1
|
Pereira M, Vale N. Ritonavir's Evolving Role: A Journey from Antiretroviral Therapy to Broader Medical Applications. Curr Oncol 2024; 31:6032-6049. [PMID: 39451754 PMCID: PMC11505664 DOI: 10.3390/curroncol31100450] [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: 08/31/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
Ritonavir is a protease inhibitor initially developed for HIV treatment that is now used as a pharmacokinetic booster for other antiretrovirals due to it being a cytochrome P450 3A4 enzyme and P-glycoprotein inhibitor. Consequently, ritonavir is of special interest for repurposing in other diseases. It had an important role in battling the COVID-19 pandemic as a part of the developed drug Paxlovid® in association with nirmatrelvir and has shown effects in hepatitis and other pathogenic diseases. Ritonavir has also shown promising results in overcoming drug resistance and enhancing the efficacy of existing chemotherapeutic agents in oncology. Evidence of cancer repurposing potential was demonstrated in cancers such as ovarian, prostate, lung, myeloma, breast, and bladder cancer, with several mechanisms of action presented. In vitro studies indicate that ritonavir alone can inhibit key pathways involved in cancer cell survival and proliferation, causing apoptosis, cell cycle arrest, endoplasmic reticulum stress, and metabolic stress due to the inhibition of molecules like heat shock protein 90 and cyclin-dependent kinases. Ritonavir also causes resistant cells to become sensitized to anticancer drugs like gemcitabine or docetaxel. These findings indicate that repurposing ritonavir, either on its own or in combination with other medications, could be a promising approach for treating various diseases. This is particularly relevant in cancer therapy, where ritonavir repurposing is the central focus of this review.
Collapse
Affiliation(s)
- Mariana Pereira
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
2
|
Permeation enhancers loaded bilosomes for improved intestinal absorption and cytotoxic activity of doxorubicin. Int J Pharm 2022; 630:122427. [PMID: 36435504 DOI: 10.1016/j.ijpharm.2022.122427] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/26/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
The clinical utility of doxorubicin is compromised due to dose related toxic side effects and limited oral bioavailability with no oral formulation being marketed. Enhancement of intestinal absorption and magnification of cytotoxicity can overcome these limitations. Accordingly, the objective was to probe penetration enhancers, bilosomes and their combinations for enhanced intestinal absorption and improved cytotoxicity of doxorubicin. Piperine and dipyridamole were tested as enhancers alone or encapsulated in bilosomes comprising Span60, cholesterol and bile salts. Bilosomes were nanosized spherical vesicles with negative zeta potential and were able to entrap doxorubicin with efficiency ranging from 45.3 % to 53 %. Intestinal absorption studies utilized in-situ rabbit intestinal perfusion which revealed site dependent doxorubicin absorption correlating with regional distribution of efflux transporters. Co-perfusion with the enhancer increased intestinal absorption with further augmentation after bilosomal encapsulation. The latter increased the % fraction absorbed by 4.5-6 and 1.8-2.5-fold from jejuno-ileum and colon, respectively, depending on bilosomes composition. Additionally, doxorubicin cytotoxicity against breast cancer cells (MCF-7) was significantly improved after bilosomal encapsulation and the recorded doxorubicin IC50 value was reduced from 13.3 μM to 0.1 μM for the best formulation. The study introduced bilosomes encapsulating absorption enhancers as promising carriers for enhanced cytotoxicity and oral absorption of doxorubicin.
Collapse
|
3
|
Rachmale M, Rajput N, Jadav T, Sahu AK, Tekade RK, Sengupta P. Implication of metabolomics and transporter modulation based strategies to minimize multidrug resistance and enhance site-specific bioavailability: a needful consideration toward modern anticancer drug discovery. Drug Metab Rev 2022; 54:101-119. [PMID: 35254954 DOI: 10.1080/03602532.2022.2048007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Induction of drug-metabolizing enzymes and efflux transporters (DMET) through activation of pregnane x receptor (PXR) is the primary factor involved in almost all bioavailability and drug resistance-related problems of anticancer drugs. PXR is a transcriptional regulator of many metabolizing enzymes and efflux transporters proteins like p-glycoprotein (p-gp), multidrug resistant protein 1 and 2 (MRP 1 and 2), and breast cancer resistant protein (BCRP), etc. Several anticancer drugs are potent activators of PXR receptors and can modulate the gene expression of DMET proteins. Involvement of anticancer drugs in transcriptional regulation of DMET can prompt increased metabolism and efflux of their own or other co-administered drugs, which leads to poor site-specific bioavailability and increased drug resistance. In this review, we have discussed several novel strategies to evade drug-induced PXR activation and p-gp efflux including assessment of PXR ligand and p-gp substrate at early stages of drug discovery. Additionally, we have critically discussed the chemical structure and drug delivery-based approaches to avoid PXR binding and inhibit the p-gp activity of the drugs at their target sites.
Collapse
Affiliation(s)
- Megha Rachmale
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Niraj Rajput
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Tarang Jadav
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Amit Kumar Sahu
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Pinaki Sengupta
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| |
Collapse
|
4
|
Luo T, Zhang FX, Zhao K, Gao HY, Zhang SG, Wang L, Dou GF, Liu T, Yu M, Zhan YQ, Chen H, Yang XM, Li CY. Preclinical Pharmacokinetics, Tissue Distribution, and Primary Safety Evaluation of Indo5, a Novel Selective Inhibitor of c-Met and Trks. Front Pharmacol 2021; 12:711126. [PMID: 34447310 PMCID: PMC8383318 DOI: 10.3389/fphar.2021.711126] [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: 05/18/2021] [Accepted: 07/30/2021] [Indexed: 11/17/2022] Open
Abstract
The compound [3-(1H-benzimidazol-2-methylene)-5-(2-methylphenylaminosulfo)-2-indolone], known as Indo5, is a novel selective inhibitor of c-Met and Trks, and it is a promising anticancer candidate against hepatocellular carcinoma (HCC). Assessing the pharmacokinetic properties, tissue distribution, and toxicity of Indo5 is critical for its medicinal evaluation. A series of sensitive and specific liquid chromatography-tandem mass spectrometry methods were developed and validated to determine the concentration of Indo5 in rat plasma and tissue homogenates. These methods were then applied to investigate the pharmacokinetics and tissue distribution of Indo5 in rats. After intravenous injection of Indo5, the maximum concentration (Cmax) and the time at which Cmax was reached (Tmax) were 1,565.3 ± 286.2 ng/ml and 1 min, respectively. After oral administration, Cmax and Tmax were 54.7 ± 10.4 ng/ml and 2.0 ± 0.48 h, respectively. We calculated the absolute oral bioavailability of Indo5 in rats to be 1.59%. Following intravenous injection, the concentrations of Indo5 in various tissues showed the following order: liver > kidney ≈ heart > lung ≈ large intestine ≈ small intestine ≈ stomach > spleen > brain ≈ testes; hence, Indo5 distributed highest in the liver and could not cross the blood–brain or blood–testes barriers. Continuous injection of Indo5 for 21 days did not lead to liver injury, considering unchanged ALT and AST levels, normal histological architecture of the liver, and normal number and frequencies of immune cells in the liver, indicating a very low toxicity of Indo5 in vivo. Collectively, our findings provide a comprehensive understanding of the biological actions of Indo5 in vivo and further support its development as an antitumor treatment for HCC patients.
Collapse
Affiliation(s)
- Teng Luo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Beijing Institute of Radiation Medicine, Beijing, China.,Institute of NBC Defence, Beijing, China
| | - Fei-Xiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | | | - Lin Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Gui-Fang Dou
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Ting Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Jackson CGCA, Hung T, Segelov E, Barlow P, Prenen H, McLaren B, Hung NA, Clarke K, Chao TY, Dai MS, Yeh HT, Cutler DL, Kramer D, He J, Zhi J, Chan WK, Kwan R, Deva S. Oral paclitaxel with encequidar compared to intravenous paclitaxel in patients with advanced cancer: A randomised crossover pharmacokinetic study. Br J Clin Pharmacol 2021; 87:4670-4680. [PMID: 33960504 DOI: 10.1111/bcp.14886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/25/2022] Open
Abstract
AIMS Paclitaxel is a widely used anti-neoplastic agent but has low oral bioavailability due to gut extrusion by P-glycoprotein (P-gp). Oral paclitaxel could be more convenient, less resource intensive, and more tolerable than intravenous administration. Encequidar (HM30181A) is a novel, minimally absorbed gut-specific P-gp inhibitor. We tested whether administration of oral paclitaxel with encequidar (oPac+E) achieved comparable AUC to intravenous paclitaxel (IVP) 80 mg/m2 . METHODS We conducted a multi-centre randomised crossover study with two treatment periods. Patients (pts) with advanced cancer received either oral paclitaxel 615 mg/m2 divided over 3 days and encequidar 15 mg orally 1 hour prior, followed by IVP 80 mg/m2 , or the reverse sequence. PK blood samples were taken up to Day 9 for oPac+E and Day 5 for IVP. RESULTS Forty-two patients were enrolled; 35 completed both treatment periods. AUC0-∞ was 5033.5 ± 1401.1 ng.h/mL for oPac+E and 5595.9 ± 1264.1 ng.h/mL with IVP. The geometric mean ratio (GMR) for AUC was 89.50% (90% CI 83.89-95.50). Mean absolute bioavailability of oPac+E was 12% (CV% = 23%). PK parameters did not change meaningfully after 4 weeks administration of oPac+E in an extension study. G3 treatment-emergent adverse events occurred in seven (18%) pts with oPac+E and two (5%) with IVP. Seventy-five per cent of patients preferred oPac+E over IVP. CONCLUSIONS GMR for AUC was within the predefined acceptable range of 80-125% for demonstrating equivalence. oPac+E is tolerable and there is no evidence of P-gp induction with repeat administration. With further study, oPac+E could be an alternative to IVP.
Collapse
Affiliation(s)
| | - Tak Hung
- Zenith Technology Corporation Limited, Dunedin, New Zealand
| | - Eva Segelov
- Monash University and Monash Health, Melbourne, Australia
| | - Paula Barlow
- Auckland District Health Board, Auckland, New Zealand
| | - Hans Prenen
- University Hospital Antwerp, Edegem, Belgium
| | - Blair McLaren
- Southern Blood and Cancer, Southern District Health Board, New Zealand
| | - Noelyn Anne Hung
- Department of Pathology, University of Otago, Dunedin, New Zealand
| | | | - Tsu-Yi Chao
- Taipei Medical University Shuang Ho Hospital, Taiwan
| | | | | | | | | | - Jimmy He
- Athenex Inc., Buffalo, NY, United States
| | - Jay Zhi
- Athenex Inc., Buffalo, NY, United States
| | | | | | - Sanjeev Deva
- Auckland District Health Board, Auckland, New Zealand
| |
Collapse
|
6
|
Shchekotikhin AE, Treshalina HM, Treshchalin MI, Pereverzeva ER, Isakova HB, Tikhomirov AS. Experimental Evaluation of Anticancer Efficiency and Acute Toxicity of Anthrafuran for Oral Administration. Pharmaceuticals (Basel) 2020; 13:ph13050081. [PMID: 32353946 PMCID: PMC7281648 DOI: 10.3390/ph13050081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/21/2020] [Accepted: 04/26/2020] [Indexed: 12/26/2022] Open
Abstract
The new antitumor agent anthrafuran has demonstrated a consistent effect in murine tumor models when administered parenterally due to the simultaneous inhibition of multiple cellular targets such as topoisomerases I/II and protein kinases. In this study, we assessed the anticancer efficiency and acute toxicity of anthrafuran administered orally. The action of anthrafuran was studied on transplanted tumor models which included P388 leukemia, Ca755 mammary adenocarcinoma, LLC lung carcinoma, and T47D human breast cancer xenografts on Balb/c nude mice. A significant antitumor efficacy of oral anthrafuran was revealed for all tested tumor models as follows: T/Cmax = 219% for P388, TGImax = 91% for Ca755, TGImax = 84% with CRmax = 54% for LLC, and T/C = 38% for T47D. The optimal treatment schedule of orally administered anthrafuran was 70–100 mg/kg given daily for five days. The LD50 value of orally administered anthrafuran (306.7 mg/kg) in mice was six times higher than that for i.p. administration (52.5 mg/kg). The rates of antitumor efficacy and acute toxicity indicate the high potential for further research on anthrafuran as a new original oral anticancer multitarget agent with an expected satisfactory tolerability and bioavailability.
Collapse
Affiliation(s)
- Andrey E. Shchekotikhin
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (M.I.T.); (E.R.P.); (H.B.I.); (A.S.T.)
- Correspondence:
| | - Helen M. Treshalina
- Federal State Budgetary Institution «National Medical Research Center of Oncology of N.N.Blokhin», Ministry of Health of Russia, 24 Kashirskoye sh., Moscow 115548, Russia;
| | - Michael I. Treshchalin
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (M.I.T.); (E.R.P.); (H.B.I.); (A.S.T.)
| | - Eleonora R. Pereverzeva
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (M.I.T.); (E.R.P.); (H.B.I.); (A.S.T.)
| | - Helen B. Isakova
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (M.I.T.); (E.R.P.); (H.B.I.); (A.S.T.)
| | - Alexander S. Tikhomirov
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (M.I.T.); (E.R.P.); (H.B.I.); (A.S.T.)
| |
Collapse
|
7
|
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
|
8
|
Abstract
Colorectal cancer is considered the third most frequent malignant neoplasm occurring in both men and women worldwide. Most approaches that have been used to fight and treat this type of malignancy are either invasive or nonselective. Noninvasive therapy using oral route can increase patient compliance and reduce treatment costs. Innovative measures such as use of nanotechnology and theranostic systems have been initiated in the oral therapy, which has been proven to be greatly advantageous in decreasing side effects, improving detection and diagnoses. This manuscript investigates recent innovative and novel therapeutic approaches through oral route and potential targets in the treatment of colorectal cancer.
Collapse
|
9
|
Rashid M, Malik MY, Singh SK, Chaturvedi S, Gayen JR, Wahajuddin M. Bioavailability Enhancement of Poorly Soluble Drugs: The Holy Grail in Pharma Industry. Curr Pharm Des 2019; 25:987-1020. [DOI: 10.2174/1381612825666190130110653] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/24/2019] [Indexed: 02/07/2023]
Abstract
Background:
Bioavailability, one of the prime pharmacokinetic properties of a drug, is defined as the
fraction of an administered dose of unchanged drug that reaches the systemic circulation and is used to describe
the systemic availability of a drug. Bioavailability assessment is imperative in order to demonstrate whether the
drug attains the desirable systemic exposure for effective therapy. In recent years, bioavailability has become
the subject of importance in drug discovery and development studies.
Methods:
A systematic literature review in the field of bioavailability and the approaches towards its enhancement
have been comprehensively done, purely focusing upon recent papers. The data mining was performed
using databases like PubMed, Science Direct and general Google searches and the collected data was exhaustively
studied and summarized in a generalized manner.
Results:
The main prospect of this review was to generate a comprehensive one-stop summary of the numerous
available approaches and their pharmaceutical applications in improving the stability concerns, physicochemical
and mechanical properties of the poorly water-soluble drugs which directly or indirectly augment their bioavailability.
Conclusion:
The use of novel methods, including but not limited to, nano-based formulations, bio-enhancers,
solid dispersions, lipid-and polymer-based formulations which provide a wide range of applications not only
increases the solubility and permeability of the poorly bioavailable drugs but also improves their stability, and
targeting efficacy. Although, these methods have drastically changed the pharmaceutical industry demand for the
newer potential methods with better outcomes in the field of pharmaceutical science to formulate various dosage
forms with adequate systemic availability and improved patient compliance, further research is required.
Collapse
Affiliation(s)
- Mamunur Rashid
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Mohd Yaseen Malik
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Sandeep K. Singh
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Swati Chaturvedi
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Jiaur R Gayen
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | | |
Collapse
|
10
|
Sohail MF, Rehman M, Sarwar HS, Naveed S, Salman O, Bukhari NI, Hussain I, Webster TJ, Shahnaz G. Advancements in the oral delivery of Docetaxel: challenges, current state-of-the-art and future trends. Int J Nanomedicine 2018; 13:3145-3161. [PMID: 29922053 PMCID: PMC5997133 DOI: 10.2147/ijn.s164518] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The oral delivery of cancer chemotherapeutic drugs is challenging due to low bioavailability, gastrointestinal side effects, first-pass metabolism and P-glycoprotein efflux pumps. Thus, chemotherapeutic drugs, including Docetaxel, are administered via an intravenous route, which poses many disadvantages of its own. Recent advances in pharmaceutical research have focused on designing new and efficient drug delivery systems for site-specific targeting, thus leading to improved bioavailability and pharmacokinetics. A decent number of studies have been reported for the safe and effective oral delivery of Docetaxel. These nanocarriers, including liposomes, polymeric nanoparticles, metallic nanoparticles, hybrid nanoparticles, dendrimers and so on, have shown promising results in research papers and clinical trials. The present article comprehensively reviews the research efforts made so far in designing various advancements in the oral delivery of Docetaxel. Different strategies to improve oral bioavailability, prevent first-pass metabolism and inhibition of efflux pumping leading to improved pharmacokinetics and anticancer activity are discussed. The final portion of this review article presents key issues such as safety of nanomaterials, regulatory approval and future trends in nanomedicine research.
Collapse
Affiliation(s)
- Muhammad Farhan Sohail
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad
- Department of Chemistry, SBA School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore
| | - Mubashar Rehman
- Department of Pharmacy, University of Lahore-Gujrat Campus, Gujrat
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Hafiz Shoaib Sarwar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad
| | - Sara Naveed
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore
| | - Omer Salman
- Department of Pharmacy, University of Lahore, Lahore Campus
| | - Nadeem Irfan Bukhari
- University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, Lahore, Pakistan
| | - Irshad Hussain
- Department of Chemistry, SBA School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Gul Shahnaz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad
| |
Collapse
|
11
|
Jebali R, Ben Salah-Abbès J, Abbès S, Hassan AM, Abdel-Aziem SH, El-Nekeety AA, Oueslati R, Abdel-Wahhab MA. Lactobacillus plantarum alleviate aflatoxins (B 1 and M 1 ) induced disturbances in the intestinal genes expression and DNA fragmentation in mice. Toxicon 2018; 146:13-23. [PMID: 29574215 DOI: 10.1016/j.toxicon.2018.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/07/2018] [Accepted: 03/20/2018] [Indexed: 01/14/2023]
|
12
|
Patel HH, Maniar M, Ren C, Dave RH. Determination of Degradation Kinetics and Effect of Anion Exchange Resin on Dissolution of Novel Anticancer Drug Rigosertib in Acidic Conditions. AAPS PharmSciTech 2018; 19:93-100. [PMID: 28600665 DOI: 10.1208/s12249-017-0820-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023] Open
Abstract
Rigosertib is a novel anticancer drug in clinical development by Onconova therapeutics, Inc. Currently, it is in pivotal phase III clinical trials for myelodysplastic syndrome (MDS) patients. Chemically, it is a sodium salt of weak acid with low solubility in lower pH solutions. In the preliminary studies, it was found that rigosertib is unstable in acidic conditions and forms multiple degradation products. In this research, drug degradation kinetics of rigosertib were studied in acidic conditions. Rigosertib follows pseudo-first-order general acid catalysis reaction. Cholestyramine, which is a strong anion exchange resin, was used to form complex with drug to improve stability and dissolution in acidic conditions. Drug complex with cholestyramine showed better dissolution profile compared to drug alone. Effect of polyethylene glycol was investigated on the release of drug from the drug resin complex. Polyethylene glycol further improved dissolution profile by improving drug solubility in acidic medium.
Collapse
|
13
|
Gou J, Feng S, Liang Y, Fang G, Zhang H, Yin T, Zhang Y, He H, Wang Y, Tang X. Polyester-Solid Lipid Mixed Nanoparticles with Improved Stability in Gastro-Intestinal Tract Facilitated Oral Delivery of Larotaxel. Mol Pharm 2017; 14:3750-3761. [PMID: 28945434 DOI: 10.1021/acs.molpharmaceut.7b00503] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The objective of this study was to investigate the role of core stability of nanoparticles on their performances in oral drug delivery. Solid lipids (Geleol Mono and Diglycerides Nf) were incorporated into nanoparticles composed of mPEG-b-PCL by the dialysis method. The prepared solid lipid loaded nanoparticles were found to be spherical nanoparticles with a core state and size distribution dependent on the amount of solid lipid incorporated. The critical aggregation concentrations of lipid-loaded nanoparticles were determined using pyrene fluorescence. Then, the stability of block copolymer in nanoparticles with different solid lipid contents was studied in simulated gastric fluid and simulated intestinal fluid. Solid lipids were found to stabilize nanoparticle cores by improving not only the thermodynamic stability (lowered CAC) of the nanoparticle but also the chemical stability of the block copolymer in the gastrointestinal environment. The stability of the loaded drug (larotaxel, LTX) in nanoparticles with different solid lipid contents was challenged by intestinal homogenate and rat liver microsome, and solid lipid loaded nanoparticles showed superior drug-protecting capability. Solid lipid incorporation exhibited limited influence on the cytotoxicity and cellular uptake but improved the transcytosis of nanoparticles in Caco-2 monolayers. The results of pharmacokinetic study indicated that core stabilization was helpful in promoting oral larotaxel absorption as the absolute bioavailability of LTX delivered by solid lipid loaded nanoparticles was found to be 13.17%, compared with that by the lipid-free nanoparticles (6.264%) and LTX solution (2.435%). Additionally, the results of biodistribution study indicated relatively higher particle integrity of solid lipid loaded nanoparticles, shown by slower liver and spleen accumulation rate, compared with its lipid-free counterpart. Overall, incorporation of solid lipids made the nanoparticles more suitable for oral drug delivery.
Collapse
Affiliation(s)
- Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Shuangshuang Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Yuheng Liang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Guihua Fang
- School of Pharmacy, Nantong University , No. 19 Qixiu Road, Nantong, China , 226001
| | - Haotian Zhang
- Department of Pharmacology, School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Tian Yin
- Department of Wine, School of Functional Food and Wine, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Yanjiao Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University , No. 103 Wenhua Road, Shenyang, China , 110016
| |
Collapse
|
14
|
Hao T, Ling Y, Wu M, Shen Y, Gao Y, Liang S, Gao Y, Qian S. Enhanced oral bioavailability of docetaxel in rats combined with myricetin: In situ and in vivo evidences. Eur J Pharm Sci 2017; 101:71-79. [DOI: 10.1016/j.ejps.2017.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 01/07/2017] [Accepted: 02/05/2017] [Indexed: 12/27/2022]
|
15
|
Ghadi R, Dand N. BCS class IV drugs: Highly notorious candidates for formulation development. J Control Release 2017; 248:71-95. [PMID: 28088572 DOI: 10.1016/j.jconrel.2017.01.014] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/08/2017] [Indexed: 12/20/2022]
Abstract
BCS class IV drugs (e.g., amphotericin B, furosemide, acetazolamide, ritonavir, paclitaxel) exhibit many characteristics that are problematic for effective oral and per oral delivery. Some of the problems associated include low aqueous solubility, poor permeability, erratic and poor absorption, inter and intra subject variability and significant positive food effect which leads to low and variable bioavailability. Also, most of the class IV drugs are substrate for P-glycoprotein (low permeability) and substrate for CYP3A4 (extensive pre systemic metabolism) which further potentiates the problem of poor therapeutic potential of these drugs. A decade back, extreme examples of class IV compounds were an exception rather than the rule, yet today many drug candidates under development pipeline fall into this category. Formulation and development of an efficacious delivery system for BCS class IV drugs are herculean tasks for any formulator. The inherent hurdles posed by these drugs hamper their translation to actual market. The importance of the formulation composition and design to successful drug development is especially illustrated by the BCS class IV case. To be clinically effective these drugs require the development of a proper delivery system for both oral and per oral delivery. Ideal oral dosage forms should produce both a reasonably high bioavailability and low inter and intra subject variability in absorption. Also, ideal systems for BCS class IV should produce a therapeutic concentration of the drug at reasonable dose volumes for intravenous administration. This article highlights the various techniques and upcoming strategies which can be employed for the development of highly notorious BCS class IV drugs. Some of the techniques employed are lipid based delivery systems, polymer based nanocarriers, crystal engineering (nanocrystals and co-crystals), liquisolid technology, self-emulsifying solid dispersions and miscellaneous techniques addressing the P-gp efflux problem. The review also focuses on the roadblocks in the clinical development of the aforementioned strategies such as problems in scale up, manufacturing under cGMP guidelines, appropriate quality control tests, validation of various processes and variable therein etc. It also brings to forefront the current lack of regulatory guidelines which poses difficulties during preclinical and clinical testing for submission of NDA and subsequent marketing. Today, the pharmaceutical industry has as its disposal a series of reliable and scalable formulation strategies for BCS Class IV drugs. However, due to lack of understanding of the basic physical chemistry behind these strategies formulation development is still driven by trial and error.
Collapse
Affiliation(s)
- Rohan Ghadi
- IPDO, Innovation Plaza, Dr Reddy's Laboratories Ltd., Bachupally, Hyderabad, 500090, India.
| | - Neha Dand
- Department of Pharmaceutics, Bharati Vidyapeeth's College of Pharmacy, CBD Belapur, Navi Mumbai, 400064, India
| |
Collapse
|
16
|
Surampalli G, Satla M, Nanjwade BK, Patil PA. In vitro and in vivo effects of morin on the intestinal absorption and pharmacokinetics of olmesartan medoxomil solid dispersions. Drug Dev Ind Pharm 2016; 43:812-829. [PMID: 27487480 DOI: 10.1080/03639045.2016.1220569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE In-situ evaluation to corroborate morin effects on the intestinal absorption and pharmacokinetic behavior of freeze-dried OLM-loaded solid dispersions with Caco-2 and in-vivo studies Methods: Intestinal transport and absorption studies were examined by Caco-2 permeability, in-situ single pass perfusion and closed-loop models along with in-vivo pharmacokinetic studies to evaluate and confirm the effect of P-gp-mediated activity of morin. We evaluated the intestinal membrane damage in the presence of morin by measuring the release of protein and lactate dehydrogenase (LDH) followed by using qualitative and quantitative morphometric analysis to describe the surface characteristics of intestinal epithelium. RESULTS Morin showed the highest Peff value 13.8 ± 0.34 × 10-6 cm/s in jejunum than ileum (p < .01) at 100 µM with absorption enhancement of 1.31-fold together with enhanced (p < .01) secretory transport of 6.27 ± 0.27 × 10 -6 cm/s in Caco-2 monolayer cells. Our findings noticed 2.37 (in-situ); 2.39 (in-vivo) and 1.43 (in-situ); 1.36 (in-vivo) fold increase in AUC0-t with elevated Cmax and shortened Tmax for freeze-dried solid dispersion in the presence of morin as compared to pure OLM and freeze-dried solid dispersions without morin, respectively. CONCLUSIONS Our study demonstrated that increased solubilization through freeze-dried OLM-loaded solid dispersion together with efflux inhibition improved intestinal permeability to one system that might lead to novel solubilization and efflux pump inhibition as a novel alternative potential to increase oral absorption and bioavailability of OLM.
Collapse
Affiliation(s)
- Gurunath Surampalli
- a Department of Pharmacology and Toxicology , Vaagdevi Institute of Pharma Sciences , Telangana, Warangal , India
| | - Madhuchander Satla
- b Department of Obstetrics and Gynecology , Kakatiya Medical College , Warangal, Telangana , India
| | - Basavaraj K Nanjwade
- c Department of Pharmacy Practice , The Oxford College of Pharmacy , Bengaluru , Karnataka , India
| | - Paragouda A Patil
- d Department of Pharmacology , USM-KLE, International Medical Program , Belgaum , Karnataka , India
| |
Collapse
|
17
|
Meanwell NA. Improving Drug Design: An Update on Recent Applications of Efficiency Metrics, Strategies for Replacing Problematic Elements, and Compounds in Nontraditional Drug Space. Chem Res Toxicol 2016; 29:564-616. [DOI: 10.1021/acs.chemrestox.6b00043] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nicholas A. Meanwell
- Department of Discovery Chemistry, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut 06492, United States
| |
Collapse
|
18
|
Li J, Wu J, Zhang J, Wang Y, Fang L, Shen Q. Oral bioavailability and evaluation of docetaxel–nicotinamide complex loaded chitosan nanoparticles. RSC Adv 2016. [DOI: 10.1039/c5ra27590c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
DTX–NA/NPs, synergism of DTX–NA complex and positively charged chitosan nanoparticles, can considerably enhance oral bioavailability.
Collapse
Affiliation(s)
- Jing Li
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Jian Wu
- Department of Pharmaceutical Sciences
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Jun Zhang
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Yiyue Wang
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Liang Fang
- Department of Pharmaceutical Sciences
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Qi Shen
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| |
Collapse
|
19
|
Yu H, Hendrikx JJMA, Rottenberg S, Schellens JHM, Beijnen JH, Huitema ADR. Development of a Tumour Growth Inhibition Model to Elucidate the Effects of Ritonavir on Intratumoural Metabolism and Anti-tumour Effect of Docetaxel in a Mouse Model for Hereditary Breast Cancer. AAPS JOURNAL 2015; 18:362-71. [PMID: 26603889 DOI: 10.1208/s12248-015-9838-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/17/2015] [Indexed: 12/31/2022]
Abstract
In a mouse tumour model for hereditary breast cancer, we previously explored the anti-cancer effects of docetaxel, ritonavir and the combination of both and studied the effect of ritonavir on the intratumoural concentration of docetaxel. The objective of the current study was to apply pharmacokinetic (PK)-pharmacodynamic (PD) modelling on this previous study to further elucidate and quantify the effects of docetaxel when co-administered with ritonavir. PK models of docetaxel and ritonavir in plasma and in tumour were developed. The effect of ritonavir on docetaxel concentration in the systemic circulation of Cyp3a knock-out mice and in the implanted tumour (with inherent Cyp3a expression) was studied, respectively. Subsequently, we designed a tumour growth inhibition model that included the inhibitory effects of both docetaxel and ritonavir. Ritonavir decreased docetaxel systemic clearance with 8% (relative standard error 0.4%) in the co-treated group compared to that in the docetaxel only-treated group. The docetaxel concentration in tumour tissues was significantly increased by ritonavir with mean area under the concentration-time curve 2.5-fold higher when combined with ritonavir. Observed tumour volume profiles in mice could be properly described by the PK/PD model. In the co-treated group, the enhanced anti-tumour effect was mainly due to increased docetaxel tumour concentration; however, we demonstrated a small but significant anti-tumour effect of ritonavir addition (p value <0.001). In conclusion, we showed that the increased anti-tumour effect observed when docetaxel is combined with ritonavir is mainly caused by enhanced docetaxel tumour concentration and to a minor extent by a direct anti-tumour effect of ritonavir.
Collapse
Affiliation(s)
- Huixin Yu
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek/Slotervaart Hospital, Louwesweg 6, PO Box 90440, 1006 BK, Amsterdam, The Netherlands.
| | - Jeroen J M A Hendrikx
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek/Slotervaart Hospital, Louwesweg 6, PO Box 90440, 1006 BK, Amsterdam, The Netherlands.,Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sven Rottenberg
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jan H M Schellens
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek/Slotervaart Hospital, Louwesweg 6, PO Box 90440, 1006 BK, 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/Slotervaart Hospital, Louwesweg 6, PO Box 90440, 1006 BK, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Hendrikx JJMA, Lagas JS, Song JY, Rosing H, Schellens JHM, Beijnen JH, Rottenberg S, Schinkel AH. Ritonavir inhibits intratumoral docetaxel metabolism and enhances docetaxel antitumor activity in an immunocompetent mouse breast cancer model. Int J Cancer 2015; 138:758-69. [PMID: 26297509 DOI: 10.1002/ijc.29812] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/03/2015] [Indexed: 12/27/2022]
Abstract
Docetaxel (Taxotere(®)) is currently used intravenously as an anticancer agent and is primarily metabolized by Cytochrome P450 3A (CYP3A). The HIV protease inhibitor ritonavir, a strong CYP3A4 inhibitor, decreased first-pass metabolism of orally administered docetaxel. Anticancer effects of ritonavir itself have also been described. We here aimed to test whether ritonavir co-administration could decrease intratumoral metabolism of intravenously administered docetaxel and thus increase the antitumor activity of docetaxel in an orthotopic, immunocompetent mouse model for breast cancer. Spontaneously arising K14cre;Brca1(F/F) ;p53(F/F) mouse mammary tumors were orthotopically implanted in syngeneic mice lacking Cyp3a (Cyp3a(-/-)) to limit ritonavir effects on systemic docetaxel clearance. Over 3 weeks, docetaxel (20 mg/kg) was administered intravenously once weekly, with or without ritonavir (12.5 mg/kg) administered orally for 5 days per week. Untreated mice were used as control for tumor growth. Ritonavir treatment alone did not significantly affect the median time of survival (14 vs. 10 days). Median time of survival in docetaxel-treated mice was 54 days. Ritonavir co-treatment significantly increased this to 66 days, and substantially reduced relative average tumor size, without altering tumor histology. Concentrations of the major docetaxel metabolite M2 in tumor tissue were reduced by ritonavir co-administration, whereas tumor RNA expression of Cyp3a was unaltered. In this breast cancer model, we observed no direct antitumor effect of ritonavir alone, but we found enhanced efficacy of docetaxel treatment when combined with ritonavir. Our data, therefore, suggest that decreased docetaxel metabolism inside the tumor as a result of Cyp3a inhibition contributes to increased antitumor activity.
Collapse
Affiliation(s)
- Jeroen J M A Hendrikx
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jurjen S Lagas
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Department of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & 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 Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Sven Rottenberg
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alfred H Schinkel
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Raza K, Thotakura N, Kumar P, Joshi M, Bhushan S, Bhatia A, Kumar V, Malik R, Sharma G, Guru SK, Katare OP. C60-fullerenes for delivery of docetaxel to breast cancer cells: A promising approach for enhanced efficacy and better pharmacokinetic profile. Int J Pharm 2015; 495:551-559. [PMID: 26383841 DOI: 10.1016/j.ijpharm.2015.09.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 01/23/2023]
Abstract
Docetaxel has always attracted the researchers owing to its promises and challenges. Despite marked efficacy, concerns like poor aqueous solubility, lower bioavailability, poor tissue penetration and dose related side-effects offer further scope of research on docetaxel. The present study aims to explore the potential of C60-fullerenes in the delivery of docetaxel to cancerous cells. C60-fullerenes were carboxylated, acylated and conjugated with the drug. The chemical processes were monitored by UV, FT-IR and NMR spectroscopy. The conjugate was further characterized for drug loading, micromeritics, drug release, morphology and evaluated for in-vitro cytotoxicity, haemolysis and in-vivo pharmacokinetic profile. The developed nanoconstruct was able to enhance the bioavailability of docetaxel by 4.2 times and decrease the drug clearance by 50%. The developed system was able to control the drug release and was found to be compatible with erythrocytes. The cytotoxic potential on studied MCF-7 and MDA-MB231 cell lines was also enhanced by many folds, indicating marked promise in efficacy enhancement and dose reduction. The present findings are encouraging and offer a technique to enhance the delivery and efficacy potential of anticancer agents, especially belonging to BCS class IV.
Collapse
Affiliation(s)
- Kaisar Raza
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Ajmer, Rajasthan 305817, India.
| | - Nagarani Thotakura
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Ajmer, Rajasthan 305817, India
| | - Pramod Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Ajmer, Rajasthan 305817, India
| | - Mayank Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Ajmer, Rajasthan 305817, India
| | - Shashi Bhushan
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Amit Bhatia
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Lovely Faculty of Applied Medical Sciences, Lovely Professional University, Jalandhar, Punjab 144806, India
| | - Vipin Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Ajmer, Rajasthan 305817, India
| | - Ruchi Malik
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Ajmer, Rajasthan 305817, India
| | - Gajanand Sharma
- Division of Pharmaceutics, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Santosh Kumar Guru
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - O P Katare
- Division of Pharmaceutics, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| |
Collapse
|
22
|
Phase I and pharmacological study of pazopanib in combination with oral topotecan in patients with advanced solid tumours. Br J Cancer 2015; 113:706-15. [PMID: 26291057 PMCID: PMC4559826 DOI: 10.1038/bjc.2015.257] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/20/2015] [Accepted: 05/27/2015] [Indexed: 01/22/2023] Open
Abstract
Background: This phase I study evaluated the safety, tolerability, maximum tolerated dose (MTD) and pharmacokinetics of two dosing schedules of oral topotecan in combination with pazopanib in patients with advanced solid tumours. Methods: Stage I of this study was to determine whether there was an impact of pazopanib on topotecan exposure. In stage II, the MTD and safety profile of oral topotecan given weekly on days 1, 8 and 15 in a 28-day cycle; or daily-times-five on days 1–5 in a 21-day cycle, both in combination with daily pazopanib, were explored. Results: In total, 67 patients were enroled. Pazopanib co-administration caused a substantial increase in exposure to total topotecan (1.7-fold) compared with topotecan alone, which is considered clinically relevant. Topotecan had no effect on pazopanib concentrations. Safety findings were consistent with the known profile of both agents. There were three drug-related deaths, liver failure, tumour haemorrhage and myelosuppression. Two patients experienced dose-limiting toxicities (DLTs; hand–foot syndrome, myelosuppression and diarrhoea) on the weekly topotecan schedule and four patients experienced DLTs (myelosuppression) on the daily-times-five topotecan schedule. When combined with pazopanib, 800 mg daily, the recommended doses for oral topotecan are: 8 mg weekly and 2.5 mg daily-times-five. Seven of eight patients with partial response had platinum-resistant ovarian cancer. In addition, 54% of patients had stable disease with 22% stable for 6 months. Conclusions: Total topotecan exposure is 1.7-fold higher when co-administered with pazopanib. Both schedules of administration were tolerated and would permit further evaluation, especially the weekly schedule.
Collapse
|
23
|
Fang G, Tang B, Chao Y, Xu H, Gou J, Zhang Y, Xu H, Tang X. Cysteine-Functionalized Nanostructured Lipid Carriers for Oral Delivery of Docetaxel: A Permeability and Pharmacokinetic Study. Mol Pharm 2015; 12:2384-95. [DOI: 10.1021/acs.molpharmaceut.5b00081] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Guihua Fang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Bo Tang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yanhui Chao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Helin Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Jingxin Gou
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Hui Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xing Tang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
24
|
Zhang P, de Gooijer MC, Buil LCM, Beijnen JH, Li G, van Tellingen O. ABCB1 and ABCG2 restrict the brain penetration of a panel of novel EZH2-Inhibitors. Int J Cancer 2015; 137:2007-18. [PMID: 25868794 DOI: 10.1002/ijc.29566] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 03/25/2015] [Indexed: 12/18/2022]
Abstract
Enhancer of Zeste Homolog 2 (EZH2) has emerged as a promising therapeutic target for treatment of a broad spectrum of tumors including gliomas. We explored the interactions of five novel, structurally similar EZH2 inhibitors (EPZ005687, EPZ-6438, UNC1999, GSK343 and GSK126) with P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2). The compounds were screened by in vitro transwell assays and EPZ005687, EPZ-6438 and GSK126 were further tested in vivo using wild-type (WT), Abcb1 and/or Abcg2 knockout mice. All EZH2 inhibitors are transported by P-gp and BCRP, although in vitro the transporter affinity of GSK126 was obscured by very low membrane permeability. Both P-gp and Bcrp1 restrict the brain penetration of EPZ005687 and GSK126, whereas the brain accumulation of EPZ-6438 is limited by P-gp only and efflux of EPZ-6438 was completely abrogated by elacridar. Intriguingly, an unknown factor present in all knockout mouse strains causes EPZ005687 and EPZ-6438 retention in plasma relative to WT mice, a phenomenon not seen with GSK126. In WT mice, the GSK126 tissue-to-plasma ratio for all tissues is lower than for EPZ005687 or EPZ-6438. Moreover, the oral bioavailability of GSK126 is only 0.2% in WT mice, which increases to 14.4% in Abcb1;Abcg2 knockout mice. These results are likely due to poor membrane permeability and question the clinical usefulness of GSK126. Although all tested EZH2 inhibitors are substrates of P-gp and BCRP, restricting the brain penetration and potential utility for treatment of glioma, EPZ-6438 would be the most suitable candidate of this series.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, People's Republic of China.,Department of Bio-Pharmacy/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek), Amsterdam, The Netherlands
| | - Mark C de Gooijer
- Department of Bio-Pharmacy/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek), Amsterdam, The Netherlands
| | - Levi C M Buil
- Department of Bio-Pharmacy/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek), Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam, The Netherlands.,Division of Drug Toxicology, Faculty of Pharmacy, Utrecht University, Utrecht, The Netherlands
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, People's Republic of China
| | - Olaf van Tellingen
- Department of Bio-Pharmacy/Mouse Cancer Clinic, The Netherlands Cancer Institute (Antoni van Leeuwenhoek), Amsterdam, The Netherlands
| |
Collapse
|
25
|
Joyce H, McCann A, Clynes M, Larkin A. Influence of multidrug resistance and drug transport proteins on chemotherapy drug metabolism. Expert Opin Drug Metab Toxicol 2015; 11:795-809. [PMID: 25836015 DOI: 10.1517/17425255.2015.1028356] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Chemotherapy involving the use of anticancer drugs remains an important strategy in the overall management of patients with metastatic cancer. Acquisition of multidrug resistance remains a major impediment to successful chemotherapy. Drug transporters in cell membranes and intracellular drug metabolizing enzymes contribute to the resistance phenotype and determine the pharmacokinetics of anticancer drugs in the body. AREAS COVERED ATP-binding cassette (ABC) transporters mediate the transport of endogenous metabolites and xenobiotics including cytotoxic drugs out of cells. Solute carrier (SLC) transporters mediate the influx of cytotoxic drugs into cells. This review focuses on the substrate interaction of these transporters, on their biology and what role they play together with drug metabolizing enzymes in eliminating therapeutic drugs from cells. EXPERT OPINION The majority of anticancer drugs are substrates for the ABC transporter and SLC transporter families. Together, these proteins have the ability to control the influx and the efflux of structurally unrelated chemotherapeutic drugs, thereby modulating the intracellular drug concentration. These interactions have important clinical implications for chemotherapy because ultimately they determine therapeutic efficacy, disease progression/relapse and the success or failure of patient treatment.
Collapse
Affiliation(s)
- Helena Joyce
- Dublin City University, National Institute for Cellular Biotechnology (NICB) , Glasnevin, Dublin 9 , Ireland +353 1 7005700 ; +353 1 7005484 ;
| | | | | | | |
Collapse
|
26
|
Durmus S, Hendrikx JJMA, Schinkel AH. Apical ABC transporters and cancer chemotherapeutic drug disposition. Adv Cancer Res 2015; 125:1-41. [PMID: 25640265 DOI: 10.1016/bs.acr.2014.10.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ATP-binding cassette (ABC) transporters are transmembrane efflux transporters that mediate cellular extrusion of a broad range of substrates ranging from amino acids, lipids, and ions to xenobiotics including many anticancer drugs. ABCB1 (P-GP) and ABCG2 (BCRP) are the most extensively studied apical ABC drug efflux transporters. They are highly expressed in apical membranes of many pharmacokinetically relevant tissues such as epithelial cells of the small intestine and endothelial cells of the blood capillaries in brain and testis, and in the placental maternal-fetal barrier. In these tissues, they have a protective function as they efflux their substrates back to the intestinal lumen or blood and thus restrict the intestinal uptake and tissue disposition of many compounds. This presents a major challenge for the use of many (anticancer) drugs, as most currently used anticancer drugs are substrates of these transporters. Herein, we review the latest findings on the role of apical ABC transporters in the disposition of anticancer drugs. We discuss that many new, rationally designed anticancer drugs are substrates of these transporters and that their oral availability and/or brain disposition are affected by this interaction. We also summarize studies that investigate the improvement of oral availability and brain disposition of many cytotoxic (e.g., taxanes) and rationally designed (e.g., tyrosine kinase inhibitor) anticancer drugs, using chemical inhibitors of these transporters. These findings provide a better understanding of the importance of apical ABC transporters in chemotherapy and may therefore advance translation of promising preclinical insights and approaches to clinical studies.
Collapse
Affiliation(s)
- Selvi Durmus
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeroen J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alfred H Schinkel
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Hendrikx JJMA, Lagas JS, Wagenaar E, Rosing H, Schellens JHM, Beijnen JH, Schinkel AH. Oral co-administration of elacridar and ritonavir enhances plasma levels of oral paclitaxel and docetaxel without affecting relative brain accumulation. Br J Cancer 2014; 110:2669-76. [PMID: 24781280 PMCID: PMC4037831 DOI: 10.1038/bjc.2014.222] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/19/2014] [Accepted: 03/27/2014] [Indexed: 11/09/2022] Open
Abstract
Background: The intestinal uptake of the taxanes paclitaxel and docetaxel is seriously hampered by drug efflux through P-glycoprotein (P-gp) and drug metabolism via cytochrome P450 (CYP) 3A. The resulting low oral bioavailability can be boosted by co-administration of P-gp or CYP3A4 inhibitors. Methods: Paclitaxel or docetaxel (10 mg/kg) was administered to CYP3A4-humanised mice after administration of the P-gp inhibitor elacridar (25 mg kg−1) and the CYP3A inhibitor ritonavir (12.5 mg kg−1). Plasma and brain concentrations of the taxanes were measured. Results: Oral co-administration of the taxanes with elacridar increased plasma concentrations of paclitaxel (10.7-fold, P<0.001) and docetaxel (four-fold, P<0.001). Co-administration with ritonavir resulted in 2.5-fold (paclitaxel, P<0.001) and 7.3-fold (docetaxel, P<0.001) increases in plasma concentrations. Co-administration with both inhibitors simultaneously resulted in further increased plasma concentrations of paclitaxel (31.9-fold, P<0.001) and docetaxel (37.4-fold, P<0.001). Although boosting of orally applied taxanes with elacridar and ritonavir potentially increases brain accumulation of taxanes, we found that only brain concentrations, but not brain-to-plasma ratios, were increased after co-administration with both inhibitors. Conclusions: The oral availability of taxanes can be enhanced by co-administration with oral elacridar and ritonavir, without increasing the brain penetration of the taxanes.
Collapse
Affiliation(s)
- J J M A Hendrikx
- 1] Department of Pharmacy and Pharmacology, Slotervaart Hospital, PO 90440, 1006 BK Amsterdam, The Netherlands [2] Division of Molecular Oncology, The Netherlands Cancer Institute, PO 90203, 1006 BE Amsterdam, The Netherlands
| | - J S Lagas
- Department of Pharmacy and Pharmacology, Slotervaart Hospital, PO 90440, 1006 BK Amsterdam, The Netherlands
| | - E Wagenaar
- Division of Molecular Oncology, The Netherlands Cancer Institute, PO 90203, 1006 BE Amsterdam, The Netherlands
| | - H Rosing
- Department of Pharmacy and Pharmacology, Slotervaart Hospital, PO 90440, 1006 BK Amsterdam, The Netherlands
| | - J H M Schellens
- 1] Department of Clinical Pharmacology, The Netherlands Cancer Institute, PO 90203, 1006 BE Amsterdam, The Netherlands [2] Department of Pharmaceutical Sciences, Utrecht University, PO 80082, 3508 TB Utrecht, The Netherlands
| | - J H Beijnen
- 1] Department of Pharmacy and Pharmacology, Slotervaart Hospital, PO 90440, 1006 BK Amsterdam, The Netherlands [2] Department of Pharmaceutical Sciences, Utrecht University, PO 80082, 3508 TB Utrecht, The Netherlands
| | - A H Schinkel
- Division of Molecular Oncology, The Netherlands Cancer Institute, PO 90203, 1006 BE Amsterdam, The Netherlands
| |
Collapse
|
28
|
Abstract
This chapter provides a review of the pharmacogenetics of membrane transporters, including ABC transporters and OATPs. Membrane transporters are heavily involved in drug disposition, by actively transporting substrate drugs between organs and tissues. As such, polymorphisms in the genes encoding these proteins may have a significant effect on the absorption, distribution, metabolism, excretion, and activity of compounds. Although few drug transporter polymorphisms have transitioned from the bench to the bedside, this chapter discusses clinical development of transporter pharmacogenetic markers. Finally, development of SLCO1B1 genotyping to avoid statin induced adverse drug reactions is discussed as a model case for transporter pharmacogenetics clinical development.
Collapse
|
29
|
Choi YH, Yu AM. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Curr Pharm Des 2014; 20:793-807. [PMID: 23688078 PMCID: PMC6341993 DOI: 10.2174/138161282005140214165212] [Citation(s) in RCA: 400] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/09/2013] [Indexed: 12/18/2022]
Abstract
Multidrug resistance (MDR) is a serious problem that hampers the success of cancer pharmacotherapy. A common mechanism is the overexpression of ATP-binding cassette (ABC) efflux transporters in cancer cells such as P-glycoprotein (P-gp/ABCB1), multidrug resistance-associated protein 1 (MRP1/ABCC1) and breast cancer resistance protein (BCRP/ABCG2) that limit the exposure to anticancer drugs. One way to overcome MDR is to develop ABC efflux transporter inhibitors to sensitize cancer cells to chemotherapeutic drugs. The complete clinical trials thus far have showen that those tested chemosensitizers only add limited or no benefits to cancer patients. Some MDR modulators are merely toxic, and others induce unwanted drug-drug interactions. Actually, many ABC transporters are also expressed abundantly in the gastrointestinal tract, liver, kidney, brain and other normal tissues, and they largely determine drug absorption, distribution and excretion, and affect the overall pharmacokinetic properties of drugs in humans. In addition, ABC transporters such as P-gp, MRP1 and BCRP co-expressed in tumors show a broad and overlapped specificity for substrates and MDR modulators. Thus reliable preclinical assays and models are required for the assessment of transporter-mediated flux and potential effects on pharmacokinetics in drug development. In this review, we provide an overview of the role of ABC efflux transporters in MDR and pharmacokinetics. Preclinical assays for the assessment of drug transport and development of MDR modulators are also discussed.
Collapse
Affiliation(s)
| | - Ai-Ming Yu
- Biochemistry & Molecular Medicine, UC Davis Medical Center, 2700 Stockton Blvd., Suite 2132, Sacramento, CA 95817, USA.
| |
Collapse
|
30
|
Jibodh RA, Lagas JS, Nuijen B, Beijnen JH, Schellens JH. Taxanes: Old drugs, new oral formulations. Eur J Pharmacol 2013; 717:40-6. [DOI: 10.1016/j.ejphar.2013.02.058] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 01/30/2013] [Accepted: 02/05/2013] [Indexed: 11/30/2022]
|
31
|
He B, Jia Z, Du W, Yu C, Fan Y, Dai W, Yuan L, Zhang H, Wang X, Wang J, Zhang X, Zhang Q. The transport pathways of polymer nanoparticles in MDCK epithelial cells. Biomaterials 2013; 34:4309-26. [DOI: 10.1016/j.biomaterials.2013.01.100] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 01/30/2013] [Indexed: 12/18/2022]
|
32
|
Chen KJ, Chen WY, Chen X, Jia YM, Peng GQ, Chen L. Increased elimination of paclitaxel by magnesium isoglycyrrhizinate in epithelial ovarian cancer patients treated with paclitaxel plus cisplatin: a pilot clinical study. Eur J Drug Metab Pharmacokinet 2013; 39:25-31. [PMID: 23681836 DOI: 10.1007/s13318-013-0136-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 05/03/2013] [Indexed: 01/20/2023]
Abstract
Magnesium isoglycyrrhizinate (MI) has been complementarily used for restoring the hepatic impairments caused by taxol plus platinum based chemotherapies in China. Due to the hepatic dependence of paclitaxel elimination, this pilot clinical study aimed to investigate the influence of MI on the pharmacokinetics of paclitaxel in epithelial ovarian cancer patients. During the standard chemotherapy of intravenous paclitaxel (125 mg/m(2) infused over a 3-h period) and intraperitoneal cisplatin (60 mg/m(2)) for patients with FIGO stage II epithelial ovarian cancer, 9 each of total 18 patients were respectively treated with intravenous MI (100 mg) or vehicle control for 4 days. Plasma paclitaxel was analyzed by HPLC and the pharmacokinetic parameters were calculated with non-compartmental analysis. The hematological, hepatic and renal status was monitored before and 3 days after paclitaxel administration. It was observed the terminal t 1/2 and MRT of paclitaxel were significantly (p = 0.002 and 0.001) reduced by MI, respectively, from 11.0 ± 2.2 and 5.6 ± 1.0 h to 7.7 ± 1.7 and 4.0 ± 0.3 h. Hematological toxicity indicated by platelet count and hepatic events marked with ALT, AST and γ-GT were significant in both groups. In spite of the insignificance of decreased system exposure of paclitaxel and recovered hepatic function by MI, they did correlate with each other. It was therefore deduced that the liver toxicities of paclitaxel plus cisplatin chemotherapy potentially decrease hepatic elimination and increase system exposure of paclitaxel, and the recovery of liver function by MI helps to restore hepatic clearance of paclitaxel. The clinical significance of this pharmacokinetic interaction requires further studies with larger population size.
Collapse
Affiliation(s)
- Kai Jie Chen
- Department of Pharmacy, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, China
| | | | | | | | | | | |
Collapse
|
33
|
Kim JE, Cho HJ, Kim JS, Shim CK, Chung SJ, Oak MH, Yoon IS, Kim DD. The limited intestinal absorption via paracellular pathway is responsible for the low oral bioavailability of doxorubicin. Xenobiotica 2012; 43:579-91. [DOI: 10.3109/00498254.2012.751140] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Liu JY, Lee KF, Sze CW, Tong Y, Tang SCW, Ng TB, Zhang YB. Intestinal absorption and bioavailability of traditional Chinese medicines: a review of recent experimental progress and implication for quality control. J Pharm Pharmacol 2012; 65:621-33. [DOI: 10.1111/j.2042-7158.2012.01608.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 09/28/2012] [Indexed: 12/18/2022]
Abstract
Abstract
Objectives
Experimental studies on the pharmacokinetics of traditional Chinese medicines (TCMs) have achieved great progress in recent years. This review aims to summarize the progress made on intestinal absorption and bioavailability of TCMs, and proposes the application of intestinal absorption assays as new tools for the quality and safety control of these medicines.
Key findings
Since only the absorbed constituents may produce possible therapeutic effect (except those that directly target the digestive tract), intestinal absorption is of utmost importance for the drug action of TCMs, which are usually taken orally. Meanwhile, complicated drug interactions may occur among the multiple ingredients in a herbal mixture. In this regard, the intestinal permeability assays not only provide useful pharmacokinetic data of TCMs, but have potential applications for quality and safety control. Moreover, knockout animals, 2/4/A1 in-vitro cell model and physiologically-based in-silico models based on the online TCM database can be quite useful for the prediction of absorption and bioavailability of TCMs.
Summary
A variety of in-vivo, in-vitro, in-situ and in-silico models for predicting the intestinal absorption and bioavailability can be applied to study the herbal interactions and screen appropriate biomarkers for the quality and safety control of TCMs.
Collapse
Affiliation(s)
- Jing-Yi Liu
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong, China
| | - Cho-Wing Sze
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Yao Tong
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Sydney Chi-Wai Tang
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tzi-Bun Ng
- The School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan-Bo Zhang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
35
|
Zee YK, Goh BC, Lee SC. Pharmacologic modulation strategies to reduce dose requirements of anticancer therapy while preserving clinical efficacy. Future Oncol 2012; 8:731-49. [PMID: 22764771 DOI: 10.2217/fon.12.53] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Drug interactions may be exploited to overcome pharmacokinetic issues in order to improve the therapeutic index of a drug, with clinical goals of reducing the dose of the active drug while preserving efficacy or reducing toxicity. This strategy has been used in infectious disease and transplant medicine, and, more recently, in oncology. Pharmacologic modulation strategies range from coadministration of either a drug that inhibits a metabolizing enzyme that would inactivate the drug of interest, a drug that induces an enzyme that activates the drug of interest or a drug that inhibits transporters that affect the uptake or elimination of the drug of interest. This review will discuss pharmacologic modulation strategies that have been tested clinically in order to increase systemic drug exposure. Important examples include ketoconazole inhibition of hepatic CYP3A4 in order to increase systemic exposure to docetaxel, irinotecan and etoposide, and cyclosporine inhibition of intestinal ATP-binding cassette transporters in order to decrease the toxicity of irinotecan and increase the bioavailability of oral docetaxel, paclitaxel and topotecan.
Collapse
Affiliation(s)
- Ying-Kiat Zee
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | | | | |
Collapse
|
36
|
Hendrikx JJ, Lagas JS, Rosing H, Schellens JH, Beijnen JH, Schinkel AH. P-glycoprotein and cytochrome P450 3A act together in restricting the oral bioavailability of paclitaxel. Int J Cancer 2012; 132:2439-47. [DOI: 10.1002/ijc.27912] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 10/10/2012] [Indexed: 12/22/2022]
|
37
|
Roger E, Kalscheuer S, Kirtane A, Guru BR, Grill AE, Whittum-Hudson J, Panyam J. Folic acid functionalized nanoparticles for enhanced oral drug delivery. Mol Pharm 2012; 9:2103-10. [PMID: 22670575 DOI: 10.1021/mp2005388] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The oral absorption of drugs that have poor bioavailability can be enhanced by encapsulation in polymeric nanoparticles. Transcellular transport of nanoparticle-encapsulated drug, possibly through transcytosis, is likely the major mechanism through which nanoparticles improve drug absorption. We hypothesized that the cellular uptake and transport of nanoparticles can be further increased by targeting the folate receptors expressed on the intestinal epithelial cells. The objective of this research was to study the effect of folic acid functionalization on transcellular transport of nanoparticle-encapsulated paclitaxel, a chemotherapeutic with poor oral bioavailability. Surface-functionalized poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles loaded with paclitaxel were prepared by the interfacial activity assisted surface functionalization technique. Transport of paclitaxel-loaded nanoparticles was investigated using Caco-2 cell monolayers as an in vitro model. Caco-2 cells were found to express folate receptor and the drug efflux protein, p-glycoprotein, to high levels. Encapsulation of paclitaxel in PLGA nanoparticles resulted in a 5-fold increase in apparent permeability (Papp) across Caco-2 cells. Functionalization of nanoparticles with folic acid further increased the transport (8-fold higher transport compared to free paclitaxel). Confocal microscopic studies showed that folic acid functionalized nanoparticles were internalized by the cells and that nanoparticles did not have any gross effects on tight junction integrity. In conclusion, our studies indicate that folic acid functionalized nanoparticles have the potential to enhance the oral absorption of drugs with poor oral bioavailability.
Collapse
Affiliation(s)
- Emilie Roger
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | | | | | | | | | | |
Collapse
|
38
|
Huang K, Hu J, Li X, Li Y. Role of P-Glycoprotein in Intestinal Absorption of FB2, a Promising Abl/Src Dual Tyrosine Kinase Inhibitor. Drug Metab Pharmacokinet 2012; 27:486-94. [DOI: 10.2133/dmpk.dmpk-11-rg-142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Hendrikx JJ, Hillebrand MJ, Thijssen B, Rosing H, Schinkel AH, Schellens JH, Beijnen JH. A sensitive combined assay for the quantification of paclitaxel, docetaxel and ritonavir in human plasma using liquid chromatography coupled with tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879:2984-90. [DOI: 10.1016/j.jchromb.2011.08.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 08/24/2011] [Accepted: 08/26/2011] [Indexed: 10/17/2022]
|
40
|
|
41
|
The role of diet on the clinical pharmacology of oral antineoplastic agents. Eur J Clin Pharmacol 2011; 68:115-22. [PMID: 21796375 DOI: 10.1007/s00228-011-1102-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 07/12/2011] [Indexed: 01/27/2023]
|
42
|
Agüeros M, Espuelas S, Esparza I, Calleja P, Peñuelas I, Ponchel G, Irache JM. Cyclodextrin-poly(anhydride) nanoparticles as new vehicles for oral drug delivery. Expert Opin Drug Deliv 2011; 8:721-34. [DOI: 10.1517/17425247.2011.572069] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Koolen SLW, Oostendorp RL, Beijnen JH, Schellens JHM, Huitema ADR. Population pharmacokinetics of intravenously and orally administered docetaxel with or without co-administration of ritonavir in patients with advanced cancer. Br J Clin Pharmacol 2011; 69:465-74. [PMID: 20573082 DOI: 10.1111/j.1365-2125.2010.03621.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
AIM Docetaxel has a low oral bioavailability due to affinity for P-glycoprotein and cytochrome P450 (CYP) 3A4 enzymes. Inhibition of the CYP3A4 enzymes by ritonavir resulted in increased oral bioavailability. The aim of this study was to develop a population pharmacokinetic (PK) model and to evaluate and quantify the influence of ritonavir on the PK of docetaxel. METHODS Data from two clinical trials were included in the data analysis, in which docetaxel (75 mg m(-2) or 100 mg) had been administered intravenously or orally (10 mg or 100 mg) with or without co-administration of oral ritonavir (100 mg). Population modelling was performed using non-linear mixed effects modelling. A three-compartment model was used to describe the i.v. data. PK data after oral administration, with or without co-administration of ritonavir, were incorporated into the model. RESULTS Gut bioavailability of docetaxel increased approximately two-fold from 19 to 39% (CV 13%) with ritonavir co-administration. The hepatic extraction ratio and the elimination rate of docetaxel were best described by estimating the intrinsic clearance. Ritonavir was found to inhibit in a concentration dependent manner the intrinsic clearance of docetaxel, which was described by an inhibition constant of 0.028 microg ml(-1) (CV 36%). A maximum inhibition of docetaxel clearance of more then 90% was reached. CONCLUSIONS A PK model describing both the PK of orally and intravenously administered docetaxel in combination with ritonavir, was successfully developed. Co-administration of ritonavir lead to increased oral absorption and reduced elimination rate of docetaxel.
Collapse
Affiliation(s)
- Stijn L W Koolen
- Department of Pharmacy & Pharmacology, the Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam, the Netherlands.
| | | | | | | | | |
Collapse
|
44
|
Nassar T, Attili-Qadri S, Harush-Frenkel O, Farber S, Lecht S, Lazarovici P, Benita S. High plasma levels and effective lymphatic uptake of docetaxel in an orally available nanotransporter formulation. Cancer Res 2011; 71:3018-28. [PMID: 21363913 DOI: 10.1158/0008-5472.can-10-3118] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Docetaxel, an efficient chemotherapeutic drug, exhibits low and variable oral bioavailability due to the active efflux by P-glycoprotein (P-gp) and more so to CYP3A4 gut metabolism. Using a spray-drying technique, docetaxel was incorporated in PLGA [poly(lactic-co-glycolic acid)] nanocapsules (NC) which were embedded in entero-coated microparticles. An oral administration of the NC formulation elicited a higher absolute bioavailability than both a docetaxel solution (276%) and a free docetaxel NC formulation (400%) injected intravenously, a 5-mg/kg dose. The batches (B) I and II NC formulations elicited C(max) values that were 1,735% and 2,254%, respectively; higher than the C(max) value of the oral docetaxel solution combined with blank microparticles, a 10-mg/kg dose. No significant difference in AUC (area under curve) was observed between the batches. These unexpected results can be explained only if the pharmacokinetics of docetaxel had been modified. It was shown that NCs released from the microparticles penetrated the enterocytes, bypassing P-gp; apparently circumventing gut metabolism and accumulating within the lymphatic system from where both intact or biodegraded NCs and free docetaxel were progressively released into the circulation as plausibly supported by the fluorescent imaging results. Furthermore, the circulating docetaxel in plasma was unencapsulated and circulated either in free form or bound to albumin. Both free docetaxel NCs and microparticles exhibited in vitro efficacy on WRC 256 cells suggesting that the activity of docetaxel was not altered. This delivery concept has potential for clinical translation, perhaps allowing docetaxel chemotherapy to be switched from intravenous to oral delivery.
Collapse
Affiliation(s)
- Taher Nassar
- Faculty of Medicine, The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
45
|
Khan F, Katara R, Ramteke S. Enhancement of bioavailability of cefpodoxime proxetil using different polymeric microparticles. AAPS PharmSciTech 2010; 11:1368-75. [PMID: 20821175 PMCID: PMC2974107 DOI: 10.1208/s12249-010-9505-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 06/30/2010] [Indexed: 11/30/2022] Open
Abstract
Poorly water-soluble drugs such as cefpodoxime proxetil (400 μg/ml) offer a challenging problem in drug formulation as poor solubility is generally associated with poor dissolution characteristics and thus poor oral bioavailability. According to these characteristics, preparation of cefpodoxime proxetil microparticle has been achieved using high-speed homogenization. Polymers (methylcellulose, sodium alginate, and chitosan) were precipitated on the surface of cefpodoxime proxetil using sodium citrate and calcium chloride as salting-out agents. The pure drug and the prepared microparticles with different concentrations of polymer (0.05-1.0%) were characterized in terms of solubility, drug content, particle size, thermal behavior (differential scanning calorimeter), surface morphology (scanning electron microscopy), in vitro drug release, and stability studies. The in vivo performance was assessed by pharmacokinetic study. The dissolution studies demonstrate a marked increase in the dissolution rate in comparison with pure drug. The considerable improvement in the dissolution rate of cefpodoxime proxetil from optimized microparticle was attributed to the wetting effect of polymers, altered surface morphology, and micronization of drug particles. The optimized microparticles exhibited excellent stability on storage at accelerated condition. The in vivo studies revealed that the optimized formulations provided improved pharmacokinetic parameter in rats as compared with pure drug. The particle size of drug was drastically reduced during formulation process of microparticles.
Collapse
Affiliation(s)
- Fahim Khan
- School of Pharmaceutical Sciences, Rajive Gandhi Technical University, Airport Bypass Road, Gandhi Nagar, Bhopal, Madhya Pradesh 462036 India
| | - Rajesh Katara
- School of Pharmaceutical Sciences, Rajive Gandhi Technical University, Airport Bypass Road, Gandhi Nagar, Bhopal, Madhya Pradesh 462036 India
| | - Suman Ramteke
- School of Pharmaceutical Sciences, Rajive Gandhi Technical University, Airport Bypass Road, Gandhi Nagar, Bhopal, Madhya Pradesh 462036 India
| |
Collapse
|
46
|
Heikkinen AT, Korjamo T, Lepikkö V, Mönkkönen J. Effects of experimental setup on the apparent concentration dependency of active efflux transport in in vitro cell permeation experiments. Mol Pharm 2010; 7:605-17. [PMID: 20163161 DOI: 10.1021/mp9003089] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
P-Glycoprotein mediated efflux is one of the barriers limiting drug absorption from the intestine. Predictions of the intestinal P-glycoprotein function need to take into account the concentration dependency because high intestinal drug concentrations may saturate P-glycoprotein. However, the substrate binding site of P-glycoprotein lies inside the cells and the drug concentration at the binding site cannot be measured directly. Therefore, rigorous determination of concentration dependent P-glycoprotein kinetics is challenging. In this study, the effects of the aqueous boundary layers, extracellular pH and cellular retention on the apparent saturation kinetics of P-glycoprotein mediated transport of quinidine in an in vitro cell permeation setting were explored. The changes in the experimental conditions caused 1 order of magnitude variation in the apparent affinity to P-glycoprotein (K(m,app)) and a 5-fold difference in the maximum effective P-glycoprotein mediated transport rate of quinidine (V(max,app)). However, fitting the concentration data into a compartmental model which accounted for the aqueous boundary layers, cell membranes and cellular retention suggested that the P-glycoprotein function per se was not altered, it was the differences in the passive transfer of quinidine which changed the apparent transport kinetics. These results provide further insight into the dynamics of the P-glycoprotein mediated transport and on the roles of several confounding factors involved in in vitro experimental setting. Further, the results confirm the applicability of compartmental model based data analysis approach in the determination of active transporter kinetics.
Collapse
Affiliation(s)
- Aki T Heikkinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| | | | | | | |
Collapse
|
47
|
Gnoth MJ, Sandmann S, Engel K, Radtke M. In vitro to in vivo comparison of the substrate characteristics of sorafenib tosylate toward P-glycoprotein. Drug Metab Dispos 2010; 38:1341-6. [PMID: 20413726 DOI: 10.1124/dmd.110.032052] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sorafenib (Nexavar) is a novel oral Raf kinase and vascular endothelial growth factor receptor inhibitor. Most anticancer drugs are substrates for ATP-binding cassette efflux pumps especially for P-glycoprotein (P-gp). To evaluate the influence of P-gp on the pharmacokinetics of sorafenib substrate properties for this transporter were investigated. Therefore, permeability of sorafenib across Caco-2 and P-gp-overexpressing cells was determined. To determine the in vivo relevance of these in vitro findings, pharmacokinetics of sorafenib in mdr1a/1b(-/-) and wild-type (WT) mice was studied. Sorafenib is highly permeable and exhibits a slight efflux across Caco-2 cells. In P-gp-overexpressing cells, a small concentration-dependent efflux was observed, which was completely blocked by the addition of ivermectin. In mdr1a/1b(-/-) and WT mice, unchanged compound represented by far the majority of radioactivity in plasma. After intravenous and oral administration, brain/plasma concentration ratios in mdr1a/1b(-/-) mice were 1.3- to 1.5-fold higher than those in WT mice. However, after intravenous or oral administration, plasma concentrations were similar in both mouse strains. In conclusion, sorafenib is highly permeable and a weak P-gp substrate in vitro. These findings were confirmed by the small factor of 1.3 to 1.5 observed for the brain/plasma ratios in mdr1a/1b(-/-) versus WT mice in vivo. Based on these in vitro and in vivo results, it is unlikely that P-gp has a major effect on the plasma concentrations of sorafenib in humans. Because of the high permeability and low P-gp-mediated transport, sorafenib might be able to cross the blood-brain barrier and target tumors within the brain.
Collapse
Affiliation(s)
- M J Gnoth
- Drug Metabolism and Pharmacokinetics, Bayer Schering Pharma AG, Wuppertal, Germany.
| | | | | | | |
Collapse
|
48
|
Kwan WS, Janneh O, Hartkoorn R, Chandler B, Khoo S, Back D, Owen A. Intracellular 'boosting' of darunavir using known transport inhibitors in primary PBMC. Br J Clin Pharmacol 2010; 68:375-80. [PMID: 19740394 DOI: 10.1111/j.1365-2125.2009.03462.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIMS ABCB1, some ABCCs and SLCOs have been reported to affect the intracellular accumulation of various protease inhibitors in vitro and ex vivo. Darunavir is the most recently licensed protease inhibitor and we sought to investigate the ability of transport inhibitors to influence its intracellular accumulation in lymphocytes from healthy volunteers. METHODS The intracellular accumulation of radiolabelled darunavir was assessed using CEM cells and ABCB1-overexpressing CEM(VBL) cells. Apical and basolateral transport of radiolabelled darunavir through MDCKII monolayers was also studied. Finally the ability of known inhibitors to influence intracellular accumulation of darunavir in peripheral blood mononuclear cells (PBMC) was investigated. RESULTS CEM(VBL) cells (1.4 +/- 0.6, P < 0.001, 95% CI for the difference = 0.46, 0.80, n= 7) had significantly lower accumulation of darunavir compared with CEM cells (5.6 +/- 0.7, n= 7) and this was reversed by addition of tariquidar (30 nm, 4.6 +/- 0.8, P < 0.001, 95% CI =-0.64, -0.41, n= 4). In MDCKII-ABCBI cells, transport from the basal to the apical compartment was observed and this was also reversible with the addition of tariquidar. In PBMCs, dipyridamole (6.9 +/- 1.3, P < 0.01, 95% CI for the difference =-1.16, -0.30, (n= 8) significantly increased whilst montelukast (5.7 +/- 1.0, P < 0.01, 95% CI for the difference = 0.16, 0.79, n= 8) significantly decreased the intracellular accumulation of darunavir when compared with control (6.2 +/- 1.1, n= 8). CONCLUSIONS Darunavir is a substrate for efflux and influx transporters in PBMC and intracellular concentrations can be manipulated using known inhibitors.
Collapse
Affiliation(s)
- Wai San Kwan
- Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | |
Collapse
|
49
|
Oostendorp RL, Buckle T, Lambert G, Garrigue JS, Beijnen JH, Schellens JHM, van Tellingen O. Paclitaxel in self-micro emulsifying formulations: oral bioavailability study in mice. Invest New Drugs 2010; 29:768-76. [PMID: 20390333 PMCID: PMC3160553 DOI: 10.1007/s10637-010-9421-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Accepted: 03/09/2010] [Indexed: 12/02/2022]
Abstract
The anticancer drug paclitaxel is formulated for i.v. administration in a mixture of Cremophor EL and ethanol. Its oral bioavailability is very low due to the action of P-glycoprotein in the gut wall and CYP450 in gut wall and liver. However, proof-of-concept studies using the i.v. formulation diluted in drinking water have demonstrated the feasibility of the oral route as an alternative when given in combination with inhibitors of P-glycoprotein and CYP450. Because of the unacceptable pharmaceutical properties of the drinking solution, a better formulation for oral application is needed. We have evaluated the suitability of various self-micro emulsifying oily formulations (SMEOF’s) of paclitaxel for oral application using wild-type and P-glycoprotein knockout mice and cyclosporin A (CsA) as P-glycoprotein and CYP450 inhibitor. The oral bioavailability of paclitaxel in all SMEOF’s without concomitant CsA was low in wild-type mice, showing that this vehicle does not enhance intestinal uptake by itself. Paclitaxel (10 mg/kg) in SMEOF#3 given with CsA resulted in plasma levels that were comparable to the Cremophor EL-ethanol containing drinking solution plus CsA. Whereas the AUC increased linearly with the oral paclitaxel dose in P-glycoprotein knockout mice, it increased less than proportional in wild-type mice given with CsA. In both strains more unchanged paclitaxel was recovered in the feces at higher doses. This observation most likely reflects more profound precipitation of paclitaxel within the gastro-intestinal tract at higher doses. The resulting absolute reduction in absorption of paclitaxel from the gut was possibly concealed by partial saturation of first-pass metabolism when P-glycoprotein was absent. In conclusion, SMEOF’s maybe a useful vehicle for oral delivery of paclitaxel in combination with CsA, although the physical stability within the gastro-intestinal tract remains a critical issue, especially when applied at higher dose levels.
Collapse
Affiliation(s)
- Roos L. Oostendorp
- Division of Experimental Therapy, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - T. Buckle
- Clinical Chemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - G. Lambert
- Novagali Pharma S.A., Batiment Genavir IV, 1 rue Pierre Fontaine, FR-91058 Evry Cedex, France
| | - J. S. Garrigue
- Novagali Pharma S.A., Batiment Genavir IV, 1 rue Pierre Fontaine, FR-91058 Evry Cedex, France
| | - J. H. Beijnen
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Biomedical Analysis, Utrecht University, Sorbonnelaan 16, 3584CA Utrecht, The Netherlands
- Depatment of Pharmacy, The Netherlands Cancer Institute, Louwesweg 6, 1066 EC Amsterdam, The Netherlands
| | - J. H. M. Schellens
- Division of Experimental Therapy, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Biomedical Analysis, Utrecht University, Sorbonnelaan 16, 3584CA Utrecht, The Netherlands
- Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - O. van Tellingen
- Clinical Chemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
50
|
Sissung TM, Baum CE, Kirkland CT, Gao R, Gardner ER, Figg WD. Pharmacogenetics of membrane transporters: an update on current approaches. Mol Biotechnol 2010; 44:152-67. [PMID: 19950006 PMCID: PMC6362991 DOI: 10.1007/s12033-009-9220-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This review provides an overview of the pharmacogenetics of membrane transporters including selected ABC transporters (ABCB1, ABCC1, ABCC2, and ABCG2) and OATPs (OATP1B1 and OATP1B3). Membrane transporters are heavily involved in drug clearance and alters drug disposition by actively transporting substrate drugs between organs and tissues. As such, polymorphisms in the genes encoding these proteins may have significant effects on the absorption, distribution, metabolism and excretion of compounds, and may alter pharmacodynamics of many agents. This review discusses the techniques used to identify substrates and inhibitors of these proteins and subsequently to assess the effect of genetic mutation on transport, both in vitro and in vivo. A comprehensive list of substrates for the major drug transporters is included. Finally, studies linking transporter genotype with clinical outcomes are discussed.
Collapse
Affiliation(s)
- Tristan M. Sissung
- Clinical Pharmacology Program, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Building 10, Room 5A01, Bethesda, MD 20892, USA,
| | - Caitlin E. Baum
- Molecular Pharmacology Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Building 10, Room 5A01, Bethesda, MD 20892, USA,
| | - C. Tyler Kirkland
- Molecular Pharmacology Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Building 10, Room 5A01, Bethesda, MD 20892, USA,
| | - Rui Gao
- Molecular Pharmacology Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Building 10, Room 5A01, Bethesda, MD 20892, USA,
| | - Erin R. Gardner
- Clinical Pharmacology Program, SAIC-Frederick, NCI-Frederick, Frederick, MD 21702, USA,
| | - William D. Figg
- Clinical Pharmacology Program, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Building 10, Room 5A01, Bethesda, MD 20892, USA; Molecular Pharmacology Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Building 10, Room 5A01, Bethesda, MD 20892, USA
| |
Collapse
|