1
|
Yu D, Li M, Hoag SW, Nie H. Understanding excipient-induced crystallization of spray-dried amorphous solid dispersion. J Pharm Sci 2025; 114:234-244. [PMID: 39306037 DOI: 10.1016/j.xphs.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024]
Abstract
This study investigates the compatibility of excipients with the model system SDI-X and their role in the induced crystallization of the amorphous compound-X in tablet formulations. We aimed to establish a straightforward and practical screening approach for evaluating excipient-induced crystallization of SDI in tablet matrices. Three methodologies-binary powder mixture, binary compact, and bilayer tablets-were employed to qualitatively and quantitatively evaluate the recrystallization of SDI-X with various excipients under accelerated storage conditions. The results demonstrated that binary compacts, providing direct physical contact between SDI-X and excipients, are superior in reflecting realistic drug-excipient contact within pharmaceutical tablets, enabling a more accurate assessment of excipient-induced crystallization for SDI-X. In contrast, the broadly used conventional binary blends can significantly underestimate this risk due to insufficient proximity. In addition, the bilayer tablets further confirmed that crystallization initiates at the contact surface between SDI-X and the excipients. The study highlighted that not only hygroscopicity but also the type of excipient and its physical contact with SDI-X significantly influence the recrystallization extent and rate of SDI-X. Interestingly, less hygroscopic diluents such as mannitol and lactose induced much higher levels of crystallization of SDIs, contrary to expectations based on moisture content alone. This suggests that the excipient type and contact surface are more critical in inducing recrystallization than just the level of moisture. The findings emphasize the need for careful excipient selection, study design, and sample preparation to enable appropriate assessments of SDI-excipient compatibility.
Collapse
Affiliation(s)
- Dongyue Yu
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, United States; Center for Materials Science and Engineering, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Meng Li
- Center for Materials Science and Engineering, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Stephen W Hoag
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, United States
| | - Haichen Nie
- Center for Materials Science and Engineering, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States.
| |
Collapse
|
2
|
Autzen Virtanen A, Myślińska M, Healy AM, Power E, Madi A, Sivén M. The challenge of downstream processing of spray dried amorphous solid dispersions into minitablets designed for the paediatric population - A sustainable product development approach. Eur J Pharm Sci 2024; 196:106752. [PMID: 38518998 DOI: 10.1016/j.ejps.2024.106752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Poorly water-soluble drugs present a significant challenge in the development of oral solid dosage forms (OSDs). In formulation development the appropriate use of excipients to adjust solubility, and the choice of manufacturing method and pharmaceutical processes to obtain a dosage form to meet the needs of the patient group, is crucial. Preparing an amorphous solid dispersion (ASD) is a well-established method for solubility enhancement, and spray drying (SD) a common manufacturing method. However, the poor flowability of spray dried materials poses a significant challenge for downstream processing. Promoting sustainability in OSD development involves embracing a versatile formulation design, which enables a broader spectrum of patients to use the product, as opposed to altering existing dosage forms retrospectively. The objective of the current study was to develop a formulation of spray dried indomethacin ASD suited to the production, by direct compression, of instant release paediatric minitablets. Excipients evaluated were PVP or HPMCAS in solid dispersions at the preformulation phase, and MCC and lactose as a filler in direct compression. From the studied formulations, a 3:1 ratio blend of Vivapur 200/Pharmatose 200 M (MCC/lactose) with 0.5% (w/w) magnesium stearate was found to be the most promising in tableting, and minitablets containing a 6.22% content of spray-dried ASD of indomethacin/PVP K 29-32 could be obtained with desired tablet hardness and pharmaceutical quality, complying with tests of weight variation and fast disintegration in an aqueous environment. As a case example, this study provides a good foundation for further studies in harnessing a sustainable approach to the development of pharmaceutical formulations that can appropriately serve different patient sub-populations.
Collapse
Affiliation(s)
- Anja Autzen Virtanen
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland.
| | - Monika Myślińska
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - Anne Marie Healy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - Eoin Power
- SK biotek Ireland, an SK pharmteco company, Ireland
| | - Atif Madi
- SK biotek Ireland, an SK pharmteco company, Ireland
| | - Mia Sivén
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland; Helsinki Institute of Sustainability Science, HELSUS, Finland
| |
Collapse
|
3
|
Vaishampayan UN, Keessen M, Dreicer R, Heath EI, Buchler T, Árkosy PF, Csöszi T, Wiechno P, Kopyltsov E, Orlov SV, Plekhanov A, Smagina M, Varlamov S, Shore ND. A global phase II randomized trial comparing oral taxane ModraDoc006/r to intravenous docetaxel in metastatic castration resistant prostate cancer. Eur J Cancer 2024; 202:114007. [PMID: 38518534 DOI: 10.1016/j.ejca.2024.114007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/24/2024]
Abstract
STUDY AIM ModraDoc006, an oral formulation of docetaxel, is co-administered with the cytochrome P450-3A4 and P-glycoprotein inhibitor, ritonavir (r): ModraDoc006/r. The preliminary efficacy and safety of oral ModraDoc006/r was evaluated in a global randomized phase II trial and compared to the current standard chemotherapy regimen of intravenous (i.v.) docetaxel and prednisone. METHODS 103 mCRPC patients, chemotherapy-naïve with/without abiraterone and/or enzalutamide pretreated, with adequate organ function and evaluable disease per RECIST v1.1 and PCWG3 guidelines were randomized 1:1 into two cohorts. In Cohort 1, 49 patients received docetaxel 75 mg/m2 i.v. every 3 weeks (Q3W). In Cohort 2, 52 patients received ModraDoc006/r; 21 patients with a starting dose of ModraDoc006 30 mg with ritonavir 200 mg in the morning and ModraDoc006 20 mg with ritonavir 100 mg in the evening (30-20/200-100 mg) bi-daily-once-weekly (BIDW) on Days 1, 8, and 15 of a 21-day cycle. To alleviate tolerability, the starting dose was amended to ModraDoc006/r 20-20/200-100 mg in another 31 patients. All patients received prednisone 10 mg daily. Primary endpoint was rPFS. RESULTS There was no significant difference in rPFS between the 2 arms (p = 0.1465). Median rPFS was 9.5 months and 11.1 months (95% CI) for ModraDoc006/r and i.v. docetaxel, respectively. Partial response was noted in 44.1% and 38.7% measurable disease patients, and 50% decline of PSA was seen in 23 (50%) and 26 (56.5%) evaluable cases treated with ModraDoc006/r and i.v. docetaxel, respectively. The safety profile of ModraDoc006/r 20-20/200-100 mg dose was significantly better than i.v. docetaxel, with mild (mostly Grade 1) gastrointestinal toxicities, no hematologic adverse events, and neuropathy and alopecia incidence of 11.5% and 25%, respectively. CONCLUSIONS ModraDoc006/r potentially represents a widely applicable, convenient, effective, and better tolerated oral taxane therapy option for mCRPC. Further investigation of ModraDoc006/r in a large randomized trial is warranted.
Collapse
Affiliation(s)
| | | | | | | | - Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czech Republic
| | | | | | - Pawel Wiechno
- Klinika Nowotworów Układu Moczowego Centrum Onkologii, Warsaw, Poland
| | | | - Sergey V Orlov
- Pavlov First St. Petersburg State Medical University, Saint Petersburg, Russian Federation
| | | | - Maria Smagina
- Leningrad Regional Oncology Dispensary, Saint Petersburg, Russian Federation
| | | | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| |
Collapse
|
4
|
Li J, Wang X, Yu D, Zhoujin Y, Wang K. Molecular complexes of drug combinations: A review of cocrystals, salts, coamorphous systems and amorphous solid dispersions. Int J Pharm 2023; 648:123555. [PMID: 37890646 DOI: 10.1016/j.ijpharm.2023.123555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
As the advancements in the medical technology and healthcare develop through the years, combinational therapy has evolved to be an important treatment modality in many disease settings, including cancer, cardiovascular disease and infectious diseases. In an effort to alleviate "pill burden" and improve patient compliance, fixed dose combinations (FDCs) have been developed to be used as effective therapeutics. Among all FDCs, the category of drug-drug molecular complexes has been proven an efficient methodology in designing and treating diseases, with many drugs being approved. Among all drug-drug molecular complexes, drug-drug cocrystals, salts, coamorphous systems and solid dispersions have been successfully developed and many have been approved by the FDA. In this review, we dwell deeply into the molecular mechanisms behind the different types of drug-drug molecular complexes, including the key functional groups involved in the intermolecular interactions, the applications of each category of molecular complexes, as well as the advantages and challenges thereof. This comprehensive review provides useful insights into the practical design and manufacture of drug-drug molecular complexes and points out the future direction for the development of new advantageous combinational therapies that benefit more patients.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Xiyan Wang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Dongyue Yu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, United States
| | - Yunping Zhoujin
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Kunlin Wang
- BeBetter Med Inc., Guangzhou, 510663, PR China; College of Pharmacy, Jinan University, Guangzhou, 510006, PR China.
| |
Collapse
|
5
|
Budiman A, Handini AL, Muslimah MN, Nurani NV, Laelasari E, Kurniawansyah IS, Aulifa DL. Amorphous Solid Dispersion as Drug Delivery Vehicles in Cancer. Polymers (Basel) 2023; 15:3380. [PMID: 37631436 PMCID: PMC10457821 DOI: 10.3390/polym15163380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer treatment has improved over the past decades, but a major challenge lies in drug formulation, specifically for oral administration. Most anticancer drugs have poor water solubility which can affect their bioavailability. This causes suboptimal pharmacokinetic performance, resulting in limited efficacy and safety when administered orally. As a result, it is essential to develop a strategy to modify the solubility of anticancer drugs in oral formulations to improve their efficacy and safety. A promising approach that can be implemented is amorphous solid dispersion (ASD) which can enhance the aqueous solubility and bioavailability of poorly water-soluble drugs. The addition of a polymer can cause stability in the formulations and maintain a high supersaturation in bulk medium. Therefore, this study aimed to summarize and elucidate the mechanisms and impact of an amorphous solid dispersion system on cancer therapy. To gather relevant information, a comprehensive search was conducted using keywords such as "anticancer drug" and "amorphous solid dispersion" in the PubMed, Scopus, and Google Scholar databases. The review provides an overview and discussion of the issues related to the ASD system used to improve the bioavailability of anticancer drugs based on molecular pharmaceutics. A thorough understanding of anticancer drugs in this system at a molecular level is imperative for the rational design of the products.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Annisa Luthfiyah Handini
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Mutia Nur Muslimah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Neng Vera Nurani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Eli Laelasari
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Insan Sunan Kurniawansyah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia;
| |
Collapse
|
6
|
Budiman A, Lailasari E, Nurani NV, Yunita EN, Anastasya G, Aulia RN, Lestari IN, Subra L, Aulifa DL. Ternary Solid Dispersions: A Review of the Preparation, Characterization, Mechanism of Drug Release, and Physical Stability. Pharmaceutics 2023; 15:2116. [PMID: 37631330 PMCID: PMC10459848 DOI: 10.3390/pharmaceutics15082116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
The prevalence of active pharmaceutical ingredients (APIs) with low water solubility has experienced a significant increase in recent years. These APIs present challenges in formulation, particularly for oral dosage forms, despite their considerable therapeutic potential. Therefore, the improvement of solubility has become a major concern for pharmaceutical enterprises to increase the bioavailability of APIs. A promising formulation approach that can effectively improve the dissolution profile and the bioavailability of poorly water-soluble drugs is the utilization of amorphous systems. Numerous formulation methods have been developed to enhance poorly water-soluble drugs through amorphization systems, including co-amorphous formulations, amorphous solid dispersions (ASDs), and the use of mesoporous silica as a carrier. Furthermore, the successful enhancement of certain drugs with poor aqueous solubility through amorphization has led to their incorporation into various commercially available preparations, such as ASDs, where the crystalline structure of APIs is transformed into an amorphous state within a hydrophilic matrix. A novel approach, known as ternary solid dispersions (TSDs), has emerged to address the solubility and bioavailability challenges associated with amorphous drugs. Meanwhile, the introduction of a third component in the ASD and co-amorphous systems has demonstrated the potential to improve performance in terms of solubility, physical stability, and processability. This comprehensive review discusses the preparation and characterization of poorly water-soluble drugs in ternary solid dispersions and their mechanisms of drug release and physical stability.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Eli Lailasari
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Neng Vera Nurani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Ellen Nathania Yunita
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Gracia Anastasya
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Rizqa Nurul Aulia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Ira Novianty Lestari
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (I.N.L.); (D.L.A.)
| | - Laila Subra
- Faculty of Bioeconomic and Health Sciences, Geomatika University College, Kuala Lumpur 54200, Malaysia;
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (I.N.L.); (D.L.A.)
| |
Collapse
|
7
|
Tong L, Zhou Z, Wang G, Wu C. A self-microemulsion enhances oral absorption of docetaxel by inhibiting P-glycoprotein and CYP metabolism. Drug Deliv Transl Res 2023; 13:983-993. [PMID: 36515864 DOI: 10.1007/s13346-022-01255-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2022] [Indexed: 12/15/2022]
Abstract
Oral absorption of docetaxel was limited by drug efflux pump p-glycoprotein (P-gp) and cytochrome P450 enzyme (CYP 450). Therefore, co-loading agent that inhibits P-gp and CYP450 in self-nanoemulsifying drug delivery systems (SMEs) is considered a promising strategy for oral delivery of docetaxel. In this study, curcumin was selected as an inhibitor of P-gp and CYP450, and it was co-encapsuled in SMEs to improve the oral bioavailability of docetaxel. SMEs quickly dispersed in water within 20 s, and the droplet size was 32.23 ± 2.21 nm. The release rate of curcumin from DC-SMEs was higher than that of docetaxel in vitro. Compared with free docetaxel, SMEs significantly increased the permeability of docetaxel by 4.6 times. And competitive experiments showed that the increased permeability was the result of inhibition of p-gp. The half-life and oral bioavailabilty of DC-SMEs increased about 1.7 times and 1.6 times than docetaxel SMEs, which indicated that its good pharmacokinetic behavior was related to the restriction of hepatic first-pass metabolism. In conclusion, DC-SME was an ideal platform to facilitate oral delivery of docetaxel through inhibited P-gp and CYP 450.
Collapse
Affiliation(s)
- Le Tong
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Zhuang Yao Medicine Center of Engineering and Technology, Guang Xi University of Chinese Medicine, Wuhe Road, Nanning, 530200, China
| | - ZeYang Zhou
- Zhuang Yao Medicine Center of Engineering and Technology, Guang Xi University of Chinese Medicine, Wuhe Road, Nanning, 530200, China
| | - Gang Wang
- Zhuang Yao Medicine Center of Engineering and Technology, Guang Xi University of Chinese Medicine, Wuhe Road, Nanning, 530200, China.
| | - Chao Wu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
8
|
van Eijk M, Yu H, Sawicki E, de Weger VA, Nuijen B, Dorlo TPC, Beijnen JH, Huitema ADR. Development of a population pharmacokinetic/pharmacodynamic model for various oral paclitaxel formulations co-administered with ritonavir and thrombospondin-1 based on data from early phase clinical studies. Cancer Chemother Pharmacol 2022; 90:71-82. [PMID: 35799067 PMCID: PMC9300539 DOI: 10.1007/s00280-022-04445-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/04/2022] [Indexed: 11/21/2022]
Abstract
Purpose Orally administered paclitaxel offers increased patient convenience while providing a method to prolong exposure without long continuous, or repeated, intravenous infusions. The oral bioavailability of paclitaxel is improved through co-administration with ritonavir and application of a suitable pharmaceutical formulation, which addresses the dissolution-limited absorption of paclitaxel. We aimed to characterize the pharmacokinetics of different paclitaxel formulations, co-administered with ritonavir, and to investigate a pharmacodynamic relationship between low-dose metronomic (LDM) treatment with oral paclitaxel and the anti-angiogenic marker thrombospondin-1 (TSP-1). Methods Fifty-eight patients treated with different oral paclitaxel formulations were included for pharmacokinetic analysis. Pharmacodynamic data was available for 36 patients. All population pharmacokinetic/pharmacodynamic modelling was performed using non-linear mixed-effects modelling. Results A pharmacokinetic model consisting of gut, liver, central, and peripheral compartments was developed for paclitaxel. The gastrointestinal absorption rate was modelled with a Weibull function. Relative gut bioavailabilities of the tablet and capsule formulations, as fractions of the gut bioavailability of the drinking solution, were estimated to be 0.97 (95%CI: 0.67–1.33) and 0.46 (95%CI: 0.34–0.61), respectively. The pharmacokinetic/pharmacodynamic relationship between paclitaxel and TSP-1 was modelled using a turnover model with paclitaxel plasma concentrations driving an increase in TSP-1 formation rate following an Emax relationship with an EC50 of 284 ng/mL (95%CI: 122–724). Conclusion The developed pharmacokinetic model adequately described the paclitaxel plasma concentrations for the different oral formulations co-administered with ritonavir. This model, and the established pharmacokinetic/pharmacodynamic relationship with TSP-1, may facilitate future development of oral paclitaxel. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-022-04445-z.
Collapse
Affiliation(s)
- Maarten van Eijk
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Huixin Yu
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Emilia Sawicki
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Modra Pharmaceuticals Holding B.V., Amsterdam, The Netherlands
| | - Vincent A de Weger
- Department of Clinical Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Thomas P C Dorlo
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
9
|
Borde S, Paul SK, Chauhan H. Ternary solid dispersions: classification and formulation considerations. Drug Dev Ind Pharm 2021; 47:1011-1028. [PMID: 33818224 DOI: 10.1080/03639045.2021.1908342] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The number of active pharmaceutical compounds from the biopharmaceutical classification system (BCS) belonging to Class II and IV have significantly increased in recent years. These compounds have high therapeutic potential but are difficult to formulate as oral dosage forms due to their poor aqueous solubility. The solubility and bioavailability of these poorly water-soluble compounds can be increased by various formulation approaches, such as amorphous solid dispersions (ASD), salt formation, complexations, etc. Out of these techniques, the ASD approach, where compounds are converted into amorphous form and embedded in the hydrophilic matrix, have been successfully used in many marketed preparations. The recent advancement of this ASD approach is the design of ternary solid dispersions (TSD), where an additional component is added to further improve their performance in terms of solubility, stability, and processability. This review discusses the classification, mechanism of performance improvement, preparation techniques, and characterizations for TSD.
Collapse
Affiliation(s)
- Shambhavi Borde
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA
| | - Sagar Kumar Paul
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA
| | - Harsh Chauhan
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA
| |
Collapse
|
10
|
Bhujbal SV, Mitra B, Jain U, Gong Y, Agrawal A, Karki S, Taylor LS, Kumar S, (Tony) Zhou Q. Pharmaceutical amorphous solid dispersion: A review of manufacturing strategies. Acta Pharm Sin B 2021; 11:2505-2536. [PMID: 34522596 PMCID: PMC8424289 DOI: 10.1016/j.apsb.2021.05.014] [Citation(s) in RCA: 236] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/05/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
Amorphous solid dispersions (ASDs) are popular for enhancing the solubility and bioavailability of poorly water-soluble drugs. Various approaches have been employed to produce ASDs and novel techniques are emerging. This review provides an updated overview of manufacturing techniques for preparing ASDs. As physical stability is a critical quality attribute for ASD, the impact of formulation, equipment, and process variables, together with the downstream processing on physical stability of ASDs have been discussed. Selection strategies are proposed to identify suitable manufacturing methods, which may aid in the development of ASDs with satisfactory physical stability.
Collapse
Key Words
- 3DP, three-dimensional printing
- ASDs, amorphous solid dispersions
- ASES, aerosol solvent extraction system
- Amorphous solid dispersions
- CAP, cellulose acetate phthalate
- CO2, carbon dioxide
- CSG, continuous-spray granulation
- Co-precipitation
- Downstream processing
- Drug delivery
- EPAS, evaporative aqueous solution precipitation
- Eudragit®, polymethacrylates derivatives
- FDM, fused deposition modeling
- GAS, gas antisolvent
- HME, hot-melt extrusion
- HPC, hydroxypropyl cellulose
- HPMC, hydroxypropyl methylcellulose
- HPMCAS, hydroxypropyl methylcellulose acetate succinate
- HPMCP, hypromellose phthalate
- Manufacturing
- Melting process
- PCA, precipitation with compressed fluid antisolvent
- PGSS, precipitation from gas-saturated solutions
- PLGA, poly(lactic-co-glycolic acid
- PVP, polyvinylpyrrolidone
- PVPVA, polyvinylpyrrolidone/vinyl acetate
- RESS, rapid expansion of a supercritical solution
- SAS, supercritical antisolvent
- SCFs, supercritical fluids
- SEDS, solution-enhanced dispersion by SCF
- SLS, selective laser sintering
- Selection criteria
- Soluplus®, polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer
- Solvent evaporation
- Stability
- Tg, glass transition temperature
- USC, ultrasound compaction
- scCO2, supercritical CO2
Collapse
Affiliation(s)
- Sonal V. Bhujbal
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Biplob Mitra
- Oral Product Development, Bristol Myers Squibb, Summit, NJ 07901, USA
| | - Uday Jain
- Material Science and Engineering, Bristol Myers Squibb, Summit, NJ 07901, USA
| | - Yuchuan Gong
- Oral Product Development, Bristol Myers Squibb, Summit, NJ 07901, USA
| | - Anjali Agrawal
- Oral Product Development, Bristol Myers Squibb, Summit, NJ 07901, USA
| | - Shyam Karki
- Oral Product Development, Bristol Myers Squibb, Summit, NJ 07901, USA
| | - Lynne S. Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Sumit Kumar
- Oral Product Development, Bristol Myers Squibb, Summit, NJ 07901, USA
| | - Qi (Tony) Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
11
|
Arnamo H, Hillebrand MJ, Huitema AD, Nuijen B, Rosing H, Beijnen JH. Development and Validation of a Stability-indicating HPLC Method for the Analysis of Cabazitaxel in Jevtana® Concentrate-solvent Leftover Samples. CURR PHARM ANAL 2021. [DOI: 10.2174/1573412916666200327144051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aim/Background:
In this study, a stability-indicating method of the anticancer agent cabazitaxel was developed and validated. This method will be used to determine the chemical stability of commercially available concentrate-solvent mixture cabazitaxel (Jevtana®) to examine the possibility of multi-dosing from the same product vial after storage. The impossibility to re-use leftovers today is contributing to an unnecessary and significant financial waste.
Methods:
A forced degradation study of cabazitaxel was performed under different conditions to produce degradation products. Acidic, basic, oxidation, heat, and ultraviolet (UV) light conditions were tested. The method to determine the stability was developed so that potential degradation products would be shown in the UV spectra after separation from cabazitaxel with a C18 column in a high-performance liquid chromatography (HPLC) system. The only degradation product occurring during storage in room temperature and ambient light was identified by accurate mass Orbitrap Mass Spectrometry.
Results:
A stability-indicating method for cabazitaxel (Jevtana®) concentrate-solvent mixture has been
developed. We demonstrated that this method can be applied to stability studies with the purpose of
multi-dosing cabazitaxel from a chemical/physical stability perspective within the tested period of time
and conditions.
Conclusion:
As an addition, the only naturally occurring degradation product found has been identified
and a degradation reaction has been suggested.
Collapse
Affiliation(s)
- Hedvig Arnamo
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam,Netherlands
| | - Michel J.X. Hillebrand
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam,Netherlands
| | - Alwin D.R. Huitema
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam,Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam,Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam,0
| | - Jos H. Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam,Netherlands
| |
Collapse
|
12
|
Vermunt MAC, van der Heijden LT, Hendrikx JJMA, Schinkel AH, de Weger VA, van der Putten E, van Triest B, Bergman AM, Beijnen JH. Pharmacokinetics of docetaxel and ritonavir after oral administration of ModraDoc006/r in patients with prostate cancer versus patients with other advanced solid tumours. Cancer Chemother Pharmacol 2021; 87:855-869. [PMID: 33744986 DOI: 10.1007/s00280-021-04259-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE ModraDoc006 is a novel oral formulation of docetaxel. The clearance of intravenous docetaxel is higher in medically castrated prostate cancer patients as compared to patients with other types of solid tumours. Oral docetaxel requires co-administration ritonavir (r), which might further impact the pharmacokinetics (PK). We now compare the PK of docetaxel and ritonavir between patients with Hormone Sensitive Prostate Cancer (HSPC), metastatic Castration-Resistant Prostate Cancer (mCRPC) and other metastatic solid tumours, treated on the same dose and weekly schedule of ModraDoc006/r. METHODS The docetaxel and ritonavir PK were compared between four patient groups from three clinical phase I trials, including eight male and eight female patients with different types of solid tumours (study 1), seven patients with HSPC (study 2) and five patients with mCRPC (study 3). All patients were treated with ModraDoc006 30 mg and ritonavir 100 mg in the morning, followed by ModraDoc006 20 mg and ritonavir 100 mg in the evening (ModraDoc006/r 30-20/100-100). For comparative purposes, the PK of six mCRPC patients that received 30-20/200-100 in study 3 were also evaluated. RESULTS The maximum plasma concentration (Cmax) was significantly lower for both docetaxel and ritonavir in the prostate cancer patients as compared to the patients with other types of solid tumours treated at ModraDoc006/r 30-20/100-100. The docetaxel area under the plasma concentration versus time curve (AUC) was significantly different at this dose, with a mean AUC0-48 of 1359 ± 374 ng/mL*h (N = 8) in female patients and 894 ± 223 ng/mL*h (N = 8) in male patients with different solid tumours (study 1), 321 ± 81 (N = 7) in HSPC (study 2) and 367 ± 182 ng/mL*h (N = 5) in mCRPC (study 3). A similar pattern was observed for ritonavir. ModraDoc006/r 30-20/200-100 in six mCRPC patients led to a comparable ritonavir exposure as compared to the patients at 30-20/100-100 in study 1 and increased the docetaxel AUC0-48 to 1266 ± 473 ng/mL*h (N = 6). CONCLUSION The exposure to docetaxel and ritonavir was significantly lower in prostate cancer patients as compared to patients with other types of solid tumours, treated on ModraDoc006/r 30-20/100-100. An increase of the ritonavir dose increased the docetaxel exposure in mCRPC patients. Therefore, a different RP2D of ModraDoc006/r is pursued in castrated prostate cancer patients as compared to patients with other types of solid tumours. TRIAL REGISTRATION Study 1: ClinicalTrials.gov Identifier NCT01173913, date of registration August 2, 2010. Study 2: ClinicalTrials.gov Identifier NCT03066154, date of registration February 28, 2017. Study 3: ClinicalTrials.gov Identifier NCT03136640, date of registration May 2, 2017.
Collapse
Affiliation(s)
- Marit A C Vermunt
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands.
| | - Lisa T van der Heijden
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Jeroen J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands.,Department of Nuclear Medicine, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Alfred H Schinkel
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Vincent A de Weger
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Eric van der Putten
- Modra Pharmaceuticals BV, Barbara Strozzilaan 201, Amsterdam, 1083HN, The Netherlands
| | - Baukelien van Triest
- Department of Radiotherapy, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Andries M Bergman
- Department of Medical Oncology and Oncogenomics, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands.,Modra Pharmaceuticals BV, Barbara Strozzilaan 201, Amsterdam, 1083HN, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Heidelberglaan 100, Utrecht, 3584CX, The Netherlands
| |
Collapse
|
13
|
Vermunt MAC, Robbrecht DGJ, Devriese LA, Janssen JM, Thijssen B, Keessen M, van Eijk M, Kessels R, Eskens FALM, Beijnen JH, Mehra N, Bergman AM. ModraDoc006, an oral docetaxel formulation in combination with ritonavir (ModraDoc006/r), in metastatic castration-resistant prostate cancer patients: A phase Ib study. Cancer Rep (Hoboken) 2021; 4:e1367. [PMID: 33709626 PMCID: PMC8388171 DOI: 10.1002/cnr2.1367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/23/2021] [Accepted: 02/19/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND ModraDoc006 is an oral formulation of docetaxel, which is co-administered with the cytochrome P450 3A4 and P-glycoprotein inhibitor ritonavir (r): ModraDoc006/r. Weekly treatment with ModraDoc006/r had been evaluated in phase I trials in patients with different types of advanced solid tumors, but up to this point in time not in patients with metastatic castration-resistant prostate cancer (mCRPC). AIM We assessed safety and pharmacokinetics (PK) of ModraDoc006/r to establish the recommended phase 2 dose (RP2D) in patients with mCRPC. METHODS mCRPC patients, treatment naïve or following abiraterone or enzalutamide treatment, were included. Dose-escalation of ModraDoc006/r was based on safety and docetaxel PK. Antitumor activity was assessed by serum prostate-specific antigen (PSA) and radiological evaluation. RESULTS Cohort 1 (n = 5) received once weekly ModraDoc006 30 mg with ritonavir 100 mg in the morning, and ModraDoc006 20 mg with ritonavir 100 mg in the evening (30-20/100-100). The mean docetaxel area under the plasma concentration-time curve (mAUC0-inf) was 461 ng/mL × h with 1 dose limiting toxicity (DLT); grade 3 alanine transferase increase. In cohort 2 (n = 6, ModraDoc006/r 30-20/200-200), the mAUC0-inf was 1687 ng/mL × h with 2 DLTs; grade 3 diarrhea and mucositis. In cohort 3A (n = 6, ModraDoc006/r 30-20/200-100), the mAUC0-inf was 1517 ng/mL × h with 1 DLT; grade 3 diarrhea. In cohort 3B (n = 3, ModraDoc006/r 20-20/200-100), the mAUC0-inf was 558 ng/mL × h without DLTs. The mAUC0-inf exceeded estimated exposures of intravenous docetaxel in cohort 2 and 3A, was lower in cohort 1 and was in range in cohort 3B. PSA decreases of >50% occurred in 6/10 evaluable patients throughout the various cohorts. In five radiological evaluable patients, two confirmed partial responses were observed. CONCLUSION The RP2D was established at weekly ModraDoc006/r 30-20/200-100. Observed PSA and radiological responses suggest promising clinical activity. These results have led to an ongoing randomized Phase 2b study, comparing weekly ModraDoc006/r with 3-weekly IV docetaxel in patients with mCRPC.
Collapse
Affiliation(s)
- Marit A C Vermunt
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Debbie G J Robbrecht
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Lot A Devriese
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Julie M Janssen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bas Thijssen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Maarten van Eijk
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rob Kessels
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ferry A L M Eskens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Modra Pharmaceuticals B.V., Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andries M Bergman
- Department of Medical Oncology and Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Vermunt MAC, de Weger VA, Janssen JM, Lopez-Yurda MI, Keessen M, Thijssen B, Rosing H, Huitema ADR, Beijnen JH, Marchetti S. Effect of Food on the Pharmacokinetics of the Oral Docetaxel Tablet Formulation ModraDoc006 Combined with Ritonavir (ModraDoc006/r) in Patients with Advanced Solid Tumours. Drugs R D 2021; 21:103-111. [PMID: 33464545 PMCID: PMC7937587 DOI: 10.1007/s40268-020-00336-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2020] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION ModraDoc006 is a novel docetaxel tablet formulation that is co-administrated with the cytochrome P450 3A4 and P-glycoprotein inhibitor ritonavir (r): ModraDoc006/r. OBJECTIVES This study evaluated the effect of food consumed prior to administration of ModraDoc006/r on the pharmacokinetics of docetaxel and ritonavir. METHODS Patients with advanced solid tumours were enrolled in this randomized crossover study to receive ModraDoc006/r in a fasted state in week 1 and after a standardized high-fat meal in week 2 and vice versa. Pharmacokinetic sampling was conducted until 48 h after both study drug administrations. Docetaxel and ritonavir plasma concentrations were determined using liquid chromatography with tandem mass spectrometry. Safety was evaluated with the Common Terminology Criteria for Adverse Events, version 4.03. RESULTS In total, 16 patients completed the food-effect study. The geometric mean ratio (GMR) for the docetaxel area under the plasma concentration-time curve (AUC)0-48, AUC0-inf and maximum concentration (Cmax) were 1.11 (90% confidence interval [CI] 0.93-1.33), 1.19 (90% CI 1.00-1.41) and 1.07 (90% CI 0.81-1.42) in fed versus fasted conditions, respectively. For the ritonavir Cmax, the GMR was 0.79 (90% CI 0.69-0.90), whereas the AUC0-48 and AUC0-inf were bioequivalent. The most frequent treatment-related toxicities were grade ≤ 2 diarrhoea and fatigue. Hypokalaemia was the only observed treatment-related grade 3 toxicity. CONCLUSIONS The docetaxel and ritonavir exposure were not bioequivalent, as consumption of a high-fat meal prior to administration of ModraDoc006/r resulted in a slightly higher docetaxel exposure and lower ritonavir Cmax. Since docetaxel exposure is the only clinically relevant parameter in our patient population, the overall conclusion is that combined ModraDoc006 and ritonavir treatment may be slightly affected by concomitant intake of a high-fat meal. In view of the small effect, it is most likely that the intake of a light meal will not affect the systemic exposure to docetaxel. CLINICALTRIALS. GOV IDENTIFIER NCT03147378, date of registration: May 10 2017.
Collapse
Affiliation(s)
- Marit A C Vermunt
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands.
| | - Vincent A de Weger
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Julie M Janssen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Marta I Lopez-Yurda
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Bas Thijssen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
- Modra Pharmaceuticals BV, Amsterdam, The Netherlands
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Serena Marchetti
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Vermunt M, Marchetti S, Beijnen J. Pharmacokinetics and Toxicities of Oral Docetaxel Formulations Co-Administered with Ritonavir in Phase I Trials. Clin Pharmacol 2021; 13:21-32. [PMID: 33536797 PMCID: PMC7850405 DOI: 10.2147/cpaa.s292746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
Introduction Docetaxel is widely used as intravenous (IV) chemotherapy. Oral docetaxel is co-administered with the cytochrome P450 3A4 and P-glycoprotein inhibitor ritonavir to increase oral bioavailability. This research explores the relationship between the pharmacokinetics (PK) and toxicity of this novel oral chemotherapy. Methods The patients in two phase I trials were treated with different oral docetaxel formulations in combination with ritonavir in different dose levels, ranging from 20 to 80 mg docetaxel with 100 to 200 mg ritonavir a day. The patients were categorized based on the absence or occurrence of severe treatment-related toxicity (grade ≥3 or any grade leading to treatment alterations). The docetaxel area under the plasma concentration–time curve (AUC) and maximum plasma concentration (Cmax) were associated with toxicity. Results Thirty-four out of 138 patients experienced severe toxicity, most frequently observed as mucositis, fatigue, diarrhea, nausea and vomiting. The severe toxicity group had a significantly higher docetaxel AUC (2231 ± 1405 vs 1011 ± 830 ng/mL*h, p<0.0001) and Cmax (218 ± 178 vs 119 ± 77 ng/mL, p<0.0001) as compared to the patients without severe toxicity. When extrapolated from IV PK data, the patients without severe toxicity had a similar cumulative docetaxel AUC as with standard 3-weekly IV docetaxel, while the Cmax was up to 10-fold lower with oral docetaxel and ritonavir. Conclusion Severe toxicity was observed in 25% of the patients treated with oral docetaxel and ritonavir. This toxicity seems related to the PK, as the docetaxel AUC0-inf and Cmax were up to twofold higher in the severe toxicity group as compared to the non-severe toxicity group. Future randomized trials will provide a further evaluation of the toxicity and efficacy of the new weekly oral docetaxel and ritonavir regimen in comparison to standard IV docetaxel.
Collapse
Affiliation(s)
- Marit Vermunt
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066, CX, the Netherlands
| | - Serena Marchetti
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066, CX, the Netherlands
| | - Jos Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066, CX, the Netherlands.,Modra Pharmaceuticals B.V., Amsterdam 1083, HN, the Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht 3584, CX, the Netherlands
| |
Collapse
|
16
|
Szabó E, Záhonyi P, Brecska D, Galata DL, Mészáros LA, Madarász L, Csorba K, Vass P, Hirsch E, Szafraniec-Szczęsny J, Csontos I, Farkas A, Van denMooter G, Nagy ZK, Marosi G. Comparison of Amorphous Solid Dispersions of Spironolactone Prepared by Spray Drying and Electrospinning: The Influence of the Preparation Method on the Dissolution Properties. Mol Pharm 2021; 18:317-327. [PMID: 33301326 PMCID: PMC7788570 DOI: 10.1021/acs.molpharmaceut.0c00965] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/02/2022]
Abstract
This research aimed to compare two solvent-based methods for the preparation of amorphous solid dispersions (ASDs) made up of poorly soluble spironolactone and poly(vinylpyrrolidone-co-vinyl acetate). The same apparatus was used to produce, in continuous mode, drug-loaded electrospun (ES) and spray-dried (SD) materials from dichloromethane and ethanol-containing solutions. The main differences between the two preparation methods were the concentration of the solution and application of high voltage. During electrospinning, a solution with a higher concentration and high voltage was used to form a fibrous product. In contrast, a dilute solution and no electrostatic force were applied during spray drying. Both ASD products showed an amorphous structure according to differential scanning calorimetry and X-ray powder diffraction results. However, the dissolution of the SD sample was not complete, while the ES sample exhibited close to 100% dissolution. The polarized microscopy images and Raman microscopy mapping of the samples highlighted that the SD particles contained crystalline traces, which can initiate precipitation during dissolution. Investigation of the dissolution media with a borescope made the precipitated particles visible while Raman spectroscopy measurements confirmed the appearance of the crystalline active pharmaceutical ingredient. To explain the micro-morphological differences, the shape and size of the prepared samples, the evaporation rate of residual solvents, and the influence of the electrostatic field during the preparation of ASDs had to be considered. This study demonstrated that the investigated factors have a great influence on the dissolution of the ASDs. Consequently, it is worth focusing on the selection of the appropriate ASD preparation method to avoid the deterioration of dissolution properties due to the presence of crystalline traces.
Collapse
Affiliation(s)
- Edina Szabó
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Petra Záhonyi
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Dániel Brecska
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Dorián L. Galata
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Lilla A. Mészáros
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Lajos Madarász
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Kristóf Csorba
- Department
of Automation and Applied Informatics, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Panna Vass
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Edit Hirsch
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Joanna Szafraniec-Szczęsny
- Department
of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Jagellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - István Csontos
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Attila Farkas
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Guy Van denMooter
- Department
of Pharmaceutical and Pharmacological Sciences, Drug Delivery and
Disposition, KU Leuven, Campus Gasthuisberg ON2, Herestraat
49 b921, 3000 Leuven, Belgium
| | - Zsombor K. Nagy
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - György Marosi
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics (BME), Műegyetem rkp. 3, H-1111 Budapest, Hungary
| |
Collapse
|
17
|
Vermunt MA, Bergman AM, der Putten EV, Beijnen JH. The intravenous to oral switch of taxanes: strategies and current clinical developments. Future Oncol 2020; 17:1379-1399. [PMID: 33356545 DOI: 10.2217/fon-2020-0876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The taxanes paclitaxel, docetaxel and cabazitaxel are important anticancer agents that are widely used as intravenous treatment for several solid tumor types. Switching from intravenous to oral treatment can be more convenient for patients, improve cost-effectiveness and reduce the demands of chemotherapy treatment on hospital care. However, oral treatment with taxanes is challenging because of pharmaceutical and pharmacological factors that lead to low oral bioavailability. This review summarizes the current clinical developments in oral taxane treatment. Intravenous parent drugs, strategies in the oral switch, individual agents in clinical trials, challenges and further perspectives on treatment with oral taxanes are subsequently discussed.
Collapse
Affiliation(s)
- Marit Ac Vermunt
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Andries M Bergman
- Department of Medical Oncology & Oncogenomics, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Eric van der Putten
- Modra Pharmaceuticals BV, Barbara Strozzilaan 201, Amsterdam, 1083HN, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands.,Modra Pharmaceuticals BV, Barbara Strozzilaan 201, Amsterdam, 1083HN, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Heidelberglaan 100, Utrecht, 3584CX, The Netherlands
| |
Collapse
|
18
|
de Weger VA, Vermunt MAC, Stuurman FE, Burylo AM, Damoiseaux D, Hendrikx JJMA, Sawicki E, Moes JJ, Huitema ADR, Nuijen B, Rosing H, Mergui-Roelvink M, Beijnen JH, Marchetti S. A Phase 1 Dose-Escalation Study of Low-Dose Metronomic Treatment With Novel Oral Paclitaxel Formulations in Combination With Ritonavir in Patients With Advanced Solid Tumors. Clin Pharmacol Drug Dev 2020; 10:607-621. [PMID: 33021083 DOI: 10.1002/cpdd.880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/08/2020] [Indexed: 01/08/2023]
Abstract
ModraPac001 (MP1) and ModraPac005 (MP5) are novel oral paclitaxel formulations that are coadministered with the cytochrome P450 3A4 inhibitor ritonavir (r), enabling daily low-dose metronomic (LDM) treatment. The primary aim of this study was to determine the safety, pharmacokinetics and maximum tolerated dose (MTD) of MP1/r and MP5/r. The second aim was to establish the recommended phase 2 dose (RP2D) as LDM treatment. This was an open-label phase 1 trial. Patients with advanced solid tumors were enrolled according to a classical 3+3 design. After initial employment of the MP1 capsule, the MP5 tablet was introduced. Safety was assessed using the Common Terminology Criteria for Adverse Events version 4.02. Pharmacokinetic sampling was performed on days 1, 2, 8, and 22 for determination of paclitaxel and ritonavir plasma concentrations. In this study, 37 patients were treated with up to twice-daily 30-mg paclitaxel combined with twice-daily 100-mg ritonavir (MP5/r 30-30/100-100) in 9 dose levels. Dose-limiting toxicities were nausea, (febrile) neutropenia, dehydration and vomiting. At the MTD/RP2D of MP5/r 20-20/100-100, the maximum paclitaxel plasma concentration and area under the concentration-time curve until 24 hours were 34.6 ng/mL (coefficient of variation, 79%) and 255 ng • h/mL (coefficient of variation, 62%), respectively. Stable disease was observed as best response in 15 of 31 evaluable patients. Based on these results, LDM therapy with oral paclitaxel coadministrated with ritonavir was considered feasible and safe. The MTD and RP2D were determined as MP5/r 20-20/100-100. Further clinical development of MP5/r as an LDM concept, including potential combination treatment, is warranted.
Collapse
Affiliation(s)
- Vincent A de Weger
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marit A C Vermunt
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frederik E Stuurman
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Artur M Burylo
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - David Damoiseaux
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeroen J M A Hendrikx
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Emilia Sawicki
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Modra Pharmaceuticals BV, Amsterdam, The Netherlands
| | - Johannes J Moes
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marja Mergui-Roelvink
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Modra Pharmaceuticals BV, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Serena Marchetti
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Kataoka M, Nakanishi R, Umesaki M, Kobayashi M, Minami K, Higashino H, Yamaguchi S, Yamashita S. An enteric polymer mitigates the effects of gastric pH on oral absorption of poorly soluble weak acid drugs from supersaturable formulations: A case study with dantrolene. Eur J Pharm Biopharm 2020; 155:29-36. [DOI: 10.1016/j.ejpb.2020.07.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 10/23/2022]
|
20
|
Hendrikx JJMA, Stuurman FE, Song J, de Weger VA, Lagas JS, Rosing H, Beijnen JH, Schinkel AH, Schellens JHM, Marchetti S. No relation between docetaxel administration route and high-grade diarrhea incidence. Pharmacol Res Perspect 2020; 8:e00633. [PMID: 32725720 PMCID: PMC7387127 DOI: 10.1002/prp2.633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 01/12/2023] Open
Abstract
Oral administration of docetaxel in combination with the CYP3A4 inhibitor ritonavir is used in clinical trials to improve oral bioavailability of docetaxel. Diarrhea was the most commonly observed and dose-limiting toxicity. This study combined preclinical and clinical data and investigated incidence, severity and cause of oral docetaxel-induced diarrhea. In this study, incidence and severity of diarrhea in patients were compared to exposure to orally administered docetaxel. Intestinal toxicity after oral or intraperitoneal administration of docetaxel was further explored in mice lacking Cyp3a and mice lacking both Cyp3a and P-glycoprotein. In patients, severity of diarrhea increased significantly with an increase in AUC and Cmax (P = .035 and P = .025, respectively), but not with an increase in the orally administered dose (P = .11). Furthermore, incidence of grade 3/4 diarrhea after oral docetaxel administration was similar as reported after intravenous docetaxel administration. Intestinal toxicity in mice was only observed at high systemic exposure to docetaxel and was similar after oral and intraperitoneal administration of docetaxel. In conclusion, our data show that the onset of severe diarrhea after oral administration of docetaxel in humans is similar after oral and intravenous administration of docetaxel and is caused by the concentration of docetaxel in the systemic blood circulation. Mouse experiments confirmed that intestinal toxicity is caused by a high systemic exposure and not by local intestinal exposure. Severe diarrhea in patients after oral docetaxel is reversible and is not related to the route of administration of docetaxel.
Collapse
Affiliation(s)
- Jeroen J. M. A. Hendrikx
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Nuclear MedicineThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Frederik E. Stuurman
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Ji‐Ying Song
- Department of Experimental Animal PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Vincent A. de Weger
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jurjen S. Lagas
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- DiakonessenhuisUtrechtThe Netherlands
| | - Hilde Rosing
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jos H. Beijnen
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Modra Pharmaceuticals BVAmsterdamThe Netherlands
| | - Alfred H. Schinkel
- Division of PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jan H. M. Schellens
- Department of Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Modra Pharmaceuticals BVAmsterdamThe Netherlands
| | - Serena Marchetti
- Department of Clinical PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
21
|
Yu H, Janssen JM, de Weger VA, Nuijen B, Stuurman RE, Marchetti S, Schellens JHM, Beijnen JH, Dorlo TPC, Huitema ADR. Quantification of the pharmacokinetic-toxicodynamic relationship of oral docetaxel co-administered with ritonavir. Invest New Drugs 2020; 38:1526-1532. [PMID: 32306204 DOI: 10.1007/s10637-020-00935-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/07/2020] [Indexed: 11/25/2022]
Abstract
Introduction Oral formulations of docetaxel have successfully been developed as an alternative for intravenous administration. Co-administration with the enzyme inhibitor ritonavir boosts the docetaxel plasma exposure. In dose-escalation trials, the maximum tolerated doses for two different dosing regimens were established and dose-limiting toxicities (DLTs) were recorded. The aim of current analysis was to develop a pharmacokinetic (PK)-toxicodynamic (TOX) model to quantify the relationship between docetaxel plasma exposure and DLTs. Methods A total of 85 patients was included in the current analysis, 18 patients showed a DLT in the four-week observation period. A PK-TOX model was developed and simulations based on the PK-TOX model were performed. Results The final PK-TOX model was characterized by an effect compartment representing the toxic effect of docetaxel, which was linked to the probability of developing a DLT. Simulations of once-weekly, once-daily 60 mg and once-weekly, twice-daily 30 mg followed by 20 mg of oral docetaxel suggested that 14% and 34% of patients, respectively, would have a probability >25% to develop a DLT in a four-week period. Conclusions A PK-TOX model was successfully developed. This model can be used to evaluate the probability of developing a DLT following treatment with oral docetaxel and ritonavir in different dosing regimens.
Collapse
Affiliation(s)
- Huixin Yu
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - Julie M Janssen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
| | - Vincent A de Weger
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - Rik E Stuurman
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
- Centre for Human Drug Research, Leiden, The Netherlands
| | - Serena Marchetti
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Thomas P C Dorlo
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
22
|
Harnessing the therapeutic potential of anticancer drugs through amorphous solid dispersions. Biochim Biophys Acta Rev Cancer 2019; 1873:188319. [PMID: 31678141 DOI: 10.1016/j.bbcan.2019.188319] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
The treatment of cancer is still a major challenge. But tremendous progress in anticancer drug discovery and development has occurred in the last few decades. However, this progress has resulted in few effective oncology products due to challenges associated with anticancer drug delivery. Oral administration is the most preferred route for anticancer drug delivery, but the majority of anticancer drugs currently in product pipelines and the majority of those that have been commercially approved have inherently poor water solubility, and this cannot be mitigated without compromising their potency and stability. The poor water solubility of anticancer drugs, in conjunction with other factors, leads to suboptimal pharmacokinetic performance. Thus, these drugs have limited efficacy and safety when administered orally. The amorphous solid dispersion (ASD) is a promising formulation technology that primarily enhances the aqueous solubility of poorly water-soluble drugs. In this review, we discuss the challenges associated with the oral administration of anticancer drugs and the use of ASD technology in alleviating these challenges. We emphasize the ability of ASDs to improve not only the pharmacokinetics of poorly water-soluble anticancer drugs, but also their efficacy and safety. The goal of this paper is to rationalize the application of ASD technology in the formulation of anticancer drugs, thereby creating superior oncology products that lead to improved therapeutic outcomes.
Collapse
|
23
|
Comparison of paclitaxel solid dispersion and polymeric micelles for improved oral bioavailability and in vitro anti-cancer effects. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:247-259. [DOI: 10.1016/j.msec.2019.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 02/21/2019] [Accepted: 03/01/2019] [Indexed: 12/24/2022]
|
24
|
de Weger VA, Stuurman FE, Koolen SLW, Moes JJ, Hendrikx JJMA, Sawicki E, Thijssen B, Keessen M, Rosing H, Mergui-Roelvink M, Huitema ADR, Nuijen B, Beijnen JH, Schellens JHM, Marchetti S. A Phase I Dose Escalation Study of Once-Weekly Oral Administration of Docetaxel as ModraDoc001 Capsule or ModraDoc006 Tablet in Combination with Ritonavir. Clin Cancer Res 2019; 25:5466-5474. [PMID: 31217201 DOI: 10.1158/1078-0432.ccr-17-2299] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/31/2017] [Accepted: 06/07/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Oral bioavailability of docetaxel is poor. Absorption could be improved by development of pharmaceutical formulations based on docetaxel solid dispersions, denoted ModraDoc001 capsule and ModraDoc006 tablet (both 10 mg) and coadministration of ritonavir, an inhibitor of CYP3A4 and P-glycoprotein. In this study, the safety, MTD, recommended phase II dose (RP2D), pharmacokinetics, and preliminary antitumor activity of oral docetaxel combined with ritonavir in a once-weekly continuous schedule was investigated. PATIENTS AND METHODS Patients with metastatic solid tumors were included. Dose escalation was performed using a classical 3+3 design. Pharmacokinetic sampling was performed for up to 48 hours after drug administration. Safety was evaluated using CTCAE v3.0. Antitumor activity was assessed according to RECIST v1.0. RESULTS Sixty-seven patients were treated at weekly docetaxel dosages ranging from 30 to 80 mg in combination with 100- or 200-mg ritonavir. Most common toxicities were nausea, vomiting, diarrhea and fatigue, mostly of grade 1-2 severity. No hypersensitivity reactions were observed. The area under the plasma concentration-time curve (AUC0-48) of docetaxel at the RP2D of once-weekly 60-mg ModraDoc001 capsule with 100-mg ritonavir was 1,000 ± 687 ng/mL/hour and for once-weekly 60-mg ModraDoc006 tablet with 100-mg ritonavir, the AUC0-48 was 1,790 ± 819 ng/mL/hour. Nine partial responses were reported as best response to treatment. CONCLUSIONS Oral administration of once-weekly docetaxel as ModraDoc001 capsule or ModraDoc006 tablet in combination with ritonavir is feasible. The RP2D for both formulations is 60-mg ModraDoc with 100-mg ritonavir. Antitumor activity is considered promising.
Collapse
Affiliation(s)
- Vincent A de Weger
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frederik E Stuurman
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Stijn L W Koolen
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Johannes J Moes
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - Jeroen J M A Hendrikx
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - Emilia Sawicki
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands.,Modra Pharmaceuticals BV, Amsterdam, the Netherlands
| | - Bas Thijssen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - Marianne Keessen
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Modra Pharmaceuticals BV, Amsterdam, the Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - Marja Mergui-Roelvink
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands.,Modra Pharmaceuticals BV, Amsterdam, the Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jan H M Schellens
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Modra Pharmaceuticals BV, Amsterdam, the Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Serena Marchetti
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Xie Y, Yao Y. Incorporation With Dendrimer-Like Biopolymer Leads to Improved Soluble Amount and In Vitro Anticancer Efficacy of Paclitaxel. J Pharm Sci 2019; 108:1984-1990. [DOI: 10.1016/j.xphs.2018.12.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 11/15/2018] [Accepted: 12/13/2018] [Indexed: 11/25/2022]
|
26
|
Ngo HV, Tran PH, Lee BJ, Tran TT. The roles of a surfactant in zein-HPMC blend solid dispersions for improving drug delivery. Int J Pharm 2019; 563:169-173. [DOI: 10.1016/j.ijpharm.2019.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/19/2022]
|
27
|
Poozesh S, Bilgili E. Scale-up of pharmaceutical spray drying using scale-up rules: A review. Int J Pharm 2019; 562:271-292. [PMID: 30910632 DOI: 10.1016/j.ijpharm.2019.03.047] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/31/2022]
Abstract
Spray drying is one of the widely used manufacturing processes in pharmaceutical industry. While there are voluminous experimental studies pertaining to the impact of various process-formulation parameters on the quality attributes of spray dried powders such as particle size, morphology, density, and crystallinity, there is scant information available in the literature regarding process scale-up. Here, we first analyze salient features of scale-up attempts in literature. Then, spray drying process is analyzed considering the fundamental physical transformations involved, i.e., atomization, drying, and gas-solid separation. Each transformation is scrutinized from a scale-up perspective with non-dimensional parameters & multi-scale analysis, and comprehensively discussed in engineering context. Successful scale-up entails similar key response variables from each transformation across various scales. These variables are identified as droplet size distribution, outlet temperature, relative humidity, separator pressure loss coefficient, and collection efficiency. Instead of trial-and-error-based approaches, this review paper advocates the use of mechanistic models and scale-up rules for establishing design spaces for the process variables involved in each transformation of spray drying. While presenting a roadmap for process development and scale-up, the paper demonstrates how to bridge the current gap in spray drying scale-up via a rational understanding of the fundamental transformations.
Collapse
Affiliation(s)
- Sadegh Poozesh
- Mechanical Engineering Department, Tuskegee University, Tuskegee, AL 36088, United States.
| | - Ecevit Bilgili
- Chemical and Materials Engineering Department, New Jersey Institute of Technology, Newark, NJ 07102, United States
| |
Collapse
|
28
|
Hot-melt extrusion process impact on polymer choice of glyburide solid dispersions: The effect of wettability and dissolution. Int J Pharm 2019; 559:245-254. [DOI: 10.1016/j.ijpharm.2019.01.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 11/23/2022]
|
29
|
Zhang E, Xing R, Liu S, Li P. Current advances in development of new docetaxel formulations. Expert Opin Drug Deliv 2019; 16:301-312. [PMID: 30773947 DOI: 10.1080/17425247.2019.1583644] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Docetaxel (DTX) is one of the most important chemotherapeutic agents and has been widely used for treatment of various types of cancers. However, the clinical chemotherapy of DTX gives many undesirable side effects due to the usage of organic solvent in the injection and its low selectivity for tumor cells. With the evolution of pharmaceutical technologies, great efforts have been paid to develop new DTX formulations to overcome these problems. AREAS COVERED This review provided an overview of the preparation and activities of new DTX formulations, which were classified by administration methods, including injection, oral, transdermal and rectal administration. Besides, up to date information of the clinical status of new DTX formulations was summarized. We also discussed the challenges and perspectives of the future development of DTX formulations. EXPERT OPINION There have been numerous studies on new DTX-based formulations in recent years, and many of them exhibited significantly enhanced anti-tumor and targeting activity compared with DTX in preclinical studies. However, only a few entered clinical trials, and none has been approved into market. The clinical translation of experimental drug faces many hurdles, including the limited knowledge of nanomedicine and oncology, safety issues, controllable and reproducible production.
Collapse
Affiliation(s)
- Enhui Zhang
- a CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology , Chinese Academy of Sciences , Qingdao , PR China.,b Laboratory for Marine Drugs and Bioproducts , Qingdao National Laboratory of Marine Science and Technology , Qingdao , PR China.,c Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao , PR China
| | - Ronge Xing
- a CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology , Chinese Academy of Sciences , Qingdao , PR China.,b Laboratory for Marine Drugs and Bioproducts , Qingdao National Laboratory of Marine Science and Technology , Qingdao , PR China.,c Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao , PR China
| | - Song Liu
- a CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology , Chinese Academy of Sciences , Qingdao , PR China.,b Laboratory for Marine Drugs and Bioproducts , Qingdao National Laboratory of Marine Science and Technology , Qingdao , PR China.,c Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao , PR China
| | - Pengcheng Li
- a CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology , Chinese Academy of Sciences , Qingdao , PR China.,b Laboratory for Marine Drugs and Bioproducts , Qingdao National Laboratory of Marine Science and Technology , Qingdao , PR China.,c Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao , PR China
| |
Collapse
|
30
|
Dong W, Su X, Xu M, Hu M, Sun Y, Zhang P. Preparation, characterization, and in vitro/vivo evaluation of polymer-assisting formulation of atorvastatin calcium based on solid dispersion technique. Asian J Pharm Sci 2018; 13:546-554. [PMID: 32211078 PMCID: PMC7081932 DOI: 10.1016/j.ajps.2018.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/04/2018] [Accepted: 08/17/2018] [Indexed: 11/29/2022] Open
Abstract
Due to low solubility and bioavailability, atorvastatin calcium is confronted with challenge in conceiving appropriate formulation. Solid dispersion of atorvastatin calcium was prepared through the solvent evaporation method, with Poloxamer 188 as hydrophilic carriers. This formulation was then characterized by scanning electron microscopy, differential scanning calorimetry, powder X-ray diffraction and fourier transform infrared spectroscopy. Moreover, all these studies suggested the conversion of crystalline atorvastatin calcium. In addition, the drug solubility studies as well as dissolution rates compared with bulk drug and market tablets Lipitor were also examined. Furthermore, the study investigated the pharmacokinetics after oral administration of Lipitor and solid dispersion. And the AUC0–8 h and Cmax increased after taking ATC-P188 solid dispersion orally compared with that of Lipitor. All these could be demonstrated that ATC-P188 solid dispersions would be prospective means for enhancing higher oral bioavailability of ATC.
Collapse
Affiliation(s)
| | | | | | | | | | - Peng Zhang
- Corresponding author. Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China. Tel.:+86 24 43520509
| |
Collapse
|
31
|
Chowdhury P, Nagesh PK, Khan S, Hafeez BB, Chauhan SC, Jaggi M, Yallapu MM. Development of polyvinylpyrrolidone/paclitaxel self-assemblies for breast cancer. Acta Pharm Sin B 2018; 8:602-614. [PMID: 30109184 PMCID: PMC6090082 DOI: 10.1016/j.apsb.2017.10.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/13/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022] Open
Abstract
The goal of this investigation was to develop and demonstrate a polymer/paclitaxel self-assembly (PTX-SA) formulation. Polymer/PTX-SAs were screened based on smaller size of formulation using dynamic light scattering analysis. Additionally, fluorescence microscopy and flow cytometry studies exhibited that polyvinylpyrrolidone (PVP)-based PTX-SAs (PVP/PTX-SAs) had superior cellular internalization capability in MCF7 and MDA-MB-231 breast cancer cells. The optimized PVP/PTX-SAs exhibited less toxicity to human red blood cells indicating a suitable formulation for reducing systemic toxicity. The formation of PVP and PTX self-assemblies was confirmed using fluorescence quenching and transmission electron microscopy which indicated that the PVP/PTX-SAs were spherical in shape with an average size range of 53.81 nm as detected by transmission electron microscopy (TEM). FTIR spectral analysis demonstrates incorporation of polymer and paclitaxel functional groups in PVP/PTX-SAs. Both proliferation (MTS) and clonogenic (colony formation) assays were used to validate superior anticancer activity of PVP/PTX-SAs in breast cancer cells over paclitaxel. Such superior anticancer activity was also demonstrated by downregulation of the expression of pro-survival protein (Bcl-xL), upregulation of apoptosis-associated proteins (Bid, Bax, cleaved caspase 7, and cleaved PARP) and β-tubulin stabilization. These results support the hypothesis that PVP/PTX-SAs improved paclitaxel delivery to cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Murali M. Yallapu
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
32
|
Baghel S, Cathcart H, O'Reilly NJ. Understanding the generation and maintenance of supersaturation during the dissolution of amorphous solid dispersions using modulated DSC and 1H NMR. Int J Pharm 2017; 536:414-425. [PMID: 29183857 DOI: 10.1016/j.ijpharm.2017.11.056] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/23/2017] [Accepted: 11/24/2017] [Indexed: 11/28/2022]
Abstract
In this study, the dissolution behaviour of dipyridamole (DPM) and cinnarizine (CNZ) spray-dried amorphous solid dispersions (ASDs) using polyvinyl pyrrolidone (PVP) and polyacrylic acid (PAA) as a carrier matrix were evaluated and compared. The drug concentrations achieved from the dissolution of PVP and PAA solid dispersions were significantly greater than the equilibrium solubility of crystalline DPM and CNZ in phosphate buffer pH 6.8 (PBS 6.8). The maximum drug concentration achieved by dissolution of PVP and PAA solid dispersions did not exceed the theoretically calculated apparent solubility of amorphous DPM and CNZ. However, the degree of supersaturation of DPM and CNZ increased considerably as the polymer weight fraction within the solid dispersion increased. In addition, the supersaturation profile of DPM and CNZ were studied in the presence and absence of the polymers. PAA was found to maintain a higher level of supersaturation compared to PVP. The enhanced drug solution concentration following dissolution of ASDs can be attributed to the reduced crystal growth rates of DPM and CNZ at an equivalent supersaturation. We have also shown that, for drugs having high crystallization tendency and weak drug-polymer interaction, the feasible way to increase dissolution might be increase the polymer weight fraction in the ASD. Solution 1H NMR spectra were used to understand dissolution mechanism and to identify drug-polymer interaction. The change in electron densities of proton attached to different groups in DPM and CNZ suggested drug-polymer interaction in solution. The relative intensities of peak shift and nature of interaction between drug and polymer in different systems are different. These different effects suggest that DPM and CNZ interacts in a different way with PVP and PAA in solution which goes some way towards explaining the different polymeric effect, particularly in terms of inhibition of drug recrystallization and dissolution of DPM and CNZ ASDs. These results established that the different drug/polymer interactions in the solid state and in solution give rise to the variation in dissolution profile observed for different systems.
Collapse
Affiliation(s)
- Shrawan Baghel
- Synthesis and Solid State Pharmaceutical Centre (SSPC), Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Cork Road, Waterford, Ireland.
| | - Helen Cathcart
- Synthesis and Solid State Pharmaceutical Centre (SSPC), Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Cork Road, Waterford, Ireland
| | - Niall J O'Reilly
- Synthesis and Solid State Pharmaceutical Centre (SSPC), Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Cork Road, Waterford, Ireland
| |
Collapse
|
33
|
Davis M, Walker G. Recent strategies in spray drying for the enhanced bioavailability of poorly water-soluble drugs. J Control Release 2017; 269:110-127. [PMID: 29117503 DOI: 10.1016/j.jconrel.2017.11.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 01/07/2023]
Abstract
Poorly water-soluble drugs are a significant and ongoing issue for the pharmaceutical industry. An overview of recent developments for the preparation of spray-dried delivery systems is presented. Examples include amorphous solid dispersions, spray dried dispersions, microparticles, nanoparticles, surfactant systems and self-emulsifying drug delivery systems. Several aspects of formulation are considered, such as pre-screening, choosing excipient(s), the effect of polymer structure on performance, formulation optimisation, ternary dispersions, fixed-dose combinations, solvent selection and component miscibility. Process optimisation techniques including nozzle selection are discussed. Comparisons are drawn with other preparation techniques such as hot melt extrusion, freeze drying, milling, electro spinning and film casting. Novel analytical and dissolution techniques for the characterization of amorphous solid dispersions are included. Progress in understanding of amorphous supersaturation or recrystallisation from solution gathered from mechanistic studies is discussed. Aspects of powder flow and compression are considered in a section on downstream processing. Overall, spray drying has a bright future due to its versatility, efficiency and the driving force of poorly soluble drugs.
Collapse
Affiliation(s)
- Mark Davis
- Synthesis and Solid State Pharmaceutical Centre (SSPC), Bernal Institute, University of Limerick, Limerick, Ireland.
| | - Gavin Walker
- Bernal Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
34
|
Démuth B, Galata DL, Szabó E, Nagy B, Farkas A, Balogh A, Hirsch E, Pataki H, Rapi Z, Bezúr L, Vigh T, Verreck G, Szalay Z, Demeter Á, Marosi G, Nagy ZK. Investigation of Deteriorated Dissolution of Amorphous Itraconazole: Description of Incompatibility with Magnesium Stearate and Possible Solutions. Mol Pharm 2017; 14:3927-3934. [PMID: 28972782 DOI: 10.1021/acs.molpharmaceut.7b00629] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Disadvantageous crystallization phenomenon of amorphous itraconazole (ITR) occurring in the course of dissolution process was investigated in this work. A perfectly amorphous form (solid dispersion) of the drug was generated by the electroblowing method (with vinylpyrrolidone-vinyl acetate copolymer), and the obtained fibers were formulated into tablets. Incomplete dissolution of the tablets was noticed under the circumstances of the standard dissolution test, after which a precipitated material could be filtered. The filtrate consisted of ITR and stearic acid since no magnesium content was detectable in it. In parallel with dissolution, ITR forms an insoluble associate, stabilized by hydrogen bonding, with stearic acid deriving from magnesium stearate. This is why dissolution curves do not have the plateaus at 100%. Two ways are viable to tackle this issue: change the lubricant (with sodium stearyl fumarate >95% dissolution can be accomplished) or alter the polymer in the solid dispersion to a type being able to form hydrogen bonds with ITR (e.g., hydroxypropyl methylcellulose). This work draws attention to one possible phenomenon that can lead to a deterioration of originally good dissolution of an amorphous solid dispersion.
Collapse
Affiliation(s)
- B Démuth
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - D L Galata
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - E Szabó
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - B Nagy
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - A Farkas
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - A Balogh
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - E Hirsch
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - H Pataki
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Z Rapi
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - L Bezúr
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics (BME) , Szent Gellért tér 4, H-1111, Budapest, Hungary
| | - T Vigh
- Drug Product Development, Janssen R&D , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - G Verreck
- Drug Product Development, Janssen R&D , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Z Szalay
- Drug Polymorphism Research, Gedeon Richter Plc. , Gyömrői út 30-32, H-1103 Budapest, Hungary
| | - Á Demeter
- Drug Polymorphism Research, Gedeon Richter Plc. , Gyömrői út 30-32, H-1103 Budapest, Hungary
| | - G Marosi
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Z K Nagy
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics (BME) , Műegyetem rkp. 3, H-1111 Budapest, Hungary
| |
Collapse
|
35
|
A dose-escalation study of bi-daily once weekly oral docetaxel either as ModraDoc001 or ModraDoc006 combined with ritonavir. Eur J Cancer 2017; 86:217-225. [PMID: 29031170 DOI: 10.1016/j.ejca.2017.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 09/14/2017] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Two solid dispersions of docetaxel (denoted ModraDoc001 capsule and ModraDoc006 tablet (both 10 mg)) were co-administered with 100 mg ritonavir (/r) and investigated in a bi-daily once weekly (BIDW) schedule. Safety, maximum tolerated dose (MTD), pharmacokinetics (PK) and preliminary activity were explored. METHODS Adult patients with metastatic solid tumours were included in two dose-escalation arms. PK sampling was performed during the first week and the second or third week. Safety was evaluated using US National Cancer Institute's Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 3.0. Antitumour activity was assessed every 6 weeks according to Response Evaluation Criteria in Solid Tumours (RECIST) version 1.0. RESULTS ModraDoc001 capsule/r and ModraDoc006 tablet/r were administered to 17 and 28 patients, respectively. The most common adverse events were nausea, vomiting, diarrhoea and fatigue, mostly of grade 1-2 severity. Grade 3/4 neutropenia/neutropenic fever was observed in 2 patients (4%). The MTD was determined as 20/20 mg ModraDoc001/r and 30/20 mg ModraDoc006/r (morning/afternoon dose) once weekly. The mean area under the plasma concentration-time curve (AUC0-48) ± standard deviation at the MTD for ModraDoc001/r and ModraDoc006/r were 686 ± 388 ng/ml*h and 1126 ± 382 ng/ml*h, respectively. Five partial responses were reported as best response to treatment. CONCLUSION Oral administration of BIDW ModraDoc001/r or ModraDoc006/r is feasible. The once weekly 30/20 mg ModraDoc006 tablet/r dose-level was selected for future clinical development. Antitumour activity is promising.
Collapse
|
36
|
Obaidat R, Alnaief M, Jaeger P. Significant solubility of carbon dioxide in Soluplus ® facilitates impregnation of ibuprofen using supercritical fluid technology. Pharm Dev Technol 2017; 23:697-705. [PMID: 28375669 DOI: 10.1080/10837450.2017.1315135] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Treatment of Soluplus® with supercritical carbon dioxide allows promising applications in preparing dispersions of amorphous solids. Several characterization techniques were employed to reveal this effect, including CO2 gas sorption under high pressure and physicochemical characterizations techniques. A gravimetric method was used to determine the solubility of carbon dioxide in the polymer at elevated pressure. The following physicochemical characterizations were used: thermal analysis, X-ray diffraction, Fourier transform, infrared spectroscopy and scanning electron microscopy. Drug loading of the polymer with ibuprofen as a model drug was also investigated. The proposed treatment with supercritical carbon dioxide allows to prepare solid solutions of Soluplus® in less than two hours at temperatures that do not exceed 45 °C, which is a great advantage to be used for thermolabile drugs. The advantages of using this technology for Soluplus® formulations lies behind the high sorption capability of carbon dioxide inside the polymer. This will ensure rapid diffusion of the dissolved/dispersed drug inside the polymer under process conditions and rapid precipitation of the drug in the amorphous form during depressurization accompanied by foaming of the polymer.
Collapse
Affiliation(s)
- Rana Obaidat
- a Pharmaceutical Technology , Jordan University of Science and Technology , Irbid , Jordan
| | | | - Philip Jaeger
- c Technische Universitat Hamburg-Harburg , Hamburg , Germany
| |
Collapse
|
37
|
Design of spray dried ternary solid dispersions comprising itraconazole, soluplus and HPMCP: Effect of constituent compositions. Int J Pharm 2017; 519:365-372. [DOI: 10.1016/j.ijpharm.2017.01.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/19/2017] [Accepted: 01/21/2017] [Indexed: 10/20/2022]
|