Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
ACS Pharmacol Transl Sci ; 7(5): 1310-1319, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38751643

ABSTRACT

The value of connected devices and health apps with features such as adherence trackers, dosing reminders, and remote communication tools for users and healthcare providers has been assessed to support home-based subcutaneous administration. A comprehensive survey was conducted with 605 participants, including users and caregivers, from eight countries. Medical conditions encompassed ankylosing spondylitis, asthma, cerebral palsy, cluster headaches, Crohn's disease, hemophilia, lupus, migraine, multiple sclerosis, Parkinson's disease, plaque psoriasis, psoriatic arthritis, rheumatoid arthritis, spasticity, spondyloarthritis, and ulcerative colitis. Utilizing a maximum difference scaling methodology, the survey gauged participant preferences regarding specific attributes and features of connected drug delivery devices. Irrespective of demographic factors like age, gender, nationality, or the specific medical condition, the device's ability to verify a successful injection stood out as universally valued. The second and third most valued attributes pertained to temperature-related indicators or warnings. These features do not necessitate the use of a connected device and can be integrated into existing autoinjector platforms. The survey findings support the development of a universal adherence tool for at-home subcutaneous dosing, independent of a specific medical condition. This tool may be gradually improved with disease-specific features. Once established as a platform, manufacturers can launch any subcutaneous medication and later integrate real-world evidence for enhanced educational, treatment, and diagnostic capabilities. This approach is crucial for advancing connected adherence tools in decentralized healthcare, aligning with user and healthcare system needs while translating scientific innovation into practical solutions.

2.
BioDrugs ; 38(1): 23-46, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37831325

ABSTRACT

In recent years, subcutaneous administration of biotherapeutics has made significant progress. The self-administration market for rheumatoid arthritis has witnessed the introduction of additional follow-on biologics, while the first subcutaneous dosing options for monoclonal antibodies have become available for multiple sclerosis. Oncology has also seen advancements with the authorization of high-volume subcutaneous formulations, facilitated by the development of high-concentration formulations and innovative delivery systems. Regulatory and Health Technology Assessment bodies increasingly consider preference data in filing dossiers, particularly in evaluating novel drug delivery methods. The adoption of a pharmacokinetic-based clinical bridging approach has become standard for transitioning from intravenous to subcutaneous administration. Non-inferiority studies with pharmacokinetics as the only primary endpoint have started deviating from traditional randomization schemes, favoring the subcutaneous route and comparing with historical intravenous data. While nonclinical and computational models made progress in predicting safety and immunogenicity for subcutaneously dosed antibodies, clinical trial evidence remains essential due to inter-individual variations and the impact of formulation parameters on anti-drug antibody formation. Ongoing technological advancements and the expanding knowledge base on pharmacokinetic-pharmacodynamic correlation across specialty areas are expected to further accelerate clinical development of subcutaneous biologics.


Subject(s)
Arthritis, Rheumatoid , Biological Products , Multiple Sclerosis , Humans , Antibodies, Monoclonal/therapeutic use , Injections, Subcutaneous , Arthritis, Rheumatoid/drug therapy , Multiple Sclerosis/drug therapy
3.
J Control Release ; 360: 335-343, 2023 08.
Article in English | MEDLINE | ID: mdl-37364797

ABSTRACT

The way a drug molecule is administered has always had a profound impact on people requiring medical interventions - from vaccine development to cancer therapeutics. In the Controlled Release Society Fall Symposium 2022, a trans-institutional group of scientists from industry, academia, and non-governmental organizations discussed what a breakthrough in the field of drug delivery constitutes. On the basis of these discussions, we classified drug delivery breakthrough technologies into three categories. In category 1, drug delivery systems enable treatment for new molecular entities per se, for instance by overcoming biological barriers. In category 2, drug delivery systems optimize efficacy and/or safety of an existing drug, for instance by directing distribution to their target tissue, by replacing toxic excipients, or by changing the dosing reqimen. In category 3, drug delivery systems improve global access by fostering use in low-resource settings, for instance by facilitating drug administration outside of a controlled health care institutional setting. We recognize that certain breakthroughs can be classified in more than one category. It was concluded that in order to create a true breakthrough technology, multidisciplinary collaboration is mandated to move from pure technical inventions to true innovations addressing key current and emerging unmet health care needs.


Subject(s)
Drug Delivery Systems , Neoplasms , Humans , Pharmaceutical Preparations , Technology
4.
Expert Opin Drug Deliv ; 20(4): 457-470, 2023 04.
Article in English | MEDLINE | ID: mdl-36855292

ABSTRACT

INTRODUCTION: The substantial acceleration in healthcare spending together with the expenditures to manage the COVID19 pandemic demand drug delivery solutions that enable a flexible care setting for high-dose monoclonal antibodies (mAbs) in oncology. AREAS COVERED: This expert opinion introduces an analogue-based framework applied to guide decision-making for associated product improvements for mAb medications that are either already authorized or in late-stage clinical development. The four pillars of this framework comprise (1) the drug delivery profile of current and emerging treatments in the market, (2) the needs and preferences of people treated with mAbs, (3) existing healthcare infrastructures, and (4) country-dependent reimbursement and procurement models. The following product optimization examples for mAb-based treatments are evaluated based on original research and review articles in the field: subcutaneous formulations, an established drug delivery modality to reduce parenteral dosing complexity, fixed-dose combinations, an emerging concept to complement combination therapy, and (connected) on-body delivery systems, an identified future opportunity to support dosing outside of a controlled healthcare institutional environment. EXPERT OPINION: Leveraging existing synergies and learnings from other disease areas is a measure to reduce associated development and commercialization costs and thus to provide sustainable product offerings already at the initial launch of a medication.


Subject(s)
Antibodies, Monoclonal , COVID-19 , Humans , Antibodies, Monoclonal/therapeutic use , Delivery of Health Care , Drug Compounding , Subcutaneous Tissue
5.
AAPS Open ; 9(1): 3, 2023.
Article in English | MEDLINE | ID: mdl-36713112

ABSTRACT

Delivering customer-centric product presentations for biotherapeutics, such as monoclonal antibodies (mAbs), represents a long-standing and paramount area of engagement for pharmaceutical scientists. Activities include improving experience with the dosing procedure, reducing drug administration-related expenditures, and ultimately shifting parenteral treatments outside of a controlled healthcare institutional setting. In times of increasingly cost-constrained markets and reinforced with the coronavirus pandemic, this discipline of "Product Optimization" in healthcare has gained momentum and changed from a nice-to-have into a must. This review summarizes latest trends in the healthcare ecosystem that inform key strategies for developing customer-centric products, including the availability of a wider array of sustainable drug delivery options and treatment management plans that support dosing in a flexible care setting. Three disease area archetypes with varying degree of implementation of customer-centric concepts are introduced to highlight relevant market differences and similarities. Namely, rheumatoid arthritis and inflammatory bowel disease, multiple sclerosis, and oncology have been chosen due to differences in the availability of subcutaneously dosed and ready-to-use self-administration products for mAb medicines and their follow-on biologics. Different launch scenarios are described from a manufacturer's perspective highlighting the necessity of platform approaches. To unfold the full potential of customer-centric care, value-based healthcare provider reimbursement schemes that incentivize the efficiency of care need to be broadly implemented.

6.
PLoS One ; 16(7): e0254765, 2021.
Article in English | MEDLINE | ID: mdl-34292990

ABSTRACT

BACKGROUND: Recombinant human hyaluronidase PH20 (rHuPH20) facilitates the dispersion and absorption of subcutaneously administered therapeutic agents. This study aimed to characterize the transient, local action of rHuPH20 in the subcutaneous (SC) space using focused biodistribution and dye dispersion studies conducted in mice. MATERIALS AND METHODS: To evaluate the biodistribution of rHuPH20, mice were intradermally administered rHuPH20 (80 U). The enzymatic activity of rHuPH20 was analyzed in the skin, lymph nodes, and plasma. Animal model sensitivity was determined by intravenous administration of rHuPH20 (80 U) to the tail vein. To evaluate local dispersion, mice received an intradermal injection of rHuPH20 followed by an intradermal injection of Trypan Blue dye at a contralateral site 45 minutes later. Dye dispersion was measured using a digital caliper. RESULTS: After intradermal rHuPH20 injection, enzymatic activity was detected within the skin near the injection site with levels decreasing rapidly after 15 minutes. There was no clear evidence of systemic exposure after administration of rHuPH20, and no discernible rHuPH20 activity was observed in lymph or plasma as a function of time after dosing. In the dye dispersion study, delivery of rHuPH20 at one site did not impact dye dispersion at a distal skin site. CONCLUSION: These observations support the classification of rHuPH20 as a transiently active and locally acting permeation enhancer.


Subject(s)
Antigens, Neoplasm , Histone Acetyltransferases , Hyaluronoglucosaminidase , Animals , Antigens, Neoplasm/pharmacology , Female , Histone Acetyltransferases/pharmacokinetics , Histone Acetyltransferases/pharmacology , Humans , Hyaluronoglucosaminidase/pharmacokinetics , Hyaluronoglucosaminidase/pharmacology , Injections, Subcutaneous , Mice , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Tissue Distribution
7.
Med Devices (Auckl) ; 12: 101-127, 2019.
Article in English | MEDLINE | ID: mdl-30881151

ABSTRACT

Connected drug delivery devices are increasingly being developed to support patient supervision and counseling in home setting. Features may include dosing reminders, adherence trackers, tools for patient education, and patient diaries to collect patient-reported outcomes, as well as monitoring tools with interfaces between patients and health care professionals (HCPs). Five connected devices have been selected as the basis for a review of the clinical evidence concerning the impact of electronic tools on treatment adherence and efficacy outcomes. Disease areas covered include multiple sclerosis, diabetes, hypertension, liver and renal transplant recipients, tuberculosis, hepatitis C, clinically isolated syndrome, asthma, and COPD. From studies comparing the use of electronic feedback tools to standard of care, there is an initial evidence for a higher adherence to treatment and better outcomes among patients who use the electronic tools. To substantiate the assumption that connected devices can improve adherence in an outpatient setting over a prolonged period of time, further data from controlled randomized studies are required. Key barriers to the broader adoption of connected devices include data privacy laws that may prevent data sharing with HCPs in some countries, as well as the need to demonstrate that the tools are consistently used and generate a high-quality and reproducible database. If these challenges can be addressed in a way that is agreeable to all stakeholders, it is expected that the future value of connected devices will be to 1) facilitate and improve patient involvement in disease management in a flexible care setting, 2) enable early treatment decisions, and 3) complement value-based reimbursement models.

8.
BioDrugs ; 32(5): 425-440, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30043229

ABSTRACT

Subcutaneous delivery of biotherapeutics has become a valuable alternative to intravenous administration across many disease areas. Although the pharmacokinetic profiles of subcutaneous and intravenous formulations differ, subcutaneous administration has proven effective, safe, well-tolerated, generally preferred by patients and healthcare providers and to result in reduced drug delivery-related healthcare costs and resource use. The aim of this article is to discuss the differences between subcutaneous and intravenous dosing from both health-economic and scientific perspectives. The article covers different indications, treatment settings, administration volumes, and injection devices. We focus on biotherapeutics in rheumatoid arthritis (RA), immunoglobulin-replacement therapy in primary immunodeficiency (PI), beta interferons in multiple sclerosis (MS), and monoclonal antibodies (mAbs) in oncology. While most subcutaneous biotherapeutics in RA, PI, and MS are self-administered at home, mAbs for oncology are still only approved for administration in a healthcare setting. Beside concerns around the safety of biotherapeutics in oncology, a key challenge for self-administration in this area is that doses and dosing volumes can be comparatively large; however, this difficulty has recently been overcome to some extent by the development of high-concentration solutions, the use of infusion pumps, and the coadministration of the dispersion enhancer hyaluronidase. Furthermore, given the increasing number of biotherapeutics being considered for combination therapy and the high dosing complexity associated with these, especially when administered intravenously, subcutaneous delivery of fixed-dose combinations might be an alternative that will diminish these burdens on healthcare systems.


Subject(s)
Biological Products/administration & dosage , Biological Products/pharmacokinetics , Injections, Subcutaneous/methods , Administration, Intravenous/economics , Administration, Intravenous/methods , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Biological Availability , Biological Products/economics , Biological Products/therapeutic use , Humans , Immunologic Deficiency Syndromes/drug therapy , Injections, Subcutaneous/adverse effects , Injections, Subcutaneous/economics , Multiple Sclerosis/drug therapy , Neoplasms/drug therapy , Trastuzumab/administration & dosage
9.
J Clin Oncol ; 32(17): 1782-91, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24821885

ABSTRACT

PURPOSE: This two-stage phase IB study investigated the pharmacokinetics and safety of subcutaneous (SC) versus intravenous (IV) administration of rituximab as maintenance therapy in follicular lymphoma. PATIENTS AND METHODS: In stage 1 (dose finding), 124 patients who responded to rituximab induction were randomly assigned to SC rituximab (375 mg/m2, 625 mg/m2, or an additional group at 800 mg/m2) or IV rituximab (375 mg/m2). The objective was to determine an SC dose that would yield a rituximab serum trough concentration (Ctrough) in the same range as that of IV rituximab. In stage 2, 154 additional patients were randomly assigned (1:1) to SC rituximab (1,400 mg) or IV rituximab (375 mg/m2) given at 2- or 3-month intervals. The objective was to demonstrate noninferior rituximab Ctrough of SC rituximab relative to IV rituximab 375 mg/m2. RESULTS: Stage 1 data predicted that a fixed dose of 1,400 mg SC rituximab would result in a serum Ctrough in the range of that of IV rituximab. Noninferiority (ie, meeting the prespecified 90% CI lower limit of 0.8) was then confirmed in stage 2, with geometric mean Ctrough SC:Ctrough IV ratios for the 2- and 3-month regimens of 1.24 (90% CI, 1.02 to 1.51) and 1.12 (90% CI, 0.86 to 1.45), respectively. Overall safety profiles were similar between formulations (in stage 2, 79% of patients experienced one or more adverse events in each group). Local administration-related reactions (mainly mild to moderate) occurred more frequently after SC administration. CONCLUSION: The fixed dose of 1,400 mg SC rituximab predicted by using stage 1 results was confirmed to have noninferior Ctrough levels relative to IV rituximab 375 mg/m2 dosing during maintenance, with a comparable safety profile. Additional investigation will be required to determine whether the SC route of administration for rituximab provides equivalent efficacy compared with that of IV administration.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Agents/administration & dosage , Lymphoma, Follicular/drug therapy , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/adverse effects , Antibodies, Monoclonal, Murine-Derived/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Disease Progression , Female , Humans , Injections, Subcutaneous , Lymphoma, Follicular/metabolism , Male , Middle Aged , Rituximab
10.
Lancet Oncol ; 15(3): 343-52, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24521993

ABSTRACT

BACKGROUND: Intravenous rituximab is a mainstay of treatment for follicular lymphoma. A subcutaneous formulation that achieves equivalent rituximab serum concentrations might improve convenience and save health-care resources without sacrificing clinical activity. We aimed to assess pharmacokinetic non-inferiority of 3 week cycles of fixed-dose subcutaneous rituximab versus standard intravenous rituximab. METHODS: In our two-stage, randomised, open-label, phase 3 trial, we enrolled patients with previously untreated grade 1-3a, CD20-positive follicular lymphoma at 67 centres in 23 countries. In stage 1, we randomly allocated patients 1:1 with the Pocock and Simon algorithm to intravenous rituximab (375 mg/m(2)) or fixed-dose subcutaneous rituximab (1400 mg), stratified by induction chemotherapy regimen (cyclophosphamide, doxorubicin, vincristine, prednisone or cyclophosphamide, vincristine, prednisone), Follicular Lymphoma International Prognostic Index score, and region. After randomisation, patients received one induction dose of intravenous rituximab in cycle 1 and then allocated treatment for cycles 2-8. Patients with a complete or partial response following induction therapy continued intravenous or subcutaneous rituximab as maintenance every 8 weeks. The primary endpoint was the ratio of observed rituximab serum trough concentrations (Ctrough) between groups at cycle 7 (before cycle 8 dosing) of induction treatment in a per-protocol population. Patients were analysed as treated for safety endpoints. Stage 2 follow-up is ongoing and is fully accrued. This study is registered with ClinicalTrials.gov, number NCT01200758. FINDINGS: Between Feb 4, 2010, and Oct 21, 2011, we enrolled 127 patients. Pharmacokinetic data were available for 48 (75%) of 64 patients randomly allocated intravenous rituximab and 54 (86%) of 63 patients randomly allocated subcutaneous rituximab. Geometric mean Ctrough was 83·13 µg/mL in the intravenous group and 134·58 µg/mL in the subcutaneous group (ratio 1·62, 90% CI 1·36-1·94), showing non-inferiority of subcutaneous rituximab. 57 (88%) of 65 patients in the intravenous rituximab safety population had adverse events (30 [46%] grade ≥3), as did 57 (92%) of 62 patients in the subcutaneous rituximab safety population (29 [47%] grade ≥3). The most common grade 3 or worse adverse event in both groups was neutropenia (14 [22%] patients in the intravenous group and 16 [26%] patients in the subcutaneous group). Adverse events related to administration were mostly grade 1-2 and occurred in 21 (32%) patients in the intravenous group and 31 (50%) patients in the subcutaneous group. INTERPRETATION: Stage 1 data show that the pharmacokinetic profile of subcutaneous rituximab was non-inferior to intravenous rituximab and was not associated with new safety concerns. Stage 2 will provide data for efficacy and safety of the subcutaneous administration. FUNDING: F Hoffmann-La Roche.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Lymphoma, Follicular/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/adverse effects , Antineoplastic Agents/adverse effects , Female , Humans , Injections, Subcutaneous , Lymphoma, Follicular/pathology , Male , Middle Aged , Neoplasm Staging , Rituximab
12.
Int J Clin Pharmacol Ther ; 51(7): 537-48, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23547849

ABSTRACT

OBJECTIVES: To investigate the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of tocilizumab with and without rHuPH20 (a recombinant human hyaluronidase) in healthy volunteers. METHODS: This was an open-label, single ascending dose study. Subjects were assigned to tocilizumab 162 mg or tocilizumab 162, 324, or 648 mg plus rHuPH20. PK and PD samples were collected after dosing and were estimated with non-compartmental methods. Geometric mean ratio (GMR) for area under the plasma concentration-time curve from zero to infinity (AUC0-∞) and (maximum serum concentration) Cmax with and without rHuPH20 was estimated using one-way analysis of variance. Safety and tolerability were monitored throughout the study. RESULTS: 48 subjects (12/cohort) received a single dose of tocilizumab with or without rHuPH20. For tocilizumab 162 mg, tocilizumab 162 mg/rHuPH20, tocilizumab 324 mg/rHuPH20, and tocilizumab 648 mg/rHuPH20, mean ± SD tocilizumab PK parameters were 2,510 ± 1,060, 2,860 ± 468, 10,800 ± 3,220, and 29,900 ± 5,280 µg×h/ml for AUC0-∞; 11.5 ± 3.7, 16.2 ± 2.8, 43.8 ± 12.4, and 77.8 ± 14.5 µg/ml for Cmax; and 89.1 ± 41.1, 54.0 ± 19.5, 66.0 ± 26.8, and 86.1 ± 50.6 h for tmax, respectively. Coadministration of tocilizumab 162 mg with rHuPH20 resulted in slightly increased exposure: GMR (90% confidence interval) for AUC0-∞, 1.20 (1.00 - 1.44) and Cmax, 1.45 (1.24 - 1.70). Increasing tocilizumab doses resulted in significant deviation from dose proportionality for tocilizumab Cmax (p = 0.0057) and AUC0-∞ (p < 0.0001). Changes in interleukin-6, soluble interleukin- 6 receptor, and C-reactive protein were also dose dependent and similar with and without rHuPH20. CONCLUSIONS: Tocilizumab in combination with rHuPH20 resulted in slightly increased tocilizumab exposure compared with tocilizumab alone, whereas PD markers were comparable. Subcutaneous administration of tocilizumab with rHuPH20 was well tolerated.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/pharmacokinetics , Hyaluronoglucosaminidase/administration & dosage , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/blood , Antirheumatic Agents/adverse effects , Antirheumatic Agents/blood , Area Under Curve , Biomarkers/blood , C-Reactive Protein/metabolism , Drug Interactions , Female , Humans , Hyaluronoglucosaminidase/adverse effects , Inflammation Mediators/blood , Injections, Subcutaneous , Interleukin-6/blood , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Models, Statistical , Receptors, Interleukin-6/blood , Recombinant Proteins/administration & dosage , Young Adult
13.
J Clin Pharmacol ; 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23504807

ABSTRACT

Trastuzumab is a key component of treatment for human epidermal growth factor receptor 2 (HER2)-positive breast cancer in both the early and metastatic settings. It is administered intravenously, with between 17 and 52 infusions in standard regimens over 1 year. Intravenous administration of trastuzumab requires substantial time commitments for patients and health care professionals and can result in patient discomfort. A subcutaneous formulation of trastuzumab, containing recombinant human hyaluronidase to overcome subcutaneous absorption barriers, would reduce the administration duration and remove the need to establish intravenous access, thus improving the overall convenience of trastuzumab administration. This open-label, 2-part, phase I/Ib study (NCT00800436) was undertaken in healthy male volunteers and female patients with HER2-positive early breast cancer to identify the dose of subcutaneous trastuzumab that resulted in exposure comparable with the approved intravenous trastuzumab dose. A subcutaneous trastuzumab dose of 8 mg/kg was found to result in exposure comparable with the intravenous trastuzumab dose of 6 mg/kg. The subcutaneous formulation was well tolerated, with a trend toward fewer adverse events versus intravenous administration; most adverse events were mild in intensity. These results support an ongoing phase III efficacy and safety study comparing a fixed subcutaneous trastuzumab dose with intravenous trastuzumab administration.

14.
J Clin Pharmacol ; 53(2): 192-201, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23436264

ABSTRACT

Trastuzumab is a key component of treatment for human epidermal growth factor receptor 2 (HER2)-positive breast cancer in both the early and metastatic settings. It is administered intravenously, with between 17 and 52 infusions in standard regimens over 1 year. Intravenous administration of trastuzumab requires substantial time commitments for patients and health care professionals and can result in patient discomfort. A subcutaneous formulation of trastuzumab, containing recombinant human hyaluronidase to overcome subcutaneous absorption barriers, would reduce the administration duration and remove the need to establish intravenous access, thus improving the overall convenience of trastuzumab administration. This open-label, 2-part, phase I/Ib study (NCT00800436) was undertaken in healthy male volunteers and female patients with HER2-positive early breast cancer to identify the dose of subcutaneous trastuzumab that resulted in exposure comparable with the approved intravenous trastuzumab dose. A subcutaneous trastuzumab dose of 8 mg/kg was found to result in exposure comparable with the intravenous trastuzumab dose of 6 mg/kg. The subcutaneous formulation was well tolerated, with a trend toward fewer adverse events versus intravenous administration; most adverse events were mild in intensity. These results support an ongoing phase III efficacy and safety study comparing a fixed subcutaneous trastuzumab dose with intravenous trastuzumab administration.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Administration, Intravenous , Adolescent , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Breast Neoplasms/blood , Female , Humans , Injections, Subcutaneous , Male , Middle Aged , Receptor, ErbB-2 , Trastuzumab , Young Adult
15.
Arzneimittelforschung ; 61(12): 707-13, 2011.
Article in English | MEDLINE | ID: mdl-22282958

ABSTRACT

The novel excipient, sodium N-[8-(2-hydroxybenzoyl)amino]caprylate (SNAC, CAS 203787-91-1) increases the oral bioavailability of co-formulated ibandronate (IBN, CAS 138926-19). The aim of this study was to investigate the effect of the IBN/SNAC formulation on the steady-state pharmacokinetics of metformin (CAS 657-24-9) and to assess safety and tolerability of IBN/SNAC when dosed in combination with metformin. Twenty-two healthy subjects received metformin on Days 1 to 6. On Day 7, subjects received metformin together with the IBN/ SNAC formulation. The safety and tolerability of IBN/SNAC co-administered with metformin was consistent with the known safety profile of the single medications. The increase in mean maximum plasma concentration (Cmax) and mean overall exposure to metformin (AUC0-tau,) was approximately 7%. The entire 90% confidence intervals for the AUC- and Cmax-ratios did fall within the acceptance region for bioequivalence (0.8-1.25). In summary, administration of the IBN/ SNAC formulation together with metformin did not lead to a significant increase in exposure to metformin. The study medication was well tolerated in healthy volunteers.


Subject(s)
Bone Density Conservation Agents/pharmacology , Diphosphonates/pharmacology , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Adolescent , Adult , Area Under Curve , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacokinetics , Chemistry, Pharmaceutical , Cross-Over Studies , Diphosphonates/administration & dosage , Diphosphonates/pharmacokinetics , Double-Blind Method , Drug Interactions , Female , Half-Life , Humans , Hypoglycemic Agents/pharmacokinetics , Ibandronic Acid , Male , Metformin/pharmacokinetics , Middle Aged , Young Adult
16.
Antivir Ther ; 10(7): 803-10, 2005.
Article in English | MEDLINE | ID: mdl-16312177

ABSTRACT

OBJECTIVE: To establish the bioequivalence of a 500 mg film-coated tablet of saquinavir mesylate (FCT SQV) to the 200 mg hard-capsule saquinavir mesylate (HC SQV), both boosted with ritonavir and administered under fed conditions. METHODS: We carried out a multi-centre, open-label, randomized, two-sequence, four-period, two-treatment, replicated crossover study in 93 healthy men and 7 healthy women. Individuals were randomly assigned to receive sequential single doses of saquinavir in one of two treatment sequences: ABAB or BABA. Individuals received 100 mg ritonavir twice daily for 24 days. On days 14,17, 20 and 23, study participants took 1000 mg of HC SQV (five 200 mg capsules, treatment A) or FCT SQV (two 500 mg tablets, treatment B) with a high-fat, high-calorie breakfast, and pharmacokinetic analyses were carried out over the next 24 hours. Area under the saquinavir concentration-time curve (AUC0-alpha), maximum saquinavir plasma concentration (Cmax), time to Cmax and terminal half-life were calculated. The relative bioavailability of FCT SOV versus HC SQV was calculated as the ratio of the respective estimated mean saquinavir AUC0-alpha and Cmax. The calculation was based on an ANOVA including the factors site, sex, sequence, period, treatment and study participant to the log-transformed parameters log(AUC0-alpha) and log(Cmax); the relative bioavailability and the 90% confidence intervals (CIs) were estimated using the treatment contrasts of the ANOVA. Bioequivalence was concluded as for both parameters, AUC0-alpha and Cmax, the 90% CIs for the relative bioavailability were entirely included in the reference region [0.80-1.25]. RESULTS: Saquinavir plasma concentration-time profiles for the two formulations were similar. Geometric mean AUC0-alpha and Cmax values were clearly increased for FCT SQV (26 826 versus 24 430 h*ng/ml; and 3644 versus 3064 ng/ml, respectively); ratios of mean exposures were estimated to be 1.10 for AUC0-alpha and 1.19 for Cmax of saquinavir. However, the corresponding two-sided 90% CIs (1.04-1.16 and 1.14-1.25, respectively) all fell within the limits set for equivalence (0.80, 1.25). The adverse event profile for FCT SQV was similar to that for HC SQV. CONCLUSION: The new 500 mg FCT SQV formulation is bioequivalent to the 200 mg HC SQV formulation, at the dose of 1000 mg, in combination with 100 mg ritonavir under fed conditions. The 500 mg FCT SQV formulation reduces pill count for boosted saquinavir (SQV/r) from six capsules to three tablets twice daily. This may increase patient acceptability of SQV/r, particularly in less treatment-experienced patients.


Subject(s)
Anti-HIV Agents/administration & dosage , Ritonavir/administration & dosage , Ritonavir/pharmacokinetics , Saquinavir/administration & dosage , Saquinavir/pharmacokinetics , Adult , Aged , Anti-HIV Agents/adverse effects , Anti-HIV Agents/blood , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Capsules , Cross-Over Studies , Drug Administration Schedule , Female , Half-Life , Health , Humans , Male , Middle Aged , Ritonavir/adverse effects , Ritonavir/blood , Saquinavir/adverse effects , Saquinavir/blood , Tablets , Therapeutic Equivalency
17.
Int J Pharm ; 279(1-2): 27-31, 2004 Jul 26.
Article in English | MEDLINE | ID: mdl-15234791

ABSTRACT

In the current investigation, the impact of the surface-active formulation ingredient Solutol HS 15 on the uptake of colchicine into freshly isolated rat hepatocytes was investigated using a centrifugal filtration technique through a silicone oil layer. Colchicine is taken up into the cells by an active transport mechanism. When conducting the experiment at 37 degrees C, it was found that at concentrations below its critical micellar concentration (CMC) of 0.021% (0.0003 and 0.003%, w/v), Solutol HS 15 did not impact the uptake of colchicine. By contrast, at a Solutol HS 15 concentration above its CMC (0.03%, w/v), the amount of colchicine taken up into the cells as well as its uptake velocity were significantly decreased. However, in control experiments performed at 4 degrees C, a temperature at which active transport processes should be significantly slowed down, Solutol HS 15 at 0.03% did not affect colchicine uptake and/or its association with the cells. The described findings might be rationalized by inhibition of colchicine transport either due to direct interaction at the transport site or due to alterations of membrane properties in the presence of Solutol HS 15 at concentrations above its CMC. Moreover, a strong molecular interaction between Solutol HS 15 and colchicine as well as an incorporation of colchicine into micelles formed by Solutol HS 15, this way resulting in a limited contact of colchicine with the cells, cannot be excluded as contributors to the observed effect.


Subject(s)
Colchicine/pharmacokinetics , Hepatocytes/metabolism , Polyethylene Glycols/pharmacology , Stearic Acids/pharmacology , Animals , Biological Transport, Active/drug effects , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Rats , Rats, Wistar , Temperature
18.
Biopharm Drug Dispos ; 25(1): 37-49, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14716751

ABSTRACT

The impact of the surface-active formulation ingredients Cremophor EL, Tween 80 and Solutol HS 15 on the intrinsic clearance (Clint) of midazolam (MDZ) was investigated in rat hepatocytes and microsomes. In rat hepatocytes with 0.003%, 0.03% and 0.3% (w/v) Solutol HS 15 already present in the incubation medium, the Clint was significantly reduced in a dose-dependent manner by about 25%, 30% and 50%, respectively. In the presence of Cremophor EL and Tween 80 a significant reduction in Clint by about 30% and 25%, respectively, was observed at 0.03% surfactant concentration. At 0.3% of Cremophor EL and Tween 80, Clint was reduced by about 50% and 20%, respectively. A reduction in Clint was also observed in experiments with rat liver microsomes. At surfactant concentrations up to 0.03%, cytotoxicity assays (lactate dehydrogenase release, adenosine triphosphate content) as well as light microscope investigations did not reveal any cytotoxic impact of the surfactants on the hepatocyte monolayer. A potential interaction of the surfactants with biological membranes was determined using phosphatidylcholine-cholesterol liposomes loaded with self-quenching concentrations of carboxyfluorescein. No marked release of carboxyfluorescein from the liposomes (that would be an indication for a surfactant-dependent disruption of membrane integrity) was observed up to concentrations of 0.03% of the different surfactants. It is concluded that cytochrome P450 3A mediated metabolism of MDZ seems to be prevented by all surfactants at concentrations above 0.03%. In our experiments the surfactants did not show toxic effects at concentrations that resulted in a decreased Clint of MDZ. Thus, a direct inhibition of the metabolizing enzymes, a molecular interaction with the microsomes as well as an alteration of membrane properties that did not yet result in a release of LDH have to be taken into consideration as reasons for the observed changes in the metabolism of MDZ.


Subject(s)
Excipients/metabolism , Glycerol/analogs & derivatives , Glycerol/metabolism , Midazolam/metabolism , Polyethylene Glycols/metabolism , Polysorbates/metabolism , Stearic Acids/metabolism , Surface-Active Agents/adverse effects , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Survival/drug effects , Cells, Cultured , Culture Media/chemistry , Cytochrome P-450 CYP3A , Drug Evaluation, Preclinical/methods , Drug Interactions , Excipients/chemistry , Excipients/pharmacology , Glycerol/chemistry , Glycerol/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Male , Membrane Proteins/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Midazolam/chemistry , Midazolam/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polysorbates/chemistry , Polysorbates/pharmacology , Rats , Rats, Wistar , Stearic Acids/chemistry , Stearic Acids/pharmacology , Surface-Active Agents/standards , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/trends
19.
Eur J Pharm Biopharm ; 56(1): 143-6, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12837492

ABSTRACT

The pharmacokinetic profile of midazolam (MDZ) and its major metabolites 1'-OH-midazolam (1'OH-MDZ) and 4-OH-midazolam (4OH-MDZ) was investigated in rats. MDZ was administered intravenously at 5 mg/kg either in the absence (NaCl 0.9%, control group) or in the presence of the surfactant Solutol HS 15, a weak inhibitor of cytochrome P450 3A (CYP3A) activity in vitro (Solutol HS 15-treated group). It was found that the pharmacokinetic profiles of MDZ, 1'OH-MDZ and 4OH MDZ did not differ significantly in the two dosing vehicles (P values above 0.2). MDZ exhibited a high plasma clearance (Cl) of 79 and 92 ml/min/kg (corresponding to a blood Cl of 64 and 75 ml/min/kg), a high volume of distribution (V(d)) of 4.0 and 3.6 l/kg, and an area under the plasma concentration-time curve (AUC(t0-tinf)) of 1062 and 932 h.ng/ml in the control group and in the Solutol HS 15-treated group, respectively. The amount of MDZ excreted unchanged into urine was below 0.01% with both dosing vehicles. AUC(t0-tinf) in the control group was 12.3 h.ng/ml for 1'OH-MDZ and 38.8 h.ng/ml 4OH-MDZ. In the Solutol HS 15-treated group, AUC(t0-tinf) was 14 h.ng/ml for 1'OH-MDZ and 35.4 h.ng/ml for 4OH-MDZ. The metabolite concentrations excreted into urine were below the limit of quantification. In the rat, MDZ has a high blood clearance that is limited by liver blood flow. Therefore, weak CYP3A inhibitors like Solutol HS 15 are not likely to affect the hepatic blood clearance of MDZ in vivo.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Hypnotics and Sedatives/pharmacokinetics , Midazolam/pharmacokinetics , Polyethylene Glycols/pharmacology , Stearic Acids/pharmacology , Surface-Active Agents/pharmacology , Administration, Oral , Animals , Area Under Curve , Half-Life , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/metabolism , Injections, Intravenous , Liver/metabolism , Male , Midazolam/administration & dosage , Midazolam/metabolism , Rats , Rats, Wistar , Solubility , Time Factors
20.
Biopharm Drug Dispos ; 24(4): 173-81, 2003 May.
Article in English | MEDLINE | ID: mdl-12698501

ABSTRACT

In the current investigation, the alkaloid colchicine was administered intravenously to male Wistar rats both as a solution in isotonic sodium chloride (NaCl 0.9%, control group) and in NaCl 0.9%:Solutol HS 15 (95:5) at 1.5 mg/kg. At predetermined time points, plasma and urine were collected from the animals and analysed for colchicine and its demethylated metabolites by LC/MS-MS. In the presence of Solutol HS 15, colchicine clearance (CI) was significantly decreased and its maximum plasma concentration (c(max)) was significantly increased as compared to the control group (CI: 15.6+/-7.0 ml/min/kg vs 34.3+/-2.3 ml/min/kg; c(max) 3055.1+/-587.4 h vs 1260.1+/-223.7 h; p<0.05). Moreover, the amount of parent colchicine excreted into urine was markedly increased in the Solutol HS 15 treated group (41.50+/-3.23 vs 1.17+/-0.41% of total dose; p<0.05). By contrast, there was no statistically significant difference but a trend to lower values only in the volume of distribution (V(d) 13.3+/-2.2 l/h vs 31.4+/-17.7 l/h, p=0.35). The half-lives for the first (t(1/2 1stphase). 0.21+/-0.02 h vs 0.20+/-0.03 h) and second phase (t(1/2 2ndphase). 18.5+/-6.9 h vs 18.3+/-7.7 h) did not differ significantly in dependence on the dosing vehicle. The free fraction in rat plasma (FF), the blood/plasma (lambda) and erythrocyte/plasma concentration ratios (K(e)) were not significantly changed in the presence of different concentrations of Solutol HS 15 compared with surfactant-free incubations (overall means: 72.25+/-0.50% for FF, 0.80+/-0.02 for lambda, 0.46+/-0.04 for K(e)). In vitro, in rat hepatocytes, the clearance of colchicine was significantly reduced at 0.003% Solutol HS 15 present in the incubation medium (0.86+/-0.15 microl/min/10(-6) cells vs 1.46+/-0.06 microl/min/10(-6) cells). As colchicine exhibits a comparatively high aqueous solubility, an impact of Solutol HS 15 on the solubility of the alkaloid is very unlikely to be a reason for the observed effect. Therefore, our results indicate that the most likely reasons for the changed pharmacokinetic behaviour of colchicine in the presence of Solutol HS 15 are alterations of metabolism and/or transport as well as distribution and elimination processes.


Subject(s)
Colchicine/administration & dosage , Colchicine/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Stearic Acids/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Colchicine/metabolism , Cytochrome P-450 Enzyme System/metabolism , Excipients/metabolism , Excipients/pharmacokinetics , Hepatocytes/chemistry , Hepatocytes/drug effects , Injections, Intravenous , Male , Polyethylene Glycols/adverse effects , Polyethylene Glycols/metabolism , Protein Binding/drug effects , Rats , Rats, Wistar , Stearic Acids/adverse effects , Stearic Acids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...