View Article

Abstract

Chrono-modulated drug delivery systems (CDDS) are innovative treatment systems that ensure peak performance with minimal side effects by coordinating drug release with the natural rhythms of the body. Diseases such as diabetes, cancer, hypertension, arthritis, asthma, and cardiovascular ailments that exhibit time-dependent symptomatology greatly benefit from CDDS. In order to dispense medications at optimal times, CDDS employs methodologies such as pulsatile, time-controlled, stimuli-responsive, multi-particulate, and implantable devices. To enhance outcomes and patient adherence, CDDS ensures that maximal drug concentration aligns with maximal disease activity through utilization of circadian fluctuations in pharmacokinetics and pharmacodynamics. Their effectiveness relies on materials such as temperature-responsive gels, pH-sensitive coatings, and biodegradable polymers. To ensure lag duration, release profile, and bioavailability, evaluation involves extensive in vitro, in vivo, and pharmacokinetic/pharmacodynamic analysis. Strong evidence of safety, efficacy, and reproducibility, supported by stability studies and in vitro–in vivo correlation, is demanded by regulatory bodies like the FDA and EMA. As wearable biosensors, personalized medicine, and chrono-pharmacology advance, CDDS offer a promising future for time-dependent, tailored treatment in preventive and chronic care.

Keywords

Circadian rhythm, Chrono-modulated drug delivery, Pulsatile release, Time-controlled release, stimuli- responsive system, Chrono-pharmacology

Introduction

Chronotherapy is a treatment method aimed at optimizing the efficacy of drugs and reducing side effects by coordinating drug administration with the body's natural biological rhythms, most notably the circadian rhythm. The suprachiasmatic nucleus of the hypothalamus controls 24-hour rhythms of physiological functions, including the release of hormones, sleep-wake cycles, fluctuations in blood pressure, and control of body temperature. The life cycles have a profound effect on the pharmacokinetics (absorption, distribution, metabolism, excretion) and pharmacodynamics (drug effect) of most drugs.  Accordingly, chronotherapy takes advantage of the circadian variations in disease manifestation and drug activity to improve therapeutic benefits.  During the day, the severity and manifestation of several conditions like cancer, rheumatoid arthritis, asthma, hypertension, and stomach ulcers change.   For example, asthma attacks are typically most common in the early morning, whereas blood pressure increases in the morning and reduces during the night.  Likewise, the symptoms of osteoarthritis typically increase over time.  Traditional drug delivery systems provide a consistent release of the drug in spite of these changes. In contrast, chrono-modulated drug delivery systems seek to maximize therapeutic effectiveness and reduce toxicity by delivering drugs when they are needed or most effective. In cardiovascular illness, where there are daily fluctuations that affect both the presentation of the disease and the efficacy of treatments, chronotherapy is important. For example, early-morning physiological changes, like elevated cortisol levels and augmented platelet aggregability, increase the risk of myocardial infarction and stroke. Research has indicated that synchronizing the administration of antihypertensive or antiplatelet drugs with these physiological events improves patient outcomes and minimizes mortality. [1,2] Tumor growth and medication metabolism in oncology tend to have predictable patterns. Because of the varying sensitivity of normal cells and malignant cells at different circadian states, chemotherapy medication administration at specific times of day can maximize efficacy and decrease toxicity. [3] Chrono-chemotherapy, or the synchronization of chemotherapy regimens with the patient's biological rhythm, is based on this concept. Diabetes management is another area that benefits from chronotherapy.  Because insulin sensitivity and glucose metabolism follow circadian patterns, timed dosing throughout the day is a potential way of improving glycemic control. [4] Based on a person's personal glucose tolerance, chronotherapeutic interventions for diabetes can consist of insulin or other antidiabetic medication delivery in either the morning or evening. At the pharmaceutical level, the creation of chrono-modulated drug delivery systems, i.e., programmable infusion pumps, time-dependent coatings, and pulsatile release tablets, has made it possible to deliver drugs with precision according to the body's biological rhythms. By providing for drug release following a given time lag, these methods ensure that the peak plasma concentration occurs at the same time as the peak activity of the disease. Chronotherapy is of utmost importance because it improves the quality of life, minimizes side effects, facilitates patient compliance, and delivers tailored treatment regimens. Chronotherapy is a promising technique to improve treatment efficacy while limiting excessive exposure to medication, especially as personalized medicine advances. As human chronobiology and medication chrono-pharmacology research continues, time-based therapy is likely to play an increasingly important role in contemporary medicine. [5]

Concept of Chrono – Modulated Drug Delivery System

Sophisticated therapeutic systems known as chrono-modulated drug delivery systems (CDDS) have been crafted to release drugs in harmony with the circadian patterns of the body. The main aim of CDDS is to deliver drugs at particular times during the day or after a pre-determined lag time, as per the physiological clock of the body and the circadian rhythms of diseases, as opposed to conventional drug delivery systems that release the drug continuously or in an immediate manner. This planned timing counteracts adverse effects when the body is more susceptible or less responsive and makes the medicine realize its optimal healing effect when most needed. Chronobiology, the study of the temporal mechanisms of biological processes, forms the basis for this phenomenon.  The majority of physiological processes within the body, such as hormone release, enzyme activity, gastric emptying, blood pressure regulation, and sensitivity to pain, are regulated by the circadian clock. The drug's pharmacokinetics (absorption, distribution, metabolism, and excretion) and pharmacodynamics (toxicity and therapeutic activity) are also affected by these rhythmic changes. For example, changes in renal clearance, gastric acidity, and liver enzyme activity during the day will change drug metabolism and bioavailability.  Giving a medication at the wrong time can cause toxicity or suboptimal therapeutic levels when the body is either more susceptible or less sensitive. [1] Chrono-modulated system is most useful for diseases that have varying symptoms throughout a span of time. For instance, blood pressure rises in the morning and falls at night, while asthma can worsen in the early morning hours. By creating delivery systems that deliver bronchodilators or antihypertensives according to these patterns, efficacy can be improved and the potential for complications can be minimized. [5] To set up CDDS, a number of technological strategies have been developed, including:

  • Pulsatile release systems: These are systems that release drugs in short periods, followed by some time of delay. They are especially appropriate for conditions like nocturnal asthma or arthritis where drugs must become effective at specific times.
  • Time-controlled systems time a delay in drug release to the right moment, using coatings or barriers that break down or dissolve progressively over time.
  • Formulations that react to internal cues, like pH, temperature, or enzyme concentration—which also change in synchrony with circadian rhythms—release drugs.
  • Programmable devices: Implantable injectors and electronic drug pumps facilitate accurate chrono-modulated administration, since they can be programmed to release drugs at specific times.

Another critical factor is patient-friendly design.  The preparation, for example, must be convenient and not need complicated storage or manipulation.  Oral controlled medicine delivery systems, such as modified tablets or capsules, are the most common because they are convenient to use.  However, transdermal, parenteral, and implanted chrono-delivery devices are becoming more common, particularly in chronic illnesses and oncology where timing of drug delivery is important. To optimize such systems, chrono-pharmacological research is also essential.  By researching the day-to-day changes in drug response, scientists can find the optimal chronopharmacokinetic profile of a medication and condition.  Through the combination of chronotherapy and controlled release technology, it is possible that therapeutic effects can be enhanced, dosing frequency lowered, and toxicity minimized. [2]

Chronobiology and Circadian Rhythms

Basics of Biological Clocks

Biological clocks, or internal timing mechanisms, control physiological and behavioral events in living organisms on a regular and predictable basis.  The most widely studied biological rhythm, the circadian rhythm, lasts around 24 hours and regulates hormone secretion, body temperature, metabolism, sleep-wake patterns, and several other vital processes. The main pacemaker of the circadian clock in animals is within the hypothalamic suprachiasmatic nucleus (SCN).  The retina provides nerve impulses to the SCN, which is entrained by the ambient light-dark cycle.  Through the transmission of neurological and endocrine signals that control rhythmic gene expression and cellular function, it synchronizes the peripheral clocks found within various tissues and organs, including the liver, heart, lungs, and kidneys. [6] The molecular basis for biological clocks relies on the transcriptional-translational feedback loops (TTFLs), which involve the core clock genes such as CLOCK, BMAL1, PER (PER1 and PER2), and CRY (CRY1 and CRY2).  Through proteins used by the feedback mechanism, these genes generate 24-hour cycles that, depending on how time has progressed, either stimulate or suppress their own transcription. [7] Disruptions in this clock system can lead to mood swings, metabolic issues, and an increased likelihood of developing chronic diseases such as diabetes and cancer. In addition to the circadian rhythm, biological clocks also govern the circalunar/circannual, ultradian (shorter than 24 hours), and infra-dian (longer than 24 hours) rhythms, which affect, for example, menstrual cycles, pattern of eating, and seasonal affective behaviors. But for its profound influence on drug absorption, metabolism, and efficacy, the circadian rhythm is the most crucial in pharmacology. [8] Environmental cues known as zeitgebers (German for “time givers”), such as light, temperature, food intake, and social interactions, are essential for entraining the biological clock to the external environment. Among these, light is the most powerful zeitgeber for humans. When the SCN is entrained by light via retinal input, it helps maintain synchrony between internal rhythms and external time, a process essential for optimal physiological functioning [9] The examination of these cycles and their role in disease and health is the area of chronobiology. These consequences of disrupted circadian rhythm have been exhibited by various studies, including impaired glucose metabolism, sleep disorders, cardiovascular diseases, and changed pharmacological responses to drugs [10]. This knowledge has driven the emergence of chrono-medicine, which aims to tailor treatment timing based on individual biological rhythms. Notably, studies have found that biological clocks are not fixed and can be changed or reset, for instance, by working night shifts, suffering from jet lag, or being exposed to artificial light at night.  Circadian misalignment caused by such changes has been linked to increased susceptibility to obesity, metabolic syndrome, and many types of cancer. [11] Scientists have discovered new clock-regulated genes involved in diverse physiological activities, owing to progress made in molecular biology and genetics, which has improved their understanding of the complex mechanisms of the clock.  This has enabled scientists to design chrono-modulated drug delivery systems, the goal of which is to time the delivery of drugs according to the natural rhythms of the body for improved therapeutic effects.

Role of Circadian Rhythms in Disease Progression and Treatment

Circadian rhythms, or 24-hour cycles in the body, are very important in controlling the timing of essential processes including secretion of hormones, metabolism, the immune system, and cellular maintenance. The suprachiasmatic nucleus (SCN) regulates these biological rhythms, synchronizing them with external signals such as temperature and light. More and more evidence indicates that onset, development, as well as responses to treatment of many diseases, such as inflammatory, cardiovascular, metabolic, and neoplastic conditions, are closely related to distortions or misalignments in circadian rhythms.

1. Cardiovascular Diseases

There are profound diurnal fluctuations in cardiovascular measurements, such as blood pressure, heart rate, and vascular tone. A high risk of acute cardiovascular events, including myocardial infarction and stroke, is typically associated with a morning increase in heart rate and blood pressure. This is thought to be a result of increased sympathetic activity, augmented cortisol release, and increased platelet aggregability in the early morning hours. [13] To maximize therapeutic outcomes and reduce adverse reactions, antihypertensive agents (such as beta-blockers or ACE inhibitors) should be administered at intervals consonant with these fluctuations. Studies have shown that evening administration of some antihypertensives reduces morning peaks and improves 24-hour blood pressure control. [14]

2. Metabolic Disorders

Circadian rhythms affect insulin sensitivity and glucose metabolism.  Important clock genes like CLOCK, BMAL1, PER, and CRY are vital in the control of metabolic activities in the liver, pancreas, and fat cells.  Abolition of these genes can cause fat deposition, elevated glucose levels, and impaired insulin secretion. Those with shift work or non-standard sleep schedules are at a much greater risk of developing type 2 diabetes, obesity, and metabolic syndrome because of chronic circadian desynchronization.  In these cases, chronotherapy is done to coordinate antidiabetic drugs and meal times with the body's metabolic rhythms in order to improve glycemic control. [15]

3. Cancer

Cell cycle, DNA repair, and apoptosis—important processes in cancer initiation and progression—are themselves subject to circadian control.  Circadian clock disruption has been linked with increased carcinogenesis and modified treatment response in a number of cancers.  Cancer cells can either maintain or alter circadian regulation, and this could impact their ability to proliferate and respond to therapy. Through the use of chronotherapy, or the delivery of chemotherapy or radiation therapy at the most favorable times, healthy tissue damage can be minimized while cancer cells are best targeted when in their most vulnerable state.  For example, some chemotherapeutic drugs exhibit increased efficacy and diminished toxicity when given during certain stages of the circadian cycle [16].

4. Inflammatory and Autoimmune Disorders

Circadian modulation is also observed in the immune system.  Leukocyte traffic, immune cell activation, and circulating levels of cytokines vary daily.  Patients with diseases like rheumatoid arthritis tend to report that the symptoms are worse in the morning, at the time of the highest circulating levels of such pro-inflammatory cytokines as TNF-α and IL-6. Using altered-release preparations or taking corticosteroids at bedtime can help relieve morning inflammation and enhance the control of symptoms.

5. Neurodegenerative and Psychiatric Disorders

Bipolar disorder, schizophrenia, depression, and Alzheimer's disease are all noted for their disruption of circadian rhythms.  In such instances, sleep-wake cycles, melatonin secretion, and expression of clock genes are usually disrupted.  Phase delay and reduced circadian amplitude, for instance, are linked with poor treatment outcomes in depression.  To promote mood control and restore circadian coordination, chronotherapy in psychiatry can encompass the timed delivery of antidepressants, light exposure, or resynchronization of sleep phases. [17]

Rationale for Chrono-Modulated Drug Delivery

The developing awareness that the toxicity and efficacy of drugs can vary considerably depending on the time when they are administered provide the basis for the creation of chrono-modulated drug delivery systems, also known as CDDS.  Their variation mainly depends on circadian rhythms, that is, biological cycles ruling various physiological processes on a 24-hour basis. These cycles will have an effect on drug absorption, distribution, metabolism, and excretion (ADME), and even on the cellular reaction to drugs.  As a result, conventional drug delivery methods that ignore these biological oscillations will cause ineffective therapeutic outcomes or worsening of side effects.

Chrono-pharmacology

The discipline of chrono-pharmacology examines how drug administration timing affects the pharmacodynamics and pharmacokinetics of drugs.  By combining the concepts of chronobiology with pharmaceutical sciences, it offers a scientific rationale for the administration of time-modulated medications.  Chrono-pharmacology acknowledges that drug actions in the body, as well as the body's metabolism of the drug, change over a daily time period according to internal biological cycles. Circadian variation affects many of the physiological processes, such as hepatic metabolism, renal clearance, gastric acidity, and enzyme activity. A drug that is metabolized quickly by the liver in the morning might necessitate a different dosing regimen, whereas a drug that is best absorbed under conditions of acidity will be better administered in the evening. Chrono-specific pharmacodynamics occur due to the variability in the sensitivity of target tissue to drugs, e.g., blood vessels to antihypertensives or cancer cells to chemotherapy, that might be time-dependent [18].

By applying chrono-pharmacological knowledge, CDDS aim to:

  • By matching the peak drug levels with the peak disease activity, therapy effects can be maximized.
  • Prevent drug release during times of increased sensitivity or low metabolism to reduce toxicity.
  • Improve patient compliance by decreasing the frequency of doses and better symptom control.

Evidence from Disease Models

Airway obstruction in asthmatic patients is higher at night, usually peaking early in the morning.  To prevent nocturnal attacks and delay the onset of symptoms, evening use of bronchodilators through a device that delivers the medication in the morning is recommended. In addition, the necessity of antihypertensive drugs that release their active components in the early morning hours is highlighted by the fact that the highest rate of cardiovascular disease events, such as myocardial infarction, occurs between 6 a.m. and noon [19]. In cases of arthritis, where joint stiffness and pain are most severe upon awakening in the morning, chrono-modulated delivery is especially useful. Altered-release corticosteroids or NSAIDs that release their active ingredients in the morning provide enhanced relief from symptoms and improve patient comfort [20]. Also, certain chemotherapeutic agents exhibit time-dependent differences in both their toxic and therapeutic effects in the area of oncology.  Patient tolerance and the results of treatment can be greatly enhanced by giving such drugs during periods when cancer cells are most sensitive and normal cells are least susceptible. [21]

Time-Dependent Variation in Drug ADME

The day-to-day variability of ADME processes according to the time of day is a strong argument for chrono-modulated drug delivery systems.  These pharmacokinetic variables change over the course of the day according to the circadian rhythm of the body, as opposed to remaining fixed.  These variations have very significant impacts on medication safety and therapeutic efficacy, and thus it is critical to time drug administration according to the biological clock for maximum benefit.

Absorption

Circadian fluctuations in intestinal motility, pH, blood flow, and enzyme activity cause variations in drug absorption during the 24 hours. The delayed gastric emptying at night and the increased transit in the morning affect Cmax and Tmax. Lipid-based and pH-sensitive drugs, depending on their administration timing, exhibit varying bioavailability. [22]

Distribution

Circadian rhythms are also found in the binding of proteins in plasma, which is a critical factor in the distribution of drugs. Acidic and basic drug-binding proteins like albumin and α1-acid glycoprotein show fluctuating levels during the day. These changes affect the amount of unbound (active) drug in plasma, thus modifying its effect and toxicity. [23]

Metabolism

Circadian genes, particularly those that encode CYP450 enzymes, are a key part of drug metabolism regulation in the liver. The enzymatic activity of these enzymes in humans is found to increase during the daytime and fall during the night. These circadian rhythms affect half-life, metabolite synthesis, and the elimination of drugs. Thus, the same dose can yield various pharmacological effects depending on the time of administration. [24]

Excretion

Circadian rhythms also affect renal functions, i.e., blood flow, glomerular filtration rate (GFR), and tubular activity, which consequently influences the clearance of drugs and metabolites. Drugs that are cleared by the kidneys, e.g., aminoglycosides and antihypertensives, are cleared faster during the day. This difference has drug accumulation and toxicity profiles implications. Thus, the dosage timing is crucial in chronotherapy. [25]

Clinical Relevance

A critical component of the chronotherapeutic strategy is the incorporation of such ADME variation into medication delivery planning. Sustained-release or pulsatile-release products, for example, might significantly enhance therapeutic effect by synchronizing drug release with peak metabolic rate or symptom onset. Additionally, to address interindividual variation in circadian timing and ADME profiles, personalized medicine techniques such as wearable biosensors and chronopharmacokinetic modeling are under development [26].

Diseases Benefiting from Chrono-Modulated Drug Delivery

The goal of CDDS is to time drug delivery in correspondence with the body's own internal biological rhythms, especially the circadian clock that governs vital physiological processes over a 24-hour time period. The development of chronotherapy a treatment approach that synchronizes healing with timing into biology to maximize therapeutic benefit and reduce side effects has been prompted by the recognition and utilization of these time-dependent patterns. Major diseases like asthma, hypertension, arthritis, cancer, diabetes, and cardiovascular disease that are known to benefit from chrono-modulated treatment are listed below in the table. The table reports the pertinent circadian rhythms in the disease pathophysiology of each disease and presents the mechanisms through which better drug scheduling can promote therapeutic responses. In the case of chronic and rhythm-dependent disorders, this method forms the basis for creating time-specific, targeted treatment plans that can significantly enhance patient treatment.

Table: Chronobiological Patterns of Major Disease and Corresponding Chrono-Based Therapeutic Strategies

Disease

Chronobiological observation

Chrono-Based Therapeutic Approach

References

 

Asthma

Airway resistance increases during night/early morning

Administering corticosteroids and bronchodilators either in the morning or at night aids in alleviating symptoms.

Selfridge JM et al., 2016 [27]

Hypertension

Blood pressure surges in early morning hours

Antihypertensive medications taken in the evening or prior to bedtime more effectively control the early morning surge.

Kario K, 2010 [28]

Arthritis

Cytokines and stiffness peak in the morning

Corticosteroids or NSAIDs with modified release that begin to take effect in the morning

Buttgereit F et al., 2008

Cancer

Cell proliferation and drug metabolism show circadian rhythms

Time-specific chemotherapy reduces toxicity while enhancing effectiveness.

Giacchetti S et al., 2006

Diabetes

Insulin sensitivity varies; lowest in evening

Glycemic control can be enhanced by the morning administration of insulin sensitizers or by modifying the timing.

Boden G et al., 1996 [31]

Cardio-vascular disease

Myocardial infarctions and strokes peak in early morning

Administering beta-blockers or statins during the night as a component of chronotherapy reduces the occurrence of cardiovascular events in the morning.

White WB et al., 2011 [32]

Principles of Chrono-Modulated Drug Delivery Systems

Through synchronizing the release profiles of drugs with the body's natural biological rhythms, chrono-modulated drug delivery systems (CDDS) offer a novel method for the improvement of therapeutic efficacy. The main objective is to improve efficacy and reduce side effects by synchronizing drug levels at the target site with the circadian rhythm of the disease or physiological activity.

Synchronizing Drug Release with Biological Rhythms

Chrono-modulated drug delivery systems (CDDS) are designed to optimize therapeutic outcomes by aligning drug release profiles with the body’s intrinsic biological rhythms. These systems aim to synchronize the timing of drug availability with circadian variations in disease symptoms and physiological processes, thus maximizing efficacy and minimizing side effects [33,34]. Biological functions such as hormone secretion, enzyme activity, gastrointestinal motility, and cardiovascular parameters follow circadian rhythms. These rhythms also affect the pharmacokinetics and pharmacodynamics of many drugs. For instance, conditions like asthma, arthritis, and hypertension exhibit time-dependent symptom severity, often peaking at specific times of day or night [35]. To address these variations, CDDS utilize formulations like pulsatile systems, which release drugs after a lag phase, time-controlled systems that ensure drug availability during periods of peak need, or stimuli-responsive systems that trigger drug release based on circadian-related changes in physiological conditions like pH or enzyme levels. [34,36] A crucial step in designing these systems is understanding the chronobiology of the disease, as well as the drug’s pharmacological profile. For example, cardiovascular events such as myocardial infarction often peak in the early morning; thus, administering antihypertensives at bedtime improves outcomes. By delivering drugs in harmony with the body's biological rhythms, CDDS not only improve therapeutic success but also enhance patient adherence and reduce dosing frequency, offering a promising strategy for managing chronic and circadian-influenced disorders [37]

Types of Chrono-Modulated Drug Delivery Systems

Sophisticated preparations called CDDS are designed to schedule drug release to coincide with body’s biological rhythms, particularly the circadian variation in disease activity. These systems enhance patient compliance, reduce side effects, and enhance treatment effectiveness by synchronizing pharmaceutical action with symptom exacerbation onset. Time-regulated, stimulus-responsive, pulsatile, multi-particulate and implantable systems are some of the primary types of CDDS illustrated by the figure. Every strategy employs a distinct mechanism to enhance drug release profiles for various disease such as cancer, cardiovascular disease, arthritis and asthma. The methodologies vary from physiological trigger reaction to lag time management by polymer. This classification illustrates the versatility of CDDS in managing time-dependent pathophysiology with tailored drug delivery methods.

Figure 1: Types of Chrono – Modulated Drug Delivery System

  1. Time –Controlled System:

These systems are designed to release the drug following a predefined lag time, aligning with circadian variations in disease activity.

  • Mechanism: Use of hydrophilic or erodible polymers that delay release
  • Application: Asthma, hypertension, and arthritis with morning symptom exacerbation.
  1. Stimuli – Responsive System

These respond to physiological triggers such as pH, temperature, or enzymatic activity to control the release.

  • Temperature-sensitive: Gel or polymer systems release drug upon temperature change. Used for colon-targeted drug delivery in conditions like ulcerative colitis.
  • pH-sensitive: Useful in targeting the gastrointestinal tract. It delivers anticancer drugs to tumor tissues exhibiting higher local temperatures.
  • Enzyme-sensitive: Designed for release in the colon or inflammatory sites. They are employed in arthritis to release corticosteroids at inflamed joints with elevated protease activity.
  1. Pulsatile Drug Delivery System:

These systems deliver the drug in one or more pulses after a lag time, matching the circadian peak of disease symptoms.

  • Mechanism: Rupturable coatings, osmotic pumps, and programmed polymers
  • Application: Cortisol therapy, cardiovascular conditions, and asthma. [38]
  1. Multi - particulate Systems

These use pellets or mini-tablets with variable coatings to deliver drugs at different times or locations in the GI tract.

  • Mechanism: Differentially coated units within a single formulation
  • Application: Suited for diseases with complex dosing schedules.
  1. Implantable Systems

These are inserted into the body for sustained or programmable drug release over weeks to months.

  • Mechanism: Biodegradable polymers or electronic-controlled pumps
  • Application: Hormone therapy, cancer, or cardiovascular drug administration. [39]

Materials Used in Chrono-Modulated Systems

Chrono-modulated drug delivery systems (CDDS) need to undergo thorough testing via various methods, including in vitro and in vivo experiments, pharmacokinetic/pharmacodynamic evaluations, as well as additional studies like stability testing, gamma scintigraphy, determination of lag time, FTIR, and muco-adhesion tests, in order to establish their ability to deliver drugs in accordance with the body's circadian rhythm. The mind map gives the major categories of materials used in CDDS, such as lipids, hydrophilic and pH-sensitive polymers, osmotic agents, temperature-sensitive polymers, coating materials, and enzyme-degradable biopolymers.Each material has a specific function, whether it is addressing intestinal problems, giving delayed release, or creating new, responsive formulations.In order to prepare effective chrono-pharmaceuticals, understanding the role and mechanism of action of each material is necessary. [40]

Figure 2: Materials used in CDDS with their functions

Evaluation and Characterization

In order to make sure that chrono-modulated drug delivery systems (CDDS) safely deliver medications in accordance with the circadian rhythms of the body, it is important to extensively evaluate their performance using a variety of methods such as in vitro, in vivo, pharmacokinetic/pharmacodynamic assessments, and additional tests such as stability studies, gamma scintigraphy, lag time determination studies, FTIR, and muco-adhesion studies.

  1. In vitro evaluation

In vitro studies simulate physiological conditions to evaluate the drug release profile of the formulation.

  • Dissolution testing: Used to quantify the lag phase and release kinetics as a function of time employing USP apparatus (I, II, or IV).
  • Coating integrity: Guarantees that stimuli-responsive or time-release coatings perform as intended in gastrointestinal simulations.
  • Swelling and erosion studies: For the study of hydrophilic matrix systems behavior over time.
  • pH and enzyme sensitivity testing: Essential for systems that mimic intestinal or colonic conditions and are pH-sensitive and enzyme-degradable. [41]
  1. In vivo evaluation

In vivo studies in animal models or humans assess the formulation’s physiological performance.

  • The measurement of the real delay prior to the release of a drug after it is administered is known as "lag time confirmation."
  • Tissue targeting: This is particularly necessary for circadian rhythm-sensitive tumors or for devices to be used for colon targeting.
  • Biological rhythm synchronization: This guarantees that the delivery of medication occurs in harmony with the circadian peaks corresponding to the disease.
  1. PK and PD evaluations:

Chrono-modulated systems require time-sensitive PK/PD evaluations to demonstrate that drug action correlates with disease rhythms.

  • Pharmacokinetic parameters are analyzed according to different dosing intervals, which include Cmax, Tmax, AUC, and half-life.
  • The influence of the circadian phase on ADME (absorption, distribution, metabolism, and excretion) is assessed through chrono-pharmacokinetics.
  • Pharmacodynamic studies: Assess the therapeutic response at various circadian phases (for instance, pain relief, reduction in blood pressure).
  • Bioavailability assessments: Confirm that the desired pharmacological properties are achieved through pulsatile or delayed release. [42, 38]

Regulatory Considerations

  • FDA and EMA perspectives

The European Medicines Agency (EMA) and the United States Food and Drug Administration (FDA) have to give serious thought to the special regulatory issues that are created by CDDS.  Both highlight the importance of thoroughly characterizing these systems from a regulatory point of view, with respect to safety, therapeutic efficacy, pharmacokinetics (PK), and pharmacodynamics (PD). The manufacturers of CDDS must prove predetermined drug release kinetics, uniform control of lag time, and bioequivalence to traditional dosage forms (whenever applicable).  Time-dependent PK profiling and full in vitro–in vivo correlation (IVIVC) testing is also required to determine whether or not the system releases the medication at the right point in the circadian cycle. Analogous specifications apply in the EMA's context to the quality, safety, and efficacy modules of the Common Technical Document (CTD).  However, personal and precision-based chronotherapy is becoming increasingly important in regulatory systems.  In order to maximize therapeutic benefits, both agencies necessitate carefully planned clinical trials, particularly for such diseases as cancer, rheumatoid arthritis, asthma, and hypertension that have circadian variability. For example, when evaluated by benefit-risk assessment committees, CDDS should prove better or at least equal treatment effects with fewer adverse effects than with the traditional dosage forms.  It is necessary to discuss relevant manufacturing parameters like uniformity of coating, release-affecting excipients, and stability under different environmental conditions in regulatory filings.  According to ICH guidelines, time-dependent release mechanisms (such as pulsatile or programmed release) must be validated using both accelerated and long-term stability testing. As far as labelling is concerned, the FDA regulates that patients are provided with clear instructions regarding the timing of the intake of medication in accordance with their sleep-wake cycle. This has special relevance for chrono-delivery systems.  For Chrono-Delivery Drug Systems (CDDS), regulatory bodies such as the FDA and EMA demand strong evidence of uniformity, therapeutic effectiveness, and safety for patients. To gain market approval, developers need to incorporate chrono-biological considerations into clinical trial design, manufacturing procedures, and labelling. Early discussion with regulatory authorities through scientific advice procedures is advisable as regulatory frameworks adapt to the incorporation of chrono - therapeutics. [43, 44]

  • Guidelines for evaluation of modified release systems

Chrono-modulated drug delivery systems (CDDS), a class of altered release (MR) formulations that include pulsatile-release and delayed-release systems, are subject to tight regulations set by international regulating bodies, such as the European Medicines Agency (EMA) and the U.S. Food and Drug Administration (FDA). The FDA offers extensive guidance for the assessment of modified release solid oral dosage forms in publications like the SUPAC-MR (Scale-Up and Post-Approval Changes for Modified Release) and general bioavailability/bioequivalence guidance. To meet such standards, CDDS must show uniform dissolution testing over a range of physiological pH levels to meet gastrointestinal transit and circadian changes in drug absorption, be able to sustain a good in vitro–in vivo correlation (IVIVC), and provide a consistent lag-time in the release of the drug. Similarly, the EMA requires evidence of pharmacokinetic consistency, particularly in cases where a chronotherapeutic benefit is claimed over traditional administration, as specified in its pharmacokinetic and clinical evaluation of modified release dosage forms.  In addition, the EMA places importance on having to determine therapeutic equivalency in formulating new formulations and in assessing the effect of circadian rhythm on the efficacy and safety of the drug. [45] In addition, the target time of dosing (e.g., bedtime or morning rise) should be explicitly defined during CDDS design and labeling, and it must be supported by pharmacodynamics rationale. Regulatory evaluations also look at whether the system results in enhanced treatment outcomes or minimizes side effects in accordance with the circadian pathophysiology of the conditions like rheumatoid arthritis, asthma, and hypertension. [35]

SUMMARY

A remarkable improvement in pharmaceutical science, chrono-modulated drug delivery systems (CDDS) are specific to coordinate drug release with the natural biological rhythms of the body, most notably circadian rhythms. These systems are notably helpful in controlling conditions whose symptoms or physiological occurrences have a regular daily cycle, i.e., asthma, hypertension, arthritis, cancer, and cardiovascular diseases. In contrast to the conventional drug-delivery systems that deliver drugs at a constant flow, CDDS has the goal of delivering medications at best times, when the patient's symptoms are worst or the body is most responsive. It maximizes therapeutic effect and minimizes side effects. To enable the accurate control of the timing of medication release, several technologies have been developed, such as time-controlled release systems, pulsatile systems, implantable devices, nanotechnology, and stimuli-sensitive platforms (i.e., pH, temperature, and enzyme-based systems). Pharmacokinetic/ pharmacodynamic (PK/PD) evaluations, in vitro tests (e.g., dissolution and coating integrity), and in vivo experiments (such as lag time and rhythmic drug release) are essential parts of Controlled Drug Delivery Systems (CDDS) evaluation. In order to achieve timed release, it is important to use materials such as temperature-sensitive gels, biodegradable polymers, osmotic agents, and pH-sensitive polymers. Regulatory-wise, the FDA and EMA demand that these systems demonstrate strict bioequivalence, uniform lag time, therapeutic benefits, and safety.  The most important areas of regulatory advice are modified release evaluation methods, stability tests, IVIVC information, and circadian-based labeling methodologies.  As personalized medicine becomes more common, CDDS are expected to become increasingly sophisticated, patient-specific solutions that are completely integrated into treatment regimens for rhythm-sensitive diseases.

CONCLUSION

CDDS development represents a significant advance in synchronizing medication with the body's biological rhythms.  By synchronizing drug release with peaks in disease activity on a circadian basis, they ensure optimum therapeutic effect.  By reducing unnecessary exposure during periods when there is minimal therapeutic requirement, this fine-tuned timing reduces adverse effects. Uses range widely across diseases, including diabetes, hypertension, cancer, arthritis, asthma, and cardiovascular disease.  Precise release scheduling is facilitated by technologies such as implantable systems, pulsatile, time-controlled, multi-particulate, and stimuli-responsive systems.  Performance is significantly impacted by the material of choice, such as temperature-responsive gels, pH-sensitive coatings, and biodegradable polymers.  Pharmacokinetic and pharmacodynamic assessments confirm that CDDS can improve patient compliance and symptom control. Robust evidence of safety, efficacy, uniform lag time, and circadian-based effects are expected by regulatory bodies like the FDA and EMA.  Regulatory approval requires stability testing and in vitro–in vivo correlation (IVIVC).  One must understand chronobiology, disease pathophysiology, and patient lifestyle factors to develop an effective CDDS. New advances in chrono-pharmacology continue to refine the timing of medicines for maximum benefit.  Wearable biosensors and other personalized medicine technologies may be combined to further individualize treatment to each patient's personal rhythms. CDDS are advantageous in that they reduce dose frequency, which enhances compliance in chronic disease treatment.  These systems maximize therapeutic effect and reduce toxicity by synchronizing drug availability with biological needs.  They can also be employed in prevention, particularly in cardiovascular events with high-risk, so that necessary precautions may be taken in a timely manner.  In cancer treatment, chronotherapy can maintain healthy tissue while exploiting tumour windows of sensitivity. The business is moving towards more individualized, patient-specific drug delivery systems.  AI release mechanisms and circadian monitoring in real time are just a couple of examples of possible future developments.  Overall, CDDS represent a blend of chronobiology and drug technology that could change the face of drug therapy.  Moving forward, safer, more effective, and more customized healthcare interventions are on the horizon.

REFERENCE

  1. Smolensky MH, Hermida RC, Reinberg A, Sackett-Lundeen L, Portaluppi F. Circadian rhythms in medicine: implications for temporal drug delivery. J Control Release. 2016 Jan 10; 216:125–43.
  2.  Lemmer B. Chronopharmacokinetics: implications for drug treatment. J Pharm Pharmacol. 1999 Aug;51(8):887–90.
  3.  Lévi F, Okyar A, Dulong S, Innominato PF, Clairambault J. Circadian timing in cancer treatments. Annu Rev Pharmacol Toxicol. 2010; 50:377–421.
  4. Gale JE, Cox HI, Qian J, Block GD, Colwell CS, Matveyenko AV. Disruption of circadian rhythms accelerates development of diabetes through pancreatic beta-cell loss and dysfunction. J Biol Rhythms. 2011 Oct;26(5):423–33.
  5. Youan BB. Chronopharmaceutical drug delivery systems: Hurdles, hype or hope? Adv Drug Deliv Rev. 2010 Apr 30;62(9–10):898–903.
  6. Mohawk JA, Green CB, Takahashi JS. Central and peripheral circadian clocks in mammals. Annu Rev Neurosci. 2012; 35:445–62.
  7. Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. 2017 Mar;18(3):164–79.
  8. Refinetti R. Circadian Physiology. 3rd ed. Boca Raton: CRC Press; 2016.
  9.  Foster RG, Kreitzman L. The rhythms of life: what your body clock means to you! Exp Physiol. 2014 Dec;99(12):599–606.
  10.   Bass J, Lazar MA. Circadian time signatures of fitness and disease. Science. 2016 Oct 14;354(6315):994–9.
  11. Archer SN, Oster H. How sleep and wakefulness influence circadian rhythmicity: effects of insufficient and mistimed sleep on the animal and human transcriptome. J Sleep Res. 2015 Feb;24(5):476–93.
  12. Panda S, Hogenesch JB, Kay SA. Circadian rhythms from flies to human. Nature. 2002 May 16;417(6886):329–35.
  13.  Tsimakouridze EV, Alibhai FJ, Martino TA. Therapeutic applications of circadian rhythms for the cardiovascular system. Front Pharmacol. 2015 Jun 2; 6:77.
  14. Hermida RC, Ayala DE, Mojón A, Fernández JR. Bedtime dosing of antihypertensive medications reduces cardiovascular risk in CKD. J Am Soc Nephrol. 2011 Dec;22(12):2313–21.
  15. Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH, et al. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature. 2010 Jul 29;466(7306):627–31.
  16. Sulli G, Manoogian ENC, Taub PR, Panda S. Training the circadian clock, clocking the drugs, and drugging the clock to prevent, manage, and treat chronic diseases. Trends Pharmacol Sci. 2018 Sep;39(9):812–27.
  17. Cutolo M, Masi AT. Circadian rhythms and arthritis. Rheum Dis Clin North Am. 2005 May;31(1):115–29.
  18. Reinberg A, Smolensky MH. Chronopharmacology: concepts and applications to drug dosing schedules. Adv Drug Deliv Rev. 1997 Mar;25(1):85–104.
  19. Bruguerolle B, Labrecque G. Rhythmic pattern in pain and analgesia: implications for chronotherapeutics. Adv Drug Deliv Rev. 2007 Dec;59(9–10):883–95.
  20. Arvidson NG, Gudbjörnsson B, Elfman L, Rydvall A, Hällgren R. Circadian rhythm of serum interleukin-6 in rheumatoid arthritis. Ann Rheum Dis. 1994 Jul;53(8):521–4.
  21. Ballesta A, Innominato PF, Dallmann R, Rand DA, Lévi FA. Systems chronotherapeutics. Pharmacol Rev. 2017 Apr;69(2):161–99.
  22. Bjarnason I, Sjoqvist F, Sundberg J, Lindström B. Chronopharmacokinetics of indomethacin in man. Eur J Clin Pharmacol. 1982;22(5):331–4.
  23.  Labrecque G, Bélanger PM. Biological rhythms in the absorption, distribution, metabolism and excretion of drugs. Pharmacol Ther. 1991;52(1):95–107.
  24. Zhang YK, Yeager RL, Klaassen CD. Circadian expression profiles of drug-processing genes and transcription factors in mouse liver. Drug Metab Dispos. 2009 Feb;37(1):106–15.
  25.  Hermida RC, Ayala DE, Calvo C, López JE, Fernández JR. Chronotherapy improves blood pressure control and reduces vascular risk in CKD. Nat Rev Nephrol. 2013 May;9(6):358–68.
  26.   Dallmann R, Brown SA, Gachon F. Chronopharmacology: new insights and therapeutic implications. Annu Rev Pharmacol Toxicol. 2014; 54:339–61.
  27. Selfridge JM, Gotoh T, Schiffhauer S, Liu J, Stauffer PE, Li Z, et al. Chronotherapy in asthma: evaluating morning versus evening dosing of inhaled corticosteroids. Chronobiol Int. 2016;33(5):553–67.
  28.  Kario K. Morning surge in blood pressure and cardiovascular risk: evidence and perspectives. Hypertension. 2010 May;56(5):765–73.
  29.  Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, et al. Efficacy of modified-release versus standard prednisone to reduce morning stiffness of the joints in rheumatoid arthritis (CAPRA-1): a double-blind, randomised controlled trial. Lancet. 2008 Feb 2;371(9608):205–14.
  30.  Giacchetti S, Bjarnason G, Garufi C, Genet D, Iacobelli S, Tampellini M, et al. Phase III trial comparing 4-day chronomodulated therapy versus conventional delivery of fluorouracil, leucovorin, and oxaliplatin as first-line chemotherapy in metastatic colorectal cancer: the EORTC 05011 trial. J Clin Oncol. 2006 Apr 1;24(10):3562–9.
  31. Boden G, Chen X, Polansky M. Disruption of circadian insulin secretion is associated with reduced glucose uptake and increased insulin resistance in type 2 diabetes. Diabetes. 1996 Dec;45(12):1543–9.
  32. White WB, Faulkner ME. Clinical relevance of the circadian rhythm in blood pressure and the effects of antihypertensive drug timing. Curr Hypertens Rep. 2011 Oct;13(5):421–8.
  33. Grigorescu R, Rus LL, Butnaru M, Andronie-Cioara FL, Oprean R, Todoran N, et al. Chronotherapy: A timed strategy in cancer treatment. Pharmaceutics. 2022;14(4):788.
  34. Ohdo S, Koyanagi S, Matsunaga N. Chronopharmacological strategies: Intra- and inter-individual variability of molecular clock. Adv Drug Deliv Rev. 2010 Jul 31;62(9-10):885-97.
  35. Bhatt P, Madhav S, Nayak P, Saha RN. Chronotherapeutics: An emerging trend in drug delivery. J Pharm Bioallied Sci. 2010;2(3):184–91.
  36. Smolensky MH, Hermida RC, Geng YJ. Chronotherapy of cardiac and vascular disease: timing medications to circadian rhythms to optimize treatment effects and outcomes. Curr Opin Pharmacol. 2021 Apr; 57:41-48.
  37. Sunil SA, Srikanth MV, Rao NS, Uhumwangho MU, Latha K, Murthy KV. Chronotherapeutic drug delivery systems: an approach to circadian rhythms diseases. Curr Drug Deliv. 2011 Nov;8(6):622-33.
  38. Mishra M, Yadav JS. Chrono-modulated drug delivery system approaches, evaluations and in vitro in vivo evaluations. YMER. 2024.
  39. Vimalraj E, Uthumansha U. A current update and future prospects of chronomodulated drug delivery systems. Afr J Biol Sci. 2024;6(15):275–91.
  40. Asiniparthi M, Anusha, Vaidehi D. Formulation and Evaluation of Chronomodulated Drug Delivery of Montelukast. Glob J Pharmaceu Sci. 2016; 1(1): 555551.
  41. Beg S, Swain S, Gahoi S, Kohli K. Design, development and evaluation of chronomodulated drug delivery systems of amoxicillin trihydrate with enhanced antimicrobial activity. Curr Drug Deliv. 2013 Apr;10(2):174-87.
  42. Alekya T, Narendar D, Mahipal D, Arjun N, Nagaraj B. Design and Evaluation of Chronomodulated Drug Delivery of Tramadol Hydrochloride. Drug Res (Stuttg). 2018 Mar;68(3):174-180.
  43. European Medicines Agency. Guideline on the Pharmacokinetic and Clinical Evaluation of Modified Release Dosage Forms. EMA/CPMP/EWP/280/96 Corr1. July 2014.
  44. Food and Drug Administration. Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products—General Considerations. March 2003.
  45. Murthy SN, Suryadevara V. Regulatory perspective of chronotherapeutics: a review. J Drug Deliv Ther. 2021;11(2):46–51.

Reference

  1. Smolensky MH, Hermida RC, Reinberg A, Sackett-Lundeen L, Portaluppi F. Circadian rhythms in medicine: implications for temporal drug delivery. J Control Release. 2016 Jan 10; 216:125–43.
  2.  Lemmer B. Chronopharmacokinetics: implications for drug treatment. J Pharm Pharmacol. 1999 Aug;51(8):887–90.
  3.  Lévi F, Okyar A, Dulong S, Innominato PF, Clairambault J. Circadian timing in cancer treatments. Annu Rev Pharmacol Toxicol. 2010; 50:377–421.
  4. Gale JE, Cox HI, Qian J, Block GD, Colwell CS, Matveyenko AV. Disruption of circadian rhythms accelerates development of diabetes through pancreatic beta-cell loss and dysfunction. J Biol Rhythms. 2011 Oct;26(5):423–33.
  5. Youan BB. Chronopharmaceutical drug delivery systems: Hurdles, hype or hope? Adv Drug Deliv Rev. 2010 Apr 30;62(9–10):898–903.
  6. Mohawk JA, Green CB, Takahashi JS. Central and peripheral circadian clocks in mammals. Annu Rev Neurosci. 2012; 35:445–62.
  7. Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. 2017 Mar;18(3):164–79.
  8. Refinetti R. Circadian Physiology. 3rd ed. Boca Raton: CRC Press; 2016.
  9.  Foster RG, Kreitzman L. The rhythms of life: what your body clock means to you! Exp Physiol. 2014 Dec;99(12):599–606.
  10.   Bass J, Lazar MA. Circadian time signatures of fitness and disease. Science. 2016 Oct 14;354(6315):994–9.
  11. Archer SN, Oster H. How sleep and wakefulness influence circadian rhythmicity: effects of insufficient and mistimed sleep on the animal and human transcriptome. J Sleep Res. 2015 Feb;24(5):476–93.
  12. Panda S, Hogenesch JB, Kay SA. Circadian rhythms from flies to human. Nature. 2002 May 16;417(6886):329–35.
  13.  Tsimakouridze EV, Alibhai FJ, Martino TA. Therapeutic applications of circadian rhythms for the cardiovascular system. Front Pharmacol. 2015 Jun 2; 6:77.
  14. Hermida RC, Ayala DE, Mojón A, Fernández JR. Bedtime dosing of antihypertensive medications reduces cardiovascular risk in CKD. J Am Soc Nephrol. 2011 Dec;22(12):2313–21.
  15. Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH, et al. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature. 2010 Jul 29;466(7306):627–31.
  16. Sulli G, Manoogian ENC, Taub PR, Panda S. Training the circadian clock, clocking the drugs, and drugging the clock to prevent, manage, and treat chronic diseases. Trends Pharmacol Sci. 2018 Sep;39(9):812–27.
  17. Cutolo M, Masi AT. Circadian rhythms and arthritis. Rheum Dis Clin North Am. 2005 May;31(1):115–29.
  18. Reinberg A, Smolensky MH. Chronopharmacology: concepts and applications to drug dosing schedules. Adv Drug Deliv Rev. 1997 Mar;25(1):85–104.
  19. Bruguerolle B, Labrecque G. Rhythmic pattern in pain and analgesia: implications for chronotherapeutics. Adv Drug Deliv Rev. 2007 Dec;59(9–10):883–95.
  20. Arvidson NG, Gudbjörnsson B, Elfman L, Rydvall A, Hällgren R. Circadian rhythm of serum interleukin-6 in rheumatoid arthritis. Ann Rheum Dis. 1994 Jul;53(8):521–4.
  21. Ballesta A, Innominato PF, Dallmann R, Rand DA, Lévi FA. Systems chronotherapeutics. Pharmacol Rev. 2017 Apr;69(2):161–99.
  22. Bjarnason I, Sjoqvist F, Sundberg J, Lindström B. Chronopharmacokinetics of indomethacin in man. Eur J Clin Pharmacol. 1982;22(5):331–4.
  23.  Labrecque G, Bélanger PM. Biological rhythms in the absorption, distribution, metabolism and excretion of drugs. Pharmacol Ther. 1991;52(1):95–107.
  24. Zhang YK, Yeager RL, Klaassen CD. Circadian expression profiles of drug-processing genes and transcription factors in mouse liver. Drug Metab Dispos. 2009 Feb;37(1):106–15.
  25.  Hermida RC, Ayala DE, Calvo C, López JE, Fernández JR. Chronotherapy improves blood pressure control and reduces vascular risk in CKD. Nat Rev Nephrol. 2013 May;9(6):358–68.
  26.   Dallmann R, Brown SA, Gachon F. Chronopharmacology: new insights and therapeutic implications. Annu Rev Pharmacol Toxicol. 2014; 54:339–61.
  27. Selfridge JM, Gotoh T, Schiffhauer S, Liu J, Stauffer PE, Li Z, et al. Chronotherapy in asthma: evaluating morning versus evening dosing of inhaled corticosteroids. Chronobiol Int. 2016;33(5):553–67.
  28.  Kario K. Morning surge in blood pressure and cardiovascular risk: evidence and perspectives. Hypertension. 2010 May;56(5):765–73.
  29.  Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, et al. Efficacy of modified-release versus standard prednisone to reduce morning stiffness of the joints in rheumatoid arthritis (CAPRA-1): a double-blind, randomised controlled trial. Lancet. 2008 Feb 2;371(9608):205–14.
  30.  Giacchetti S, Bjarnason G, Garufi C, Genet D, Iacobelli S, Tampellini M, et al. Phase III trial comparing 4-day chronomodulated therapy versus conventional delivery of fluorouracil, leucovorin, and oxaliplatin as first-line chemotherapy in metastatic colorectal cancer: the EORTC 05011 trial. J Clin Oncol. 2006 Apr 1;24(10):3562–9.
  31. Boden G, Chen X, Polansky M. Disruption of circadian insulin secretion is associated with reduced glucose uptake and increased insulin resistance in type 2 diabetes. Diabetes. 1996 Dec;45(12):1543–9.
  32. White WB, Faulkner ME. Clinical relevance of the circadian rhythm in blood pressure and the effects of antihypertensive drug timing. Curr Hypertens Rep. 2011 Oct;13(5):421–8.
  33. Grigorescu R, Rus LL, Butnaru M, Andronie-Cioara FL, Oprean R, Todoran N, et al. Chronotherapy: A timed strategy in cancer treatment. Pharmaceutics. 2022;14(4):788.
  34. Ohdo S, Koyanagi S, Matsunaga N. Chronopharmacological strategies: Intra- and inter-individual variability of molecular clock. Adv Drug Deliv Rev. 2010 Jul 31;62(9-10):885-97.
  35. Bhatt P, Madhav S, Nayak P, Saha RN. Chronotherapeutics: An emerging trend in drug delivery. J Pharm Bioallied Sci. 2010;2(3):184–91.
  36. Smolensky MH, Hermida RC, Geng YJ. Chronotherapy of cardiac and vascular disease: timing medications to circadian rhythms to optimize treatment effects and outcomes. Curr Opin Pharmacol. 2021 Apr; 57:41-48.
  37. Sunil SA, Srikanth MV, Rao NS, Uhumwangho MU, Latha K, Murthy KV. Chronotherapeutic drug delivery systems: an approach to circadian rhythms diseases. Curr Drug Deliv. 2011 Nov;8(6):622-33.
  38. Mishra M, Yadav JS. Chrono-modulated drug delivery system approaches, evaluations and in vitro in vivo evaluations. YMER. 2024.
  39. Vimalraj E, Uthumansha U. A current update and future prospects of chronomodulated drug delivery systems. Afr J Biol Sci. 2024;6(15):275–91.
  40. Asiniparthi M, Anusha, Vaidehi D. Formulation and Evaluation of Chronomodulated Drug Delivery of Montelukast. Glob J Pharmaceu Sci. 2016; 1(1): 555551.
  41. Beg S, Swain S, Gahoi S, Kohli K. Design, development and evaluation of chronomodulated drug delivery systems of amoxicillin trihydrate with enhanced antimicrobial activity. Curr Drug Deliv. 2013 Apr;10(2):174-87.
  42. Alekya T, Narendar D, Mahipal D, Arjun N, Nagaraj B. Design and Evaluation of Chronomodulated Drug Delivery of Tramadol Hydrochloride. Drug Res (Stuttg). 2018 Mar;68(3):174-180.
  43. European Medicines Agency. Guideline on the Pharmacokinetic and Clinical Evaluation of Modified Release Dosage Forms. EMA/CPMP/EWP/280/96 Corr1. July 2014.
  44. Food and Drug Administration. Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products—General Considerations. March 2003.
  45. Murthy SN, Suryadevara V. Regulatory perspective of chronotherapeutics: a review. J Drug Deliv Ther. 2021;11(2):46–51.

Photo
Sathish Kumar T.
Corresponding author

Department of Pharmaceutics, C. L. Baid Metha College of Pharmacy

Photo
Tarun S.
Co-author

Department of Pharmaceutics, C. L. Baid Metha College of Pharmacy

Photo
Sivakumar R.
Co-author

Department of Pharmaceutics, C. L. Baid Metha College of Pharmacy

Tarun S., Sathish Kumar T.*, Sivakumar R., Chrono– Drug Delivery System– A Comprehensive Review, Int. J. Sci. R. Tech., 2025, 2 (10), 67-80. https://doi.org/10.5281/zenodo.17284223

More related articles
Evaluation and Preparation of Joint Pain & Muscle ...
Poonam Bansode, Shital Palkar, Sakshi Ingle, Sanika Borpi, Sayli ...
Dissolve Discomfort Instantly: Herbal Sublingual F...
Aniket Thul, Pooja Rasal, Shruti Naik, Sneha Nishad, Onkar Shepal...
Nano Based Drug Delivery Systems: Recent Developme...
Tejaswini Shinde, Kanchan Deshmukh, Vaibhavi Gavali, Shradha Deok...
CRISPR and Gene Therapy Applications– A Comprehensive Review...
Amit Kumar Patel, Sachchidanand Pathak, Dhruv Maurya, Diwakar Raj, Devendra Yadav, ...
A Review on Vinca Alkaloids in Cancer Therapy: Mechanisms, Cultivation and Futur...
Priti Bhure, Rajashri Wagh, Trupti Bankar, Shital Karhale, Vikram Saruk, Manoj Garad, ...
A Review on Vinca Alkaloids in Cancer Therapy: Mechanisms, Cultivation and Futur...
Priti Bhure, Rajashri Wagh, Trupti Bankar, Shital Karhale, Vikram Saruk, Manoj Garad, ...
Related Articles
Determination of Sex from the Sternum and Fourth Rib Measurements (A Cross-Secti...
Nitin Kumar, Sandhya Verma, Jyoti Yadav, Shubhanshi Rani, Shivam Kumar, ...
Chrono– Drug Delivery System– A Comprehensive Review...
Sathish Kumar T., Tarun S., Sivakumar R., ...
Recent Advances in Nanoparticles-Based Drug Delivery Systems...
Pokale Shraddha, Bhise Gorakhnath , Salve Aniket , Ghuge Tanuja , Kolhe Vishakha , ...
Evaluation and Preparation of Joint Pain & Muscle Pain Releasing Spray...
Poonam Bansode, Shital Palkar, Sakshi Ingle, Sanika Borpi, Sayli Tayde, Sakshi Dhote, Rupali Shelke,...
More related articles
Evaluation and Preparation of Joint Pain & Muscle Pain Releasing Spray...
Poonam Bansode, Shital Palkar, Sakshi Ingle, Sanika Borpi, Sayli Tayde, Sakshi Dhote, Rupali Shelke,...
Dissolve Discomfort Instantly: Herbal Sublingual Films as A Natural Cure for Aci...
Aniket Thul, Pooja Rasal, Shruti Naik, Sneha Nishad, Onkar Shepal, ...
Nano Based Drug Delivery Systems: Recent Developments and Future Prospects...
Tejaswini Shinde, Kanchan Deshmukh, Vaibhavi Gavali, Shradha Deokar, ...
Evaluation and Preparation of Joint Pain & Muscle Pain Releasing Spray...
Poonam Bansode, Shital Palkar, Sakshi Ingle, Sanika Borpi, Sayli Tayde, Sakshi Dhote, Rupali Shelke,...
Dissolve Discomfort Instantly: Herbal Sublingual Films as A Natural Cure for Aci...
Aniket Thul, Pooja Rasal, Shruti Naik, Sneha Nishad, Onkar Shepal, ...
Nano Based Drug Delivery Systems: Recent Developments and Future Prospects...
Tejaswini Shinde, Kanchan Deshmukh, Vaibhavi Gavali, Shradha Deokar, ...