Next Article in Journal
Development Perspectives for Curative Technologies in Primary Demyelinating Disorders of the Central Nervous System with Neuromyelitis Optica Spectrum Disorder (NMOSD) and Myelin Oligodendrocyte Glycoprotein Antibody-Associated Disease (MOGAD) at the Forefront
Previous Article in Journal
Mid- and Late-Life Chronic Kidney Disease Is Associated with Parkinson’s Disease, Not with an Increased Risk of Alzheimer’s Disease
Previous Article in Special Issue
Effects of Dry Needling on Active Myofascial Trigger Points and Pain Intensity in Tension-Type Headache: A Randomized Controlled Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Revolutionizing Sleep Health: The Emergence and Impact of Personalized Sleep Medicine

by
Sergio Garbarino
1,2 and
Nicola Luigi Bragazzi
3,4,*
1
Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences (DINOGMI), University of Genoa, 16126 Genoa, Italy
2
Post-Graduate School of Occupational Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
3
Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON M3J 1P3, Canada
4
Human Nutrition Unit (HNU), Department of Food and Drugs, University of Parma, 43125 Parma, Italy
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2024, 14(6), 598; https://doi.org/10.3390/jpm14060598
Submission received: 23 February 2024 / Revised: 11 May 2024 / Accepted: 3 June 2024 / Published: 4 June 2024
(This article belongs to the Special Issue New Advances in the Prevention and Treatment of Neurological Diseases)

Abstract

:
Personalized sleep medicine represents a transformative shift in healthcare, emphasizing individualized approaches to optimizing sleep health, considering the bidirectional relationship between sleep and health. This field moves beyond conventional methods, tailoring care to the unique physiological and psychological needs of individuals to improve sleep quality and manage disorders. Key to this approach is the consideration of diverse factors like genetic predispositions, lifestyle habits, environmental factors, and underlying health conditions. This enables more accurate diagnoses, targeted treatments, and proactive management. Technological advancements play a pivotal role in this field: wearable devices, mobile health applications, and advanced diagnostic tools collect detailed sleep data for continuous monitoring and analysis. The integration of machine learning and artificial intelligence enhances data interpretation, offering personalized treatment plans based on individual sleep profiles. Moreover, research on circadian rhythms and sleep physiology is advancing our understanding of sleep’s impact on overall health. The next generation of wearable technology will integrate more seamlessly with IoT and smart home systems, facilitating holistic sleep environment management. Telemedicine and virtual healthcare platforms will increase accessibility to specialized care, especially in remote areas. Advancements will also focus on integrating various data sources for comprehensive assessments and treatments. Genomic and molecular research could lead to breakthroughs in understanding individual sleep disorders, informing highly personalized treatment plans. Sophisticated methods for sleep stage estimation, including machine learning techniques, are improving diagnostic precision. Computational models, particularly for conditions like obstructive sleep apnea, are enabling patient-specific treatment strategies. The future of personalized sleep medicine will likely involve cross-disciplinary collaborations, integrating cognitive behavioral therapy and mental health interventions. Public awareness and education about personalized sleep approaches, alongside updated regulatory frameworks for data security and privacy, are essential. Longitudinal studies will provide insights into evolving sleep patterns, further refining treatment approaches. In conclusion, personalized sleep medicine is revolutionizing sleep disorder treatment, leveraging individual characteristics and advanced technologies for improved diagnosis, treatment, and management. This shift towards individualized care marks a significant advancement in healthcare, enhancing life quality for those with sleep disorders.

1. Introduction

Sleep is a ubiquitous natural phenomenon [1,2], yet extremely complex [3], unique [2,3], and with a significant amount of inter- and intra-individual variability [4,5,6,7,8], which has been found to correlate with age, race/ethnicity, body mass index (BMI), physical and mental health, and chronotype [9], among others.
By embracing such a complexity, “personalized sleep medicine” represents a cutting-edge, deeply transformative field in healthcare that caters to the specific sleep-related healthcare needs of individuals, considering the bidirectional relationship between sleep and health [10]. This novel framework goes beyond traditional one-size-fits-all approaches and underscores the distinct physiological and psychological characteristics of each person to optimize sleep quantity and quality and effectively address sleep disorders [11].
The foundation of this approach lies in recognizing and responding to a range of diverse factors that influence sleep, such as genetic predispositions, behavior, lifestyle habits (nutrition, physical activity, and use of drugs and stimulants), environmental influences, and underlying health conditions [12,13]. By considering these variables, personalized sleep medicine can offer more accurate diagnoses, targeted treatments, and proactive management strategies that better fit each individual’s needs.
The rapid development and integration of new technologies in this field are noteworthy. The advent of new tools, including digital ones, and a deeper understanding of sleep patterns are propelling personalized sleep medicine to the forefront, offering innovative solutions for sleep-related issues [14]. Wearable devices, mobile health applications, and advanced diagnostic tools are increasingly employed to gather, store, and process detailed data on sleep patterns, quantity, quality, and disturbances. These technologies enable continuous monitoring and analysis, providing a wealth of information that can be used to tailor treatments and interventions effectively [15,16,17].
Moreover, the advent of sophisticated data analysis techniques, particularly machine learning (ML) and artificial intelligence (AI), including generative conversational AI [18], has further enhanced and augmented the capabilities of personalized sleep medicine [19,20,21]. These technologies facilitate the interpretation of large datasets, revealing nuanced insights into sleep behaviors and potential disorders. They can empower healthcare providers to craft highly customized treatment plans that align closely with an individual’s specific sleep profile [22].
Additionally, a deeper understanding of sleep science, including the study of circadian rhythms and sleep physiology, plays a crucial role in this field [23]. Research in these areas is uncovering new layers of complexity in how sleep affects overall health and well-being, contributing valuable knowledge that can inform personalized treatment approaches [24]. In essence, personalized sleep medicine stands in the vanguard of a healthcare revolution, one that emphasizes individualized care and harnesses the power of technology and scientific knowledge to improve sleep health [25].
This approach not only addresses sleep disorders more effectively but also enhances the overall quality of life, recognizing that good sleep is a cornerstone of health and well-being [26].
The research aim of the present review is to better frame and conceptualize the emerging field of personalized sleep medicine, envisaging how this disruptive approach in healthcare, tailoring sleep-related treatments and interventions to individual needs and leveraging new technologies, sophisticated data analyses, and a more profound and nuanced understanding of sleep science, can improve sleep health and overall well-being (Table 1).

2. Wearable Technology, IoT/IoMT Integration, Telemedicine, and Virtual Healthcare Platforms

The next generation of wearable technology is already offering and will offer increasingly sophisticated and non-intrusive monitoring of sleep patterns. Integration with the Internet of Things (IoT), the Internet of Medical Things (IoMT), and smart home systems could enable a more holistic approach to managing sleep environments, optimizing factors like lighting, temperature, and noise based on individual preferences and needs [27].
Telemedicine in sleep medicine and more broadly in healthcare has grown exponentially since 2015, driven initially by the need for specialty care access and advancing technologies. The SARS-CoV-2 pandemic accelerated its adoption as health systems sought safe care delivery methods amid restrictions on non-emergent care [28].
Regarding its effectiveness and cost-effectiveness, a number of reviews have underscored the nuanced benefits of telemedicine in sleep disease management and opened up discussions for its optimized use in clinical settings, alongside its impact on medical staff workload, suggesting potential benefits in healthcare resource allocation but also raising questions about its implementation.
For instance, according to a recently published systematic review and meta-analysis [29], synthesizing 33 articles and totaling 8689 participants, the impact of telemedicine-based follow-up management on patients with obstructive sleep apnea (OSA) was found to be beneficial. More in detail, telemedicine increased the average daily usage of continuous positive airway pressure (CPAP) by 36 min, raising the weighted mean difference to 0.61 (95% confidence interval: 0.39 to 0.83). It also increased the percentage of days when CPAP was used for more than four hours by 10.67%, even if telemedicine did not significantly enhance overall CPAP compliance, with an odds ratio of 1.13 (95% confidence interval: 0.72 to 1.76). The analysis showed marginal effects on sleep quality, with a standardized mean difference of 0.15 (95% confidence interval: −0.03 to 0.32) and a decrease in daytime sleepiness by a weighted mean difference of −0.26 (95% confidence interval: −0.79 to 0.28). The reduction in the apnea–hypopnea index (AHI), one of the key metrics of OSA, was −0.53 (95% confidence interval: −3.58 to 2.51). In terms of the overall quality of life, the standardized mean difference was −0.25 (95% confidence interval: −0.25 to 0.76). The conclusion drawn was that while telemedicine-based follow-up within six months helped improve CPAP usage in patients, it did not enhance sleep quality, daytime sleepiness, apnea severity, or quality of life compared to traditional follow-up methods. Additionally, it was found to be more cost-effective, although there was no consensus on whether it increased the workload for medical staff.
The areas of concern identified in this systematic review and meta-analysis as well as in other reviews should be addressed to optimize the use of telemedicine in the management of sleep diseases, such as OSA. While telemedicine has shown some advantages, it does not significantly improve or improves only marginally other sleep-related characteristics and domains. Efforts should focus on enhancing the comprehensive benefits of telemedicine, ensuring that it not only promotes device usage but also contributes to overall patient health outcomes. Further research and improvements in telemedicine protocols could explore more integrated approaches that combine technology with traditional care methods to address these gaps. Additionally, the economic and operational impacts of implementing telemedicine, such as its cost-effectiveness and potential to increase the workload for medical staff, need a thorough evaluation. By addressing these concerns, healthcare providers can better leverage telemedicine as a supportive tool in the management of sleep disorders, making it a more effective and sustainable option in clinical practice.
Finally, expanding telemedicine services and virtual healthcare platforms will make personalized sleep medicine more accessible. This will be especially beneficial for patients in remote or underserved areas, allowing them to receive specialized care without the need for physical travel [30].
For example, the TeleSleep Enterprise-Wide Initiative (EWI) represents an important, successful initiative aimed at enhancing access to sleep care for rural veterans through the development and implementation of national telehealth networks [31,32].

3. Advanced Data Integration and Analysis and Biomarker (Genomic and Molecular) Research

Future developments in personalized sleep medicine will likely emphasize the integration of diverse data sources, reflecting the impact of the complex, non-linear interplay of genetic, environmental, and lifestyle factors on sleep health. Advanced data analytics and AI will play a crucial role in synthesizing this information to provide more comprehensive and personalized sleep assessments and treatments [33].
The exploration of genetic and molecular aspects of sleep [34,35,36] will deepen, potentially leading to breakthroughs in understanding individual sleep disorders as well as the determinants of sleep health.
Recent advancements in the field of genomics on sleep, including genome-wide association studies (GWASs), have provided substantial insights into the genetic underpinnings of various sleep-related traits and disorders. Studies have identified numerous genetic loci associated with the architecture of sleep in terms of sleep duration, quality, and timing. For example, a GWAS conducted by the UK Biobank and other cohorts identified genes related to neurotransmitter regulation, such as PAX8, which influence sleep duration, suggesting that genetic factors can significantly affect how long and how well individuals sleep [37].
Other GWASs on insomnia have identified several risk loci, highlighting the involvement of brain regions and pathways that regulate sleep, stress responses, and circadian rhythm. For instance, variants in genes like MEIS1 and others that are also implicated in neurodevelopmental and psychiatric disorders suggest a complex interplay between sleep disruption and mental health [38].
Studies focusing on circadian rhythms have discovered genes that affect both the timing of sleep and physiological processes that follow a daily cycle. Notably, variants near genes such as PER1 and CRY2 are significant, underscoring the genetic basis of circadian regulation that can influence sleep patterns and overall health [39,40].
Specific sleep disorders like narcolepsy have been linked to genetic variants through GWASs. A notable example is the identification of a strong association between narcolepsy and variants near the HLA-DQB1 and HLA-DQA1 genes [41], highlighting immune system involvement and underscoring the bidirectional relationship between sleep and immunity [42,43].
Recently, some multi-trait GWASs have started to explore the genetic correlations between sleep traits and other health outcomes, besides psychiatric disorders, such as metabolic health and cardiovascular disease. These studies suggest that poor sleep can be both a cause and consequence of other health issues, driven by shared genetic pathways [44,45].
Furthermore, comparative studies across different populations, including cross-population and multi-ethnic/multi-ancestry meta-analyses, have begun to reveal how genetic determinants of sleep traits may vary by ancestry, which is critical for understanding the global epidemiology of sleep disorders and tailoring interventions appropriately. These insights from GWASs are crucial for developing personalized medicine approaches and could lead to better therapeutic strategies targeting sleep disorders and related health conditions [46,47].
Finally, a recently published study [48] utilized Genomic Structural Equation Modeling (Genomic SEM) to explore the genetic foundations of sleep health and its links to psychopathology. This method involves using diverse GWAS data to construct models estimating the genetic relationships and latent variables among various sleep-related traits. By comparing different genetic models, the study identified how multiple dimensions of sleep health genetically correlate and interact with psychological well-being and psychiatric disorders, providing insights into potential targets for improving mental health outcomes. More specifically, six sleep health factors (namely, sleep efficiency, sleep duration, sleep regularity, daytime alertness, non-insomnia, and circadian preference) showed significant genetic correlations with each other and with various psychopathological conditions, highlighting that these sleep health components are genetically distinct yet interrelated, and they differently associate with psychological and psychopathological outcomes. Taken together, these findings underscore the complexity of sleep as a multivariate health behavior with implications for mental health, suggesting that interventions targeting specific sleep health components could differentially influence multiple health outcomes [49].
All this could pave the way for highly personalized treatment plans based on genetic predispositions and molecular pathways: as research continues, the integration of GWAS data with other biological data (like epigenetics and gene expression studies) [50] is likely to deepen our understanding of sleep and its complex relationship with human health.

4. Personalized Sleep Stage Estimation and Circadian Rhythm Analysis

Accurate sleep stage estimation is essential for the effective diagnosis and management of sleep disorders. Recently, there have been significant advancements in the methodologies used to assess sleep stages, including the adoption of non-contact devices [51,52] and techniques based on ML and deep learning [53]. These innovations enhance the precision of sleep stage detection, which is crucial for determining the overall quality and quantity of sleep, as well as its various impacts on health. The integration of ML and deep learning in sleep medicine is transformative: these technologies analyze large datasets to reveal patterns and anomalies that were previously difficult or impossible to detect. This capability allows for a deeper understanding of sleep processes and aids in the identification of specific sleep disorders. By providing a clearer picture of an individual’s sleep patterns, these advanced analytical tools can lead to better-targeted and more effective treatments [53,54]. ML and deep learning are particularly advantageous in that they allow for personalized sleep stage estimation. These technologies can adapt to the unique sleep patterns of individuals, offering a customized analysis that aligns closely with personal sleep characteristics. This individualized approach is critical in optimizing treatment strategies for sleep disorders, potentially improving clinical outcomes [53,54].
For instance, in a recent study [54], the authors crafted a personalized sleep intervention framework leveraging real-world sleep–wake data from wearable technology to enhance alertness during designated times. This innovative framework employed a mathematical model to monitor dynamic sleep pressure (homeostatic process or process S) and circadian rhythms (process C) [55], drawing on the user’s sleep history to provide accurate real-time alertness predictions, even for shift workers with irregular sleep and work schedules. This approach enabled the identification of a novel sleep–wake pattern known as adaptive circadian split sleep (ACSS), which combines a primary sleep period with a late nap to sustain high alertness throughout both work and leisure times for shift workers. The authors also developed a mobile app integrating this model to suggest personalized sleep schedules tailored to maximizing alertness during specific activities, considering each user’s preferred sleep onset and total sleep time. This innovation not only minimizes the risk of errors during times when high alertness is needed but also enhances the health and quality of life for those with shift-work-like schedules.
Another notable application of these technologies is their use in analyzing ultradian rhythms—natural cycles within the sleep period typically lasting 60 to 120 min. One innovative study [56] explored the integration of ML algorithms with predicted ultradian rhythms to refine sleep stage estimation. This approach involves weighting the ML results according to these rhythms, enhancing the system’s ability to adapt to individual sleep cycle variations. Validated through experiments involving human subjects, this method demonstrated its efficacy by producing results that were competitive with, or superior to, traditional ML techniques. This highlights its potential clinical relevance and the value of incorporating such rhythms into sleep analysis models. The focus on individual sleep patterns and the adjustment for ultradian rhythms illustrate a significant shift towards more customized healthcare solutions in sleep medicine.
These advancements highlight the potential of personalized sleep medicine, where treatment is tailored not just to the specific sleep disorder but to the individual characteristics and sleep profile of each patient. As ML and deep learning continue to evolve, their impact on the field of sleep medicine is likely to grow, leading to more precise diagnostics, better patient outcomes, and a deeper understanding of the complexities of sleep.

5. Computational Models of Sleep Disorders

Computational models represent a cutting-edge approach in the customized treatment of sleep disorders: these models offer a sophisticated method of personalizing management plans based on individual patient needs and conditions [57].
By using detailed 3D models of a patient’s anatomical structures, clinicians can accurately simulate how different treatment strategies might affect the patient. For example, in OSA, such models can predict how changes in airway structure due to various interventions (like CPAP, dental appliances, or surgery) impact airflow and tissue displacement during sleep. This level of detail allows for highly personalized therapy plans that are tailored not just to the disease but to the patient’s unique physiological characteristics, as previously mentioned. Also, computational modeling can facilitate the optimization of treatment outcomes by allowing for the simulation and assessment of multiple therapeutic scenarios. This approach helps in selecting the most effective treatment strategy with potentially higher success rates and reduced side effects or complications. Further, these models can enhance the understanding of the pathophysiological mechanisms underlying sleep disorders. By observing how different variables affect sleep dynamics and respiratory patterns, researchers can identify new therapeutic targets and improve existing treatment modalities.
For instance, a study [57] explored the use of computational models for the individualized treatment of sleep disorders, such as OSA, employing patient-specific partial 3D finite element studies. This approach underscores the effectiveness of tailor-made treatment strategies, especially in managing complex sleep disorders. By adapting to the specific conditions of each patient, these computational models pave the way for more effective, customized treatment plans. Moreover, computational models can be integrated with other technologies such as ML and real-time data monitoring (from wearables or bedside monitors). Such integration can lead to dynamic models that not only predict but also adapt to changes in a patient’s condition over time, potentially leading to continuously optimized treatment plans [57].
Despite these advantages, significant challenges remain in the broader implementation of computational models. These include the need for high-quality, patient-specific data, the computational cost of detailed simulations, and the necessity for robust validation of the models against clinical outcomes. Future research should focus on addressing these challenges, improving the accuracy of the models, and integrating them more seamlessly into clinical workflows. By addressing these aspects, computational models could revolutionize the management of sleep disorders, making treatments not only more effective but also more responsive to the changing health landscape of each patient [57].

6. Customized Sleep Pharmacogenetics and Pharmacotherapy

Personalized sleep medicine is poised to revolutionize pharmacotherapy, with medications being tailored to individual patient profiles [58,59]. This could involve adjusting dosages and the timing of administration and choosing specific drug types based on individual responses and tolerances.
Traditional one-size-fits-all dosing strategies are often inadequate because individuals vary in their responses to medications. In personalized sleep medicine, dosages are carefully adjusted based on factors like the patient’s metabolism, age, sex/gender, BMI, and underlying health status. This ensures maximum effectiveness while minimizing potential side effects.
The timing of medication [60,61] can have a profound impact on its effectiveness, particularly in the realm of sleep disorders. Personalized sleep medicine takes into account the patient’s circadian rhythms and sleep patterns. Medications might be administered at specific times to align with these patterns, enhancing their efficacy.
Different patients may respond better to different types of sleep medications [60,61]. Some might benefit more from sedatives, while others may need medications that alter sleep architecture. Factors like underlying health conditions, concurrent medications, and even genetic makeup can influence which drug is most suitable for a patient.
Melatonin serves as a prime example of customization in sleep medications through the lens of personalized pharmacogenetics and chronotherapy. As a naturally occurring hormone that regulates the sleep–wake cycle, melatonin supplements are commonly used to treat sleep disorders such as insomnia and jet lag. However, its effectiveness can vary widely among individuals, a variation that can be attributed to genetic differences [62,63]. Research in pharmacogenetics has revealed that certain genetic variants in melatonin receptor genes, such as MTNR1A and MTNR1B, influence the efficacy of endogenous melatonin [64,65]. By analyzing these genetic markers, as well as other biomarkers, healthcare providers can tailor melatonin treatment plans to enhance effectiveness and reduce side effects, offering a more personalized approach to sleep management, representing a major shift towards individualized medicine, where treatments can be optimized based on a person’s genetic profile [66,67].
Melatonin’s role in sleep management also highlights its importance from a chronotherapy perspective. Melatonin, integral in regulating circadian rhythms, is particularly well suited for this approach. By administering melatonin at specific times, clinicians can effectively shift and re-synchronize these rhythms, thereby aiding individuals whose sleep patterns are misaligned with their environmental cues, such as shift workers or those experiencing jet lag. This approach based on strategic timing not only improves sleep quality but also harnesses the body’s clock to enhance overall well-being. Thus, melatonin exemplifies how understanding and integrating chronobiology into treatment plans can significantly improve the management of sleep disorders and potentially other circadian rhythm-related conditions [63].
Furthermore, continuous monitoring and feedback are essential components of personalized sleep medicine [68]. This could involve regular check-ups, longitudinally designed sleep studies, and the use of wearable technology to track sleep patterns and medication effects [69,70]. Adjustments to the treatment plan are made based on these observations to ensure optimal outcomes. Advances in genetics and biomarker research can provide insights into how a patient might respond to certain sleep medications. This can guide the selection of drugs and dosages for better, more personalized treatment outcomes [71].
Furthermore, personalized sleep medicine also recognizes the importance of non-pharmacological interventions. This may include tailored advice on sleep hygiene, dietary changes, and stress management techniques that can complement the pharmacological treatment [72].
Overall, personalized sleep medicine aims to move beyond the traditional trial-and-error approach, offering more precise, effective, and safer treatment options for individuals suffering from sleep disorders. This represents a significant shift towards a more patient-centered approach in healthcare.

7. Obstructive Sleep Apnea as a Case Study

OSA is a complex disorder characterized by repeated episodes of partial or complete obstruction of the upper airway during sleep, leading to impaired sleep, disrupted sleep patterns, and decreased oxygen levels in the blood [73,74]. OSA represents a significant global health concern, generating a dramatic epidemiological and clinical burden [75]. It is estimated, indeed, to affect nearly one billion adults worldwide with enormous individual and public health consequences. From a gender perspective, this pathology affects 17% and 34% of women and men, respectively, and excessive daytime sleepiness is its most common presenting symptom, although it is reported by as few as 15–50% of people with OSA in the general population [73,74,75,76]. The traditional one-size-fits-all approach often fails to address the multifaceted nature of OSA, which is where the concept of clinical phenotypes and endotypes (representing subtypes of a condition, defined by distinct functional or pathobiological mechanisms) becomes crucial [77,78,79,80].
Certain craniofacial structures and alignments, like retrognathia (receded jaw), high-arched palate, and overbite, can predispose individuals to OSA [81]. By taking into account these features, personalized treatment may involve dental or orthodontic interventions such as mandibular advancement devices (MADs) or surgeries to modify the craniofacial structure, thereby improving airway patency [82,83]. Skeletal features, including the size and positioning of the jaw and other facial bones, can significantly affect airway size and shape [81]. As such, personalized interventions might include orthognathic surgery to reposition skeletal components for patients with severe skeletal discrepancies [81,82,83].
The neurological control of upper airway muscles plays a role in OSA too, as reduced muscle tone during sleep can lead to airway collapse [84,85,86,87]. Personalized treatment targeting this pathophysiological mechanism could include therapies to enhance the tone and function of these muscles, possibly through neuromuscular electrical stimulation or targeted exercises [88].
Lung function and respiratory control mechanisms also contribute to OSA, particularly in individuals with underlying respiratory diseases [84,85,86,87]. Management might involve using positive airway pressure therapies, such as CPAP, to keep the airway open, or addressing underlying respiratory conditions.
Obesity, particularly central obesity, is a significant risk factor for OSA. Fat deposits around the neck and upper airway can exacerbate airway obstruction. Weight loss programs, bariatric surgery, or other metabolic interventions could be part of a personalized treatment plan for such individuals [89,90,91,92].
Personalized treatment begins with a comprehensive evaluation of the patient, including detailed physical examinations, imaging studies, and sleep studies [93]. Genetic testing and molecular profiling might also play a role in understanding individual susceptibilities and responses to treatment [94,95,96,97].
Based on the assessment, a combination of therapies may be recommended [98,99]. This could include medical devices, surgical interventions, lifestyle changes, and pharmacotherapy [100,101,102,103]. For example, a patient with a pronounced craniofacial endophenotype might benefit more from orthodontic or surgical interventions than from CPAP therapy alone. Besides clinical subtypes, comorbidities and patients’ individual preferences should be taken into account when customizing the treatment plan [104,105,106,107]. Continuous monitoring and regular follow-up are essential to assess the effectiveness of the treatment and make adjustments as needed [108]. Wearable technology and telemedicine platforms can facilitate ongoing monitoring and communication between patients and healthcare providers [108].
Existing strategic frameworks for managing OSA adopt a uniform approach that applies to all patients—the so-called “one-size-fits-all models” [105,106,107,109]. Ultimately, the diagnosis and severity of OSA are based on key indicators, like the AHI, the oxygen desaturation index (ODI), minimum oxygen saturation, the Epworth Sleepiness Scale (ESS), the duration of apneas and hypopneas, and treatment initiated with CPAP, followed by alternative treatment options (e.g., MAD) if CPAP “fails”. As previously mentioned, this will have to consider the heterogeneity of patients with OSA, characterized by different risk factors, pathophysiological causes, clinical manifestations, and comorbidities. A range of analytical methods, such as cluster analysis, can be utilized to discern patterns within clinical heterogeneity and OSA phenotypes. Identifying patient subtypes with distinct characteristics through these techniques may facilitate more tailored and effective strategies for prognosis and treatment (e.g., symptom-based subtypes such as “minimally symptomatic”, “excessive sleepiness”, and “disturbed sleep”) [106,110].
In precision medicine for OSA, polysomnographic subtypes, distinguished by the association of respiratory events with hypoxemia, arousals, or both, also present variable risks of cardiovascular disease and responses to therapy [106,110]. By linking various phenotypes to clinically relevant outcomes, a correct and rigorous assessment of phenotype reproducibility can be ensured, and the need for tools that categorize and interpret these differences effectively can be satisfied. Thus, clinicians can better tailor treatments to individual patient profiles. This approach emphasizes the importance of a nuanced understanding of each patient’s specific sleep disorder characteristics, enabling more targeted and potentially more effective therapeutic interventions.
Treating OSA effectively often requires a multidisciplinary team, including sleep specialists, dentists, surgeons, neurologists, pulmonologists, otorhinolaryngologists, and dietitians [111]. Collaboration among these specialists ensures a comprehensive approach to addressing the various endophenotypes of the patient [111].
Finally, educating patients about their specific endophenotypes and involving them in treatment decisions are crucial for adherence and success [112,113,114]. Personalized treatment plans should consider the patient’s preferences, lifestyle, and ability to adhere to the recommended therapies [115]. In summary, recognizing and addressing the multiple endophenotypes in OSA are fundamental to providing personalized and effective treatment. This approach not only targets the specific contributing factors of the disorder for each individual but also offers a more holistic and patient-centered strategy for managing this complex condition.

8. Future Directions

The field of personalized sleep medicine is anticipated to benefit from cross-disciplinary collaborations, involving sleep scientists, neurologists, psychologists, data scientists, bioengineers, and other experts. Such collaborations could lead to more innovative solutions and a deeper understanding of complex sleep disorders [111].
There will be an increased focus on integrating “customized” cognitive behavioral therapy (CBT) and other mental health interventions into personalized sleep medicine [113]. This holistic approach will address both the psychological and physiological aspects of sleep disorders, offering a more integrated and effective treatment strategy.
Increased public awareness and educational initiatives about the importance of personalized approaches to sleep health will be essential. Educating both healthcare providers and the general public can lead to earlier recognition of sleep disorders and more proactive management [112,113,114].
As personalized sleep medicine evolves, there will be a need for updated regulatory frameworks to address privacy, data security, and ethical considerations, especially concerning genetic and biometric data [116].
Finally, emphasis on longitudinal studies and continuous monitoring will provide deeper insights into how sleep patterns evolve over time in relation to aging, lifestyle changes, and health status, further refining personalized treatment approaches. The process of monitoring and managing sleep disorders, notably OSA, as well as ensuring that patients consistently adhere to and comply with their prescribed treatment plans, poses substantial challenges. This involves regular follow-up appointments to assess the patient’s progress, adjustments to treatment protocols based on the patient’s response, and continuous education and support to encourage patient compliance. Effective management of sleep disorders like OSA requires a multifaceted approach, including the use of medical devices such as CPAP machines, lifestyle modifications, and possibly medication. Ensuring patient engagement and adherence to these treatment strategies is crucial for the successful management of the condition and to prevent associated risks such as cardiovascular problems, daytime fatigue, and decreased quality of life. The complexity of these tasks highlights the need for a coordinated approach involving healthcare professionals, patient education, and support systems to overcome these challenges [117,118].

9. Conclusions

Within the broader P4–P6 (personalized, predictive, preventive, participatory, psycho-cognitive, and public) framework of contemporary medicine [119,120,121,122], “personalized sleep medicine” has great potential to revolutionize the understanding and treatment of sleep disorders. By focusing on individual characteristics and employing advanced technologies like ML and computational modeling, this field offers a more nuanced and effective approach to sleep health. The promise of personalized sleep medicine lies in its ability to provide better diagnosis, treatment, and management of sleep disorders, leading to improved sleep quality and overall health outcomes.
This shift towards individualized care is a significant step forward in the realm of healthcare, showcasing the potential of personalized approaches to enhance the quality of life for individuals with sleep disorders.

Author Contributions

S.G. and N.L.B. designed and wrote the review. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Vyazovskiy, V.V. Sleep, recovery, and metaregulation: Explaining the benefits of sleep. Nat. Sci. Sleep 2015, 7, 171–184. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  2. Assefa, S.Z.; Diaz-Abad, M.; Wickwire, E.M.; Scharf, S.M. The Functions of Sleep. AIMS Neurosci. 2015, 2, 155–171. [Google Scholar] [CrossRef]
  3. Eban-Rothschild, A. Beyond model organisms: Diversifying experimental species and ecological complexity to reveal the evolutionary history and functions of sleep. Sleep 2022, 45, zsac102. [Google Scholar] [CrossRef] [PubMed]
  4. Becker, S.P.; Sidol, C.A.; Van Dyk, T.R.; Epstein, J.N.; Beebe, D.W. Intraindividual variability of sleep/wake patterns in relation to child and adolescent functioning: A systematic review. Sleep Med. Rev. 2017, 34, 94–121. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  5. Johnson, D.A.; Jackson, C.L.; Williams, N.J.; Alcántara, C. Are sleep patterns influenced by race/ethnicity—A marker of relative advantage or disadvantage? Evidence to date. Nat. Sci. Sleep 2019, 11, 79–95. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  6. Lemola, S.; Ledermann, T.; Friedman, E.M. Variability of sleep duration is related to subjective sleep quality and subjective well-being: An actigraphy study. PLoS ONE 2013, 8, e71292. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  7. Ng, A.S.C.; Massar, S.A.A.; Bei, B.; Chee, M.W.L. Assessing ‘readiness’ by tracking fluctuations in daily sleep duration and their effects on daily mood, motivation, and sleepiness. Sleep Med. 2023, 112, 30–38. [Google Scholar] [CrossRef] [PubMed]
  8. Fang, Y.; Forger, D.B.; Frank, E.; Sen, S.; Goldstein, C. Day-to-day variability in sleep parameters and depression risk: A prospective cohort study of training physicians. NPJ Digit. Med. 2021, 4, 28. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  9. Bei, B.; Wiley, J.F.; Trinder, J.; Manber, R. Beyond the mean: A systematic review on the correlates of daily intraindividual variability of sleep/wake patterns. Sleep Med. Rev. 2016, 28, 108–124. [Google Scholar] [CrossRef] [PubMed]
  10. Stewart, E.M.; Landry, S.; Edwards, B.A.; Drummond, S.P. The bidirectional relationship between sleep and health. In The Wiley Encyclopedia of Health Psychology; Wiley: New York, NY, USA, 2020; pp. 165–188. [Google Scholar]
  11. Bragazzi, N.L.; Guglielmi, O.; Garbarino, S. SleepOMICS: How Big Data Can Revolutionize Sleep Science. Int. J. Environ. Res. Public Health 2019, 16, 291. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Genderson, M.R.; Rana, B.K.; Panizzon, M.S.; Grant, M.D.; Toomey, R.; Jacobson, K.C.; Xian, H.; Cronin-Golomb, A.; Franz, C.E.; Kremen, W.S.; et al. Genetic and environmental influences on sleep quality in middle-aged men: A twin study. J. Sleep Res. 2013, 22, 519–526. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  13. Sejbuk, M.; Mirończuk-Chodakowska, I.; Witkowska, A.M. Sleep Quality: A Narrative Review on Nutrition, Stimulants, and Physical Activity as Important Factors. Nutrients 2022, 14, 1912. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  14. Redline, S.; Purcell, S.M. Sleep and Big Data: Harnessing data, technology, and analytics for monitoring sleep and improving diagnostics, prediction, and interventions-an era for Sleep-Omics? Sleep 2021, 44, zsab107. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  15. Cheung, J.M.; Menczel Schrire, Z.; Aji, M.; Rahimi, M.; Salomon, H.; Doggett, I.; Glozier, N.; Bartlett, D.J.; Wong, K.; Grunstein, R.R.; et al. Embedding digital sleep health into primary care practice: A triangulation of perspectives from general practitioners, nurses, and pharmacists. Digit. Health 2023, 9, 20552076231180970. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  16. Vijayan, V.; Connolly, J.P.; Condell, J.; McKelvey, N.; Gardiner, P. Review of Wearable Devices and Data Collection Considerations for Connected Health. Sensors 2021, 21, 5589. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  17. de Zambotti, M.; Cellini, N.; Goldstone, A.; Colrain, I.M.; Baker, F.C. Wearable Sleep Technology in Clinical and Research Settings. Med. Sci. Sports Exerc. 2019, 51, 1538–1557. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Bragazzi, N.L.; Garbarino, S. Toward Clinical Generative AI: Conceptual Framework. JMIR AI 2024. [Google Scholar]
  19. Bragazzi, N.L.; Garbarino, S. Assessing the Accuracy of Generative Conversational Artificial Intelligence in Debunking Sleep Health Myths: Mixed Methods Comparative Study with Expert Analysis. JMIR Form. Res. 2024, 8, e55762. [Google Scholar] [CrossRef] [PubMed]
  20. Garbarino, S.; Bragazzi, N.L. Evaluating the effectiveness of artificial intelligence-based tools in detecting and understanding sleep health misinformation: Comparative analysis using Google Bard and OpenAI ChatGPT-4. J. Sleep Res. 2024, 5, e14210. [Google Scholar] [CrossRef] [PubMed]
  21. Watson, N.F.; Fernandez, C.R. Artificial intelligence and sleep: Advancing sleep medicine. Sleep Med. Rev. 2021, 59, 101512. [Google Scholar] [CrossRef] [PubMed]
  22. Perez-Pozuelo, I.; Zhai, B.; Palotti, J.; Mall, R.; Aupetit, M.; Garcia-Gomez, J.M.; Taheri, S.; Guan, Y.; Fernandez-Luque, L. The future of sleep health: A data-driven revolution in sleep science and medicine. NPJ Digit. Med. 2020, 3, 42. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Foster, R.G. Sleep, circadian rhythms and health. Interface Focus. 2020, 10, 20190098. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Thomas, C.L.; Collen, J. A Systematic Approach to Central Sleep Apnea in an Era of Medical Complexity. Fed. Pract. 2023, 40, 76–77. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  25. Ryba-White, B.; Gupta, S.; Khalil, W.; Irfan, M. Integrating telemedicine into sleep medicine: A learner’s perspective. J. Clin. Sleep Med. 2021, 17, 2573–2576. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  26. Ramar, K.; Malhotra, R.K.; Carden, K.A.; Martin, J.L.; Abbasi-Feinberg, F.; Aurora, R.N.; Kapur, V.K.; Olson, E.J.; Rosen, C.L.; Rowley, J.A.; et al. Sleep is essential to health: An American Academy of Sleep Medicine position statement. J. Clin. Sleep Med. 2021, 17, 2115–2119. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  27. Jaworski, D.J.; Park, A.; Park, E.J. Internet of Things for Sleep Monitoring. IEEE Instrum. Meas. Mag. 2021, 24, 30–36. [Google Scholar] [CrossRef]
  28. Haleem, A.; Javaid, M.; Singh, R.P.; Suman, R. Telemedicine for healthcare: Capabilities, features, barriers, and applications. Sens. Int. 2021, 2, 100117. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Niu, Y.; Xi, H.; Zhu, R.; Guo, Y.; Wang, S.; Xiong, X.; Wang, S.; Guo, L. Effects of telemedicine-based follow-up management on adults with obstructive sleep apnea: A systematic review and meta-analysis. Int. J. Med. Inf. 2023, 176, 105108. [Google Scholar] [CrossRef] [PubMed]
  30. Shamim-Uzzaman, Q.A.; Bae, C.J.; Ehsan, Z.; Setty, A.R.; Devine, M.; Dhankikar, S.; Donskoy, I.; Fields, B.; Hearn, H.; Hwang, D.; et al. The use of telemedicine for the diagnosis and treatment of sleep disorders: An American Academy of Sleep Medicine update. J. Clin. Sleep Med. 2021, 17, 1103–1107. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  31. Belkora, J.K.; Ortiz DeBoque, L.; Folmer, R.L.; Totten, A.M.; Williams, K.; Whooley, M.A.; Boudreau, E.; Atwood, C.W.; Zeidler, M.; Rezayat, T.; et al. Sustainment of the TeleSleep program for rural veterans. Front. Health Serv. 2023, 3, 1214071. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  32. Chun, V.S.; Whooley, M.A.; Williams, K.; Zhang, N.; Zeidler, M.R.; Atwood, C.W.; Folmer, R.L.; Totten, A.M.; Smith, C.J.; Boudreau, E.A.; et al. Veterans Health Administration TeleSleep Enterprise-Wide Initiative 2017–2020: Bringing sleep care to our nation’s veterans. J. Clin. Sleep Med. 2023, 19, 913–923. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  33. Verma, R.K.; Dhillon, G.; Grewal, H.; Prasad, V.; Munjal, R.S.; Sharma, P.; Buddhavarapu, V.; Devadoss, R.; Kashyap, R.; Surani, S. Artificial intelligence in sleep medicine: Present and future. World J. Clin. Cases 2023, 11, 8106–8110. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Jan, M.; O’Hara, B.F.; Franken, P. Recent advances in understanding the genetics of Sleep. F1000Res 2020, 9, 214. [Google Scholar] [CrossRef] [PubMed]
  35. Lane, J.M.; Qian, J.; Mignot, E.; Redline, S.; Scheer, F.A.J.L.; Saxena, R. Genetics of circadian rhythms and sleep in human health and disease. Nat. Rev. Genet. 2023, 24, 4–20. [Google Scholar] [CrossRef] [PubMed]
  36. Gottlieb, D.J.; Hek, K.; Chen, T.H.; Watson, N.F.; Eiriksdottir, G.; Byrne, E.M.; Cornelis, M.; Warby, S.C.; Bandinelli, S.; Cherkas, L.; et al. Novel loci associated with usual sleep duration: The CHARGE Consortium Genome-Wide Association Study. Mol. Psychiatry 2015, 20, 1232–1239. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  37. Foo, H.; Thalamuthu, A.; Jiang, J.; Koch, F.C.; Mather, K.A.; Wen, W.; Sachdev, P.S. Novel genetic variants associated with brain functional networks in 18,445 adults from the UK Biobank. Sci. Rep. 2021, 11, 14633. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Song, W.; Torous, J.; Kossowsky, J.; Chen, C.Y.; Huang, H.; Wright, A. Genome-wide association analysis of insomnia using data from Partners Biobank. Sci. Rep. 2020, 10, 6928. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. von Schantz, M.; Archer, S.N. Clocks, genes and Sleep. J. R. Soc. Med. 2003, 96, 486–489. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  40. Tao, Y.; Qin, Y.; Chen, S.; Xu, T.; Lin, J.; Su, D.; Yu, W.; Chen, X. Emerging trends and hot spots of sleep and genetic research: A bibliometric analysis of publications from 2002 to 2022 in the field. Front. Neurol. 2023, 14, 1264177. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  41. Mignot, E.; Lin, L.; Rogers, W.; Honda, Y.; Qiu, X.; Lin, X.; Okun, M.; Hohjoh, H.; Miki, T.; Hsu, S.; et al. Complex HLA-DR and -DQ interactions confer risk of narcolepsy-cataplexy in three ethnic groups. Am. J. Hum. Genet. 2001, 68, 686–699. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  42. Veler, H. Sleep and Inflammation: Bidirectional Relationship. Sleep Med. Clin. 2023, 18, 213–218. [Google Scholar] [CrossRef] [PubMed]
  43. Garbarino, S.; Lanteri, P.; Bragazzi, N.L.; Magnavita, N.; Scoditti, E. Role of sleep deprivation in immune-related disease risk and outcomes. Commun. Biol. 2021, 4, 1304. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Campos, A.I.; Ingold, N.; Huang, Y.; Mitchell, B.L.; Kho, P.F.; Han, X.; García-Marín, L.M.; Ong, J.S.; 23andMe Research Team; Law, M.H.; et al. Discovery of genomic loci associated with sleep apnea risk through multi-trait GWAS analysis with snoring. Sleep 2023, 46, zsac308. [Google Scholar] [CrossRef] [PubMed]
  45. Sofer, T.; Kurniansyah, N.; Murray, M.; Ho, Y.L.; Abner, E.; Esko, T.; Estonian Biobank Research Team; Huffman, J.E.; Cho, K.; Wilson, P.W.F.; et al. Genome-wide association study of obstructive sleep apnoea in the Million Veteran Program uncovers genetic heterogeneity by sex. EBioMedicine 2023, 90, 104536. [Google Scholar] [CrossRef] [PubMed]
  46. Austin-Zimmerman, I.; Levey, D.F.; Giannakopoulou, O.; Deak, J.D.; Galimberti, M.; Adhikari, K.; Zhou, H.; Denaxas, S.; Irizar, H.; Kuchenbaecker, K.; et al. Genome-wide association studies and cross-population meta-analyses investigating short and long sleep duration. Nat. Commun. 2023, 14, 6059. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Khoury, S.; Wang, Q.P.; Parisien, M.; Gris, P.; Bortsov, A.V.; Linnstaedt, S.D.; McLean, S.A.; Tungate, A.S.; Sofer, T.; Lee, J.; et al. Multi-ethnic GWAS and meta-analysis of sleep quality identify MPP6 as a novel gene that functions in sleep center neurons. Sleep 2021, 44, zsaa211. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  48. Morrison, C.L.; Winiger, E.A.; Rieselbach, M.M.; Vetter, C.; Wright, K.P., Jr.; LeBourgeois, M.K.; Friedman, N.P. Sleep Health at the Genomic Level: Six Distinct Factors and Their Relationships with Psychopathology. Biol. Psychiatry Glob. Open Sci. 2022, 3, 530–540. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Chung, J.; Goodman, M.; Huang, T.; Wallace, M.L.; Lutsey, P.L.; Chen, J.T.; Castro-Diehl, C.; Bertisch, S.; Redline, S. Multi-dimensional sleep and mortality: The Multi-Ethnic Study of Atherosclerosis. Sleep 2023, 46, zsad048. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  50. Jan, M.; Gobet, N.; Diessler, S.; Franken, P.; Xenarios, I. A multi-omics digital research object for the genetics of sleep regulation. Sci. Data 2019, 6, 258. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  51. Watanabe, T.; Watanabe, K. Noncontact method for sleep stage estimation. IEEE Trans. Biomed. Eng. 2004, 51, 1735–1748. [Google Scholar] [CrossRef] [PubMed]
  52. Kamon, M.; Okada, S.; Furuta, M.; Yoshida, K. Development of a non-contact sleep monitoring system for children. Front. Digit. Health 2022, 4, 877234. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Sridhar, N.; Shoeb, A.; Stephens, P.; Kharbouch, A.; Shimol, D.B.; Burkart, J.; Ghoreyshi, A.; Myers, L. Deep learning for automated sleep staging using instantaneous heart rate. NPJ Digit. Med. 2020, 3, 106, Erratum in NPJ Digit. Med. 2020, 3, 131. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Song, Y.M.; Choi, S.J.; Park, S.H.; Lee, S.J.; Joo, E.Y.; Kim, J.K. A real-time, personalized sleep intervention using mathematical modeling and wearable devices. Sleep 2023, 46, zsad179. [Google Scholar] [CrossRef] [PubMed]
  55. Borbély, A. The two-process model of sleep regulation: Beginnings and outlook. J. Sleep Res. 2022, 31, e13598. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. Nakari, I.; Takadama, K. Personalized Non-contact Sleep Stage Estimation with Weighted Probability Estimation by Ultradian Rhythm. In Proceedings of the 2023 45th Annual International Conference of the IEEE Engineering in Medicine & Biology Society (EMBC), Sydney, Australia, 24–27 July 2023; pp. 1–4. [Google Scholar] [CrossRef] [PubMed]
  57. Ayyalasomayajula, V.; Moxness, M.; Skallerud, B. Potential of computational models in personalized treatment of obstructive sleep apnea: A patient-specific partial 3D finite element study. Biomech. Model. Mechanobiol. 2023. ahead of print. [Google Scholar] [CrossRef] [PubMed]
  58. Holst, S.C.; Valomon, A.; Landolt, H.P. Sleep Pharmacogenetics: Personalized Sleep-Wake Therapy. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 577–603. [Google Scholar] [CrossRef] [PubMed]
  59. Landolt, H.P.; Holst, S.C.; Valomon, A. Clinical and Experimental Human Sleep-Wake Pharmacogenetics. Handb. Exp. Pharmacol. 2019, 253, 207–241. [Google Scholar] [CrossRef] [PubMed]
  60. Cardinali, D.P.; Brown, G.M.; Pandi-Perumal, S.R. Chronotherapy. Handb. Clin. Neurol. 2021, 179, 357–370. [Google Scholar] [CrossRef] [PubMed]
  61. Zečević, K.; Popović, N.; Vuksanović Božarić, A.; Vukmirović, M.; Rizzo, M.; Muzurović, E. Timing Is Important-Management of Metabolic Syndrome According to the Circadian Rhythm. Biomedicines 2023, 11, 1171. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Erland, L.A.; Saxena, P.K. Melatonin Natural Health Products and Supplements: Presence of Serotonin and Significant Variability of Melatonin Content. J. Clin. Sleep Med. 2017, 13, 275–281. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  63. Moroni, I.; Garcia-Bennett, A.; Chapman, J.; Grunstein, R.R.; Gordon, C.J.; Comas, M. Pharmacokinetics of exogenous melatonin in relation to formulation, and effects on sleep: A systematic review. Sleep Med. Rev. 2021, 57, 101431. [Google Scholar] [CrossRef] [PubMed]
  64. Karamitri, A.; Renault, N.; Clement, N.; Guillaume, J.L.; Jockers, R. Minireview: Toward the establishment of a link between melatonin and glucose homeostasis: Association of melatonin MT2 receptor variants with type 2 diabetes. Mol. Endocrinol. 2013, 27, 1217–1233. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Kolomeichuk, S.N.; Korneva, V.A.; Kuznetsova, T.Y.; Korostovtseva, L.S.; Bochkarev, M.V.; Sviryaev, Y.V.; Blagonravov, M.L. MTNR1A and MTNR1B Gene Variants of the Melatonin Receptor and Arterial Stiffness in Persons without Arterial Hypertension. Bull. Exp. Biol. Med. 2023, 174, 460–463. [Google Scholar] [CrossRef] [PubMed]
  66. Skelton, R.L.; Kornhauser, J.M.; Tate, B.A. Personalized medicine for pathological circadian dysfunctions. Front. Pharmacol. 2015, 6, 125. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Torres, R.; Kramer, W.G.; Baroldi, P. Pharmacokinetics of the dual melatonin receptor agonist tasimelteon in subjects with hepatic or renal impairment. J. Clin. Pharmacol. 2015, 55, 525–533. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Bar, N.; Sobel, J.A.; Penzel, T.; Shamay, Y.; Behar, J.A. From sleep medicine to medicine during sleep—A clinical perspective. Physiol. Meas. 2021, 42, 044006. [Google Scholar] [CrossRef] [PubMed]
  69. Smith, A.A.; Li, R.; Tse, Z.T.H. Reshaping healthcare with wearable biosensors. Sci. Rep. 2023, 13, 4998. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Ho, D.; Quake, S.R.; McCabe, E.R.B.; Chng, W.J.; Chow, E.K.; Ding, X.; Gelb, B.D.; Ginsburg, G.S.; Hassenstab, J.; Ho, C.M.; et al. Enabling Technologies for Personalized and Precision Medicine. Trends Biotechnol. 2020, 38, 497–518. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  71. Alroobaea, R.; Rubaiee, S.; Hanbazazah, A.S.; Jahrami, H.; Garbarino, S.; Damiani, G.; Wu, J.; Bragazzi, N.L. IL-4/13 Blockade and sleep-related adverse drug reactions in over 37,000 Dupilumab reports from the World Health Organization Individual Case Safety reporting pharmacovigilance database (VigiBase™): A big data and machine learning analysis. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 4074–4081. [Google Scholar] [CrossRef] [PubMed]
  72. Leonidis, A.; Korozi, M.; Sykianaki, E.; Tsolakou, E.; Kouroumalis, V.; Ioannidi, D.; Stavridakis, A.; Antona, M.; Stephanidis, C. Improving Stress Management and Sleep Hygiene in Intelligent Homes. Sensors 2021, 21, 2398. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  73. Abbasi, A.; Gupta, S.S.; Sabharwal, N.; Meghrajani, V.; Sharma, S.; Kamholz, S.; Kupfer, Y. A comprehensive review of obstructive sleep apnea. Sleep Sci. 2021, 14, 142–154. [Google Scholar] [PubMed] [PubMed Central]
  74. Veasey, S.C.; Rosen, I.M. Obstructive Sleep Apnea in Adults. N. Engl. J. Med. 2019, 380, 1442–1449. [Google Scholar] [CrossRef] [PubMed]
  75. Garbarino, S.; Magnavita, N.; Sanna, A.; Bragazzi, N.L. Estimating the hidden burden of obstructive sleep apnoea: Challenges and pitfalls. Lancet Respir. Med. 2020, 8, e1. [Google Scholar] [CrossRef] [PubMed]
  76. Gottlieb, D.J.; Punjabi, N.M. Diagnosis and Management of Obstructive Sleep Apnea: A Review. JAMA 2020, 323, 1389–1400. [Google Scholar] [CrossRef] [PubMed]
  77. McNicholas, W.T.; Korkalainen, H. Translation of obstructive sleep apnea pathophysiology and phenotypes to personalized treatment: A narrative review. Front. Neurol. 2023, 14, 1239016. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  78. Subramani, Y.; Singh, M.; Wong, J.; Kushida, C.A.; Malhotra, A.; Chung, F. Understanding Phenotypes of Obstructive Sleep Apnea: Applications in Anesthesia, Surgery, and Perioperative Medicine. Anesth. Analg. 2017, 124, 179–191. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  79. Malhotra, A.; Mesarwi, O.; Pepin, J.L.; Owens, R.L. Endotypes and phenotypes in obstructive sleep apnea. Curr. Opin. Pulm. Med. 2020, 26, 609–614. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  80. Bailly, S.; Grote, L.; Hedner, J.; Schiza, S.; McNicholas, W.T.; Basoglu, O.K.; Lombardi, C.; Dogas, Z.; Roisman, G.; Pataka, A.; et al. Clusters of sleep apnoea phenotypes: A large pan-European study from the European Sleep Apnoea Database (ESADA). Respirology 2021, 26, 378–387. [Google Scholar] [CrossRef] [PubMed]
  81. Schellenberg, J.B.; Maislin, G.; Schwab, R.J. Physical findings and the risk for obstructive sleep apnea. The importance of oropharyngeal structures. Am. J. Respir. Crit. Care Med. 2000, 162 Pt 1, 740–748. [Google Scholar] [CrossRef] [PubMed]
  82. Uniken Venema, J.A.M.; Rosenmöller, B.R.A.M.; de Vries, N.; de Lange, J.; Aarab, G.; Lobbezoo, F.; Hoekema, A. Mandibular advancement device design: A systematic review on outcomes in obstructive sleep apnea treatment. Sleep Med. Rev. 2021, 60, 101557. [Google Scholar] [CrossRef] [PubMed]
  83. Anitua, E.; Mayoral, P.; Almeida, G.Z.; Durán-Cantolla, J.; Alkhraisat, M.H. A Multicenter Prospective Study on the Use of a Mandibular Advancement Device in the Treatment of Obstructive Sleep Apnea. Dent. J. 2023, 11, 247. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Zheng, D.H.; Wang, X.X.; Ma, D.; Zhou, Y.; Zhang, J. Upper airway asymmetry in skeletal Class III malocclusions with mandibular deviation. Sci. Rep. 2017, 7, 12185. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  85. Carberry, J.C.; Amatoury, J.; Eckert, D.J. Personalized Management Approach for OSA. Chest 2018, 153, 744–755. [Google Scholar] [CrossRef] [PubMed]
  86. Kubin, L. Neural Control of the Upper Airway: Respiratory and State-Dependent Mechanisms. Compr. Physiol. 2016, 6, 1801–1850. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  87. Rukhadze, I.; Fenik, V.B. Neuroanatomical Basis of State-Dependent Activity of Upper Airway Muscles. Front. Neurol. 2018, 9, 752. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  88. Duong-Quy, S.; Nguyen-Huu, H.; Hoang-Chau-Bao, D.; Tran-Duc, S.; Nguyen-Thi-Hong, L.; Nguyen-Duy, T.; Tang-Thi-Thao, T.; Phan, C.; Bui-Diem, K.; Vu-Tran-Thien, Q.; et al. Personalized Medicine and Obstructive Sleep Apnea. J. Pers. Med. 2022, 12, 2034. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Nokes, B.; Baptista, P.M.; de Apodaca, P.M.R.; Carrasco-Llatas, M.; Fernandez, S.; Kotecha, B.; Wong, P.Y.; Zhang, H.; Hassaan, A.; Malhotra, A. Transoral awake state neuromuscular electrical stimulation therapy for mild obstructive sleep apnea. Sleep Breath. 2023, 27, 527–534. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  90. Labarca, G.; Schmidt, A.; Dreyse, J.; Jorquera, J.; Enos, D.; Torres, G.; Barbe, F. Efficacy of continuous positive airway pressure (CPAP) in patients with obstructive sleep apnea (OSA) and resistant hypertension (RH): Systematic review and meta-analysis. Sleep Med. Rev. 2021, 58, 101446. [Google Scholar] [CrossRef] [PubMed]
  91. Labarca, G.; Schmidt, A.; Dreyse, J.; Jorquera, J.; Barbe, F. Telemedicine interventions for CPAP adherence in obstructive sleep apnea patients: Systematic review and meta-analysis. Sleep Med. Rev. 2021, 60, 101543. [Google Scholar] [CrossRef] [PubMed]
  92. Perdomo, C.M.; Cohen, R.V.; Sumithran, P.; Clément, K.; Frühbeck, G. Contemporary medical, device, and surgical therapies for obesity in adults. Lancet 2023, 401, 1116–1130. [Google Scholar] [CrossRef] [PubMed]
  93. An, H.J.; Baek, S.H.; Kim, S.W.; Kim, S.J.; Park, Y.G. Clustering-based characterization of clinical phenotypes in obstructive sleep apnoea using severity, obesity, and craniofacial pattern. Eur. J. Orthod. 2020, 42, 93–100. [Google Scholar] [CrossRef] [PubMed]
  94. Palmer, L.J.; Redline, S. Genomic approaches to understanding obstructive sleep apnea. Respir. Physiol. Neurobiol. 2003, 135, 187–205. [Google Scholar] [CrossRef] [PubMed]
  95. Mukherjee, S.; Saxena, R.; Palmer, L.J. The genetics of obstructive sleep apnoea. Respirology 2018, 23, 18–27. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  96. Conte, L.; Greco, M.; Toraldo, D.M.; Arigliani, M.; Maffia, M.; De Benedetto, M. A review of the “OMICS” for management of patients with obstructive sleep apnoea. Acta Otorhinolaryngol. Ital. 2020, 40, 164–172. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  97. Khalyfa, A.; Marin, J.M.; Sanz-Rubio, D.; Lyu, Z.; Joshi, T.; Gozal, D. Multi-Omics Analysis of Circulating Exosomes in Adherent Long-Term Treated OSA Patients. Int. J. Mol. Sci. 2023, 24, 16074. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  98. Templier, L.; Rossi, C.; Miguez, M.; Pérez, J.C.; Curto, A.; Albaladejo, A.; Vich, M.L. Combined Surgical and Orthodontic Treatments in Children with OSA: A Systematic Review. J. Clin. Med. 2020, 9, 2387. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  99. Fank, F.; Artismo, R.S.; de Santana, M.G.; Esteves, A.M.; Matte, D.L.; Mazo, G.Z. Effects of combined exercise training with sleep education in older adults with obstructive sleep apnea: Protocol for a randomized clinical trial. Front. Psychol. 2024, 15, 1322545. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  100. Rokou, A.; Eleftheriou, A.; Tsigalou, C.; Apessos, I.; Nena, E.; Dalamaga, M.; Voulgaris, A.; Steiropoulos, P. Effect of the Implementation of a Structured Diet Management Plan on the Severity of Obstructive Sleep Apnea: A Systematic Review. Curr. Nutr. Rep. 2023, 12, 26–38. [Google Scholar] [CrossRef] [PubMed]
  101. Al Oweidat, K.; Toubasi, A.A.; Tawileh, R.B.A.; Tawileh, H.B.A.; Hasuneh, M.M. Bariatric surgery and obstructive sleep apnea: A systematic review and meta-analysis. Sleep Breath 2023, 27, 2283–2294. [Google Scholar] [CrossRef] [PubMed]
  102. Randerath, W.; Verbraecken, J.; de Raaff, C.A.L.; Hedner, J.; Herkenrath, S.; Hohenhorst, W.; Jakob, T.; Marrone, O.; Marklund, M.; McNicholas, W.T.; et al. European Respiratory Society guideline on non-CPAP therapies for obstructive sleep apnoea. Eur. Respir. Rev. 2021, 30, 210200. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  103. Lee, Y.C.; Lu, C.T.; Chuang, L.P.; Lee, L.A.; Fang, T.J.; Cheng, W.N.; Li, H.Y. Pharmacotherapy for obstructive sleep apnea—A systematic review and meta-analysis of randomized controlled trials. Sleep Med. Rev. 2023, 70, 101809. [Google Scholar] [CrossRef] [PubMed]
  104. Sutherland, K.; Almeida, F.R.; de Chazal, P.; Cistulli, P.A. Prediction in obstructive sleep apnoea: Diagnosis, comorbidity risk, and treatment outcomes. Expert. Rev. Respir. Med. 2018, 12, 293–307. [Google Scholar] [CrossRef] [PubMed]
  105. Lim, D.C.; Sutherland, K.; Cistulli, P.A.; Pack, A.I. P4 medicine approach to obstructive sleep apnoea. Respirology 2017, 22, 849–860. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  106. Zinchuk, A.V.; Gentry, M.J.; Concato, J.; Yaggi, H.K. Phenotypes in obstructive sleep apnea: A definition, examples and evolution of approaches. Sleep Med. Rev. 2017, 35, 113–123. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  107. Arachchige, M.A.; Steier, J. Beyond Usual Care: A Multidisciplinary Approach Towards the Treatment of Obstructive Sleep Apnoea. Front. Cardiovasc. Med. 2022, 8, 747495. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  108. Hwang, D. Monitoring Progress and Adherence with Positive Airway Pressure Therapy for Obstructive Sleep Apnea: The Roles of Telemedicine and Mobile Health Applications. Sleep Med. Clin. 2016, 11, 161–171. [Google Scholar] [CrossRef] [PubMed]
  109. Spicuzza, L.; Caruso, D.; Di Maria, G. Obstructive sleep apnoea syndrome and its management. Ther. Adv. Chronic Dis. 2015, 6, 273–285. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  110. Zinchuk, A.; Yaggi, H.K. Phenotypic Subtypes of OSA: A Challenge and Opportunity for Precision Medicine. Chest 2020, 157, 403–420. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  111. Parrino, L.; Halasz, P.; Szucs, A.; Thomas, R.J.; Azzi, N.; Rausa, F.; Pizzarotti, S.; Zilioli, A.; Misirocchi, F.; Mutti, C. Sleep medicine: Practice, challenges and new frontiers. Front. Neurol. 2022, 13, 966659. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  112. Askland, K.; Wright, L.; Wozniak, D.R.; Emmanuel, T.; Caston, J.; Smith, I. Educational, supportive and behavioural interventions to improve usage of continuous positive airway pressure machines in adults with obstructive sleep apnoea. Cochrane Database Syst. Rev. 2020, 4, CD007736. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  113. Mijnster, T.; Boersma, G.J.; Meijer, E.; Lancel, M. Effectivity of (Personalized) Cognitive Behavioral Therapy for Insomnia in Mental Health Populations and the Elderly: An Overview. J. Pers. Med. 2022, 12, 1070. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  114. Haiech, J.; Kilhoffer, M.C. Personalized medicine and education: The challenge. Croat. Med. J. 2012, 53, 298–300. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  115. Aguilera-Cobos, L.; García-Sanz, P.; Rosario-Lozano, M.P.; Claros, M.G.; Blasco-Amaro, J.A. An innovative framework to determine the implementation level of personalized medicine: A systematic review. Front. Public Health 2023, 11, 1039688. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  116. Glackin, S.N.; d’Ancona, G. Ethical, philosophical, and practical considerations in adherence to therapy in sleep medicine. Curr. Opin. Pulm. Med. 2019, 25, 609–613. [Google Scholar] [CrossRef] [PubMed]
  117. Martin, L.R.; Williams, S.L.; Haskard, K.B.; Dimatteo, M.R. The challenge of patient adherence. Ther. Clin. Risk Manag. 2005, 1, 189–199. [Google Scholar] [PubMed] [PubMed Central]
  118. Martin, L.R.; Feig, C.; Maksoudian, C.R.; Wysong, K.; Faasse, K. A perspective on nonadherence to drug therapy: Psychological barriers and strategies to overcome nonadherence. Patient Prefer. Adherence 2018, 12, 1527–1535. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  119. Flores, M.; Glusman, G.; Brogaard, K.; Price, N.D.; Hood, L. P4 medicine: How systems medicine will transform the healthcare sector and society. Pers. Med. 2013, 10, 565–576. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  120. Amram, D.; Cignoni, A.; Banfi, T.; Ciuti, G. From P4 medicine to P5 medicine: Transitional times for a more human-centric approach to AI-based tools for hospitals of tomorrow. Open Res. Eur. 2022, 2, 33. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  121. Gorini, A.; Pravettoni, G. P5 medicine: A plus for a personalized approach to oncology. Nat. Rev. Clin. Oncol. 2011, 8, 444. [Google Scholar] [CrossRef] [PubMed]
  122. Bragazzi, N.L. From P0 to P6 medicine, a model of highly participatory, narrative, interactive, and “augmented” medicine: Some considerations on Salvatore Iaconesi’s clinical story. Patient Prefer. Adherence 2013, 7, 353–359. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Table 1. Conceptualization of personalized sleep medicine: an overview of major concepts, advancements, applications, and technologies in patient-centered sleep medicine.
Table 1. Conceptualization of personalized sleep medicine: an overview of major concepts, advancements, applications, and technologies in patient-centered sleep medicine.
Concept/Advancement/Application/TechnologyDescription
Personalized ApproachTailoring sleep-related healthcare to individual needs, recognizing the unique physiological and psychological characteristics of each person
Taking into Account Different Influencing FactorsConsidering genetic predispositions, lifestyle habits, environmental influences, and underlying health conditions
Technology Integration (Wearable Devices and IoT/IoMT)Making use of sophisticated devices for non-intrusive monitoring of sleep patterns, including wearable devices, mobile health applications, and advanced diagnostic tools for detailed data collection and analysis; integration with IoT/IoMT for optimizing sleep environments
Data Analysis Techniques (Machine Learning and AI in Sleep Analysis)Employing machine learning and AI for interpreting large datasets and providing customized treatment plans, including utilization of non-contact devices and advanced algorithms (machine learning/deep learning techniques) for personalized sleep stage estimation, analysis of sleep patterns, and circadian rhythm analysis
Circadian Rhythms and Sleep PhysiologyResearching these areas and contributing to a deeper understanding of sleep’s impact on health to inform personalized treatment approaches in an evidence-based, data-driven fashion
Advanced Data Integration and AnalysisEmphasizing the integration of genetic, environmental, and lifestyle data for comprehensive sleep assessments and treatments
Biomarker (Genomic and Molecular) ResearchExploring individual sleep disorders and determinants of sleep health, leading to personalized treatment plans
Computational and Mathematical Models for Sleep Disorders, Including Patient-specific 3D Computational ModelsCreating patient-specific treatments for conditions like obstructive sleep apnea using computational models
Customized Sleep Pharmacogenetics and PharmacotherapyDelivering personalized and data-driven insights for the selection of drugs; customizing dosages and timing of sleep medication based on genetic predispositions, molecular pathways, metabolism, and underlying health status; and providing continuous pharmacological monitoring and feedback
Customized Non-pharmacological InterventionsIncorporating advice on sleep hygiene, dietary changes, and stress management techniques to complement pharmacological treatments
Cross-disciplinary and Multidisciplinary CollaborationsCollaboration among various experts and specialists (like dentists, surgeons, neurologists, pulmonologists, otorhinolaryngologists, and dietitians) for a comprehensive approach to treating complex sleep disorders, innovative solutions, and a deeper understanding of sleep issues
Regulatory and Ethical Considerations.Addressing concerns related to privacy, data security, and the use of genetic/biometric data
Educational Initiatives and Public AwarenessIncreasing awareness about personalized sleep health, leading to earlier recognition and proactive management of sleep disorders
Compliance and Patient EngagementEnsuring adherence to treatment plans through education, support systems, and regular follow-up
Longitudinal Studies and Continuous MonitoringProviding insights into the evolution of sleep patterns over time, refining personalized treatment approaches
Telemedicine and Virtual Healthcare PlatformsExpanding services for accessible personalized sleep medicine, especially beneficial for remote or underserved areas.
Holistic and Integrative ApproachesIntegrating mental health interventions and cognitive behavioral therapy in sleep medicine
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Garbarino, S.; Bragazzi, N.L. Revolutionizing Sleep Health: The Emergence and Impact of Personalized Sleep Medicine. J. Pers. Med. 2024, 14, 598. https://doi.org/10.3390/jpm14060598

AMA Style

Garbarino S, Bragazzi NL. Revolutionizing Sleep Health: The Emergence and Impact of Personalized Sleep Medicine. Journal of Personalized Medicine. 2024; 14(6):598. https://doi.org/10.3390/jpm14060598

Chicago/Turabian Style

Garbarino, Sergio, and Nicola Luigi Bragazzi. 2024. "Revolutionizing Sleep Health: The Emergence and Impact of Personalized Sleep Medicine" Journal of Personalized Medicine 14, no. 6: 598. https://doi.org/10.3390/jpm14060598

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop