Next Article in Journal
Population Pharmacokinetics and Dosing Regimen Optimization of Latamoxef in Chinese Children
Next Article in Special Issue
Enhancing Dissolution and Oral Bioavailability of Ursodeoxycholic Acid with a Spray-Dried pH-Modified Extended Release Formulation
Previous Article in Journal
Formulation and Evaluation of SNEDDS Loaded with Original Lipophenol for the Oral Route to Prevent Dry AMD and Stragardt’s Disease
Previous Article in Special Issue
Benchmarking the Solubility Enhancement and Storage Stability of Amorphous Drug–Polyelectrolyte Nanoplex against Co-Amorphous Formulation of the Same Drug
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Compounded Nonsterile Preparations and FDA-Approved Commercially Available Liquid Products for Children: A North American Update

by
Richard H. Parrish II
1,*,
Lisa D. Ashworth
2,
Raimar Löbenberg
3,
Sandra Benavides
4,
Jeffrey J. Cies
5,6 and
Robert B. MacArthur
7
1
Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA 31902, USA
2
Department of Pharmacy Services, Children’s Health System of Texas, Dallas, TX 75235, USA
3
Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
4
School of Pharmacy, Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, USA
5
Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA 19129, USA
6
Department of Pharmacy Services, St. Christopher’s Hospital for Children/Tower Health, Philadelphia, PA 19134, USA
7
Department of Pharmacy Services, Rockefeller University Hospital, New York, NY 10065, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(5), 1032; https://doi.org/10.3390/pharmaceutics14051032
Submission received: 3 March 2022 / Revised: 6 May 2022 / Accepted: 7 May 2022 / Published: 10 May 2022
(This article belongs to the Special Issue Feature Papers in Physical Pharmacy and Formulation)

Abstract

:
The purpose of this work was to evaluate the suitability of recent US Food and Drug Administration (US-FDA)-approved and marketed oral liquid, powder, or granule products for children in North America, to identify the next group of Active Pharmaceutical Ingredients (APIs) that have high potential for development as commercially available FDA-approved finished liquid dosage forms, and to propose lists of compounded nonsterile preparations (CNSPs) that should be developed as commercially available FDA-approved finished liquid dosage forms, as well as those that pharmacists should continue to compound extemporaneously. Through this identification and categorization process, the pharmaceutical industry, government, and professionals are encouraged to continue to work together to improve the likelihood that patients will receive high-quality standardized extemporaneously compounded CNSPs and US-FDA-approved products.

1. Introduction

Since the publication of our last paper, a number of novel, commercially scaled pediatric formulations have been approved by the FDA that incorporate candidate molecules (active pharmaceutical ingredients—API), which in the past were primarily available only as extemporaneously compounded nonsterile preparations (CNSPs) [1]. In that paper, a list of 16 candidate active pharmaceutical ingredients (APIs) were selected from the universe of CNSPs described in the professional literature, which were commonly dispensed in pediatric clinical practice. Bearing in mind the guidance from the U.S. Food and Drug Administration (US-FDA) that discourages the compounding of drug preparations that are “essentially copies of approved products”, pharmacists should refrain from compounding any preparation that is now available in an approved finished dosage form appropriate for an individual patient [2]. The following discussion illustrates the variety and formulation characteristics of seven recently marketed mass-produced final oral liquid dosage forms submitted to and approved by the US-FDA under section 505(b)(2) of the Federal Food, Drug, and Cosmetics Act [3]. Additionally, based on the author’s experience and expertise, this report will: (1) evaluate the appropriateness of these newly marketed products for use in neonatal and pediatric populations: (2) identify the next wave of candidate APIs using the algorithm previously developed and described in the manuscript; (3) propose two lists of API candidates: (a) those that are listed currently as compounding monographs in the United States Pharmacopeia (USP) Compounding Compendium (CC) and should be considered for development as approved mass produced finished liquid dosage forms [4]; and (b) those not currently listed in USP CC that have limited mass market potential. Currently listed monographs should remain in the USP CC to provide guidance globally as well as to mitigate the impacts of drug shortages on the approved pharmaceutical supply chain.

2. FDA-Approved Manufactured Dosage Forms Marketed in the U.S. since 2016

Several authors recently have reviewed the array of pediatric oral formulations on the worldwide market and in development, and it has been suggested that the future of formulation development for children lies in mini-tablets and other solid dispersible dosage forms [5,6,7,8]. Many, if not most, of these dose forms provide only “close enough” dosing flexibility for individual patients. However, “close enough” often is not good enough, especially for the most vulnerable of patients [9]. In our experience, how to measure exact milligram per kilogram (mg/kg) doses from these solid orals in the in-patient setting would be problematic, considering that the pharmacies in most U.S. pediatric institutions provide a measured, ready-to-give liquid dose form for the nurse to administer for improved patient safety. Table 1 outlines the eight liquid finished dosage forms from the AA list that have become available since 2016, including two amlodipine salts.

3. Suitability of Recently Marketed Manufactured Liquids for Children

USP defines excipients, often called ’inactive ingredients’, “(As comprising) everything except the active pharmaceutical ingredients (APIs). Excipient functions range from helping to guarantee the stability and bioavailability of the API to the drug product’s manufacturability to its texture and taste. Excipients are major components of almost all drugs, as well as foods, cosmetics, and dietary supplements” [10]. For children, not all inactive ingredients are inactive. Because a number of excipients are inappropriate for children, the European Pediatric Formulation Initiative (EPFI) and United States Pediatric Formulation Initiative (USPFI) have created a searchable common database to assist manufacturers and compounders alike in identifying age-suitable excipients. Common excipients and preservatives found in the labeling of manufactured products marketed in the US include water, citric acid, glycerin, methyl- and propylparaben, sodium benzoate, and sodium citrate, as well as various forms of cellulose, sugars and sweeteners, and sugar alcohols. Based on the EPFI’s S.T.E.P database and PubChem, as well as a recent comprehensive review, Table 2 lists recently manufactured oral liquid products, their excipients and preservatives, and comments about the suitability for use in pediatric patients [11]. Caution should be exercised in the use of these newly approved oral liquids, especially in neonates.

4. Additional APIs Marketed in Manufactured Oral Liquid or Granule Dosage Forms

An additional 20 APIs have been FDA-approved and marketed in the US in an oral liquid or granule or powder for reconstitution (Table 3). Of the products listed in Table 3, several are approved for adult populations only (bolded), and their suitability for use as off-label treatment in children needs to be established. Of note, Azurity Pharmaceuticals has INDs submitted for two antiepileptic medications in oral liquids—lamotrigine and zonisamide. Two other products, topiramate 25 mg/mL (Eprontia™) and baclofen 5 mg/mL oral liquid formulations, were recently FDA-approved. Of note, an unapproved but marketed metronidazole 50 mg/mL product also became available as a compounding kit from Azurity.

5. The Next Wave of Oral Liquid Formulation Development—Formulation Considerations

The following nine APIs were included on the list of 16 and represent the next wave of candidates for conversion from extemporaneously CNSPs to commercially available FDA-approved finished liquid dosage form products, in order of their potential for development as a commercial market. Those CNSPs include ursodiol, bosentan, captopril, pantoprazole, valacyclovir, clopidogrel, acetazolamide, warfarin, and nifedipine. The suggested concentrations of oral liquid dosage forms are based on four factors: (1) the usual dosage range; (2) the maximum adult dose and volume (not to exceed 20 mL); (3) the expected water solubility; and (4) the standardized concentration for the extemporaneously compounded preparation, if applicable (Table 4).
  • Ursodiol or ursodeoxycholic acid (UDCA) [12,13,14,15,16,17,18,19] inhibits the hepatic synthesis and secretion of cholesterol and its intestinal absorption. It is indicated in primary biliary cirrhosis and for the prevention and treatment of gallstones. It is BCS class II due to low solubility and high permeability. Development of an oral liquid may be facilitated using methylcellulose and glycerin as excipients and via incorporation into polymeric nanoparticle carriers [20,21].
  • Bosentan [22,23], a hazardous medication, is a sulfonamide-derived, dual endothelin receptor antagonist used in the treatment of pulmonary arterial hypertension [22,24]. It belongs to BCS class II, and is available in solid and quadrisected dispersible tablets. Its oral bioavailability may be increased through nanosuspension [25] and water-rich co-solvent mixtures using propylene glycol [26].
  • Captopril, [4,27,28,29,30,31,32,33,34] is an angiotensin I-converting enzyme inhibitor (ACE-I) indicated in the treatment of heart failure and hypertension [34,35,36]. It is a BCS class I agent and is freely soluble in water (160 mg/mL), although its stability is limited due to disulfide formation. Fast-dispersing tablet formulations in 2.5 and 10 mg strengths for reconstitution have been suggested [37].
  • Pantoprazole [4,38] is a proton pump inhibitor prodrug in the benzimidazole family with a provisional BCS class III designation due to its high solubility and low permeability. It is currently available in a delayed release granule for the preparation of an oral suspension used to treat erosive esophagitis, gastroesophageal reflux disease, and Zollinger–Ellison syndrome. Dividing the granule formulation into smaller doses in children is not recommended in the product’s labeling. Formulation options include alginate–pectin polymeric raft-forming systems and divisible buccal films [39,40,41].
  • Valacyclovir [4,42,43,44,45] is a valyl ester prodrug that is converted to acyclovir, and is indicated for herpes labialis and zoster in children. A BCS class III agent, its stability in solution is pH-dependent at concentration ranges from 2.2 to 174 mg/mL [46]. Valacyclovir tablet product labeling includes instructions for preparing a CNSP at concentrations of 25 and 50 mg/mL in 100 mL quantities, using a suspension-structured vehicle and cherry flavor to mask the bitter taste. This product has beyond use dating of up to 28 days when stored under refrigeration. However, solution formulations using powdered API with combinations of glycerol and maltodextrin have been suggested [47,48].
  • Clopidogrel [4,49,50,51], a prodrug activated in two steps primarily by CYP 2C19, is a BCS class II agent with a bioavailability of about 50%. Its uses in children are for arterial ischemic stroke, heart disease, and management of endovascular stents [52,53]. Stabilization with stearoyl polyoxylglycerides (Gelucire® 50/13) and polyoxyethylated castor oil (Cremophor® RH40) have been suggested for self-emulsifying oral drug delivery formulations to improve the bioavailability and storage duration [54].
  • Acetazolamide [4] is a potent inhibitor of the enzyme carbonic anhydrase, which catalyzes the reversible hydration of carbon dioxide and dehydration of carbonic acid, resulting in the renal loss of bicarbonate anion, sodium, and water. Its primary uses in children are for the treatment of metabolic alkalosis, seizure disorder, glaucoma (topically), and intracranial hypertension [55,56,57,58]. It is available as a lyophilized powder for injection. Acetazolamide is very slightly soluble in water (BCS class IV), and its oral formulation bioavailability may be enhanced through application of mucoadhesive nanoparticles and the use of spray drying techniques [59,60,61].
  • Warfarin is an epoxide reductase inhibitor of the synthesis of vitamin K-dependent clotting factors, including the anticoagulant proteins C and S, as well as factor II, VII, IX, and X. Its indications include the prophylaxis and treatment of venous thromboembolism, pulmonary embolism, and complications associated with atrial fibrillation, myocardial infarction, and ischemic stroke. Warfarin is listed as BCS class I. While the labeled stability of a reconstituted intravenous injection is 4 h, more dilute oral solutions (1 mg/mL in aqueous media) may have longer stability [62,63]. The use of a semisolid extrusion of orodispersible hydroxypropylcellulose films created through the use of 3D printers may hold promise for both the preparation and individualization of doses [63,64].
  • Nifedipine [65] is a dihydropyridine calcium channel blocker formulated as a liquid-filled capsule. It is indicated for chronic hypertension and vasospastic or chronic unstable angina [34,35]. It is BCS class II with poor water solubility, and undergoes extensive first pass metabolism. The formulation of a powder for reconstitution may be facilitated by reductions in particle size through high-pressure homogenization or fabricated nanosponge encapsulation [32,66,67,68,69].
Table 4. Next-wave mass-manufactured API candidates.
Table 4. Next-wave mass-manufactured API candidates.
MedicationDosing Range in Children (mg/kg/day)Solubility in Water at RT and Neutral pH (mg/mL)Suggested Mass Production ConcentrationSuggested Mass Production Liquid Dose Form
ursodiol10–400.0260 mg/mL *Nanoparticle suspension
bosentan3–40.436.25 mg/mLPowder for reconstitution
captopril0.02–0.31601 mg/mL *Powder for reconstitution
pantoprazole0.5–10.0482 mg/mLPowder for reconstitution
valacyclovir60–12017450 mg/mL *Solution
clopidogrel0.20.0515 mg/mLPowder for reconstitution
acetazolamide8–300.925 mg/mLNanoparticle powder for reconstitution
warfarin0.05–0.350.0171 mg/mLPowder for reconstitution
nifedipine1–20.00594 mg/mL *Nanoparticle powder for reconstitution
(Note: * indicates ASHP standardized concentration [70]).

5.1. Additional API Candidates with High Potential for Development as Commercially Available FDA-Approved Finished Liquid Dosage Forms

Table 5 lists an additional 26 mass production candidate APIs, eight of which are on the hazardous drugs list [71]. BCS class I APIs with the highest mass marketing potential include hydroxyurea, allopurinol, flecainide, and cyclophosphamide. For BCS class II, amiodarone, carvedilol, isradipine, quetiapine, and losartan have the highest potential marketability. In BCS class III, clonidine, hydralazine, apixaban, atenolol, and ganciclovir top the list. Finally, BCS class IV API include hydrochlorothiazide and azathioprine. Hazardous APIs with mass marketing potential included hydroxyurea, ganciclovir, cyclophosphamide, and azathioprine. The commercial availability of finished liquid dosage forms for hazardous medications for oral administration affords an opportunity to reduce overall inadvertent toxic exposure due to dose manipulation in institutional settings and at home, irrespective of other evaluation criteria. These APIs:
  • Have the highest marketing potential for pediatric populations in terms of off-label use in prioritized therapeutic categories (antiarrhythmics, antibiotics, antihypertensives, antineoplastics, central nervous system agents, and proton pump inhibitors) [72];
  • Appear on the FDA and World Health Organization list of essential medicines [73];
  • Have a standardized concentration identified on the ASHP Standardize 4 safety list [70,74];
  • Are available in an intravenous formulation that would support potential oral formulation feasibility; and
  • Have a pediatric Biopharmaceutical Classification System (BCS) classification indicating relative ease of generating suitable oral liquid formulations intended primarily for children [75].
BCS class II and IV APIs have low solubility and high or low gastrointestinal permeability, and several techniques have been forwarded to address them. In many cases, the optimal solubility–permeability balance is based on the extent to which membrane–aqueous partitioning is maximized; that is, the intersection where both are at their highest relative values [76]. Formulation methods to increase either solubility or dissolution rates for BCS class II and IV APIs identified in the literature include:
  • 3D printing [63,77,78,79];
  • Amorphous solid dispersion [59,80,81];
  • Complexation [82,83];
  • Fusion [84,85];
  • Hot-melt extrusion [84,86];
  • Lipid microemlusion [54];
  • Lyophilization [84,87,88];
  • Micelles [87];
  • Nanosizing [60,89]; and
  • Spray drying [59,60].
Excipients that have been shown to increase gastrointestinal tract permeability include:
  • Cyclodextrins [67,82];
  • Surfactants [53,88]; and
  • Cosolvents [90].
A brief product profile for the APIs with the highest potential for development as commercially available FDA-approved finished liquid dosage forms includes: (1) clinical pharmacology/PK/PD/PG; (2) indications and dosing regimens; and (3) clinical pharmaceutics and potential formulation characteristics.

5.1.1. High Solubility/High Permeability (BCS Class I)

  • Hydroxyurea [91] is an antimetabolite used to treat sickle cell anemia crisis; management of melanoma; resistant chronic myelocytic leukemia; and recurrent, metastatic, or inoperable carcinoma of the ovaries. It inhibits DNA synthesis through the inhibition of ribonucleoside diphosphate reductase. It is well absorbed orally and the water solubility is 100 mg/mL. An oral liquid with a concentration of 100 mg/mL has been studied [92].
  • Allopurinol is a xanthine oxidase inhibitor used to reduce urinary and serum uric acid concentrations in patients with gout, recurrent calcium oxalate calculi, and various malignancies. Children 6 to 10 years of age with secondary hyperuricemia associated with malignancies may be given 300 mg allopurinol daily, while those under 6 years are generally given 150 mg daily. The response is evaluated after approximately 48 h of therapy and a dosage adjustment is made if necessary. The solubility in water at room temperature is between 0.48 and 0.57 mg/mL. A 500 mg lyophilized injection is available. Hydrophilic carriers such as polyvinylpyrrolidone/polyethylene glycol 6000 at ratios of 1:1, 1:2, and 1:4 (drug to carrier ratio) have been shown to increase aqueous solubility [80].
  • Flecainide [93] is a class Ic antiarrhythmic agent used to manage atrial fibrillation and paroxysmal supraventricular tachycardias (PSVT). Its water solubility is 48.4 mg/mL at 37 °C. Dosing in children is usually less than 100 mg per dose. A 20 mg/mL formulation using bulk powder and purified water and simple syrup (50:50) resulted in a transparent solution [94].
  • Cyclophosphamide [95] is an alkylating nitrogen mustard antineoplastic and immunosuppressive agent that must be activated in the liver to aldophosphamide. In addition to solid tumors, it is also indicated for the treatment of biopsy-proven minimal change nephrotic syndrome and membranous nephritis in pediatric patients [96]. For nononcologic indications, oral cyclophosphamide dosing usually in the range of 2.5 to 3 mg/kg daily for a period of 60 to 90 days is recommended for both initial and maintenance dosing, and can be used in place of a low-dose intravenous formulation. It is soluble in water at between 10 and 50 mg/mL, and liquid formulations using the lyophilized intravenous formulation may be prepared in a 10 mg/mL concentration [97].

5.1.2. Low Solubility/High Permeability (BCS Class II)

  • Amiodarone, considered a class III antiarrhythmic with α- and β-receptor antagonism, is a benzofuran derivative indicated for initiation of treatment and prophylaxis of frequently recurring ventricular fibrillation (VF) and hemodynamically unstable ventricular tachycardia (VT) in patients that are refractory to other therapy. Most patients will require this therapy for 48 to 96 h, but amiodarone may be safely administered for longer periods if necessary. Pediatric dosing ranges from 10 to 15 mg/kg/day in 1 to 2 divided doses/day for 4 to 14 days or until adequate control of arrhythmia or prominent adverse effects occur. Dosage should then be reduced to 5 mg/kg/day given once daily for several weeks. If arrhythmia does not recur, reduce to the lowest effective dosage possible. Usual daily minimal dose: 2.5 mg/kg/day; maintenance doses may be given for 5 or 7 days/week. Amiodarone is available in a 50 mg/mL intravenous injection, exhibits 0.72 mg/mL solubility in water, and is highly lipophilic. A 5 mg/mL formulation at pH = 4 with cherry flavoring has been suggested [33].
  • Carvedilol [12] is a racemic mixture where the S(-) enantiomer is a beta adrenoceptor blocker and the R(+) enantiomer is both a beta and alpha-1 adrenoceptor blocker. It is currently used to treat heart failure, left ventricular dysfunction, and hypertension. Pediatric dosing ranges from 0.4 to 0.8 mg/kg/day in 2 divided doses. It is virtually insoluble in water, and is highly lipophilic. A 1–1.25 mg/mL oral suspension has been suggested [31].
  • Isradipine [35,36] belongs to the dihydropyridine (DHP) class of calcium channel blockers (CCBs), the most widely used class of CCBs. It is structurally related to felodipine, nifedipine, and nimodipine, and is the most potent calcium channel blocking agent of the DHP class. Isradipine binds to calcium channels with high affinity and specificity and inhibits calcium flux into cardiac and arterial smooth muscle cells. It exhibits greater selectivity towards arterial smooth muscle cells owing to alternative splicing of the alpha-1 subunit of the channel and increased prevalence of inactive channels in smooth muscle cells. Isradipine may be used to treat mild to moderate essential hypertension. Pediatric dosing ranges from 0.15 to 0.2 mg/kg/day divided every 6 to 8 h with a maximum dosage of 0.8 mg/kg/day, not to exceed 20 mg/day. It is practically insoluble in water. A 1 mg/mL suspension has been compounded since its initial marketing [98].
  • Quetiapine is a dopamine type 2 and serotonin 2A receptor antagonist and binds to the norepinephrine transporter. Additional effects of quetiapine, including somnolence, orthostatic hypotension, and anticholinergic effects, may result from the antagonism of histamine-1, adrenergic α1, and muscarinic-1 receptors, respectively. It is used in the management of bipolar disorder, schizophrenia, major depressive disorder, and delirium [99]. Quetiapine is rapidly and well absorbed after administration of an oral dose, and a steady state is achieved within 48 h. It is metabolized by CPY 2D6 and 3A4. The water solubility of quetiapine is 0.6 mg/mL with a pKa of 7.06. Pediatric dosing ranges from 0.5 to 6 mg/kg/day. Nanotechnology formulations of 2.5, 5, 10, 20 and 40 mg/mL have been suggested [100,101].
  • Losartan [35] is an angiotensin receptor antagonist indicated to treat hypertension in patients older than 6 years, to reduce the risk of stroke in patients with hypertension and left ventricular hypertrophy, and to treat diabetic nephropathy with elevated serum creatinine and proteinuria in patients with type 2 diabetes and hypertension. It reversibly and competitively prevents angiotensin II binding to the AT1 receptor in tissues like vascular smooth muscles and the adrenal gland. The usual recommended starting dose is 0.7 mg/kg once daily (up to 50 mg total), and dosages above 1.4 mg/kg (or in excess of 100 mg) daily have not been studied in pediatric patients. Losartan has an 8 mg/mL water solubility level with a pKa of 5.5. A 5 mg/mL formulation has recently been studied [102], and directions for the preparation of a 2.5 mg/mL suspension are contained in the package insert [103].

5.1.3. High Solubility/Low Permeability (BCS Class III)

  • Clonidine [104,105,106,107,108] is an imidazole derivate that acts as an agonist of alpha-2 adrenoceptors used to treat hypertension and severe cancer pain, among other conditions, and to treat withdrawal symptoms from various substances. It is available in a 0.1 mg/mL liquid solution for injection and is insoluble in water. Its bioavailability is between 55 and 87%, and the primary metabolism includes hydroxylation via CYP2D6, CYP1A2, CYP3A4, CYP1A1, and CYP3A5 enzymes. The usual pediatric dose range is between 5 and 10 mcg/kg/day orally in divided doses every 8 to 12 h then titrated based on clinical response, with a maximum dose of 25 mcg/kg/day or 0.9 mg/day.
  • Hydralazine [35] is a direct-acting vasodilator that is used as an antihypertensive agent. It inhibits the phosphorylation of myosin protein and chelation of trace metals required for smooth muscle contraction, resulting in increases in heart rate, stroke volume, and cardiac output. Available in a 20 mg/mL injection solution, hydralazine is freely soluble in water. The initial dose is 0.75 mg/kg/day in 4 divided doses, with gradual increases over 3 to 4 weeks to a maximum of 7.5 mg/kg/day or 200 mg/day. Taking oral hydralazine with food improves the bioavailability of the drug. A 4 mg/mL suspension using crushed hydralazine tablets has been suggested [49].
  • Apixaban [109] is an oral, direct, and highly selective factor Xa (FXa) inhibitor of both free and bound FXa, as well as prothrombinase, independent of antithrombin III, for the prevention and treatment of thromboembolic diseases. Children 12 to <18 years old weighing less than 40 kg have received an apixaban dose of 0.2 mg/kg twice daily for 7 days followed by 0.1 mg/kg twice daily, whereas children at the same age weighing more than 40 kg receive the adult VTE treatment dose (i.e., 10 mg twice daily for 7 days followed by 5 mg twice daily). Apixaban is approximately 50% bioavailable, and is mainly metabolized by cytochrome CYP 3A4 and to a lesser extent by CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2J2. It has a water solubility of 0.11 mg/mL. A 0.25 mg/mL suspension with a 7-day stability has been reported [110].
  • Atenolol [36] is a synthetic beta-1 selective blocker used in the management of hypertension and chronic angina, and to reduce mortality in known or suspected myocardial infarction in hemodynamically stable patients. It is available in a liquid injection at a concentration of 0.5 mg/mL. Approximately 50% of an oral dose is absorbed from the gastrointestinal tract, the remainder being excreted unchanged in the feces. Its water solubility is 13.3 mg/mL. The typical dosage range for children is 0.5–1 mg/kg/day given once daily or divided in 2 doses per day with a maximum dose of 2 mg/kg/day [111].
  • Ganciclovir, a DNA polymerase inhibitor prodrug used to treat cytomegalovirus (CMV) and herpetic keratitis of the eye and to prevent CMV in organ transplantation, must be converted to the active form by a virus-encoded cellular enzyme, thymidine kinase [112]. Oral administration follows intravenous induction. It has a solubility of 3 mg/mL in water with pKa values of 2.2 and 9.4. Pediatric dosing in normal renal function ranges 20–40 mg/kg or 500–700 mg/m2 every 8 h [113,114].

5.1.4. Low Solubility/Low Permeability (BCS Class IV)

  • Hydrochlorothiazide [34,115] is a thiazide diuretic used alone or in combination for the management of edema associated with congestive heart failure, hepatic cirrhosis, nephrotic syndrome, acute glomerulonephritis, chronic renal failure, and hypertension. Hydrochlorothiazide acts on the proximal region of the distal convoluted tubule, inhibiting reabsorption by the sodium-chloride symporter, also known as solute carrier family 12 member 3 (SLC12A3). It has a water solubility level of 0.7 mg/mL. Because of its poor oral absorption, several novel dosage forms have been proposed, including orally disintegrating mini-tablets, liquid complexations with cyclodextrin, and nanostructured lipid carriers [116,117,118].
  • Azathioprine, a prodrug of 6-mercaptopurine, is used to treat inflammatory conditions such as rheumatoid arthritis, systemic lupus erythematosus, and inflammatory bowel diseases, as well as being an immunosuppressant in the prevention of renal transplant rejection [119]. The usual daily dose ranges from 1 to 5 mg/kg orally. It is insoluble in water, with a pKa of 7.8. Both 10 mg/mL and 50 mg/mL suspensions have been studied [49,120].

5.2. Extemporaneous CNSP APIs Included in USP CC Monographs

Extemporaneous CNSP APIs included in USP CC monographs that could be developed as FDA-approved manufactured products, as well as those without a mass market, are outlined below [121]. The list is broken down into those APIs whose compounding recipes are found currently (1) in the USP CC and (2) those available in other sources. A total of 45 APIs are included in these two lists (23 found in USP CC). Those APIs listed in the USP CC that are suitable for development as commercially available FDA-approved finished liquid dosage forms include clonazepam, desmopressin phytonadione, pyridoxine, rifampin, and ethambutol. Of note, desmopressin, phytonadione, and pyridoxine are available in liquid injections. Those that should remain as extemporaneously CNSPs include pyrazinamide, dapsone, diltiazem, ketoconazole, metolazone, pyrimethamine, rifabutin, bethanechol, propylthiouracil, dipyridamole, chloroquine, quinidine, temozolomide, terbutaline, tetracycline, tiagabine, dolasetron, and phenoxybenzamine.

5.3. Potentially Approvable Products from Extemporaneously Compounded APIs Not Included in USP CC

Zinc, buprenorphine, naltrexone, and everolimus represent API CNSP formulations available from other sources that are suitable for development and approval as commercially available finished liquid dosage forms. After appropriate testing for stability, those that could be incorporated into USP CC included amitriptyline, hydroxychloroquine, thiamine, rifaximin, valsartan, venlafaxine, buspirone, phenazopyridine, dantrolene, mexiletine, nadolol, pravastatin, topotecan, tretinoin, chlorpromazine, ethacrynic acid, flucytosine, amiloride, primaquine, procarbazine, and disopyramide. As a result of validating the monographs of over 59 listed APIs, USP identified seven CNSPs that failed stability testing using the present formulation recipe, including methimazole, phenazopyridine, probenecid, and trazodone CNSP liquids [122]. While none are considered to have high mass market potential, efforts to reformulate these APIs as stable CNSPs should be undertaken.

6. Discussion

The current pace of the creation and production of suitable oral dosage forms for pediatrics, whether through FDCA 505A or 503A and 503B mechanisms, could in part represent what some would call an “unfulfilled’ promise involving personalized medicine [123]. Efforts to develop oral liquid formulations for vulnerable populations, including pediatric and geriatric patients, especially for rare diseases, must continue to accelerate so that the uncertainties associated with off-label drug preparation and use are minimized and therapeutic benefits are optimized [124]. Moreover, patients and their families need assurances that formulations prepared for them have scientific evidence of safety and effectiveness based on governmental approval or professional authorization mechanisms. This is especially true with the emergence of targeted therapies for childhood cancers. For example, the BCR-ABL1 target tyrosine kinase inhibitor (TKI) imatinib was the only TKI approved for front-line treatment of pediatric patients with chronic myeloid leukemia (CML) in chronic phase (CML-CP) until dasatinib demonstrated superior efficacy [125]. A bioequivalence study involving 50 mg dispersable tablets and a 10 mg/mL suspension conducted in adults demonstrated that the bioavailability of dasatinib powder for oral suspension (PFOS) was 19% lower than that of dasatinib tablets. However, while available in oral tablets strengths ranging from 20 to 140 mg, the innovator company markets neither PFOS nor dispersible tablet formulations [126]. Further, the labeling of the product prohibits tablet crushing, making an extemporaneous oral formulation “off-label” [127]. Table 6 lists common pediatric cancers treated with targeted therapies and the availability of manufactured or compounded oral liquid formulations. This list reveals the commercial and professional gaps in the development and availability of high-quality formulations for many commonly used targeted therapies from the published scientific literature and routine clinical practice.

7. Summary

The purpose of this work was to evaluate the suitability of recent FDA-approved and marketed oral liquid, powder, or granule products; to identify the next group of APIs with potential for mass marketing and FDA approval; and to propose CNSPs that should be developed as approved and manufactured products, as well as those that should continue to be extemporaneously prepared. A total of 25 APIs with the highest potential for mass marketing as FDA-approved products were identified. Table 7 summarizes the remaining 48 identified APIs based on their potential for further development, either as manufactured products (n = 10) and compounded preparations (n = 38). Six USP CC-listed APIs are suitable for development and approved as commercially available FDA-approved finished liquid dosage forms, and four non-USP CC-listed CNSPs should be developed as approved mass manufactured products. Those CNSPs listed in USP CC without high mass marketing and approval potential have also been identified, as well as APIs that USP should be considered for addition to the CC, perhaps through active solicitation for the formulation monograph donation program (CPMDonate@usp.org) [138]. The current state of development of formulations for targeted therapies represents an identified gap in personalized treatments for vulnerable populations (Table 6). Through this identification and categorization process, the authors encourage industry, government, and the professions to continue to work together to improve the likelihood that patients will receive high-quality standardized extemporaneously prepared CNSPs and FDA-approved mass manufactured products.

Author Contributions

Conceptualization, R.H.P.II; Project administration, R.H.P.II; Writing—original draft, R.H.P.II; Writing—review & editing, R.H.P.II, L.D.A., R.L., S.B., J.J.C. and R.B.M. All authors have read and agreed to the published version of the manuscript.

Funding

RM’s work was funded in part by The Rockefeller University Center for Clinical and Translational Science grant # UL1 TR001866 from the National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH) Clinical and Translational Science Award (CTSA) program.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors report no conflict of interest.

References

  1. Bhatt-Mehta, V.; MacArthur, R.B.; Löbenberg, R.; Cies, J.J.; Cernak, I.; Parrish, R.H. Development of an algorithm to identify mass production candidate molecules to develop children’s oral medicines: A North American perspective. AAPS Open 2016, 2, 8. [Google Scholar] [CrossRef] [Green Version]
  2. United States Food and Drug Administration. Compounded Drug Products That Are Essentially Copies of a Commercially Available Drug Product under Section 503A of the Federal Food, Drug, and Cosmetic Act: Guidance for Industry. 2016; pp. 1–11. Available online: https://www.fda.gov/media/98973/download (accessed on 3 March 2022).
  3. United States Food and Drug Administration. Guidance for Industry: Applications Covered by Section 505(b)(2). Guidance. 1999. Available online: https://www.fda.gov/media/72419/download (accessed on 3 March 2022).
  4. United States Pharmacopeial Convention. June 2020/USP Compounding Compendium. 2020. Available online: https://www.usp.org/products/usp-compounding-compendium (accessed on 23 October 2020).
  5. Ivanovska, V.; Rademaker, C.M.A.; Van Dijk, L.; Mantel-Teeuwisse, A.K. Pediatric drug formulations: A review of challenges and progress. Pediatrics 2014, 134, 361–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lopez, F.L.; Ernest, T.B.; Tuleu, C.; Gul, M.O. Formulation approaches to pediatric oral drug delivery: Benefits and limitations of current platforms. Expert Opin. Drug Deliv. 2015, 12, 1727–1740. [Google Scholar] [CrossRef] [PubMed]
  7. Strickley, R.G. Pediatric Oral Formulations: An Updated Review of Commercially Available Pediatric Oral Formulations Since 2007. J. Pharm. Sci. 2019, 108, 1335–1365. [Google Scholar] [CrossRef] [PubMed]
  8. Bar-Shalom, D. Necessity of rethinking oral pediatric formulations. Clin. Ther. 2014, 36, 180–183. [Google Scholar] [CrossRef] [PubMed]
  9. Binson, G.; Sanchez, C.; Waton, K.; Chanat, A.; Di Maio, M.; Beuzit, K.; Dupuis, A. Accuracy of Dose Administered to Children Using Off-Labelled or Unlicensed Oral Dosage Forms. Pharmaceutics 2021, 13, 1014. [Google Scholar] [CrossRef]
  10. United States Pharmacopeia. FAQs: Excipients. Available online: https://www.usp.org/frequently-asked-questions/excipents (accessed on 22 February 2022).
  11. Rouaz, K.; Chiclana-Rodríguez, B.; Nardi-Ricart, A.; Suñé-Pou, M.; Mercadé-Frutos, D.; Suñé-Negre, J.M.; Pérez-Lozano, P.; Garcia-Montoya, M.E. Excipients in the Paediatric Population: A Review. Pharmaceutics 2021, 13, 387. [Google Scholar] [CrossRef]
  12. Polonini, H.; da Silva, S.L.; Brandão, M.A.F.; Bauters, T.; De Moerloose, B.; Ferreira, A.O. Compatibility of Baclofen, Carvedilol, Hydrochlorothiazide, Mercaptopurine, Methadone Hydrochloride, Oseltamivir Phosphate, Phenobarbital, Propranolol Hydrochloride, Pyrazinamide, Sotalol Hydrochloride, Spironolactone, Tacrolimus Monohydrate, Ursodeoxycholic Acid, and Vancomycin Hydrochloride Oral Suspensions Compounded with SyrSpend SF pH4. Int. J. Pharm. Compd. 2018, 22, 516–526. [Google Scholar]
  13. Johnson, C.E.; Nesbitt, J. Stability of ursodiol in an extemporaneously compounded oral liquid. Am. J. Health Syst. Pharm. 1995, 52, 1798–1800. [Google Scholar] [CrossRef]
  14. Rudic, J.S.; Poropat, G.; Krstic, M.N.; Bjelakovic, G.; Gluud, C. Ursodeoxycholic acid for primary biliary cirrhosis. Cochrane Database Syst. Rev. 2012, 12, CD000551. [Google Scholar] [CrossRef]
  15. Trissel, L.A.; Ashworth, L.D.; Ashworth, J. (Eds.) Ubidecarenone–Ursodiol. In Trissel’s Stability of Compounded Formulations, 6th ed.; American Pharmacists Asociation: Washington, DC, USA, 2018. [Google Scholar] [CrossRef]
  16. Mallett, M.S.; Hagan, R.L.; Peters, D.A. Stability of ursodiol 25 mg/mL in an extemporaneously prepared oral liquid. Am. J. Health Syst. Pharm. 1997, 54, 1401–1404. [Google Scholar] [CrossRef] [PubMed]
  17. Pramar, Y.V.; Mandal, T.K.; Bostanian, L.A.; Nguyen, A.T.; Miller, V.; Morris, T.C.; Graves, R.A. Stability of Compounded Ursodiol Suspensions in PCCA Base, SuspendIt. Int. J. Pharm. Compd. 2019, 23, 70–76. [Google Scholar] [PubMed]
  18. de Oliveira Junior, E.R.; Truzzi, E.; Ferraro, L.; Fogagnolo, M.; Pavan, B.; Beggiato, S.; Rustichelli, C.; Marettim, E.; Lima, E.M.; Leo, E.; et al. Nasal administration of nanoencapsulated geraniol/ursodeoxycholic acid conjugate: Towards a new approach for the management of Parkinson’s disease. J. Control. Release 2020, 21, 540–552. [Google Scholar] [CrossRef]
  19. Geiger, C.M.; Voudrie, M.A., 2nd; Sorenson, B. Stability of ursodiol in SyrSpend SF Cherry flavored. Int. J. Pharm. Compd. 2012, 16, 510–512. [Google Scholar] [PubMed]
  20. Tereshchenko, O.G.; Nikolskaya, E.D.; Zhunina, O.A.; Zavarzina, V.; Yabbarov, N.G.; Fomicheva, M.V.; Zubkov, E.V.; Sokol, M.B.; Gukasova, N.V.; Severin, E.S. Formulation of perspective hepatoprotector polymeric forms based on silybin and ursodeoxycholic acid. Russ. Chem. Bull. 2019, 67, 2290–2296. [Google Scholar] [CrossRef]
  21. Santoveña, A.; Sánchez, E.; Charola, L.; Llabrés, M.; Fariña, J.B. Study of quality and stability of ursodeoxycholic acid formulations for oral pediatric administration. Int. J. Pharm. 2014, 477, 32–38. [Google Scholar] [CrossRef] [PubMed]
  22. Berger, R.M.F.; Hawort, S.G.; Bonnet, D.; Dulac, Y.; Fraisse, A.; Galiè, N.; Ivy, D.D.; Jaïs, X.; Miera, O.; Rosenzweig, E.B.; et al. FUTURE-2: Results from an open-label, long-term safety and tolerability extension study using the pediatric FormUlation of bosenTan in pUlmonary arterial hypeRtEnsion. Int. J. Cardiol. 2016, 202, 52–58. [Google Scholar] [CrossRef]
  23. Carter, N.J.; Keating, G.M. Bosentan: In pediatric patients with pulmonary arterial hypertension. Pediatr. Drugs 2010, 12, 63–73. [Google Scholar] [CrossRef]
  24. Beghetti, M.; Haworth, S.G.; Bonnet, D.; Barst, R.J.; Acar, P.; Fraisse, A.; Ivy, D.D.; Jais, X.; Schulze-Neick, I.; Galiè, N.; et al. Pharmacokinetic and clinical profile of a novel formulation of bosentan in children with pulmonary arterial hypertension: The FUTURE-1 study. Br. J. Clin. Pharmacol. 2009, 68, 948–955. [Google Scholar] [CrossRef] [Green Version]
  25. Ghasemian, E.; Motaghian, P.; Vatanara, A. D-optimal design for preparation and optimization of fast dissolving bosentan nanosuspension. Adv. Pharm. Bull. 2016, 6, 211–218. [Google Scholar] [CrossRef] [Green Version]
  26. Babaei, M.; Shayanfar, A.; Rahimpour, E.; Barzegar-Jalali, M.; Martinez, F.; Jouyban, A. Solubility of bosentan in {propylene glycol + water} mixtures at various temperatures: Experimental data and mathematical modelling. Phys. Chem. Liq. 2019, 57, 3. [Google Scholar] [CrossRef]
  27. Chan, D.S.; Sato, A.K.; Claybaugh, J.R. Degradation of captopril in solutions compounded from tablets and standard powder. Am. J. Hosp. Pharm. 1994, 51, 1205–1207. [Google Scholar] [CrossRef] [PubMed]
  28. Allen, L.V.; Erickson, M.A. Stability of baclofen, captopril, diltiazem hydrochloride, dipyridamole and flecainide acetate in extemporaneously compounded oral liquids. Am. J. Health Syst. Pharm. 1996, 53, 2179–2184. [Google Scholar] [CrossRef]
  29. Lin, T.R.; Yang, Y.H.K.; Huang, Y.F.; Tsai, J.C. Content uniformity of captopril, furosemide, nadolol and propranolol hydrochloride powder packets extemporaneously compounded from tablets. Chin. Pharm. J. 2000, 52, 58–63. [Google Scholar] [CrossRef]
  30. Geiger, C.M.; Sorenson, B.; Whaley, P.A. Stability of captopril in SyrSpend SF. Int. J. Pharm. Compd. 2013, 17, 336–338. [Google Scholar] [PubMed]
  31. Kuriata, E.; Sawicki, W. Evaluation of cases with the usage of commercially available tablets in the pediatric formula. Acta Pol. Pharm. 2015, 72, 551–558. [Google Scholar]
  32. Harris Tieca, T.; Adebayo Amusa, S.; Deon, B.; Oluwayomi, O.; Charmaine, S. Extemporaneous compounding for children with coronary heart diseases in Jamaica. Eur. J. Clin. Pharm. 2018, 20, 204–212. [Google Scholar]
  33. Thrimawithana, T.R.; D’Amore, S.; Dib, Y.; Fadavi Firooz, N.; Fakhouri, W.; Saeed, A.; Allahham, A. Critical appraisal of commercially available suspending vehicles for extemporaneous compounding of cardiovascular medicines: Physical and chemical stability mini review. Pharm. Dev. Technol. 2019, 24, 529–538. [Google Scholar] [CrossRef]
  34. Momma, K. ACE inhibitors in pediatric patients with heart failure. Pediatr. Drugs 2006, 8, 55–69. [Google Scholar] [CrossRef]
  35. Meyers, R.S.; Siu, A. Pharmacotherapy Review of Chronic Pediatric Hypertension. Clin. Ther. 2011, 33, 1331–1356. [Google Scholar] [CrossRef]
  36. Robinson, R.F.; Nahata, M.C.; Batisky, D.L.; Mahan, J.D. Pharmacologic treatment of chronic pediatric hypertension. Pediatr. Drugs 2005, 7, 27–40. [Google Scholar] [CrossRef] [PubMed]
  37. Pabari, R.M.; McDermott, C.; Barlow, J.; Ramtoola, Z. Stability of an Alternative Extemporaneous Captopril Fast-Dispersing Tablet Formulation versus an Extemporaneous Oral Liquid Formulation. Clin. Ther. 2012, 34, 2221–2229. [Google Scholar] [CrossRef] [PubMed]
  38. Dentinger, P.J.; Swenson, C.F.; Anaizi, N.H. Stability of pantoprazole in an extemporaneously compounded oral liquid. Am. J. Health Syst. Pharm. 2002, 59, 953–956. [Google Scholar] [CrossRef] [PubMed]
  39. Abbas, G.; Hanif, M. Development and pharmacokinetic evaluation of alginate-pectin polymeric rafts forming tablets using box behnken design. Drug Dev. Ind. Pharm. 2018, 44, 2026–2037. [Google Scholar] [CrossRef]
  40. Thombre, S.K.; Gaikwad, S.S. Design and development of mucoadhesive buccal delivery for Pantoprazole with stability enhancement in human saliva. Int. J. Pharm. Pharm. Sci. 2013, 5, 122–127. [Google Scholar]
  41. Dheerajvarma, K.; Sai Krishna, P.; Prasanna, L. Formulation and evaluation of Pantoprazole sodium sesquihydrate IR buccal films. Res. J. Pharm. Biol. Chem. Sci. 2014, 5, 881–886. [Google Scholar]
  42. Kimberlin, D.W.; Jacobs, R.F.; Weller, S.; van der Walt, J.S.; Heitman, C.K.; Man, C.Y.; Bradley, J.S. Pharmacokinetics and safety of extemporaneously compounded valacyclovir oral suspension in pediatric patients from 1 month through 11 years of age. Clin. Infect. Dis. 2010, 50, 221–228. [Google Scholar] [CrossRef]
  43. Jew, R.K.; Soo-Hoo, W.; Erush, S.C.; Amiri, E. (Eds.) Valacyclovir Suspension 50 mg/mL. In Extemporaneous Formulations for Pediatric, Geriatric, and Special Needs Patients, 3rd ed.; ASHP: Bethesda, MD, USA, 2020. [Google Scholar] [CrossRef]
  44. Fish, D.N.; Vidaurri, V.A.; Deeter, R.G. Stability of valacyclovir hydrochloride in extemporaneously prepared oral liquids. Am. J. Health Syst. Pharm. 1999, 56, 1957–1960. [Google Scholar] [CrossRef]
  45. Granero, G.E.; Amidon, G.L. Stability of valacyclovir: Implications for its oral bioavailability. Int. J. Pharm. 2006, 317, 14–18. [Google Scholar] [CrossRef]
  46. Granero, G.E.; Longhi, M.R.; Becker, C.; Junginger, H.E.; Kopp, S.; Midha, K.K.; Shah, V.P.; Stavchansky, S.; Dressman, J.B.; Barends, D.M. Biowaiver monographs for immediate release solid oral dosage forms: Acetazolamide. J. Pharm. Sci. 2008, 97, 3691–3699. [Google Scholar] [CrossRef]
  47. Bastiaans, D.E.T.; Bartels-Wilmer, C.M.; Colbers, A.P.H.; Heijens, C.A.; Velthoven-Graafland, K.; Smeets, O.S.; Vink, N.; Harbers, V.E.; Warris, A.; Burger, D.M. A new paediatric formulation of valaciclovir: Development and bioequivalence assessment. Arch. Dis. Child. 2016, 101, 971–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Bastiaans, D.E.T.; Immohr, L.I.; Zeinstra, G.G.; Strik-Albers, R.; Pein-Hackelbusch, M.; van der Flier, M.; de Haan, A.F.J.; Boelens, J.J.; Lankester, A.C.; Burger, D.M.; et al. In vivo and in vitro palatability testing of a new paediatric formulation of valaciclovir. Br. J. Clin. Pharmacol. 2017, 83, 2789–2797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Polonini, H.; da Silva, S.L.; Cunha, C.N.; Ferreira, A.O.; Anagnostou, K.; Dijkers, E. Stability of Azathioprine, Clonidine Hydrochloride, Clopidogrel Bisulfate, Ethambutol Hydrochloride, Griseofulvin, Hydralazine Hydrochloride, Nitrofurantoin, and Thioguanine Oral Suspensions Compounded with SyrSpend SF pH4. Int. J. Pharm. Compd. 2020, 24, 252–262. [Google Scholar] [PubMed]
  50. Yamreudeewong, W.; Dolence, E.K.; Teixeira, M.G. Stability of clopidogrel in three extemporaneously compounded oral liquid preparations. Int. J. Pharm. Compd. 2011, 14, 435–437. [Google Scholar]
  51. Ellis, D.P.; Rozek, T.; Milne, R.W. Stability of a compounded oral liquid formulation of clopidogrel for infants. J. Pharm. Pract. Res. 2020, 50, 321–328. [Google Scholar] [CrossRef]
  52. Soman, T.; Rafay, M.F.; Hune, S.; Allen, A.; MacGregor, D.; DeVeber, G. The risks and safety of clopidogrel in pediatric arterial ischemic stroke. Stroke 2006, 37, 1120–1122. [Google Scholar] [CrossRef]
  53. Hatai, Y.; Nykanen, D.G.; Williams, W.G.; Freedom, R.M.; Benson, L.N. Endovascular stents in children under 1 year of age: Acute impact and late results. Br. Heart J. 1995, 74, 689–695. [Google Scholar] [CrossRef] [Green Version]
  54. Kim, D.W.; Kwon, M.S.; Yousaf, A.M.; Balakrishnan, P.; Park, J.H.; Kim, D.S.; Lee, B.J.; Park, Y.J.; Yong, C.S.; Kim, J.O.; et al. Comparison of a solid SMEDDS and solid dispersion for enhanced stability and bioavailability of clopidogrel napadisilate. Carbohydr. Polym. 2014, 114, 365–374. [Google Scholar] [CrossRef]
  55. Cheung, M.S. Drugs Used in Paediatric Bone and Calcium Disorders. Endocr. Dev. 2015, 28, 277–290. [Google Scholar] [CrossRef]
  56. Cleves-Bayon, C. Idiopathic Intracranial Hypertension in Children and Adolescents: An Update. Headache 2018, 58, 485–493. [Google Scholar] [CrossRef]
  57. Bar, A.; Cies, J.; Stapleton, K.; Tauber, D.; Chopra, A.; Shore, P.M. Acetazolamide therapy for metabolic alkalosis in critically ill pediatric patients. Pediatr. Crit. Care Med. 2015, 16, e34–e40. [Google Scholar] [CrossRef] [PubMed]
  58. López, C.; Alcaraz, A.J.; Toledo, B.; Cortejoso, L.; Gil-Ruiz, M.A. Acetazolamide Therapy for Metabolic Alkalosis in Pediatric Intensive Care Patients. Pediatr. Crit. Care Med. 2016, 17, e551–e558. [Google Scholar] [CrossRef] [PubMed]
  59. Manchanda, S.; Sahoo, P.K. Topical delivery of acetazolamide by encapsulating in mucoadhesive nanoparticles. Asian J. Pharm. Sci. 2017, 12, 550–557. [Google Scholar] [CrossRef]
  60. Dureja, H.; Pandey, P. Optimizing the spray-drying parameters for a formulation of nanoparticles-in-microparticles system of acetazolamide. Drug Dev. Deliv. 2017. Available online: https://drug-dev.com/formulation-development-optimizing-the-spray-drying-parameters-for-a-formulation-of-nanoparticles-in-microparticles-system-of-acetazolamide/?msclkid=bd09d3bacfa011ecbdc02a358541c8d4 (accessed on 9 May 2022).
  61. Pandey, P.; Marwaha, R.K.; Nanda, A.; Dureja, H. Spray-Dried Nanoparticles-in-Microparticles System (NiMS) of Acetazolamide Using Central Composite Design. Nanosci. Nanotechnol. Asia 2017, 6, 146–156. [Google Scholar] [CrossRef]
  62. Sharley, N.A.; Yu, A.M.C.; Williams, D.B. Stability of mixtures formulated from warfarin tablets or powder. J. Pharm. Pract. Res. 2007, 37, 95–97. [Google Scholar] [CrossRef]
  63. Schlatter, J.; Cisternino, S. Stability of warfarin sodium flavoured preservative-free oral liquid formulations. Eur. J. Hosp. Pharm. 2018, 25, e98–e101. [Google Scholar] [CrossRef]
  64. Öblom, H.; Sjöholm, E.; Rautamo, M.; Sandler, N. Towards printed pediatric medicines in hospital pharmacies: Comparison of 2d and 3d-printed orodispersible warfarin films with conventional oral powders in unit dose sachets. Pharmaceutics 2019, 11, 334. [Google Scholar] [CrossRef] [Green Version]
  65. Gajendran, J.; Krämer, J.; Shah, V.P.; Langguth, P.; Polli, J.; Mehta, M.; Groot, D.; Cristofoletti, R.; Abrahamsson, B.; Dressman, J.B. Biowaiver Monographs for Immediate-Release Solid Oral Dosage Forms: Nifedipine. J. Pharm. Sci. 2015, 104, 3289–3298. [Google Scholar] [CrossRef]
  66. Geiger, C.M.; Sorenson, B.; Whaley, P. Stability Assessment of 10 Active Pharmaceutical Ingredients Compounded in SyrSpend SF. Int. J. Pharm. Compd. 2015, 19, 420–427. [Google Scholar]
  67. Helin-Tanninen, M.; Naaranlahti, T.; Kontra, K.; Ojanen, T. Enteral suspension of nifedipine for neonates. Part 2. Stability of an extemporaneously compounded nifedipine suspension. J. Clin. Pharm. Ther. 2001, 26, 59–66. [Google Scholar] [CrossRef] [PubMed]
  68. Shringirishi, M.; Mahor, A.; Gupta, R.; Prajapati, S.K.; Bansal, K.; Kesharwani, P. Fabrication and characterization of nifedipine loaded β-cyclodextrin nanosponges: An in vitro and in vivo evaluation. J. Drug Deliv. Sci. Technol. 2017, 41, 344–350. [Google Scholar] [CrossRef]
  69. Naaranlahti, T.; Kontra, K.; Wallenius, K.; Helin-Tanninen, M. Enteral suspension of nifedipine for neonates. Part 1. Formulation of nifedipine suspension for hospital use. J. Clin. Pharm. Ther. 2001, 26, 49–57. [Google Scholar] [CrossRef]
  70. American Society of Health-system Pharmacists. Standardize 4 Safety Initiative. 2020. Available online: https://www.ashp.org/-/media/assets/pharmacy-practice/s4s/docs/Compound-Oral-Liquid.pdf (accessed on 23 February 2022).
  71. United States Department of Health and Human Services. Centers for Disease Control and Prevention. National Institute for Occupational Safety and Health. NIOSH List of Antineoplastic and Other Hazardous Drugs in Healthcare Settings, 2016. Publication no. 2016-161. Available online: https://www.cdc.gov/niosh/docs/2016-161/pdfs/2016-161.pdf?id=10.26616/NIOSHPUB2016161 (accessed on 23 February 2022).
  72. Department of Health and Human Services. National Institutes of Health. Best Pharmaceuticals for Children Act (BPCA) Priority List of Needs in Pediatric Therapeutics 2017–2018. Available online: https://www.nichd.nih.gov/sites/default/files/inline-files/priority_list_063017_0.pdf (accessed on 23 February 2022).
  73. United States Food and Drug Administration. Drug and Biologic Essential Medicines, Medical Countermeasures, and Critical Inputs for the List Described in Section 3(c) of the Executive Order 13944. 2020. Available online: https://www.fda.gov/media/143406/download (accessed on 23 February 2022).
  74. World Health Organization. Model List of Essential Medicines in Children. Available online: https://www.who.int/selection_medicines/committees/expert/20/EMLc_2015_FINAL_amended_JUN2015.pdf (accessed on 23 February 2022).
  75. Bhatt-Mehta, V.; Hammoud, H.; Amidon, G.L. A proposed pediatric biopharmaceutical classification system for medications for chronic diseases in children. Eur. J. Pharm. Sci. 2020, 152, 105437. [Google Scholar] [CrossRef] [PubMed]
  76. Dahan, A.; Miller, J.M. The solubility-permeability interplay and its implications in formulation design and development for poorly soluble drugs. AAPS J. 2012, 14, 244–251. [Google Scholar] [CrossRef] [Green Version]
  77. Ursan, I.; Chiu, L.; Pierce, A. Three-dimensional drug printing: A structured review. J. Am. Pharm. Assoc. 2013, 53, 136–144. [Google Scholar] [CrossRef]
  78. Beg, S.; Almalki, W.H.; Malik, A.; Farhan, M.; Aatif, M.; Rahman, Z.; Alruwaili, N.K.; Alrobaian, M.; Tarique, M.; Rahman, M. 3D printing for drug delivery and biomedical applications. Drug Discov. Today 2020, 25, 1668–1681. [Google Scholar] [CrossRef]
  79. Wadher, K.; Trivedi, R.; Wankhede, N.; Kale, M.; Umekar, M. 3D printing in pharmaceuticals: An emerging technology full of challenges. Ann. Pharm. Fr. 2021, 79, 107–118. [Google Scholar] [CrossRef]
  80. Changdeo, J.S.; Vinod, M.; Shankar, K.B.; Rajaram, C.A. Physicochemical characterization and solubility enhancement studies of allopurinol solid dispersions. Braz. J. Pharm. Sci. 2011, 47, 3. [Google Scholar] [CrossRef] [Green Version]
  81. Hecq, J.; Deleers, M.; Fanara, D.; Vranckx, H.; Amighi, K. Preparation and characterization of nanocrystals for solubility and dissolution rate enhancement of nifedipine. Int. J. Pharm. 2005, 299, 167–177. [Google Scholar] [CrossRef]
  82. Jansook, P.; Ogawa, N.; Loftsson, T. Cyclodextrins: Structure, physicochemical properties and pharmaceutical applications. Int. J. Pharm. 2018, 535, 272–284. [Google Scholar] [CrossRef] [PubMed]
  83. Nehm, S.J.; Rodríguez-Spong, B.; Rodríguez-Hornedo, N. Phase solubility diagrams of cocrystals are explained by solubility product and solution complexation. Cryst. Growth Des. 2006, 6, 592–600. [Google Scholar] [CrossRef]
  84. Nikghalb, L.A.; Singh, G.; Singh, G.; Kahkeshan, K.F. Solid Dispersion: Methods and Polymers to increase the solubility of poorly soluble drugs. J. Appl. Pharm. Sci. 2012, 2, 170–175. [Google Scholar] [CrossRef]
  85. Bali, D.E.; Osman, M.A.; El Maghraby, G.M. Enhancement of Dissolution Rate and Intestinal Stability of Clopidogrel Hydrogen Sulfate. Eur. J. Drug Metab. Pharmacokinet. 2016, 41, 807–818. [Google Scholar] [CrossRef] [PubMed]
  86. Palekar, S.; Nukala, P.K.; Mishra, S.M.; Kipping, T.; Patel, K. Application of 3D printing technology and quality by design approach for development of age-appropriate pediatric formulation of baclofen. Int. J. Pharm. 2019, 556, 106–116. [Google Scholar] [CrossRef]
  87. Bromberg, L. Polymeric micelles in oral chemotherapy. J. Control Release 2008, 128, 99–112. [Google Scholar] [CrossRef]
  88. Hosny, K.M.; Alhakamy, N.A.; Almodhwahi, M.A.; Kurakula, M.; Almehmady, A.M.; Elgebaly, S.S. Self-Nanoemulsifying System Loaded with Sildenafil Citrate and Incorporated within Oral Lyophilized Flash Tablets: Preparation, Optimization, and In Vivo Evaluation. Pharmaceutics 2020, 12, 1124. [Google Scholar] [CrossRef]
  89. Ma, X.H.; Yao, Z.K.; Yang, Z.; Guo, H.; Xu, L.; Tang, C.Y.; Elimelech, M. Nanofoaming of Polyamide Desalination Membranes to Tune Permeability and Selectivity. Environ. Sci. Technol. Lett. 2018, 5, 123–130. [Google Scholar] [CrossRef]
  90. Nakano, Y.; Tajima, M.; Sugiyama, E.; Sato, V.; Sato, H. Development of a Novel Nano-emulsion Formulation to Improve Intestinal Absorption of Cannabidiol. Med. Cannabis Cannabinoids 2019, 2, 35–42. [Google Scholar] [CrossRef]
  91. Wiczling, P.; Liem, R.I.; Panepinto, J.A.; Garg, U.; Abdel-Rahman, S.M.; Kearns, G.L.; Neville, K.A. Population pharmacokinetics of hydroxyurea for children and adolescents with sickle cell disease. J. Clin. Pharmacol. 2014, 54, 1016–1022. [Google Scholar] [CrossRef]
  92. Estepp, J.H.; Melloni, C.; Thornburg, C.D.; Wiczling, P.; Rogers, Z.; Rothman, J.A.; Green, N.S.; Liem, R.; Brandow, A.M.; Crary, S.E.; et al. Pharmacokinetics and bioequivalence of a liquid formulation of hydroxyurea in children with sickle cell anemia. J. Clin. Pharmacol. 2016, 56, 298–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Jang, D.H.; Hoffman, R.S.; Nelson, L.S. A case of near-fatal flecainide medication error in a neonate. Clin. Toxicol. 2010, 44, 781–783. [Google Scholar] [CrossRef] [Green Version]
  94. Santoveña, A.; Charola, I.; Suárez-González, J.; Teigell-Pérez, N.; García-van Nood, S.; Soriano, M.F.J. Development of a novel physico-chemically and microbiologically stable oral solution of flecainide for pediatrics. Pharm. Dev. Technol. 2018, 23, 978–985. [Google Scholar] [CrossRef] [PubMed]
  95. Bomback, A.S.; Fervenza, F.C. Membranous Nephropathy: Approaches to Treatment. Am. J. Nephrol. 2018, 47 (Suppl. S1), 30–42. [Google Scholar] [CrossRef] [PubMed]
  96. Silvariño, R.; Ferreiro, A.; González-Martínez, F.; Gadola, L.; Noboa, O.; Caorsi, H. Substitution of Oral for Intravenous Cyclophosphamide in Membranous Nephropathy. Kidney360 2020, 1, 943–949. [Google Scholar] [CrossRef]
  97. Kennedy, R.; Groepper, D.; Tagen, M.; Christensen, R.; Navid, F.; Gajjar, A.; Stewart, C.F. Stability of cyclophosphamide in extemporaneous oral suspensions. Ann. Pharmacother. 2010, 44, 295–301. [Google Scholar] [CrossRef] [Green Version]
  98. MacDonald, J.L.; Johnson, C.E.; Jacobson, P. Stability of isradipine in an extemporaneously compounded oral liquid. Am. J. Hosp. Pharm. 1994, 51, 2409–2411. [Google Scholar] [CrossRef]
  99. Masi, G.; Milone, A.; Veltri, S.; Iuliano, R.; Pfanner, C.; Pisano, S. Use of quetiapine in children and adolescents. Paediatr. Drugs 2015, 17, 125–140. [Google Scholar] [CrossRef]
  100. Papazisis, G.; Siafis, S. The Added Value of Liquid Antipsychotics: The Case of Quetiapine. Curr. Clin. Pharmacol. 2019, 14, 101–107. [Google Scholar] [CrossRef]
  101. Agarwal, S.; HariKumar, S.L.; Negi, P.; Upadhyay, N.; Garg, R. Quetiapine Fumarate Loaded Nanostructured Lipid Carrier for Enhancing Oral Bioavailability: Design, Development and Pharmacokinetic Assessment. Curr. Drug Deliv. 2021, 18, 184–198. [Google Scholar] [CrossRef]
  102. Foley, L.; Toney, J.; Barlow, J.W.; O’Connor, M.; Fitzgerald-Hughes, D.; Ramtoola, Z. Investigation of the Physical, Chemical and Microbiological Stability of Losartan Potassium 5 mg/mL Extemporaneous Oral Liquid Suspension. Molecules 2021, 26, 301. [Google Scholar] [CrossRef] [PubMed]
  103. United States Food and Drug Administration. Cozaar Package Insert. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/020386s058lbl.pdf (accessed on 28 April 2022).
  104. Kraft, W.K.; Van den Anker, J.N. Pharmacologic Management of the Opioid Neonatal Abstinence Syndrome. Pediatr. Clin. N. Am. 2012, 59, 1147–1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Disher, T.; Gullickson, C.; Singh, B.; Cameron, C.; Boulos, L.; Beaubien, L.; Campbell-Yeo, M. Pharmacological Treatments for Neonatal Abstinence Syndrome: A Systematic Review and Network Meta-analysis. JAMA Pediatr. 2019, 173, 234–243. [Google Scholar] [CrossRef] [PubMed]
  106. Frazier, L.M.; Bobby, L.E.; Gawronski, K.M. Emerging therapies for the treatment of neonatal abstinence syndrome. J. Matern. Neonatal Med. 2020, 35, 987–995. [Google Scholar] [CrossRef] [PubMed]
  107. van Hoogdalem, M.W.; McPhail, B.T.; Hahn, D.; Wexelblatt, S.L.; Akinbi, H.T.; Vinks, A.A.; Mizuno, T. Pharmacotherapy of neonatal opioid withdrawal syndrome: A review of pharmacokinetics and pharmacodynamics. Expert Opin. Drug Metab. Toxicol. 2021, 17, 87–103. [Google Scholar] [CrossRef] [PubMed]
  108. Sutter, M.B.; Leeman, L.; His, A. Neonatal opioid withdrawal syndrome. Obstet. Gynecol. Clin. N. Am. 2014, 41, 317–334. [Google Scholar] [CrossRef]
  109. Esch, J.J.; Hellinger, A.; Friedman, K.G.; VanderPluym, C.J. Apixaban for treatment of intracardiac thrombosis in children with congenital heart disease. Interact. Cardiovasc. Thorac. Surg. 2020, 30, 950–951. [Google Scholar] [CrossRef]
  110. Caraballo, M.L.; Donmez, S.; Nathan, K.; Zhao, F. Compounded apixaban suspensions for enteral feeding tubes. Hosp. Pharm. 2017, 52, 478–482. [Google Scholar] [CrossRef]
  111. Wheeler, J.; Francis, P.; Kim, J. Atenolol compounding and atrioventricular block: A case report. J. Investig. Med. 2012, 60, 172. [Google Scholar] [CrossRef]
  112. Franck, B.; Autmizguine, J.; Marquet, P.; Ovetchkine, P.; Woillard, J.B. Pharmacokinetics, Pharmacodynamics, and Therapeutic Drug Monitoring of Valganciclovir and Ganciclovir in Transplantation. Clin. Pharmacol. Ther. 2021. [Google Scholar] [CrossRef]
  113. Jorga, K.; Reigner, B.; Chavanne, C.; Alvaro, G.; Frey, N. Pediatric Dosing of Ganciclovir and Valganciclovir: How Model-Based Simulations Can Prevent Underexposure and Potential Treatment Failure. CPT Pharmacomet. Syst. Pharmacol. 2019, 8, 167–176. [Google Scholar] [CrossRef] [PubMed]
  114. Pescovitz, M.D.; Brook, B.; Jindal, R.M.; Leapman, S.B.; Milgrom, M.L.; Filo, R.S. Oral ganciclovir in pediatric transplant recipients: A pharmacokinetic study. Clin. Transplant. 1997, 11, 613–617. [Google Scholar] [PubMed]
  115. Tan, C.; Sehgal, K.; Sehgal, K.; Krishnappa, S.B.; Sehgal, A. Diuretic use in infants with developing or established chronic lung disease: A practice looking for evidence. Paediatr. Child Health 2020, 56, 1189–1193. [Google Scholar] [CrossRef] [PubMed]
  116. Stoltenberg, I.; Breitkreutz, J. Orally disintegrating mini-tablets (ODMTs)—A novel solid oral dosage form for paediatric use. Eur. J. Pharm. Biopharm. 2011, 78, 462–469. [Google Scholar] [CrossRef] [PubMed]
  117. Uriel, M.; Gomez-Rincon, C.; Marro, D. Stability of regularly prescribed oral liquids formulated with SyrSpend® SF. Pharmazie 2018, 73, 196–201. [Google Scholar] [CrossRef]
  118. Cirri, M.; Maestrini, L.; Maestrelli, F.; Mennini, N.; Mura, P.; Ghelardini, C.; Di Cesare Mannelli, L. Design, characterization and in vivo evaluation of nanostructured lipid carriers (NLC) as a new drug delivery system for hydrochlorothiazide oral administration in pediatric therapy. Drug Deliv. 2018, 25, 1910–1921. [Google Scholar] [CrossRef]
  119. Filler, G. Calcineurin inhibitors in pediatric renal transplant recipients. Paediatr. Drugs 2007, 9, 165–174. [Google Scholar] [CrossRef]
  120. Amin, A.; Bourget, P.; Vidal, F.; Cartier, F.; Beauvais, R.; Afonso Vdo, N. Physicochemical and microbiological stability of azathioprine in InOrpha suspending agent studied under various conditions. Int. J. Pharm. Compd. 2015, 19, 73–77. [Google Scholar]
  121. Stumpf, J.L.; Leja, N.; Ciarkowski, S.L.; Schaeffler, K.L.; Salah, S. Updating formulations for compounded oral liquid medications in a university health system. Am. J. Health Syst. Pharm. 2018, 75, 1394–1398. [Google Scholar] [CrossRef]
  122. United States Pharmacopeia. Compounded Monographs Failed Studies. 2020. Available online: https://www.usp.org/sites/default/files/usp/document/get-involved/partner/usp-compounded-preparation-monographs-failed-studies.pdf (accessed on 3 March 2022).
  123. Riley, M.F. An Unfulfilled Promise: Changes Needed to the Drug Approval Process to Make Personalized Medicine a Reality. Food Drug Law J. 2015, 70, 289–314. [Google Scholar]
  124. Singer, D.R.; Zaïr, Z.M. Clinical Perspectives on Targeting Therapies for Personalized Medicine. Adv. Protein Chem. Struct. Biol. 2016, 102, 79–114. [Google Scholar] [CrossRef] [PubMed]
  125. Gore, L.; Kearns, P.R.; de Martino, M.L.; Lee De Souza, C.A.; Bertrand, Y.; Hijiya, N.; Stork, L.C.; Chung, N.G.; Cardos, R.C.; Saikia, T.; et al. Dasatinib in Pediatric Patients with Chronic Myeloid Leukemia in Chronic Phase: Results from a Phase II Trial. J. Clin. Oncol. 2018, 36, 1330–1338. [Google Scholar] [CrossRef] [PubMed]
  126. ClinicalTrials.gov: Pharmacokinetic Study Comparing Blood Levels of Dasatinib in Healthy Participants Who Received the Tablet Formulation with Those Who Received Liquid and Tablet-Dispersed Formulations. Available online: https://clinicaltrials.gov/ct2/show/NCT01392703 (accessed on 28 April 2022).
  127. Srycel (Dasatinib Tablets for Oral Use) Package Insert. Bristol Myers Squibb. Available online: https://packageinserts.bms.com/pi/pi_sprycel.pdf (accessed on 28 April 2022).
  128. Hinterlang, M.; Gendron, A.; Fleury, T.; Rieutord, A.; Vrana, A.; Schlatter, J.; Annereau, M. Design and stability of pediatric oral formulation of imatinib. J. Oncol. Pharm. Pract. 2022, 28, 337–342. [Google Scholar] [CrossRef] [PubMed]
  129. Carofiglio, F.; Lopalco, A.; Lopedota, A.; Cutrignelli, A.; Nicolotti, O.; Denora, N.; Stefanachi, A.; Leonetti, F. Bcr-Abl Tyrosine Kinase Inhibitors in the Treatment of Pediatric CML. Int. J. Mol. Sci. 2020, 21, 4469. [Google Scholar] [CrossRef] [PubMed]
  130. Clinical Trials.gov. A Study of the Safety and Preliminary Efficacy of Oral Midostaurin (PKC412) in Relapsed or Refractory Pediatric Leukemia. Available online: https://clinicaltrials.gov/ct2/show/NCT00866281 (accessed on 28 April 2022).
  131. Fouladi, M.; Park, J.R.; Stewart, C.F.; Gilbertson, R.J.; Schaiquevich, P.; Sun, J.; Reid, J.M.; Ames, M.M.; Speights, R.; Ingle, A.M.; et al. Pediatric phase I trial and pharmacokinetic study of vorinostat: A Children’s Oncology Group phase I consortium report. J. Clin. Oncol. 2010, 28, 3623–3629. [Google Scholar] [CrossRef] [Green Version]
  132. Hong, D.S.; DuBois, S.G.; Kummar, S.; Farago, A.F.; Albert, C.M.; Rohrberg, K.S.; van Tilburg, C.M.; Nagasubramanian, R.; Berlin, J.D.; Federman, N.; et al. Larotrectinib in patients with TRK fusion-positive solid tumours: A pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020, 21, 531–540. [Google Scholar] [CrossRef]
  133. Balis, F.M.; Thompson, P.A.; Mosse, Y.P.; Blaney, S.M.; Minard, C.G.; Weigel, B.J.; Fox, E. First-dose and steady-state pharmacokinetics of orally administered crizotinib in children with solid tumors: A report on ADVL0912 from the Children’s Oncology Group Phase 1/Pilot Consortium. Cancer Chemother. Pharmacol. 2017, 79, 181–187. [Google Scholar] [CrossRef] [Green Version]
  134. ClinicalTrials.gov: A Phase I Study of Pazopanib as a Single Agent for Children with Relapsed or Refractory Solid Tumors, Including CNS Tumors. Available online: https://clinicaltrials.gov/ct2/show/NCT01130623 (accessed on 3 May 2022).
  135. Cohen-Rabbie, S.; Mattinson, A.; So, K.; Wang, N.; Goldwater, R. Effect of food on capsule and granule formulations of selumetinib. Clin. Transl. Sci. 2022, 15, 878–888. [Google Scholar] [CrossRef]
  136. Cox, D.S.; Allred, A.; Zhou, Y.; Infante, J.R.; Gordon, M.S.; Bendell, J.; Jones, S.; Burris, H., 3rd; Orford, K. Relative bioavailability of pediatric oral solution and tablet formulations of trametinib in adult patients with solid tumors. Clin. Pharmacol. Drug Dev. 2015, 4, 287–294. [Google Scholar] [CrossRef]
  137. Mamdouhi, T.; Vagrecha, A.; Johnson, A.A.; Levy, C.F.; Atlas, M.; Krystal, J.I. Successful use of crushed formulation of dabrafenib and trametinib in a pediatric glioneural tumor. Pediatr. Blood Cancer 2021, 68, e29187. [Google Scholar] [CrossRef]
  138. United States Pharmacopeia. USP Compounded Preparation Monograph Donation Program. Available online: https://www.usp.org/get-involved/partner/compounding-monographs (accessed on 23 February 2022).
Table 1. FDA-approved commercially available finished liquid dosage forms from the 2016 AA list.
Table 1. FDA-approved commercially available finished liquid dosage forms from the 2016 AA list.
Active Pharmaceutical IngredientBrand NameStrength and Dosage FormNDA#Company and Year of Approval
lisinoprilQbrelis™1 mg/mL oral solution208401Azurity, Woburn, MA, USA—2016
spironolactoneCarospir™5 mg/mL oral suspension209478CMP Pharma, Farmville, NC, USA—2017
metoprolol succinateKapspargo™25, 50, 100, and 200 mg extended release capsules210428Sun Pharma, Mumbai, India—2018
amlodipine benzoateKaterzia™1.3 mg/mL oral suspension (=1 mg amlodipine)211340Azurity—2019
baclofenOzobax™1 mg/mL oral solution208193Metacel, Athens, GA, USA—2019
sildenafilRevatio™ and generics10 mg/mL powder for oral suspension203109Pfizer, Brooklyn, NY, USA, Cipla USA, Warren, NJ, USA and Novadoz, Piscataway, NJ, USA—2017–2019
levothyroxine sodiumTirosint-SOL™13, 25, 50, 75, 88, 100, 112, 125, 137, 150, 175, and 200 mcg/mL unit dose oral solution206977IBSA Pharma, Parsippany, NJ, USA—2019
amlodipine besylateNorliqva™1.385 mg/mL oral solution (=1 mg/mL amodipine)214439CMP Pharma—2022
Table 2. Suitability of recently approved and marketed manufactured liquids for children.
Table 2. Suitability of recently approved and marketed manufactured liquids for children.
ProductExcipients Listed in the Package InsertRationale for Caution
Kapspargo™ (metoprolol succinate) sprinkles 25, 50, 100, 200 mg capsulesethyl cellulose; hypromellose (hydroxypropyl methylcellulose); polyethylene glycol 400; polyethylene glycol 6000; sugar spheres (corn starch and sucrose); talc; triethyl citrateFixed dose may not be suitable for neonates
Qbrelis™ (lisinopril) 1 mg/mL oral liquidwater; xylitol; sodium citrate; citric acid; sodium benzoate; hydrochloric acid; sodium hydroxideContains sodium benzoate
Katerzia™ (amlodipine benzoate) 1 mg/mL oral suspensioncitric acid monohydrate; silicone dioxide; hypromellose; maltodextrin; polysorbate 80; sodium benzoate; sodium citrate; sodium hydroxide; sucralose; waterContains sodium benzoate and sucralose
Carospir™ (spironolactone) 5 mg/mL oral suspensionxanthan gum; dimethicone; sorbic acid; potassium sorbate; saccharin sodium; anhydrous citric acid; trisodium citrate dihydrate; ammonium glycyrrhizate (licorice); glycerin; waterContains ammonium glycyrrhizate and saccharin
Ozobax™ (baclofen) 1 mg/mL oral solutionanhydrous citric acid; glycerin; methylparaben; propylparaben; trisodium citrate dihydrate; sucralose; waterContains methyl- and propylparaben and sucralose
Revatio™ (sildenafil citrate) 10 mg/mL powder for oral liquid suspensionmicronized cellulose; anhydrous dibasic calcium phosphate; croscarmellose sodium; magnesium stearate; hypromellose; titanium dioxide; lactose monohydrate; triacetin Contains lactose; not labeled for children
Tirosint-SOL™—unit dose oral solution (levothyroxine sodium)—12 strengths between 13 and 200 mcg/mLglycerin; waterMany endocrinologists prefer crushing and dissolving tablets; multiple strengths may lead to medication errors
Table 3. FDA-approved and marketed in finished oral liquid or granule dosage forms—2014 to present.
Table 3. FDA-approved and marketed in finished oral liquid or granule dosage forms—2014 to present.
Active Pharmaceutical IngredientBrand NameNDA#US-FDA Approval YearLabeled Pediatric Indication
(<12 Years of Age)
glycopyrrolateCuvposa0225712018Yes
cannabidiolEpidiolex2103652018Yes
vancomycinFirvanq2099102018Yes
mercaptopurinePurixan2059192014Yes
aprepitantEmend2078652015Yes
rivaroxabanXarelto2024392021Yes
methotrexateXatmep2084002017Yes
hydrocortisoneAlkindi2138762020Yes
deflazacortEmflaza2086852017Yes
dronabinolSyndros2055252016Yes
fenfluramineFintepla2121022020Yes
tacrolimus Prograf2101152019Yes
tofacitinibXeljanz2130822020Yes
tramadolQdolo2140442020No
stiripentolDiacomit2072232018Yes
colchicineGloperba2109422019No
celecoxibElyxyb2121572020No
sodium zirconium cyclosilicateLokelma2070782018No
triheptanoinDojolvi2136872020Yes
topiramate Eprontia2146792021Yes
Table 5. APIs with the highest potential for development as commercially available FDA-approved finished liquid dosage forms by BCS class.
Table 5. APIs with the highest potential for development as commercially available FDA-approved finished liquid dosage forms by BCS class.
Active Pharmaceutical IngredientBiopharmaceutical Classification (BCS)Potential Pediatric Indication
clonidineIIIADHD, neonatal abstinence syndrome, hypertension
hydroxyureaISickle cell anemia
hydrochlorothiazideIVHypertension, nephrolithiasis, diabetes insipidus
amiodaroneIISupraventricular tachycardia (SVT), atrial flutter
carvedilolIIHeart failure, dilated cardiomyopathy
hydralazineIIIHypertension
isradipineIIHypertension
allopurinolITumor lysis syndrome, urate nephropathy, hyperuricemia
flecainideISVT, atrial fibrillation
quetiapineIIPsychosis, delirium
apixabanIIIVenous thromboembolism
atenololIIIHypertension
cyclophosphamideISarcoma, brain tumor, vasculitis, systemic lupus erythematosus, nephrotic syndrome, juvenile rheumatoid arthritis
losartanIIHypertension
ganciclovirIIICytomegalovirus prevention and treatment
azathioprineIVSolid organ transplantation, inflammatory bowel disease, juvenile idiopathic arthritis, lupus nephritis, autoimmune hepatitis
Table 6. Selected pediatric cancers treated with targeted therapies and availability of oral liquids.
Table 6. Selected pediatric cancers treated with targeted therapies and availability of oral liquids.
Active Pharmaceutical IngredientConcentration (mg/mL) StudiedIndicationOral Liquid or Powder for Reconstitution Marketed (M) or Compounded (C)?
dasatinib [127]10Chronic myeloid leukemia (CML)No
imatinib mesylate [128]30CMLC
nilotinib [129]n.a.CMLNo
midostaurin [130]25Acute myeloid leukemia (AML)No
vorinostat [131]50Cutaneous T-cell lymphomaC
larotrectinib [132]20Tropomyosin receptor kinase-positive solid tumorsM
crizotinib [133]25ALK+ anaplastic large cell lymphoma (ALCL)No
pazopanib [134]50Solid tumors in children (sarcoma, neuroblastoma, Wilms, osteosarcoma, and brain)No
selumetinib [135]granuleNeurofibromatosis type 1No (in development)
trametinib [136]0.05Unresectable or metastatic melanomaNo
dabrafenib [137]n.a.Unresectable or metastatic melanomaNo
Note: n.a. = not available.
Table 7. APIs with potential for development as manufactured products or compounded preparations.
Table 7. APIs with potential for development as manufactured products or compounded preparations.
USP CC-Listed APIsNon-USP CC-Listed APIs
with Mass Market Potentialwithout Mass Market Potentialwith Mass Market Potentialwithout Mass Market Potential
desmopressinpyrazinamidezincamitriptyline
phytonadionedapsonebuprenorphinehydroxychloroquine
pyridoxinediltiazemnaltrexonethiamine
rifampinketoconazoleeverolimusrifaxamin
ethambutolmetolazone valsartan
clonazepampyrimethamine venlafaxine
rifabutin buspirone
bethanechol dantrolene
propylthiouracil mexiletine
dipyridamole nadolol
chloroquine pravastatin
quinidine topotecan
temozolomide tretinoin
terbutaline chlorpromazine
tetracycline ethacrynic acid
tiagabine flucytosine
dolasetron amiloride
phenoxybenzamine primaquine
procarbazine
disopyramide
USP—United States Pharmacopeia; CC—compounding compendium; API—active pharmaceutical ingredient.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Parrish, R.H., II; Ashworth, L.D.; Löbenberg, R.; Benavides, S.; Cies, J.J.; MacArthur, R.B. Compounded Nonsterile Preparations and FDA-Approved Commercially Available Liquid Products for Children: A North American Update. Pharmaceutics 2022, 14, 1032. https://doi.org/10.3390/pharmaceutics14051032

AMA Style

Parrish RH II, Ashworth LD, Löbenberg R, Benavides S, Cies JJ, MacArthur RB. Compounded Nonsterile Preparations and FDA-Approved Commercially Available Liquid Products for Children: A North American Update. Pharmaceutics. 2022; 14(5):1032. https://doi.org/10.3390/pharmaceutics14051032

Chicago/Turabian Style

Parrish, Richard H., II, Lisa D. Ashworth, Raimar Löbenberg, Sandra Benavides, Jeffrey J. Cies, and Robert B. MacArthur. 2022. "Compounded Nonsterile Preparations and FDA-Approved Commercially Available Liquid Products for Children: A North American Update" Pharmaceutics 14, no. 5: 1032. https://doi.org/10.3390/pharmaceutics14051032

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop