may enhance treatment efficacy and minimize adverse effects [88].
Combination Therapies: Combining immunomodulatory drugs with other therapeutic approaches, such as chemotherapy, radiation, or targeted therapies, holds great potential for improving treatment outcomes. Synergistic effects and overcoming resistance mechanisms can be achieved by combining multiple modalities [89].
Novel Targets and Pathways: Identifying and targeting new immune checkpoints, signaling pathways, or cellular components involved in immune responses can expand the repertoire of immunomodulatory drugs. Advancements in immunology research and high-throughput screening techniques offer opportunities to discover novel targets for therapeutic intervention [90].
Overcoming Resistance: Resistance to immunomodulatory therapies can limit their long-term efficacy. Understanding the mechanisms underlying resistance and developing strategies to overcome it are crucial. Combinatorial approaches, adaptive treatment regimens, and modulation of immune cell interactions are potential strategies to tackle resistance [91].
Safety and Adverse Effects: While immunomodulatory therapies have shown remarkable efficacy, they can also lead to immune-related adverse effects. Further research is needed to better understand and mitigate these side effects. Monitoring and management strategies for immune-related toxicities are essential to ensure patient safety [91].
Regulatory Challenges: As immunomodulatory therapies evolve, there are regulatory challenges to consider. The development of appropriate guidelines, biomarkers, and clinical trial designs specific to immunomodulation is crucial to facilitate drug development and regulatory approvals [92-96].
Cost and Accessibility: The cost of immunomodulatory therapies can pose challenges to their accessibility. Developing cost-effective strategies, improving manufacturing processes, and ensuring equitable access to these therapies are important considerations for the future [97-107].
By addressing these future directions and potential challenges, the field of immunomodulation and drug discovery can continue to advance and revolutionize the treatment of various diseases.
Conclusion
Reflection on the potential impact of immunomodulation and drug discovery on the future of medicine
Immunomodulation and drug discovery have the potential to revolutionize the future of medicine by offering novel treatment approaches and improving patient outcomes. The ability to modulate the immune system opens up new avenues for tackling a wide range of diseases, including autoimmune disorders, cancer, infectious diseases, and inflammatory conditions. The impact of immunomodulation and drug discovery can be seen across several aspects of medicine:
Precision Medicine: Immunomodulatory therapies have the potential to be tailored to individual patients based on their immune profile and disease characteristics. This personalized approach can lead to more targeted and effective treatments, minimizing adverse effects and optimizing outcomes. By understanding the intricate interactions within the immune system, researchers and clinicians can develop therapies that are specifically designed for each patient’s unique needs.
Cancer Treatment: The emergence of immune checkpoint inhibitors and other immunomodulatory agents has revolutionized cancer treatment. These therapies unleash the power of the immune system to recognize and destroy cancer cells. They have demonstrated remarkable success in various types of cancer, offering long-term remission and survival benefits to patients who previously had limited treatment options. The field of immuno-oncology continues to evolve rapidly, with ongoing research focused on identifying new targets and developing combination therapies to enhance responses and overcome resistance.
Infectious Diseases: Immunomodulation can also play a crucial role in combating infectious diseases. By modulating the immune response, researchers aim to enhance the body’s natural defense mechanisms against pathogens. This includes the development of vaccines that stimulate specific immune responses or the use of immunomodulatory drugs to restore immune function in individuals with compromised immunity. Immunomodulation has the potential to improve the prevention, treatment, and management of infectious diseases, contributing to global health efforts.
Autoimmune and Inflammatory Disorders: The development of immunomodulatory drugs has transformed the treatment landscape for autoimmune and inflammatory disorders. By targeting specific components of the immune system, these therapies help regulate immune responses and reduce inflammation. This has led to improved disease control, reduced symptoms, and enhanced quality of life for patients with conditions such as rheumatoid arthritis, psoriasis, inflammatory bowel disease, and multiple sclerosis.
Future Therapeutic Targets: Continued research and drug discovery efforts are uncovering new therapeutic targets and pathways within the immune system. This opens up exciting possibilities for the development of innovative immunomodulatory therapies. By understanding the complex interplay between immune cells, cytokines, and signaling molecules, researchers can identify novel targets for intervention and design drugs with enhanced efficacy and safety profiles.
While there are challenges to overcome, such as resistance, safety concerns, and cost, the potential impact of immunomodulation and drug discovery on the future of medicine cannot be overstated. With ongoing advancements in our understanding of the immune system, combined with technological innovations, collaboration between disciplines, and a patient-centered approach, the field holds great promise for transforming healthcare and improving patient outcomes in the years to come.
Conflict of interest: The authors declare that there is no conflict of interest
References
- Abbas AK, Lichtman AH, Pillai S. Cellular and Molecular Immunology. 9th edition. Philadelphia: Elsevier; 2018.
- Mayer-Barber KD, Andrade BB, Barber DL, et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature. 2014;511(7507):99-103.
- María M, Collado MDC, Llopis F, et al. New therapeutic opportunities targeting the innate immune response during sepsis. Front Cell Infect Microbiol. 2021;11:661567.
- Sterne JAC, Murthy S, Diaz JV, et al. Association between administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: a meta-analysis. JAMA. 2020;324(13):1330-1341.
- The RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384(8):693-704.
- Pilcher CD, Joaki G, Hoffman IF, et al. Amplified transmission of HIV-1: comparison of HIV-1 concentrations in semen and blood during acute and chronic infection. AIDS. 2007;21(13):1723-1730.
- Huang L, Xu X, Li R, et al. The effect of thalidomide on hepatitis B virus replication in vitro and in vivo. AntivirTher. 2015;20(1):55-64.
- Khan MW, Saadalla A, Ewida AH, et al. Personalized Immunotherapy for Cancer: A Novel Classification System Based on Tumor Immunogenomic Profiling. Front Immunol. 2020;11:1442.
- Galluzzi L, Buqué A, Kepp O, et al. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17(2):97-111.
- Swain SL, McKinstry KK, Strutt TM. Expanding roles for CD4(+) T cells in immunity to viruses. Nat Rev Immunol. 2012;12(2):136-148.
- Zugazagoitia J, Guedes C, Ponce S, et al. Current Challenges in Cancer Treatment. ClinTher. 2020;42(4):609-627.
- Johnson DB, Sullivan RJ, Menzies AM. Immune checkpoint inhibitors in challenging populations. Cancer. 2018;124(4):727-737.
- Gomez-Roca C, Michot JM, Stathis A, et al. Innovative clinical trial designs in immuno-oncology: Current challenges and future perspectives. Cancer Treat Rev. 2021;92:102124.
- Doroshow DB, Sanmamed MF, Hastings K, et al. Immunotherapy in non-small cell lung cancer: Facts and hopes. Clin Cancer Res. 2019;25(15):4592-4602.
- Liu J, Hamrouni A, Wolowiec D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-γ and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 2007;110(1):296-304.
- Barrett P, Beasley N, Wang F, et al. A phase II study of an anti-CTLA-4 monoclonal antibody (MDX-010) in metastatic melanoma. Proc Am SocClinOncol. 2002;21:197a.
- Plotkin SA. Vaccines: The Fourth Century. Clin Vaccine Immunol. 2014;21(9):1152-1154.
- Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383(27):2603-2615.
- Smolen JS, Aletaha D, McInnes IB. Rheumatoid arthritis. Lancet. 2016;388(10055):2023-2038.
- Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161(2):205-214.
- Plotkin SA. Correlates of protection induced by vaccination. Clin Vaccine Immunol. 2010;17(7):1055-1065.
- Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119-1131.
- Liao KP, Playford MP, Frits M, et al. The association between reduction in inflammation and changes in lipoprotein levels and HDL cholesterol efflux capacity in rheumatoid arthritis. J Am Heart Assoc. 2018;7(24):e010580.
- Weinblatt ME, Keystone EC, Furst DE, et al. Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum. 2003;48(1):35-45.
- Sandborn WJ, Hanauer SB, Katz S, et al. Etanercept for active Crohn’s disease: a randomized, double-blind, placebo-controlled trial. Gastroenterology. 2001;121(5):1088-1094.
- Griffiths CE, Strober BE, van de Kerkhof P, et al. Comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl J Med. 2010;362(2):118-128.
- De Benedetti F, Brunner HI, Ruperto N, et al. Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med. 2012;367(25):2385-2395.
- Ruperto N, Brunner HI, Quartier P, et al. Two randomized trials of canakinumab in systemic juvenile idiopathic arthritis. N Engl J Med. 2012;367(25):2396-2406.
- Hoffman HM, Throne ML, Amar NJ, et al. Efficacy and safety of rilonacept (interleukin-1 trap) in patients with cryopyrin-associated periodic syndromes: results from two sequential placebo-controlled studies. Arthritis Rheum. 2008;58(8):2443-2452.
- Emery P, Keystone E, Tony HP, et al. IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentrerandomised placebo-controlled trial. Ann Rheum Dis. 2008;67(11):1516-1523.
- Yokota S, Imagawa T, Nishikomori R, et al. Long-term safety and efficacy of tocilizumab, an anti-IL-6 receptor monoclonal antibody, in monotherapy, in children with systemic juvenile idiopathic arthritis (sJIA): 5-year results of the phase 3 clinical trial. Ann Rheum Dis. 2015;74(9): 1690-1699.
- Edwards JC, Szczepanski L, Szechiński J, et al. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med. 2004;350(25):2572-2581.
- Hauser SL, Waubant E, Arnold DL, et al. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358(7):676-688.
- Furie R, Petri M, Zamani O, et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63(12):3918-3930.
- Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364(26):2517-2526.
- Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627-1639.
- Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-723.
- Fleischmann R, Kremer J, Cush J, et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med. 2012;367(6):495-507.
- Kavanaugh A, Kremer J, Ponce L, et al. Filgotinib (GLPG0634/GS-6034), an oral selective JAK1 inhibitor, is effective as monotherapy in patients with active rheumatoid arthritis: results from a randomised, dose-finding study (Darwin 2). Ann Rheum Dis. 2017;76(6):1009-1019.
- Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799-807.
- Nanji AA, Jokelainen K, Tipoe GL, et al. Curcumin prevents alcohol-induced liver disease in rats by inhibiting the expression of NF-kappa B-dependent genes. Am J PhysiolGastrointest Liver Physiol. 2003;284(2):G321-7.
- Rainsford KD, Parke AL, Clifford-Rashotte M, Kean WF. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology. 2015;23(5):231-269.
- Saini R, Saini S, Sharma S. Potential drug targets for prevention and treatment of cancer by aspirin: A narrative review. Indian J Cancer. 2014;51(3): 524-531.
- Opelz G, Döhler B. Lymphomas after solid organ transplantation: a collaborative transplant study report. Am J Transplant. 2004;4(2):222-230.
- Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119-1131.
- Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-264.
- Gonzalez-Cao M, Iduma P, Karachaliou N, et al. Human cancer immunotherapy with PD-1/PD-L1 blockade. Anticancer Drugs. 2015;26(5):427-434.
- Mesa RA, Yasothan U, Kirkpatrick P. Fedratinib. Nat Rev Drug Discov. 2019;18(4):267-268.
- Pulendran B, Ahmed R. Immunological mechanisms of vaccination. Nat Immunol. 2011;12(6):509-517.
- June CH, O’Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):eaar6711.
- Thomson AW, Humar A, Lakkis FG, et al. Immune regulation in transplantation: recent developments and future directions. Am J Transplant. 2021;21(Suppl 2):S1-S10.
- Drews J. Drug discovery: A historical perspective. Science. 2000;287(5460):1960-1964.
- Galluzzi L, Vacchelli E, Eggermont A, et al. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1(5):699-716.
- Akdis CA, Agache I. Global Atlas of Allergic Rhinitis and Chronic Rhinosinusitis. European Academy of Allergy and Clinical Immunology. 2019.
- Simpson EL, Bieber T, Guttman-Yassky E, et al. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N Engl J Med. 2016;375(24):2335-2348.
- Bieber T. Interleukin-13: Targeting an underestimated cytokine in atopic dermatitis. Allergy. 2017;72(6):825-831.
- Bantz SK, Zhu Z, Zheng T. The Atopic March: Progression from Atopic Dermatitis to Allergic Rhinitis and Asthma. J Clin Cell Immunol. 2014;5(2):202.
- Wong SS, Nankervis H. Immunomodulatory therapy for severe atopic dermatitis. Australas J Dermatol. 2019;60(4):e281-e290.
- Cuello-Garcia CA, Brozek JL, Fiocchi A, et al. Probiotics for the prevention of allergy: A systematic review and meta-analysis of randomized controlled trials. J Allergy ClinImmunol. 2015;136(4):952-961.
- Scott LJ. Baricitinib: A review in rheumatoid arthritis. Drugs. 2018;78(8):761-772.
- Abbas AK, Lichtman AH, Pillai S. Cellular and Molecular Immunology. 9th edition. Philadelphia: Elsevier; 2018.
- Bluestone JA, Tang Q. Tolerance and immunotherapy: Mechanisms of immune regulation in autoimmune disease and transplantation. Transplantation. 2011;91(4):377-381.
- Malek TR. The main function of IL-2 is to promote the development of T regulatory cells. J Leukoc Biol. 2003;74(6):961-965.
- Karaman MW, Herrgard S, Treiber DK, et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2018;26(1):127-132.
- Drews J, Böttcher C. Target identification and validation in drug design. Methods Mol Biol. 2015;1263:3-22.
- Kitchen DB, Decornez H, Furr JR, et al. Docking and scoring in virtual screening for drug discovery: methods and applications. Nat Rev Drug Discov. 2004;3(11):935-949.
- Kerns EH, Di L. Drug-like Properties: Concepts, Structure Design and Methods from ADME to Toxicity Optimization. 2nd ed. Academic Press; 2008.
- Arrowsmith J, Miller P. Trial watch: phase II and phase III attrition rates 2011-2012. Nat Rev Drug Discov. 2013;12(8):569.
- Drews J. Drug discovery: A historical perspective. Science. 2000;287(5460):1960-1964.
- Gribbon P, Jones S. A pipeline approach to drug discovery. Drug Discov Today. 2012;17(7-8):303-305.
- Hann MM, Keseru GM, Leeson PD, et al. Why are some targets “undruggable”? Drug Discov Today. 2011;16(7-8):294-299.
- Swinney DC, Anthony J. How were new medicines discovered? Nat Rev Drug Discov. 2011;10(7):507-519.
- Arrowsmith J. Trial watch: Phase III and submission failures: 2007-2010. Nat Rev Drug Discov. 2011;10(2):87.
- Hatsis P, Boyer EW, Sacks CA. Regulatory watch: Post-marketing surveillance of pharmaceuticals. ClinToxicol (Phila). 2016;54(5):345-355.
- Pollard KM, Cauvi DM, Toomey CB, Morris KV, Kono DH. Interferon-gamma and systemic autoimmunity. Discov Med. 2013;16(87):123-131.
- Firestein GS. Immunologic mechanisms in the pathogenesis of rheumatoid arthritis. J ClinRheumatol. 2005;11(3 Suppl):S39-44.
- Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011;365(22):2110-2121.
- Gold R, Wolinsky JS. Pathophysiology of multiple sclerosis and the place of teriflunomide. Acta Neurol Scand. 2011;124(2):75-84.
- Neurath MF. Current and emerging therapeutic targets for IBD. Nat Rev GastroenterolHepatol. 2017;14(5):269-278.
- Morris PJ, Shorthouse R. Immunological principles of immunosuppression in organ transplantation. Surgery (Oxford). 2018;36(6):305-310.
- Webster AC, Woodroffe RC. Handbook of Renal Transplantation. Wiley-Blackwell; 2017.
- Chen W, Liu P, Wang Y, et al. Induction of transplantation tolerance by allogeneic donor-derived CD4+CD25+Foxp3+ regulatory T cells. TransplImmunol. 2018;46:1-6.
- Vincenti F, Larsen C, Durrbach A, et al. Costimulation blockade with belatacept in renal transplantation. N Engl J Med. 2005;353(8):770-781.
- Macarron R, Banks MN, Bojanic D, et al. Impact of high-throughput screening in biomedical research. Nat Rev Drug Discov. 2011;10(3):188-195.
- Hopkins AL, Groom CR. The druggable genome. Nat Rev Drug Discov. 2002;1(9):727-730.
- Swamidass SJ, Azencott CA, Lin TW, et al. Compound activity prediction using models of activity landscapes. J ChemInf Model. 2014;54(2):472-482.
- Harvey AL, Edrada-Ebel R, Quinn RJ. The re-emergence of natural products for drug discovery in the genomics era. Nat Rev Drug Discov. 2015;14(2):111-129.
- Lipinski CA, Hopkins AL. Navigating chemical space for biology and medicine. Nature. 2004;432(7019):855-861.
- Reinders ME, de Fijter JW, Roelofs H, et al. Autologous bone marrow-derived mesenchymal stromal cells for the treatment of allograft rejection after renal transplantation: results of a phase I study. Stem Cells Transl Med. 2013;2(2):107-111.
- Kawai T, Sachs DH. Tolerance induction: hematopoietic chimerism. CurrOpin Organ Transplant. 2014;19(6):581-587.
- Ching T, Himmelstein DS, Beaulieu-Jones BK, et al. Opportunities and obstacles for deep learning in biology and medicine. J R Soc Interface. 2018;15(141):20170387.
- Cortés-Ciriano I, Bender A, Malliavin TE. Artificial intelligence in drug discovery: Hard lessons for these are the world. Drug Discov Today. 2020;25(4):660-665.
- Aliper A, Plis S, Artemov A, et al. Deep Learning Applications for Predicting Pharmacological Properties of Drugs and Drug Repurposing Using Transcriptomic Data. Mol Pharm. 2016;13(7):2524-2530.
- Muegge I, Oloff S. Advances in computational medicinal chemistry and drug design. Methods Mol Biol. 2016;1414:1-18.
- Ginsburg GS, Phillips KA. Precision medicine: From science to value. Health Aff (Millwood). 2018.
- Leventhal JR, Mathew JM, Salomon DR, et al. Genomic biomarkers correlate with HLA-identical renal transplant tolerance. J Am SocNephrol. 2016;27(2):545-555.
- Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-264.
- June CH, O’Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):eaar6711.
- Sackmann EK, Fulton AL, Beebe DJ. The present and future role of microfluidics in biomedical research. Nature. 2014;507(7491):181-189.
- Pantazis P, Supatto W. Advances in whole-organ imaging: a quantitative transition is underway. Nat Rev Mol Cell Biol. 2014;15(5):327-339.
- Swamidass SJ, et al. Trends in high-throughput phenotyping of small molecules. Nat Commun. 2019;10(1):2383.
- Tetko IV, et al. How deep is your screening? A perspective on the deep screening of chemical libraries. ChemSoc Rev. 2020;49(23):8163-8174.
- Zhang Y, et al. DNA-Encoded Dynamic Combinatorial Chemical Libraries for Exploring the Interactions between Chemical Space, Biology, and Drug Discovery. Chem Rev. 2019;119(19):10360-10393.
- Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14(10):1014-1022.
- Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17(13):4550-4557.
- Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer. 2012;12(4):237-251.
- Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004;3(8):673-683.
- Gurwitz D. “Epidemiology of medication-related problems in elderly patients in the community” Eur J ClinPharmacol. 2017;7253(8):993-1006.
- Deeks SG, Lewin SR, Havlir DV. The end of AIDS: HIV infection as a chronic disease. Lancet. 2013;382(9903):1525-1533.