a sulfadimidine model to evaluate pharmacokinetics and residues at

140
Aus dem Institut für Pharmakologie, Toxikologie und Phamazie der Stiftung Tierärztliche Hochschule Hannover A sulfadimidine model to evaluate pharmacokinetics and residues at various concentrations in laying hens INAUGURAL DISSERTATION Zur Erlagung des Grades eines Doktors der Veterinärmedizin (Dr. med. vet.) durch die Stiftung Tierärztliche Hochschule Hannover Vorgelegt von Natthasit Tansakul aus Lopburi/ Thailand Hannover 2008

Upload: others

Post on 15-Feb-2022

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: A sulfadimidine model to evaluate pharmacokinetics and residues at

Aus dem Institut für Pharmakologie, Toxikologie und Phamazie

der Stiftung Tierärztliche Hochschule Hannover

A sulfadimidine model to evaluate pharmacokinetics and residues at various concentrations in laying hens

INAUGURAL DISSERTATION

Zur Erlagung des Grades eines

Doktors der Veterinärmedizin

(Dr. med. vet.)

durch die Stiftung Tierärztliche Hochschule Hannover

Vorgelegt von

Natthasit Tansakul

aus Lopburi/ Thailand

Hannover 2008

Page 2: A sulfadimidine model to evaluate pharmacokinetics and residues at

Wissenchaftliche Betreuung: Univ.-Prof. Dr. Manfred Kietzmann

1. Gutachter: Univ.-Prof. Dr. Manfred Kietzmann

2. Gutachter: Prof. Dr. Gerd Hamscher

Tag der mündlichen Prüfung: 11 November 2008

Page 3: A sulfadimidine model to evaluate pharmacokinetics and residues at

Dedicated to my father, with love and never ending faith &

my beloved mother

Page 4: A sulfadimidine model to evaluate pharmacokinetics and residues at

Contents

Contents

1 Introduction.................................................................................................................7

2 Literature review.........................................................................................................8

2.1 The principle of pharmacokinetics.......................................................................8

2.1.1 Definition and principle ................................................................................8

2.1.2 Pharmacokinetic parameters .........................................................................9

2.2 General considerations of sulfonamides ............................................................14

2.2.1 History and properties of sulfonamides ......................................................14

2.2.2 Pharmacokinetic of sulfonamides ...............................................................15

2.3 Veterinary drug residues ....................................................................................20

2.4 Sulfonamide residues in animals .......................................................................27

2.4.1 Sulfonamide residues in livestock ..............................................................27

2.4.2 Sulfonamides residues in poultry................................................................29

2.5 Sulfadimidime....................................................................................................35

2.5.1 Properties of sulfadimidine .........................................................................35

2.5.2 Pharmacokinetics of sulfadimidine in laying hens .....................................35

2.5.3 Residues of sulfadimidine in laying hens ...................................................38

3 Aims and objectives..................................................................................................40

4 Materials and methods ..............................................................................................41

4.1 Materials ............................................................................................................41

4.2 Methods..............................................................................................................43

4.2.1 Drug administration and sample collection ................................................43

4.2.2 Analysis (HPLC).........................................................................................45

4.2.3 Pharmacokinetic parameters and statistical analyses..................................49

5 Results.......................................................................................................................50

5.1 Method of analysis.............................................................................................50

5.1.1 Selectivity ...................................................................................................50

5.1.2 Linearity......................................................................................................52

5.1.3 Limit of detection (LOD) and limit of quantification (LOQ) .....................54

5.1.4 Recovery .....................................................................................................54

Page 5: A sulfadimidine model to evaluate pharmacokinetics and residues at

Contents

5.1.5 Precision......................................................................................................54

5.2 Plasma concentrations of sulfadimidine following various doses administration

by intravenous and oral in laying hens ....................................................................56

5.3 Pharmacokinetic characteristics of sulfadimidine in laying hens ......................58

5.3.1 Pharmacokinetic characteristics of sulfadimidine following a single

intravenous bolus administration in laying hens..................................................58

5.3.2 Pharmacokinetic characteristics of sulfadimidine following a single oral

bolus administration at different concentrations in laying hens...........................60

5.4 Sulfadimidine residue in eggs after a single oral bolus administration at various

doses in laying hens .................................................................................................65

5.4.1 Sulfadimidine residue in yolk .....................................................................65

5.4.2 Sulfadimidine residue in albumen ..............................................................66

5.4.3 Transfer rate of sulfadimidine into eggs after a single oral administration 67

5.5 Sulfadimidine residue in eggs after medication via mixed feed at various doses

for seven consecutive days in laying hens ...............................................................68

5.5.1 Sulfadimidine residue in yolk .....................................................................68

5.5.2 Sulfadimidine residue in albumen ..............................................................70

5.5.3 Sulfadimidine concentrations in plasma .....................................................73

5.6 Comparative pharmacokinetic behavior and residues of sulfadimidine between

healthy and impaired kidney hens............................................................................74

5.6.1 Comparison pharmacokinetic behaviors of sulfadimidine between healthy

and impaired kidney hens ....................................................................................74

5.6.2 Comparison sulfadimidine residue in eggs between healthy and impaired

kidney hens ..........................................................................................................77

6 Discussion .................................................................................................................80

6.1 Method of analysis.............................................................................................80

6.2 Pharmacokinetic behavior of sulfadimidine following single bolus via

intravenous injection and various doses of single bolus via oral administration in

laying hens ...............................................................................................................82

6.3 Sulfadimidine residues in egg components after a single bolus via oral and

medicated feed at different concentrations ..............................................................86

6.4 Comparison pharmacokinetic characteristics and residues of sulfadimidine

between healthy and impaired kidney laying hens following intravenously

administration ..........................................................................................................89

Page 6: A sulfadimidine model to evaluate pharmacokinetics and residues at

Contents

7 Summary ...................................................................................................................92

8 Zusammenfassung.....................................................................................................94

9 References.................................................................................................................96

10 Appendix...............................................................................................................127

10.1 Glossary of abbreviations ..............................................................................127

10.2 Tables.............................................................................................................130

11 Acknowledgement ................................................................................................139

Parts of the thesis have been published:

TANSAKUL N., F. NIEDORF and M. KIETZMANN (2007):

A sulfadimidine model to evaluate pharmacokinetics and residues at various

concentrations in laying hen.

Food Addit. Contam. 24(6), 598–604

Page 7: A sulfadimidine model to evaluate pharmacokinetics and residues at

Introduction

7

1 Introduction

Currently, more attention is being paid to food safety. Veterinary drug residue is one

of many global issues concerning food contamination. The use of veterinary drugs in

food-producing animals has the potential to generate residues in animal-derived

products and poses a health hazard to the customer. Low-level drug contamination of

animal’s feed is one causative channel of residues in animal products. Even very low

levels of antibiotic contaminations may result in relevant residues in food, as

confirmed by studies on coccidiostats in poultry feed (ROKKA et al., 2005;

KENNEDY et al., 1998) and sulfadimidine in swine (BIEHL et al., 1981;

McCAUGHEY et al., 1990a). In the present study, sulfadimidine (SDD) was used as

a model to determine the pharmacokinetic characteristics and residue risk at various

doses in laying hens.

In former research, pharmacokinetic characteristics of SDD in chicken have been

described (KIETZMANN, 1980; GEERTSMA et al., 1987; NOUWS et al., 1988;

JIAN and PUNIA, 2001). Different doses and routes of SDD residue have also been

studied in laying hens (BLOM, 1975; SEIB, 1991; ROUDAUT, 1993; ROUDAUT

and GARNIER, 2002; FURUSAWA, 2001; KAN, 2003). These studies indicated a

transfer of sulfonamides into the eggs, but mainly provided data focusing on

therapeutic doses. Therefore, information about the pharmacokinetic behavior and

residue risk following the administration of SDD at low doses are scarce. Specially,

very little data are available on residue levels in hens after low-level sulfonamides

contaminations of layer feed. In order to determine whether substance carry-over may

result in relevant residues even in untreated animals, pharmacokinetic characteristics

and residue of SDD at low to high doses in laying hens after a single bolus oral

administration and medication via feed were observed. Moreover, the effect of kidney

dysfunction on kinetic behaviour and residue as compared to healthy laying hens was

also investigated.

Page 8: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

8

2 Literature review 2.1 The principle of pharmacokinetics 2.1.1 Definition and principle

The term pharmacokinetics was first introduced by DOST (1953) in order to describe

the characteristic of drugs after an aplication. It is the study of the time course of drug

and xenobiotic concentration in the body. To increase understanding of a drug’s

behaviour, it is important to examine the four major processes after drug

consumption: absorption, distribution, biotransformation and excretion.

Absorption is described as the process which a compound passes from its site of

administration into the bloodstream. Absorption is influenced by many factors such as

the properties of cell membrane, drug properties and route of administration and

physio-pathological state of the animal (CURRY, 1974). An indication of the rate of

drug absorption is obtained from the peak plasma concentration (Cmax) and time

reaching the maximum concentration (Tmax). Distribution is the process whereby a drug is transported to all the tissues and organs

(LABAUNE, 1989). After entering the systemic circulation, in whatever route of

administration, drugs are conveyed throughout the body and reach their site of action.

There are four major factors responsible for the extent and rate of distribution. These

are the physicochemical properties of the drug, the concentration gradient established

between the blood and tissue, the ratio of blood flow to tissue mass, and the affinity of

the drug for tissue constituents and serum protein binding (RIVIERE et al., 1991).

Only the fraction free-form (unbound) of drug is capable of exiting the circulation to

distribute through the body and exert activity at the site of action (BOTSOGLOU and

FLETOURIS, 2001). The parameter which defines the process of distribution is the

volume of distribution (Vd).

Page 9: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

9

Biotransformation (Metabolism) is the principle mechanism of elimination for the

transformation of drugs or xenobiotics into metabolites by the chemical reaction

(LABAUNE, 1989). The two classes of enzymatic pathways of biotransformation are

phase I (non-synthesis) and phase II (conjugation). Phase I corresponds to

functionalisation processes including oxidation, reduction, hydrolysis, hydration and

isomerisation reactions. Phase II reactions involve conjugation of the drug or phase I

metabolite with the endogenous substrate such as glucuronic acid, sulfate, acetate and

methyl group (RIVIERE et al., 1991). Although some drugs are eliminated from the

body by uncharged, most drugs undergo metabolism where liver is the main organ of

reaction (BOTSOGLOU and FLETOURIS, 2001). In addition, the liver’s function

may change the drug’s form to being inactive and easy to excrete but some drugs may

be converted to an activating form.

Excretion is the process by which the parent drug or its metabolites are removed from

the body fluids. The kidney is the most important site of drug excretion. There are

three renal mechachnisms: glomerular filtration, carrier-mediated proximal tubular

secretion and pH-dependent passive tubular resorption in the distal nephron

(RIVIERE et al., 1991). Renal insufficiency usually significantly affects drug

excretion (BOTSOGLOU and FLETOURIS, 2001). The systemic clearance (CL) and

elimination half-life (T1/2el) are important parameters referring to the overall rate of

elimination (metabolism and excretion). Although most compounds are excreted

primarily by the renal, some drugs are partially or completely excreted through the

bile. It has been reported that there is an extensive species variation among animals in

their general ability to secrete drugs in the bile e.g. chicken are characterised as good

biliary excreters, whereas sheep and rabbit are characterised as moderate and poor

excreters (BOTSOGLOU and FLETOURIS, 2001).

2.1.2 Pharmacokinetic parameters

The drug plasma concentration-time profile can be mathematically predicted if

quantitative estimates of the rate and extent of absorption, distribution and elimination

are available (RIVIERE et al., 1991). After administration, the drug may distribute

into the entire accessible region instantly. In such a case, the body is considered as a

Page 10: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

10

homogeneous container. The disposition of the drug can be described as a one-

compartment open model, as generalised in figure 1.

Administration K01 K10

Figure 1: Schematic of one-compartment open model

Mathematical expression of one-compartment with first-order input, first-order output

and no lag time can be performed following this equation:

C(T) = D*K10 / V*(K01-K10) * (exp(-K10*T)-exp(-K01*T))

C(T) is the plasma drug concentration at any time (T) and D is the dose (amount) of

drug to be administered. K01 and K10 are the apparent first-order absorption and

distribution (elimination) rate constants, respectively. V is an apparent volume of

distribution based on the area under the curve and the coefficient represents the zero-

time intercept (in unit of concentration) of the back-extrapolated disposition phase

corresponding to the zero-time intercept of the absorption phase (BAGGOT, 2001).

2.1.2.1 Area under the curve

The area under the curve (AUC) is the area under a concentration of the analysis

versus time curve. Theoretically, if C(t) denotes the concentration of analysis at time

t, then AUC can be described in equation 1.1. Moreover, the AUC can be simply

obtained from the relationship between dose (D) and total clearance (CL), following

equation 1.2.

AUCab =a ∫b c(t)dt (1.1)

AUC = D/ CL (1.2)

0 1

Page 11: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

11

2.1.2.2 Elimination half-life

The elimination half-life time (T1/2el) indicates the rate of elimination. It is the time

required drug plasma concentration at 50 % of the dose present in the circulation. It is

related to the elimination rate constant (Kel or K10) by the equation 1.3. However,

with the relationship between the volume of distribution and the systemic (body)

clearance of the drug, the general equation for calculating of the elimination half-life

is given in equation 1.4 (BAGGOT, 2001).

T1/2el = 0.693/ Kel (1.3)

T1/2el = 0.693 . Vd(area)/ CL (1.4)

After carrying out the elimination half-life five times, 97 % of a drug could be

eliminated or only 3 % of the dose may remain in the body. However, the hybrid

parameter corresponds with both Vd and CL. Thus, the body can be affected

pathologically making the derived T1/2el an unreliable indicator of drug elimination

(RIVIERE et al., 1991). Regarding the time unit, the Tmax is the observed time after

drug administration at which peak plasma concentration occurs. Generally, the Tmax

can theoretically be calculated by equation 1.5. However, estimating Tmax in equation

1.6 was obtained by using the WinNonLin® program. According to the relationship

with Tmax, Cmax is the maximum concentration at Tmax, this being calculated by

equation 1.7.

Tmax = (ln . Ka/Kel) / Ka- Kel (1.5)

Tmax = ln(K01/K10)/ (K01-K10) (1.6)

ln Cmax = ln Yo – (Kel . Tmax) (1.7)

Yo is the theoretically calculated zero time plasma concentration.

Page 12: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

12

2.1.2.3 Volume of distribution

The apparent volume of distribution (Vd) indicates the apparent space (L/kg) in the

body available to contain the drug. Thus, the volume of distribution at the steady state

Vss can be calculated after intravenous administration. Vd is the apparent distribution

which relates the plasma concentration of a drug to the total amount of drug in the

body at any time after pseudodistribution equilibrium has been attained (equation

1.7).

Vd = Dose/Cp0 (1.7)

Cp0 is the drug concentration at time zero. The volume of distribution can be

calculated by equation 1.8 (BAGGOT, 2001). The volume of distribution can also be

calculated from the total clearance and half-life by using equation 1.9 (LABAUNE,

1989).

Vd(area) = Dose/ AUC . β (1.8)

Vd = T1/2el . CL / 0.693 (1.9)

2.1.2.4 Clearance

The systemic clearance (CL) of a drug represents the sum of the clearances by the

various organs that contribute to eliminating of the drug. Total body clearance is the

sum of the clearances of each organ (renal, hepatic and others) of elimination

(RIVIERE et al., 1991; BAGGOT, 2001). The systemic clearance can be generalised

by the following equation:

CL = F . Dose/ AUC (1.10)

F is the fraction of absorption which assumes complete systemic availability after

intravenous administration. From the relationship with volume of distribution, the

systemic clearance can be obtained by:

Page 13: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

13

CL = Vd(area) . β (1.11)

β is the apparent first-order rate constant fraction associated with elimination per unit

of time after intravenous or extra-vascular administration.

2.1.2.5 Bioavailability

The absolute bioavailability is determined by comparing the area under the curve

(AUC) after extra-vascular administration, with the AUC after intravascular

administration. The “first-pass effect” can substantially reduce the systemic

availability (absolute oral bioavailability) of lipid soluble drugs undergo extensive

biotransformation in the liver (BAGGOT, 2001). The usual method for estimating

systemic availability (F) of drugs after extra-vascular administration of the dose is the

method of corresponding areas, following the equation:

F = AUC(extra-vascular) / AUC (intravascular) (1.12)

2.1.2.6 Mean residence time

The mean residence time (MRT) represents the average time the molecules of a drug

reside in the body following the administration of a single dose (BAGGOT, 2001).

The molecules introduced into the body when the drug is administered, have the same

probability of being absorbed, distributed, metabolised or eliminated. The distribution

curve resulting from this (plasma concentration vs time) can be characterised by its

mean value (MRT) and the variance of this mean (LABAUNE, 1989). Based on the

area under the curve, MRT can be calculated by the following equation:

MRT = AUMC/ AUC (1.13)

AUMC is the area under the (first) moment curve obtained from the product of plasma

concentration and time versus time from time zero to infinity and AUC is the total

area under the curve (zero moment) (BAGGOT, 2001).

Page 14: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

14

2.2 General considerations of sulfonamides

2.2.1 History and properties of sulfonamides

After the discovery of the prototype of sulfonamides (Prontosil) by Gerhard Domagk

in 1935, the active portion known as sulfanilamide was found. Sulfonamides are a

group of compounds containing the structure SO2NH2 (see table 4 page 35). Over

5400 derivatives of sulfanilamide were created, but approximately 20 derivative

forms are generally available. Sulfonamides, the first synthesis chemotherapeutic,

play an important role as effective antimicrobials inhibiting both gram-positive and

gram-negative bacteria, as well as some protozoa, such as coccidials (PRESCOTT,

2000; BARRAGRY, 1994). The fundamental act of sulfonamides was described by

Woods and Fildes in 1940, who discovered para - aminobenzoic acid (PABA) as a

metabolic substrate for folic acid (WOODS, 1940). Sulfonamides interfere with

multiplication of the bacterial cell by completely competitively binding the PABA

molecule and prevent the folic acid formation required for DNA synthesis. Therefore,

sulfonamides are usually bacteriostatics.

Generally, sulfonamides are weak organic acids, relatively insoluble in water, more

soluble in alkaline than acid pH, with their variability and extent bound to plasma

protein (15%-90%) having a wide range of pKa values from 2.65 to 10.4 (SPOO and

RIVIERE, 2001). Sulfonamides are relatively nontoxic when used in normal

therapeutic doses (BARRAGRY, 1994). Nevertheless, except for the sulfapyrimidine

group (sulfamethazine, sulfadiazine and sulfamerazine; KAHN, 2005), sulfonamides

are predisposed to precipitation in renal tubules due to low water solubility of

acetylated forms (RIVIERE et al., 1991; PRESCOTT, 2000). Some other toxic effects

have been documented e.g. allergies in humans (CHOQUET-KASTYLEVSKY et al.,

2002), keratoconjunctivitis sicca in dogs (COLLINS et al., 1986), and haemorrhagic

syndrome in chickens from sulfaquinoxaline administration (DAFT et al., 1989).

Sulfadimidine has been reported to exhibit carcinogenicity in mice (LITTLEFIELD et

al., 1989 and 1990), but a recent report showed that it would not occur at a normal or

low dose (LIONEL et al., 1999).

Page 15: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

15

In the veterinary field, sulfonamides were proposed for treatment and, in the past, as

growth promoters for some decades. In addition, the indications of sulfonamides for

many species of animals are well established (SPOO and RIVIERE, 2001; PLUMB,

2005). It is reportedly effective against a wide variety of infectious diseases and

commonly therapeutic especially for cattle and swine. In poultry, it has been used for

treating of cocidiosis, infectious coryza, pullorum disease and flow typhoid

(GIGUÈRE et al., 2006). However, owing to theirs low cost, ease of administration

and a wide range of application, sulfonamides have been used extensively; resulting

in a rapidly rise of bacteria-resistance, cross-resistance among sulfonamides and

residues appearing in animal products (EINSTEIN, 1994; AIELLO, 1998; JEMMI

and KÖNIG, 1999; PRESCOTT, 2000; SMITH et al., 2007).

2.2.2 Pharmacokinetic of sulfonamides

2.2.2.1 Absorption

Various routes of administration such as p.o. via drinking water and mixed feed, i.v.,

i.m., i.p., intrauterine and topical are generally used for sulfonamides, indicating that

they are extensively applied and well absorbed (KAHN, 2005). Most sulfonamides

are well absorbed by the gastrointestinal tract except some long-acting compounds

such as sulfamethoxypyridazine (RIVIERE et al., 1991; EINSTEIN, 1994).

Nonetheless, the other exceptions are some so-called enteric drugs e.g.

phthalylsulfathiazole, succinylsulfathiazole, sulfaquinoxaline and sulfaguanidine

(SPOO and RIVIERE, 2001; FREY et al., 1996). However, the absorption may be

delayed in pathological animals (BOTSOGLOU and FLETOURIS, 2001), in adult

ruminants and when administered with food to monogastric animals (PRESCOTT and

BAGGOT, 1993; PLUMB, 1991).

Bioavailability shows the percentage fraction of a drug that reaches systemic

circulation following non i.v. application. In chickens, oral bioavailability of

sulfadiazine has been reported at approximately 100 % (LÖSCHER et al., 1990) and

80%, respectively (BAERT et al., 2003). In a study on the bioavailability of

sulfadimidine administered to yearling cattle in different oral dosage forms, it was

Page 16: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

16

shown that 81% of the dose is available systemically from an oral solution, 63% from

a rapid-release bolus and 32% from a slow-release bolus (BEVILL et. al., 1977). The

estimation of bioavailability of sulfadimidine was based on the cumulative urinary

excretion of the drug (BAGGOT, 2001). Varying times required to reach maximum

plasma concentration in broiler for other sulfonamides have been recorded; for

sulfaquinoxaline of 5.5 h (EL-SAYED et al., 1995), for sulfadiazine of 1.6 h to 2.7 h

(LÖSCHER et al., 1990; BEART et al., 2003), for sulfadimidine approximately 2 h

(KIETZMANN, 1980; JAIN and PUNIA, 2001).

2.2.2.2 Distribution

It has been reported that sulfonamides penetrate widely into tissue and fluid

throughout the body including CNS, synovial fluid (joint), urine, bile and milk (SPOO

and RIVIERE, 2001). However, distribution of individual sulfonamides depends on

many factors such as pKa value, ionisation state, the vascularity of the absorption site,

lipophilicity, plasma protein-binding properties and species (BOTSOGLOU and

FLETOURIS, 2001). In addition, total plasma protein concentration in domestic

mammals is similar to that in humans (range of 6.0-8.5 g/dL), but it is approximately

3.8-5.2 g/dL in galliform species due to the low albumin concentration (BAGGOT,

2001). The different degree of plasma-binding protein in ruminants ranging from less

than 20% sulfanilamide (NIELSON and RASMUSSEN, 1977) to 94%

sulfadimethoxine (VAN GOGH, 1980) has been reported. In chickes, the percentage

of plasma protein-binding has been recorded as being 22 % for sulfadimidine

(NOUWS et al., 1988) and 40-60% for sulfamerazine (ATEF et al., 1978; OSHIMA

et al., 1964). Moreover, it has been reported that plasma protein-binding of the

metabolites of sulfamonomethoxine and sulfadimethoxine was higher than that of the

parents in laying hens, possibly affecting their excretion rates (VREE et al., 1987).

The volume of distribution (Vd) of SDD following single oral administration at 100

mg/kg in broilers was 0.55 L/kg (JAIN and PUNIA, 2001). The Vd of sulfadiazine in

chickens has been recorded as being 0.43 L/kg after i.v. administration of 33.34

mg/kg BW (BAERT et al., 2003) and 0.96 L/kg after i.v. injection of 100 mg/kg BW

(LÖSCHER et al., 1990).

Page 17: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

17

Following feeding trials to laying hens, sulfamonomethoxine and sulfadimethoxine

were reported which quickly distribute through the body, reaching a constant level in

various tissues 8 hours after the application, the highest concentration being in the

kidney and the lowest occurring in adipose tissues (FURUSAWA and MUKAI,

1995). This is supported by a latter paper which revealed that sulfadiazine,

sulfadimidine, sulfadimethoxazole, sulfaquinoxaline and sulfamonomethoxine were

well distributed though tissues of laying hens (FURUSAWA and KISHIDA, 2002).

Sulfadimidine has been found to significantly distribute into milk and eggs, remaining

in milk up to 2 days (PAULSON et al, 1992) and for weeks in eggs (SEIB, 1991) after

withdrawal of the drugs. Moreover, transfer and distribution of certain veterinary

drugs including sulfonamides in laying hens have been interestingly studied

(FURUSAWA, 2001).

2.2.2.3 Biotransformation

Sulfonamides are usually easily and extensively metabolised (KAHN, 2005). They are

primarily metabolised in the liver, but they are also biotransformed in other tissues

e.g. kidney, lung, brain, adrenal, blood, neuron, skin and gastrointestinal tract,

BOTSOGLOU and FLETOURIS, 2001). Acetylation, at the N4-position, is the major

process of most sulfonamides, the other metabolism including aromatic

hydroxylation, glucoronidation, O-dealkylation, deamination and sulfatation (VREE

et al., 1985).

Metabolisation is influenced by several factors e.g. species, age, pathological effect

and the specific sulfonamides administrated. Canine species are considered to lack the

ability to acetylate due to a high rate of deacetelation (VREE et al., 1983; TRETTIEN

et al., 1994; CAMPBELL, 1999), while swine are unable to hydroxylate this drug

(SPOO and RIVIERE, 2001; BOTSOGLOU and FLETOURIS, 2001). The

acetylation pathway of sulfadimidine is predominant in swine due to the 6-

hydroxymethylsulfadimidine metabolite in plasma, edible tissues and urine not being

able to be detected (NOUWS et al, 1986). Some different metabolism processes have

been reported e.g. sulfadimethoxine is mainly glucoronidated at the N1-position in

human whereas it is acetylated at the N4-position in pigs (VREE et al., 1985). In

general, metabolites of sulfonamides are more water-soluble and rapidly eliminated

Page 18: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

18

than the parent drug (NOUWS et al., 1988). However, the desaminosulfadimidine

half-life of elimination can vary from 1-9 days, while sulfadimidine and other

metabolites have a shorter half-life of 10-20 hours in pigs (MITCHELL and

PAULSON, 1986). NOUWS et al., (1991) have observed that the N4-acetyl

metabolite of sulfamethoxazole in plasma was 24.5% for cows and 100% for newborn

calves.

In poultry, approximately 6-20 % of most sulfonamides are metabolised by

acetylation (KIETZMANN, 1980, VREE and HEKSTER, 1985; NOUWS et al.,

1988). The low yield of plasma N4-acetylsulfonamide may be due to a high rate of

deacetylation in chickens (OIKAWA et. al., 1977), a high faecal excretion rate or to a

preferred metabolic pathway by hydroxylation or deamination (JAIN, 1994).

However, FURUSAWA (2000) found that the main pathway of in vitro hepatic

biotransformation of sulfamonomethoxine was hydroxylation (2, 6-diOH-SMM) in

laying hens. N4-acetyl metabolites of sulfonamides have no antimicrobial activity and

most are less water-soluble (except for sulfadiazine, sulfamerazine and

sulfadimidine).

2.2.2.4 Excretion

Kidneys play the primarily route of elimination for most sulfonamides either as parent

substance or metabolite; by glomerular filtration (uncharged), active carrier-mediated

proximal tubular secretion and passive reabsorption of nonionised drugs from the

distal part of the renal tubules (PRESCOTT, 1993 and 2000; PLUMB, 1991 and

2005). Other less significant routes of excretion are via milk, bile, faeces, tears and

sweat (SPOO and RIVERE, 2001). In the case of sulfadimidine it has been reported

that 11-37% and 24.5% of the dose was excreted uncharged into the urine in cattle

and swine, respectively (NOUWS et al., 1988; BEVILL et al., 1977; DUFFEE et al.,

1984).

It has been indicated that sulfadimidine might be extensively metabolised in turkeys

where only 8.6% (of oral dose) and about 17% (of i.v. dose) of the parent compound

was found in urine and faeces (HEATH et al., 1975). In addition, it has been reported

Page 19: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

19

that sulfadimethoxine was renally excreted at approximately 17.9% and 58.4% in the

form of a parent drug and its metabolites, respectively, in cattle (BOURNE et al.,

1981). In swine, approximately 48% of the dose was excreted into urine as uncharged

sulfathiazole and about 19% of the dose was excreted into urine as acetylsulfathiazole

(BOURNE et al., 1978).

The elimination half-life of sulfonamides varies according to the species and

individual drug; for example, sulfadimethoxine has an approximate half-life of 12.5 h

in cattle, 8.6 h in goats, 11.3 h in horses, 15.5 h in swine, 13.2 h in dogs and 10.2 h in

cats (GIGUÈRE et al., 2006), whereas sulfadimidine has an estimate half-life of 8-11

h in cattle, 3-8 h in goats, 10-13 h in horses, 9-16 h in swine, 4-17 h in dogs and 7-10

h in chickens (FREY and LÖSCHER, 2002). Varying withdrawal time of

sulfonamides from edible tissues of poultry have been reported; 2 days for

sulfadimethoxine from broiler tissues (NAGATA et al., 1994) and approximately 4-7

days for sulfadimidine, sulfanilamide and sulfaguanidine in fattening chicks

(VLAHOVIĆ et al., 1996).

Sulfadimidine has been documented as being generally more rapidly eliminated after

injection than compared to oral medication by mixing feed or drinking water

(BOTSOGLOU and FLETOURIS, 2001). Approval drugs treatment for coccidiosis in

chickens have been established. For example, the withdrawal time of sulfadimidine is

10 days after administration via drinking water of 0.1% for 2 days or 0.05% for 4

days, whereas it is 5 days after a dose of 0.05% for 6 days of sulfadimethoxine

(KAHN, 2005).

Page 20: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

20

2.3 Veterinary drug residues

At present, quality and safety of food from animal products are widely concerning

public and health agencies around the world. Since veterinary drugs have played an

important role in the field of animal husbandry and agro-industry for some decades,

increasing occurrence of residues and resistance have become interesting issues.

Residues of veterinary medicinal products, as defined by the European Union, are

"pharmacologically active substances (whether active principles, excipients or

degradation products) and their metabolites which remain in foodstuffs obtained from

animals to which the veterinary medicinal product in question has been administered",

(ANONYMOUS 1, 1990). In addition, the Center for Veterinary Medicine (CVM), an

agency under the Food and Drug organization (FDA) in the USA, has defined

residues as follows: “A residue means any compound present in edible tissues that

results from the use of a drug, and includes the drug, its metabolites, and any other

substance formed in or on food because of the drug's use”, (ANONYMOUS 2, 2004).

Recently, among positive samples, the EU reported that 51-52 % non-compliant

results of residues in animals were antibacterial (ANONYMOUS 3, 2004 and

ANONYMOUS 10, 2006). Accordingly, prudent usage of drugs should be strongly

considered (BOTSOGLOU and FLETOURIS, 2001).

2.3.1 Maximum Residue Limits (MRLs)

The European Commission sets Maximum Residue Limits (MRLs) after adoption by

the Standing Committee, following advice from the Committee for Veterinary

Medicinal Products (CVMP). MRL is the maximum concentration of a residue

following administration of a veterinary drug which is legally permitted or acceptable

in food. Determining MRL is a requirement of European legislation under a Council

Regulation (2377/90). Under this legislation, substances must be entered into one of

four annexes to the Regulation; Annex I: The data in the dossier are considered

adequate to establish a final MRL. Annex II: No MRLs required on the basis of the

data supplied. Annex III: established provisional MRLs but some clarification of

further studies must be supplied before final MRLs can be set. Annex IV: No MRLs

due to unacceptable risk to public health and prohibited for use in food producing

animals. In the EU, the MRL of sulfonamides for edible tissues is 100 µg/kg

Page 21: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

21

(ANONYMOUS 1, 1990). The MRLs values of sulfonamides established by some

health agencies are given in the table 1. However, MRLs values of sulfonamides have

not been set for eggs only. The withdrawal period is defined as the time taken for the

level of residues in the tissues (muscle, liver, kidney, skin/fat) or products (milk, eggs,

honey) to be depleted below the MRLs after the last dose has been given to the

animals. Regulatory requirements for withdrawal time vary among countries. The

selection of times listed in table 2 (KAHN, 2005) serves only as a general guideline.

Drug Target animal

species

Target tissue MRLs

µg/kg

Health

agency

All sulfonamides All animals

Bovine, Ovine,

Caprine

Muscle, fat, liver,

kidney

Milk

100

25

JECFA

JECFA

Sulfadimethoxine Turkey, Chicken,

Duck, Cattle

Not specified

Edible tissues

Milk

100

10

USDA

USDA

Sulfadiazine Swine, Poultry,

Fish

Muscle, liver, kidney,

milk (not egg)

100 EMEA

Sulfamerazine Trout Edible tissues 0 USDA

Sulfamethazine Cattle, Swine,

Chicken, Turkey

Swine

Edible tissues

Muscle, liver, kidney,

milk (not egg)

100

100

USDA

EMEA

Sulfaquinoxaline Cattle, Chicken,

Turkey

Poultry

Edible tissues

Muscle, liver, kidney,

milk (not egg)

100

100

USDA

EMEA

Sulfathiazole Swine Edible tissues 100 USDA

Table 1: Example of MRLs of sulfonamides produced by health agencies

Page 22: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

22

Drug Species Withdrawal time

(days)

Milk discard

(h)

Sulfadimidine Cattle

Swine

10a

14

96

-

Sulfabromethazine Cattle 10 96

Sulfadimethoxine Cattle 7 60

Sulfonamide combinationb Cattle 10 96

Trimethoprim/Sulfadiazine Cattle 3 7

Trimethoprim/Sulfadoxine Cattle 5 (p.o.)c -

Table 2: Drug withdrawal and milk discard times of sulfonamides (Modified from

KAHN, 2005)

a) 28 days for slow-release bolus

b) 8% sodium sulfadimidine: 8% sodium sulfapyridine: 8% sodium sulfathiazole

c) 28 days following parenteral administration

2.3.2 Possible explanation causes of veterinary drug residues in animals

Veterinary drug residues are one of the major problems for food contamination, if

natural forms of contamination (mycotoxins, microorganism or etc.) and organic

pollutant (dioxin etc.) are excluded. There are many factors influencing the build-up

of residues in animal products such as drug’s properties and their pharmacokinetic

characteristics, physicochemical or biological processes of animals and their products

(tissues, milk and eggs). Drug residues are usually accumulated in the liver or kidney

rather than other tissues (DOYLE, 2006). It has been noted that different residue

levels can be found in different tissue positions such as site and route of

administration (KORSRUD et al., 1994; KAARTINEN et al., 1999) or even between

breast and thigh of chickens (REYES-HERRERA et al., 2005).The most likely reason

for drug residues may result from human management such as improper usage

including extra-label or illegal drug applications (KENNEDY et al., 2000; FAJT,

2001; STOLKER et al., 2004; LUNESTAD and GRAVE, 2005; LI et al., 2006).

However, the most obvious reason for unacceptable residues might be due to failure

to keep to the withdrawal period including using overdose and long-acting drugs

Page 23: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

23

(SHEARER, 1999). Inadequate good sanitary care during animal or product

transportation (WHIPPLE et al., 1980) including the cross-contamination of animal

feeding stuffs with inadvertently applied drugs (BIEHL et al., 1981; McCAUGHEY

et al., 1990c), environmental and animal to animal transfer of drugs (McCAUGHEY

et al., 1990b; KIETZMANN et al., 1995; KENNEDY et al., 2000) may also cause

residues.

As mentioned earlier, a low-level drug contamination of animal feed is one large

causative channel of residues in animal products. Drug cross-contamination or drug

carry-over is a form of feed contamination that may result when the substance in

question is transferred (carried) from an acceptable to an unacceptable location or

feeding stuff. Drugs carry-over may come from dust and drug powder that have

electrostatic or moisture properties (HARNER et al., 1996). In addition, a recently

report showed various antibiotic agents including tylosin, tetracycline, sulfadimidine

and chloramphenicol were detected in dust on a swine farm (HAMSCHER et al.,

2003). Drug carry-over may also arise from medicated feed remaining in the mixers,

bins, conveyors, elevators and bulk trucks during or between batches of

manufacturing or delivering processes (HARNER et al., 1996). However, it seems

that approximately 5% carry-over between feeding batches is technically unavoidable

even with good manufacturing practice (BOTSOGLOU and FLETOURIS, 2001). The

potential contamination of some veterinary drugs in swine, broilers and layers feeds

have been documented (AERTS, 1990). The result of this study revealed that more

than 50% of the investigated feeds were generally contaminated with drugs at low

levels.

Studies carried out drug cross-contamination in feed have been published; low-level

contamination of sulfadimidine, ca. 1% of normal dosage, with detectable residues in

pig’s tissues (CROMWELL et al., 1980; McCAUGHEY et al., 1990a). Recently,

unintentionally contaminated polyether ionophores (coccidiostats) in animal feed,

resulting in detectable residues in eggs are more interesting in the EU

(ANONYMOUS 4, 2007). Carry-over of some anticoccidials e.g. diclazuril,

robenidine, halofuginone and nicarbazin in combination with narasin into eggs have

been studied (MORTIER et al., 2005). This demonstration showed that some drugs

presented at 5% in feed could cause residues in eggs. This value was also similar in

Page 24: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

24

the study of monensin carried out on feed mill (KENNEDY et al., 1998). In addition,

violative residues of nicarbazin in eggs and broiler liver have been reported following

ingestion of low-level contaminated feed (McEVOY et al., 2003). Furthermore,

finding of feed containing lasalocid at 0.1 mg/kg produced detectable residues at 6-8

µg/kg in egg when fed to hens have also been published (KENNEDY et al., 1996).

The transfer of some veterinary substances exposed at a very low-level into animal

products has been documented; McCRACKEN and KENNEDY (1997) showed a

metabolite of furazolidone (AOZ) residue in pig’s tissues after low-dose treatment

(0.5-2.3 mg/kg) for five days. Drug cross-contamination may create serious problems

among different species. The horse is a crucial example since it takes a very serious

problem of carry-over of monensin due to its LD50 at only 2-3 mg/kg (LANGTHON

et al., 1985). Thus, mortality of the horse can occur from an inadvertent low-level

contamination of monensin.

Drug low-level contamination generally may not generate a violation problem on

public health. However, extensive use of drugs may increase the risk of an adverse

effect of residues on the customer including the occurrence of antibiotic resistance.

Therefore, prudent use in the manner of preventing feed contamination is necessary.

2.3.3 Adverse effect of veterinary drug residues

Although the adverse effects of veterinary drug residues have been discussed, in this

thesis only four hazards will be considered according to DOYLE (2006). Firstly,

veterinary drug residues can cause allergic or toxic reactions in humans; penicillin is

one crucial example of this. The occurrences of penicillin-induced hypersensitivity

after milk or beef consumption have been cited (ORMEROD et al., 1987; REYES-

HERRERA et al., 2005). However, with regard to other antibiotics, macrolide and

cepharosporin residues are unlikely to be an allergenic hazard (DEWDNEY et al.,

1991). Although sulfonamides are potentially induced skin hypersensitivity reactions

(CHOQUET-KASTYLEVSKY et al., 2002), however, no allergic cases involving of

exposure to residues of sulfonamides in food have been reported (PAIGE et al., 1999).

Fortunately, exposure of antibiotics via oral channels usually sensitises to a lesser

extent than the parenteral route does (DEWDNEY et al., 1991).

Page 25: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

25

Secondly, chronic toxic effects occurring with prolonged exposure to a low level of

antibiotics also might generate side effects or tissue-degeneration. It has been noted

that most residues of veterinary drugs appear at a low level in animal products but

adverse effects are likely to occur due to acute rather than long-time exposure

(PAIGE et al., 1997). Although sulfadimidine reportedly has the potential to induce

thyroid teratogenic in mice (LITTLEFIELD et al., 1989 and 1990), it would not occur

if received at therapeutic level (LIONEL et al., 1999), thus implying that it might be

safe when there is low-dose exposure. Recently, however, there have been

cooperation studies within EU countries to evaluate risks of long-term or low-dose

exposure in foods (ANOYMOUS 5, 2004). So far, no report has been confirmed.

Thirdly, veterinary drug residues potentially develop antibiotic-resistant bacteria.

Since the initial widespread use of antibiotics over 60 years ago, situations have been

recognised in which an antibiotic has lost its effectiveness in controlling infection,

even when the dose is increased. These may increase the risk of treatment failures in

humans. LANZ et al. (2003) observed 581 cases of clinical Escherichia coli isolated

from different animal species in Switzerland between 1999 and 2001. They found that

the most frequent antimicrobial resistance was sulfonamides, tetracycline and

streptomycin. There are several mechanisms of bacteria resistance to sulfonamides

such as development of a form of dihydropteroate synthase, which is highly selective

for PABA and has less affinity to sulfonamides (MANDELL et al., 1996; ALLEN et

al., 1993). Moreover, an altering the recycling or utilising of dihydropteroic acid

within the cell, (CRAIG and WHITE, 1976), increasing synthesis of PABA

(GREENE, 1998) and destroying the sulfonamides (WHILCKE, 1988) have been

reported. Resistance to sulfonamides is usually persistent and irreversible.

Fortunately, this does not imply cross-resistance to other drug groups (CAMPBELL,

1999). The relationship of antibiotic-resistance between animals and humans has been

observed e.g., in the Netherlands the emergence of fluoroquinolones-resistant

Campylobacter species among poultry and humans has been reported (ENDTZ et al.,

1991). In Germany and the USA, increasing incidents of its fluoroquinolones-

resistance have also been found (HELMUTH and PROTZ 1997; SMITH et al., 1998).

Finally, there is disruption of normal human flora in the intestine. Antibiotic residues

may decrease the amount of commensal bacteria (normal flora) in humans, therefore

Page 26: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

26

creating opportunity for pathogenic bacteria to invade and cause illness. An increase

in E. coli resistance in faeces of mice given a low-dose of ampicillin and

chlortetracycline was observed. However, this failed to show the effect on humans

studied (CORPET, 1987). Recently, one study has reported that tetracycline-

resistance of commensal E. coli in swine is frequently linked to ampicillin- and

trimethoprim-sulfonamides- resistance (DEWULF et al., 2007). However, this was

controversial since several other reports stated that antibiotic residues in food of

animal origin would probably not disrupt the intestinal microflora in humans

(CERNIGLIA and KOTARSKI, 2005). The resistance gene of commensal flora in the

animal gut, which is a large reservoir, may be one probable route of transmission

leading to impacts on human health (DEWULF et al., 2007).

Page 27: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

27

2.4 Sulfonamide residues in animals

2.4.1 Sulfonamide residues in livestock

Sulfonamides have been classified in group B1 (veterinary medicines and

contaminants) of annex I of Directive 96/23/EC (1996), (ANONYMOUS 6, 1996).

The sulfonamide group is primarily approved for swine and cattle production. It has

potentially raised a residue problem in edible tissues. It has been found in animal

products for decades, as shown in the following reports. In Germany, sulfonamides

residues found during 1991-1992 ranged from 14-33,600 µg/kg (BERGNER et al.,

1993). The violation sulfonamides residues were found at 2.2-5.4% (1992-1995) in

Switzerland (ROLAND et al., 1996), 5.2-11.3% (1986-1990) in the UK (KAY and

PENNY, 1996), 0.2-1.3% (1990-1995) in Canada (NEIDERT and

SASCHENBRECKER, 1996) and 0.3-5.6% in the USA, respectively

(ANONYMOUS 7, 2003). The report of the EU in 2003 followed by the monitoring

program, SANCO, indicated that the sulfonamide group is one of the most commonly

occurring contaminating drug (ANONYMOUS 8, 2004). This conclusion agrees with

the recent reports by KENNEDY et al., (2000) and WANG et al., (2006). Fortunately,

a recent report by the EU showed a large decrease in sulfonamides detected in animal

products amounting to approximately 0.03% (106 samples out of 362,551 targeted

samples) of non-compliant results (ANONYMOUS 10, 2006). Additionally, most

sulfonamides found in animal products which included suspected samples were

sulfadiazine (47 samples), sulfadimidine (30 samples) and sulfadimethoxine (21

samples) (ANONYMOUS 10, 2006). Nonetheless, the remaining of unexpected

substances found in animal products proved that adequate residue prevention for the

consumer has not yet been achieved.

In swine production, an emergence of sulfonamides residues in pork and its products

has been worryingly observed (CROMWELL et al., 1980; ASHWORTH et al., 1986;

McCAUGHEY et al., 1990a, b, c). Sulfonamides have generated a huge problem

concerning their residues in animal products in the USA for decades (CORDEL,

1989). Recently, it has been reported that in the USA that sulfonamides contamination

still amounts to over 4% (DEY et al., 2003). Sulfadimidine is one classical example of

Page 28: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

28

drug residue in pork since it has been used at approximately 75% in hogs markets in

the USA (AUGSBURG, 1989) and frequently found in swine tissues (BEVILL et al.,

1986). Sulfadimidine needs nine times more for withdrawal time than sulfathiazole,

therefore deducing that it is prone to accumulate more than sulfathiazole as the data

showed, (BEVILL et al., 1986). The withdrawal time of sulfadimidine from swine

tissues have been reported starting at 7-8 days (GARWACKI et al., 1996; KORSRUD

et al., 1996), 14-15 days (WHIPPLE et al., 1980; AUGSBURG, 1989; PRIPWAI et

al., 2004) or even 30 days (PAPAPANAGIOTOU et al., 2005). Studies by

ASHWORTH et al. (1986) have shown that swine fed with sulfadimidine

concentrations greater than 8 mg/kg produced residues at a level greater than 100

µg/kg in muscle and 400 µg/kg in the liver, whereas, being fed up to 2 mg/kg could

be tolerated in withdrawal feeds before liver residues exceeded 100 µg/kg. However,

this study was contradicted those of the University of Kentucky and University of

Nebraska in 1981 (ANONYMOUS 9, 1981) which showed that a very little amount of

contamination of sulfamethazine (sulfadimidine) at 1 mg/kg in pig feed could

generate a high incidence of residues in pig’s liver. The contamination of

sulfadimidine in pig has been studied which transfers via cross-contamination

(McCAUGHEY et al. (1990c), and animal to animal (WHIPPLE et al., 1980;

McCAUGHEY et al., 1990b). The result revealed violative residues in an

unmedicated group, detected in the kidney and diaphragm, after exposure to the

contaminated housing for 6-24 h. In addition, they found the contaminating of

sulfadimidine in the faeces or urine in untreated pigs. This result was supported by

KIETZMANN et al., (1995). Furthermore, it was suggested that sulfadimidine was

preferentially retained in the mixer (McCAUGHEY et al., 1990a) and detected in

tissues of swine being found in feed at 2 g/tonne (BIEHL et al, 1981).

Sulfonamides residues in milk have also been observed. Sulfadimidine has created a

public health problem concerning its residue, this being highly detectable

approximately 40-70% in milk samples from supermarkets in the USA (PAIGE and

KENT, 1987; GRASSIE, 1988). A recent report showed that sulfadimidine was the

second most frequently drug presented in dairy products following Beta-lactams

(ANONYMOUS 7, 2003). Though, BLÜTHGEN et al., (2000) reported a carry-over

percentage of sulfadimidine from feed to milk of less than 1%. In contrast to the

studies on lactating diary cows by McEVOY et al., (1999) who indicated no violative

Page 29: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

29

contamination of sulfadimidine detected in milk samples after 7 days of withdrawal,

taken from the cows fed the concentrate containing 2 or 10 mg/kg. Unfortunately, the

peak level of sulfonamides in milk were found around the MRL value established by

the EU following the experiment that fed cows consist of sulfadiazine, sulfadimidine

or chlortetracycline at 250 mg/kg for 21 days, which is the normal dose used

(McEVOY et al., 1999 and 2000), In Japan, a survey of sulfadimidine,

sulfamethoxypyrimidine, sulfachloropyridazine, sulfathiazole and sulfamethoxazole

residues in milk were found below the detectable level (FURUSAWA and KISHIDA,

2005).

2.4.2 Sulfonamides residues in poultry

Poultry meat and eggs are very commonly consumed by humans. However, there may

be situations where contaminations of drug residues in poultry products occur.

Veterinary drugs and feed additives, especially anticoccidials and antibacterials e.g.

sulfonamides are drugs most commonly used on poultry farms. They can be easily

absorbed and distributed through the body of chickens, accumulated in various tissues

and transferred into their products (KAN and PETZ, 2000; WEISS et al, 2007).

Interestingly, sulfonamides were detected in 41.6% (5 out of 12 non-compliant

results) among antibacterials found in eggs (ANONYMOUS 10, 2006).

Residue of sulfadimethoxine has been observed in broiler tissues following dietary

administration at 25, 50 and 100 mg/kg (NAGATA, 1994). The residue was very

rapidly eliminated from tissues with less than 0.1 µg/g remaining within two days

after the drug withdrawal. This result is in the range of the study of TAKAHASHI

(1991), who reported that sulfadimethoxine was rapidly disappeared from plasma and

tissues (except skin) on the 3rd-5th day post dosed via drinking water. Compared to

sulfadimethoxine residue in laying hens, it has been found that it quickly distributed

through the body and reached a constant level in various tissues at 8 hours after feed

(FURUSAWA and MUKAI, 1995). However, it may appear below 0.1 µg/g in yolk

and albumen up to 7 and 3 days after the drug withdrawal, respectively (NAGATA,

1992). The comparison experiment of depletion of 3 sulfonamides; sulfadimidine,

sulfanilamide and sulfaguanidine, from muscle and liver after six days continuous

feeding in fattening chicks has been demonstrated (VLAHOVIĆ et al., 1996). The

Page 30: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

30

result showed that sulfaguanidine was the fastest eliminated, 4 days for all tissues,

whereas sulfanilamide and sulfadimidine needed 3 days longer to sink to below 0.1

µg/g in the liver. In turkeies, sulfadimidine given by drinking water was also

observed. It was found that concentrations fluctuated between 0.1-0.4 ppm in the

kidney, liver, and skin after 72 hours of withdrawal and as long as 14 days (HEATH

et al., 1975). Feed containing sulfadimethoxine (60 mg/kg) and sulfaquinoxaline (100

mg/kg) in turkeies was also studied by EPSTEIN and ASHWORTH (1989); presented

sulfadimethoxine and sulfaquinoxaline needed 2-3 and 4-6 days respectively for liver

and muscle tissues to decrease to the level of 0.1 µg/g.

In daily egg production incidents of residues in this product occur easily if withdrawal

time is ignored. There might be some parent drugs, including theirs metabolites

remaining in eggs, resulting from improper or extra-label used as feed additives for

laying hens older than 16 weeks (SHAIKH et al., 1999). However, cross-

contamination of the medicated feed into the feed free-drugs may be the cause of

residues in eggs (SERRATOSA et al., 2006). The distribution of 5 sulfonamides;

sulfadimidine, sulfadiazine, sulfamethoxazole, sulfamonomethoxine and

sulfaquinoxaline, following administration via medicated feed at 100 mg/kg in laying

hens has been studied (FURUSAWA and KISHIDA, 2002). The result showed that

sulfaquinoxaline was the highest residue found in samples (plasma, muscle, liver,

ovary and oviduct) whereas sulfadimidine was the lowest, after seven days of the

treatment.

Sulfonamide residues occurring in egg compositions have previously been studied.

Therefore, variations of withdrawal times of sulfonamides for eggs have been

reported; 9 to 10 days for sulfaquinoxaline (FURUSAWA et al., 1998; CAVALIERE

et al., 2003; SHAIKH et al., 2004) and for a combination of sulfaquinoxaline:

sulfadimidine: sulfamerazine (ROMAVARY and SIMON, 1992), 7 days for

sulfadimethoxine (NAGATA et al., 1992) and at least 4 days of sulfadiazine given by

drinking water (ATTA and EL-ZEINI, 2001). Additionally, KAN and PETZ (2000)

have interestingly summarised the concentrations and ratio of sulfonamide transfer

into egg white and egg yolk, given in table 3. However, up-to-date data are added in

this table.

Page 31: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

31

Content (mg/kg) Ratio

Drug White

(W)

Yolk

(Y) W/Y

Exposure

method Reference

Sulfadimethoxine4.6 1.7 2.71

400 mg/kg in

feed, 5 days

Furusawa

et al., 1994

0.86 0.37 2.32

100 mg/kg in

feed, 14 days

Kan and Jacobs,

1998

0.15 0.05 3

25 mg/kg in

feed, 21 days

Nagata

et al., 1989

0.25 0.1 2.5

50 mg/kg in

feed, 21 days

Nagata

et al., 1989

0.5 0.2 2.5

100 mg/kg in

feed, 21 days

Nagata

et al., 1989

0.04 0.02 2

10 mg/kg in

feed, 14 days

Nagata

et al., 1992

35 9 3.89

5000 mg/L in

water, 5 days

Roudaut, 1993

Sulfadiazine 0.14 0.015 9.33

20 mg/kg in

feed, 14 days

Kan and Jacobs,

1998

0.015 <0.008 >1.8

1.3 mg/kg in

feed , 21 days

Tomassen

et al., 1996

0.04 <0.008 >5

3.8 mg/kg in

feed, 21 days

Tomassen

et al., 1996

0.10 0.022 4.55

8.1 mg/kg in

feed, 21 days

Tomassen

et al., 1996

0.22 0.15 1.47

0.2 g/L drinking

water, 5 days

Atta et al., 2001

0.32 0.18 1.78

0.4 g/L drinking

water, 5 days

Atta et al., 2001

Table 3: Sulfonamide residues in egg yolk and albumen (modified from KAN and

PETZ, 2000)

Page 32: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

32

Content (mg/kg) Ratio

Drug White

(W)

Yolk

(Y) W/Y

Exposure

method Reference

Sulfanilamide 35 43 0.81

1000 mg/L in

water, 8 days

Bloom, 1975

0.15 0.14 1.07

20 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

Sulfachlorpyrazine 0.52 0.17 3.05

50 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

Sulfadimidine 56 45 1.24

1000 mg/L in

water, 8 days

Blom, 1975

97 83 1.17

2000 mg/L in

water, 8 days

Blom, 1975

20 5 4

5x 100 mg/kg

BW, 5 days

Geertsma

et al., 1987

0.03 0.01 3

20 mg/kg in

feed,14 days

Kan and

Jacobs, 1998

70 30 2.33

1000 mg/L in

water, 5 days

Krieg, 1966

51 34 1.5

1000 mg/L in

water, 5 days

Rodaut, 1993

38 23 1.65

94 mg/kg in

feed, 5 days

Sieb, 1991

Table 3(cont): Sulfonamide residues in egg yolk and albumen (modified from KAN

and PETZ, 2000)

Page 33: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

33

Content (mg/kg) Ratio

Drug White

(W)

Yolk

(Y) W/Y

Exposure

method Reference

Sulfaquinoxaline 50 36 1.39

400 mg/L in

water, 8 days

Blom, 1975

2.3 2.0 1.15

200 mg/L in

feed, 7 days

Furusawa

et al., 1998

0.54 0.26 2.07

20 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

0.95 1.57 0.61

60 mg/kg in

feed, 14 days

Nose et al.,

1982

3.7 1.4 2.64

100 mg/kg in

feed, 7 days

Petz, 1993

80 20 4

350 mg/L in

water, 3 days

Rana et al.,

1993

3.4 2.9 1.17

500 mg/kg in

feed, 12 days

(intermittent)

Righter et al.,

1970

8 2 4

400 mg/L in

water, 3 days

Romvary and

Simon, 1992

8.3 2 4.15

6000 mg/L in

water

Sakono et al.,

1981

Sulfapyridine <0.1 <0.005 -

50 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

Sulfisoxazole 0.05 0.02 2.5

100 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

Table 3 (cont): Sulfonamide residues in egg yolk and albumen (modified from KAN

and PETZ, 2000)

Page 34: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

34

Table 3(cont): Sulfonamide residues in egg yolk and albumen (modified from KAN

and PETZ, 2000)

Content (mg/kg) Ratio

Drug White

(W)

Yolk

(Y) W/Y

Exposure

method Reference

Sulfaguanidine 0.32 0.64 0.5

100 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

Sulfamerazine 0.22 0.03 7.33

100 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

23 6 3.83

2000 mg/kg in

feed, 25 days

Onodera

et al., 1970

Sulfamethoxazole 20 1.8 11.11

2000 mg/kg in

feed, 5 days

Oikawa et al.,

1977

39 2.9 13.45

4000 mg/kg in

feed, 5 days

Oikawa et al.,

1977

0.42 0.12 3.5

50 mg/kg in

feed, 14 days

Kan and

Jacobs, 1998

Sulfamono-

methoxine 6.8 1.5 4.53

400 mg/kg in

feed, 5 days

Furusawa and

Mukai, 1995

0.20 0.04 5

25 mg/kg in

feed, 21 days

Nagata et al.,

1989

0.35 0.07 5

50 mg/kg in

feed, 21 days

Nagata et al.,

1989

1.0 0.20 5

100 mg/kg in

feed, 21 days

Nagata et al.,

1989

Page 35: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

35

2.5 Sulfadimidine

2.5.1 Properties of sulfadimidine

Sulfadimidine (SDD), known as sulfamethazine, is one of the most widely used

sulfonamides in animals. It has reportedly been effectively used against a wide variety

of infectious diseases, a common therapeutic drug, especially for cattle, swine and

poultry. It is approved for treating coccidiosis and has been used as a growth promoter

in swine and cattle. It is not approved for use in lactating dairy cows and laying hens

older than 16 weeks (BOTSOGLOU and FLETOURIS, 2001). General properties of

sulfadimidine are given in table 4 (SUKUL and SPITELLER, 2006).

Chemical structure

Nomenclature 2-(4-Aminobenzensulfonamido)-4,6-dimethylpyrimidine

Other names Sulfamethazine, Sulfadimethylpyrimidine, Sulfamethiazine,

Sulfadine, sulfodimesin, ect.

Formula C12H14N4O2S

Molecular weight 278.33

pKa value 7.45± 0.50 (most acid)

2.79± 0.24 (most basic)

Koc value (L/kg) 1 at pH 1, 61.2 at pH 4, 48.2 at pH 7, 1 at pH 10

Table 4: General properties of SDD (modified from SUKUL and SPITELLER, 2006)

2.5.2 Pharmacokinetics of sulfadimidine in laying hens

Pharmacokinetic characteristics of SDD in different species have been interestingly

summarised by SPOO and RIVIERE, (2001) but complete pharmacokinetic values of

SDD in poultry are limited. However, in this present study the focus on SDD in laying

hens will be reviewed. Sulfadimidine has been reported as being rapidly absorbed by

the digestive tract of the hen, after it being given 2 g/L of SDD via drinking water

Page 36: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

36

(GOREN et al., 1984).This is in agreement with ROUDAUT and GARNIER (2002)

who studied SDD at the same route and dose. Furthermore, GOREN et al. (1984)

showed no significant difference in the SDD concentration in the plasma between

broilers and layers after intravenous or crop inoculation. Following a single oral

dosage of SDD at 100 mg/kg in laying hens, GEERTSMA et al. (1987) found that its

peak plasma concentration reached the range of between 44-73 µg/mL within 2-3

hours. Therefore, rapid absorption is confirmed. Compared to SDD treated in broilers,

following oral application at 100 mg/kg, the absorption rate constant (Ka) at 1.29 h-1

with the Tmax of 2.07 h (JAIN and PUNIA, 2001), Ka at 0.28 h-1 with T max of 1.42

(recalculated) (REDDY et al., 1988; CRAIGMILL el al., 2006) and Tmax of 2.0 h

(KIETZMANN, 1980) have been reported. Thus, all studies indicated that SDD was

also quickly absorbed in broilers.

Disposition of SDD into tissues and eggs has been presented (BLOM, 1975;

GEERTSMA et al., 1987; SHAIKH and CHU, 2000; ROUDAUT and GARNIER,

2002). BLOM (1975) has interestedly reported that plasma protein binding of SDD

was about 24-27%. This is in agreement with later results documented by NOUWS et

al. (1988), who showed the percentage degree of plasma protein-binding at 22% when

SDD concentration in plasma was greater than 50 µg/mL. However, at the low plasma

concentration (less than 50 µg/mL) the degree of plasma protein-binding was

increased to 53%.

The metabolism pathway of SDD in laying hens is similar to that of other

sulfonamides. Acetylation and hydroxylation are the major processes but their

metabolites may not be higher than 20% (VREE and HEKSTER, 1985). However,

GEERTSMA et al. (1987) and NOUWS et al. (1988) described that there may have

been an additional unknown metabolic pathway in poultry, since 57% of the dose

remained unaccounted for in the faecal excretion. In addition, NOUWS et al. (1988)

have demonstrated that N4-acetyl-sulfadimidine followed by hydroxylmethyl-

sulfadimidine and 5-hydroxy-sulfadimine in plasma were found only at about 6.6-

7.2%, 1.7-4.5% and 0.91-2.0% of the dose administered at 100 mg/kg, respectively.

However, its acetylation occurred only at 12% in broilers (KIETZMANN, 1980).

Moreover, the finding of SDD metabolites being higher than metabolites of

sulfadiazine in broilers has been documented (LÖSCHER et al., 1990).

Page 37: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

37

Sulfadimidine is generally more rapidly eliminated after injection than by oral

application via feed or drinking water (BOTSOGLOU and FLETOURIS, 2001).

Some previous papers reported that SDD decreased more rapidly than

sulfaquinoxaline (LÜDERS et al., 1974) and sulfadiazine (LÖSCHER et al., 1990) in

broilers. FURUSAWA and KISHIDA (2002) have studied the comparison of the

distribution among sulfadimidine, sulfadiazine, sulfamethoxazole, sulfaquinoxaline

and sulfamonomethoxine, following administration via medicated feed at 100 mg/kg

in laying hens. The results revealed SDD as the lowest concentration found in plasma

and tissues of these drugs after stop treatment. Thus, SDD was the most rapidly

excreted of all those drugs considered. Regarding the studies by NOUWS et al. (1988)

and GEERTSMA et al. (1987), the findings of the renal clearance of the metabolites

of SDD was approximately 10 times greater than SDD as a parent drug, indicating

that its metabolites were excreted faster than those of the parent drug.

However, varying data of the biological half-life values of SDD in laying hens were

recorded; NOUWS et al. (1988) and GEERTSMA et al. (1987) reported the final half-

life of 2.0 h, 2.9 h and 6.4 h post single intravenous, single oral and multiple dosing

orally, respectively, whereas the elimination half-life of SDD at 4.7 h (GOREN et al.,

1984) and as long as 16 h (BLOM, 1975) has also been reported. Interestingly,

REDDY et al. (1988) have reported that the elimination half-life was 15.5 h.

Nevertheless, it was only 8.9 h when recalculated by the WinNonlin® program

(CRAIGMILL el al., 2006). However, these results were in a similar range as those

from previous studies on broilers; KIETZMANN (1980) reported that SDD reached

peak plasma level at 87.4 µg/mL within 4-8 h with a calculated half-life of 5.1 h after

a single oral treatment of 150 mg/kg. Furthermore, JAIN and PUNIA (2001) have

showed the profile of SDD following oral treatment at 100 mg/kg as achieving its

peak level (Cmax at 83 µg/mL) of 2.1 h and the elimination half-life of 6.1 h. In

general, a wide range of SDD residues remained in various edible tissues of poultry,

the fluctuated level in muscle, skin, kidney, fat and liver being 0.01-0.47 ppm on day

ten (RIGHTER et al., 1971.). In the liver and skin of turkey SDD residues were

detected at 0.1-0.4 ppm after 14 days post treatment (HEATH, 1975). Variation of

withdrawal times of SDD has also been reported, approximately three days for

broilers (LÜDERS et al., 1974.), about 5-6 days for muscle and 7 days for the liver of

Page 38: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

38

fattening chickens (VLAHOVIĆ et al., 1996) and four days for tissues of hens

(NOUWS et al., 1988) post administration.

2.5.3 Residues of sulfadimidine in laying hens

Sulfadimidine residues in tissues and eggs of chickens have been studied. RIGHTER

(1971) has studied the comparison of tissues residues of SDD between calves and

chickens after application of SDD at 0.4% in mixed feed and 0.1% in drinking water.

The results showed that the residue was lower than 0.1 ppm after 8 days for calves but

longer than 10 days for chickens. Moreover, the report stated that residue in hepatic,

renal and skin tissues of chickens given medicated feed was more slowly eliminated

than residue in tissues in those given drinking water. Sulfadimidine residue in eggs

following drug administration via drinking water was also documented elsewhere

(KRIEG et al., 1966; BLOM, 1975; ROUDUAT, 1993 and 2000).

KRIEG et al. (1966) and BLOM (1975) have observed SDD residue in eggs after

administration via drinking water at 1 g/L. The result revealed that SDD concentration

in albumen was 70 and 56 µg/mL, respectively, which was higher than that found in

plasma, 50 and 47 µg/mL, respectively. GEERTSMA and colleagues (1987) have

studied residue of SDD and its metabolites in eggs following a single and multiple

oral applications (gelatine capsule form) at 100 mg/kg. Similar ranges of maximum

concentration residues in whole egg were found at 20.1 and 37 µg/g for both

experiments, respectively. They also found the residue in eggs after treatment up to 8

days. This value is similarly reported by PENSABENE et al. (1998), who observed an

occurrence of SDD in eggs fed by a single dose capsule of 75 mg/kg. In addition, they

demonstrated that SDD deposition in the whole eggs quickly declined from 13.9-15.5

ppm, during the first two days, reaching 0.6 ppm on the third day after treatment. This

range of time was also supported by later studies; KAN and PETZ (2000), SHAIKH

and CHU (2000). SDD residue in eggs and tissues, given at 100 mg/kg by feed

medication was also studied (SIEB, 1991). This observation detected SDD in albumen

greater than 100 ppb even 15 days post administration and presented the residue

preferential accumulated in the kidney and liver. This is in agreement with

FURUSAWA and KISCHIDA (2002), who found a greater amount of SDD in the

liver than in other tissues (muscle, plasma, and ovary), a comparison study of the

Page 39: A sulfadimidine model to evaluate pharmacokinetics and residues at

Literature review

39

distribution of sulfadimidine, sulfadiazine, sulfamethoxazole, sulfamonomethoxine

and sulfaquinoxaline, administered via medicated feed at 100 mg/kg in laying hens.

Moreover, the ratio of sulfadimidine in the tissues concerning egg formation to that in

the plasma was found to be significantly greater than those drugs. Thus, it may be

deduced that SDD is prone to transfer into egg components more easily than those

drugs. In addition, SDD may excrete into egg ca. 1% of the dose after a single

application (GEERTSMA et al, 1987; ROUDUAT et al., 2001).

Distribution of total 14C residues of SDD in eggs and tissues, after single (121.4 mg of

[14C] SDD) or multiple (274.6 mg/day of [14C] SDD) oral doses has been studied

(SHAIKH and CHU, 2000). They reported a higher concentration of SDD in the

albumen than in the yolk on the first two days. Thereafter, it was the opposite case,

this remaining detectable for up to 9 days in yolk. No metabolites of SDD were

detectable in either albumen or yolk, indicating that the parent drug was the main

marker residue in eggs. This result was attributed to support the study of SHAIHK et

al., (1999) and GEERTSMA (1987), who concluded that known metabolites of SDD

residues in eggs could be ignored. ROUDAUT and GARNIER (2002) observed drug

residues of sulfadimethoxine (0.5 g/L) and sulfadimidine (1.0 and 2.0 g/L) in eggs

post administration via drinking water for 5 days. The results revealed that 0.9-1.4%

of the application dose was deposited in the eggs and sulfadimidine was more easily

transferred to the eggs than sulfadimethoxine (1.4% versus 1.0%). Moreover,

approximately 3 times the amount of residues than that in yolk was found in albumen.

Thus, the albumen was recommended as the component for monitoring the drug

residues in eggs. This is in contrast to a recent study which suggested that yolk is an

appropriate matrix for monitoring sulfaquinoxaline in eggs (SHAIKH et al., 2004). In

general, the depletion of SDD and its metabolites in eggs may take at least 7-8 days

following the reports below; after cessation of medication via drinking water (BLOM,

1975; ROUDAUT and GARNIER, 2002), or by oral administration in gelatine

capsule form (NOUWS et al., 1986; GEERTSMA et al., 1987; PENSABENE et al,

1998; SHAIKH and CHU, 2000). However, it has also been documented that its

residue was detected for up to 15 days (SIEB, 1991).

Page 40: A sulfadimidine model to evaluate pharmacokinetics and residues at

Aims and objectives

40

3 Aims and objectives

The present studies were undertaken to elucidate on pharmacokinetic characteristics

of the veterinary drug and its residues in egg components following application at

various doses. Sulfadimidine and laying hens were used as models. The specific aims

were as follows:

- To develop the analytic procedure of sulfadimidine in the incurred residue samples.

- To evaluate the pharmacokinetic profiles at various doses.

- To observe the drug residues in plasma and eggs components from high to low-level

via oral and medicated feed administration.

- To investigate cross-contamination (drug carry-over) of the drug, in particular,

exposure at low levels.

- To compare pharmacokinetic behaviours and residue levels in egg components

between healthy and hens with impaired kidney function.

Page 41: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

41

4 Materials and methods

4.1 Materials

4.1.1 Chemical agent

Acetic acid 85% Applichem, Darmstadt Germany

Acetonitrile (HPLC far UV) Labscan, Dublin Ireland

Cyclohexane (extra pure) Merck, Darmstadt Germany

Ethyl acetate (HPLC grade) Labscan, Dublin Ireland

Formalin Merck, Darmstadt Germany

Hydrochloric acid 37% Merck, Darmstadt Germany

Methanol (HPLC grade) Labscan, Dublin Ireland

n-hexane (HPCL grade) Labscan, Dublin Ireland

Phosphate hydrogen monohydrate Merck, Darmstadt Germany

Phosphoric acid 85% Merck, Darmstadt Germany

Potassium dichromate Merck, Darmstadt Germany

Sodium chloride (analytical grade) Merck, Darmstadt Germany

Sulfadimidine standard (≥ 99%) Sigma-Aldrich, Steinheim Germany

Sulfadoxine standard (analytical grade) Sigma-Aldrich, Steinheim Germany

Water (HPLC grade) Karl Hecht KG, WD 3V, Germany

4.1.2 Instrument and material

Agitator (micro tube, eppendorf) Heildolph® REAK top, Germany

Agitator horizontal (Tube) IKA® Vibrax VXR basic, Brazil

Analytical column LiChropher®100 RP-18, end capped,

125×4, 5 µm, Darmstadt Germany

Auto sampler Gilson, model 231, USA

Centrifuge tube 2 mL Eppendorf® safe-lock, Germany

Chromatographic software 32-Karat™ Software, version 3.0.,USA

Column oven Spark Holland B.V., The Netherlands

Page 42: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

42

Cooling centrifuge Centrifuge 5403, Eppendorf, Hamburg

Germany

Detector UV detector, Varian® 2050, USA

Evaporator Labor technik Barkey, Bielefeld

Germany

Glass pasture pipettes 150 mm Brand, Wertheim Germany

Guard cartridge LiChorCart® 100 RP-18e (5µm),

Darmstadt Germany

Heparin sodium Euro OTC, Kamen Germany

HPLC vial WICOM, Darmstadt Germany

Nitrogen gas Westfaland, Germany

pH meter WTW pH320, Weilheim Germany

Pipett tips 20 µl, 100 µl, 2 mL Greiner, Frickenhausen Germany

Polypropylene test tube 10 mL Sarstedt, Nümbrecht Germany

Solvent delivery system Beckman®, Programmable solvent

module 116, USA

Sonicator Bandelin sonorex®, Berlin Germany

4.1.3 Composition of buffer solution

Mobile phase 0.05 mol/L phosphate buffer pH 3

13.8 g NaH2PO4 · H2O

2000 mL Water (HPLC grade)

pH 3 adjust with Phosphoric acid

4.1.4 Animals

Six healthy laying hens each group (Lohmann Brown), 22-26 weeks of ages (1.95 ±

0.15 kg), were housed at controlled temperature (20 ± 2°C) and light (14 h/day) in

accordance with the animal welfare acts (33-42502-04/917, LAVES). Water and a

commercial standard laying hen mash (deuka Deutsche Tiernahrung, Germany) were

supplied ad libitum.

Page 43: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

43

4.2 Methods 4.2.1 Drug administration and sample collection

4.2.1.1 Sulfadimidine experiment I

Groups of six hens each were treated with SDD via single intravenous injection (i.v.),

a single oral bolus administration (p.o.) and medicated feed. Animals were fasted

(feed) overnight before treatment. For a group of single i.v. administration, SDD was

injected as a single bolus via the vena axillaris at 100 mg/kg body weight (BW). For a

single oral bolus application (5 groups), each group of hens was treated at 1, 3, 10, 30

and 100 mg/kg BW. For the three groups of medicated feed, SDD was mixed into

feed resulting in a dose of 3, 10 and 30 mg/kg BW and fed for seven consecutive

days.

Blood samples of 1.5 mL were taken from the brachial vein at predetermined

intervals, as shown in table 5 and 6. All blood was heparinised to prevent blood

clotting. This was centrifuged at 4100 g for 10 min to separate the plasma, which was

placed into a 2 mL centrifuge vial for drug extraction. Eggs were collected for 7 days,

starting at 20 h after the treatment. Egg yolk and egg albumen were separated, and

placed into 10 mL polypropylene test tubes. All samples were stored at –20°C

pending analysis.

Table 5: Experiment method of single bolus administration of SDD

Route of

administration

Dose

(mg/kg BW)

Time determined

for plasma sample

Egg sample

Intravenous 100

Oral 1

Oral 3

Oral 10

Oral 30

0.25 (only for i.v.),

0.5, 1, 2, 4, 6, 8,

12, 24, 32, 48, 60,

72 h.

Collected daily

after dosed when

hens had laid eggs.

Oral 100

Page 44: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

44

Route of

administration

Dose

(mg/kg BW)

Feed duration

(day)

Plasma sample Egg sample

Mixed feed 3 7

Mixed feed 10 7

Mixed feed 30 7

Collected daily Collected daily

after dosed if

eggs were laid.

Table 6: Experiment method of medicated feed of SDD

4.2.1.2 Sulfadimidine experiment II

Hens were divided into two groups, one for healthy hens (n=4) and another for

impaired kidney hens (n=6). Potassium dichromate (K2Cr2O7), a renal toxic

substance, was prepared as a solution in water at 200 mg/mL (EICHLER, 1965). To

induce kidney damage hens were subcutaneously injected with K2Cr2O7 at 15 mg/kg

BW and allowed a stage of kidney damage for 7 days. Thereafter, an i.v.

administration of SDD at 100 mg/kg BW was applied for both groups. Blood samples

were collected following experiment I (see 4.2.1.1). Eggs were collected for 7 days,

starting at 6 h after the treatment. Egg yolk and egg albumen were separated, and

placed into 10 mL polypropylene test tubes. All samples were stored at –20°C prior to

analysis. At the end of the experiment, birds were sacrificed and their renal tissues

were examined histologically to confirm pathological lesions. The protocol for

experiment II is presented in table 7.

Hen Route of

administration

Dose

(mg/kg BW.)

Time determined

for plasma sample

Egg sample

Healthy i.v. 100

Impaired

kidney

i.v. 100

0.5, 1, 2, 4, 6, 8,

12, 24, 32, 48, 60,

72 h.

Collected daily

after dosed

when hens

have laid eggs.

Table 7: Protocol for experiment II of SDD

Page 45: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

45

4.2.2 Analysis (HPLC)

4.2.2.1 Preparing standard solution

Sulfadimidine (SDD), standard solutions were prepared in distilled water at a

concentration of 0.001 mg/mL to 1 mg/mL. Sulfadoxine (SDX), as an internal

standard, was dissolved in methanol at concentrations of 50 µg/mL.

4.2.2.2 Extraction process

Plasma: A 10 µL aliquot of SDX solution (50µg/mL) was added to 490 µL of

plasma. Then, 100 µL of 0.1 N HCl and 1 mL of ethyl acetate were added. Thereafter,

the sample was mixed for 1 min using a vortex mixer and centrifuged at 23000 g for 5

min. 600 µL supernatant was taken into a centrifuge cup and evaporated under a

gentle N2 steam at 40°C. The residue was mixed for 1 min with 700 µL eluent and

centrifuged at 23000 g for 5 min. A 500 µL aliquot of the clearly upper phase was

collected into a HPLC vial for injection (100 µL) into the HPLC system.

Yolk: A 1 g accurately weighed sample of yolk was put into a 10 mL test tube. The

yolk sample was mixed with 2.9 mL distilled water and internal standard (10 µL of

100µg/mL) of SDX. A 500 µL of 0.1 N HCl and 2 ml of ethyl acetate were added

before mixing the sample for 2 min with a vortex mixer. The amount of excessive

NaCl (approximately 1.2 g) along with 100 µL of 1 N HCl and 500 µL of n-Hexane

were added into the colloid. Afterwards it was mixed for 2 min and centrifuged at

4100 g for 5 min: 1.5 mL supernatant was put into 10 ml test tube. The remainder was

re-extracted with 1.5 mL ethyl acetate, then mixed for 2 min and centrifuged at 4100

g for 5 min to collect the 1.5 mL upper phase. Thereafter, combined with the previous

one, the extract was evaporated to dryness with a N2-evaporator. The residue was

reconstituted with 700 µL of eluent. After mixing for 2 min, the solution was

transferred into a centrifuge cup and centrifuged at 23100 g for 5 min. 500 µL of the

supernatant was filled in the HPLC vial for analysis.

Albumen: A 1 g portion of albumen sample was weighed into test tube, and then 0.5

mL distilled water and10 µL of SDX (100 µg/mL) were added. The mixed sample

Page 46: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

46

was added with 0.5 mL of 0.1 N HCl, 0.9 mL of acetonitrile and 0.1 mL of n-Hexane,

respectively. After mixing for 2 min, excessive NaCl (approximately 1.2 g) and 2 mL

of ethyl acetate were added. The sample was mixed for 2 min with a vortex mixer and

centrifuged at 4100 for 5 min. 2 mL of the upper phase was evaporated under N2

steam at 40°C. The remnant was dissolved with 700 µL mobile phase, mixed for 1

min and centrifuged at 4100 g for 5 min, and then 500 µL of the clearly phase was put

into the HPLC vial for the HPLC injection at 100 µL.

4.2.2.3 HPLC conditions

A mobile phase consisting of 0.05 mol/L phosphate buffer (pH 3.0) and acetonitrile at

85:15, v/v and 89:11, v/v was used for plasma and egg analysis, respectively. A flow

rate of 1.5 mL/min and a wavelength of 268 nm were applied and the temperature of

the column oven was set to 40°C throughout.

The validation method is involves the checking of data or programs for correctness,

compliance with standards and conformance with the requirement specification. To fit

the intended purpose, the typical validation characteristics considered are selectivity,

linearity, limit of detection, limit of quantification, recovery, accuracy, and precision.

Selectivity: The ability of the bioanalytical method to measure and differentiate the

analyte in presence of components that may be expected to present regarded as

“selectivity”. The selectivity of the analytical method must be demonstrated by

providing data to show the absence of interference peaks with regard to degradation

products, synthetic impurities and the matrices.

Linearity: Linearity is defined of an analytical procedure as the ability (within a

given range) to obtain test results of variable data which are directly proportional to

the concentration in the sample. At least five different concentration levels should be

used. The linearity is expressed by the linear regression coefficient (R2). The

calibration curve is the relationship between experimental response value and known

concentration of the analyte. At least duplication of five different concentrations was

used for the curve. Table 8 provide the concentrations used for linear calibration curve

of sulfadimidine in plasma, yolk and albumen.

Page 47: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

47

Matrix Concentration

Plasma 25, 100, 500, 1000, 2000, 5000 (ng/mL)

Yolk 50, 200, 1000, 5000, 10000, 20000 (ng/g)

Albumen 50, 200, 1000, 5000, 10000, 20000 (ng/g)

Table 8: Matrix and concentration used for linearity curve of SDD

Limit of detection: The limit of detection (LOD) is the lowest concentration of an

analyte that the bioanalytical procedure can reliably differentiate from background

noise. However, the LOD may not necessarily quantities as an exact value. The LOD

was calculated from the confidence interval of the calibration curve according to DIN

32645.

Limit of quantification: The limit of quantification (LOQ) is the lowest

concentration of an analyte in sample that can be determined quantitatively with

suitable precision (less than 20% of SD) and accuracy (80-120%). The LOQ was

determined by preparing standard solutions at the estimated LOQ level (based on the

preliminary tests which provide precision and accuracy values).

Recovery: Recovery is the extraction efficiency of an analytical process, reported as

the yield of an analyst obtained after complete sample extraction and processing steps

of the method in comparison to the theoretical maximum defined by complete transfer

from the natural matrix into the solution actually measured. The recovery of the

analyte need not be 100%, but the extent of recovery of an analyte and or internal

standard should be consistent, precise and reproducible.

Recovery = [Cfrt – C unf / Cadd ] . 100

Cfrt = concentration determined in fortified sample

Cunf = concentration determined in unfortified sample

Cadd = concentration of fortification

Page 48: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

48

Accuracy: Accuracy of an analytical procedure expresses the closeness of agreement

between the expected and the measured mean value. Accuracy is determined by

replicate analysis of samples containing known amounts of the analyte. The

measurements of 5 samples per concentration were calculated. Table 9 is given

different spiking concentrations in plasma, yolk and albumen. The mean value should

be within 15% of the actual value except at the LOQ, where it should not deviate by

more than 20%.

Matrix Sample amount Concentration

Plasma 5 0.025, 0.10, 0.50 (µg/mL)

Yolk 5 0.10, 1.0 (µg/g)

Albumen 5 0.10, 1.0 (µg/g)

Table 9: Spiked SDD at different concentrations in plasma, yolk and albumen

Precision: Precision presents the reproducibility of repeated measurements at the

same homogeneous sample under the prescribed condition. Precision is usually

expressed in term of the relative standard deviation (RSD), which is useful in case of

the comparison of samples with different concentrations.

The precision may be called “intra-day” if operated within short interval of time or

within batch and “inter-day” when operated in different day or between batches. The

measurements of 6 replicate determinations per concentration per day which at least 3

different days were operated. The intra- and inter-day precision determined at each

concentration level should not exceed 15% except for the LOQ level, where it should

not exceed 20%. Table 10 provided matrix, sample amounts and concentration for

precision test of SDD. Intra-day precision, sw and inter-day precision, sb were

calculated as:

∑∑ ⋅

=Tagesserie

TagesserieTagesseriew f

sfs

)( 2

Tage

gesamtTagesserie

b fXX

s ∑ −=

2)(

Page 49: A sulfadimidine model to evaluate pharmacokinetics and residues at

Materials and methods

49

Matrix Sample amount/ day Concentration

Plasma 6 0.025, 0.050, 0.075, 0.10 (µg/mL)

Yolk 6 0.050, 0.125 (µg/g)

Albumen 6 0.050, 0.125 (µg/g)

Table 10: Matrix, sample amounts and concentrations for precision test of SDD

4.2.3 Pharmacokinetic parameters and statistical analyses

Pharmacokinetic parameters were calculated using a program WinNonLin®version

4.1, Pharsight® USA. A first-order one-compartment model was used throughout the

calculation. The area under the curve (AUC), half-life of absorption (K01_HL),

elimination half-life (K10_HL), total body clearance (CL), time of maximum plasma

concentration (Tmax) and maximum plasma concentration (Cmax) were calculated.

In the experiment II, comparison between health and impaired kidney hens, statistical

data were performed using a software package, Prism 4 for Windows version 4.01

(GraphPad Software, Inc, San Diego USA). The data were evaluated using Mann-

Whitney test (two-tailed). The significance value was presented at p ≤ 0.05.

Page 50: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

50

5 Results

5.1 Method of analysis

5.1.1 Selectivity

To determine the selectivity of the analysis of SDD in the matrices; plasma, albumen

and yolk, the control and fortified samples were examined. Figure 2, 3 and 4

illustrates the typical chromatograms, which showed no interference and clearly

differentiate between SDD spiked into the matrices at the LOQ level (black arrow)

and the internal standard (I.S.; SDX).

Figure 2: Typical chromatogram obtained from fortified plasma at 0.025 µg/mL (A)

and control plasma (B)

Page 51: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

51

Figure 3: Typical chromatogram obtained from fortified albumen at 0.05 µg/g (C) and

control albumen (D)

Figure 4: Typical chromatogram obtained from fortified yolk at 0.05 µg/g (E) and

control yolk (F)

Page 52: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

52

5.1.2 Linearity The linear calibration curve of SDD in plasma, within the range of 25- 5000 ng/mL,

was obtained by plotting the peak area against the amount. For yolk and albumen, the

linearity was determined within the range of 50- 20000 ng/g. The calculation of linear

regression showed R2= 0.9990 for plasma (Fig. 5), R2= 0.9994 for yolk (Fig. 6) and

R2= 0.9991 for albumen (Fig. 7).

y = 0.0014x + 0.0628R2 = 0.999

0

2

4

6

8

0 1000 2000 3000 4000 5000 6000Concentration (ng/mL)

Res

pons

e

Figure 5: The linear calibration curve of SDD in plasma

Page 53: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

53

y = 0.0008x + 0.0204R2 = 0.9994

0

5

10

15

20

0 5000 10000 15000 20000 25000Concentration (ng/g)

Res

pons

e

Figure 6: The linear calibration curve of SDD in yolk

y = 0.0009x - 0.0401R2 = 0.9991

0

5

10

15

20

0 5000 10000 15000 20000 25000Concentration (ng/g)

Res

pons

e

Figure 7: The linear calibration curve of SDD in albumen

Page 54: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

54

5.1.3 Limit of detection (LOD) and limit of quantification (LOQ) The method provided the LOD of SDD in plasma at 0.01 µg/mL and 0.02 µg/g for

both yolk and albumen. According to the results, the LOQ concentration of SDD at

0.025 µg/mL for plasma and 0.05 µg/g for both yolk and albumen were found,

respectively.

5.1.4 Recovery

Collated average recoveries of SDD from different spiking concentrations in plasma,

yolk and albumen are presented in table 11. The values of recoveries greater than 80

% were satisfied with relative standard deviation (RSD) between 2.22% and 6.66%

except at the LOQ level of plasma (10.20 %).

Matrix Spiked concentration

(µg/mL or µg/g) n

Recovery

(%)

Relative standard

deviation, RSD. (%)

0.025 5 94.57 10.20

0.1 5 86.81 2.22 Plasma

0.5 5 84.70 3.29

0.1 5 97.06 6.66 Yolk

1.0 5 88.74 3.54

0.1 5 90.05 5.09 Albumen

1.0 5 90.14 6.15

Table 11: Average recoveries of SDD at different spiking concentrations of plasma

(µg/mL), yolk (µg/g) and albumen (µg/g)

5.1.5 Precision

The precision of SDD in plasma, yolk and albumen are given in table 12. An

overview, the intra-day and inter-day repeatability are less than 15 % except at the

LOQ level (0.025 µg/mL) of plasma for intra-day (16.95%).

Page 55: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

55

Table 12: Precision data of SDD (intra-day and inter-day) by different spiking

concentrations of plasma (µg/mL), yolk (µg/g) and albumen (µg/g)

Matrix Concentration

(µg/mL or µg/g)

Intra-day

(%)

Inter-day

(%)

0.025 16.95 8.37

0.050 6.80 4.47

0.075 5.18 2.02 Plasma

0.100 4.46 1.77

0.050 6.82 7.60 Yolk

0.125 8.07 4.20

0.050 9.58 3.35 Albumen

0.125 5.42 1.66

Page 56: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

56

5.2 Plasma concentrations of sulfadimidine following various doses

administered intravenously and orally in laying hens

Average plasma concentrations (Mean ± SD) of SDD in laying hen at predetermined

interval times following a single intravenous bolus and various doses via oral

treatment are given in table 13. Following i.v. administration at 100 mg/kg BW, the

maximum concentration of SDD was 129.40 ± 19.69 µg/mL rapidly declining to 0.06

± 0.06 µg/mL at 48 h after injection. Thereafter it was undetectable.

For the groups treated orally, the maximum plasma level of SDD reached 72.35 ±

15.49 µg/mL after 4 h of oral application at 100 mg/kg BW, whereas the maximum

concentration of 23.33 ± 1.36 µg/mL was detected 2 h after oral treatment with 30

mg/kg BW. For all low doses of oral administration at 1, 3 and 10 mg/kg BW, the

maximum plasma levels achieved 0.5 h after application were 0.53 ± 0.23, 2.09 ± 1.06

and 5.29 ± 2.49 µg/mL, respectively (see table 13). Semi-logarithmic plots of plasma

concentration-time curve of SDD (Mean ± SD) following i.v. and p.o. treatment are

given in figures 8 and 9, respectively. As shown in the plots, SDD was rapidly

depleted from the plasma, which showed concentrations lower than 0.025 µg/mL

within 48 h after i.v. injection and within 6-32 h after p.o. administration.

Page 57: A sulfadimidine model to evaluate pharmacokinetics and residues at

Dose (mg/kg BW.) 100 (i.v.) 100 (p.o.) 30 (p.o.) 10 (p.o.) 3 (p.o.) 1 (p.o.)

Time (h)

0.25 129.40 ± 19.69 - - - - -

0.5 121.68 ± 16.25 30.78 ± 15.82 18.60 ±11.14 5.29 ± 2.49 2.09 ±1.06 0.53 ±0.23

1 113.16 ± 15.03 40.96 ± 8.79 20.92 ± 8.93 5.08 ± 1.71 1.65 ± 0.35 0.51 ± 0.10

2 101.18 ± 11.11 68.27 ± 15.91 23.33 ± 1.36 4.41 ± 1.15 0.78 ± 0.18 0.21 ± 0.02

4 83.93 ± 16.34 72.35 ± 15.49 18.32 ± 4.25 1.64 ± 0.50 0.21 ± 0.07 0.06 ± 0.02

6 64.36 ± 13.45 69.08 ± 23.77 10.76 ± 2.06 0.56 ± 0.27 0.07 ± 0.01 <0.025

8 52.89 ± 12.40 65.53 ± 19.55 5.54 ± 1.33 0.33 ± 0.13 <0.025 ND

12 29.98 ± 9.92 51.00 ± 18.70 1.27 ± 0.55 0.03 ± 0.04 ND ND

24 4.37 ± 2.01 23.08 ± 10.72 <0.025 ND ND ND

32 0.03 ± 0.09 2.67 ± 2.10 ND ND ND ND

48 0.06 ± 0.06 <0.025 ND ND ND ND

Table 13: Average SDD plasma concentration (µg/mL, Mean ± SD), (n=6)

Drug administered via i.v. and p.o. route at different doses (1, 3, 10, 30 and 100 mg/kg BW)

ND = not detected, < 0.025 = below the LOQ

57

Page 58: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

58

5.3 Pharmacokinetic characteristics of sulfadimidine in laying hens 5.3.1 Pharmacokinetic characteristics of sulfadimidine following a single

intravenous bolus administration in laying hens

Pharmacokinetics values following plasma concentrations in laying hens after a single

intravenous bolus injection at 100 mg/kg BW were calculated by first-order one

compartment model. Figure 8 depicts a semi-logarithmic plot of average plasma

concentration of six hens versus time. As presented in the plot, SDD was rapidly

depleted from the plasma, which lowered to less than 0.025 µg/mL within 48 h. Table

14 presents the calculation of the pharmacokinetics parameters of SDD after i.v.

application. The calculated maximum plasma concentration (Cmax) of 132.20 ± 16.95

µg/mL was found 15 minutes after the dose and could be detected over LOQ (0.025

µg/mL) up to 48 hours. The elimination half-life (K01_HL) of 5.87 ± 1.14 h and the

total body clearance (CL) of 94.15 ± 27.84 mL/h/kg were found.

0.010

0.100

1.000

10.000

100.000

1000.000

0 6 12 18 24 30 36 42 48

Time (h)

Plas

ma

conc

entra

tion

( µ

g/m

L)

Figure 8: Plasma concentration of sulfadimidine (Mean ± SD) versus time after a

single intravenous bolus injection in laying hens (n=6)

Page 59: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

59

Hen (number) Parameter Unit 1 2 3 4 5 6 Mean SD

AUC µg*h/mL 1285.54 674.95 1212.25 997.63 1254.40 1307.43 1122.03 245.81

K10_HL h 6.78 4.44 6.14 4.82 5.65 7.41 5.87 1.14

CL mL/h/kg 77.79 148.16 82.49 100.24 79.72 76.49 94.15 27.85

Cmax µg/mL 131.38 105.41 136.94 143.37 153.90 122.23 132.20 16.95

Vss mL/kg 761.15 948.70 730.22 679.77 649.77 818.14 767.58 105.44

MRT h 9.78 6.40 8.85 6.96 8.15 10.70 8.47 1.64

Table 14: Pharmacokinetic values (Mean ± SD) of sulfadimidine after intravenous

injection of 100 mg/kg BW in laying hens

AUC = area under the curve, K01_HL = the absorption half-life,

K10_HL = the elimination half-life, CL = the systemic clearance,

CL_F = the total body clearance (for extra vascular models),

Tmax = the time of peak concentration, Cmax = the maximum concentration,

Vss = the volume of distribution at steady state, K01 = the apparent first-order

absorption rate constant, K10 = the apparent first-order elimination rate constant

Page 60: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

60

5.3.2 Pharmacokinetic characteristics of sulfadimidine following a single oral

bolus administration at different concentrations in laying hens

Based on plasma concentrations following a single oral bolus administration of

various doses (1, 3, 10, 30 and 100 mg/kg BW.), pharmacokinetic profiles were

calculated which are presented in tables 15-18. However, an oral application group of

1 mg/kg BW was not calculated due to inadequate quality data for performing kenetic

values. Figure 9 shows semi-logarithmic plots of average plasma concentration versus

time with six hens in each group. SDD was very quickly eliminated from the plasma

within 6-32 h. The calculated pharmacokinetic parameter showed the maximum

plasma concentrations as being 2.42 ± 1.15, 6.20 ± 1.79, 25.32 ± 5.78 and 76.50 ±

19.85 µg/mL following oral application of 3, 10, 30 and 100 mg/kg BW, respectively.

However, a wide range of the AUC values between 4.18 ± 1.22 to 1379.30 ± 428.13

h*µg/mL was observed. SDD had rapid absorption behaviour as presented in the

results of the rate of absorption of half-life (K01_HL) are 0.21 ± 0.26 to 1.20 ± 0.44 h

after various doses administered orally. Moreover, the biological half-life (K10_HL)

of SDD range was between 0.89 ± 0.16 h to 9.10 ± 1.58 h.

0.001

0.010

0.100

1.000

10.000

100.000

1000.000

0 6 12 18 24 30 36 42 48

Time (h)

Plas

ma

conc

entra

tion

( µ

g/m

L)

p.o. 100 mg/kg

p.o. 30 mg/kg

p.o. 10 mg/kg

p.o. 3 mg/kg

p.o. 1 mg/kg

Figure 9: Plasma concentration of sulfadimidine (Mean ± SD) versus time after a

single oral bolus administration of different doses in laying hens

Page 61: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

61

Hen (number) Parameter Unit 1 2 3 4 5 6 Mean SD

AUC µg*h/mL 1533.92 1400.57 1210.57 1734.85 1777.25 618.43 1379.30 428.13

K01_HL h 0.90 0.70 1.28 1.42 1.93 10.99 1.20 0.44

K10_HL h 10.49 9.67 10.80 8.63 8.53 6.48 9.10 1.58

CL_F mL/h/kg 65.19 71.40 82.59 57.64 56.27 161.70 82.47 40.00

Tmax h 3.48 2.87 4.46 4.42 5.35 3.16 3.96 0.94

Cmax µg/mL 80.55 81.74 58.39 97.68 93.49 47.15 76.50 40.00

K01 1/h 0.77 0.99 0.54 0.49 0.36 0.70 0.64 0.22

K10 1/h 0.77 0.07 0.06 0.08 0.08 0.11 0.08 0.02

Table 15: Pharmacokinetic properties of SDD after a single oral bolus administration

at 100 mg/kg BW

Hen (number) Parameter Unit 1 2 3 4 5 6 Mean SD

AUC µg*h/mL 160.65 122.04 163.32 175.77 175.25 112.20 151.54 27.53

K01_HL h 0.89 0.47 0.18 0.07 0.62 1.82 0.67 0.63

K10_HL h 2.37 2.48 3.56 3.39 3.00 1.86 2.78 0.65

CL_F mL/h/kg 186.75 245.82 183.69 170.68 171.18 267.38 204.25 41.63

Tmax h 1.98 1.40 0.80 0.42 1.77 2.66 1.51 0.81

Cmax µg/mL 26.34 23.03 27.16 33.00 26.86 15.54 25.32 5.78

K01 1/h 0.80 1.46 3.93 9.44 1.12 0.38 2.86 3.46

K10 1/h 0.29 0.28 0.19 0.20 0.23 0.37 0.26 0.07

Table 16: Pharmacokinetic properties of SDD after a single oral bolus administration

at 30 mg/kg BW

Page 62: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

62

Hen (number) Parameter Unit 1 2 3 4 5 6 Mean SD

AUC µg*h/mL 17.30 25.49 17.10 17.82 19.80 13.20 18.45 4.06

K01_HL h 0.96 0.03 0.03 0.76 0.58 0.03 0.40 0.42

K10_HL h 0.97 2.42 1.49 0.79 0.60 2.27 1.42 0.78

CL_F mL/h/kg 577.96 392.31 584.83 561.17 504.99 757.66 563.15 119.24

Tmax h 1.39 0.20 0.17 1.10 0.85 0.21 0.65 0.53

Cmax µg/mL 4.58 6.89 7.35 5.96 8.60 3.78 6.20 1.79

K01 1/h 0.72 21.55 24.47 0.91 1.20 19.81 1.44 11.60

K10 1/h 0.72 0.29 0.46 0.91 1.16 0.31 0.64 0.35

Table 17: Pharmacokinetic properties of SDD after a single oral bolus administration

at 10 mg/kg BW

Hen (number) Parameter Unit 1 2 3 4 5 6 Mean SD

AUC µg*h/mL 3.50 4.68 5.16 5.26 4.38 2.07 4.18 1.22

K01_HL h 0.28 0.03 0.05 0.05 0.15 0.70 0.21 0.26

K10_HL h 0.94 1.15 0.91 0.76 0.87 0.70 0.89 0.16

CL_F mL/h/kg 858.03 640.84 581.65 567.58 684.27 1451.47 797.31 337.09

Tmax h 0.70 0.17 0.23 0.22 0.46 1.01 0.46 0.33

Cmax µg/mL 1.54 2.55 3.30 3.93 2.42 0.75 2.42 1.15

K01 1/h 2.48 21.69 13.52 12.89 4.53 0.99 9.35 8.03

K10 1/h 0.74 0.60 0.76 0.91 0.80 0.99 0.80 0.14

Table 18: Pharmacokinetic properties of SDD after a single oral bolus administration

at 3 mg/kg BW

To observe the relation of dose (mg/kg BW) and some pharmacokinetic parameters,

the Cmax, K10_HL, Tmax and CL_F values were plotted. Figures 10-13 elucidate that

after a single oral bolus administration of SDD at different doses, the Cmax, K10_HL

and Tmax increased while the CL_F decreased with increasing doses.

Page 63: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

63

0

20

40

60

80

100

120

0 20 40 60 80 100 120dose [mg/kg BW]

Cm

ax [ µ

g/m

L]

Figure 10: Cmax values versus dose after a single oral bolus administration of SDD at

different doses (Mean ± SD, n = 6)

0

2

4

6

8

10

12

0 20 40 60 80 100 120 dose [mg/kg BW]

K10

_HL

[h]

Figure 11: K10_HL values versus dose after a single oral bolus administration of SDD

at different doses (Mean ± SD, n = 6)

Page 64: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

64

0

1

2

3

4

5

6

0 20 40 60 80 100 120 dose [mg/kg BW]

Tm

ax [h

]

Figure 12: Tmax values versus dose after a single oral bolus administration of SDD at

different doses (Mean ± SD, n = 6)

0

200

400

600

800

1000

1200

0 20 40 60 80 100 120 dose [mg/kg BW]

CL

[ml/h

/kg]

Figure 13: CL values versus dose after a single oral bolus administration of SDD at

different doses (Mean ± SD, n = 6)

Page 65: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

65

5.4 Sulfadimidine residue in eggs after a single oral bolus

administration at various doses in laying hens

5.4.1 Sulfadimidine residue in yolk

Following various single oral dose treatments of SDD at 1, 3, 10, 30 and 100 mg/kg

BW, the residues in yolk are summarised in table 19. The residue of 14.32 ± 6.47

µg/g was found in yolk one day after application at 100 mg/kg BW. For sub-

therapeutic doses at 3, 10 and 30 mg/kg BW, the residues on the first day after the

dose treatment were 0.11 ± 0.05, 0.42 ± 0.08 and 3.08 ± 1.52 µg/g, respectively.

However, no drug residue in yolk above LOQ (0.05 µg/g) was detected after oral

treatment of 1 mg/kg BW.

The residue was detectable above LOQ up to 8 days after administration at 100 mg/kg

BW. In addition, it could be found up to 7, 3 and 1 day for the dose of 30, 10 and 3

mg/kg BW, respectively.

Day after Dose (mg/kg BW)

treatment 100 30 10 3 1

1 14.32 ± 6.47 3.08 ± 1.52 0.42 ± 0.08 0.11 ± 0.05 ND

2 6.62 ± 2.72 0.51 ± 0.01 0.07 ± 0.02 <0.05 ND

3 3.67 ± 1.17 0.44 ± 0.12 0.06 ± 0.02 ND ND

4 2.74 ± 0.69 0.32 ± 0.05 0.04 ± 0.01 ND ND

5 1.98 ± 0.56 0.20 ± 0.04 <0.05 ND ND

6 1.21 ± 0.59 0.13 ± 0.02 ND ND ND

7 0.43 ± 0.32 0.07 ± 0.03 ND ND ND

8 0.12 ± 0.14 <0.05 ND ND ND

9 <0.05 <0.05 ND ND ND

10 ND ND ND ND ND

Table 19: Sulfadimidine residue in yolk (µg/g, Mean ± SD) after a single oral

administration of different doses; ND = not detected, < 0.05 = below the LOQ

Page 66: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

66

5.4.2 Sulfadimidine residue in albumen

Table 20 shows SDD residue in albumen following a single oral bolus administration

at different concentrations in laying hens. Within the normal dose of 100 mg/kg BW

the residue of SDD was found at a high amount in albumen at 49.34 ± 7.03 µg/g, one

day after of the dose, and detectable above the LOQ for up to 6 days. In the case of

the low administered doses, the residue levels of 0.28 ± 0.12, 0.98 ± 0.24 and 6.25 ±

1.85 µg/g were detected on day one following oral application with 3, 10 and 30

mg/kg BW, respectively. Interestingly, the detection of SDD residue at 0.07 ± 0.02

µg/g in albumen was observed after a dose of 1 mg/kg BW.

Day after Dose (mg/kg BW)

treatment 100 30 10 3 1

1 49.34 ± 7.03 6.25 ± 1.85 0.98 ± 0.24 0.28 ± 0.12 0.07 ± 0.02

2 6.10 ± 0.43 0.16 ± 0.04 ND ND ND

3 0.83 ± 0.16 0.15 ± 0.14 ND ND ND

4 0.38 ± 0.12 ND ND ND ND

5 0.20 ± 0.17 ND ND ND ND

6 0.12 ± 0.03 ND ND ND ND

7 <0.05 ND ND ND ND

8 ND ND ND ND ND

Table 20: Sulfadimidine residue in albumen (µg/g, Mean ± SD) after a single oral

administration of different doses; ND = not detected, < 0.05 = below the LOQ

Page 67: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

67

5.4.3 Transfer rate of sulfadimidine into eggs after a single oral administration

In the presented residues the calculation of the drug transfer rate (TR) into the eggs

was equated. The sum of residues (Rtotal) in the albumen and yolk were calculated

using the measured concentration in the albumen and yolk (Calbumen, Cyolk) and the

mass of the albumen and yolk (malbumen, myolk) at day i as:

[1] iyolkiyolkialbumenialbumenitotal mcmcR ,,,,, ⋅+⋅=

For the mass of yolk and albumen, an average weight of six representative eggs was

used (13.84 g and 36.28 g, respectively). The transfer rate was calculated from the

sum of residues of all eggs measured up to 10 days after administration as:

[2] 100[%]

10

1,

⋅=∑=

=

dose

RTR

t

iitotal

Interestingly, the calculation reveals that the transfer rates of SDD into the eggs after

single oral doses are dose-dependent (table 21). Decreasing doses are related with a

diminishing transfer rate.

Dose Drug Residue Transfer-ratec

intakea in whole eggb

(mg kg-1 BW) (mg) (µg) (%)100 195 2497.16 1.2830 61.5 303.60 0.4910 19.1 43.17 0.233 5.8 11.68 0.201 1.8 2.54 0.14

Table 21: Transfer rate of SDD into eggs after single oral administration

a calculated from dose × average BW, (data not shown)

b calculated according to equation [1]

c calculated according to equation [2]

Page 68: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

68

5.5 Sulfadimidine residue in eggs after medication via mixed feed at

various doses for seven consecutive days in laying hens

5.5.1 Sulfadimidine residue in yolk

In table 22 SDD residue levels in yolk are presented after application via mixed feed.

The average residue levels following medicated feed administration of 10 and 30

mg/kg BW for 7 days were 0.14 ± 0.06 and 1.43 ± 0.57 µg/g, respectively. However,

no residue level was found in the group medicated with 3 mg/kg BW. Figure 14 and

15 elucidate residue levels of each hen per day. The residues in yolk following

medication with 10 and 30 mg/kg BW gradually rose after the first day of application.

However, as compared to the albumen (see table 23), all SDD residues were slowly

accumulated in the yolk.

Matrix Yolk (µg/g)

Dose (mg/kg.BW) 30 10 3

Day 1 0.26 ± 0.14 0.04 ± 0.01 ND

Day 2 1.23 ± 0.52 0.10 ± 0.03 ND

Day 3 1.50 ± 0.70 0.13 ± 0.03 ND

Day 4 2.04 ± 0.60 0.15 ± 0.03 ND

Day 5 1.80 ± 0.52 0.16 ± 0.02 ND

Day 6 1.55 ± 0.45 0.20 ± 0.02 ND

Day 7 1.64 ± 0.55 0.20 ± 0.02 ND

Mean ± SD 1.43 ± 0.57 0.14 ± 0.06 ND

Table 22: SDD residue level in the yolk (µg/g, Mean ± SD) following medication via

feed for seven consecutive days in laying hens; ND = not detected

Page 69: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

69

0

1

2

3

0 1 2 3 4 5 6 7

Time (day)

Con

cent

ratio

n ( µ

g/g)

Hen 1Hen 2Hen 3Hen 4Hen 5Hen 6

Figure 14: SDD residue level in the yolk, each hen following medicated via feed for 7

days of 30 mg/kg BW

0

0.1

0.2

0.3

0 1 2 3 4 5 6 7

Time (day)

Con

cent

ratio

n ( µ

g/g)

Hen 1Hen 2Hen 3Hen 4Hen 5Hen 6

Figure 15: SDD residue level in the yolk each hen following medicated via feed of 10

mg/kg BW for seven days

Page 70: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

70

5.5.2 Sulfadimidine residue in albumen

Table 23 presents SDD residue concentrations in albumen following medication via

feed. Figure 16 to 18 give the residue levels for each hen per day. High concentrations

of SDD residue in the albumen of 2.50 ± 1.10 µg/g were found after the first day of

application at 30 mg/kg BW via medicated feed. For the group administration of 3 and

10 mg/kg BW, the average residue levels of day one were 0.13 ± 0.03 and 0.42 ± 0.07

µg/g, respectively. Interestingly, the residue levels in the albumen were estimated as

being equal to those found in the plasma (see table 24), which confirm a rapid

accumulation of SDD residue in the albumen.

Matrix Albumen (µg/g)

Dose (mg/kg BW) 30 10 3

Day 1 2.50 ± 1.10 0.42 ± 0.07 0.13 ± 0.03

Day 2 5.40 ± 2.58 0.62 ± 0.15 0.15 ± 0.02

Day 3 4.86 ± 1.44 0.61 ± 0.15 0.15 ± 0.01

Day 4 4.99 ± 1.93 0.64 ± 0.05 0.14 ± 0.01

Day 5 3.77 ± 1.25 0.45 ± 0.06 0.13 ± 0.02

Day 6 2.84 ± 1.25 0.40 ± 0.12 0.14 ± 0.02

Day 7 2.97 ± 0.90 0.43 ± 0.14 0.13 ± 0.01

Mean ± SD 3.90 ± 1.18 0.51 ± 0.10 0.14 ± 0.01

Table 23: SDD residue level in the albumen (µg/g, Mean ± SD) following medication

via feed for seven consecutive days in laying hens

Page 71: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

71

0

2

4

6

8

10

0 1 2 3 4 5 6 7Time (day)

Con

cent

ratio

n ( µ

g/g)

Hen 1Hen 2Hen 3Hen 4Hen 5Hen 6

Figure 16: SDD residue levels in the albumen of each hen following medication via

feed for seven days of 30 mg/kg BW

0.0

0.2

0.4

0.6

0.8

1.0

0 1 2 3 4 5 6 7Time (day)

Con

cent

ratio

n ( µ

g/g)

Hen 1Hen 2Hen 3Hen 4Hen 5Hen 6

Figure 17: SDD residue levels in the albumen of each hen following medication via

feed for seven days of 10 mg/kg BW

Page 72: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

72

0.0

0.1

0.2

0.3

0 1 2 3 4 5 6 7

Time (day)

Con

cent

ratio

n ( µ

g/g)

Hen 1Hen 2Hen 3Hen 4Hen 5Hen 6

Figure 18: SDD residue levels in the albumen of each hen following medication via

feed for seven days of 3 mg/kg BW

Page 73: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

73

5.5.3 Sulfadimidine concentrations in plasma

The residue concentrations of SDD in the plasma following seven consecutive days of

medication via feed are given in table 24. The average residue concentrations of 0.09

± 0.01, 0.45 ± 0.09 and 3.38 ± 1.24 µg/g were found after medication via feed with 3,

10 and 30 mg/kg BW, respectively. All residue levels appeared approximately equal

in the albumen (see table 23).

Matrix Plasma (µg/mL)

Dose (mg/kg BW) 30 10 3

Day 1 4.87 ± 2.05 0.58 ± 0.17 0.11 ± 0.03

Day 2 3.33 ± 1.97 0.52 ± 0.11 0.09 ± 0.03

Day 3 4.70 ± 1.88 0.47 ± 0.07 0.09 ± 0.02

Day 4 3.99 ± 1.52 0.44 ± 0.05 0.09 ± 0.03

Day 5 2.36 ± 2.41 0.43 ± 0.07 0.10 ± 0.03

Day 6 2.91 ± 1.08 0.31 ± 0.10 0.07 ± 0.01

Day 7 1.48 ± 0.95 0.37 ± 0.12 0.09 ± 0.02

Mean ± SD 3.38 ± 1.24 0.45 ± 0.09 0.09 ± 0.01

Table 24: SDD residue in the plasma (µg/mL, Mean ± SD) following medication via

feed for seven consecutive days in laying hens

Page 74: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

74

5.6 Comparative pharmacokinetic behavior and residues of

sulfadimidine between hen with healthy and impaired kidneys

5.6.1 Comparison of pharmacokinetic behaviours of sulfadimidine between hen

with healthy and impaired kidneys

Kidney damage was induce in those hens with impaired kidneys by subcutaneously

injection of K2Cr2O7 at 15 mg/kg BW and the stage of kidney damage allowed for

seven days. Thereafter, SDD was injected via i.v. at 100 mg/kg BW in the same day

for both healthy and impaired kidney groups. Most birds in the group of impaired

kidney hens showed depression, lethargy and increased water intake while feed intake

was decreased. At the end of the experiment, the birds were sacrificed. Kidneys gross

lesion in the renal dysfunction group showed a swollen kidney. According to the

histopathological confirmation by the Institute of Pathology (TiHo, Hannover), all

hens in the impaired kidney group were found to have multifocal fibrosis, interstitial

nephritis, tubular epithelial cell degeneration and necrosis, depicting of the induced

kidney damage.

Figure 19 depicts the comparison of the plasma concentration in semi-logarithmic

plots among normal kidney hens and kidney-damaged hens. The graphs show some

hens in the impaired kidney group having longer elimination than the normal hens.

Based on those plasma concentrations, the kinetics parameters were calculated and

presented in tables 25 and 26. Similar values of certain pharmacokinetic parameters

between normal kidney hens and impaired kidney hens were Cmax of 131.15 ± 13.88

and 124.51 ± 30.12 µg/mL, K01_HL of 8.23 ± 1.32 and 12.37 ± 3.86 h, CL of 66.52 ±

16.60 and 53.16 ± 24.35 mL/h/kg, respectively. Statistical significance of healthy

hens as compared to the impaired kidney hens was determined using a Mann-

Whitney, two-tailed test and applied for all pharmacokinetic values. The results

revealed p > 0.05 for all pharmacokinetic parameters. Therefore, the kinetic

behaviours of SDD were not statistically significantly different in healthy hens as

compared to kidney damaged hens.

Page 75: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

75

0.001

0.01

0.1

1

10

100

1000

0 10 20 30 40 50 60 70

Time (h)

Con

cent

ratio

n ( µ

g/m

L)

Healthy hens

Impaired kidney hens

Figure 19: Plasma concentration of sulfadimidine (Mean ± SD) versus times after a

single i.v. bolus administration at 100 mg/kg BW in laying hens

Hen (number)

Parameter Unit 1 2 3 4 Mean SD

AUC µg*h/mL 1846.40 1124.80 1863.07 1442.26 1569.14 354.41

K10_HL h 8.83 6.71 8.62 7.77 8.23 1.32

Cmax µg/mL 130.12 116.12 149.71 128.62 131.15 13.88

CL_F mL/h/kg 54.19 88.90 53.67 69.33 66.52 16.60

MRT h 14.18 9.68 12.44 11.21 11.88 1.91

Vss mL/kg 768.51 861.12 667.90 777.44 768.75 79.11

Table 25: Pharmacokinetic parameters of healthy hens

Page 76: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

76

Hen (number)

Parameter Unit 1 2 3 4 5 6 Mean SD

AUC µg*h/mL 1455 1116 1716 3884 2090 3467 2288 1128

K10_HL h 13.24 7.30 9.34 16.58 10.98 16.77 12.37 3.86

Cmax µg/mL 76.13 105.95 127.35 162.37 131.93 143.31 124.51 30.12

CL_F mL/h/kg 68.72 89.55 58.26 25.74 47.83 28.83 53.16 24.35

MRT h 19.11 10.53 13.47 23.92 15.84 24.19 17.84 5.58

Vss mL/kg 1313.44 943.79 785.23 615.87 757.96 697.77 852.34 250.68

Table 26: Pharmacokinetic parameters of impaired kidney hens

Page 77: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

77

5.6.2 Comparison of sulfadimidine residue in eggs between healthy and impaired

kidney hens

The residue levels of SDD in yolk following a single i.v. bolus treatment at 100 mg/kg

BW for normal kidney hens are given in table 27. Unfortunately, the hens had laid

only a few eggs. The residue in the yolk of two hens was 44.03 and 35.16 µg/g after

the first day of an application. The concentration of 0.53 µg/g was found in one hen

on day nine. The table 28 presents SDD levels in the yolk of each impaired kidney

hen after i.v. dosed with 100 mg/kg BW. Lower residue levels were found on the first

day after application compared to those of normal group. However, SDD residue

could be detected up to the 8th day after injection.

Hen (Number)

Day 1 2 3 4 Mean SD

1 44.03 - - 35.16 35.59 6.27

2 13.73 - - - - -

3 10.19 - 8.08 7.71 8.66 1.33

4 - - 5.67 - - -

5 4.53 - 4.64 3.27 4.15 0.76

6 3.13 - 2.5 - 2.81 0.31

7 1.58 - 0.68 - 1.13 0.45

8 0.12 - - 1.63 0.87 0.75

9 - - - 0.53 - -

Table 27: SDD residue levels in yolk (µg/g) after i.v. administration

at 100 mg/kg BW in healthy hens (n=4)

Page 78: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

78

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 14.82 15.88 21.80 1.5 - 6.23 12.05 8.10

2 7.03 - - - - - - -

3 - 4.66 7.01 5.84 - - 5.84 1.17

4 3.86 4.59 5.51 12.98 - - 6.73 4.21

5 3.41 3.37 4.62 6.94 - - 4.58 1.67

6 1.16 1.16 2.90 3.49 - - 2.18 1.20

7 - 0.57 - 1.49 - - 1.03 0.65

8 - - 0.90 0.13 - - 0.51 0.54

Table 28: SDD residue levels in yolk (µg/g) after i.v. administration

at 100 mg/kg BW in impaired kidney hens (n=6)

Tables 29 and 30 indicate residue levels of SDD in the albumen following a single i.v

bolus treatment at 100 mg/kg BW for normal kidney and impaired kidney hens,

respectively. The residue in the albumen of two normal hens was 18.78 and 44.96

µg/g on the first day after application. The concentration of 0.09 µg/g was found on

day eight in one hen. For the impaired kidney group, the residue levels in the albumen

on the first day after the dose were 3.24 to 25.77 µg/g. Very rapid depletion of the

residue in the albumen on the second day after dose was observed for both groups.

Page 79: A sulfadimidine model to evaluate pharmacokinetics and residues at

Results

79

Hen (Number)

Day 1 2 3 4 Mean SD

1 18.78 - - 44.96 31.87 18.51

2 4.01 - - - - -

3 1.01 - 0.71 0.66 0.19 0.19

4 - - 0.66 - - -

5 0.19 - 0.18 0.14 0.17 0.026

6 0.20 - 0.09 - 0.055 0.050

7 0.04 - - - - -

8 - - - 0.09 - -

Table 29: SDD residue levels in albumen (µg/g) after i.v. administration

at 100 mg/kg BW in healthy hens (n=4)

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 25.77 9.12 18.94 3.24 - 10.16 13.45 8.88

2 7.03 - - - - - - -

3 - 0.77 0.35 5.47 - - 0.66 0.10

4 0.22 0.23 0.23 0.76 - - 0.36 0.26

5 0.09 0.14 0.14 - - - 0.12 0.03

6 - - 0.10 0.11 - - 0.105 0.007

7 - - - 0.07 - - - -

Table 30: SDD residue levels in albumen (µg/g) after i.v. administration

at 100 mg/kg BW in impaired kidney hens (n=6)

Page 80: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

80

6 Discussion Unintentionally cross-contamination of veterinary drugs into feeding stuffs may

generate a violative residue problem in animal products. Therefore, the studies in this

thesis were conducted to investigate the potential residue deposition, in particular that

derived from low levels of contaminated feed. The pharmacokinetic characteristics

and residue depletion were studied, using sulfadimidine (SDD) application by

different doses and routes in laying hens as a model. The drug was applied as a single

i.v. injection (100 mg/kg BW), as a single oral bolus administration (1, 3, 10, 30 and

100 mg/kg BW) and medicated in mixed feed for 7 consecutive days (3, 10 and 30

mg/kg BW). Plasma and egg components (egg yolk and egg albumen) were samples

of interest. A comparison between healthy and impaired kidneys from those animals

was also observed.

The following discussion is divided into four major parts. The first part covers on the

methods of analysis, the second part dealing with drug plasma concentrations at

various doses via i.v. and p.o application. Based on plasma concentration versus time,

the pharmacokinetic parameters were calculated. In the third part, following an oral

and medicated feed application at different doses, drug residues were analysed in the

matrices of interest. A focus on drug concentrations as to whether residue in eggs at

low levels can be created residue in eggs was investigated. Finally, the data profiles

comparing of pharmacokinetic properties and the residues between the normal and

kidney dysfunction hens were also evaluated.

6.1 Method of analysis

In the present study, high performance liquid chromatography with spectroscopic UV

detection (HPLC-UV) was employed. The HPLC is one of the most widely used

techniques for determining sulfonamide residues in plasma, milk, meat and poultry

(AGARWAL, 1992; WANG et al., 2006). In this experiment, the UV wavelength was

carried out at 268 nm, which was in the range (265-275 nm) of those in previous

research (PETZ, 1983; HORII et al., 1990; PENSABENE et al., 1998; SHAIKH et al.,

1999). The retention behaviour of sulfonamides was dependent on its ionisation and

Page 81: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

81

polarity of mobile phase. Therefore, the pH of mobile phase plays an important role in

the chromatographic separations (WANG et al., 2006). For this, the optimum pH of

the buffer of 3.0 consisting of 0.05 mol/L phosphate hydrogen monohydrate was used

throughout. Traditionally, extraction and sample clean-up process involving the

determination were laborious, time-consuming, using a large amount of both volume

of samples and hazardous organic solvent, in particular for biological matrices such as

egg components which are rich in protein and lipid (AGARWAL, 1992; AERTS et

al., 1995). Therefore, a new approach was developed which had only a few steps,

using less volume of organics solvent and needing a volume of samples of only 0.5

mL of plasma and 1 g of egg components for an extraction.

To meet the criteria of validation, this present method provided good specificity,

linearity, limit of detection, limit of quantification, accuracy, precision and recovery.

The calculation of linear regression for plasma (0.025-5 µg/mL), yolk and albumen

(0.05-20 µg/g) showed R2 = 0.9990, 0.9994 and 0.9991, respectively. The limit of

detection (LOD) was 0.01 µg/mL and 0.025 µg/g for plasma and egg components,

respectively. However, these values were within the range for sulfonamides presented

in previous papers (FURUSAWA, 1999b; FURUSAWA, 2003b; KISHIDA et al.,

2004). The methods found the limit of quantification (LOQ) at 0.025 µg/mL for

plasma and 0.05 µg/g for both yolk and albumen. It has been recorded that the LOQ

value of sulfonamides in the plasma and eggs ranges from 0.02-0.10 µg/mL or µg/g

(FURUSAWA, 2002; KISHIDA et al., 2004; KISHIDA, 2007).

Several previous studies showed a variation in recovery values of approximately 60–

99% (TSAI et al., 1995; SHAIKH et al., 1999; FURUSAWA, 2002; ROUDAUT and

GARNIER, 2002). In this study, there was a high yield recovery of 84-97% with RSD

values less than 10% for all samples except for plasma spiked at 0.025 µg/mL

(10.20%), indicating that the methods were extreamly accurate. Moreover, these

methods provided intra- and interday precision values less than 10% except at the

LOQ of plasma (16.9%).

Page 82: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

82

6.2 Pharmacokinetic behaviour of sulfadimidine following single

bolus via intravenous injection and various doses of single bolus via

oral administration in laying hens

Due to different methods, dosage, route of administration, drug forms and duration of

medication the comparison of kinetic profiles of the drug are prevented (ROUDAUT

and GARNIAR, 2002). Little information on sulfonamides kinetic profiles of different

concentrations in laying hens is available. Therefore, the present experiment was

studied.

The pharmacokinetic values in this experiment were best described mathematically by

a first-order one compartment model. As showed in the results, the maximum plasma

concentration (Cmax) of SDD following an i.v. application at 100 mg/kg reached

132.20 ± 16.95 µg/mL, whereas it was detected at 76.50 ± 19.85 µg/mL after 4 h of

p.o. application. The results were of a similar level to those in former studies on

chickens; the Cmax value of SDD after i.v. injection has been recorded at 130 µg/mL

(NOUWS et al., 1988) or after p.o. application at 98.63 µg/mL and 82.98 µg/mL,

respectively, (REDDY et al., 1988; JAIN and PUNIA, 2001). However, the smaller

Cmax value at 59 µg/mL following p.o. treatment has also been reported (GEERTSMA

et al., 1987). In agreement with other studies, the time need to reach a peak was 2-4 h

(NOUWS et al., 1988; JAIN and PUNIA, 2001). The results confirmed that SDD was

rapidly absorbed. Consequently, SDD concentrations were found to be at an equal

level after 4-6 h for both i.v. and p.o administration. Thereafter, drug level following

i.v. injection showed more rapid depletion than p.o. application.

Following i.v. and p.o. administration, the calculated AUC showed similar levels at

1122.03 ± 245.81 and 1379.30 ± 428.13 µg*h/mL, respectively. It is important to note

that the slightly higher value in the case of oral rather than i.v. application may vary

due to the physiological aspect of each hen since different hens were used in these

two experiments. Additionally, the large amount reabsorbed within the cloaca in most

avian species (NOUWS et al., 1988 and LIERZ, 2003) or the occurrence of an

entrohepatic recircular pattern (EHC) may account for the higher AUC value after

Page 83: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

83

oral rather than for i.v. application. The findings, nonetheless, revealed that SDD has

very good absorption properties following oral treatment.

The present experiment provided the volume distribution of SDD following i.v.

injection at 0.77 ± 0.10 L/kg. According to NIELSON and RASMUSSEN (1975), the

observed volume lesser than 1 L/kg indicated that the concentration of the drug in

tissues was similar or less than in the plasma. Thus, the apparent volume of

distribution in this study was less than 1, indicating that the extent of SDD

concentration distributed in the plasma or blood was more than in the chicken body.

As compared to the same route and dose of application, a lower amount of the Vss of

SDD following i.v. injection has been reported by BLOM, 1975 (0.61 L/kg) and

REDDY et al., 1988 (0.43 L/kg). Regarding this present result, it is assumed that

intra-vessel administration of SDD penetrates better in chickens than some other

species since lower Vss values (0.24-0.40 L/kg) were reported in cattle and sheep

(BEVILL et al., 1977; WITCAMP et al., 1992; ELSHEIKH et al., 1991). However,

similar levels of Vss (0.50-0.62 L/kg) have also been reported in pigs and dogs

(RIFFAT et al., 1982; DUFFEE et al., 1984; YUAN et al., 1997).

BOTSOGLOU and FLETOURIS (2001) reported that SDD is more quickly

eliminated after injection than oral application via feed or drinking water. This is in

accordance with the results established in this present study where the elimination

half-life (T1/2el) after i.v. injection (5.87 ± 1.14 h) was shorter than p.o. administration

(9.10 ± 1.58 h). Thus, that the level of SDD decreases more quickly after i.v. injection

than p.o. application can be confirmed. Correspondingly, these values correlated well

with CL values, since the finding CL value in oral treatment was 82.47 ± 40 mL/h/kg,

relatively lower than that in i.v. application (94.15 ± 27.85 mL/h/kg). In contrast to

other reports, this presented CL value of SDD in hens was lower than that recorded at

around 200 mL/h/kg (GOREN et al., 1984) but of a slightly larger volume than that

reported at around 60 mL/h/kg (NOUWS et al., 1988).

The current findings showed that SDD, after oral administration in chickens, has a

relatively longer T1/2el than those published to date (2.9-6.4 h) in hens (NOUWS et al.,

1988; GEERTSMA et al., 1987; JAIN and PUNIA, 2001) and in broilers

(KIETZMANN, 1980). Nevertheless, a wide range of the T1/2el of 1.4-16 h following

Page 84: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

84

i.v. injection has also been reported (LÖSCHER et al., 1990; GOREN et al., 1984;

REDDY et al., 1988; BLOM, 1975). Thus, different experimental conditions such as

pharmacokinetic model, animal physiologys, species, breed, age, drug formulation

and dosage may be an explanation for these varying data (EL-SAYED et al., 1995;

BOTSOGLOU and FLETOURIS, 2001; FURUSAWA, 2001; ROUDAUT and

GARNIER, 2002). Furthermore, the absorption rate of the drug following oral

treatment depends on the concentration and duration time remaining in the crop which

consequents affects on the T1/2el value (KIETZMANN, 1980).

Other sulfonamides profiles studied in poultry have also been reported. Sulfadiazine

applications via i.v. in broilers have been recorded; the T1/2el of 2.7 h with an apparent

volume of distribution at 0.96 L/kg (LÖSCHER et al., 1990) and another recent

report, the T1/2el at 3.2 h with the Vss value of 0.43 L/kg (BAERT et al., 2003). As

compared to this present data, SDD showed a longer time of elimination since the

T1/2el (5.9 h) was higher than those previously reported. This is also supported by an

article of REDDY et al. (1988) who presented the T1/2el of SDD at 3.9 h, whereas T1/2el

of suldiazine was 2.3 h. On the contrary, from those kinetic profiles, it could be

deduced that SDD applied in hens is faster excreted than sulfaquinoxaline treated in

broilers since sulfaquinoxaline treatment in broilers has been reported a T1/2el of 12.6

h, a Vss value of 0.44 L/kg and a CL value of 17 mL/h/kg (EL-SAYED et al., 1995).

When comparing the same dose (100 mg/kg) following oral administration both

results of this present study and of REDDY et al. (1988) documented that the T1/2el of

SDD was 9.1 h and 15.6 h, respectively. This was much higher than the T1/2el reported

at 6 h for sulfadiazine (LÖSCHER et al., 1990) and at 4.5 h (REDDY et al., 1988).

Thus, the finding of this experiment are inconsistent with those previously reported

which presented SDD as being eliminated faster than sulfadiazine in broilers

following an oral dose (LÖSCHER et al., 1990). Furthermore, from this it can be

deduced that SDD has a longer elimination time than sulfadiazine, not only after i.v.

injection but also after oral application. In addition, the T1/2el of SDD following oral

application at 100 mg/kg as established in this experiment was comparatively higher

than that of sulfadoxine, 6 h, (JAIN and PUNIA, 2000) but lower in comparison to

sulfamethoxypyridazine, 13.2 h, (BAJWA and SINGH, 1977; CRAIGMILL et., al

2006) and sulfadimethoxine, 11.9 h, (BAJWA and SINGH, 1977) or 15.1 h (REDDY

Page 85: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

85

et al., 1988). Nevertheless, in this present case no flip-flop phenomenon was shown

since the entire absorption rate constant values (K01) were higher than the elimination

rate constant values (K10), following an overnight fast before treatment.

According to SPOO and RIVIERE (2006), sulfonamides are classified as short-acting

when the drug level in blood remains above 50 µg/mL for less than 12 h, and as

intermediate-acting if drug levels are obtained between 12-24 h. Thus, SDD in this

present study may be considered as short-acting in the case of administration via i.v.,

and intermediate-acting following p.o. application at 100 mg/kg.

In the experiments of sub-therapeutic concentrations administered via oral route, the

kinetic profiles of the dose of 3, 10, 30 mg/kg were calculated except for the group

application at 1 mg/kg due to its insufficiency data. A comparison of pharmacokinetic

parameters of SDD at different concentrations following oral administration showed

dose-dependent behaviours of Cmax, Tmax and T1/2 el. The SDD characteristic has more

rapidly absorption even at a low dose of administration, proved by a decrease of the

absorption rate half-life (K01_HL) to 0.2 h of 3 mg/kg compared to 1.2 h of 100

mg/kg. For this finding, the Tmax values were reached more quickly from 3.96 h to

0.46 h after reducing the drug concentrations. Moreover, the plot in figure 10

indicating high correlation between Cmax and different doses demonstrates a dose-

dependent trend. Furthermore, decreasing doses were consorted with larger volumes

of total body clearances. Correspondingly, the elimination half-life was shorter at a

low dose (0.9 h of 3 mg/kg) rather than at a high concentration (9.1 h of 100 mg/kg).

The rate-limited absorption may explain the deviations from dose-independent total

clearance, which is expected when using first-order kinetics. However, the almost

equal clearance value following i.v. and p.o. application with the same dose (100

mg/kg) suggested that the excretion is rate-limited, with both processes resulting in an

over-proportional decrease of the AUC after decreasing doses of SDD. In addition,

the extent and the rate of metabolism which have an influence on the elimination of

sulfonamides (MENGELERS et al., 2001) and capacity-metabolism or extensive

reabsorption from the cloaca (NOUWS et al., 1988 and LIERZ, 2003) may also

partially support the present profiles.

Page 86: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

86

SDD was mainly excreted by the renal. It has been reported that its major metabolite

form is N4-actyl in laying hens (NOUWS et al., 1988; KIETZMANN, 1980), but this

form does not occur over 20% (VREE, 1985). The underlying physiological

mechanisms involved in this complex pattern of SDD absorption and excretion in

chickens remain to be clarified. The limited data gathered in this study, however, do

not allow me to make further interpretations or create more complex pharmacokinetic

models. Nevertheless, as first-order kinetics may not be readily assumed for SDD in

chickens, pharmacokinetic characteristics formerly established on therapeutic levels

could not be extrapolated on sub-therapeutic levels.

6.3 Sulfadimidine residues in egg components after a single bolus via

oral and medicated feed at different concentrations

This study focused on residues incurred in eggs from the treatment of SDD in hens,

specifically exposed to low levels. The experiment was divided into two parts; firstly,

six laying hens in each group were orally treated with a single bolus at 1, 3, 10, 30 or

100 mg/kg, and secondly, they received medicated feed at 3, 10 or 30 mg/kg for seven

successive days. The results of the first experiment demonstrated that the residues of

SDD could be quickly detected as early as the first day in both yolk and albumen,

except for yolk at the lowest dose (1 mg/kg BW). The results obviously showed

higher level residue concentration, in particular when received at the highest dose in

the albumen (49.34 µg/g) than yolk (14.32 µg/g). It should be noted that only on the

first two days post oral dose did the residue level remain around 2-4 times higher in

the albumen than in the yolk. Thereafter, the residue level in the albumen rapidly

declined, most obviously found in the group administered the highest dose (100

mg/kg BW). The residue level in the albumen of this group quickly fell from 49.34

µg/g to 6.10 µg/g on the following day, and remaining at a low concentration (< 1

µg/g) for 5 days before being undetectable. Compared to yolk, the residue was

detected at 14.32 µg/g on the first day, then dropping to 6.62 µg/g on the following

day. Thereafter, the residue level in yolk decreased slightly and could be detected

below 1 µg/g on day 7 after dose. These findings indicate that SDD is prone to

accumulate during the initial phase in the albumen, the reverse being true for the yolk.

This behaviour may occur during the drug exchange within egg components,

Page 87: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

87

especially during shell formation (KAN and PETZ, 2000; ROUDAUT and

GARNIER, 2002). The rapid depletion pattern of SDD in the albumen was supported

by the previous research (GEERTSMA et al., 1987; PENSABENE et al., 1998;

SHAIKH and CHU, 2000; FURUSAWA, 2001; ROUDAUT and GARNIER, 2002).

In the present study, SDD residue in yolk was detectable over a longer period after a

single oral administration of the therapeutic dose (up to 8th day, compared to 6th day in

albumen, whereas it was eliminated from the plasma after only 32 h). Consequently,

the SDD level in yolk exceeded that in the albumen after about 2 days, which is in

agreement with previous reports (GEERTSMA et al., 1987; SHAIKH and CHU

2000). Moreover, slower depletion behaviour of residue in yolk than in albumen could

be seen even in hens exposed to the drug at sub-therapeutic doses (30 and 10 mg/kg

BW). Therefore, the yolk should be used in general as an appropriate matrix for

monitoring SDD residue in eggs, as previously suggested for sulfaquinoxaline

(SHAIKH et al, 2004). Nevertheless, we found no residues in yolk after receiving the

drug at 1 mg/kg. This may suggest that using of the albumen as an alternative matrix

is the best choice in case of egg contamination of SDD at very low levels, i.e. 1-3 %

of the recommended dose.

In the second experiment the SDD residue rapidly appeared in egg components within

the first day after starting the feed medication treatment. This tallied with the previous

study by GEERTSMA et al. (1987), the present results showing that residue levels of

the SDD in albumen reflected the concentration in the plasma rather than in the yolk.

A gradual increase then a levelling off of the SDD levels in those matrices 2-3 days

after feeding start should be noted. KAN and PETZ (2000) described that residue

concentration of drugs in plasma and albumen generally becomes constant 2-3 days

after the start treatment, whereas 8-10 days are needed in case of the yolk.

Nevertheless, the present findings may partly support these results due to the constant

level of SDD in yolk being achieved in only 2 days with medicated feed with sub-

therapeutic concentration. An appropriate withdrawal time of 8 days for SDD is

suggested for laying hens following treatment of the recommended dose. This

duration is in agreement with the results of GEERTSMA et al. (1987). This contrast

with previous research which reported that the withdrawal time should be longer than

2 weeks since the residue in eggs was detected up to 15-20 days (SIEB, 1991;

Page 88: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

88

ROUDAUT and GARNIER, 2002) or longer than 10 days for chickens (RIGHTER et

al., 1971). However, my findings showed prolonged withdrawal time of SDD rather

than of other sulfonamides e.g. sulfdimethoxine (7 days; NAGATA et al., 1992;

FURUSAWA et al., 1994) or sulfadiazine (4 days; ATTA and EL-ZEINI, 2001),

nonetheless shorter than sulfaquinoxaline (9-10 days; FURUSAWA et al., 1998;

CAVALIERE et al., 2003; SHAIKH et al., 2004).

Unfortunately, this experiment did not determine the SDD metabolites in eggs.

However, it has been documented that the parent drug was the major substance

residue found in both albumen and yolk since trace level metabolites were detectable

(GEERTSMA et al., 1987; SHAIKH et al., 1999) or not found (SHAIKH and CHU,

2000). Furthermore, it has been shown that SDD rather than other sulfonamides was

found preferentially in the tissues involving egg formation e.g. oviduct (FURUSAWA

and KISCHIDA, 2002). This indicated that the drug might be easily accumulated in

eggs where drug deposition in eggs may occur during the plumping or calcification

phase (DONOHUE and HAIRSTON, 2000; ROUDAUT and GARNIER, 2002).

Following oral treatment, the transfer rates of SDD into eggs were calculated. This

was approximately 1 % for the normal dose. The transfer rates of SDD in the present

study commensurated with those reported by GEERTSMA et al., (1987);

FURUSAWA (2001) and ROUDAUT and GARNIER (2002). Comparison transfer

rates between SDD and certain antibiotics into eggs have been noted (FURUSAWA,

2001). Furusawa reported that SDD had the highest transference (1.54 %) of all

studied agents, i.e. sulfadimethoxine (0.65 %), sulfamonomethoxine (0.77 %),

amprolium (0.21 %) and tylosin (0.005 %). Nonetheless, as stated in the present

results, the transfer rate of SDD application at the low concentrations was less than

0.5 %. Thus, this information is reflected in the reduced risk incurred in the residue

level in eggs when hens are exposed to drug at low levels.

Drugs such as sulfonamides, which have adequate water solubility properties, were

mostly had residues at a higher level in the albumen than in the yolk (FURUSAWA,

2001). Similar observations were also reported in studies on some antibiotics, e.g.

oxolinic acid (ROUDAUT, 1989), enrofloxacin (GORLAR et al., 1997) and

olaquidox (KEUKENS et al., 1996). Recently, KAN (2003) studied the relation of

Page 89: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

89

physicochemical properties such as pKa value, lipid solubility or protein binding in

order to predict the distribution pattern of drugs into and between egg components.

However, none of those properties could clearly explain the drug penetration

behaviours. Thus, the patterns of drug deposited in eggs remain to be elucidated.

Cross-contamination awareness of veterinary drugs in feed and transference to animal

products has been described elsewhere. However, little information on drug cross-

contamination on layer feed and eggs is available (KENNEDY et al., 2000;

McEVOY, 2002). Furthermore, studies carried out on low levels in contaminated feed

inducing residue problems in animal products, specifically in eggs, are scarce. To our

knowledge, the above cited studies failed to observe residues occurring in eggs after

receiving various concentrations of drugs via oral or medicated feed. This is the first

study of its kind showing measurable residues of SDD in egg components even at the

intake at a very low concentration of 1% of the therapeutic dose. Moreover, the

pharmacokinetics findings of dose-dependence are a noticeable feature of interest

namely that residue occurring in eggs should be determined at the relevant

concentrations and should not be predicted from known high-level kinetics. These

data may be of benefit concerning risk assessment, reducing and controlling residues

occurring in animal products.

6.4 Comparison of pharmacokinetic characteristics and residues of

sulfadimidine between healthy and impaired kidney laying hens

following intravenous administration

Renal excretion is the principle process for eliminating of drugs that are

predominantly ionised at physiological pH and for compounds with limited solubility

in lipid (drug metabolites) (BAGGOT, 2001). There are many causes of kidney

disease in birds e.g. viral infection, bacterial infection, toxins, obstruction (gout) and

vitamin A deficiency. Avian renal tubular cells in particular are susceptible to injuries,

renal dysfunction and are frequently encountered (SCHMIDT, 2006). However,

pathognomonic signs are rare when renal disorder occurs (LIERZ, 2003). Unlike

other species, chickens have two complex types of nephorns, reptilian (cortical) and

mammalian (meddullary)-type (MORILD et al., 1985). Most birds excrete urate as a

Page 90: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

90

waste product, instead of toxic urea. This process requires a lesser amount of water. In

birds this is probably a method for conserving water by resorption within the coaca

(LIERZ, 2003). The manifestation of renal damage in birds is different from

mammals in that it is usually associated with deposition of gout crystals within the

lumen and cytoplasm of tubular cells due to the regular production of urate as an end-

product of nitrogen metabolism in avians. The deposition of urate is in turn harmful to

the tubular cells, causing deterioration of tubular lesions. However, avian renal

tubular cells and kidney function may be reversible from the impaired stage. The bird

is one species that has a relatively good chance of recovery as long as the disability

caused by injury is removed in time.

Generally, SDD is mainly excreted by the kidney (PRESCOTT, 2000). In this

experiment, similar values of the pharmacokinetic parameters between hens with

normal kidney functions and those with impaired kidney functions were found: Cmax

of 131.15 ± 13.88 and 124.51 ± 30.12 µg/mL, K01_HL of 8.23 ± 1.32 and 12.37 ±

3.86 h, CL of 66.52 ± 16.60 and 53.16 ± 24.35 µg/mL, respectively. However,

relatively slow elimination of SDD in kidney damaged hens than in healthy hens

could be observed. As stated in the results, the SDD level was detected over the LOQ

value for up to 60 h for the group of impaired kidney hens, whereas it was

undetectable at 48 h for healthy hens after drug application (see Fig. 21). A noticeable

feature of this experiment following Mann-Whitney (two-tailed) calculation,

nonetheless, showed no statistically significant (p > 0.05) of the SDD kinetic

behaviours between healthy and kidney damaged hens.

SDD, as a parent drug, has been observed in some species (humans, ruminants and

swine) mainly being excreted via glomerular filtration, whereas its metabolites were

predominantly eliminated by tubular secretion (VREE and HEKSTER, 1985;

NOUWS et al., 1986). It has been reported that the use of potassium dichromate as a

renal damage agent mainly induces tubular lesion in animals e.g. rabbits and dogs

(EICHLER, 1965). According to the results in this present study, the histo-pathology

of impaired kidney hens was also mainly tubular lesion. Thus, it might be deemed that

the SDD application in hens is preferably excreted by glomerular filtration rather than

by tubular secretion when similar SDD excretion behaviour in the healthy group as in

the impaired kidney hens is presented in the results. Additionally, NOUWS et al.

Page 91: A sulfadimidine model to evaluate pharmacokinetics and residues at

Discussion

91

(1988) reported that an additional metabolic pathway exists for sulfonamides in

chickens. Unfortunately, this present study did not consider SDD metabolites.

However, limited rates of absorption and excretion of SDD were found in experiment

I of this study. Thus, the capacity-limited absorption, metabolism and excretion of

SDD in hens may also be involved. Therefore, these reasons may partly explain the

insignificant characteristics occurring between both groups of hens in this present

experiment.

For SDD residue in eggs, unfortunately few eggs were laid. However, this correlated

with experiment I of this study which found that SDD residue highly accumulated in

both albumen and yolk on the first day of the dose. Additionally, it should be noted

that the high concentration of SDD residue was detected in the albumen (31.87 µg/g)

and yolk (35.59 µg/g) on the first day after treatment in the healthy hens group,

whereas it was found at a lower level in both albumen (13.45 µg/g) and yolk (12.05

µg/g) for the kidney dysfunction hens group. This results from the pathological effect

maybe also occurring in the tissues involving egg formation. It is noteworthy that the

maximum SDD residue in the yolk of hen number four in the impaired kidney group

was found later on the third day after drug application. Individual kinetic profiles of

this hen may partly account for its CL value being low, whereas AUC, T1/2el and Cmax

were greater than other hens. However, a similar withdrawal time of SDD to the

experiment I could be documented due to the SDD residue detectable up to 8th-9th day

post dose for both groups. Furthermore, no obviously different level of SDD residue

among those hens could be seen after a few days of treatment. Correspondingly, no

statistically significant difference (p > 0.05) in the SDD kinetic behaviours between

healthy and kidney damaged hens was found.

Page 92: A sulfadimidine model to evaluate pharmacokinetics and residues at

Summary

92

7 Summary

The studies presented in this thesis were undertaken to elucidate the potential residue

risk derived from low levels of contaminated feed. Sulfadimidine (SDD) was used as

a model to determine the residue risk at various doses and routes of application in

laying hens. To achieve the aims of the studies, groups of six hens each aged between

22-26 weeks old were used for two experiments. In experiment I, six groups of hens

received either a single intravenous (i.v.) bolus of 100 mg/kg BW or per oral (p.o.) at

1, 3, 10, 30 and 100 mg/kg BW. Three additional groups of hens were fed a feed

consisting of SDD with a resulting dose of 3, 10 and 30 mg/kg BW for seven

consecutive days. Experiment II dealt with the comparison of pharmacokinetic

behaviours and residue of SDD between healthy and impaired kidney hens after i.v.

injection at 100 mg/kg. Drug levels were determined with HPLC-UV for the matrices

of interest (plasma, yolk and albumen).

The methods of analysis provided good specificity, linearity, limit of detection, limit

of quantification, accuracy, precision and recovery. The calculations of linear

regression for all the matrices were R2 > 0.9990. The limit of detection (LOD) was

0.01 µg/g and 0.025 µg/mL and the limit of quantification (LOQ) was 0.025 µg/mL

and 0.05 µg/g for plasma and egg components, respectively. The intra- and interday

precision values were less than 10% except at the LOQ of the plasma (16.9%). The

pharmacokinetic parameters were calculated assuming first-order kinetics, except for

one group (1 mg/kg BW.) due to insufficient data quality. The results revealed that

SDD was rapidly absorbed after treatment at the therapeutic dose. Furthermore, at

sub-therapeutic doses, our data demonstrated that SDD was absorbed even more

rapidly with a decrease in the calculated absorption rate half-life value (K 01_HL) to

0.2 h at 3 mg/kg BW as compared to 1.2 h at 100 mg/kg BW. Correspondingly, the

Tmax value decreased from 4 h to 0.5 h. The elimination of SDD was quickly depleted

from the plasma. Within 48 h after a single bolus treatment via i.v. and 6-32 h after

p.o. administration at various doses, the concentrations were lower than the LOQ.

Following single oral doses, the Cmax, Tmax and K10_HL increased while the CL

decreased with increasing doses. Therefore, these characteristics might be deemed

that dose-dependent. Regarding the SDD residue in egg components, our data

Page 93: A sulfadimidine model to evaluate pharmacokinetics and residues at

Summary

93

revealed that, at the initial phase after a single oral administration and during

medication via feed the SDD concentration in the albumen was 2 to 4 times higher

than in the yolk. However, the SDD residue in yolk could be detected longer over

LOQ, up to the 8th day as compared to the 6th day in the albumen, whereas it was

eliminated from the plasma after only 32 h. For other lower concentrations, SDD

levels were detected approximately 2 days longer in the yolk than in the albumen. The

ingestion of sub-therapeutic doses of SDD at 1% to 3% of the recommended dose led

to measurable residue in eggs. The transfer rates of SDD deposited into egg

components were dose-dependent, which might be deemed to reduce the risk of

residue occurring in eggs. Results of the comparison between the kinetic profiles and

residues in eggs of SDD obtained from healthy and impaired kidney hens showed no

statistically significant difference in the kinetic behaviours. This may link to rate-

limited absorption and elimination. Thus, moderate kidney damaged in laying hen

may have no affect on kinetic behaviours of SDD.

In conclusion, our study demonstrated that low levels intake of SDD by ingesting of

contaminated feed may generate a residue problem in eggs. The data showed that an

application at only 1% of the therapeutic dose resulted in a measurable concentration

in eggs. These results suggest that residue risks should be examined at relevant

concentrations and may not be estimated from known high-level kinetics. Thus,

similar protocol approaches should be applied to other drug substances in order to

avoid a residue risk for the consumer.

Page 94: A sulfadimidine model to evaluate pharmacokinetics and residues at

Zusammenfassung

94

8 Zusammenfassung In der vorliegenden Studie wurde das potentielle Riskiko einer Rückstandsbildung

durch eine Arzneistoffkontamination des Futters mit niedrigen Dosen untersucht.

Dazu wurde in einem Modellversuch Sulfadimidin an 22 bis 26 Wochen alte

Legehennen verabreicht und die Rückstände im Ei nach verschiedenen Dosierungen

gemessen. Darüberhinaus wurde geprüft, ob die Induktion eines Nierenschadens und

damit einhergehende verminderte Sulfadimidinelimination zu höheren Rückständen

im Ei führen kann. Zunächst wurden pharmakokinetische Grunddaten an einer

Gruppe von 6 Hühnern ermittelt, die mit 100 mg/kg KW Sulfadimidin i.v. behandelt

wurden. Danach wurde die Rückstandsbildung im Ei nach einmaliger oraler

Sulfadimidin-Applikation (5 Gruppen von jeweils 6 Hühnern in Dosierungen von 1,

3, 10, 30 and 100 mg/kg KW) sowie nach einwöchiger Dauermedikation mit dem

Futter (3 Gruppen von jeweils 6 Hühnern in Dosierungen von 3, 10 and 30 mg/kg

KW) untersucht. Zur Induktion eines Nierenschadens wurde Kaliumdichromat

benutzt und der Effekt durch klinische und pathohistologische Parameter abgeschätzt.

Bei allen Experimenten erfolgte die Analytik interessierender Matrices (Plasma,

Eigelb und Eiweiss) mittels einer HPLC-UV-Methode, welche hinsichtlich Spezifität,

Linearität, Nachweis- und Quantifizierungsgrenze, Richtigkeit, Präzision und

Wiederfindung validiert wurde. Dabei erwies sich die Methode als linear (R²>0.999 in

allen untersuchten Matrices). Die Nachweisgrenze wurde bei 0,01 µg/ml (Plasma)

bzw. 0,025 µg/g (Eigelb und Eiweiss) und die Quantifizierungsgrenze bei 0,025

µg/ml (Plasma) und 0,05 µg/g (Eigelb und Eiweiss) etabliert. Bei allen Matrices

zeigte sich bei allen untersuchten Konzentrationen eine Tages- und

Zwischentagespräzision mit Abweichungen von <10% vom Mittelwert (Ausnahme:

Plasma an der Quantifizierungsgrenze: 16,9%). Für die Berechnung

pharmakokinetischer Parameter wurde eine Kinetik erster Ordnung im 1-

Kompartiment-Modell angenommen. Dabei zeigte sich bei steigenden Dosierungen

eine Zunahme der Tmax, Cmax und der Halbwertszeit (K01) (nach 3 mg/kg KW: Tmax:

0,4 h, K01: 0,2 h; nach 100 mg/kg KW: Tmax: 4 h, K01: 1,2 h) sowie eine Abnahme

der Clearence. Nach i.v. Gabe war im Plasma nach 48 h und nach oraler Gabe nach 6-

32 h Sulfadimidin nicht mehr quantifizierbar. Messbare Sulfadimidinkonzentrationen

Page 95: A sulfadimidine model to evaluate pharmacokinetics and residues at

Zusammenfassung

95

wurden im Ei nach oraler Gabe von lediglich 1% der empfohlenen Dosis erreicht.

Diese wiesen sowohl nach einmaliger oraler Gabe als auch nach oraler Behandlung

über eine Woche im Eiweiss eine 2- bis 4-fach höhere Konzentration als im Eigelb

auf. Allerdings war Sulfadimidin nach Beendigung der Behandlung im Eigelb länger

nachweisbar (8 Tage) als im Eiweiss (6 Tage). Vergleichbar zu anderen

pharmakokinetischen Daten wies auch die Transferrate von Sulfadimidin ins Ei eine

Dosisabhängigkeit auf, wobei steigende Dosen mit einem höheren relativen Transfer

der Substanz ins Ei einhergingen, wodurch sich das Risiko einer Rückstandsbildung

im Ei bei subtherapeutischen Dosen zu verringern scheint. Beim Vergleich der

Kinetik von Sulfadimidin in gesunden und nierengeschädigten Hühnern zeigte sich

kein statistisch abzusicherner Unterschied. Dies könnte auf eine Sättigungskinetik

entweder der Absorption oder der Elimination hindeuten, wobei moderate

Nierenschäden zu keiner Veräbnderung des kinetischen Verhaltens von Sulfadimidin

führen würden.

Zusammenfassend zeigt die vorliegende Studie, dass die Aufnahme von

kontaminiertem Futter auch bei geringen Arzneistoffgehalten zu Rückständen in

Hühnereiern führen kann. Dabei wurden messbare Rückstände bereits nach

einmaliger Aufnahme eines Futters gemessen, welches lediglich 1% der

therapeutischen Dosis enthielt. Die deutliche Dosisabhängigkeit der Kinetik von

Sulfadimidin legt zudem nahe, dass das Rückstandsrisiko durch kontaminiertes Futter

auch von anderen Arzneimitteln bei relevanten Konzentrationen bestimmt, und nicht

von höheren Konzentrationen extrapoliert werden sollte.

Page 96: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

96

9 References

ABIOLA F.A., and K. AKA (1990):

Preliminary pharmacokinetics of sulfadimidine in healthy Sahelian sheep and cow.

J. Vet. Pharmacol. Ther. 13, 220-222

AERTS M.M.L. (1990):

Residues of veterinary drugs in edible products, an analytical approach.

Thesis, University of Amsterdam

AERTS M.M.L., A.C. HOGENBOOM and U.A. BRINKMAN (1995):

Analytical strategies for the screening of veterinary drugs and their residues in edible

products.

J. Chromatogr. B. 667, 1-40

AGARWAL V.K. (1992):

High-performance liquid chromatographic methods for the determination of

sulphonamides in tissues, milk and eggs.

J. Chromatogr. 624(1-2), 411-423

AIELLO S.E. (1998):

The Merck veterinary manual, 8th edn.

Merck & Co., inc. Whitehouse station, N.J., USA

ALLEN D.G., J.K. PRINGLE, D.A. SMITH, P.D. CONLON and P.M. BURGMANN

(1993):

Hand book of veterinary drugs.

Philadelphia, J.B. LIPPICOTT, USA

Page 97: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

97

ANONYMOUS 1 (1990):

Council regulation (EEC) No 2377/90.

Available at:

http://ec.europa.eu/food/food/chemicalsafety/residues/third_countries_en.htm

(accessed on 11.08.2006)

ANONYMOUS 2 (2004):

FDA, Subpart A-general provision; Definition-21 CFA 530.3 (i).

Available at:

http://a257.g.akamaitech.net/7/257/2422/14mar20010800/edocket.access.gpo.gov/cfr_

2004/aprqtr/21cfr530.3.htm (accessed on 17.08.2006)

ANONYMOUS 3 (2004):

Commission staff working document on the implementation of nation residue

monitoring plans in the member states in 2004 (Council Directive 96/23/EC).

Available at:

http://ec.europa.eu/food/food/chemicalsafety/residues/control_en.htm

(accessed on 21.7.2006)

ANONYMOUS 4 (2007):

EFSA, European food safety authority.

Cross-contamination of non-target feedingstuffs by lasalocid authorised for use as a

feed additive [1] - Scientific Opinion of the Panel on Contaminants in the food Chain.

Available at: http://www.efsa.europa.eu/EFSA/efsa_locale-

1178620753812_1178652751622.htm (accessed on 20.09.2007)

ANONYMOUS 5 (2004):

EMEA, European Medicines Agency, Veterinary Medicines and Inspections.

Studies to evaluate the safety of residues of veterinary drugs in human food:

reproduction testing.

Available at: http://www.emea.eu.int/pdfs/vet/vich/052500en.pdf (accessed on 16

June 2004)

Page 98: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

98

ANONYMOUS 6 (1996):

Council directive 96/23/EC

Available at:

http://ec.europa.eu/food/food/chemicalsafety/residues/third_countries_en.htm

(accessed on 11.08.2006)

ANONYMOUS 7 (2003):

FSIS, Food safety and inspection service in: National residue program data.

US department of agriculture, Office of public health and science, Chemistry and

toxicology division, Washington, DC.

Available at: http://www.fsis.usda.gov/Science/2003_Red_Book/index.asp

(accessed on 09.09.2006)

ANONYMOUS 8 (2004):

SANCO, the Directorate General for Health and Consumer Affairs.

Report for 2003 on the results of residue monitoring in food of animal origin in the

Member State (SANCO/2810/2004).

Available at: http://ec.europa.eu/food/chemicalsafety/residues/workdoc_2003_en.pdf

(accessed on 14.02.2007)

ANONYMOUS 9 (1981):

University of Kentucky and University of Nebraska, Preventing sulfa residues in pork

in: MU Extension.

Available at: http://muextension.missouri.edu/xplor/agguides/ansci/g02358.htm

(accessed on 10.3.2006)

ANONYMOUS 10 (2006):

Commission staff working document on the implementation of nation residue

monitoring plans in the member states in 2004 (Council Directive 96/23/EC).

Available at:

http://ec.europa.eu/food/food/chemicalsafety/residues/workdoc_2006_en.pdf

(accessed on 11.10.2008)

Page 99: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

99

ASHWORTH R.B., R.L. EPSTEIN, M.H. THOMAS and L.T. FROBISH (1986):

Sulfamethazine blood/tissue correlation study in swine.

Amer. J. Vet. Res. 47, 2596-2603

ATEF M., M.A. MOHAMMED and S.M. SOKKAR (1978):

Blood and tissue level of sulphamerazine in chickens.

Zentbl. Vet. Med. 25, 149-154

ATTA A.H., and S.A. EL-ZEINI (2001):

Depletion of trimethoprim and sulphadiazine from eggs of laying hens receiving

trimethoprim/sulphadiazine combination.

Food Control. 12, 269-274

AUGSBURG J.K. (1989):

Sulfa residues in pork: an update.

J. Anim. Sci. 67(10), 2817-2821

BAERT K., S. De BAERE, S. CROUBELS and P. De BACKER (2003):

Pharmacokinetics and oral bioavailability of sulfadiazine and trimethoprim in broiler

chickens.

Vet. Res. Commun. 27(4), 301-309

BAGGOT J.D. (2001):

The physiological basis of veterinary clinical pharmacology.

Blackwell Science, Oxford, England

BAJWA R.S., and SINGH J. (1977):

Studies on the levels of sulphadimethoxine and sulphamethoxypyridazine in blood of

poultry.

Indian J. Anim. Sci. 47, 549-553

Page 100: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

100

BARRAGRY T.B. (1994):

Veterinary drug therapy.

Lea & Febiger, Philadelphia, USA

BERGNER B., B. BOURGEOIS, M. EDELHAUSER, E. KLEIN, R. LIPPOLD, M.

MOLLERS and D. PLETSCHER (1993):

Chemical drug residue analysis of inhibitor-positive samples of meat, kidney and liver

in: Proceeding of the Euroresidue II conference on residue of veterinary drugs in food.

2(1), 186-190

BEVILL R.F. (1986):

Sulfonamide residues in swine

in: Practices in veterinary public health and preventive medicine in the United States.

G.T. Woods, Ames Iowa, Iowa state University Press

BEVILL R.F., L.W. DITTERT and D.W.A. BOURNE (1977):

Disposition of sulfonamides in food producing animals. IV. Pharmacokinetics of

sulfamethazine in cattle following administration of an intravenous dose and three

oral dosage forms.

J. Pharma. Sci. 66(5), 619-623

BIEHL L.G., R.F. BEVILL, M. LIMPOKA and G.D. KORITZ (1981):

Sufamethazine residues in swine.

J. Vet. Pharmacol. Ther. 4, 285-290

BLOM L. (1975):

Plasma half-lives and the excretion into egg-white and –yolk of three sulphonamides

and pyrimethamine after medication of laying hens.

Acta Pharmacol. Toxicol. (Copenh). 37(1), 79-93

Page 101: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

101

BLÜTHGEN A.H. (2000):

Contamination of milk from feed.

Bull. Int. Dairy Feder. 356, 43-47

BOTSOGLOU N.A., and D.J. FLETOURIS (2001):

Drug residues in foods pharmacology: food safety, and analysis.

Marcel dekker, New York, USA

BOURNE D.W.A., L.W. DITTERT, G.D. KORITZ and R.F. BEVILL (1978):

Disposition of sulfonamides in food-producing animals, VII. Disposition of

sulfathiazole in tissue, urine, and plasma of swine following intravenous

administration.

J. Pharmacokinet. Biopharm. 6(2), 123-134

BOURNE D.W.A., M. BIALER, L.W. DITTERT, M. HAYASHI, G. RUDAWSKY,

G.D. KORITZ and R.F. BEVILL (1981):

Disposition of sulfadimethoxine in cattle: inclusion of protein binding factors in a

pharmacokinetic model.

J. Pharm. Sci. 79(9), 1068-1072

CAMBELL K.L. (1999):

Sulphonamides: updates on use in veterinary medicine.

Vet. Dermatol. 10, 205-215

CAVALIERE C., R. CURINI, A.D. CORCIA, M. NAZZARI and R. SAMPERI

(2003):

A simple and sensitive liquid chromatography-mass spectrometry confirmatory

method for analyzing sulfonamide antibacterials in milk and egg.

J. Agric. Food Chem. 51,558-566

Page 102: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

102

CERNIGLIA C.E., and S. KOTARSKI (2005):

Approaches in the safety evaluations of veterinary antimicrobial agents in food to

determine the effects on the human intestinal microflora.

J. Vet. Pharmacol. Ther. 28, 3-20

CHOQUET-KASTYLEVSKY G., T. VIAL, and J. DESCOTES (2002):

Allergic adverse reactions to sulfonamides.

Curr. Allergy Asthma Rep. 2(1), 16-25

COLLINS B.K., C.P. MOORE and J.H. HAGEE (1986):

Sulfonamide-associated keratoconjunctivitis sicca and corneal ulceration in dysuric

dog.

J. Am. Vet. Med. Assoc. 189(8), 924-6

CORDLE M.K. (1989):

Sulfonamide residues in pork: past, present, and future.

J. Anim. Sci. 67(10), 2810-2816

CORPET D.E. (1987):

Antibiotic residues and drug resistance in human intestinal flora.

Antimicrob. Agents Chemother. 31(4), 587-593

CRAIG G.R., and G. WHITE (1976):

Studies in dogs and cats dosed with trimethoprim and sulphadiazine.

Vet. Rec. 98, 82-86

CRAIGMILL A.L., J. RIVIERE and A.I. WEBB (2006):

Tabulation of FARAD comparative and veterinary pharmacokinetic data.

Blackwell Publishing Ltd, UK

Page 103: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

103

CROMWELL G., T. STAHLY and T. MONEGUE (1980):

Effect of level of sulfamethazine in finishing feed for swine on sulfa residues in pork.

J. Anim. Sci. 51(1), 79

CURRY S.H. (1974):

Drug Disposition and Pharmacokinetics: with a consideration of pharmacological and

clinical relationship.

Blackwell Scientific Publications, Oxford, England

DAFT B.M., A.A. BICKFORD and M.A. HAMMARLUND (1989):

Experimental and field sulfaquinoxaline toxicosis in leghorn chickens.

Avian Dis. 33, 30-4

DEWDNEY J.M., L. MAES, J.P. RAYNAUD, F. BLANC, J.P. SCHEID, T.

JACKSON, S. LENS and C. VERSCHUEREN (1991):

Risk assessment of antibiotic residues of beta-lactams and macrolides in food

products with regard to their immuno-allergic potential.

Food Chem. Toxicol. 29(7), 477 -483

DEWULF J., B. CATRY, T. TIMMERMAN, G. OPSOMER, A. de KRUIF and D.

MAES (2007):

Tetracycline-resistance in lactose-positive enteric coliforms originating from Belgian

fattening pigs: Degree of resistance, multiple resistance and risk factors.

Prev. Vet. Med. 78, 339–351

DEY B.P., A. THALER and F. GWOZDZ (2003):

Analysis of microbiological screen test data for antimicrobial residues in food

animals.

J. Environ. Sci. Health B. 38(3), 391-404

Page 104: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

104

DONOHUE D.J. and H. HAIRSTON (2000):

Food safety implication: certain antibiotics may rapidly contaminate egg albumen

during the process of its formation.

Br. Poult. Sci. 41(2), 174-177

DOST F.H. (1953):

Der Blutspiegel, Kinetik der Konzentrationsabläufe in der Kreislaufflüssigkeit.

Georg Thieme Verlag, Leipzig

DOYLE M.E. (2006):

Veterinary drug residues in processed meats, potential health risk. Food research

institute.

Available at: http://www.wisc.edu/fri/briefs/FRIBrief_VetDrgRes.pdf (accessed on

March 2006)

DUFFEE N.E., R.F. BEVILL, J.C. THURMAN, H.G. LUTHER, D.E. NELSON and

F.E. HACKER (1984):

Pharmacokinetics of sulfamethazine in male, female and castrated male swine.

J. Vet. Pharmacol. Ther. 7, 203-211

EICHER O., A. FARAH, H. HERKEN and A.D. WELCH (1965):

Erzeugung von Krankheitszuständen durch das experment.

in: Handbook der experimentellen pharmacologie, vol. XVI/4.

Springer-verlag, Berlin

EINSTEIN R. (1994):

Principles of veterinary therapeutics.

Harlow, Essex: Longman Sciencetific & Technical

EL-SAYED M.G., M.I. ABD EL-AZIZ and M.H. EL-KHOLY (1995):

Kinetic behaviour of sulphaquinoxaline and amprolium in chickens.

Dtsch. Tierarztl. Wochenschr. 102(12), 481-485

Page 105: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

105

ELSHEIKH H.A., B.H. ALI, A.M. HOMEIDA, T. HASSAN and H.J. HAPKE

(1991):

Pharmacokinetics of antipyrine and sulphadimidine (sulphamethazine) in camel,

sheep and goat.

J. Vet. Pharmacol. Ther. 14, 269-275

ENDTZ H.P., G.J. RUIJS, B. VAN KLINGEREN, W.H. JANSEN, T. VAN DER

REYDEN and R.P. MOUTON (1991):

Quinolone resistance in campylobacter isolated from man and poultry following the

introduction of fluoroquinolones in veterinary medicine.

J. Antimicrob. Chemother. 27, 199-208

EPSTEIN R.L. and R.B. ASHWORTH (1989):

Tissue sulfonamide concentration and correlation in turkeys.

Am. J. Vet. Res. 50(6), 926-928

FAJT, V.R. (2001):

Label and extra label drug use in small ruminants.

Vet. Clin. North Am. Food Anim. Pract. 17(2), 403-420

FREY H.H. and W. LÖSCHER (2002):

Lerhbuch der Pharmakologie und Toxikologie für die Veterinärmedizin.

Enke, Stuttgart

FREY H.H., F. BERSCHAUER and W. LÖSCHER (1996):

Lerhbuch der Pharmakologie und Toxikologie für die Veterinärmedizin.

Enke, Stuttgart

FURUSAWA N. (1999b):

Liquid-chromatographic determination of sulfadimidine in milk and eggs.

Fresenius J. Anal. Chem. 364, 270-272

Page 106: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

106

FURUSAWA N. (2002):

Determination of sulfonamide residues in eggs by liquid chromatography.

J. AOAC Int. 85(4), 848-851

FURUSAWA N. (2003b):

Rapid high-performance liquid chromatographic determination technique of

sulphamonomethoxine, sulphadimethoxine, and sulphaquinoxaline in eggs without

use of organic solvents.

Chromatographia. 57, 317-320

FURUSAWA N. and K. KISHIDA (2002):

Transfer and distribution profiles of dietary sulphonamides in the tissues of the laying

hen.

Food Addit. Contam. 19(4), 368-372

FURUSAWA N. and K. KISHIDA (2005):

A green, low-cost method for determining sulphonamides in milk.

LC.GC Europe, November, 600-605

FURUSAWA N. and T. MUKAI (1995):

Easiness of Transfer of Dietary Sulfamonomethoxine into Eggs.

Jpn. Poult. Sci. 32, 26-33

FURUSAWA N. (2001):

Transference of dietary veterinary drugs into eggs.

Vet. Res. Commun. 25, 651-662

FURUSAWA N., T. MUKAI and M. YOSHIDA (1994):

Easiness to transfer of dietary sulfadimethoxine into eggs and its disappearance

pattern from eggs.

Jpn. Poult. Sci. 31, 168-180

Page 107: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

107

FURUSAWA N., Y. TSUZUKIDA and H. YAMAGUCHI (1998):

Decreasing profile of residual sulphaquinoxaline in eggs.

Br. Poult. Sci. 39, 241-244

FURUSAWA N. and T. MUKAI (1995):

Transfer of dietary sulphamonomethoxine and sulphadimethoxine into various tissues

of laying hens.

Br. Poult. Sci. 36(2), 313-316

FURUSAWA, N. (2000):

Hepatic biotransformation profiles of sulphamonomethoxine in food-producing

animals and rats in vitro.

Acta Vet. Hung. 48(3), 293-300

GARWACKI S., J. LEWICKI, M. WIECHETEK, S. GRYS, J. RUTKOWSI and M.

ZAREMBA (1996):

A study of the pharmacokinetics and tissue residues of an oral

trimethoprim/sulfadiazine formulation in healthy pigs.

J. Vet. Pharmacol. Ther. 19(6), 423-430

GEERTSMA M.F., J.F.M. NOUWS, J.L. GRONDEL, M.M.L. AERTS, T.B. VREE

and C.A. KAN (1987):

Residues of sulphadimidine and its metabolites in eggs following oral sulphadimidine

medication of hens.

Vet. Q. 9, 67-75

GIGUÈRE S., J.F. PRESSCOTT, J.D. BAGGOT, R.D. WALKER and P.M.

DOWLING (2006):

Antimicrobial therapy in veterinary medicine. 4th edition

Blackwell Publishing Ltd, Oxford, UK

Page 108: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

108

GOREN E., W.A. de JUNG and P. DOORNENBAL (1984):

Some pharmacokinetics aspects of four sulphonamides and trimethoprim, and their

therapeutic efficacy in experimental Escherichia coli infection in poultry.

Vet. Q. 6(3), 134-140

GRASSIE L. (1998):

Eliminating sulfamethazine residues in milk.

FDA Vet. 3(4), 1

GREENE C.E. (1998):

Infectious disease of the dog and cat. 2nd edn.

W.B. Saunders Co, Philadelphia

HAMSCHER G., H. T. PAWELZICK, S. SCZESNY, H. NAU and J. HARTUNG

(2003):

Antibiotics in dust originating from a pig-fattening farm: A new source of health

hazard for farmers?

Environ. Health Perspect. 111(13), 1590-1594.

HARNER III J.P., T. HERRMAN, J. FALK and K. BEHNKE (1996):

Avoiding drug carryover during feed processing and delivery.

Kansas State University, Available on

http://www.oznet.ksu.edu/library/grsci2/mf2055.pdf (accessed on March 2006)

HEATH G.E., D.A. KLINE, C.J. BARNES and D.H. SHOWALTER (1975):

Elimination of sulfamethazine from edible tissues, blood, urine, and feces of turkey

poults.

Am. J. Vet. Res. 36(7), 913-917

Page 109: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

109

HELMUTH R. and D. PROTZ (1997):

How to modify conditions limiting resistance in bacteria in animals and other

reservoirs.

Clin. Infect. Dis. 24(1), 136-138

HORII S., C. MOMMA, K. MIYAHARA, T. MARUYAMA and M.

MASTSUMOTO (1990):

Liquid chromatographic determination of three sulfonamides in animal tissue and egg.

J. Assoc. Off. Anal. Chem. 73(6), 990-992

JAIN S.K. and J.S. PUNIA (2000):

Disposition kenetics of sulfadoxine following oral administration in poultry.

Indian J. Anim. Sci. 70(4), 399-400

JAIN S.K. and J.S. PUNIA (2001):

Pharmacokinetic profile of sulfadimidine following oral administration in poultry.

Indian Vet. Med. Jour. 25, 149-151

JAIN S.K. (1994):

Studies on metabolism and disposition kinetics of sulfonamides in sheep.

Dissertation. Tierärztliche Hochschule Hannover

JEMMI T. and M. KÖNIG (1999):

Residues of antibiotics and chemotherapeutics in meat [Rückstände von Antibiotika

und Chemotherapeutika in Fleisch].

Schweiz. Arch. Tierheilkd. 141(3), 109-113

KAARTINEN L., K. LÖHÖNEN, B. WIESE, A. FRANKLIN, S. PYÖRÄLÄ (1999):

Pharmacokinetics of sulphadiazine-trimethoprim in lactating dairy cows.

Acta Vet. Scand. 40(3), 271-278

Page 110: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

110

KAHN C.M. (2005):

The Merck veterinary manual. 9th edn.

Whitehouse station, NJ: Merck

KAN C. A., and J.G. JACOBS (1998):

Bepalen van de verdelingscoefficient van een aantal sulfonamiden over

water/organisch oplosmiddel, alsmede de verdeling over dooier/wit na toediening via

voer aan leghennen.

ID-DLO Internal Report 98.001, 37

KAN C.A. (2003):

Residues of veterinary drugs in eggs and possible explanation for their distribution

between egg white and yolk.

Dissertation. University of Wuppertal, Germany

KAN C. A., and M. PETZ (2000):

Residues of veterinary drugs in eggs and their distribution between yolk and white.

J. Agric. Food Chem. 48, 6397-6403

KAY J. F., and C.E. PENNY (1996):

Surveillance scheme for veterinary drug residues in the United Kingdom

in: Proceeding of the Euroresidue III conference on residue of veterinary drugs in

food. 2, 596-600

KENNEDY D.G., W.J. BLANCHFLOWER, P.J. HUGHES and W.J. McCAUGHEY

(1996):

The incidence and cause of lasalocid residues in eggs in Northern Ireland.

Food Addit Contam. 13, 787-794

KENNEDY D.G., A. CANNAVAN and R.J. McCRACKEN (2000):

Regulatory problems caused by contamination, a frequently overlooked cause of

veterinary drug residues.

J. Chromatogr. A. 882, 37-52

Page 111: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

111

KENNEDY D.G., P.J. HUGHES and W.J. BLANCHFLOWER (1998):

Ionophore residues in eggs in Northern Ireland: Incidence and cause.

Food Addit. Contam. 15(5), 535-541

KENNEDY D.G., W.G. SMYTH, S.A. HEWITT and J.D.G. McEVOY (1998):

Monensin carry-over into unmedicated broiler feeds.

Analyst. 123, 2529-2533

KEUKENS H. J., C.A. KAN and M.J.H. TOMASSEN (1996):

Study on the presence of olaquindox residues in eggs after administration of feeds

containing low levels of olaquindox to laying hens

in: Proceeding of the Euroresidue III conference on residue of veterinary drugs in

food. 2, 611-615

KIETZMANN M. (1980):

Beitrag zur Pharmakokinetik von Sulfonamiden beim Geflügel.

Dissertation. Tierärztliche Hochschule Hannover

KIETZMANN M., W. MARKUS, J. CHAVEZ and W. BOLLWAHN (1995):

Drug residues in untreated swine.

Dtsch. Tierarztl. Wochenschr. 102(11),441-442

KISHIDA K. (2007):

Restricted-access media liquid chromatography for determine of

sulfamonomethoxine, sulfadimethoxine, and their N4-acetyl metabolites in eggs.

Food Chem. 101, 281-285

KISHIDA K., K. NISHINARI and N. FURUSAWA (2004):

Liquid chromatographic determination of sulphamonomethoxine, sulphadimethoxine

and their n4-metabolites in chicken plasma.

Chromatographia. 61, 81-84

Page 112: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

112

KORSRUD G.O., J.O. BOISON, PAPICH, W.D.G. YATES, J.D. MACNEIL, E.D.

JANZEN, J.J. McKINNON, D.A. LAUNDY, G. LAMBERT, M.S. YOUNG and L.

RITTER (1994):

Depletion of pennicillin G residues in tissues and injection sites of yearling beef steers

dosed with benzathine pennilcillin G alone or in combination with procaine

pennicillin G.

Food Addit Contam. 11, 1-6

KORSRUD G.O., M.G. PAPICH, A.C. FESSER, C.D. SALISBURY and J.D.

MACNEIL (1996):

Residue depletion in tissues and fluids from swine fed sulfamethazine,

chlortetracycline and penicillin G in combination.

Food Addit Contam. 13(3), 287-292

KRIEG V.R. (1966):

Der Sulfonamidgehalt in Eiern, Blut und Organen von Weissen Leghorn während und

nach einer Behandlung mit dem Sulfonamid Sulmet (Sulfadimidin).

Archiv für Geflügelkunde. 30, 299-308

LABAUNE J.P. (1989):

Hand book of pharmacokinetics.

Ellis Horwood Limited, West Sussex, England

LANGSTON V.C., F. GALEY, R. LOVELL and W.B. BUCK (1985):

Toxicity and therapeutics of monensin.

Vet. Med. 80, 75-84

LARNZ R., P. KUHNERT and P. BOERLIN (2003):

Antimicrobial resistance and resistance determinants in clinical Escherichia coli from

different animal species in Switzerland.

Vet. Microbiol. 91(1), 73-84

Page 113: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

113

LI H., P.J. KIJAK, S.B. TURNIPSEED and W. CUI (2006):

Analysis of veterinary drug residues in shrimp: A multi-class method by liquid

chromatography-quadrupole ion trap mass spectrometry.

J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci. 836(1-2), 22-38

LI T., G.L. QIAO, G.Z. HU, F.D. MENG, Y.S. QIU, X.Y. ZHANG. W.X. GUO, H.L.

YIE, S.F. LI and S.Y. LI (1995):

Comparative plasma and tissue pharmacokinetics and drug residue profiles of

different chemotherapeutants in fowls and rabbits.

J. Vet. Pharmacol. Ther. 18(4), 260-273

LIERZ M. (2003):

Avian renal disease: pathogenesis, diagnosis, and therapy.

Vet. Clin. Exot. Anim. 6, 29-55

LIONEL A.P., R.D. DANIEL, W.G. DAVID, A.M. MARGARET, J.L. RONALD,

A.C. DANIEL, F.K. FREDD and A.S. BERNARD (1999):

An FDA Review of Sulfamethazine Toxicity.

Regul. Toxicol. Pharmacol. 30, 217-222

LITTLEFIELD N.A., D.W. GAYLOR, B.N. BLACKWELL and R.R. ALLEN

(1989):

Chronic toxicity/carcinogenicity studies of sulphamethazine in B6C3F1 mice.

Food Chem. Toxicol. 27(7), 455-63

LITTLEFIELD N.A., W.G. SHELDON, R.R. ALLEN and D.W. GAYLOR (1990):

Chronic toxicity/carcinogenicity studies of sulphamethazine in Fischer 344/N rats:

two-generation exposure.

Food Chem. Toxicol. 28(3), 157-67

Page 114: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

114

LÖSCHER W., G.P. FASSBENDER, M. WEISSING and M. KIETZMANN (1990):

Drug plasma levels following administration of trimethoprim and sulphinamide

combinations to broilers.

J.Vet. Pharmacol. Ther. 13, 309-319

LÜDERS H., K.W. LAI and K.H. HINZ (1974):

Blood and tissue content of sulfamethazine and sulfaquinoxaline in broilers following

medication with drinking water. A combination to mass medication in poultry.

Zentralbl. Veterinärmed. B. 21, 110-118

LUNESTAD B. T., and K. GRAVE (2005):

Therapeutic agents in Norwegian aquaculture from 2000 to 2004: Usage and residue

control.

Bull. Eur. Assoc. Fish Pathol. 25(6), 284-290

MANDELL G. L., and W.A. PETRI (1996):

Sulfonamides, trimethoprim-sulfamethoxazole, quinolones and agent for urinary tract.

in: Hardman J.G., LIMBIRD L.E., MOLINOFF P.B., RUDDON R.W., GILMAN

A.G., eds. Goodman and Gilman’s the Pharmacological basis of therapeutics, 9th edn.

New York: McGraw-Hill, 1057-72

McCAUGHEY W.J., C.T. EILLOT, J.N. CAMPBELL, W.J. BLANCHFLOWER and

D.A. RICE (1990a):

Tissue residues in pigs fed on meal contaminated with sulphadimidine during mixing.

Ir. Vet. J. 48, 127-130

McCAUGHEY W.J., C.T. ELLIOTT and S.R.H. CROOK (1990b):

Carry-over of sulphadimidine in the faeces and urine of pigs fed medicated feed.

Vet. Rec. 126, 351-354

Page 115: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

115

McCAUGHEY W.J., J.N. CAMPBELL and C.T. ELLIOTT (1990c):

Reduction of sulphadimidine contamination in pig feedingstuffs.

Vet. Rec. 126, 113

McCRACKEN R.J., and D.G. KENNEDY (1997):

Determination of the furazolidone metabolite, 3-amino-2-oxazolidinone, in porcine

tissues using liquid chromatography-thermospray mass spectrometry and the

occurrence of residues in pigs produced in Northern Ireland.

J. Chromatogr. B. Biomed. Appl. 691(1), 87-94

McEVOY J.D. (2002):

Contamination of animal feedingstuffs as a cause of residues in food: a review of

regulatory aspects, incidence and control.

Anal. Chim. Acta 473, 3-26

McEVOY J.D., C.S. MAYNE, H.C. HIGGINS and D.G. KENNEDY (1999):

Transfer of sulphamethazine from contaminated dairy feed to cows' milk.

Vet Rec. 144, 470-475

McEVOY J.D., H.C. HIGGINS, C.S. MAYNE and D.G. KENNEDY (2000):

Transfer of sulphamethazine from contaminated dairy feed to cows' milk

in: Proceeding of the Euroresidue IV conference on residue of veterinary drugs in

food. 2,753-757

McEVOY J.D.G., W.G. SMYTH and D.G. KENNEDY (2003):

Contamination of animal feedingstuffs with nicarbazin: Investigations in a feed mill.

Food Addit Contam. 20(2), 136-140

MENGELERS M.J., E.R. VAN GOGH, M.B. HUVENEERS, P.E. HOUGEE, H.A.

KUIPER, A. PIJPERS, J.H. VERHEIJDEN and A.S. VAN MIERT (2001):

Pharmacokinetics of sulfadimethoxine and sulfamethoxazole in combination with

trimethoprim after oral single- and multiple- dose administration to healthy pigs.

Vet. Res. Commun. 25(6), 461-481

Page 116: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

116

MITCHELL A.D., and G.D. PAULSON (1986):

Depletion kinetics of 14C-sulfamethzine {4-amino-N-(4,6-dimethly-2-

pyrimidnyl)benzene[U-14C]sulfonamide} metabolism in swine.

Drug Metab. Dispo. 14, 155-160

MORILD I., A. BOHLE and J.A. CHRISTENSEN (1985):

Structure of the avian kidney.

Anat. Rec. 212, 33-40

MORTIER L., A.C. HUET, C. CHARLIER, E. DAESELEIRE, P. DELAHAUT and

C. VAN PETEGHEM (2005):

Incidence of residues of nine anticoccidials in eggs.

Food Addit Contam. 22(11), 1120-1125

NAGATA T., M. SAEKI, T. LIDA, M. KATAOKA and R. INO (1989):

Transfer of dietary sulfadimethoxine and sulfamonomethoxine into eggs and their

disappearance from eggs.

J. Food Hyg. Soc. Jpn. 30, 375-383

NAGATA T., M. SAEKI, M. WAKI, M. KATAOKA and S. SHIKANO (1994):

Tissue residue of sulfadimethoxine following dietary administration to broiler-

chickens.

J. Vet. Med. Sci. 56(4), 795-797

NAGATA T., M. SAEKI, T. IIDA, M. KATAOKA and S. SHIKANO (1992):

Determination of pyrimethamine and sulphadimethoxine residues in eggs by high

performance liquid chromatography.

Br. Poult. Sci. 33, 953-961

NEIDERT E., and P.W. SASCHENBRECKER (1996):

Over view of the Canadian veterinary drug residue control program

in: Proceeding of the Euroresidue III conference on residue of veterinary drugs in

food. 2, 185-190

Page 117: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

117

NIELSON P., and F. RASMUSSEN (1977):

Half-life, apparent volume of distribution and protein-binding for some

sulphonamides in cows.

Res.Vet.Sci. 22, 205-208

NIELSON P., and F. RASMUSSEN (1975):

Elimination of trimethoprim in swine: Comparison of results obtained by three

analytical methods.

Acta Pharmacol. Toxicol. (Copenh). 37, 309-319

NOSE N., Y. HOSHINO, Y. KIKUCHI, H. MASAKI, S. HORIE and S.

KAWAUCKI (1982):

Residues of synthetic antibacterial food additives in tissues and eggs of chickens.

J. Food Hyg. Soc. Jpn. 23, 246-252

NOUWS J.F.M., M.F. GEERTSMA, J.L. GRONDEL, M.M.L. AERTS, T.B. VREE

and C.A. KAN (1988):

Plasma disposition and renal clearance of sulphadimidine and its metabolites in laying

hens.

Res. Vet. Sci. 44, 202-207

NOUWS J.F.M., T.B. VREE, R. AERTS and J. GRONDEL (1986):

Pharmacokinetics and residues of sulfadimidine and its N4-acetyl and hydroxyl

metabolites in food-producing animals

in: Agriculture uses of antibiotics (W.A. Moats, Ed.).

American Chemical Society, Washington, DC, 168-182

NOUWS, J.F.M., T.B. VREE, M. DEGEN and D. MEVIUS (1991):

Pharmacokinetics of sulphamethoxazole in calves and cows.

Vet. Q. 13(1), 10-15

OIKAWA H., K. NAKAMOTO, K. HIROTA and K. KATAGIRI (1977):

Clearance of sulfomethoxazole in eggs and tissues of chickens.

Poult. Sci. 56, 813-821

Page 118: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

118

ONODERA T., S.I. INOUE, A. KASAHARA and Y. OSHIMA (1970):

Experimental studies on sulfadimethoxine in fowls III. egg- and other tissue-levels of

sulfonamides.

Jpn. J. Vet. Sci. 32, 275-283

ORMEROD A.D., T. M. S. REID and R. A. MAIN (1987):

Penicillin in milk—its importance in urticaria.

Clinical & Experimental Allergy. 17 (3), 229–234

OSHIMA Y., A. KASAHARA, T. ONODERA and M. MOGI (1964):

Experimental studies on sulfadimethoxine in fowls. I. Plasma concentration in hens

after oral administration.

Jap. J. Vet. Sci. 26, 115-120

PAIGE J.C., and R. KENT (1987):

Tissue residue briefs.

FDA Vet, 11, 10

PAIGE J.C., L. TOLLEFSON and M. MILLER (1997):

Public health impact on drug residues in animal tissues.

Vet. Human. Toxicol. 39(3), 162-169

PAIGE J.C., L. TOLLEFSON and M.A. MILLER (1999):

Health implications of residues of veterinary drugs and chemicals in animal tissues.

Vet. Clin. N. Amer. Food Anim. Pract. 15, 31-43

PAPAPANAGIOTOU E.P., D.J. FLETOURIS and I.E. PSOMAS (2005):

Sulphamethazine residue depletion study in piglets after oral administration.

Anal. Chim. Acta. 529, 325-329

PAULSON G.D., V.J. FEIL, P.J. SOMMER and C.H. LAMOUREUX (1992):

Xenobiotics and food-producing animals

in: HUTSON D.H., D.R. HAWKINS, G.D. PAULSON and C.B. STRUBLE

Eds. American Chemical Society, Washington, DC, 190

Page 119: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

119

PENSABENE J.W., W. FIDDLER and D.J. DONOGHUE (1998):

Supercritical fluid extraction compared with solvent method for incurred

sulfamethazine in chicken eggs.

J. Food Sci. 63(1), 25-26

PETZ M. (1983):

Hochdruckflüssigchromatographische rücksandsanalyse von chloramphenicol,

furazolidon und fünf sulfonamiden in eiern, fleisch und milch

Z. Lebesm. Unters. Forsch. 176, 289-293

PETZ M. (1993):

Distribution of sulfaquinoxaline and three nitrofurans between yolk and egg white

during medication and depletion.

in: Proceeding of the Euroresidue II conference on residue of veterinary drugs in food.

2, 528-532

PLUMB D.C. (1991):

Vetreinary drug hand book.

White Bear lake, Minn: PharmaVet Publ. USA

PLUMB D.C. (2005):

Veterinary drug hand book. 5th edn.

Ames, Iowa, Blackwell, USA

PRESCOTT J.F., and J.D. BAGGOT (1993):

Antimicrobials therapy in veterinary medicine. 2nd edn.

Ames, Iowa, Blackwell, USA

PRESCOTT J.F. (2000):

Antimicrobials therapy in veterinary medicine. 3rd edn.

Ames, Iowa, Blackwell, USA

Page 120: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

120

PRIPWAI N., N CHAOWITTAYANGKUL, T. SUPPADIT and S. JATURASITHA

(2004):

Sulfamethazine residues in pork with different withdrawal periods and pig tissues in

Chiang Mai market.

Journal of Agriculture and Rural Development in the Tropics and Subtropics,

Supplement. 80, 214-223

RANA R., M.S. AKHTAR and M. NAWAZ (1993):

Residues of sulfaquinoxaline in poultry products.

Pakistan Vet. J. 13, 161-166

REDDY K.S., S.K. JAIN and R.P. UPPAL (1988):

Pharmacokinetics studies of sulphonamides in poultry.

Indain J. Anim. Sci. 58, 437-439

REYES-HERRERA I., M.J. SCHNEIDER, K. COLE, M.B. FARNELL, P.J. BLORE

and D.J. DONOGHUE (2005):

Concentrations of antibiotic residues vary between different edible muscle tissues in

poultry.

J. Food Prot. 68(10), 2217-2219

RIFFA S., M. NAWAZ and ZIA-UR-REHMAN (1982):

Pharmacokinetics of sulphadimidine in normal and febrile dogs.

J. Vet. Pharmacol. Ther. 5(2), 131-135

RIGHTER H. F., J. M. WORTHINGTON, H.E. ZIMMERMAN and H. D. MERCER

(1970):

Tissue-Residue Depletion of Sulfaquinoxaline in Poultry.

Am. J. Vet. Res. 31(6), 1051-1054

RIGHTER H.F., J.M. WORTHINGTON and H.D. MERCER (1971):

Tissue-residue depletion of Sulfamethazine in calves and chickens.

Am. J. Vet. Res. 32(7), 1003-1006

Page 121: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

121

RIVIERE J. E., A.L. CRAIGMILL and S.F. SUNDLOF (1991):

Handbook of comparative pharmacokinetics and residues of veterinary antimicrobials.

Boca Raton, FL: CRC press, Inc., Florida, USA

ROKKA M., EEROLA S., PERTTILA U., ROSSOW L., VENALAINEN E.,

VALKONEN E., VALAJA J. and K. PELTONEN (2005):

The residue levels of narasin in eggs of laying hens fed with unmedicated and

medicated feed.

Mol. Nutr. Food Res. 49(1), 38-42

ROLAND C., L. WILLIAM and D. ROLAND (1996):

Determination of veterinary drugs residues: our experience during the last five years.

in: Proceeding of the Euroresidue III conference on residue of veterinary drugs in

food. 2, 320-325

ROMVARY A., and F. SIMON (1992):

Sulfonamide residues in eggs.

Acta Vet. Hung. 40, 99-106

ROUDAUT B., and M. GARNIER (2002):

Sulphonamide residues in eggs following drug administration via the drinking water.

Food Addit. Contam. 19(4), 373-378

ROUDAUT B. (1989):

Residues of aminoglycoside antibiotics in eggs after medication of laying hen.

Br. Poult. Sci. 30(2), 265-271

ROUDAUT B. (1993):

Residues of sulphonamides in eggs following oral medication of laying hens

in: Proceeding of the Euroresidue II conference on residue of veterinary drugs in food,

2, 591-595

Page 122: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

122

SAKANO T., S. MASUDA and T. AMANO (1981):

Determination of residual diaveridine and sulfaquinoxaline in hen’s egg, chicken

plasma and tissues by high-performance liquid chromatography.

Chem. Pharm. Bull. 29, 2290-2295

SCHMIDT R.E. (2006):

Types of renal disease in avian species.

Vet. Clin. Exot. Anim. 9, 97-106

SERRATOSA J., A. BLASS, B. RIGAU, B. MONGRELL, T. RIGAU. M.

TORTADÈS, E. TOLOSA, C. AGUILAR, O. RIBÒ and J. BALAGUÉ (2006):

Residues from veterinary medicinal products, growth promoters and performance

enhancers in food-producing animal: a European Union perspective.

Rev.sci.tech.Off.int.Epiz. 25(2), 637-653

SHAIKH B., and P.S. CHU (2000):

Distribution of total 14C residue in egg yolk, albumen, and tissues following oral [14C]

sulfamethazine administration to hen.

J.Agric.Food Chem. 48, 6404-6408

SHAIKH B., N. RUMMEL and D. DONOGHUE (1999):

Determination of sulfamethazine and its major metabolites in egg albumen and egg

yolk by high performance liquid chromatography.

J. Liq. Chromatogr. Relat. Technol. 22(17), 2651-2662

SHAIKH B., N. RUMMEL and D. SMITH (2004):

Determination of 14C residue in egg of laying hens administered orally with (14C)

sulfaquinoxaline.

Food Addit. Contam. 21(6), 545-54

Page 123: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

123

SHEARER G. (1999):

Veterinary drugs including antibiotics

in: Enviromental contaminants in food: MOFFAT C.F. and WHITTLE K.J. eds. 1st

edn.

Sheffield Academic Press Ltd. England, 273-304

SIEB U. (1991):

Zum Rückstandsverhalten von Sulfadimidin in Eieren, Follikeln und Geweben von

Legenhennen.

Dissertation, University of Kiel

SIMUNEK J., and E. HEGEROVA (1990):

Influence of coccidiostats on the pharmacokinetics of orally administered

sulfonamides.

Eur. Assoc. Vet. Pharm. Tox. 4th Con. 171-175

SMITH J.L., D.J.V. DRUM, Y. DAI, J.M. KIM, S. SANCHEZ, J.J. MAURER, C.L.

HOFACRE and M.D. LEE (2007):

Impact of antimicrobial usage on antimicrobial resistance in commensal Escherichia

coli strains colonizing broiler chickens.

Appl. Environ. Microb. 73(5), 1404-1414

SMITH K.E., J.M. BESSER and F. LEANO (1998):

Flouroquinolone resistant Campylobacter isolated from humans and poultry in

Minnesota

in: Proc. International Conference on Emerging Infectious Diseases

Atlanta, GA, USA, March 8–11, 69

SPOO J.W., and J.E. RIVIERE (2001):

Sulfonamides

in: Veterinary pharmacology and therapeutics.

ADAMS H.R., Iowa, State University Press. Ames, 796-817

Page 124: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

124

STOLKER A.A.M., LINDERS S.H.M.A., Van GINKEL L.A. and U.A.T.

BRINKMAN (2004):

Application of the revised EU criteria for the confirmation of anabolic steroids in

meat using GC-MS.

Anal. Bioanal. Chem. 378(5), 1313-1321

SUKUL P. and M. SPITELLER (2006):

Sulfonamides in the environment as veterinary drugs

in: Reviews of environmental contamination and toxicology, volume 187.

Springer New York Inc, New York, USA, 67-101

TAKAHASHI Y., A.A. SAID, M. HASHIZUME and Y. KIDO (1991):

Sulfadimethoxine residue in broiler-chicken skin.

J. Vet. Med. Sci. 53(1), 33-36

TOMASSEN M.J.H., H.J. KEUKENS and C.A. KAN (1996):

Overdracht van lage doseringen sulfadiazine van voer naar ei.

RIKILT rapport. 96(37), 10 pp

TRETTIEN A.L., and M.A. KRAM (1994):

Primor® once-a-day canine antibacterial has important advantages.

SmithKline Beecham animal health technical bulletin. July, 1-6

VAN GOGH H. (1980):

Pharmacokinetics of nine sulphonamides in goats.

J. Vet. Pharmacol. Ther. 3, 69-81

VLAHOVIĆ M., I. RAJIĆ, R. CMILJANIĆ and B. ŽIVKOVIĆ (1996):

Dynamics of excretion of sulfonamides from the tissue of fattening chicks.

Acta Vet. Brno. 46(4), 221-226

Page 125: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

125

VREE T.B., and Y.A. HEKSTER (1985):

Pharmacokinetics of Sulfonamides revisited.

Antibiot. Chemother. 34, 1-208

VREE T.B., J.J. REEKERS- KETTING, Y.A. HEKSTER and J.F.M. NOUWS,

(1883):

Acetylation and deacetylation of sulfonamides in dogs.

J. Vet. Pharmacol. Therap. 6,153-156

VREE T.B., Y.A. HEKSTER, J.F.M. NOUWS and G.M. DORRESTEIN (1987):

Pharmacokinetics of sulfonamides in animals.

Antibiot. Chemother. 37, 131-177

WANG S., H.Y. ZHANG, L. WANG, Z. J. DUAN and I. KENNEDY (2006):

Analysis of sulphonamide residues in edible animal products: A review.

Food Addit. Contam. 23(4), 362-384

WEISS C., A. CONTE, C. MILANDRI, G. SCORTICHINI, P. SEMPRINI, R.

USBERTI and G. MIGLIORATI (2007):

Veterinary drugs residue monitoring in Italian poultry: Current strategies and possible

developments.

Food Control. 18, 1068-1076

WHIPPLE D.M., G. SAMUELSON, G.E. HEATH and D.H. SHOWALTER (1980):

Tissue residue depletion and recycling of sulfamethazine in swine.

J. Am. Vet. Med. Assoc. 176, 1348-1352

WILCKE J.R. (1988):

Therapeutic application of sulfadiazine/ trimethoprim in dogs and cats: a review.

Companion Anim. Pract. 2, 3-8

Page 126: A sulfadimidine model to evaluate pharmacokinetics and residues at

References

126

WITKAMP R.F., H.I. YUN, G.A.E. VAN’T KLOOSTER, J.F. VAN MOSEL, J.M.

ENSINK, J. NOORDHOEK and A.S.J.P.A.M. VAN MIERT (1992):

Comparative aspect and sex differentiation of plasma sulfamethazine elimination and

metabolite formation in rats, rabbits, dwarf goats and cattle.

Am. J. Vet. Res. 53(10), 1830-1835

WITKAMP R.F., S.M. NIJMEIJER, C.T.M. VAN DUIN, M.M. BEVERS, T.

WENSING, H. VAN GOUH and A.S.J.P.A.M. VAN MIERT (1989):

Has bovine somatotropin (BST) an effect upon drug disposition? Comparative studies

in goat and cattle with sulphadimidine and antipyrine after parenteral administration

of BST, zeranol and proligestone.

J. Vet. Pharmacol. Ther. 12, 163-178

WOODS D.D. (1940):

The relation of p-aminobenzoic acid to the mechanism of the action of sulfanilamide.

Br. J. Exp. Path. 21 , 74-90

YUAN Z.H., X.Q. MIAO and Y.H. YIN (1997):

Pharmacokinetics of ampicillin and sulfadimidine in pigs infected experimentally

with Streptococcus suum.

J. Vet. Pharmacol. Ther. 20, 318-322

Page 127: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

127

10 Appendix

10.1 Glossary of abbreviations

β the apparent first-order rate constant fraction

µL microliter

µg microgram

AUC area under the curve

AUMC the area under the (first) moment curve

BW body weight

C(T) plasma drug concentration at any time

C° degree celsius

Calbumen the measured concentration in albumen

CL the systemic clearance

CL_F the total body clearance (extra-vascular models)

Cmax the peak plasma concentration

concn concentration

Cp0 drug concentration at time zero

Cyolk the measured concentration in yolk

D dose

dL deciliter

F the fraction of absorption

g gram

h hour

HCl hydrochloric acid

HPLC High pressure liquid chromatography

i.m. intramuscular

i.p. intraperitoneal

I.S. internal standard

i.v. intravenous

K01 the apparent first-order absorption rate constant

K01_HL the absorption half-life

Page 128: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

128

K10 the apparent first-order elimination rate constant

K10_HL the elimination half-life

K2Cr2O7 Potassium dichromate

Ka the rate constant of absorption

Kel the rate constant of elimination

Koc the organic carbon adsorption coefficient

kg kilogram

ln natural logarithm

L liter

LOD limit of detection

LOQ limit of quantification

malbumen mass of albumen

mg milligram

min minute

mL milliliter

Mol mole

MRL maximum residue limit MRT the mean residence time

myolk mass of yolk

N normal

n number

N2 nitrogen

NaCl sodium chloride

NaH2PO4 · H2O Phosphate hydrogen monohydrate

ND not detected

ng nanogram

nm nanometer

p.o. per os

R2 correlation coefficient

RSD relative standard deviation

Rtotal the sum of residues in albumen and yolk

SD standard deviation

SDD sulfadimidine

SDX sulfadoxine

Page 129: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

129

T time

T1/2el the elimination half-life

Tmax the maximum concentration

TR drug transfer rate into the eggs

UV Ultraviolet

V an apparent volume of distribution

v/v volume/volume

Vd the volume of distribution

Vss the volume of distribution at steady state

Page 130: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

130

10.2 Tables Hen (Number)

Time (h) 1 2 3 4 5 6 Mean SD

0.25 124.29 101.68 137.71 141.14 156.46 115.11 129.40 19.69

0.5 126.63 91.97 126.19 129.17 139.36 116.78 121.68 16.25

1 114.12 86.25 116.80 122.90 129.86 108.99 113.16 15.03

2 109.03 89.47 111.77 117.01 119.87 101.92 108.18 11.11

4 93.40 57.19 87.48 71.99 101.55 91.97 83.93 16.34

6 76.00 40.93 70.81 55.99 73.92 68.49 64.36 13.45

8 55.06 29.23 59.84 50.93 58.40 63.91 52.89 12.40

12 37.54 10.97 32.12 28.76 32.80 37.68 29.98 9.92

24 5.91 0.67 6.10 3.86 4.37 5.28 4.37 2.01

32 0.46 0.22 0.39 0.25 0.25 0.26 0.30 0.09

48 0.08 0.13 0.13 ND ND ND 0.06 0.06

Table 1: SDD plasma concentration (µg/mL) after i.v. administration at 100

mg/kg BW

Page 131: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

131

Hen (Number)

Time (h) 1 2 3 4 5 6 Mean SD

0.5 38.94 48.99 5.57 40.84 30.35 20.01 30.78 15.82

1 45.20 53.04 31.84 44.70 41.18 29.78 40.96 8.79

2 78.04 76.94 52.42 86.75 69.57 45.90 68.27 15.49

4 76.39 80.06 63.89 84.78 84.17 44.81 72.35 15.49

6 70.43 66.63 51.26 96.40 94.21 35.58 69.08 23.77

8 62.06 66.75 52.83 86.58 87.72 37.22 65.53 19.55

12 63.68 46.39 34.94 67.67 68.95 24.38 51.00 18.70

24 23.75 23.56 30.98 26.37 31.49 2.34 23.08 10.72

32 1.83 1.64 2.77 3.64 6.15 ND 2.67 2.10

Table 2: SDD plasma concentration (µg/mL) after p.o. administration at 100

mg/kg BW

Hen (Number)

Time (h) 1 2 3 4 5 6 Mean SD

0.5 20.00 16.78 26.37 33.37 14.34 0.75 18.60 11.14

1 20.14 21.70 24.35 28.43 27.08 3.84 20.92 8.93

2 21.68 21.56 23.90 24.77 24.25 23.82 23.33 1.36

4 25.83 14.16 17.35 17.47 20.18 14.95 18.32 4.25

6 10.38 8.13 10.92 11.66 14.13 9.36 10.76 2.06

8 5.52 4.28 5.70 6.23 7.60 3.94 5.54 1.33

12 1.95 0.66 1.19 1.38 1.80 0.63 1.27 0.55

24 ND ND ND ND ND ND - -

Table 3: SDD plasma concentration (µg/mL) after p.o. administration at 30

mg/kg BW

Page 132: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

132

Hen (Number)

Time (h) 1 2 3 4 5 6 Mean SD

0.5 1.78 6.45 7.09 4.25 8.52 3.67 5.29 2.49

1 5.41 4.92 3.95 6.31 7.35 2.54 5.08 1.71

2 4.38 5.06 3.57 4.82 5.95 2.68 4.41 1.15

4 1.69 2.49 1.44 1.30 - 1.29 1.64 0.50

6 0.46 0.98 0.44 0.30 0.81 0.39 0.56 0.27

8 0.12 0.40 0.51 0.36 0.27 0.31 0.33 0.13

12 - 0.06 ND ND 0.11 0.03 0.03 0.04

24 0.08 0.04 ND ND 0.03 0.06 0.03 0.03

Table 4: SDD plasma concentration (µg/mL) after p.o. administration at 10

mg/kg BW

Hen (Number)

Time (h) 1 2 3 4 5 6 Mean SD

0.5 1.47 2.17 2.88 3.29 2.41 0.32 2.09 1.06

1 1.46 1.56 1.84 2.06 1.89 1.11 1.65 0.35

2 0.81 0.87 0.97 0.81 0.82 0.43 0.78 0.18

4 0.19 0.29 0.22 0.24 0.22 0.09 0.21 0.07

6 0.07 0.07 0.06 0.09 0.06 0.06 0.07 0.01

8 <0.025 0.03 ND <0.025 <0.025 <0.025 <0.025 -

Table 5: SDD plasma concentration (µg/mL) after p.o. administration at 3

mg/kg BW

Page 133: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

133

Hen (Number)

Time (h) 1 2 3 4 5 6 Mean SD

0.5 0.78 0.75 0.55 0.13 0.52 0.47 0.53 0.23

1 0.47 0.44 0.43 0.56 0.69 0.50 0.51 0.10

2 0.23 0.20 0.18 0.25 0.20 0.23 0.21 0.02

4 0.05 0.04 0.06 0.10 0.05 0.06 0.06 0.02

6 0.03 <0.025 0.03 0.04 <0.025 0.03 0.02 0.01

8 ND ND ND ND ND ND - -

Table 6: SDD plasma concentration (µg/mL) after p.o. administration at 1

mg/kg BW

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 15.40 19.68 10.51 20.75 5.28 - 14.32 6.47

2 6.86 7.27 6.17 10.19 2.60 - 6.62 2.72

3 3.90 4.69 3.45 4.55 1.78 - 3.68 1.17

4 3.18 3.01 2.83 3.16 1.54 - 2.75 0.69

5 2.33 2.15 1.66 2.58 1.18 - 1.98 0.56

6 1.94 1.18 0.59 1.67 0.70 - 1.21 0.59

7 0.88 0.24 0.06 0.16 0.36 - 0.43 0.32

8 0.28 0.02 - - 0.06 - 0.12 0.14

9 <0.05 <0.05 <0.05 <0.05 <0.05 - <0.05 -

Table 7: SDD residue concentration (µg/g) in yolk following p.o.

administration at 100 mg/kg BW

Page 134: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

134

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 1.81 1.93 3.46 5.53 2.67 - 3.08 1.52

2 0.46 - 0.63 0.41 0.56 - 0.51 0.10

3 0.44 0.36 0.45 0.63 0.31 - 0.44 0.12

4 0.38 0.34 0.26 0.31 - - 0.32 0.05

5 0.25 0.17 0.18 - - - 0.20 0.04

6 0.16 0.14 0.10 0.13 0.13 - 0.13 0.02

7 0.09 0.04 0.08 - - - 0.07 0.03

8 <0.05 <0.05 <0.05 <0.05 <0.05 - <0.05 -

Table 8: SDD residue concentration (µg/g) in yolk following p.o.

administration at 30 mg/kg BW

Hen (Numer)

Day 1 2 3 4 5 6 Mean SD

1 0.44 0.33 0.39 0.48 0.54 0.34 0.42 0.08

2 0.07 0.08 0.06 0.06 0.10 0.04 0.07 0.02

3 0.05 0.08 0.06 0.04 0.09 - 0.06 0.02

4 0.03 0.05 0.04 0.03 0.04 - 0.04 0.01

5 <0.05 <0.05 <0.05 <0.05 <0.05 - <0.05 -

Table 9: SDD residue concentration (µg/g) in yolk following p.o.

administration at 10 mg/kg BW

Page 135: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

135

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 0.08 0.16 0.07 0.14 0.15 0.06 0.11 0.05

2 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 -

Table 10: SDD residue concentration (µg/g) in yolk following p.o.

administration at 3 mg/kg BW

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 53.36 57.61 39.05 50.05 46.64 - 49.34 7.04

2 6.33 6.69 5.56 5.97 5.93 - 6.10 0.43

3 0.85 0.78 0.62 0.81 1.07 - 0.83 0.16

4 0.45 0.49 0.33 0.42 0.20 - 0.38 0.12

5 0.28 0.26 0.14 0.18 0.12 - 0.20 0.07

6 0.16 0.12 0.09 0.14 0.08 - 0.12 0.03

7 <0.05 <0.05 <0.05 <0.05 <0.05 - <0.05 -

Table 11: SDD residue concentration (µg/g) in albumen following p.o.

administration at 100 mg/kg BW

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 6.34 3.97 8.30 7.77 4.85 - 6.25 1.85

2 0.13 - 0.17 0.22 0.13 - 0.16 0.04

3 0.08 0.41 0.07 0.12 0.07 - 0.15 0.15

4 <0.05 <0.05 <0.05 <0.05 <0.05 - <0.05 -

Table 12: SDD residue concentration (µg/g) in albumen following p.o.

administration at 30 mg/kg BW

Page 136: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

136

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 1.02 1.23 0.73 0.96 1.24 0.68 0.98 0.24

2 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 -

Table 13: SDD residue concentration (µg/g) in albumen following p.o.

administration at 10 mg/kg BW

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 0.02 0.42 0.22 0.36 0.35 0.12 0.28 0.12

2 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 -

Table 14: SDD residue concentration (µg/g) in albumen following p.o.

administration at 3 mg/kg BW

Hen (Number)

Day 1 2 3 4 5 6 Mean SD

1 0.09 0.07 0.04 0.08 0.10 0.08 0.08 0.02

2 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 -

Table 15: SDD residue concentration (µg/g) in albumen following p.o.

administration at 1 mg/kg BW

Page 137: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

137

Hen (Number)

Time (h) 1 2 3 4 Mean SD

0.5 137.65 112.57 153.44 137.20 135.21 16.88

1 121.93 104.11 134.75 112.96 118.44 13.08

2 100.41 92.08 116.82 96.87 101.55 10.74

4 95.16 77.34 104.42 85.20 90.53 11.79

6 80.61 59.16 88.93 72.81 75.37 12.66

8 70.44 50.79 87.35 66.49 68.77 15.02

12 70.51 41.55 74.15 54.11 60.08 15.12

24 28.71 6.18 11.14 14.83 15.2 2 9.67

32 5.86 0.05 0.43 1.10 1.86 2.70

48 ND ND ND ND ND -

Table 16: SDD plasma concentration (µg/mL) after i.v. administration at

100 mg/kg BW in healthy hen, experiment II, (n=4)

Page 138: A sulfadimidine model to evaluate pharmacokinetics and residues at

Appendix

138

Impaired kidney hen (Number)

Time (h) 1 2 3 4 5 6 Mean SD

0.5 59.52 98.16 118.45 161.01 124.53 140.63 117.05 35.25

1 64.51 101.53 121.95 152.42 125.13 134.21 116.62 30.44

2 67.04 76.71 106.22 145.59 114.88 119.73 105.03 29.00

4 71.19 80.11 96.99 137.71 101.36 124.17 101.92 25.38

6 71.52 61.96 78.16 126.74 87.33 115.93 90.27 25.68

8 58.56 53.52 74.31 118.30 84.93 107.66 82.88 26.07

12 50.72 33.75 61.99 105.14 73.92 104.88 71.73 28.98

24 7.72 4.23 16.88 62.65 24.68 49.08 27.54 23.47

32 0.30 0.09 2.69 36.80 8.41 27.57 12.64 15.70

48 ND ND ND 0.34 ND 0.19 0.26 0.11

60 ND ND ND 0.11 ND 0.09 0.10 0.01

Table 17: SDD plasma concentration (µg/mL) after i.v. administration at 100 mg/kg

BW in kidney damaged hens, experiment II, (n=6)

Page 139: A sulfadimidine model to evaluate pharmacokinetics and residues at

Acknowledgement

139

11 Acknowledgement

I wish to express deep gratitude to my supervisor, Univ. Prof. Dr. Manfred

Kietzmann, who provided me with a great opportunity to undertake this study. I am

also highly indebted to him for his excellent guidance, supervision and advice not

only on academic aspects but also on a more personal level.

I would like to extend my grateful thanks to Dr. Frank Niedorf for his valuable

technical support, kind advice on various aspects and good corporation. Grateful

appreciation is also extended to Dr. Wolfgang Baümer and Dr. Michael Braun, for

their kind willingness to help me every time I asked for it. I will not forget with their

help with improving my German laguage skills, idiomatic and slang words and

inviting me to join them at parties, talking about culture and telling about humorous

stories.

My special thanks go to Mr. H.-H. Bohr and Ms.Victoria Garder for their technical

assistance, preparing some solutions and handling the chickens.

I express sincere thanks to Prof. Dr. Dr. Wulf Winkenwerder and his family for their

warm hospitality, helping all Thai veterinary students in Germany.

My deeply heartfelt thanks go to Assoc. Dr. Chartchai Noonpugdee, who passed away

before I finished doctoral degree. His suggestions and inspiration, helping me during

my studies at the TiHo will always remain in my memory.

My thanks go to Thai government under Prime Minister Dr. Thaksin Shinnawatra

(2003) for their full scholarship support of my Doctoral study

I would like to thank the staff of Foreign Student Affair of the TiHo, Hannover for

looking after overseas students and taking us on interesting cultural tours.

Page 140: A sulfadimidine model to evaluate pharmacokinetics and residues at

Acknowledgement

140

I also wish to thank for all doctoral students, trainees and laboratory technicians of the

Institute of Pharmacology, Toxicology and Pharmacy, TiHo Hannover, who helped

me improve my German language skills and gave me a fun time.

My immense thanks go to Asist. Prof. Dr. Ukadej Boonprakob and Asist. Prof. Dr.

Chaiyan Kasorndorkbua for proof-reading the first English version.

I am thankful to the administrators and colleagues of the Faculty of Veterinary

Medicine and Kasetsart University for their supported and devoted time during my

study period.

An exciting, funny and enjoyable life in Germany would not have occurred without

my Thai friends (P´ Wan, P´ Chu, Tom, Knot, Pae, Benz among others) who shared

experiences, happiness and activities such as Thai cooking, short trips, parties, and

sports activities.

Special thanks go to Dr. Pornchai Sanyathitiseree (P´ Rug), who helped me from the

very first day I came to Germany until the day he returned to Thailand.

Last but not least, I am heartily grateful to my wife, for her help, patience and

encouragement during the tenure of this study.

Nevertheless, any weaknesses in this dissertation are entirely my own responsibility.

Lastly, the encouragement and support of my beloved family, mother, older sisters

and brother are gratefully acknowledged.