Previous Article in Journal
Multimodal Wearable Monitoring of Exercise in Isolated, Confined, and Extreme Environments: A Standardized Method
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

RP-DAD-HPLC Method for Quantitative Analysis of Clofazimine and Pyrazinamide for Inclusion in Fixed-Dose Combination Topical Drug Delivery System

Faculty of Health Sciences, Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), North-West University, Building G16, 11 Hoffman Street, Potchefstroom 2520, South Africa
*
Author to whom correspondence should be addressed.
Methods Protoc. 2026, 9(1), 16; https://doi.org/10.3390/mps9010016
Submission received: 11 November 2025 / Revised: 12 December 2025 / Accepted: 15 December 2025 / Published: 21 January 2026
(This article belongs to the Section Biochemical and Chemical Analysis & Synthesis)

Abstract

Reversed-phase high-performance liquid chromatography (RP-HPLC) remains one of the most widely applied analytical techniques in the development and quality control testing of finished pharmaceutical products. The combination of gradient chromatographic methods with diode-array detection (DAD) enhances selectivity, ensuring accuracy and reliability when testing drugs with diverse chemical properties in a single dosage form (i.e., fixed-dose combination (FDC) products). In this study, an RP-DAD-HPLC method was developed for the quantitative analysis of clofazimine (CFZ) and pyrazinamide (PZA) for inclusion in an FDC topical drug delivery system. Chromatographic separation was achieved using a C18 column (4.6 mm × 150 mm, 5 µm particle size) with gradient elution at 1 mL/min, employing 0.1% aqueous formic acid and acetonitrile (mobile phases). PZA and CFZ were detected at 254 nm and 284 nm, respectively. The method was validated in accordance with ICH Q2 guidelines, assessing specificity (considering interference from solvents, product matrix, and degradation products), linearity (7.8–500.0 µg/mL, r2 = 0.9999), system repeatability (%RSD ≤ 2.7%), and intermediate precision (25–500 µg/mL, %RSD ≤ 0.85%). Method robustness was evaluated using a three-level Box–Behnken design (BBD) with response surface methodology (RSM) to assess the effects of variations in detection wavelength, mobile phase flow rate, and column temperature.

1. Introduction

In 1826, Théophile Laennec, the inventor of the stethoscope [1], reported the first case of an extrapulmonary tuberculosis (EPTB) variant known as true cutaneous tuberculosis (CTB), a rare skin infection primarily caused by Mycobacterium tuberculosis, though other Mycobacterium species may also be implicated. CTB remains difficult to diagnose due to its diverse and often deceptive clinical manifestations, which frequently mimic other dermatological conditions. As a result, misdiagnosis is common, treatment is delayed, and tissue damage may progress [2]. Despite increasing research attention, CTB still lacks a universally accepted definition or diagnostic standard. The current classification systems attempt to integrate pathogenesis, clinical morphology, and histopathological findings, without converging on a single standardized framework [3].
Although the World Health Organization (WHO) has no CTB-specific guideline, the global tuberculosis (TB) policy documents do recognize the skin as one of the possible sites of extrapulmonary involvement [4]. Current WHO operational handbooks and treatment regimens address EPTB broadly, yet disease-specific criteria for CTB remain undeveloped [4]. Thus, CTB continues to be discussed mainly in scientific literature rather than in global policy [5].
TB is generally preventable and curable [5], yet WHO statistics show that in 2023 it again became the leading cause of death worldwide from a single infectious agent, surpassing COVID-19 [5]. TB caused nearly twice as many deaths as the human immunodeficiency virus (HIV) and acquired immune deficiency syndrome (AIDS) in the same year, and its global incidence has continued to rise, with over 10 million new cases annually [5]. Although CTB accounts for only 1.5–3% of all EPTB cases [2,6,7], increasing multidrug-resistant TB has contributed to its growing clinical relevance [8]. The global TB burden translates to an estimated 150,000 CTB cases annually. In high-TB-burden regions, CTB accounts for roughly 2% of dermatology outpatient visits, and about one-third of affected patients also present with systemic TB, underscoring the importance of timely recognition by dermatologists [2].
CTB management generally follows WHO regimens for drug-susceptible TB, as no CTB-specific regimens exist. Standard therapy consists of a two-month intensive phase using isoniazid, rifampicin, pyrazinamide (PZA), and ethambutol, followed by at least four months of treatment with isoniazid and rifampicin [2,9]. For multidrug-resistant or rifampicin-resistant TB, the WHO recommends all-oral regimens incorporating bedaquiline, pretomanid, linezolid, and moxifloxacin. In an instance where fluoroquinolone resistance is present, moxifloxacin is excluded [10]. National TB programs, such as those in South Africa, adopt these protocols considering drug availability, HIV co-infection, and disease severity [11]. Treatment adherence remains a significant challenge due to long treatment durations and severe adverse effects experienced [5,12].
Topical drug delivery offers a promising alternative for CTB treatment, as it is non-invasive, targets the site of infection directly, and may reduce systemic side effects [13,14].
Recent research has focused on fixed-dose drug combinations (FDCs), which exploit drug synergy to enhance therapeutic outcomes, reduce dosing frequency, and lower drug resistance risks [15,16,17]. However, FDC development requires careful dosing, compatible pharmacokinetics, and avoidance of drug-induced toxicity [16].
In another study, we explored the feasibility of developing a topical drug delivery system for CTB treatment, employing FDC therapy incorporating clofazimine (CFZ) and pyrazinamide (PZA). Oral CFZ–PZA combinations have been shown to significantly reduce the required CFZ dose from 25 mg/kg to 6.25 mg/kg [18] supporting their synergistic potential [19,20]. CFZ is effective against multi-drug-resistant TB strains, but its use is limited owing to its dose-dependent skin discoloration [19,20,21,22,23], severe gastrointestinal side effects, ocular deposition, and dark-brown fingernail discoloration. These side effects arise from CFZ’s highly lipophilic nature (Log P of 7.66) [24], which causes prolonged accumulation in adipose tissue for up to 70 days [25,26]. Consequently, incorporating a reduced quantity of CFZ into a topical drug delivery system would be beneficial, as it could decrease systemic adverse effects by limiting systemic absorption [25,26,27].
However, CFZ’s poor aqueous solubility complicates formulation development [27,28]. Furthermore, PZA, a nicotinamide analog used in combination with drugs such as rifampicin and isoniazid during the initial treatment phase [29,30,31], is a relatively hydrophilic drug (Log P of −0.71) [24]. Although its physicochemical properties are not ideal for dermal drug diffusion, PZA remains essential due to its primarily bacteriostatic nature, with bactericidal effects against Mycobacterium tuberculosis in acidic environments (pH ≤ 5.6) [32], and it shortens the treatment duration [33]. Orally, PZA is associated with gastrointestinal discomfort, hepatotoxicity, nausea, and vomiting [34]. Therefore, an alternative administration route, such as dermal delivery, could provide substantial benefits by directly targeting the infection site, lowering the CFZ concentration, reducing individually caused side effects, and ultimately improving patient compliance.
In developing an FDC combining a highly lipophilic drug (i.e., CFZ) with a highly hydrophilic drug (i.e., PZA), solubilizing both drugs is challenging. Lipid-based formulations, such as spontaneously forming emulsions, are commonly used as carriers for poorly water-soluble drugs [35,36,37,38]. The manufacturing process allows for the incorporation of existing anti-tubercular drugs into an FDC within a topical formulation. In this system, the highly lipophilic CFZ is dissolved in the oil phase and the highly hydrophilic PZA in the aqueous phase. As a result, both CFZ and PZA are dissolved and available for absorption within the topical drug delivery system, thereby overcoming solubilization challenges and improving drug delivery [39].
To co-deliver the highly lipophilic CFZ and hydrophilic PZA, topical microemulsion formulations using different oil phases were manufactured. Olive oil (OLV) was chosen as the “base” oil, and was also combined with 1.5% (v/v) of different essential oils, namely eucalyptus oil (EUC), peppermint oil (PPO), and tea tree oil (TTO). The use of plant-based oils in pharmaceutical formulations has become increasingly prevalent, as they have been shown to enhance skin penetration by temporarily altering the barrier function of the stratum corneum [40]. Furthermore, these oils can promote enhanced dermal drug penetration by creating an occlusive effect that reduces transepidermal water loss and improves skin hydration [41]. Additionally, to ensure proper solubilization of CFZ (0.7% w/v) and PZA (2% w/v) and to form stable microemulsions, a surfactant/cosurfactant combination comprising equal volumes of Span®83 (surfactant) and Tween®60 (cosurfactant) was included.
An existing HPLC method [42] was initially considered for quantifying PZA and CFZ in solubility studies (in water, in OLV, and in the mixtures of OLV with 1.5% (v/v) EUC, PPO, or TTO), emulsions, and in vitro drug release studies. However, injections of CFZ-PZA standards produced unusable chromatograms (Figure 1) showing peak deformities, unknown peaks, and baseline drifts. Consequently, this method could not be successfully transferred to our laboratory.
As reported in the literature, a common reason for HPLC method non-transferability is its lack of robustness [43]. Unfortunately, the cited article [42] did not provide any optimization or robustness data. This necessitated the development and validation of a new HPLC method, which is presented in this article.
The objective of this study was to develop and validate a rapid and reliable HPLC method capable of accurately quantifying the solubility of CFZ and PZA, assaying the amounts of each drug in the topical FDC emulsions, and determining the concentration of the FDC on, in, and through the skin following dermal drug delivery from the various emulsions tested. Method development and validation were performed in accordance with the International Council for Harmonization (ICH) and Good Manufacturing Practice (GMP) requirements [44] and presented in the following sections. The detailed experimental setup and results from using the developed method for solubility studies, assays, and transdermal diffusion studies are presented in a companion publication entitled: “Quality-by-Design Optimization of Self-Emulsifying CFZ/PZA Emulsions and Comparative Diffusion Across Strat-M® and Human Skin”.

2. Experimental Design

2.1. Materials

CFZ was acquired from Cipla (Cape Town, South Africa), and PZA was obtained from DB Fine Chemicals Pty Ltd. (Sandton, South Africa). The various oil phases used (i.e., OLV, EUC, PPO, and TTO) were obtained from Scatters Oils (Johannesburg, South Africa). Hydrochloric acid, sodium chloride, and hydrogen peroxide (3%), all analytical grade, were sourced from Merck (Modderfontein, Johannesburg, South Africa). Ultra-purified water was produced using a Rephile Bioscience Ltd. Genie U12 water purification system (Boston, MA, USA).
Span®83 (sorbitan sesquioleate) and Tween®60 (polyoxyethylene sorbitan monostearate or polysorbate 60) were purchased from Sigma-Aldrich Chemistry GmbH (Steinheim, Germany). The HPLC-grade methanol and acetonitrile used were supplied by Fisher Chemical as Fisher Scientific Company L.L.C. (Pittsburgh, PA, USA). EMSURE® ACS Ph. Eur. grade Formic acid (98–100%) was procured from Merck KGaA (Darmstadt, Germany).

2.2. Equipment

A Hitachi Chromaster HPLC system (Hitachi High-Tech Science Corporation, Tokyo, Japan) was employed in this study. The HPLC system was equipped with a Chromaster 5160 quaternary pump, a Chromaster 5280 temperature-controlled autosampler, a Chromaster 5310 column oven, and a Chromaster 5430 diode array detector with a variable wavelength detector. A Luna® C18(2) 100 Å CC column (4.6 mm × 150 mm, 5 µm particle size) from Phenomenex (Torrance, CA, USA) was used. The injection volume was set to 1 µL, and the flow rate was set to 1.0 mL/min. Mobile phase A was a 0.1% aqueous formic acid solution, and mobile phase B was HPLC-grade acetonitrile. The gradient program employed during the analysis is summarized in Table 1.

2.3. HPLC Operating Conditions

Data acquisition and analysis were performed using OpenLab CDS PLUS software version 2.7. The chromatographic system was operated with an injection volume of 1 µL and a flow rate of 1.0 mL/min. The mobile phases consisted of 0.1% aqueous formic acid solution (mobile phase A) and HPLC-grade acetonitrile (mobile phase B). The gradient elution program applied during the analysis is summarized in Table 1.

3. Procedures

3.1. Preparation of Stock and Standard Solutions

Stock standard solutions containing approximately 500.0 µg/mL of PZA and CFZ in methanol were prepared. Each stock solution was serially diluted with methanol to obtain solutions containing approximately 400.0, 250.0, 125.0, 62.5, 15.6, and 7.8 µg/mL of PZA and CFZ. These solutions served to validate the method, as described in the following sections.

3.2. Chromatographic Properties of the Eluted Peaks

The symmetry of the eluted peaks was assessed by calculating the tailing factor (TF) using Equation (1), as described in the available literature [45]:
T F   =   W 0.05 2 d
where: (W0.05) represents the width of the peak at one-twentieth of the total peak height, and (d) denotes the distance between the perpendicular line dropped from the peak maximum and the leading edge of the peak at one-twentieth of the total peak height.
The resolution (RS) between the PZA and CFZ peaks was calculated using Equation (2) [45]:
R s   =   1.18   ×   t R 2     t R 1 W h 1 +   W h 2
where: (Rs) indicates the RS between the peaks of PZA (Peak 1) and CFZ (Peak 2), (tR1) and (tR2) are the retention times of the PZA and CFZ peaks, respectively, and (Wh1) and (Wh2) represent the widths at half-height for the PZA and CFZ peaks, individually.

3.3. Method Validation Procedure

Method validation was performed by assessing the following parameters: system repeatability, specificity, linearity and range, limit of detection, limit of quantitation, accuracy, intermediate precision, and robustness.

3.3.1. System Repeatability

System repeatability was evaluated by injecting a standard solution containing approximately 250 µg/mL of PZA and CFZ in methanol six times and calculating the relative standard deviation (%RSD) values of the instrument responses, namely the area under the curve (AUC) and the retention time (TR) for both drugs. In accordance with the United States Pharmacopoeia (USP) requirements, the %RSD for system repeatability must not exceed 3.3% [45].

3.3.2. Specificity

Method specificity was assessed in accordance with the ICH Q2(R2) guidelines, considering chromatographic RS and peak purity [44]. Specificity studies determine the ability of the analytical method to unambiguously detect and quantify analytes in the presence of other components, such as solvents, formulation matrices, and potential analyte degradation products.
To demonstrate selectivity, the solvents (i.e., water, OLV, and OLV essential oil mixtures consisting of OLV+ 1.5% (v/v) EUC; OLV + 1.5% (v/v) PPO; and OLV + 1.5% (v/v) TTO), as well as placebos of the various PZA-CFZ FDC topical emulsions, were analyzed using methanol as the solvent to detect potential interference with the RTs of the PZA and CFZ peaks.
Forced degradation studies were conducted under acidic (0.1 N HCl), basic (0.1 N NaOH), and oxidative (3% (v/v) H2O2) conditions at an elevated temperature of 60 °C to detect potential interference from degradation products. Individual standard solutions of PZA and CFZ were prepared by dissolving 100 mg of the respective reference standard in 50 mL of methanol and diluting it to a final volume of 100 mL with the indicated stressor solutions. The resulting solutions were incubated at 60 °C for 3 h, after which aliquots were diluted with methanol to an approximate final concentration of 500 µg/mL before HPLC analysis.

3.3.3. Linearity, Range, Limit of Detection, and Limit of Quantitation

Linearity and range of the analytical method were established by preparing serial dilutions from the stock solutions described in Section 3.1. Calibration curves were constructed using the six calibration points, which ranged from 7.8 to 500.0 µg/mL. The limit of detection (LOD) and limit of quantitation (LOQ) were determined as described in the ICH Q2 guideline [44].

3.3.4. Accuracy

Accuracy and precision of the analytical method were evaluated by preparing spiked solutions at three concentration levels (25, 250, and 500 µg/mL) and calculating the recovery of PZA and CFZ using Equation (3):
%   Recovery   =   Experimental   concentration Theoretical   concentration   × 100

3.3.5. Intermediate Precision

Intermediate precision was investigated by analyzing spiked solutions at three concentration levels (25, 250, and 500 µg/mL) prepared on three different days (Day 1, Day 2, and Day 3) as part of the accuracy study. The % recovery values of PZA and CFZ were determined from six replicate injections, and the %RSD values for the individual and combined data sets were calculated.

3.3.6. Robustness

Method robustness was examined by introducing deliberate changes to the system parameters, including detection wavelength, mobile phase flow rate, and column temperature. A standard solution containing approximately 250 µg/mL of PZA and CFZ in methanol was used for all the experimental runs. The effects of these variations were evaluated using a three-level Box–Behnken design (BBD) and response surface methodology (RSM) approach commonly applied within Quality-by-Design (QbD) principles, which has gained popularity in assessing method robustness of HPLC methods [46,47,48,49]. The benefits of using RSM include the fact that it allows for a more efficient experimental design (requiring fewer runs than a full factorial design), and it evaluates interaction effects and aligns with Analytical QbD (AQbD) approaches commonly required in regulatory submissions.
The BBD incorporated three independent and six dependent variables. In this study, the independent variables included the wavelength of detection (x1, nm), flow rate (x2, mL/min), and column temperature (x3, °C). The dependent factors included the RT (y1, min) of PZA or CFZ, the TF (y2) of PZA or CFZ, and the RS factor (y3) observed under the respective conditions. Table 2 summarizes the levels of the independent variables, while Table 4 outlines the experimental design used in this study and the results obtained.
The effects of the independent variables on the responses were analyzed using Statistica software (version 14.0.0.15; TIBCO®, San Ramon, CA, USA), which generated the polynomial equations describing the relationships among variables. The model quality was assessed using the correlation coefficient (r2), adjusted correlation coefficient (Adj. r2), and the analysis of variance (F-test). Acceptable model criteria include r2 > 0.900, a difference between r2 and Adj. r2 ≤ 0.2, and a p-value < 0.05 for the F-test. Contour plots of the tested responses were constructed from the fitted polynomial models to visualize changes in the response surfaces under varying analytical conditions.

4. Results and Discussion

4.1. Chromatographic Analysis

As illustrated in Figure 1, the analytical method proposed by van Staden et al. [42] was unfortunately not inter-laboratory transferable. The method was subsequently adjusted, retaining the use of 0.1% aqueous formic acid and acetonitrile as mobile phases, since this combination is commonly employed in HPLC gradient method development and provides adequate solubility for both analytes [50]. Formic acid establishes an acidic environment that promotes the ionization of weakly basic drugs such as PZA, thereby improving the shapes by suppressing the ionization of silanol groups in the stationary phase. Furthermore, acetonitrile serves as an ideal organic modifier to facilitate the separation between the hydrophilic (PZA) and lipophilic (CFZ) drugs.
Upon reviewing the gradient conditions of the published method, rapid changes in the mobile phase composition were identified as a potential source for the observed problems. Consequently, the gradient was modified as described in the Methodology section (Table 1).
In the study by Van Staden et al. [42] the detection wavelengths of 254 nm and 320 nm were used for isoniazid/PZA and rifampicin/CFZ, respectively. Considering the UVmax values reported in the literature for the two drugs (UVmax − PZA = 269 nm and UVmax − CFZ = 284 nm) [51], the detection wavelength for PZA was maintained at 254 nm, as the peak response obtained was sufficient to prevent detector overload at higher concentrations. However, the detection wavelength for CFZ was adjusted to 284 nm to enhance detector sensitivity.
The chromatogram obtained using the adapted method (Figure 2) for a standard solution containing both analytes (PZA and CFZ) showed two distinct peaks. PZA, the hydrophilic drug, eluted at approximately 3.493 min, while CFZ, the lipophilic drug, eluted at around 9.847 min.

4.2. Validation of the Analytical Method

The system repeatability results complied with the USP specifications, as the %RSD values of both the AUC and TR for both PZA and CFZ were below 3.3: %RSDAUC-PZA = 1.0%, %RSDTR-PZA = 0.1%, %RSDAUC-CFZ = 1.0%, and %RSDTR-CFZ = 0.4%.
The calculated TFs for the two peaks were 1.1 and 1.5, respectively, both of which fall within the acceptable TF range of 1.0–1.5. The RS between the PZA and CFZ peaks was determined to be 44.4, indicating complete baseline-to-baseline separation (discussed in Section 4.3).
As previously mentioned, the selectivity of the method was evaluated in accordance with the ICH Q2(R2) guidelines, considering chromatographic RS and peak purity analysis [44]. According to ICH Q2(R2), the specificity of an HPLC method is demonstrated by the absence of interference from excipients, solvents, and potential degradation products [44].
The chromatograms of the solvents used during the solubility studies, namely water, OLV, and OLV mixtures containing 1.5% (v/v) of selected essential oils (EUC, PPO, and TTO), each diluted with methanol in a 50:50 ratio, did not display any peaks. Therefore, no interference with the PZA and CFZ peaks was detected.
Figure 3 illustrates examples of chromatograms obtained during the assay testing of the microemulsions prepared in OLV, and the OLV mixtures containing the mentioned essential oils. These chromatograms illustrate that the excipients present in the various microemulsions did not co-elute with either drug.
As mentioned earlier, in a companion publication entitled “Quality-by-Design Optimization of Self-Emulsifying CFZ/PZA Emulsions and Comparative Diffusion Across Strat-M® and Human Skin”, the release of CFZ and PZA from the most optimal microemulsion (i.e., OLV&PPO base), and subsequent transport onto, into, and through human skin (Caucasian and African skin) are discussed. Figure 4 depicts examples of HPLC chromatograms of samples analyzed during the transdermal diffusion studies, showing baseline separation of PZA and CFZ from endogenous skin-derived components under the validated conditions for Caucasian skin. Similar chromatographic behavior was observed when African skin was used. The reasons for the limited or no transport of CFZ and/or PZA into the epidermal-dermal region and through the skin have been discussed in the companion publication.
The method’s selectivity toward degradation products was verified through forced degradation studies. In these studies, PZA and CFZ were exposed to acidic (0.1 N HCl), basic (0.1 N NaOH), and oxidative (3% H2O2) stressors at 60 °C for 3 h. The percentage degradation observed in PZA and CFZ under the various stress conditions is summarized in Table 3.
PZA was found to be more susceptible to basic than acid stress conditions, consistent with findings reported by Lakshmi and Jacob [52]. Under acidic conditions, PZA showed only 0.4% degradation. A small peak appearing at 3.191 min in the chromatogram (Figure 5a) could be attributed to a degradation product. The RS between PZA and this peak was 1.8, indicating that the two adjacent peaks are baseline-resolved (RS ≥ 1.5) [45]. Under basic conditions, PZA showed approximately 26.7% degradation, with the degradation product eluting at 3.183 min (Figure 5b) and an RS of 1.7 relative to the PZA peak. Exposure of PZA to oxidative stress conditions for 3 h did not result in detectable degradation. The purity (spectral match factor) for the PZA peak (Figure 5b) was 99.98%, confirming that the peak is spectrally pure and unlikely to contain co-eluting impurities or degradation products.
Similarly, CFZ was found to be more susceptible to basic (95.2%) than to acidic (0.6%) degradation, consistent with the observations of Patil and Deshpande [53]. The spectral purity of the CFZ peak (Figure 6a) was 99.97%. No degradation peaks were detected for CFZ under acidic or basic stress conditions. The spectral purity of the CFZ peak (Figure 6b) was slightly lower (97.49%), possibly due to system noise dominating spectral variations, as the peak was relatively small. A degradation peak was detected in the chromatogram of the oxidized sample (Figure 6c) at an RT of 1.626 min. This degradation product is highly polar and appears as an unretained peak. The spectral purity of the corresponding CFZ peak was 99.96%.
It can thus be concluded that the method demonstrated acceptable specificity, as no solvent, matrices, degradation product, or human skin (and receptor-fluid) interference was detected. The degradation products formed under the tested conditions were effectively detected and well separated from the two analytes of interest.
The external calibration curves (with 95% confidence intervals for the regression lines) for both drugs exhibited linear responses, as depicted in Figure 7. The calculated r2 supported strong linear relationships. ANOVA results indicated that the intercepts were statistically insignificant for both PZA and CFZ, with p-values > 0.05.
The LOQ of PZA was slightly lower than that of CFZ, which may be attributed to the lower response factor of PZA (2.05 mAU/µg·mL−1) compared to CFZ (7.23 mAU/µg·mL−1). The recovery of PZA ranged from 99.0% to 100.3% across the tested concentration levels, with a %RSD ≤ 0.98%, indicating excellent accuracy and precision. The recovery of CFZ ranged from 99.0% to 100.6%, with %RSD ≤ 0.61%, further supporting the method’s accuracy and precision.
Considering that the assay specification for the formulation requires that it contain no less than 90.0% and not more than 110.0% of the labeled amounts of PZA and CFZ, the %RSD required by the HPLC method at a 95% confidence interval (n = 3) was calculated to be no greater than 4.0% [54]. The intermediate precision results confirmed that the %RSD values for both individual sets and combined data were well within this limit (≤4.0%), demonstrating that the method is robust and produces consistent results across the working range and on different days. Table 4 summarizes the validation parameters and the results obtained.
Table 4. Validation parameters and results for PZA and CFZ quantitation.
Table 4. Validation parameters and results for PZA and CFZ quantitation.
ParameterPZACFZ
Concentration range (µg/mL)7.8–5007.8–500
Regression
Correlation coefficient (r2)
Regression equation
p-value for intercept

0.9999
y = 2.0219x + 0.4574
0.86

0.9999
y = 6.9080x + 17.1681
0.07
Limit of detection (LOD)
(µg/mL)
7.146.33
Limit of quantitation (LOQ) (µg/mL)21.6219.18
Accuracy
Expressed as % recovery at ~500, 250, and 25 µg/mL
Mps 09 00016 i001Mps 09 00016 i002
Intermediate precision
Determined at 3 levels, over 3 days
Day 1Day 2Day 3 Day 1Day 2Day 3
Level 1
(~25 µg/mL)




x ¯ :
%RSD:
99.3
98.3
99.0
98.4
99.5
99.0
99.0
0.37
99.3
99.1
99.3
99.1
98.8
98.9
99.1
0.21
99.1
98.9
99.3
99.1
98.8
98.8
99.0
0.22
Level 1
(~25 µg/mL)




x ¯ :
%RSD:
100.0
100.2
100.0
99.8
99.6
99.4
99.8
0.28
99.7
100.0
99.8
100.2
102.0
100.5
100.4
0.85
100.3
101.2
100.7
100.7
100.5
101.0
100.7
0.32
Level 2
(~250 µg/mL)




  x ¯ :
%RSD:
101.0
101.1
100.9
101.0
100.9
100.9
101.0
0.12
101.3
101.0
100.7
100.3
100.5
100.6
100.7
0.36
98.8
98.7
99.2
99.5
99.4
99.4
99.2
0.36
Level 2
(~250 µg/mL)




  x ¯ :
%RSD:
100.7
100.8
100.6
100.5
100.7
100.4
100.6
0.14
99.8
100.3
99.6
100.1
99.8
100.2
100.0
0.26
100.4
101.2
101.7
101.6
100.9
101.3
101.2
0.48
Level 3
(~500 µg/mL)




x ¯ :
%RSD:
100.3
100.7
100.9
100.6
101.1
101.2
100.8
0.33
100.8
100.9
101.2
100.7
100.7
100.9
100.9
0.19
99.1
99.3
99.5
99.4
99.3
99.4
99.3
0.16
Level 3
(~500 µg/mL)




  x ¯ :
%RSD:
98.6
99.9
100.1
99.7
99.8
99.5
99.6
0.53
98.2
98.8
98.3
98.6
98.8
98.8
98.6
0.26
99.1
98.8
98.7
98.5
99.0
99.0
98.8
0.24

4.3. Robustness of the Method—A Box–Behnken Experimental Design

A BBD was applied in the robustness study to investigate how deliberate changes to experimental parameters affect the instrument response and overall method performance. The relationships between the selected detection wavelength, mobile phase flow rate, and column temperature (each evaluated at three levels) and their impact on the responses of RT, peak tailing, and resolution between the PZA and CFZ peaks were investigated using 3D response surface plots.
Based on the chromatogram obtained (Figure 2) and pharmacopoeial requirements [45], the following desired chromatographic properties (i.e., critical quality attributes) were identified: (i) the desired RT for PZA should range between 3 and 4 min to avoid potential interference from solvent or mobile phase peaks (1.7 to 2.5 min and 7.0 to 8.5 min), and that for CLF between 9 and 10 min to reduce potential interference from the slight baseline drift resulting from gradient changes (i.e., refractive index fluctuations in the eluent) [55]; (ii) the TF should range between 1.0 and 1.5 [45], and (iii) the RS between the PZA and CFZ peaks should not be less than 2.
The responses obtained following the experimental design are presented in Table 5. The data were subjected to statistical analysis and mathematical model fitting. A second-order polynomial model provided the best fit of all the responses. Table 6 presents a summary of the model-fit statistics for all dependent variables.
The values of the correlation coefficients and predicted correlation coefficients for the effects on the retention times of PZA and CFZ, PZA peak tailing, and peak RS were found to be close (<0.2 difference), indicating acceptable prognostic efficiency of the experimental design for these response variables. It can thus be concluded that the retention time, tailing, and RS of the PZA peak are influenced by the mobile phase flow rate, column temperature, and detection wavelength. Interestingly, while retention time is typically unaffected by the detection wavelength, variations in wavelength may alter the peak shape and intensity, influencing the detection of the peak apex and, consequently, the apparent retention time.
The r2 value describing the model fit for the TF of the CFZ peak was low, indicating poor data fit. Furthermore, discrepancies between the r2 and adjusted r2 values suggested that the r2 value was artificially inflated due to the absence of statistically significant main and interaction effects at a 95% confidence level (p < 0.05) for all regression coefficients. This finding was confirmed by the ANOVA results, which showed no evidence that changes to the detection wavelength, flow rate, or column temperature—either individually or in combination—had a statistically significant effect on the tailing of the CFZ peak. This could be attributed to strong lipophilic interactions between the larger CFZ molecules and the stationary phase, which result in slower diffusion and greater mass-transfer resistance, making CFZ less sensitive to deliberate variations in the experimental parameters. PZA, on the other hand, is a smaller, hydrophilic molecule with weaker retention, eluting earlier in the gradient where changes in the flow rate, mobile phase composition, and column temperature are more pronounced. Consequently, its chromatographic behavior is more sensitive to changes in these independent variables, consistent with the behavior described by the van Deemter equation [56].
The 3D response surface plots illustrating the effects of the independent variables on the RT of PZA (Figure 8a) were relatively flat with minimal curvature, indicating little interaction effects. Changes in the detection wavelength had little influence on the RT of PZA. However, variations in the flow rate had the most significant impact on RT, with higher flow rates leading to shorter RTs. Column temperature also had a moderate effect, where increasing the temperature shortened the RT of PZA, where increasing the column temperature resulted in a shorter RT, similar to literature findings [57]. The observed PZA RT values (Table 5) ranged from 3.19 to 4.06 min across all experimental conditions, with an average RT of 3.58 ± 0.14 min, which remained within the targeted RT range (3–4 min). A similar trend was observed for CFZ, with RT values ranging from 9.11 to 10.23 min, showing less variation (%RSD = 3.4%) compared to PZA (%RSD = 7.1%), supporting the robustness of the analytical method.
Interestingly, all three response-surface plots exhibit small but measurable curvature. The wavelength-temperature plot shows a gentle concave curvature with a central low region, suggesting optimal peak symmetry at a column temperature between 24 and 26 °C. The wavelength-flow rate plot displays a distinct saddle-shaped curvature [58] with a symmetry optimum (1 ≤ TF ≤ 1.04) around the central flow rate (~1 mL/min). The temperature-flowrate plot displays a gentle valley with optimal symmetry at a 0.98 mL/min ≤ flowrate ≤ 1.12 mL/min at 32 °C ≤ column temperature ≤ 21 °C, confirming the optimized state of the new method. The symmetry of the CFZ peak (Table 5) remained acceptable as 1.0 ≤ TF ≤ 1.1 under all the deliberate parameter variations.
The resolution surfaces shown in the 3D surface plots for PZA (Figure 8c) and CFZ (Figure 9b) portrayed strong curvature, indicating interaction effects among the independent variables. The flow rate was identified as the most critical factor influencing the resolution between PZA and CFZ, while the detection wavelength had the least effect, as expected, followed by column temperature. Across all combination scenarios, the RS values ranged between 40 and 47. Although this represents an unusually high resolution, it may prompt the chromatographer to further optimize the method to achieve shorter elution times. However, doing so would require more rapid changes in the organic phase composition of the gradient due to the significantly large difference in the lipophilicity of the two drugs (PZA: Log P = −0.71 [24]; CFZ: Log P = 7.66 [31]). Preliminary tests showed that such adjustments resulted in the same transferability issues discussed earlier. Moreover, a slightly longer elution time was deemed advantageous to ensure the removal of nonpolar matrix components from the dermal formulation, thereby preventing increased backpressure and loss of column efficiency [59].

5. Conclusions

A rapid and reliable RP-DAD-HPLC method was developed to accurately quantify the solubility of CFZ and PZA in water, OLV, and selected mixtures of OLV + 1.5% (v/v) of a chosen essential oil (i.e., EUC; PPO; and TTO). The method may also be applied to quantify the amounts of PZA and CFZ in topical FDC emulsions prepared using the mentioned oil phases. Furthermore, this method can and has been used to measure dermal drug delivery from these emulsions. Therefore, it can be concluded that this versatile method is suitable not only for final quality control testing of formulated PZA and CFZ topical FDC emulsions, but also for pre-formulation studies (e.g., solubility analysis) and evaluation of drug release from the finished formulated product.
The use of a more conservative gradient program, combined with detection of both drugs at their optimal wavelengths using DAD, enhances method robustness by reducing the sensitivity to dwell-volume differences between chromatographic systems. The method was successfully validated in accordance with the ICH Q2 (R2) requirements.

Author Contributions

Conceptualization, J.M.V. and M.B.; methodology, M.B., J.M.V. and F.B.; software, MB, J.M.V. and F.B.; validation, M.B. and F.B.; formal analysis, M.B., F.B. and J.M.V.; investigation, F.B.; resources, J.M.V. and M.B.; data curation, M.B., F.B. and J.M.V.; writing—original draft preparation, M.B., J.M.V. and F.B.; writing—review and editing, J.M.V., M.B. and F.B.; visualization, M.B., J.M.V. and F.B.; supervision, J.M.V. and M.B.; project administration, J.M.V. and M.B.; funding acquisition, J.M.V. and M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The research was conducted in accordance with the Declaration of Helsinki and approved by the North-West University Health Research Ethics Committee (NWU-HREC) on 5 May 2025 (NWU-00114-25-A1) for studies involving human abdominoplasty tissue (biological waste tissue).

Informed Consent Statement

This study did not make use of any human or animal subjects.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors upon request.

Acknowledgments

The authors would like to thank the Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), Faculty of Health Sciences, North-West University, South Africa, for the financial contribution to this project. Applications in MS-Word, namely, ‘Writefull’, as well as ‘Grammarly’, were used to assist with proper grammar in-text. No AI was used to create any text, paragraphs, or results.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AIDSAcquired Immune Deficiency Syndrome
Adj. r2Adjusted correlation coefficient
ANOVAAnalysis of Variance
AQbDAnalytical Quality-by-Design
AUCArea Under the Curve
BBDBox–Behnken Design
CFZClofazimine
r2Correlation coefficient
CTBCutaneous Tuberculosis
EUCEucalyptus oil
EPTBExtrapulmonary Tuberculosis
FDCFixed-Dose Drug Combination
GMPGood Manufacturing Practice
HIVHuman Immunodeficiency Virus
ICHInternational Council for Harmonization
LODLimit of Detection
LOQLimit of Quantitation
OLVOlive oil
PPOPeppermint oil
PZAPyrazinamide
QbDQuality-by-Design
%RSDRelative Standard Deviation
RSResolution
RSMResponse Surface Methodology
TRRetention Time
TFTailing Factor
TTOTea tree oil
TBTuberculosis
USPUnited States Pharmacopoeia
WHOWorld Health Organization

References

  1. Rullán, J.; Seijo-Montes, R.E.; Vaillant, A.; Sánchez, N.P. Cutaneous manifestations of pulmonary disease. In Atlas of Dermatology in Internal Medicine; Springer: Berlin/Heidelberg, Germany, 2011; pp. 17–30. [Google Scholar]
  2. Gramminger, C.; Biedermann, T. Recognising cutaneous tuberculosis. J. Dtsch. Dermatol. Ges. 2025, 23, 793–802. [Google Scholar] [CrossRef]
  3. Kaul, S.; Kaur, I.; Mehta, S.; Singal, A. Cutaneous tuberculosis. Part I: Pathogenesis, classification, and clinical features. J. Am. Acad. Dermatol. 2023, 89, 1091–1103. [Google Scholar] [CrossRef]
  4. World Health Organization (WHO). WHO Operational Handbook on Tuberculosis. Module 4: Treatment-Drug-Susceptible Tuberculosis Treatment; World Health Organization: Geneva, Switzerland, 2021.
  5. World Health Organization (WHO). Global Tuberculosis Report 2024; World Health Organization: Geneva, Switzerland, 2024. Available online: https://www.who.int/teams/global-programme-on-tuberculosis-and-lung-health/tb-reports/global-tuberculosis-report-2024 (accessed on 10 October 2025).
  6. Chen, Q.; Chen, W.; Hao, F. Cutaneous tuberculosis: A great imitator. Clin. Dermatol. 2019, 37, 192–199. [Google Scholar] [CrossRef]
  7. Kumar, B.; Muralidhar, S. Cutaneous tuberculosis: A twenty-year prospective study. Int. J. Tuberc. Lung Dis. 1999, 3, 494–500. [Google Scholar]
  8. Bhandari, A.; Mahajan, R.; Ramesh, V. Drug-resistance and its impact on cutaneous tuberculosis. Indian Dermatol. Online J. 2022, 13, 570–577. [Google Scholar] [CrossRef]
  9. Bravo, F.G.; Gotuzzo, E. Cutaneous tuberculosis. Clin. Dermatol. 2007, 25, 173–180. [Google Scholar] [CrossRef]
  10. WHO. WHO Consolidated Guidelines on Tuberculosis. Module 4: Treatment-Drug-Resistant Tuberculosis Treatment, 2022 Update; World Health Organization: Geneva, Switzerland, 2022.
  11. DOH. National Guidelines on the Treatment of Tuberculosis Infection; National Department of Health: Canberra, Australia, 2023. Available online: https://knowledgehub.health.gov.za/elibrary/national-guidelines-treatment-tuberculosis-infection (accessed on 10 October 2025).
  12. Pradipta, I.S.; Houtsma, D.; van Boven, J.F.M.; Alffenaar, J.C.; Hak, E. Interventions to improve medication adherence in tuberculosis patients: A systematic review of randomized controlled studies. NPJ Prim. Care Respir. Med. 2020, 30, 21. [Google Scholar] [CrossRef] [PubMed]
  13. Tapfumaneyi, P.; Imran, M.; Mohammed, Y.; Roberts, M.S. Recent advances and future prospective of topical and transdermal delivery systems. Front. Drug Deliv. 2022, 2, 957732. [Google Scholar] [CrossRef]
  14. Lunter, D.; Klang, V.; Eichner, A.; Savic, S.M.; Savic, S.; Lian, G.; Erdő, F. Progress in Topical and Transdermal Drug Delivery Research-Focus on Nanoformulations. Pharmaceutics 2024, 16, 817. [Google Scholar] [CrossRef]
  15. Godman, B.; McCabe, H.; Leong, T.D.; Mueller, D.; Martin, A.P.; Hoxha, I.; Mwita, J.C.; Rwegerera, G.M.; Massele, A.; de Oliveira Costa, J. Fixed dose drug combinations—Are they pharmacoeconomically sound? Findings and implications especially for lower- and middle-income countries. Expert. Rev. Pharmacoecon Outcomes Res. 2020, 20, 1–26. [Google Scholar] [CrossRef] [PubMed]
  16. John, M.; Gopinath, D.; Kalra, S. Triple fixed drug combinations in type 2 diabetes. Indian J. Endocrinol. Metab. 2015, 19, 311–313. [Google Scholar] [CrossRef]
  17. Lange, C.; Chesov, D.; Heyckendorf, J. Clofazimine for the treatment of multidrug-resistant tuberculosis. Clin. Microbiol. Infect. 2019, 25, 128–130. [Google Scholar] [CrossRef]
  18. Ammerman, N.C.; Swanson, R.V.; Bautista, E.M.; Almeida, D.V.; Saini, V.; Omansen, T.F.; Guo, H.; Chang, Y.S.; Li, S.Y.; Tapley, A.; et al. Impact of Clofazimine Dosing on Treatment Shortening of the First-Line Regimen in a Mouse Model of Tuberculosis. Antimicrob. Agents Chemother. 2018, 62, 7. [Google Scholar] [CrossRef]
  19. Dheda, K.; Chang, K.C.; Guglielmetti, L.; Furin, J.; Schaaf, H.S.; Chesov, D.; Esmail, A.; Lange, C. Clinical management of adults and children with multidrug-resistant and extensively drug-resistant tuberculosis. Clin. Microbiol. Infect. 2017, 23, 131–140. [Google Scholar] [CrossRef] [PubMed]
  20. Duan, H.; Chen, X.; Li, Z.; Pang, Y.; Jing, W.; Liu, P.; Wu, T.; Cai, C.; Shi, J.; Qin, Z.; et al. Clofazimine improves clinical outcomes in multidrug-resistant tuberculosis: A randomized controlled trial. Clin. Microbiol. Infect. 2019, 25, 190–195. [Google Scholar] [CrossRef]
  21. Li, Q.; Song, Y.; Chen, H.; Xie, L.; Gao, M.; Ma, L.; Huang, Y. Retrospective Analysis of 28 Cases of Tuberculosis in Pregnant Women in China. Sci. Rep. 2019, 9, 15347. [Google Scholar] [CrossRef]
  22. Dey, T.; Brigden, G.; Cox, H.; Shubber, Z.; Cooke, G.; Ford, N. Outcomes of clofazimine for the treatment of drug-resistant tuberculosis: A systematic review and meta-analysis. J. Antimicrob. Chemother. 2013, 68, 284–293. [Google Scholar] [CrossRef] [PubMed]
  23. Jagannath, C.; Reddy, M.V.; Kailasam, S.; O’Sullivan, J.F.; Gangadharam, P.R. Chemotherapeutic activity of clofazimine and its analogues against Mycobacterium tuberculosis. In vitro, intracellular, and in vivo studies. Am. J. Respir. Crit. Care Med. 1995, 151, 1083–1086. [Google Scholar] [CrossRef]
  24. Villagracia, A.R.; Ong, H.L.; Lagua, F.M.; Alea, G. Chemical reactivity and bioactivity properties of pyrazinamide analogs of acetylsalicylic acid and salicylic acid using conceptual density functional theory. Heliyon 2020, 6, e04239. [Google Scholar] [CrossRef] [PubMed]
  25. van Staden, D.; Haynes, R.K.; Viljoen, J.M. Adapting Clofazimine for Treatment of Cutaneous Tuberculosis by Using Self-Double-Emulsifying Drug Delivery Systems. Antibiotics 2022, 11, 806. [Google Scholar] [CrossRef]
  26. Cunliffe, W.J.; Glass, D.; Goode, K.; Stables, G.I.; Boorman, G.C. A double-blind investigation of the potential systemic absorption of isotretinoin, when combined with chemical sunscreens, following topical application to patients with widespread acne of the face and trunk. Acta Derm. Venereol. 2001, 81, 14–17. [Google Scholar] [CrossRef]
  27. van Staden, D.; du Plessis, J.; Viljoen, J. Development of a Self-Emulsifying Drug Delivery System for Optimized Topical Delivery of Clofazimine. Pharmaceutics 2020, 12, 523. [Google Scholar] [CrossRef]
  28. Becker, C.; Dressman, J.B.; Amidon, G.L.; Junginger, H.E.; Kopp, S.; Midha, K.K.; Shah, V.P.; Stavchansky, S.; Barends, D.M. Biowaiver monographs for immediate release solid oral dosage forms: Isoniazid. J. Pharm. Sci. 2007, 96, 522–531. [Google Scholar] [CrossRef]
  29. Ali, A.E.; Elasala, G.S.; Mohamed, E.A.; Kolkaila, S.A. Spectral, thermal studies and biological activity of pyrazinamide complexes. Heliyon 2019, 5, e02912. [Google Scholar] [CrossRef]
  30. Lamont, E.A.; Dillon, N.A.; Baughn, A.D. The Bewildering Antitubercular Action of Pyrazinamide. Microbiol. Mol. Biol. Rev. 2020, 84, 2. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, Y.; Feng, J.; McManus, S.A.; Lu, H.D.; Ristroph, K.D.; Cho, E.J.; Dobrijevic, E.L.; Chan, H.-K.; Prud’homme, R.K. Design and Solidification of Fast-Releasing Clofazimine Nanoparticles for Treatment of Cryptosporidiosis. Mol. Pharm. 2017, 14, 3480–3488. [Google Scholar] [CrossRef] [PubMed]
  32. Laudouze, J.; Rokitskaya, T.I.; Abolet, A.; Point, V.; Firsov, A.M.; Khailova, L.S.; Cavalier, J.F.; Canaan, S.; Baulard, A.R.; Antonenko, Y.N.; et al. Pyrazinamide kills Mycobacterium tuberculosis via pH-driven weak-acid permeation and cytosolic acidification. bioRxiv 2025. Preprint. [Google Scholar] [CrossRef]
  33. Zhang, J.; Ge, D.; Wang, X.; Wang, W.; Cui, D.; Yuan, G.; Wang, K.; Zhang, W. Influence of Surfactant and Weak-Alkali Concentrations on the Stability of O/W Emulsion in an Alkali-Surfactant-Polymer Compound System. ACS Omega 2021, 6, 5001–5008. [Google Scholar] [CrossRef]
  34. Ramappa, V.; Aithal, G.P. Hepatotoxicity Related to Anti-tuberculosis Drugs: Mechanisms and Management. J. Clin. Exp. Hepatol. 2013, 3, 37–49. [Google Scholar] [CrossRef]
  35. Dokania, S.; Joshi, A.K. Self-microemulsifying drug delivery system (SMEDDS)—Challenges and road ahead. Drug Deliv. 2015, 22, 675–690. [Google Scholar] [CrossRef]
  36. Kauss, T.; Gaubert, A.; Tabaran, L.; Tonelli, G.; Phoeung, T.; Langlois, M.H.; White, N.; Cartwright, A.; Gomes, M.; Gaudin, K. Development of rectal self-emulsifying suspension of a moisture-labile water-soluble drug. Int. J. Pharm. 2018, 536, 283–291. [Google Scholar] [CrossRef] [PubMed]
  37. Kazi, M.; Al-Swairi, M.; Ahmad, A.; Raish, M.; Alanazi, F.K.; Badran, M.M.; Khan, A.A.; Alanazi, A.M.; Hussain, M.D. Evaluation of self-nanoemulsifying drug delivery systems (SNEDDS) for poorly water-soluble talinolol: Preparation, in vitro and in vivo assessment. Front. Pharmacol. 2019, 10, 459. [Google Scholar] [CrossRef] [PubMed]
  38. Rohrer, J.; Zupančič, O.; Hetenyi, G.; Kurpiers, M.; Bernkop-Schnuerch, A. Design and evaluation of SEDDS exhibiting high emulsifying properties. J. Drug Deliv. Sci. Technol. 2018, 44, 366–372. [Google Scholar] [CrossRef]
  39. Morgen, M.; Saxena, A.; Chen, X.Q.; Miller, W.; Nkansah, R.; Goodwin, A.; Cape, J.; Haskell, R.; Su, C.; Gudmundsson, O.; et al. Lipophilic salts of poorly soluble compounds to enable high-dose lipidic SEDDS formulations in drug discovery. Eur. J. Pharm. Biopharm. 2017, 117, 212–223. [Google Scholar] [CrossRef]
  40. Chen, J.; Jiang, Q.D.; Wu, Y.M.; Liu, P.; Yao, J.H.; Lu, Q.; Zhang, H.; Duan, J.A. Potential of Essential Oils as Penetration Enhancers for Transdermal Administration of Ibuprofen to Treat Dysmenorrhoea. Molecules 2015, 20, 18219–18236. [Google Scholar] [CrossRef]
  41. Pivetta, T.P.; Simões, S.; Araújo, M.M.; Carvalho, T.; Arruda, C.; Marcato, P.D. Development of nanoparticles from natural lipids for topical delivery of thymol: Investigation of its anti-inflammatory properties. Colloids Surf. B Biointerfaces 2018, 164, 281–290. [Google Scholar] [CrossRef]
  42. Staden, D.V.; Haynes, R.K.; Van der Kooy, F.; Viljoen, J.M. Development of a HPLC Method for Analysis of a Combination of Clofazimine, Isoniazid, Pyrazinamide, and Rifampicin Incorporated into a Dermal Self-Double-Emulsifying Drug Delivery System. Methods Protoc. 2023, 6, 104. [Google Scholar] [CrossRef] [PubMed]
  43. Liao, J.J.Z.; Capen, R.C.; Schofield, T.L. Assessing the Reproducibility of an Analytical Method. J. Chromatogr. Sci. 2006, 44, 119–122. [Google Scholar] [CrossRef]
  44. ICH. Validation of Analytical Procedures Q2(R2); U.S. Food and Drug Administration: Silver Spring, MD, USA, 2023. Available online: https://database.ich.org/sites/default/files/ICH_Q2-R2_Document_Step2_Guideline_2022_0324.pdf (accessed on 14 October 2025).
  45. United States Pharmacopoeia (USP). Chromatography. 2025. Available online: https://online.uspnf.com/uspnf/document/1_GUID-6C3DF8B8-D12E-4253-A0E7-6855670CDB7B_9_en-US#C621S12 (accessed on 14 October 2025).
  46. Patel, K.; Shah, U.A.; Patel, C.N. Box–Behnken design-assisted optimization of RP-HPLC method for the estimation of evogliptin tartrate by analytical quality by design. Future J. Pharm. Sci. 2023, 9, 57. [Google Scholar] [CrossRef]
  47. Boppy, N.V.V.D.P.; Haridasyam, S.B.; Vadagam, N.; Sara, N.; Sara, K.; Tamma, E. Stability-indicating HPLC method optimization using quality by design with design of experiments approach for quantitative estimation of organic related impurities of Doripenem in pharmaceutical formulations. J. Appl. Pharm. Sci. 2025, 15, 114–126. [Google Scholar]
  48. Tomić, J.; Djajić, N.; Agbaba, D.; Otašević, B.; Malenović, A.; Protić, A. Robust optimization of gradient RP HPLC method for simultaneous determination of ivabradine and its eleven related substances by AQbD approach. Acta Chromatogr. 2021, 34, 1–11. [Google Scholar] [CrossRef]
  49. Afzal, M.; Muddassir, M.; Alarifi, A.; Ansari, M.T. Box-Behnken Assisted Validation and Optimization of an RP-HPLC Method for Simultaneous Determination of Domperidone and Lansoprazole. Separations 2021, 8, 5. [Google Scholar] [CrossRef]
  50. Dong, M.W.; Boyes, B.E. Modern Trends and Best Practices in Mobile-Phase Selection in Reversed-Phase Chromatography. LCGC N. Am. 2018, 36, 752–767. [Google Scholar]
  51. O’Neil, M.J. The Merck Index: An Encyclopedia of Chemicals, Drugs, and Biologicals, 14th ed.; Merck: Whitehouse Station, NJ, USA, 2006. [Google Scholar]
  52. Lakshmi, S.D.; Jacob, J.T. Validated Degradation studies for the estimation of Pyrazinamide, Ethambutol, Isoniazid and Rifampacin in a fixed dose combination by UPLC. Res. J. Pharm. Technol. 2018, 11, 2869–2875. [Google Scholar] [CrossRef]
  53. Patil, T.S.; Deshpande, A.S. Development of an Innovative Quality by Design (QbD) Based Stability-Indicating HPLC Method and its Validation for Clofazimine from its Bulk and Pharmaceutical Dosage Forms. Chromatographia 2019, 82, 579–590. [Google Scholar] [CrossRef]
  54. Catalano, T. Essential Elements for a GMP Analytical Chemistry Department; Springer: New York, NY, USA, 2013; p. 195. [Google Scholar]
  55. LCGCs Chomacademy. HPLC Gradient Elution—Baseline Drift. Available online: https://www.chromatographyonline.com/view/hplc-gradient-elution-baseline-drift (accessed on 7 October 2025).
  56. van Deemter, J.J.; Zuiderweg, F.J.; Klinkenberg, A. Longitudinal diffusion and resistance to mass transfer as causes of nonideality in chromatography. Chem. Eng. Sci. 1956, 5, 271–289. [Google Scholar] [CrossRef]
  57. Gagliardi, L.G.; Castells, C.B.; Ràfols, C.; Rosés, M.; Bosch, E. Effect of temperature on the chromatographic retention of ionizable compounds. II. Acetonitrile-water mobile phases. J. Chromatogr. A 2005, 1077, 159–169. [Google Scholar] [CrossRef]
  58. Montgomery, D.C. Design and Analysis of Experiments, 8th ed.; John Wiley & Sons: Hoboken, NJ, USA, 2012. [Google Scholar]
  59. Snyder, L.R.; Kirklank, J.J. Introduction to Modern Liquid Chromatography, 2nd ed.; Wiley-Interscience Publication, John Wiley & Sons Inc.: New York, NY, USA, 1979; p. 862. [Google Scholar]
Figure 1. Chromatograms obtained for a standard solution containing ~250 µg/mL CFZ and PZA in methanol recorded at two wavelengths: 320 nm (cyan) and 254 nm (dark blue), respectively.
Figure 1. Chromatograms obtained for a standard solution containing ~250 µg/mL CFZ and PZA in methanol recorded at two wavelengths: 320 nm (cyan) and 254 nm (dark blue), respectively.
Mps 09 00016 g001
Figure 2. Chromatograms obtained for a standard solution containing PZA and CFZ recorded at two wavelengths—254 nm (blue) and 284 nm (purple)—individually.
Figure 2. Chromatograms obtained for a standard solution containing PZA and CFZ recorded at two wavelengths—254 nm (blue) and 284 nm (purple)—individually.
Mps 09 00016 g002
Figure 3. HPLC chromatograms of (a) CFZ-PZA reference standard solution, (b) CFZ-PZA microemulsion in the OLV base, (c) CFZ-PZA microemulsion in the OLV&EUC base, (d) CFZ-PZA microemulsion in the OLV&TTO base, and (e) CFZ-PZA microemulsion in the OLV&PPO base. Each was recorded at two wavelengths: 254 nm (dark blue) and 284 nm (cyan), respectively.
Figure 3. HPLC chromatograms of (a) CFZ-PZA reference standard solution, (b) CFZ-PZA microemulsion in the OLV base, (c) CFZ-PZA microemulsion in the OLV&EUC base, (d) CFZ-PZA microemulsion in the OLV&TTO base, and (e) CFZ-PZA microemulsion in the OLV&PPO base. Each was recorded at two wavelengths: 254 nm (dark blue) and 284 nm (cyan), respectively.
Mps 09 00016 g003
Figure 4. HPLC chromatograms of the samples analyzed during the transdermal diffusion studies to quantify (a) CFZ-PZA reference standard solution, (b) CFZ-PZA diffused onto the skin (stratum corneum tape striped samples), (c) CFZ-PZA diffused into the epidermal-dermal layer, and (d) CFZ-PZA diffused through the human skin into the acceptor chamber. Each was recorded at two wavelengths: 254 nm (dark blue) and 284 nm (cyan), respectively.
Figure 4. HPLC chromatograms of the samples analyzed during the transdermal diffusion studies to quantify (a) CFZ-PZA reference standard solution, (b) CFZ-PZA diffused onto the skin (stratum corneum tape striped samples), (c) CFZ-PZA diffused into the epidermal-dermal layer, and (d) CFZ-PZA diffused through the human skin into the acceptor chamber. Each was recorded at two wavelengths: 254 nm (dark blue) and 284 nm (cyan), respectively.
Mps 09 00016 g004
Figure 5. HPLC chromatograms showing PZA and its degradation products following exposure to (a) acidic (0.1 N HCl), (b) basic (0.1 N NaOH), and (c) oxidative (3% H2O2) stress conditions for 3 h at 60 °C.
Figure 5. HPLC chromatograms showing PZA and its degradation products following exposure to (a) acidic (0.1 N HCl), (b) basic (0.1 N NaOH), and (c) oxidative (3% H2O2) stress conditions for 3 h at 60 °C.
Mps 09 00016 g005
Figure 6. HPLC chromatograms showing CFZ and its degradation products exposed to (a) acidic (0.1 N HCl), (b) basic (0.1 N NaOH), and (c) oxidative (3% H2O2) stress conditions for 3 h at 60 °C.
Figure 6. HPLC chromatograms showing CFZ and its degradation products exposed to (a) acidic (0.1 N HCl), (b) basic (0.1 N NaOH), and (c) oxidative (3% H2O2) stress conditions for 3 h at 60 °C.
Mps 09 00016 g006
Figure 7. Calibration plots of (a) PZA and (b) CFZ across the working concentration range of 7.8–500.0 µg/mL. The 95% confidence intervals of the calibration lines are indicated by red dashed lines.
Figure 7. Calibration plots of (a) PZA and (b) CFZ across the working concentration range of 7.8–500.0 µg/mL. The 95% confidence intervals of the calibration lines are indicated by red dashed lines.
Mps 09 00016 g007
Figure 8. 3D response surface plots illustrating the effects of the selected independent variables (i.e., detector wavelength, mobile phase flow rate, and column temperature) on the (a) retention time (RT) and (b) tailing factor of PZA and on the (c) resolution of PZA.
Figure 8. 3D response surface plots illustrating the effects of the selected independent variables (i.e., detector wavelength, mobile phase flow rate, and column temperature) on the (a) retention time (RT) and (b) tailing factor of PZA and on the (c) resolution of PZA.
Mps 09 00016 g008aMps 09 00016 g008b
Figure 9. 3D response surface plots demonstrating the effects of the designated independent variables (i.e., detector wavelength, mobile phase flow rate, and column temperature) on the (a) Retention time (RT) and (b) Resolution of CFZ.
Figure 9. 3D response surface plots demonstrating the effects of the designated independent variables (i.e., detector wavelength, mobile phase flow rate, and column temperature) on the (a) Retention time (RT) and (b) Resolution of CFZ.
Mps 09 00016 g009
Table 1. Gradient program employed during the analysis.
Table 1. Gradient program employed during the analysis.
Time
(Minutes)
Mobile Phase A
(% v/v)
Mobile Phase B
(% v/v)
Comment
0–493.0–90.07.0–10.0Linear gradient
4–690.0–50.010.0–50.0Linear gradient
6–1050.050.0Isocratic
10–1250.0–93.050.0–7.0Linear gradient
12–1593.07.0Re-equilibration
Table 2. Levels of the independent variables used in the BBD optimization of the HPLC method.
Table 2. Levels of the independent variables used in the BBD optimization of the HPLC method.
Independent VariableLevels
LowMediumHigh
x1
Wavelength for PZA (nm)
250255260
Wavelength for CFZ (nm)280285290
x2
Flow rate (mL/min)
0.901.001.10
x3
Column temperature (°C)
202530
Table 3. Results of forced PZA and CFZ degradation studies under the defined stress conditions.
Table 3. Results of forced PZA and CFZ degradation studies under the defined stress conditions.
Stress
Conditions
Treatment
of Samples
% Degradation Observed
PZACFZ
Acid degradation0.1 N HCl for 3 h at 60 °C0.40.6
Base degradation0.1 N NaOH for 3 h at 60 °C26.795.2
Oxidation3% H2O2 for 3 h at 60 °C0.082.8
Table 5. Robustness BBD experimental setup and results.
Table 5. Robustness BBD experimental setup and results.
PZACFZ
Run No.Wave Length (nm)Flow Rate (mL/min)Column
Temperature
(°C)
RT
(min)
TFRSRun No.Wave Length (nm)Flow Rate (mL/min)Column
Temperature
(°C)
RT
(min)
TFRS
12500.9254.001.04012800.92510.231.240
22600.9253.901.04222900.92510.231.142
32501.1253.331.04432801.1259.371.144
42601.1253.191.14742901.1259.411.147
52501.0203.661.14252801.0209.731.242
62601.0203.651.14262901.0209.741.142
72501.0303.471.14572801.0309.781.145
82601.0303.441.14582901.0309.781.145
92550.9204.061.14092850.92010.151.140
102551.1203.461.143102851.1209.111.143
112550.9303.831.143112850.93010.201.243
122551.1303.221.044122851.1309.371.144
132551.0253.541.044132851.0259.791.144
142551.0253.531.145142851.0259.781.145
152551.0253.531.044152851.0259.781.244
162551.0253.531.044162851.0259.761.244
Average:3.581.0543 Average:9.761.143
Standard deviation:0.260.021.87 Standard deviation:0.330.031.87
Relative standard deviation (%):7.132.384.3 Relative standard deviation (%):3.373.004.3
Table 6. Summary of the model-fit statistics for all dependent variables.
Table 6. Summary of the model-fit statistics for all dependent variables.
Dependent Variabler2 ValueAdjusted r2 Value
RT of PZA0.999950.99975
TF of PZA0.950080.75404
RS of PZA and CFZ0.99210.97700
RT of CFZ0.999760.99879
TF of CFZ0.69829 *0.00000 *
RS of PZA and CFZ0.99540.97700
* Poor fit—model not used.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Brits, M.; Bouwer, F.; Viljoen, J.M. RP-DAD-HPLC Method for Quantitative Analysis of Clofazimine and Pyrazinamide for Inclusion in Fixed-Dose Combination Topical Drug Delivery System. Methods Protoc. 2026, 9, 16. https://doi.org/10.3390/mps9010016

AMA Style

Brits M, Bouwer F, Viljoen JM. RP-DAD-HPLC Method for Quantitative Analysis of Clofazimine and Pyrazinamide for Inclusion in Fixed-Dose Combination Topical Drug Delivery System. Methods and Protocols. 2026; 9(1):16. https://doi.org/10.3390/mps9010016

Chicago/Turabian Style

Brits, Marius, Francelle Bouwer, and Joe M. Viljoen. 2026. "RP-DAD-HPLC Method for Quantitative Analysis of Clofazimine and Pyrazinamide for Inclusion in Fixed-Dose Combination Topical Drug Delivery System" Methods and Protocols 9, no. 1: 16. https://doi.org/10.3390/mps9010016

APA Style

Brits, M., Bouwer, F., & Viljoen, J. M. (2026). RP-DAD-HPLC Method for Quantitative Analysis of Clofazimine and Pyrazinamide for Inclusion in Fixed-Dose Combination Topical Drug Delivery System. Methods and Protocols, 9(1), 16. https://doi.org/10.3390/mps9010016

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop