Next Article in Journal
Mepolizumab Improves Outcomes of Chronic Rhinosinusitis with Nasal Polyps in Severe Asthmatic Patients: A Multicentric Real-Life Study
Previous Article in Journal
Escaping the Reality of the Pandemic: The Role of Hopelessness and Dissociation in COVID-19 Denialism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Next-Generation Personalized Medicine: Implementation of Variability Patterns for Overcoming Drug Resistance in Chronic Diseases

Department of Medicine, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem POB12000, Israel
J. Pers. Med. 2022, 12(8), 1303; https://doi.org/10.3390/jpm12081303
Submission received: 30 June 2022 / Revised: 5 August 2022 / Accepted: 8 August 2022 / Published: 10 August 2022

Abstract

:
Chronic diseases are a significant healthcare problem. Partial or complete non-responsiveness to chronic therapies is a significant obstacle to maintaining the long-term effect of drugs in these patients. A high degree of intra- and inter-patient variability defines pharmacodynamics, drug metabolism, and medication response. This variability is associated with partial or complete loss of drug effectiveness. Regular drug dosing schedules do not comply with physiological variability and contribute to resistance to chronic therapies. In this review, we describe a three-phase platform for overcoming drug resistance: introducing irregularity for improving drug response; establishing a deep learning, closed-loop algorithm for generating a personalized pattern of irregularity for overcoming drug resistance; and upscaling the algorithm by implementing quantified personal variability patterns along with other individualized genetic and proteomic-based ways. The closed-loop, dynamic, subject-tailored variability-based machinery can improve the efficacy of existing therapies in patients with chronic diseases.

1. Introduction

Treatment of chronic diseases is a significant health burden. Over a third of patients with various chronic diseases develop partial or complete resistance to chronic therapies, necessitating increased dosages, additional treatments, or replacing medications with newer, typically expensive drugs, which are not always associated with a better response [1].
Variability patterns are inherent to many biological systems [2]. We review the concept of using a closed-loop, dynamic, individualized variability-based machinery for improving the efficacy of existing drugs in patients with chronic diseases.

2. Treatment of Chronic Diseases Is a Significant Worldwide Health Burden

The World Health Organization (WHO) defines chronic disease as slow progression, long duration, and not transferable from person to person [3]. The Global Burden of Disease study reported a significant growth in the years lived with disability (YLD) over the last decade [4,5]. The prevalence of chronic condition multi-morbidity increases with age. About 40% of subjects over 44 suffer from a chronic disease. The percentages rise to 50% for 65–74-year olds and 70% for individuals over 85 years of age [5,6]. In 2014, 60% of Americans suffered from at least one chronic condition, and 42% had multiple chronic conditions [7]. Chronic diseases are the most significant cause of death globally, accounting for 7 of 10 deaths each year [8,9]. Cardiovascular diseases, hypertension, diabetes, stroke, depression, cancer, chronic respiratory diseases, epilepsy, fatty liver, obesity, mental health, and rheumatologic diseases denote some of the most common and costly chronic diseases [8,10,11]. Population aging leads to a surge in people suffering from chronic diseases [12]. The Center for Disease Control (CDC) estimates that 90% of the world’s USD 3.3 trillion in yearly health care expenditure is for individuals with chronic disease [13]. The direct cost of handling chronic disease was USD 1.1 trillion in the U.S. in 2016, equal to 5.8 percent of the gross domestic product (GDP) [14].

3. Drug Resistance Is Associated with Partial or Complete Loss of Effect and with Poor Prognosis in Patients with Chronic Diseases

Adaptation to prolonged exposure to drugs or development of any tolerance and tachyphylaxis prohibit the maximal and long-lasting effects of drugs [1]. Drug tolerance describes a subject’s reduced reaction to a drug after repeated use [15]. Resistance refers to the ability of a target subcellular, cellular, or whole organ to resist the effects of a drug that is usually effective against them [16]. The reduced response to medication following recurrent dosing reflects an adaptive body’s response to continued exposure [15]. While augmenting dosages may re-increase the response, it can accelerate the tolerance or increase side effects [17].
Several mechanisms underlie resistance, which is not identical for all drugs/subjects/diseases [18,19]. Adaptation and habituation to therapy can be at the levels of subcellular, cell organelles, and organs. The extent of adaptation depends on the disease, the drug, individual genetic and other factors, and the type of medication, dosage, and duration of treatment [20,21,22,23]. Circadian rhythms due to endocrinological or other mechanisms partly underlie drug resistance [24,25].
Pharmacokinetic or metabolic types of tolerance may occur due to the induction of drug-metabolizing enzymes. This type of tolerance happens due to a reduced amount of the substance reaching its target [26,27]. The therapeutic effect directed to an exact target at the molecular level depends on four variables: absorption, distribution, metabolism, and excretion (ADME). These parameters determine drug properties’ efficacy, duration of action, and side effects. The disturbance of one or more ADME elements can lead to drug resistance [28]. Polymorphic variants in several ADME genes are recognized for their role in toxicity [29,30]. The induction of enzymes is partially accountable for the tolerance; a recurrent drug administration reduces its outcome. For some medicines, metabolism with UDP-glucuronosyltransferases (UGTs) can transform drug substrates into polar metabolites, which are improved compounds for the various transporters compared with the original drug. UGT-linked resistance is related to enzyme overexpression. Studies showed that multidrug resistance is glucuronidation for medications used to manage hypercholesterolemia, hypertension, epilepsy, and psychiatric diseases [28]. The potential role of UGT1A1 *28/*28 in reducing the conversion of irinotecan to inactive metabolites requires testing and dosing adjustments based on the UGT1A1 genotypes before starting the chemotherapy schedule based on irinotecan [31].
Induction of CYP 450 enzymes by smoking (CYP1A2) and exposure to other drugs (CYP3A4) are additional examples of tolerance [32].
Pharmacodynamics or functional tolerance is due to “adaptation” of the drugs’ targets, such as losing receptor sensitivity. Repeated use reduces the cellular response to a substance. A common cause is increased compound concentrations continually binding to the receptor, desensitizing it via continual interaction [17]. A reduction in receptor density and mechanisms leading to action potential firing rates are additional causes. The majority of pharmacodynamic tolerance ensues following continued exposure to a medication. However, occurrences of rapid tolerance can occur [33]. Functional tolerance can end up with a complete loss of drug activity.
Behavioral tolerance happens following the administration of psychoactive drugs. In these circumstances, tolerance to a behavioral effect of a medication, for example, methamphetamine-associated increased motor activity, is a result of regular use. It happens due to a pharmacodynamic tolerance in the brain or via drug-independent learning [34]. Tachyphylaxis is an abrupt onset of tolerance to medications, which is not dose-dependent. It is a form of short-term tolerance following the dosing [35]. Opioid-induced hyperalgesia is another common form of tolerance [36].
Partial or complete drug resistance to chronic medications significantly impacts the outcome of chronic diseases and is associated with poor prognosis. Attempts to improve adherence to chronic therapy do not necessarily improve the disease outcome; they may increase non-responsiveness. Examples include the higher expenditures and hospitalization rates for patients with insulin resistance compared to non-insulin-resistant patients [37] and the increased mortality of patients with resistance to pain killers [38].

4. Drug Resistance in Common Chronic Diseases

Described below are typical examples of drug resistance that significantly impact both patients and society.

4.1. Loss of Effect for Antiepileptic Drugs

Epilepsy is diagnosed in more than 70 million people worldwide. Introducing new anti-seizure drugs did not assist one-third of patients who continue to suffer from seizures that are refractory to pharmacotherapy [39]. Nearly all first-, second-, and third-generation antiepileptic medications lose their efficacy, to some extent, during continued treatment [40]. The pharmacokinetics, target, intrinsic severity, gene variant, neural network, and transporter hypotheses may explain this phenomenon. Increasing doses, replacing the drugs, and brain stimulation and surgery are used in these patients with modest success [41,42,43].

4.2. Loss of Effect for Drugs That Work on the Heart and Blood Vessels

Nitrates effectively relieve angina pectoris symptoms in acute settings and prevent angina. However, their chronic use is associated with tolerance [44]. Tolerance to the effect of beta-blockers is well established following prolonged use. A dose-related loss of cardioselectivity of metoprolol has also been described [25,45].

4.3. Diuretic Resistance in Patients with Congestive Heart Failure Showing Diuretic Resistance

Heart failure (HF) is a growing epidemic associated with high morbidity, mortality, and healthcare expenditures. It affects 6 million people in the United States and represents a leading cause of cardiovascular mortality [46]. Volume overload symptoms are considered part of the morbidity, and reduced quality of life associated with HF. Loop diuretics are the mainstay in the treatment of volume overload. Diuretic resistance is a failure to reach congestion relief despite proper or escalating doses of diuretics [25,47]. Resistance to loop diuretics results from multiple variables associated with constant daily administration of equal or higher dosages [48]. Some of the measures taken in these patients are using higher dosages of diuretics, intravenous administration, or adding sequential nephron blockade by using combinations of two or more compounds from other classes of diuretics. These measures can further increase resistance [47].

4.4. Insulin Resistance in Subjects with Diabetes

Impaired response to exogenous insulin characterizes insulin resistance in type 2 diabetes mellitus. Patients who require more than 1 unit/kg/day are classified as having insulin resistance, and those requiring above 2 units/kg/day are classified as severe resistance. Alternatively, a daily insulin dose above 200 units is considered severe insulin resistance [49,50]. A growing number of subjects develop severe insulin resistance necessitating large doses of insulin [49]. Insulin resistance is due to an effect on the insulin receptor requiring an increase in insulin dose in patients with type 2 diabetes over time [51,52]. Treating subjects with severe resistance is a significant challenge as it is hard to control their glucose levels reasonably. Loss of the effect of glucagon-like peptide-1 (GLP-1) analogs, glucokinase activators, and dipeptidyl peptidase 4 (DPP4) inhibitors has been described in some patients [53,54].

4.5. Loss of Effect in Antidepressant and Anti-Psychotic Medications

Antidepressant resistance occurs in depressed subjects who lose their response to a formerly effective antidepressant therapy while receiving similar drugs and dosages [55,56]. Depression is classified as resistant when at least two attempts with antidepressants from different classes, proper doses, duration, and compliance do not achieve remission. A substantial proportion of subjects with depression ultimately develop treatment-resistant or refractory depression (TRD) [57,58]. The current therapy for TRD involves evaluating comorbid medical and psychiatric conditions and attempts to enhance antidepressant efficacy by augmentation, combination, or switching of anti-depressants [57,59]. However, a majority of TRD patients do not adequately respond. Anti-psychotic tolerance refers to a decrease in drug effect resulting from chronic use and the brain’s adaptive response to the foreign drug property [60]. “Drug learning and memory” notions were suggested to underlie the development of resistance, reflecting associative and non-associative processing that are affected by behavioral, environmental, and pharmacological variables. Drug-induced neuroplasticity, associated with functional alterations of the receptors and signaling pathways of prefrontal serotonin (5-HT)2A and striatal dopamine D2 receptors are possible mechanisms [60]. An “irregular” or “pulse therapy” approach might not be appropriate for some conditions, such as depression, where inconsistent drug use is a risk factor for disease relapse and other complications.

4.6. Loss of the Effect of Anti-Cancer Medications

Drug resistance in oncology is a multifactorial phenomenon [61]. Tumors show an acceptable response following the initial administration of chemotherapeutic drugs. However, they may become resistant after repeated treatments [62,63]. Cancer cells may resist chemotherapy due to spontaneous mutations in any growing cell subset, whether exposed to treatment or not [17]. When a drug destroys healthy cells, a higher percentage of the survivors may become resistant. Pharmacogenomic, pharmacokinetic, pharmacodynamic factors, and drug selection and dosing determine resistance. Exosomal miRNAs also contribute to the development of drug resistance [62]. Epidermal growth factor receptor (EGFR) inhibitors, panitumumab, and cetuximab used to treat metastatic colorectal cancer expressing wild-type KRAS show a beneficial effect in only 10–20% of patients [64,65]. Drug resistance limits the effectiveness of therapies for lung cancer and is associated with tumor heterogeneity [66]. The resistance acquired after partial administration of chemotherapy is associated with developing more aggressive clones. Mutations may be spontaneous but are induced mainly by exposure to drugs [61]. Intratumoral heterogeneity contributes to the inconsistency of responses to chemotherapy, which is not captured by most current approaches based on cancer cell biomarkers. Some contributing factors to the development of drug resistance are as follows: development of alternative pathways for growth activation; interpatient variability of genetic and epigenetic factors; altered differentiation pathways; alterations in drug targets; alterations of the local physiology of tumor, such as blood supply; behavior of neighboring cells; the anti-tumor immune response; drug transport; intracellular distribution; and apoptosis inhibition and increased enzymatic activity [67,68].

4.7. Loss of Effectivity in Treating Neurological Disorders

Disease-modifying therapies for multiple sclerosis, such as immunomodulatory drugs, have high variability in efficacy. Individual response to current medications significantly varies across patients; moreover, 30–80% discontinue therapy [69,70].

4.8. Loss of Effect for Painkillers

Drug tolerance to chronic analgesics necessitates increasing dosages [14]. Following tobacco, alcohol, and marijuana, the most commonly abused drugs are methylphenidate (Ritalin), Diazepam (Valium®), and oxycodone (OxyContin®). Opiates are the primary approach for chronic pain therapy in cancer patients suffering from moderate to severe pain and in patients with non-cancer chronic pain. Patients with chronic pain often develop tolerance to opioids over time with aggravated pain [71]. Prolonged administration of opiates is associated with developing antinociceptive tolerance. Higher doses are mandatory overtime for reaching the same degree of analgesia [72]. Sustained exposure to morphine results in paradoxical pain, which occurs in regions not affected by the original pain resulting in dose escalation, termed ‘analgesic tolerance’ [72,73]. Narcotic medications downregulate the Mu receptors in the brain. With fewer receptors, it takes more narcotic-like molecules for subjects to obtain the same feeling. This down-regulation leads to tolerance and a need for increased dosages over time to achieve pain relief [74,75,76].

4.9. Loss of the Effect of Immunomodulatory and Anti-Inflammatory Drugs

Partial or complete loss of anti-TNF drugs’ impact occurs in patients with inflammatory bowel diseases (IBD), rheumatoid arthritis, and psoriasis [77,78]. Loss of response (LOR) to anti-TNF therapy is common in IBD patients [79]. The incidence of LOR among adult IBD patients is 36% [80,81,82]. About 50% of subjects with Crohn’s disease or ulcerative colitis develop LOR to infliximab following an early response to the drug [82]. There is no difference in time to LOR between subjects treated using regimens comprising several drugs or different anti-TNF agents [83]. Switching between anti-TNF agents, dose intensification, and adding an immunomodulator to suppress immunogenicity may overcome LOR with moderate success [84,85].

5. Averages-Based Treatment Regimens Are Associated with Non-Responsiveness, and Current Measures of Personalized Medicine Are Insufficient to Overcome It

Treatment of chronic diseases commonly follows a pre-determined regimen. It is carried out based on protocols within the therapeutic and efficacy windows. Once a treatment regimen is prescribed/configured, it stays identical until complete or non-responsiveness occurs. Most drugs and therapies developed for the “average patient” are insufficient for most subjects. However, patients respond differently to similar treatments. Using averages to determine drugs’ effects leaves high proportions of patients as partial or complete non-responders [86,87,88]. Only a relatively small number of subjects respond to the medication, exposing others to the risk of side effects through ineffective therapies [89]. Using averages in medicine is insufficient for developing personalized approaches. However, there are no valid methods for most diseases to determine which treatment is best for an individual patient [1,90].
Differences in multiple parameters drive inter-individual variation in drug response. Gene interactions are studied using gene regulatory networks, RNA velocity, and single-cell sequencing of thousands of cells. Single-cell data combined with big data can reconstruct personalized, cell-type, and context-specific gene regulatory networks [89,91]. However, environmental exposures, proportions of cell types involved, patients’ genetic background, and variabilities in proteomics and metabolomics are nearly impossible to control simultaneously.
Therefore, attempts to use “personalized” measures, such as genomics, proteomics, metabolomics, and others, are only partially successful in overcoming the challenge of improving response to chronic therapies.

6. Variability Is Inherent to Biological Systems: Loss/Change in Variability Patterns Leads to Poor Prognosis

Intra- inter-patient variabilities are inherent to biological systems, with their dynamics affected by both intrinsic and extrinsic sources [92,93]. These systems manifest a high rate of uncertainties regarding the specific sources of variabilities evolving from multiple genetic, biochemical, and metabolic variables [94,95,96,97].
Variability exists to form the levels of genes and molecules to that of whole organs [2,94,95,96,98,99,100,101]. The stochastic behavior resulted from collisions among molecules within the entire cellular compartment and characterized all living systems [102]. Intrinsic and extrinsic stochasticity leads to single-cell variability in gene expression—the intrinsic stochasticity results from randomness in gene expression processes and mRNA and protein synthesis pathways. Extrinsic alterations mirror the status of systems and their communications with the intracellular and extracellular environments [102].
Variability is the hallmark of microtubules’ function. The microtubules constitute the cellular cytoskeleton, and their dynamic instability is a feature of biological variability that characterizes their function. Their dynamic behavior constitutes the basis for multiple biological processes contributing to cellular plasticity and cell signaling [100,101].
Cell death processes manifest variability. Not all cells expire at the same treatment dose or at the same time of a chemotherapeutic drug. This cell-to-cell variability results from differences in apoptosis signaling networks and intracellular and extracellular parameters [103]. Intra- and inter-cell variabilities occur to express cell epitopes, cell singling pathways, and cytokine secretion, suggesting a marked heterogeneity in cells from the same person [91].
Variability is inherent to the function of whole organs. Patterns of variability are associated with normal physiology and health. Examples of variability inherent to healthy organs’ functions include variations in heart rate [104], breathing [105,106], gate, and blood glucose levels [107]. Changes in normal physiologic variability loss lead to disease states and bad outcomes [107,108,109,110].

7. Intra- and Inter-Patient Variabilities for Pharmacodynamics, Drug Metabolism, and Response

A high inter- and intra-patient variability characterizes drug responsiveness, metabolism, and pharmacodynamics. This variability is associated with partial or even complete loss of drug effectiveness [111,112,113,114]. The cell is crowded with uneven distribution of macromolecules that interrelate with a drug in multiple specific and non-specific ways. This phenomenon results in a high heterogeneity in drug response between cells [115]. A study showed marked daily variability in antiepileptic levels in subjects stabilized with the same drug over time [116]. High intra-patient variability to tacrolimus, an immunosuppressive drug, was associated with graft rejection [114].
Interindividual variability in drug efficacy and toxicity is a significant challenge in designing personalized therapeutic regimens. Variations in drug pharmacokinetics (PKs) and pharmacodynamics (PDs) are partially a result of the polymorphic variants in genes encoding drug metabolizing enzymes and transporters and genes encoding drug receptors. Pharmacogenomics (PGx) assists in selecting biomarkers of the pharmacology variables of genome-drug interactions enabling personalizing therapies [117].
Regular drug dosing regimens are incompatible with physiological variability and may contribute to resistance to the effect of medications. A “drug holiday” can sometimes overcome tolerance [118]. The successful design of therapies necessitates looking into the intrinsic variabilities in the responses to medications [1,119].

8. Applying Variability to Biological Systems

The chaos theory describes deterministic systems with predictable behavior; however, these systems may become random [120,121]. The theory focuses on activities of non-linear dynamic systems that are sensitive to the primary settings [122,123,124]. In chaotic systems, minor differences in the opening conditions may lead to different outcomes. Whether these systems are deterministic, their long-term behavior prediction is difficult [125].
The interplay between disorder and order in a chaotic system is a significant task in biology [94,95,96]. Chaos theory enables quantifying the degree of order in biological systems. Mathematical formulations based on chaos theory are somewhat appropriate to biology [126]. Several concepts of this theory, including fractal dimension, entropy, and algorithms for obtaining quantitative characteristics of the degree of the order, have been applied [127].
Based on the complexity and chaos theories, systems biology applies models to forecast the associations between genes, proteins, and different variables in the external milieus of these systems [128]. Network medicine further adapts systems biology to clinical sciences [128]. Combining high-throughput data collection within molecular or higher-order systems modeling can improve the results [129]. Data-driven techniques can determine chaotic and random systems and assess their dynamics [94,95,96,130,131]. Network models can establish scale-free features, reproducing the networks’ structures for the interactions between proteins [132]. Studies showed this concept applies to several physiological processes, including neuronal activity, breathing, heart rate, and electroencephalograms [127].
Recent studies described the chaotic modeling approaches from the molecular to whole organs, including cardiac rhythms, brain dynamics systems [133], eye tracking disorder in schizophrenics [134], and warning signs of fetal hypoxia [135]. Genome chaos is the process of complex, rapid genome re-organization, leading to chaotic genomes, followed by establishing stable genomes [91,136]. Mathematical models predict calcium oscillations in vascular smooth muscle [137]. An algorithm of the bone-density stress adaptation model involves a chaos mechanism [138]. Chaos approaches helped uncover a peptide or RNA state of integrated protometabolism. Chaos theory revealed the mechanism that drives genetic heterogeneity observed in tumors [139].

9. A Three-Phase Roadmap for Developing a Platform for Overcoming Drug Resistance in Patients with Chronic Diseases

Recent studies described the development of a platform for overcoming drug resistance based on the following three phases [1,140,141]: introducing irregularity in therapeutic regimens for improving drug responsiveness; establishing a closed-loop algorithm for generating individualized patterns of irregularity to overcome drug resistance; upscaling the algorithm by implementing quantified personal variability patterns along with additional personalized signatures based on genes, proteins, and other disease or host relevant variables [1,25,43,68,76,85,98,99,100,101,142,143,144,145,146,147,148,149,150,151].
Figure 1 illustrates a three-step approach for introducing a system for overcoming resistance to chronic drugs.
  • Step A: Using irregularity to overcome drug resistance: Implementing treatment regimens based on aperiodic regimens of taking the drugs at irregular intervals and strengths.
  • Step B: Establish a closed-loop algorithm for generating individualized patterns of irregularity for overcoming drug resistance: The closed-loop algorithm provides a method for overcoming the loss of response to drugs by setting up an irregularity within a specific range that is determined in a subject-tailored way. The algorithm reaches a physiological target. The algorithm receives inputs from the user and other users to update the treatment regimen.
  • Step C: Upscaling the algorithm by implementing quantified personal variability patterns and additional personalized signatures: Cellular and whole organ patterns of variability are quantified and implemented into the algorithm. A dataset of variability patterns at the cellular levels using single cell-based techniques is generated from cells harvested from patients before and after chronic disease therapy, including single-cell RNA sequencing, proteomics, metabolomics, and epitope expression. Methods for quantifying these inherent variability patterns and combining single cells with whole organ variability patterns generate an individualized factor implemented into the treatment algorithm. The quantified variability patterns are implemented into individualized-based treatment regimens. The algorithm continuously alters the irregular regimen based on the patient’s closed-loop feedback on the therapy’s effect.

9.1. Using Irregularity to Overcome Drug Resistance

Regular administration of a constant daily dose, or a continuous increase in drug dose, is more likely to be associated with resistance to medications when compared with irregular administration of the same or altering dose [152]. Using treatment regimens based on nonperiodic routines of drug administration, and using variability in the intervals and strengths can reduce the likelihood of resistance and improve drug effectiveness [152].
A recent study described a pre-clinical model documenting variability in response to immunomodulatory drugs. Using the immune-mediated hepatitis model of Concanavalin A (ConA) and treatment with two immunomodulators (e.g., anti-CD3 or glucosylceramide (GC) were studied [142]. In multiple consecutive studies, the study showed an individualized response pattern of reaction to an injection of ConA and oral administration of immunomodulatory agents. As measured by liver enzymes, improvement of the liver injury showed marked intra-group and inter-experiment variabilities. Similarly, the data showed marked variability in the response for serum cytokine levels and lymphocyte subsets. Using irregularity in administering steroids and anti-LPS antibodies was superior in alleviating immune-mediated damage in this model compared to regular drug administration [142].
Clinical trials are ongoing to determine the use of irregularity for overcoming drug resistance in patients who have lost their response to drugs [25,43,68,76,85,143,144,145,146,147,148,149,150,151,153,154]. Patients are treated according to a regimen that introduces irregularity in the dose and intervals of drug administration, maintaining them within the drug’s therapeutic window and a physician’s pre-determined range [141].

9.2. Establishing a Closed-Loop Algorithm for Generating Individualized Patterns of Irregularity for Overcoming Drug Resistance

One research challenge is implementing subject-specific, disease-tailored, and drug-tailored features within closed-loop deep machine learning algorithms. The closed-loop algorithm provides a platform for reducing the loss of drug efficacy by setting up an irregularity within a specific range determined in an individualized form [141]. The algorithm aims to reach a physiological goal or target for each patient and disease. The patients or their care providers update the machine with inputs indicative of progress towards the desired goal. The learning machine provides updated dose and administration time parameters based on the data learned. It receives inputs from the user and other users to update the algorithm to enable redirecting or further define the treatment regimen [1,140,141].

9.3. Upscaling the Algorithm by Implementing Quantified Personal Variability Patterns along with Additional Personalized Signatures

It is essential to establish and quantify cellular and whole organ patterns of variability and then implement them into the algorithm for improving accuracy [25,43,68,76,85,97,100,101,141,142,143,144,145,146,147,148,149,150,151,153,154,155,156,157,158]. Important information is also obtained by evaluating single-cell variabilities in gene expression, proteomics, metabolomics, and epitope expression performed on cells harvested from patients before and after therapy for chronic disease. A dataset of variability patterns at the cellular levels using single cell-based techniques is generated from cells harvested from patients before and after chronic disease therapy, including single-cell RNA sequencing, proteomics, metabolomics, and epitope expression [159,160,161,162].
Quantifying variability signatures at the molecular level, such as the dynamic instability of microtubules, cytokine secretion, and others. These platforms can be used to implement novel therapies, improve response to chronic cytokine microtubules, overcome drug resistance, exert gut-based systemic immune responses, and generate patient-tailored dynamic therapeutic regimens [68,100,141].
Similarly, assessment of whole organ variability patterns also provides vital data. For example, patients with congestive heart failure are followed for their heart rate variabilities over a 24 h measurement period [25,163,164]. Patterns of variability changes are defined and correlated with patient clinical status and by comparing patients’ current pattern of heart rate variability to heart recordings before deterioration. Similarly, diabetes patients are followed for glucose level variabilities, and their patterns of variabilities were defined. An analysis of the data received from continuous glucose monitor (CGM) sensors in patients with type 2 diabetes showed that the variations in CGM data analyzed every 5 min are not simply “uncorrelated noise”. Quantification of the complexity of the CGM time series temporal structure using multiscale entropy analysis showed that the fluctuations in serum glucose levels from control subjects are more complex when compared with the data from patients with type 2 diabetes [107]. Methods for quantifying these inherent variability patterns, and combining single cells with whole organ variability patterns, must be established for generating an individualized factor to be implemented into the treatment algorithm.
The quantified inherent variability patterns, along with other personalized ways based on genomics, proteomics, microbiome-based, and different signatures, are implemented into individualized-based treatment regimens for overcoming drug resistance. The algorithm continuously alters the irregular regimen based on the patient’s closed-loop feedback on the therapy’s effect. The generated insightful database evolves and supports the dynamic individualized-irregular treatment regimens [1,140,141].
Study limitations: This paper does not present the results of using the described platform in patients with chronic disease. Ongoing studies will shed light on its use for developing personalized therapeutic regimens for patients with chronic diseases.
In summary, overcoming the partial or complete loss of responsiveness to chronic drug use is a significant health problem linked with high morbidity and mortality. The described closed-loop, deep machine learning algorithms generate an individualized, dynamic irregularity, which implements quantified variability and other personalized patterns into therapeutic regimens. This method may answer the unmet need for next-generation personalized medicine enabling a sustainable long-term effect for therapies targeting chronic diseases.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are available in public resources.

Conflicts of Interest

Y. Ilan is the founder of Oberon Sciences.

Abbreviations

ADMEabsorption: distribution: metabolism, and excretion
UGTUDP-glucuronosyltransferases
HFheart failure
TRDtreatment-resistant depression
IBDinflammatory bowel diseases
LORloss of response
CGMglucose monitor

References

  1. Ilan, Y. Overcoming Compensatory Mechanisms toward Chronic Drug Administration to Ensure Long-Term, Sustainable Beneficial Effects. Mol. Ther. Methods Clin. Dev. 2020, 18, 335–344. [Google Scholar] [CrossRef] [PubMed]
  2. Ilan, Y. Order through Disorder: The Characteristic Variability of Systems. Front. Cell Dev. Biol. 2020, 8, 186. [Google Scholar] [CrossRef] [PubMed]
  3. WHOGsrond. 2014. Available online: http://www.who.int/nmh/publications/ncd-status-report2014/en/ (accessed on 29 June 2022).
  4. Gbods. 2017. Available online: http://ghdx.healthdata.org/gbd-2017 (accessed on 29 June 2022).
  5. Reynolds, R.; Dennis, S.; Hasan, I.; Slewa, J.; Chen, W.; Tian, D.; Bobba, S.; Zwar, N. A systematic review of chronic disease management interventions in primary care. BMC Fam. Pract. 2018, 19, 11. [Google Scholar] [CrossRef] [PubMed]
  6. AIoHaWCd. 2015. Available online: http://www.aihw.gov.au/chronic-diseases/ (accessed on 29 June 2022).
  7. Buttorff, C.; Ruder, T.; Bauman, M. Multiple Chronic Conditions in the United States; Rand Corp.: Santa Monica, CA, USA, 2017; Available online: https://www.rand.org/content/dam/rand/pubs/tools/TL200/TL221/RAND_TL221.pdf (accessed on 29 June 2022).
  8. About Chronic Diseases. 2022. Available online: https://www.cdc.gov/chronicdisease/about/index.htm (accessed on 29 June 2022).
  9. Yach, D.; Hawkes, C.; Gould, C.L.; Hofman, K.J. The global burden of chronic diseases: Overcoming impediments to prevention and control. JAMA 2004, 291, 2616–2622. [Google Scholar] [CrossRef] [PubMed]
  10. Bousquet, J.; Jorgensen, C.; Dauzat, M.; Cesario, A.; Camuzat, T.; Bourret, R.; Best, N.; Anto, J.M.; Abecassis, F.; Aubas, P.; et al. Systems medicine approaches for the definition of complex phenotypes in chronic diseases and ageing. From concept to implementation and policies. Curr. Pharm. Des. 2014, 20, 5928–5944. [Google Scholar] [CrossRef]
  11. Sinnige, J.; Braspenning, J.; Schellevis, F.; Stirbu-Wagner, I.; Westert, G.; Korevaar, J. The prevalence of disease clusters in older adults with multiple chronic diseases-a systematic literature review. PLoS ONE 2013, 8, e79641. [Google Scholar] [CrossRef]
  12. Global Burden of Disease Study 2013 Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2015, 386, 743–800. [Google Scholar] [CrossRef] [Green Version]
  13. Health and Economic Costs of Chronic Diseases. 2022. Available online: https://www.cdc.gov/chronicdisease/about/costs/index.htm#ref1C (accessed on 29 June 2022).
  14. M WHaG. The Cost of Chronic Disease in the U.S. 2018. Available online: https://assets1b.milkeninstitute.org/assets/Publication/Viewpoint/PDF/Chronic-Disease-Executive-Summary-r2.pdf (accessed on 29 June 2022).
  15. Loscher, W.; Schmidt, D. Experimental and clinical evidence for loss of effect (tolerance) during prolonged treatment with antiepileptic drugs. Epilepsia 2006, 47, 1253–1284. [Google Scholar] [CrossRef]
  16. Tolerance and Resistance to Drugs. 2022. Available online: https://www.msdmanuals.com/home/drugs/factors-affecting-response-to-drugs/tolerance-and-resistance-to-drugs (accessed on 29 June 2022).
  17. Bespalov, A.; Muller, R.; Relo, A.L.; Hudzik, T. Drug Tolerance: A Known Unknown in Translational Neuroscience. Trends Pharmacol. Sci. 2016, 37, 364–378. [Google Scholar] [CrossRef] [PubMed]
  18. Wojtkowiak, J.W.; Verduzco, D.; Schramm, K.J.; Gillies, R.J. Drug resistance and cellular adaptation to tumor acidic pH microenvironment. Mol. Pharm. 2011, 8, 2032–2038. [Google Scholar] [CrossRef]
  19. de Jonge, M.E.; Huitema, A.D.; Schellens, J.H.; Rodenhuis, S.; Beijnen, J.H. Individualised cancer chemotherapy: Strategies and performance of prospective studies on therapeutic drug monitoring with dose adaptation: A review. Clin. Pharm. 2005, 44, 147–173. [Google Scholar] [CrossRef]
  20. Kennedy, D.A.; Read, A.F. Why does drug resistance readily evolve but vaccine resistance does not? Proc. Biol. Sci. 2017, 284, 20162562. [Google Scholar] [CrossRef] [PubMed]
  21. Nussinov, R.; Tsai, C.J.; Jang, H. A New View of Pathway-Driven Drug Resistance in Tumor Proliferation. Trends Pharmacol. Sci. 2017, 38, 427–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Katayama, R. Therapeutic strategies and mechanisms of drug resistance in anaplastic lymphoma kinase (ALK)-rearranged lung cancer. Pharmacol. Ther. 2017, 177, 1–8. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, L.; Wang, H.; Song, D.; Xu, M.; Liebmen, M. New strategies for targeting drug combinations to overcome mutation-driven drug resistance. Semin. Cancer Biol. 2017, 42, 44–51. [Google Scholar] [CrossRef] [PubMed]
  24. Shinoda, H.; Nakano, S. Pharmacokinetic activity of drugs and circadian rhythm. Nihon Yakurigaku Zasshi 1996, 108, 152–154. [Google Scholar] [PubMed]
  25. Kenig, A.; Kolben, Y.; Asleh, R.; Amir, O.; Ilan, Y. Improving Diuretic Response in Heart Failure by Implementing a Patient-Tailored Variability and Chronotherapy-Guided Algorithm. Front. Cardiovasc. Med. 2021, 8, 695547. [Google Scholar] [CrossRef] [PubMed]
  26. Herbette, L.G.; Vecchiarelli, M.; Sartani, A.; Leonardi, A. Lercanidipine: Short plasma half-life, long duration of action and high cholesterol tolerance. Updated molecular model to rationalize its pharmacokinetic properties. Blood Press. Suppl. 1998, 2, 10–17. [Google Scholar] [CrossRef] [PubMed]
  27. Greenblatt, D.J.; Shader, R.I. Dependence, tolerance, and addiction to benzodiazepines: Clinical and pharmacokinetic considerations. Drug Metab. Rev. 1978, 8, 13–28. [Google Scholar] [CrossRef]
  28. Mazerska, Z.; Mroz, A.; Pawlowska, M.; Augustin, E. The role of glucuronidation in drug resistance. Pharmacol. Ther. 2016, 159, 35–55. [Google Scholar] [CrossRef]
  29. Arbitrio, M.; Di Martino, M.T.; Scionti, F.; Barbieri, V.; Pensabene, L.; Tagliaferri, P. Pharmacogenomic Profiling of ADME Gene Variants: Current Challenges and Validation Perspectives. High. Throughput. 2018, 7, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Rodrigues, J.C.G.; Fernandes, M.R.; Guerreiro, J.F.; da Costa da Silva, A.L.; Ribeiro-dos-Santos, Â.; Santos, S.; Carneiro dos Santos, N.P. Polymorphisms of ADME-related genes and their implications for drug safety and efficacy in Amazonian Amerindians. Sci. Rep. 2019, 9, 7201. [Google Scholar] [CrossRef] [PubMed]
  31. Argevani, L.; Hughes, C.; Schuh, M.J. Dosage Adjustment of Irinotecan in Patients with UGT1A1 Polymorphisms: A Review of Current Literature. Innov. Pharm. 2020, 11, 10.24926. [Google Scholar] [CrossRef] [PubMed]
  32. Hukkanen, J.; Jacob, P., 3rd; Peng, M.; Dempsey, D.; Benowitz, N.L. Effect of nicotine on cytochrome P450 1A2 activity. Br. J. Clin. Pharmacol. 2011, 72, 836–838. [Google Scholar] [CrossRef] [Green Version]
  33. Swanson, J.; Gupta, S.; Guinta, D.; Flynn, D.; Agler, D.; Lerner, M.; Williams, L.; Shoulson, I.; Wigal, S. Acute tolerance to methylphenidate in the treatment of attention deficit hyperactivity disorder in children. Clin. Pharmacol. Ther. 1999, 66, 295–305. [Google Scholar] [CrossRef]
  34. Wolgin, D.L. Contingent tolerance to amphetamine hypophagia: New insights into the role of environmental context in the expression of stereotypy. Neurosci. Biobehav. Rev. 2000, 24, 279–294. [Google Scholar] [CrossRef]
  35. Katz, G. Tachyphylaxis/tolerance to antidepressive medications: A review. Isr. J. Psychiatry Relat. Sci. 2011, 48, 129–135. [Google Scholar]
  36. Lee, M.; Silverman, S.M.; Hansen, H.; Patel, V.B.; Manchikanti, L. A comprehensive review of opioid-induced hyperalgesia. Pain Physician 2011, 14, 145–161. [Google Scholar] [CrossRef]
  37. Sacks, N. The Economic Burden of Insulin Resistance, Obesity, and Cardiovascular Disease in Medicare Beneficiaries 65 Years of Age and Older. Circulation 2018, 136, A15099. [Google Scholar]
  38. Powell, D.; Pacula, R.L.; Jacobson, M. Do medical marijuana laws reduce addictions and deaths related to pain killers? J. Health Econ. 2018, 58, 29–42. [Google Scholar] [CrossRef] [Green Version]
  39. Tang, F.; Hartz, A.M.S.; Bauer, B. Drug-Resistant Epilepsy: Multiple Hypotheses, Few Answers. Front. Neurol. 2017, 8, 301. [Google Scholar] [CrossRef]
  40. Widdess-Walsh, P.; Devinsky, O. Antiepileptic drug resistance and tolerance in epilepsy. Rev. Neurol. Dis. 2007, 4, 194–202. [Google Scholar]
  41. Brodie, M.J. Pharmacological Treatment of Drug-Resistant Epilepsy in Adults: A Practical Guide. Curr. Neurol. Neurosci. Rep. 2016, 16, 82. [Google Scholar] [CrossRef]
  42. Liu, J.T.; Liu, B.; Zhang, H. Surgical versus medical treatment of drug-resistant epilepsy: A systematic review and meta-analysis. Epilepsy Behav. 2018, 82, 179–188. [Google Scholar] [CrossRef]
  43. Potruch, A.; Khoury, S.T.; Ilan, Y. The role of chronobiology in drug-resistance epilepsy: The potential use of a variability and chronotherapy-based individualized platform for improving the response to anti-seizure drugs. Seizure 2020, 80, 201–211. [Google Scholar] [CrossRef]
  44. Facchini, E.; Degiovanni, A.; Cavallino, C.; Lupi, A.; Rognoni, A.; Bongo, A.S. Beta-Blockers and Nitrates: Pharmacotherapy and Indications. Cardiovasc. Hematol. Agents Med. Chem. 2015, 13, 25–30. [Google Scholar] [CrossRef]
  45. Ben-Abraham, R.; Stepensky, D.; Assoulin-Dayan, Y.; Efrati, O.; Lotan, D.; Manisterski, Y.; Berkovitch, M.; Barzilay, Z.; Paret, G. Beta1- or beta2-blockers to improve hemodynamics following endotracheal adrenaline administration. Drug Metabol. Drug Interact. 2005, 21, 31–39. [Google Scholar] [CrossRef]
  46. Tran, H.A.; Lin, F.; Greenberg, B.H. Potential new drug treatments for congestive heart failure. Expert Opin. Investig. Drugs 2016, 25, 811–826. [Google Scholar] [CrossRef]
  47. Jardim, S.I.; Ramos Dos Santos, L.; Araujo, I.; Marques, F.; Branco, P.; Gaspar, A.; Fonseca, C. A 2018 overview of diuretic resistance in heart failure. Rev. Port. Cardiol. 2018, 37, 935–945. [Google Scholar] [CrossRef]
  48. Bowman, B.N.; Nawarskas, J.J.; Anderson, J.R. Treating Diuretic Resistance: An Overview. Cardiol. Rev. 2016, 24, 256–260. [Google Scholar] [CrossRef]
  49. Church, T.J.; Haines, S.T. Treatment Approach to Patients with Severe Insulin Resistance. Clin. Diabetes 2016, 34, 97–104. [Google Scholar] [CrossRef] [Green Version]
  50. Czech, M.P. Insulin action and resistance in obesity and type 2 diabetes. Nat. Med. 2017, 23, 804–814. [Google Scholar] [CrossRef] [PubMed]
  51. Boucher, J.; Kleinridders, A.; Kahn, C.R. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb. Perspect. Biol. 2014, 6, a009191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Turtle, J.R. The economic burden of insulin resistance. Int. J. Clin. Pract. Suppl. 2000, 113, 23–28. [Google Scholar]
  53. Roussel, M.; Mathieu, J.; Dalle, S. Molecular mechanisms redirecting the GLP-1 receptor signalling profile in pancreatic beta-cells during type 2 diabetes. Horm. Mol. Biol. Clin. Investig. 2016, 26, 87–95. [Google Scholar]
  54. Nakamura, A.; Terauchi, Y. Present status of clinical deployment of glucokinase activators. J. Diabetes Investig. 2015, 6, 124–132. [Google Scholar] [CrossRef]
  55. Lauterbach, E.C. Treatment Resistant Depression with Loss of Antidepressant Response: Rapid-Acting Antidepressant Action of Dextromethorphan, A Possible Treatment Bridging Molecule. Psychopharmacol. Bull. 2016, 46, 53–58. [Google Scholar]
  56. Targum, S.D. Identification and treatment of antidepressant tachyphylaxis. Innov. Clin. Neurosci. 2014, 11, 24–28. [Google Scholar]
  57. Berlim, M.T.; Fleck, M.P.; Turecki, G. Current trends in the assessment and somatic treatment of resistant/refractory major depression: An overview. Ann. Med. 2008, 40, 149–159. [Google Scholar] [CrossRef]
  58. Johnston, K.M.; Powell, L.C.; Anderson, I.M.; Szabo, S.; Cline, S. The burden of treatment-resistant depression: A systematic review of the economic and quality of life literature. J. Affect. Disord. 2019, 242, 195–210. [Google Scholar] [CrossRef]
  59. Kolar, D.; Kolar, M.V. Critical Review of Available Treatment Options for Treatment Refractory Depression and Anxiety-Clinical and Ethical Dilemmas. Med. Pregl. 2016, 69, 171–176. [Google Scholar] [CrossRef] [PubMed]
  60. Li, M. Antipsychotic-induced sensitization and tolerance: Behavioral characteristics, developmental impacts, and neurobiological mechanisms. J. Psychopharmacol. 2016, 30, 749–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Fraczek, N.; Bronisz, I.; Pietryka, M.; Kępińska, D.; Strzała, P.; Mielnicka, K.; Korga, A.; Dudka, J. An outline of main factors of drug resistance influencing cancer therapy. J. Chemother. 2016, 28, 457–464. [Google Scholar] [CrossRef] [PubMed]
  62. Bach, D.H.; Hong, J.Y.; Park, H.J.; Lee, S.K. The role of exosomes and miRNAs in drug-resistance of cancer cells. Int. J. Cancer 2017, 141, 220–230. [Google Scholar] [CrossRef] [Green Version]
  63. Calvo, E.; Walko, C.; Dees, E.C.; Valenzuela, B. Pharmacogenomics, Pharmacokinetics, and Pharmacodynamics in the Era of Targeted Therapies. Am. Soc. Clin. Oncol. Educ. Book 2016, 35, e175–e184. [Google Scholar] [CrossRef]
  64. Gottesman, M.M. Mechanisms of cancer drug resistance. Annu. Rev. Med. 2002, 53, 615–627. [Google Scholar] [CrossRef] [Green Version]
  65. Majumder, B.; Baraneedharan, U.; Thiyagarajan, S.; Radhakrishnan, P.; Narasimhan, H.; Dhandapani, M.; Brijwani, N.; Pinto, D.D.; Prasath, A.; Shanthappa, B.U.; et al. Predicting clinical response to anti-cancer drugs using an ex vivo platform that captures tumour heterogeneity. Nat. Commun. 2015, 6, 6169. [Google Scholar] [CrossRef] [Green Version]
  66. Wu, D.; Wang, D.C.; Cheng, Y.; Qian, M.; Zhang, M.; Shen, Q.; Wang, X. Roles of tumor heterogeneity in the development of drug resistance: A call for precision therapy. Semin. Cancer Biol. 2017, 42, 13–19. [Google Scholar] [CrossRef]
  67. Gottesman, M.M.; Lavi, O.; Hall, M.D.; Gillet, J.-P. Toward a Better Understanding of the Complexity of Cancer Drug Resistance. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 85–102. [Google Scholar] [CrossRef]
  68. Ilan, Y.; Spigelman, Z. Establishing patient-tailored variability-based paradigms for anti-cancer therapy: Using the inherent trajectories which underlie cancer for overcoming drug resistance. Cancer Treat. Res. Commun. 2020, 25, 100240. [Google Scholar] [CrossRef]
  69. Gajofatto, A.; Benedetti, M.D. Treatment strategies for multiple sclerosis: When to start, when to change, when to stop? World J. Clin. Cases 2015, 3, 545–555. [Google Scholar] [CrossRef] [PubMed]
  70. Dolati, S.; Babaloo, Z.; Jadidi-Niaragh, F.; Ayromlou, H.; Sadreddini, S.; Yousefi, M. Multiple sclerosis: Therapeutic applications of advancing drug delivery systems. Biomed. Pharmacother. 2017, 86, 343–353. [Google Scholar] [CrossRef] [PubMed]
  71. Dai, Z.; Chu, H.; Ma, J.; Yan, Y.; Zhang, X.; Liang, Y. The Regulatory Mechanisms and Therapeutic Potential of MicroRNAs: From Chronic Pain to Morphine Tolerance. Front. Mol. Neurosci. 2018, 11, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. King, T.; Ossipov, M.H.; Vanderah, T.W.; Porreca, F.; Lai, J. Is paradoxical pain induced by sustained opioid exposure an underlying mechanism of opioid antinociceptive tolerance? Neurosignals 2005, 14, 194–205. [Google Scholar] [CrossRef] [PubMed]
  73. Vadivelu, N.; Singh-Gill, H.; Kodumudi, G.; Kaye, A.J.; Urman, R.D.; Kaye, A.D. Practical guide to the management of acute and chronic pain in the presence of drug tolerance for the healthcare practitioner. Ochsner J. 2014, 14, 426–433. [Google Scholar]
  74. Rudd, R.A.; Aleshire, N.; Zibbell, J.E.; Gladden, R.M. Increases in Drug and Opioid Overdose Deaths—United States. Morb. Mortal. Wkly. Rep. 2016, 64, 1378–1382. [Google Scholar] [CrossRef] [Green Version]
  75. CDC Grand Rounds: Prescription Drug Overdoses–A U.S. Epidemic CfDCaP. Morb. Mortal. Wkly. Rep. 2012, 61, 10–13.
  76. Azmanov, H.; Ross, E.L.; Ilan, Y. Establishment of an Individualized Chronotherapy, Autonomic Nervous System, and Variability-Based Dynamic Platform for Overcoming the Loss of Response to Analgesics. Pain Physician 2021, 24, 243–252. [Google Scholar]
  77. Shmidt, E.; Kochhar, G.; Hartke, J.; Chilukuri, P.; Meserve, J.; Chaudrey, K.; Koliani-Pace, J.L.; Hirten, R.; Faleck, D.; Barocas, M.; et al. Predictors and Management of Loss of Response to Vedolizumab in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2018, 24, 2461–2467. [Google Scholar] [CrossRef]
  78. Kalden, J.R.; Schulze-Koops, H. Immunogenicity and loss of response to TNF inhibitors: Implications for rheumatoid arthritis treatment. Nat. Rev. Rheumatol. 2017, 13, 707–718. [Google Scholar] [CrossRef]
  79. Yanai, H.; Hanauer, S.B. Assessing response and loss of response to biological therapies in IBD. Am. J. Gastroenterol. 2011, 106, 685–698. [Google Scholar] [CrossRef]
  80. Zhang, Q.W.; Shen, J.; Zheng, Q.; Ran, Z.H. Loss of response to scheduled infliximab therapy for Crohn’s disease in adults: A systematic review and meta-analysis. J. Dig. Dis. 2018, 20, 65–72. [Google Scholar] [CrossRef]
  81. Qiu, Y.; Chen, B.L.; Mao, R.; Zhang, S.-H.; He, Y.; Zeng, Z.-R.; Ben-Horin, S.; Chen, M.-H. Systematic review with meta-analysis: Loss of response and requirement of anti-TNFalpha dose intensification in Crohn’s disease. J. Gastroenterol. 2017, 52, 535–554. [Google Scholar] [CrossRef]
  82. Yokoyama, Y.; Kamikozuru, K.; Watanabe, K.; Nakamura, S. Inflammatory bowel disease patients experiencing a loss of response to infliximab regain long-term response after undergoing granulocyte/monocyte apheresis: A case series. Cytokine 2018, 103, 25–28. [Google Scholar] [CrossRef] [PubMed]
  83. Varma, P.; Rajadurai, A.S.; Holt, D.Q.; Devonshire, D.A.; Desmond, C.P.; Swan, M.P.; Nathan, D.; Shelton, E.T.; Prideaux, L.; Sorrell, C.; et al. Immunomodulator Use Does Not Prevent First Loss of Response to Anti-TNF Therapy in Inflammatory Bowel Disease: Long Term Outcomes in a Real-World Cohort. Intern. Med. J. 2019, 49, 753–760. [Google Scholar] [CrossRef]
  84. Ben-Horin, S. Loss of response to anti-tumor necrosis factors: What is the next step? Dig. Dis. 2014, 32, 384–388. [Google Scholar] [CrossRef]
  85. Khoury, T.; Ilan, Y. Introducing Patterns of Variability for Overcoming Compensatory Adaptation of the Immune System to Immunomodulatory Agents: A Novel Method for Improving Clinical Response to Anti-TNF Therapies. Front. Immunol. 2019, 10, 2726. [Google Scholar] [CrossRef] [Green Version]
  86. Djulbegovic, B.; Ioannidis, J.P.A. Precision medicine for individual patients should use population group averages and larger, not smaller, groups. Eur. J. Clin. Investig. 2019, 49, e13031. [Google Scholar] [CrossRef] [Green Version]
  87. Khatry, D.B. Precision medicine in clinical practice. Per. Med. 2018, 15, 413–417. [Google Scholar] [CrossRef]
  88. Leopold, J.A.; Loscalzo, J. Emerging Role of Precision Medicine in Cardiovascular Disease. Circ. Res. 2018, 122, 1302–1315. [Google Scholar] [CrossRef]
  89. van der Wijst, M.G.P.; de Vries, D.H.; Brugge, H.; Westra, H.-J.; Franke, L. An integrative approach for building personalized gene regulatory networks for precision medicine. Genome Med. 2018, 10, 96. [Google Scholar] [CrossRef] [Green Version]
  90. Vinks, A.A. Precision Medicine-Nobody Is Average. Clin. Pharmacol. Ther. 2017, 101, 304–307. [Google Scholar] [CrossRef]
  91. Finn, E.H.; Misteli, T. Molecular basis and biological function of variability in spatial genome organization. Science 2019, 365, eaaw9498. [Google Scholar] [CrossRef] [PubMed]
  92. Goldberger, A.L. Non-linear dynamics for clinicians: Chaos theory, fractals, and complexity at the bedside. Lancet 1996, 347, 1312–1314. [Google Scholar] [CrossRef]
  93. Nijhout, H.F. The nature of robustness in development. Bioessays 2002, 24, 553–563. [Google Scholar] [CrossRef]
  94. Ilan, Y. Overcoming randomness does not rule out the importance of inherent randomness for functionality. J. Biosci. 2019, 44, 132. [Google Scholar] [CrossRef]
  95. Ilan, Y. Advanced Tailored Randomness: A Novel Approach for Improving the Efficacy of Biological Systems. J. Comput. Biol. 2020, 27, 20–29. [Google Scholar] [CrossRef]
  96. Ilan, Y. Generating randomness: Making the most out of disordering a false order into a real one. J. Transl. Med. 2019, 17, 49. [Google Scholar] [CrossRef] [Green Version]
  97. Shabat, Y.; Lichtenstein, Y.; Ilan, Y. Short-Term Cohousing of Sick with Healthy or Treated Mice Alleviates the Inflammatory Response and Liver Damage. Inflammation 2021, 44, 518–525. [Google Scholar] [CrossRef]
  98. Ilan, Y. Randomness in microtubule dynamics: An error that requires correction or an inherent plasticity required for normal cellular function? Cell Biol. Int. 2019, 43, 739–748. [Google Scholar] [CrossRef]
  99. Ilan, Y. Microtubules: From understanding their dynamics to using them as potential therapeutic targets. J. Cell. Physiol. 2019, 234, 7923–7937. [Google Scholar] [CrossRef] [Green Version]
  100. Ilan-Ber, T.; Ilan, Y. The role of microtubules in the immune system and as potential targets for gut-based immunotherapy. Mol. Immunol. 2019, 111, 73–82. [Google Scholar] [CrossRef] [PubMed]
  101. Forkosh, E.; Kenig, A.; Ilan, Y. Introducing variability in targeting the microtubules: Review of current mechanisms and future directions in colchicine therapy. Pharmacol. Res. Perspect. 2020, 8, e00616. [Google Scholar] [CrossRef]
  102. Bandiera, L.; Furini, S.; Giordano, E. Phenotypic Variability in Synthetic Biology Applications: Dealing with Noise in Microbial Gene Expression. Front. Microbiol. 2016, 7, 479. [Google Scholar] [CrossRef] [Green Version]
  103. Xia, X.; Owen, M.S.; Lee, R.E.; Gaudet, S. Cell-to-cell variability in cell death: Can systems biology help us make sense of it all? Cell Death Dis. 2014, 5, e1261. [Google Scholar] [CrossRef]
  104. Singh, N.; Moneghetti, K.J.; Christle, J.W.; Hadley, D.; Froelicher, V.; Plews, D. Heart Rate Variability: An Old Metric with New Meaning in the Era of using mHealth Technologies for Health and Exercise Training Guidance. Part One: Physiology and Methods. Arrhythm. Electrophysiol. Rev. 2018, 7, 193–198. [Google Scholar] [CrossRef] [Green Version]
  105. Shields, R.W., Jr. Heart rate variability with deep breathing as a clinical test of cardiovagal function. Cleve Clin. J. Med. 2009, 76 (Suppl. 2), S37–S40. [Google Scholar] [CrossRef]
  106. Kox, M.; Pompe, J.C.; van der Hoeven, J.G.; Hoedemaekers, C.W.; Pickkers, P. Influence of different breathing patterns on heart rate variability indices and reproducibility during experimental endotoxaemia in human subjects. Clin. Sci. 2011, 121, 215–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Costa, M.D.; Henriques, T.; Munshi, M.N.; Segal, A.R.; Goldberger, A.L. Dynamical glucometry: Use of multiscale entropy analysis in diabetes. Chaos 2014, 24, 033139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Nayyar, S.; Hasan, M.A.; Roberts-Thomson, K.C.; Sullivan, T.; Baumert, M. Effect of Loss of Heart Rate Variability on T-Wave Heterogeneity and QT Variability in Heart Failure Patients: Implications in Ventricular Arrhythmogenesis. Cardiovasc. Eng. Technol. 2017, 8, 219–228. [Google Scholar] [CrossRef]
  109. Avolio, A. Heart rate variability and stroke: Strange attractors with loss of complexity. J. Hypertens. 2013, 31, 1529–1531. [Google Scholar] [CrossRef]
  110. Moon, Y.; Sung, J.; An, R.; Hernandez, M.E.; Sosnoff, J.J. Gait variability in people with neurological disorders: A systematic review and meta-analysis. Hum. Mov. Sci. 2016, 47, 197–208. [Google Scholar] [CrossRef]
  111. Leino, A.D.; King, E.C.; Jiang, W.; Vinks, A.A.; Klawitter, J.; Christians, U.; Woodle, E.S.; Alloway, R.R.; Rohan, J.M. Assessment of tacrolimus intrapatient variability in stable adherent transplant recipients: Establishing baseline values. Am. J. Transplant. 2018, 19, 1410–1420. [Google Scholar] [CrossRef] [PubMed]
  112. Gueta, I.; Markovits, N.; Yarden-Bilavsky, H.; Raichlin, E.; Freimark, D.; Lavee, J.; Loebstein, R.; Peled, Y. High tacrolimus trough level variability is associated with rejections after heart transplant. Am. J. Transplant. 2018, 18, 2571–2578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Gueta, I.; Markovits, N.; Yarden-Bilavsky, H.; Raichlin, E.; Freimark, D.; Lavee, J.; Loebstein, R.; Peled, Y. Intrapatient variability in tacrolimus trough levels after solid organ transplantation varies at different postoperative time periods. Am. J. Transplant. 2019, 19, 611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Del Bello, A.; Congy-Jolivet, N.; Danjoux, M.; Muscari, F.; Lavayssière, L.; Esposito, L.; Hebral, A.-L.; Bellière, J.; Kamar, N. High tacrolimus intra-patient variability is associated with graft rejection, and de novo donor-specific antibodies occurrence after liver transplantation. World J. Gastroenterol. 2018, 24, 1795–1802. [Google Scholar] [CrossRef]
  115. Elgart, V.; Lin, J.R.; Loscalzo, J. Determinants of drug-target interactions at the single cell level. PLoS Comput. Biol. 2018, 14, e1006601. [Google Scholar] [CrossRef] [Green Version]
  116. Contin, M.; Alberghini, L.; Candela, C.; Benini, G.; Riva, R. Intrapatient variation in antiepileptic drug plasma concentration after generic substitution vs stable brand-name drug regimens. Epilepsy Res. 2016, 122, 79–83. [Google Scholar] [CrossRef]
  117. Arbitrio, M.; Scionti, F.; Di Martino, M.T.; Caracciolo, D.; Pensabene, L.; Tassone, P.; Tagliaferri, P. Pharmacogenomics Biomarker Discovery and Validation for Translation in Clinical Practice. Clin. Transl. Sci. 2021, 14, 113–119. [Google Scholar] [CrossRef]
  118. Weiner, W.J.; Koller, W.C.; Perlik, S.; Nausieda, P.A.; Klawans, H.L. Drug holiday and management of Parkinson disease. Neurology 1980, 30, 1257–1261. [Google Scholar] [CrossRef]
  119. Toni, T.; Tidor, B. Combined model of intrinsic and extrinsic variability for computational network design with application to synthetic biology. PLoS Comput. Biol. 2013, 9, e1002960. [Google Scholar] [CrossRef] [Green Version]
  120. Boeing, G. Visual Analysis of Nonlinear Dynamical Systems: Chaos, Fractals, Self-Similarity and the Limits of Prediction. Systems 2016, 4, 37. [Google Scholar] [CrossRef] [Green Version]
  121. Lorenz, E.N. Deterministic non-periodic flow. J. Atmos. Sci. 1963, 20, 130–141. [Google Scholar] [CrossRef] [Green Version]
  122. Gunaratne, G.H.; Linsay, P.S.; Vinson, M.J. Chaos beyond onset: A comparison of theory and experiment. Phys. Rev. Lett. 1989, 63, 1–4. [Google Scholar] [CrossRef]
  123. Poole, R. Chaos theory: How big an advance? Science 1989, 245, 26–28. [Google Scholar] [CrossRef]
  124. Shinbrot, T.; Ditto, W.; Grebogi, C.; Ott, E.; Spano, M.; Yorke, J.A. Using the sensitive dependence of chaos (the “butterfly effect”) to direct trajectories in an experimental chaotic system. Phys. Rev. Lett. 1992, 68, 2863–2866. [Google Scholar] [CrossRef] [Green Version]
  125. Werndl, C. What are the New Implications of Chaos for Unpredictability? Br. J. Philos. Sci. 2009, 60, 195–220. [Google Scholar] [CrossRef] [Green Version]
  126. Skinner, J.E. Low-dimensional chaos in biological systems. Biotechnology 1994, 12, 596–600. [Google Scholar] [CrossRef]
  127. Umriukhin, E.A.; Sudakov, K.V. Chaos theory: The transforming role of functional systems. Ross. Fiziol. Zh. Im. IM Sechenova 1997, 83, 190–216. [Google Scholar]
  128. Baffy, G. The impact of network medicine in gastroenterology and hepatology. Clin. Gastroenterol. Hepatol. 2013, 11, 1240–1244. [Google Scholar] [CrossRef] [Green Version]
  129. Gebicke-Haerter, P.J. Systems biology in molecular psychiatry. Pharmacopsychiatry 2008, 41 (Suppl. 1), S19–S27. [Google Scholar] [CrossRef] [PubMed]
  130. Brunton, S.L.; Brunton, B.W.; Proctor, J.L.; Kaiser, E.; Kutz, J.N. Chaos as an intermittently forced linear system. Nat. Commun. 2017, 8, 19. [Google Scholar] [CrossRef] [PubMed]
  131. Sternad, D.; Hasson, C.J. Predictability and Robustness in the Manipulation of Dynamically Complex Objects. Adv. Exp. Med. Biol. 2016, 957, 55–77. [Google Scholar]
  132. Cai, S.; Liu, Z.; Lee, H.C. Mean field theory for biology inspired duplication-divergence network model. Chaos 2015, 25, 083106. [Google Scholar] [CrossRef]
  133. Lesne, A.A. Chaos in biology. Riv. Biol. 2006, 99, 467–481. [Google Scholar]
  134. Huberman, B.A. A Model for Dysfunctions in Smooth Pursuit Eye Movement. Ann. N. Y. Acad. Sci. 1987, 504, 260–273. [Google Scholar] [CrossRef] [PubMed]
  135. Bozoki, Z. Chaos theory and power spectrum analysis in computerized cardiotocography. Eur. J. Obstet. Gynecol. Reprod. Biol. 1997, 71, 163–168. [Google Scholar] [CrossRef]
  136. Liu, G.; Stevens, J.B.; Horne, S.D.; Abdallah, B.; Ye, K.; Bremer, S.; Ye, C.; Chen, D.J.; Heng, H. Genome chaos: Survival strategy during crisis. Cell Cycle 2014, 13, 528–537. [Google Scholar] [CrossRef] [Green Version]
  137. Edwards, D.H. Local, integrated control of blood flow: Professor Tudor Griffith Memorial. Auton Neurosci. 2013, 178, 4–8. [Google Scholar] [CrossRef] [PubMed]
  138. Cowin, S.C.; Arramon, Y.P.; Luo, G.M.; Sadegh, A.M. Chaos in the discrete-time algorithm for bone-density remodeling rate equations. J. Biomech. 1993, 26, 1077–1089. [Google Scholar] [CrossRef]
  139. Schneider, B.L.; Kulesz-Martin, M. Destructive cycles: The role of genomic instability and adaptation in carcinogenesis. Carcinogenesis 2004, 25, 2033–2044. [Google Scholar] [CrossRef] [Green Version]
  140. Ilan, Y. Second-Generation Digital Health Platforms: Placing the Patient at the Center and Focusing on Clinical Outcomes. Front. Digit. Health. 2020, 2, 569178. [Google Scholar] [CrossRef]
  141. Ilan, Y. Improving Global Healthcare and Reducing Costs Using Second-Generation Artificial Intelligence-Based Digital Pills: A Market Disruptor. Int. J. Environ. Res. Public Health 2021, 18, 811. [Google Scholar] [CrossRef]
  142. El-Haj, M.; Kanovitch, D.; Ilan, Y. Personalized inherent randomness of the immune system is manifested by an individualized response to immune triggers and immunomodulatory therapies: A novel platform for designing personalized immunotherapies. Immunol. Res. 2019, 67, 337–347. [Google Scholar] [CrossRef]
  143. Ilan, Y. Why targeting the microbiome is not so successful: Can randomness overcome the adaptation that occurs following gut manipulation? Clin. Exp. Gastroenterol. 2019, 12, 209–217. [Google Scholar] [CrossRef] [Green Version]
  144. Ilan, Y. beta-Glycosphingolipids as Mediators of Both Inflammation and Immune Tolerance: A Manifestation of Randomness in Biological Systems. Front. Immunol. 2019, 10, 1143. [Google Scholar] [CrossRef] [Green Version]
  145. Kenig, A.; Ilan, Y. A Personalized Signature and Chronotherapy-Based Platform for Improving the Efficacy of Sepsis Treatment. Front. Physiol. 2019, 10, 1542. [Google Scholar] [CrossRef] [PubMed]
  146. Kessler, A.; Weksler-Zangen, S.; Ilan, Y. Role of the Immune System and the Circadian Rhythm in the Pathogenesis of Chronic Pancreatitis: Establishing a Personalized Signature for Improving the Effect of Immunotherapies for Chronic Pancreatitis. Pancreas 2020, 49, 1024–1032. [Google Scholar] [CrossRef] [PubMed]
  147. Kolben, Y.; Weksler-Zangen, S.; Ilan, Y. Adropin as a potential mediator of the metabolic system-autonomic nervous system-chronobiology axis: Implementing a personalized signature-based platform for chronotherapy. Obes. Rev. 2021, 22, e13108. [Google Scholar] [CrossRef]
  148. Gelman, R.; Bayatra, A.; Kessler, A.; Schwartz, A.; Ilan, Y. Targeting SARS-CoV-2 receptors as a means for reducing infectivity and improving antiviral and immune response: An algorithm-based method for overcoming resistance to antiviral agents. Emerg. Microbes Infect. 2020, 9, 1397–1406. [Google Scholar] [CrossRef]
  149. Ishay, Y.; Kolben, Y.; Kessler, A.; Ilan, Y. Role of circadian rhythm and autonomic nervous system in liver function: A hypothetical basis for improving the management of hepatic encephalopathy. Am. J. Physiol.-Gastrointest. Liver Physiol. 2021, 321, G400–G412. [Google Scholar] [CrossRef]
  150. Hurvitz, N.; Azmanov, H.; Kesler, A.; Ilan, Y. Establishing a second-generation artificial intelligence-based system for improving diagnosis, treatment, and monitoring of patients with rare diseases. Eur. J. Hum. Genet. 2021, 29, 1485–1490. [Google Scholar] [CrossRef]
  151. Khoury, T.; Ilan, Y. Platform introducing individually tailored variability in nerve stimulations and dietary regimen to prevent weight regain following weight loss in patients with obesity. Obes. Res. Clin. Pract. 2021, 15, 114–123. [Google Scholar] [CrossRef]
  152. Kyriazis, M. Practical applications of chaos theory to the modulation of human ageing: Nature prefers chaos to regularity. Biogerontology 2003, 4, 75–90. [Google Scholar] [CrossRef]
  153. Isahy, Y.; Ilan, Y. Improving the long-term response to antidepressants by establishing an individualized platform based on variability and chronotherapy. Int. J. Clin. Pharmacol. Ther. 2021, 59, 768–774. [Google Scholar] [CrossRef]
  154. Ishay, Y.; Potruch, A.; Schwartz, A.; Berg, M.; Jamil, K.; Agus, S.; Ilan, Y. A digital health platform for assisting the diagnosis and monitoring of COVID-19 progression: An adjuvant approach for augmenting the antiviral response and mitigating the immune-mediated target organ damage. Biomed. Pharmacother. 2021, 143, 112228. [Google Scholar] [CrossRef]
  155. Ishay, Y.; Kessler, A.; Schwarts, A.; Ilan, Y. Antibody response to SARS-Co-V-2, diagnostic and therapeutic implications. Hepatol. Commun. 2020, 4, 1731–1743. [Google Scholar] [CrossRef]
  156. Ilan, Y. Digital Medical Cannabis as Market Differentiator: Second-Generation Artificial Intelligence Systems to Improve Response. Front. Med. 2021, 8, 788777. [Google Scholar] [CrossRef]
  157. Gelman, R.; Berg, M.; Ilan, Y. A Subject-Tailored Variability-Based Platform for Overcoming the Plateau Effect in Sports Training: A Narrative Review. Int. J. Environ. Res. Public Health 2022, 19, 1722. [Google Scholar] [CrossRef]
  158. Azmanov, H.; Bayatra, A.; Ilan, Y. Digital Analgesic Comprising a Second-Generation Digital Health System: Increasing Effectiveness by Optimizing the Dosing and Minimizing Side Effects. J. Pain Res. 2022, 15, 1051–1060. [Google Scholar] [CrossRef]
  159. Emara, S.; Amer, S.; Ali, A.; Abouleila, Y.; Oga, A.; Masujima, S. Single-Cell Metabolomics. Adv. Exp. Med. Biol. 2017, 965, 323–343. [Google Scholar] [PubMed]
  160. Su, Y.; Shi, Q.; Wei, W. Single cell proteomics in biomedicine: High-dimensional data acquisition, visualization, and analysis. Proteomics 2017, 17, 1600267. [Google Scholar] [CrossRef] [PubMed]
  161. Lafzi, A.; Moutinho, C.; Picelli, S.; Heyn, H. Tutorial: Guidelines for the experimental design of single-cell RNA sequencing studies. Nat. Protoc. 2018, 13, 2742–2757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Skanland, S.S. Phospho Flow Cytometry with Fluorescent Cell Barcoding for Single Cell Signaling Analysis and Biomarker Discovery. J. Vis. Exp. 2018, 140, e58386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Huikuri, H.V.; Stein, P.K. Heart rate variability in risk stratification of cardiac patients. Prog Cardiovasc. Dis. 2013, 56, 153–159. [Google Scholar] [CrossRef] [PubMed]
  164. Adamson, P.B. Continuous heart rate variability from an implanted device: A practical guide for clinical use. Congest. Heart Fail. 2005, 11, 327–330. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A three-step approach for introducing a system for overcoming drug resistance.
Figure 1. A three-step approach for introducing a system for overcoming drug resistance.
Jpm 12 01303 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ilan, Y. Next-Generation Personalized Medicine: Implementation of Variability Patterns for Overcoming Drug Resistance in Chronic Diseases. J. Pers. Med. 2022, 12, 1303. https://doi.org/10.3390/jpm12081303

AMA Style

Ilan Y. Next-Generation Personalized Medicine: Implementation of Variability Patterns for Overcoming Drug Resistance in Chronic Diseases. Journal of Personalized Medicine. 2022; 12(8):1303. https://doi.org/10.3390/jpm12081303

Chicago/Turabian Style

Ilan, Yaron. 2022. "Next-Generation Personalized Medicine: Implementation of Variability Patterns for Overcoming Drug Resistance in Chronic Diseases" Journal of Personalized Medicine 12, no. 8: 1303. https://doi.org/10.3390/jpm12081303

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