1
|
Alphonse N, Sécher T, Heuzé-Vourc'h N. A breath of fresh air: inhaled antibodies to combat respiratory infectious diseases - a clinical trial overview. Expert Opin Drug Deliv 2025; 22:197-218. [PMID: 39711323 DOI: 10.1080/17425247.2024.2446608] [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: 09/18/2024] [Revised: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION With the worldwide growing burden of respiratory tract infections (RTIs), innovative therapeutic approaches are in high demand. Inhaled antibodies (Abs) represent a promising avenue, offering targeted treatment options with potentially better therapeutic index compared to traditional delivery methods. AREAS COVERED This comprehensive review summarizes the challenges faced in delivering Abs by (intranasal and pulmonary) inhalation. It outlines the physiological and biological barriers encountered by inhaled drugs, as well as the influence of delivery devices and formulation on the deposition and efficacy of inhaled molecules. Moreover, it provides a detailed overview of the current clinical trial landscape of inhaled anti-RTI Abs, highlighting the progress in the development of inhaled Abs targeting a range of pathogens, such as severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus. The mechanism of action, therapeutic targets, and clinical outcomes of these novel therapies are detailed. EXPERT OPINION Delivery of Abs by inhalation faces several challenges. Addressing these challenges and developing specific approaches to deliver inhaled Abs represent a promising avenue for the development of the next generation of inhaled Abs. By offering targeted, localized therapy with the potential for a better therapeutic index, inhaled Abs could significantly improve outcomes for patients with RTIs.
Collapse
Affiliation(s)
- Noémie Alphonse
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
| | - Thomas Sécher
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
| | - Nathalie Heuzé-Vourc'h
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
| |
Collapse
|
2
|
Sécher T, Cortes M, Boisseau C, Barba Goudiaby MT, Pitiot A, Parent C, Thomas M, Heuzé-Vourc’h N. Synergy between Lactobacillus murinus and anti-PcrV antibody delivered in the airways to boost protection against Pseudomonas aeruginosa. Mol Ther Methods Clin Dev 2024; 32:101330. [PMID: 39314638 PMCID: PMC11418128 DOI: 10.1016/j.omtm.2024.101330] [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: 12/14/2023] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
Therapeutic antibodies (Ab) have revolutionized the management of multiple illnesses including respiratory tract infections (RTIs). However, anti-infectious Ab displayed several limitations including antigen restrictiveness, narrowed therapeutic windows, and limited dose in the vicinity of the target when delivered by parenteral routes. Strategies enhancing further Ab-dependent containment of infection are currently needed. Here we showed that a combination of inhaled anti-infectious Ab and probiotics is an efficient formulation to protect against lung infection. Using a mouse model of Pseudomonas aeruginosa-induced pneumonia, we demonstrated a synergistic effect reducing both bacterial burden and pro-inflammatory response affording protection against primary and secondary infections. This is the first study showing that the local combination in the airways of anti-infective Ab and probiotics subverts suboptimal potency of Ab monotherapy and provides protection against respiratory pathogen.
Collapse
Affiliation(s)
- Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Mélanie Cortes
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Chloé Boisseau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Marie-Thérèse Barba Goudiaby
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, UMR1319 Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Aubin Pitiot
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Christelle Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Muriel Thomas
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, UMR1319 Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Nathalie Heuzé-Vourc’h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| |
Collapse
|
3
|
Blay V, Pandiella A. Strategies to boost antibody selectivity in oncology. Trends Pharmacol Sci 2024; 45:1135-1149. [PMID: 39609227 DOI: 10.1016/j.tips.2024.10.005] [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: 07/22/2024] [Revised: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 11/30/2024]
Abstract
Antibodies in oncology are being equipped with toxic cargoes and effector functions that can kill cells at very low concentrations. A key challenge is that most targets on cancer cells are also present on at least some healthy cells. Shared targets can result in off-tumor binding and compromise the safety and potential of therapeutic candidates. In this review, we survey strategies that can help direct biologics to cancer sites more selectively. These strategies are becoming increasingly feasible thanks to advances in molecular design and engineering. The objective is to create therapeutics that exploit changes in cancer and leverage the human body infrastructure, enabling therapeutics that discriminate not just self from non-self but diseased from healthy tissue.
Collapse
Affiliation(s)
- Vincent Blay
- University of California Santa Cruz, Department of Microbiology and Environmental Toxicology, Santa Cruz, CA 95064, USA.
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, CIBERONC and IBSAL, 37007 Salamanca, Spain
| |
Collapse
|
4
|
Puccetti M, Pariano M, Schoubben A, Giovagnoli S, Ricci M. Biologics, theranostics, and personalized medicine in drug delivery systems. Pharmacol Res 2024; 201:107086. [PMID: 38295917 DOI: 10.1016/j.phrs.2024.107086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
The progress in human disease treatment can be greatly advanced through the implementation of nanomedicine. This approach involves targeted and cell-specific therapy, controlled drug release, personalized dosage forms, wearable drug delivery, and companion diagnostics. By integrating cutting-edge technologies with drug delivery systems, greater precision can be achieved at the tissue and cellular levels through the use of stimuli-responsive nanoparticles, and the development of electrochemical sensor systems. This precision targeting - by virtue of nanotechnology - allows for therapy to be directed specifically to affected tissues while greatly reducing side effects on healthy tissues. As such, nanomedicine has the potential to transform the treatment of conditions such as cancer, genetic diseases, and chronic illnesses by facilitating precise and cell-specific drug delivery. Additionally, personalized dosage forms and wearable devices offer the ability to tailor treatment to the unique needs of each patient, thereby increasing therapeutic effectiveness and compliance. Companion diagnostics further enable efficient monitoring of treatment response, enabling customized adjustments to the treatment plan. The question of whether all the potential therapeutic approaches outlined here are viable alternatives to current treatments is also discussed. In general, the application of nanotechnology in the field of biomedicine may provide a strong alternative to existing treatments for several reasons. In this review, we aim to present evidence that, although in early stages, fully merging advanced technology with innovative drug delivery shows promise for successful implementation across various disease areas, including cancer and genetic or chronic diseases.
Collapse
Affiliation(s)
- Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Italy,.
| | | | | | | | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Italy,.
| |
Collapse
|
5
|
Ryan M, Lam N, Wright K, Siderov J. Clinical Oncology Society of Australia Position Statement: 2022 update to the safe handling of monoclonal antibodies in healthcare settings. Asia Pac J Clin Oncol 2023; 19:723-730. [PMID: 36899469 DOI: 10.1111/ajco.13943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 03/12/2023]
Abstract
AIM The aims were to (a) review the scientific literature on occupational risk, including exposure mechanisms and risk assessment, with regards to handling monoclonal antibodies (mABs) in healthcare settings; and (b) update the recommendations in the Clinical Oncology Society of Australia (COSA) safe handling of monoclonal antibodies in healthcare settings position statement, published in 2013. METHODS A literature search was conducted between April 24, 2022, and July 3, 2022, to identify evidence relating to occupational exposure and handling of mABs in healthcare settings. Evidence in the literature was compared to the Position Statement published in 2013, and any potential additions, deletions, or revisions were discussed by the authors, and then agreed changes were made. RESULTS Thirty-nine references were included in this update, comprising of the 2013 Position Statement itself and 10 of its references, as well as 28 new references. The risks to healthcare workers in the preparation and administration of mABs arise from four distinct exposure mechanisms: dermal, mucosal, inhalation, and oral. Updates included recommendations on using protective eyewear during the preparation and administration of mABs, developing a local institutional risk assessment tool and handling recommendations, considerations for using closed system transfer devices, and to have awareness of the nomenclature change from 2021 for new mABs. CONCLUSION Practitioners should follow the 14 recommendations to lower occupational risk when handling mABs. Another Position Statement update should occur in 5-10 years to ensure the currency of recommendations.
Collapse
Affiliation(s)
- Marissa Ryan
- Department of Pharmacy, Princess Alexandra Hospital, Brisbane, Australia
- Centre for Health Services Research, The University of Queensland, Brisbane, Australia
- Centre for Online Health, The University of Queensland, Brisbane, Australia
| | - Neil Lam
- Icon Cancer Centre River City Pharmacy, Brisbane, Australia
| | - Kate Wright
- Central West Cancer Care Centre, Orange, Australia
- Department of Pharmacy, Orange Health Service, Orange, Australia
| | - Jim Siderov
- Department of Pharmacy, Austin Health, Melbourne, Australia
| |
Collapse
|
6
|
Simal I, Bauters T, Paepens C, Clottens N, Ramaut P, Kestens E. Developing a flowchart to evaluate the use of Closed System Drug-Transfer Devices with monoclonal antibodies: Focus on the clinical trial setting. J Oncol Pharm Pract 2023; 29:2014-2022. [PMID: 37680124 DOI: 10.1177/10781552231199412] [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: 09/09/2023]
Abstract
OBJECTIVE Available guidelines are ambiguous about safe handling monoclonal antibodies (MABs) and whether or not to use a Closed System Drug-Transfer Device (CSTD). In this article we want to describe a standardized working method on handling MABs in a clinical trial setting. DATA SOURCES The current workflow at the clinical trial unit of the Ghent University Hospital was critically analyzed, after which an extensive literature review was performed using the National Institute for Occupational Safety and Health Working Group guidelines and the database PubMed (Keywords: monoclonal antibodies, closed system transfer devices, safety guidelines, safe handling, management, administration, (bio)compatibility, volume loss, contamination, clinical trial unit. Period: 2020-2022). DATA SUMMARY Literature data are ambiguous. CSTDs can reduce cross-contamination and minimize exposure to potential hazardous drugs for healthcare professionals. However, in recent years more questions have been raised about their in-use compatibility and their impact on final product quality. This makes the debate on implementing CSTDs a hot topic in daily pharmacy practice and demands a holistic and standardized approach when deciding whether or not to use a CSTD when handling MABs. In a clinical trial setting, where safety data are frequently not available and the compatibility of CSTDs and investigational product is often unknown, this poses additional challenges that need to be taken into account. CONCLUSION We developed a flowchart which standardizes the use of a CSTD when handling MABs. It allows other healthcare professionals and clinical trial sponsors to define and evaluate the necessary criteria when standardizing the position of a CSTD in their safe handling procedures.
Collapse
Affiliation(s)
- Ine Simal
- Pharmacy Department, Ghent University Hospital, Ghent, Belgium
| | - Tiene Bauters
- Pharmacy Department, Ghent University Hospital, Ghent, Belgium
| | | | - Nele Clottens
- Pharmacy Department, Ghent University Hospital, Ghent, Belgium
| | - Pieter Ramaut
- Pharmacy Department, Ghent University Hospital, Ghent, Belgium
| | - Els Kestens
- Pharmacy Department, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
7
|
Sécher T, Heuzé-Vourc'h N. Barriers for orally inhaled therapeutic antibodies. Expert Opin Drug Deliv 2023; 20:1071-1084. [PMID: 37609943 DOI: 10.1080/17425247.2023.2249821] [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: 05/12/2023] [Revised: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023]
Abstract
INTRODUCTION Respiratory diseases represent a worldwide health issue. The recent Sars-CoV-2 pandemic, the burden of lung cancer, and inflammatory respiratory diseases urged the development of innovative therapeutic solutions. In this context, therapeutic antibodies (Abs) offer a tremendous opportunity to benefit patients with respiratory diseases. Delivering Ab through the airways has been demonstrated to be relevant to improve their therapeutic index. However, few inhaled Abs are on the market. AREAS COVERED This review describes the different barriers that may alter the fate of inhaled therapeutic Abs in the lungs at steady state. It addresses both physical and biological barriers and discusses the importance of taking into consideration the pathological changes occurring during respiratory disease, which may reinforce these barriers. EXPERT OPINION The pulmonary route remains rare for delivering therapeutic Abs, with few approved inhaled molecules, despite promising evidence. Efforts must focus on the intertwined barriers associated with lung diseases to develop appropriate Ab-formulation-device combo, ensuring optimal Ab deposition in the respiratory tract. Finally, randomized controlled clinical trials should be carried out to establish inhaled Ab therapy as prominent against respiratory diseases.
Collapse
Affiliation(s)
- Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université de Tours, Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université de Tours, Tours, France
| |
Collapse
|
8
|
Matera MG, Calzetta L, Rinaldi B, Cazzola M, Rogliani P. Strategies for overcoming the biological barriers associated with the administration of inhaled monoclonal antibodies for lung diseases. Expert Opin Drug Deliv 2023; 20:1085-1095. [PMID: 37715502 DOI: 10.1080/17425247.2023.2260310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/10/2023] [Accepted: 09/14/2023] [Indexed: 09/17/2023]
Abstract
INTRODUCTION Monoclonal antibodies (mAbs) should be administered by inhalation rather than parenterally to improve their efficiency in lung diseases. However, the pulmonary administration of mAbs in terms of aerosol technology and the formulation for inhalation is difficult. AREAS COVERED The feasible or suitable strategies for overcoming the barriers associated with administering mAbs are described. EXPERT OPINION Providing mAbs via inhalation to individuals with lung disorders is still difficult. However, inhalation is a desirable method for mAb delivery. Inhaled mAb production needs to be well thought out. The illness, the patient group(s), the therapeutic molecule selected, its interaction with the biological barriers in the lungs, the formulation, excipients, and administration systems must all be thoroughly investigated. Therefore, to create inhaled mAbs that are stable and efficacious, it will be essential to thoroughly examine the problems linked to instability and protein aggregation. More excipients will also need to be manufactured, expanding the range of formulation design choices. Another crucial requirement is for novel carriers for topical delivery to the lungs since carriers might significantly enhance proteins' stability and pharmacokinetic profile.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Luigino Calzetta
- Unit of Respiratory Diseases and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Barbara Rinaldi
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
9
|
Pitiot A, Ferreira M, Parent C, Boisseau C, Cortes M, Bouvart L, Paget C, Heuzé-Vourc'h N, Sécher T. Mucosal administration of anti-bacterial antibodies provide long-term cross-protection against Pseudomonas aeruginosa respiratory infection. Mucosal Immunol 2023; 16:312-325. [PMID: 36990281 DOI: 10.1016/j.mucimm.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/22/2023] [Accepted: 03/12/2023] [Indexed: 03/30/2023]
Abstract
Bacterial respiratory infections, either acute or chronic, are major threats to human health. Direct mucosal administration, through the airways, of therapeutic antibodies (Abs) offers a tremendous opportunity to benefit patients with respiratory infections. The mode of action of anti-infective Abs relies on pathogen neutralization and crystallizable fragment (Fc)-mediated recruitment of immune effectors to facilitate their elimination. Using a mouse model of acute pneumonia induced by Pseudomonas aeruginosa, we depicted the immunomodulatory mode of action of a neutralizing anti-bacterial Abs. Beyond the rapid and efficient containment of the primary infection, the Abs delivered through the airways harnessed genuine innate and adaptive immune responses to provide long-term protection, preventing secondary bacterial infection. In vitro antigen-presenting cells stimulation assay, as well as in vivo bacterial challenges and serum transfer experiments indicate an essential contribution of immune complexes with the Abs and pathogen in the induction of the sustained and protective anti-bacterial humoral response. Interestingly, the long-lasting response protected partially against secondary infections with heterologous P. aeruginosa strains. Overall, our findings suggest that Abs delivered mucosally promotes bacteria neutralization and provides protection against secondary infection. This opens novel perspectives for the development of anti-infective Abs delivered to the lung mucosa, to treat respiratory infections.
Collapse
Affiliation(s)
- Aubin Pitiot
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Marion Ferreira
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Christelle Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Chloé Boisseau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Mélanie Cortes
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Laura Bouvart
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Christophe Paget
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France
| | - Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France.
| |
Collapse
|
10
|
Hye T, Moinuddin SM, Sarkar T, Nguyen T, Saha D, Ahsan F. An evolving perspective on novel modified release drug delivery systems for inhalational therapy. Expert Opin Drug Deliv 2023; 20:335-348. [PMID: 36720629 PMCID: PMC10699164 DOI: 10.1080/17425247.2023.2175814] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/30/2023] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Drugs delivered via the lungs are predominantly used to treat various respiratory disorders, including asthma, chronic obstructive pulmonary diseases, respiratory tract infections and lung cancers, and pulmonary vascular diseases such as pulmonary hypertension. To treat respiratory diseases, targeted, modified or controlled release inhalation formulations are desirable for improved patient compliance and superior therapeutic outcome. AREAS COVERED This review summarizes the important factors that have an impact on the inhalable modified release formulation approaches with a focus toward various formulation strategies, including dissolution rate-controlled systems, drug complexes, site-specific delivery, drug-polymer conjugates, and drug-polymer matrix systems, lipid matrix particles, nanosystems, and formulations that can bypass clearance via mucociliary system and alveolar macrophages. EXPERT OPINION Inhaled modified release formulations can potentially reduce dosing frequency by extending drug's residence time in the lungs. However, inhalable modified or controlled release drug delivery systems remain unexplored and underdeveloped from the commercialization perspective. This review paper addresses the current state-of-the-art of inhaled controlled release formulations, elaborates on the avenues for developing newer technologies for formulating various drugs with tailored release profiles after inhalational delivery and explains the challenges associated with translational feasibility of modified release inhalable formulations.
Collapse
Affiliation(s)
- Tanvirul Hye
- Oakland University William Beaumont School of Medicine, 586 Pioneer Dr, 48309, Rochester, MI, USA
| | - Sakib M. Moinuddin
- California Northstate University, College of Pharmacy, 9700 West Taron Drive, 95757, Elk Grove, CA, USA
- East Bay Institute for Research & Education (EBIRE), 95655, Mather, CA, USA
| | - Tanoy Sarkar
- California Northstate University, College of Pharmacy, 9700 West Taron Drive, 95757, Elk Grove, CA, USA
- East Bay Institute for Research & Education (EBIRE), 95655, Mather, CA, USA
| | - Trieu Nguyen
- California Northstate University, College of Pharmacy, 9700 West Taron Drive, 95757, Elk Grove, CA, USA
- East Bay Institute for Research & Education (EBIRE), 95655, Mather, CA, USA
| | - Dipongkor Saha
- California Northstate University, College of Pharmacy, 9700 West Taron Drive, 95757, Elk Grove, CA, USA
| | - Fakhrul Ahsan
- California Northstate University, College of Pharmacy, 9700 West Taron Drive, 95757, Elk Grove, CA, USA
- East Bay Institute for Research & Education (EBIRE), 95655, Mather, CA, USA
- MedLuidics, 95757, Elk Grove, CA, USA
| |
Collapse
|
11
|
Montefusco-Pereira CV. Steps toward nebulization in-use studies to understand the stability of new biological entities. Drug Discov Today 2023; 28:103461. [PMID: 36455828 PMCID: PMC9770090 DOI: 10.1016/j.drudis.2022.103461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
The need for novel biological drugs against respiratory diseases has been highlighted during the Coronavirus (COVID-19) pandemic. The use of inhalation presents challenges to drug product stability, which is especially true for delivery using nebulizers (jet versus mesh technologies). The late-stage process of drug development in the pharmaceutical industry requires the investigation of in-use stability. In-use studies generate data that are guided by the requirements of regulatory authorities for inclusion in the clinical trial application dossier. In this review, I introduce the initial aspects of in-use stability studies during the development of an aerosol formulation to deliver biologics with a nebulizer. Lessons learned from this experience can guide future development and planning for formulation, analytics, material compatibility, nebulization process, and clinical trial preparations.
Collapse
|
12
|
Man F, Tang J, Swedrowska M, Forbes B, T M de Rosales R. Imaging drug delivery to the lungs: Methods and applications in oncology. Adv Drug Deliv Rev 2023; 192:114641. [PMID: 36509173 PMCID: PMC10227194 DOI: 10.1016/j.addr.2022.114641] [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: 08/31/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/14/2022]
Abstract
Direct delivery to the lung via inhalation is arguably one of the most logical approaches to treat lung cancer using drugs. However, despite significant efforts and investment in this area, this strategy has not progressed in clinical trials. Imaging drug delivery is a powerful tool to understand and develop novel drug delivery strategies. In this review we focus on imaging studies of drug delivery by the inhalation route, to provide a broad overview of the field to date and attempt to better understand the complexities of this route of administration and the significant barriers that it faces, as well as its advantages. We start with a discussion of the specific challenges for drug delivery to the lung via inhalation. We focus on the barriers that have prevented progress of this approach in oncology, as well as the most recent developments in this area. This is followed by a comprehensive overview of the different imaging modalities that are relevant to lung drug delivery, including nuclear imaging, X-ray imaging, magnetic resonance imaging, optical imaging and mass spectrometry imaging. For each of these modalities, examples from the literature where these techniques have been explored are provided. Finally the different applications of these technologies in oncology are discussed, focusing separately on small molecules and nanomedicines. We hope that this comprehensive review will be informative to the field and will guide the future preclinical and clinical development of this promising drug delivery strategy to maximise its therapeutic potential.
Collapse
Affiliation(s)
- Francis Man
- School of Cancer & Pharmaceutical Sciences, King's College London, London, SE1 9NH, United Kingdom
| | - Jie Tang
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Magda Swedrowska
- School of Cancer & Pharmaceutical Sciences, King's College London, London, SE1 9NH, United Kingdom
| | - Ben Forbes
- School of Cancer & Pharmaceutical Sciences, King's College London, London, SE1 9NH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, United Kingdom.
| |
Collapse
|
13
|
Reinders LMH, Noelle D, Klassen MD, Jaeger M, Schmidt TC, Tuerk J, Teutenberg T. Development and validation of a method for airborne monoclonal antibodies to quantify workplace exposure. J Pharm Biomed Anal 2022; 221:115046. [PMID: 36152489 DOI: 10.1016/j.jpba.2022.115046] [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: 07/26/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 10/31/2022]
Abstract
Modern therapy strategies are based on patient-specific treatment where the drug and dose are optimally adapted to the patient's needs. In recent drugs, monoclonal antibodies (mAbs) are increasingly used as active ingredients. Their patient-specific formulations are not part of the pharmaceutical industry's manufacturing process but are prepared from concentrates by pharmaceutical personnel. During the manufacturing process, however, active pharmaceutical ingredients are released in trace amounts or, in the case of accidents and spills, also in high concentrations. Regardless of the source of entry, mAbs can become airborne, be inhaled, and cause undesirable side-effects such as sensitization. To assess the risk for pharmaceutical personnel, a personal air sampling method was developed and validated for bevacizumab, cetuximab, daratumumab, omalizumab, rituximab and trastuzumab. The method is based on the combination of high-performance liquid chromatography with tandem mass spectrometry (HPLC-MS/MS). The analytical method achieves a limit of detection of 0.30-8.8 ng mL-1, recoveries of 83-96 % (intra-day assay) and 75-89 % (inter-day assay), with no detectable carry-over. A polycarbonate filter proved suitable for sampling airborne monoclonal antibodies, as it achieved 80-104 % recovery across all mAbs. It also showed concentration-independent desorption efficiency. The sampling duration can be up to 480 min without negatively affecting the recovery. MAbs are stable on the polycarbonate filter at 5 °C for 3 days (recovery: 94 % ± 5 %) and at - 20 °C for 14 days (recovery: 97 % ± 4 %). Our method demonstrated that there is a potential for release when handling monoclonal antibodies. However, this can be reduced below the limit of detection by using pressure equalization systems (spikes).
Collapse
Affiliation(s)
- Lars M H Reinders
- Institut für Energie und Umwelttechnik e. V. (IUTA, Institute of Energy and Environmental Technology), Bliersheimer Str. 58-60, 47229 Duisburg, Germany; Hochschule Niederrhein (University of Applied Science), Reinarzstr. 49, 47805 Krefeld, Germany; University Duisburg-Essen, Faculty of Chemistry, Instrumental Analytical Chemistry, Universitätsstr. 5, 45141 Essen, Germany
| | - Dennis Noelle
- Institut für Energie und Umwelttechnik e. V. (IUTA, Institute of Energy and Environmental Technology), Bliersheimer Str. 58-60, 47229 Duisburg, Germany; Hochschule Niederrhein (University of Applied Science), Reinarzstr. 49, 47805 Krefeld, Germany
| | - Martin D Klassen
- Institut für Energie und Umwelttechnik e. V. (IUTA, Institute of Energy and Environmental Technology), Bliersheimer Str. 58-60, 47229 Duisburg, Germany
| | - Martin Jaeger
- Hochschule Niederrhein (University of Applied Science), Reinarzstr. 49, 47805 Krefeld, Germany
| | - Torsten C Schmidt
- University Duisburg-Essen, Faculty of Chemistry, Instrumental Analytical Chemistry, Universitätsstr. 5, 45141 Essen, Germany
| | - Jochen Tuerk
- Institut für Energie und Umwelttechnik e. V. (IUTA, Institute of Energy and Environmental Technology), Bliersheimer Str. 58-60, 47229 Duisburg, Germany
| | - Thorsten Teutenberg
- Institut für Energie und Umwelttechnik e. V. (IUTA, Institute of Energy and Environmental Technology), Bliersheimer Str. 58-60, 47229 Duisburg, Germany.
| |
Collapse
|
14
|
Hufnagel S, Xu H, Sahakijpijarn S, Moon C, Chow LQ, Williams III RO, Cui Z. Dry Powders for Inhalation Containing Monoclonal Antibodies Made by Thin-Film Freeze-Drying. Int J Pharm 2022; 618:121637. [DOI: 10.1016/j.ijpharm.2022.121637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
|
15
|
Cazzola M, Ora J, Cavalli F, Rogliani P, Matera MG. An Overview of the Safety and Efficacy of Monoclonal Antibodies for the Chronic Obstructive Pulmonary Disease. Biologics 2021; 15:363-374. [PMID: 34475751 PMCID: PMC8407524 DOI: 10.2147/btt.s295409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022]
Abstract
Several mAbs have been tested or are currently under clinical evaluation for the treatment of COPD. They can be subdivided into those that aim to block specific pro-inflammatory and pro-neutrophilic cytokines and chemokines, such as TNF-α, IL-1β, CXCL8 and IL-1β, and those that act on T2-mediated inflammation, respectively, by blocking IL-5 and/or its receptor, preventing IL-4 and IL-13 signaling, affecting IL-33 pathway and blocking TSLP. None of these approaches has proved to be effective, probably because in COPD there is no dominant cytokine or chemokine and, therefore, a single mAb cannot be effective on all pathways. With a more in-depth understanding of the numerous pheno/endotypic pathways that play a role in COPD, it may eventually be possible to identify those specific patients in whom some of these cytokines or chemokines might predominate. In this case, it will be possible to implement a personalized treatment, but the use of each mAb will only be reserved for a very limited number of subjects.
Collapse
Affiliation(s)
- Mario Cazzola
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Francesco Cavalli
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Maria Gabriella Matera
- Chair of Pharmacology, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
16
|
Parray HA, Shukla S, Perween R, Khatri R, Shrivastava T, Singh V, Murugavelu P, Ahmed S, Samal S, Sharma C, Sinha S, Luthra K, Kumar R. Inhalation monoclonal antibody therapy: a new way to treat and manage respiratory infections. Appl Microbiol Biotechnol 2021; 105:6315-6332. [PMID: 34423407 PMCID: PMC8380517 DOI: 10.1007/s00253-021-11488-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/14/2021] [Accepted: 07/30/2021] [Indexed: 12/23/2022]
Abstract
The route of administration of a therapeutic agent has a substantial impact on its success. Therapeutic antibodies are usually administered systemically, either directly by intravenous route, or indirectly by intramuscular or subcutaneous injection. However, treatment of diseases contained within a specific tissue necessitates a better alternate route of administration for targeting localised infections. Inhalation is a promising non-invasive strategy for antibody delivery to treat respiratory maladies because it provides higher concentrations of antibody in the respiratory airways overcoming the constraints of entry through systemic circulation and uncertainity in the amount reaching the target tissue. The nasal drug delivery route is one of the extensively researched modes of administration, and nasal sprays for molecular drugs are deemed successful and are presently commercially marketed. This review highlights the current state and future prospects of inhaled therapies, with an emphasis on the use of monoclonal antibodies for the treatment of respiratory infections, as well as an overview of their importance, practical challenges, and clinical trial outcomes.Key points• Immunologic strategies for preventing mucosal transmission of respiratory pathogens.• Mucosal-mediated immunoprophylaxis could play a major role in COVID-19 prevention.• Applications of monoclonal antibodies in passive immunisation.
Collapse
Affiliation(s)
- Hilal Ahmad Parray
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Shivangi Shukla
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Reshma Perween
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Ritika Khatri
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Tripti Shrivastava
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Vanshika Singh
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Praveenkumar Murugavelu
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Shubbir Ahmed
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Sweety Samal
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Chandresh Sharma
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India
| | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rajesh Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, PO Box # 04, Faridabad, Haryana, 121001, India.
| |
Collapse
|
17
|
Fatima N, Kaushik V, Ayoub A. A Narrative Review of a Pulmonary Aerosolized Formulation or a Nasal Drop Using Sera Containing Neutralizing Antibodies Collected from COVID-19-Recovered Patients as a Probable Therapy for COVID-19. IRANIAN JOURNAL OF MEDICAL SCIENCES 2021; 46:151-168. [PMID: 34083848 PMCID: PMC8163704 DOI: 10.30476/ijms.2020.86417.1624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/29/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) emerged as a new contagion during December 2019, since which time it has triggered a rampant spike in fatality rates worldwide due to insufficient medical treatments and a lack of counteragents and prompted the World Health Organization to declare COVID-19 a public health emergency. It is, therefore, vital to accelerate the screening of new molecules or vaccines to win the battle against this pandemic. Experiences from previous epidemiological data on coronaviruses guide investigators in designing and exploring new compounds for a safe and cost-effective treatment. Several reports on the severe acute respiratory syndrome (SARS) epidemic indicate that severe acute respiratory syndrome coronavirus (SARS-CoV) and the novel COVID-19 use angiotensin-converting enzyme 2 (ACE2) as a receptor for binding to the host cell in the lung epithelia through the spike protein on their virion surface. ACE2 is a mono-carboxypeptidase best known for cleaving major peptides and substrates. Its degree in human airway epithelia positively correlates with coronavirus infection. The treatment approach can be the neutralization of the virus entering lung epithelial cells by using sera containing antibodies collected from COVID-19-recovered patients. Hence, we herein propose a pulmonary aerosolized formulation or a nasal drop using sera, which contain antibodies to prevent, treat, or immunize against COVID-19 infection.
Collapse
Affiliation(s)
- Nishat Fatima
- School of Pharmacy, Al-Hawash Private University, Homs, Syria
| | | | - Amjad Ayoub
- School of Pharmacy, Al-Hawash Private University, Homs, Syria
| |
Collapse
|
18
|
Novel formulations and drug delivery systems to administer biological solids. Adv Drug Deliv Rev 2021; 172:183-210. [PMID: 33705873 DOI: 10.1016/j.addr.2021.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in formulation sciences have expanded the previously limited design space for biological modalities, including peptide, protein, and vaccine products. At the same time, the discovery and application of new modalities, such as cellular therapies and gene therapies, have presented formidable challenges to formulation scientists. We explore these challenges and highlight the opportunities to overcome them through the development of novel formulations and drug delivery systems as biological solids. We review the current progress in both industry and academic laboratories, and we provide expert perspectives in those settings. Formulation scientists have made a tremendous effort to accommodate the needs of these novel delivery routes. These include stability-preserving formulations and dehydration processes as well as dosing regimes and dosage forms that improve patient compliance.
Collapse
|
19
|
Mayor A, Thibert B, Huille S, Respaud R, Audat H, Heuzé-Vourc'h N. Inhaled antibodies: formulations require specific development to overcome instability due to nebulization. Drug Deliv Transl Res 2021; 11:1625-1633. [PMID: 33768475 PMCID: PMC7993445 DOI: 10.1007/s13346-021-00967-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
Abstract Respiratory infections are life-threatening and therapeutic antibodies (Ab) have a tremendous opportunity to benefit to patients with pneumonia due to multidrug resistance bacteria or emergent virus, before a vaccine is manufactured. In respiratory infections, inhalation of anti-infectious Ab may be more relevant than intravenous (IV) injection-the standard route-to target the site of infection and improve Ab therapeutic index. One major challenge associated to Ab inhalation is to prevent protein instability during the aerosolization process. Ab drug development for IV injection aims to design a high-quality product, stable to different environment stress. In this study, we evaluated the suitability of Ab formulations developed for IV injection to be extended for inhalation delivery. We studied the aerosol characteristics and the aggregation profile of three Ab formulations developed for IV injection after nebulization, with two mesh nebulizers. Although the formulations for IV injection were compatible with mesh nebulization and deposition into the respiratory tract, the Ab were more unstable during nebulization than exposition to a vigorous shaking. Overall, our findings indicate that Ab formulations developed for IV delivery may not easily be repurposed for inhalation delivery and point to the requirement of a specific formulation development for inhaled Ab. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s13346-021-00967-w.
Collapse
Affiliation(s)
- Alexie Mayor
- INSERM, Centre D'Etude Des Pathologies Respiratoires, Universite François Rabelais de Tours, 10 Boulevard Tonnellé, U1100F-37032, Tours, France
- University of Tours, Tours, France
- Sanofi, Formulation and Process Development, Impasse Des Ateliers, 94400, Vitry-sur-Seine, France
| | - Béatrice Thibert
- Sanofi, Formulation and Process Development, Impasse Des Ateliers, 94400, Vitry-sur-Seine, France
| | - Sylvain Huille
- Sanofi, Formulation and Process Development, Impasse Des Ateliers, 94400, Vitry-sur-Seine, France
| | | | - Héloïse Audat
- Sanofi, Formulation and Process Development, Impasse Des Ateliers, 94400, Vitry-sur-Seine, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Centre D'Etude Des Pathologies Respiratoires, Universite François Rabelais de Tours, 10 Boulevard Tonnellé, U1100F-37032, Tours, France.
- University of Tours, Tours, France.
| |
Collapse
|
20
|
Okuda T, Okamoto H. Present Situation and Future Progress of Inhaled Lung Cancer Therapy: Necessity of Inhaled Formulations with Drug Delivery Functions. Chem Pharm Bull (Tokyo) 2021; 68:589-602. [PMID: 32611996 DOI: 10.1248/cpb.c20-00086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Inhaled lung cancer therapy is promising because of direct and noninvasive drug delivery to the lungs with low potential for severe systemic toxicity. Thus chemotherapeutic drugs have been administered clinically by nebulization of solution or suspension formulations, which demonstrated their limited pulmonary absorption and relatively mild systemic toxicity. In all these clinical trials, however, there was no obviously superior anticancer efficacy in lung cancer patients even at the maximum doses of drugs limited by pulmonary toxicity. Therefore methods that deliver both higher anticancer efficacy and lower pulmonary toxicity are strongly desired. In addition to the worldwide availability of pressured metered dose inhalers (pMDIs) and dry powder inhalers (DPIs) to treat local respiratory diseases, recent innovations in medicines and technologies are encouraging next steps toward effective inhaled lung cancer therapy with new therapeutic or drug delivery concepts. These include the discovery of target cells/molecules and drug candidates for novel cancer therapy, the development of high-performance inhalation devices for effective pulmonary drug delivery, and the establishment of manufacturing technologies for functional nanoparticles/microparticles. This review highlights the present situation and future progress of inhaled drugs for lung cancer therapy, including an overview of available inhalation devices, pharmacokinetics, and outcomes in clinical trials so far and some novel formulation strategies based on drug delivery systems to achieve enhanced anticancer efficacy and attenuated pulmonary toxicity.
Collapse
|
21
|
Jin Q, Zhu W, Zhu J, Zhu J, Shen J, Liu Z, Yang Y, Chen Q. Nanoparticle-Mediated Delivery of Inhaled Immunotherapeutics for Treating Lung Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007557. [PMID: 33448035 DOI: 10.1002/adma.202007557] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/24/2020] [Indexed: 05/18/2023]
Abstract
Despite the critical breakthrough achieved by immune checkpoint blockade (ICB), the clinical benefits are usually restricted by inefficient infiltration of immune cells and immune-associated adverse effects. Noninvasive aerosol inhalation, as a definitive procedure for treatment of respiratory diseases, for ICB immunotherapy against lung metastasis, has not been realized to the best knowledge. Herein, an inhaled immunotherapeutic chitosan (CS)-antibody complex is developed for immunotherapy against lung cancer. In this system, CS is used as a carrier to assemble with anti-programmed cell death protein ligand 1 (aPD-L1) to enable efficient transmucosal delivery. Moreover, CS exhibits adjuvant effects to drive potent immune responses via activating the cyclic-di-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. Interestingly, repeated inhalation of CS/aPD-L1 complex can effectively activate the immune system by promoting the infiltration of different immune cells especially CD8+ T cells around tumor lesions, and finally prolongs the survival of mice to 60 days. Thus, the work presents a unique aerosol inhalation delivery system for ICB antibody, which is promising for immunotherapy against lung metastasis without the concern of systemic toxicity.
Collapse
Affiliation(s)
- Qiutong Jin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Wenjun Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Jiafei Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jingjing Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
22
|
Establishment of a Mouse Ovarian Cancer and Peritoneal Metastasis Model to Study Intraperitoneal Chemotherapy. Cancers (Basel) 2020; 12:cancers12123818. [PMID: 33348855 PMCID: PMC7766637 DOI: 10.3390/cancers12123818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Intraperitoneal chemotherapy (IPC) is a locoregional treatment option in patients with peritoneal metastases (PM). Small animal models are valuable research tools allowing for rapid, reproducible, and inexpensive study and optimization of different forms of IPC. Here, we present a mouse model of ovarian cancer-derived PM and demonstrate its suitability for various modes of IPC, including pressurized intraperitoneal chemotherapy (PIPAC) using a micro-nozzle for aerosolized drug delivery. Abstract Intraperitoneal chemotherapy (IPC) is a locoregional treatment option in patients with peritoneal metastases (PM). Here, we present an ovarian cancer (OC)-derived PM mouse model for the study of different forms of IPC. Xenograft cell proliferation (luciferase-transfected OVCAR3 and SKOV3 clones) and growth kinetics were assessed using PET scan, bioluminescence imaging (BLI), and histological tumor analysis. Liquid IPC was achieved by intraperitoneal injection with/without capnoperitoneum (6–7 mmHg). Pressurized intraperitoneal aerosol chemotherapy (PIPAC) was mimicked using an intratracheal drug aerosol administration system (micro-nozzle), which, as demonstrated by ex vivo granulometric analysis using laser diffraction spectrometry, produced a polydisperse, bimodal aerosol with a volume-weighted median diameter of (26.49 ± 2.76) µm. Distribution of Tc-99m-labeled doxorubicin in mice was characterized using SPECT and was dependent on the delivery mode and most homogeneous when the micro-nozzle was used. A total of 2 mg doxorubicin per kg body weight was determined to be the optimally effective and tolerable dose to achieve at least 50% tumor reduction. Repeated PIPAC (four times at seven-day-intervals) with doxorubicin in SKOV3-luc tumor-bearing mice resulted in halted tumor proliferation and tumor load reduced after the second round of PIPAC versus controls and the number of tumor nodules was significantly reduced (27.7 ± 9.5 vs. 57.3 ± 9.5; p = 0.0003). Thus, we established the first mouse model of OC PM for the study of IPC using a human xenograft with SKOV3 cells and an experimental IPC setup with a miniaturized nozzle. Repeated IPC was feasible and demonstrated time-dependent anti-tumor activity.
Collapse
|
23
|
Matthews AA, Ee PLR, Ge R. Developing inhaled protein therapeutics for lung diseases. MOLECULAR BIOMEDICINE 2020; 1:11. [PMID: 34765995 PMCID: PMC7595758 DOI: 10.1186/s43556-020-00014-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/15/2020] [Indexed: 12/28/2022] Open
Abstract
Biologic therapeutics such as protein/polypeptide drugs are conventionally administered systemically via intravenous injection for the treatment of diseases including lung diseases, although this approach leads to low target site accumulation and the potential risk for systemic side effects. In comparison, topical delivery of protein drugs to the lung via inhalation is deemed to be a more effective approach for lung diseases, as proteins would directly reach the target in the lung while exhibiting poor diffusion into the systemic circulation, leading to higher lung drug retention and efficacy while minimising toxicity to other organs. This review examines the important considerations and challenges in designing an inhaled protein therapeutics for local lung delivery: the choice of inhalation device, structural changes affecting drug deposition in diseased lungs, clearance mechanisms affecting an inhaled protein drug’s lung accumulation, protein stability, and immunogenicity. Possible approaches to overcoming these issues will also be discussed.
Collapse
|
24
|
Airway Delivery of Anti-influenza Monoclonal Antibodies Results in Enhanced Antiviral Activities and Enables Broad-Coverage Combination Therapies. J Virol 2020; 94:JVI.00052-20. [PMID: 32847855 PMCID: PMC7592225 DOI: 10.1128/jvi.00052-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022] Open
Abstract
Influenza causes widespread illness in humans and can result in morbidity and death, especially in the very young and elderly populations. Because influenza vaccination can be poorly effective some years, and the immune systems of the most susceptible populations are often compromised, passive immunization treatments using broadly neutralizing antibodies is a promising therapeutic approach. However, large amounts of a single antibody are required for effectiveness when delivered through systemic administration (typically intravenous infusion), precluding the feasible dosing of antibody combinations via this route. The significance of our research is the demonstration that effective therapeutic treatments of multiple relevant influenza types (H1N1, H3N2, and B) can be achieved by airway administration of a single combination of relatively small amounts of three anti-influenza antibodies. This advance exploits the discovery that airway delivery is a more potent way of administering anti-influenza antibodies compared to systemic delivery, making this a feasible and cost-effective therapeutic approach. Effective and reliable anti-influenza treatments are acutely needed and passive immunizations using broadly neutralizing anti-influenza monoclonal antibodies (bNAbs) are a promising emerging approach. Because influenza infections are initiated in and localized to the pulmonary tract, and newly formed viral particles egress from the apical side of the lung epithelium, we compared the effectiveness of hemagglutinin (HA) stalk-binding bNAbs administered through the airway (intranasal or via nebulization) versus the systemic route (intraperitoneal or intravenous). Airway deliveries of various bNAbs were 10- to 50-fold more effective than systemic deliveries of the same bNAbs in treating H1N1, H3N2, B/Victoria-, and B/Yamagata-lineage influenza viral infections in mouse models. The potency of airway-delivered anti-HA bNAbs was highly dependent on antiviral neutralization activity, with little dependence on the effector function of the antibody. In contrast, the effectiveness of systemically delivered anti-HA bNAbs was not dependent on antiviral neutralization, but critically dependent on antibody effector functions. Concurrent administration of a neutralizing/effector function-positive bNAb via the airway and systemic routes showed increased effectiveness. The small amount of airway-delivered bNAbs needed for effective influenza treatment creates the opportunity to combine potent bNAbs with different anti-influenza specificities to generate a cost-effective antiviral therapy that provides broad coverage against all circulating influenza strains infecting humans. A 3 mg/kg dose of the novel triple antibody combination CF-404 (i.e., 1 mg/kg of each component bNAb) delivered to the airway was shown to effectively prevent weight loss and death in mice challenged with ten 50% lethal dose (LD50) inoculums of either H1N1, H3N2, B/Victoria-lineage, or B/Yamagata-lineage influenza viruses. IMPORTANCE Influenza causes widespread illness in humans and can result in morbidity and death, especially in the very young and elderly populations. Because influenza vaccination can be poorly effective some years, and the immune systems of the most susceptible populations are often compromised, passive immunization treatments using broadly neutralizing antibodies is a promising therapeutic approach. However, large amounts of a single antibody are required for effectiveness when delivered through systemic administration (typically intravenous infusion), precluding the feasible dosing of antibody combinations via this route. The significance of our research is the demonstration that effective therapeutic treatments of multiple relevant influenza types (H1N1, H3N2, and B) can be achieved by airway administration of a single combination of relatively small amounts of three anti-influenza antibodies. This advance exploits the discovery that airway delivery is a more potent way of administering anti-influenza antibodies compared to systemic delivery, making this a feasible and cost-effective therapeutic approach.
Collapse
|
25
|
Liang W, Pan HW, Vllasaliu D, Lam JKW. Pulmonary Delivery of Biological Drugs. Pharmaceutics 2020; 12:E1025. [PMID: 33114726 PMCID: PMC7693150 DOI: 10.3390/pharmaceutics12111025] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/13/2020] [Accepted: 10/20/2020] [Indexed: 12/19/2022] Open
Abstract
In the last decade, biological drugs have rapidly proliferated and have now become an important therapeutic modality. This is because of their high potency, high specificity and desirable safety profile. The majority of biological drugs are peptide- and protein-based therapeutics with poor oral bioavailability. They are normally administered by parenteral injection (with a very few exceptions). Pulmonary delivery is an attractive non-invasive alternative route of administration for local and systemic delivery of biologics with immense potential to treat various diseases, including diabetes, cystic fibrosis, respiratory viral infection and asthma, etc. The massive surface area and extensive vascularisation in the lungs enable rapid absorption and fast onset of action. Despite the benefits of pulmonary delivery, development of inhalable biological drug is a challenging task. There are various anatomical, physiological and immunological barriers that affect the therapeutic efficacy of inhaled formulations. This review assesses the characteristics of biological drugs and the barriers to pulmonary drug delivery. The main challenges in the formulation and inhalation devices are discussed, together with the possible strategies that can be applied to address these challenges. Current clinical developments in inhaled biological drugs for both local and systemic applications are also discussed to provide an insight for further research.
Collapse
Affiliation(s)
- Wanling Liang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, China; (H.W.P.); (J.K.W.L.)
| | - Harry W. Pan
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, China; (H.W.P.); (J.K.W.L.)
| | - Driton Vllasaliu
- School of Cancer and Pharmaceutical Sciences, King’s College London, 150 Stamford Street, London SE1 9NH, UK;
| | - Jenny K. W. Lam
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, China; (H.W.P.); (J.K.W.L.)
| |
Collapse
|
26
|
Abstract
Today, bio-medical efforts are entering the subcellular level, which is witnessed with the fast-developing fields of nanomedicine, nanodiagnostics and nanotherapy in conjunction with the implementation of nanoparticles for disease prevention, diagnosis, therapy and follow-up. Nanoparticles or nanocontainers offer advantages including high sensitivity, lower toxicity and improved safety—characteristics that are especially valued in the oncology field. Cancer cells develop and proliferate in complex microenvironments leading to heterogeneous diseases, often with a fatal outcome for the patient. Although antibody-based therapy is widely used in the clinical care of patients with solid tumours, its efficiency definitely needs improvement. Limitations of antibodies result mainly from their big size and poor penetration in solid tissues. Nanobodies are a novel and unique class of antigen-binding fragments, derived from naturally occurring heavy-chain-only antibodies present in the serum of camelids. Their superior properties such as small size, high stability, strong antigen-binding affinity, water solubility and natural origin make them suitable for development into next-generation biodrugs. Less than 30 years after the discovery of functional heavy-chain-only antibodies, the nanobody derivatives are already extensively used by the biotechnology research community. Moreover, a number of nanobodies are under clinical investigation for a wide spectrum of human diseases including inflammation, breast cancer, brain tumours, lung diseases and infectious diseases. Recently, caplacizumab, a bivalent nanobody, received approval from the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) for treatment of patients with thrombotic thrombocytopenic purpura.
Collapse
Affiliation(s)
- Ivana Jovčevska
- Medical Center for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Serge Muyldermans
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium.
| |
Collapse
|
27
|
Comparison between Colistin Sulfate Dry Powder and Solution for Pulmonary Delivery. Pharmaceutics 2020; 12:pharmaceutics12060557. [PMID: 32560289 PMCID: PMC7356940 DOI: 10.3390/pharmaceutics12060557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 11/16/2022] Open
Abstract
To assess the difference in the fate of the antibiotic colistin (COLI) after its pulmonary delivery as a powder or a solution, we developed a COLI powder and evaluated the COLI pharmacokinetic properties in rats after pulmonary administration of the powder or the solution. The amorphous COLI powder prepared by spray drying was characterized by a mass median aerodynamic diameter and fine particle fraction of 2.68 ± 0.07 µm and 59.5 ± 5.4%, respectively, when emitted from a Handihaler®. After intratracheal administration, the average pulmonary epithelial lining fluid (ELF): plasma area under the concentration versus time curves (AUC) ratios were 570 and 95 for the COLI solution and powder, respectively. However, the same COLI plasma concentration profiles were obtained with the two formulations. According to our pharmacokinetic model, this difference in ELF COLI concentration could be due to faster systemic absorption of COLI after the powder inhalation than for the solution. In addition, the COLI apparent permeability (Papp) across a Calu-3 epithelium model increased 10-fold when its concentration changed from 100 to 4000 mg/L. Based on this last result, we propose that the difference observed in vivo between the COLI solution and powder could be due to a high local ELF COLI concentration being obtained at the site where the dry particles impact the lung. This high local COLI concentration can lead to a local increase in COLI Papp, which is associated with a high concentration gradient and could produce a high local transfer of COLI across the epithelium and a consequent increase in the overall absorption rate of COLI.
Collapse
|
28
|
Rong Y, Pauly M, Guthals A, Pham H, Ehrbar D, Zeitlin L, Mantis NJ. A Humanized Monoclonal Antibody Cocktail to Prevent Pulmonary Ricin Intoxication. Toxins (Basel) 2020. [PMID: 32235318 DOI: 10.3390/toxins1204215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
PB10 IgG1, a monoclonal antibody (MAb) directed against an immunodominant epitope on the enzymatic subunit (RTA) of ricin toxin (RT), has been shown to passively protect mice and non-human primates from an aerosolized lethal-dose RT challenge. However, it was recently demonstrated that the therapeutic efficacy of PB10 IgG1 is significantly improved when co-administered with a second MAb, SylH3, targeting RT's binding subunit (RTB). Here we report that the PB10/SylH3 cocktail is also superior to PB10 alone when used as a pre-exposure prophylactic (PrEP) in a mouse model of intranasal RT challenge. The benefit of the PB10/SylH3 cocktail prompted us to engineer a humanized IgG1 version of SylH3 (huSylH3). The huPB10/huSylH3 cocktail proved highly efficacious in the mouse model, thereby opening the door to future testing in non-human primates.
Collapse
MESH Headings
- Administration, Inhalation
- Animals
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/pharmacology
- Antidotes/administration & dosage
- Antidotes/pharmacology
- Chlorocebus aethiops
- Disease Models, Animal
- Drug Therapy, Combination
- Female
- Lung Diseases/chemically induced
- Lung Diseases/prevention & control
- Mice, Inbred BALB C
- Pre-Exposure Prophylaxis
- Ricin/antagonists & inhibitors
- Ricin/immunology
- Vero Cells
Collapse
Affiliation(s)
- Yinghui Rong
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - Michael Pauly
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Adrian Guthals
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Henry Pham
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Dylan Ehrbar
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| |
Collapse
|
29
|
A Humanized Monoclonal Antibody Cocktail to Prevent Pulmonary Ricin Intoxication. Toxins (Basel) 2020; 12:toxins12040215. [PMID: 32235318 PMCID: PMC7232472 DOI: 10.3390/toxins12040215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
PB10 IgG1, a monoclonal antibody (MAb) directed against an immunodominant epitope on the enzymatic subunit (RTA) of ricin toxin (RT), has been shown to passively protect mice and non-human primates from an aerosolized lethal-dose RT challenge. However, it was recently demonstrated that the therapeutic efficacy of PB10 IgG1 is significantly improved when co-administered with a second MAb, SylH3, targeting RT’s binding subunit (RTB). Here we report that the PB10/SylH3 cocktail is also superior to PB10 alone when used as a pre-exposure prophylactic (PrEP) in a mouse model of intranasal RT challenge. The benefit of the PB10/SylH3 cocktail prompted us to engineer a humanized IgG1 version of SylH3 (huSylH3). The huPB10/huSylH3 cocktail proved highly efficacious in the mouse model, thereby opening the door to future testing in non-human primates.
Collapse
|
30
|
Cazzola M, Cavalli F, Usmani OS, Rogliani P. Advances in pulmonary drug delivery devices for the treatment of chronic obstructive pulmonary disease. Expert Opin Drug Deliv 2020; 17:635-646. [DOI: 10.1080/17425247.2020.1739021] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Mario Cazzola
- Department of Experimental Medicine, Unit of Respiratory Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Francesco Cavalli
- Department of Experimental Medicine, Unit of Respiratory Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Omar S. Usmani
- Imperial College London and Royal Brompton Hospital, Airways Disease Section, National Heart and Lung Institute (NHLI), London, UK
| | - Paola Rogliani
- Department of Experimental Medicine, Unit of Respiratory Medicine, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
31
|
Topical application of nebulized human IgG, IgA and IgAM in the lungs of rats and non-human primates. Respir Res 2019; 20:99. [PMID: 31118031 PMCID: PMC6532128 DOI: 10.1186/s12931-019-1057-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/24/2019] [Indexed: 01/31/2023] Open
Abstract
Background Recurrent and persistent infections are known to affect airways of patients with Primary Immunodeficiency despite appropriate replacement immunoglobulin serum levels. Interestingly, patients with Chronic Obstructive Pulmonary Disease or with non-CF bronchiectasis also show similar susceptibility to such infections. This may be due to the limited availability of immunoglobulins from the systemic circulation in the conductive airways, resulting in local immunodeficiency. Topical application of nebulized plasma-derived immunoglobulins may represent a means to address this deficiency. In this study, we assessed the feasibility of nebulizing plasma-derived immunoglobulins and delivering them into the airways of rats and non-human primates. Methods Distinct human plasma-derived immunoglobulin isotype preparations were nebulized with an investigational eFlow® nebulizer and analyzed in vitro or deposited into animals. Biochemical and immunohistological analysis of nebulized immunoglobulins were then performed. Lastly, efficacy of topically applied human plasma-derived immunoglobulins was assessed in an acute Streptococcus pneumoniae respiratory infection in mice. Results Characteristics of the resulting aerosols were comparable between preparations, even when using solutions with elevated viscosity. Neither the structural integrity nor the biological function of nebulized immunoglobulins were compromised by the nebulization process. In animal studies, immunoglobulins levels were assessed in plasma, broncho-alveolar lavages (BAL) and on lung sections of rats and non-human primates in samples collected up to 72 h following application. Nebulized immunoglobulins were detectable over 48 h in the BAL samples and up to 72 h on lung sections. Immunoglobulins recovered from BAL fluid up to 24 h after inhalation remained structurally and functionally intact. Importantly, topical application of human plasma-derived immunoglobulin G into the airways of mice offered significant protection against acute pneumococcal pneumonia. Conclusion Taken together our data demonstrate the feasibility of topically applying plasma-derived immunoglobulins into the lungs using a nebulized liquid formulation. Moreover, topically administered human plasma-derived immunoglobulins prevented acute respiratory infection. Electronic supplementary material The online version of this article (10.1186/s12931-019-1057-3) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Matera MG, Calzetta L, Rogliani P, Cazzola M. Monoclonal antibodies for severe asthma: Pharmacokinetic profiles. Respir Med 2019; 153:3-13. [PMID: 31136930 DOI: 10.1016/j.rmed.2019.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/05/2019] [Accepted: 05/09/2019] [Indexed: 12/27/2022]
Abstract
Several monoclonal antibodies (mAbs) (omalizumab, mepolizumab, reslizumab, benralizumab, and dupilumab) are currently approved for the treatment of severe asthma. They have complex pharmacokinetic profiles. These profiles are unique in that they are dependent on their structure as well as can be markedly influenced by the biology of their target antigen, but their general behaviour can still be considered a class property, similar to their endogenous IgG counterpart. They cannot be administered by oral route, have a slow distribution into tissue, are metabolized to peptides and amino acids in several tissues but are protected from degradation by binding to protective receptors (the FcRn), which explains their long elimination half-lives. Their clearance is nonlinear because of the saturation of the target-mediated elimination. Also anti-drug antibody (ADA) response and off-target binding, as well as their glycosylation pattern, can influence the pharmacokinetics of mAbs.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigino Calzetta
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
33
|
Sécher T, Dalonneau E, Ferreira M, Parent C, Azzopardi N, Paintaud G, Si-Tahar M, Heuzé-Vourc'h N. In a murine model of acute lung infection, airway administration of a therapeutic antibody confers greater protection than parenteral administration. J Control Release 2019; 303:24-33. [PMID: 30981816 DOI: 10.1016/j.jconrel.2019.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/06/2019] [Accepted: 04/03/2019] [Indexed: 11/28/2022]
Abstract
Due to growing antibiotic resistance, pneumonia caused by Pseudomonas aeruginosa is a major threat to human health and is driving the development of novel anti-infectious agents. Preventively or curatively administered pathogen-specific therapeutic antibodies (Abs) have several advantages, including a low level of toxicity and a unique pharmacological profile. At present, most Abs against respiratory infections are administered parenterally; this may not be optimal for therapeutics that have to reach the lungs to be effective. Although the airways constitute a logical delivery route for biologics designed to treat respiratory diseases, there are few scientific data on the advantages or disadvantages of this route in the context of pneumonia treatment. The objective of the present study was to evaluate the efficacy and fate of an anti-P. aeruginosa Ab targeting pcrV (mAb166) as a function of the administration route during pneumonia. The airway-administered mAb166 displayed a favorable pharmacokinetic profile during the acute phase of the infection, and was associated with greater protection (relative to other delivery routes) of infected animals. Airway administration was associated with lower levels of lung inflammation, greater bacterial clearance, and recruitment of neutrophils in the airways. In conclusion, the present study is the first to have compared the pharmacokinetics and efficacy of an anti-infectious Ab administered by different routes in an animal model of pneumonia. Our findings suggest that local delivery to the airways is associated with a more potent anti-bacterial response (relative to parenteral administration), and thus open up new perspectives for the prevention and treatment of pneumonia with Abs.
Collapse
Affiliation(s)
- Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Emilie Dalonneau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Marion Ferreira
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France; CHRU de Tours, Département de Pneumologie et d'exploration respiratoire fonctionnelle, F-37032 Tours, France
| | - Christelle Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | | | - Gilles Paintaud
- Université de Tours, GICC, PATCH Team, F-37032 Tours, France; CHRU de Tours, Laboratoire de Pharmacologie-Toxicologie, F-37032 Tours, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France.
| |
Collapse
|
34
|
Koernig S, Campbell IK, Mackenzie-Kludas C, Schaub A, Loetscher M, Ching Ng W, Zehnder R, Pelczar P, Sanli I, Alhamdoosh M, Ng M, Brown LE, Käsermann F, Vonarburg C, Zuercher AW. Topical application of human-derived Ig isotypes for the control of acute respiratory infection evaluated in a human CD89-expressing mouse model. Mucosal Immunol 2019; 12:1013-1024. [PMID: 31105268 PMCID: PMC7746524 DOI: 10.1038/s41385-019-0167-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/11/2019] [Accepted: 03/27/2019] [Indexed: 02/04/2023]
Abstract
Recurrent and persistent airway infections remain prevalent in patients with primary immunodeficiency (PID), despite restoration of serum immunoglobulin levels by intravenous or subcutaneous plasma-derived IgG. We investigated the effectiveness of different human Ig isotype preparations to protect mice against influenza when delivered directly to the respiratory mucosa. Four polyvalent Ig preparations from pooled plasma were compared: IgG, monomeric IgA (mIgA), polymeric IgA-containing IgM (IgAM) and IgAM associated with the secretory component (SIgAM). To evaluate these preparations, a transgenic mouse expressing human FcαRI/CD89 within the myeloid lineage was created. CD89 was expressed on all myeloid cells in the lung and blood except eosinophils, reflecting human CD89 expression. Intranasal administration of IgA-containing preparations was less effective than IgG in reducing pulmonary viral titres after infection of mice with A/California/7/09 (Cal7) or the antigenically distant A/Puerto Rico/8/34 (PR8) viruses. However, IgA reduced weight loss and inflammatory mediator expression. Both IgG and IgA protected mice from a lethal dose of PR8 virus and for mIgA, this effect was partially CD89 dependent. Our data support the beneficial effect of topically applied Ig purified from pooled human plasma for controlling circulating and non-circulating influenza virus infections. This may be important for reducing morbidity in PID patients.
Collapse
Affiliation(s)
- Sandra Koernig
- 0000 0001 1512 2287grid.1135.6CSL Limited, Bio21 Institute, 30 Flemington Rd, Parkville, VIC 3010 Australia
| | - Ian K. Campbell
- 0000 0001 1512 2287grid.1135.6CSL Limited, Bio21 Institute, 30 Flemington Rd, Parkville, VIC 3010 Australia
| | - Charley Mackenzie-Kludas
- 0000 0001 2179 088Xgrid.1008.9Department of Microbiology and Immunology The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, 792 Elizabeth St, Melbourne, VIC 3000 Australia
| | - Alexander Schaub
- 0000 0004 0646 1916grid.488260.0CSL Behring AG, Wankdorfstrasse 10, 3010 Bern, Switzerland
| | - Marius Loetscher
- 0000 0004 0646 1916grid.488260.0CSL Behring AG, Wankdorfstrasse 10, 3010 Bern, Switzerland
| | - Wy Ching Ng
- 0000 0001 2179 088Xgrid.1008.9Department of Microbiology and Immunology The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, 792 Elizabeth St, Melbourne, VIC 3000 Australia
| | - Roland Zehnder
- 0000 0004 0646 1916grid.488260.0CSL Behring AG, Wankdorfstrasse 10, 3010 Bern, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, Mattenstrasse 22, 4002 Basel, Switzerland
| | - Ildem Sanli
- Center for Transgenic Models, Mattenstrasse 22, 4002 Basel, Switzerland
| | - Monther Alhamdoosh
- 0000 0001 1512 2287grid.1135.6CSL Limited, Bio21 Institute, 30 Flemington Rd, Parkville, VIC 3010 Australia
| | - Milica Ng
- 0000 0001 1512 2287grid.1135.6CSL Limited, Bio21 Institute, 30 Flemington Rd, Parkville, VIC 3010 Australia
| | - Lorena E. Brown
- 0000 0001 2179 088Xgrid.1008.9Department of Microbiology and Immunology The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, 792 Elizabeth St, Melbourne, VIC 3000 Australia
| | - Fabian Käsermann
- 0000 0004 0646 1916grid.488260.0CSL Behring AG, Wankdorfstrasse 10, 3010 Bern, Switzerland
| | - Cédric Vonarburg
- 0000 0004 0646 1916grid.488260.0CSL Behring AG, Wankdorfstrasse 10, 3010 Bern, Switzerland
| | - Adrian W. Zuercher
- 0000 0004 0646 1916grid.488260.0CSL Behring AG, Wankdorfstrasse 10, 3010 Bern, Switzerland
| |
Collapse
|
35
|
Bioavailability of protein therapeutics in rats following inhalation exposure: Relevance to occupational exposure limit calculations. Regul Toxicol Pharmacol 2018; 100:35-44. [PMID: 30291877 DOI: 10.1016/j.yrtph.2018.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/27/2018] [Accepted: 10/02/2018] [Indexed: 11/23/2022]
Abstract
Protein therapeutics represent a rapidly growing proportion of new medicines being developed by the pharmaceutical industry. As with any new drug, an Occupational Exposure Limit (OEL) should be developed to ensure worker safety. Part of the OEL determination addresses bioavailability (BA) after inhalation, which is poorly understood for protein therapeutics. To explore this, male Sprague-Dawley rats were exposed intravenously or by nose-only inhalation to one of five test proteins of varying molecular size (10-150 kDa), including a polyethylene glycol-conjugated protein. Blood, lung tissue and bronchoalveolar lavage (BAL) fluid were collected over various time-points depending on the expected test protein clearance (8 minutes-56 days), and analyzed to determine the pharmacokinetic profiles. Since the BAL half-life of the test proteins was observed to be > 4.5 h after an inhalation exposure, accumulation and direct lung effects should be considered in the hazard assessment for protein therapeutics with lung-specific targets. The key finding was the low systemic bioavailability after inhalation exposure for all test proteins (∼≤1%) which did not appear molecular weight-dependent. Given that this study examined the inhalation of typical protein therapeutics in a manner mimicking worker exposure, a default 1% BA assumption is reasonable to utilize when calculating OELs for protein therapeutics.
Collapse
|
36
|
Ferreira LM, Alonso JD, Kiill CP, Ferreira NN, Buzzá HH, Martins de Godoi DR, de Britto D, Assis OBG, Seraphim TV, Borges JC, Gremião MPD. Exploiting supramolecular interactions to produce bevacizumab-loaded nanoparticles for potential mucosal delivery. Eur Polym J 2018. [DOI: 10.1016/j.eurpolymj.2018.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
37
|
Jeannot V, Gauche C, Mazzaferro S, Couvet M, Vanwonterghem L, Henry M, Didier C, Vollaire J, Josserand V, Coll JL, Schatz C, Lecommandoux S, Hurbin A. Anti-tumor efficacy of hyaluronan-based nanoparticles for the co-delivery of drugs in lung cancer. J Control Release 2018; 275:117-128. [DOI: 10.1016/j.jconrel.2018.02.024] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/09/2018] [Accepted: 02/16/2018] [Indexed: 12/20/2022]
|
38
|
Heng X, Yeates DB. Generation of High Concentrations of Respirable Solid-Phase Aerosols from Viscous Fluids. AEROSOL SCIENCE AND TECHNOLOGY : THE JOURNAL OF THE AMERICAN ASSOCIATION FOR AEROSOL RESEARCH 2018; 52:933-952. [PMID: 30718938 PMCID: PMC6358172 DOI: 10.1080/02786826.2018.1488078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
High outputs of respirable solid-phase aerosols were generated from viscous solutions or suspensions of low and high molecular weight polyvinylprrolidone (PVP) solutions, 10% (w/v) albumin and, gamma-globulin solutions as well as 10.3% (w/v) surfactant suspensions. A central fluid flow was aerosolized by coaxial converging compressed air. The water was evaporated from the droplets using warm dilution air and infrared radiation. The resulting aerosol particles were concentrated using a virtual impactor. The aerosols were generated at fluid flow rates between 1 and 3 ml/min and delivered at a flow rate of 44 l/min as 2.6 - 3.6 μm MMAD aerosols with geometric standard deviations between 1.5 and 2. Increases in viscosity over the range of 4 to 39 cSt caused a modest increase in MMAD. Increases in aerosol exit orifice diameter was associated with a decrease in aerosol diameter. Increases in compressed air pressure caused a decrease in aerosol diameter. Increases in fluid flow rate resulted modest increases in MMAD together with proportional increases in output mass. Aerosolizing 10% 8 kDa PVP at 3 ml/min resulted in the delivery of 193 mg/min of PVP at 64% efficiency enabling 1.2 g to be collected in 7 min. Aerosolizing 10.3% surfactant suspensions at 3 ml/min resulted in the delivery of up to 163 mg/min with 59% efficiency. The surface tension of the surfactant was not changed by these processes. SEM showed dimpled particles of PVP, albumin and gamma globulin indicating that their aerodynamic diameter was less than their morphometric diameter.
Collapse
Affiliation(s)
| | - Donovan B. Yeates
- Corresponding Author: Dr. Donovan B. Yeates, KAER Biotherapeutics Corporation, 926 S. Andreasen Dr., Suite 105, Escondido, CA 92029, United States, , www.linkedin.com/in/donovan-yeates
| |
Collapse
|
39
|
Carter PJ, Lazar GA. Next generation antibody drugs: pursuit of the 'high-hanging fruit'. Nat Rev Drug Discov 2017; 17:197-223. [DOI: 10.1038/nrd.2017.227] [Citation(s) in RCA: 447] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Guillon A, Sécher T, Dailey LA, Vecellio L, de Monte M, Si-Tahar M, Diot P, Page CP, Heuzé-Vourc'h N. Insights on animal models to investigate inhalation therapy: Relevance for biotherapeutics. Int J Pharm 2017; 536:116-126. [PMID: 29180257 DOI: 10.1016/j.ijpharm.2017.11.049] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022]
Abstract
Acute and chronic respiratory diseases account for major causes of illness and deaths worldwide. Recent developments of biotherapeutics opened a new era in the treatment and management of patients with respiratory diseases. When considering the delivery of therapeutics, the inhaled route offers great promises with a direct, non-invasive access to the diseased organ and has already proven efficient for several molecules. To assist in the future development of inhaled biotherapeutics, experimental models are crucial to assess lung deposition, pharmacokinetics, pharmacodynamics and safety. This review describes the animal models used in pulmonary research for aerosol drug delivery, highlighting their advantages and limitations for inhaled biologics. Overall, non-clinical species must be selected with relevant scientific arguments while taking into account their complexities and interspecies differences, to help in the development of inhaled medicines and ensure their successful transposition in the clinics.
Collapse
Affiliation(s)
- A Guillon
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France; Université François Rabelais de Tours, F-37032, Tours, France; CHRU de Tours, Service de Médecine Intensive - Réanimation, F-37000, Tours, France
| | - T Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France; Université François Rabelais de Tours, F-37032, Tours, France
| | - L A Dailey
- Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck Str. 4, 06122, Halle (Saale), Germany
| | - L Vecellio
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France; Aerodrug, Université François Rabelais - Faculté de Médecine, Tours, France
| | - M de Monte
- Plateforme Scientifique et Technique (PST) Animaleries, Université F. Rabelais, F-37000, Tours, France
| | - M Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France; Université François Rabelais de Tours, F-37032, Tours, France
| | - P Diot
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France; Université François Rabelais de Tours, F-37032, Tours, France; CHRU de Tours, Service de Pneumologie, F-37000, Tours, France
| | - C P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - N Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France; Université François Rabelais de Tours, F-37032, Tours, France.
| |
Collapse
|
41
|
Bioley G, Monnerat J, Lötscher M, Vonarburg C, Zuercher A, Corthésy B. Plasma-Derived Polyreactive Secretory-Like IgA and IgM Opsonizing Salmonella enterica Typhimurium Reduces Invasion and Gut Tissue Inflammation through Agglutination. Front Immunol 2017; 8:1043. [PMID: 28900429 PMCID: PMC5581814 DOI: 10.3389/fimmu.2017.01043] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/11/2017] [Indexed: 12/27/2022] Open
Abstract
Due to the increasing emergence of antibiotic-resistant strains of enteropathogenic bacteria, development of alternative treatments to fight against gut infections is a major health issue. While vaccination requires that a proper combination of antigen, adjuvant, and delivery route is defined to elicit protective immunity at mucosae, oral delivery of directly active antibody preparations, referred to as passive immunization, sounds like a valuable alternative. Along the gut, the strategy suffers, however, from the difficulty to obtain sufficient amounts of antibodies with the appropriate specificity and molecular structure for mucosal delivery. Physiologically, at the antibody level, the protection of gastrointestinal mucosal surfaces against enteropathogens is principally mediated by secretory IgA and secretory IgM. We previously demonstrated that purified human plasma-derived IgA and IgM can be associated with secretory component to generate biologically active secretory-like IgA and IgM (SCIgA/M) that can protect epithelial cells from infection by Shigella flexneri in vitro. In this study, we aimed at evaluating the protective potential of these antibody preparations in vivo. We now establish that such polyreactive preparations bind efficiently to Salmonella enterica Typhimurium and trigger bacterial agglutination, as observed by laser scanning confocal microscopy. Upon delivery into a mouse ligated intestinal loop, SCIgA/M-mediated aggregates persist in the intestinal environment and limit the entry of bacteria into intestinal Peyer’s patches via immune exclusion. Moreover, oral administration to mice of immune complexes composed of S. Typhimurium and SCIgA/M reduces mucosal infection, systemic dissemination, and local inflammation. Altogether, our data provide valuable clues for the future appraisal of passive oral administration of polyreactive plasma-derived SCIgA/M to combat infection by a variety of enteropathogens.
Collapse
Affiliation(s)
- Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Justine Monnerat
- R&D Laboratory, Division of Immunology and Allergy, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | | | | | | | - Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
42
|
Costa-Gouveia J, Pancani E, Jouny S, Machelart A, Delorme V, Salzano G, Iantomasi R, Piveteau C, Queval CJ, Song OR, Flipo M, Deprez B, Saint-André JP, Hureaux J, Majlessi L, Willand N, Baulard A, Brodin P, Gref R. Combination therapy for tuberculosis treatment: pulmonary administration of ethionamide and booster co-loaded nanoparticles. Sci Rep 2017; 7:5390. [PMID: 28710351 PMCID: PMC5511234 DOI: 10.1038/s41598-017-05453-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/24/2017] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB) is a leading infectious cause of death worldwide. The use of ethionamide (ETH), a main second line anti-TB drug, is hampered by its severe side effects. Recently discovered "booster" molecules strongly increase the ETH efficacy, opening new perspectives to improve the current clinical outcome of drug-resistant TB. To investigate the simultaneous delivery of ETH and its booster BDM41906 in the lungs, we co-encapsulated these compounds in biodegradable polymeric nanoparticles (NPs), overcoming the bottlenecks inherent to the strong tendency of ETH to crystallize and the limited water solubility of this Booster. The efficacy of the designed formulations was evaluated in TB infected macrophages using an automated confocal high-content screening platform, showing that the drugs maintained their activity after incorporation in NPs. Among tested formulations, "green" β-cyclodextrin (pCD) based NPs displayed the best physico-chemical characteristics and were selected for in vivo studies. The NPs suspension, administered directly into mouse lungs using a Microsprayer®, was proved to be well-tolerated and led to a 3-log decrease of the pulmonary mycobacterial load after 6 administrations as compared to untreated mice. This study paves the way for a future use of pCD NPs for the pulmonary delivery of the [ETH:Booster] pair in TB chemotherapy.
Collapse
MESH Headings
- Administration, Inhalation
- Animals
- Antitubercular Agents/pharmacology
- Disease Models, Animal
- Drug Carriers
- Drug Compounding/methods
- Drug Synergism
- Drug Therapy, Combination/methods
- Ethionamide/pharmacology
- Female
- Humans
- Mice
- Mice, Inbred BALB C
- Mycobacterium tuberculosis/drug effects
- Mycobacterium tuberculosis/growth & development
- Mycobacterium tuberculosis/pathogenicity
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- Oxadiazoles/pharmacology
- Piperidines/pharmacology
- Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
- RAW 264.7 Cells
- Solubility
- Treatment Outcome
- Tuberculosis, Multidrug-Resistant/drug therapy
- Tuberculosis, Multidrug-Resistant/microbiology
- Tuberculosis, Multidrug-Resistant/pathology
- Tuberculosis, Pulmonary/drug therapy
- Tuberculosis, Pulmonary/microbiology
- Tuberculosis, Pulmonary/pathology
- beta-Cyclodextrins/chemistry
Collapse
Affiliation(s)
- Joana Costa-Gouveia
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Elisabetta Pancani
- University of Paris-Sud, University Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), 91405, Orsay, France
| | - Samuel Jouny
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Arnaud Machelart
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Vincent Delorme
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Giuseppina Salzano
- University of Paris-Sud, University Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), 91405, Orsay, France
| | - Raffaella Iantomasi
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Catherine Piveteau
- Univ. Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000, Lille, France
| | - Christophe J Queval
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Ok-Ryul Song
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Marion Flipo
- Univ. Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000, Lille, France
| | - Benoit Deprez
- Univ. Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000, Lille, France
| | | | - José Hureaux
- University Hospital Center of Angers, 49000, Angers, France
| | - Laleh Majlessi
- Pathogénomique Mycobactérienne Intégrée, Département de Génomes et Génétique, Institut Pasteur, Paris, France
| | - Nicolas Willand
- Univ. Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000, Lille, France
| | - Alain Baulard
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Priscille Brodin
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France.
| | - Ruxandra Gref
- University of Paris-Sud, University Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), 91405, Orsay, France.
| |
Collapse
|
43
|
de Lemos ML, Badry N, Kletas V, Fabbro J, Tew A. Safe handling of monoclonal antibodies: Too large to be hazardous? J Oncol Pharm Pract 2017; 24:218-220. [PMID: 29284346 DOI: 10.1177/1078155217698846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It has been argued that the larger molecular weight of hazardous monoclonal antibodies may prevent their dermal absorption via occupational exposure. However, this assertion does not seem to be supported by direct evidence. Although the larger molecular weight may render monoclonal antibodies less probable to achieve therapeutic systemic level through dermal absorption, the concern in occupational health is whether these drugs can possibly attain a detectable level through repeated dermal exposure. Currently, there is no direct evidence to support a particular molecular weight above which a drug cannot achieve a detectable level following repeated occupational exposure. Therefore, the precautionary principle would dictate that repeated exposure of healthcare workers to hazardous monoclonal antibodies should be kept to a minimum.
Collapse
Affiliation(s)
- Mário L de Lemos
- 1 Provincial Pharmacy, Systemic Therapy Program, BC Cancer Agency, Vancouver, BC, Canada
| | - Nadine Badry
- 2 Provincial Pharmacy Systemic Therapy Program, BC Cancer Agency, Victoria, BC, Canada
| | - Victoria Kletas
- 1 Provincial Pharmacy, Systemic Therapy Program, BC Cancer Agency, Vancouver, BC, Canada
| | - Joan Fabbro
- 3 Provincial Pharmacy Systemic Therapy Program, BC Cancer Agency, Kelowna, BC, Canada
| | - Amber Tew
- 3 Provincial Pharmacy Systemic Therapy Program, BC Cancer Agency, Kelowna, BC, Canada
| |
Collapse
|
44
|
Van Heeke G, Allosery K, De Brabandere V, De Smedt T, Detalle L, de Fougerolles A. Nanobodies® † †Nanobody is a registered trademark of Ablynx NV. as inhaled biotherapeutics for lung diseases. Pharmacol Ther 2017; 169:47-56. [DOI: 10.1016/j.pharmthera.2016.06.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2016] [Indexed: 02/06/2023]
|
45
|
Respaud R, Marchand D, Pelat T, Tchou-Wong KM, Roy CJ, Parent C, Cabrera M, Guillemain J, Mac Loughlin R, Levacher E, Fontayne A, Douziech-Eyrolles L, Junqua-Moullet A, Guilleminault L, Thullier P, Guillot-Combe E, Vecellio L, Heuzé-Vourc'h N. Development of a drug delivery system for efficient alveolar delivery of a neutralizing monoclonal antibody to treat pulmonary intoxication to ricin. J Control Release 2016; 234:21-32. [PMID: 27173943 DOI: 10.1016/j.jconrel.2016.05.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 12/13/2022]
Abstract
The high toxicity of ricin and its ease of production have made it a major bioterrorism threat worldwide. There is however no efficient and approved treatment for poisoning by ricin inhalation, although there have been major improvements in diagnosis and therapeutic strategies. We describe the development of an anti-ricin neutralizing monoclonal antibody (IgG 43RCA-G1) and a device for its rapid and effective delivery into the lungs for an application in humans. The antibody is a full-length IgG and binds to the ricin A-chain subunit with a high affinity (KD=53pM). Local administration of the antibody into the respiratory tract of mice 6h after pulmonary ricin intoxication allowed the rescue of 100% of intoxicated animals. Specific operational constraints and aerosolization stresses, resulting in protein aggregation and loss of activity, were overcome by formulating the drug as a dry-powder that is solubilized extemporaneously in a stabilizing solution to be nebulized. Inhalation studies in mice showed that this formulation of IgG 43RCA-G1 did not induce pulmonary inflammation. A mesh nebulizer was customized to improve IgG 43RCA-G1 deposition into the alveolar region of human lungs, where ricin aerosol particles mostly accumulate. The drug delivery system also comprises a semi-automatic reconstitution system to facilitate its use and a specific holding chamber to maximize aerosol delivery deep into the lung. In vivo studies in monkeys showed that drug delivery with the device resulted in a high concentration of IgG 43RCA-G1 in the airways for at least 6h after local deposition, which is consistent with the therapeutic window and limited passage into the bloodstream.
Collapse
Affiliation(s)
- Renaud Respaud
- Université François-Rabelais de Tours, UMR 1100, CHRU de Tours, Service de Pharmacie, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Denis Marchand
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France; Aerodrug, F-37032 Tours, France
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; Brétigny sur Orge, France; BIOTEM, Parc d'activité Bièvre Dauphine, Apprieu, France
| | - Kam-Meng Tchou-Wong
- NYU School of Medicine, Department of Environmental Medicine, 57 Old Forge Road, Tuxedo, New York 10987, USA
| | - Chad J Roy
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Christelle Parent
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Maria Cabrera
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Joël Guillemain
- SESAME, Expertise en toxicologie, Chambray-les-tours, France
| | | | | | | | | | | | - Laurent Guilleminault
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; Brétigny sur Orge, France
| | - Emmanuelle Guillot-Combe
- DGA, Direction de la Stratégie (DS), Mission pour la recherche et l'Innovation scientifique (MRIS), France
| | - Laurent Vecellio
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France; Aerodrug, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France.
| |
Collapse
|
46
|
Fellner RC, Terryah ST, Tarran R. Inhaled protein/peptide-based therapies for respiratory disease. Mol Cell Pediatr 2016; 3:16. [PMID: 27098663 PMCID: PMC4839019 DOI: 10.1186/s40348-016-0044-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/13/2016] [Indexed: 11/10/2022] Open
Abstract
Asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF) are all chronic pulmonary diseases, albeit with different etiologies, that are characterized by airflow limitation, chronic inflammation, and abnormal mucus production/rheology. Small synthetic molecule-based therapies are commonly prescribed for all three diseases. However, there has been increased interest in “biologicals” to treat these diseases. Biologicals typically constitute protein- or peptide-based therapies and are often more potent than small molecule-based drugs. In this review, we shall describe the pros and cons of several different biological-based therapies for respiratory disease, including dornase alfa, a recombinant DNAase that reduces mucus viscosity and short palate lung and nasal epithelial clone 1 (SPLUNC1)-derived peptides that treat Na+ hyperabsorption and rebalance CF airway surface liquid homeostasis.
Collapse
Affiliation(s)
- Robert C Fellner
- Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, University of North Carolina, 7102 Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC, 27599-7248, USA
| | - Shawn T Terryah
- Spyryx Biosciences, 801-9 Capitola Drive, Durham, NC, 27713, USA
| | - Robert Tarran
- Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, University of North Carolina, 7102 Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC, 27599-7248, USA. .,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
47
|
Raes F, Sobilo J, Le Mée M, Rétif S, Natkunarajah S, Lerondel S, Le Pape A. High Resolution Ultrasound and Photoacoustic Imaging of Orthotopic Lung Cancer in Mice: New Perspectives for Onco-Pharmacology. PLoS One 2016; 11:e0153532. [PMID: 27070548 PMCID: PMC4829195 DOI: 10.1371/journal.pone.0153532] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/30/2016] [Indexed: 11/18/2022] Open
Abstract
Objectives We have developed a relevant preclinical model associated with a specific imaging protocol dedicated to onco-pharmacology studies in mice. Materials and Methods We optimized both the animal model and an ultrasound imaging procedure to follow up longitudinally the lung tumor growth in mice. Moreover we proposed to measure by photoacoustic imaging the intratumoral hypoxia, which is a crucial parameter responsible for resistance to therapies. Finally, we compared ultrasound data to x-ray micro computed tomography and volumetric measurements to validate the relevance of this approach on the NCI-H460 human orthotopic lung tumor. Results This study demonstrates the ability of ultrasound imaging to detect and monitor the in vivo orthotopic lung tumor growth by high resolution ultrasound imaging. This approach enabled us to characterize key biological parameters such as oxygenation, perfusion status and vascularization of tumors. Conclusion Such an experimental approach has never been reported previously and it would provide a nonradiative tool for assessment of anticancer therapeutic efficacy in mice. Considering the absence of ultrasound propagation through the lung parenchyma, this strategy requires the implantation of tumors strictly located in the superficial posterior part of the lung.
Collapse
Affiliation(s)
- Florian Raes
- PHENOMIN-TAAM-UPS44, CIPA (Centre d’Imagerie du Petit Animal), CNRS Orléans, France
- * E-mail:
| | - Julien Sobilo
- PHENOMIN-TAAM-UPS44, CIPA (Centre d’Imagerie du Petit Animal), CNRS Orléans, France
| | - Marilyne Le Mée
- PHENOMIN-TAAM-UPS44, CIPA (Centre d’Imagerie du Petit Animal), CNRS Orléans, France
| | - Stéphanie Rétif
- PHENOMIN-TAAM-UPS44, CIPA (Centre d’Imagerie du Petit Animal), CNRS Orléans, France
| | - Sharuja Natkunarajah
- PHENOMIN-TAAM-UPS44, CIPA (Centre d’Imagerie du Petit Animal), CNRS Orléans, France
| | - Stéphanie Lerondel
- PHENOMIN-TAAM-UPS44, CIPA (Centre d’Imagerie du Petit Animal), CNRS Orléans, France
| | - Alain Le Pape
- PHENOMIN-TAAM-UPS44, CIPA (Centre d’Imagerie du Petit Animal), CNRS Orléans, France
- INSERM U1100, CEPR, University of Tours, France
| |
Collapse
|
48
|
Hertel SP, Winter G, Friess W. Protein stability in pulmonary drug delivery via nebulization. Adv Drug Deliv Rev 2015; 93:79-94. [PMID: 25312674 DOI: 10.1016/j.addr.2014.10.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 08/22/2014] [Accepted: 10/01/2014] [Indexed: 10/24/2022]
Abstract
Protein inhalation is a delivery route which offers high potential for direct local lung application of proteins. Liquid formulations are usually available in early stages of biopharmaceutical development and nebulizers are the device of choice for atomization avoiding additional process steps like drying and enabling fast progression to clinical trials. While some proteins were proven to remain stable throughout aerosolization e.g. DNase, many biopharmaceuticals are more susceptible towards the stresses encountered during nebulization. The main reason for protein instability is unfolding and aggregation at the air-liquid interface, a problem which is of particular challenge in the case of ultrasound and jet nebulizers due to recirculation of much of the generated droplets. Surfactants are an important formulation component to protect the sensitive biomolecules. A second important challenge is warming of ultrasound and vibrating mesh devices, which can be overcome by overfilling, precooled solutions or cooling of the reservoir. Ultimately, formulation development has to go hand in hand with device evaluation.
Collapse
|
49
|
Cortez-Jugo C, Qi A, Rajapaksa A, Friend JR, Yeo LY. Pulmonary monoclonal antibody delivery via a portable microfluidic nebulization platform. BIOMICROFLUIDICS 2015; 9:052603. [PMID: 25945147 PMCID: PMC4393410 DOI: 10.1063/1.4917181] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/27/2015] [Indexed: 05/06/2023]
Abstract
Nebulizers have considerable advantages over conventional inhalers for pulmonary drug administration, particularly because they do not require coordinated breath actuation to generate and deliver the aerosols. Nevertheless, besides being less amenable to miniaturization and hence portability, some nebulizers are prone to denature macromolecular drugs due to the large forces generated during aerosolization. Here, we demonstrate a novel portable acoustomicrofluidic device capable of nebulizing epidermal growth factor receptor (EGFR) monoclonal antibodies into a fine aerosol mist with a mass median aerodynamic diameter of approximately 1.1 μm, optimal for deep lung deposition via inhalation. The nebulized monoclonal antibodies were tested for their stability, immunoactivity, and pharmacological properties, which confirmed that nebulization did not cause significant degradation of the antibody. In particular, flow cytometry demonstrated that the antigen binding capability of the antibody is retained and able to reduce phosphorylation in cells overexpressing the EGFR, indicating that the aerosols generated by the device were loaded with stable and active monoclonal antibodies. The delivery of antibodies via inhalation, particularly for the treatment of lung cancer, is thus expected to enhance the efficacy of this protein therapeutic by increasing the local concentration where they are needed.
Collapse
Affiliation(s)
| | - Aisha Qi
- Micro/Nanophysics Research Laboratory, RMIT University , Melbourne, Victoria 3001, Australia
| | - Anushi Rajapaksa
- Murdoch Children's Research Institute , Parkville, Victoria 3052, Australia
| | - James R Friend
- Micro/Nanophysics Research Laboratory, RMIT University , Melbourne, Victoria 3001, Australia
| | - Leslie Y Yeo
- Micro/Nanophysics Research Laboratory, RMIT University , Melbourne, Victoria 3001, Australia
| |
Collapse
|
50
|
Hervé V, Rabbe N, Guilleminault L, Paul F, Schlick L, Azzopardi N, Duruisseaux M, Fouquenet D, Montharu J, Redini F, Paintaud G, Lemarié E, Cadranel J, Wislez M, Heuzé-Vourc'h N. VEGF neutralizing aerosol therapy in primary pulmonary adenocarcinoma with K-ras activating-mutations. MAbs 2015; 6:1638-48. [PMID: 25484066 DOI: 10.4161/mabs.34454] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
K-ras mutations promote angiogenesis in lung cancer and contribute to the drug resistance of cancer cells. It is not clear whether K-ras mutated adenocarcinomas are sensitive to anti-angiogenic therapy with monoclonal antibodies (mAbs) that target vascular endothelial growth factor (VEGF). Anti-angiogenic mAbs are usually delivered systemically, but only a small proportion reaches the lung after intravenous injection. We investigated the relevance of a non-invasive pulmonary route for the delivery of anti-VEGF mAbs in the mouse K-ras(LA1) model. We found that pulmonary delivery of these mAbs significantly reduced the number of tumor lesions and inhibited malignant progression. The antitumor effect involves the VEGFR2-dependent inhibition of blood vessel growth, which impairs tumor proliferation. Pharmacokinetic analysis of aerosolized anti-VEGF showed its low rate of passage into the bloodstream, suggesting that this delivery route is associated with reduced systemic side effects. Our findings highlight the value of the aerosol route for administration of anti-angiogenic mAbs in pulmonary adenocarcinoma with K-ras activating-mutations.
Collapse
|