1
|
Im C, Ahn JH, Lim H, Kim D, Lee YJ, Kang CS, Kang CM. Development of a Novel Prosthetic Click-Linker for Radioiodination of Antibody-Based Radiopharmaceuticals with High Stability and Specificity. Mol Pharm 2025; 22:295-303. [PMID: 39579150 DOI: 10.1021/acs.molpharmaceut.4c00897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Radioiodine has been exploited in nuclear medicine for diagnostic and therapeutic purposes in various diseases. There are two radioiodination methods for biomolecules, that is, (1) direct radioiodination of tyrosine or histidine residue in a biomolecule and (2) indirect radioiodination by using a prosthetic group, which bridges radioiodine and the biomolecule. While directly radioiodinated biomolecules suffer from deiodination in vivo, the most commonly used indirect labeling method based on N-succinimidyl-3-[*I]iodobenzoate has a problem of inconvenience due to an high-performance liquid chromatography (HPLC) purification process. To tackle both issues, a novel prosthetic click-linker-antibody conjugate (3-[123/125I]iodobenzoyl-PEG4-tetrazine-TCO-PEG4-trastuzumab (3-[123/125I]IBTTT)) with favorable radiochemical yield (>57%) and purity (>99%) was developed using a fluorous tin-based organotin precursor with streamlined purification process utilizing fluorous solid-phase extraction (FSPE) cartridge and spin column. In vitro binding studies demonstrated that 3-[125I]IBTTT maintained its biological activity with a KD value (5.606 nM) comparable to that of unmodified trastuzumab (5.0 nM). In vivo imaging of 3-[123I]IBTTT in a human epidermal growth factor receptor 2 (HER2)-expressing gastric cancer mouse model revealed favorable tumor accumulation and negligible thyroid uptake compared to directly radioiodinated trastuzumab ([123I]trastuzumab). It was also confirmed, by blocking experiments and a biodistribution study, that the tumor accumulation of 3-[123I]IBTTT was attributed to HER2-specific binding. In summary, we developed a novel radioiodinated prosthetic click-linker agent (3-[123/125I]IBTTT) with favorable radiochemical yield, purity, stability, and in vivo behavior, providing a highly promising tool for targeted imaging and potential therapy of HER2-positive cancers.
Collapse
Affiliation(s)
- Changkeun Im
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul 01812, Korea
| | - Jae Hun Ahn
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hwisoo Lim
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul 01812, Korea
| | - Dohyeon Kim
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
- Korea Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
| | - Chi Soo Kang
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul 01812, Korea
| | - Choong Mo Kang
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul 01812, Korea
| |
Collapse
|
2
|
Agrohia DK, Goswami R, Jantarat T, Çiçek YA, Thongsukh K, Jeon T, Bell JM, Rotello VM, Vachet RW. Suborgan Level Quantitation of Proteins in Tissues Delivered by Polymeric Nanocarriers. ACS NANO 2024; 18:16808-16818. [PMID: 38870478 PMCID: PMC11497159 DOI: 10.1021/acsnano.4c02344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Amidst the rapid growth of protein therapeutics as a drug class, there is an increased focus on designing systems to effectively deliver proteins to target organs. Quantitative monitoring of protein distributions in tissues is essential for optimal development of delivery systems; however, existing strategies can have limited accuracy, making it difficult to assess suborgan dosing. Here, we describe a quantitative imaging approach that utilizes metal-coded mass tags and laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) to quantify the suborgan distributions of proteins in tissues that have been delivered by polymeric nanocarriers. Using this approach, we measure nanomole per gram levels of proteins as delivered by guanidinium-functionalized poly(oxanorborneneimide) (PONI) polymers to various tissues, including the alveolar region of the lung. Due to the multiplexing capability of the LA-ICP-MS imaging, we are also able to simultaneously quantify protein and polymer distributions, obtaining valuable information about the relative excretion pathways of the protein cargo and carrier. This imaging approach will facilitate quantitative correlations between nanocarrier properties and protein cargo biodistributions.
Collapse
Affiliation(s)
- Dheeraj K. Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Teerapong Jantarat
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Yağız Anil Çiçek
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Korndanai Thongsukh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jonathan M. Bell
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
3
|
Iaboni M, Coppo A, Remotti D, Queliti R, Blasi F, Bussi S, Cabella C, Poggi L. Fluorescence-based absolute quantification of near-infrared probes in tissue extracts for biodistribution analyses. Anal Biochem 2023; 677:115251. [PMID: 37473979 DOI: 10.1016/j.ab.2023.115251] [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: 04/26/2023] [Revised: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
In recent years, significant progress has been made in the development of fluorescent contrast agents for clinical applications. For the development of a fluorescent probe, it is crucial to evaluate its safety profile, including biodistribution. Specific methods need to be developed for the absolute quantification of fluorescent probes in tissue specimens from animals administered with test compounds in the framework of biodistribution/efficacy/toxicity studies. Here, we describe a new method for the absolute quantification of fluorescent probes in tissue specimens from animals administered with compounds that have absorption and emission wavelength in the Near-Infrared region (600-800 nm). The protocol is based on the standard addition approach in order to minimize the interference of the matrix on the analyte signal causing inaccuracy in the absolute determination of the concentration. The measurement of the fluorescence intensity is done via a microplate reader. The method has been fully validated and applied for the quantification of a fluorescence-guided surgery targeted contrast agent in a Good Laboratory Practice (GLP) biodistribution study. Results clearly demonstrate that this procedure is fully applicable in a preclinical setting and that it overcomes common issues associated with fluorescence signal quantification in tissue extracts.
Collapse
Affiliation(s)
- Margherita Iaboni
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy.
| | - Alessandra Coppo
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Davide Remotti
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Roberta Queliti
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Francesco Blasi
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Simona Bussi
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Claudia Cabella
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| | - Luisa Poggi
- Bracco Imaging SpA, Centro Ricerche Bracco, Via Ribes 5, 10010, Colleretto Giacosa, Turin, Italy
| |
Collapse
|
4
|
van Dijk B, Hooning van Duyvenbode JFF, de Vor L, Nurmohamed FRHA, Lam MGEH, Poot AJ, Ramakers RM, Koustoulidou S, Beekman FJ, van Strijp J, Rooijakkers SHM, Dadachova E, Vogely HC, Weinans H, van der Wal BCH. Evaluating the Targeting of a Staphylococcus-aureus-Infected Implant with a Radiolabeled Antibody In Vivo. Int J Mol Sci 2023; 24:ijms24054374. [PMID: 36901805 PMCID: PMC10002501 DOI: 10.3390/ijms24054374] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/06/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Implant infections caused by Staphylococcus aureus are difficult to treat due to biofilm formation, which complicates surgical and antibiotic treatment. We introduce an alternative approach using monoclonal antibodies (mAbs) targeting S. aureus and provide evidence of the specificity and biodistribution of S.-aureus-targeting antibodies in a mouse implant infection model. The monoclonal antibody 4497-IgG1 targeting wall teichoic acid in S. aureus was labeled with indium-111 using CHX-A"-DTPA as a chelator. Single Photon Emission Computed Tomography/computed tomographyscans were performed at 24, 72 and 120 h after administration of the 111In-4497 mAb in Balb/cAnNCrl mice with a subcutaneous implant that was pre-colonized with S. aureus biofilm. The biodistribution of this labelled antibody over various organs was visualized and quantified using SPECT/CT imaging, and was compared to the uptake at the target tissue with the implanted infection. Uptake of the 111In-4497 mAbs at the infected implant gradually increased from 8.34 %ID/cm3 at 24 h to 9.22 %ID/cm3 at 120 h. Uptake at the heart/blood pool decreased over time from 11.60 to 7.58 %ID/cm3, whereas the uptake in the other organs decreased from 7.26 to less than 4.66 %ID/cm3 at 120 h. The effective half-life of 111In-4497 mAbs was determined to be 59 h. In conclusion, 111In-4497 mAbs were found to specifically detect S. aureus and its biofilm with excellent and prolonged accumulation at the site of the colonized implant. Therefore, it has the potential to serve as a drug delivery system for the diagnostic and bactericidal treatment of biofilm.
Collapse
Affiliation(s)
- Bruce van Dijk
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-88-75-569-71
| | | | - Lisanne de Vor
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | | | - Marnix G. E. H. Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Alex J. Poot
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ruud M. Ramakers
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Sofia Koustoulidou
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Freek J. Beekman
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Jos van Strijp
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - H. Charles Vogely
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of BioMechanical Engineering, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Bart C. H. van der Wal
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
5
|
Cancer therapy by antibody-targeted Cerenkov light and metabolism-selective photosensitization. J Control Release 2022; 352:25-34. [PMID: 36243234 DOI: 10.1016/j.jconrel.2022.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/29/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022]
Abstract
Photodynamic therapy (PDT) is an effective cancer treatment option, but it suffers from penetration limit of light, making it available only for superficial and endoscopically accessible cancers. Recently, there have been reports that Cerenkov luminescence originated from radioisotopes can be utilized as an excitation source for PDT without external light illumination. Here, cancer-selective agents, i.e., (1) clinically available 5-aminolevulinic acid (5-ALA), which promotes cancer metabolism-specific accumulation of protoporphyrin IX (PpIX), and (2) 64Cu-DOTA-trastuzumab, which has HER2-expressing cancer selective uptake, are separately applied as a photosensitizer and an in situ radiator, respectively, to potentiate tumor-specific Cerenkov luminescence energy transfer (CLET) from 64Cu to PpIX for high-precision PDT of cancer. It is shown that the combinational administration and tumor colocalization of 5-ALA and 64Cu-DOTA-trastuzumab exert significant in vitro cytotoxicity (cell viability <9%) as well as in vivo antitumor effects (tumor volume ratio of 0.50 on 14 days post-injection) on HER2-expressing breast and gastric cancer models. This study proves that high-precision treatment regimen using dual-targeted CLET-based PDT is feasible for HER2-expressing cancers. Furthermore, the results offer great potential for clinical translation to the dual-targeted CLET-based PDT because the treatment regimen uses components, 5-ALA and 64Cu-DOTA-trastuzumab, which are already in clinical uses.
Collapse
|
6
|
Allen KJH, Jiao R, Li J, Beckford-Vera DR, Dadachova E. In Vitro and In Vivo Characterization of 89Zirconium-Labeled Lintuzumab Molecule. Molecules 2022; 27:molecules27196589. [PMID: 36235126 PMCID: PMC9571394 DOI: 10.3390/molecules27196589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Positron emission tomography (PET) imaging is a powerful non-invasive method to determine the in vivo behavior of biomolecules. Determining biodistribution and pharmacokinetic (PK) properties of targeted therapeutics can enable a better understanding of in vivo drug mechanisms such as tumor uptake, off target accumulation and clearance. Zirconium-89 (89Zr) is a readily available tetravalent PET-enabling radiometal that has been used to evaluate the biodistribution and PK of monoclonal antibodies. In the current study, we performed in vitro and in vivo characterization of 89Zr-lintuzumab, a radiolabeled anti-CD33 antibody, as a model to evaluate the in vivo binding properties in preclinical models of AML. Methods: Lintuzumab was conjugated to p-SCN-Bn-deferoxamine (DFO) and labeled with 89Zr using a 5:1 µCi:µg specific activity at 37 °C for 1h. The biological activity of 89Zr-lintuzumab was evaluated in a panel of CD33 positive cells using flow cytometry. Fox Chase SCID mice were injected with 2 × 106 OCI-AML3 cells into the right flank. After 12 days, a cohort of mice (n = 4) were injected with 89Zr-lintuzumab via tail vein. PET/CT scans of mice were acquired on days 1, 2, 3 and 7 post 89Zr-lintuzumab injection. To demonstrate 89Zr-lintuzumab specific binding to CD33 expressing tumors in vivo, a blocking study was performed. This cohort of mice (n = 4) was injected with native lintuzumab and 24 h later 89Zr-lintuzumab was administered. This group was imaged 3 and 7 days after injection of 89Zr-lintuzumab. A full ex vivo biodistribution study on both cohorts was performed on day 7. The results from the PET image and ex vivo biodistribution studies were compared. Results: Lintuzumab was successfully radiolabeled with 89Zr resulting in a 99% radiochemical yield. The 89Zr-lintuzumab radioconjugate specifically binds CD33 positive cells in a similar manner to native lintuzumab as observed by flow cytometry. PET imaging revealed high accumulation of 89Zr-lintuzumab in OCI-AML3 tumors within 24h post-injection of the radioconjugate. The 89Zr-lintuzumab high tumor uptake remains for up to 7 days. Tumor analysis of the PET data using volume of interest (VOI) showed significant blocking of 89Zr-lintuzumab in the group pre-treated with native lintuzumab (pre-blocked group), thus indicating specific targeting of CD33 on OCI-AML3 cells in vivo. The tumor uptake findings from the PET imaging study are in agreement with those from the ex vivo biodistribution results. Conclusions: PET imaging of 89Zr-lintuzumab shows high specific uptake in CD33 positive human OCI-AML3 tumors. The results from the image study agree with the observations from the ex vivo biodistribution study. Our findings collectively suggest that PET imaging using 89Zr-lintuzumab could be a powerful drug development tool to evaluate binding properties of anti-CD33 monoclonal antibodies in preclinical cancer models.
Collapse
Affiliation(s)
- Kevin J. H. Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Rubin Jiao
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Jason Li
- Actinium Pharmaceuticals, New York, NY 10016, USA
| | | | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Correspondence: ; Tel.: +1-306-966-5163
| |
Collapse
|
7
|
Targeting Melanin in Melanoma with Radionuclide Therapy. Int J Mol Sci 2022; 23:ijms23179520. [PMID: 36076924 PMCID: PMC9455397 DOI: 10.3390/ijms23179520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/13/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Nearly 100,000 individuals are expected to be diagnosed with melanoma in the United States in 2022. Treatment options for late-stage metastatic disease up until the 2010s were few and offered only slight improvement to the overall survival. The introduction of B-RAF inhibitors and anti-CTLA4 and anti-PD-1/PD-L1 immunotherapies into standard of care brought measurable increases in the overall survival across all stages of melanoma. Despite the improvement in the survival statistics, patients treated with targeted therapies and immunotherapies are subject to very serious side effects, the development of drug resistance, and the high costs of treatment. This leaves room for the development of novel approaches as well as for the exploration of novel combination therapies for the treatment of metastatic melanoma. One such approach is targeting melanin pigment with radionuclide therapy. Advances in melanin-targeting radionuclide therapy of melanoma can be viewed from two spheres: (1) radioimmunotherapy (RIT) and (2) radiolabeled small molecules. The investigation of mechanisms of the action and efficacy of targeting melanin in melanoma treatment by RIT points to the involvement of the immune system such as complement dependent cytotoxicity. The combination of RIT with immunotherapy presents synergistic killing in mouse melanoma models. The field of radiolabeled small molecules is focused on radioiodinated compounds that have the ability to cross the cellular membranes to access intracellular melanin and can be applied in both therapy and imaging as theranostics. Clinical applications of targeting melanin with radionuclide therapies have produced encouraging results and clinical work is on-going. Continued work on targeting melanin with radionuclide therapy as a monotherapy, or possibly in combination with standard of care agents, has the potential to strengthen the current treatment options for melanoma patients.
Collapse
|
8
|
de Vor L, van Dijk B, van Kessel K, Kavanaugh JS, de Haas C, Aerts PC, Viveen MC, Boel EC, Fluit AC, Kwiecinski JM, Krijger GC, Ramakers RM, Beekman FJ, Dadachova E, Lam MGEH, Vogely HC, van der Wal BCH, van Strijp JAG, Horswill AR, Weinans H, Rooijakkers SHM. Human monoclonal antibodies against Staphylococcus aureus surface antigens recognize in vitro and in vivo biofilm. eLife 2022; 11:e67301. [PMID: 34989676 PMCID: PMC8751199 DOI: 10.7554/elife.67301] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 12/06/2021] [Indexed: 12/22/2022] Open
Abstract
Implant-associated Staphylococcus aureus infections are difficult to treat because of biofilm formation. Bacteria in a biofilm are often insensitive to antibiotics and host immunity. Monoclonal antibodies (mAbs) could provide an alternative approach to improve the diagnosis and potential treatment of biofilm-related infections. Here, we show that mAbs targeting common surface components of S. aureus can recognize clinically relevant biofilm types. The mAbs were also shown to bind a collection of clinical isolates derived from different biofilm-associated infections (endocarditis, prosthetic joint, catheter). We identify two groups of antibodies: one group that uniquely binds S. aureus in biofilm state and one that recognizes S. aureus in both biofilm and planktonic state. Furthermore, we show that a mAb recognizing wall teichoic acid (clone 4497) specifically localizes to a subcutaneously implanted pre-colonized catheter in mice. In conclusion, we demonstrate the capacity of several human mAbs to detect S. aureus biofilms in vitro and in vivo.
Collapse
Affiliation(s)
- Lisanne de Vor
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Bruce van Dijk
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
| | - Kok van Kessel
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Jeffrey S Kavanaugh
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Carla de Haas
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Piet C Aerts
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Marco C Viveen
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Edwin C Boel
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Ad C Fluit
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Jakub M Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Gerard C Krijger
- Department of Radiology and Nuclear Medicine, University Medical Centre UtrechtUtrechtNetherlands
| | - Ruud M Ramakers
- MILabs B.VUtrechtNetherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical CenterUtrechtNetherlands
- Department of Radiation Science and Technology, Delft University of TechnologyDelftNetherlands
| | - Freek J Beekman
- MILabs B.VUtrechtNetherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical CenterUtrechtNetherlands
- Department of Radiation Science and Technology, Delft University of TechnologyDelftNetherlands
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of SaskatchewanSaskatoonCanada
| | - Marnix GEH Lam
- Department of Radiology and Nuclear Medicine, University Medical Centre UtrechtUtrechtNetherlands
| | - H Charles Vogely
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
| | - Bart CH van der Wal
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
| | - Jos AG van Strijp
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
- Department of Veterans Affairs, Eastern Colorado Health Care SystemDenverUnited States
| | - Harrie Weinans
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
- Department of Biomechanical engineering, TU DelftDelftNetherlands
| | - Suzan HM Rooijakkers
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| |
Collapse
|
9
|
Usama SM, Marker SC, Caldwell DR, Patel NL, Feng Y, Kalen JD, St Croix B, Schnermann MJ. Targeted Fluorogenic Cyanine Carbamates Enable In Vivo Analysis of Antibody-Drug Conjugate Linker Chemistry. J Am Chem Soc 2021; 143:21667-21675. [PMID: 34928588 DOI: 10.1021/jacs.1c10482] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibody-drug conjugates (ADCs) are a rapidly emerging therapeutic platform. The chemical linker between the antibody and the drug payload plays an essential role in the efficacy and tolerability of these agents. New methods that quantitatively assess the cleavage efficiency in complex tissue settings could provide valuable insights into the ADC design process. Here we report the development of a near-infrared (NIR) optical imaging approach that measures the site and extent of linker cleavage in mouse models. This approach is enabled by a superior variant of our recently devised cyanine carbamate (CyBam) platform. We identify a novel tertiary amine-containing norcyanine, the product of CyBam cleavage, that exhibits a dramatically increased cellular signal due to an improved cellular permeability and lysosomal accumulation. The resulting cyanine lysosome-targeting carbamates (CyLBams) are ∼50× brighter in cells, and we find this strategy is essential for high-contrast in vivo targeted imaging. Finally, we compare a panel of several common ADC linkers across two antibodies and tumor models. These studies indicate that cathepsin-cleavable linkers provide dramatically higher tumor activation relative to hindered or nonhindered disulfides, an observation that is only apparent with in vivo imaging. This strategy enables quantitative comparisons of cleavable linker chemistries in complex tissue settings with implications across the drug delivery landscape.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Sierra C Marker
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Donald R Caldwell
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Nimit L Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
10
|
Functional in vitro assessment of modified antibodies: Impact of label on protein properties. PLoS One 2021; 16:e0257342. [PMID: 34529712 PMCID: PMC8445452 DOI: 10.1371/journal.pone.0257342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 12/16/2022] Open
Abstract
Labelling of therapeutic antibodies with radionuclides or fluorophores is routinely used to study their pharmacokinetic properties. A critical assumption in utilizing labelled therapeutic antibodies is that the label has no unfavourable effects on antibody charge, hydrophobicity, or receptor affinity. Ideally, the labelled protein should not have any significant deviations from the physiological properties of the original molecule. This article describes an established quality in vitro assessment workflow for labelled antibodies that ensures better prediction of changes in antibody pharmacokinetic (PK) properties after modifications. This analysis package considers degradation and aggregation analysis by size-exclusion chromatography, changes in neonatal-Fc-receptor (FcRn) affinity, and heparin interaction. FcRn binding is important for antibody recycling and half-life extension, whereas heparin affinity provides estimates on the rate of endocytosis through unspecific cell surface binding. Additionally, mass spectrometric analysis to determine the degree of labelling (DoL) completes the package and the combined analysis data allow to predict the label contribution to the PK properties of the modified antibody. This analytical strategy for labelling 11 IgGs has been investigated using 2 different IgG1 constructs and applying 7 different types of labels. Each labelling resulted in a change in the physicochemical properties of the protein. Not only can the DoL of modified IgGs lead to a change in protein properties, but the type of label also can. Furthermore, it was demonstrated that the labelling process can also influence the behaviour of labelled mAbs. An identical label on different constructs of IgG1 can cause different affinities for FcRn and heparin. Considering the assessment data, only 6 of the 11 modified antibodies from this study can be recommended for subsequent experiments. In conclusion, a suitability assessment of labelled antibodies prior to any pharmacokinetic studies is essential to reduce cost, allocate resources and reduce the number of animal experiments during pre-clinical drug development.
Collapse
|
11
|
Lee JY, Mushtaq S, Park JE, Shin HS, Lee SY, Jeon J. Radioanalytical Techniques to Quantitatively Assess the Biological Uptake and In Vivo Behavior of Hazardous Substances. Molecules 2020; 25:molecules25173985. [PMID: 32882977 PMCID: PMC7504758 DOI: 10.3390/molecules25173985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/25/2022] Open
Abstract
Concern about environmental exposure to hazardous substances has grown over the past several decades, because these substances have adverse effects on human health. Methods used to monitor the biological uptake of hazardous substances and their spatiotemporal behavior in vivo must be accurate and reliable. Recent advances in radiolabeling chemistry and radioanalytical methodologies have facilitated the quantitative analysis of toxic substances, and whole-body imaging can be achieved using nuclear imaging instruments. Herein, we review recent literature on the radioanalytical methods used to study the biological distribution, changes in the uptake and accumulation of hazardous substances, including industrial chemicals, nanomaterials, and microorganisms. We begin with an overview of the radioisotopes used to prepare radiotracers for in vivo experiments. We then summarize the results of molecular imaging studies involving radiolabeled toxins and their quantitative assessment. We conclude the review with perspectives on the use of radioanalytical methods for future environmental research.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Environmental and Safety Engineering, Ajou University, Suwon 16499, Korea;
| | - Sajid Mushtaq
- Department of Nuclear Engineering, Pakistan Institute of Engineering and Applied Sciences, Islamabad 45650, Pakistan;
| | - Jung Eun Park
- Department of Applied Chemistry, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea;
| | - Hee Soon Shin
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Korea; (H.S.S.); (S.-Y.L.)
- Food Biotechnology Program, University of Science and Technology, Daejeon 34113, Korea
| | - So-Young Lee
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Korea; (H.S.S.); (S.-Y.L.)
- Food Biotechnology Program, University of Science and Technology, Daejeon 34113, Korea
| | - Jongho Jeon
- Department of Applied Chemistry, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea;
- Correspondence: ; Tel.: +82-53-950-5584
| |
Collapse
|
12
|
Safety Evaluation of an Alpha-Emitter Bismuth-213 Labeled Antibody to (1→3)-β-Glucan in Healthy Dogs as a Prelude for a Trial in Companion Dogs with Invasive Fungal Infections. Molecules 2020; 25:molecules25163604. [PMID: 32784359 PMCID: PMC7465188 DOI: 10.3390/molecules25163604] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 01/30/2023] Open
Abstract
Background: With the limited options available for therapy to treat invasive fungal infections (IFI), radioimmunotherapy (RIT) can potentially offer an effective alternative treatment. Microorganism-specific monoclonal antibodies have shown promising results in the experimental treatment of fungal, bacterial, and viral infections, including our recent and encouraging results from treating mice infected with Blastomyces dermatitidis with 213Bi-labeled antibody 400-2 to (1→3)-β-glucan. In this work, we performed a safety study of 213Bi-400-2 antibody in healthy dogs as a prelude for a clinical trial in companion dogs with acquired invasive fungal infections and later on in human patients with IFI. Methods: Three female beagle dogs (≈6.1 kg body weight) were treated intravenously with 155.3, 142.5, or 133.2 MBq of 213Bi-400-2 given as three subfractions over an 8 h period. RBC, WBC, platelet, and blood serum biochemistry parameters were measured periodically for 6 months post injection. Results: No significant acute or long-term side effects were observed after RIT injections; only a few parameters were mildly and transiently outside reference change value limits, and a transient atypical morphology was observed in the circulating lymphocyte population of two dogs. Conclusions: These results demonstrate the safety of systemic 213Bi-400-2 administration in dogs and provide encouragement to pursue evaluation of RIT of IFI in companion dogs.
Collapse
|
13
|
van Dijk B, Allen KJH, Helal M, Vogely HC, Lam MGEH, de Klerk JMH, Weinans H, van der Wal BCH, Dadachova E. Radioimmunotherapy of methicillin-resistant Staphylococcus aureus in planktonic state and biofilms. PLoS One 2020; 15:e0233086. [PMID: 32407350 PMCID: PMC7224548 DOI: 10.1371/journal.pone.0233086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Background Implant associated infections such as periprosthetic joint infections are difficult to treat as the bacteria form a biofilm on the prosthetic material. This biofilm complicates surgical and antibiotic treatment. With rising antibiotic resistance, alternative treatment options are needed to treat these infections in the future. The aim of this article is to provide proof-of-principle data required for further development of radioimmunotherapy for non-invasive treatment of implant associated infections. Methods Planktonic cells and biofilms of Methicillin-resistant staphylococcus aureus are grown and treated with radioimmunotherapy. The monoclonal antibodies used, target wall teichoic acids that are cell and biofilm specific. Three different radionuclides in different doses were used. Viability and metabolic activity of the bacterial cells and biofilms were measured by CFU dilution and XTT reduction. Results Alpha-RIT with Bismuth-213 showed significant and dose dependent killing in both planktonic MRSA and biofilm. When planktonic bacteria were treated with 370 kBq of 213Bi-RIT 99% of the bacteria were killed. Complete killing of the bacteria in the biofilm was seen at 185 kBq. Beta-RIT with Lutetium-177 and Actinium-225 showed little to no significant killing. Conclusion Our results demonstrate the ability of specific antibodies loaded with an alpha-emitter Bismuth-213 to selectively kill staphylococcus aureus cells in vitro in both planktonic and biofilm state. RIT could therefore be a potentially alternative treatment modality against planktonic and biofilm-related microbial infections.
Collapse
Affiliation(s)
- B. van Dijk
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - K. J. H. Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - M. Helal
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - H. C. Vogely
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M. G. E. H. Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J. M. H. de Klerk
- Department of Nuclear Medicine, Meander Medical Center Amersfoort, Amersfoort, The Netherlands
| | - H. Weinans
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biomechanical engineering, TU Delft, Delft, The Netherlands
| | - B. C. H. van der Wal
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E. Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
- * E-mail:
| |
Collapse
|
14
|
Garg R, Mills K, Allen KJH, Causey P, Perron RW, Gendron D, Sanche S, Berman JW, Gorny MK, Dadachova E. Comparison of various radioactive payloads for a human monoclonal antibody to glycoprotein 41 for elimination of HIV-infected cells. Nucl Med Biol 2020; 82-83:80-88. [PMID: 32113033 DOI: 10.1016/j.nucmedbio.2020.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/10/2020] [Accepted: 02/16/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND cART has significantly improved the life expectancy of people living with HIV (PLWH). However, it fails to eliminate the long-lived reservoir of latent HIV-infected cells. Radioimmunotherapy (RIT) relies on antigen-specific monoclonal antibodies (mAbs) for targeted delivery of lethal doses of ionizing radiation to cells. Previously, we have demonstrated that human mAb 2556 against HIV gp41 conjugated with 213Bismuth radioisotope (t1/2 = 46 min, alpha-emitter) selectively killed HIV-infected cells. 225Actinium (t1/2 = 9.92 d, alpha-emitter) and 177Lutetium (t1/2 = 6.7 d, beta-emitter) are two long-lived clinically proven radioisotopes for cancer treatment which might be more effective in killing infected cells systemically and in CNS. METHODS In this study we have conjugated 2556 mAb with 213Bi, 225Ac and 177Lu, and compared their ability to kill HIV-infected human peripheral blood mononuclear cells (PBMCs) and monocytes. PBMCs and monocytes from healthy donors were infected with HIVp49.5 and treated in vitro with increasing concentrations of 213Bi (4-20 μCi)-, 225Ac (20-100 nCi)- and 177Lu (4-50 μCi)-2556 mAb. RESULTS After three days post-treatment of infected PBMCs and monocytes, 213Bi- and 177Lu-conjugated 2556 mAb reduced virus production measured by p24 level in a dose-dependent manner, whereas, 225Ac-2556 showed minimal effect. However, seven days post-treatment all three radioisotopes showed significantly more pronounced reduction of virus replication as compared to control labeled mAb with 225Ac-2556 showing the least non-specific killing. CONCLUSION These results indicate that RIT holds promise as a novel treatment option for the eradication of HIV-infected cells that merits further study in combination with cART and reactivation drugs.
Collapse
Affiliation(s)
- Ravendra Garg
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kienna Mills
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kevin J H Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | | | | | | | - Stephen Sanche
- College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Miroslaw K Gorny
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
15
|
Helal M, Allen KJH, van Dijk B, Nosanchuk JD, Snead E, Dadachova E. Radioimmunotherapy of Blastomycosis in a Mouse Model With a (1→3)-β-Glucans Targeting Antibody. Front Microbiol 2020; 11:147. [PMID: 32117166 PMCID: PMC7019017 DOI: 10.3389/fmicb.2020.00147] [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: 07/30/2019] [Accepted: 01/21/2020] [Indexed: 12/16/2022] Open
Abstract
Invasive fungal infections (IFI) cause devastating morbidity and mortality, with the number of IFIs more than tripling since 1979. Our laboratories were the first to demonstrate that radiolabeled microorganism-specific monoclonal antibodies are highly effective for treatment of experimental fungal, bacterial and viral infections. Later we proposed to utilize surface expressed pan-antigens shared by major IFI-causing pathogens such as beta-glucans as RIT targets. Here we evaluated in vivo RIT targeting beta-glucan in Blastomyces dermatitidis which causes serious infections in immunocompromised and immunocompetent individuals and in companion dogs. B. dermatitidis cells were treated with the 400-2 antibody to (1→3)-β-glucans radiolabeled with the beta-emitter 177Lutetium (177Lu) and alpha-emitter 213Bismuth (213Bi) and the efficacy of cell kill was determined by colony forming units (CFUs). To determine the antigen-specific localization of the 400-2 antibody in vivo, C57BL6 mice were infected intratracheally with 2 × 105B. dermatitidis cells and given 111In-400-2 antibody 24 h later. To evaluate the killing of B. dermatitidis cells with RIT, intratracheally infected mice were treated with 150 μCi 213Bi-400-2 and their lungs analyzed for CFUs 96 h post-infection. 213Bi-400-2 proved to be more effective in killing B. dermatitidis cells in vitro than 177Lu-400-2. Three times more 111In-400-2 accumulated in the lungs of infected mice, than in the non-infected ones. 213Bi-400-2 lowered the fungal burden in the lungs of infected mice more than 2 logs in comparison with non-treated infected controls. In conclusion, our results demonstrate the ability of an anti-(1-3)-beta-D-glucan antibody armed with an alpha-emitter 213Bi to selectively kill B. dermatitidis cells in vitro and in vivo. These first in vivo results of the effectiveness of RIT targeting pan-antigens on fungal pathogens warrant further investigation.
Collapse
Affiliation(s)
- Muath Helal
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kevin J H Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Bruce van Dijk
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joshua D Nosanchuk
- Department of Medicine, Albert Einstein College of Medicine, The Bronx, NY, United States
| | - Elisabeth Snead
- Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
16
|
Evaluating the Combination of Radioimmunotherapy and Immunotherapy in a Melanoma Mouse Model. Int J Mol Sci 2020; 21:ijms21030773. [PMID: 31991626 PMCID: PMC7037880 DOI: 10.3390/ijms21030773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 01/02/2023] Open
Abstract
Immunotherapy has changed the oncology landscape during the last decade and become standard of care for several cancers. The combinations of immunotherapy with other treatment modalities are also being investigated. One of the challenges to investigate such combinations is to identify suitable mouse models for the pre-clinical experiments. In the past, we and other researchers showed that murine B16-F10 melanoma in C57Bl6 mice is refractory to treatment with immune checkpoint inhibitors. In this work we studied the suitability of an alternative syngeneic model, Cloudman S91 murine melanoma in DBA/2 mouse (DBA/2NCrl), to study the combination of immunotherapy targeting PD-1 and radioimmunotherapy targeting melanin. DBA/2 male and female mice were injected subcutaneously with 3-6 million Cloudman S91 cells. When the tumors reached ~150 mm3 volume, the animals were treated intraperitoneally with PBS (sham), h8C3 unlabeled (cold) antibody to melanin, immunotherapy with anti-PD-1 antibody, radioimmunotherapy with 213Bismuth (213Bi)-labeled h8C3 antibody, or several combinations of immunotherapy and radioimmunotherapy. Treatments with immunotherapy alone produced very modest effect on the tumor size, while combination therapy resulted in significant slowing down of the tumor growth, increased animal survival, and no decrease in animal body weight. We conclude that Cloudman S91 murine melanoma in DBA/2 mouse is a suitable model to evaluate combination of immunotherapy of melanoma with tangentially targeted treatments.
Collapse
|
17
|
Drug Delivery Technology Development in Canada. Pharmaceutics 2019; 11:pharmaceutics11100541. [PMID: 31627471 PMCID: PMC6835823 DOI: 10.3390/pharmaceutics11100541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 11/29/2022] Open
Abstract
Canada has a long and rich history of ground-breaking research in drug delivery within academic institutions, pharmaceutical industry and the biotechnology community. Drug delivery refers to approaches, formulations, technologies, and systems for transporting a pharmaceutical compound in the body as needed to safely achieve its desired therapeutic effect. It may involve rational site-targeting, or facilitating systemic pharmacokinetics; in any case, it is typically concerned with both quantity and duration of the presence of the drug in the body. Drug delivery is often approached through a drug’s chemical formulation, medical devices or drug-device combination products. Drug delivery is a concept heavily integrated with dosage form development and selection of route of administration; the latter sometimes even being considered part of the definition. Drug delivery technologies modify drug release profile, absorption, distribution and elimination for the benefit of improving product efficacy and safety, as well as patient convenience and adherence. Over the past 30 years, numerous Canadian-based biotechnology companies have been formed stemming from the inventions conceived and developed within academic institutions. Many have led to the development of important drug delivery products that have enhanced the landscape of drug therapy in the treatment of cancer to infectious diseases. This Special Issue serves to highlight the progress of drug delivery within Canada. We invited articles on all aspects of drug delivery sciences from pre-clinical formulation development to human clinical trials that bring to light the world-class research currently undertaken in Canada for this Special Issue.
Collapse
|