Int J App Pharm, Vol 15, Issue 6, 2023, 1-13Review Article

IRINOTECAN-REVIEW OF ANALYTICAL METHODS DEVELOPED FOR PHARMACEUTICAL DOSAGE FORMS AND BIOLOGICAL FLUIDS

HRUTA SUNDAR SWAIN1, RUCHI VERMA1*, LALIT KUMAR2

1Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Karnataka-576104, India. 2Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
*Corresponding author: Ruchi Verma; *Email: ruchiverma.pharma@gmail.com

Received: 18 Jul 2023, Revised and Accepted: 09 Sep 2023


ABSTRAC

Irinotecan (IRI) is utilised as a first line anticancer medication in the cure of cancer having extraordinary ability to block DNA-topoisomerase-I. It is used as a monotherapy and adjunct therapy in the treatment of metastatic colorectal cancer and other cancers, and it differs chemically and pharmacologically from other anticancer medications. The proposed review is divided into two main sections i.e. a) Different analytical methods for estimating irinotecan in pharmaceutical formulations, b) Diverse analytical methods for detecting irinotecan in biological matrices. This work also considers the development of numerous analytical methods based on various parameters, as well as the validation of the methods used. Estimated validation characteristics such as Linearity, Limit of Detection (LOD), and Limit of Quantitation (LOQ) are considered for each. Applying bioanalytical methods, the wavelength of detection, mobile phase, columns, flow rate, retention duration, and sample preparation processes are all evaluated as essential quality variables for estimating Irinotecan.

Keywords: Irinotecan, Analytical methods, Biological matrices


INTRODUCTION

Irinotecan is a cytotoxic medication used as 1st line therapy in conjunction with 5-fluorouracil for the treatment of colorectal cancer that has spread to other parts of the body [1, 2]. CPT-11 and Camptothecin-11 are other names for irinotecan. Camptothecin, a cytotoxic alkaloid found in plants like Camptotheca acuminata, is a water-soluble semisynthetic derivative. Irinotecan and other camptothecin analogues/derivatives have demonstrated anticancer activities in in vitro as well as in vivo against a number of experimental tumour forms, including multidrug-resistant cell lines [2, 3]. Irinotecan was approved for cancer treatment in Japan for the first time around 25 y ago. IRI has made important contributions to the treatment of solid tumours all around the world for more than two decades [4]. It is a therapy for colorectal and small cell lung cancer that has been approved by the United States–Food and Drug Act (US-FDA) [3, 5]. Chemically, IRI is a Camptothecin analogue, available with trade names CAMPTOSAR, IRINOMIL, IRNOCAM, TORSIRIN, IRINOTEL, IRBEST [1-6]. Its molecular formula is C33H38N4O6, molecular weight is 586.7 g/mol, available in solid state as pale yellow color powder. Its melting point is 222 °C. It is hygroscopic, soluble in water with a water solubility of 0.107 g/l. It is also reported to be light-sensitive compound [6]. IRI inhibits DNA-topoisomerase-I and causes cell death by stabilising the complexes generated during DNA replication. It is a pro-drug that is hydrolysed by enzymes to produce a metabolite that is active (7-ethyl-10-hydroxycamptothecin, SN-38). Both of these molecules exist in two states at the same time: a lactone form that is active and a carboxylate form that is inactive with no inhibitory activity against topoisomerase I. At lower pH, lactone forms predominate, whereas carboxylate forms predominate at above pH 8 [5]. As IRI exhibits cytotoxic activity, it is employed in the cure of different types of cancers. IRI is available as an intravenous dosage form in two strengths i.e., 20 mg/ml and 4.3 mg/ml, respectively. Each patient's determined volume is taken from the vial and diluted with 500 ml of dextrose 5 percent water (D5W) or normal saline (NS) [1]. IRI's volume of distribution ranges from 110 to 234 L, with protein binding ranging from 30 to 68% [1]. It is extensively metabolised by the liver via several enzyme systems, culminating in the generation of SN-38, a potent active metabolite. The enzyme carboxylesterase breaks down the Di-piperidinocarbonyloxy group of irinotecan to create carboxylic acid and SN-38. Hepatic UDP-glucuronyl transferase can convert SN-38 to SN-38 glucuronide [7]. With a total body clearance of 13.3 to 13.9 L/h, it is eliminated via the biliary and renal routes (about 25 to 50% in 48 h). Irinotecan has a half-life of 6 to 12 hr. Because of considerable inter-individual pharmacokinetic variability, the exact therapeutic range of IRI is unknown. Alopecia, vomiting, neutropenia, asthenia, diarrhoea, fever, myelosuppression, and thrombocytopenia are among side effects of IRI [1].

Because IRI is so important in cancer treatment, rigorous quality control methods should be adopted to ensure drug purity and efficacy. First and foremost, analytical methods for certifying and quantifying pharmaceuticals in pharmaceutical formulations and biological matrices are reviewed. It becomes critical for compiling the numerous analytical methodologies established for calculating IRI.

Fig. 1: Reported methods of analysis from 1996–2020

The numerous analytical approaches for calculating IRI in pharmaceutical formulations are discussed in this article. The literature was obtained from various databases i.e. Science Direct, Scopus, Taylor and Francis, Web of Sciences, Elsevier, Springer, PubMed. The data collected was from 1996-2020.

Fig. 1 shows a visual representation of key studies on analytical approaches used for IRI estimation published between 1996 and 2020. IRI has been calculated in a number of formulations and biological fluids over the years.

The graphical representation in fig. 1 is primarily intended to address various approaches that have been established as well as to provide insight into what other methods may be produced in the future. Fig. 2 depicts the position of various analytical methodologies presented for estimating IRI. This review also looks at how approaches have progressed, from ultra violet (UV) spectrophotometry through fluori-metric detection to hyphenated techniques.

Spectroscopic methods

Ultraviolet and spectrophotometric methods 

UV spectrophotometry, which uses UV absorption and chemical reactions to identify and quantify IRI, is an important technology. For routine analysis, it is a low-cost, simple, quick, selective, and accurate approach. This approach is often used in laboratories, because it is versatile and cost-effective. The numerous spectrophotometry methods for detecting and estimating IRI in pharmaceutical formulations are shown in table 1 [8].

Fig. 2: An outlook of various analytical methods proposed for the estimation of IRI

Hyphenated techniques

LC-MS-MS AND UPLC-MS-MS

Liquid chromatography with tandem mass spectrometry (LC-MS-MS) is a potent analytical technology that combines liquid chromatography's separating ability with triple quadrupole mass spectrometry's sensitive and selective mass analysis capability. The anticancer drug 7-ethyl-10-hydroxycamptothecin (SN-38) has been measured intracellularly in tumour cells using an ultra-high performance liquid-chromatography–mass spectrometry-mass spectrometry (UPLC–MS-MS) approach with camptothecin (CPT) as an internal reference. The SN-38 was extracted using acidified acetonitrile (ACN). SN-38 and CPT were separated on a pentafluorophenyl (PFP) column using gradient elution with acidified water and ACN. SN-38 and CPT were measured using a triple quadrupole mass spectrometry instrument. Least-square regression calibration lines were obtained with average correlation coefficients of R2 = 0.9993±0.0016. Under low-quality control (LQC, 5 ng/ml) and high-quality control (HQC, 500 ng/ml), SN-38 had a lower limit of detection (LOD) of 0.1 ng/ml and a lower limit of quantification (LOQ) of 0.3 ng/ml, while CPT recovery was 98.5±13 percent and SN-38 recovery was 89±6 percent and 95±8 percent, respectively [9-11].

HPLC-FLD

High-Performance Liquid Chromatography (HPLC) with fluorescence detection is one of the most widely used techniques for drug estimation since it is more reliable than other methods. In order to analyse the amount of lactone and lactone plus carboxylate (total) forms of anticancer medicine irinotecan (CPT-11) and its active metabolite SN-38 in human plasma, sensitive high-performance liquid chromatographic tests have been devised. Camptothecin, a chemical made from irinotecan, was used as an internal standard. Lactone forms were pre-treated using ACN-n-butyl chloride (1:4, %v/v) solvent extraction, whereas total forms were cleaned by aqueous perchloric acid: methanol-(1:1, %v/v) protein precipitation, resulting in carboxylate conversion to lactone forms. For chromatography, a Hypersil ODS column was employed, and fluorimetric detection was used. Lactone and total form quantification limits are 0.5 and 2.0 ng/ml, respectively [13, 14].

Table 1: Analytical methods for the estimation of IRI in analytical samples

Year Drug Method Matrix Sample preparation Column/Mobile phase Retention time Detection Linearity range Ref.
2020 USP IRI-HCL HPLC API Dilution

Column: 4.6-mm × 25 cm; 5 µm packing L1

Flow rate: 1.5 ml/min, Mobile phase: Acetonitrile, methanol, and Solution A (17:24:59 %v/v/v), Solution A: 2.8 g/l of monobasic sodium phosphate monohydrate and 1.8 g/l of 1-octanesulfonic acid sodium salt monohydrate in water

NA UV 255 nm NA [64]
2020USP IRI-HCL HPLC API (RS) Dilution Column: 4.6 mm × 25 cm; 10 µm packing L40, Flow rate: 1.0 ml/min, Mobile phase: Hexane, dehydrated alcohol, and di-ethylamine (250:250:1) 0.55 min (for IRI related compound B), 0.60 min (for IRI related compound C), and 1.00 min (for IRI) UV 370 nm NA [64]
2020USP IRI-HCL HPLC Injection (Assay) Dilution

Column: 4.6 mm × 25 cm; 5 µm packing L7

Mobile phase-Acetonitrile (ACN) and buffer (34:66 %v/v). Adjust with phosphoric acid to a pH of 2.5

NA UV 254 nm NA [63]
2020USP IRI-HCL HPLC Injection (RS) Dilution

Column: 4.6 mm × 25 cm; 5 µm packing L1

Elution: Gradient, Mobile phase: Solution A: Dissolve 2 g of sodium 1 hexane sulfonate and 1 ml of triethylamine in 1 L of water. Adjust with phosphoric acid to a pH of 2.5., Solution B: Acetonitrile

0.53 min (for IRI related compound B), 0.65 min (for Camptothecin), 1.00 min (for IRI), and 1.16 min (7-Ethylcamptothecin) UV 254 nm NA [63]
2021 IRI HPLC-DAD Standard solution Dilution Column: Purosher® STAR RP-18 end-capped 3 µm Hibar® RT 150-4.6 mm, Mobile phase: 20 mmol Phosphate buffer with pH 1.7: Methanol (45:55) (%v/v) 2.715 min UV 233 nm 0.04 to 1.80 mg/ml [18]
2018 IRI (infusion) HPLC-DAD Infusion solution concentrates Dilution

Column: Zorbax Extend C18 (250 mm × 4.6 mm; 5 µm particle size); Security Guard Column (4.0 × 3.0 mm; 5 µm particle size).

Mobile phase: Milli Q water and acetonitrile (96:04) (%v/v).

8.97±0.38 min 376 nm 0.1 to 18 µg/ml [19]
2016 Tablet and parenteral formulations UV spectrophotometry, HPTLC Infusion solution concentrates Dilution Chromatography plates: Silica gel plates (Pre-Coated), Mobile phase: (Toluene: ethyl acetate: MeOH: carbon tetrachloride (CCl4)) (9.2: 5: 0.9: 0.8) (v/v/v/v) 0.34±0.08 (Rf value) UV 317 nm (densitometric), 247 nm (UV spectrophotometry) 200–1200 ng/spot (HPTLC), 2–10 µg/ml, (UV spectrophotometry) [8]
2012 IRI HCl working standard injection and its impurities UPLC-UV Infusion solution concentrates Dilution Column: Waters Acquity BEH C8 column (100 × 2.1 mm, 1.7 µm particle size) Mobile phase: Solvent A-(0.02 Mol/l potassium dihydrogen ortho-phosphate, pH adjusted to 3.4 with ortho-phosphoric acid), Solvent B-(a mixture of ACN and MeOH in the ratio of 62:38 %v/v) (UPLC) 2.165 min UV 220 nm 0.063 to 83.2 µg/ml [26]
2021 IRI Infusion HPLC-UV/VIS photometer Infusion solution concentrates Dilution Column: ACE 5 column (Hichrom Ltd, UK) (110 Å, C18, 5 µm, 250 × 4.6 mm) (ACE 5 column, 110 Å), Mobile phase: ACN: 20 mmol phosphoric acid/sodium phosphate buffer pH 3.2 (28:72) (%v/v) NA 220 nm (PDA) 0.90-37.00 mg/ml [29]
2021 IRI HPLC-UV Injection formulation Dilution Column: HyperClone (Phenomenex®) C18 column (250 × 4.6 mm id, particle size 5 µm, ODS 130 Å)., Mobile phase: Acetonitrile: 20 mmol/l potassium phosphate buffer (pH 2.5) containing 0.1% triethylamine in the ratio (45:55) (%v/v). 4.09 min UV 254 mn 0.5 to 18 µg/ml [9]
2019 IRI UHPLC-MS-MS Raw material (pure Irinotecan) Solid-phase extraction Column: Kinetex XB-C18 column (3.0 mm × 100 mm, 1.7 µm)., Mobile phase: Solvent A-water with 0.1% formic acid., Solvent B-MeOH with 0.1% formic acid 5.96 min Mass/Charge 587.3 → Mass/Charge 543.3 NA [30]
2018 IRI HPLC-FLD Hospital effluent Liquid-liquid microextraction, solid-phase extraction Column: Alltima (Deerfield, USA) C18 (150 × 3.2 mm, 5 µm particle size), Elution–Gradient, Mobile phase-Solvent A–Acetonitrile, Solvent B-(100 mmol Ammonium formate, with 0.02% triethylamine) pH-4.5 NA Fluorescence 0.8 to 100
µg/l
[14]
2019 IRI UPLC-MS-MS Hospital chemotherapy compounding units Dilution Column: Cortecs UPLC T 3, (1.6 µm, 2.1 mm × 100 mm), Elution–Gradient, Mobile phase: freshly prepared 10 mmol ammonium acetate (pH 5.1) (Solvent A) pure ACN as organic modifiers (Solvent B). 10.9 min Precursor ion 587.3 m/z 1-400 ng/ml [41]
2013 IRI LC-MS-MS Wipe samples Extraction Column: Octadecyl silyl column Inertsil® (GL Sciences, Tokyo, Japan) ODS-3; 50 mm × 2.1 mm; particle size 3 μm; guard column (cartridge guard-column® (GL Sciences, Tokyo, Japan) 20 mm × 2.0 mm; particle size, 3 μm)., Mobile phases: 0.1% formic acid–water (Solvent A), ACN (Solvent B) 5.77 min Mass/Charge 587.7→167.3 (IRI) 5–1000 ng/wipe [45]
2006 SN-38 HPLC-DAD Novel liposome-based formulation Dilution Column: Agilent Zorbax SB-C18 (4.6 mm × 250 mm, 5 μm), Mobile phase: 25 mmol Sodium dihydrogen phosphate with pH 3.1 and ACN in the ratio 50:50, (%v/v). 4.9 min 265 nm 1–25 μg/ml [46]
2015 IRI HPLC-UV Cancer chemotherapy infusions Dilution Column: Synergi® Max-RP, (Phenomenex)C12-(4 μm, 150 mm × 4.6 mm) with guard-column-Max-RP (4 mm × 3 mm)., Elution–GradientMobile phase: Solvent A (6.2 mmol Nonafluoropentanoic acid (NFPA) aqueous solution, with pH 2.5) and Solvent B (MeOH) 16.6 min 254 nm 6–120 μg/ml [49]
2012 IRI LC-MS-MS Wipe samples Desorption Column: ZORBAX SB-C18 (RR-2.1 × 100 mm, 3.5 µm), Mobile phase: Ultrapure water (solution A), ACN (solution B) and FA 1% (solution C). 13.66 min Mass/Charge 587.9→ 587.3 NA [51]
2007 IRI RP-HPLC-UV Injections Dilution Column: Reverse phase cyano column (4.6 mm × 25 cm, 5 µm)., Mobile phase: Phosphate buffer with pH 2.5: ACN (75:25) (%v/v). 5.82 min 225 nm 20.0 to 80.0 μg/ml [52]
2011 IRI HPLC-UV Bulk and tablets Dilution Column: Inertsil ODS C-18 column-(250 × 4.6 mm, 5 µm), Elution-isocratic mode, Mobile phase-ACN: MeOH: 0.01 M Potassium dihydrogen ortho-phosphate in the ratio of 55:18:27 (%v/v/v) 2.9 min 220 nm 30-90 µg/ml [2]
2009 IRI HCL RP-HPLC-UV Injections Dilution Column: Kromasil C18 analytical column-(4.6 mm × 250 mm, 5 μm particle size), Mobile phase-(10 mmol Potassium dihydrogen ortho-phosphate buffer (pH 3.5): ACN: MeOH (55:25:20) (%v/v/v) 7.6±0.18 254 nm 30–70 μg/ml [53]
2013 IRI RP-HPLC-UV Cleaning validation swab samples Extraction Column: Waters symmetry shield RP-18-(250 mm × 4.6 mm, 5 µm) column, Elution-Isocratic, Mobile phase-0.02 M Potassium di-hydrogen orthophosphate, with pH 3.5: MeOH: ACN (60:20:20) (%v/v/v).

4.779

min

220 nm 0.024 to 0.143 µg/ml [55]
2019 IRI (bulk) and dosage forms RP-HPLC-UV Bulk and dosage forms Dilution Column: Reverse phase Inertsil ODS (250 × 4.6 mm, 5 μm particle size), Mobile phase-0.02 M potassium dihydrogen ortho-phosphate buffer and ACN (40: 60) (%v/v) 2.1 min 222 nm 40-120 µg/ml [56]
2020 IRI HCl HPLC-UV Irinotecan Hydrochloride and Curcumin in Co-delivered Polymeric Nanoparticles Irinotecan hydrochloride and curcumin co-delivered nanoparticle (ICN) were prepared by combinatorial entrapping them into polyethylene glycol–polylactic acid-co-glycolic acid (PEG–PLGA) polymeric nanoparticles. Column: C18 column Kinetex-(150 mm × 4.6 mm, 2.6 µm particle size, 100 Å), Pre-column (WATREX 50 mm × 4 mm, ReproSil 100 C18, 5 µm particle size), Mobile phase-ACN and ultrapure water containing sodium dodecyl sulfate (0.08 mol/l), disodium phosphate (Na2HPO4, 0.002 mol/l) and acetic acid (4 %v/v) in the ratio 50:50 (%v/v).

3.317

min

256 nm 2.05-1050 μg/ml. [57]
2012 IRI UPLC-UV and LC-MS-MS Injections Dilution UPLC: Column: Waters Acquity BEH C8-(100 × 2.1 mm, 1.7 μm), Mobile phase-Solvent A (0.02 mol/l potassium dihydrogen ortho-phosphate, pH with 3.4), Solvent B (a mixture of ACN and MeOH in the ratio of 62:38 %v/v), LC-MS conditions: Column: Waters Symmetry Shield RP 18, -(250 × 4.6 mm, 5 μm) columnMobile phase-Ammonium formate buffer (0.1 mol/l, pH 3.5) (Solvent A) ACN and MeOH in the ratio of 60:40 %v/v, (Solvent B) 1.9-2.4 min (IRI) 220 nm (UPLC) and Mass/Charge 605.5 (LC-MS-MS) 0.063 to 83.2 µg/ml [58]

HPLC-UV

HPLC (High-Performance Liquid Chromatography) is a process where separation is done based on the size, surface charge, and other factors. After separation, UV spectroscopy combined with HPLC allows for the assessment of molecular concentrations. It is straight forward, particularly, accurate, and precise method. To begin, the method was utilised to successfully determine the amount of irinotecan in 100 mg Irnocam pills. In isocratic mode, a mobile phase that contains 55:18:27 (%v/v/v) acetonitrile (ACN): methanol (MeOH): 0.01M Potassium dihydrogen phosphate (KH2PO4) was run on an Inertsil ODS C-18, column with a 250 × 4.6 mm, 5 µm internal diameter. At a flow rate of 1.5 ml/min, the effluents were measured at 220 nm. The retention time of irinotecan was 2.9 min. Linearity, accuracy, precision, specificity, detection limit, quantification limit, and resilience were also evaluated. Limit of quantification and limit of detection values was found to be 35 ng/ml and 10 ng/ml, respectively [2, 12].

Biological matrices

A variety of bioanalytical techniques have been developed to estimate the IRI in biological matrices such as blood, plasma, tissue, serum, urine, and so on. By permitting the identification and quantification of substances and their metabolites in sample specimens, bioanalytical techniques aid in the interpretation of the results of bioequivalence, pharmacokinetic, and toxicokinetic studies [15, 16]. Table 1 summarises the many analytical approaches developed for assessing IRI in biological matrices both alone and in combination with other medicines, including hyphenated techniques that allow for faster analysis than previous techniques [2, 8, 9, 11-13, 16-59].

Table 2: Bioanalytical methods for the estimation of IRI in samples

Year Drug Method Matrix Sample preparation Column/Mobile phase Retention time Detection Linearity Ref.
2018 IRI HPLC-FLD Dried blood spots Liquid extraction

Column: Eclipse plus C8 column (150 × 4.6 mm, 5 µm particle size)

Mobile phase: 0.1 M phosphate buffer with pH 4.0 and ACN (80:20 %v/v)

8.20 min Excitation wavelength: 370 nm, Emission wavelength: 420 nm 10 to 3000 ng/ml [16]
2020 IRI, SN-38 HPLC-FLD Plasma and tissue homogenates (kidney, spleen, small intestine, liver, colon) Protein precipitation

Column: Zorbax SB C18 column (150 × 2.1 mm ID, 5 µm particle size) with C18 guard (Phenomenex, USA) column (4 × 2 mm)

Elution: Gradient

Mobile phase: 20 mmol ammonium acetate buffer with pH 3.5 (Eluent A)

ACN (Eluent B)

Between 4.5 to 5.0 min Excitation wavelength: 368 nm, Emission wavelength: 515 nm 7.5 to 1500 ng/ml (IRI),5 to 1000 ng/ml (SN-38) [17]
2018 IRI HPLC-DAD Plasma Precipitation of protein sample

Column: Zorbax
Extend C18 column (250 mm × 4.6 mm; 5 µm) with security guard column (4.0 × 3.0 mm; 5 µm particle size)

Elution: Gradient

Mobile phase: Milli Q water (Solvent A) and methanol (Solvent B)

8.97 min 376 nm 0.1 to 1.8 µg/ml [19]
2019 Irinotecan in a liposomal form (IRI,SN-38 and SN-38G) Triple quadrapole mass analyser with ESI Blank plasma, blank liver homogenates, and homogenates mixture of blank tissue (heart, spleen, lung, and kidney) Solid Phase extraction

Column:

UPLC BEH C18 (Waters, USA) column (2.1 mm × 50 mm; 1.7 µm particle size)

Elution: Isocratic

Mobile phase: ACN: 0.1% formic acid in water (27:73) (%v/v)

0.67 min (CPT-11), 1.63 min (IS) and 1.28 min (SN-38)

m/z 393.2 → 349 (SN-38)

m/z 587.4 → 167.1 (IRI)

m/z 349.2 → 249.0 (IS)

10 to 10,000 ng/ml (Free IRI), 4.4 to 20,000 ng/ml (Total IRI), 5 to 1000 ng/ml (SN-38), 25 to 1000 ng/ml (Others) [20]
2016

Irinotecan, SN-38 glucuronide (SN-38G),

SN-38

UPLC-MS-MS Biological matrices-Plasma, urine, faeces, and tissues are all samples that can be taken (liver and kidney) Liquid-liquid extraction

Column: Acquity UPLC BEH C18 column (2.1 mm × 50 mm, 300 Å, 1.7 µm particle size)

Elution: Gradient

Mobile phase:

0.1% formic acid (Solvent A) and ACN (Solvent B)

1.8 to 2.0 min (for IRI); 2.1 to 2.2 min (for SN-38); and 1.6 to 1.8 min (for SN-38G) m/z 587.6-124.04 (for IRI); 393.1-349.06 (for SN-38); and 569.05-393 (for SN-38G)

In plasma samples:

4.88 to 10,000 nM (for IRI),

4.88 to 10,000 nM (for SN-38), 6.25 to 2,000 nM (for SN-38G); In faeces samples: 39 to 5,000 nM (for IRI), 39 to 5,000 nM (for SN-38), 4.88 to 1,250 nM (for SN-38G); In liver and kidney samples: 48.8 to 6,250 nM (for IRI), 48.8 to 6,250 nM (for SN-38), 9.8 to 1,250 nM (for SN-38G).

[21]
2012 IRI, SN-38, and SN-38G UPLC–MS-MS Blank human plasma in cryovials Solid-phase extraction

Column: Acquity UPLC BEH RP18 column-(2.1 × 50 mm, 1.7 µm)
Elution: Gradient

Mobile phase: 0.1% formic acid (Solvent A)

MeOH (Solvent B).

0.8 min (IRI), 1.1 min (SN-38G), 1.4 min (Camptothecin), and 1.57 min (SN-38)

Precursor ion m/z: 587.3 (IRI), 393.2 (SN-38), 569.2 (SN-38-G), and 349.1 (Camptothecin).

Product ion m/z: 124.2 (IRI), 349.2 (SN-38), 393.2 (SN-38G), and 305.2 (Camptothecin)

5 to 1,000 ng/ml for IRI; 0.5–100 ng/ml for SN-38 and SN-38G [22]
2010 IRI, SN-38 HPLC-FLD Plasma Single-step protein precipitation with ACN

Column: Wakosil II C18 RS column (250 mm × 4.5 mm, 5 µm particle size)

Elution: Isocratic

Mobile phase: 36 mmol sodium dihydrogen phosphate dehydrate and 4 mmol sodium 1 heptane sulfonate: ACN (72:28) (%v/v)

4.70 min (for IRI),

6.5 min (for SN-38), and 8.00 min for (CPT)

Excitation wavelength: 355 nm, Emission wavelength: 515 nm 5 to 5,000 ng/ml (IRI) and 5 to 240 ng/ml (SN-38) [23]
2015 IRI HPLC-UV Tissue Tyrode’s
solution collected after perfusion of intestinal segment and Permeation through everted gut sac

Column: Supelco RP C18 column-(250 mm × 4.6 mm, 5 µm particle size)

Elution: Isocratic

Mobile phase: 0.045 µM sodium dihydrogen phosphate dehydrate buffer with pH 3.0: ACN (72:28) (%v/v)

8.75±0.15 min 254.9 nm 0.060-10.0 µg/ml [24]
2016 IRI, SN-38, and SN38-G HPLC-MS-MS Blood, Plasma Protein precipitation

Column: Thermo Scientific Hypersil GOLD column (50 × 2.1 mm, 3 µm particle size)

Mobile phase: 0.1% formic acid in water: 0.1% formic acid in ACN (80:20) (%v/v).

4.57 min (for IRI),

4.67 min (for SN-38), and 4.68 min (for Camptothecin)

m/z 587.3→124.1 (for IRI),

m/z 393.2→349.3 (for SN-38), and 349.1→305.1 (for Camptothecin)

25–2500 ng/ml (IRI) and 5–500 ng/ml (SN-38) [25]
2012 IRI, SN-38 UPLC-MS-MS Plasma and brain Liquid-liquid extraction

Column: Waters Acqity UPLC BEH C18 column (2.1 mm × 50 mm, 1.7 µm particle size)

Elution: Gradient

Mobile phase-10 mmol Ammonium acetate buffer (pH 3.5) with 0.1% formic acid (Solvent A) and ACN (Solvent B)

1.8 min (for IRI),

2.1 min (for SN-38), and 2.3 min (for IS)

m/z 587.49→167.11 (for IRI Lactone)

m/z 393.37→249.23 (for SN-38), and m/z 349.28→305.30 (for IS)

Both IRI and SN-38:

5–5,000 ng/ml (Plasma) and 1.25–1,250 ng/g (Brain)

[27]
2021 IRI, SN-38, and SN38-G LC-MS-MS Plasma and tissue samples Protein precipitation

Colum: Waters Symmetry C18 (150 × 3.9 mm, 5 µm, particle size) column

Elution: Gradient

Mobile phase:

Mobile phase A-MeOH-water solution (10:90), containing 0.1 % formic acid and 0.1 % ammonium formate (%v/v)

Mobile phase B-MeOH solution containing 0.1 % formic acid and 0.1 % ammonium formate

3.01 min (IRI), 3.56 min (SN-38), and 3.03 min (SN-38G). m/z 605.20 → 543.30 (for IRI), and 411.20 → 347.10 (for SN-38)

For both IRI and SN-38

9–9,000 ng/ml (for Plasma samples) and 10–4,500 ng/ml (for tissue samples)

[28]
2009 IRI, SN-38, SN-38G LC-MS-MS Plasma Protein precipitation

Column: ZORBAX SB (Agilent, USA) C18 column (100 mm × 2.1 mm, 3.5 µm particle size)

Mobile phase-water with 0.05% formic acid (Solvent A), ACN (Solvent B)

11.2 min (for IRI), 13.0 min (for SN-38), and 7.3 min (for SN-38G). m/z 587.3→ 167.1 (for IRI), m/z 393.2→ 349.3 (for SN-38), m/z 569.0→ 393.2 (for SN-38G), and m/z 349.1 → 305.1 (for Camptothecin) 10.0 to 2000.0 ng/ml (for IRI) and 0.5 to 200.0 ng/ml (for SN-38) [31]
2008 IRI, SN-38 HPLC-UV Plasma and bile Single protein precipitation (plasma) and liquid-liquid extraction (bile)

Column: YMC C-18 (ODS-A RP column)-(250 mm × 4.6 mm, 4 µm) stainless steel column.

Elution: Gradient

Mobile phase: ACN (Solvent A)

Milli-Q water, adjusted to pH 3.0 with 20% o-phosphoric acid (Solvent B)

11.4 min (for Topotecan), 13.4 min (for IRI), and 15.5 min (for SN-38) 254 nm and 365 nm (for IRI in plasma and bile, respectively), 380 nm (for SN-38)

For both IRI and SN-38:

25–10,000 ng/ml
(Plasma) and 0.5–100 µg/ml (Bile)

[32]
2014 IRI, SN-38 HPLC-MS-MS Plasma Protein precipitation

Column: XBridge C18 analytical column (50 × 2.1 mm, 3.5 µ m)

Elution: Isocratic

Mobile phase: 5 mmol ammonium formate buffer (pH 3.5): ACN (25:75) (%v/v)

1.085 min (for IRI),

1.567 min (for SN-38), and 1.934 min (for Camptothecin)

m/z 587.2→124.1 (for IRI), m/z 393.0→349.0 (for SN-38), and m/z 349.0→305.1 (for Camptothecin)

For both IRI and SN-38:

5 to 1000 ng/ml

[33]
2016 IRI MALDI-MS, LC-MS-MS Plasma Protein precipitation NA NA

m/z 587.4 →124.2 (for IRI)

m/z

349.2 → 305.1 (for IS)

300 to 10,000 ng/ml (MALDI-MS), 10–10,000 ng/ml (LC-MS-MS) [34]
2008 IRI, SN-38, 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxy-camptothecin (APC) LC-MS-MS Human liver microsomal fractions and plasma Protein precipitation

Column: Alltima C18 column (150 × 2.1 mm, 5 µm) with guard column-5 µm Alltima C18 5 µ (7.5 × 2.1 mm) cartridge.

Elution: Gradient

Mobile phase:

Water (Solvent A)

ACN (Solvent B) containing 0.25% formic acid.

4.57 min (for IRI), 4.92 min (for SN-38), 4.53 min (for APC), and 5.09 min (for IS) m/z 587.20 (for IRI), m/z 392.90 (for SN-38), m/z 619.20 (for APC), and m/z 349.20 (for IS).

Microsomal fractions:

1.56–100 ng/ml (for IRI), 3.13–150 ng/ml (for SN-38), and 0.78–100 ng/ml (for APC)

Plasma samples:

1.56–25 ng/ml (for IRI), 3.13–150 ng/ml (for SN-38), and 0.78–25 ng/ml (for APC)

[35]
1999 IRI, SN-38 LC-ES-MS Serum Protein precipitation

Column: Symmetry C18, 3.5 µm (150 × 1 mm I.D.)

Elution: Gradient

Mobile phase-Acetonitrile and 5 mmol

Ammonium formate with pH 3.0.

3.9 min (for IRI-Lactone),

3.5 min (for IRI-Carboxylate),

5.1 min (for SN-38–Lactone)

5.5 min (for AN-38–Carboxylate), and

5.4 min (for IS)

m/z 587.3 (for IRI-Lactone),

m/z 605.3 (for IRI-Carboxylate),

m/z 393.0 (for SN-38-Lactone),

m/z 411.2 (for IRI-Carboxylate), and

m/z 349.2 (for IS)

0 to 10,000 ng/ml (for IRI), and

0 to 100 ng/ml (for SN-38)

[36]
2014 IRI, SN-38 LC-MS-MS Plasma, tumours Tissue homogenization

Column: Reverse phase Waters Xterra® MS C18 column (150 × 2.1 mm)

Elution: Isocratic

Mobile phase: ACN-MeOH-buffer (5 mmol ammonium formate with 0.1% formic acid) (3:4:3)

(%v/v/v)

1.8 min (for IRI), 3.0 (for SN-38), and 3.1 min (for Camptothecin) m/z 587.6→167.2 (for IRI), 393.6→349.3 (for SN-38), and 349.4→305.2 (for Camptothecin) 50 to 10,000 ng/ml (for IRI), and 10 to 2,500 ng/ml (for SN-38) [37]
2005 IRI, SN-38, Camptothecin (active lactone and inactive carboxylate forms) HPLC-TBAHS Plasma Protein precipitation

Column: Hypersil ODS Column (200 mm × 4.6 mm, 5 µm) with

guard column-Phenomenex C18

Elution: Isocratic

Mobile phase: 0.1 M potassium dihydrogen phosphate containing 0.01 M TBAHS (pH 6.4): ACN (75:25) (%v/v).

5.33±0.14 min (for IRI lactone), 11.60±0.20 min (for SN-38 lactone),13.52±0.21 min (for Camptothecin–lactone form), 3.97±0.10 min (for carboxylate form), 7.05±0.15 min (for SN-38 carboxylate form), and 8.12±0.18 min (for camptothecin-carboxaylate form) Excitation wavelength: 380 nm and Emission wavelength: 540 nm

For all compounds:

0.01 to 10 µM

[38]
2000 IRI, SN-38, Camptothecin HPLC-FLD Plasma De-proteinisation

Column: Nucleosil C18 (Phenomenex) (250 × 4.0 mm, 5 µm particle size)

Elution: Isocratic

Mobile phase: 0.1 M Potassium dihydrogen phosphate with pH 4.2: ACN (67:33) (%v/v)

0.00 to 5.80 min (for IRI), 5.80 to 7.90 min (for SN-38), and 7.90 to 9.50 (for Camptothecin) Excitation: 228 nm and Emission: 450 nm (for IRI), 543 nm (for SN-38) and 433 nm (for Camptothecin) 1 to 10,000 ng/ml (for IRI) 0.5 to 400 ng/ml (for SN-38) [39]
2007 IRI, SN-38, Camptothecin (IS) HPLC-FLD Tissue culture media and cancer cells Solid-phase extraction

Column:

Hyperclon (Phenomenex) ODS C18 (200 mm × 4.6 mm) with Phenomenex C18 guard column

Elution: Isocratic

Mobile phase: ACN: 50 mmol disodium hydrogen phosphate buffer containing 10 mmol sodium 1-heptane-sulfonate, with pH 3.0 with 85% (w/v) ortho-phosphoric acid (27:73), (%v/v)

10.04±1.06 min (for IRI), 6.15±0.06 min (for SN-38), and 7.80±0.07 min (for Camptothecin) Excitation wavelength: 380 nm and Emission wavelength: 540 nm 5–42,000 ng/ml (IRI) and 1–1500 ng/ml (SN-38) in culture medium and 2–42,000 ng/ml (IRI) and 0.5–1500 ng/ml (SN-38) in cell lysates [40]
2013 SN-38 Microfluidic chip-based Nano-LC-MS-MS Plasma Protein precipitation

Column: Zorbax 80 SB-C18 (150 mm × 75 μm, 5 μm particle size)

Elution: Gradient

Mobile phase: (Solvent A) 0. 1% formic acid in water

(Solvent B) 0.1% formic acid in ACN.

5.325 min (for IRI)

5.067 min (for SN-38)

m/z

393.0 → 349.0 (for SN-38),

m/z

349.0 → 305.1 (for IRI)

50 to 10,000 pg/ml [42]
2005 IRI, SN-38 LC-MS-MS Plasma and tissues Protein precipitation

Method 1:

Column: Zorbax (Agilent Technologies, USA) SB-phenyl (2.0 mm × 50 mm, 5 μm)

Elution: Isocratic

Mobile phase: 20 mmol ammonium acetate with pH 3.5: ACN (65:35) (%v/v)

Method 2:

Xterra (Waters, USA) C18 (2.0 mm× 50 mm, 5 μm)

Elution: Isocratic

Mobile phase: 20 mmol ammonium acetate with pH 3.5: ACN (67:33) (%v/v))

In plasma/tissue homogenate:

1.2 to 1.4 min (for SN-38),

1.1 to 1.3 min (for IRI), and

1.4 to 1.6 min (for IS)

m/z

393.4 → 349.3 (for SN-38),

m/z

587.6 → 167.2 (for IRI), and m/z

349.3 → 305.3 (for IS)

For both IRI and SN-38:

0.5 to 500 ng/ml

[43]
2019 IRI HPLC-(MRM-IDA-EPI) Plasma Dilution

Column: Xbridge C18 column (150 mm × 4.6 mm, 3.5 μm particle size)

Mobile phase: ACN and water, both acidified with 0.1% formic acid

7.67 min Mass/Charge 587→ 502 (IRI) 0.1–50 ng/ml [44]
2013 SN-38 LC-MS-MS Micro dialysates from rat brain Protein precipitation

Column: Agilent Eclipse Plus RP-18-(2.1 mm × 100 mm, 1.8 μm)

Elution: Gradient

Mobile phase: ACN: 0.1% methanoic acid

NA Mass/Charge 393.1→349.1 0.1015-1015 ng/ml [11]
2003 IR,SN-38,SN-38G,APCand NPC HPLC-FLD Plasma and saliva Protein precipitation

Column: Xterra (Waters, USA) RP18 (250 mm × 4.6 mm, 5 μm) with guard Xterra (Waters, USA) RP18 (20 mm × 3.9 mm, 5 μm)

Mobile phase: ACN: sterile water (75:25) (%v/v) (Solvent A)

Phosphate buffer, pH 4.0 (Solvent B)

5.54 min (for SN-38G), 13.9 min (for NPC), 16.0 min (for APC), 19.6 min (for IRI) and 22.5 min (for SN-38) Excitation wavelength: 370 nm, and Emission wavelength: 470 nm for the first 24 min and then at 534 nm for the next 4 min 0.5–1000 μg/l [47]
2003 SN-38, Camptothecin (IS) LC-MS-MS Plasma containing liposome-based SN-38 Protein precipitation

Column: Synergi (Phenomene, USA) Hydro-RP column C18, (50 × 2 mm, 4 µm),

Elution-gradient elution

Mobile phase: 0.1% acetic acid (Solvent A) and ACN (Solvent B)

1.55 min (for SN-38), and 1.78 min (for IS) m/z 393.1→349.2 (for SN-38), and m/z 349.1→305.1 (for IS) 0.05–400 ng/ml [48]
2014 SN-38, Camptothecin (IS) UPLC–MS-MS Tumor cells Extraction

Column: Kinetex (Phenomenex, UK) 1.7 μm PFP, LC Column-(100 mm × 2.1 mm)

Elution: Gradient

Mobile phase: 0.1% formic acid in water (Solvent A) and 0.1% formic acid in ACN (Solvent B)

1.75 min (for SN-38), and 1.89 min (for IS) m/z 393.1→349.4 (for SN-38), and m/z 349.1→305.4 (for IS) 0.1–1,000 ng/ml [9]
2015 IRI, SN-38, camptothecin, (IS), tegafur (TF), 5-fluorouracil (5-FU), and leucovorin (LV) Capillary electrophoresis with UV and fluorimetric detection Urine Solid-supported liquid-liquid extraction

Capillary:

Fused-silica (BGB Analytik, Switzerland) capillary of (58.5 cm × 75 µm id × 375 µm od)

Solvents for conditioning:

1.0 M NaOH, ultrapure water, and BGE (phosphate buffer, pH 11.34; 20 mmol)

Eluted with:

Ethyl acetate: methanol ratio (95:5), (%v/v)

IF: 2.5 to 2.7 min (for IRI), 2.8 to 3.0 min (for IS), and 3.8 to 4.0 min (for SN-38)

A265:3.1 to 3.3 min (for IRI), 3.5 to 3.7 min (for IS), and 4.1 to 4.2 min (for TF), 4.5 to 4.7 min (for SN-38), 5.1 to 5.2 min (for 5-FU), and 5.8 5.9 min (for LV)

360 nm (for IRI), 410 nm (for SN-38), 265 nm (for 5-FU), 272 nm (for TF) and 288 nm (for LV)

Excitation: 360 nm (IRI) and Emission: 440 nm (IRI). Excitation: 340 and
410 nm (SN-38), and Emission: 548 nm (SN-38)

1 to 30 mg/l (for IRI and LV), 2 to 20 mg/l (for TF and 5-FU) and 0.05 to 3.5 mg/l (for SN-38) [50]
1997 IRI, SN-38, and Camptothecin (IS) RP-HPLC-FLD Plasma Protein precipitation

Column: Hypersil (LC Service, The Netherlands) ODS Column-(100 × 4.6 mm, 5 μm), With guard column-LiChroCART 4-4 end-capped pre-column (4 × 4 mm, 5 μm) LiChrospher 100 RP-18 material (Merck), Elution: Isocratic

Mobile phase: MeOH-0.1 M, ammonium acetate containing 0.01 M tetrabutylammonium sulphate (40:60) (%v/v) for the lactone forms, and (35:65) (%v/v) for the total forms with the pH 5.5 adjusted using hydrochloric acid.

For lactone forms:

4.9 min (for IRI), 8.1 min (for SN-38) and 6.5 min (for IS); and

For total forms:

8.3 min (for IRI) and 15.0 min (for SN-38)

Excitation wavelength: 355 nm, and Emission wavelength: 515 nm

For lactone form of IRI and SN-38: 0.5 to 15 ng/ml

For total forms of both IRI and SN-38: 2.0 to 200 ng/ml

[13]
1998 IRI, SN-38, SN-38G, and 7-ethyl-10[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin (APC) HPLC-FLD Plasma, urine, and faeces Protein precipitation

Column: Hypersil (LC Service, Emmen, The Netherlands) ODS (100 × 4.6 mm, 5 μm)with LiChroCart 4-4 end-capped (RP 18) guard column

Elution: Isocratic

Mobile phase: MeOH: 0.1 M ammonium acetate containing 10 mmol tetrabutylammonium sulphate (30:70), (%v/v), pH adjusted to 5.3 with hydrochloric acid.

6.95±0.21 min (for SN-38G), 11.86±1.27 min (for APC), 16.1±2.55 min (for IRI) and 28.6±1.77 min (for SN-38) Excitation wavelength: 355 nm Emission wavelength: 515 nm

For plasma:

10 to 400 ng/ml (for IRI and SN-38), and

For urine and faeces:

100 to 5,000 ng/ml) (for IRI and SN-38)

[54]
2015 IRI, SN-38, SN-38G, camptothecin (IS) and APC HPLC-MS-MS Plasma Extraction

Column: Gemini (Phenomenex, Torrance, USA) C18 (100 × 2.0 mm, 110 Å, 3 μM) with

Security guard cartridge (Gemini-NX C18; 4.0 × 2.0 mm)

Mobile phase: 0.1% acetic acid (CH3COOH)/Bi-distilled water (Solvent A)

0.1% CH3COOH/ACN (Solvent B)

5.05 min (for IRI), 6.43 min (for SN-38), 7.90 min (for SN-38G), 5.07 min (for APC), and 6.57 min (for IS)

IRI-Mass/Charge 587.4>124.2

SN-38-Mass/Charge 393.3>349.3

SN-38G-Mass/Charge 569.3>393.2

APC-Mass/Charge 619.2>393.3

10 to 10,000 ng/ml (for IRI), 1 to 500 ng/ml (for SN-38 and SN-38G) and

1 to 5,000 ng/ml (for APC)

[59]

DISCUSSION

Irinotecan is a semi-synthetic analogue of camptothecin, a quinolone-based alkaloid found in the native Chinese/Tibetan ornamental tree Camptotheca acuminata, also known as Xi Shu in Chinese and Tibetan ("happy tree"). Irinotecan is a prodrug that is metabolised to its active metabolite SN-38 (7-ethyl-10-hydroxycamptothecin) by a carboxylesterase enzyme found mostly in the liver, gut mucosa, and tumour tissue. SN-38 is a powerful inhibitor of the nuclear enzyme topoisomerase I, which is involved in a variety of key nuclear functions, including DNA replication, and is 100 to 1,000 times more hazardous than irinotecan [60] CPT-11 can also be metabolised through cytochrome P-450 (CYP) 3A-mediated oxidation pathways. 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1 piperidino] 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1 piperidino] 7-ethyl-10-carbonyloxycamptothecin (APC) and carbonyloxycamptothecin (APC) (4-amino-1-piperidine). The principal oxidation products catalysed by CYP3A4 are carbonyl oxycamptothecin (NPC), both of which are weak inhibitors of topoisomerase [61].

Irinotecan was discovered and produced in 1983. In the laboratories, it initially demonstrated promising effects against a wide range of cancers [62]. Phase of clinical trials I began my research in Japan in 1986, Europe in 1990, and the United States in 1991. In Japan, irinotecan was first approved in 1994 for the treatment of non-small-cell lung cancer, small-cell lung cancer, and gynaecologic malignancies. In 1995, irinotecan was approved as a second-line treatment for colorectal cancer in France, and a year later in the United States [60].

HPLC with fluorescence or UV detector is the most often used analytical technique [65] for quantifying IRI and its active metabolites in pharmaceutical dosage form, pure form, and plasma [65,66]. TLC, RP-HPLC, HPLC-MS-MS, LC-MS-MS, and UV spectrophotometric approaches have all been reported for quantifying IRI and its metabolites in pure form and in combination with other drugs in bulk and pharmaceutical formulations. In this review article, UV detector wavelengths range from 220 to 400 nm, while fluorescence detector emission wavelengths range from 450 to 550 nm and excitation wavelengths from 355 to 400 nm.

This article reviews several HPLC methods, including HPLC-UV, HPLC-FLD, HPLC-DAD, HPLC-MS-MS, and HPLC-TBAHS; UPLC methods, including UPLC-MS-MS and UPLC-UV; RP-HPLC methods, including RP-HPLC-FLD and RP-HPLC-UV; LC-MS-MS methods, including microfluidic chip-based Nano-LC-MS-MS and LC-MS-MS, MALDI-MS-MS method, capillary electrophoresis with UV and fluorometric detection, and Triple quadrupole mass spectrometer with electrospray ionization methods. Also reviews the simple sample preparation techniques such as solid-phase extraction, liquid-liquid extractions, and protein precipitation, various columns and mobile phase, the retention time of each method, and linearity range of the various methods.

CONCLUSION

Irinotecan is the drug of choice and is frequently used as monotherapy and adjuvant therapy in the treatment of metastatic colorectal cancer. Irinotecan must be estimated in analytical and bioanalytical samples, because it is structurally and pharmacologically distinct from other camptothecin analogues. This study covers a wide range of analytical methods for estimating Irinotecan in analytical and bioanalytical samples as a separate entity, as well as its metabolites as alone and in combination with other anticancer drugs. Over time, a variety of analytical techniques have been developed, including UV visible spectrophotometry, spectrofluorimetric techniques, chromatographic techniques, and hyphenated procedures. LC-MS-MS, HPLC-FLD, HPLC-DAD, HPLC-UV, and RP-HPLC-UV are hyphenated techniques that combine chromatographic and spectral techniques to enable for facile separation and detection of even minute fractions of the drug in both pure and combined form. The recent development in this area includes the development of the sensitive UHPLC-MS-MS method for more accurate and reliable results.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICT OF INTERESTS

Declared none

REFERENCES

  1. Ayoub Z, Mehta A, Mishra SK. Ethnopharmacological review of natural products in cancer prevention and therapy. Asian J Pharm Clin Res. 2018;11(6):32-44, doi: 10.22159/ajpcr.2018.v11i6.24792.

  2. Konda RK, Chandu BR, Chandra Sekhar KB. An improved HPLC-UV method for the estimation of irinotecan in bulk and tablet dosage form. Int J Pharm Biomed Res. 2011;2(3):188-91.

  3. Bhaskaran NA, Jitta SR, Salwa, Cheruku S, Kumar N, Kumar L. Orally delivered solid lipid nanoparticles of irinotecan coupled with chitosan surface modification to treat colon cancer: preparation, in vitro and in vivo evaluations. Int J Biol Macromol. 2022;211:301-15. doi: 10.1016/j.ijbiomac.2022.05.060, PMID 35568152.

  4. Bailly C. Irinotecan: 25 y of cancer treatment. Pharmacol Res. 2019;148:104398. doi: 10.1016/j.phrs.2019.104398, PMID 31415916.

  5. Tariq M, Negi LM, Talegaonkar S, Ahmad FJ, Iqbal Z, Khan AM. Liquid chromatographic method for irinotecan estimation: screening of p-gp modulators. Indian J Pharm Sci. 2015;77(1):14-23. doi: 10.4103/0250-474x.151577, PMID 25767314.

  6. Irinotecan; 2021. PubChem. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/irinotecan. [Last accessed on 01 Nov 2021]

  7. Chabot GG. Clinical pharmacokinetics of irinotecan. Clin Pharmacokinet. 1997;33(4):245-59. doi: 10.2165/00003088-199733040-00001, PMID 9342501.

  8. Sharma S, Sharma MC. Development and validation of spectrophotometric method and TLC densitometric determination of irinotecan HCl in pharmaceutical dosage forms. Arab J Chem. 2016;9:S1368-72. doi: 10.1016/j.arabjc.2012.02.012, PMID 23781456.

  9. Ghazaly E, Perry J, Kitromilidou C, Powles T, Joel S. Development and validation of an ultra-high performance LC-MS/MS assay for intracellular SN-38 in human solid tumour cell lines: comparison with a validated HPLC-fluorescence method. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;969:213-8. doi: 10.1016/j.jchromb.2014.08.024, PMID 25195021.

  10. Nussbaumer S, Geiser L, Sadeghipour F, Hochstrasser D, Bonnabry P, Veuthey JL. Wipe sampling procedure coupled to LC-MS/MS analysis for the simultaneous determination of 10 cytotoxic drugs on different surfaces. Anal Bioanal Chem. 2012;402(8):2499-509. doi: 10.1007/s00216-011-5157-2, PMID 21701850.

  11. Cao Y, Cheng Q, Lu H, Zhang H. Determination of 7-ethyl-10-hydroxycamptothecin in microdialysates from rat brain with LC-MS/MS. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2013;42(1):98-102. doi: 10.3785/j.issn.1008-9292.2013.01.016, PMID 23505115.

  12. Bhaskaran NA, Kumar L, Reddy MS, Pai GK. An analytical ”quality by design” approach in RP-HPLC method development and validation for reliable and rapid estimation of irinotecan in an injectable formulation. Acta Pharm. 2021;71(1):57-79. doi: 10.2478/acph-2021-0008, PMID 32697749.

  13. de Bruijn P, Verweij J, Loos WJ, Nooter K, Stoter G, Sparreboom A. Determination of irinotecan (CPT-11) and its active metabolite SN-38 in human plasma by reversed-phase high-performance liquid chromatography with fluorescence detection. J Chromatogr B Biomed Sci Appl. 1997;698(1-2):277-85. doi: 10.1016/s0378-4347(97)00290-9, PMID 9367218.

  14. Souza DM, Reichert JF, Martins AF. A simultaneous determination of anti-cancer drugs in hospital effluent by DLLME HPLC-FLD, together with a risk assessment. Chemosphere. 2018;201:178-88. doi: 10.1016/j.chemosphere.2018.02.164, PMID 29524818.

  15. Wal P, Kumar B, Bhandari A, Rai AK, Wal A. Bioanalytical method development–determination of drugs in biological fluids. J Pharm Sci Technol. 2010;2(10):333-47.

  16. Hahn RZ, Arnhold PC, Andriguetti NB, Schneider A, Kluck HM, Dos Reis SL. Determination of irinotecan and its metabolite SN-38 in dried blood spots using high-performance liquid-chromatography with fluorescence detection. J Pharm Biomed Anal. 2018;150:51-8. doi: 10.1016/j.jpba.2017.11.079, PMID 29216585.

  17. Martinez Chavez A, Rosing H, Gan C, Wang Y, Schinkel AH, Beijnen JH. Bioanalytical method for the simultaneous quantification of irinotecan and its active metabolite SN-38 in mouse plasma and tissue homogenates using HPLC-fluorescence. J Chromatogr B Analyt Technol Biomed Life Sci. 2020;1149:122177. doi: 10.1016/j.jchromb.2020.122177, PMID 32464539.

  18. Sanogo S, Silimbani P, Gaggeri R, Masini C. Development and validation of an HPLC-DAD method for the simultaneous identification and quantification of Topotecan, Irinotecan, Etoposide, Doxorubicin and Epirubicin. Arab J Chem. 2021;14(1):102896. doi: 10.1016/j.arabjc.2020.11.002.

  19. Malatesta L, Cosco D, Paolino D, Cilurzo F, Costa N, Di Tullio A. Simultaneous quantification of Gemcitabine and Irinotecan hydrochloride in rat plasma by using high performance liquid chromatography-diode array detector. J Pharm Biomed Anal. 2018;159:192-9. doi: 10.1016/j.jpba.2018.06.060, PMID 29990886.

  20. Yang W, Yang Z, Liu J, Liu D, Wang Y. Development of a method to quantify total and free irinotecan and 7-ethyl-10-hydroxycamptothecin (SN-38) for pharmacokinetic and bio-distribution studies after administration of irinotecan liposomal formulation. Asian J Pharm Sci. 2019;14(6):687-97. doi: 10.1016/j.ajps.2018.08.003. PMID 32104495.

  21. Basu S, Zeng M, Yin T, Gao S, Hu M. Development and validation of an UPLC-MS/MS method for the quantification of irinotecan, SN-38 and SN-38 glucuronide in plasma, urine, feces, liver and kidney: application to a pharmacokinetic study of irinotecan in rats. J Chromatogr B Analyt Technol Biomed Life Sci. 2016;1015-1016:34-41. doi: 10.1016/j.jchromb.2016.02.012, PMID 26894853.

  22. Chen X, Peer CJ, Alfaro R, Tian T, Spencer SD, Figg WD. Quantification of irinotecan, SN38, and SN38G in human and porcine plasma by ultra-high-performance liquid chromatography–tandem mass spectrometry and its application to hepatic chemoembolization. J Pharm Biomed Anal. 2012;62:140-48. doi: 10.1016/j.jpba.2012.01.008, PMID 22305081.

  23. Baylatry MT, Joly AC, Pelage JP, Bengrine Lefevre L, Prugnaud JL, Laurent A. Simple liquid chromatography method for the quantification of irinotecan and SN38 in sheep plasma: application to in vivo pharmacokinetics after pulmonary artery chemoembolization using drug eluting beads. J Chromatogr B Analyt Technol Biomed Life Sci. 2010;878(9-10):738-42. doi: 10.1016/j.jchromb.2010.01.017, PMID 20171941.

  24. Tariq M, Negi LM, Talegaonkar S, Ahmad FJ, Iqbal Z, Khan AM. Liquid chromatographic method for irinotecan estimation: screening of p-gp modulators. Indian J Pharm Sci. 2015;77(1):14-23. doi: 10.4103/0250-474x.151577, PMID 25767314.

  25. Herviou P, Richard D, Roche L, Pinguet J, Libert F, Eschalier A. Determination of irinotecan and SN38 in human plasma by TurboFlow™ liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal. 2016;118:284-91. doi: 10.1016/j.jpba.2015.10.044, PMID 26580826.

  26. Kumar N, Sangeetha D, Reddy SP. UPLC and LC-MS studies on degradation behavior of irinotecan hydrochloride and development of a validated stability-indicating ultra-performance liquid chromatographic method for determination of irinotecan hydrochloride and its impurities in pharmaceutical dosage forms. J Chromatogr Sci. 2012;50(9):810-9. doi: 10.1093/chromsci/bms075, PMID 22661461.

  27. Goldwirt L, Lemaitre F, Zahr N, Farinotti R, Fernandez C. A new UPLC-MS/MS method for the determination of irinotecan and 7-ethyl-10-hydroxycamptothecin (SN-38) in mice: application to plasma and brain pharmacokinetics. J Pharm Biomed Anal. 2012;66:325-33. doi: 10.1016/j.jpba.2012.04.003, PMID 22551773.

  28. Chen Y, Hu Z, Qi W, Gao S, Jiang J, Wang S. Pharmacovigilance of herb-drug interactions: a pharmacokinetic study on the combination administration of herbal Kang’ai injection and chemotherapy irinotecan hydrochloride injection by LC-MS/MS. J Pharm Biomed Anal. 2021;194:113784. doi: 10.1016/j.jpba.2020.113784, PMID 33280996.

  29. Tsotsou GE, Gkotzamani P, Petro V, Argyropoulou A, Karkalousos P. A simple, rapid and low-cost spectrophotometric method for irinotecan quantification in human plasma and in pharmaceutical dosage forms. Anal Methods. 2021;13(2):258-66. doi: 10.1039/d0ay02201b, PMID 33367449.

  30. Gosetti F, Belay MH, Marengo E, Robotti E. Development and validation of a UHPLC-MS/MS method for the identification of irinotecan photodegradation products in water samples. Environ Pollut. 2020;256:113370. doi: 10.1016/j.envpol.2019.113370, PMID 31662244.

  31. Zhang W, Dutschman GE, Li X, Ye M, Cheng YC. Quantitation of irinotecan and its two major metabolites using a liquid chromatography–electrospray ionization tandem mass spectrometric. J Chromatogr B Analyt Technol Biomed Life Sci. 2009;877(27):3038-44. doi: 10.1016/j.jchromb.2009.07.025, PMID 19648066.

  32. Bansal T, Awasthi A, Jaggi M, Khar RK, Talegaonkar S. Development and validation of reversed phase liquid chromatographic method utilizing ultraviolet detection for quantification of irinotecan (CPT-11) and its active metabolite, SN-38, in rat plasma and bile samples: application to pharmacokinetic studies. Talanta. 2008;76(5):1015-21. doi: 10.1016/j.talanta.2008.04.058, PMID 18761148.

  33. Ahn G, Park DM, Park JW, Kim HY, Cho JY, Rhee SJ. A rapid, simple and reliable HPLC-triple quadrupole tandem mass spectrometer method for a simultaneous quantification of irinotecan and its active metabolite 7-ethyl-10-hydroxycamptothecin (SN38) in mouse plasma. Biomed Chromatogr. 2014;28(7):919-22. doi: 10.1002/bmc.3134, PMID 24458571.

  34. Calandra E, Posocco B, Crotti S, Marangon E, Giodini L, Nitti D. Cross-validation of a mass spectrometric-based method for the therapeutic drug monitoring of irinotecan: implementation of matrix-assisted laser desorption/ionization mass spectrometry in pharmacokinetic measurements. Anal Bioanal Chem. 2016;408(19):5369-77. doi: 10.1007/s00216-016-9634-5, PMID 27235158.

  35. D’Esposito F, Tattam BN, Ramzan I, Murray M. A liquid chromatography/electrospray ionization mass spectrometry (LC-MS/MS) assay for the determination of irinotecan (CPT-11) and its two major metabolites in human liver microsomal incubations and human plasma samples. J Chromatogr B Analyt Technol Biomed Life Sci. 2008;875(2):522-30. doi: 10.1016/j.jchromb.2008.10.011, PMID 18952506.

  36. Ragot S, Marquet P, Lachâtre F, Rousseau A, Lacassie E, Gaulier JM. Sensitive determination of irinotecan (CPT-11) and its active metabolite SN-38 in human serum using liquid chromatography-electrospray mass spectrometry. J Chromatogr B Biomed Sci Appl. 1999;736(1-2):175-84. doi: 10.1016/s0378-4347(99)00452-1, PMID 10676997.

  37. Park DJ, Won JH, Cho AR, Yun HJ, Heo JH, Hwhang TH. Determination of irinotecan and its metabolite SN-38 in rabbit plasma and tumors using a validated method of tandem mass spectrometry coupled with liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;962:147-52. doi: 10.1016/j.jchromb.2014.05.042, PMID 24927278.

  38. Yang X, Hu Z, Chan SY, Goh BC, Duan W, Chan E. Simultaneous determination of the lactone and carboxylate forms of irinotecan (CPT-11) and its active metabolite SN-38 by high-performance liquid chromatography: application to plasma pharmacokinetic studies in the rat. J Chromatogr B Analyt Technol Biomed Life Sci. 2005;821(2):221-8. doi: 10.1016/j.jchromb.2005.05.010, PMID 15936253.

  39. Escoriaza J, Aldaz A, Castellanos C, Calvo E, Giraldez J. Simple and rapid determination of irinotecan and its metabolite SN-38 in plasma by high-performance liquid-chromatography: application to clinical pharmacokinetic studies. J Chromatogr B Biomed Sci Appl. 2000;740(2):159-68. doi: 10.1016/s0378-4347(00)00048-7, PMID 10821401.

  40. Hu ZP, Yang XX, Chen X, Chan E, Duan W, Zhou SF. Simultaneous determination of irinotecan (CPT-11) and SN-38 in tissue culture media and cancer cells by high performance liquid chromatography: application to cellular metabolism and accumulation studies. J Chromatogr B Analyt Technol Biomed Life Sci. 2007;850(1-2):575-80. doi: 10.1016/j.jchromb.2006.12.056, PMID 17270505.

  41. Guichard N, Rudaz S, Bonnabry P, Fleury Souverain S. Validation and uncertainty estimation for trace amounts determination of 25 drugs used in hospital chemotherapy compounding units. J Pharm Biomed Anal. 2019;172:139-48. doi: 10.1016/j.jpba.2019.04.042, PMID 31035095.

  42. Ahn G, Park DM, Park JW, Cho JY, Rhee SJ, Kim HY. Development and validation of a microfluidic chip-based nano-liquid chromatography–triple quadrupole tandem mass spectrometry method for a sensitive and reliable quantification of 7-ethyl-10-hydroxycamptothecin (SN38) in mouse plasma. Anal Bioanal Chem. 2013;405(30):9817-24. doi: 10.1007/s00216-013-7411-2, PMID 24126840.

  43. Bardin S, Guo W, Johnson JL, Khan S, Ahmad A, Duggan JX. Liquid chromatographic-tandem mass spectrometric assay for the simultaneous quantification of Camptosar and its metabolite SN-38 in mouse plasma and tissues. J Chromatogr A. 2005;1073(1-2):249-55. doi: 10.1016/j.chroma.2004.08.060, PMID 15909526.

  44. Shu P, Zhao T, Wen B, Mendelsohn Victor K, Sun D, Friese CR. Application of an innovative high-throughput liquid chromatography-tandem mass spectrometry method for simultaneous analysis of 18 hazardous drugs to rule out accidental acute chemotherapy exposures in health care workers. J Oncol Pharm Pract. 2020;26(4):794-802. doi: 10.1177/1078155219870591, PMID 31483750.

  45. Maeda S, Miwa Y. Multicomponent high-performance liquid chromatography/tandem mass spectrometry analysis of ten chemotherapeutic drugs in wipe samples. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;921-922:43-8. doi: 10.1016/j.jchromb.2013.01.014, PMID 23419953.

  46. Xuan T, Zhang JA, Ahmad I. HPLC method for determination of SN-38 content and SN-38 entrapment efficiency in a novel liposome-based formulation, LE-SN38. J Pharm Biomed Anal. 2006;41(2):582-8. doi: 10.1016/j.jpba.2005.10.051, PMID 16386867.

  47. Poujol S, Pinguet F, Malosse F, Astre C, Ychou M, Culine S. Sensitive HPLC-fluorescence method for irinotecan and four major metabolites in human plasma and saliva: application to pharmacokinetic studies. Clin Chem. 2003;49(11):1900-8. doi: 10.1373/clinchem.2003.023481, PMID 14578322.

  48. Khan S, Ahmad A, Ahmad I. A sensitive and rapid liquid chromatography tandem mass spectrometry method for quantitative determination of 7-ethyl-10-hydroxycamptothecin (SN-38) in human plasma containing liposome-based SN-38 (LE-SN38). Biomed Chromatogr. 2003;17(8):493-9. doi: 10.1002/bmc.257, PMID 14648604.

  49. Brachet G, Bruno C, Boulay D, Tournamille JF, Gyan E, Viaud Massuard MC. An ion-pairing, reversed-phase liquid chromatography method to assess the cross-contamination of cancer chemotherapy infusions prepared in a dual-operator aseptic isolator. Drug Test Anal. 2016;8(9):985-90. doi: 10.1002/dta.1902, PMID 26480955.

  50. Hurtado Sanchez M, Acedo Valenzuela MI, Duran Meras I, Rodriguez Caceres MI. Determination of chemotherapeutic drugs in human urine by capillary electrophoresis with UV and fluorimetric detection using solid-supported liquid-liquid extraction for sample clean-up. J Sep Sci. 2015;38(11):1990-7. doi: 10.1002/jssc.201401443, PMID 25820908.

  51. Nussbaumer S, Geiser L, Sadeghipour F, Hochstrasser D, Bonnabry P, Veuthey JL. Wipe sampling procedure coupled to LC-MS/MS analysis for the simultaneous determination of 10 cytotoxic drugs on different surfaces. Anal Bioanal Chem. 2012;402(8):2499-509. doi: 10.1007/s00216-011-5157-2, PMID 21701850.

  52. Balaram VM, Rao JV, Ganesh RSS, Krishna TB. Validated reverse phase HPLC method for the determination of irinotecan in pharmaceutical dosage forms. E-Journal of Chemistry. 2007;4(1):128-36. doi: 10.1155/2007/597409.

  53. Shende P, Gaud R. Validated RP-HPLC analysis of irinotecan HCl in the bulk material and in pharmaceutical formulations. Acta Chromatogr. 2009;21(1):71-82. doi: 10.1556/AChrom.21.2009.1.6.

  54. Sparreboom A, de Bruijn P, de Jonge MJ, Loos WJ, Stoter G, Verweij J. Liquid chromatographic determination of irinotecan and three major metabolites in human plasma, urine and feces. J Chromatogr B Biomed Sci Appl. 1998;712(1-2):225-35. doi: 10.1016/s0378-4347(98)00147-9, PMID 9698245.

  55. Reddy PS, Babu KS, Kumar N. A validated RP-HPLC method for the determination of irinotecan hydrochloride residues for cleaning validation area. Rev Cuba Farm. 2013;47(1):41-50.

  56. Kumudhavalli MV, Prakash C, Venkateshwarlu BS, Kumar M. A novel technique for the quantification of irinotecan HCl injection. Res J Pharm Technol. 2019;12(12):6027-30. doi: 10.5958/0974-360X.2019.01046.1.

  57. Xiao H, Sedlarik V. A rapid and sensitive HPLC method for simultaneous determination of irinotecan hydrochloride and curcumin in co-delivered polymeric nanoparticles. J Chromatogr Sci. 2020;58(7):651-60. doi: 10.1093/chromsci/bmaa033, PMID 32627829.

  58. Kumar N, Sangeetha D, Reddy SP. UPLC and LC-MS studies on degradation behavior of irinotecan hydrochloride and development of a validated stability-indicating ultra-performance liquid chromatographic method for determination of irinotecan hydrochloride and its impurities in pharmaceutical dosage forms. J Chromatogr Sci. 2012;50(9):810-9. doi: 10.1093/chromsci/bms075, PMID 22661461.

  59. Marangon E, Posocco B, Mazzega E, Toffoli G. Development and validation of a high-performance liquid chromatography-tandem mass spectrometry method for the simultaneous determination of irinotecan and its main metabolites in human plasma and its application in a clinical pharmacokinetic study. PLOS ONE. 2015;10(2):e0118194. doi: 10.1371/journal.pone.0118194, PMID 25689738.

  60. Fukuoka M. Current status of irinotecan in lung cancer. Oncology (Williston Park). 2001;15 Suppl 1:6-7. PMID 11221022.

  61. Sai K, Kaniwa N, Ozawa S, Sawada JI. An analytical method for irinotecan (CPT‐11) and its metabolites using a high‐performance liquid chromatography: parallel detection with fluorescence and mass spectrometry. Biomed Chromatogr. 2002;16(3):209-18. doi: 10.1002/bmc.137, PMID 11920947.

  62. Kunimoto T, Nitta K, Tanaka T, Uehara N, Baba H, Takeuchi M. Antitumor activity of 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothec in, a novel water-soluble derivative of camptothecin, against murine tumors. Cancer Res. 1987;47(22):5944-7. PMID 3664496.

  63. USP monograph of irinotecan hydrochloride injection, USP-NF/PF abstract; 2023. doi: 10.31003/USPNF_M42489_04_01.

  64. USP monograph of irinotecan hydrochloride, USP-NF/PF abstract; 2023. doi: 10.31003/USPNF_M42485_05_01.

  65. Rodriguez ET, Szijj JV, Cachia M, Falzon P, Axisa K, Serracino Inglott AS. An efficient HPLC-UV method for determination of tetrahydrocannabinol in oil. Asian J Pharm Clin Res. 2023;16(3):110-15. doi: 10.22159/ajpcr.2023.v16i3.47462.

  66. Damle MC, Sonule JA. Hydrolytic degradation study of roxadustat by RP-HPLC and HPTLC. Int J Pharm Pharm Sci. 2023;15(8):36-49. doi: 10.22159/ijpps.2023v15i8.48355.