Top Banner
DOI 10.1515/revac-2012-0020 Rev Anal Chem 2013; 32(1): 15–34 Ana M. Botana, Paz Otero, Paula Rodríguez, Amparo Alfonso and Luis M. Botana* Current situation on analysis of marine toxins Abstract: Marine toxins are a food safety concern world- wide. This review discusses current analytical methods for those toxins that are legally regulated in Europe, namely domoic acid, saxitoxin, okadaic acid, yessotoxin, pectenotoxin and azaspiracids, and all their analogues, and those that are not regularly monitored but are a threat to humans, such as tetrodotoxin, ciguatoxins and cyclic imines. Because of legislative changes that were imple- mented in 2011, most legally required methods are chro- matographic. Saxitoxin and domoic acid are monitored by high-performance liquid chromatography with fluo- rescent and ultraviolet detection, respectively. All other toxins are monitored nowadays by means of chromato- graphic separation and mass spectrometry detection. Keywords: analytical standards; chromatography; marine toxins; mass spectrometry. *Corresponding author: Luis M. Botana, Department of Pharmacology, Veterinary School, Faculty Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, Lugo 27002, Spain, e-mail: [email protected] Ana M. Botana: Department of Analytical Chemistry, Universidad de Santiago de Compostela, Campus de Lugo, Lugo 27002, Spain Paz Otero: Department of Pharmacology, Veterinary School, Faculty Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, Lugo 27002, Spain Paula Rodríguez: Department of Pharmacology, Veterinary School, Faculty Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, Lugo 27002, Spain Amparo Alfonso: Department of Pharmacology, Veterinary School, Faculty Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, Lugo 27002, Spain Introduction Phycotoxins are an acute worldwide seafood safety concern; hence, their detection and quantification in food with reliable methods is important. The methods must be validated according to international guidelines to ensure adequate results and to protect public health and reduce their economic impact. Harmful algal blooms is the term used to describe a proliferation of marine microalgae (dinoflagellates, diatoms) that carries a toxic effect and, in many cases, economic losses. The microalgae are usually present in the plankton in low concentrations, but under some envi- ronmental conditions they can rapidly multiply in dense blooms. These microalgae produce phycotoxins, which accumulate in the digestive organs of filter-feeding shell- fish, zooplankton and herbivorous fishes and through the trophic chain can produce human intoxications. The toxins are generally considered to be secondary metabo- lites and therefore not essential to the basic metabolism and growth of the microorganism (Van Dolah 2000). These episodes previously regarded as sporadic and localised phenomena and restricted to certain geographic areas have started to show up as recurrent and ubiquitous in any coast of the world and sometimes cause great damage to the local fishing industry and also to the natural com- munities of marine organisms (Vieites and Cabado 2008). Different countries have established standards about the total amount of phycotoxins that can be present in seafood products and also about the testing methods for detecting these molecules. Legal requirements for paralytic shellfish toxins [usually named paralytic shell- fish poisoning (PSP)], amnesic shellfish toxins [amnesic shellfish poisoning (ASP)], lipophilic toxins, ciguatoxins (CTXs), brevetoxins or tetrodotoxins (TTXs) have been set around the world (Rodríguez-Velasco 2008). The Euro- pean legislation establishes a maximum level of toxins in bivalve molluscs, echinoderms, tunicates and marine gastropods (the whole body or any part edible separately) of 800 μg of PSP equivalent/kg, 20 mg of domoic acid (DA)/kg, 160 μg of okadaic acid (OA) equivalents/kg, 160 μg of azaspiracids (AZAs) equivalents/kg and 1 mg of yes- sotoxin equivalent/kg, and the total absence of TTX and CTX (European Commission 2004a) (see structures in Figure 1). To check compliance with the limits laid down, several methods including mouse bioassay (MBA), high- performance liquid chromatography (HPLC) with fluo- rescence (FL), ultraviolet (UV) or mass spectrometry (MS) detection, immune assays or functional assays have been proposed. However, not all these methods are officially accepted as a validation process is necessary (European Commission 2007). MBA has been for many years consid- ered as the reference monitoring method for phycotoxins, although the European Commission established that to be in accordance with animal health protection, all possible replacement, refinement and reduction of animals must be taken into account when laboratory animal methods are
20

Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

Dec 29, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

DOI 10.1515/revac-2012-0020      Rev Anal Chem 2013; 32(1): 15–34

Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo Alfonso and Luis M. Botana *

Current situation on analysis of marine toxins Abstract: Marine toxins are a food safety concern world-

wide. This review discusses current analytical methods

for those toxins that are legally regulated in Europe,

namely domoic acid, saxitoxin, okadaic acid, yessotoxin,

pectenotoxin and azaspiracids, and all their analogues,

and those that are not regularly monitored but are a threat

to humans, such as tetrodotoxin, ciguatoxins and cyclic

imines. Because of legislative changes that were imple-

mented in 2011, most legally required methods are chro-

matographic. Saxitoxin and domoic acid are monitored

by high-performance liquid chromatography with fluo-

rescent and ultraviolet detection, respectively. All other

toxins are monitored nowadays by means of chromato-

graphic separation and mass spectrometry detection.

Keywords: analytical standards; chromatography; marine

toxins; mass spectrometry.

*Corresponding author: Luis M. Botana, Department of

Pharmacology, Veterinary School , Faculty Veterinaria, Universidad

de Santiago de Compostela, Campus de Lugo, Lugo 27002 , Spain ,

e-mail: [email protected]

Ana M. Botana: Department of Analytical Chemistry , Universidad de

Santiago de Compostela, Campus de Lugo, Lugo 27002 , Spain

Paz Otero: Department of Pharmacology, Veterinary School , Faculty

Veterinaria, Universidad de Santiago de Compostela, Campus de

Lugo, Lugo 27002 , Spain

Paula Rodr í guez: Department of Pharmacology, Veterinary School ,

Faculty Veterinaria, Universidad de Santiago de Compostela,

Campus de Lugo, Lugo 27002 , Spain

Amparo Alfonso: Department of Pharmacology, Veterinary School ,

Faculty Veterinaria, Universidad de Santiago de Compostela,

Campus de Lugo, Lugo 27002 , Spain

Introduction Phycotoxins are an acute worldwide seafood safety

concern; hence, their detection and quantification in food

with reliable methods is important. The methods must be

validated according to international guidelines to ensure

adequate results and to protect public health and reduce

their economic impact.

Harmful algal blooms is the term used to describe

a proliferation of marine microalgae (dinoflagellates,

diatoms) that carries a toxic effect and, in many cases,

economic losses. The microalgae are usually present in

the plankton in low concentrations, but under some envi-

ronmental conditions they can rapidly multiply in dense

blooms. These microalgae produce phycotoxins, which

accumulate in the digestive organs of filter-feeding shell-

fish, zooplankton and herbivorous fishes and through

the trophic chain can produce human intoxications. The

toxins are generally considered to be secondary metabo-

lites and therefore not essential to the basic metabolism

and growth of the microorganism (Van Dolah 2000 ). These

episodes previously regarded as sporadic and localised

phenomena and restricted to certain geographic areas

have started to show up as recurrent and ubiquitous in

any coast of the world and sometimes cause great damage

to the local fishing industry and also to the natural com-

munities of marine organisms (Vieites and Cabado 2008 ).

Different countries have established standards about

the total amount of phycotoxins that can be present in

seafood products and also about the testing methods

for detecting these molecules. Legal requirements for

paralytic shellfish toxins [usually named paralytic shell-

fish poisoning (PSP)], amnesic shellfish toxins [amnesic

shellfish poisoning (ASP)], lipophilic toxins, ciguatoxins

(CTXs), brevetoxins or tetrodotoxins (TTXs) have been set

around the world (Rodr í guez-Velasco 2008 ). The Euro-

pean legislation establishes a maximum level of toxins

in bivalve molluscs, echinoderms, tunicates and marine

gastropods (the whole body or any part edible separately)

of 800 μ g of PSP equivalent/kg, 20 mg of domoic acid

(DA)/kg, 160 μ g of okadaic acid (OA) equivalents/kg, 160

μ g of azaspiracids (AZAs) equivalents/kg and 1 mg of yes-

sotoxin equivalent/kg, and the total absence of TTX and

CTX (European Commission 2004a ) (see structures in

Figure 1 ). To check compliance with the limits laid down,

several methods including mouse bioassay (MBA), high-

performance liquid chromatography (HPLC) with fluo-

rescence (FL), ultraviolet (UV) or mass spectrometry (MS)

detection, immune assays or functional assays have been

proposed. However, not all these methods are officially

accepted as a validation process is necessary (European

Commission 2007 ). MBA has been for many years consid-

ered as the reference monitoring method for phycotoxins,

although the European Commission established that to be

in accordance with animal health protection, all possible

replacement, refinement and reduction of animals must be

taken into account when laboratory animal methods are

Page 2: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

16      A.M. Botana et al.: Analysis of marine toxins

Palytoxin

Azaspiracid1

Saxitoxin

Ciguatoxin3C Okadaic acid

Pectenotoxin

Yessotoxin

N

NH

O

R1

+NH2

R3

R4

OH

HO

H

R2 H

H

HH

HO

O

H

109

2

1

5

7

4

Tetrodotoxin

Brevetoxin

Figure 1   Structures of representative toxins.

Page 3: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      17

used (Hess et al. 2006 ), and therefore alternative methods

should be applied. Any method proposed as an alternative

to the biological assay should not be less effective than

the biological method, and the implementation should

provide an equivalent level of public health protection.

Following these premises, several methods were validated

as alternatives to the MBA (Botana et al. 2009 , 2010, Camp-

bell et al. 2011 ). In the case of PSP toxins, the total content

of toxins in the edible parts of molluscs must be officially

detected by MBA (European Commission 2005 ) and also by

the HPLC Lawrence method with pre-column oxidation as

published in the Association of Official Analytical Chemi-

sts Official Method2205.06 (European Commission 2006 ).

If the results are challenged, the reference method shall be

the biological method. To detect PSP toxins, other analyti-

cal non-validated methods are available, such as an HPLC

method with post-column oxidation (Oshima et  al. 1976 ,

Rodriguez et  al. 2010 ), plasmon resonance biosensors

(Fonfria et  al. 2007 ) or enzyme immunoassays [enzyme-

linked immunoassay (ELISA)] (Huang et  al. 1996 , Garet

et al. 2010 ), based on both the use of antibodies and several

functional assays based on the mechanism of action of the

toxins (Vieytes et al. 1993 , Louzao et al. 2001 ). In the case of

ASP, the total content of toxins in edible parts of molluscs

may be detected by using either an HPLC method with UV

detection (HPLC-UV) or an ELISA method (European Com-

mission 2007 ). In this case, if results are challenged, the

reference method shall be the HPLC method. For the detec-

tion of lipophilic toxins, a series of MBA procedures, dif-

fering in the test portion of flesh and the solvents used for

extraction, were the official method for many years (Euro-

pean Commission 2005 ); however, after July 2011 a liquid

chromatography (LC) tandem MS (LC-MS/MS) method was

approved to be applied as the reference method for the

detection of these toxins (Commission Regulation 2011 ). In

addition to this, several methods based on the mechanism

of action of each lipophilic toxin group have been devel-

oped (Vieytes et al. 1997 , Alfonso et al. 2004 , Vilarino et al.

2009 , Otero et al. 2011b ). There are no official methods to

detect the presence of TTX or CTX even though several

LC-MS/MS methods can be used to detect these toxins

(Otero et al. 2010 , Rodr í guez et al. 2012 ).

As was mentioned earlier, the presence of phycotox-

ins is a dynamic and ubiquitous process in any coast of

the world and therefore the presence of new analogues,

new toxins or toxins from different locations are issues

that must be taken into account when any analytical

method is used. However, the legislation also established

that in addition to the amount of toxin, the total toxicity,

which is calculated by using conversion factors based

on the available toxicity data of each toxin, should be

calculated (Commission Regulation 2011 ). In this sense,

the toxicity equivalent factor (TEF) of each compound

compared with the potency of the reference compound

must be applied to calculate the total toxicity of a sample

(Botana et al. 2010 ). The Working Group on marine bio-

toxins, as part of the Contaminants Panel at the European

Food Safety Agency (EFSA), has defined, on the basis of

current knowledge, the TEF that should be used for the

conversion of analytical results into toxic concentrations

in each toxin group (European Commission Panel 2009b ).

Another essential item to implement and to validate any

chemical detection method is the availability of standards

as the identification of one toxin by using other analogues

can lead to gross errors (Otero et al. 2011a ). In addition,

pure toxins in high amount are necessary to calculate the

toxicity and TEF of each molecule to achieve results to

protect human health. It can be concluded that analytical

methods are useful tools to quantify the amount of known

marine toxins in seafood samples, but several important

details such as toxicity, calibrants and new toxins should

be considered.

Analysis of PSP toxins PSP is a fatal human syndrome produced by a naturally

occurring group of neurotoxic alkaloids present in several

species of dinoflagellates. This group of toxins constitutes

the most spread worldwide affecting the whole of Europe,

South Africa, India, Morocco, eastern Asian coast and

North and South America as well.

Saxitoxin (STX) is the first described and most studied

compound of the group (it is the representative com-

pound), and 57 other analogues also have been researched

(Wiese et al. 2010 ). Traditionally, STX analogues have been

composed only of hydrophilic compounds and divided

into subgroups on the basis of substituent side chains:

carbamate [STX, neo-STX and gonyautoxins (GTX1 – 4)],

decarbamoyl (dc-STX, dc-neo-STX and dc-GTX1 – 4) and N -

sulfocarbamoyl (GTX5 – 6 and C1 – 4). These groups present

different toxicities, with the carbamoyl analogues being

the most toxic, followed by decarbamoyl analogues with

intermediate toxicity, and N -sulfocarbamoyl analogues

the least toxic (Vale et  al. 2008a,b ). Figure 1 shows the

chemical structure of the more often found compounds in

toxic dynoflagellates and poisoned seafood.

More recently, analogues from a novel family of PSP

toxins containing a hydrophobic side chain were isolated

and structurally characterised from Australian strains of

Gymnodinium catenatum and designated GC1-GC3 (Negri

Page 4: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

18      A.M. Botana et al.: Analysis of marine toxins

et al. 2003 ). Other hydrophilic analogues of STX with an

acetate R 4 side chain were reported in the freshwater fila-

mentous cyanobacterium Lyngbya wollei but not in the

marine environment so far (Onodera et al. 1997 ).

PSP toxins specifically block the excitation current in

nerve and muscle cells by means of site one of the sodium

channel (Messner and Catterall 1986 ); therefore, the accu-

mulation of PSP toxins in shellfish creates a serious public

health problem and affects the fisheries industry. Sommer

and Meyer (1937) were the first to develop a method to

determine PSP toxins: their MBA method established the

basis for that. This is the reference method internationally

accepted to quantify PSP toxicity, and it is used worldwide

in monitoring programmes. The chemicals methods used

to determine PSP toxins are fluorimetric assays, HPLC

with fluorimetric detection (either pre-column or post-

column oxidation), LC/MS and capillary electrophoresis

(CE) methods.

The HPLC methods are widely used to quantify PSP

toxins present in seafood samples, and they are also

useful to provide the PSP profile because chromatographic

methods are identification methods as well. These toxins

have only a weak chromophore group, and it must be

modified before detection: when they are oxidised in alka-

line solution, a purine is formed that becomes fluorescent

at acidic pH. This reaction can be either a pre-column one

or a post-column one, and obtained purines are monito-

rised with an FL detector.

Bates and Rapoport (1975) first studied the oxidative

alkaline conditions of PSP toxins to get fluorescent com-

pounds; then, Buckley et  al. (1978) added them to their

method and established the basis of the HPLC method for

these compounds with the post-column reaction.

In 1984, Oshima et  al. (1984) proposed a method

based on alkaline oxidation to produce highly fluores-

cent derivatives, but with problems in separating toxins

such as GTX1, GTX4 and GTX3, GTX5. Afterwards, they

described a method that was able to separate almost all

the PSP toxins with three different eluents according to

the basicity of the three groups of toxins (group I: C1 – C4;

group II: GTX1,4, GTX5 (B1) and GTX6 (B2), dc-GTX1,4;

group III: NEO, dc-STX and STX) (Oshima et  al. 1987 ).

In a later work, Oshima (1995) improved the system and

included an extract cleaning procedure. The first dis-

advantage is that it is a time-consuming method for the

analysis of PSP toxins.

In the group of pre-column methods, the method

developed by Lawrence and Menard (1991) needs to be

highlighted. One of the biggest disadvantages was the ina-

bility to easily distinguish between different toxin groups

with similar toxicity.

Following this, the method was modified by chang-

ing chromatographic conditions to reduce analysis time

and improve performance (Lawrence et al. 1995 ). In 2005,

the Lawrence method was adopted as the official method

to detect PSP toxins and then approved by the European

Union (EU) for monitoring these toxins (Association of

Official Analytical Chemists 2005, European Commis-

sion 2006 ). It is based on the pre-column oxidation of PSP

toxins with hydrogen peroxide and sodium periodate fol-

lowed by fluorimetric detection. It was validated for the

determination of STX, NEO, GTX2,3, GTX1,4, dc-STX, GTX5

(B1), C1,2 and C3,4 in molluscs (mussels, clams, oysters

and scallops). In 2009 a validation study was under-

taken for the extension of this method to two more toxins:

dc-NEO and dc-GTX2,3 (Turner et al. 2009 ).

Pre- and post-column HPLC methods, despite the

many benefits of each, which includes an increased sen-

sitivity to low concentrations of toxins and less variability

in results, also present some drawbacks that should be

resolved. In the case of hydrophobic analogues, they are

retained by C18 resins (Negri et al. 2003 ), because of which

the use of HPLC methods in monitoring programmes will

miss the presence of hydrophobic analogues (Vale 2008 ,

Vale et al. 2009 ). LC-MS methods are also being developed

to obtain a good characterisation of these compounds:

Electrospray ionisation MS (ESI-MS) is effective for the

detection of polar PSP toxins (Quilliam 2003 , Dell ’ Aversano

et  al. 2005 ); however, variable retention times for PSP

toxins in different seafood matrixes were observed. At

present, it is not possible to achieve a complete separa-

tion of all PSP toxins, and the sensitivity of MS detection is

not good enough to control seafood toxin at the regulatory

limit established (Diener et al. 2007 ).

Although the analysis of PSP toxins provides a profile

of the composition of a given toxic episode, the use of

surface plasmon resonance biosensors has reached a con-

siderable level of development (Campbell et al. 2007 ). The

high sensitivity of this approach allows fast throughput,

as sample extraction and detection is rather simple, plus

the chips can be used many times (Fonfria et  al. 2007 ).

For this techonology, an antibody-based method has been

described and validated both at the single laboratory level

(Campbell et  al. 2010 ) and in an interlabotoratory study

(van den Top et al. 2011b ).

Analysis of DA DA, belonging to the kainoid class of compounds, is the

chemical responsible for ASP. It was originally isolated as

Page 5: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      19

a product from the red algae Chondria armata and subse-

quently isolated from other red algae and several species of

diatoms, mainly of the genus Pseudonitzschia (Bates et al.

1989 , Lundholm et  al. 1994 ). DA has also been detected

in several species of molluscs worldwide including the

United States, New Zealand, Mexico, several European

countries such as France, Portugal, Ireland and Spain and

the Croatian coast (Horner et al. 1993 , M í guez et al. 1996 ,

Vale, 2001, Ujevic et al. 2010 ). Although the DA is the main

toxin found in a variety of shellfish species, other minor

analogues, about 10 isomers of DA (isodomoic acids A – H

and DA 5 ′ diasteriomer), have been identified in marine

samples (Wright et al. 1990 , Zaman et al. 1997 ).

The first method developed to detect the compound

was LC-UV (Lawrence et al. 1989 ), and a protocol involv-

ing aqueous methanol extraction and strong anion-

exchange (SAX) clean-up has been applied extensively to

the analysis of DA and its known isomers in shellfish and

fish tissues on a regulatory basis. Nowadays, the LC-UV

remains the most widely used method, and it has been

validated and standardised as the reference method for

DA quantification (International 2000 , European Com-

mittee for Standardization 2003 ). Furthermore, ELISA

has also been validated (Kleivdal et al. 2007 ) and is used

officially in the EU for screening purposes. These methods

have limits of detection (LODs) low enough to detect DA at

a concentration of 4.5 mg/kg of shellfish meat. This dose

was established as the maximum amount that a portion of

400 g of meat should contain for not exceeding the acute

reference dose of 30 mg DA/kg body weight (European

Commission Panel 2009a ).

Several other biological methods have been deve-

loped to detect and quantify DA in seawater and tissue

samples, such as electrochemical ELISA (Micheli et  al.

2004 ), radioimmunoassay (Lawrence et  al. 1994 ), recep-

tor binging assay (Lefebvre et al. 2010 ), cytotoxicity assay

(Beani et al. 2000 , Leira et al. 2003 ) and surface plasmon

resonance (Le Berre and Kane 2006 , Traynor et al. 2006 ,

Stevens et  al. 2007 ). Some of these techniques, such as

enzyme immunoassay and ELISA, sometimes may require

little clean-up of shellfish samples and have the advan-

tages of being easy and fast (Yu et  al. 2004 ). However,

the cross-reactivity with similar toxins will result in false

positives and limit the demonstration of toxicity (He et al.

2010 ).

DA contains a characteristic conjugated diene chromo-

phore with strong absorbance at 242 nm, which permits

its detection by LC-UV at concentrations as low as 4 – 80

ng/ml, depending on the sensitivity of the detector (Mafra

et  al. 2009 ). This is suitable for regulatory purposes;

however, false positives are frequently encountered

because of interferences from crude extracts (Hess et  al.

2001 ). Mainly, tryptophan and its derivatives, normally

contained in shellfish and finfish tissues, may be eluted

close to DA with some columns and chromatographic con-

ditions. Thus, there are other alternatives, such as FL (James

et  al. 2000 , Chan et  al. 2007 ) and chemilumine scence

detection (Kodamatani et  al. 2004 ). As DA concentra-

tions in Pseudonitzschia cultures and phytoplankton field

samples are often much lower, a more sensitive method

of detection is required. The most common technique for

the ana lysis of DA in seawater samples uses a derivatising

agent, which results in low LODs (Pocklington et al. 1990 ),

but this procedure involves some problems. Among them,

a poor selectivity because many compounds in the sample,

such as amino acids, can be derivatised and interfere with

DA detection. Also, the handling during the derivatisation

step is time consuming for the use of LC-FL on a large scale.

In addition to the LC-UV and LC-FL, various extraction and

screening techniques, such as thin-layer chromatography

(Quilliam et al. 1998 ), CE (Gago -Martinez et al. 2003 ), gas

chromatography coupled to MS (GC-MS) (Pleasance et al.

1990 ), LC-MS/MS (Wang et al. 2007 , Mafra et al. 2009 ) and

ultraperformance liquid chromatography with tandem MS

detection (UPLC-MS/MS) (de la Iglesia et  al. 2008 ), have

allowed the detection of novel compounds inside of the DA

group in new coastal areas of the world.

As the seafood matrix can interfere in the detection

of trace levels, pre-treatment steps to concentrate and

purify DA are needed. Solid-phase extraction (SPE) is the

most common method used for the clean-up of shellfish

samples before using a chromatography or other analyti-

cal technique to quantify the toxin amount (Zhao et  al.

1997 , Quilliam et al. 1998 , James et al. 2000 , Pardo et al.

2007 , He et al. 2010 ]. So, several methods have been based

on SAX and strong cation exchange (Pleasance et al. 1990 ,

Furey et  al. 2001 , Gago -Martinez et  al. 2003 , Ciminiello

et al. 2005 ). The purification with SPE cartridges results in

a valid approach for the routine monitoring of DA in shell-

fish to prevent the matrix effect and signal suppression.

However, this is a manual time-consuming procedure;

even some authors concluded that an additional step

with an SPE cartridge did not significantly improve the

recovery of DA from shellfish samples (Powell et al. 2002 ).

Therefore, there are many analytical methods that do use

a sample clean-up and could be avoided when the pH of

the mobile phase in the LC methods is properly adjusted

(He et  al. 2010 ). Table 1 summarises several chemical

methods, specifying the sample clean-up procedure in

different matrices and the LOD obtained in each method.

MS techniques are being increasingly used in some

laboratories, and today, LC-MS/MS is perhaps the most

Page 6: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

20      A.M. Botana et al.: Analysis of marine toxins

Method Matrices Treatment sample (clean-up) LODs References

LC-UV Mussels Molecularly imprinted polymer-

solid-phase extraction (MISPE)

0.1 mg/ml (Ciminiello et al. 2005)

LC-UV Fur seals (fluidics) SPE 8 ng/ml (European Commission Panel

2009a )

UPLC-MS/

MS

< 1 ng/ml

LC-UV Phytoplankton, seawater

samples

Without sample pre-

concentration and derivatisation

42 pg/ml

(on column)

(Traynor et al. 2006 )

LC-ESI-MS 15 pg/ml

LC-FL ∼ 15 pg/ml

LC-FL Mussels SPE and post-column

derivatisation

25 ng/ml (Powell et al. 2002 )

LC-FL Shellfish and phytoplankton SPE and pre-column

derivatisation (shellfish) Without

SPE (phytoplankton)

  ≤  1 ng/ml (He et al. 2010 )

UPLC-MS/

MS

Seawater SPE-C18 (with Empore disks) 0.02 ng/ml (Gago -Martinez et al. 2003 )

UPLC-MS/

MS

Crude extracts Without clean-up 0.05 – 0.09 mg/kg (McNabb et al. 2005 )

LC-MS/MS Seawater and phytoplankton SPE-C18 cartridges 30 pg/ml (Quilliam et al. 1998 )

LC-MS/MS Mussels, clams, cockles Pressurised liquid extraction

(PLC) with purification inside the

extraction cell

0.2 μ g/ml (Wang et al. 2007 )

LC-UV Mussels and scallops Without SPE clean-up 25 ng/ml (Doucette et al. 2009 )

LC-ESI-MS 0.008 μ g/ml

LC-UV-CLD Mussels Without any pre-concentration or

derivatisation steps

8 pg/ml (Hess et al. 2001 )

LC-MS/MS Seawater, phytoplankton,

mammalian fluids and tissues

SPE-C18 cartridges without pre-

concentration

1 – 4 pg/ml

(on column)

(Mosher et al. 1965 )

TLC Mussels SAX clean-up ∼ 10 μ g/g (Miyazawa and Noguchi 2001 )

GC-MS Mussels SPE clean-up: reversed phase

and SCX cartridges

1 μ g/ml (Pocklington et al. 1990 )

LC-UV Clams and scallops SAX cartridges 0.47 μ g/ml (Kodamatani et al. 2004 )

CE-EIA Mussels, oysters, clams and

scallops

– 0.02 ng/ml (Noguchi and Arakawa 2008 )

CE Mussels, razor clams and

anchovy

SAX clean-up 0.15 μ g/g (Pleasance et al. 1990 )

Table 1   Some representative analytical chemical methods for the detection of DA in a range of matrices.

CE, capillary electrophoresis; CE-EIA, capillary electrophoresis-enzyme immunoassay; DA, domoic acid; EIA, enzyme immunoassay;

GC-MS, gas chromatography-mass spectrometry; LC-ESI-MS, liquid chromatography-enzyme immunoassay-tandem mass spectrometry;

LC-FL, liquid chromatography with fluorescence detection; LC-MS/MS, liquid chromatography with tandem mass spectrometry; LC-UV, liquid

chromatography with ultraviolet detection; LC-UV-CLD, liquid chromatography-ultraviolet-chemiluminescence detection; LODs, limits

of detection; SAX, strong anion exchange; SCX, strong cation exchange; UPLC-MS/MS, ultraperformance liquid chromatography with

tandem mass spectrometry; TLC, thin layer chromatography.

important and legally accepted confirmatory tool, provid-

ing a high sensitivity, accuracy and selectivity for DA and

its isomers in crude extracts (McNabb et al. 2005 ). Nev-

ertheless, LC-UV is often the only analytical instrument

available in many research institutes and regulatory agen-

cies responsible for monitoring the occurrence of marine

toxins. Although the LOD of DA can be improved by using

different injection volumes in the LC-UV, the matrix com-

plexity may result in some degree of uncertainty about

the peak ’ s identity at low DA levels ( < 3 ng/ml), when a

sensible UV spectrum cannot be obtained (Mafra et  al.

2009 ). In this case, confirmation by LC-MS is sometimes

necessary and should also be required whenever DA is

reported in a new geographical area. Some studies on

algal production of DA to track the toxin in seawater, as an

early alert for toxin accumulation in marine organisms,

are available for remote, subsurface detection (Doucette

et al. 2009 ).

Page 7: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      21

Overall, an improvement in techniques based on

LC and/or MS to achieve reliable, simple and low-cost

methods is essential for DA detection in any sample

matrix.

Analysis of TTX TTX, a well-known potent neurotoxin, was first isolated

from puffer fish (Mosher et  al. 1965 ) and has also been

found in a wide variety of animals including arthropods,

echinoderms, mollusks, worms, newts or frogs (Miyazawa

and Noguchi 2001 , Noguchi and Arakawa 2008 ). Neither

its biochemical path nor its true origin is fully clarified, as

three hypotheses point to its origin: endogenous (Lehman

et  al. 2004 ), through food chain (Lin and Hwang 2001 ,

Kono et al. 2008 ) or through symbionts (Narita et al. 1987 ,

Wang et al. 2008 ).

Although TTX exists mainly in tropical waters

worldwide, this toxin has appeared on European coasts

(Katikou et al. 2009 ). It is possibly due to the migration of

toxic species from the Red Sea to the Mediterranean Sea

through the Suez Canal, a phenomenon known as Lessep-

sian migration (Golani 1998 , Galil and Zenetos 2002 ).

This migration may happen because of the opening of

new corridors allied to the increase in water temperature

as a result of climate change. So, marine species typical

of tropical and subtropical waters are adapted to waters

less warm, and changes in environmental conditions

lead to poisoning incidents in new geographical areas. In

fact, recent intoxication cases caused by the consumption

of TTX bearers have been described in the Israeli coast

(puffer fish: Lagocephalus sceleratus ) and in Spain (gastro-

pod: Charonia lampas ) (Zaki and Mossa 2005 , Bentur et al.

2008 , Rodriguez et  al. 2008 ). This last episode was the

first report of TTX occurrence in autochthonous species in

Atlantic and Mediterranean waters. It triggered an investi-

gation monitoring different invertebrate species in several

sites of Portuguese coast, where low amounts of TTXs were

found in two intertidal gastropod species (Silva et al. 2012 ).

As a consequence of the migration of TTX bearers, interna-

tional fisheries agreements in the EU could be modified.

At the moment, the importation of puffer fish and other

toxic species is not permitted in some countries, including

the United States and Europe, and no regulatory limits for

TTX and its analogues have been established yet (Gessner

and McLaughlin 2008 ). With regard to current EU legis-

lation, toxic fish belonging to the family Tetraodontidae

or their products should not be placed on the European

markets (European Commission 2004b ). However, there

are regulations for TTX in countries such as Japan and

Korea, where official control does not regulate the amount

of toxin in fish that can be placed on the market, but res-

taurants that want to serve species containing TTX require

special licenses. EFSA has not published any documents

related to the risks involving TTX, but some authors claim

that the minimum lethal dose in humans is 2 mg, though

it may vary with factors such as age, health and sensitivity

to the toxin (Noguchi and Ebesu 2001 , Cohen et al. 2009 ).

Currently, there is no official method for TTX detection,

but the MBA has been used in many cases to determine

TTX toxicity in food matrices (Kawabata 1978 , Yasumoto

1991 ). However, the MBA is not completely satisfactory,

due to its low sensibility. MBA is not suitable for meas-

uring TTX in biological samples as it cannot distinguish

between TTX and STX or TTX analogues. Other biological

methods have been developed for TTX detection, such as

ELISA, methods that use antibodies specific to the toxin

(Raybould et al. 1992 , Neagu et al. 2006 , Zhou et al. 2007 ),

tests with cultured neuroblastoma, hemolytic assays

(Hamasaki et al. 1996 ) and also techniques using biosen-

sors (Kreuzer et al. 2002 , Neagu et al. 2006 , Yakes et al.

2011 ). Nevertheless, to obtain specific information of a

sample, as the toxic profile or the amount of an individual

toxin, chemical methods have been developed, including

immunoaffinity chromatography (Kawatsu et  al. 1999 ),

GC-MS (Man et al. 2010 ), HPLC with FL detection (HPLC-

FL) ( O ’ Leary et  al. 2004 , Jen et  al. 2007 ) and HPLC-UV

(Yu et  al. 2010 ). Although these techniques ensure low

LODs and high sensitivity to detect TTX, they have some

limitations. In the case of immunoaffinity or ELISA, both

methods require a costly monoclonal antibody or reagent

(Leung et  al. 2011 ), and the GC-MS requires a complex

extraction procedure (Jen et al. 2008 ). However, HPLC-FL

shows differences in the FL intensities of some TTX ana-

logues compared with TTX itself, which causes problems

in quantifying (Shoji et al. 2001 ). For these reasons, ana-

lytical methods based on LC-MS and LC-MS/MS have been

developed (Jang et  al. 2010 , Chen et  al. 2011 , Rodr í guez

et  al. 2012 ). These LC methods, together with appropri-

ate extraction procedures, have been able to determine

the presence of TTX, not only in the remains of fish and

other animals but also in the blood and urine of poisoned

patients (Tsai et  al. 2006 , Rodriguez et  al. 2008 ). More-

over, these methods provide better LODs when compared

with HPLC-FL or UV (Jen et al. 2008 ). Most of the LC-MS

instruments are equipped with an atmospheric pressured

ionisation (API) fitted with an ESI source based on colli-

sion-induced dissociation (CID) of a triple quadrupole

mass analyser. For the detection of TTXs, the ESI interface

operates in the positive ionisation mode. So, TTX and its

Page 8: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

22      A.M. Botana et al.: Analysis of marine toxins

analogues are analysed by MS, in which positive ionisa-

tion produces a typical molecular ion of [M + H] + for each

one. Because of the continuous emergence of new TTX

analogues, the most recent 8-epi-type TTX analogues

found in newts (Kudo et al. 2012 ), some authors prefer to

operate with the MS in the multiple reaction monitoring

(MRM) mode to quantify the individual toxins. However,

others use the MS operating in the single ion monitoring

mode or the single ion recording mode, such that only a

selected m/z value is detected in the analysis. On the con-

trary, the MRM mode not only allows detecting the precur-

sor molecule but also tracks multiple product ions from

the fragmentation pattern in the same run. In this case,

only one product ion is used in the quantification and the

others to confirm the toxin.

Today, the investigation is focused in developing

LC-MS methods that lead to a good separation and iden-

tification of all TTX analogues in any sample with low

LODs. So, different analytical columns and LC condi-

tions are tested by researchers to improve the detection

of TTX. Table 2 summarises several LC-MS/MS methods

to detect TTX and its analogues. The toxin separation is

typically achieved in reverse-phase columns and with

solvents containing an ion pair reagent (i.e., ammonium

heptafluorobutyrate). But this ion pair sometimes tends

to remain in the MS, causing spectral noise (Shoji et al.

2001 , Rodr í guez et  al. 2012 ). Also, because of the pola-

rity of TTXs, some of them are not well retained in these

columns. To solve all these problems, the hydrophilic

interaction liquid chromatography (HILIC) is becoming

more useful in the analysis of TTXs (Chen et  al. 2011 ,

Chulanetra et al. 2011 , Cho et al. 2012 , Kudo et al. 2012 ,

Rodr í guez et al. 2012 , Silva et al. 2012 ). The HILIC tech-

nique employs low aqueous/high polar organic solvents

without ion pair reagents, resulting in increased sensitiv-

ity compared with the reverse phase. So, TTX, which is

the more polar compound between its analogues, elutes

with the higher proportion of water in a gradient elution.

Also, problems with noise peaks are reduced in HILIC

columns. Almost all TTX analogues show a common

major product ion, which is usually 162 (m/z) formed

from the molecular ions in MS/MS, suggesting that this

product ion can be used in the quantification. However,

there are analogues such as TTX and 4-epi-TTX that have

the same molecular weight and the same fragmentation

pattern; therefore, a complete separation is needed to

quantify them. As a result, the HILIC separation system

is very useful for the simultaneous quantification of

TTXs by LC-MS/MS in the MRM mode. Figure 2 shows an

example of a chromatogram obtained by LC-ESI-CID-MS/

MS from puffer fish sample. The TTX analogues eluted in

a HILIC column and detected under the MRM mode were

the following: 5,6,11-trideoxy-TTX (m/z 272 > 254/162),

5-deoxy-TTX, and 11-deoxy-TTX (m/z 304 > 286/176),

4,9-anhydro-TTX (m/z 302 > 256/162), 11-nor-TTX-6(R)-ol

and 11-nor-TTX-6(S)-ol (m/z 290 > 272/162),4-epi-TTX and

TTX (m/z 320 > 302/162).

In summary, the LC-MS/MS technology is the most

appropriate and chosen by some laboratories world-

wide for detecting TTXs. One problem is the difficulty in

obtaining purified TTX analogues for calibration. TTX is

frequently used as an internal standard for their quantifi-

cation, and this could lead to errors in the toxin amount.

Nevertheless, because of the timely appearance of TTX in

new geographical areas, monitoring programmes coupled

to a suitable and standardised analytical method should

be developed and for this, certified standards of TTXs

would be necessary.

Detection of lipophilic marine toxins Marine lipophilic toxins are a heterogeneous group of

toxins with different molecular weights and specific

chemical characteristics (Table 3 ). This group includes

yessotoxins (YTXs), AZAs, pectenotoxins, gymnodimines

(GYMs), spirolides (SPXs), CTXs and diarrhetic shellfish

poisoning (DSP). Members of the DSP toxin group are OA

and its derivatives dinophysistoxin-1 (DTX-1), dinophysis-

toxin-2 (DTX-2) and dinophysistoxin-3. To detect them, a

number of analytical methods have been developed in the

last decades, comprising MBAs, in vitro assays and chemi-

cal assays (Botana et al. 2012 ). Each method has its merits

and drawbacks, and ultimately it is the responsibility of

regulators and analysts to decide upon the most appropri-

ate technology for their particular situation. The regula-

tion of lipophilic marine toxins worldwide differs widely.

In Japan, Canada and South America, MBA is the method

used for the official control of these toxins, while in New

Zealand, chromatographic methods have been used for

their monitoring programmes (McNabb 2008 , Gerssen

et  al. 2011 ). In Europe, chromatographic techniques

coupled to MS were also the method of choice to replace

MBAs to detect lipophilic marine toxins (Commission Reg-

ulation 2011 ). MBA, which has been used for many years

as an exclusive reference method for lipohilic marine

toxin detection, was replaced by the LC-MS/MS approach

with the new regulation.

To date, more than 200 lipophilic marine toxins

have been described in the literature (Gerssen et  al.

Page 9: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      23

Separation Eluents Toxins [M + H] + (m/z) Matrices LODs References

Reverse

phase

30 m m ammonium heptafluorobutyrate in

1 m m ammonium acetate buffer (pH 5.0)

320, 336, 302, 290,

304 and unknown

analogues (348, 330)

Amphibian – (Stobo et al.

2005)

ZIC-HILIC (A) 10 m m ammonium formate and 10 m m

formic acid in water

(B) acetronitrile:water (80:20) in 5 m m

ammonium formate and 2 m m formic acid

320, 302, 304, 272 Puffer fish 0.09 – 0.2

ng/ml

(Villar -Gonzalez

et al. 2008 )

HILIC 16 m m ammonium formate/acetonitrile

(3:7 v/v)

256, 272, 288, 302,

304, 320, 336

Puffer fish 0.9 pmol/

injection

(Wang et al.

2008)

HILIC (A) 0.1 % formic acid/water and (B) 100 %

methanol

320 Blood serum 0.1 ng/ml (Jang et al. 2010)

Reverse

phase

1 % acetonitrile, 10 m m trimethylamine and

10 m m ammonia formate in water

320, 272 Gastropods and human

fluids (blood and urine)

< 10 ng/ml (Gessner and

McLaughlin 2008)

HILIC 16 m m ammonium formate/acetonitrile

(3:7 v/v)

320, 302, 304, 288,

272

Puffer fish < 0.5

nmol/g

(Cho et al. 2012)

ZIC-HILIC (A) 10 m m ammonium formate and 10 m m

formic acid in water

(B) acetronitrile:water (80:20) in 5 m m

ammonium formate and 2 m m formic acid

320, 302, 304 Puffer fish – (McNabb 2008)

HILIC 10 m m ammonioum formate/acetonitrile

(22:78 v/v)

272, 288, 290, 302,

304, 320, 336

Puffer fish 0.10 ng/ml (Kudo et al. 2012)

HILIC 1 % acetic acid in acetonitrile/1 % acetic

acid in water (88:12 v/v, pH 4.1)

320 Blood 0.32 ng/ml (Gerssen et al.

2011)

Reverse

phase

(A) 1 % acetonitrile, 20 m m

heptafluorobutyric acid, 20 m m ammonium

hydroxide and 10 m m ammonium formate

in water (B) and same mixture (A) with 5 %

acetonitrile

320, 290, 304, 302 Puffer fish – (Rodr í guez et al.

2012)

HILIC (A) 10 m m ammonium formate and 10 m m

formic acid in water

(B) acetronitrile:water (95:5) in 5 m m

ammonium formate and 2 m m formic acid

320, 290, 304, 302,

272

Puffer fish 16 ng/ml (Rodr í guez et al.

2012)

HILIC Not clear 254, 270, 272, 286,

288, 290, 302, 304,

320

Netws – (Botana et al.

2012)

HILIC (A) 10 m m ammonium formate and 10 m m

formic acid in water

(B) acetronitrile:water (95:5) in 5 m m

ammonium formate and 2 m m formic acid

320, 272 Gastropods 1.7 ng/ml (Cohen et al.

2009)

Table 2   Summary of several LC-MS/MS methods to detect TTX and its analogues.

HILIC, hydrophilic interaction liquid chromatography; LC-MS/MS, liquid chromatography with tandem mass spectrometry; LODs, limits of

detection; TTX, tetrodotoxin; ZIC-HILIC, Highly polar Zwitterionic hydrophilic interaction liquid chromatography.

2011 ). However, most LC-MS/MS methods are focused

on the analysis of the 13 toxins that are legislated in the

EU (Table 3). GYMs and SPXs are usually included in the

multi-toxin detection methods despite the fact that they

are not legislated in the EU. Hence, LC-MS/MS methods

detect different marine toxin groups in a short period of

time (Stobo et al. 2005 , McNabb 2008 , Gerssen et al. 2011 ),

and they are classified according to the chromatographic

solvents used. Basic or acid mobile phases can be used

with an elution gradient. Table 4 summarises the LC char-

acteristics of two types of chromatography developed

for the monitoring of lipophilic marine toxin. If an acid

chromatography is employed, the mobile phase is com-

posed of water and ACN/water (95:5), both containing

50 m m formic acid and 2 m m ammonium formate (Alfonso

et al. 2008 , Villar -Gonzalez et al. 2008 , Otero et al. 2010 ,

Blay et  al. 2011 ). Mobile phases for basic chromatogra-

phy are composed of water and ACN/water (90:10), both

containing 0.05 % ammonia, 2 m m bicarbonate or 6.7 m m

ammonium hydroxide (pH 11) (Gerssen et al. 2009 , 2011).

MRM data are detailed in Table 4. The transition with

the highest intensity is used for quantification, while the

Page 10: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

24      A.M. Botana et al.: Analysis of marine toxins

100 TIC of +MRM

Max. 4.5 x 105

1: 5-deoxyTTX2: 11-deoxyTTX3: 11-norTTX-6(R)-ol4: 4,9-anhydroTTX5: 11-norTTX-6(S)-ol6: 4-epiTTX7: TTX

5,6,11-trideoxyTTX

02 4 6 8 10 12 14

Time (min)16

14 6

7

5

3

2

18 20 22 24

Rel

ativ

e in

tens

ity (%

)

Figure 2   Mass chromatogram of the LC-ESI-CID-MS/MS obtained

under MRM operation from the puffer fish sample. Toxins eluted

in an XBridge Amide column (i.d. 2.1 × 150 mm; 3.5 μ m) at 25 ° C,

0.2 ml/min. The LC was operated in gradient with eluent A consisting

of 10 m m ammonium formate and 10 m m formic acid in water. Eluent

B contained 95 % acetonitrile and 5 % water with a final concentration

of 5 m m ammonium formate and 2 m m formic acid. Minutes 16 to

22 represent a broadening of the chromatogram where the toxins

are eluted. CID, collision-induced dissociation; ESI, electrospray

ionisation; i.d., inner diameter; LC, liquid chromatography; MRM,

multiple reaction monitoring; MS, mass spectrometry; TIC, total ion

chromatogram.

Toxin groups Chemical class Main compounds ( M r )

Formula Toxin analogues covered by the EU legislation

Okadaic acid (OA) Polyether, spiro-keto

assembly

OA (804) C 44

H 68

O 13

OA, DTX-1, DTX-2, DTX-3

Azaspiracid (AZA) Polyether, second amine,

3-spiro ring

AZA-1 (841.5) C 47

H 71

NO 12

AZA-1, AZA-2, AZA-3

Petecnotoxin (PTX) Polyeter, ester

macrocycle

PTX-1 (874.5) C 47

H 70

O 14

PTX-1, PTX-2

Yessotoxin (YTX) Ladder-shaped polyether YTX (1141) C 55

H 82

O 21

S 2 YTX, 45-OH-YTX, homo-YTX,

45-homo-YTX

Gymnodimines (GYMs) Macrocycle; cyclic imine GYM (507) C 32

H 45

NO 4 None

Spirolides (SPXs) Macrocycle; cyclic imine SPX-1 (691.5) C 41

H 61

NO 7 None

Table 3   Chemical characteristics of lipophilic marine toxins.

EU, European Union.

transition with the lowest intensity is used for confirma-

tory purposes. With the exception of OA and YTX toxins

groups, which are ionised in the negative mode, the

remaining lipophilic toxins are preferably ionised in the

positive mode. If the instrument is able to simultaneously

operate in positive and negative ionisation modes, the MS

methods include transitions for the toxins that are ionised

in both ionisation modes. If it is not capable of detecting

all toxins in a single injection, compounds are detected in

two separated runs, one with the equipment operating in

the positive mode and the other with the equipment oper-

ating in the negative mode.

This technique has been evaluated, and it is consid-

ered to be successful by many laboratories (Gerssen et al.

2009 , Blay et  al. 2011 ). The short, narrow-bore column

packed with 3 μ m Hypersil-BDS-C8 phase and the X-Bridge

C18 are two of the most widely used columns, which are

capable of separating a wide range of toxins by using rapid

gradient (Villar -Gonzalez et al. 2007 , Ciminiello et al. 2010 ,

Gerssen et al. 2011 ). The method with both basic and acid

mobile phases was also adjusted to the new technologies,

UPLC-MS/MS (Fux et al. 2007 , Rundberget et al. 2011 ), and

more than 20 analogues were separated in only 6.6 min

(Fux et al. 2007 ). One advantage of the acid against basic

chromatography is that the first one facilitates good sepa-

ration of acidic OA analogues by suppressing the ionisa-

tion of the carboxyl groups and preventing deleterious ion

exchange interactions with residual silanol groups in the

stationary phase (Suzuki and Quilliam 2011 ). Neverthe-

less, the chromatography of compounds included in the

YTX group can be problematic under acidic conditions

(Gerssen et al. 2009 ). LODs for lipophilic toxins achieved

by the LC-MS/MS approach are low (Table 4), and toxins

can be detected at levels below the current regulatory

limit. For both acid and basic chromatography conditions,

these LODs are lower for toxins ionised in the positive

mode than in the negative one, specially for SPXs (Blay

et al. 2011 , van den Top et al. 2011a ).

Although analytical methods are by far the target of

most of the international efforts for the detection of this

group of compounds, it should be highlighted that enzyme-

based assays are available for high-throughput detection

of these compounds (Vieytes et  al. 1997 , Gonz á lez et  al.

2002 , Rubiolo et al. 2012 ), and biosensor methods that use

the surface plasmon resonance technology (Llamas et al.

Page 11: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      25

Crom

atog

raph

y un

der a

cid

cond

ition

sCr

omat

ogra

phy

unde

r bas

ic co

nditi

ons

Co

lum

nB

DS

-Hyp

ers

il C

8,

50

mm

×2 m

m,

3 μ

m p

art

icle

siz

eX

-Bri

dg

e C

18

, 1

50

mm

×3 m

m,

5 μ

m p

art

icle

siz

e

Flo

w0

.2 m

l/m

in0

.25

–0

.4 m

l/m

in

Inje

ctio

n v

ol.

5 μ

l5

–1

0 μ

l

Co

lum

n T

25

°C2

5°C

–4

0°C

Mo

bil

e p

ha

se

A:

Wa

ter

(bo

th c

on

tain

ing

2 m

m a

mm

on

ium

fo

rma

te

A:

Wa

ter

(bo

th c

on

tain

ing

6.7

mm

am

mo

niu

m h

ydro

xid

e)

B:

AC

N (

95

%)

an

d 5

0 m

m f

orm

ic a

cid

) B

: A

CN

(9

0%

)(p

H 1

1)

Gra

die

nt

Tim

e (

min

)M

ob

ile

ph

as

e A

(%

)M

ob

ile

ph

as

e B

(%

)Ti

me

(m

in)

Mo

bil

e p

ha

se

A (

%)

Mo

bil

e p

ha

se

B (

%)

07

03

00

–1

90

10

81

09

01

01

09

0

11

10

90

13

10

90

11

.57

03

01

59

01

0

17

70

30

19

90

10

LOD

sO

A:

22

.1 p

g,

YTX

: 6

.1 p

g,

AZ

A-1

: 1

.1 p

g;

PTX

-2:

6.9

pg

; S

PX

-1:

1.9

 pg

; G

YM

: 1

4.1

pg

(Ge

rss

en

et

al.

20

09

)

OA

: 9

.1 p

g,

YTX

: 2

.2 p

g,

AZ

A-1

: 1

.1 p

g;

PTX

-2:

7.4

pg

; S

PX

-1:

0.8

pg

; G

YM

: 3

.7 p

g

(Ge

rss

en

et

al.

20

09

)

OA

: 1

0 μ

g/k

g,

AZ

A-1

: 0

.3 μ

g/k

g

(Kil

coyn

e,

20

10

#2

4)

OA

: 5

μg

/kg

, A

ZA

-1:

0.5

μg

/kg

(Kil

coyn

e,

20

10

)

OA

: 1

5 μ

g/k

g,

PTX

-2:

10

μg

/kg

, S

PX

-1:

4 μ

g/k

g

(Vil

lar-

Go

nza

lez

et 

al.

20

07

)

OA

: 3

μg

/kg

, D

TX-1

: 5

μg

/kg

; D

TX-2

: 3

μg

/kg

;

AZ

A-1

: 2

μg

/kg

; A

ZA

-2:

2 μ

g/k

g,

AZ

A-3

:

3 μ

g/k

g,

PTX

-2:

2 μ

g/k

g,

YTX

: 1

6 μ

g/k

g

(va

n d

en

To

p e

t a

l.

20

11

)

Tabl

e 4 

Ma

in c

ha

ract

eri

sti

cs o

f tw

o t

ype

s o

f li

qu

id c

hro

ma

tog

rap

hy

(LC

) fo

r th

e d

ete

ctio

n o

f li

po

hil

ic m

ari

ne

to

xin

s:

(1)

chro

ma

tog

rap

hy

un

de

r a

cid

ic c

on

dit

ion

s a

nd

(2

) ch

rom

ato

gra

ph

y

un

de

r b

as

ic c

on

dit

ion

s.

AC

N,

ace

ton

itri

le;

AZ

A,

aza

sp

ira

cid

; D

TX,

din

op

hys

isto

xin

; G

YM

, g

ymn

od

imin

e;

OA

, o

ka

da

ic a

cid

; P

TX,

pe

tecn

oto

xin

; S

PX

, s

pir

oli

de

; Y

TX,

yes

so

toxi

n.

aLi

mit

s o

f d

ete

ctio

n (

LOD

s)

of

se

vera

l

lip

op

hil

ic t

oxi

ns

are

als

o i

nd

ica

ted

on

it.

MS

/MS

de

tect

ion

wa

s p

erf

orm

ed

in

th

e m

ult

iple

re

act

ion

mo

nit

ori

ng

(M

RM

) m

od

e b

y u

sin

g t

wo

tra

ns

itio

ns

pe

r to

xin

: O

A a

nd

DTX

-2 (

m/z

80

3.5

/25

5.0

,

m/z

 80

3.5

/11

3.0

), D

TX-1

(m

/z 8

17

.5/2

55

.0,

m/z

81

7.5

/11

3.0

), Y

TX (

m/z

11

41

.5/1

06

1.7

, m

/z 1

14

1.5

/85

5.0

), 4

5-O

H-Y

TX (

m/z

11

57

.5/1

07

7.7

, m

/z 1

15

7.5

/87

1.5

), h

om

o-Y

TX

(m/z

11

55

.5/1

07

5.5

, m

/z 1

15

5.5

/86

9.5

), 4

5-O

H-h

om

o-Y

TX (

m/z

11

71

.5/1

09

1.5

, m

/z 1

17

1.5

/86

9.5

), P

TX-1

(m

/z 8

92

.5/8

21

.0,

m/z

89

2.5

/21

3.2

), P

TX-2

(m

/z 8

76

.5/8

23

.4,

m/z

87

6.5

/21

3.2

), A

ZA

-1 (

m/z

84

2.5

/82

4.5

, m

/z 8

42

.5/8

06

.5),

AZ

A-2

(m

/z 8

56

.5/8

38

.5,

m/z

85

6.5

/82

0.5

) a

nd

AZ

A-3

(m

/z 8

28

.5/8

10

.5,

m/z

82

8.5

/79

2.5

).

Page 12: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

26      A.M. Botana et al.: Analysis of marine toxins

2007 ), in combination with antibodies that match their

cross-reactivity to the relative toxicity of each analogue

(Stewart et al. 2009a,b ).

In general, methods based on LC-MS/MS are spe-

cific, have sufficient LODs and also have the possibility

for multi-toxin group detection in a single run. However,

one of the major problems found using this approach is

the standard availability. When toxins are not available,

the calibration curve of one toxin standard is often used

to quantify other toxins from the same group, assum-

ing a given ionisation conversion factor. This important

issue was checked by using the DSP toxin group (OA,

DTX-1 and DTX-2) (Otero et  al. 2011a ), and no compara-

ble results were obtained. When each one of the three

analogues was quantified by using the calibration

curve of the other ones, the amounts were increased

or decreased; that is, wrong results were obtained. The

highest variability was obtained when DTX-2 was quanti-

fied by using a DTX-1 calibration curve. In this case, the

overestimation was up to 204 % . When DTX-2 was quanti-

fied by using an OA calibration curve, the overestimation

was 88 % . This last quantification type is very usual in

many laboratories as the OA standard is more available

than DTX-2. Therefore, the use of quality standards or

certified reference materials of each one of the marine

toxins is essential for the exact quantification of these

products by MS, and all compounds for which standards

are not available or unknown should not be monitored

in the samples.

Analysis of CTX This toxin group is the only one that does accumulate

in fish of certain species, such as G. javanicus , Lutjanus bohar , Plectropomus leopardus and Epinephelus fuscogut-tatus (Caillaud et al. 2010 ), not in bivalves. CTXs are potent

polyether toxins issued from Gambierdiscus species of

dinoflagellates. These toxins are usually found in areas

between 35 ° N and 35 ° S latitude, mainly Indo-Pacific and

Caribbean areas. However, in recent years, these toxins

are increasingly appearing in countries not expected for

their latitude such as waters close to the European and

African continents (Perez -Arellano et al. 2005 , Bentur and

Spanier 2007 , Otero et al. 2010 ). Its distribution is expected

to shift towards other countries, and in fact this seems to

have begun the trend. For this reason, some laboratories

that did not normally include these compounds in the

toxin analysis are now in need of improving the methods

for detecting them.

Nowadays, the most used chemical method used for

detecting CTXs in shellfish samples is LC-MS/MS. Methods

based on HPLC-UV and HPLC-FL have been also used for

many years, but if they are compared with LC-MS/MS, it

is obvious that both have disadvantages. As CTXs do not

have characteristic chromophore groups in their struc-

tures (like DA), the use of HPLC-UV results in methods

with poor sensitivity and selectivity (Caillaud et al. 2010 ).

Despite this, several authors have used this technique as a

strategy for the isolation and purification of CTXs prior to

their characterisation in fish tissues and extracts of dino-

flagellates (Lewis and Jones 1997; Lewis and Vernoux 1998 ,

Satake et al. 1998 , Hamilton et al. 2002a,b ). However, the

presence of primary hydroxy groups in some CTXs (P-CTX-

1, P-CTX-2, P-CTX-3 and 2,3-dihydroxy-CTX-3C) suggested

that these toxins could be derivatised into fluorescent

esters and thus become suitable for HPLC-FL (Yasumoto

et al. 1995 ). These analyses are performed in a Develosil

ODS-5 column (4.6 × 250 mm) using an isocratic elution

gradient with around 90 % acetonitrile (Yasumoto et  al.

1995 ). This method was successfully applied to samples

of Gambierdiscus toxicus and to different carnivorous fish

species, including G. javanicus , Lutjanus bohar , Plectro-pomus leopardus and Epinephelus fuscoguttatus (Caillaud

et  al. 2010 ). However, this approach lacks applicability

as many CTX analogues such as P-CTX-3C do not exhibit

this primary hydroxy group, and therefore they cannot be

derivatised.

Later, the application of MS played a critical role in the

structure elucidation of many CTX congeners (Lewis and

Jones 1997 , Hamilton et al. 2002a,b , Pottier et al. 2002a ).

Today, LC-MS/MS technology is widely used by many

laboratories for their detection in contaminated samples.

This technique is not the reference method, as happens in

the case of other lipophilic toxins. Each laboratory uses

LC conditions that are considered more properly and thus,

the bibliography describes different methods of analy-

sis. Prior to the chromatographic analysis of CTXs in fish

samples, strict clean protocols are used (Hamilton et  al.

2002a,b , Pottier et al. 2002a , Otero et al. 2010 ). Although

these steps improve the sensitivity and reduce the matrix

effects, it is certain that they do not remove metallic impu-

rities, which result in the formation of adducts (Hamilton

et al. 2002a , Otero et al. 2010 ).

LC systems comprise C18 columns, such as 5- μ m

Phenomenex Luna (Lewis et al. 2009 ) or 3.5- μ m Zorbax

300SB (Hamilton et  al. 2002a,b ) and C8 columns such

as 3- μ m Phenomenex Hyperclone (Roeder et  al. 2010 )

or 3- μ m Phenomenex Luna (Dechraoui et  al. 2005 ).

Mobile phases are composed generally by acetonitrile

and water, and the use of gradients of these organic

Page 13: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      27

solvents buffered with 1 m m ammonium acetate improve

the separation and detection of CTXs compared with

an acetonitrile:water gradient modified with 0.1 % tri-

fluorocetic acid (Lewis and Jones 1997 ). MS detection

is usually performed in the positive MRM mode (Perez -

Arellano et al. 2005 , Roeder et al. 2010 ) or in the Q1 mode

selecting a range of mass (Hamilton et al. 2002a , Pottier

et al. 2002a ). In the last case, the MS spectrum will show

the characteristic pattern of ion formation for CTX poly-

ethers (multiple losses of water and the formation of

sodium and ammonium adducts) (Pottier et  al. 2002a ).

Each CTX gives rise to prominent ammonium, potassium

and sodium adducts and losses of waters. An example

of a typical chromatogram and spectrum of a CTX stand-

ard is shown in Figure 3 . The compound is the synthetic

51-hydroxy-CTX-3C with M r 1038.5, and the identification

was performed in a UPLC system coupled to an Xevo TQ

MS mass spectrometer from Waters (Manchester, UK).

The column used was a Waters Acquity UPLC BEH C 18

(100 × 2.1 mm; 1.7 μ m), and the mobile phase consisted of

a gradient of acetonitrile/water. As can be observed, the

ion 1039.5 m/z due to the [M + H] + is not the most promi-

nent. However, the ions m/z 1077.5 [M + K] + and m/z 1061.5

[M + Na] + have a high intensity. In addition, the spectrum

shows two water losses, m/z 1021.5, which corresponds

to [M + H-H 2 O], and m/z 1003.5, which corresponds to

[M + H-H 2 O].

Another consideration to be taken into account in the

analysis of CTXs by LC-MS/MS is the big number of ana-

logues belonging to this toxin group and the few stand-

ards available. To date, more than 50 different congeners

have been identified (Litaker et  al. 2010 ), and the pres-

ence of several CTXs with equal molecular weight in the

same extract is very frequent. Up to six different analogues

for the compound 1111.6 m/z (CTX-1B) eluting in different

retention times have been described (Lewis and Jones

1997 ). This issue makes the identification of CTX difficult.

Moreover, because of the lack of certified standards and

reference materials and the limited amounts of contami-

nated material available for method development, the vali-

dation status of LC-MS/MS methods is very restricted and

up to now no collaborative study has been undertaken.

Recently, a method with 14 reference toxins prepared

by either synthesis or isolation from natural sources was

used to identify and quantify 16 toxins from the CTXs

group (Yogi et  al. 2011 ). LC separation was performed in

a Zorbax Eclipse Plus C18 column in < 14 min by using

a linear gradient of mobile phases: 5 m m ammonium

formate and 0.1 % formic acid in water and in methanol.

The mass spectrometer operates in the positive mode

for the monitoring of sodium adduct ions [M + Na] + . This

method has greatly reduced the time of analysis as up to

now almost all methods described use run times of around

50 min ( Hamilton et al. 2002a,b, 2009 , Pottier et al. 2002b ,

100A

B 100[M+H - 2H2O]+

1003.5

[M+H - 2H2O]+

1021.5

1077.5 [M+H]+ MS2 ES+1.09e5

[M+H]+

1039.5

[M+Na]+

1061.5

%

%

XEVO-TQMS51-OH-CTX-3C

0

01000 1020 1040 1060 1080 1100 1120 1140 1160

2.00 4.00 6.00

5.801039.5

SIR of 1 channel ES+TIC

6.36e4

8.00 10.00 12.00Time (min)

m/z

Figure 3   Chromatogram (A) and mass spectrum (B) of 51-OH-CTX-3C ( M r 1038.5) standard at 1000 ng/ml, using selected ion recording

(SIR) UPLC mode of 1039.5 m/z [M + H] + . Analysis was performed in an ACQUITY UPLC system coupled with Xevo TQ MS mass spectrometer.

Chromatographic identification was achieved in a Waters Acquity UPLC BEH C 18

column (100 × 2.1 mm; 1.7 μ m). The mobile phase consisted

of two components: acetonitrile/water (95:5) (A) and water (B), both containing 50 m m formic acid and 2 m m ammonium formate using a

gradient elution. 51-OH-CTX-3C, 51-hydroxy-CTX-3C; UPLC, ultraperformance liquid chromatography.

Page 14: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

28      A.M. Botana et al.: Analysis of marine toxins

Dechraoui et al. 2005 , Roeder et al. 2010 ). This method pre-

sents limits of 0.25 pg for CTX detection. This means that

it can detect CTX-1B at 45 pg/g in fish flesh. However, an

obstacle to the widespread use and further validation of

this method is the difficulty in obtaining standard toxins.

In summary, the LC-MS/MS methodology has proven

to be extremely useful for identifying characteristic profiles

of CTXs in different strains of Gambierdiscus spp. (Rhodes

et al. 2010 , Roeder et al. 2010 ) and fish (Pottier et al. 2002a ,

Dechraoui et al. 2005 , Otero et al. 2010 ) worldwide. The LC

and MS methods and instruments have greatly improved

in recent years. Because of the high selectivity, accurate

mass spectra are obtained, susceptible to being compared

with spectra of known molecules or standard spectra in

the case they are available. These advances give rise to the

confirmation of many suspected cases of ciguateric fish

by selecting the transitions object study. Techniques such

as the UPLC-MS/MS improve sensitivity; however, at the

time of protecting the human health they are still far from

being used as a routine method in the laboratories for the

detection of CTX.

Conclusions Although there are many choices for the analysis and

detection of marine toxins, the current legal requirement

demands the use of LC-MS for most of the groups, with

the exception of the DA and saxitoxin groups, which can

be detected by HPLC. The chemical complexity of each

toxin group, with new analogues being added every year,

and their different profiles depending on the geographi-

cal area and the climate conditions highlight the impor-

tance of robust analytical methods and a good supply of

analytical standards. Along with this demand, it is very

important to continue with the development of high-

throughput methods that may provide quick results about

the presence of these very toxic compounds in food, as

food safety will always be a concern for industries that

deal with certain mollusks and certain types of fish.

Received June 7, 2012; accepted October 5, 2012; previously

published online November 23, 2012

References Alfonso, A.; Vieytes, M. R.; Yasumoto, T.; Botana, L. M. A rapid

microplate fluorescence method to detect yessotoxins based

on their capacity to activate phosphodiesterases. Anal. Biochem . 2004 , 326 , 93 – 99.

Alfonso, C.; Rehmann, N.; Hess, P.; Alfonso, A.; Wandscheer, C.

B.; Abuin, M.; Vale, C.; Otero, P.; Vieytes, M. R.; Botana, L. M.

Evaluation of various pH and temperature conditions on the

stability of Azaspiracids and their importance in preparative

isolation and toxicological studies. Anal. Chem. 2008 , 80, 9672 – 9680.

Association of Official Analytical Chemists. Official Methods of Analysis of the Association of Official Analytical Chemists Method 2005.06 ; First Action. MD, USA: AOAC, 2005.

Bates, H. A.; Rapoport, H. Chemical assay for saxitoxin, the paralytic

shellfish poison. J. Agr. Food Chem . 1975 , 23 , 237 – 239.

Bates, S. S.; Bird, C. J.; De Freitas, A. S. W.; Foxall, R. A.; Gilgan,

M.; Hanic, L. A.; Johnson, G. R.; McCulloch, A. W.; Odense, P.;

Pocklington, R.; Quilliam, M. A.; Sim, P. G.; Smith, J. C.; Subba

Rao, D. V.; Todd, E. C. D.; Walter, J. A.; Wright, J. L. C. Pennate

Diatom Nitzschia pungens as the Primary Source of Domoic

Acid, a Toxin in Shellfish from Eastern Prince Edward Island,

Canada. Can. J. Fish Aquat. Sci . 1989 , 46 , 1203 – 1215.

Beani, L.; Bianchi, C.; Guerrini, F.; Marani, L.; Pistocchi, R.;

Tomasini, M. C.; Ceredi, A.; Milandri, A.; Poletti, R.; Boni, L.

High sensitivity bioassay of paralytic (PSP) and amnesic (ASP)

algal toxins based on the fluorimetric detection of [Ca 2 + ] i in rat

cortical primary cultures. Toxicon . 2000 , 38 , 1283 – 1297.

Bentur, Y.; Spanier, E. Ciguatoxin-like substances in edible fish

on the eastern Mediterranean. Clin. Toxicol. (Phila ) 2007 , 45 ,

695 – 700.

Bentur, Y.; Ashkar, J.; Lurie, Y.; Levy, Y.; Azzam, Z. S.; Litmanovich, M.;

Golik, M.; Gurevych, B.; Golani, D.; Eisenman, A. Lessepsian

migration and tetrodotoxin poisoning due to Lagocephalus sceleratus in the eastern Mediterranean. Toxicon 2008 , 52, 964 – 968.

Blay, P.; Hui, J. P.; Chang, J.; Melanson, J. E. Screening for multiple

classes of marine biotoxins by liquid chromatography – high-

resolution mass spectrometry. Anal. Bioanal. Chem. 2011 , 400, 577 – 585.

Botana, L. M.; Alfonso, A.; Botana, A.; Vieytes, M. R.; Vale, C.;

Vilari ñ o, N.; Louzao, M. C. Functional assays for marine toxins

as an alternative, high-throughput-screening solution to

animal tests. Trends Anal. Chem. 2009 , 28 , 603 – 611.

Botana, L. M.; Vilari ñ o, N.; Elliott, C. T.; Campbell, K.; Alfonso, A.;

Vale, C.; Louzao, M. C.; Botana, A. M. The problem of toxicity

equivalent factors in developing alternative methods to animal

bioassays for marine-toxin detection. Trends Anal. Chem. 2010 , 29 , 1316 – 1325.

Botana, L. M.; Vieytes, M. R.; Botana, A. M.; Vale, C.; Vilari ñ o, N.

In: Cabado, A.G; Vieites, J. M., eds. New Trends in Marine and Freshwater Toxins . New York: Nova Science, Inc., 2012,

p. 203.

Buckley, L. J.; Oshima, Y.; Shimizu, Y. Construction of a paralytic

shellfish toxin analyzer and its application. Anal. Biochem .

1978 , 85 , 157 – 164.

Caillaud, A.; de la Iglesia, P.; Darius, H. T.; Pauillac, S.; Aligizaki, K.;

Fraga, S.; Chinain, M.; Diogene, J. Update on methodologies

available for ciguatoxin determination: perspectives to

confront the onset of ciguatera fish poisoning in Europe.

Mar. Drugs 2010 , 8 , 1838 – 1907.

Page 15: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      29

Campbell, K.; Stewart, L. D.; Doucette, G. J.; Fodey, T. L.; Haughey, S.

A.; Vilarino, N.; Kawatsu, K.; Elliott, C. T. Assessment of specific

binding proteins suitable for the detection of paralytic shellfish

poisons using optical biosensor technology. Anal. Chem . 2007 ,

79 , 5906 – 5914.

Campbell, K.; Haughey, S. A.; van den Top, H.; van Egmond, H.;

Vilarino, N.; Botana, L. M.; Elliott, C. T. Single laboratory

validation of a surface plasmon resonance biosensor screening

method for paralytic shellfish poisoning toxins. Anal. Chem .

2010 , 82 , 2977 – 2988.

Campbell, A.; Vilari ñ o, N.; Botana, L. M.; Elliott, C. T. A European

perspective on progress in moving away from the mouse

bioassay for marine-toxin analysis. Trends Anal. Chem. 2011 ,

30 , 239 – 253.

C.E. (CEN). European Committee for Standardization (CEN), 2003 .

Chan, I. O.; Tsang, V. W.; Chu, K. K.; Leung, S. K.; Lam, M. H.; Lau,

T. C.; Lam, P. K.; Wu, R. S. Solid-phase extraction-fluorimetric

high performance liquid chromatographic determination of

domoic acid in natural seawater mediated by an amorphous

titania sorbent. Anal. Chim. Acta 2007 , 583 , 111 – 117.

Chen, X. W.; Liu, H. X.; Jin, Y. B.; Li, S. F.; Bi, X.; Chung, S.; Zhang, S. S.;

Jiang, Y. Y. Separation, identification and quantification of

tetrodotoxin and its analogs by LC-MS without calibration of

individual analogs. Toxicon. 2011 , 57, 938 – 943.

Cho, H. E.; Ahn, S. Y.; Son, I. S.; In, S.; Hong, R. S.; Kim, D. W.;

Woo, S. H.; Moon, D. C.; Kim, S. Determination and validation

of tetrodotoxin in human whole blood using hydrophilic inter-

action liquid chromatography – tandem mass spectroscopy and

its application. Forensic Sci. Int. 2012 , 217 , 76 – 80.

Chulanetra, M.; Sookrung, N.; Srimanote, P.; ndrawattana, N.;

Thanongsaksrikul, J.; Sakolvaree, Y.; Chongsa-Nguan, M.;

Kurazono, H.; Chaicumpa, W. Toxic marine puffer fish in

thailand seas and tetrodotoxin they contained. Toxins. (Basel)

2011 , 3, 1249 – 1262.

Ciminiello, P.; Dell ’ Aversano, C.; Fattorusso, E.; Forino, M.; Magno,

G. S.; Tartaglione, L.; Quilliam, M. A.; Tubaro, A.; Poletti, R.

Hydrophilic interaction liquid chromatography/mass spec-

trometry for determination of domoic acid in Adriatic shellfish.

Rapid Commun. Mass. Spectrom . 2005 , 19 , 2030 – 2038.

Ciminiello, P.; Dell ’ Aversano, C.; Dello Iacovo, E.; Fattorusso, E.;

Forino, M.; Grauso, L.; Tartaglione, L.; Guerrini, F.; Pistocchi, R.

Complex palytoxin-like profile of Ostreopsis ovata . Identifica-

tion of four new ovatoxins by high-resolution liquid chroma-

tography/mass spectrometry. Rapid Commun. Mass Spectrom. 2010 , 24 , 2735 – 2744.

Cohen, N. J.; Deeds, J. R.; Wong, E. S.; Hanner, R. H.; Yancy, H. F.;

White, K. D.; Thompson, T. M.; Wahl, M.; Pham, T. D.; Guichard,

F. M.; Huh, I.; Austin, C.; Dizikes, G.; Gerber, S. I. J. Food Prot. 2009 , 72 , 810.

Commission Regulation. Off. J. Eur. Commun . 2011 , L6 , 3.

de la Iglesia, P.; Gimenez, G.; Diogene, J. Determination of dissolved

domoic acid in seawater with reversed-phase extraction disks

and rapid resolution liquid chromatography tandem mass

spectrometry with head-column trapping. J. Chromatogr. A

2008 , 1215 , 116 – 124.

Dechraoui, M. Y.; Tiedeken, J. A.; Persad, R.; Wang, Z.; Granade, H.

R.; Dickey, R. W.; Ramsdell, J. S. Use of two detection methods

to discriminate ciguatoxins from brevetoxins: Application to

great barracuda from Florida Keys. Toxicon. 2005 , 46 ,

261 – 270.

Dell ’ Aversano, C.; Hess, P.; Quilliam, M. A. Hydrophilic interaction

liquid chromatography – mass spectrometry for the analysis

of paralytic shellfish poisoning (PSP) toxins. J. Chromatogr. A

2005 , 1081 , 190 – 201.

Diener, M.; Erler, K.; Christian, B.; Luckas, B. Application of a new

zwitterionic hydrophilic interaction chromatography column

for determination of paralytic shellfish poisoning toxins. J . Sep. Sci . 2007 , 30 , 1821 – 1826.

Doucette, G.; Mikulski, C.; Jones, K.; King, K.; Marin, R.; Jensen, S.;

Roman, B.; Elliott, C.; Scholin, C. Remote, subsurface detec-

tion of the algal toxin domoic acid onboard the environmental

sample processor: assay development and field trials. Harmful Algae 2009 , 8 , 880 – 888.

European Commission. Off. J. Eur. Union 2004a , L 226 , 22.

European Commission. Off. J. Eur Union L 2004b , 226, 83.

European Commission. Off. J. Eur. Union 2005 , L 338 , 27.

European Commission. Off. J. Eur. Union 2006 , L 320 , 13.

European Commission. Off. J. Eur. Union 2007 , L 281 , 12.

European Commission Panel. EFSA J . 2009a , 1181 , 1.

European Commission Panel. EFSA J . 2009b , 1306 , 1.

Fonfria, E. S.; Vilarino, N.; Campbell, K.; Elliott, C.; Haughey, S. A.;

Ben-Gigirey, B.; Vieites, J. M.; Kawatsu, K.; Botana, L. M.

Paralytic shellfish poisoning detection by surface plasmon

resonance-based biosensors in shellfish matrixes. Anal. Chem .

2007 , 79 , 6303 – 6311.

Furey, A.; Lehane, M.; Gillman, M.; Fernandez-Puente, P.; James, K. J.

Determination of domoic acid in shellfish by liquid chromato-

graphy with electrospray ionization and multiple tandem mass

spectrometry. J. Chromatogr. A 2001 , 938 , 167–174.

Fux, E.; McMillan, D.; Bire, R.; Hess, P. Development of an ultra-

performance liquid chromatography – mass spectrometry

method for the detection of lipophilic marine toxins.

J.  Chromatogr. A 2007 , 1157, 273 – 280.

Gago-Martinez, A.; Pineiro, N.; Aguete, E. C.; Vaquero, E.;

Nogueiras, M.; Leao, J. M.; Rodriguez-Vazquez, J. A.; Dabek-

Zlotorzynska, E. Further improvements in the application of

high-performance liquid chromatography, capillary electropho-

resis and capillary electrochromatography to the analysis of

algal toxins in the aquatic environment. J. Chromatogr. A 2003 ,

992 , 159 – 168.

Galil, B. S.; Zenetos, A. In: Leppa, E.; Koski, S.; Gollasch, S.; Olenin,

eds. Invasive Aquatic Species of Europe: Distribution, Impacts and Management . Dordrecht, The Netherlands: Kluwer

Academic, 2002, p. 325.

Garet, E.; Gonz á lez-Fern á ndez, A.; Lago, J.; Vieites, J. M.; Cabado, A. G.

Comparative evaluation of Enzyme-Linked Immunoassay and

reference methods for the detection of shellfish hydrophilic toxins

in several presentations of seafood. Toxicon 2010 , 58 , 1410–1415.

Gerssen, A.; McElhinney, M. A.; Mulder, P. P.; Bire, R.; Hess, P.; de

Boer, J. Solid phase extraction for removal of matrix effects

in lipophilic marine toxin analysis by liquid chromatography-

tandem mass spectrometry. Anal. Bioanal. Chem. 2009 , 394 ,

1213 – 1226.

Gerssen, A.; Mulder, P. P.; de Boer, J. Screening of lipophilic marine

toxins in shellfish and algae: development of a library using

liquid chromatography coupled to orbitrap mass spectrometry.

Anal. Chim. Acta. 2011 , 685 , 176 – 185.

Gessner, B. D.; McLaughlin, J. B. In: Botana, L. M. ed. Seafood and Freshwater Toxins: Pharmacology, Physiology and Detection .

Boca Raton, FL: CRC Press (Taylor and Francis Group), 2008, p. 77.

Page 16: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

30      A.M. Botana et al.: Analysis of marine toxins

Golani, D. Distribution of Lessepsian migrant fish in the Mediterra-

nean. Ital. J. Zool. 1998 , 65, 95 – 99.

Gonz á lez, J. C.; Leira, F.; Fontal, O. I.; Vieytes, M. R.; Ar é valo, F. F.;

Vieites, J. M.; Berm ú dez-Puente, M.; Mu ñ iz, S.; Salgado, C.;

Yasumoto, T.; Botana, L. M. Inter-laboratory validation of the

fluorescent protein phosphatase inhibition assay to determine

diarrhetic shellfish toxins: intercomparison with liquid

chromatography and mouse bioassay. Anal. Chim. Acta. 2002 ,

466, 233 – 246.

Hamasaki, K.; Kogure, K.; Ohwada, K. A biological method for the

quantitative measurement of tetrodotoxin (TTX): Tissue culture

bioassay in combination with a water-soluble tetrazolium salt.

Toxicon. 1996 , 34 , 490 – 495.

Hamilton, B.; Hurbungs, M.; Jones, A.; Lewis, R. J. Multiple cigua-

toxins present in Indian Ocean reef fish. Toxicon. 2002a , 40 ,

1347 – 1353.

Hamilton, B.; Hurbungs, M.; Vernoux, J. P.; Jones, A.; Lewis, R. J.

Isolation and characterisation of Indian Ocean ciguatoxin.

Toxicon. 2002b , 40 , 685 – 693.

Hamilton, B., Whittle, N., Shaw, G., Eaglesham, G., Moore, M. R.,

Lewis, R. J. Human fatality associated with Pacific ciguatoxin

contaminated fish. Toxicon . 2010 , 56 , 668–673.

He, Y.; Fekete, A.; Chen, G.; Harir, M.; Zhang, L.; Tong, P.; Schmitt-

Kopplin, P. Analytical approaches for an important shellfish

poisoning agent: domoic acid. J. Agric. Food Chem . 2010 , 58 ,

11525 – 11533.

Hess, P.; Gallacher, S.; Bates, L. A.; Brown, N.; Quilliam, M. A.

Determination and confirmation of the amnesic shellfish

poisoning toxin, domoic acid, in shellfish from Scotland by

liquid chromatography and mass spectrometry. J. AOAC Int . 2001 , 84 , 1657–1667.

Hess, P.; Grune, B.; Anderson, D. B.; Aune, T.; Botana, L. M.;

Caricato, P.; van Egmond, H. P.; Halder, M.; Hall, S.;

Lawrence, J. F.; Moffat, C.; Poletti, R.; Richmond, J.; Rossini, G.

P.; Seamer, C.; Vilageliu, J. S. Altern. Lab. Anim. 2006 , 34 , 193.

Horner, R. A.; Kusske, M. B.; Moynihan, B. P.; Skinner, R. N.; Wekell,

J. C. Retention of domoic acid by Pacific razor clams, Siliqua patula (Dixon, 1798): preliminary study J. Shellfish Res . 1993 ,

12 , 451.

Huang, X.; Hsu, K. H.; Chu, F. S. Direct competitive enzyme-linked

immunosorbent assay for saxitoxin and neosaxitoxin. J. Agric. Food Chem . 1996 , 44 , 1029 – 1035.

International, A. In: Horowitz, ed. Official Methods of Analysis of the AOAC . Gaithersburg, Maryland: AOAC, 2000.

James, K. J.; Gillman, M.; Lehane, M.; Gago-Martinez, A. New fluori-

metric method of liquid chromatography for the determination

of the neurotoxin domoic acid in seafood and marine phyto-

plankton. J. Chromatogr. A. 2000 , 871 , 1 – 6.

Jang, J. H.; Lee, J. S.; Yotsu-Yamashita, M. LC/MS analysis of

tetrodotoxin and its deoxy analogs in the marine puffer fish

Fugu niphobles from the southern coast of Korea, and in

the brackishwater puffer fishes Tetraodon nigroviridis and

Tetraodon biocellatus from Southeast Asia. Mar. Drugs 2010 , 8, 1049 – 1058.

Jen, H. C.; Lin, S. J.; Lin, S. Y.; Huang, Y. W.; Liao, I. C.; Arakawa, O.;

Hwang, D. F. Occurrence of tetrodotoxin and paralytic

shellfish poisons in a gastropod implicated in food poisoning

in southern Taiwan. Food Addit. Contam. 2007 , 24, 902 – 909.

Jen, H. C.; Lin, S. J.; Tsai, Y. H.; Chen, C. H.; Lin, Z. C.; Hwang, D. F.

Tetrodotoxin poisoning evidenced by solid-phase extraction

combining with liquid chromatography\xe2\u20ac\u201ctan-

dem mass spectrometry. J. Chromatogr. B 2008 , 871, 95 – 100.

Katikou, P.; Georgantelis, D.; Sinouris, N.; Petsi, A.; Fotaras, T. First

report on toxicity assessment of the Lessepsian migrant puff-

erfish Lagocephalus sceleratus (Gmelin, 1789) from European

waters (Aegean Sea, Greece). Toxicon. 2009 , 54, 50 – 55.

Kawabata, T. Food Hygiene Examination Manual . Tokyo: Japan Food

Hygiene Association, 1978, p. 232.

Kawatsu, K.; Shibata, T.; Hamano, Y. Application of immunoaffinity

chromatography for detection of tetrodotoxin from urine sam-

ples of poisoned patients. Toxicon. 1999 , 37, 325 – 333.

Kilcoyne, J.; Fux, E. Strategies for the elimination of matrix effects in

the liquid chromatography tandem mass spectrometry analysis

of the lipophilic toxins okadaic acid and azaspiracid-1 in

molluscan shellfish. J.Chromatogr. A. 2010, 1217, 7123–7130.

Kleivdal, H.; Kristiansen, S. I.; Nilsen, M. V.; Goksoyr, A.; Briggs, L.;

Holland, P.; McNabb, P. Determination of domoic acid toxins in

shellfish by biosense ASP ELISA – a direct competitive enzyme-

linked immunosorbent assay: collaborative study. J. AOAC Int . 2007 , 90 , 1011.

Kodamatani, H.; Saito, K.; Niina, N.; Yamazaki, S.; Muromatsu, A.;

Sakurada, I. Sensitive determination of domoic acid using

high-performance liquid chromatography with electrogen-

erated Tris(2,2 ′ -bipyridine)ruthenium(III) chemiluminescence

detection. Anal. Sci . 2004 , 20 , 1065 – 1068.

Kono, M.; Matsui, T.; Furukawa, K.; Takase, T.; Yamamori, K.;

Kaneda, H.; Aoki, D.; Jang, J. H.; Yotsu-Yamashita, M. Examina-

tion of transformation among tetrodotoxin and its analogs

in the living cultured juvenile puffer fish, kusafugu, Fugu niphobles by intramuscular administration. Toxicon. 2008 , 52 ,

714 – 720.

Kreuzer, M. P.; Pravda, M.; O ’ Sullivan, C. K.; Guilbault, G. G. Novel

electrochemical immunosensors for seafood toxin analysis.

Toxicon. 2002 , 40, 1267 – 1274.

Kudo, Y.; Yasumoto, T.; Konoki, K.; Cho, Y.; Yotsu-Yamashita, M.

Isolation and structural determination of the first 8- epi- type

tetrodotoxin analogs from the newt, cynops ensicauda popei, and comparison of tetrodotoxin analogs profiles of this newt

and the puffer fish, Fugu poecilonotus. Mar. Drugs 2012 , 10, 655 – 667.

Lawrence, J. F.; Menard, C. Liquid chromatographic determination

of paralytic shellfish poisons in shellfish after prechromato-

graphic oxidation. J. AOAC Int . 1991 , 74 , 1006–1012.

Lawrence, J. F.; Charbonneau, C. F.; Menard, C.; Quilliam, M. A.;

Sim, P. G. Liquid chromatographic determination of domoic

acid in shellfish products using the paralytic shellfish poison

extraction procedure of the association of official analytical

chemists. J. Chromatogr . 1989 , 462 , 349 – 356.

Lawrence, J. F.; Cleroux, C.; Truelove, J. F. Comparison of high-

performance liquid chromatography with radioimmunoassay

for the determination of domoic acid in biological samples.

J. Chromatogr. A 1994 , 662 , 173 – 177.

Lawrence, J. F.; Menard, C.; Cleroux, C. Evaluation of prechromato-

graphic oxidation for liquid chromatographic determination of

paralytic shellfish poisons in shellfish. J. AOAC Int . 1995 , 78 , 514.

Le Berre, M.; Kane, M. Biosensor-based assay for domoic acid:

comparison of performance using polyclonal, monoclonal, and

recombinant antibodies. Anal. Lett. 2006 , 39 , 1587 – 1598.

Lefebvre, K. A.; Robertson, A.; Frame, E. R.; Colegrove, K. M.; Nance,

S.; Baugh, K. A.; Wiedenhoft, H.; Gulland, F. M. D. Clinical signs

Page 17: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      31

and histopathology associated with domoic acid poisoning

in northern fur seals (callorhinus ursinus) and comparison of

toxin detection methods. Harmful Algae 2010 , 9 , 374.

Lehman, E. M.; Brodie, E. D. Jr.; Brodie, E. D. III. No evidence for

an endosymbiotic bacterial origin of tetrodotoxin in the newt

Taricha granulosa. Toxicon. 2004 , 44 , 243 – 249.

Leira, F.; Alvarez, C.; Cabado, A. G.; Vieites, J. M.; Vieytes, M. R.;

Botana, L. M. Development of a F actin-based live-cell fluori-

metric microplate assay for diarrhetic shellfish toxins. Anal. Biochem . 2003 , 317 , 129 – 135.

Leung, K. S.-Y.; Fong, B. M.-W.; Tsoi, Y.-K. Analytical challenges:

determination of tetrodotoxin in human urine and plasma by

LC-MS/MS. Mar. Drugs 2011 , 9 , 2291 – 2303.

Lewis, R. J.; Jones, A. Characterization of ciguatoxins and ciguatoxin

congeners present in ciguateric fish by gradient reverse-phase

high-performance liquid chromatography/mass spectrometry.

Toxicon. 1997 , 35, 159 – 168.

Lewis, R. J.; Vernoux, B. I. M. Structure of Caribbean ciguatoxin

lsolated from Caranx latus. J. Am. Chem. Soc. 1998 , 120, 5914 – 5920.

Lewis, R. J.; Yang, A.; Jones, A. Rapid extraction combined with

LC-tandem mass spectrometry (CREM-LC/MS/MS) for the

determination of ciguatoxins in ciguateric fish flesh. Toxicon. 2009 , 54, 62 – 66.

Lin, S. J.; Hwang, D. F. Possible source of tetrodotoxin in the starfish

Astropecten scoparius. Toxicon. 2001 , 39 , 573 – 579.

Litaker, R. W.; Vandersea, M. W.; Faust, M. A.; Kibler, S. R.; Nau,

A. W.; Holland, W. C.; Chinain, M.; Holmes, M. J.; Tester, P. A.

Toxicon. 2010 , 56, 711 – 730.

Llamas, N. M.; Stewart, L.; Fodey, T.; Higgins, H. C.; Velasco, M. L.;

Botana, L. M.; Elliott, C. T. Development of a novel immuno-

biosensor method for the rapid detection of okadaic acid

contamination in shellfish extracts. Anal. Bioanal. Chem. 2007 ,

389, 581 – 587.

Louzao, M. C.; Vieytes, M. R.; Baptista de Sousa, J. M.; Leira, F.;

Botana, L. M. A fluorimetric method based on changes in

membrane potential for screening paralytic shellfish toxins in

mussels. Anal. Biochem . 2001 , 289 , 246 – 250.

Lundholm, N.; Skov, J.; Pocklington, R.; Moestrup, Ø . Domoic acid,

the toxic amino acid responsible for amnesic shellfish poison-

ing, now in Pseudonitzschia seriata (Bacillariophyceae) in

Europe. Phycologia 1994 , 33 , 475 – 478.

Mafra, L. L. Jr.; Leger, C.; Bates, S. S.; Quilliam, M. A. Analysis

of trace levels of domoic acid in seawater and plankton by

liquid chromatography without derivatization, using UV or

mass spectrometry detection. J. Chromatogr. A. 2009 , 1216 ,

6003 – 6011.

Man, C. N.; Noor, N. M.; Harn, G. L.; Lajis, R.; Mohamad, S. Screen-

ing of tetrodotoxin in puffers using gas chromatography –

mass spectrometry. J. Chromatogr. A 2010 , 1217, 7455 – 7459.

McNabb, P. In: Botana, L. M., ed. Seafood and Freshwater Toxins: Pharmacology, Physiology and Detection . Boca Raton, FL: CRC

Press (Taylor and Francis Group), 2008, p. 209.

McNabb, P.; Selwood, A. I.; Holland, P. T.; Aasen, J.; Aune, T.;

Eaglesham, G.; Hess, P.; Igarishi, M.; Quilliam, M.; Slattery, D.;

Van de Riet, J.; Van Egmond, H.; Van den Top, H.; Yasumoto, T.

J. AOAC Int . 2005 , 88 , 761.

Messner, D. J.; Catterall, W. A. The sodium channel from rat brain.

Role of the beta 1 and beta 2 subunits in saxitoxin binding.

J. Biol. Chem . 1986 , 261 , 211.

Micheli, L.; Radoi, A.; Guarrina, R.; Massaud, R.; Bala, C.; Moscone,

D.; Palleschi, G. Disposable immunosensor for the determina-

tion of domoic acid in shellfish. Biosens. Bioelectron . 2004 ,

20 , 190 – 196.

M í guez, A.; Fernandez, M. L.; Fraga, S. First detection of domoic

acid in Galicia (NW of Spain). In: Yasumoto, T.; Oshima, Y.;

Fukuyo, Y., eds. Harmful and Toxic Algal Blooms: Intergovern-mental Oceanographic Commission of UNESCO . Paris, France:

UNESCO, 1996, p. 143.

Miyazawa, K.; Noguchi, T. J. Toxicol. Toxin. Rev . 2001 , 20 , 11.

Mosher, H.S.; Fuhrman, G.J.; Fuhrman, F.A.; Fischer, H.G. Tarichatoxin-

tetrodotoxin: a potent neurotoxin. Science 1965 , 144 , 1100 – 1110.

Narita, H.; Martsubara, S.; Miwa, N.; Akahane, S.; Murakami, M.;

Goto, T.; Nara, M.; Noguchi, T.; Saito, T.; Shida, Y.; Hashimoto, K.

Occurrence of Tetrodotoxin in a Trumpet Shell, “ Boshubora ”

Charonia sauliae . Bull. Japan Soc. Sci. Fish . 1987 , 47 , 935 – 941.

Neagu, D.; Micheli, L.; Palleschi, G. Study of a toxin – alkaline

phosphatase conjugate for the development of an immunosen-

sor for tetrodotoxin determination. Anal. Bioanal. Chem. 2006 ,

385, 1068 – 1074.

Negri, A.; Stirling, D.; Quilliam, M.; Blackburn, S.; Bolch, C.;

Burton, I.; Eaglesham, G.; Thomas, K.; Walter, J.; Willis, R.

Three novel hydroxybenzoate saxitoxin analogues isolated

from the dinoflagellate Gymnodinium catenatum. Chem. Res. Toxicol . 2003 , 16 , 1029 – 1033.

Noguchi, T.; Ebesu, J. S. M. Puffer poisoning: epidemiology and

treatment. J. Toxicol. Toxin Rev. 2001 , 20, 1–10.

Noguchi, T.; Arakawa, O. Tetrodotoxin – distribution and accumula-

tion in aquatic organisms, and cases of human intoxication.

Mar. Drugs 2008 , 6 , 220 – 242.

O ’ Leary, M. A.; Schneider, J. J.; Isbister, G. K. Use of high performance

liquid chromatography to measure tetrodotoxin in serum and

urine of poisoned patients. Toxicon. 2004 , 44, 549 – 553.

Onodera, H.; Satake, M.; Oshima, Y.; Yasumoto, T.; Carmichael, W. W.

New saxitoxin analogues from the freshwater filamentous

cyanobacterium Lyngbya wolle Nat. Toxins 1997 , 5 , 146 – 151.

Oshima, Y. Postcolumn derivatization liquid chromatographic

method for paralytic shellfish toxins. J. AOAC Int . 1995 , 78 , 528.

Oshima, Y.; Fallon, W. E.; Shimizu, Y.; Noguchi, T.; Hashimoto, Y.

Toxins of the Gonyaulax sp. and Infested Bivalves in Owase

Bay. Bull. Jpn. Soc. Sci. Fish 1976 , 42 , 851 – 856.

Oshima, Y.; Machida, M.; Sasaki, K.; Tamaoki, Y.; Yasumoto, T.

Liquid chromatographic-fluorometric analysis of paralytic

shellfish toxins. Agric. Biol. Chem . 1984 , 48 , 1707 – 1711.

Oshima, Y.; Hasegawa, M.; Yasumoto, T.; Hallegraeff, G.;

Blackburn, S. Dinoflagellate Gymnodinium catenatum as the

source of paralytic shellfish toxins in Tasmanian shellfish.

Toxicon 1987 , 25 , 1105 – 1111.

Otero, P.; Perez, S.; Alfonso, A.; Vale, C.; Rodriguez, P.; Gouveia, N. N.;

Gouveia, N.; Delgado, J.; Vale, P.; Hirama, M.; Ishihara, Y.;

Molg ó , J.; Botana, L. M. First toxin profile of ciguateric fish

in Madeira arquipelago (Europe). Anal. Chem . 2010 , 82 ,

6032 – 6039.

Otero, P.; Alfonso, A.; Alfonso, C.; Rodriguez, P.; Vieytes, M. R.;

Botana, L. M. Effect of uncontrolled factors in a validated liquid

chromatography – tandem mass spectrometry method question

its use as a reference method for marine toxins: major causes

for concern. Anal. Chem . 2011a , 83 , 5903 – 5911.

Otero, P.; Alfonso, A.; Alfonso, C.; Araoz, R.; Molg ó , J.; Vieytes, M.

R.; Botana, L. M. First direct fluorescence polarization assay

Page 18: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

32      A.M. Botana et al.: Analysis of marine toxins

for the detection and quantification of spirolides in mussel

samples. Anal. Chim. Acta 2011b , 701 , 200 – 208.

Pardo, O.; Yusa, V.; Leon, N.; Pastor, A. Development of a pres-

surised liquid extraction and liquid chromatography with

electrospray ionization-tandem mass spectrometry method for

the determination of domoic acid in shellfish. J. Chromatogr. A   2007 , 1154 , 287 – 294.

Perez-Arellano, J. L.; Luzardo, O. P.; Perez Brito, A.; Hernandez

Cabrera, M.; Zumbado, M.; Carranza, C.; Angel-Moreno, A.;

Dickey, R. W.; Boada, L. D. Ciguatera fish poisoning, Canary

Islands. Emerg. Infect. Dis. 2005 , 11, 1981.

Pleasance, S.; Xie, M.; LeBlanc, Y.; Quilliam, M. A. Analysis of

domoic acid and related compounds by mass spectrometry and

gas chromatography/mass spectrometry as N -trifluoroacetyl-

O -silyl derivatives. Biomed . Environ. Mass. Spectrom . 1990 , 19 ,

420 – 427.

Pocklington, R.; Milley, J. E.; Bates, S. S.; Bird, C. J.; De Freitas, A.

S. W.; Quilliam, M. A. Trace determination of domoic

acid in sea water and phytoplankton by high-performance

liquid chromatography of the fluorenylmethoxycarbonyl

(FMOC) derivative. Int. J. Environ. Anal. Chem . 1990 , 38 ,

351 – 368.

Pottier, I.; Vernoux, J. P.; Jones, A.; Lewis, R. J. Analysis of toxin

profiles in three different fish species causing ciguatera fish

poisoning in Guadeloupe, French West Indies. Food Addit. Contam. 2002a , 19, 1034 – 1042.

Pottier, I.; Vernoux, J. P.; Jones, A.; Lewis, R. J. Characterisation of

multiple Caribbean ciguatoxins and congeners in individual

specimens of horse-eye jack ( Caranx latus ) by high-perfor-

mance liquid chromatography/mass spectrometry. Toxicon. 2002b , 40, 929 – 939.

Powell, C. L.; Ferdin, M. E.; Busman, M.; Kvitek, R. G.; Doucette, G. J.

Development of a protocol for determination of domoic acid

in the sand crab ( Emerita analoga ): a possible new indicator

species. Toxicon. 2002 , 40 , 485 – 492.

Quilliam, M. A. The role of chromatography in the hunt for red tide

toxins. J. Chromatogr. A 2003 , 1000 , 527 – 548.

Quilliam, M. A.; Thomas, K.; Wright, J. L. Analysis of domoic acid in

shellfish by thin-layer chromatography. Nat. Toxins 1998 , 6 ,

147 – 152.

Raybould, T.; Bignami, G. S.; Inouye, L. K.; Simpson, S. B.; Byrnes,

J. B.; Grothaus, P. G.; Vann, D. C. A monoclonal antibody-based

immunoassay for detecting tetrodotoxin in biological samples.

J. Clin. Lab. Anal . 1992 , 6 , 65 – 72.

Rhodes, L. L.; Smith, K. F.; Munday, R.; Selwood, A. I.; McNabb, P. S.;

Holland, P. T.; Bottein, M. Y. Toxic dinoflagellates ( Dinophyceae)

from Rarotonga, Cook Islands. Toxicon. 2010 , 56 , 751 – 758.

Rodriguez, P.; Alfonso, A.; Vale, C.; Alfonso, C.; Vale, P.; Tellez, A.;

Botana, L. M. First toxicity report of tetrodotoxin and

5,6,11-trideoxyTTX in the trumpet shell Charonia lampas lampas in Europe. Anal. Chem. 2008 , 80, 5622 – 5629.

Rodriguez, P.; Alfonso, A.; Botana, A. M.; Vieytes, M. R.; Botana, L. M.

Comparative analysis of pre- and post-column oxidation

methods for detection of paralytic shellfish toxins. Toxicon. 2010 , 56 , 448 – 457.

Rodr í guez, P.; Alfonso, A.; Otero, P.; Katikou, P.; Georgantelis, D.;

Botana, L. M. Liquid chromatography – mass spectrometry

method to detect tetrodotoxin and its analogues in the puffer

fish Lagocephalus sceleratus (Gmelin, 1789) from European

waters. Food Chem . 2012 , 132 , 1103 – 1111.

Rodr í guez-Velasco, M. L. Toxin monitoring programs and regulatory

review. In: Botana, L. M., ed. Seafood and Freshwater Toxins: Pharmacology, Physiology and Detection . Boca Raton, FL: CRC

Press (Taylor and Francis Group), 2008, p. 919.

Roeder, K.; Erler, K.; Kibler, S.; Tester, P.; Van The, H.; Nguyen-Ngoc,

L.; Gerdts, G.; Luckas, B. Characteristic profiles of Ciguatera

toxins in different strains of Gambierdiscus spp. Toxicon. 2010 ,

56 , 731 – 738.

Rubiolo, J. A.; Lopez-Alonso, H.; Alfonso, A.; Vega, F. V.; Vieytes, M. R.;

Botana, L. M. Characterization and activity determination of

the human protein phosphatase 2a catalytic subunit a

expressed in insect larvae. Appl. Biochem. Biotechnol. 2012 ,

167, 918 – 928.

Rundberget, T.; Aasen, J. A.; Selwood, A. I.; Miles, C. O. Pinnatoxins

and spirolides in Norwegian blue mussels and seawater.

Toxicon. 2011 , 58, 700 – 711.

Satake, M.; Ishibashi, Y.; Legrand, A. M.; Yasumoto, T. Biosci. Biotechnol. Biochem. 1997 , 60, 2103 – 2105.

Shoji, Y.; Yotsu-Yamashita, M.; Miyazawa, T.; Yasumoto, T.

Electrospray ionization mass spectrometry of tetrodotoxin

and its analogs: liquid chromatography/mass spectrometry,

tandem mass spectrometry, and liquid chromatography/

tandem mass spectrometry. Anal. Biochem . 2001 , 290, 10.

Silva, M.; Azevedo, J.; Rodriguez, P.; Alfonso, A.; Botana, L. M.;

Vasconcelos, V. New gastropod vectors and tetrodotoxin

potential expansion in temperate waters of the Atlantic Ocean.

Mar. Drugs 2012 , 10, 712 – 726.

Sommer, H.; Meyer, K. F. Paralytic shellfish poisoning. Arch. Path .

1937 , 24 , 560.

Stevens, R. C.; Soelberg, S. D.; Eberhart, B.-T. L.; Spencer, S.;

Wekell, J. C.; Chinowsky, T. M.; Trainer, V. L.; Furlong, C. E.

Detection of the toxin domoic acid from clam extracts using a

portable surface plasmon resonance biosensor. Harmful Algae

2007 , 6 , 166 – 174.

Stewart, L.; Elliott, C.; Walker, A.; Curran, R.; Connolly, L.

Development of a monoclonal antibody binding okadaic acid

and dinophysistoxins-1, -2 in proportion to their toxicity

equivalence factors. Toxicon. 2009a , 54 , 491 – 498.

Stewart, L. D.; Hess, P.; Connolly, L.; Elliott, C. T. Development and

single-laboratory validation of a pseudofunctional biosensor

immunoassay for the detection of the okadaic acid group of

toxins. Anal. Chem. 2009b , 81 , 10208 – 10214.

Stobo, L. A.; Lacaze, J. P.; Scott, A. C.; Gallacher, S.; Smith, E. A.;

Quilliam, M. A. J. AOAC Int. 2005 , 88, 1371.

Suzuki, T.; Quilliam, M. A. LC-MS/MS analysis of diarrhetic shellfish

poisoning (DSP) toxins, okadaic acid and dinophysistoxin

analogues, and other lipophilic toxins. Anal. Sci. 2011 , 27, 571.

Traynor, I. M.; Plumpton, L.; Fodey, T. L.; Higgins, C.; Elliott, C. T.

J. AOAC Int . 2006 , 89 , 868.

Tsai, Y. H.; Hwang, D. F.; Cheng, C. A.; Hwang, C. C.; Deng, J. F.

Determination of tetrodotoxin in human urine and blood using

C18 cartridge column, ultrafiltration and LC-MS. J. Chromatogr. B 2006 , 832, 75 – 80.

Turner, A. D.; Norton, D. M.; Hatfield, R. G.; Morris, S.; Reese, A.

R.; Algoet, M.; Lees, D. N. Refinement and extension of AOAC

method 2005.06 to include additional toxins in mussels:

single-laboratory validation. J. AOAC Int . 2009 , 92 , 190.

Ujevic, I.; Nincevic-Gladan, Z.; Roje, R.; Skejic, S.; Arapov, J.;

Marasovic, I. Domoic acid – a new toxin in the croatian adriatic

shellfish toxin profile. Molecules 2010 , 15 , 6835 – 6849.

Page 19: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

A.M. Botana et al.: Analysis of marine toxins      33

Vale, C.; Alfonso, A.; Vieytes, M. R.; Romaris, X. M.; Arevalo, F.;

Botana, A. M.; Botana, L. M. In vitro and in vivo evaluation

of paralytic shellfish poisoning toxin potency and the

influence of the pH of extraction. Anal. Chem . 2008a , 80 ,

1770 – 1776.

Vale, P. Complex profiles of hydrophobic paralytic shellfish

poisoning compounds in Gymnodinium catenatum identified

by liquid chromatography with fluorescence detection and

mass spectrometry. J. Chromatogr. A 2008b , 1195 , 85 – 93.

Vale, P.; Sampayo, M. A. M. Domoic acid in Portuguese shellfish and

fish. Toxicon. 2001, 39, 893–904.

Vale, P.; Rangel, I.; Silva, B.; Coelho, P.; Vilar, A. Atypical profiles of

paralytic shellfish poisoning toxins in shellfish from Luanda

and Mussulo bays, Angola. Toxicon. 2009 , 53 , 176 – 183.

van den Top, H. J.; Gerssen, A.; McCarron, P.; van Egmond, H. P.

Quantitative determination of marine lipophilic toxins in

mussels, oysters and cockles using liquid chromatography-

mass spectrometry: inter-laboratory validation study. Food Addit. Contam. 2011a , 28, 1745–1757.

van den Top, H. J.; Elliott, C. T.; Haughey, S. A.; Vilarino, N.; van

Egmond, H. P.; Botana, L. M.; Campbell, K. Surface plasmon

resonance biosensor screening method for paralytic shellfish

poisoning toxins: a pilot interlaboratory study. Anal. Chem .

2011b , 83 , 4206 – 4213.

Van Dolah, F. M. Diversity of marine and freshwater algal toxins.

In: Botana, L. M., ed. Seafood and Freshwater Toxins: Pharmacology, Physiology and Detection . New York: Marcel

Dekker, 2000, p. 19.

Vieites, J. M.; Cabado, A. G. Incidence of marine toxins on industrial

activity. In: Botana, L. M., ed. Seafood and Freshwater Toxins: Pharmacology, Physiology and Detection . Boca Raton, FL: CRC

Press (Taylor and Francis Group), 2008, p. 899.

Vieytes, M. R.; Cabado, A. G.; Alfonso, A.; Louzao, M. C.;

Botana, A. M.; Botana, L. M. Solid-phase radioreceptor assay

for paralytic shellfish toxins. Anal. Biochem . 1993 , 211 ,

87 – 93.

Vieytes, M. R.; Fontal, O. I.; Leira, F.; Baptista de Sousa, J. M.;

Botana, L. M. A fluorescent microplate assay for diarrheic

shellfish toxins. Anal. Biochem . 1997 , 248 , 258 – 264.

Vilarino, N.; Fonfria, E. S.; Molgo, J.; Araoz, R.; Botana, L. M. Detection

of gymnodimine-A and 13-desmethyl C spirolide phycotoxins by

fluorescence polarization. Anal. Chem . 2009 , 81 , 2708 – 2714.

Villar-Gonzalez, A.; Rodriguez-Velasco, M. L.; Ben-Gigirey, B.;

Botana, L. M. Lipophilic toxin profile in Galicia (Spain): 2005

toxic episode. Toxicon. 2007 , 49, 1129.

Villar-Gonzalez, A.; Rodriguez-Velasco, M. L.; Ben-Gigirey, B.;

Yasumoto, T.; Botana, L. M. Assessment of the hydrolysis pro-

cess for the determination of okadaic acid-group toxin ester:

Presence of okadaic acid 7- O -acyl-ester derivates in Spanish

shellfish. Toxicon. 2008 , 51 , 765 – 773.

Wang, Z.; King, K. L.; Ramsdell, J. S.; Doucette, G. J. Determina-

tion of domoic acid in seawater and phytoplankton by liquid

chromatography – tandem mass spectrometry. J. Chromatogr. A 2007 , 1163 , 169 – 176.

Wang, X. J.; Yu, R. C.; Luo, X.; Zhou, M. J.; Lin, X. T. Toxin-screening

and identification of bacteria isolated from highly toxic

marine gastropod Nassarius semiplicatus. Toxicon. 2008 , 52 ,

55 – 61.

Wiese, M.; D ’ Agostino, P. M.; Mihali, T. K.; Moffitt, M. C.; Neilan, B. A.

Neurotoxic alkaloids: saxitoxin and its analogs. Mar. Drugs

2010 , 8 , 2185 – 2211.

Wright, J. L. C.; Falk, M.; McInnes, A. G.; Walter, J. A. Identification of

isodomoic acid D and two new geometrical isomers of domoic

acid in toxic mussels. Can. J. Chem . 1990 , 68 , 22 – 25.

Yakes, B. J.; Deeds, J.; White, K.; DeGrasse, S. L. Evaluation of

surface plasmon resonance biosensors for detection of

tetrodotoxin in food matrices and comparison to analytical

methods. J. Agric. Food Chem. 2011 , 59 , 839 – 846.

Yasumoto, Y. The Manual for the Methods of Food Sanitation Tests.

Japan Food Hygienic Association: Tokyo, Japan, 1991, p. 296.

Yasumoto, T.; Fukui, M.; Sasaki, K.; Sugiyama, K. Determinations of

marine toxins in foods. J. AOAC Int. 1995 , 78, 574–582.

Yogi, K.; Oshiro, N.; Inafuku, Y.; Hirama, M.; Yasumoto, T. Detailed

LC-MS/MS analysis of ciguatoxins revealing distinct regional

and species characteristics in fish and causative alga from the

Pacific. Anal. Chem. 2011 , 83, 8886 – 8891.

Yu, C. H.; Yu, C. F.; Tam, S.; Yu, P. H. Rapid screening of tetrodotoxin

in urine and plasma of patients with puffer fish poisoning by

HPLC with creatinine correction. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2010 , 27, 89–96.

Yu, F.-Y.; Liu, B.-H.; Wu, T.-S.; Chi, T.-F.; Su, M.-C. Development of a

sensitive enzyme-linked immunosorbent assay for the determi-

nation of domoic acid in shellfish. J. Agric. Food Chem . 2004 ,

52 , 5334 – 5339.

Zaki, M. A.; Mossa, A. E. W. Red Sea puffer fish poisoning:

Emergency diagnosis and management of human intoxication.

Egypt. J. Aquat. Res. 2005 , 31, 370–378.

Zaman, L.; Arakawa, O.; Shimosu, A.; Onoue, Y.; Nishio, S.; Shida, Y.;

Noguchi, T. Two new isomers of domoic acid from a red alga,

Chondria armata. Toxicon. 1997 , 35 , 205 – 212.

Zhao, J. Y.; Thibault, P.; Quilliam, M. A. Analysis of domoic acid iso-

mers in seafood by capillary electrophoresis. Electrophoresis

1997 , 18 , 268 – 276.

Zhou, Y.; Li, Y.-S.; Pan, F.-G.; Liu, Z.-S.; Wang, Z. The development

and optimization of ELISA for the determination of tetrodo-

toxin. J. Med. Coll. of PLA 2007 , 22, 347 – 351.

Page 20: Ana M. Botana , Paz Otero , Paula Rodr í guez , Amparo ...

34      A.M. Botana et al.: Analysis of marine toxins

Ana M. Botana professor of Analytical Chemistry; 60 international

papers.

Luis M. Botana full professor of Pharmacology at the University

of Santiago de Compostela (USC), expert in marine toxin

pharmacology; 200 international papers, editor of 8 books and

author of 25 patents.

Paula Rodr í guez and Paz Otero Postdoctoral researchers;

20 international papers.

Amparo Alfonso Professor of Pharmacology at the USC;

90 International papers, and 6 patents.