1
|
Sepp A, Stader F, Derbalah A, Liu C, Zyla A, Gardner I, Jamei M. The physiological limits of bispecific monoclonal antibody tissue targeting specificity. MAbs 2025; 17:2492236. [PMID: 40223272 PMCID: PMC12005452 DOI: 10.1080/19420862.2025.2492236] [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: 01/22/2025] [Revised: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025] Open
Abstract
Bispecific monoclonal antibodies (bsmAbs) are expected to provide targeted drug delivery that overcomes the dose-limiting toxicities often accompanying antibody-drug conjugates (ADC) in clinical practice. Much attention has been paid in the past to target selection, mAb affinities and the payload linker design, but challenges remain. Here, we demonstrate, by physiologically based pharmacokinetic (PBPK) in silico modeling and simulation, that the tissue-targeting accuracy of mono- and bispecific antibody therapeutics is substantially limited by normal physiological characteristics like organ volumes, blood flow rates, lymphatic circulation, and rates of extravasation. Only a small fraction of blood flows through solid tumor, where the diffusion-driven extravasation is relatively slow compared with many other organs. EGFR and HER2 are used as model antigens based on their experimentally measured tissue and tumor expression levels, but the approach is generic and can account for the cellular expression variation of targets. The model confirms experimental observations that only about 0.1-1% of the dosed mAb is likely to reach the tumor, while the rest ends up in healthy tissues due to target-mediated internalization and nonspecific uptake. The model suggests that the dual-positive tumor cell targeting specificity with bispecific antibodies is likely to be higher at lower drug concentrations and doses. However, this can be offset by elevated drug exposure in more accessible healthy tissues, primarily endothelium. The balance of exposure can be shifted toward tumor cells by using higher doses, albeit at the expense of more extensive target engagement elsewhere in the body, suggesting the need to adapt the toxicity of the payload if ADCs are considered. We suggest that PBPK modeling can guide and support biologics and bsmAb development, from target evaluation and drug optimization to therapeutic dose selection.
Collapse
Affiliation(s)
- Armin Sepp
- Certara Predictive Technologies division, Certara UK Ltd, Sheffield, UK
| | - Felix Stader
- Certara Predictive Technologies division, Certara UK Ltd, Sheffield, UK
| | - Abdallah Derbalah
- Certara Predictive Technologies division, Certara UK Ltd, Sheffield, UK
| | - Cong Liu
- Certara Predictive Technologies division, Certara UK Ltd, Sheffield, UK
| | - Adriana Zyla
- Certara Predictive Technologies division, Certara UK Ltd, Sheffield, UK
| | - Iain Gardner
- Certara Predictive Technologies division, Certara UK Ltd, Sheffield, UK
| | - Masoud Jamei
- Certara Predictive Technologies division, Certara UK Ltd, Sheffield, UK
| |
Collapse
|
2
|
Liu Y, Zong Q, Tu Y, Zhang X, Tan Q, Ullah I, Yuan Y. A tumor heterogeneity-independent antigen-responsive nanocarrier enabled by bioorthogonal pre-targeting and click-activated self-immolative polymer. Biomaterials 2025; 319:123200. [PMID: 39987854 DOI: 10.1016/j.biomaterials.2025.123200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Bioorthogonal pre-targeting alleviate the limitations of traditional nanomedicines in passive and active targeting delivery. However, the high selectivity of bioorthogonal pre-targeting depends on the high expression level of antigens in lesion sites, and there are very limited targets with sufficient overexpression. Herein, we propose a tumor heterogeneity-independent antigen-responsive nanocarrier utilizing bioorthogonal pre-targeting and click-activated self-immolative polymers for stimulus signal conversion and amplification. This approach comprises a tetrazine (Tz) conjugated with trastuzumab (T-Tz), and a bioorthogonally activatable nanocarrier CONP which self-assembled by isocyanide and polyethylene glycol-modified poly (thiocarbamate) (NC-PTC-PEG) and hydrogen sulfide (H2S)-responsive self-immolative polymers. In practice, T-Tz is first injected to actively pretarget HER2-positive tumor cells and followed by the second injection of nanocarrier CONP. The NC-PTC-PEG in CONP undergoes a click reaction with Tz to generate H2S, thereby achieving the transformation from antigen signal to H2S signal. Finally, NO2-PTC-PEG responds to H2S stimulation and undergoes a head-to-tail depolymerization process similar to dominoes to produce a large amount of H2S, further amplifying the stimulus signal. This bioorthogonal pre-targeting combine with click-activated self-immolative polymers is anticipated to enhance the effectiveness of existing pre-targeting strategies for tumor imaging and therapy, with the potential to overcome challenges posed by tumor heterogeneity.
Collapse
Affiliation(s)
- Ye Liu
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Qingyu Zong
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Yalan Tu
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, Sichuan, 610061, PR China.
| | - Xingzu Zhang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 511442, PR China
| | - Qiaoling Tan
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Ihsan Ullah
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Youyong Yuan
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
3
|
Verdonk JDJ, Piet B, Ter Heine R, van den Heuvel MM, Smeets RL, Koenen HJPM. Ex vivo pembrolizumab pharmacology for personalized PD-1 inhibitor therapy reveals a critical gap between receptor occupancy and T cell functionality. Int Immunopharmacol 2025; 157:114754. [PMID: 40318274 DOI: 10.1016/j.intimp.2025.114754] [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: 03/04/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
PURPOSE Targeting the programmed death (ligand)-1 (PD-1/PD-L1) axis with immune checkpoint inhibitors (ICIs), like pembrolizumab, has improved cancer survival. Unfortunately, the optimal dose remains unknown and less than 50 % of patients respond. Understanding PD-1 receptor pharmacology and developing early-response biomarkers are crucial to personalize therapy. We hypothesize that characterizing individual pre-treatment variability in immune responses to pembrolizumab will enhance PD-1 receptor pharmacology insights and improve treatment response prediction. Hence, this study evaluates the performance of a newly developed ex vivo immunopharmacological bioassay under healthy and pathological states. METHODS Peripheral blood mononuclear cells from healthy individuals and non-small cell lung cancer (NSCLC) patients were stimulated in vitro in the presence of pembrolizumab. PD-1 expression, interleukin-2 (IL-2) secretion, T cell differentiation, expression of activation markers and phosphorylation of T cell signalling molecules were analysed. RESULTS In healthy individuals, receptor saturation occurred at >0.025 μg/mL pembrolizumab. Yet IL-2 production still increased significantly beyond this concentration. NSCLC patients showed significantly higher PD-1 expression and IL-2 production than healthy individuals. Nevertheless, pembrolizumab induced IL-2 production similarly in both cohorts. In NSCLC patients, pembrolizumab inhibited differentiation towards CD4 central memory T cells. Remarkably, phosphorylation of proximal phospho-markers in response to pembrolizumab varied between NSCLC patients, potentially discriminating responders from non-responders. CONCLUSIONS We highlight the importance of evaluating T cell functionality alongside PD-1 receptor occupancy. We identified PD-1-induced modulation of phosphorylation of proximal signalling molecules as potential predictors for ICI treatment response in NSCLC. We recommend further development of this immunopharmacological bioassay for personalization of ICI treatment.
Collapse
Affiliation(s)
- Judith D J Verdonk
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Berber Piet
- Department of Pulmonary Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
| | | | - Ruben L Smeets
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
4
|
Weinstein LA, Wei B. Hiding in Plain Sight: Cell Biomimicry for Improving Hematological Cancer Outcomes. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:739. [PMID: 40423130 DOI: 10.3390/nano15100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025]
Abstract
The field of nanomedicine has been fruitful in creating novel drug delivery ideas to battle hematologic cancers. However, one persistent barrier to efficient nanoparticle treatment is phagocytic uptake or the clearance of nanoparticles by immune cells. To prevent this immune uptake, scientists have utilized biomimicry, the emulation of natural structures for engineered applications, to create particles that are able to remain unrecognized by immune cells. This method aims to improve the overall circulation time of nanoparticles by decreasing the amount of particles filtered out of the blood. It can even lead to homotypic cancer cell targeting, decreasing cancer cell vitality. This review summarizes recent in vivo and in vitro studies to prove that biomimetic cargo delivery is a unique and tenable way of increasing survival outcomes in patients with hematologic cancers.
Collapse
Affiliation(s)
- Laura A Weinstein
- Department of Biomedical Engineering, University of Delaware, Newark 19716, DE, USA
| | - Bingqing Wei
- Department of Mechanical Engineering, University of Delaware, Newark 19716, DE, USA
| |
Collapse
|
5
|
Chen P, Bordeau BM, Zhang W, Balthasar JP. Investigations of Influence of Antibody Binding Kinetics on Tumor Distribution and Anti-Tumor Efficacy. AAPS J 2025; 27:91. [PMID: 40341444 DOI: 10.1208/s12248-025-01076-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/19/2025] [Indexed: 05/10/2025] Open
Abstract
The pharmacokinetics of antibodies with varied binding kinetics were simulated to assess the role of affinity and binding microconstants (kon, koff) on tumor exposure and intra-tumoral distribution. Anti-HER2 constructs (trastuzumab, pertuzumab, VK3VH6, and conjugates with DM1 and gelonin) were produced, purified, and tested for binding and cytotoxicity in vitro, and for intra-tumoral distribution and anti-tumor efficacy in mice. Simulations demonstrated that homogeneity in intra-tumoral distribution increases with increases in koff and with decreases in kon. Interestingly, simulations also predicted that homogeneity in tumor distribution may be improved by decreasing kon and koff in parallel (without changing affinity). Relative to trastuzumab, pertuzumab exhibits similar affinity but a ~ fivefold smaller kon and koff, while VK3VH6 exhibits a similar koff but a ~ 30-fold lower kon and affinity. Conjugate concentrations associated with 50% inhibition of cell proliferation (IC50s) were found to vary with affinity, where IC50 values were similar for pertuzumab and trastuzumab, and higher for VK3VH6. Consistent with model simulations, VK3VH6 and pertuzumab demonstrated more homogeneous tumor distribution than trastuzumab. Although treatment differences were not statistically significant, pertuzumab and VK3VH6 conjugates showed trends for increased survival time relative to mice treated with trastuzumab conjugates. Our simulation and experimental results demonstrate complex relationships between antibody-antigen binding kinetics, intratumoral distribution, and efficacy. The rate constant of association, kon, is an underappreciated determinant of intra-tumoral distribution; among high-affinity antibodies, those with lower values of kon may be expected to exhibit improved intra-tumoral distribution and, potentially, efficacy.
Collapse
MESH Headings
- Animals
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/metabolism
- Mice
- Humans
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/administration & dosage
- Trastuzumab/pharmacokinetics
- Cell Line, Tumor
- Kinetics
- Female
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/pharmacology
- Xenograft Model Antitumor Assays
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/pharmacology
Collapse
Affiliation(s)
- Ping Chen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 452 Pharmacy Building, Buffalo, New York, 14214, USA
| | - Brandon M Bordeau
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 452 Pharmacy Building, Buffalo, New York, 14214, USA
| | - Wenqiu Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 452 Pharmacy Building, Buffalo, New York, 14214, USA
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, 452 Pharmacy Building, Buffalo, New York, 14214, USA.
| |
Collapse
|
6
|
Ngô TM, Vágner A, Nagy G, Ország G, Nagy T, Szoboszlai Z, Csikos C, Váradi B, Trencsényi G, Tircsó G, Garai I. HER2 expression in different cell lines at different inoculation sites assessed by [ 52Mn]Mn-DOTAGA(anhydride)-trastuzumab. Pathol Oncol Res 2025; 31:1611999. [PMID: 40365451 PMCID: PMC12069034 DOI: 10.3389/pore.2025.1611999] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025]
Abstract
Purpose Positron emission tomography (PET) hybrid imaging targeting HER2 requires antibodies labelled with longer half-life isotopes. With a suitable radiation profile, 52Mn coupled with DOTAGA as a bifunctional chelator is a potential candidate. In this study, we investigated the tumor HER2 specificity and the temporal biodistribution of the [52Mn]Mn-DOTAGA(anhydride)-trastuzumab in preclinical models. Methods PET/MRI and PET/CT were performed on SCID mice bearing orthotopic and ectopic HER2-positive and ectopic HER2-negative tumors at 4, 24, 48, 72, and 120 h post-injection with [52Mn]Mn-DOTAGA(anhydride)-trastuzumab. Melanoma xenografts were included for comparison of specificity. Results In vivo biodistribution demonstrated strong contrast in HER2-positive tumors, particularly in orthotopic tumors, where uptake was significantly higher than in the blood pool and other organs from 24 h onwards and consistently higher than in ectopic HER2-positive tumors at all time points. Significantly higher tumor-to-blood and tumor-to-muscle ratios were observed in HER2-positive ectopic tumors compared to HER2-negative tumors but only at 4 and 24 h; the differences were likely due to non-specific binding of the tracer. The ratios for orthotopic HER2-positive tumors were significantly higher than those for ectopic HER2-negative tumors and melanoma at all time points. However, the differences between HER2-positive and HER2-negative tumors decreased at later time points. Conclusion These results suggest that [52Mn]Mn-DOTAGA(anhydride)-trastuzumab demonstrates efficient tumor-to-background contrast, emphasize the higher tumor uptake observed in orthotopic tumors, and highlight the influence of tumor environment characteristics on uptake.
Collapse
Affiliation(s)
- Toàn Minh Ngô
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | | | | | | | - Tamás Nagy
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
- Scanomed Ltd., Debrecen, Hungary
| | | | - Csaba Csikos
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | - Balázs Váradi
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - György Trencsényi
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
- Scanomed Ltd., Debrecen, Hungary
| | - Gyula Tircsó
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Ildikó Garai
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
- Scanomed Ltd., Debrecen, Hungary
| |
Collapse
|
7
|
Curry SD, Bower BM, Saemundsson SA, Goodwin AP, Cha JN. Binding affinity and transport studies of engineered photocrosslinkable affibody-enzyme-nanoparticle constructs. NANOSCALE ADVANCES 2025; 7:2239-2247. [PMID: 40028492 PMCID: PMC11866575 DOI: 10.1039/d4na00823e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Nanoparticle accumulation at tumor sites has been well reported in vivo, where targeting typically shows increased retention, but challenges remain for clinical translation. This work examines the effect of targeting ligand binding affinities and nanoparticle size on retention and transport through a solid tumor. We first show using cell affinity assays that modifying a wildtype (WT) anti-epidermal growth factor receptor (EGFR) affibody-enzyme fusion protein into a UV-photocrosslinkable (N23BP) version led to a significant decrease in affinity, whether as a free protein or as a conjugate to silica nanoparticles. Despite the reduced EGFR affinity, all protein conjugated nanoparticles showed binding and uptake to EGFR-overexpressing HTB9 bladder cancer cells as detected by confocal microscopy and flow cytometry. Next, transport studies of the protein conjugated nanoparticles using monoculture spheroids revealed that spheroid binding was higher for 17 nm particles bound with the WT proteins than N23BP, which was expected based on their respective K D values. However, the 17 nm particles conjugated with the photocrosslinkable N23BP affibody-enzymes showed an altered distribution profile that peaked further into the spheroid than the WT nanoparticle conjugates or in the absence of UV treatment. We correlate this finding with increased transport and retention of the photocrosslinked N23BP-nanoparticle conjugates in 3D spheroids to both the lower binding affinity of the affibodies for EGFR and the ability to introduce covalent linkages between the affibody and cell receptor. The larger 40 nm protein-conjugated nanoparticles showed limited penetration regardless of affinity or photocrosslinking on a 12 h timescale but did show overall increased transport after 24 h.
Collapse
Affiliation(s)
- Shane D Curry
- Department of Chemical and Biological Engineering, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
| | - Bryce M Bower
- Department of Chemical and Biological Engineering, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
| | - Sven A Saemundsson
- Department of Chemical and Biological Engineering, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
| | - Andrew P Goodwin
- Department of Chemical and Biological Engineering, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
- Materials Science and Engineering Program, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
| | - Jennifer N Cha
- Department of Chemical and Biological Engineering, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
- Materials Science and Engineering Program, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
- Biomedical Engineering Program, University of Colorado, Boulder 596 UCB Boulder CO 80303 USA
| |
Collapse
|
8
|
Li N, Yang L, Zhao Z, Du T, Liang G, Li N, Tang J. Antibody-drug conjugates in breast cancer: current evidence and future directions. Exp Hematol Oncol 2025; 14:41. [PMID: 40114224 PMCID: PMC11924693 DOI: 10.1186/s40164-025-00632-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are a rapidly evolving class of antitumor drugs and have already revolutionized the treatment strategy of many hematologic and solid cancers. So far, trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), sacituzumab govitecan (SG) and datopotamab deruxtecan (Dato-DXd) are the four ADCs that have been approved by US food and drug administration (FDA) in treatment of breast cancer, and SKB264 has been approved by Chinese national medical products administration (NMPA). Many ADCs for treatment of breast cancer are currently being tested in late-phase clinical trials, with several encouraging results achieved recently. However, major issues arise during the use of ADCs, including emergence of acquired resistance, occurrence of treated-related toxicities, and identification of biomarkers of response and resistance. ADCs are being increasingly tested in combination with other agents, and novel next-generation ADC development is progressing rapidly. A better understanding of the design and development of ADCs will promote ADC development for cancer treatment. In this review, we aim to provide a broad overview of the design and the recent advances of ADCs in breast cancer. We also propose several notable future directions of ADCs in treatment of breast cancer.
Collapse
Affiliation(s)
- Ning Li
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lu Yang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
- Shantou University Medical College, Shantou University, Shantou, 515000, China
| | - Zixuan Zhao
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Tian Du
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Gehao Liang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Na Li
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Jun Tang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
9
|
Sun L, Sun Y, Zuo K, Fan L, Wang X, Zhang J, Hu S, Liu X, Li J, Li Y, Shao Z, Xu X, Wu A, Song S. Pilot Study of Nectin-4-Targeted PET Imaging Agent 68Ga-FZ-NR-1 in Triple-Negative Breast Cancer from Bench to First-in-Human. J Nucl Med 2025; 66:473-479. [PMID: 39947908 DOI: 10.2967/jnumed.124.269024] [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: 10/21/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
Nectin cell adhesion molecule 4 (Nectin-4) is an emerging biomarker for cancer diagnosis and therapy. We developed a Nectin-4-targeted 68Ga-DOTA-Sar10-Nectin-4 (68Ga-FZ-NR-1) PET/CT radiotracer for detecting Nectin-4 expression in a tumor model and in triple-negative breast cancer (TNBC) patients. Methods: A series of Nectin-4-targeted radiotracers-68Ga-FZ-NR-1, 68Ga-DOTA-polyethylene glycol 5-Nectin-4 (68Ga-FZ-NR-2), and 68Ga-DOTA-polyethylene glycol 10-Nectin-4 (68Ga-FZ-NR-3)-were synthesized, and their targeting ability and specificity were evaluated in vitro and in vivo. In vitro experiments were performed in the MDA-MB-468 (Nectin-4-positive) and MDA-MB-231 (Nectin-4-negative) cell lines. PET/CT imaging in tumor models was performed to assess the Nectin-4-targeting ability of the radiotracers. After preclinical experiments and screening, the 68Ga-FZ-NR-1 radiotracer was selected for safety and efficacy evaluation in a first-in-human trial in TNBC patients. Positive lesions were biopsied and analyzed by immunohistochemistry to determine Nectin-4 expression levels. Results: The 3 68Ga-labeled radiotracers exhibited high radiochemical purity, stability, and strong affinity for Nectin-4. In vitro cell uptake studies showed that the radiotracers effectively targeted Nectin-4 in MDA-MB-468 cells, and 68Ga-FZ-NR-1 showed the highest targeting efficacy. In the MDA-MB-468 tumor model, PET/CT imaging showed that 68Ga-FZ-NR-1 was taken up at higher rates than 68Ga-FZ-NR-2 and 68Ga-FZ-NR-3, and it exhibited favorable pharmacokinetics and safety profiles. 68Ga-FZ-NR-1 was thus selected for subsequent clinical trials. 68Ga-FZ-NR-1 PET/CT effectively identified tumors in 9 patients with TNBC, which was confirmed by 18F-FDG PET/CT. Biopsy samples of the tumor lesions revealed that the positive lesions identified by 68Ga-FZ-NR-1 PET/CT corresponded to areas of high Nectin-4 expression. Conclusion: A series of Nectin-4-targeted radiotracers (68Ga-FZ-NR-1, 68Ga-FZ-NR-2, and 68Ga-FZ-NR-3) was developed and evaluated. Preclinical studies demonstrated that 68Ga-FZ-NR-1 can identify tumors with high Nectin-4 expression. In a preliminary clinical study, 68Ga-FZ-NR-1 was used to effectively identify and visualize Nectin-4-expressing tumor lesions in patients with TNBC, which was confirmed by immunohistochemistry. This radiotracer provides a noninvasive approach to the assessment of Nectin-4 and a potential basis for the development of Nectin-4-targeted treatments for TNBC.
Collapse
Affiliation(s)
- Li Sun
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, CAS Key Laboratory of Magnetic Materials and Devices, and Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Yuyun Sun
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| | - Ke Zuo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Fan
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| | - Silong Hu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| | - Xiaosheng Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| | - Jindian Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| | - Ye Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| | - Zhiming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China;
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, CAS Key Laboratory of Magnetic Materials and Devices, and Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China;
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China; and
| |
Collapse
|
10
|
Liu Y, Wang T, Chi X, Yu S, He W, He H, Wang G, Hao K, Zhang J. Modeling based dynamics mechanism and pathway of liposome penetration in multicellular tumor spheroid for liposome optimization. Int J Pharm 2025; 671:125237. [PMID: 39842737 DOI: 10.1016/j.ijpharm.2025.125237] [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: 10/29/2024] [Revised: 12/26/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
Liposomes are widely recognized as effective drug delivery systems, characterized by biodegradability, biocompatibility, and ability to minimize toxicity. However, liposome-based nanotechnology has not demonstrated superior anti-tumor efficacy due to their limited intratumor penetration. Strategies to improve the tumor delivery efficiency of nanomedicine remain to be developed. Moreover, the specific steps involving inter-/intra-cellular pathways in delivery could not be fully revealed by real experiment. Mathematical modeling is a great choice. Hence, this study analyzed the roles of physicochemical properties of liposomes and tumorassociated environment in intratumoral penetration, using ten anti-tumor liposomes datasets to develop two kinetic models in tumor spheroid cells based on transcytosis and paracellular transport mechanisms. Modeling results reveal the dominated penetration pathway of liposomes studied through the paracellular pathway compared to transcytosis. Liposomes with positive surface charge and high membrane fluidity enhance the maximal binding capacity on the cell membrane. Smaller liposome sizes promote internalization on the cell membrane, leading to increased drug accumulation within the cell. The pattern of liposome penetration remained consistent across different tumor-associated environments. Our developed kinetic models accurately described the penetration process of liposomes in multicellular tumor spheroid, offering valuable insights for the development of new nano antitumor medications with similar characteristics from a pharmacokinetic perspective at the tissue level.
Collapse
Affiliation(s)
- Yinuo Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xingyu Chi
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Simiao Yu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Kun Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Jingwei Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
11
|
Chowdhury A, Shrestha P, Jois SD. Molecular Chimera in Cancer Drug Discovery: Beyond Antibody Therapy, Designing Grafted Stable Peptides Targeting Cancer. Int J Pept Res Ther 2025; 31:38. [PMID: 39974747 PMCID: PMC11832722 DOI: 10.1007/s10989-025-10690-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2025] [Indexed: 02/21/2025]
Abstract
Background Several cancer therapies are being developed, and given the variability of different cancer types, the goal of these therapies is to remove the invasive tumor from the body, kill the cancer cells, or else retard the growth. These include chemotherapeutic agents and targeted therapy using small molecules and antibodies. However, antibodies can generate an immune response upon repeated administration, and producing antibodies could be expensive. Purpose The purpose of this review is to describe different therapeutic approaches utilized for cancer therapy, the current therapeutic approaches, and their limitations. As a novel strategy to combat cancer, designing new stable peptide scaffolds such as cyclotides and sunflower trypsin inhibitors (SFTI) is described. Results Stable peptides that can target proteins can be used as therapeutic agents. Here, we review the utilization and amalgamation of plant-based peptides with biological epitopes in designing molecules called "Molecular Chimeras" using a grafted peptide strategy. These cyclic peptides can bind to target receptors or modulate protein-protein interactions as they bind with high affinity and selectivity. Grafted peptides also possess better serum stability owing to the head-to-tail cyclization and other structural modifications. Conclusion Stable cyclic peptides outweigh the other biologicals in terms of stability and manufacturing process. Peptides and peptidomimetics can be used as therapeutic agents, and these molecules provide alternatives for biologicals and small molecule inhibitors as drugs.
Collapse
Affiliation(s)
- Arpan Chowdhury
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, Skip Bertman Drive, Baton Rouge, LA-70803 USA
| | - Prajesh Shrestha
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, Skip Bertman Drive, Baton Rouge, LA-70803 USA
| | - Seetharama D. Jois
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, Skip Bertman Drive, Baton Rouge, LA-70803 USA
| |
Collapse
|
12
|
Li K, Guo B, Gu J, Ta N, Gu J, Yu H, Sun M, Han T. Emerging advances in drug delivery systems (DDSs) for optimizing cancer complications. Mater Today Bio 2025; 30:101375. [PMID: 39759851 PMCID: PMC11699619 DOI: 10.1016/j.mtbio.2024.101375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
The management and treatment of tumor complications pose continuous challenges due to the inherent complexity. However, the advent of drug delivery systems (DDSs) brings promising opportunities to address the tumor complications using innovative technological approaches. This review focuses on common oncological complications, including cancer thrombosis, malignant serous effusion, tumor-associated infections, cancer pain, and treatment-related complications. Emphasis was placed on the application and potential of DDSs in mitigating and treating these tumor complications, and we delved into the underlying mechanisms of common cancer-associated complications, discussed the limitations of conventional treatments, and outlined the current status and potential development of DDSs for various complications in this review. Moreover, we have discussed the existing challenges in DDSs research, underscoring the need for addressing issues related to biocompatibility and targeting of DDSs, optimizing drug delivery routes, and enhancing delivery efficiency and precision. In conclusion, DDSs offer promising avenues for treating cancer complications, offering the potential for the development of more effective and safer drug delivery strategies, thereby improving the quality of life and survival rates of cancer patients.
Collapse
Affiliation(s)
- Kerui Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Bei Guo
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Junmou Gu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Na Ta
- Department of Neurology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
| | - Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Hao Yu
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
13
|
Cardle II, Raman J, Nguyen DC, Wang T, Wu AY, Sellers DL, Pichon TJ, Cheng EL, Kacherovsky N, Salipante SJ, Jensen MC, Pun SH. DNA Aptamer-Polymer Conjugates for Selective Targeting of Integrin α4β1 + T-Lineage Cancers. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4543-4561. [PMID: 39788927 PMCID: PMC11995848 DOI: 10.1021/acsami.4c17788] [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: 01/12/2025]
Abstract
Selective therapeutic targeting of T-cell malignancies is difficult due to the shared lineage between healthy and malignant T cells. Current front-line chemotherapy for these cancers is largely nonspecific, resulting in frequent cases of relapsed/refractory disease. The development of targeting approaches for effectively treating T-cell leukemia and lymphoma thus remains a critical goal for the oncology field. Here, we report the discovery of a DNA aptamer, named HR7A1, that displays low nanomolar affinity for the integrin α4β1 (VLA-4), a marker associated with chemoresistance and relapse in leukemia patients. After truncation of HR7A1 to a minimal binding motif, we demonstrate elevated binding of the aptamer to T-lineage cancer cells over healthy immune cells. Using cryo-EM and competition studies, we find that HR7A1 shares an overlapping binding site on α4β1 with fibronectin and VCAM-1, which has implications for sensitizing blood cancers to chemotherapy. We last characterize barriers to in vivo aptamer translation, including serum stability, temperature-sensitive binding, and short circulation half-life, and synthesize an aptamer-polymer conjugate that addresses these challenges. Future work will seek to validate in vivo targeting of α4β1+ tumors with the conjugate, establishing an aptamer-based biomaterial that can be readily adapted for targeted treatment of T-cell malignancies.
Collapse
Affiliation(s)
- Ian I. Cardle
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Seattle Children’s Therapeutics, Seattle, WA 98101, USA
| | - Jai Raman
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Dinh Chuong Nguyen
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Tong Wang
- Nanoscience Initiative, CUNY Advanced Science Research Center, City University of New York, NY 10031, USA
| | - Abe Y. Wu
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Trey J. Pichon
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Emmeline L. Cheng
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Nataly Kacherovsky
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Michael C. Jensen
- Seattle Children’s Therapeutics, Seattle, WA 98101, USA
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
14
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 PMCID: PMC11817814 DOI: 10.3390/cells14030170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A. Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia (V.G.T.)
| | | | | | | | | |
Collapse
|
15
|
Knight K, Park JB, Oot RA, Khan MM, Roh SH, Wilkens S. Monoclonal nanobodies alter the activity and assembly of the yeast vacuolar H +-ATPase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632502. [PMID: 39829782 PMCID: PMC11741422 DOI: 10.1101/2025.01.10.632502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The vacuolar ATPase (V-ATPase; V1Vo) is a multi-subunit rotary nanomotor proton pump that acidifies organelles in virtually all eukaryotic cells, and extracellular spaces in some specialized tissues of higher organisms. Evidence suggests that metastatic breast cancers mislocalize V-ATPase to the plasma membrane to promote cell survival and facilitate metastasis, making the V-ATPase a potential drug target. We have generated a library of camelid single-domain antibodies (Nanobodies; Nbs) against lipid-nanodisc reconstituted yeast V-ATPase Vo proton channel subcomplex. Here, we present an in-depth characterization of three anti-Vo Nbs using biochemical and biophysical in vitro experiments. We find that the Nbs bind Vo with high affinity, with one Nb inhibiting holoenzyme activity and another one preventing enzyme assembly. Using cryoEM, we find that two of the Nbs bind the c subunit ring of the Vo on the lumen side of the complex. Additionally, we show that one of the Nbs raised against yeast Vo can pull down human V-ATPase (HsV1Vo). Our research demonstrates Nb versatility to target and modulate the activity of the V-ATPase, and highlights the potential for future therapeutic Nb development.
Collapse
Affiliation(s)
- Kassidy Knight
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Jun Bae Park
- Department of Biological Sciences, Seoul National University, Seoul, Korea
- Present address: Department of Cancer Biology, Lerner research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rebecca A. Oot
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Md. Murad Khan
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Present address: Howard Hughes Medical Institute, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Soung-Hun Roh
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| | - Stephan Wilkens
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
16
|
Prodi E, Corbellari R, Ghezzi L, Ciambellotti S, Catastini JE, Rappuoli M, Rotta G, Sakic I, Weiss T, Weller M, Pellegrino C, Manz MG, Neri D, De Luca R. Tripokin: A multi-specific immunocytokine for cancer immunotherapy. Int J Cancer 2025; 156:216-228. [PMID: 39177452 DOI: 10.1002/ijc.35145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/10/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024]
Abstract
Antibodies that target the tumor microenvironment can be used to deliver pro-inflammatory payloads, such as cytokines. Cytokines are small proteins able to modulate the activity of the immune system, and antibody-cytokine fusion proteins have been tested in preclinical and clinical settings. In this study, we describe Tripokin, a novel multi-specific fusion protein that combines interleukin-2 and a single amino acid mutant of tumor necrosis factor. The two pro-inflammatory payloads were fused to the L19 antibody, a clinical-grade antibody against the extradomain B of fibronectin. The human payloads were used for clinical applications, while the corresponding murine cytokines were used for preclinical studies. The resulting fusion proteins were produced in mammalian cells and purified to homogeneity. The murine Tripokin product was well tolerated in tumor-bearing mice at three doses of 30 μg in a 2-day interval and promoted rapid tumor eradication in murine models, more efficiently than single-agent immunocytokines. Tripokin induced rapid tumor necrosis and stimulated a robust immune response, impacting innate and adaptive immune pathways. In addition, the combination with immune checkpoint inhibitors further boosted the therapeutic efficacy of our molecule. Tripokin represents a promising clinical candidate for the simultaneous delivery of interleukin-2 and tumor necrosis factor to neoplastic sites.
Collapse
Affiliation(s)
- Eleonora Prodi
- Philochem AG, Otelfingen, Switzerland
- CiBIO (Department of Cellular, Computational and Integrative Biology), University of Trento, Trento, Italy
| | - Riccardo Corbellari
- Philochem AG, Otelfingen, Switzerland
- CiBIO (Department of Cellular, Computational and Integrative Biology), University of Trento, Trento, Italy
| | | | | | | | | | | | - Irma Sakic
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Dario Neri
- Philogen Spa, Siena, Italy
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
17
|
Montagner A, Arleo A, Suzzi F, D’Assoro AB, Piscaglia F, Gramantieri L, Giovannini C. Notch Signaling and PD-1/PD-L1 Interaction in Hepatocellular Carcinoma: Potentialities of Combined Therapies. Biomolecules 2024; 14:1581. [PMID: 39766289 PMCID: PMC11674819 DOI: 10.3390/biom14121581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy has shown significant improvement in the survival of patients with hepatocellular carcinoma (HCC) compared to TKIs as first-line treatment. Unfortunately, approximately 30% of HCC exhibits intrinsic resistance to ICIs, making new therapeutic combinations urgently needed. The dysregulation of the Notch signaling pathway observed in HCC can affect immune cell response, reducing the efficacy of cancer immunotherapy. Here, we provide an overview of how Notch signaling regulates immune responses and present the therapeutic rationale for combining Notch signaling inhibition with ICIs to improve HCC treatment. Moreover, we propose using exosomes as non-invasive tools to assess Notch signaling activation in hepatic cancer cells, enabling accurate stratification of patients who can benefit from combined strategies.
Collapse
Affiliation(s)
- Annapaola Montagner
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA;
| | - Andrea Arleo
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
| | - Fabrizia Suzzi
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
| | - Antonino B. D’Assoro
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Catia Giovannini
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
18
|
Park HB, An EK, Kim SJ, Ryu D, Zhang W, Pack CG, Kim H, Kwak M, Im W, Ryu JH, Lee PCW, Jin JO. Anti-PD-L1 Antibody Fragment Linked to Tumor-Targeting Lipid Nanoparticle Can Eliminate Cancer and Its Metastasis via Photoimmunotherapy. ACS NANO 2024; 18:33366-33380. [PMID: 39603816 DOI: 10.1021/acsnano.4c08448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Effective cancer therapy aims to treat primary tumors and metastatic and recurrent cancer. Immune checkpoint blockade-mediated immunotherapy has shown promising effects against tumors; however, its efficacy in metastatic or recurrent cancer is limited. Here, based on the advantages of nanomedicine, lipid nanoparticles (LNPs) that can target tumors are synthesized for photothermal therapy (PTT) and immunotherapy to treat primary and metastatic recurrent cancer. These LNPs, termed piLNPs, are encapsulated with indocyanine green and incorporated with the antigen (Ag)-binding fragment of the anti-PD-L1 antibody for targeting tumors and immunotherapy. Intravenously injected piLNPs in 4T1 breast tumor-bearing BALB/c mice effectively target the 4T1 tumor and are suitable for performing PTT using a near-infrared laser. Moreover, lung metastatic 4T1 tumor growth is completely prevented in mice previously cured of the 4T1 breast tumor by piLNP treatment and rechallenged with lung 4T1 metastatic cancer. Blockage of the second challenged metastatic 4T1 breast cancer by piLNP is due to the activation of Ag-specific T cells. Cytotoxic T lymphocytes from piLNP-cured mice selectively attack 4T1 breast cancer cells. Therefore, piLNP can be used as a multifunctional breast cancer treatment composition that can target tumors, treat primary tumors, and prevent metastasis and recurrence.
Collapse
Affiliation(s)
- Hae-Bin Park
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Eun-Koung An
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - So-Jung Kim
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Dayoung Ryu
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Wei Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Chan-Gi Pack
- Department of Biomedical Engineering, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, South Korea
| | - Minseok Kwak
- Department of Chemistry and Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, South Korea
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Peter C W Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Jun-O Jin
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| |
Collapse
|
19
|
Cuiffo B, Maxwell M, Yan D, Guemiri R, Boone A, Bellet D, Rivest B, Cardia J, Robert C, Fricker SP. Self-delivering RNAi immunotherapeutic PH-762 silences PD-1 to generate local and abscopal antitumor efficacy. Front Immunol 2024; 15:1501679. [PMID: 39697325 PMCID: PMC11652358 DOI: 10.3389/fimmu.2024.1501679] [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: 09/25/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024] Open
Abstract
Objective Immunotherapeutic inhibition of PD-1 by systemically administered monoclonal antibodies is widely used in cancer treatment, but it may cause severe immune-related adverse events (irSAEs). Neoadjuvant PD-1 inhibition before surgery has shown promise in reducing recurrence by stimulating durable antitumor immunity. Local intratumoral (IT) immunotherapy is a potential strategy to minimize irSAEs, but antibodies have limited tumor penetration, making them less suitable for this approach. Therapeutic self-delivering RNAi (INTASYL) is an emerging modality well-suited for neoadjuvant immunotherapy. This study presents preclinical proof-of-concept for PH-762, an INTASYL designed to silence PD-1, currently in clinical development for advanced cutaneous malignancies (ClinicalTrials.gov#NCT06014086). Methods and analysis PH-762 pharmacology was characterized in vitro, and in vivo antitumor efficacy was evaluated using a murine analogue (mPH-762) in syngeneic tumor models with varying PD-1 responsiveness. Bilateral Hepa1-6 models assessed abscopal effects of local treatment. Ex vivo analyses explored mechanisms of direct and abscopal efficacy. Results PH-762 was rapidly internalized by human T cells, silencing PD-1 mRNA and decreasing PD-1 surface protein, enhancing TCR-stimulated IFN-γ and CXCL10 secretion. In vivo, IT mPH-762 provided robust antitumor efficacy, local and lymphatic biodistribution, and was well tolerated. Ex vivo analyses revealed that IT mPH-762 depleted PD-1 protein, promoted leukocyte and T cell infiltration, and correlated with tumor control. IT mPH-762 also demonstrated efficacy against untreated distal tumors (abscopal effect) by priming systemic antitumor immunity. Conclusion These data support PH-762 as a promising candidate for neoadjuvant immunotherapy in clinical studies.
Collapse
Affiliation(s)
| | | | - Dingxue Yan
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Ramdane Guemiri
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | - Andrew Boone
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Deborah Bellet
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | | - James Cardia
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Caroline Robert
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | |
Collapse
|
20
|
Bower BM, Curry SD, Goodwin AP, Cha JN. Photocrosslinkable, Low-Affinity Affibodies Show Improved Transport and Retention in 3D Tumor Spheroids. Biomacromolecules 2024; 25:7511-7517. [PMID: 39453818 DOI: 10.1021/acs.biomac.4c01225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
The efficacy of affinity-based treatments for cancer and other diseases is often limited by poor distribution throughout the targeted tissue. Although lower-affinity antibodies will penetrate more uniformly, these often reach lower concentrations because of their rapid clearance from the tissue. To increase retention and improve distribution, we created low-affinity photocrosslinkable affibodies that can diffuse into dense tumor matrices with limited tumor barrier formation and then be photocrosslinked in place to cell receptors to increase retention. In testing with 3D tumor spheroids, the addition of a 50 nM photocrosslinkable affibody showed a similar level of accumulation at the edges of the spheroid but a higher level near the middle of the spheroid than the wild-type (non-photocrosslinkable) affibody. These results show that target affinity affects protein transport in tumor microenvironments and that covalently cross-linking the ligands to cells may improve both their transport and retention.
Collapse
Affiliation(s)
- Bryce M Bower
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
| | - Shane D Curry
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
| | - Andrew P Goodwin
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
- Materials Science and Engineering Program, University of Colorado, Boulder, Colorado 80303, United States
| | - Jennifer N Cha
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
- Materials Science and Engineering Program, University of Colorado, Boulder, Colorado 80303, United States
- Biomedical Engineering Program, University of Colorado, Boulder, Colorado 80303, United States
| |
Collapse
|
21
|
Pangua C, Espuelas S, Simón JA, Álvarez S, Martínez-Ohárriz C, Collantes M, Peñuelas I, Calvo A, Irache JM. Enhancing bevacizumab efficacy in a colorectal tumor mice model using dextran-coated albumin nanoparticles. Drug Deliv Transl Res 2024:10.1007/s13346-024-01734-3. [PMID: 39455507 DOI: 10.1007/s13346-024-01734-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Bevacizumab is a monoclonal antibody (mAb) that prevents the growth of new blood vessels and is currently employed in the treatment of colorectal cancer (CRC). However, like other mAb, bevacizumab shows a limited penetration in the tumors, hampering their effectiveness and inducing adverse reactions. The aim of this work was to design and evaluate albumin-based nanoparticles, coated with dextran, as carriers for bevacizumab in order to promote its accumulation in the tumor and, thus, improve its antiangiogenic activity. These nanoparticles (B-NP-DEX50) displayed a mean size of about 250 nm and a payload of about 110 µg/mg. In a CRC mice model, these nanoparticles significantly reduced tumor growth and increased tumor doubling time, tumor necrosis and apoptosis more effectively than free bevacizumab. At the end of study, bevacizumab plasma levels were higher in the free drug group, while tumor levels were higher in the B-NP-DEX50 group (2.5-time higher). In line with this, the biodistribution study revealed that nanoparticles accumulated in the tumor core, potentially improving therapeutic efficacy while reducing systemic exposure. In summary, B-NP-DEX can be an adequate alternative to improve the therapeutic efficiency of biologically active molecules, offering a more specific biodistribution to the site of action.
Collapse
Affiliation(s)
- Cristina Pangua
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | - Socorro Espuelas
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Jon Ander Simón
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
| | - Samuel Álvarez
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | | | - María Collantes
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Iván Peñuelas
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Juan M Irache
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain.
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain.
| |
Collapse
|
22
|
Scheuher B, Ghusinga KR, McGirr K, Nowak M, Panday S, Apgar J, Subramanian K, Betts A. Towards a platform quantitative systems pharmacology (QSP) model for preclinical to clinical translation of antibody drug conjugates (ADCs). J Pharmacokinet Pharmacodyn 2024; 51:429-447. [PMID: 37787918 DOI: 10.1007/s10928-023-09884-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/16/2023] [Indexed: 10/04/2023]
Abstract
A next generation multiscale quantitative systems pharmacology (QSP) model for antibody drug conjugates (ADCs) is presented, for preclinical to clinical translation of ADC efficacy. Two HER2 ADCs (trastuzumab-DM1 and trastuzumab-DXd) were used for model development, calibration, and validation. The model integrates drug specific experimental data including in vitro cellular disposition data, pharmacokinetic (PK) and tumor growth inhibition (TGI) data for T-DM1 and T-DXd, as well as system specific data such as properties of HER2, tumor growth rates, and volumes. The model incorporates mechanistic detail at the intracellular level, to account for different mechanisms of ADC processing and payload release. It describes the disposition of the ADC, antibody, and payload inside and outside of the tumor, including binding to off-tumor, on-target sinks. The resulting multiscale PK model predicts plasma and tumor concentrations of ADC and payload. Tumor payload concentrations predicted by the model were linked to a TGI model and used to describe responses following ADC administration to xenograft mice. The model was translated to humans and virtual clinical trial simulations were performed that successfully predicted progression free survival response for T-DM1 and T-DXd for the treatment of HER2+ metastatic breast cancer, including differential efficacy based upon HER2 expression status. In conclusion, the presented model is a step toward a platform QSP model and strategy for ADCs, integrating multiple types of data and knowledge to predict ADC efficacy. The model has potential application to facilitate ADC design, lead candidate selection, and clinical dosing schedule optimization.
Collapse
Affiliation(s)
- Bruna Scheuher
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
- DMPK and Modeling, Takeda, Boston, MA, United States
| | | | - Kimiko McGirr
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
| | | | - Sheetal Panday
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
| | - Joshua Apgar
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
| | - Kalyanasundaram Subramanian
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
- Differentia Bio, Pleasanton, California, United States
| | - Alison Betts
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA.
- DMPK and Modeling, Takeda, Boston, MA, United States.
| |
Collapse
|
23
|
Anaki A, Tzror-Azankot C, Motiei M, Sadan T, Popovtzer R. Impact of synthesis methods on the functionality of antibody-conjugated gold nanoparticles for targeted therapy. NANOSCALE ADVANCES 2024:d4na00134f. [PMID: 39247853 PMCID: PMC11372556 DOI: 10.1039/d4na00134f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/25/2024] [Indexed: 09/10/2024]
Abstract
Gold nanoparticles (GNPs) are emerging as promising modular platforms for antibody-based cancer therapeutics. Their unique physiochemical properties enable efficient binding of multiple antibodies upon a single particle, thereby enhancing therapeutic potential. However, the effect of widely used synthesis techniques on the characteristics and functionality of antibody-GNP platforms has yet to be fully understood. Here, we investigated the effect of key synthesis approaches, namely, covalent binding and physical adsorption, on the properties and anti-cancer functionality of antibody-coated GNPs. By carefully manipulating synthesis variables, including antibody mass in reaction and linker compositions, we revealed a direct impact of these synthesis methods on antibody binding efficiency and anti-cancer functionality. We found that covalent binding of antibodies to GNPs generated a platform with increased cancer cell killing functionality as compared to the adsorption approach. Additionally, a higher antibody mass in the synthesis reaction and a higher polyethylene glycol linker ratio upon covalently bound antibody-GNPs led to increased cell death. Our findings emphasize the critical role of synthesis strategies in determining the functionality of targeted GNPs for effective cancer therapy.
Collapse
Affiliation(s)
- Adi Anaki
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University Ramat Gan 5290002 Israel
| | - Chen Tzror-Azankot
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University Ramat Gan 5290002 Israel
| | - Menachem Motiei
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University Ramat Gan 5290002 Israel
| | - Tamar Sadan
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University Ramat Gan 5290002 Israel
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University Ramat Gan 5290002 Israel
| |
Collapse
|
24
|
Haikal Y, Blazeck J. Exploiting protein domain modularity to enable synthetic control of engineered cells. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 31:100550. [PMID: 39430298 PMCID: PMC11486415 DOI: 10.1016/j.cobme.2024.100550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
The ability to precisely control cellular function in response to external stimuli can enhance the function and safety of cell therapies. In this review, we will detail how the modularity of protein domains has been exploited for cellular control applications, specifically through design of multifunctional synthetic constructs and controllable split moieties. These advances, which build on techniques developed by biologists, protein chemists and drug developers, harness natural evolutionary tendencies of protein domain fusion and fission. In this light, we will highlight recent advances towards the development of novel immunoreceptors, base editors, and cytokines that have achieved intriguing therapeutic potential by taking advantage of well-known protein evolutionary phenomena and have helped cells learn new tricks via synthetic biology. In general, protein modularity, i.e., the relatively facile separation or (re)assembly of functional single protein domains or subdomains, is becoming an enabling phenomenon for cellular engineering by allowing enhanced control of phenotypic responses.
Collapse
Affiliation(s)
- Yusef Haikal
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA
| |
Collapse
|
25
|
Höffgen KS, Dabel J, Konken CP, Depke DA, Hermann S, Dörner W, Schelhaas S, Schäfers M, Mootz HD. Combining poly-epitope MoonTags and labeled nanobodies for signal amplification in cell-specific PET imaging in vivo. Nucl Med Biol 2024; 136-137:108937. [PMID: 38964257 DOI: 10.1016/j.nucmedbio.2024.108937] [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: 03/08/2024] [Revised: 05/31/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Immunorecognition provides an excellent basis for targeted imaging techniques covering a wide range from basic research to diagnostics and from single cells to whole organisms. Fluorescence- or radioisotope-labeled antibodies, antibody fragments or nanobodies enable a direct signal readout upon binding and allow for versatile imaging from microscopy to whole-body imaging. However, as the signal intensity directly correlates with the number of labeled antibodies bound to their epitopes (1:1 binding), sensitivity for low-expressing epitopes can be limiting for visualization. For the first time, we developed poly-epitope tags with multiple copies (1 to 7) of a short peptide epitope, specifically the MoonTag, that are recognized by a labeled nanobody and aimed at signal amplification in microscopy and cell-specific PET imaging. In transiently transfected HeLa cells or stably transduced A4573 cells we characterized complex formation and in vitro signal amplification. Indeed, using fluorescently and radioactively labeled nanobodies we found an approximately linear signal amplification with increasing numbers of epitope copies in vitro. To test the poly-epitope approach in vivo, A4573 tumor cells were injected subcutaneously into the shoulder of NSG mice, with A4573 tumor cells expressing a poly-epitope of 7 MoonTags on one side and WT cells on the other side. Using a [68Ga]-labeled NODAGA-conjugated MoonTag nanobody, we performed PET/CT imaging at day 8-9 after tumor implantation. Specific binding of a [68Ga]-labeled NODAGA-conjugated MoonTag nanobody was observed in 7xMoonTag tumors (1.7 ± 0.5%ID/mL) by PET imaging, showing significantly higher radiotracer accumulation compared to the WT tumors (1.1 ± 0.3%ID/mL; p < 0.01). Ex vivo gamma counter measurements confirmed significantly higher uptake in 7xMoonTag tumors compared to WT tumors (p < 0.001). In addition, MoonTag nanobody binding was detected by autoradiography which was spatially matched with histological analysis of the tumor tissues. In conclusion, we expect nanobody-based poly-epitope tag strategies to be widely applicable for multimodal imaging techniques given the advantageous properties of nanobodies and their amenability to genetic and chemical engineering.
Collapse
Affiliation(s)
| | - Jennifer Dabel
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Christian P Konken
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany; University Hospital Münster, Department of Nuclear Medicine, Münster, Germany
| | - Dominic A Depke
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Sven Hermann
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Wolfgang Dörner
- University of Münster, Institute of Biochemistry, Münster, Germany
| | - Sonja Schelhaas
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Michael Schäfers
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany; University Hospital Münster, Department of Nuclear Medicine, Münster, Germany.
| | - Henning D Mootz
- University of Münster, Institute of Biochemistry, Münster, Germany.
| |
Collapse
|
26
|
Martin S, Wendlinger L, Zitti B, Hicham M, Postupalenko V, Marx L, Giordano-Attianese G, Cribioli E, Irving M, Litvinenko A, Faizova R, Viertl D, Schottelius M. Validation of the C-X-C chemokine receptor 3 (CXCR3) as a target for PET imaging of T cell activation. EJNMMI Res 2024; 14:77. [PMID: 39196448 PMCID: PMC11358572 DOI: 10.1186/s13550-024-01142-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
PURPOSE CXCR3 is expressed on activated T cells and plays a crucial role in T-cell recruitment to the tumor microenvironment (TME) during cell-based and immune checkpoint inhibitor (ICI) immunotherapy. This study utilized a 64Cu-labeled NOTA-α-CXCR3 antibody to assess CXCR3 expression in the TME and validate it as a potential T cell activation biomarker in vivo. PROCEDURES CXCR3+ cells infiltrating MC38 tumors (B57BL/6 mice, untreated and treated with αPD-1/αCTLA-4 ICI) were quantified using fluorescence microscopy and flow cytometry. A commercial anti-mouse CXCR3 antibody (α-CXCR3) was site-specifically conjugated with 2,2,2-(1,4,7-triazacyclononane-1,4,7-triyl)triacetic acid (NOTA) and radiolabeled with 64Cu. Saturation binding of [64Cu]Cu-NOTA-α-CXCR3 was investigated using CHO cells stably transfected with murine CXCR3. Biodistribution and PET imaging studies both at baseline and after 1 to 3 cycles of ICI, respectively, were carried out using different molar activities (10 GBq/µmol to 300 GBq/µmol) of [64Cu]Cu-NOTA-α-CXCR3. RESULTS Flow cytometry analysis at baseline confirmed the presence of CXCR3 + T-cells in MC38 tumors, which was significantly increased at day five after ICI (treated 33.8 ± 17.4 vs. control 8.8 ± 6.2 CD3+CXCR3+ cells/mg). These results were qualitatively and quantitatively confirmed by immunofluorescence of tumor cryoslices. In vivo PET imaging of MC38 tumor bearing mice before, during and after ICI using [64Cu]Cu-NOTA-α-CXCR3 (Kd = 3.3 nM) revealed a strong dependence of CXCR3-specificity of tracer accumulation in secondary lymphoid organs on molar activity. At 300 GBq/µmol (1.5 µg of antibody/mouse), a specific signal was observed in lymph nodes (6.33 ± 1.25 control vs. 3.95 ± 1.23%IA/g blocking) and the spleen (6.04 ± 1.02 control vs. 3.84 ± 0.79%IA/g blocking) at 48 h p.i. Spleen-to-liver ratios indicated a time dependent systemic immune response showing a steady increase from 1.08 ± 0.19 (untreated control) to 1.54 ± 0.14 (three ICI cycles). CONCLUSIONS This study demonstrates the feasibility of in vivo imaging of CXCR3 upregulation under immunotherapy using antibodies. However, high molar activities and low antibody doses are essential for sensitive detection in lymph nodes and spleen. Detecting therapy-induced changes in CXCR3+ T cell numbers in tumors was challenging due to secondary antibody-related effects. Nonetheless, CXCR3 remains a promising target for imaging T cell activation, with anticipated improvements in sensitivity using alternative tracers with high affinities and favorable pharmacokinetics.
Collapse
Affiliation(s)
- Sebastian Martin
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, 1011, Switzerland
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland
- SCCL Swiss Cancer Center Leman, Lausanne, 1011, Switzerland
| | - Lennard Wendlinger
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, 1011, Switzerland
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland
- SCCL Swiss Cancer Center Leman, Lausanne, 1011, Switzerland
| | - Béatrice Zitti
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Mehdi Hicham
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Viktoriia Postupalenko
- Debiopharm Research & Manufacturing SA, Campus "après-demain", Rue du Levant 146, Martigny, 1920, Switzerland
| | - Léo Marx
- Debiopharm Research & Manufacturing SA, Campus "après-demain", Rue du Levant 146, Martigny, 1920, Switzerland
| | - Greta Giordano-Attianese
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, 1066, Switzerland
- Department of Oncology, University Hospital of Lausanne, Lausanne, 1011, Switzerland
| | - Elisabetta Cribioli
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, 1066, Switzerland
- Department of Oncology, University Hospital of Lausanne, Lausanne, 1011, Switzerland
| | - Melita Irving
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, 1066, Switzerland
- Department of Oncology, University Hospital of Lausanne, Lausanne, 1011, Switzerland
| | - Alexandra Litvinenko
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, 1011, Switzerland
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland
- SCCL Swiss Cancer Center Leman, Lausanne, 1011, Switzerland
| | - Radmila Faizova
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, 1011, Switzerland
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland
- SCCL Swiss Cancer Center Leman, Lausanne, 1011, Switzerland
| | - David Viertl
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, 1011, Switzerland
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland
- SCCL Swiss Cancer Center Leman, Lausanne, 1011, Switzerland
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, 1011, Switzerland.
- AGORA, Pôle de recherche sur le cancer, Lausanne, 1011, Switzerland.
- SCCL Swiss Cancer Center Leman, Lausanne, 1011, Switzerland.
| |
Collapse
|
27
|
Shoemaker RL, Larsen RJ, Larsen PA. Single-domain antibodies and aptamers drive new opportunities for neurodegenerative disease research. Front Immunol 2024; 15:1426656. [PMID: 39238639 PMCID: PMC11374656 DOI: 10.3389/fimmu.2024.1426656] [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: 05/01/2024] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
Neurodegenerative diseases (NDs) in mammals, such as Alzheimer's disease (AD), Parkinson's disease (PD), and transmissible spongiform encephalopathies (TSEs), are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Despite the presence of these pathogenic proteins, the immune response in affected individuals remains notably muted. Traditional immunological strategies, particularly those reliant on monoclonal antibodies (mAbs), face challenges related to tissue penetration, blood-brain barrier (BBB) crossing, and maintaining protein stability. This has led to a burgeoning interest in alternative immunotherapeutic avenues. Notably, single-domain antibodies (or nanobodies) and aptamers have emerged as promising candidates, as their reduced size facilitates high affinity antigen binding and they exhibit superior biophysical stability compared to mAbs. Aptamers, synthetic molecules generated from DNA or RNA ligands, present both rapid production times and cost-effective solutions. Both nanobodies and aptamers exhibit inherent qualities suitable for ND research and therapeutic development. Cross-seeding events must be considered in both traditional and small-molecule-based immunodiagnostic and therapeutic approaches, as well as subsequent neurotoxic impacts and complications beyond protein aggregates. This review delineates the challenges traditional immunological methods pose in ND research and underscores the potential of nanobodies and aptamers in advancing next-generation ND diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rachel L Shoemaker
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| | - Roxanne J Larsen
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
- Priogen Corp., St. Paul, MN, United States
| | - Peter A Larsen
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| |
Collapse
|
28
|
Pais DAM, Mayer JPA, Felderer K, Batalha MB, Eichner T, Santos ST, Kumar R, Silva SD, Kaufmann H. Holistic in silico developability assessment of novel classes of small proteins using publicly available sequence-based predictors. J Comput Aided Mol Des 2024; 38:30. [PMID: 39164492 DOI: 10.1007/s10822-024-00569-x] [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: 01/16/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024]
Abstract
The development of novel therapeutic proteins is a lengthy and costly process, with an average attrition rate of 91% (Thomas et al. Clinical Development Success Rates and Contributing Factors 2011-2020, 2021). To increase the probability of success and ensure robust drug supply beyond approval, it is essential to assess the developability profile of new potential drug candidates as early and broadly as possible in development (Jain et al. MAbs, 2023. https://doi.org/10.1016/j.copbio.2011.06.002 ). Predicting these properties in silico is expected to be the next leap in innovation as it would enable significantly reduced development timelines combined with broader screens at lower costs. However, developing predictive algorithms typically requires substantial datasets generated under very defined conditions, a limiting factor especially for new classes of therapeutic proteins that hold immense clinical promise. Here we describe a strategy for assessing the developability of a novel class of small therapeutic Anticalin® proteins using machine learning in conjunction with a knowledge-driven approach. The knowledge-driven approach considers developability attributes such as aggregation propensity, charge variants, immunogenicity, specificity, thermal stability, hydrophobicity, and potential post-translational modifications, to calculate a holistic developability score. Based on sequence-derived descriptors as input parameters we established novel statistical models designed to predict the developability scores for Anticalin proteins. The best models yielded low root mean square errors across the entire dataset and were further validated by removing input data from individual screening campaigns and predicting developability scores for those drug candidates. The adoption of the described workflow will enable significantly streamlined preclinical development of Anticalin drug candidates and could potentially be applied to other therapeutic protein scaffolds.
Collapse
Affiliation(s)
- Daniel A M Pais
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Jan-Peter A Mayer
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Karin Felderer
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Maria B Batalha
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Timo Eichner
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Sofia T Santos
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Raman Kumar
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Sandra D Silva
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Hitto Kaufmann
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany.
| |
Collapse
|
29
|
Gondaliya P, Sayyed AA, Yan IK, Driscoll J, Ziemer A, Patel T. Targeting PD-L1 in cholangiocarcinoma using nanovesicle-based immunotherapy. Mol Ther 2024; 32:2762-2777. [PMID: 38859589 PMCID: PMC11405167 DOI: 10.1016/j.ymthe.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/07/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
This study demonstrates the potential of using biological nanoparticles to deliver RNA therapeutics targeting programmed death-ligand 1 (PD-L1) as a treatment strategy for cholangiocarcinoma (CCA). RNA therapeutics offer prospects for intracellular immune modulation, but effective clinical translation requires appropriate delivery strategies. Milk-derived nanovesicles were decorated with epithelial cellular adhesion molecule (EpCAM) aptamers and used to deliver PD-L1 small interfering RNA (siRNA) or Cas9 ribonucleoproteins directly to CCA cells. In vitro, nanovesicle treatments reduced PD-L1 expression in CCA cells while increasing degranulation, cytokine release, and tumor cell cytotoxicity when tumor cells were co-cultured with T cells or natural killer cells. Similarly, immunomodulation was observed in multicellular spheroids that mimicked the tumor microenvironment. Combining targeted therapeutic vesicles loaded with siRNA to PD-L1 with gemcitabine effectively reduced tumor burden in an immunocompetent mouse CCA model compared with controls. This proof-of-concept study demonstrates the potential of engineered targeted nanovesicle platforms for delivering therapeutic RNA cargoes to tumors, as well as their use in generating effective targeted immunomodulatory therapies for difficult-to-treat cancers such as CCA.
Collapse
Affiliation(s)
- Piyush Gondaliya
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Adil Ali Sayyed
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Irene K Yan
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Julia Driscoll
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Abbye Ziemer
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
30
|
Ding L, Deng B, Chen G. RNA therapeutics targeting PD-L1 is a promising immune-activation strategy against difficult-to-treat cancers. Mol Ther 2024; 32:2439-2440. [PMID: 39038454 PMCID: PMC11405145 DOI: 10.1016/j.ymthe.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Affiliation(s)
- Li Ding
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Bin Deng
- Department of Gastroenterology, Northern Jiangsu People's Hospital, Yangzhou 225001, China.
| | - Gang Chen
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| |
Collapse
|
31
|
Kenakin T. Know your molecule: pharmacological characterization of drug candidates to enhance efficacy and reduce late-stage attrition. Nat Rev Drug Discov 2024; 23:626-644. [PMID: 38890494 DOI: 10.1038/s41573-024-00958-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2024] [Indexed: 06/20/2024]
Abstract
Despite advances in chemical, computational and biological sciences, the rate of attrition of drug candidates in clinical development is still high. A key point in the small-molecule discovery process that could provide opportunities to help address this challenge is the pharmacological characterization of hit and lead compounds, culminating in the selection of a drug candidate. Deeper characterization is increasingly important, because the 'quality' of drug efficacy, at least for G protein-coupled receptors (GPCRs), is now understood to be much more than activation of commonly evaluated pathways such as cAMP signalling, with many more 'efficacies' of ligands that could be harnessed therapeutically. Such characterization is being enabled by novel assays to characterize the complex behaviour of GPCRs, such as biased signalling and allosteric modulation, as well as advances in structural biology, such as cryo-electron microscopy. This article discusses key factors in the assessments of the pharmacology of hit and lead compounds in the context of GPCRs as a target class, highlighting opportunities to identify drug candidates with the potential to address limitations of current therapies and to improve the probability of them succeeding in clinical development.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
32
|
Medina Pérez VM, Baselga M, Schuhmacher AJ. Single-Domain Antibodies as Antibody-Drug Conjugates: From Promise to Practice-A Systematic Review. Cancers (Basel) 2024; 16:2681. [PMID: 39123409 PMCID: PMC11311928 DOI: 10.3390/cancers16152681] [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: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) represent potent cancer therapies that deliver highly toxic drugs to tumor cells precisely, thus allowing for targeted treatment and significantly reducing off-target effects. Despite their effectiveness, ADCs can face limitations due to acquired resistance and potential side effects. OBJECTIVES This study focuses on advances in various ADC components to improve both the efficacy and safety of these agents, and includes the analysis of several novel ADC formats. This work assesses whether the unique features of VHHs-such as their small size, enhanced tissue penetration, stability, and cost-effectiveness-make them a viable alternative to conventional antibodies for ADCs and reviews their current status in ADC development. METHODS Following PRISMA guidelines, this study focused on VHHs as components of ADCs, examining advancements and prospects from 1 January 2014 to 30 June 2024. Searches were conducted in PubMed, Cochrane Library, ScienceDirect and LILACS using specific terms related to ADCs and single-domain antibodies. Retrieved articles were rigorously evaluated, excluding duplicates and non-qualifying studies. The selected peer-reviewed articles were analyzed for quality and synthesized to highlight advancements, methods, payloads, and future directions in ADC research. RESULTS VHHs offer significant advantages for drug conjugation over conventional antibodies due to their smaller size and structure, which enhance tissue penetration and enable access to previously inaccessible epitopes. Their superior stability, solubility, and manufacturability facilitate cost-effective production and expand the range of targetable antigens. Additionally, some VHHs can naturally cross the blood-brain barrier or be easily modified to favor their penetration, making them promising for targeting brain tumors and metastases. Although no VHH-drug conjugates (nADC or nanoADC) are currently in the clinical arena, preclinical studies have explored various conjugation methods and linkers. CONCLUSIONS While ADCs are transforming cancer treatment, their unique mechanisms and associated toxicities challenge traditional views on bioavailability and vary with different tumor types. Severe toxicities, often linked to compound instability, off-target effects, and nonspecific blood cell interactions, highlight the need for better understanding. Conversely, the rapid distribution, tumor penetration, and clearance of VHHs could be advantageous, potentially reducing toxicity by minimizing prolonged exposure. These attributes make single-domain antibodies strong candidates for the next generation of ADCs, potentially enhancing both efficacy and safety.
Collapse
Affiliation(s)
- Víctor Manuel Medina Pérez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Marta Baselga
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
33
|
Nair RR, Prasad A, Bhatavdekar O, Sarkar A, Gabrielson KL, Sofou S. Combined, yet separate: cocktails of carriers (not drugs) for actinium-225 α-particle therapy of solid tumors expressing moderate-to-low levels of targetable markers. Eur J Nucl Med Mol Imaging 2024; 51:2649-2662. [PMID: 38641714 DOI: 10.1007/s00259-024-06710-0] [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: 11/07/2023] [Accepted: 03/28/2024] [Indexed: 04/21/2024]
Abstract
Alpha-particle radionuclide-antibody conjugates are being clinically evaluated against solid tumors even when they moderately express the targeted markers. At this limit of lower tumor-absorbed doses, to maintain efficacy, the few(er) intratumorally delivered alpha-particles need to traverse/hit as many different cancer cells as possible. We complement antibody-radioconjugate therapies with a separate nanocarrier delivering a fraction of the same total injected radioactivity to tumor regions geographically different than those affected by targeting antibodies; these carrier-cocktails collectively distribute the alpha-particle emitters better. METHODS The efficacy of actinium-225 delivered by our carrier-cocktails was assessed in vitro and on mice with orthotopic MDA-MB-436 and/or MDA-MB-231 triple-negative breast cancers and/or an ectopic BxPC3 pancreatic cancer. Cells/tumors were chosen to express low-to-moderate levels of HER1, as model antibody-targeted marker. RESULTS Independent of cell line, antibody-radioconjugates were most lethal on cell monolayers. On spheroids, with radii greater than alpha-particles' range, carrier-cocktails improved killing efficacy (p < 0.0500). Treatment with carrier-cocktails decreased the MDA-MB-436 and MDA-MB-231 orthotopic tumor volumes by 73.7% and 72.1%, respectively, relative to treatment with antibody-radioconjugates alone, at same total injected radioactivity; these carrier-cocktails completely eliminated formation of spontaneous metastases vs. 50% and 25% elimination in mice treated with antibody-radioconjugates alone. In BxPC3 tumor-bearing mice, carrier-cocktails increased the median survival to 25-26 days (in male-female animals) vs. 20-21 days of mice treated with antibody-radioconjugates alone (vs. 17 days for non-treated animals). Survival with carrier-cocktail radiotherapy was further prolonged by pre-injecting low-dose, standard-of-care, gemcitabine (p = 0.0390). CONCLUSION Tumor-agnostic carrier-cocktails significantly enhance the therapeutic efficacy of existing alpha-particle radionuclide-antibody treatments.
Collapse
Affiliation(s)
- Rajiv Ranjit Nair
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Aprameya Prasad
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Omkar Bhatavdekar
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Aira Sarkar
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Kathleen L Gabrielson
- Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, USA
| | - Stavroula Sofou
- Chemical and Biomolecular Engineering (ChemBE), Institute for NanoBioTechnology (INBT), Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Cancer Invasion & Metastasis Program, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
34
|
Schlegel LS, Werbrouck C, Boettcher M, Schlegel P. Universal CAR 2.0 to overcome current limitations in CAR therapy. Front Immunol 2024; 15:1383894. [PMID: 38962014 PMCID: PMC11219820 DOI: 10.3389/fimmu.2024.1383894] [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: 02/08/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has effectively complemented the treatment of advanced relapsed and refractory hematological cancers. The remarkable achievements of CD19- and BCMA-CAR T therapies have raised high expectations within the fields of hematology and oncology. These groundbreaking successes are propelling a collective aspiration to extend the reach of CAR therapies beyond B-lineage malignancies. Advanced CAR technologies have created a momentum to surmount the limitations of conventional CAR concepts. Most importantly, innovations that enable combinatorial targeting to address target antigen heterogeneity, using versatile adapter CAR concepts in conjunction with recent transformative next-generation CAR design, offer the promise to overcome both the bottleneck associated with CAR manufacturing and patient-individualized treatment regimens. In this comprehensive review, we delineate the fundamental prerequisites, navigate through pivotal challenges, and elucidate strategic approaches, all aimed at paving the way for the future establishment of multitargeted immunotherapies using universal CAR technologies.
Collapse
Affiliation(s)
- Lara Sophie Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Coralie Werbrouck
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Pediatric Hematology and Oncology, Westmead Children’s Hospital, Sydney, NSW, Australia
| |
Collapse
|
35
|
O'Connell RP, Liaw K, Wellhausen N, Chuckran CA, Bhojnagarwala PS, Bordoloi D, Park D, Shupin N, Kulp D, June CH, Weiner D. Format-tuning of in vivo-launched bispecific T cell engager enhances efficacy against renal cell carcinoma. J Immunother Cancer 2024; 12:e008733. [PMID: 38834201 PMCID: PMC11163651 DOI: 10.1136/jitc-2023-008733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Advanced clear cell renal cell carcinoma (ccRCC) is a prevalent kidney cancer for which long-term survival rates are abysmal, though immunotherapies are showing potential. Not yet clinically vetted are bispecific T cell engagers (BTEs) that activate T cell-mediated cancer killing through intercellular synapsing. Multiple BTE formats exist, however, with limited cross-characterizations to help optimize new drug design. Here, we developed BTEs to treat ccRCC by targeting carbonic anhydrase 9 (CA9) while characterizing the persistent BTE (PBTE) format and comparing it to a new format, the persistent multivalent T cell engager (PMTE). These antibody therapies against ccRCC are developed as both recombinant and synthetic DNA (synDNA) medicines. METHODS Antibody formatting effects on binding kinetics were assessed by flow cytometry and intercellular synaptic strength assays while potency was tested using T-cell activation and cytotoxicity assays. Mouse models were used to study antibody plasma and tumor pharmacokinetics, as well as antitumor efficacy as both recombinant and synDNA medicines. Specifically, three models using ccRCC cell line xenografts and human donor T cells in immunodeficient mice were used to support this study. RESULTS Compared with a first-generation BTE, we show that the PBTE reduced avidity, intercellular synaptic strength, cytotoxic potency by as much as 33-fold, and ultimately efficacy against ccRCC tumors in vivo. However, compared with the PBTE, we demonstrate that the PMTE improved cell avidity, restored intercellular synapses, augmented cytotoxic potency by 40-fold, improved tumor distribution pharmacokinetics by 2-fold, and recovered synDNA efficacy in mouse tumor models by 20-fold. All the while, the PMTE displayed a desirable half-life of 4 days in mice compared with the conventional BTE's 2 hours. CONCLUSIONS With impressive efficacy, the CA9-targeted PMTE is a promising new therapy for advanced ccRCC, which can be effectively delivered through synDNA. The highly potent PMTE format itself is a promising new tool for future applications in the multispecific antibody space.
Collapse
Affiliation(s)
- Ryan P O'Connell
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Vaccine & Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kevin Liaw
- Vaccine & Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Nils Wellhausen
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | - Devivasha Bordoloi
- Vaccine & Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Daniel Park
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Vaccine & Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Nicholas Shupin
- Vaccine & Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Daniel Kulp
- Vaccine & Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David Weiner
- Vaccine & Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| |
Collapse
|
36
|
Rubahamya B, Dong S, Thurber GM. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. SCIENCE ADVANCES 2024; 10:eadk1894. [PMID: 38820153 PMCID: PMC11141632 DOI: 10.1126/sciadv.adk1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Antibody drug conjugates (ADCs) have made impressive strides in the clinic in recent years with 11 Food and Drug Administration approvals, including 6 for the treatment of patients with solid tumors. Despite this success, the development of new agents remains challenging with a high failure rate in the clinic. Here, we show that current approved ADCs for the treatment of patients with solid tumors can all show substantial efficacy in some mouse models when administered at a similar weight-based [milligrams per kilogram (mg/kg)] dosing in mice that is tolerated in the clinic. Mechanistically, equivalent mg/kg dosing results in a similar drug concentration in the tumor and a similar tissue penetration into the tumor due to the unique delivery features of ADCs. Combined with computational approaches, which can account for the complex distribution within the tumor microenvironment, these scaling concepts may aid in the evaluation of new agents and help design therapeutics with maximum clinical efficacy.
Collapse
Affiliation(s)
- Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
37
|
Toàn NM, Vágner A, Nagy G, Ország G, Nagy T, Csikos C, Váradi B, Sajtos GZ, Kapus I, Szoboszlai Z, Szikra D, Trencsényi G, Tircsó G, Garai I. [ 52Mn]Mn-BPPA-Trastuzumab: A Promising HER2-Specific PET Radiotracer. J Med Chem 2024; 67:8261-8270. [PMID: 38690886 DOI: 10.1021/acs.jmedchem.4c00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
This study aimed to develop a novel radiotracer using trastuzumab and the long-lived [52Mn]Mn isotope for HER2-targeted therapy selection and monitoring. A new Mn(II) chelator, BPPA, synthesized from a rigid bispyclen platform possessing a picolinate pendant arm, formed a stable and inert Mn(II) complex with favorable relaxation properties. BPPA was converted into a bifunctional chelator (BFC), conjugated to trastuzumab, and labeled with [52Mn]Mn isotope. In comparison to DOTA-GA-trastuzumab, the BPPA-trastuzumab conjugate exhibits a labeling efficiency with [52Mn]Mn approximately 2 orders of magnitude higher. In female CB17 SCID mice bearing 4T1 (HER2-) and MDA-MB-HER2+ (HER2+) xenografts, [52Mn]Mn-BPPA-trastuzumab demonstrated superior uptake in HER2+ cells on day 3, with a 3-4 fold difference observed on day 7. Overall, the hexadentate BPPA chelator proves to be exceptional in binding Mn(II). Upon coupling with trastuzumab as a BFC ligand, it becomes an excellent imaging probe for HER2-positive tumors. [52Mn]Mn-BPPA-trastuzumab enables an extended imaging time window and earlier detection of HER2-positive tumors with superior tumor-to-background contrast.
Collapse
Affiliation(s)
- Ngô Minh Toàn
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen H-4032, Hungary
| | | | | | | | - Tamás Nagy
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen H-4032, Hungary
- Scanomed Ltd., Debrecen H-4032, Hungary
| | - Csaba Csikos
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen H-4032, Hungary
| | - Balázs Váradi
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen H-4032, Hungary
- Doctoral School of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen H-4032, Hungary
| | - Gergő Zoltán Sajtos
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen H-4032, Hungary
- Doctoral School of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen H-4032, Hungary
| | - István Kapus
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen H-4032, Hungary
- Doctoral School of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen H-4032, Hungary
| | | | - Dezső Szikra
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen H-4032, Hungary
- Scanomed Ltd., Debrecen H-4032, Hungary
| | - György Trencsényi
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen H-4032, Hungary
- Scanomed Ltd., Debrecen H-4032, Hungary
| | - Gyula Tircsó
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen H-4032, Hungary
| | - Ildikó Garai
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen H-4032, Hungary
- Scanomed Ltd., Debrecen H-4032, Hungary
| |
Collapse
|
38
|
Jolly KJ, Zhang F. IVT-mRNA reprogramming of myeloid cells for cancer immunotherapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:247-288. [PMID: 39034054 DOI: 10.1016/bs.apha.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
In the past decade, in vitro transcribed messenger RNAs (IVT-mRNAs) have emerged as promising therapeutic molecules. The clinical success of COVID-19 mRNA vaccines developed by Pfizer-BioNTech and Moderna, have demonstrated that IVT-mRNAs can be safely and successfully used in a clinical setting, and efforts are underway to develop IVT-mRNAs for therapeutic applications. Current applications of mRNA-based therapy have been focused on (1) mRNA vaccines for infectious diseases and cancer treatment; (2) protein replacement therapy; (3) gene editing therapy; and (4) cell-reprogramming therapies. Due to the recent clinical progress of cell-based immunotherapies, the last direction-the use of IVT-mRNAs as a therapeutic approach to program immune cells for the treatment of cancer has received extensive attention from the cancer immunotherapy field. Myeloid cells are important components of our immune system, and they play critical roles in mediating disease progression and regulating immunity against diseases. In this chapter, we discussed the progress of using IVT-mRNAs as a therapeutic approach to program myeloid cells against cancer and other immune-related diseases. Towards this direction, we first reviewed the pharmacology of IVT-mRNAs and the biology of myeloid cells as well as myeloid cell-targeting therapeutics. We then presented a few cases of current IVT-mRNA-based approaches to target and reprogram myeloid cells for disease treatment and discussed the advantages and limitations of these approaches. Finally, we presented our considerations in designing mRNA-based approaches to target myeloid cells for disease treatment.
Collapse
Affiliation(s)
- Kevon J Jolly
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Fan Zhang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Chemical Engineering, College of Engineering, University of Florida, Gainesville, FL, United States; Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
39
|
Ludwig SD, Meksiriporn B, Tan J, Kureshi R, Mishra A, Kaeo KJ, Zhu A, Stavrakis G, Lee SJ, Schodt DJ, Wester MJ, Kumar D, Lidke KA, Cox AL, Dooley HM, Nimmagadda S, Spangler JB. Multiparatopic antibodies induce targeted downregulation of programmed death-ligand 1. Cell Chem Biol 2024; 31:904-919.e11. [PMID: 38547863 PMCID: PMC11102303 DOI: 10.1016/j.chembiol.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/28/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024]
Abstract
Programmed death-ligand 1 (PD-L1) drives inhibition of antigen-specific T cell responses through engagement of its receptor programmed death-1 (PD-1) on activated T cells. Overexpression of these immune checkpoint proteins in the tumor microenvironment has motivated the design of targeted antibodies that disrupt this interaction. Despite clinical success of these antibodies, response rates remain low, necessitating novel approaches to enhance performance. Here, we report the development of antibody fusion proteins that block immune checkpoint pathways through a distinct mechanism targeting molecular trafficking. By engaging multiple receptor epitopes on PD-L1, our engineered multiparatopic antibodies induce rapid clustering, internalization, and degradation in an epitope- and topology-dependent manner. The complementary mechanisms of ligand blockade and receptor downregulation led to more durable immune cell activation and dramatically reduced PD-L1 availability in mouse tumors. Collectively, these multiparatopic antibodies offer mechanistic insight into immune checkpoint protein trafficking and how it may be manipulated to reprogram immune outcomes.
Collapse
Affiliation(s)
- Seth D Ludwig
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bunyarit Meksiriporn
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biology, School of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - Jiacheng Tan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rakeeb Kureshi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Akhilesh Mishra
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kyle J Kaeo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Angela Zhu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Georgia Stavrakis
- Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, MD 21205, USA
| | - Stephen J Lee
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - David J Schodt
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Michael J Wester
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Dhiraj Kumar
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Andrea L Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Helen M Dooley
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology (IMET), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sridhar Nimmagadda
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jamie B Spangler
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
40
|
Feng X, Wen Z, Zhu X, Yan X, Duan Y, Huang Y. Anti-HER2 Immunoliposomes: Antitumor Efficacy Attributable to Targeted Delivery of Anthraquinone-Fused Enediyne. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307865. [PMID: 38355309 PMCID: PMC11077693 DOI: 10.1002/advs.202307865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/24/2023] [Indexed: 02/16/2024]
Abstract
Although natural products are essential sources of small-molecule antitumor drugs, some can exert substantial toxicities, limiting their clinical utility. Anthraquinone-fused enediyne natural products are remarkably potent antitumor drug candidates, and uncialamycin and tiancimycin (TNM) A are under development as antibody-drug conjugates. Herein, a novel drug delivery system is introduced for TNM A using anti-human epidermal growth factor receptor 2 (HER2) immunoliposomes (ILs). Trastuzumab-coated TNM A-loaded ILs (HER2-TNM A-ILs) is engineered with an average particle size of 182.8 ± 2.1 nm and a zeta potential of 1.75 ± 0.12 mV. Compared with liposomes lacking trastuzumab, HER2-TNM A-ILs exhibited selective toxicity against HER2-positive KPL-4 and SKBR3 cells. Coumarin-6, a fluorescent TNM A surrogate, is encapsulated within anti-HER2 ILs; the resultant ILs have enhanced cellular uptake in KPL-4 and SKBR3 cells when compared with control liposomes. Furthermore, ILs loaded with more Cy5.5 accumulated in KPL-4 mouse tumors. A single HER2-TNM A-IL dose (0.02 mg kg-1) suppressed the growth of HER2-positive KPL-4 mouse tumors without apparent toxicity. This study not only provides a straightforward method for the effective delivery of TNM A against HER2-positive breast tumors but also underscores the potential of IL-based drug delivery systems when employing highly potent cytotoxins as payloads.
Collapse
Affiliation(s)
- Xueqiong Feng
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
| | - Zhongqing Wen
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
| | - Xiangcheng Zhu
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug DiscoverChangshaHunan410011China
| | - Xiaohui Yan
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Yanwen Duan
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug DiscoverChangshaHunan410011China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug DiscoveryChangshaHunan410011China
| | - Yong Huang
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug DiscoveryChangshaHunan410011China
- Institute of Health and MedicineHefei Comprehensive National Science CenterHefeiAnhui230093China
| |
Collapse
|
41
|
Kim HS, Hariri K, Zhang X, Chen L, Katz BB, Pei H, Louie SG, Zhang Y. Synthesis of site-specific Fab-drug conjugates using ADP-ribosyl cyclases. Protein Sci 2024; 33:e4924. [PMID: 38501590 PMCID: PMC10949397 DOI: 10.1002/pro.4924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 03/20/2024]
Abstract
Targeted delivery of small-molecule drugs via covalent attachments to monoclonal antibodies has proved successful in clinic. For this purpose, full-length antibodies are mainly used as drug-carrying vehicles. Despite their flexible conjugation sites and versatile biological activities, intact immunoglobulins with conjugated drugs, which feature relatively large molecular weights, tend to have restricted tissue distribution and penetration and low fractions of payloads. Linking small-molecule therapeutics to other formats of antibody may lead to conjugates with optimal properties. Here, we designed and synthesized ADP-ribosyl cyclase-enabled fragment antigen-binding (Fab) drug conjugates (ARC-FDCs) by utilizing CD38 catalytic activity. Through rapidly forming a stable covalent bond with a nicotinamide adenine dinucleotide (NAD+ )-based drug linker at its active site, CD38 genetically fused with Fab mediates robust site-specific drug conjugations via enzymatic reactions. Generated ARC-FDCs with defined drug-to-Fab ratios display potent and antigen-dependent cytotoxicity against breast cancer cells. This work demonstrates a new strategy for developing site-specific FDCs. It may be applicable to different antibody scaffolds for therapeutic conjugations, leading to novel targeted agents.
Collapse
Affiliation(s)
- Hyo Sun Kim
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Kimia Hariri
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Xiao‐Nan Zhang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Liang‐Chieh Chen
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Benjamin B. Katz
- Department of ChemistryUniversity of California, IrvineIrvineCaliforniaUSA
| | - Hua Pei
- Titus Family Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Stan G. Louie
- Titus Family Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Norris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Norris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Chemistry, Dornsife College of Letters, Arts and SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Research Center for Liver DiseasesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
42
|
Liu K, Li M, Li Y, Li Y, Chen Z, Tang Y, Yang M, Deng G, Liu H. A review of the clinical efficacy of FDA-approved antibody‒drug conjugates in human cancers. Mol Cancer 2024; 23:62. [PMID: 38519953 PMCID: PMC10960395 DOI: 10.1186/s12943-024-01963-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/21/2024] [Indexed: 03/25/2024] Open
Abstract
While strategies such as chemotherapy and immunotherapy have become the first-line standard therapies for patients with advanced or metastatic cancer, acquired resistance is still inevitable in most cases. The introduction of antibody‒drug conjugates (ADCs) provides a novel alternative. ADCs are a new class of anticancer drugs comprising the coupling of antitumor mAbs with cytotoxic drugs. Compared with chemotherapeutic drugs, ADCs have the advantages of good tolerance, accurate target recognition, and small effects on noncancerous cells. ADCs occupy an increasingly important position in the therapeutic field. Currently, there are 13 Food and Drug Administration (FDA)‒approved ADCs and more than 100 ADC drugs at different stages of clinical trials. This review briefly describes the efficacy and safety of FDA-approved ADCs, and discusses the related problems and challenges to provide a reference for clinical work.
Collapse
Affiliation(s)
- Kaifeng Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Meijia Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yudong Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yutong Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Zixin Chen
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yiqi Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Meitian Yang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Guoquan Deng
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Hongwei Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China.
| |
Collapse
|
43
|
Guo Y, Li X, Xie Y, Wang Y. What influences the activity of Degrader-Antibody conjugates (DACs). Eur J Med Chem 2024; 268:116216. [PMID: 38387330 DOI: 10.1016/j.ejmech.2024.116216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024]
Abstract
The targeted protein degradation (TPD) technology employing proteolysis-targeting chimeras (PROTACs) has been widely applied in drug chemistry and chemical biology for the treatment of cancer and other diseases. PROTACs have demonstrated significant advantages in targeting undruggable targets and overcoming drug resistance. However, despite the efficient degradation of targeted proteins achieved by PROTACs, they still face challenges related to selectivity between normal and cancer cells, as well as issues with poor membrane permeability due to their substantial molecular weight. Additionally, the noteworthy toxicity resulting from off-target effects also needs to be addressed. To solve these issues, Degrader-Antibody Conjugates (DACs) have been developed, leveraging the targeting and internalization capabilities of antibodies. In this review, we elucidates the characteristics and distinctions between DACs, and traditional Antibody-drug conjugates (ADCs). Meanwhile, we emphasizes the significance of DACs in facilitating the delivery of PROTACs and delves into the impact of various components on DAC activity. These components include antibody targets, drug-antibody ratio (DAR), linker types, PROTACs targets, PROTACs connections, and E3 ligase ligands. The review also explores the suitability of different targets (antibody targets or PROTACs targets) for DACs, providing insights to guide the design of PROTACs better suited for antibody conjugation.
Collapse
Affiliation(s)
- Yaolin Guo
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yang Xie
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
44
|
Tsuchikama K, Anami Y, Ha SYY, Yamazaki CM. Exploring the next generation of antibody-drug conjugates. Nat Rev Clin Oncol 2024; 21:203-223. [PMID: 38191923 DOI: 10.1038/s41571-023-00850-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 149.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Antibody-drug conjugates (ADCs) are a promising cancer treatment modality that enables the selective delivery of highly cytotoxic payloads to tumours. However, realizing the full potential of this platform necessitates innovative molecular designs to tackle several clinical challenges such as drug resistance, tumour heterogeneity and treatment-related adverse effects. Several emerging ADC formats exist, including bispecific ADCs, conditionally active ADCs (also known as probody-drug conjugates), immune-stimulating ADCs, protein-degrader ADCs and dual-drug ADCs, and each offers unique capabilities for tackling these various challenges. For example, probody-drug conjugates can enhance tumour specificity, whereas bispecific ADCs and dual-drug ADCs can address resistance and heterogeneity with enhanced activity. The incorporation of immune-stimulating and protein-degrader ADCs, which have distinct mechanisms of action, into existing treatment strategies could enable multimodal cancer treatment. Despite the promising outlook, the importance of patient stratification and biomarker identification cannot be overstated for these emerging ADCs, as these factors are crucial to identify patients who are most likely to derive benefit. As we continue to deepen our understanding of tumour biology and refine ADC design, we will edge closer to developing truly effective and safe ADCs for patients with treatment-refractory cancers. In this Review, we highlight advances in each ADC component (the monoclonal antibody, payload, linker and conjugation chemistry) and provide more-detailed discussions on selected examples of emerging novel ADCs of each format, enabled by engineering of one or more of these components.
Collapse
Affiliation(s)
- Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Yasuaki Anami
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Summer Y Y Ha
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chisato M Yamazaki
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
45
|
Mohr P, van Sluis J, Lub-de Hooge MN, Lammertsma AA, Brouwers AH, Tsoumpas C. Advances and challenges in immunoPET methodology. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1360710. [PMID: 39355220 PMCID: PMC11440922 DOI: 10.3389/fnume.2024.1360710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/05/2024] [Indexed: 10/03/2024]
Abstract
Immuno-positron emission tomography (immunoPET) enables imaging of specific targets that play a role in targeted therapy and immunotherapy, such as antigens on cell membranes, targets in the disease microenvironment, or immune cells. The most common immunoPET applications use a monoclonal antibody labeled with a relatively long-lived positron emitter such as 89Zr (T 1/2 = 78.4 h), but smaller antibody-based constructs labeled with various other positron emitting radionuclides are also being investigated. This molecular imaging technique can thus guide the development of new drugs and may have a pivotal role in selecting patients for a particular therapy. In early phase immunoPET trials, multiple imaging time points are used to examine the time-dependent biodistribution and to determine the optimal imaging time point, which may be several days after tracer injection due to the slow kinetics of larger molecules. Once this has been established, usually only one static scan is performed and semi-quantitative values are reported. However, total PET uptake of a tracer is the sum of specific and nonspecific uptake. In addition, uptake may be affected by other factors such as perfusion, pre-/co-administration of the unlabeled molecule, and the treatment schedule. This article reviews imaging methodologies used in immunoPET studies and is divided into two parts. The first part summarizes the vast majority of clinical immunoPET studies applying semi-quantitative methodologies. The second part focuses on a handful of studies applying pharmacokinetic models and includes preclinical and simulation studies. Finally, the potential and challenges of immunoPET quantification methodologies are discussed within the context of the recent technological advancements provided by long axial field of view PET/CT scanners.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
46
|
Li D, Ding L, Chen Y, Wang Z, Zeng Z, Ma X, Huang H, Li H, Qian X, Yang Z, Zhu H. Exploration of radionuclide labeling of a novel scFv-Fc fusion protein targeting CLDN18.2 for tumor diagnosis and treatment. Eur J Med Chem 2024; 266:116134. [PMID: 38266552 DOI: 10.1016/j.ejmech.2024.116134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/04/2024] [Accepted: 01/07/2024] [Indexed: 01/26/2024]
Abstract
PURPOSE Claudin 18.2 (CLDN18.2), due to its highly selective expression in tumor cells, has made breakthrough progress in clinical research and is expected to be integrated into routine tumor diagnosis and treatment. METHODS In this research, we obtained an scFv-Fc fusion protein (SF106) targeting CLDN18.2 through hybridoma technology. The scFv-Fc fusion protein was labeled with radioactive isotopes (124I and 177Lu) to generate the radio-probes. The targeting and specificity of the radio-probes were tested in cellular models, and its diagnostic and therapeutic potential was further evaluated in tumor-bearing models. RESULTS The molecular probes [124I]I-SF106 and [177Lu]Lu-DOTA-SF106 possess high radiochemical purity (RCP, 98.18 ± 0.93 % and 97.05 ± 1.1 %) and exhibit good stability in phosphate buffer saline and 5 % human serum albumin (92.44 ± 4.68 % and 91.03 ± 2.42 % at 120 h). [124I]I-SF106 uptake in cells expressing CLDN18.2 was well targeted and specific, and the dissociation constant was 17.74 nM [124I]I-SF106 micro-PET imaging showed that the maximum standardized uptake value (SUVmax) was significantly higher than CLDN18.2-negative tumors (1.83 ± 0.02 vs. 1.23 ± 0.04, p < 0.001). The maximum uptake was attained in tumors expressing CLDN18.2 at 48 h after injection. [124I]I-SF106 and [177Lu]Lu-DOTA-SF106 dosimetric study showed that the effective dose in humans complies with the medical safety standards required for their clinical application. The results of treatment experiments showed that 3 MBq of [177Lu]Lu-DOTA-SF106 in CLDN18.2-expressing tumor-bearing mice could significantly inhibit tumor growth. CONCLUSION These results indicate that radionuclide-labeled scFv-Fc molecular probes ([124I]I-SF106 and [177Lu]Lu-DOTA-SF106) provide a new possibility for the diagnosis and treatment of CLDN18.2-positive cancer patients in clinical practice.
Collapse
Affiliation(s)
- Dapeng Li
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lei Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yan Chen
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zilei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Ziqing Zeng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaopan Ma
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Haifeng Huang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550025, Guizhou, China
| | - Hongjun Li
- Suzhou Transcenta Therapeutics Co., Ltd, Suzhou, 215000, China
| | - Xueming Qian
- Suzhou Transcenta Therapeutics Co., Ltd, Suzhou, 215000, China.
| | - Zhi Yang
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Hua Zhu
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
47
|
Douez E, Allard-Vannier E, Amar IAM, Jolivet L, Boursin F, Maisonial-Besset A, Witkowski T, Chezal JM, Colas C, Letast S, Auvert E, Denevault-Sabourin C, Aubrey N, Joubert N. Branched pegylated linker-auristatin to control hydrophobicity for the production of homogeneous minibody-drug conjugate against HER2-positive breast cancer. J Control Release 2024; 366:567-584. [PMID: 38215985 DOI: 10.1016/j.jconrel.2024.01.012] [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: 07/21/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Trastuzumab emtansine (Kadcyla®) was the first antibody-drug conjugate (ADC) approved by the Food and Drug Administration in 2013 against a solid tumor, and the first ADC to treat human epidermal growth factor receptor 2 positive (HER2+) breast cancer. However, this second generation ADC is burden by several limitations included heterogeneity, limited activity against heterogeneous tumor (regarding antigen expression) and suboptimal tumor penetration. To address this, different development strategies are oriented towards homogeneous conjugation, new drugs, optimized linkers and/or smaller antibody formats. To reach better developed next generation ADCs, a key parameter to consider is the management of the hydrophobicity associated with the linker-drug, increasing with and limiting the drug-to-antibody ratio (DAR) of the ADC. Here, an innovative branched pegylated linker was developed, to control the hydrophobicity of the monomethyl auristatin E (MMAE) and its cathepsin B-sensitive trigger. This branched pegylated linker-MMAE was then used for the efficient generation of internalizing homogeneous ADC of DAR 8 and minibody-drug conjugate of DAR 4, targeting HER2. Both immunoconjugates were then evaluated in vitro and in vivo on breast cancer models. Interestingly, this study highlighted that the minibody-MMAE conjugate of DAR 4 was the best immunoconjugate regarding in vitro cellular internalization and cytotoxicity, gamma imaging, ex vivo biodistribution profile in mice and efficient reduction of tumor size in vivo. These results are very promising and encourage us to explore further fragment-drug conjugate development.
Collapse
Affiliation(s)
- Emmanuel Douez
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France; Pharmacy Department, Tours University Hospital, F-37200 Tours, France
| | - Emilie Allard-Vannier
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France.
| | | | - Louis Jolivet
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Fanny Boursin
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Aurélie Maisonial-Besset
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Tiffany Witkowski
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Jean-Michel Chezal
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Cyril Colas
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France; UMR 7311 ICOA, CNRS, University of Orléans, F-45067 Orléans, France
| | - Stéphanie Letast
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France
| | - Etienne Auvert
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France
| | | | - Nicolas Aubrey
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Nicolas Joubert
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France.
| |
Collapse
|
48
|
Sharma R, Mukherjee A, Kumar A, Sarma HD. Evaluation of 177Lu-Labeled Pertuzumab F(ab') 2 Fragments for HER2-Positive Cancer Targeting: A Comparative In Vitro and In Vivo Study. Cancer Biother Radiopharm 2024; 39:64-74. [PMID: 38363819 DOI: 10.1089/cbr.2023.0108] [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: 02/18/2024] Open
Abstract
Background: Radiolabeled antibody fragments present a promising opportunity as theranostic agents, offering distinct advantages over whole antibodies. In this study, the authors investigate the potential of [177Lu]Lu-DTPA-F(ab')2-pertuzumab as a theranostic agent for precise targeting of human epidermal growth factor receptor 2 (HER2)-positive cancers. Additionally, the authors aim to quantitatively assess the binding synergism in the presence of cold trastuzumab. Materials and Methods: F(ab')2-pertuzumab was prepared by pepsin digestion and conjugated with a bifunctional chelator. The immunoconjugate was radiolabeled with 177Lu and characterized by chromatography techniques. Binding parameters (affinity, specificity, and immunoreactivity) and cellular binding enhancement studies were evaluated in HER2-overexpressing and triple-negative cell lines. The in vivo enhancement in tumor uptake of the radiolabeled immunoformulation was assessed in severe combined immunodeficient (SCID) mice bearing tumors, both in the presence and absence of unlabeled trastuzumab. Results: The formulation of [177Lu]Lu-DTPA-F(ab')2-pertuzumab could be prepared in high yields and with consistent radiochemical purity, ensuring reproducibility. Comprehensive in vitro and in vivo evaluation studies confirmed high specificity and immunoreactivity of the formulation toward HER2 receptors. Binding synergism of radiolabeled pertuzumab fragments in the presence of trastuzumab to HER2 receptors was observed. Conclusions: The radioformulation of [177Lu]Lu-DTPA-F(ab')2-pertuzumab holds great promise as a targeted approach for addressing HER2-positive cancers. A potentially effective strategy to amplify therapeutic efficacy involves dual epitope targeting by combining radiolabeled pertuzumab with cold trastuzumab.
Collapse
Affiliation(s)
- Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Anuj Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| | - Haladhar Dev Sarma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| |
Collapse
|
49
|
Shen X, Pan D, Gong Q, Gu Z, Luo K. Enhancing drug penetration in solid tumors via nanomedicine: Evaluation models, strategies and perspectives. Bioact Mater 2024; 32:445-472. [PMID: 37965242 PMCID: PMC10641097 DOI: 10.1016/j.bioactmat.2023.10.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Effective tumor treatment depends on optimizing drug penetration and accumulation in tumor tissue while minimizing systemic toxicity. Nanomedicine has emerged as a key solution that addresses the rapid clearance of free drugs, but achieving deep drug penetration into solid tumors remains elusive. This review discusses various strategies to enhance drug penetration, including manipulation of the tumor microenvironment, exploitation of both external and internal stimuli, pioneering nanocarrier surface engineering, and development of innovative tactics for active tumor penetration. One outstanding strategy is organelle-affinitive transfer, which exploits the unique properties of specific tumor cell organelles and heralds a potentially transformative approach to active transcellular transfer for deep tumor penetration. Rigorous models are essential to evaluate the efficacy of these strategies. The patient-derived xenograft (PDX) model is gaining traction as a bridge between laboratory discovery and clinical application. However, the journey from bench to bedside for nanomedicines is fraught with challenges. Future efforts should prioritize deepening our understanding of nanoparticle-tumor interactions, re-evaluating the EPR effect, and exploring novel nanoparticle transport mechanisms.
Collapse
Affiliation(s)
- Xiaoding Shen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
50
|
Nyesiga B, Levin M, Säll A, Rosén A, Jansson K, Fritzell S, Hägerbrand K, Weilguny D, von Schantz L. RUBY® - a tetravalent (2+2) bispecific antibody format with excellent functionality and IgG-like stability, pharmacology and developability properties. MAbs 2024; 16:2330113. [PMID: 38527972 DOI: 10.1080/19420862.2024.2330113] [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: 11/02/2023] [Accepted: 03/08/2024] [Indexed: 03/27/2024] Open
Abstract
Despite the large number of existing bispecific antibody (bsAb) formats, the generation of novel bsAbs is still associated with development and bioprocessing challenges. Here, we present RUBY, a novel bispecific antibody format that allows rapid generation of bsAbs that fulfill key development criteria. The RUBYTM format has a 2 + 2 geometry, where two Fab fragments are linked via their light chains to the C-termini of an IgG, and carries mutations for optimal chain pairing. The unique design enables generation of bsAbs with mAb-like attributes. Our data demonstrate that RUBY bsAbs are compatible with small-scale production systems for screening purposes and can be produced at high yields (>3 g/L) from stable cell lines. The bsAbs produced are shown to, in general, contain low amounts of aggregates and display favorable solubility and stress endurance profiles. Further, compatibility with various IgG isotypes is shown and tailored Fc gamma receptor binding confirmed. Also, retained interaction with FcRn is demonstrated to translate into a pharmacokinetic profile in mice and non-human primates that is comparable to mAb controls. Functionality of conditional active RUBY bsAbs is confirmed in vitro. Anti-tumor effects in vivo have previously been demonstrated, and shown to be superior to a comparable mAb, and here it is further shown that RUBY bsAbs penetrate and localize to tumor tissue in vivo. In all, the RUBY format has attractive mAb-like attributes and offers the possibility to mitigate many of the development challenges linked to other bsAb formats, facilitating both high functionality and developability.
Collapse
Affiliation(s)
- Barnabas Nyesiga
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
- Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Mattias Levin
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Anna Säll
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Anna Rosén
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Kim Jansson
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Sara Fritzell
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | | | | |
Collapse
|