1
|
Park K. PLGA-based long-acting injectable (LAI) formulations. J Control Release 2025; 382:113758. [PMID: 40268201 DOI: 10.1016/j.jconrel.2025.113758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
Long-acting injectable (LAI) formulations, which deliver drugs over weeks or months, have been in use for more than three decades. Most clinically approved LAI products are formulated using poly(lactide-co-glycolide) (PLGA) polymers. Historically, the development of PLGA-based LAI formulations has relied predominantly on trial-and-error methods, primarily due to a limited understanding of the complex factors involved in LAI formulations and insufficient analytical techniques available for characterizing individual PLGA polymers of the prepared formulations. This article offers a personal perspective on recent advancements in characterization methods for PLGA polymers within final formulations, i.e., products, as well as enhanced insights into the drug release mechanisms associated with LAI products. With a deeper understanding of PLGA polymer properties and drug release mechanisms, the formulation development process can transition from traditional trial-and-error practices to a more systematic Quality by Design (QbD) approach. Additionally, this article explores the emerging role of artificial intelligence (AI) in formulation science and its potential, when applied carefully, to enhance the future development of PLGA-based LAI formulations.
Collapse
Affiliation(s)
- Kinam Park
- Purdue University, Weldon School of Biomedical Engineering and Department of Industrial and Molecular Pharmaceutics, West Lafayette, IN 47907, USA; Akina, Inc., 3495 Kent Avenue, West Lafayette, IN 47906, United States of America.
| |
Collapse
|
2
|
Hu T, Yang S, Yu J, Yang Z, Jin Y, Zhu Z. Transitions in treatment: A systematic review and meta-aggregation of preferences and barriers in switching from oral to long-acting injectable antiretroviral therapy among people living with HIV and stakeholders. HIV Med 2025; 26:512-535. [PMID: 39748161 DOI: 10.1111/hiv.13756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION Antiretroviral treatment (ART) has significantly enhanced health outcomes for people living with HIV (PLWH). With the evolution of treatment options, there is an increasing interest in the development of long-acting injectable formulations of antiretroviral drugs. These formulations present a promising alternative to oral ART. METHODS The methodology and reporting of this systematic review followed the guidance of the Joanna Briggs Institute Reviewer's Manual and Enhancing Transparency in Reporting the Synthesis of Qualitative Research (ENTREQ). The comprehensive searches involved multiple databases, including PubMed, MEDLINE (Ovid), Embase (Ovid), CINHAL (EBSCO), ProQuest Dissertations and Theses, Web of Science, Wanfang (Chinese), CNKI (Chinese), Google Scholar and Baidu Scholar (Chinese). RESULTS In all, 142 studies were identified and 20 eligible studies were included in the meta-aggregation. A total of 141 findings, 20 categories and nine synthesized findings were extracted from 20 studies. The nine synthesized findings identified from the 20 studies focused on the following topics: benefits, flexibility and practicality of long-acting injectable (LAI) treatment; scepticism about the use of LAI treatment; management challenges; logistical challenges; potential for protecting marginalized populations; concerns about side effects; financial issue; suggestions for improvement. PLWH's geographical distribution, backgrounds, demographics and clinical characteristics were limited. CONCLUSION We recommend considering the needs and experiences of PLWH in the transition from oral ART to LAI treatment. For marginalized populations, it is crucial to maintain regular communication with healthcare providers and institutions. Additionally, at the community level, engaging diverse stakeholders with valuable insights is vital, as is enhancing health education programmes and intensifying efforts to combat discrimination. These measures will play a key role in addressing the needs of PLWH, enhancing public awareness and promoting better understanding of LAI treatment.
Collapse
Affiliation(s)
- Tiantian Hu
- Fudan University School of Nursing, Shanghai, China
| | - Shuqi Yang
- Fudan University School of Nursing, Shanghai, China
| | - Junwen Yu
- Fudan University School of Nursing, Shanghai, China
| | | | - Yilin Jin
- Fudan University School of Nursing, Shanghai, China
| | - Zheng Zhu
- Fudan University School of Nursing, Shanghai, China
- Rory Meyers College of Nursing, New York University, New York, New York, USA
| |
Collapse
|
3
|
Anjani QK, Johnson AR, Sabri AH, Lutz R, Tignor S, Ballard J, Rudd N, Zhao L, Vora LK, Barrett SE, Wagner A, Donnelly RF. Delivery of Islatravir via High Drug-Load, Long-acting Microarray Patches for the Prevention or Treatment of Human Immunodeficiency Virus. Adv Healthc Mater 2025; 14:e2403615. [PMID: 39840486 PMCID: PMC11912095 DOI: 10.1002/adhm.202403615] [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: 09/21/2024] [Revised: 01/07/2025] [Indexed: 01/23/2025]
Abstract
This research focuses on developing and characterizing islatravir-loaded dissolving microarray patches (MAPs) to provide an effective, minimally invasive treatment option for human immunodeficiency virus (HIV-1) prevention and treatment. The research involves manufacturing these MAPs using a double-casting approach, and conducting in vitro and in vivo evaluations. Results show that the MAPs have excellent needle fidelity, structural integrity, and mechanical strength. in vitro studies demonstrate that the MAPs can penetrate skin up to 580 µm and dissolve within 2 hours. Permeation studies reveal that the delivery efficiency of islatravir across the skin is around 40%. In rodent models, these dissolving MAPs sustain islatravir delivery for up to 3 months. Scaling up the MAPs and increasing drug loading produced detectable levels in minipig. Projections from animal data suggest that these dissolving MAPs can achieve effective islatravir levels for a month after a single application in humans. These findings indicate dissolving MAPs as a minimally invasive approach to sustained release of islatravir.
Collapse
Affiliation(s)
- Qonita Kurnia Anjani
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Ashley R Johnson
- Sterile and Specialty Products, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Akmal H Sabri
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Ryan Lutz
- Absorption, Distribution, Metabolism & Excretion, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Steven Tignor
- Small Molecule Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Jeanine Ballard
- Absorption, Distribution, Metabolism & Excretion, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Nathan Rudd
- Pharmaceutical Sciences R&D, Merck Animal Health, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Li Zhao
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Stephanie E Barrett
- Sterile and Specialty Products, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Angela Wagner
- Sterile and Specialty Products, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| |
Collapse
|
4
|
Zulbeari N, Lund NE, Holm R. Small-scale aqueous suspension preparation using dual centrifugation: the effect of process parameters on the sizes of drug particles. Eur J Pharm Sci 2025; 205:106980. [PMID: 39643126 DOI: 10.1016/j.ejps.2024.106980] [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: 09/07/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
The dual centrifugation approach has in the recent years emerged as a powerful milling tool to prepare pharmaceutical suspensions in submicron range with a fast-milling capacity by milling 40 samples simultaneously in 2 mL vials. While there is some standardized milling conditions described in the literature when preparing aqueous suspensions with dual centrifugation, a more systematic and experimental understanding of the milling process to evaluate the impact of different process variables in the dual centrifuge on the final sizes of the suspended drug particles independent of the drug compound used was desired. Overall, the present study demonstrated the applicability of the dual centrifuge for small-scale screening purposes and showed the impact of process parameters on the physical attributes of prepared suspensions. In the present work, the rate of size reduction on three different model compounds, i.e., cinnarizine, haloperidol, and indomethacin, was found to be mostly influenced by the milling speed, size of milling beads, and the bead loading during milling, whereas the rotor temperature did not affect the particle size profiles when stabilized with polysorbate 20 during milling with dual centrifugation. Smaller particle sizes were in general obtained at the highest milling intensity, i.e., 1500 rpm, smallest bead size, i.e., 0.2 mm, and higher bead loadings (42 %, 56 %, and 83 %). The grinding limit of approximately 0.50 µm, 0.70 µm, and 0.35 µm for cinnarizine, haloperidol, and indomethacin, respectively, was achieved relatively fast, i.e., 30 min of milling at the specified conditions, compared to when suspensions were milled with larger bead sizes (i.e., 1.0 mm), lower milling intensities (e.g., 1000 rpm), and lower bead loadings (e.g., 14 or 28 %). The study further confirmed that a higher milling intensity was necessary during milling of haloperidol suspensions probably due to the compounds predominantly plastic properties. Sizes of indomethacin particles increased with longer milling runs up to 240 min and also higher bead loadings of 56 % and 83 %. These observations were further supported by the color conversion from white to yellow of indomethacin suspensions which indicated generation of small quantities of amorphic material after milling with a high milling intensity. Upscale investigations showed comparable particle size profiles for all three model compounds while milling at 1500 rpm for five minutes.
Collapse
Affiliation(s)
- Nadina Zulbeari
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55 5230, Odense, Denmark
| | - Nanna Einshøj Lund
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55 5230, Odense, Denmark
| | - René Holm
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55 5230, Odense, Denmark.
| |
Collapse
|
5
|
Farooq MA, Johnston APR, Trevaskis NL. Impact of nanoparticle properties on immune cell interactions in the lymph node. Acta Biomater 2025; 193:65-82. [PMID: 39701340 DOI: 10.1016/j.actbio.2024.12.039] [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: 08/07/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
The lymphatic system plays an important role in health and many diseases, such as cancer, autoimmune, cardiovascular, metabolic, hepatic, viral, and other infectious diseases. The lymphatic system is, therefore, an important treatment target site for a range of diseases. Lymph nodes (LNs), rich in T cells, B cells, dendritic cells, and macrophages, are also primary sites of action for vaccines and immunotherapies. Promoting the delivery of therapeutics and vaccines to LNs can, therefore, enhance treatment efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. Several nanoparticle (NP) based delivery systems, such as polymeric NPs, lipid NPs, liposomes, micelles, and dendrimers, have been reported to enhance the delivery of therapeutics and/or vaccines to LNs. Specific uptake into the lymph following injection into tissues is highly dependent on particle properties, particularly particle size, as small molecules are more likely to be taken up by blood capillaries due to higher blood flow rates, whereas larger molecules and NPs can be specifically transported via the lymphatic vessels to LNs as the initial lymphatic capillaries are more permeable than blood capillaries. Once NPs enter LNs, particle properties also have an important influence on their disposition within the node and association with immune cells, which has significant implications for the design of vaccines and immunotherapies. This review article focuses on the impact of NP properties, such as size, surface charge and modification, and route of administration, on lymphatic uptake, retention, and interactions with immune cells in LNs. We suggest that optimizing all these factors can enhance the efficacy of vaccines or therapeutics with targets in the lymphatics and also be helpful for the rational design of vaccines. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an essential role in health and is an important treatment target site for a range of diseases. Promoting the delivery of immunotherapies and vaccines to immune cells in lymph nodes can enhance efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. One of the major approaches used to deliver therapeutics and vaccines to lymph nodes is via injection in nanoparticle delivery systems. This review aims to provide an overview of the impact of nanoparticle properties, such as size, surface charge, modification, and route of administration, on lymphatic uptake, lymph node retention, and interactions with immune cells in lymph nodes. This will inform the design of future improved nanoparticle systems for vaccines and immunotherapies.
Collapse
Affiliation(s)
- Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
6
|
Zulbeari N, Wang F, Mustafova SS, Parhizkar M, Holm R. Machine learning strengthened formulation design of pharmaceutical suspensions. Int J Pharm 2025; 668:124967. [PMID: 39566699 DOI: 10.1016/j.ijpharm.2024.124967] [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: 09/14/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Many different formulation strategies have been investigated to oppose suboptimal treatment of long-term or chronic conditions, one of which are the nano- and microsuspensions prepared as long-acting injectables to prolong the release of an active pharmaceutical compound for a defined period of time by regulating the size of particles by milling. Typically, surfactant and/or polymers are added in the dispersion medium of the suspension during processing for stabilization purposes. However, current formulation investigations with milling are heavily based on prior expertise and trial-and-error approaches. Various interacting parameters such as the milling bead size, stabilizer type and concentration have confounded the investigation of milling process. The present study systematically exploited statistical and machine learning (ML) strategies to understand the relationship between suspension characteristics and formulation parameters under full-factorial milling experiments. Stabilizer concentration was identified as a significant factor (p < 0.001) for median suspension diameter (D50). A formulation stability classification ML model with high prediction accuracy (0.91) and F1-score (0.91) under 10-fold cross-validation was constructed based on 72 formulation datapoints. Model interpretation through Shapley additive explanations (SHAP) revealed the prominent impact of stabilizer concentration and milling bead size on formulation stability. The present work demonstrated the potential to achieve a deeper understanding of the design and optimization of nano- and microsuspensions through explainable ML modelling on formulation screening data.
Collapse
Affiliation(s)
- Nadina Zulbeari
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Fanjin Wang
- Deparment of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, WC1N 1AX London, United Kingdom
| | - Sibel Selyatinova Mustafova
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Maryam Parhizkar
- Deparment of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, WC1N 1AX London, United Kingdom
| | - René Holm
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
7
|
Zulbeari N, Ulrich Kristensen L, Mende S, Holm R. Temperature mapping of milling by dual centrifugation: A systematic investigation. Int J Pharm 2024; 666:124760. [PMID: 39332461 DOI: 10.1016/j.ijpharm.2024.124760] [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: 08/02/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Using low quantities of drug compounds is often favorable in the early stages of drug development, especially for what require a large screening investigation to define the final formulation composition, such as nano- and microsuspensions. For that reason, the dual centrifugation approach has in the recent years been used due to its reproducible and fast-milling capacity with 40 samples in 2 mL vials simultaneously without the addition of cooling breaks due to a built-in cooling system. Nonetheless, heat can be dissipated into the samples during high-intensity milling, resulting in increased sample temperatures that potentially can affect thermolabile compounds and potential influence the obtained suspensions in the screening experiments if the used stabilizer has temperature dependent variations in the performance. Hence, a systematic investigation of the influence of different process parameters on the heat dissipation in samples during milling by the dual centrifugation approach was performed in the present study. It was found that the milling speed had the highest impact on the final sample temperature, but also other parameters, such as the bead loading, bead size, and placement in the centrifuge during milling had significantly influenced the final mean temperature of the milling media. Higher temperatures were obtained with higher bead loadings, i.e., 3000 mg milling beads/mL and milling speeds (1500 rpm), and when smaller milling beads, i.e., 0.1 mm, were used during production. The study further showed that higher temperatures were measured for samples located on the bottom disk during milling, and also when located on the outer placement on the sample disk. Upscale investigations showed immensely increased sample temperatures (almost up to boiling point) when samples were prepared under similar formulation parameters and milling speed as small-volume vials. Furthermore, the study indicated that the addition of drug compounds during suspension preparation decreased the final sample temperature compared to samples that only contained purified water due to energy absorption of the drug compound.
Collapse
Affiliation(s)
- Nadina Zulbeari
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Louise Ulrich Kristensen
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Stefan Mende
- NETZSCH-Feinmahltechnik GmbH, Sedanstrasse 70, 95100 Selb, Germany
| | - René Holm
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
8
|
Zulbeari N, Hansen SM, Holm R. Two in One: Size Characterization and Accelerated Short-Term Physical Stability of Dual-Drug Suspensions with Two Acidic Compounds (Indomethacin and Naproxen). Pharmaceutics 2024; 16:1495. [PMID: 39771474 PMCID: PMC11676886 DOI: 10.3390/pharmaceutics16121495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Co-delivering dual-drug systems have proven to be effective in, for example, anticancer therapy or HIV prophylaxis due to a higher target selectivity and therapeutic efficacy from compound synergism. However, various challenges regarding physical stability can arise during the formulation definition when multiple drug compounds are included in the same formulation. In this work, the focus was on aqueous suspensions, which could be applied as long-acting injectable formulations to release the drug compounds over weeks to months after administration. Methods: It was possible to gain insights into dual-drug nano- and microsuspensions containing two acidic compounds (indomethacin and naproxen) prepared by milling with dual centrifugation. Information regarding the physical stability of individual suspensions was subtracted and compared to dual-drug suspensions when prepared with the same milling conditions and stored at elevated temperatures of 40 °C. Results: Distinct particle size profiles after milling were obtained dependent on the stabilizer used in both individual and dual-drug suspensions. Most notably, the combination of indomethacin and naproxen in one formulation resulted in smaller sizes of drug particles compared to individual suspensions under the presence of some stabilizers. The obtained particle size profiles further indicated that at least one of the model compounds needed to be sufficiently stabilized from a stabilizer to obtain physically stable dual-drug suspensions over 28 days when stored at 40 °C. Similarly, the particle size distribution was dependent on the individual distribution of the suspensions, which showed a monomodal distribution could be achieved for dual-drug suspensions when at least one of the individual suspensions demonstrated a monomodal distribution in the presence of the stabilizer alone. Over a 28-day period, the smallest particle size was obtained in dual-drug suspensions stabilized with a combination of polysorbate 85 and poloxamer 338 compared to dual-drug suspensions stabilized with only a single stabilizer during preparation, indicating tendencies towards stabilization synergism from a combination of stabilizers as well as the model compounds. Conclusion: Overall, the study showed insights into the preparation and physical stability of dual-drug suspensions containing indomethacin and naproxen.
Collapse
Affiliation(s)
| | | | - René Holm
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (N.Z.); (S.M.H.)
| |
Collapse
|
9
|
Zulbeari N, Mustafova SS, Simonsen AC, Lund FW, Holm R. The Langmuir-Blodgett trough (Langmuir film balance) can be used to understand the stabilizer concentrations in aqueous nano- and microsuspensions. Int J Pharm 2024; 665:124726. [PMID: 39293577 DOI: 10.1016/j.ijpharm.2024.124726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
Aqueous suspensions of poorly soluble, crystalline drug particles in the sub-micron range hold the ability to regulate the drug release for a defined period of time after e.g., intramuscular, or subcutaneous administration, working as an eminent formulation strategy for the preparation of long-acting injectables. Aqueous suspensions are typically prepared by top-down approaches, e.g., wet bead media milling or high-pressure homogenization, containing the active pharmaceutical compound and surfactants and/or polymers for stabilization purposes. Currently, the screening of proper stabilizers and adequate stabilizer concentration during formulation investigations is based on a trial-and-error approach with variations in combinations, concentrations, and/or ratios. To obtain a more efficient methodology during formulation screening, the present study investigated the correlation between the surface activity of two different surfactants, i.e., poloxamer 188 and polysorbate 20, by drop profile tensiometry and Langmuir trough monolayer, and the obtained sizes of cinnarizine particles as a tool to predict the optimal surfactant concentration to prepare physical stable nano- and microsuspensions. The obtained results demonstrated that the molecular area determined as the area per surfactant molecule measured in the Langmuir trough combined with the specific surface area of the prepared suspensions could be used to predict the suitable concentration of the surfactant based upon short-term stress stability data. The results further showed that higher concentrations of poloxamer 188 were necessary to stabilize the suspensions when compared to the needed concentration of polysorbate 20. In addition, it was observed that there was a need for a slightly higher surfactant concentration when the suspensions were milled with the smallest bead size of 0.5 mm instead of larger sizes of bead (0.8 and 1.0 mm), which could not be accounted for by differences in specific surface area.
Collapse
Affiliation(s)
- Nadina Zulbeari
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Sibel Selyatinova Mustafova
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Adam Cohen Simonsen
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Frederik Wendelboe Lund
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - René Holm
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
10
|
M.Ravichandran S, M.McFadden W, A.Snyder A, G.Sarafianos S. State of the ART (antiretroviral therapy): Long-acting HIV-1 therapeutics. Glob Health Med 2024; 6:285-294. [PMID: 39483451 PMCID: PMC11514626 DOI: 10.35772/ghm.2024.01049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 11/03/2024]
Abstract
Human immunodeficiency virus (HIV) impacts millions of individuals worldwide, and well over 2/3 of those living with HIV are accessing antiviral therapies that are successfully repressing viral replication. Most often, HIV treatments and prevention are administered in the form of daily pills as combinations of multiple drugs. An emergent and effective strategy for suppressing viral replication is the application of long-acting antiretroviral therapy (LAART), or antivirals that require less-frequent, non-daily doses. Thus far, the repertoire of LAARTs includes the widely used antiviral classes of non-nucleoside reverse transcriptase inhibitors (NNRTIs) and integrase strand transfer inhibitors (INSTIs) and has recently expanded to include a capsid-targeting antiviral. Possible future additions are nucleoside reverse transcriptase inhibitors (NRTIs) and nucleoside reverse transcriptase translocation inhibitors (NRTTIs). Here, we discuss the different strategies of using long-acting compounds to treat or prevent HIV-1 infection by targeting reverse transcriptase, integrase, and capsid.
Collapse
Affiliation(s)
- Shreya M.Ravichandran
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - William M.McFadden
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Alexa A.Snyder
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Stefan G.Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
11
|
Hernandez JL, Chien ST, Doan MA, Suydam IT, Woodrow KA. Antiretroviral (ARV) Properties Dictate Long-Acting Release and Tissue Partitioning Behaviors in Multidrug Subcutaneous Implants. ACS Biomater Sci Eng 2024; 10:6363-6376. [PMID: 39231268 DOI: 10.1021/acsbiomaterials.4c01290] [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] [Indexed: 09/06/2024]
Abstract
Subcutaneous implants can provide patients with long-acting, compliance-independent drug dosing. For this reason, subcutaneous implants have shown emerging interest in human immunodeficiency virus (HIV) prevention. However, any successful long-acting HIV-prevention device will require multidrug dosing, which poses a challenge for formulation considering the physicochemically diverse selection of antiretroviral (ARV) candidates. As a method that has shown the capacity of efficient multidrug delivery, we assessed electrospun fiber implants composed of three synergistically potent ARVs and a biodegradable polymer selected by in vitro release studies. In mice, subcutaneous electrospun fiber implants exhibit burst release of the more hydrophilic drugs maraviroc (MVC) and raltegravir (RAL), which could be reduced via simple prewash treatments of the implants. Over an extended 120 day time frame, fiber implants show drug-specific differences in release time frames and magnitudes in blood serum. However, end-point drug tissue concentrations show that the most hydrophobic drug etravirine (ETR) remains in high concentrations within the implant and in local skin tissue biopsies. Furthermore, ETR is found to be capable of significant partitioning into lymph nodes, the lower female reproductive tract, and the rectum. Topologically smooth film implants also exhibit the same drug-dependent trends. Therefore, we illustrate that drug release and drug tissue partitioning are largely dictated by drug properties. Further, we find that the properties of ETR enable significant drug quantities within the tissues most relevant to HIV protection. Evidence from this work emphasizes the need for a greater focus on drug properties and prodrug strategies to enable relevant, extended, and targeted drug release.
Collapse
Affiliation(s)
- Jamie L Hernandez
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195, United States
| | - Shin-Tian Chien
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195, United States
| | - My-Anh Doan
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195, United States
| | - Ian T Suydam
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195, United States
| |
Collapse
|
12
|
Nguyen HD, Ngo HV, Lee B. Novel pH-Responsive Structural Rearrangement of Myristic Acid-Conjugated Quetiapine Nanosuspension for Enhanced Long-Acting Delivery Performance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405200. [PMID: 39225461 PMCID: PMC11516153 DOI: 10.1002/advs.202405200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Quetiapine myristate (QM), an ester-bonded lipophilic prodrug of quetiapine (QTP), is synthesized and converted into an amphiphilic structure in acidic pH to trigger a novel self-assembled QM nanosuspension (QMN). Following injection, this QMN rearranges within physiological pH to form nanoaggregates in structure, resulting in enhanced physicochemical properties and in vivo therapeutic performance without an initial burst release. The 200-nm-sized QMN exhibits less invasive injection, higher drug content, and better storage stability profile than conventional poly(lactide-co-glycolide) (PLGA) nanosuspensions containing QTP or QM. Following a single intramuscular injection to beagle dogs (35 mg kg-1 QTP), QMN undergoes pH-responsive nanoaggregation to form the lipophilic prodrug, providing esterase-oriented sustained release for five weeks compared with the two-week period of PLGA nanosuspensions. Notably, QMN exhibits improved in vivo pharmacokinetic performance with long-acting delivery while minimizing issues associated with polymeric PLGA formulations, including the initial massive burst release, cellular toxicity, and adverse side effects. These results support the further development of QMN as a novel long-acting injectable to improve patient compliance and dosing frequency.
Collapse
Affiliation(s)
- Hy Dinh Nguyen
- Department of Pharmacy, College of PharmacyAjou UniversitySuwon16499Republic of Korea
| | - Hai Van Ngo
- Department of Pharmacy, College of PharmacyAjou UniversitySuwon16499Republic of Korea
| | - Beom‐Jin Lee
- Department of Pharmacy, College of PharmacyAjou UniversitySuwon16499Republic of Korea
- Institute of Pharmaceutical Science and TechnologyAjou UniversitySuwon16499Republic of Korea
| |
Collapse
|
13
|
Alidori S, Subramanian R, Holm R. Patient-Centric Long-Acting Injectable and Implantable Platforms─An Industrial Perspective. Mol Pharm 2024; 21:4238-4258. [PMID: 39160132 PMCID: PMC11372838 DOI: 10.1021/acs.molpharmaceut.4c00665] [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] [Indexed: 08/21/2024]
Abstract
The increasing focus on patient centricity in the pharmaceutical industry over the past decade and the changing healthcare landscape, driven by factors such as increased access to information, social media, and evolving patient demands, has necessitated a shift toward greater connectivity and understanding of patients' unique treatment needs. One pharmaceutical technology that has supported these efforts is long acting injectables (LAIs), which lower the administration frequency for the patient's provided convenience, better compliance, and hence better therapeutical treatment for the patients. Furthermore, patients with conditions like the human immunodeficiency virus and schizophrenia have positively expressed the desire for less frequent dosing, such as that obtained through LAI formulations. In this work, a comprehensive analysis of marketed LAIs across therapeutic classes and technologies is conducted. The analysis demonstrated an increasing number of new LAIs being brought to the market, recently most as aqueous suspensions and one as a solution, but many other technology platforms were applied as well, in particular, polymeric microspheres and in situ forming gels. The analysis across the technologies provided an insight into to the physicochemical properties the compounds had per technology class as well as knowledge of the excipients typically used within the individual formulation technology. The principle behind the formulation technologies was discussed with respect to the release mechanism, manufacturing approaches, and the possibility of defining predictive in vitro release methods to obtain in vitro in vivo correlations with an industrial angle. The gaps in the field are still numerous, including better systematic formulation and manufacturing investigations to get a better understanding of potential innovations, but also development of new polymers could facilitate the development of additional compounds. The biggest and most important gaps, however, seem to be the development of predictive in vitro dissolution methods utilizing pharmacopoeia described equipment to enable their use for product development and later in the product cycle for quality-based purposes.
Collapse
Affiliation(s)
- Simone Alidori
- Independent Researcher, Havertown, Pennsylvania 19083, United States
| | - Raju Subramanian
- Gilead Sciences, 333 Lakeside Drive, Foster City, California 94403, United States
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
14
|
Bibi D, Bilgraer R, Steiner L, Hallak H. Physiologically-Based Pharmacokinetic Modeling and In Vitro-In Vivo Correlation of TV-46000 (Risperidone LAI): Prediction from Dog to Human. Pharmaceutics 2024; 16:896. [PMID: 39065593 PMCID: PMC11279950 DOI: 10.3390/pharmaceutics16070896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
The interest in the development and therapeutic application of long-acting injectable products for chronic or long-term treatments has experienced exponential growth in recent decades. TV-46000 (Uzedy, Teva) is a long-acting subcutaneous (sc) injectable formulation of risperidone, approved for the treatment of schizophrenia in adults. Following sc injection, the copolymers together with risperidone precipitate to form a sc depot under the skin to deliver therapeutic levels of risperidone over a prolonged period of either 1 month or 2 months, depending upon the dose. This work presents the strategy and the results of the physiologically-based pharmacokinetic (PBPK) modeling and establishing of in vitro-in vivo correlation (IVIVC) for the prediction of TV-46000 pharmacokinetic profile in humans, using in vitro release, intravenous (iv), and sc single-dose pharmacokinetic data in beagle dogs. The resulting simulated TV-46000 PK profile in humans showed that the shape of the predicted risperidone and its active metabolite 9-OH-risperidone PK profiles was different from the observed one, thus suggesting that the TV-46000 release profile was species-dependent and cannot be directly extrapolated from dog to human. In conclusion, while level A IVIVC cannot be claimed, this work combining PBPK and IVIVC modeling represents an interesting alternative approach for complex injectable formulations where classical methods are not applicable.
Collapse
Affiliation(s)
- David Bibi
- Non-Clinical Development, Teva Pharmaceutical Industries Ltd., Netanya 4250400, Israel
| | | | | | | |
Collapse
|
15
|
Jeong MY, Ho MJ, Park JS, Jeong H, Kim JH, Jang YJ, Shin DM, Yang IG, Kim HR, Song WH, Lee S, Song SH, Choi YS, Han YT, Kang MJ. Tricaprylin-based drug crystalline suspension for intramuscular long-acting delivery of entecavir with alleviated local inflammation. Bioeng Transl Med 2024; 9:e10649. [PMID: 39036080 PMCID: PMC11256175 DOI: 10.1002/btm2.10649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 07/23/2024] Open
Abstract
In order to ensure prolonged pharmacokinetic profile along with local tolerability at the injection site, tricaprylin-based drug crystalline suspension (TS) was designed and its local distribution, pharmacokinetics, and inflammatory response, were evaluated with conventional aqueous suspension (AS). As model drug particles, entecavir 3-palmitate (EV-P), an ester lipidic prodrug for entecavir (EV), was employed. The EV-P-loaded TS was prepared by ultra-sonication method. Prepared TS and conventional AS exhibited comparable morphology (rod or rectangular), median diameter (2.7 and 2.6 μm), crystallinity (melting point of 160-165°C), and in vitro dissolution profile. However, in vivo performances of drug microparticles were markedly different, depending on delivery vehicle. At AS-injected site, drug aggregates of up to 500 μm were formed upon intramuscular injection, and were surrounded with inflammatory cells and fibroblastic bands. In contrast, no distinct particle aggregation and adjacent granulation was observed at TS-injected site, with >4 weeks remaining of the oily vehicle in micro-computed tomographic observation. Surprisingly, TS exhibited markedly alleviated local inflammation compared to AS, endowing markedly lessened necrosis, fibrosis thickness, inflammatory area, and macrophage infiltration. The higher initial systemic exposure was observed with TS compared to AS, but TS provided prolonged delivery of EV for 3 weeks. Therefore, we suggest that the novel TS system can be a promising tool in designing parenteral long-acting delivery, with improved local tolerability.
Collapse
Affiliation(s)
- Min Young Jeong
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Myoung Jin Ho
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Joon Soo Park
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Hoetaek Jeong
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Jin Hee Kim
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Yong Jin Jang
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Doe Myung Shin
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - In Gyu Yang
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Hye Rim Kim
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Woo Heon Song
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Sangkil Lee
- College of Pharmacy, Chung‐Ang UniversitySeoulRepublic of Korea
| | - Seh Hyon Song
- College of Pharmacy, Kyungsung UniversityBusanRepublic of Korea
| | - Yong Seok Choi
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Young Taek Han
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| | - Myung Joo Kang
- College of Pharmacy, Dankook UniversityCheonanChungnamRepublic of Korea
| |
Collapse
|
16
|
Tang Y, Liu B, Zhang Y, Liu Y, Huang Y, Fan W. Interactions between nanoparticles and lymphatic systems: Mechanisms and applications in drug delivery. Adv Drug Deliv Rev 2024; 209:115304. [PMID: 38599495 DOI: 10.1016/j.addr.2024.115304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
The lymphatic system has garnered significant attention in drug delivery research due to the advantages it offers, such as enhancing systemic exposure and enabling lymph node targeting for nanomedicines via the lymphatic delivery route. The journey of drug carriers involves transport from the administration site to the lymphatic vessels, traversing the lymph before entering the bloodstream or targeting specific lymph nodes. However, the anatomical and physiological barriers of the lymphatic system play a pivotal role in influencing the behavior and efficiency of carriers. To expedite research and subsequent clinical translation, this review begins by introducing the composition and classification of the lymphatic system. Subsequently, we explore the routes and mechanisms through which nanoparticles enter lymphatic vessels and lymph nodes. The review further delves into the interactions between nanomedicine and body fluids at the administration site or within lymphatic vessels. Finally, we provide a comprehensive overview of recent advancements in lymphatic delivery systems, addressing the challenges and opportunities inherent in current systems for delivering macromolecules and vaccines.
Collapse
Affiliation(s)
- Yisi Tang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| | - Wufa Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
17
|
Bettonte S, Berton M, Battegay M, Stader F, Marzolini C. Development of a physiologically-based pharmacokinetic model to simulate the pharmacokinetics of intramuscular antiretroviral drugs. CPT Pharmacometrics Syst Pharmacol 2024; 13:781-794. [PMID: 38429889 PMCID: PMC11098154 DOI: 10.1002/psp4.13118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
There is growing interest in the use of long-acting (LA) injectable drugs to improve treatment adherence. However, their long elimination half-life complicates the conduct of clinical trials. Physiologically-based pharmacokinetic (PBPK) modeling is a mathematical tool that allows to simulate unknown clinical scenarios for LA formulations. Thus, this work aimed to develop and verify a mechanistic intramuscular PBPK model. The framework describing the release of a LA drug from the depot was developed by including both the physiology of the injection site and the physicochemical properties of the drug. The framework was coded in Matlab® 2020a and implemented in our existing PBPK model for the verification step using clinical data for LA cabotegravir, rilpivirine, and paliperidone. The model was considered verified when the simulations were within twofold of observed data. Furthermore, a local sensitivity analysis was conducted to assess the impact of various factors relevant for the drug release from the depot on pharmacokinetics. The PBPK model was successfully verified since all predictions were within twofold of observed clinical data. Peak concentration, area under the concentration-time curve, and trough concentration were sensitive to media viscosity, drug solubility, drug density, and diffusion layer thickness. Additionally, inflammation was shown to impact the drug release from the depot. The developed framework correctly described the release and the drug disposition of LA formulations upon intramuscular administration. It can be implemented in PBPK models to address pharmacological questions related to the use of LA formulations.
Collapse
Affiliation(s)
- Sara Bettonte
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical ResearchUniversity Hospital BaselBaselSwitzerland
- Faculty of MedicineUniversity of BaselBaselSwitzerland
| | - Mattia Berton
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical ResearchUniversity Hospital BaselBaselSwitzerland
- Faculty of MedicineUniversity of BaselBaselSwitzerland
| | - Manuel Battegay
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical ResearchUniversity Hospital BaselBaselSwitzerland
- Faculty of MedicineUniversity of BaselBaselSwitzerland
| | | | - Catia Marzolini
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical ResearchUniversity Hospital BaselBaselSwitzerland
- Faculty of MedicineUniversity of BaselBaselSwitzerland
- Department of Molecular and Clinical PharmacologyUniversity of LiverpoolLiverpoolUK
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and PathologyUniversity Hospital Lausanne and University of LausanneLausanneSwitzerland
| |
Collapse
|
18
|
Thoueille P, Saldanha SA, Schaller F, Choong E, Munting A, Cavassini M, Braun D, Günthard HF, Kusejko K, Surial B, Furrer H, Rauch A, Rougemont M, Ustero P, Calmy A, Stöckle M, Marzolini C, Di Benedetto C, Bernasconi E, Schmid P, Piso RJ, Andre P, Girardin FR, Guidi M, Buclin T, Decosterd LA. Real-world trough concentrations and effectiveness of long-acting cabotegravir and rilpivirine: a multicenter prospective observational study in Switzerland. THE LANCET REGIONAL HEALTH. EUROPE 2024; 36:100793. [PMID: 38162253 PMCID: PMC10757247 DOI: 10.1016/j.lanepe.2023.100793] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024]
Abstract
Background The efficacy and tolerability of long-acting cabotegravir and rilpivirine were demonstrated in Phase III trials. However, low concentrations combined with other risk factors have been associated with an increased risk of virologic failure. This study aims to verify whether drug concentrations measured in a real-world setting are consistent with those previously reported. Methods SHCS-879 is a nationwide observational study within the Swiss HIV Cohort Study for the monitoring of people with HIV (PWH) on long-acting cabotegravir plus rilpivirine. Samples were collected from March 2022 to March 2023. Findings Overall, 725 samples were obtained from 186 PWH. Our data show a large inter-individual variability in cabotegravir and rilpivirine concentrations, with some individuals exhibiting repeatedly low concentrations. Rilpivirine trough concentrations were consistent with those from Phase III trials, while cabotegravir concentrations were lower. The first concentrations quartile was only slightly above the target of 664 ng/mL. Exploratory statistical analyses found 35% (p < 0·01) lower cabotegravir trough in males compared to females. Overall, 172 PWH (92%) remained suppressed and three experienced virologic failures (1·6%), of those, two had sub-optimal drug exposure. No association was found between low trough levels and detectable viral load. Interpretation Real-world cabotegravir concentrations are substantially lower than previously reported. However, these concentrations appear sufficient to ensure sustained virological suppression in almost every PWH. These reassuring data challenge the rather conservative thresholds adopted to date, which may raise unnecessary concerns. Yet, our study reveals that some PWH have repeatedly very low drug levels, for reasons that remain to be elucidated. Funding This work was funded by the Swiss National Science Foundation, grant number N◦ 324730_192449. This study received no support from pharmaceutical industries. This study was performed within the framework of the Swiss HIV Cohort Study, supported by the Swiss National Science Foundation (grant #201369), by SHCS project #879, and by the SHCS research foundation. The SHCS data were gathered by the Five Swiss University Hospitals, two Cantonal Hospitals, 15 affiliated hospitals and 36 private physicians (listed in http://www.shcs.ch/180-health-care-providers).
Collapse
Affiliation(s)
- Paul Thoueille
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Susana Alves Saldanha
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fabian Schaller
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eva Choong
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Aline Munting
- Service of Infectious Diseases, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Matthias Cavassini
- Service of Infectious Diseases, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dominique Braun
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Katharina Kusejko
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Bernard Surial
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hansjakob Furrer
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andri Rauch
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Mathieu Rougemont
- Primary Care Medicine Division, University Hospital Geneva, Geneva, Switzerland
| | - Pilar Ustero
- Division of Infectious Diseases, Geneva University Hospitals, Faculty of Medicine, Geneva, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, Geneva University Hospitals, Faculty of Medicine, Geneva, Switzerland
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marcel Stöckle
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Catia Marzolini
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Enos Bernasconi
- Division of Infectious Diseases, Ente Ospedaliero Cantonale, Lugano, Switzerland
- University of Geneva, University of Southern Switzerland, Lugano, Switzerland
| | - Patrick Schmid
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Rein Jan Piso
- Department of Internal Medicine, Infectious Diseases and Hospital Epidemiology, Cantonal Hospital Olten, Switzerland
| | - Pascal Andre
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - François R. Girardin
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Monia Guidi
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Thierry Buclin
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurent A. Decosterd
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - The Swiss HIV Cohort Study
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Infectious Diseases, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Primary Care Medicine Division, University Hospital Geneva, Geneva, Switzerland
- Division of Infectious Diseases, Geneva University Hospitals, Faculty of Medicine, Geneva, Switzerland
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Division of Infectious Diseases, Ente Ospedaliero Cantonale, Lugano, Switzerland
- University of Geneva, University of Southern Switzerland, Lugano, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
- Department of Internal Medicine, Infectious Diseases and Hospital Epidemiology, Cantonal Hospital Olten, Switzerland
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
| |
Collapse
|
19
|
McGowan A, Gennemark P, Akieh-Pirkanniemi M, Wirman L, Davies N, Elebring M, Tivesten A, Strimfors M, Hölttä M, Söderberg M, Berntsson V, Balas D, Koskinen M, Leino L, Abrahmsén-Alami S. Injectable Biodegradable Silica Depot for Controlled Subcutaneous Delivery of Antisense Oligonucleotides with beyond Monthly Administration. Mol Pharm 2024; 21:143-151. [PMID: 38126776 DOI: 10.1021/acs.molpharmaceut.3c00663] [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] [Indexed: 12/23/2023]
Abstract
Single-stranded antisense oligonucleotides (ASOs) are typically administered subcutaneously once per week or monthly. Less frequent dosing would have strong potential to improve patient convenience and increase adherence and thereby for some diseases result in more optimal therapeutic outcomes. Several technologies are available to provide sustained drug release via subcutaneous (SC) administration. ASOs have a high aqueous solubility and require relatively high doses, which limits the options available substantially. In the present work, we show that an innovative biodegradable, nonporous silica-based matrix provides zero-order release in vivo (rats) for at least 4 weeks for compositions with ASO loads of up to about 100 mg/mL (0.5 mL injection) without any sign of initial burst. This implies that administration beyond once monthly can be feasible. For higher drug loads, substantial burst release was observed during the first week. The concentrations of unconjugated ASO levels in the liver were found to be comparable to corresponding bolus doses. Additionally, infusion using a minipump shows a higher liver exposure than SC bolus administration at the same dose level and, in addition, clear mRNA knockdown and circulating protein reduction comparable to SC bolus dosing, hence suggesting productive liver uptake for a slow-release administration.
Collapse
Affiliation(s)
- Asmaa McGowan
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Peter Gennemark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
- Department of Biomedical Engineering, Linköping University, Linköping 581 83, Sweden
| | | | - Linda Wirman
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Marie Elebring
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Anna Tivesten
- CVRM CMC Projects, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Marie Strimfors
- Bioscience metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Mikko Hölttä
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Magnus Söderberg
- Pathology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Veronica Berntsson
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Daniela Balas
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| | - Mika Koskinen
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Lasse Leino
- DelSiTech Ltd., PharmaCity, Itäinen Pitkäkatu 4 B, 20520 Turku, Finland
| | - Susanna Abrahmsén-Alami
- Sustainable Innovation & Transformational Excellence, Pharmaceutical Technology & Development, Operations, AstraZeneca, Mölndal, Gothenburg SE-431 83, Sweden
| |
Collapse
|
20
|
Liu C, Hern FY, Shakil A, Temburnikar K, Chambon P, Liptrott N, McDonald TO, Neary M, Flexner C, Owen A, Meyers CF, Rannard SP. Polymer-prodrug conjugates as candidates for degradable, long-acting implants, releasing the water-soluble nucleoside reverse-transcriptase inhibitor emtricitabine. J Mater Chem B 2023; 11:11532-11543. [PMID: 37955203 PMCID: PMC10718295 DOI: 10.1039/d3tb02268d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Circulating, soluble polymer-drug conjugates have been utilised for many years to aid the delivery of sensitive, poorly-soluble or cytotoxic drugs, prolong circulation times or minimise side effects. Long-acting therapeutics are increasing in their healthcare importance, with intramuscular and subcutaneous administration of liquid formulations being most common. Degradable implants also offer opportunities and the use of polymer-prodrug conjugates as implant materials has not been widely reported in this context. Here, the potential for polymer-prodrug conjugates of the water soluble nucleoside reverse transciption inhibitor emtricitabine (FTC) is studied. A novel diol monomer scaffold, allowing variation of prodrug substitution, has been used to form polyesters and polycarbonates by step-growth polymerisation. Materials have been screened for physical properties that enable implant formation, studied for drug release to provide mechanistic insights, and tunable prolonged release of FTC has been demonstrated over a period of at least two weeks under relevant physiological conditions.
Collapse
Affiliation(s)
- Chung Liu
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Faye Y Hern
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Anika Shakil
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Kartik Temburnikar
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Pierre Chambon
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Neill Liptrott
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Tom O McDonald
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Megan Neary
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Charles Flexner
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Andrew Owen
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Caren Freel Meyers
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Steve P Rannard
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| |
Collapse
|
21
|
Subramanian R, Tang J, Zheng J, Lu B, Wang K, Yant SR, Stepan GJ, Mulato A, Yu H, Schroeder S, Shaik N, Singh R, Wolckenhauer S, Chester A, Tse WC, Chiu A, Rhee M, Cihlar T, Rowe W, Smith BJ. Lenacapavir: A Novel, Potent, and Selective First-in-Class Inhibitor of HIV-1 Capsid Function Exhibits Optimal Pharmacokinetic Properties for a Long-Acting Injectable Antiretroviral Agent. Mol Pharm 2023; 20:6213-6225. [PMID: 37917742 PMCID: PMC10698746 DOI: 10.1021/acs.molpharmaceut.3c00626] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/04/2023]
Abstract
Lenacapavir (LEN) is a picomolar first-in-class capsid inhibitor of human immunodeficiency virus type 1 (HIV-1) with a multistage mechanism of action and no known cross resistance to other existing antiretroviral (ARV) drug classes. LEN exhibits a low aqueous solubility and exceptionally low systemic clearance following intravenous (IV) administration in nonclinical species and humans. LEN formulated in an aqueous suspension or a PEG/water solution formulation showed sustained plasma exposure levels with no unintended rapid drug release following subcutaneous (SC) administration to rats and dogs. A high total fraction dose release was observed with both formulations. The long-acting pharmacokinetics (PK) were recapitulated in humans following SC administration of both formulations. The SC PK profiles displayed two-phase absorption kinetics in both animals and humans with an initial fast-release absorption phase, followed by a slow-release absorption phase. Noncompartmental and compartmental analyses informed the LEN systemic input rate from the SC depot and exit rate from the body. Modeling-enabled deconvolution of the input rates from two processes: absorption of the soluble fraction (minor) from a direct fast-release process leading to the early PK phase and absorption of the precipitated fraction (major) from an indirect slow-release process leading to the later PK phase. LEN SC PK showed flip-flop kinetics due to the input rate being substantially slower than the systemic exit rate. LEN input rates via the slow-release process in humans were slower than those in both rats and dogs. Overall, the combination of high potency, exceptional stability, and optimal release rate from the injection depot make LEN well suited for a parenteral long-acting formulation that can be administered once up to every 6 months in humans for the prevention and treatment of HIV-1.
Collapse
Affiliation(s)
- Raju Subramanian
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jennifer Tang
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | | | - Bing Lu
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Kelly Wang
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Stephen R. Yant
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - George J. Stepan
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Andrew Mulato
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Helen Yu
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Scott Schroeder
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Naveed Shaik
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Renu Singh
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | | | - Anne Chester
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | | | - Anna Chiu
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Martin Rhee
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Tomas Cihlar
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - William Rowe
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Bill J. Smith
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| |
Collapse
|
22
|
Balta VA, Stiffler D, Sayeed A, Tripathi AK, Elahi R, Mlambo G, Bakshi RP, Dziedzic AG, Jedlicka AE, Nenortas E, Romero-Rodriguez K, Canonizado MA, Mann A, Owen A, Sullivan DJ, Prigge ST, Sinnis P, Shapiro TA. Clinically relevant atovaquone-resistant human malaria parasites fail to transmit by mosquito. Nat Commun 2023; 14:6415. [PMID: 37828012 PMCID: PMC10570281 DOI: 10.1038/s41467-023-42030-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Long-acting injectable medications, such as atovaquone, offer the prospect of a "chemical vaccine" for malaria, combining drug efficacy with vaccine durability. However, selection and transmission of drug-resistant parasites is of concern. Laboratory studies have indicated that atovaquone resistance disadvantages parasites in mosquitoes, but lack of data on clinically relevant Plasmodium falciparum has hampered integration of these variable findings into drug development decisions. Here we generate atovaquone-resistant parasites that differ from wild type parent by only a Y268S mutation in cytochrome b, a modification associated with atovaquone treatment failure in humans. Relative to wild type, Y268S parasites evidence multiple defects, most marked in their development in mosquitoes, whether from Southeast Asia (Anopheles stephensi) or Africa (An. gambiae). Growth of asexual Y268S P. falciparum in human red cells is impaired, but parasite loss in the mosquito is progressive, from reduced gametocyte exflagellation, to smaller number and size of oocysts, and finally to absence of sporozoites. The Y268S mutant fails to transmit from mosquitoes to mice engrafted with human liver cells and erythrocytes. The severe-to-lethal fitness cost of clinically relevant atovaquone resistance to P. falciparum in the mosquito substantially lessens the likelihood of its transmission in the field.
Collapse
Affiliation(s)
- Victoria A Balta
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Deborah Stiffler
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Abeer Sayeed
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Abhai K Tripathi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Rubayet Elahi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Godfree Mlambo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Rahul P Bakshi
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186, USA
| | - Amanda G Dziedzic
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Anne E Jedlicka
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Elizabeth Nenortas
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186, USA
| | - Keyla Romero-Rodriguez
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186, USA
| | - Matthew A Canonizado
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186, USA
| | - Alexis Mann
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Andrew Owen
- Centre of Excellence in Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, L69 3BX, UK
| | - David J Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Sean T Prigge
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Photini Sinnis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Theresa A Shapiro
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA.
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186, USA.
| |
Collapse
|
23
|
Pandya AK, Vora LK, Umeyor C, Surve D, Patel A, Biswas S, Patel K, Patravale VB. Polymeric in situ forming depots for long-acting drug delivery systems. Adv Drug Deliv Rev 2023; 200:115003. [PMID: 37422267 DOI: 10.1016/j.addr.2023.115003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023]
Abstract
Polymeric in situ forming depots have emerged as highly promising drug delivery systems for long-acting applications. Their effectiveness is attributed to essential characteristics such as biocompatibility, biodegradability, and the ability to form a stable gel or solid upon injection. Moreover, they provide added versatility by complementing existing polymeric drug delivery systems like micro- and nanoparticles. The formulation's low viscosity facilitates manufacturing unit operations and enhances delivery efficiency, as it can be easily administered via hypodermic needles. The release mechanism of drugs from these systems can be predetermined using various functional polymers. To enable unique depot design, numerous strategies involving physiological and chemical stimuli have been explored. Important assessment criteria for in situ forming depots include biocompatibility, gel strength and syringeability, texture, biodegradation, release profile, and sterility. This review focuses on the fabrication approaches, key evaluation parameters, and pharmaceutical applications of in situ forming depots, considering perspectives from academia and industry. Additionally, insights about the future prospects of this technology are discussed.
Collapse
Affiliation(s)
- Anjali K Pandya
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India; School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Chukwuebuka Umeyor
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 422001, Anambra State, Nigeria
| | - Dhanashree Surve
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Akanksha Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, Telangana 500078, India
| | - Ketankumar Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Vandana B Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India.
| |
Collapse
|
24
|
Sharma R, Yadav S, Yadav V, Akhtar J, Katari O, Kuche K, Jain S. Recent advances in lipid-based long-acting injectable depot formulations. Adv Drug Deliv Rev 2023; 199:114901. [PMID: 37257756 DOI: 10.1016/j.addr.2023.114901] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/28/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Long-acting injectable (LAIs) delivery systems sustain the drug therapeutic action in the body, resulting in reduced dosage regimen, toxicity, and improved patient compliance. Lipid-based depots are biocompatible, provide extended drug release, and improve drug stability, making them suitable for systemic and localized treatment of various chronic ailments, including psychosis, diabetes, hormonal disorders, arthritis, ocular diseases, and cancer. These depots include oil solutions, suspensions, oleogels, liquid crystalline systems, liposomes, solid lipid nanoparticles, nanostructured lipid carriers, phospholipid phase separation gel, vesicular phospholipid gel etc. This review summarizes recent advancements in lipid-based LAIs for delivering small and macromolecules, and their potential in managing chronic diseases. It also provides an overview of the lipid depots available in market or clinical phase, as well as patents for lipid-based LAIs. Furthermore, this review critically discusses the current scenario of using in vitro release methods to establish IVIVC and highlights the challenges involved in developing lipid-based LAIs.
Collapse
Affiliation(s)
- Reena Sharma
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Sheetal Yadav
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Vivek Yadav
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Junia Akhtar
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Oly Katari
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Kaushik Kuche
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Sanyog Jain
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India.
| |
Collapse
|
25
|
Bauer A, Berben P, Chakravarthi SS, Chattorraj S, Garg A, Gourdon B, Heimbach T, Huang Y, Morrison C, Mundhra D, Palaparthy R, Saha P, Siemons M, Shaik NA, Shi Y, Shum S, Thakral NK, Urva S, Vargo R, Koganti VR, Barrett SE. Current State and Opportunities with Long-acting Injectables: Industry Perspectives from the Innovation and Quality Consortium "Long-Acting Injectables" Working Group. Pharm Res 2023; 40:1601-1631. [PMID: 36811809 DOI: 10.1007/s11095-022-03391-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/06/2022] [Indexed: 02/24/2023]
Abstract
Long-acting injectable (LAI) formulations can provide several advantages over the more traditional oral formulation as drug product opportunities. LAI formulations can achieve sustained drug release for extended periods of time, which results in less frequent dosing requirements leading to higher patient adherence and more optimal therapeutic outcomes. This review article will provide an industry perspective on the development and associated challenges of long-acting injectable formulations. The LAIs described herein include polymer-based formulations, oil-based formulations, and crystalline drug suspensions. The review discusses manufacturing processes, including quality controls, considerations of the Active Pharmaceutical Ingredient (API), biopharmaceutical properties and clinical requirements pertaining to LAI technology selection, and characterization of LAIs through in vitro, in vivo and in silico approaches. Lastly, the article includes a discussion around the current lack of suitable compendial and biorelevant in vitro models for the evaluation of LAIs and its subsequent impact on LAI product development and approval.
Collapse
Affiliation(s)
- Andrea Bauer
- Sunovion Pharmaceuticals, Marlborough, MA, 01752, USA
| | | | | | | | - Ashish Garg
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Ye Huang
- AbbVie Inc., North Chicago, IL, 60064, USA
| | | | | | | | - Pratik Saha
- GlaxoSmithKline, Collegeville, PA, 19426, USA
| | - Maxime Siemons
- Janssen R&D, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | | | - Yi Shi
- AbbVie Inc., North Chicago, IL, 60064, USA
| | - Sara Shum
- Takeda Development Center Americas, Inc., Cambridge, MA, 02139, USA
| | | | - Shweta Urva
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Ryan Vargo
- Merck & Co., Inc., Rahway, NJ, 07065, USA
| | | | | |
Collapse
|
26
|
Coulter SM, Pentlavalli S, Vora LK, An Y, Cross ER, Peng K, McAulay K, Schweins R, Donnelly RF, McCarthy HO, Laverty G. Enzyme-Triggered l-α/d-Peptide Hydrogels as a Long-Acting Injectable Platform for Systemic Delivery of HIV/AIDS Drugs. Adv Healthc Mater 2023; 12:e2203198. [PMID: 36880399 PMCID: PMC11469249 DOI: 10.1002/adhm.202203198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/24/2023] [Indexed: 03/08/2023]
Abstract
Eradicating HIV/AIDS by 2030 is a central goal of the World Health Organization. Patient adherence to complicated dosage regimens remains a key barrier. There is a need for convenient long-acting formulations that deliver drugs over sustained periods. This paper presents an alternative platform, an injectable in situ forming hydrogel implant to deliver a model antiretroviral drug (zidovudine [AZT]) over 28 days. The formulation is a self-assembling ultrashort d or l-α peptide hydrogelator, namely phosphorylated (naphthalene-2-ly)-acetyl-diphenylalanine-lysine-tyrosine-OH (NapFFKY[p]-OH), covalently conjugated to zidovudine via an ester linkage. Rheological analysis demonstrates phosphatase enzyme instructed self-assembly, with hydrogels forming within minutes. Small angle neutron scattering data suggest hydrogels form narrow radius (≈2 nm), large length fibers closely fitting the flexible cylinder elliptical model. d-Peptides are particularly promising for long-acting delivery, displaying protease resistance for 28 days. Drug release, via hydrolysis of the ester linkage, progress under physiological conditions (37 °C, pH 7.4, H2 O). Subcutaneous administration of Napffk(AZT)Y[p]G-OH in Sprague Dawley rats demonstrate zidovudine blood plasma concentrations within the half maximal inhibitory concentration (IC50 ) range (30-130 ng mL-1 ) for 35 days. This work is a proof-of-concept for the development of a long-acting combined injectable in situ forming peptide hydrogel implant. These products are imperative given their potential impact on society.
Collapse
Affiliation(s)
- Sophie M. Coulter
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Sreekanth Pentlavalli
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Lalitkumar K. Vora
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Yuming An
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Emily R. Cross
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Ke Peng
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Kate McAulay
- School of ChemistryUniversity of GlasgowJoseph Black BuildingGlasgowScotlandG12 8QQUK
- School of Computing, Engineering and Built EnvironmentGlasgow Caledonian UniversityGlasgowScotlandG4 0BAUK
| | - Ralf Schweins
- Large Scale Structures GroupInstitut Laue – Langevin71 Avenue des Martyrs, CS 20156Grenoble Cedex 938042France
| | - Ryan F. Donnelly
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Helen O. McCarthy
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| | - Garry Laverty
- School of PharmacyQueen's University BelfastMedical Biology Centre97 Lisburn RoadBelfast, Co. AntrimNorthern IrelandBT9 7BLUK
| |
Collapse
|
27
|
Chien ST, Suydam IT, Woodrow KA. Prodrug approaches for the development of a long-acting drug delivery systems. Adv Drug Deliv Rev 2023; 198:114860. [PMID: 37160248 PMCID: PMC10498988 DOI: 10.1016/j.addr.2023.114860] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Long-acting formulations are designed to reduce dosing frequency and simplify dosing schedules by providing an extended duration of action. One approach to obtain long-acting formulations is to combine long-acting prodrugs (LA-prodrug) with existing or emerging drug delivery technologies (DDS). The design criteria for long-acting prodrugs are distinct from conventional prodrug strategies that alter absorption, distribution, metabolism, and excretion (ADME) parameters. Our review focuses on long-acting prodrug delivery systems (LA-prodrug DDS), which is a subcategory of long-acting formulations where prodrug design enables DDS formulation to achieve an extended duration of action that is greater than the parent drug. Here, we define LA-prodrugs as the conjugation of an active pharmaceutical ingredient (API) to a promoiety group via a cleavable covalent linker, where both the promoiety and linker are selected to enable formulation and administration from a drug delivery system (DDS) to achieve an extended duration of action. These LA-prodrug DDS results in an extended interval where the API is within a therapeutic range without necessarily altering ADME as is typical of conventional prodrugs. The conversion of the LA-prodrug to the API is dependent on linker cleavage, which can occur before or after release from the DDS. The requirement for linker cleavage provides an additional tool to prolong release from these LA-prodrug DDS. In addition, the physicochemical properties of drugs can be tuned by promoiety selection for a particular DDS. Conjugation with promoieties that are carriers or amenable to assembly into carriers can also provide access to formulations designed for extending duration of action. LA-prodrugs have been applied to a wide variety of drug delivery strategies and are categorized in this review by promoiety size and complexity. Small molecule promoieties (typically MW < 1000 Da) have been used to improve encapsulation or partitioning as well as broaden APIs for use with traditional long-acting formulations such as solid drug dispersions. Macromolecular promoieties (typically MW > 1000 Da) have been applied to hydrogels, nanoparticles, micelles, dendrimers, and polymerized prodrug monomers. The resulting LA-prodrug DDS enable extended duration of action for active pharmaceuticals across a wide range of applications, with target release timescales spanning days to years.
Collapse
Affiliation(s)
- Shin-Tian Chien
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Ian T Suydam
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| |
Collapse
|
28
|
Corpstein CD, Li T. A Perspective on Model-Informed IVIVC for Development of Subcutaneous Injectables. Pharm Res 2023; 40:1633-1639. [PMID: 37523013 DOI: 10.1007/s11095-023-03572-3] [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: 02/27/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Subcutaneously administered drugs are growing in popularity for both large and small molecule drugs. However, development of these systems - particularly generics - is slowed due to a lack of formal guidance regarding preclinical testing and in vitro - in vivo correlations (IVIVC). Many of these methods, while appropriate for oral drugs, may not be optimized for the complex injection site physiologies, and release rate and absorption mechanisms of subcutaneous drugs. Current limitations for formulation design and IVIVC can be supported by implementing mechanistic, computational methods. These methods can help to inform drug development by identifying key drug and formulation attributes, and their effects on drug release rates. This perspective, therefore, addresses current guidelines in place for oral IVIVC development, how they may differ for subcutaneously administered compounds, and how modeling and simulation can be implemented to inform design of these products. As such, integration of modeling and simulation with current IVIVC systems can help in driving the development of subcutaneous injectables.
Collapse
Affiliation(s)
- Clairissa D Corpstein
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | - Tonglei Li
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
29
|
Hugo Silva M, Hudson SP, Tajber L, Garin M, Dong W, Khamiakova T, Holm R. Osmolality of Excipients for Parenteral Formulation Measured by Freezing Point Depression and Vapor Pressure - A Comparative Analysis. Pharm Res 2023; 40:1709-1722. [PMID: 35460023 DOI: 10.1007/s11095-022-03262-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE To investigate the difference in methods to determine the osmolality in solutions of stabilizers used for long-acting injectable suspensions. METHODS The osmolality was measured by freezing point depression and vapor pressure for 11 different polymers and surfactants (PEG 3350, 4000, 6000, 8000, 20,000, PVP K12, K17 and K30, poloxamer 188, 388 and 407, HPMC E5, Na-CMC, polysorbate 20 and 80, vitamin E-TPGS, phospholipid, DOSS and SDS) in different concentrations. RESULTS Independently of the measuring method, an increase in osmolality with increasing concentration was observed for all polymers and surfactants, as would be expected due to the physicochemical origin of the osmolality. No correlation was found between the molecular weight of the polymers and the measured osmolality. The osmolality values were different for PVPs, PEGs, and Na-CMC using the two different measurement methods. The values obtained by the freezing point depression method tended to be similar or higher than the ones provided by vapor pressure, overall showing a significant difference in the osmolality measured by the two investigated methods. CONCLUSIONS For lower osmolality values (e.g. surfactants), the choice of the measuring method was not critical, both the freezing point depression and vapor pressure could be used. However, when the formulations contained higher concentrations of excipients and/or thermosensitive excipients, the data suggests that the vapor pressure method would be more suited.
Collapse
Affiliation(s)
- Mariana Hugo Silva
- Pharmaceutical Product Development and Supply, Janssen Research and Development, Johnson & Johnson, Beerse, Belgium
- Department of Chemical Sciences, SSPC the Science Foundation Ireland Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Castletroy, Co. Limerick, Ireland
| | - Sarah P Hudson
- Department of Chemical Sciences, SSPC the Science Foundation Ireland Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Castletroy, Co. Limerick, Ireland
| | - Lidia Tajber
- School of Pharmacy and Pharmaceutical Sciences, SSPC the Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Dublin 2, College Green, Ireland
| | - Matthieu Garin
- Pharmaceutical Product Development and Supply, Janssen Research and Development, Johnson & Johnson, Beerse, Belgium
| | - Wenyu Dong
- Pharmaceutical Product Development and Supply, Janssen Research and Development, Johnson & Johnson, Beerse, Belgium
| | - Tatsiana Khamiakova
- Pharmaceutical Product Development and Supply, Janssen Research and Development, Johnson & Johnson, Beerse, Belgium
| | - René Holm
- Pharmaceutical Product Development and Supply, Janssen Research and Development, Johnson & Johnson, Beerse, Belgium.
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
| |
Collapse
|
30
|
Holm R, Lee RW, Glassco J, DiFranco N, Bao Q, Burgess DJ, Lukacova V, Alidori S. Long-Acting Injectable Aqueous Suspensions-Summary From an AAPS Workshop. AAPS J 2023; 25:49. [PMID: 37118621 DOI: 10.1208/s12248-023-00811-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/12/2023] [Indexed: 04/30/2023] Open
Abstract
Through many years of clinical application of long-acting injectables, there is clear proof that this type of formulation does not just provide the patient with convenience, but more importantly a more effective treatment of the medication provided. The formulation approach therefore contains huge untapped potential to improve the quality of life of many patients with a variety of different diseases. This review provides a summary of some of the central talks provided at the workshop with focus on aqueous suspensions and their use as a long-acting injectable. Elements as formulation, manufacturing, in vitro dissolution methods, in vitro and in vivo correlation, in silico modelling provide an insight into some of the current understandings, learnings, and not least gaps in the field.
Collapse
Affiliation(s)
- René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
| | - Robert W Lee
- Lubrizol Life Science, Health, CDMO Division, 3894 Courtney St., Bethlehem, Pennsylvania, 18017, USA
| | - Joey Glassco
- Lubrizol Life Science, Health: 9911 Brecksville Road, Cleveland, Ohio, 44141, USA
| | - Nicholas DiFranco
- Lubrizol Life Science, Health: 9911 Brecksville Road, Cleveland, Ohio, 44141, USA
| | - Quanying Bao
- School of Pharmacy, University of Connecticut, Storrs, Connecticut, 06269, USA
| | - Diane J Burgess
- School of Pharmacy, University of Connecticut, Storrs, Connecticut, 06269, USA
| | - Viera Lukacova
- Simulations Plus, Inc., 42505 10Th Street, Lancaster, California, 93534, USA
| | - Simone Alidori
- GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, Pennsylvania, 19426-2990, USA
| |
Collapse
|
31
|
Balta VA, Stiffler D, Sayeed A, Tripathi AK, Elahi R, Mlambo G, Bakshi RP, Dziedzic AG, Jedlicka AE, Nenortas E, Romero-Rodriguez K, Canonizado MA, Mann A, Owen A, Sullivan DJ, Prigge ST, Sinnis P, Shapiro TA. Transmissibility of clinically relevant atovaquone-resistant Plasmodium falciparum by anopheline mosquitoes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527535. [PMID: 36798298 PMCID: PMC9934642 DOI: 10.1101/2023.02.07.527535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Rising numbers of malaria cases and deaths underscore the need for new interventions. Long-acting injectable medications, such as those now in use for HIV prophylaxis, offer the prospect of a malaria "chemical vaccine", combining the efficacy of a drug (like atovaquone) with the durability of a biological vaccine. Of concern, however, is the possible selection and transmission of drug-resistant parasites. We addressed this question by generating clinically relevant, highly atovaquone-resistant, Plasmodium falciparum mutants competent to infect mosquitoes. Isogenic paired strains, that differ only by a single Y268S mutation in cytochrome b, were evaluated in parallel in southeast Asian (Anopheles stephensi) or African (Anopheles gambiae) mosquitoes, and thence in humanized mice. Fitness costs of the mutation were evident along the lifecycle, in asexual parasite growth in vitro and in a progressive loss of parasites in the mosquito. In numerous independent experiments, microscopic exam of salivary glands from hundreds of mosquitoes failed to detect even one Y268S sporozoite, a defect not rescued by coinfection with wild type parasites. Furthermore, despite uniformly successful transmission of wild type parasites from An. stephensi to FRG NOD huHep mice bearing human hepatocytes and erythrocytes, multiple attempts with Y268S-fed mosquitoes failed: there was no evidence of parasites in mouse tissues by microscopy, in vitro culture, or PCR. These studies confirm a severe-to-lethal fitness cost of clinically relevant atovaquone-resistant P. falciparum in the mosquito, and they significantly lessen the likelihood of their transmission in the field.
Collapse
Affiliation(s)
- Victoria A. Balta
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Deborah Stiffler
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Abeer Sayeed
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Abhai K. Tripathi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Rubayet Elahi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Godfree Mlambo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Rahul P. Bakshi
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186
| | - Amanda G. Dziedzic
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
| | - Anne E. Jedlicka
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
| | - Elizabeth Nenortas
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186
| | - Keyla Romero-Rodriguez
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186
| | - Matthew A. Canonizado
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186
| | - Alexis Mann
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Andrew Owen
- Centre of Excellence in Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3BX, UK
| | - David J. Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Sean T. Prigge
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Photini Sinnis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
| | - Theresa A. Shapiro
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, 21205
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, 21205-2186
| |
Collapse
|
32
|
Nguyen VTT, Darville N, Vermeulen A. Pharmacokinetics of Long-Acting Aqueous Nano-/Microsuspensions After Intramuscular Administration in Different Animal Species and Humans-a Review. AAPS J 2022; 25:4. [PMID: 36456852 DOI: 10.1208/s12248-022-00771-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/21/2022] [Indexed: 12/04/2022] Open
Abstract
Formulating aqueous suspensions is an attractive strategy to incorporate poorly water-soluble drugs, where the drug release can be tailored to maintain desired release profiles of several weeks to months after parenteral (i.e., intramuscular or subcutaneous) administration. A sustained drug release can be desirable to combat chronic diseases by overcoming pill fatigue of a daily oral intake, hence, improving patient compliance. Although the marketed aqueous suspensions for intramuscular injection efficiently relieve the daily pill burden in chronic diseases, the exact drug release mechanisms remain to be fully unraveled. The in vivo drug release and subsequent absorption to the systemic circulation are influenced by a plethora of variables, resulting in a complex in vivo behavior of aqueous suspensions after intramuscular administration. A better understanding of the factors influencing the in vivo performance of aqueous suspensions could advance their drug development. An overview of the potential influential variables on the drug release after intramuscular injection of aqueous suspensions is provided with, where possible, available pharmacokinetic parameters in humans or other species derived from literature, patents, and clinical trials. These variables can be categorized into drug substance and formulation properties, administration site properties, and the host response towards drug particles. Based on the findings, the most critical factors are particle size, dose level, stabilizing excipient, drug lipophilicity, gender, body mass index, and host response.
Collapse
Affiliation(s)
- Vy Thi Thanh Nguyen
- Ghent University, Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ottergemsesteenweg 460, B-9000, Ghent, Belgium.
| | - Nicolas Darville
- Pharmaceutical Product Development & Supply, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Antwerp, Belgium
| | - An Vermeulen
- Ghent University, Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| |
Collapse
|
33
|
Abstract
In 2019, more than 4 years after the widespread availability of safe, oral, curative treatments, an estimated 58 million people were living with hepatitis C virus infections (PLWHC). Additional tools may enable those not yet reached to be treated. One such tool could be long-acting parenteral formulations of HCV treatments, which may allow PLWHC to be diagnosed and cured in a single encounter. Although existing highly effective oral medications might be formulated as long-acting parenteral treatments, pharmacological, regulatory, patent, and medical challenges have to be overcome; this requires the concerted efforts of PLWHC, researchers, funding agencies, industry, the World Health Organization, and other stakeholders.
Collapse
Affiliation(s)
- David L Thomas
- Department of Medicine, Division of Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Owen
- Department of Pharmacology and Therapeutics, Centre of Excellence for Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, United Kingdom
| | - Jennifer J Kiser
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
34
|
Flexner C, Thomas DL, Clayden P, Swindells S. What Clinicians Need to Know About the Development of Long-Acting Formulations. Clin Infect Dis 2022; 75:S487-S489. [PMID: 36410382 PMCID: PMC10200319 DOI: 10.1093/cid/ciac749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We encourage readers of this Supplement to find articles that are relevant to the current and future practice of infectious diseases medicine. Access to long-acting products and formulations in low- and middle-income countries remains a key determinant of the impact of these advances on global health.
Collapse
Affiliation(s)
- Charles Flexner
- Divisions of Clinical Pharmacology, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Infectious Diseases, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - David L Thomas
- Infectious Diseases, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
35
|
Neary M, Owen A, Olagunju A. A Holistic Review of the Preclinical Landscape for Long-Acting Anti-infective Drugs Using HIV as a Paradigm. Clin Infect Dis 2022; 75:S490-S497. [PMID: 36410386 PMCID: PMC10200324 DOI: 10.1093/cid/ciac685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lack of predictive preclinical models is a key contributor to the steep attrition rate in drug development. Successful clinical translation may be higher for new chemical entities or existing approved drugs reformulated for long-acting (LA) administration if preclinical studies designed to identify any new uncertainties are predictive of human exposure and response. In this review, we present an overview of standard preclinical assessments deployed for LA formulations and delivery systems, using human immunodeficiency virus LA therapeutics preclinical development as a paradigm. Key progress in the preclinical development of novel LA antiretrovirals formulations and delivery systems are summarized, including bispecific broadly neutralizing monoclonal antibody and small molecule technologies for codelivery of multiple drugs with disparate solubility properties. There are new opportunities to take advantage of recent developments in tissue engineering and 3-dimensional in vitro modeling to advance preclinical modeling of anti-infective activity, developmental and reproductive toxicity assessment, and to apply quantitative modeling and simulation strategies. These developments are likely to drive the progression of more LA anti-infective drugs and multipurpose technologies into clinical development in the coming years.
Collapse
Affiliation(s)
- Megan Neary
- Department of Pharmacology and Therapeutics, Centre of Excellence for Long-acting Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Andrew Owen
- Department of Pharmacology and Therapeutics, Centre of Excellence for Long-acting Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Adeniyi Olagunju
- Department of Pharmacology and Therapeutics, Centre of Excellence for Long-acting Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, Merseyside, United Kingdom
| |
Collapse
|
36
|
Kozlina F, Meštrović I, Novak V, Marjanović N, Cetina-Čižmek B. Development of fiber optic in vitro release testing method for dexamethasone release from the oil solutions. ADMET & DMPK 2022; 10:315-329. [PMID: 36578560 PMCID: PMC9793461 DOI: 10.5599/admet.1465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/17/2022] [Indexed: 12/31/2022]
Abstract
For many parenteral drugs, there is still no standardized method for in vitro release (IVR) testing available. This article presents the development of a new IVR method for oil solutions using a dialysis membrane and USP II apparatus coupled to a fiber optic UV-Vis spectrometer. Experiments were performed using dexamethasone formulations containing castor oil as a solvent with the addition of cosolvents, 20 % (v/v) of isopropanol or Capryol® 90. Based on solubility testing results, castor oil was chosen as the best solvent amongst other vegetable oils, while a significant increase in solubility was obtained by adding either of the two cosolvents. Partitioning experiments were performed to ensure these formulations could achieve prolonged drug release. IVR testing was performed with model formulations and critical test parameters were varied in order to examine the method's sensitivity. The developed method was sensitive to temperature and stirring rate, while coupling the USP II apparatus with a fiber optic UV-Vis spectrometer enabled complete automation. Moreover, due to the interference of excipients on fiber optic detection of dexamethasone during the release testing, derivative spectroscopy was successfully introduced for the elimination of the interference. The developed IVR method described herein could be useful in preformulation investigations and the early development of novel formulations.
Collapse
Affiliation(s)
- Filip Kozlina
- University of Zagreb, Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Ivan Meštrović
- University of Zagreb, Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Viktor Novak
- University of Zagreb, Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Nikola Marjanović
- PLIVA Croatia Ltd, TEVA Group Member, Prilaz baruna Filipovića 25, 10 000 Zagreb, Croatia
| | - Biserka Cetina-Čižmek
- PLIVA Croatia Ltd, TEVA Group Member, Prilaz baruna Filipovića 25, 10 000 Zagreb, Croatia,*Corresponding Author: E-mail: ; Tel.: +385-1-372-2648
| |
Collapse
|
37
|
Monroe MK, Wang H, Anderson CF, Jia H, Flexner C, Cui H. Leveraging the therapeutic, biological, and self-assembling potential of peptides for the treatment of viral infections. J Control Release 2022; 348:1028-1049. [PMID: 35752254 PMCID: PMC11022941 DOI: 10.1016/j.jconrel.2022.06.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022]
Abstract
Peptides and peptide-based materials have an increasing role in the treatment of viral infections through their use as active pharmaceutical ingredients, targeting moieties, excipients, carriers, or structural components in drug delivery systems. The discovery of peptide-based therapeutic compounds, coupled with the development of new stabilization and formulation strategies, has led to a resurgence of antiviral peptide therapeutics over the past two decades. The ability of peptides to bind cell receptors and to facilitate membrane penetration and subsequent intracellular trafficking enables their use in various antiviral systems for improved targeting efficiency and treatment efficacy. Importantly, the self-assembly of peptides into well-defined nanostructures provides a vast library of discrete constructs and supramolecular biomaterials for systemic and local delivery of antiviral agents. We review here the recent progress in exploiting the therapeutic, biological, and self-assembling potential of peptides, peptide conjugates, and their supramolecular assemblies in treating human viral infections, with an emphasis on the treatment strategies for Human Immunodeficiency Virus (HIV).
Collapse
Affiliation(s)
- Maya K Monroe
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Caleb F Anderson
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Hongpeng Jia
- Department of Surgery, The Johns Hopkins University School of Medicine, United States of America
| | - Charles Flexner
- Divisions of Clinical Pharmacology and Infectious Diseases, The Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, MD 21205, United States of America.
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Deptartment of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America; Center for Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, United States of America.
| |
Collapse
|
38
|
Shakil A, Hern FY, Liu C, Temburnikar K, Chambon P, Liptrott N, McDonald TO, Neary M, Owen A, Freel Meyers C, Rannard SP. Linear and branched polymer prodrugs of the water-soluble nucleoside reverse-transcriptase inhibitor emtricitabine as structural materials for long-acting implants. J Mater Chem B 2022; 10:4395-4404. [PMID: 35604111 PMCID: PMC9199480 DOI: 10.1039/d2tb00825d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022]
Abstract
Long-acting drug delivery is a growing area of interest as it overcomes many challenges related to patient adherence to therapy and the pill burden associated with chronic illness. Injectable formulations are becoming more common and drug-releasing implants also provide several opportunities. Highly water soluble drug compounds are poor candidates for long-acting delivery. Here, the water-soluble nucleoside reverse transcriptase inhibitor emtricitabine (FTC) has been used as a novel A-B monomer in step-growth polymerisation with chloroformate functional Cn monomers, to produce new poly(carbamate/carbonate) structures with varying architecture. The polymer prodrugs were all solid at ambient temperature and have been shown to release FTC when subjected to mixed gender human plasma. Vacuum compression moulding has been used to form solid rod implants without polymer degradation; the rods show FTC release over long periods in the presence of microsomes, establishing the basis of a polymer prodrug strategy for FTC delivery.
Collapse
Affiliation(s)
- Anika Shakil
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Faye Y Hern
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Chung Liu
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Kartik Temburnikar
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Pierre Chambon
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Neill Liptrott
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Tom O McDonald
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Megan Neary
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Andrew Owen
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Caren Freel Meyers
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Steve P Rannard
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| |
Collapse
|
39
|
Navrátil O, Lizoňová D, Slonková K, Mašková L, Zadražil A, Sedmidubský D, Štěpánek F. Antibiotic depot system with radiofrequency controlled drug release. Colloids Surf B Biointerfaces 2022; 217:112618. [PMID: 35738076 DOI: 10.1016/j.colsurfb.2022.112618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/20/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Abstract
Drug depot systems have traditionally relied on the spontaneous dissolution and diffusion of drugs or prodrugs from a reservoir with constant exposure to the surrounding physiological fluids. While this is appropriate for clinical scenarios that require constant plasma concentration of the drug over time, there are also situations where multiple bursts of the drug at well-defined time intervals are preferred. This work presents a drug depot system that enables repeated on-demand release of antibiotics in precise doses, controlled by an external radiofrequency magnetic field. The remotely controlled depot system consists of composite microcapsules with a core-shell structure. The core contains micronized drug particles embedded in a low-melting hydrophobic matrix. The shell is formed by a hydrogel with immobilised magnetic nanoparticles that facilitate local heat dissipation after exposure to a radiofrequency magnetic field. When the melting point of the core material is locally exceeded, the embedded drug particles are mobilised and their surface is exposed to the external aqueous phase. It is shown that drug release can be controlled in an on/off manner by a chosen sequence and duration of radiofrequency pulses. The capacity of the depot system is shown to be significantly higher than that of purely diffusion-controlled systems containing a pre-dissolved drug. The functionality of the depot system is demonstrated in vitro for the specific case of norfloxacin acting on E. coli.
Collapse
Affiliation(s)
- Ondřej Navrátil
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Denisa Lizoňová
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Karolína Slonková
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Lucie Mašková
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Aleš Zadražil
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - David Sedmidubský
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - František Štěpánek
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
40
|
Muddineti OS, Omri A. Current trends in PLGA based long-acting injectable products: The industry perspective. Expert Opin Drug Deliv 2022; 19:559-576. [PMID: 35534912 DOI: 10.1080/17425247.2022.2075845] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Poly (lactic-co-glycolic acid) (PLGA) has been used in many long-acting drug formulations, which have been approved by the US Food and Drug Administration (FDA). PLGA has unique physicochemical properties, which results in complexities in the formulation, characterization, and evaluation of generic products. To address the challenges of generic development of PLGA-based products, the FDA has established an extensive research program to investigate novel methods and tools to aid product development and regulatory review. AREAS COVERED This review article intends to provide a comprehensive review on physicochemical properties of PLGA polymer, characterization, formulation, and analytical aspects, manufacturing conditions on product performance, in-vitro release testing, and bioequivalence. Current research on formulation development as per QbD in vitro release testing methods, regulatory research outcomes, and bioequivalence. EXPERT OPINION The development of PLGA based long-acting injectables is promising and challenging when considering the numerous interrelated delivery-related factors. Achieving a successful formulation requires a thorough understanding of the critical interactions between polymer/drug properties, release profiles over time, up-to-date knowledge on regulatory guidance, and elucidation of the impact of multiple in vivo conditions to methodically evaluate the eventual clinical efficacy.
Collapse
Affiliation(s)
- Omkara Swami Muddineti
- Formulation Research & Development, Vimta Labs Limited, Plot No.5, M N Park, Genome Valley, Shameerpet, Hyderabad, Telangana, 500101, India
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON, Canada
| |
Collapse
|
41
|
Magnitude of Drug–Drug Interactions in Special Populations. Pharmaceutics 2022; 14:pharmaceutics14040789. [PMID: 35456623 PMCID: PMC9027396 DOI: 10.3390/pharmaceutics14040789] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
Drug–drug interactions (DDIs) are one of the most frequent causes of adverse drug reactions or loss of treatment efficacy. The risk of DDIs increases with polypharmacy and is therefore of particular concern in individuals likely to present comorbidities (i.e., elderly or obese individuals). These special populations, and the population of pregnant women, are characterized by physiological changes that can impact drug pharmacokinetics and consequently the magnitude of DDIs. This review compiles existing DDI studies in elderly, obese, and pregnant populations that include a control group without the condition of interest. The impact of physiological changes on the magnitude of DDIs was then analyzed by comparing the exposure of a medication in presence and absence of an interacting drug for the special population relative to the control population. Aging does not alter the magnitude of DDIs as the related physiological changes impact the victim and perpetrator drugs to a similar extent, regardless of their elimination pathway. Conversely, the magnitude of DDIs can be changed in obese individuals or pregnant women, as these conditions impact drugs to different extents depending on their metabolic pathway.
Collapse
|
42
|
Brah AT, Barthold D, Hauber B, Collier AC, Ho RJY, Marconi VC, Simoni JM, Graham SM. The systematic development of attributes and levels for a discrete choice experiment of HIV patient preferences for long-acting antiretroviral therapies in the United States. AIDS Res Ther 2022; 19:13. [PMID: 35216610 PMCID: PMC8881811 DOI: 10.1186/s12981-022-00435-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 02/04/2022] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Patient preferences for long-acting antiretroviral therapies (LA-ART) should inform development of regimens with optimal adherence and acceptability. We describe a systematic process used to identify attributes and levels for a discrete choice experiment (DCE) designed to elicit preferences for potential LA-ART options in the US. METHODS Our approach was conducted in four stages: data collection, data reduction, removing inappropriate attributes, and optimizing wording. We started with 8 attributes defining potential LA-ART products based on existing literature and knowledge of products in development. We conducted 12 key informant interviews with experts in HIV treatment. The list of attributes, the set of plausible levels for each attribute, and restrictions on combinations of attribute levels were updated iteratively. RESULTS Despite uncertainty about which products will become available, key informant discussions converged on 4 delivery modes (infusions and patches were not considered immediately feasible) and 6 additional attributes. Treatment effectiveness and frequency of clinical monitoring were dropped. Oral lead-in therapy was split into two attributes: pre-treatment time undetectable and pre-treatment negative reaction testing. We omitted product-specific systemic and local side effects. In addition to mode, the final set of attributes included: frequency of dosing; location of treatment; pain; pre-treatment time undetectable; pre-treatment negative reaction testing; and late-dose leeway. CONCLUSIONS A systematic process successfully captured elements that are both feasible and relevant to evaluating the acceptability of potential LA-ART alternatives to patients.
Collapse
Affiliation(s)
- Aaron T. Brah
- grid.34477.330000000122986657Division of Allergy & Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA USA
| | - Douglas Barthold
- grid.34477.330000000122986657The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington, Seattle, WA USA
| | - Brett Hauber
- grid.34477.330000000122986657The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington, Seattle, WA USA ,grid.410513.20000 0000 8800 7493Pfizer, Inc, New York, NY USA
| | - Ann C. Collier
- grid.34477.330000000122986657Division of Allergy & Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA USA
| | - Rodney J. Y. Ho
- grid.34477.330000000122986657Department of Pharmaceutics and Bioengineering, University of Washington, Seattle, WA USA
| | - Vincent C. Marconi
- grid.189967.80000 0001 0941 6502Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Jane M. Simoni
- grid.34477.330000000122986657Department of Psychology, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Department of Global Health, University of Washington, Seattle, WA USA
| | - Susan M. Graham
- grid.34477.330000000122986657Division of Allergy & Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Department of Global Health, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Department of Epidemiology, University of Washington, Seattle, WA USA
| |
Collapse
|
43
|
Paredes AJ, Volpe-Zanutto F, Vora LK, Tekko IA, Permana AD, Picco CJ, McCarthy HO, Donnelly RF. Systemic delivery of tenofovir alafenamide using dissolving and implantable microneedle patches. Mater Today Bio 2022; 13:100217. [PMID: 35243292 PMCID: PMC8858997 DOI: 10.1016/j.mtbio.2022.100217] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/30/2022] Open
Abstract
The human immunodeficiency virus (HIV) remains a global health concern, with 37.7 million people currently living with the infection and 1.5 million new cases every year. Current antiretroviral (ARV) therapies are administered through the oral route daily, often in lifelong treatments, leading to pill fatigue and poor treatment adherence. Therefore, the development of novel formulations for the administration ARV drugs using alternative routes is actively sought out. In this sense, microneedle array patches (MAPs) offer a unique user-centric platform that can be painlessly self-applied to the skin and deliver drugs to the systemic circulation. In this work, dissolving and implantable MAPs loaded with the tenofovir alafenamide (TAF) were developed with the aim of releasing the drug systemically. Both MAPs were sufficiently strong to pierce excised neonatal full-thickness porcine skin and form drug depots. In vitro release experiments performed in dialysis membrane models, demonstrated a relatively fast delivery of the drug in all cases. Franz cells experiments revealed that dissolving and implantable MAPs deposited 47.87 ± 16.33 μg and 1208.04 ± 417.9 μg of TAF in the skin after 24 h. Pharmacokinetic experiments in rats demonstrated a fast metabolization of TAF into tenofovir, with a rapid elimination of the metabolite from the plasma. The MAPs described in this work could be used as an alternative to current oral treatments for HIV management.
Collapse
Affiliation(s)
- Alejandro J. Paredes
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Fabiana Volpe-Zanutto
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Lalitkumar K. Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Ismaiel A. Tekko
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Andi Dian Permana
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
- Department of Pharmaceutics, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Camila J. Picco
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Helen O. McCarthy
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Ryan F. Donnelly
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| |
Collapse
|
44
|
Surve DH, Jindal AB. Development of cationic Isometamidium chloride loaded long-acting lipid nanoformulation: optimization, cellular uptake, pharmacokinetics, biodistribution, and immunohistochemical evaluation. Eur J Pharm Sci 2021; 167:106024. [PMID: 34592462 DOI: 10.1016/j.ejps.2021.106024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
The aim of the present work involved the development and evaluation of long-acting Isometamidium chloride (ISMM)-Docusate sodium (DS) complex loaded lipid nanoparticles (LA ISMM-DS LNP). The development involved screening various anionic complexing agents, including DS, dextran sulphate, and sodium alginate. Anionic DS was selected to synthesize hydrophobic ionic complex (ISMM-DS HIC), which was loaded into lipid nanoparticles (LA ISMM-DS LNP) by in situ complexation followed by the solvent evaporation method. 35-5-folds increase in the drug loading of hydrophilic cationic ISMM within nanoparticles was observed due to ISMM-DS HIC. The LA ISMM-DS LNP were non-hemolytic (0-2.52%), cytocompatible (80.6-47.5% cell viability), and enhanced THP-1 cellular uptake (2.3-folds higher) compared with free ISMM. The LA ISMM-DS LNP engender protracted in vivo plasma drug concentration for seven days with enhanced AUC0-ꝏ, MRT0-ꝏ, and t1/2, along with reduced Cl compared with free ISMM. Interestingly, the amount of ISMM was 2.9-, 4.2- and 2.0-folds higher in target reticuloendothelial (RES) organs like liver (Kupffer cells), spleen (spleenotropic macrophages and 15% T-lymphocytes), and lymph nodes (75% T-lymphocytes), respectively in LA ISMM-DS LNP group compared with free ISMM. Furthermore, LA ISMM-DS LNP caused higher peripheral blood mononuclear cells (PBMC) infiltration with diminished toxicity and inflammation. Therefore, the in vitro and in vivo studies predicted enhanced safety and efficacy of LA ISMM-DS LNP compared with free ISMM. To conclude, successfully developed LA ISMM-DS LNP would elicit a tremendous clinical potential for treatment and prevention against trypanosomiasis.
Collapse
Affiliation(s)
- Dhanashree H Surve
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Jhunjhunu, Rajasthan 333031, India
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Jhunjhunu, Rajasthan 333031, India.
| |
Collapse
|
45
|
Thoueille P, Choong E, Cavassini M, Buclin T, Decosterd LA. Long-acting antiretrovirals: a new era for the management and prevention of HIV infection. J Antimicrob Chemother 2021; 77:290-302. [PMID: 34499731 PMCID: PMC8809192 DOI: 10.1093/jac/dkab324] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The long-acting antiretroviral cabotegravir and rilpivirine combination has just received FDA, EMA and Health Canada approval. This novel drug delivery approach is about to revolutionize the therapy of people living with HIV, decreasing the 365 daily pill burden to only six intramuscular injections per year. In addition, islatravir, a first-in-class nucleoside reverse transcriptase translocation inhibitor, is intended to be formulated as an implant with a dosing interval of 1 year or more. At present, long-acting antiretroviral therapies (LA-ARTs) are given at fixed standard doses, irrespectively of the patient's weight and BMI, and without consideration for host genetic and non-genetic factors likely influencing their systemic disposition. Despite a few remaining challenges related to administration (e.g. pain, dedicated medical procedure), the development and implementation of LA-ARTs can overcome long-term adherence issues by improving patients' privacy and reducing social stigma associated with the daily oral intake of anti-HIV treatments. Yet, the current 'one-size-fits-all' approach does not account for the recognized significant inter-individual variability in LA-ART pharmacokinetics. Therapeutic drug monitoring (TDM), an important tool for precision medicine, may provide physicians with valuable information on actual drug exposure in patients, contributing to improve their management in real life. The present review aims to update the current state of knowledge on these novel promising LA-ARTs and discusses their implications, particularly from a clinical pharmacokinetics perspective, for the future management and prevention of HIV infection, issues of ongoing importance in the absence of curative treatment or an effective vaccine.
Collapse
Affiliation(s)
- Paul Thoueille
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eva Choong
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Matthias Cavassini
- Service of Infectious Diseases, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Thierry Buclin
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurent A Decosterd
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
46
|
Simoni JM, Beima-Sofie K, Wanje G, Mohamed ZH, Tapia K, McClelland RS, Ho RJY, Collier AC, Graham SM. "Lighten This Burden of Ours": Acceptability and Preferences Regarding Injectable Antiretroviral Treatment Among Adults and Youth Living With HIV in Coastal Kenya. J Int Assoc Provid AIDS Care 2021; 20:23259582211000517. [PMID: 33685272 PMCID: PMC7952847 DOI: 10.1177/23259582211000517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: Long-acting injectable (LAI) antiretroviral therapy (ART) may offer persons living with HIV (PLWH) an attractive alternative to pill-based treatment options, yet acceptability data remain scant, especially in sub-Saharan Africa. Methods: We conducted 6 focus group discussions with PLWH, including key stake holder groups, and analyzed data with content analysis. Results: Initial reactions to the idea of LAI-ART were often positive. The primary advantages voiced were potential to facilitate improved adherence and alleviate the burden of daily pill-taking while avoiding inadvertent disclosure and HIV stigma. Potential side effects were a particular concern of the women. Most participants preferred clinic-based administration over self-injections at home due to concerns about safety, privacy, and potential need for refrigeration. Conclusions: LAI-ART may be acceptable in Kenya, provided injections are infrequent and delivered in a clinic setting. However, HIV stigma, fear of potential side effects, and limited clinical capacity would need to be addressed.
Collapse
Affiliation(s)
- Jane M Simoni
- Department of Psychology, 7284University of Washington, Seattle Washington, USA.,Department of Global Health, 7284University of Washington, Seattle Washington, USA.,Department of Gender, Women and Sexuality Studies, 7284University of Washington, Seattle Washington, USA
| | - Kristin Beima-Sofie
- Department of Global Health, 7284University of Washington, Seattle Washington, USA
| | - George Wanje
- Department of Medical Microbiology, 107854University of Nairobi, Nairobi, Kenya
| | - Zahra H Mohamed
- Department of Global Health, 7284University of Washington, Seattle Washington, USA
| | - Kenneth Tapia
- Department of Global Health, 7284University of Washington, Seattle Washington, USA
| | - R Scott McClelland
- Department of Global Health, 7284University of Washington, Seattle Washington, USA.,Department of Medical Microbiology, 107854University of Nairobi, Nairobi, Kenya.,Department of Medicine, 7284University of Washington, Seattle Washington, USA.,Department of Epidemiology, 7284University of Washington, Seattle Washington, USA
| | - Rodney J Y Ho
- Department of Pharmaceutics, 7284University of Washington, Seattle Washington, USA.,Department of Bioengineering, 7284University of Washington, Seattle Washington, USA
| | - Ann C Collier
- Department of Medicine, 7284University of Washington, Seattle Washington, USA
| | - Susan M Graham
- Department of Global Health, 7284University of Washington, Seattle Washington, USA.,Department of Medicine, 7284University of Washington, Seattle Washington, USA.,Department of Epidemiology, 7284University of Washington, Seattle Washington, USA
| |
Collapse
|
47
|
Jucker BM, Fuchs EJ, Lee S, Damian V, Galette P, Janiczek R, Macura KJ, Jacobs MA, Weld ED, Solaiyappan M, D'Amico R, Shaik JS, Bakshi K, Han K, Ford S, Margolis D, Spreen W, Gupta MK, Hendrix CW, Patel P. Multiparametric magnetic resonance imaging to characterize cabotegravir long-acting formulation depot kinetics in healthy adult volunteers. Br J Clin Pharmacol 2021; 88:1655-1666. [PMID: 34240449 PMCID: PMC9290983 DOI: 10.1111/bcp.14977] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/20/2021] [Accepted: 05/01/2021] [Indexed: 12/26/2022] Open
Abstract
AIM Cabotegravir long-acting (LA) intramuscular (IM) injection is being investigated for HIV preexposure prophylaxis due to its potent antiretroviral activity and infrequent dosing requirement. A subset of healthy adult volunteers participating in a Phase I study assessing cabotegravir tissue pharmacokinetics underwent serial magnetic resonance imaging (MRI) to assess drug depot localization and kinetics following a single cabotegravir LA IM targeted injection. METHODS Eight participants (four men, four women) were administered cabotegravir LA 600 mg under ultrasonographic-guided injection targeting the gluteal muscles. MRI was performed to determine injection-site location in gluteal muscle (IM), subcutaneous (SC) adipose tissue and combined IM/SC compartments, and to quantify drug depot characteristics, including volume and surface area, on Days 1 (≤2 hours postinjection), 3 and 8. Linear regression analysis examined correlations between MRI-derived parameters and plasma cabotegravir exposure metrics, including maximum observed concentration (Cmax ) and partial area under the concentration-time curve (AUC) through Weeks 4 and 8. RESULTS Cabotegravir LA depot locations varied by participant and were identified in the IM compartment (n = 2), combined IM/SC compartments (n = 4), SC compartment (n = 1) and retroperitoneal cavity (n = 1). Although several MRI parameter and exposure metric correlations were determined, total depot surface area on Day 1 strongly correlated with plasma cabotegravir concentration at Days 3 and 8, Cmax and partial AUC through Weeks 4 and 8. CONCLUSION MRI clearly delineated cabotegravir LA injection-site location and depot kinetics in healthy adults. Although injection-site variability was observed, drug depot surface area correlated with both plasma Cmax and partial AUC independently of anatomical distribution.
Collapse
Affiliation(s)
| | - Edward J Fuchs
- Departments of Internal Medicine and Radiology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | | - Katarzyna J Macura
- Departments of Internal Medicine and Radiology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael A Jacobs
- Departments of Internal Medicine and Radiology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ethel D Weld
- Departments of Internal Medicine and Radiology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Meiyappan Solaiyappan
- Departments of Internal Medicine and Radiology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | | - Susan Ford
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | | | | | | | - Craig W Hendrix
- Departments of Internal Medicine and Radiology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Parul Patel
- ViiV Healthcare, Research Triangle Park, NC, USA
| |
Collapse
|
48
|
Rojekar S, Vora LK, Tekko IA, Volpe-Zanutto F, McCarthy HO, Vavia PR, Donnelly RF. Etravirine-loaded dissolving microneedle arrays for long-acting delivery. Eur J Pharm Biopharm 2021; 165:41-51. [PMID: 33971273 DOI: 10.1016/j.ejpb.2021.04.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 04/12/2021] [Accepted: 04/18/2021] [Indexed: 12/11/2022]
Abstract
A key challenge of HIV treatment with multiple antiretroviral drugs is patient adherence. Thus, there is an urgent need for long-acting depot systems for delivering drugs over an extended duration. Although the parenteral route is preferred for depot systems, it is associated with obvious drawbacks, such as painful injections, potentially-contaminated sharps waste, and the necessity of trained healthcare personnel for administration. Amongst a small number of alternatives in development microneedles are versatile delivery systems enabling systemic drug delivery and potentially improving patient adherence due to their capacity for self-administration. We have developed dissolving microneedle (DMNs) embedded with etravirine nanosuspension (ETR NS) as a long-acting HIV therapy to improve patient adherence. The ETR NS prepared by sonoprecipitation yielded particle sizes of 764 ± 96.2 nm, polydispersity indices of of 0.23 ± 0.02, and zeta potentials of -19.75 ± 0.55 mV. The DMNs loaded with ETR NS demonstrated 12.84 ± 1.33% ETR deposition in ex-vivo neonatal porcine skin after 6 h application. In in vivo rat pharmacokinetic studies, the Cmax exhibited by DMNs loaded with ETR powder and ETR NS were 158 ± 10 ng/mL and 177 ± 30 ng/mL, respectively. DMN groups revealed a higher t1/2, Tmax, and mean residence time compared to intravenous ETR solutions, suggesting the long-acting potential of etravirine delivered intradermally using DMNs.
Collapse
Affiliation(s)
- Satish Rojekar
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, University Under Section 3 of UGC Act - 1956, Elite Status and Center of Excellence - Govt. of Maharashtra, TEQIP Phase III Funded, Mumbai 400019, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ismaiel A Tekko
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Aleppo University, Syria
| | - Fabiana Volpe-Zanutto
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Pradeep R Vavia
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, University Under Section 3 of UGC Act - 1956, Elite Status and Center of Excellence - Govt. of Maharashtra, TEQIP Phase III Funded, Mumbai 400019, India.
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
49
|
Impact of Therapeutic Drug Monitoring of Antiretroviral Drugs in Routine Clinical Management of People Living With HIV: A Narrative Review. Ther Drug Monit 2021; 42:64-74. [PMID: 31393332 DOI: 10.1097/ftd.0000000000000684] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The treatment of HIV infection has evolved significantly since the advent of highly active antiretroviral therapy. As a result, a response rate of 90%-95% now represents a realistically achievable target. Given this background, it is difficult to imagine the additional benefits that therapeutic drug monitoring (TDM) could provide in the management of HIV infection. METHODS This article is not intended to provide a systematic literature review on TDM of antiretroviral agents; rather, the authors aim to discuss the potential added value of TDM in the optimal management of people living with HIV (PLWH) in selected real-life clinical scenarios based on data collected over 10 years by their TDM service. RESULTS Some clinical situations, in which the selection of the optimal antiretroviral therapy is challenging, have been identified. These include poorly compliant patients, suboptimal antiretroviral therapies (in terms of both efficacy and toxicity), polypharmacy with a high risk of drug-drug interactions, and different patient populations, such as pregnant women. CONCLUSIONS The transformation of HIV infection from a near-universally fatal illness to a lifelong chronic disease has resulted in an HIV population that is growing and aging, placing new and increasing demands on public programs and health services. Increasingly, the management of comorbidities, polypharmacy, and drug-drug interaction, and their impact on antiretroviral therapy will have to be undertaken. These clinical settings represent some of the new frontiers for the use of TDM with the goal of achieving optimal prescription and outcome for PLWH.
Collapse
|
50
|
Kumar SR, Mehta CH, Nayak UY. Long-Acting Formulations: A Promising Approach for the Treatment of Chronic Diseases. Curr Pharm Des 2021; 27:876-889. [PMID: 32634073 DOI: 10.2174/1381612826666200707122012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/28/2020] [Indexed: 11/22/2022]
Abstract
Medication and patient adherence are the two main aspects of any successful treatment of chronic disease. Even though diseases and its treatment existed for several hundred years, the treatment optimization for a given patient is still a researcher question for scientists. There are differences in treatment duration, prognostic signs and symptoms between patient to patient. Hence, designing ideal formulation to suit individual patient is a challenging task. The conventional formulations like oral solids and liquids gives a partial or incomplete treatment because the patient needs to follow the daily pills for a longer time. In such cases, the long-acting formulations will have better patient compliances as drug will be released for a longer duration. Many such approaches are under the clinical investigation. The favorable pharmacokinetic and pharmacodynamic relationships, will be promising option for the treatment of chronic diseases. In this review, we have highlighted the importance of long-acting formulations in the treatment of chronic diseases and the advent of newer formulation technologies.
Collapse
Affiliation(s)
- Somaraju R Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Chetan H Mehta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|