Información de la revista
Vol. 45. Núm. S1.
Páginas 45-55 (Diciembre 2021)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
302
Vol. 45. Núm. S1.
Páginas 45-55 (Diciembre 2021)
REVIEW
Acceso a texto completo
Personalized pharmacotherapy in oncology: Application of pharmacokinetic-pharmacodynamic criteria
Farmacoterapia personalizada en oncología: Aplicación de criterios farmacocinéticos-farmacodinámicos
Visitas
302
Begoña Porta-Oltra1,
Autor para correspondencia
porta_beg@gva.es

Author of correspondence Begona Porta Oltra, Servicio de Farmacia, Hospital Universitario Doctor Peset, Avda. Gaspar Aguilar, 90, 46017 Valencia. Spain
, Matilde Merino-Sanjuán2,3
1 Pharmacy Department, Hospital Universitario Dr. Peset, Valencia. Spain
2 Department of Pharmacy, Pharmaceutical Technology and Parasitology, Universidad de Valencia, Valencia. Spain
3 Interuniversity Institute for Molecular Recognition Research and Technological Development, Universidad de Valencia, Universidad Politécnica de Valencia, Valencia. Spain
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Tablas (4)
Table 1. Causes limiting the implementation of pharmacokinetic monitoring of antineoplastic drugs in clinical practice
Table 2. Criteria to be met by antineoplastic drugs for pharmacokinetic monitoring-based dosing individualization
Table 3. Evidence of the contribution of therapeutic drug monitoring for oral antineoplastic agents in clinical practice (adapted from Mueller-Schoell et al.42, Groenland et al.44 and Verheijen et al.47)
Table 4. PK and PD characteristics of monoclonal antibodies, monoclonal antibody-drug conjugates and bispecific T-cell engagers (BiTEs) (adapted from Paci et al.64 and Desnoyer et al.66)
Mostrar másMostrar menos
Suplemento especial
Este artículo forma parte de:
Vol. 45. Núm S1
Más datos
Abstract
Objective

Indication of personalized pharmacotherapy in oncologic patients is based on the selection of the optimal treatment (drugs, dosing, routes and methods of administration and duration) and on the most appropriate dosing method to achieve maximum antineoplastic efficacy, expressed in terms of remission or relapse-free time and acceptable toxicity for the patients. The aim of this study was to explore the contribution of therapeutic monitoring of plasma concentrations and of the application of the pharmacokinetic and pharmacodynamic information available for some widely used drugs to therapeutic personalization to the care of oncologic patients.

Method

A complete non-systematic literature review was carried out of the pharmacokinetic and pharmacodynamic properties of antineoplastic agents, as well as of the results of their use in clinical practice. The search for high quality articles included primary and secondary bibliographic sources. The benefits of therapeutic monitoring were evaluated for parenteral cytotoxic drugs, oral antineoplastic drugs, monoclonal antibodies and other biological therapies used in clinical practice.

Results

Therapeutic personalization of antineoplastic drugs based on therapeutic monitoring of plasma concentrations together with the informotion provided by pharmacokinetic-pharmacodynamic models makes it possible to reduce toxicity and increase the effectiveness of treatment. When personalized treatment is established with high-dose methotrexate in patients with osteosarcoma, target maximum concentrations are reached in 70% of the cycles (49% when fixed doses are used). When 5-fluorouracil is used in patients with colorectal cancer, the response rate is 33.7% (18.3% with fixed doses). Similar benefit rates are obtained with asparaginase, busulfan, oral antineoplastics and monoclonal antibodies.

Conclusions

Due to the narrow therapeutic range of antineoplastic drugs and the highly variable clinical response they elicit, both in terms of effectiveness and safety, the monitoring of their plasma concentrations and the application of pharmacokinetic and pharmacodynamic principles and models constitute feasible and promising tools in the personalization of oncologic treatment.

KEYWORDS:
Antineoplastic drugs
Pharmacokinetics
Pharmacodynamics
Therapeutic drug monitoring
Resumen
Objetivo

La indicación de una farmacoterapia personalizada en oncología se sustenta en la selección del tratamiento óptimo (fármacos, dosis, vías y métodos de administración y duración) y en el método de ajuste de la dosis para alcanzar la máxima eficacia antineoplásica, expresada en términos de remisión de la enfermedad o de tiempo libre de recaída, con una toxicidad aceptable para el paciente. El objetivo de este trabajo es explorar la contribución, en la personalización terapéutica en oncología clínica asistencial, de la monitorización terapéutica de las concentraciones plasmáticas y la aplicación de la información farmacocinética y farmacodinámica disponible para algunos fármacos ampliamente utilizados.

Método

Se ha realizado una revisión bibliográfica no sistemática completa de los criterios farmacocinéticos y farmacodinámicos de los antineoplásicos, así como de los resultados derivados de su utilización en la práctica clínica asistencial. En la búsqueda de artículos de alta calidad sobre los temas planteados se han incluido fuentes bibliográficas primarias y secundarias. La utilidad de la monitorización terapéutica se ha centrado en fármacos citotóxicos parenterales, antineoplásicos orales, anticuerpos monoclonales y otras terapias biológicas utilizadas en la práctica clínica asistencial.

Resultados

La personalización terapéutica de fármacos antineoplásicos basada en la monitorización terapéutica de las concentraciones plasmáticas, y la información que proporcionan los modelos farmaco-cinéticos-farmacodinámicos, permite reducir la toxicidad y aumentar la efectividad asociada al tratamiento. Cuando se instaura un tratamiento personalizado con metotrexato a altas dosis en pacientes con osteosarcoma se alcanza la concentración máxima objetivo en un 70% de los ciclos (49% en dosis fijas), y con 5-fluorouracilo en pacientes con cáncer colorrectal la tasa de respuesta es del 33,7% (18,3% en dosis fijas). Con asparaginasa, busulfán, antineoplásicos orales y anticuerpos monoclonales se obtienen tasas de beneficios similares.

Conclusiones

Debido al bajo intervalo terapéutico de los medicamentos antineoplásicos y a su alta variabilidad en la respuesta clínica, tanto en términos de efectividad como de seguridad, la monitorización de sus concentraciones plasmáticas, y la aplicación de los principios y de los modelos farmacocinéticos y farmacodinámicos, constituyen herramientas factibles y prometedoras en la personalización de los tratamientos en oncología.

PALABRAS CLAVE:
Fármacos antineoplásicos
Farmacocinética
Farmacodinamia
Monitorización terapéutica de fármacos
Texto completo
Introduction

A wide range of drugs is currently available for the treatment of cancer, with dosing in most cases being calculated based on the patient's body surface area (mg/m2) or weight (mg/kg). However, this approach is associated with significant differences in terms of clinical outcomes, which is largely explained by the high inter-individual variability in the plasma (or serum) concentrations (Cp) achieved. Coefficients of variation higher than 50% in the plasma clearance of antineoplastic drugs, expressed in units of body surface area (L/h/m2) or in terms of L/h, are commonly observed, indicating that this pharmacokinetic parameter is poorly correlated with body surface area. Occasionally, differences observed in Cp may result from genetic differences or alterations in the functional status of cancer patients that affect the absorption and/or bioavailability of drugs1,2. Therefore, personalization of administration regimens of antineoplastic drugs, taking into account the patients’ characteristics, the current disease and the pharmacokinetic information provided by the drug Cp could constitute a valuable tool to reduce variability in the response observed.

According to the International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT) therapeutic drug monitoring (TDM) is “a multidisciplinary clinical specialty aimed at improving patient care by individually adjusting the dose of drugs for which clinical experience or clinical trials have shown it improves the outcome in the general or specific populations. It can be based on a priori pharmacogenetic, demographic or clinical information and/or on the a posteriori measurement of blood concentrations of drugs and/or biomarkers”. The usefulness of this concept in the context of antineoplastic drugs is indisputable as it can be instrumental in personalizing the dosing or administration regimen and maximizing the therapeutic benefit obtained by each patient3. However, several factors have stood in the way of its implementation in the clinical setting4,5 (Table 1). Likewise, for pharmacokinetic monitoring of antineoplastic agents to be useful, the drug must meet several criteria, some of which are listed in Table 2.

Table 1.

Causes limiting the implementation of pharmacokinetic monitoring of antineoplastic drugs in clinical practice

1. Limited availability of simple, rapid and inexpensive analytical techniques for drugs and/or metabolites. 
2. Delays between obtaining the biological sample and quantification of the drug and/or metabolite. 
3. Lack of knowledge about the target values (Cp, AUC, etc.) of the drug, as a single agent and in combination chemo-immunotherapy schemes, which would allow prediction of response (effectiveness and toxicity). 
4. Shortage of clinical trials validating the clinical significance of introducing covariates or predictors of inter- and intra-individual pharmacokinetic variability. 
5. Need to obtain a large number of biological samples to accurately estimate the AUC value. 

AUC: plasma concentration-time area under the curve; Cp: drug (or metabolite) concentration in plasma or serum.

Table 2.

Criteria to be met by antineoplastic drugs for pharmacokinetic monitoring-based dosing individualization

1. High inter-individual variability of kinetic and/or dynamic processes. 
2. Moderate inter-occasion (or intra-individual) variability of kinetic processes. 
3. Narrow therapeutic window. 
4. Evidence that the relationship between drug exposure and therapeutic effect is superior to that between dose and response. 
5. Difficulty in monitoring clinical response, distinguishing between clinical response and adverse effects, or delayed clinical response over time. 
6. Availability of accurate analytical techniques for the determination of drug and/or metabolite levels. 
7. Existence of validated dosing individualization methods based on the increase of treatment effectiveness, reduction of associated morbidity and mortality and reduction of per-process costs. 

The drugs used for the treatment of cancer tend to have a narrow therapeutic range and high interindividual variability in Cp, which can lead to a suboptimal therapeutic response or increased toxicity6. These factors are magnified in cancer, who are often administered antineoplastic drugs as prodrugs or as part of combination chemo-immunotherapy schemes, which makes it difficult to establish a specific therapeutic range target for an individual drug as its achievement will depend on to the overall composition of the antineoplastic scheme. In summary, the therapeutic range of a drug may depend on whether it is administered on its own or in combination with other drugs.

The contribution that a sound understanding of pharmacokinetic and pharmacodynamic factors can make to individualized dosing in the context of oncology seems evident and unquestionably warrants the development of integrated pharmacokinetic, pharmacogenetic and pharmacodynamic models7. Such models should allow identification of specific subpopulations of patients with different clinical responses to antineoplastics resulting from their varying individual factors (genotype, renal or hepatic insufficiency, stage of disease, associated treatments, etc.), or from the presence of a resistance to the antineoplastic agent. They should also allow quantification of the influence of these covariates or predictors of the variability in the clinical response of the target population8.

Indication of personalized pharmacotherapy in oncologic patients is based on the selection of an optimal treatment (drugs, dosing, routes and methods of administration and duration) and on the dosing adjustment used to achieve maximum antineoplastic efficacy, expressed in terms of disease remission or relapse-free time, and acceptable toxicity for the patients. So far, pharmacokinetic monitoring of antineoplastic drugs has been limited if compared with other drug families. However, an increasing number of studies have demonstrated the feasibility of implementing pharmacokinetic monitoring to optimize treatment with several antineoplastics, including 5-fluorouracil, abiraterone, everolimus, imatinib, methotrexate, pazopanib, sunitinib, and tamoxifen9. Against this background, the purpose of this study was to explore the contribution of therapeutic personalization in the care of oncological patients, such personalization should be based on the therapeutic monitoring of plasma concentrations and the application of the pharmacokinetic and pharmacodynamic information available for some of the most widely used drugs.

Methods

To explore the current situation of personalization of oncologic treatment based on therapeutic monitoring of plasma concentrations, a complete non-systematic literature review of the pharmacokinetic and pharmacodynamic characteristics of antineoplastic agents and of the evidence on their clinical use was carried out. A search for high quality articles on the topic of inquiry was carried out in PubMed without limitation of publication date and up to July 2020. The search included primary and secondary bibliographic sources (reviews) and was based on the following terms: antineoplastic agents, pharmacokinetics, pharmacodynamics, therapeutic drug monitoring as well as the different drugs and drug families.

Results

The following sections describe the pharmacokinetic (PK) and pharmacodynamic (PD) models supporting the development of pharmacokinetic/pharmacodynamic (PK/PD) models, as well as the current evidence of the usefulness of pharmacokinetic monitoring of antineoplastic drugs in clinical practice.

Pharmacokinetic models

Pharmacokinetic analysis of Cp-time (t) curves comprises (1) a non-compartmental analysis, in which pharmacokinetic parameters such as AUC, minimum (Cmin) or maximum (Cmax) plasma concentration and time to maximum plasma concentration (tmax) are obtained from the experimental data obtained for each individual; and (2) a compartmental analysis where the pharmacokinetic parameters that best describe the selected pharmacokinetic model (one- or bi-compartmental) are estimated from pairs of experimental Cp-t values.

Non-compartmental analysis provides a good approximation to the exposure to the drug when a large amount of data is available, but prediction of the Cp-t profiles that would be obtained using other dosing regimens is limited. In contrast, the compartmental analysis provides insight into pharmacokinetic parameters [absorption rate constant (ka), bioavailability (F), apparent volume of distribution (Vd), total clearance (Cl), etc.], whose values can be related to the pathophysiological situation of the patient and from which it is possible to predict the Cp-t curves in different dosing scenarios.

When a drug is administered intravenously (IV) the entire dose administered reaches systemic circulation and, consequently, bioavailability is rapid and complete. However, the absorption process of orally administered antineoplastic drugs is complex and is exposed to multiple factors (intestinal and/or hepatic first-pass effect, simultaneous administration with food, dosage, interactions between concomitantly administered drugs and the patient's nutritional status) that can compromise their bioavailability and may explain the significant variability in their pharmacokinetic parameters10.

Volume of distribution is the pharmacokinetic parameter that provides information on the distribution of the drug in the body. Its value depends on different factors (the patient's hydration status and plasma protein and body fat content). Circulating drug metabolites can bind to different blood components, including plasma proteins such as albumin and α-1-acid glycoprotein (AGP), and reach the different organs and tissues through various transport processes. The balance that is established between the free fraction of the drug in plasma and the fraction bound to plasma proteins is crucial for the disposition of the drug. Sometimes, differences in plasma protein levels can explain the pharmacokinetic variability observed in this parameter. For example, in a state of malnutrition and liver disorder, plasma albumin levels decrease, which leads to an increase in the free fraction of the drug. On the other hand, in acute inflammatory processes the free fraction of the drug may decrease because of increased plasma levels of AGP and other components11.

Obesity is a physiological situation that results in a significant interindividual variability in the disposition for the different antineoplastic drugs. Indeed, it is difficult to establish which weight value (actual, adjusted or ideal) should be used to calculate the body surface area and select the dose to be administered. This situation may lead to increased toxicity or a decrease in efficacy. For example, the elimination half-life of ifosfamide is prolonged in obese patients with bronchial carcinoma, which may reflect a diffusion of ifosfamide into fatty tissue12. However, it has not been demonstrated that limiting the dose of antineoplastic agents in obese patients (dosing based on ideal weight rather than actual weight) to prevent toxicity may have beneficial effects. On the contrary, it could have negative effects on the survival of this population group13.

The liver is the main organ responsible for metabolic biotransformation and, consequently, diseases affecting its metabolic and synthetic capacity may explain part of the pharmacokinetic variability observed in drug bioavailability parameters. The genetic polymorphisms of enzymes involved in phase I reactions (cytochrome P [CYP] 450), phase II reactions (glucuronosyltransferases [UGTs]) and hepatic reuptake processes (organic anion transport polyproteins [OATP]) account for a high percentage of this variability.

Excretion of antineoplastic drugs mainly occurs via the biliary and renal routes. Biliary excretion of amphiphilic and lipid-soluble antineoplastic drugs is carried out through the ABCB1 and ABCC2 transporters belonging to the ATP-binding cassette (ABC) family14, whose specific polymorphisms influence the bioavailability and anti-tumor activity of antineoplastic drugs15. Renal excretion is, together with hepatic biotransformation, the main clearance route of antineoplastic drugs. Two drugs exhibiting high pharmacokinetic variability that are cleared primarily via the kidney are carboplatin and methotrexate.

Pharmacodynamic models

PD models are equations that establish the relationship between drug concentrations in the biophase and a (therapeutic or toxic) response. The goal is to provide the information required to explain and predict the evolution of the response to a drug after its administration (onset, intensity and duration of effect) and optimize its therapeutic benefit in an individual patient.

In oncology, the effectiveness of a treatment is evaluated based on final and intermediate clinical outcome variables. Intermediate clinical outcome variables include indicators that provide information on tumor size (RECIST criteria in solid tumors16), biomarkers, and measures that allow quantification of the clinical response over time (disease-free time, progression-free survival, overall survival, etc.). As regards the toxicity of antineoplastic drugs, the National Cancer Institute's17Common Toxicity Criteria (CTC) for the treatment of cancer are currently considered a common standard. Of all pharmacokinetic parameters, AUC is the one most frequently correlated with the efficacy and/or toxicity of antineoplastic drugs. However, most of the correlations established for pharmacokinetic parameters account for less than 70% of the evaluated response and refer mainly to toxicity.

PD models are constructed according to the mathematical characteristics of the variable used to quantify response (continuous or discrete). When the variable (effectiveness or toxicity) is continuous, the most common PD models are linear, semi-logarithmic, maximum-effect and sigmoid. The most widely used continuous response PD models include models obtained with different biomarkers, tumor size models18 and neutropenia models19. When they report a specific effect and their evolution is closely related to the final clinical outcome, biomarkers allow personalized monitoring of a disease, personalized treatment selection and/or designing the most optimal follow-up strategies for each patient20. Selection of predictive biomarkers is based on exploring biological variables that allow a priori identification of patients likely to show a good response to a given treatment and on an understanding of which of them will have a high risk of relapse. The reference biomarker for clinical outcomes in oncology is tumor size, as it forms the basis for some clinical endpoints such as disease progression, overall survival or disease-free survival, among others.

In the case of binary variables, the usual PD models are the logistic regression model, used when the time it takes for the variable to manifests itself is unimportant, and the Cox regression model, employed in the opposite case. When the variable assumes a limited number of values greater than two (CTCAE or RECIST criteria), other more complex models are used, especially when the variable is measured repeatedly over the period of time during which the patient receives treatment. If the result of two consecutive assessments can be correlated, they are studied using Markov models, which consider that the result obtained over a specific period of time are influenced by the value expressed by the variable at a previous point in time. These models are widely accepted in oncology to explain the evolution of the disease, since they can be used to model the changes observed in the patient between one stage of the disease and another, this transition being exclusively dependent on the last determination of the variable's value21–24.

Other models using clinical endpoint variables (overall survival) are based on Kaplan Meier curves and time to event models25. The latter are currently the models of choice, since they make it possible to identify an underlying risk function, obtain the risk ratio between two groups, perform simulations in different scenarios to personalize the drug administration schedule, and include covariates that may or may not change over time. Although time to event models are often referred to as survival models, other variables such as disease-free survival, progression-free survival and time to manifestation of an adverse effect can also be used in their construction.

Pharmacokinetic-pharmacodynamic (PK/PD) models can be obtained by integrating the pharmacokinetic and pharmacodynamic information obtained for the drug. Depending on the characteristics of the equilibrium established between the drug Cp (or drug concentration in another pharmacokinetic compartment) and the drug concentration in the biophase, direct effect models are obtained when the equilibrium is fast, and indirect effect models are obtained when the equilibrium takes longer to occur26.

Current evidence on the use of therapeutic monitoring of antineoplastic agents in clinical practiceMethotrexate

The paradigm for implementing pharmacokinetic monitoring of antineoplastic drugs in patients with osteosarcoma is represented by methotrexate. Given the high interocassion (intra-individual) variability in methotrexate clearance, monitoring must be performed at each administration cycle. For this drug there is an analytical technique of proven reliability as well as a PK/PD model, derived from a bicompartmental pharmacokinetic model, which allows personalized dose adjustment. The purpose of methotrexate Cp monitoring is to guarantee that the treatment is effective [by obtaining a Cmax of 1,000 µmol/L at the end of the infusion (8th hour)] and safe (by preventing the development of toxicity preventing Cp from remaining above 0.08 µmol/L for more than 96 hours)27. During pharmacokinetic monitoring it is essential to ensure proper hydration (3 L/m2), diuresis (300 mL/h) and urine alkalinization (pH ≥ 8) in order to promote adequate clearance and prevent the risk of precipitation and associated renal damage. Folinic rescue is always mandatory 12 hours after discontinuation of methotrexate and should be maintained for at least 60 hours or until Cp falls below 0.05 µmol/L. Likewise, when apparent half-life at 12 hours post-infusion is longer than 3 hours, the patient is considered to be at high risk of toxicity. To avoid this, rescue with colestipol should be initiated. Finally, the dosing regimen for the next cycle is established according to the Cp obtained during the clearance phase (24, 48 and 72 h post-infusion). Using this method means that patients with osteosarcoma treated with high-dose methotrexate reach the target Cmax in 70% of the cycles compared to 49% when fixed doses are administered28.

In the treatment of acute lymphoblastic leukemia (ALL), a correlation between methotrexate exposure and clinical effectiveness has been established. Evans et al. showed a relationship between the risk of relapse and methotrexate clearance in 108 children with ALL. Adjusting the methotrexate dose according to the patient's drug clearance profile during 24-hour infusion significantly improves clinical outcomes in this subpopulation of patients29.

However, in clinical practice, monitoring of methotrexate at high doses (> 500 mg/m2) is mainly applied in the prevention of toxicity rather than for adjusting the dose adjustment and optimizing the drug's effectiveness. Currently, delayed clearance of methotrexate is diagnosed when Cp are above 10 µmol/L at 24 hours, 1 µmol/L at 48 hours or 0.1 µmol/L at 72 hours30.

A recent study set about genotyping the organic anion polypeptide transporter 1B1 (OATP1B1) since this transporter allows uptake of methotrexate from circulating blood by hepatocytes. This transporter's genetic variants are associated with significant variability in drug clearance. The rs4149056 variant in particular is associated with decreased hepatic transport activity and has been shown to cause a moderate increase (13-26%) in the plasma clearance of the drug31,32.

5-fluorouracil

Heterogeneity in the polymorphisms identified in the metabolism of 5-fluorouracil (5-FU), especially those related to dihydropyridine dehydrogenase (DPD), and their inherent intra- and inter-patient variability in patients receiving 5-FU continuous intravenous infusion, warrant pharmacokinetic monitoring of 5-FU. Nomograms, typically based on the establishment of a relationship between the Cp measured at a given time (or the AUC) and clinical response, have been used to individualize the dose of 5-FU to be administered. Thus, after determining the AUC 48 hours after the start of IV infusion, the appropriate dose for the patient is calculated to ensure the drug's effectiveness and safety as adjuvant and neoadjuvant therapy and as treatment for advanced disease in patients diagnosed with colorectal carcinoma (CRC) and squamous cell carcinoma of the head and neck (SCCHN)33. The AUC analysis has shown a good correlation with grade 3-4 toxicity, tumor response and survival, and several studies suggest better patient outcomes when a value between 20-24 mg-h/L is achieved34 (extended to 20-30 mg-h/L in continuous 46-hour infusions35). Using conventional dosing criteria for 5-FU based on body surface area, 20-30% of patients would achieve the therapeutic target for AUC and approximately 60% of patients would have lower exposure33. Pharmacokinetic monitoring of 5-FU in clinical practice has increased due to the incorporation of an automated immunoassay technique for the determination of 5-FU in plasma and has contributed to reducing variability in its exposure and toxicity rates (mainly diarrhea and hand-foot syndrome), although the results in terms of efficacy are more limited, with an acceptable increase in the response rate (33.7% vs. 18.3%)34 but poor survival improvement results in randomized clinical trials36.

The information provided by genetic analyses for the personalization of doses of fluoropyrimidines (5-FU and capecitabine) is complementary to monitoring exposure to 5-FU. Polymorphisms have been identified in the DPYD gene, which synthesizes the DPD involved in its metabolism and inactivation, resulting in a complete (0.1% of the population) or partial (3-8% of the population) deficiency of this enzyme. Different expert groups (ESMO, CPIC and DPWG)37–39 have recommended that, prior to treatment with fluoropyrimidines, the presence of four variants of the DPYD gene that have been shown to have a considerable functional and clinical impact should be analyzed: DPYD*2A and c. 1679T> G (complete deficiency), which result in complete DPD deficiency, and c.2846A> T and c.1236G> A, which result in partial deficiency. Based on the presence of these polymorphisms, recommendations are made regarding personalization of the 5-FU dose prior to initiation of treatment to reduce the potential of myelosuppression, gastrointestinal toxicity (diarrhea and/or mucositis) and neurotoxicity among other adverse events. Patients heterozygous for any of these 4 allelic variants (8% of the population) presented higher grade ≥ 3 toxicity with respect to native patients (39% vs. 23%; p = 0.0013), and DPYD genotype-guided dose personalization reduced the relative risk of developing this toxicity40. Nonetheless, it is estimated that only about 50% of cases with impaired DPD enzyme activity can be identified by these 4 variants.

The Spanish Medicines and Health Products Agency (AEMPS) has recently issued a safety note recommending genotype and/or phenotype testing for DPD deficiency in patients who are candidates for treatment with fluoropyrimidines as well as pharmacokinetic monitoring of 5-FU in patients with partial DPD deficiency. AEMPS also declared its use to be contraindicated in patients with complete DPD deficiency41.

Oral antineoplastic agents

Oral antineoplastics (OAs), which are used at fixed doses in most patients, present with high inter-individual variability in their pharmacokinetic parameters, which also include release and absorption phases that are highly influenced by various pathophysiological and/or external factors. Thus, the inter-individual variability in exposure when standard doses of tyrosine kinase inhibitors are administered is between 19 and 100%. Examples of sources of pharmacokinetic variability are the first-pass effect; the presence of interactions at a metabolic level (especially via cytochrome P450) and with food; the existence of polymorphisms in transporters; renal or hepatic insufficiency; and potential problems of non-adherence to treatment, which is especially important in long-term treatments. Consequently, when standard doses are used, exposure is very likely to be below (> 30%) or above (> 15%) the therapeutic target42.

Significant exposure-response and/or exposure-toxicity relationships have been demonstrated for most OAs43. However, despite compliance with the premises required for therapeutic monitoring44, the latter's usefulness in the context of dose personalization, or in the application of alternative interventions to ensure therapeutic concentrations and increase success rates, has not been demonstrated in prospective randomized studies. Nevertheless, for some OAs pharmacokinetic monitoring is considered useful as retrospective studies have associated it with a significant clinical benefit and the maintenance of drug plasma levels within the therapeutic range, and its implementation has been shown to be feasible in clinical practice (Table 3). A prospective study is currently underway of 23 OAs in a population of more than 600 patients (with historical controls) with the aim of determining the feasibility, effectiveness (proportion of patients below the therapeutic range, objective response rate according to RECIST 1.1, progression-free survival) and safety of dose personalization based on therapeutic monitoring45. Likewise, the lack of optimal standards or circuits for implementing pharmacokinetic monitoring as part of the therapeutic personalization of OAs in clinical oncology care is evident46,47.

Table 3.

Evidence of the contribution of therapeutic drug monitoring for oral antineoplastic agents in clinical practice (adapted from Mueller-Schoell et al.42, Groenland et al.44 and Verheijen et al.47)

Drug  Target (ng/ml)  Patients < target with standard dose (%)  Mean/median Css, min at standard dose [ng/mL] (IIV)  Exposure-response ratio (S/N)  Exposure-toxicity ratio (Y/N)  NLME model  Comments 
Abiraterone  Css, min ≥ 8.448  35-42  9.3 (70%) 
  • Yes

  • PFS: 12.2 vs 7.4 months (HR: 0.55; p = 0.044)

 
No  Yes  Ingestion with food increases exposure with no additional toxicity 
Axitinib  AUC ≥ 30049 Css, min ≥ 550  38  AUC: 367 (77%) 
  • Yes

  • PR: increases the probability of achieving a PR by 1.5 times for every increase of 100 in the AUC

  • PFS: 13.8 vs 7.4 months (HR: 0.558; p = 0.003) OS: 37.4 vs 15.8 months (HR: 0.489; p < 0.001)

 
Yes (hypertension, hyperthyroidism, proteinuria, fatigue, diarrhea)  Yes  Evidence in monotherapy. Phase 2 randomized dose escalation trial with clinical benefit 
Everolimus  Css, min 10 (11.9)-26.351  37  15.65 (IC90: 14.79-16.55) 
  • Yes

  • PFS: Css, min < 11.9 ng/mL was associated with a 3-fold increased risk of progression (HR = 3.2; p = 0.001)

 
Yes (pulmonary events, stomatitis)  Yes  5 mg BID decreases Cmax and decreases the risk of toxicity with respect to 10 mg QD 
Gefitinib  Css, min ≥ 20052  30  266 (41%) 
  • Yes

  • OS: 14.6 vs 4.7 months (p = 0.007)

 
Yes (dermal toxicity, diarrhea, hepatotoxicity)  No  Higher Css, min of gefitinib associated with disease control 
Imatinib 
  • Css, min CML:

  • 1,000-1,60053, 54 GIST:

  • 1,100-1,60055

 
73  979 (54%) 926 (52%) 
  • Yes

  • CCR 2 years: 80 vs 73%. MMR 2 years: 78 vs 73%.

  • Yes

  • PT: 30 vs 11.3 months (p = 0.003)

 
Yes (neutropenia, rash, diarrhea, arthralgia, edema)  Yes  Alternative to GIST Css, min ≥ 960 Evidence from randomized cohort and randomized CE studies 
Pazopanib  Css, min ≥ 20,50056, 57  16-20  28.100 (40%) 
  • Yes

  • PFS: 52 vs 19.6 weeks (p = 0.004) Tumor reduction: 37.9 vs 6.9%.

 
Yes (fatigue, anorexia, hypertension)  Yes  Dose of 400 mg BID and concomitant intake with food increases exposure with respect to 800 mg QD 
Sunitinib 
  • Drug + active metabolite: With rest (4 + 2 weeks):

  • Css, min: 50-10058

  • No rest:

  • Css, min ≥ 37.559

 
49-52  51.6 (39%) 
  • Yes

  • AUC associated with longer TTP and OS in GIST and mRCC. Correlation Css, min and AUC (r2: 0.8-0.9)

 
Yes (hypertension, fatigue, anorexia, myelosuppression, hand-foot syndrome, taste alteration, mucositis)  Yes  Long biological half-life of drug and metabolite: TDM in the last week of treatment prior to rest 
Tamoxifen 
  • Active metabolite endoxifen:

  • Css, min ≥ 5.9760

 
20  9.72 (1.73-30.8) 
  • Yes

  • TR: 30% lower risk of recurrence (HR = 0.70, CI95: 0.52-0.94)

 
No  Yes  Genotype-guided dose escalation (CYP2D6) has been shown to be safe 
Trametinib  Css, min ≥ 10.461  27  12.1 (6-34) 
  • Yes

  • Higher response rate (CR and PR) and higher SLP

 
No  Yes  Evidence in monotherapy 

AUC: plasma concentration versus time area under the curve (ng-h/ml); BID: twice daily; CCR: complete cytogenetic response; CR: complete response; Css, min: minimum concentration (pre-dose) at steady state; HR: hazard ratio; IIV: inter-individual variability; MMR: major molecular response; NDME: nonlinear mixed effect; OS: overall survival; PFS: progression-free survival; PR: partial response; PT: time to progression; QD: once daily; RR: recurrence rate; vs: versus.

Note: The Exposure-Response Ratio (Y/N) column shows the values of the indicator available under pharmacokinetic monitoring conditions and, in their absence, the available information referring to the contribution of pharmacokinetic monitoring to the referenced indicator.

Monoclonal antibodies and other targeted biologic therapies

Monoclonal antibodies (mAb) directed against cellular antigens have complex pharmacokinetic profiles characterized by the presence of multiple clearance pathways (proteolysis, internalization in effector cells and autoantibodies), with both dose- and time-dependent elimination half-live. Thus, clearance of the mAb may be lower at high doses than when lower doses are administered due to saturation of the antigenic receptors. On the other hand, when the antigen concentration is high, half-life decreases because the mAb binds to its epitope and is more rapidly eliminated from the body; as the antigen is depleted, clearance from plasma decreases and the plasma clearance half-life increases. Consequently, mAb clearance is a kinetic parameter that seems to play a major role in serum concentrations at the end of the dosing interval. Moreover, its high inter-individual variability (between 29 and 97%) seems to determine the risk of placing the patient outside the proposed therapeutic window62. Generally speaking, a high inter-individual variability has been observed in pharmacokinetic parameters following exposure to mAbs depending on various biometric (gender, weight or body surface area) and pathophysiological factors (glomerular filtration rate, performance status, tumor burden, histology, bone marrow infiltration, etc.). Thus, only 32% of the inter-individual variability in the rituximab clearance rate has been associated with the amount of circulating CD20 antigen, demonstrating the presence of other covariates that influence the drug's elimination63. On the other hand, the complexity is greater for cytotoxic mAb-drug conjugates or bispecific T-cell engagers (BiTEs) or multispecific mAbs. In addition to the complex pharmacokinetics of mAb, there are the characteristics of the cytotoxic agent and its release kinetics. Several studies have established significant relationships between exposure and response, which would justify the performance of prospective studies to validate the application of pharmacokinetic monitoring in clinical practice, since exposure is a predictive variable of the efficacy and/or toxicity of these targeted therapies64.

Immune checkpoint inhibitor mAbs (anti-CTLA-4, PD-1 and PD-L1) exhibit pharmacokinetic properties typical of mAbs along with a wide inter-individual variability in their kinetic parameters (30-50%) and therapeutic and toxic response65. Exposure-response relationships have been described for anti-CTL4 (ipilimumab and tremelimumab) and anti-PD-1 (nivolumab, pembrolizumab and cemiplimab) mAbs although limited data is available for anti-PD-L1 (atezolizumab, avelumab, durvalumab). For some of these mAbs, time-dependent clearance has been demonstrated (nivolumab, pembrolizumab and anti-PD-L1) and, in the case of atezolizumab, the high rate of immunogenicity observed could affect its pharmacokinetic properties. In any case, additional PK/PD studies for this group of mAbs would allow a more precise definition of the association between the concentration and/or clearance of these drugs and clinical response and would confirm the usefulness of monitoring them in clinical practice66. In addition, the correlation between weight and clearance observed in the case of ipilimumab, pembrolizumab and avelumab should be confirmed to legitimize certain claims related to the choice of a fixed dose versus a dose normalized to body weight. Variations in their nature (chimeric, humanized or human), type (IgG1-4) and even binding sites (monospecific, bispecific) and affinity may explain the differences between these mAbs’ various pharmacokinetic profiles (Table 4).

Table 4.

PK and PD characteristics of monoclonal antibodies, monoclonal antibody-drug conjugates and bispecific T-cell engagers (BiTEs) (adapted from Paci et al.64 and Desnoyer et al.66)

Drug (trade name)  Ig type  Vd (L)  Clearance (mL/h)  T1/2  Target: Cmin or AUC 
Rituximab (Mabthera®, Rixathon®, Truxima®)  IgG1  2.7  25  22 days  Cmin: > 25 μg/mL (NHL) 67, 68 AUC: > 9,400 mg-h/L (DLBCL) 69 
Obinutuzumab (Gazyvaro®)  IgG1  3.0  3.3-4.6  26-37 days  Cmin: > 244 μg/mL when tumor > 1,750 mm270 
Ofatumumab (Arzerra®)  IgG1  5.3  7.5  21.8 days  n.a.71 
Bevacizumab (Avastin®)  IgG1  2.7-3.3  7.9-9.2  18-20 days  Cmin: > 15.5 μg/mL72 
Cetuximab (Erbitux)  IgG1  5.0  22  4 days  Cmin: > 33.8 μg/mL (SCCHN)73 Cmin: > 40 μg/mL (CCR)74 
Panitumumab (Vectibix®)  IgG2  6.54  14  7 days  n.a.75 
Trastuzumab (Herceptin®, Herzuma®)  IgG1  2.6-3.6  4.7-7.3  n.a.  Cmin: > 20 μg/mL (BC)76, 77 
Pertuzumab (Perjeta®)  IgG1  2.50-3.11  9.8  18 days  n.a. 78 
Alemtuzumab (Campath®)  IgG1  10.5  n.a.  6 days  AUC0-12 hours: > 5 mg-h/L79 Cmin: > 13.2 mg/mL80 
Daratumumab (Darzalex®)  IgG1  4.2  n.a.  18-23 days  Cmin: > 274 μg/mL81 
Gentuzumab-ozogamicin (Mylotarg®)  IgG4  21.0 (mAb)  265 (mAb)  72 hours (mAb)  n.a. 82 
Brentuximab-vedotin (Adcetris®)  IgG1  6-10 (conj)  73 (conj)  4-6 days (conj)  n.a. 83 
Trastuzumab-emtasina (Kadcyla®)  IgG1  4.0 (conj)  38 (conj)  3-4 days (conj)  n.a. 84 
Inotuzumab-ozogamicin (Besponsa®)  IgG4  12 (conj)  33.3 (conj)  12.3 days (conj)  n.a. 85 
Blinatumomab (Blyncyto®)  n.d.  4.5  122  2 hours  Cmin: > 1,830 pg/mL86 
Ipilimumab (Yervoy®)  IgG1K  7.5  15  15 days (8-15 days for children)  Cmin: > 10 μg/mL (in vitro) 87 
Tremelimumab  IgG2  6.0  8-9.9  19-24 days  Cmin: > 30 μg/mL88 
Nivolumab (Opdivo®)  IgG4  8.0  7.9-9.5  25-26.7 days  n.a. 89 
Pembrolizumab (Keytruda®)  IgG4K  7.4  8.3  27 days  Cmin: > 20 μg/mL90 
Cemiplimab (Libtayo®)  IgG4  5.2  8.8  19 days  > 60 μg/mL91 
Atezolizumab (Tecentriq®)  IgG1  6.9  8.3  27 days  > 6 μg/mL92 
Avelumab (Bavencio®)  IgG1A  4.7  24.6  6.1 days  > 1 μg/mL93 
Durvalumab (Imfinzi®)  IgG1  5.6  8.2  17-18 days  n.a.94 

BC: breast cancer; conj: conjugated mAb-cytotoxic drug; CRC: colorectal carcinoma; DLBCL: diffuse large B-cell lymphoma; IG: inmunoglobuline; n.a.: not available; NHL: non-Hodgkin's lymphoma; SCCHN: squamous cell carcinoma of the head and neck.

Other drugs

Asparaginase is used in chemotherapeutic regimens for the treatment of acute lymphoblastic leukemia (ALL) with increased cure rates, especially in the pediatric population. Achievement of optimal results with this therapy depends on complete and sustained depletion of serum asparagine. However, high inter-individual variability in exposure, differences in pharmacokinetic properties between different asparaginases and the formation of anti-asparaginase antibodies make it difficult to predict the degree of asparagine depletion that occurs after administration of a given dose. Under physiological conditions, the range of circulating asparagine concentrations is between 40 and 80 µM and, although there is no universal consensus, some investigators have established that asparagine deficiency is complete when plasma concentration is less than 0.1-0.2 µM. Thus, in a study in 214 children in first relapse of ALL, patients with serum asparagine levels < 1 µM on day 14 were more likely to achieve a second remission compared to patients with higher levels95. However, due to the rapid ex vivo metabolism of asparagine in the presence of asparaginase, and the complexity of the analysis, direct determination of asparagine concentration in vivo is challenging. The ALL therapeutic monitoring program should also include determination of asparaginase activity (≥ 100 IU/L; between 100-250 IU/L), and of anti-asparaginase antibodies, thus identifying situations of subclinical hypersensitivity and suboptimal enzyme activity (silent inactivation), personalizing current treatment, or evaluating the benefit of a change of formulation96. This would allow identification of situations where patients develop allergic-type reactions, but no enzymatic inactivation, and therefore do not require a change of formulation to ensure adequate treatment.

Busulfan represents the cornerstone of many myeloablative regimens. It is frequently administered in a two-hour infusion every 6 hours for 4 days. Monitoring of the therapy is recommended during the first dose to individualize subsequent doses by calculating the AUC from three plasma samples (end of infusion, 4 and 6 hours post-infusion). Currently, the therapeutic target is an AUC between 900-1,350 µM-min in pediatric patients and 900-1,500 µM-min in adults (four times higher for daily dosing schedules). AUC can be estimated by non-compartmental analysis, population pharmacokinetic models, or pooled sampling methods with a small number of samples97,98. Monitoring minimizes the incidence of hepatic veno-occlusive disease and neurologic toxicity, as well as graft failure and disease recurrence. Pediatric patients present with greater pharmacokinetic variability than adult patients. The variability is highest among newborns and infants < 9 kg, 40% of whom reach off-target AUC values, which makes monitoring particularly necessary in this subpopulation of patients99.

Carboplatin dosing is based on patients’ renal function (the main source of pharmacokinetic variability) and exposure to this drug (AUC) has been shown to correlate well with therapeutic response and toxicity in both pediatric and adult patients. Carboplatin monitoring is recommended mainly in patient subpopulations where exposure is more unpredictable, a positive impact of the treatment having been demonstrated for example in children with cancer on intensification regimes based on high carboplatin doses. In such cases, there is a higher risk of severe toxicity, and the degree of bioavailability may be more variable than in adult patients100. Likewise, monitoring can have a significant impact on anuric patients and those with renal failure, where non-renal clearance predominates. In obese patients, dosing based on total body weight (TBW) overestimates the renal clearance of carboplatin by more than 20%; conversely, ideal body weight (IBW) may underestimate clearance. The best weight value for carboplatin dosing in obese patients is the intermediate value between ideal and total weight (IBW*0.512x[TBW-IBW])101. Regarding myelotoxicity, population PK/PD models have described the impact of exposure to carboplatin on thrombocytopenia to be 1.45 times higher when administered in schedules with etoposide; 2.33-times higher with gemcitabine; and 0.764 times higher (protective effect) with paclitaxel102.

Several PK/PD studies have analyzed exposure-effectiveness relationships or exposure-toxicity relationships for taxanes such as paclitaxel and docetaxel. The usefulness of paclitaxel monitoring using Bayesian methods in reducing inter-individual variability has been demonstrated. However, although the time during which paclitaxel concentration exceeds 0.05 µM (42.7 µg/mL) may be considered a predictor of hematologic toxicity, neuropathy, and therapeutic response, further studies are required to confirm its clinical benefit99. In a prospective randomized controlled trial conducted to evaluate the feasibility and performance of docetaxel dosing based on AUC as a predictor of myelotoxicity and febrile neutropenia, inter-individual variability in the individualized treatment group was over 50% lower than in the body surface area-based dosing group103.

Irinotecan is another antineoplastic that is a good candidate to monitoring due to the high inter-individual variability in the metabolism of its active metabolite SN-38 mediated by UDP glucuronosyltransferase 1A1 (UGT1A1). In this case, a genetic approach is the most appropriate way of reducing the risk of toxicity (neutropenia and digestive toxicity). Several functional polymorphisms have been identified in the UGT1A1 gene (UGT1A1*28 in European, African and Latin populations and UGT1A1*6 in Asian populations mainly; UGT1A1*36 and UGT1A1*37 almost exclusively in African populations), leading to low enzyme expression and decreased glucuronidation activity, which have a significant impact on the incidence of toxicity. As a result, determination of these polymorphisms should be routinely carried out to identify patients at risk of severe toxicity where lower doses should be administered at initiation of treatment104,105. This strategy is especially useful in dose-intensive irinotecan regimens that have only been shown to be safe in patients homozygous for the UGT1A1*1 variant. The Royal Dutch Pharmacists Association - Pharmacogenetics Working Group has recommended an initial dose reduction of 30% for individuals homozygous for the UGT1A1*28 variant, or poor metabolizers, and a subsequent increase based on neutrophil count106. On the other hand, the Group of Clinical Onco-pharmacology (GPCO-Unicancer) and the National Pharmacogenetics Network (RNPGx) have recommended an initial dose reduction of 30% in UGT1A1*28 homozygous individuals for dosing schedules between 180 and 230 mg/m2 (if dose is lower than 180 mg/m2 no dose adjustment is proposed). In contrast, administration of the full dose in dosing schedules ≥ 240 mg/m2 is only recommended in patients homozygous for the UGT1A1*1 variant and in UGT1A1*1/*28 heterozygous patients in the absence of risk factors and under strict surveillance107.

Final considerations and conclusions

Historically, therapeutic drug monitoring or TDM has focused primarily on measuring drug exposure and analyzing pharmacokinetics in an individual patient. The examples given above are a representation of the application of PK/PD criteria in TDM, increasingly established in the management of oncologic drugs. However, application of pharmacokinetic monitoring in clinical practice is still infrequent, perhaps because there is still much to be learned about the clinical benefits, in terms of the toxicity and especially the effectiveness of antineoplastic treatments, provided by personalizing therapy on the basis on the pharmacokinetic information provided by the PC determination.

In this sense, interpretation of Cp is a key aspect as it is serves as a basis for personalized dose adjustments. It is therefore necessary to know which pharmacokinetic exposure parameter (Cmin, Cmax, AUC, etc.) is best correlated with the response to each antineoplastic drug. Considering the principles behind PD models, the PK parameter of choice should be AUC, but the pharmacokinetic analyses performed in various clinical trials have identified Cmin as a surrogate measure of the exposure to a given drug. This measure is particularly well suited to dose adjustment in the case of drugs with a prolonged terminal half-life. However, for drugs with nonlinear (concentration-dependent or time-dependent) pharmacokinetic profiles including nivolumab, whose clearance varies over time and is related to treatment efficacy, or asparaginase, whose pharmacokinetics is related to the formulation used, or several mAbs, personalized dose based on Cp monitoring is more complex.

Currently, PK parameters are obtained from the available databases, using population pharmacokinetic (or pharmacometric) resources, where the models obtained characterize drugs’ pharmacokinetic profiles for the typical individual and quantify the effect of the covariates that explain part of the inter-individual pharmacokinetic variability. In the context of therapeutic personalization, the benefits of pharmacokinetic monitoring are indisputable, not only because it increases the probability of achieving therapeutic benefits but also because PK, PD and/or PK/PD models make it possible to predict, through simulation exercises, the drug exposure achieved by patients on other dosing regimens and select the optimal scenario, after comparing the results obtained in different situations. However, these benefits can only be achieved when the models have been obtained from a minimum number of Cp. Only then will it be possible to overcome the inaccuracies inherent in the models and take advantage of the capacity and potential of current computing tools to characterize the PK parameters and their sources of variability with a view to personalizing oncological treatments so that the majority of treated patients achieve the maximum therapeutic benefit.

The present review of studies on the personalization of antineoplastic drugs based on pharmacokinetic-pharmacodynamic model-based therapeutic drug monitoring indicates that such individualization of treatment allows a reduction in the toxicity and an increase in the effectiveness associated with the treatment. Specifically, when personalized treatment with high-dose methotrexate is administered to patients with osteosarcoma, the target Cmax is reached in 70% of cycles (49% in fixed doses) and when personalized treatment with 5-fluorouracil is administered to patients with colorectal cancer the response rate is 33.7% (18.3% in fixed doses). Similar benefit rates are obtained with asparaginase, busulfan, oral antineoplastics and monoclonal antibodies. Therefore, due to the narrow therapeutic window of antineoplastic drugs and the high variable clinical response they elicit, both in terms of effectiveness and safety, monitoring their plasma concentrations, and applying pharmacokinetic and pharmacodynamic principles and models, constitute feasible and promising tools in the personalization of treatments in oncologic patients.

Funding

No funding.

Conflict of interests

No conflict of interests.

Uncited references
[53]
Larson RA , Druker BJ , Guilhot F , O’Brien SG , Riviere GJ , Krahnke T , et al.
Imatinib pharmacokinetics and its correlation with response and safety in chronicphase chronic myeloid leukemia: a subanalysis of the IRIS study.
Blood., 111 (2008), pp. 4022-4028
[54]
Rousselot P , Johnson-Ansah H , Huguet F , Legros L , Escoffre-Barbe M , Gardembas M , et al.
Personalized Daily Doses of Imatinib By Therapeutic Drug Monitoring Increase the Rates of Molecular Responses in Patients with Chronic Myeloid Leukemia. Final Results of the Randomized OPTIM Imatinib Study.
Blood [Internet]., 126 (2015), pp. 133
[56]
Suttle AB , Ball HA , Molimard M , Hutson TE , Carpenter C , Rajagopalan D , et al.
Relationships between pazopanib exposure and clinical safety and efficacy in patients with advanced renal cell carcinoma.
Br J Cancer., 111 (2014), pp. 1-8
[57]
Verheijen RB , Bins S , Mathijssen RHJ , Lolkema MP , Van Doorn L , Schellens JHM , on behalf of the Dutch Pharmacology Oncology Group, et al.
Individualized Pazopanib Dosing: A Prospective Feasibility Study in Cancer Patients.
Clin Cancer Res., 22 (2016), pp. 5738-5746
[68]
Jäger U , Fridrik M , Zeitlinger M , Heintel D , Hopfinger G , Burgstaller S , et al.
Rituximab serum concentrations during immuno-chemotherapy of follicular lymphoma correlate with patient gender, bone marrow infiltration and clinical response.
Haematologica., 97 (2012), pp. 1431-1438
[71]
Nightingale G .
Ofatumumab: a novel anti-CD20 monoclonal antibody for treatment of refractory chronic lymphocytic leukemia.
Ann Pharmacother., 45 (2011), pp. 1248-1255
[76]
Pegram M , Hsu S , Lewis G , Pietras R , Beryt M , Sliwkowski M , et al.
Inhibitory effects of combinations of HER-2/neu antibody and chemotherapeutic agents used for treatment of human breast cancers.
Oncogene., 18 (1999), pp. 2241-2251
[77]
González García J , Gutiérrez Nicolás F , Nazco Casariego GJ , Batista López JN , Ceballos Lenza I , Ramos Díaz R , et al.
Influence of anthropometric characteristics in patients with her2-positive breast cancer on initial plasma concentrations of trastuzumab.
Ann Pharmacother., 51 (2017), pp. 976-980
[81]
Xu XS , Yan X , Puchalski T , Lonial S , Lokhorst HM , Voorhees PM , et al.
Clinical implications of complex pharmacokinetics for daratumumab dose regimen in patients with relapsed/refractory multiple myeloma.
Clin Pharmacol Ther., 101 (2017), pp. 721-724
[87]
Small EJ , Tchekmedyian NS , Rini BI , Fong L , Lowy I , Allison JP .
A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer.
Clin Cancer Res., 13 (2007), pp. 1810-1815
[91]
Libtayo: EPAR – Product information [Internet] [accessed 08/09/2021]. Available at: https://www.ema.europa.eu/en/documents/product-information/libtayo-eparproduct-information_en.pdf
Bibliography
[1]
Grochow LB .
Individualized dosing of anti-cancer drugs and the role of therapeutic monitoring.
A clinician’s guide to chemotherapy pharmacokinetics and pharmacodynamics, pp. 3-53
[2]
Zandvliet AS , Schellens JHM , Beijnen JH , Huitema ADR .
Population pharmacoki-netics and pharmacodynamics for treatment optimization in clinical oncology.
Clin Pharmacokinet., 47 (2008), pp. 487-513
[3]
Wilkinson DS .
Therapeutic Drug Monitoring in Oncology.
Ther Drug Monit., 41 (2019), pp. 551-552
[4]
De Jonge ME , Huitema DR , Schellens JHM , Rodenhuis S , Beijnen JH .
Individualised cancer chemotherapy: strategies and performance of prospective studies on therapeutic drug monitoring with dose adaptation.
Clin Pharmacokinet., 44 (2005), pp. 147-173
[5]
Rousseau A , Marquet P , Debord J , Sabot C , Lachátre G .
Adaptive contro methods for the dose individualisation of anticancer agents.
Clin Pharmacokinetic., 38 (2000), pp. 315-353
[6]
Sassen SDT , Zwaan CM , Van der Sluis IM , Mathót RAA .
Pharmacokinetics and population pharmacokinetics in pediatric oncology.
Pediatr Blood Cancer., 67 (2020), pp. e28132
[7]
ICH Expert Working Group. International Conference of Harmonization of technical requirements for registration of pharmaceuticals for human use. General considerations for clinical trails E8 [Internet]. Step 4 version, July 17, 1997 [accessed 23/06/2021]: [17 p.]. Available at: https://database.ich.org/sites/default/files/E8_Guideline.pdf
[8]
Mandema JW .
Population pharmacokinetics and pharmacodinamics.
Pharmacokinetics: regulatory, industrial, academic perspective, pp. 441-450
[9]
Menz BD , Stocker SL , Verougstraete N , Kocic D , Galettis P , Stove CP , et al.
Barriers and opportunities for the clinical implementation of therapeutic drug monitoring in oncology.
Br J Clin Pharmacol., 87 (2021), pp. 227-233
[10]
Jiménez Torres NV , Casabó Alós VG , Sancho Chust V .
Manual de procedimientos para farmacocinética clínica,
[11]
Slaviero KA , Clarke SJ , Rivory LP .
Inflammatory response: an unrecognised source of variability in the pharmacokinetics and pharmacodynamics of cancer chemotherapy.
Lancet Oncol., 4 (2003), pp. 224-232
[12]
Cheymol G .
Effects of Obesity on Pharmacokinetics. Implications for Drug Therapy.
Clin Pharmacokinet., 39 (2000), pp. 215-231
[13]
Hunter RJ , Navo MA , Thaker PH , Bodurka DC , Wolf JK , Smith JA .
Doping chemotherapy in obese patients: actual versus assigned body surface area (BSA).
Cancer Treat Rev., 35 (2009), pp. 69-78
[14]
Nozawa T , Minami H , Sugiura S , Tsuji A , Tamai I .
Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7-ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms.
Drug Metab Dispos., 33 (2005), pp. 434-439
[15]
Han JY , Lim HS , Yoo YK , Shin ES , Park YH , Lee SY , et al.
Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer.
Cancer., 110 (2007), pp. 138-147
[16]
Eisenhauer EA , Therasse P , Bogaerts J , Schwartz LH , Sargent D , Ford R , et al.
New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1).
Eur J Cancer., 45 (2009), pp. 228-247
[17]
U.S. Department of Health and Human Services. National Institutes of Health. National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) [Internet]. Version 5.0, november 27, 2017 [accessed 23/06/2021]: [147 p.]. Available at: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_8.5x11.pdf
[18]
Simeoni M , Magni P , Cammia C , De Nicolao G , Croci V , Pesenti E , et al.
Predictive Pharmacokinetic-Pharmacodynamic Modeling of Tumor Growth Kinetics in Xenograft Models after Administration of Anticancer Agents.
Cancer Res., 64 (2004), pp. 1094-1101
[19]
Friberg LE , Henningsson A , Maas H , Nguyen L , Karlsson MO .
Model of chemotherapy-induced myelosuppression with parameter consistency across drugs.
J Clin Oncol., 20 (2002), pp. 4713-4721
[20]
Biomarkers Definitions Working Group.
Biomarkers and surrogate endpoints: preferred definitions and conceptual framework.
Clin Pharmacol Ther., 69 (2001), pp. 89-95
[21]
Schindler E , Karlsson MO .
A Minimal Continuous-Time Markov Pharmacometric Model.
AAPS J., 19 (2017), pp. 1424-1435
[22]
Xu C , Ravva P , Dang JS , Laurent J , Adessi C , McIntyre C , et al.
A continuous-time multistate Markov model to describe the occurrence and severity of diarrhea events in metastatic breast cancer patients treated with lumretuzumab in combination with oertuzumab and paclitaxel.
Cancer Chemother Pharmacol., 82 (2018), pp. 395-406
[23]
Xie F , Van Bocxlaer J , Colin P , Carlier C , Van Kerschaver O , Weerts J , et al.
PKPD Modeling and Dosing Considerations in Advanced Ovarian Cancer Patients Treated with Cisplatin-Based Intraoperative Intraperitoneal Chemotherapy.
[24]
Hansson EK , Ma G , Amantea MA , French J , Milligan PA , Friberg LE , et al.
PKPD Modeling of Predictors for Adverse Effects and Overall Survival in Sunitinib-Treated Patients With GIST.
CPT Pharmacometrics Syst Pharmacol., 2 (2013), pp. e85
[25]
Austin PC .
A Tutorial on Multilevel Survival Analysis: Methods, Models and Applications.
Int Stat Rev., 85 (2017), pp. 185-203
[26]
Bonate PL .
Pharmacokinetic-pharmacodynamic modeling and simulation,
[27]
Pignon T , Lacarelle B , Duffaud F , Guillet P , Durand A , Monjanel S , et al.
Pharmaco-kinetics of high dose methotrexate in adult osteogenic sarcoma.
Cancer Chemother Pharmacol., 33 (1994), pp. 420-424
[28]
Legido Perdices E, González Álvarez A, Borrás Almenar C, Albert Marí A, Porta Oltra B, Jiménez Torres NV. Individualización posológica de metotrexato a dosis altas en pacientes con osteosarcoma. Póster n° 750. Presentado en 55 Congreso Nacional de la Sociedad Española de Farmacia Hospitalaria; 2010. Madrid
[29]
Evans W , Crom WR , Stewart CF , Bowman WP , Chen CH , Abromowitch M , et al.
Methotrexate systemic clearance influences probability of relapse in children with standard-risk acute lymphocytic leukaemia.
Lancet., 323 (1984), pp. 359-362
[30]
Paci A , Veal G , Bardin C , Levêque D , Widmer N , Beijnen J , et al.
Review of therapeutic drug monitoring of anticancer drugs part 1–cytotoxics.
Eur J Cancer., 50 (2014), pp. 2010-2019
[31]
Ramsey LB , Panetta JC , Smith C , Yang W , Fan Y , Winick NJ , et al.
Genome-wide study of methotrexate clearance replicates SLCO1 B1.
Blood., 121 (2013), pp. 898-904
[32]
Radtke S , Zolk O , Renner B , Paulides M , Zimmermann M , Möricke A , et al.
Germline genetic variations in methotrexate candidate genes are associated with phar-macokinetics, toxicity, and outcome in childhood acute lymphoblastic leukemia.
Blood., 121 (2013), pp. 5145-5153
[33]
Beumer JH , Chu E , Allegra C , Tanigawara Y , Milano G , Diasio R , et al.
Therapeutic Drug Monitoring in Oncology: IATDMCT Recommendations for 5-Fluorouracil Therapy.
Clin Pharmacol Ther., 105 (2019), pp. 598-613
[34]
Gamelin E , Delva R , Jacob J , Merrouche Y , Raoul JL , Pezet D , et al.
Individua fluorouracil dose adjustment based on pharmacokinetic follow-up compared with conventional dosage: results of a multicenter randomized trial of patients with metas-tatic colorectal cancer.
J Clin Oncol., 26 (2008), pp. 2099-2105
[35]
Kaldate RR , Haregewoin A , Grier CE , Hamilton SA , McLeod HL .
Modeling the 5-Fluorouracil Area Under the Curve Versus Dose Relationship to Develop a Phar-macokinetic Dosing Algorithm for Colorectal Cancer Patients Receiving FOLFOX6.
The Oncologist., 17 (2012), pp. 296-302
[36]
Beumer JH , Boisdron-Celle M , Clarke W , Courtney JB , Egorin MJ , Gamelin E , et al.
Multicenter Evaluation of a Novel Nanoparticle Immunoassay for 5-Fluorouracil on the Olympus AU400 Analyzer.
Ther Drug Monit., 31 (2009), pp. 688-694
[37]
Henricks LM , Opdam FL , Beijnen JH , Cats A , Schellens JHM .
DPYD genotype-guided dose individualization to improve patient safety of fluoropyrimidine therapy: call for a drug label update.
Ann Oncol., 28 (2017), pp. 2915-2922
[38]
Amstutz U , Henricks LM , Offer SM , Barbarino J , Schellens JHM , Swen JJ , et al.
Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydro-pyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing. 2017 Update.
Clin Pharmacol Ther., 103 (2018), pp. 210-216
[39]
Lunenburg CATC , Van der Wouden CH , Nijenhuis M , Crommentuijn-van Rhenen MH , De Boer-Veger NJ , Buunk AM , et al.
Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction of DPYD and fluoropyrimidines.
Eur J Hum Genet., 28 (2020), pp. 508-517
[40]
Henricks LM , Lunenburg CATC , De Man FM , Meulendijks D , Frederix GWJ , Kienhuis E , et al.
DPYD genotype-guided dose individualisation of fluoropyrimi-dine therapy in patients with cancer: a prospective safety analysis.
Lancet Oncol., 19 (2018), pp. 459-467
[41]
Agencia Española del Medicamento. Nota de seguridad: Fluorouracilo, cape-citabina, tegafur y flucitosina en pacientes con déficit de dihidropirimidina deshidrogenasa [Internet]. May 11, 2020 [accessed 23/06/2021]. Available at: https://www.aemps.gob.es/informa/notasInformativas/medicamen-tosUsoHumano/seguridad/2020/docs/NI_MUH_FV-8-2020-Fluorouracilo.pdf?x16990
[42]
Groenland SL , Van Eerden RAG , Verheijen RB , Koolen SLW , Moes DJAR , Desar ME , et al.
Therapeutic Drug Monitoring of Oral Anticancer Drugs: The Dutch Pharmacology macology Oncology Group–Therapeutic Drug Monitoring Protocol for a Prospective Study.
Ther Drug Monit., 41 (2019), pp. 561-567
[43]
Widmer N , Bardin C , Chatelut E , Paci A , Beijnen J , Levêque D , et al.
Review of therapeutic drug monitoring of anticancer drugs part two – Targeted therapies.
Eur J Cancer., 50 (2014), pp. 2020-2036
[44]
Groenland SL , Mathijssen RHJ , Beijnen JH , Huitema ADR , Steeghs N .
Individualized dosing of oral targeted therapies in oncology is crucial in the era of precision medicine.
Eur J Clin Pharmacol., 75 (2019), pp. 1309-1318
[45]
Netherlands Trial Register NTR6866. Therapeutic drug monitoring for oral anti-cancer drugs [Internet]. Dec 2017 [accessed 08/09/2021]. Available at: https://www.trialregister.nl/trial/6695
[46]
Mueller-Schoell A , Groenland SL , Scherf-Clavel O , Van Dyk M , Huisinga W , Michelet R , et al.
Therapeutic drug monitoring of oral targeted antineoplastic drugs.
Eur J Clin Pharmacol., 77 (2021), pp. 441-464
[47]
Verheijen RB , Yu H , Schellens JHM , Beijnen JH , Steeghs N , Huitema ADR .
Practical Recommendations for Therapeutic Drug Monitoring of Kinase Inhibitors in Oncology.
Clin Pharmacol Ther., 102 (2017), pp. 765-776
[48]
Carton E , Noe G , Huillard O , Golmard L , Giroux J , Cessot A , et al.
Relation between plasma trough concentration of abiraterone and prostate-specific antigen response in metastatic castration-resistant prostate cancer patients.
Eur J Cancer., 72 (2017), pp. 54-61
[49]
Rini BI , Garrett M , Poland B , Dutcher JP , Rixe O , Wilding G , et al.
Axitinib in Metastatic Renal Cell Carcinoma: Results of a Pharmacokinetic and Pharmacodynamic Analysis.
J Clin Pharmacol., 53 (2013), pp. 491-504
[50]
Tsuchiya N, Igarashi R, Suzuki-Honma N, Fujiyama N, Narita S, Inoue T, et al. Association of pharmacokinetics of axitinib with treatment outcome and adverse events in advanced renal cell carcinoma patients. J Clin Oncol [Internet]. 2015; 33: suppl 7; abstract 506 [accessed 09/09/2021]. Available at: https://ascopubs.org/doi/10.1200/jco.2015.33.7_suppl.506
[51]
Verheijen RB , Atrafi F , Schellens JHS , Beijnen JH , Huitema ADR , Mathijssen RHJ , et al.
Pharmacokinetic Optimization of Everolimus Dosing in Oncology: A Randomized Crossover Trial.
Clin Pharmacokinet., 57 (2018), pp. 637-644
[52]
Zhao YY , Li S , Zhang Y , Zhao HY , Liao H , Guo Y , et al.
The relationship between drug exposure and clinical outcomes of non-small cell lung cancer patients treated with gefitinib.
Med Oncol., 28 (2011), pp. 697-702
[55]
Demetri GD , Wang Y , Wehrle E , Racine A , Nikolova Z , Blanke CD , et al.
Imatinib plasma levels are correlated with clinical benefit in patients with unresectable/metastatic gastrointestinal stromal tumors.
J Clin Oncol., 27 (2009), pp. 3141-3147
[58]
Houk BE , Bello CL , Poland B , Rosen LS , Demetri GD , Motzer RJ .
Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis.
Cancer Chemother Pharmacol., 66 (2010), pp. 357-371
[59]
Yu H , Steeghs N , Nijenhuis CM , Schellens JHM , Beijnen JH , Huitema ADR .
Practical guidelines for therapeutic drug monitoring of anticancer tyrosine kinase inhibitors: focus on the pharmacokinetic targets.
Clin Pharmacokinet., 53 (2014), pp. 305-325
[60]
Madlensky L , Natarajan L , Tchu S , Pu M , Mortimer J , Flatt SW , et al.
Tamoxifen metabolite concentrations, CYP2D6 genotype, and breast cancer outcomes.
Clin Pharmacol Ther., 89 (2011), pp. 718-725
[61]
Ouellet D , Kassir N , Chiu J , Mouksassi MS , Leonowens C , Cox D , et al.
Population pharmacokinetics and exposure-response of trametinib, a MEK inhibitor, in patients with BRAF V600 mutation-positive melanoma.
Cancer Chemother Pharmacol., 77 (2016), pp. 807-817
[62]
Fleisher B , Ait-Oudhia S .
A retrospective examination of the US Food and Drug Administration’s clinical pharmacology reviews of oncology biologics for potential use of therapeutic drug monitoring.
Onco Targets Ther., 11 (2017), pp. 113-121
[63]
Cartron G , Letestu R , Dartigeas C , Tout M , Mahé B , Gagez AL , et al.
Increased rituximab exposure does not improve response and outcome of patients with chronic lymphocytic leukemia after fludarabine, cyclophosphamide, rituximab. A French Innovative Leukemia Organization (FILO) study.
Haematologica., 103 (2018), pp. e356e9
[64]
Paci A , Desnoyer A , Delahousse J , Blondel L , Maritaz C , Chaput N , et al.
Pharmacokinetic/pharmacodynamic relationship of therapeutic monoclonal antibodies used in oncology: Part 1, monoclonal antibodies, antibody-drug conjugates and bispecific T-cell engagers.
Eur J Cancer., 128 (2020), pp. 107-118
[65]
Le Louedec F , Leenhardt F , Marin C , Chatelut E , Evrard A , Ciccolini J .
Cancer Immunotherapy Dosing: A Pharmacokinetic/Pharmacodynamic Perspective.
Vaccines., 8 (2020), pp. 632
[66]
Desnoyer A , Broutin S , Delahousse J , Maritaz C , Blondel L , Mir O , et al.
Pharmacokinetic/pharmacodynamic relationship of therapeutic monoclonal antibodies used in oncology: Part 2, immune checkpoint inhibitor antibodies.
Eur J Cancer., 128 (2020), pp. 119-128
[67]
Berinstein NL , Grillo-López AJ , White CA , Bence-Bruckler I , Maloney D , Czuczman M , et al.
Association of serum Rituximab (IDEC-C2B8) concentration and anti-tumor response in the treatment of recurrent low-grade or follicular non-Hodgkin’s lymphoma.
Ann Oncol., 9 (1998), pp. 995-1001
[69]
Tout M , Casasnovas O , Meignan M , Lamy T , Morschhauser F , Salles G , et al.
Rituximab exposure is influenced by baseline metabolic tumor volume and predicts outcome of DLBCL patients: a Lymphoma Study Association report.
Blood., 129 (2017), pp. 2616-2623
[70]
Gibiansky E , Gibiansky L , Carlile DJ , Jamois C , Buchheit V , Frey N .
Population pharmacokinetics of obinutuzumab (GA101) in chronic lymphocytic leukemia (CLL) and non-Hodgkin’s lymphoma and exposure-response in CLL.
CPT Pharmacometrics Syst Pharmacol., 3 (2014), pp. 1-11
[72]
Caulet M , Lecomte T , Bouché O , Rollin J , Gouilleux-Gruart V , Azzopardi N , et al.
Bevacizumab pharmacokinetics influence overall and progression-free survival in metastatic colorectal cancer patients.
Clin Pharmacokinet., 55 (2016), pp. 1381-1394
[73]
Becher F , Ciccolini J , Imbs DC , Marin C , Fournel C , Dupuis C , et al.
A simple and rapid LC-MS/MS method for therapeutic drug monitoring of cetuximab: a GPCOUNICANCER proof of concept study in head-and-neck cancer patients.
[74]
Azzopardi N , Lecomte T , Ternant D , Boisdron-Celle M , Piller F , Morel A , et al.
Cetuximab pharmacokinetics influences progression-free survival of metastatic colorectal cancer patients.
Clin Cancer Res., 17 (2011), pp. 6329-6337
[75]
Yang BB , Lum P , Chen A , Arends R , Roskos L , Smith B , et al.
Pharmacokinetic and pharmacodynamic perspectives on the clinical drug development of panitumumab.
Clin Pharmacokinet., 49 (2010), pp. 729-740
[78]
Rocca A , Andreis D , Fedeli A , Maltoni R , Sarti S , Cecconetto L , et al.
Pharmacokinetics, pharmacodynamics and clinical efficacy of pertuzumab in breast cancer therapy.
Expert Opin Drug Metabol Toxicol., 11 (2015), pp. 1647-1663
[79]
Montillo M , Tedeschi A , Miqueleiz S , Veronese S , Cairoli R , Intropido L , et al.
Alemtuzumab as consolidation after a response to fludarabine is effective in purging residual disease in patients with chronic lymphocytic leukemia.
J Clin Oncol., 24 (2006), pp. 2337-2342
[80]
Mould DR , Baumann A , Kuhlmann J , Keating MJ , Weitman S , Hillmen P , et al.
Population pharmacokinetics-pharmacodynamics of alemtuzumab (Campath) in patients with chronic lymphocytic leukaemia and its link to treatment response.
Br J Clin Pharmacol., 64 (2007), pp. 278-291
[82]
Dowell JA , Korth-Bradley J , Liu H , King SP , Berger MS .
Pharmacokinetics of gemtuzumab ozogamicin, an antibodytargeted chemotherapy agent for the treatment of patients with acute myeloid leukemia in first relapse.
J Clin Pharmacol., 41 (2001), pp. 1206-1214
[83]
Younes A , Bartlett NL , Leonard JP , Kennedy DA , Lynch CM , Sievers EL , et al.
Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas.
N Engl J Med., 363 (2010), pp. 1812-1821
[84]
Krop IE , Beeram M , Modi S , Jones SF , Holden SN , Yu W , et al.
Phase I study of trastuzumab-DM1, an HER2 antibody-drug conjugate, given every 3 Weeks to patients with HER2-positive metastatic breast cancer.
J Clin Oncol., 28 (2010), pp. 2698-2704
[85]
Besponsa: EPAR – Product Information [Internet] [accessed 08/09/2021]. Available at: https://www.ema.europa.eu/en/documents/product-information/besponsa-epar-product-information_en.pdf
[86]
Lee KJ , Chow V , Weissman A , Tulpule S , Aldoss I , Akhtari M .
Clinical use of blinatumomab for B-cell acute lymphoblastic leukemia in adults.
Ther Clin Risk Manag., 12 (2016), pp. 1301-1310
[88]
Wang E , Kang D , Bae KS , Marshall MA , Pavlov D , Parivar K .
Population pharmacokinetic and pharmacodynamic analysis of tremelimumab in patients with metastatic melanoma.
J Clin Pharmacol., 54 (2014), pp. 1108-1116
[89]
Bajaj G , Wang X , Agrawal S , Gupta M , Roy A , Feng Y .
Model-based population pharmacokinetic analysis of nivolumab in patients with solid tumors.
CPT Pharmacometrics Syst Pharmacol., 6 (2017), pp. 58-66
[90]
Freshwater, T., Kondic, A., Ahamadi, M. et al. Evaluation of dosing strategy for pembrolizumab for oncology indications. J. Immunotherapy Cancer 5, 43 (2017). https://doi.org/10.1186/s40425-017-0242-5
[92]
Stroh M , Winter H , Marchand M , Claret L , Eppler S , Ruppel J , et al.
Clinical pharmacokinetics and pharmacodynamics of atezolizumab in metastatic urothelial carcinoma.
Clin Pharmacol Ther., 102 (2017), pp. 305-312
[93]
Kim ES .
Avelumab: first global approval.
Drugs., 77 (2017), pp. 929-937
[94]
Imfinzi: EPAR – product information [Internet] [accessed 08/09/2021]. Available at: https://www.ema.europa.eu/en/documents/product-information/imfizi-eparproductinformation_en.pdf
[95]
Asselin B , Rizzari C .
Asparaginase pharmacokinetics and implications of therapeutic drug monitoring.
Leuk Lymphoma., 56 (2015), pp. 2273-2280
[96]
Kloos RQH , Pieters R , Jumelet FMV , De Groot-Kruseman HA , Van den Bos C , Van der Sluis IM .
Individualized Asparaginase Dosing in Childhood Acute Lymphoblastic Leukemia.
J Clin Oncol., 38 (2020), pp. 715-724
[97]
Russell JA , Kangarloo SB .
Therapeutic Drug Monitoring of Busulfan in Transplantation.
Curr Pharm Des., 14 (2008), pp. 1936-1949
[98]
Kishimoto K , Hasegawa D , Irie K , Okada A , Nakamura S , Tamura A , et al.
Pharmacokinetic analysis for model-supported therapeutic drug monitoring of busulfan in Japanese pediatric hematopoietic stem cell transplantation recipients.
Pediatr Transplant., 24 (2020), pp. e13696
[99]
Paci A , Veal G , Bardin C , Levêque D , Widmer N , Beijnen J , et al.
Review of therapeutic drug monitoring of anticancer drugs part 1 – Cytotoxics.
Eur J Cancer., 50 (2014), pp. 2010-2019
[100]
Barnett S , Kong J , Makin G , Veal GJ .
Over a decade of experience with carboplatin therapeutic drug monitoring in a childhood cancer setting in the United Kingdom.
Br J Clin Pharmacol., 87 (2021), pp. 256-262
[101]
Bénézet S , Guimbaud R , Chatelut E , Chevreau C , Bugat R , Canal P .
How to predict carboplatin clearance from standard morphological and biological characteristics in obese patients.
Ann Oncol., 8 (1997), pp. 607-609
[102]
Maillard M , Le Louedec F , Thomas F , Chatelut E .
Diversity of dose-individualization and therapeutic drug monitoring practices of platinum compounds: a review.
Expert Opin Drug Metab Toxicol., 16 (2020), pp. 907-925
[103]
Gerritsen-van Schieveen P , Royer B .
Therapeutic drug monitoring group of the French Society of Pharmacology and Therapeutics. Level of evidence for therapeutic drug monitoring of taxanes.
Fundam Clin Pharmacol., 25 (2011), pp. 414-424
[104]
Takano M , Sugiyama T .
UGT1A1 polymorphisms in cancer: impact on irinotecan treatment.
Pharmgenomics Pers Med., 10 (2017), pp. 61-68
[105]
Hulshof EC , Deenen MJ , Guchelaar HJ , Gelderblom H .
Pre-therapeutic UGT1A1 genotyping to reduce the risk of irinotecan-induced severe toxicity: Ready for prime time.
Eur J Cancer., 141 (2020), pp. 9-20
[106]
The Royal Dutch Pharmacists Association – Pharmacogenetics Working Group. Dutch guidelines [Internet] November 2018 update [accessed 08/09/2021]. Available at: https://www.pharmgkb.org/guidelineAnnotation/PA166104951
[107]
Etienne-Grimaldi MC , Boyer JC , Thomas F , Quaranta S , Picard N , Loriot MA , Collective work by Groupe de Pharmacologie Clinique Oncologique (GPCO-Unicancer); French Réseau National de Pharmacogénétique Hospitalière (RNPGx), et al.
UGT1A1 genotype and irinotecan therapy: general review and implementation in routine practice.
Fundam Clin Pharmacol., 29 (2015), pp. 219-237
Copyright © 2021. Sociedad Española de Farmacia Hospitalaria
Idiomas
Farmacia Hospitalaria
Opciones de artículo
Herramientas
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

es en
Política de cookies Cookies policy
Utilizamos cookies propias y de terceros para mejorar nuestros servicios y mostrarle publicidad relacionada con sus preferencias mediante el análisis de sus hábitos de navegación. Si continua navegando, consideramos que acepta su uso. Puede cambiar la configuración u obtener más información aquí. To improve our services and products, we use "cookies" (own or third parties authorized) to show advertising related to client preferences through the analyses of navigation customer behavior. Continuing navigation will be considered as acceptance of this use. You can change the settings or obtain more information by clicking here.