Traditional and Non-Traditional Cardiovascular Risk Factor Profiles in Young Patients with Coronary Artery Disease

Research Article | DOI: https://doi.org/10.31579/2693-4779/203

Traditional and Non-Traditional Cardiovascular Risk Factor Profiles in Young Patients with Coronary Artery Disease

  • Rehan Haider 1*
  • Geetha Kumari Das 2
  • Asghar Mehdi 3
  • Zameer Ahmed 4
  • Sambreen Zameer 4

1Riggs Pharmaceuticals, Karachi. Department of Pharmacy, University of Karachi-Pakistan

2GD Pharmaceutical Inc OPJS University Rajasthan India   

 3Assistant Professor Dow University of Health Sciences Karachi Pakistan

4Associate Prof Department of Pathology Dow University of Health Sciences Karachi Pakistan

*Corresponding Author: Rehan Haider, Riggs Pharmaceuticals, Karachi. Department of Pharmacy, University of Karachi-Pakistan.

Citation: Rehan Haider, Geetha Kumari Das, Asghar Mehdi, Zameer Ahmed, Sambreen Zameer, (2024), Traditional and Non-Traditional Cardiovascular Risk Factor Profiles in Young Patients with Coronary Artery Disease, Clinical Research and Clinical Trials, 10(2); DOI:10.31579/2693-4779/203

Copyright: © 2024, Rehan Haider. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: 18 April 2024 | Accepted: 03 May 2024 | Published: 20 June 2024

Keywords: phase i clinical trials; extrapolation; animal models; human exposure; drug development preclinical studies; translational research; methodologies; challenges; predictive modeling

Abstract

Phase I dispassionate troubles comprise critical achievements in drug happening, contributing to the first freedom to convert findings from preclinical animal studies to human cases. However, connecting the gap between the animal dossier and human uncovering poses meaningful challenges. This abstract investigates the methods and concerns involved in inferring data from animal models to anticipate human effects during Phase I tests. Phase I dispassionate trials are important heavily in judging the security and tolerability of new drugs in people. Yet, they commit heavily to preclinical dossiers derived from animal studies. The change from animal models to human issues includes inferring pharmacokinetic and pharmacodynamic limits in the way that drug absorption, disposal, and productiveness are measured while considering interspecies dissimilarities in the study of animals, absorption, and study of animals. Methodologies for prediction include physiologically located pharmacokinetic shaping, allo metric measuring, and artificial-in vivo prediction methods. However, challenges endure on account of the basic differences between classes and the complexity of the human study of animals.

Understanding the restraints and doubts in inferring animal dossier to human uncovering is essential for plotting Phase I troubles that plan outpatient security and increase the probability of boom-in-after aspects of drug happening. This abstract underline the detracting role of translational research in optimizing the transition from preclinical studies to dispassionate troubles and eventually reconstructing the effectiveness and influence of drug processes.

Introduction

Successful preclinical drug-finding programs commonly reach a point where there is a need to pick an individual or two aspirants from between a whole pharmacological class of new drugs for point, I testing (Welling and Tse, 1995) [1]. There is so much important need to create trustworthy and breakneck guesses of human responses from the animal data.

Although drug finding is generally devised to find compounds accompanying desired productiveness, the choice from with diversified compounds conceivably offer insult efficacy frequently worsens to those accompanying the most benign pharmacokinetics (Welling and Tse, 1995). Thus, compounds are preferred when utilizing animal data, at least by way of appropriate bioavailability, half-life, and tissue penetration characteristics. As we are going to review beneath, the possibility of multi phasic skin level decay patterns following the venous drug is a main piece of this draft process.

Pharmacokinetics, connected when attainable to the noticed drug belongings, is effective and critical component of an important step in animal research to human research in the drug growth process. Data for preferred compounds will usually also have happened bear hardship concurrent forming of pharmacokinetic and pharmacodynamic dossier from animals, repeated in work to increase the chances that the drugs preferred will have the correct combined humans particularized in a pre-discovery crop sketch. Meanwhile, the pharmacodynamic information usually includes data from receptor-binding studies, artificial working assays and in vivo pharmacological protection experiments. The significance of this critical step in drug growth, communicable the new drug into human

beings is the making of the right indicators of in vivo drug effects from the artificial data.

The accumulation of vitro dossier from animal materials and prediction (a) from material properties to the artificial dossier, (b) from the artificial dossier to nonhuman in vivo data, and (c) from nonhuman in vivo data to dispassionate in vivo answers may be accomplished more efficiently-ciently utilizing online reasoning and simulations. This stage inquiries to explain expeditious progress may be completed for new synthetic bodies through this process, utilizing artificial and in vivo data and leading posing processes. This must be visualized in the circumstances of the entire drug finding process that, on the best scale, is planned to find effective, safe drugs (in persons), established ani mal dossier (Figure 8.1). We predict a period when in vitro pharmacodynamic dossier will be routinely linked accompanying artificial drug absorption dossier in a rational guess of drug answers in athletic human steps forward, accompanying consequent quickening of the drug-finding work, and therefore a general trend for more adept use of money in early dispassionate incident.

Figure 8.1: General scheme showing the pharmacokinetic prediction pathway from physicochemical properties to human drug response via in vitro and in vivo studies in laboratory animals

In Vitro and Invivo Prediction

The challenge is to search out and predict systemic clearance volume of distribution and oral bioavailability in humans from a combination if vitro and vivo preclinical data. If this forecasting can become reliable, before development, I study to enhance more confirmed conservative ness. The use of human hepatocytes and isolated enzymes can form a fault-finding containing the artificial table. The clearance of almost all drugs is by renal, metabolic, and/or biliary means. There are excellent irregularities, to a degree, sleep-inducer gases that are breathed unaltered. However, in this place phase, we shall apply ourselves to the usual position. Physicochemical characteristics, especially lipophilicity, frequently rule the go-ahead route; lipophilicity is usually calculated as record D7:4, where this changeable equals log10 ([drug in octanol]/[drug in liquid safeguard]) at pH ¼ 7:4, in an independent system at balance. Generally, compounds accompanying a record D7:4 worth beneath 0 have significant renal consent principles, since compounds accompanying record D7:4 values above 0 will customarily be removed mainly by hepatic absorption (Smith et al., 1996) [2]. Molecular size still has few effects on these authorization routes. For example, compounds accompanying microscopic weights greater than 400 Da are frequently removed through the hostility unaltered, whilst tinier lipophilic com pounds will mainly be metabolized Elementary facets of consent the prevailing, clinical calculation of drug clearance includes attractive sequential unmodified blood samples. As time passes later Tmax (moment of truth when drug aggregation reaches allure peak), parent drug concentrations steadily decline. Modeling of Drug vanishing is an explanatory process that requires real human uncovering. Unsa-curated removal machines, in the absence of drug seclusion may be designed as natural, first-order removal using a fixed (k) accompanying parts of h 1; skin aggregation (C) is then designed by equations of the approximate form:

C ¼ Ae kt 

place A is the concentration of the drug earlier (t). (Provided that there was immediate and homogenous equilibration of the application into the circulating subdivision). As the number of compartments increases, therefore so does the number of agreements of the form proved on the right-hand side of the equation proved above.

The removal rate forever has wholes of (bulk/ time) for some removal process. For first-order processes, the removal rate at some individual moment is depicted by touching the removal curve for some specified period t or drug aggregation C. In contrast, nothing-order removal processes are periodically encountered. These customarily show satiation for one drug of the elimination mechanics group(s). These ‘drug vanishing’ curves are straight and accordingly interpreted plainly by:

C ¼ A bt

where the elimination rate (b) does not change with time or drug concentration. If followed for long enough, most drugs that are subject to zero-order elimination eventually fall to such low concentrations that the elimination mechanism becomes unsaturated, and first-order elimination then supervenes; good examples include ethanol and sodium dichloroacetate (Hawkins and Kalant,1972; Curry et al., 1985; Fox et al., 1996) [3,4,5]

The elimination rate for zero-order processes may also be treated as a maximal rate of reaction

(Vmax), and thus this type of data may be subject to ordinary Michaelis-Menten analysis (see further, below). Note that first-order elimination curves are so common that ‘drug disappearance’ curves routinely analyzed as semi-logarithmic plots (which linearizes the curve). The literature is sometimes ambiguous in its use of the term ‘linear data’, authors may or may not assume that the semi-logarithmic transformation is to be taken as read. When the elimination rate is known, then clearance (Cl) is defined simply as:                                                                                                                                          Cl ¼ elimination rate=C where C is again the drug concentration. Note that in first-order elimination processes, the elimination rate of the drug (with units of mass/time) changes with time (and drug concentration), and thus only instantaneous clearances, specifying time or drug concentration can be stated.

Urinary clearance, obviously, may only partly explain the rate of drug disappearance from plasma. In any case, the urinary clearance of an agent may be found from the familiar equation:

Cl ¼ ðU VÞ=P

where U is the urinary concentration, V is the volume of urine excreted during a specified time period and P is the average plasma concentration during that period. For inulin and sodium iothalamate, but not for creatinine or urea, the urinary clearance is a good measure of glomerular filtration rate. These elementary aspects of clearance may be revised in any textbook (e.g., Curry, 1980; Benet et al., 1996) [6,7]. The purpose of the remainder of this section is to show how much more informative the concept of clearance may be and to provide an illustration of its use.

Prediction of human drug clearance for those compounds that are predominantly cleared by metabolism, human blood clearance can be predicted using simple enzyme kinetic data (Houston, 1994; Ashforth et al., 1995; Iwatsubs et al., 1996; Obach, 1996a) [8,9,10,11]. These predictions may be strengthened by comparing preclinical in vivo data with the predictions made from in in vitro data using tissues from the same preclinical species (Rane et al., 1977) [12]. As an illustration, consider compound X (anonymized but real). This compound has a molecular weight of less than 400 and a log D7.4 value of approximately 0.5, suggesting that it could undergo both renal and hepatic clearances. Preclinical in vivo studies indicate that compound X is eliminated largely unchanged in the urine in the rat (90%). Several oxidative bio transformation pathways have nonetheless been identified. In common with studies of compound X clearance in humans, simple in vitro enzyme kinetic studies were used in conjunction with knowledge from rat in vivo data. The general strategy for prediction of kinetic studies is shown in Figure 8.2.

Using liver microsomes from different species, the intrinsic clearance (Cl0 int) for each species can be determined and then scaled to hepatic clearance. This is typically done by first determining in vitro Km (the Michaelis–Menten constant) and Vmax (the

Figure 8.2

Strategy for the in vitro–in vivo scaling of hepatic clearance (see for example Iwatsubo et al., 1996) maximal rate of metabolism) for each metabolic reaction, using substrate saturation plots (using the familiar algebra and, because of enzyme saturation, finding that Cl0 int ¼ Vmax=Km). However, for compound X, the situation is more complicated because we know that the Cl0 int (drug disappearance) is due to several combined transformational pathways (i.e. Cl0).intðtotalÞ ¼ Cl0 int1þ Cl0 int2 þ Cl0 int3 þ L), thus complicating any Km and Vmax determinations from a simple substrate saturation plot.

To determine the Cl0 int of compound X, we are able to use the in vitro half-life method, which is simpler than finding all the components of Cl0 int values.

When the substrate concentration is much smaller than Km, the Michaelis-Menten equation is simple. fies from velocity ðVÞ ¼ Vmaxð½SÞ=ðKm þ ½SÞ, because [S] (substrate concentration) becomes negligible. Furthermore, under these conditions, the in vitro half-life (T1 = 2 ¼ 0:693/Kel) can be measured, and this, in turn, is related to the Michaelis–Menten equation through the relationship velocity (V) ¼ volume Kel (where volume is standardized) for the volume containing 1 mg of microsomal protein). When both V and Vmax are known, then Km is also found. Although simpler than finding a complicated Cint, one caveat of the in vitro half-life method is that one assumes that the substrate concentration is much smaller than Km. It may be necessary to repeat the half-life determinations at several substrate concentrations, and even model the asymptotic of this relationship, because every low substrate concentration that are beneath biochemical detection may be needed to fulfill the assumptions needed to simplify the Michaelis– Menten equation.

Material and Methods

Note also that in this in vitro application, intrinsic clearance, like all conventional mathematical evaluation of clearances has units of volume, time, 1. It is obtained from Vmax and measurements, where Vmax has units of mass time 1. The definition of intrinsic clearance is Vmax/Km 1 should not be confused with the historically prevalent calculation of Kel (the first-order rate constant of decay of concentration in plasma), calculated from kel ¼ Vmax/Km, where Vmax is the zero-order rate of plasma concentration decay observed at high concentrations and Kmax is the concentration of plasma at a half-maximal rate of plasma level decay.

Once the in vitro intrinsic clearance has been determined, the next step, scaling in vitro intrinsic Clearance to the whole liver proceeds as follows: in vivo Cl0 int ¼ in vitro Cl0 int weight microsomal protein=g liver weight liver=kg body weight the amount of microsomal protein per gram the liver is constant across mammalian species (45 mg g1 liver). Thus, the only species-dependent variable is the weight of liver tissue per kilogram body weight.

In vivo, hepatic clearance is determined by factoring in the hepatic blood flow (Q), the fraction of drug unbound in the blood (fu) and the fraction of drug unbound in the microsomal incubations (fuðincÞ) against the intrinsic clearance of the drug by the whole liver (the in vivo C0 int). The fun and fuðincÞ are included when the drug shows consideredable plasma or microsomal protein binding (Obach, 1996b) [13]. Several models are available for scaling in vivo intrinsic clearance to hepatic clearance, including the parallel tube model or sinusoidal perfusion model, the well-stirred model or venous equilibration model and the distributed sinusoidal perfusion model (Wilkinson, 1987) [14].

Thus far, for compound X, we have obtained good results in this context with the simplest of these, the well-stirred model (Table 8.1 for the equations, with and without significant plasma

Table 8.1: Equations for predicting hepatic clearance using the well-stirred model

Table 8.2: Comparison of the predicted in vivo hepatic clearance and the actual clearance values for compound X

Predicted values were scaled from in vitro half-life data using liver microsomes and the well-stirred model of hepatic extraction. Hepatic Cl predictions were corrected for plasma and microsomal protein binding. Predicted total Cl was obtained by adding in renal Cl estimates which were, in turn, scaled allometrically (Y ¼ aW0:75) and/or microsomal protein binding). Using this well-stirred model, it has proved possible to predict the hepatic clearance from in vitro intrinsic clearance rates in rats, dogs, and humans (Table 8.2). The hepatic clearance value for the rat (0.972 ml/min) mg (1 protein) was approximately one-tenth the actual clearance found in vivo; well in agreement with the observation that in vivo compound X was eliminated by the rat, largely unchanged, by the kidneys (90%). To predict hepatic clearance of compound X in humans, human in vitro intrinsic clearance could then be scaled to hepatic clearance, using a technique that had been validated in rats (Ashfortt et al., 1995). Renal clearance is subject to an allometric relationship and can generally be scaled across species (see below). The predicted in vivo renal Cl for rat (estimated by multiplying the predicted hepatic Cl by 9) may be scaled allometrically to obtain a prediction for the human in vivo renal clearance. Total or systemic Cl in humans can then be estimated by adding the two clearances parameters (hepatic and renal) together; in practice, for compound X, later first-in-human data revealed an actual in vivo Cl nearly identical to the predicted total Cl (2.15 vs. 1.87–2.45 mlmin 1 mg 1 , respectively; Table 8.2). Here, then, is a real-world example of, first, how rats in vitro and in vivo, preclinical data were used to develop and validate a scaling method for compound X in rats; and second, how the scaling method successfully predicted in vivo overall drug clearance in humans.

However, if the same methods are used for compound X in dogs, things initially appear to be different. Scaling the in vitro intrinsic clearance to hepatic Cl using the rat-validated method, in conjunction with allometric scaling of renal Cl, resulted in a five-fold under-prediction of the total or systemic clearance in vivo. However, further metabolism studies in the dog in vivo revealed that compound X undergoes significant additional biotransformation, particularly N methylation, which is unique (as far as we are aware) to this species, and invalidates some of our in vitro assumptions. This canine bio-motion pathway was not detected by our initial microsomal studies because there are no N-methyl transferases in microsomes. Thus, although we did not successfully predict dog systemic clearance for compound X, our scaling tactics did eventually teach us about a new clearance mechanism, and how important this was for the systemic clearance of compound X in the dog.This is an example of how in vitro studies can be combined with in vivo preclinical data, leading to the useful prediction of human systemic drug clearance. Nonetheless, several caveats are encountered in such scaling exercises, which warrant restating. The first caveat is that all clearance pathways (hepatic, renal, biliary, or other) must be taken into consideration. If a compound undergoes a high level of hepatic clearance, then in vitro and in vivo scaling may be used to predict the fraction of Systemic clearance is expected from this pathway. If a compound undergoes a high level of renal elimination, allometric scaling may be also used to predict the clearance attributed to this pathway.

The second caveat is that, to accurately To predict hepatic clearance, the correct in vitro system must be chosen. If the candidate drug is primarily oxidatively metabolized, then the liver microsomes will be sufficient. However, if the potential for non-microsomal biotransformation exists, then a different in vitro system, such as hepatocyte suspensions should be used. In the illustration above, it turned out, as far as clearance as far as compound X is concerned, humans are specifically like a rat and unlike a dog.

The third caveat is that one must consider the variability in the expression of metabolizing enzymes between individuals. Oxidative metabolism ( in vivo and microsomal enzymes), and especially cytochrome P450s, vary tremendously between human individuals (Meyer, 1994; Shimada et al., 1994){15,16}.Had we used a single donor microsomal sample rather than pooled liver microsomes? (a pool consisting of at least eight individual donors), to scale in vitro data to in vivo hepatic clearance, we might have made greatly misleading predictions (note that oxidative, initial drug metabolism is sometimes called ‘phase I metabolism’ in the literature, causing ambiguity with the stage of drug development or type of clinical trial).Volumes of distribution Review of elementary concepts The volume of distribution is a theoretical concept that may or may not correspond to the anatomical compartment(s) which drugs or metabolites may access after dosing. When the size of the dose (D) is known, and when drug concentration (C) may be found by sampling biological fluids, then, in the simplest case, the volume of distribution (VD) is:

VD ¼ D=C Clinical protocols can usually only prescribe the sampling of a subset of compartments when a drug is known to distribute widely in the body. For example, a lipophilic drug may penetrate lipophilic organs such as the brain, and brain playing simply for pharmacokinetic purposes is usually possible only in animals. In such cases, blood concentrations fall far lower than if the the dose had been distributed solely into the circulation compartment; C becomes very small, and VD becomes correspondingly very large. The opposite effect would require the drug to be restricted to a fraction of the compartment that is sampled, essentially suggesting that too few compartments have been postulated, and the effect is rarely encountered. Again, Curry (1980) or Benet et al. (1996) for the expansion of these elementary aspects of the volume of distribution.Prediction of human volumes of distribution The free (not plasma protein bound) volume of the distribution of experimental drugs is generally considered to be constant for all species. Thus, the volume of distribution in humans can easily be predicted through a simple proportionality between in vitro plasma protein binding data in humans and in a preclinical species, and in vivo the volume of distribution in that same preclinical species: VDhuman ¼ VDpre-clinical species human future-clinical species where fu is a fraction of unbound V0 plasma proteins.

Table 8.3 shows the predicted volume of distribution of a single intravenous bolus dose of compound X in humans; this is found by using the above equation, an in vitro estimate of protein binding data for rat and dog plasmas, and the observed volumes of distribution for these two species in vivo. For humans, VDhuman was predicted to be 3.48–4.591 kg1 using the rat data and 3.01–5.061 kg1 using the dog data.

Table 8.3: In vitro plasma protein binding, in vivo volume of distribution and predicted volume of  distribution in humans

Elementary aspects of oral bioavailability

The oral bioavailability (F) of a drug is dependent on (a) the absorption of the drug from the gastrointestinal (GI) tract and (b) the capability of the liver to clear the drug during its first pass-through the portal venous system. Oral bioavailability may be described as the fraction of the total oral dose for which systemic exposure is achieved. It is a measurement of the extent of exposure and contrasts with the rates of absorption or elimination discussed above.

Clinically, F is found by comparing the systemic exposures that result after intravenous and (usually) oral doses of the same drug. Note that this comparison need not be for doses of the same size (an important consideration when assessing the tolerability aspects of a proposed normal volunteer study). It is, in fact, preferable to achieve concentrations in the same range as the two doses. Typically, Cmax for a standard dose is going to be higher after bolus intravenous dosing

(IV) than after oral administration (PO), and adverse effects of new agents are likely to be concentration-dependent. The relevant equation is: Fð%Þ ¼ ½ðAUCPO DoseIVÞ=ðAUCIV DosePOÞ� 100% where AUC is the area under the time–plasma concentration curve after each of the respective administrations (the dose terms cancel when equally sized doses are administered by both routes of administration). A residual of less than 15% (sometimes 10% of the total AUC is a commonly used standard for timing the last plasma sample. These studies are usually conducted under standard conditions and using crossover protocols, although, occasionally, a double-label study may be used to measure F instantaneously. Comparison of generic within no gator's formulations, and slow-release with rapidly absorbed formulations may be done using equations of the same form. Similarly, subcutaneous and intravenous injections can be compared. With very rare exceptions, the intravenous administration of a dose is assumed to be 100% bioavailable. For example, very short-acting drugs, for example, some arachidonate derivatives, remifentanil, esmolol, and adenosine, may be metabolized during their first return circulation after intravenous administration and still not achieve 100% ‘bioavailability’. Also, the concept does not apply to topically acting drugs. However, assessing the bioavailability of these drugs by any other route of administration is usually pointless, unless there is some highly specialized issue, for example, absorption after intrathecal administration or potential for drug abuse.

Fluctuation of plasma drug concentration is an important aspect of the bioavailability of slow-release formulations, which almost always have lower Cmax values for a standard dose size than, albeit similar AUC to, a more rapidly absorbed tablet. Assuming that the assay can handle the inevitably lower plasma concentrations, a useful measure of fluctuation, after the initial absorption phase of the curve and during the next four half-lives of elimination is:

ðCmax  CminÞ=Cavg

where Cavg is the average concentration during the specified period; whether to use the arithmetic or geometric average is a controversy, with respected protagonists on both sides. Prediction of oral bioavailability Oral bioavailability can be predicted using the following equation: F ¼ Fað1  Cl=QÞ where Fa represents the fraction of drug absorbed through the intestinal lining, Cl is the hepatic clearance (predicted from in vitro studies; see earlier section) and Q is the hepatic blood flow

in humans (, Rane et al., 1977). Octanol/water partitioning has traditionally been used to predict the fraction absorbed through the intestinal lining. Recently, Caco-2 cell permeability studies have replaced the use of octanol and buffer partitioning studies. Yee (1997){17} established a relationship between Fa and Caco-2 cell permeability, expressed as the apparent permeability constant (Papp), as follows:

The use of Caco-2 cell permeability studies has resulted in more accurate oral bioavailability predictions. Using the predicted hepatic clearance for compound X in humans (see above), estimating Fa by extrapolation from the Caco-2 cell Papp and assuming hepatic blood flow for humans (Rane et al., 1977) of 20 ml min 1 kg the human oral bioavailability of 69–98% is predicted for compound X. This compares well with the known oral bioavailability of this compound in rats and dogs (83 and 72%, respectively).                                                                                                                                                    

Prediction from animals to humans in vivo Elementary aspects

Allometric scaling is an empirical method for predicting physiological, anatomical, and pharmacokinetic measures across species in relation to time and size (Boxenbaum, 1982; Ings, 1990;

Boxenbaum and DiLea, 1995) {18,19,20}. Allometric scaling is based on similarities among species in their physiology, anatomy, and biochemistry, coupled with the observation that smaller animals perform physiological functions that are similar to larger animals, but at a faster rate. The allometric equation is Y ¼ aWb and a log transformation of this the formula yields the straight line:

where Y is the pharmacokinetic or physiological the variable of interest, a, is the allometric coefficient (And log a is the intercept of the line), W is the body weight and b is the allometric exponent (slope of the line) One of the first applications of allometric scaling was the use of the toxicity of anticancer agents in animals to predict toxicity in human children. It was observed that the toxic dose of a drug is similar among species when the dose is compared on the basis of body surface area (Freireich et al., 1966) {21}. For most vertebrate species, the body weight/ volume ratio varies very little, but the surface area/volume ratio increases as species become smaller. Allometric correction of dose multiples in toxicology (compared with proposed human doses) is thus important, especially when small rodents provide the principal toxicology coverage. Body surface area (Y) is related to body weight (W, in kg) by the formula:

This allometric relationship between body surface area and species body weight then allows for a simple conversion of drug doses across species (Figure 8.3), and allometrically equivalent doses of drugs (mg/kg) can be calculated for any species (Table 8.4). The conversion factor (km) is simply the body weight divided by the body surface area. Thus, using the km factors, the dose in Species 1 (in mg/kg) is equivalent to (kmspecies2/kmspecies1) times the dose in Species 2 (in mg kg 1 For example, a 50 mg kg1 dose of the drug in mice would be equivalent to a 4.1 mg kg-1 dose in humans, that is approximately one-twelfth of the dose (Table 8.4). Likewise, the conversion factor can be used to calculate equivalent doses between any species. An equivalent dose in milligrams per kilogram in rats would be twice that in mice.

Allometric approaches to drug discovery Using limited data, allometric scaling may be useful in drug discovery. We assume that, for the formula Y ¼ aWb, the value of the power function ‘b’ (or slope of the line from a log vs. log plot) is drug independent, unlike the intercept ‘a’, which is drug dependent. By doing this, we can use data from a single species (rat) to successfully predict the pharmacokinetics of compound X in humans

Figure 8.4 : Complex Dedrick plot of rat and human data for compound X again showing very good scaling

between rat and human half-life would be 14.5 h, and the plasma clearance would be 0.138 l h 1 kg 1 and the V1, Vdss and Vdb be 1.01, 2.37 and 2.56 kg, respectively. The predictions using rat data were within 15% of the actual mean values in human volunteers. A complex Dedrick plot of the rat and the human data showed nearly superimposed concentration-time curves (Figure 8.4).

This illustrates how allometric scaling is a useful part of the drug discovery process: we avoided studying irrelevant doses and saved time. Ideally, allometric scaling should be done using pharmacokinetic data from at least four species, even though accurate predictions can be made using data from a single species. If possible, information about differences in metabolism among species should be considered when making predictions.

Pharmacokinetic/ pharmacodynamic models’ Elementary aspects The possibility that times since the dose changes the relationship between pharmacological effect size and drug concentrations in plasma have been known for a long time (Levy, 1964, 1966; Levy and

Nelson, 1965; Wagner, 1968; Curry, 1980) {22,23,24,25}, The pioneering work was done by Levy and his colleagues in the 1960s on single-dose plasma levels. effect relationships and on the duration of action of drugs as a function of dose. Brodie and colleagues had shown even earlier how complicated relationships are when drugs have multiple components. distribution is studied in this context (. Brodie, 1967) {26}. Lasagna and colleagues, using diuretics, found that, depending on whether a cumulative effect (24-hour urine production) or an ‘instant’ effect (rate of urine flow at a particular time) was measured, different relationships of response were possible (Murphy et al., 1961) [27]. Nagashima et al. (1969) [28] demonstrated the relative time courses of anticoagulant concentration and effect. Thus, the relationship between effect size and concentration of drug in plasma should not be expected to be constant or simple, and it can vary with time.

The objectives of modern analysis of drug action are to delineate the chemical or physical interactions between the drug and target cell and to characterize the full sequence and scope of actions of each drug (Ross, 1996) [29]. Preclinical models describing the relationship between the concentration of drug in blood or plasma and drug-receptor occupancy or functional response provides clinically useful tools regarding potency, efficacy, and the time course of effect.

Potency is an expression of the activity of a compound, in terms of either the concentration or amount needed to produce a defined effect. Emax is the maximal drug-induced effect. EC50 is the concentration of an agonist that produces 50% of the maximal possible response. An EC50 can be described for drug concentrations using in vitro assays or as a plasma concentration in vivo. IC50 is the concentration of an antagonist that reduces a specified response to 50% of its former value?

A measure of the tendency of a ligand and its receptor to bind to each other is expressed as Kd in receptor occupancy studies. Kd is the equilibrium constant for the two processes of drug-receptor combination and dissociation. Kd may be found for both agonists and antagonists, although sometimes the former poses a more technical challenge due to alterations to the conformation of the binding site. In contrast, efficacy is a relative measure. amongst different agonists, describing the response size for a standard degree of receptor occupation (Jenkinson et al., 1995). When an agonist must occupy 100% of available receptors to cause Emax, its efficacy may be said to be unity. If occupation of all receptors achieves a response that is less than Emax, then the agonist’s efficacy is less than 1 and equal to the ratio of observed maximal effect to maximal effect for an agonist with efficacy 1 (we call these partial agonists or agonist–antagonists). Some agonists need to occupy only a subset of the available receptors, to achieve Emax, and these have efficacy greater than unity. In the latter case, the concentration-response curve lies to the left of the concentration-receptor occupancy curve (e.g., Minneman et al., 1983). Drugs with efficacy 1 are also called full agonists.

Below, we present some model relationships between observed concentration and effect size, as examples from a considerable volume of literature. The reader is referred to key texts for comprehensive coverage of this topic (. Smolen, 1971; Gibaldi and Perrier, 1982, Dayneka et al.,

1993; Levy, 1993; Lesko and Williams, 1994; Colburn, 1995; Derendorf and Hochhaus, 1995; Gabrielsson and Weiner, 1997; Sharma and Jusko, 1997{30}

Pharmacokinetic-pharmacodynamic (PK/PD) modeling Single-compartment, time-independent PK/PD models

The simplest model is where (a) the drug is divided into a single compartment, represented by plasma, and (b) the effect is instantaneous. the direct function of the concentration in that compartment. In this situation, the relationship between drug concentration (C) and a pharmacological effect (E) can be simply described by the linear function:

where S is a slope parameter. If the measured effect has some baseline value (E0), when the drug is absent e.g. physiological, diastolic blood pressure, or resting tension on the tissue in an organ bath), then the model may be expressed as:

The parameters of this model, S and E0, maybe estimated by linear regression. This model does not contain any information about the efficacy and potency, cannot identify the maximum effect and thus cannot be used to find EC50.

When the effect can be measured for a wide concentration range, the relationship between the effect and concentration is often observed to be curved-linear. A semi-logarithmic plot of effect versus log concentration commonly linearizes these data within the approximate range of 20–80% of maximal effect. This log transformation of the concentration axis facilitates a graphical estimation of the slope of the linear segment of the curve:

Where m and C0 are the slope and the hypothetical baseline concentration (usually zero, but not for experiments of add-on therapy or when administering molecules that are also present endogenously), respectively. In this equation, the pharmacological effect may be expressed, when the drug concentration is zero, as:

As mentioned earlier, for functional data based on biophase, plasma, or tissue measurements, we often represent potency as EC50, and when two compounds are compared concerning potency, the one with the lowest EC50 value has the highest potency. A general expression for the observed effect is by analogy with the Michaelis–Menten equation (above) is:                                         

There are various forms of this function for agonist (stimulatory) and antagonist (inhibitory) effects. For example, if there is a baseline effect (E0), then this may be added to the right-hand side of the

equation:

Alternatively, the relationship between concentration and effect for an antagonist, including a baseline value, is:

In the Emax model above, plasma concentration and EC50 is raised to the power of n (Hill factor). to 1. A more general form of the equation is the sigmoid curve:

where, by addition of a single parameter (n) to the Emax model, it is possible to account for curves which are both shallower and steeper than when n ¼ 1 (i.e. unlike the ordinary Emax models). Note that the sigmoidicity parameter (n) does not necessarily have a direct biological interpretation and should be viewed as an extension of the original Emax model to account for curvature. The larger the value of the exponent, the more curved (steeper, concave downwards) is the line. A very high exponent can be viewed as indicating an all-or-none effect (e.g. the development of an action potential in a nerve). Within a narrow concentration range, the observed effect goes from all to nothing or vice versa. An exponent less than unity (<1>

In vivo, these models, analogous to the classical dose or log dose-response curves of in vitro pharmacology are limited to direct effects in single-compartment systems. These models make no concession in the intervening time-established occurrences in drug response. complex PK/PD and event-susceptible models

The maximum common technique for in vivo pharmaco energetic and pharmacodynamic displaying includes next evaluation of the aggregation towards duration and effect towards time file, aforementioned that the lively model specifies an independent, changeable, to a point aggregation, forceful the dynamics. most effective in limited positions manage it and predict that the effect affects the kinetics, e.g. assets on ancestry flow or drug approval itself.

Levy (1964), Jusko (1971) and Smolen (1971), 1976) described the reasoning of dosage-reaction possibility data. They grew a hypothetical footing for the act of this reasoning from the dossier were given from the statement of the time direction of pharmacological response, later a sole prescription of the drug, by using any path of presidency. Smolen (1976) lengthened the reasoning to request of dose–solution length file for bioequivalence experiment.

In dose-response occasion models, the latent the assumption is that the pharmacodynamic file gives us records of the movement of drugs inside the biophase (that is, the tissue or subdivision exactly places the drug well-known shows allure effect). In other words, obvious half growth, bioavailability, and effectiveness maybe acquired concurrently from the measure solution–length dossier. thinking about the sort of version, presumptuous (a) first-order advice/benefit processes and (b) extravascular drug, the kinetic version before driving the limit feature of the critical version. it is an essential behavior this is illustrated apiece answer version. A zero-order advice and first-order productivity rule the change of the response. This allows us to acknowledge the situations place the crimson frame fluid aggregation represents transfer of the drug to an impact phase; the opportunity course of drug aggregation and effect (each within the biophase) is diverse from that, definitely observed in pores and skin concentrations. the amount of drug in a single hypothetic section, eventually an endovenous (IV) dose is typically designed to accompany the mono-competitive decline and is analogous to the ‘plasma disappearance’

curve (above):

The amount of drug in a single hypothetical compartment after an extravascular dose is then modeled with first-order input/output kinetics:

Concentration–time effect modeling is illustrated by the following example, which was chosen to illustrate a single dose of the drug causing the reversal of a symptom (pain). Many other types of examples exist. The plasma kinetics of the analgesic were describable by the following expression after the intravenous bolus dose, with C0 ¼ 45:0 and K ¼ 0:50 h 1 :

In the same study, effect measurements were recorded during 80 minutes, as shown in Figure 8.5. Often, drug effects do not parallel changes in plasma concentration. This can result from distribution phenomena, such as when the effect occurs outside the plasma compartment (e.g. the sedative effect of a dose of benzodiazepine which occurs in the brain), or when the effect recorded reflects, for example, a chain of biochemical events triggered by the presence of a drug (e.g. the aborting of a migraine attack by a serotoninergic drug). About the first of these possibilities, a model, sometimes called a ‘link model’ (also called the ‘effect-compartment’ or the ‘effect-distribution’ model), allows estimation of the in vivo pharmacodynamic effect from non-steady-state effect € versus time and concentration (C) versus time data, within which potential exists for observed E and C to display temporal displacement concerning each other (Segre, 1968; Wagner, 1968; Dahlstrom et al., 1978; Sheiner et al., 1979). The rate of change of drug amount (Ae) in a hypothetical effect compartment can be expressed as:

where A is the amount of drug in the central compartment of a pharmacokinetic model, linked to the effect compartment, with first-order rate constants k1e and ke0. The corresponding expression for the amount of drug in the effect compartment, for a


 

 

Figure 8.5 Observed effect-time data for an analgesic a one-compartment model with bolus input of a dose

(D) is:

where K is the elimination rate constant. The concentration of the drug in the effect compartment, Ce, is obtained by dividing Ae by the effect compartment volume, Ve:

At equilibrium, the rates of drug transfer between the central and effect compartments are equal:

If the partition coefficient, Kp, equals Ce/C in the equation (i.e. kale ¼ ke0) yields:

 At equilibrium, C will be equal to Ce/Kp by definition, and thus:

 This is how the link model relates to the kinetics in plasma to the kinetics of the drug in the effect compartment. When used together with the Emax model for estimation of the maximal drug-induced effect, the concentration at half-maximal effect (apparent EC50) and the rate constant of the disappearance of the effect (ke0):

Computer fitting of the equations to the effect data and estimation of the rate constant for the disappearance of the effect, ke0, EC50, and Emax follows, assuming the sigmoidicity factor (n) to be equal to unity.

At steady state, Ce is directly proportional to the plasma concentration (C), as Ce ¼ KpC. Consequently, the potency (EC50) obtained by regressing the last two equations represents the steady-state plasma concentration producing 50% of Emax. Note that the effect equilibration rate constant (ke0) may be viewed as a first-order distribution rate constant. It can also be thought of in terms of the rate of presentation of a drug to a specific tissue, determined by, for example, tissue perfusion rate, apparent volume of the tissue, and eventual diffusion into the tissue. The results of the data fitting in this exercise with the analgesic are Emax 4.5; EC50 0.61 ng ml 1 and ke0 0.07 h 1 Effect compartment or link models are limited by their applicability to situations in which the equilibrium between plasma and response is due to distributional phenomena. In reality, there is often a delay between the occurrence of maximum drug concentration in the effect compartment and the maximum intensity of the effect caused by slow development of the effect rather than slow distribution to the site of action. In this situation, indirect or ‘physiological substance’ models are more appropriate (Dayneka et al., 1993; Levy, 1994; Sharma and Jusko, 1997). Warfarin is a good example, where this drug inhibits the prothrombin complex activity (PCA) (inhibition of production of effect). This is illustrated by the following example, which relates changes in S-warfarin concentration to the observed PCA. The dose was intravenous. The change in PCA is shown in Figure 8.6. The plasma kinetics of (S)-warfarin were described by the following mono-exponential expression: CwðsÞ ¼ 1:05 e0:0228 t and the equation for the turnover of clotting factor


 

Figure 8.6: Observed PCA time course following the administration of an intravenous bolus dose of warfarin

In this equation, kd is the apparent first-order degradation rate constant (also called kout). This constant can be obtained experimentally from the slope of a ln(P) versus time plot, after administration of a synthesis-blocking dose of coumarin anticoagulant (Nagashima et al., 1969; Pitsui et al., 1993). P0 is the baseline value of the prothrombin time, Cw(s) is the concentration of (S)-warfarin and IC50s is the the concentration of warfarin at 50% of maximal blocking effect. It was also possible to estimate the half-life of the apparent first-order degradation. An alternative model, including a lag time to allow for distributional effects embedded in the observed time delay of the onset of the effect after warfarin administration, was published by Pitsui et al. (1993). Setting the baseline value of clotting factor activity in the absence of warfarin (P0) to a fixed mean of three predose measurements, the program can estimate that parameter.

The model equations are as follows:

where I(CwðsÞ) is the inhibition function of warfarin. It is appropriate to substitute Kin with kd P0. Inhibition of synthesis (rate in) rate of drug infusion into a one-compartment system. The time to steady state is only governed by the elimination rate constant and not the rate of infusion. At steady state: If the baseline condition for PCAwith no inhibition of drug is:

 

then the steady-state condition for the pharmacological response (PCAss) with drug present becomes:

and where I(CwðsÞ) am a function of Cw(s), n and IC50s, then:

As stated before, the intensity of a pharmacological response may not be due to a direct effect of the drug on the receptor. Rather, it may be the net result of several processes only one of which is influenced by the drug. The process that is influenced by the drug must be identified and an attempt be made to relate plasma drug concentration to changes in that process. Warfarin provides a good example of this, as the anticoagulant (hypo thrombinemic) effect is an inhibition of the synthesis of certain vitamin K-dependent clotting factors.

Initial parameter estimates were obtained from the PC Aversus time data. The baseline value (120 s) was obtained from the intercept on the effect axis.

This value is the ratio Kin/kd. From the intercept and slope, Kinwas calculated to be 3.5 s h 1. The plasma concentration at the time of the trough of the effect corresponded approximately with the EC50 value. Thus,

has an impact on the peak (trough) level rather than the time to the peak. This is similar to a constant P0 ¼ 130 s and tlag ¼ 0 h. The computer fitting gave 0.262 9.46 for the IC50, 0.033 17.9 for kd, 2.68 39.6 for n and 121 58 for P0 (limits are CV%) with no lag time. Precision increased when a finite lag time was included in the fitting. As stated earlier, these are two of the many examples that can be chosen to illustrate principles. These two cases, however, are especially relevant to the relationship between animal work and phase I studies in which only the simplest effects, such as counteraction of a painful stimulus or raising/lowering of a physiological parameter such as PCA, are likely to be commonly measured. The reader is again referred to standard texts for a more thorough treatment of models of this kind (Sharma and Jusko, 1997).

Prospectus

In the future, models will exist which will link constants for in vitro binding to cloned human receptors (Kd), data from in vitro functional assays (IC50) and animal and human in vivo EC50 values.

A composite prediction matrix will be applied rapidly and accurately to the process of synthesis of new compounds for phase I testing. In the shorter term, what can we now do to expedite the drug selection process? Figure 8.8 represents a flow chart illustrating one form of metabolism/pharmacokinetics input into the drug discovery process. Arrows (indicating the flow of work and communication) pointing to the right represent perceived progress, whereas arrows pointing to the left represent ‘disappointments’ (and other feedback) leading to corrections and revisions. The numbered asterisks indicate continuations. The ‘flow of time’ is from left to right and from the top panel to the bottom panel. The rectangles indicate tasks that are to be completed, and rectangles in a a column within a panel represents work done by different departments which may be simultaneous or not simultaneous but does not require much interaction between the investigators involved.

Unlike the flow chart of a computer program, after which the diagram is modeled, most of the 

Decisions

PHASE I: THE FIRST OPPORTUNITY FOR EXTRAPOLATION FROM ANIMAL DATA

Figure 8.7Correlation (r ¼ 0:993, p < 0:001) between benzodiazepine-free drug concentrations EC50 units producing 50% of the maximal EEG effect (change in amplitudes in the aˆ frequency band, as determined by aperiodic EEG analysis) and affinity to the GABA–benzodiazepine receptor complex 

(Ki). Binding to the benzodiazepine receptor was determined on basis of displacement of [3 H] flumazenil in washed brain homogenate at 37 C for six drugs B, IA, M, F, O, and C. (Reproduced with permission from Danhof and Mandema, 1995) are made in discussions among committee members and may not necessarily be based on hard and fast criteria. Also, unlike a computer flow chart, a decision concerning a particular drug will usually be based in part on the results of work with other compounds that have the same indication.

In the boxes representing tasks to complete in the phase I study in humans, we have used the symbol 1 to represent work that can be expedited by good 

validated preclinical data. The symbol 2 represents the tasks that can be expedited by online pharmacokinetic modeling. Among the pharmacokinetic questions that will be asked online in the phase I trial are the following:

1. As the doses are escalated, do the kinetics of the drug appear to be linear or nonlinear over the dose range?

2. With repeated dosing, is there any evidence of a change in kinetics, for example, a higher elimination rate that might be indicative of auto induction?

3. Does the drug accumulate in tissues more than predicted with repeated dosing?

4. If preclinical work identified metabolite(s) to measure in humans, are the pharmacokinetics of metabolite(s) linear and as predicted?

5. Does the relationship between concentration and effect change with dose, time, and duration of treatment?

We expect that the task lists represented by some of the boxes will increase. For example, within the box including ‘in vitro intrinsic clearance’, there may be in vitro predictors of oral availability and measures of potentially toxic metabolites. The ‘in vivo pharmacokinetics’ in rats may include an increasing the number of compartments whose concentrations are measured by microdialysis and may include measures of a few selected metabolite concentrations.

This diagram is not a comprehensive guide to drug discovery. However, it does show that the chemists discover new chemical entities with desirable properties. In vitro, biochemistry is followed by initial in vivo work in the rat which is conducted with pharmacokinetic support and in vitro drug metabolism in parallel. Compounds meeting pre-arranged criteria proceed through

Figure 8.8: Flow diagram for involvement of pharmacokinetic and pharmacodynamic mode/computer-generated

feedback into the iterative process of drug discovery from medicinal chemistry to the decision to enter phase II trials. This is not a comprehensive flow diagram for all aspects of drug discovery – it is restricted to the components of the process discussed in this chapter. This flow diagram emphasizes efficient involvement of in vitro and in vivo experimental science and computer modeling, in review of data obtained in phase I studies, in the decisions related to selection of the best compound for patient studies pharmacological screening to general pharmacology and toxicology, all with pharmacokinetic support, which involves the development of pharmacokinetic and pharmacodynamic models.

As a chemical series develops, correlations such as that in Figure 8.6 are developed. Eventually, a compound or compounds is/are chosen for phase I studies.

In this scheme, phase I is influenced by pharmacokinetic and pharmacodynamic modeling. This modeling is used to refine the phase I  protocol, providing advice on sampling times, doses and warning signs of difficulty if they occur, as well as permitting comparison of, for example, EC50 data from humans with EC50 data from animals and in vitro/in vivo comparisons. The objective is the judicious choice of the best compound, with the ever-present limitations on the information available. Note that this scheme can involve feedback from Phase I to renewed chemical synthesis, as well as a choice of a second or third compound for human testing. Currently, phase I studies themselves tend to be quite straightforward and focused on single compounds. Typically, after adequate preclinical characterization of a candidate drug and 14-day and/or 3-month multiple-dose toxicology studies in two mammalian species, a very low dose is chosen for the first human exposure to the drug. In later exposures, the dose is escalated according to some pre-arranged criteria until the drug concentrations in plasma associated with undesirable properties in animals are reached and/or until some other limiting the response is threatened or observed in the human volunteers. Doses may be single or short multiple-dose series. Simple physiological and biochemical measurements are routinely made to monitor for safety. If possible, responses to the drug are also measured when relevant to the intended therapeutic use. A drug successfully passes to phase II if with appropriate plasma levels, responses are predictable, reversible, and related to the known pharmacological mechanisms of the drug and there is a viewpoint among the investigators concerned that the drug could safely be given in initial studies to patients from its target population. Hopefully, all or most of what is observed in phase I is in line with predictions based on the pharmacokinetic and pharmacodynamic properties of the drug in animals. Once phase I is complete, the humans become the first-choice test species, under all but the most specialized of circumstances (e.g. effects on reproduction). In this context, phase I serves as the interface between preclinical research and clinical development, and the validity of the predictions from animals to humans involved is of paramount importance. We believe that with enhanced integrated study of animals and humans and with data feedback based on computer models, the process of drug discovery from synthesis to proof of safety in humans could be dramatically improved in its efficiency. This is beyond what has traditionally been expected from departments of drug metabolism and pharmacokinetics (Welling and Tse, 1995). The time saved could be used to permit a a larger number of compounds with better prospects, from a single research program, to be compared to phase I studies. Consequently, the extremely costly testing programs for patients which follows phase I could be started sooner and conducted better.

Research Methodology:

The research methods for this study complicated an orderly approach to gathering appropriate essays and case studies concerning the prediction of data from animal models to human uncovering in Phase I clinical troubles. Key databases in the way that PubMed, Scopus, and Web of Science were fully hunted for utilizing a combination of particular keywords containing "Phase I dispassionate troubles," "prediction," "animal models," and "human exposure." The additional tests circumscribed studies that determined judgments into methods, challenges, and outcomes guide translating preclinical verdicts to human issues. Both exploratory studies and hypothetical models were considered, guaranteeing an inclusive test of the material.

Results

The results of the history review disclosed a diverse array of methods working in inferring dossier from animal models to think human exposure all the while Phase I troubles. Notable approaches contained physiologically-located pharmacokinetic posing, allometric scaling, artificial-in vivo prediction methods, and populace pharmacokinetic posing. Case studies were recognized to exemplify the realistic request of these methods in foreseeing human pharmacokinetics and pharmacodynamics from animal data. Furthermore, the results highlighted important challenges, to a degree interspecies dissimilarity in plant structure, absorption, and plant structure, as well as disadvantages owned by preclinical models.

Discussion

The consultation portion precariously analyzed the suggestions of inferring animal dossier to human uncovering in Phase I troubles. It provided a meticulous test of the substances and disadvantages guiding various extrapolation methods, stressing the impact of interspecies instability on predicting veracity. Additionally, ethical concerns encircling the use of animal models in drug growth were discussed, alongside the need for surveying alternative approaches. The discussion emphasized the significance of cooperative exertions with researchers, clinicians, and supervisory instrumentalities to address the challenges formal by interspecies dissimilarities and embellish the translational validity of the preclinical dossier.

Conclusion

In conclusion, connecting the breach middle from two points animal dossier and human uncovering in Phase I trials is authoritative for guaranteeing the security and productiveness of novel cures. The study emphasized the need for constant cultivation of prediction methods, confirmation of predicting models, and exploration of alternative methods to decrease confidence in animal studies. By reconstructing the translational genuineness of preclinical data, the gain rate of Phase I tests may be embellished, eventually facilitating the development of creative analyses for dispassionate use

Acknowledgement

We no longer have our genuine appreciation for the many things and arrangements whose offerings and support fashioned this research project attainable. We are intensely appreciative to all those the one donated to the happiness concerning this project. We would like to express our straightforward recognition to our tutors, Naweed Imam Syed, Professor in the Department of Cell Biology at the University of Calgary, and Dr. Sadaf Ahmed from the Psychophysiology Lab at the University of Karachi, for their priceless counseling and support during the whole of the wholeness concerning this research endeavor. Their acumens and knowledge played an important act in forming the course concerning this project.

Declaration of Interest

I herewith reveal that I have no monetary or different private interests, either direct or roundabout, in some matter that conceivably contradict my maturities as an investigator concerning this study.

Management Conflicts of Interest

The authors prove that they have no conflicts of interest to reveal.

Financial Support and Protection

No extrinsic capital was wanted or taken to support the growth concerning this study.

References

Clearly Auctoresonline and particularly Psychology and Mental Health Care Journal is dedicated to improving health care services for individuals and populations. The editorial boards' ability to efficiently recognize and share the global importance of health literacy with a variety of stakeholders. Auctoresonline publishing platform can be used to facilitate of optimal client-based services and should be added to health care professionals' repertoire of evidence-based health care resources.

img

Virginia E. Koenig

Journal of Clinical Cardiology and Cardiovascular Intervention The submission and review process was adequate. However I think that the publication total value should have been enlightened in early fases. Thank you for all.

img

Delcio G Silva Junior

Journal of Women Health Care and Issues By the present mail, I want to say thank to you and tour colleagues for facilitating my published article. Specially thank you for the peer review process, support from the editorial office. I appreciate positively the quality of your journal.

img

Ziemlé Clément Méda

Journal of Clinical Research and Reports I would be very delighted to submit my testimonial regarding the reviewer board and the editorial office. The reviewer board were accurate and helpful regarding any modifications for my manuscript. And the editorial office were very helpful and supportive in contacting and monitoring with any update and offering help. It was my pleasure to contribute with your promising Journal and I am looking forward for more collaboration.

img

Mina Sherif Soliman Georgy

We would like to thank the Journal of Thoracic Disease and Cardiothoracic Surgery because of the services they provided us for our articles. The peer-review process was done in a very excellent time manner, and the opinions of the reviewers helped us to improve our manuscript further. The editorial office had an outstanding correspondence with us and guided us in many ways. During a hard time of the pandemic that is affecting every one of us tremendously, the editorial office helped us make everything easier for publishing scientific work. Hope for a more scientific relationship with your Journal.

img

Layla Shojaie

The peer-review process which consisted high quality queries on the paper. I did answer six reviewers’ questions and comments before the paper was accepted. The support from the editorial office is excellent.

img

Sing-yung Wu

Journal of Neuroscience and Neurological Surgery. I had the experience of publishing a research article recently. The whole process was simple from submission to publication. The reviewers made specific and valuable recommendations and corrections that improved the quality of my publication. I strongly recommend this Journal.

img

Orlando Villarreal

Dr. Katarzyna Byczkowska My testimonial covering: "The peer review process is quick and effective. The support from the editorial office is very professional and friendly. Quality of the Clinical Cardiology and Cardiovascular Interventions is scientific and publishes ground-breaking research on cardiology that is useful for other professionals in the field.

img

Katarzyna Byczkowska

Thank you most sincerely, with regard to the support you have given in relation to the reviewing process and the processing of my article entitled "Large Cell Neuroendocrine Carcinoma of The Prostate Gland: A Review and Update" for publication in your esteemed Journal, Journal of Cancer Research and Cellular Therapeutics". The editorial team has been very supportive.

img

Anthony Kodzo-Grey Venyo

Testimony of Journal of Clinical Otorhinolaryngology: work with your Reviews has been a educational and constructive experience. The editorial office were very helpful and supportive. It was a pleasure to contribute to your Journal.

img

Pedro Marques Gomes

Dr. Bernard Terkimbi Utoo, I am happy to publish my scientific work in Journal of Women Health Care and Issues (JWHCI). The manuscript submission was seamless and peer review process was top notch. I was amazed that 4 reviewers worked on the manuscript which made it a highly technical, standard and excellent quality paper. I appreciate the format and consideration for the APC as well as the speed of publication. It is my pleasure to continue with this scientific relationship with the esteem JWHCI.

img

Bernard Terkimbi Utoo

This is an acknowledgment for peer reviewers, editorial board of Journal of Clinical Research and Reports. They show a lot of consideration for us as publishers for our research article “Evaluation of the different factors associated with side effects of COVID-19 vaccination on medical students, Mutah university, Al-Karak, Jordan”, in a very professional and easy way. This journal is one of outstanding medical journal.

img

Prof Sherif W Mansour

Dear Hao Jiang, to Journal of Nutrition and Food Processing We greatly appreciate the efficient, professional and rapid processing of our paper by your team. If there is anything else we should do, please do not hesitate to let us know. On behalf of my co-authors, we would like to express our great appreciation to editor and reviewers.

img

Hao Jiang

As an author who has recently published in the journal "Brain and Neurological Disorders". I am delighted to provide a testimonial on the peer review process, editorial office support, and the overall quality of the journal. The peer review process at Brain and Neurological Disorders is rigorous and meticulous, ensuring that only high-quality, evidence-based research is published. The reviewers are experts in their fields, and their comments and suggestions were constructive and helped improve the quality of my manuscript. The review process was timely and efficient, with clear communication from the editorial office at each stage. The support from the editorial office was exceptional throughout the entire process. The editorial staff was responsive, professional, and always willing to help. They provided valuable guidance on formatting, structure, and ethical considerations, making the submission process seamless. Moreover, they kept me informed about the status of my manuscript and provided timely updates, which made the process less stressful. The journal Brain and Neurological Disorders is of the highest quality, with a strong focus on publishing cutting-edge research in the field of neurology. The articles published in this journal are well-researched, rigorously peer-reviewed, and written by experts in the field. The journal maintains high standards, ensuring that readers are provided with the most up-to-date and reliable information on brain and neurological disorders. In conclusion, I had a wonderful experience publishing in Brain and Neurological Disorders. The peer review process was thorough, the editorial office provided exceptional support, and the journal's quality is second to none. I would highly recommend this journal to any researcher working in the field of neurology and brain disorders.

img

Dr Shiming Tang

Dear Agrippa Hilda, Journal of Neuroscience and Neurological Surgery, Editorial Coordinator, I trust this message finds you well. I want to extend my appreciation for considering my article for publication in your esteemed journal. I am pleased to provide a testimonial regarding the peer review process and the support received from your editorial office. The peer review process for my paper was carried out in a highly professional and thorough manner. The feedback and comments provided by the authors were constructive and very useful in improving the quality of the manuscript. This rigorous assessment process undoubtedly contributes to the high standards maintained by your journal.

img

Raed Mualem

International Journal of Clinical Case Reports and Reviews. I strongly recommend to consider submitting your work to this high-quality journal. The support and availability of the Editorial staff is outstanding and the review process was both efficient and rigorous.

img

Andreas Filippaios

Thank you very much for publishing my Research Article titled “Comparing Treatment Outcome Of Allergic Rhinitis Patients After Using Fluticasone Nasal Spray And Nasal Douching" in the Journal of Clinical Otorhinolaryngology. As Medical Professionals we are immensely benefited from study of various informative Articles and Papers published in this high quality Journal. I look forward to enriching my knowledge by regular study of the Journal and contribute my future work in the field of ENT through the Journal for use by the medical fraternity. The support from the Editorial office was excellent and very prompt. I also welcome the comments received from the readers of my Research Article.

img

Dr Suramya Dhamija

Dear Erica Kelsey, Editorial Coordinator of Cancer Research and Cellular Therapeutics Our team is very satisfied with the processing of our paper by your journal. That was fast, efficient, rigorous, but without unnecessary complications. We appreciated the very short time between the submission of the paper and its publication on line on your site.

img

Bruno Chauffert

I am very glad to say that the peer review process is very successful and fast and support from the Editorial Office. Therefore, I would like to continue our scientific relationship for a long time. And I especially thank you for your kindly attention towards my article. Have a good day!

img

Baheci Selen

"We recently published an article entitled “Influence of beta-Cyclodextrins upon the Degradation of Carbofuran Derivatives under Alkaline Conditions" in the Journal of “Pesticides and Biofertilizers” to show that the cyclodextrins protect the carbamates increasing their half-life time in the presence of basic conditions This will be very helpful to understand carbofuran behaviour in the analytical, agro-environmental and food areas. We greatly appreciated the interaction with the editor and the editorial team; we were particularly well accompanied during the course of the revision process, since all various steps towards publication were short and without delay".

img

Jesus Simal-Gandara

I would like to express my gratitude towards you process of article review and submission. I found this to be very fair and expedient. Your follow up has been excellent. I have many publications in national and international journal and your process has been one of the best so far. Keep up the great work.

img

Douglas Miyazaki

We are grateful for this opportunity to provide a glowing recommendation to the Journal of Psychiatry and Psychotherapy. We found that the editorial team were very supportive, helpful, kept us abreast of timelines and over all very professional in nature. The peer review process was rigorous, efficient and constructive that really enhanced our article submission. The experience with this journal remains one of our best ever and we look forward to providing future submissions in the near future.

img

Dr Griffith

I am very pleased to serve as EBM of the journal, I hope many years of my experience in stem cells can help the journal from one way or another. As we know, stem cells hold great potential for regenerative medicine, which are mostly used to promote the repair response of diseased, dysfunctional or injured tissue using stem cells or their derivatives. I think Stem Cell Research and Therapeutics International is a great platform to publish and share the understanding towards the biology and translational or clinical application of stem cells.

img

Dr Tong Ming Liu

I would like to give my testimony in the support I have got by the peer review process and to support the editorial office where they were of asset to support young author like me to be encouraged to publish their work in your respected journal and globalize and share knowledge across the globe. I really give my great gratitude to your journal and the peer review including the editorial office.

img

Husain Taha Radhi

I am delighted to publish our manuscript entitled "A Perspective on Cocaine Induced Stroke - Its Mechanisms and Management" in the Journal of Neuroscience and Neurological Surgery. The peer review process, support from the editorial office, and quality of the journal are excellent. The manuscripts published are of high quality and of excellent scientific value. I recommend this journal very much to colleagues.

img

S Munshi

Dr.Tania Muñoz, My experience as researcher and author of a review article in The Journal Clinical Cardiology and Interventions has been very enriching and stimulating. The editorial team is excellent, performs its work with absolute responsibility and delivery. They are proactive, dynamic and receptive to all proposals. Supporting at all times the vast universe of authors who choose them as an option for publication. The team of review specialists, members of the editorial board, are brilliant professionals, with remarkable performance in medical research and scientific methodology. Together they form a frontline team that consolidates the JCCI as a magnificent option for the publication and review of high-level medical articles and broad collective interest. I am honored to be able to share my review article and open to receive all your comments.

img

Tania Munoz

“The peer review process of JPMHC is quick and effective. Authors are benefited by good and professional reviewers with huge experience in the field of psychology and mental health. The support from the editorial office is very professional. People to contact to are friendly and happy to help and assist any query authors might have. Quality of the Journal is scientific and publishes ground-breaking research on mental health that is useful for other professionals in the field”.

img

George Varvatsoulias

Dear editorial department: On behalf of our team, I hereby certify the reliability and superiority of the International Journal of Clinical Case Reports and Reviews in the peer review process, editorial support, and journal quality. Firstly, the peer review process of the International Journal of Clinical Case Reports and Reviews is rigorous, fair, transparent, fast, and of high quality. The editorial department invites experts from relevant fields as anonymous reviewers to review all submitted manuscripts. These experts have rich academic backgrounds and experience, and can accurately evaluate the academic quality, originality, and suitability of manuscripts. The editorial department is committed to ensuring the rigor of the peer review process, while also making every effort to ensure a fast review cycle to meet the needs of authors and the academic community. Secondly, the editorial team of the International Journal of Clinical Case Reports and Reviews is composed of a group of senior scholars and professionals with rich experience and professional knowledge in related fields. The editorial department is committed to assisting authors in improving their manuscripts, ensuring their academic accuracy, clarity, and completeness. Editors actively collaborate with authors, providing useful suggestions and feedback to promote the improvement and development of the manuscript. We believe that the support of the editorial department is one of the key factors in ensuring the quality of the journal. Finally, the International Journal of Clinical Case Reports and Reviews is renowned for its high- quality articles and strict academic standards. The editorial department is committed to publishing innovative and academically valuable research results to promote the development and progress of related fields. The International Journal of Clinical Case Reports and Reviews is reasonably priced and ensures excellent service and quality ratio, allowing authors to obtain high-level academic publishing opportunities in an affordable manner. I hereby solemnly declare that the International Journal of Clinical Case Reports and Reviews has a high level of credibility and superiority in terms of peer review process, editorial support, reasonable fees, and journal quality. Sincerely, Rui Tao.

img

Rui Tao

Clinical Cardiology and Cardiovascular Interventions I testity the covering of the peer review process, support from the editorial office, and quality of the journal.

img

Khurram Arshad

Clinical Cardiology and Cardiovascular Interventions, we deeply appreciate the interest shown in our work and its publication. It has been a true pleasure to collaborate with you. The peer review process, as well as the support provided by the editorial office, have been exceptional, and the quality of the journal is very high, which was a determining factor in our decision to publish with you.

img

Gomez Barriga Maria Dolores

The peer reviewers process is quick and effective, the supports from editorial office is excellent, the quality of journal is high. I would like to collabroate with Internatioanl journal of Clinical Case Reports and Reviews journal clinically in the future time.

img

Lin Shaw Chin

Clinical Cardiology and Cardiovascular Interventions, I would like to express my sincerest gratitude for the trust placed in our team for the publication in your journal. It has been a true pleasure to collaborate with you on this project. I am pleased to inform you that both the peer review process and the attention from the editorial coordination have been excellent. Your team has worked with dedication and professionalism to ensure that your publication meets the highest standards of quality. We are confident that this collaboration will result in mutual success, and we are eager to see the fruits of this shared effort.

img

Maria Dolores Gomez Barriga