1
|
Kazuta N, Nakashima K, Tarumizu Y, Sato T, Maya Y, Watanabe H, Ono M. Novel Radiotheranostic Ligands Targeting Prostate-Specific Membrane Antigen Based on Dual Linker Approach. Mol Pharm 2025; 22:377-386. [PMID: 39614820 DOI: 10.1021/acs.molpharmaceut.4c00974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Radiotheranostics using prostate-specific membrane antigen (PSMA)-targeting radioligands offers precision medicine by performing radionuclide therapy based on results of diagnosis. Albumin binder (ALB) binds to albumin reversibly and contributes to effective radiotheranostics by enhancing tumor accumulation of PSMA-targeting radioligands. We newly developed two ALB-containing PSMA-targeting radioligands including dual functional linkers, a hydrophilic linker, d-glutamic acid, and a hydrophobic linker, 4-(aminomethyl)benzoic acid, with the opposite arrangement (PNT-DA6 and PNT-DA7). A biodistribution study of [111In]In-PNT-DA6 indicated that the introduction and arrangement of dual functional linkers contributed to improved pharmacokinetics. A single photon emission computed tomography study of [111In]In-PNT-DA6 produced a clear PSMA-expressing tumor image. Moreover, [225Ac]Ac-PNT-DA6 showed the inhibition of tumor growth in targeted radionuclide therapy in PSMA-expressing tumor-bearing mice. These results indicated that [111In]In-PNT-DA6 and [225Ac]Ac-PNT-DA6 exhibited useful characteristics as PSMA-targeting radiotheranostic ligands.
Collapse
Affiliation(s)
- Nobuki Kazuta
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuta Tarumizu
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Kitasode, Sodegaura-shi, Chiba 299-0266, Japan
| | - Takumi Sato
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Kitasode, Sodegaura-shi, Chiba 299-0266, Japan
| | - Yoshifumi Maya
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Kitasode, Sodegaura-shi, Chiba 299-0266, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
2
|
Linciano S, Vigolo E, Rosato A, Kumada Y, Angelini A. Albumin-based strategies to effectively prolong the circulation half-life of small immunomodulatory payloads in cancer therapy. Curr Opin Biotechnol 2024; 90:103218. [PMID: 39481162 DOI: 10.1016/j.copbio.2024.103218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 08/14/2024] [Accepted: 09/26/2024] [Indexed: 11/02/2024]
Abstract
Small immunomodulatory payloads (IMMs) such as peptide vaccines and cytokines have the capability to activate and boost the immune response against cancer. However, their clinical use has often been hindered by their poor stability and short circulating half-lives. To enhance the pharmacokinetic properties of small IMMs and promote their trafficking and accumulation in lymphatic and tumor tissues, a large variety of strategies have been developed. One of the most successful relies on the use of serum albumin (SA), the most abundant protein in the circulatory and lymphatic system. Here, we report a comparative analysis of the different covalent and noncovalent SA-based strategies applied so far to improve the efficacy of small IMMs in cancer therapy.
Collapse
Affiliation(s)
- Sara Linciano
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; Department of Functional Chemistry and Engineering, Kyoto Institute of Technology, 1 Matsugasaki-Hashikami-Cho, Sakyo-ku, Kyoto 606-0951, Japan
| | - Emilia Vigolo
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy
| | - Antonio Rosato
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy; Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, Via Giustiniani 2, 35124 Padua, Italy
| | - Yoichi Kumada
- Department of Functional Chemistry and Engineering, Kyoto Institute of Technology, 1 Matsugasaki-Hashikami-Cho, Sakyo-ku, Kyoto 606-0951, Japan
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; European Centre for Living Technology (ECLT), Ca' Bottacin, Dorsoduro 3911, Calle Crosera, 30123 Venice, Italy.
| |
Collapse
|
3
|
Yamaguchi K, Kazuta N, Tsuchihashi S, Watanabe H, Ono M. Structure-affinity-pharmacokinetics relationships of 111In-labeled PSMA-targeted ligands with different albumin binders. Nucl Med Biol 2024; 138-139:108945. [PMID: 39153354 DOI: 10.1016/j.nucmedbio.2024.108945] [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: 04/20/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA) is a promising target for treating metastatic castration-resistant prostate cancer. Our previous report presented 111In- or 225Ac-labeled PSMA-NAT-DA1 (PNT-DA1) as a PSMA-targeted ligand. To improve its therapeutic efficiency, PNT-DA1 contains 4-(p-iodophenyl)butyric acid (IPBA), which is known as an albumin binder (ALB) moiety. However, few reports have examined the relationship between the chemical modification of the ALB moiety and pharmacokinetics of PSMA-targeted radioligands. To assess this relationship, we designed, synthesized, and evaluated four [111In]In-PNT-DA1 analogues with ALB moieties different from IPBA. METHODS The [111In]In-PNT-DA1 analogues were synthesized from their corresponding precursors through ligand substitution reaction. The stability of [111In]In-PNT-DA1 analogues in mouse plasma, their affinity for human serum albumin (HSA), their binding to mouse plasma proteins, and their affinity for PSMA were evaluated in vitro. The tissue distribution profile of the radioligands was assessed in biodistribution studies using LNCaP tumor-bearing nude mice. RESULTS All [111In]In-PNT-DA1 analogues were obtained at a high radiochemical yield and purity. These analogues were highly stable in mouse plasma after 24 h. The binding affinity for HSA significantly varied among the different ALB moieties. Moreover, high affinity for mouse plasma proteins was observed for all [111In]In-PNT-DA1 analogues compared with their counterparts without an ALB moiety. The affinity for PSMA was comparable for all radioligands. In the biodistribution assay, the pharmacokinetics of [111In]In-PNT-DA1 analogues varied markedly depending on the type of ALB moiety. In particular, tumor area under the curve (AUC) values were increased for radioligands with higher blood retention, while some previous studies reported that compounds with moderate blood retention exhibited the highest tumor AUC values. CONCLUSION The introduction of an appropriate ALB moiety into the ligand may lead to the development of more useful PSMA-targeted radioligands with higher tumor accumulation.
Collapse
Affiliation(s)
- Keisei Yamaguchi
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Nobuki Kazuta
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shohei Tsuchihashi
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
4
|
Shirian J, Hockla A, Gleba JJ, Coban M, Rotenberg N, Strik LM, Alasonyalilar Demirer A, Pawlush ML, Copland JA, Radisky ES, Shifman JM. Improving Circulation Half-Life of Therapeutic Candidate N-TIMP2 by Unfolded Peptide Extension. Biomolecules 2024; 14:1187. [PMID: 39334953 PMCID: PMC11429640 DOI: 10.3390/biom14091187] [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: 07/02/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Matrix metalloproteinases (MMPs) are significant drivers of many diseases, including cancer, and are established targets for drug development. Tissue inhibitors of metalloproteinases (TIMPs) are endogenous MMP inhibitors and are being pursued for the development of anti-MMP therapeutics. TIMPs possess many attractive properties for drug candidates, such as complete MMP inhibition, low toxicity, low immunogenicity, and high tissue permeability. However, a major challenge with TIMPs is their rapid clearance from the bloodstream due to their small size. This study explores a method for extending the plasma half-life of the N-terminal domain of TIMP2 (N-TIMP2) by appending it with a long, intrinsically unfolded tail containing Pro, Ala, and Thr (PATylation). We designed and produced two PATylated N-TIMP2 constructs with tail lengths of 100 and 200 amino acids (N-TIMP2-PAT100 and N-TIMP2-PAT200). Both constructs demonstrated higher apparent molecular weights and retained high inhibitory activity against MMP-9. N-TIMP2-PAT200 significantly increased plasma half-life in mice compared to the non-PATylated variant, enhancing its therapeutic potential. PATylation offers distinct advantages for half-life extension, such as fully genetic encoding, monodispersion, and biodegradability. It can be easily applied to N-TIMP2 variants engineered for high affinity and selectivity toward individual MMPs, creating promising candidates for drug development against MMP-related diseases.
Collapse
Affiliation(s)
- Jason Shirian
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Alexandra Hockla
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Justyna J. Gleba
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Matt Coban
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Naama Rotenberg
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Laura M. Strik
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Aylin Alasonyalilar Demirer
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Matt L. Pawlush
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Evette S. Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Julia M. Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
5
|
Kim SJ, Jung CW, Anh NH, Yoon YC, Long NP, Hong SS, Cho EJ, Kwon SW. Metabolic phenotyping combined with transcriptomics metadata fortifies the diagnosis of early-stage Hepatocellular carcinoma. J Adv Res 2024:S2090-1232(24)00391-6. [PMID: 39243943 DOI: 10.1016/j.jare.2024.09.007] [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: 01/28/2024] [Revised: 07/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
INTRODUCTION The low sensitivity of alpha-fetoprotein (AFP) renders it unsuitable as a stand-alone marker for early hepatocellular carcinoma (eHCC) surveillance. Therefore, additional blood-based biomarkers with enhanced sensitivities are required. OBJECTIVES In light of the metabolic changes that are distinctive to eHCC development, the current study presents a panel of serum metabolites that may serve as noninvasive diagnostic indicators for patients with eHCC. METHODS Serum samples obtained from normal control (NC), cirrhosis, and eHCC patients were analyzed by four different metabolomic platforms. A meta-analysis of very early-stage HCC transcriptomic datasets retrieved from public sources supports the integrated interpretation with metabolic changes. RESULTS A total of 94 metabolites were significantly correlated with a progressive disease status. Integrated analysis of the significant metabolites and differentially expressed genes from meta-analysis emphasized metabolic pathways including bile acid biosynthesis, phenylalanine and tyrosine metabolism, and butanoate metabolism. The 11 metabolites associated with these pathways were compiled into a metabolite panel for use as diagnostic signatures. With an accuracy of 81.8%, compared with 45.4% for a model trained solely on AFP, the model enhanced its ability to differentiate between the three groups by incorporating a metabolite panel and AFP. Upon examining the trained models using receiver operating characteristic curves, the AFP and metabolite panel combined model exhibited greater area under the curve values in comparisons between NC and eHCC (1.000 versus 0.810) and cirrhosis and eHCC (0.926 versus 0.556). The result was consistent in an independent validation cohort. CONCLUSION This study emphasizes the role of circulating metabolite markers in the diagnosis of eHCC.
Collapse
Affiliation(s)
- Sun Jo Kim
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Cheol Woon Jung
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Nguyen Hoang Anh
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Cheol Yoon
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Science, College of Medicine, and Program in Biomedical Sciences and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Eun Ju Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sung Won Kwon
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
6
|
Vidal-Calvo EE, Martin-Salazar A, Choudhary S, Dagil R, Raghavan SSR, Duvnjak L, Nordmaj MA, Clausen TM, Skafte A, Oberkofler J, Wang K, Agerbæk MØ, Løppke C, Jørgensen AM, Ropac D, Mujollari J, Willis S, Garcias López A, Miller RL, Karlsson RTG, Goerdeler F, Chen YH, Colaço AR, Wang Y, Lavstsen T, Martowicz A, Nelepcu I, Marzban M, Oo HZ, Ørum-Madsen MS, Wang Y, Nielsen MA, Clausen H, Wierer M, Wolf D, Gögenur I, Theander TG, Al-Nakouzi N, Gustavsson T, Daugaard M, Salanti A. Tumor-agnostic cancer therapy using antibodies targeting oncofetal chondroitin sulfate. Nat Commun 2024; 15:7553. [PMID: 39215044 PMCID: PMC11364678 DOI: 10.1038/s41467-024-51781-0] [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: 10/09/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Molecular similarities between embryonic and malignant cells can be exploited to target tumors through specific signatures absent in healthy adult tissues. One such embryonic signature tumors express is oncofetal chondroitin sulfate (ofCS), which supports disease progression and dissemination in cancer. Here, we report the identification and characterization of phage display-derived antibody fragments recognizing two distinct ofCS epitopes. These antibody fragments show binding affinity to ofCS in the low nanomolar range across a broad selection of solid tumor types in vitro and in vivo with minimal binding to normal, inflamed, or benign tumor tissues. Anti-ofCS antibody drug conjugates and bispecific immune cell engagers based on these targeting moieties disrupt tumor progression in animal models of human and murine cancers. Thus, anti-ofCS antibody fragments hold promise for the development of broadly effective therapeutic and diagnostic applications targeting human malignancies.
Collapse
Affiliation(s)
- Elena Ethel Vidal-Calvo
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark.
| | - Anne Martin-Salazar
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Swati Choudhary
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Robert Dagil
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Sai Sundar Rajan Raghavan
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lara Duvnjak
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Mie Anemone Nordmaj
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Ann Skafte
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Jan Oberkofler
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kaituo Wang
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Ø Agerbæk
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VARCT Diagnostics, Copenhagen, Denmark
| | - Caroline Løppke
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Amalie Mundt Jørgensen
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VARCT Diagnostics, Copenhagen, Denmark
| | - Daria Ropac
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Joana Mujollari
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Shona Willis
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Agnès Garcias López
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rebecca Louise Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Richard Torbjörn Gustav Karlsson
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Felix Goerdeler
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Ana R Colaço
- Proteomics Research Infrastructure, University of Copenhagen, Copenhagen, Denmark
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Thomas Lavstsen
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Agnieszka Martowicz
- Department of Internal Medicine V, Haematology & Oncology, Comprehensive Cancer Center Innsbruck (CCCI) and Tyrolean Cancer Research Institute (TKFI), Medical University of Innsbruck, Innsbruck, Austria
| | - Irina Nelepcu
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mona Marzban
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Maj Sofie Ørum-Madsen
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Morten A Nielsen
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Wierer
- Proteomics Research Infrastructure, University of Copenhagen, Copenhagen, Denmark
| | - Dominik Wolf
- Department of Internal Medicine V, Haematology & Oncology, Comprehensive Cancer Center Innsbruck (CCCI) and Tyrolean Cancer Research Institute (TKFI), Medical University of Innsbruck, Innsbruck, Austria
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital Køge, Køge, Denmark
| | - Thor G Theander
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Nader Al-Nakouzi
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias Gustavsson
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Mads Daugaard
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark.
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark.
| |
Collapse
|
7
|
Guzzi R, Bartucci R. Thermal effects and drugs competition on the palmitate binding capacity of human serum albumin. Biochem Biophys Res Commun 2024; 722:150168. [PMID: 38797156 DOI: 10.1016/j.bbrc.2024.150168] [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: 03/14/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Human serum albumin (HSA) is the most abundant plasma protein of the circulatory system. It is a multidomain, multifunctional protein that, combining diverse affinities and wide specificity, binds, stores, and transports a variety of biological compounds, pharmacores, and fatty acids. HSA is finding increasing uses in drug-delivery due to its ability to carry functionalized ligands and prodrugs. All this raises the question of competition for binding sites occupancy in case of multiple ligands, which in turn influences the protein structure/dynamic/function relationship and also has an impact on the biomedical applications. In this work, the effects of interactive binding of palmitic acid (PA), warfarin (War) and ibuprofen (Ibu) on the thermal stability of HSA were studied using DSC, ATR-FTIR, and EPR. PA is a high-affinity physiological ligand, while the two drugs are widely used for their anticoagulant (War) and anti-inflammatory (Ibu) efficacy, and are exogenous compounds that accommodate in the deputed drug site DS1 and DS2, respectively overlapping with some of the fatty acid binding sites. The results indicate that HSA acquires the highest thermal stability when it is fully saturated with PA. The binding of this physiological ligand does not hamper the binding of War or Ibu to the native state of the protein. In addition, the three ligands bind simultaneously, suggesting a synergic cooperative influence due to allosteric effects. The increased thermal stability subsequent to binary and multiple ligands binding moderates protein aggregation propensity and restricts protein dynamics. The biophysics findings provide interesting features about protein stability, aggregation, and dynamics in interaction with multiple ligands and are relevant in drug-delivery.
Collapse
Affiliation(s)
- Rita Guzzi
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036, Rende, Italy; CNR-NANOTEC, Department of Physics, University of Calabria, 87036, Rende, Italy.
| | - Rosa Bartucci
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036, Rende, Italy
| |
Collapse
|
8
|
Kazuta N, Nakashima K, Watanabe H, Ono M. Effect of Linker Entities on Pharmacokinetics of 111In-Labeled Prostate-Specific Membrane Antigen-Targeting Ligands with an Albumin Binder. ACS Pharmacol Transl Sci 2024; 7:2401-2413. [PMID: 39144550 PMCID: PMC11320743 DOI: 10.1021/acsptsci.4c00257] [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: 05/02/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024]
Abstract
In the field of radiopharmaceutical development targeting cancer, an albumin binder (ALB) is commonly used to improve accumulation of radioligands in tumors because it has high binding affinity for albumin and extends the circulation time of radioligands. The further development of ALB-containing radioligands is also expected to regulate their pharmacokinetics. In this study, we newly designed and synthesized [111In]In-PNT-DA1 derivatives, prostate-specific membrane antigen (PSMA)-targeting radioligands including a functional linker (d-glutamic acid or 4-(aminomethyl)benzoic acid), and evaluated the relationships among the structure, albumin-binding affinity, and pharmacokinetics. These derivatives showed a different binding affinity for albumin by the introduction of a linker. Biodistribution studies revealed that the introduction of a linker affects the pharmacokinetics of each derivative. The biodistribution studies also suggested that moderate albumin-binding affinity enhances the tumor/kidney ratio of the derivative. SPECT imaging using [111In]In-PNT-DA3 with the highest tumor/kidney ratio among [111In]In-PNT-DA1 derivatives led to clear visualization of a PSMA-positive LNCaP tumor. The results suggest that the appropriate introduction of linker entities may be necessary to improve the pharmacokinetics of PSMA-targeting radioligands.
Collapse
Affiliation(s)
- Nobuki Kazuta
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| |
Collapse
|
9
|
Boersma B, Poinot H, Pommier A. Stimulating the Antitumor Immune Response Using Immunocytokines: A Preclinical and Clinical Overview. Pharmaceutics 2024; 16:974. [PMID: 39204319 PMCID: PMC11357675 DOI: 10.3390/pharmaceutics16080974] [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: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are immune modulators which can enhance the immune response and have been proven to be an effective class of immunotherapy. Nevertheless, the clinical use of cytokines in cancer treatment has faced several challenges associated with poor pharmacokinetic properties and the occurrence of adverse effects. Immunocytokines (ICKs) have emerged as a promising approach to overcome the pharmacological limitations observed with cytokines. ICKs are fusion proteins designed to deliver cytokines in the tumor microenvironment by taking advantage of the stability and specificity of immunoglobulin-based scaffolds. Several technological approaches have been developed. This review focuses on ICKs designed with the most impactful cytokines in the cancer field: IL-2, TNFα, IL-10, IL-12, IL-15, IL-21, IFNγ, GM-CSF, and IFNα. An overview of the pharmacological effects of the naked cytokines and ICKs tested for cancer therapy is detailed. A particular emphasis is given on the immunomodulatory effects of ICKs associated with their technological design. In conclusion, this review highlights active ways of development of ICKs. Their already promising results observed in clinical trials are likely to be improved with the advances in targeting technologies such as cytokine/linker engineering and the design of multispecific antibodies with tumor targeting and immunostimulatory functional properties.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland;
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Hélène Poinot
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Research Centre in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Aurélien Pommier
- UMR1240 Imagerie Moléculaire et Stratégies Théranostiques INSERM, Université Clermont Auvergne, BP 184, F-63005 Clermont-Ferrand, France
| |
Collapse
|
10
|
Shirian J, Hockla A, Gleba JJ, Coban M, Rotenberg N, Strik LM, Alasonyalilar Demirer A, Pawlush ML, Copland JA, Radisky ES, Shifman JM. Improving Circulation Half-Life of Therapeutic Candidate N-TIMP2 by Unfolded Peptide Extension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600979. [PMID: 38979353 PMCID: PMC11230438 DOI: 10.1101/2024.06.27.600979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Matrix Metalloproteinases (MMPs) are drivers of many diseases including cancer and are established targets for drug development. Tissue inhibitors of metalloproteinases (TIMPs) are human proteins that inhibit MMPs and are being pursued for the development of anti-MMP therapeutics. TIMPs possess many attractive properties of a drug candidate, such as complete MMP inhibition, low toxicity and immunogenicity, high tissue permeability and others. A major challenge with TIMPs, however, is their formulation and delivery, as these proteins are quickly cleared from the bloodstream due to their small size. In this study, we explore a new method for plasma half-life extension for the N-terminal domain of TIMP2 (N-TIMP2) through appending it with a long intrinsically unfolded tail containing a random combination of Pro, Ala, and Thr (PATylation). We design, produce and explore two PATylated N-TIMP2 constructs with a tail length of 100- and 200-amino acids (N-TIMP2-PAT100 and N-TIMP2-PAT200, respectively). We demonstrate that both PATylated N-TIMP2 constructs possess apparent higher molecular weights compared to the wild-type protein and retain high inhibitory activity against MMP-9. Furthermore, when injected into mice, N-TIMP2-PAT200 exhibited a significant increase in plasma half-life compared to the non-PATylated variant, enhancing the therapeutic potential of the protein. Thus, we establish that PATylation could be successfully applied to TIMP-based therapeutics and offers distinct advantages as an approach for half-life extension, such as fully genetic encoding of the gene construct, mono-dispersion, and biodegradability. Furthermore, PATylation could be easily applied to N-TIMP2 variants engineered to possess high affinity and selectivity toward individual MMP family members, thus creating attractive candidates for drug development against MMP-related diseases.
Collapse
Affiliation(s)
- Jason Shirian
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Alexandra Hockla
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Justyna J. Gleba
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Matt Coban
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Naama Rotenberg
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Laura M. Strik
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Aylin Alasonyalilar Demirer
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Matt L. Pawlush
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Evette S. Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Julia M. Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| |
Collapse
|
11
|
Iwamoto N, Kai T, Inuki S, Ohno H, Maeda H, Watanabe H, Maruyama T, Oishi S. Mirror-Image Human Serum Albumin Domain III as a Tool for Analyzing Site II-Dependent Molecular Recognition. Bioconjug Chem 2024; 35:816-825. [PMID: 38781049 DOI: 10.1021/acs.bioconjchem.4c00150] [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: 05/25/2024]
Abstract
Human serum albumin (HSA) as a drug carrier can significantly improve the pharmacokinetic profiles of short-lived therapeutics. Conjugation of albumin-binding moieties (ABMs) to therapeutic agents may prolong their serum half-life by promoting their association with endogenous HSA. To discover a new molecular class of ABMs from mirror-image chemical space, a preparation protocol for bioactive HSA domain III and its d-enantiomer (d-HSA domain III) was established. Structural and functional analyses suggested that the synthetic protein enantiomers exhibited mirror-image structures and stereoselective neonatal fragement crystallizable receptor (FcRn) recognition. Additionally, the ligand-binding properties of synthetic l-HSA domain III were comparable with those of site II in native HSA, as confirmed using site II-selective fluorescent probes and an esterase substrate. Synthetic d-HSA domain III is an attractive tool for analyzing the site II-dependent molecular recognition properties of HSA.
Collapse
Affiliation(s)
- Naoya Iwamoto
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo ku, Kyoto 606-8501, Japan
| | - Takuma Kai
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo ku, Kyoto 606-8501, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo ku, Kyoto 606-8501, Japan
| | - Hitoshi Maeda
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Hiroshi Watanabe
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Toru Maruyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo ku, Kyoto 606-8501, Japan
- Laboratory of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina ku, Kyoto 607-8412, Japan
| |
Collapse
|
12
|
Shangguan W, Li X, Wang Y, Huang Z, Dong Y, Feng M, Feng J. Design and Biological Evaluation of the Long-Acting C5-Inhibited Ornithodoros moubata Complement Inhibitor (OmCI) Modified with Fatty Acid. Bioconjug Chem 2024; 35:653-664. [PMID: 38593046 DOI: 10.1021/acs.bioconjchem.4c00126] [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: 04/11/2024]
Abstract
Disorder of complement response is a significant pathogenic factor causing some autoimmune and inflammation diseases. The Ornithodoros moubata Complement Inhibitor (OmCI), a small 17 kDa natural protein, was initially extracted from soft tick salivary glands. The protein was found binding to complement C5 specifically, inhibiting the activation of the complement pathway, which is a successful therapeutic basis of complement-mediated diseases. However, a short half-life due to rapid renal clearance is a common limitation of small proteins for clinical application. In this study, we extended the half-life of OmCI by modifying it with fatty acid, which was a method used to improve the pharmacokinetics of native peptides and proteins. Five OmCI mutants were initially designed, and single-site cysteine mutation was introduced to each of them. After purification, four OmCI mutants were obtained that showed similar in vitro biological activities. Three mutants of them were subsequently coupled with different fatty acids by nucleophilic substitution. In total, 15 modified derivatives were screened and tested for anticomplement activity in vitro. The results showed that coupling with fatty acid would not significantly affect their complement-inhibitory activity (CH50 and AH50). OmCIT90C-CM02 and OmCIT90C-CM05 were validated as the applicable OmCI bioconjugates for further pharmacokinetic assessments, and both showed improved plasma half-life in mice compared with unmodified OmCI (15.86, 17.96 vs 2.57 h). In summary, our data demonstrated that OmCI conjugated with fatty acid could be developed as the potential long-acting C5 complement inhibitor in the clinic.
Collapse
Affiliation(s)
- Wenwen Shangguan
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Xiaowan Li
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Yandan Wang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 310014 Hangzhou, China
| | - Zongqing Huang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Yuanzhen Dong
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Meiqing Feng
- School of Pharmacy, Fudan University, 201203 Shanghai, China
| | - Jun Feng
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| |
Collapse
|
13
|
Abdallah M, Lin L, Styles IK, Mörsdorf A, Grace JL, Gracia G, Landersdorfer CB, Nowell CJ, Quinn JF, Whittaker MR, Trevaskis NL. Impact of conjugation to different lipids on the lymphatic uptake and biodistribution of brush PEG polymers. J Control Release 2024; 369:146-162. [PMID: 38513730 DOI: 10.1016/j.jconrel.2024.03.032] [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: 01/03/2024] [Revised: 02/28/2024] [Accepted: 03/16/2024] [Indexed: 03/23/2024]
Abstract
Delivery to peripheral lymphatics can be achieved following interstitial administration of nano-sized delivery systems (nanoparticles, liposomes, dendrimers etc) or molecules that hitchhike on endogenous nano-sized carriers (such as albumin). The published work concerning the hitchhiking approach has mostly focussed on the lymphatic uptake of vaccines conjugated directly to albumin binding moieties (ABMs such as lipids, Evans blue dye derivatives or peptides) and their subsequent trafficking into draining lymph nodes. The mechanisms underpinning access and transport of these constructs into lymph fluid, including potential interaction with other endogenous nanocarriers such as lipoproteins, have largely been ignored. Recently, we described a series of brush polyethylene glycol (PEG) polymers containing end terminal short-chain or medium-chain hydrocarbon tails (1C2 or 1C12, respectively), cholesterol moiety (Cho), or medium-chain or long-chain diacylglycerols (2C12 or 2C18, respectively). We evaluated the association of these materials with albumin and lipoprotein in rat plasma, and their intravenous (IV) and subcutaneous (SC) pharmacokinetic profiles. Here we fully detail the association of this suite of polymers with albumin and lipoproteins in rat lymph, which is expected to facilitate lymph transport of the materials from the SC injection site. Additionally, we characterise the thoracic lymph uptake, tissue and lymph node biodistribution of the lipidated brush PEG polymers following SC administration to thoracic lymph cannulated rats. All polymers had moderate lymphatic uptake in rats following SC dosing with the lymph uptake higher for 1C2-PEG, 2C12-PEG and 2C18-PEG (5.8%, 5.9% and 6.7% dose in lymph, respectively) compared with 1C12-PEG and Cho-PEG (both 1.5% dose in lymph). The enhanced lymph uptake of 1C2-PEG, 2C12-PEG and 2C18-PEG appeared related to their association profile with different lipoproteins. The five polymers displayed different biodistribution patterns in major organs and tissues in mice. All polymers reached immune cells deep within the inguinal lymph nodes of mice following SC dosing. The ability to access these immune cells suggests the potential of the polymers as platforms for the delivery of vaccines and immunotherapies. Future studies will focus on evaluating the lymphatic targeting and therapeutic potential of drug or vaccine-loaded polymers in pre-clinical disease models.
Collapse
Affiliation(s)
- Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Lihuan Lin
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Alexander Mörsdorf
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - James L Grace
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Cornelia B Landersdorfer
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - John F Quinn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia; Department of Chemical Engineering, Faculty of Engineering, Monash University, Clayton, VIC, Australia
| | - Michael R Whittaker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia; Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Li Q, Kong Y, Zhong Y, Huang A, Ying T, Wu Y. Half-life extension of single-domain antibody-drug conjugates by albumin binding moiety enhances antitumor efficacy. MedComm (Beijing) 2024; 5:e557. [PMID: 38737471 PMCID: PMC11082534 DOI: 10.1002/mco2.557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 05/14/2024] Open
Abstract
Single-domain antibody-drug conjugates (sdADCs) have been proven to have deeper solid tumor penetration and intratumor accumulation capabilities due to their smaller size compared with traditional IgG format ADCs. However, one of the key challenges for improving clinical outcomes of sdADCs is their abbreviated in vivo half-life. In this study, we innovatively fused an antihuman serum albumin (αHSA) nanobody to a sdADCs targeting oncofetal antigen 5T4, conferring serum albumin binding to enhance the pharmacokinetic profiles of sdADCs. The fusion protein was conjugated with monomethyl auristatin E (MMAE) at s224c site mutation. The conjugate exhibited potent cytotoxicity against various tumor cells. Compared with the nonalbumin-binding counterparts, the conjugate exhibited a 10-fold extended half-life in wild-type mice and fivefold prolonged serum half-life in BxPC-3 xenograft tumor models as well as enhanced tumor accumulation and retention in mice. Consequently, n501-αHSA-MMAE showed potent antitumor effects, which were comparable to n501-MMAE in pancreatic cancer BxPC-3 xenograft tumor models; however, in human ovarian teratoma PA-1 xenograft tumor models, n501-αHSA-MMAE significantly improved antitumor efficacy. Moreover, the conjugate showed mitigated hepatotoxicity. In summary, our results suggested that fusion to albumin-binding moiety as a viable strategy can enhance the therapeutic potential of sdADCs through optimized pharmacokinetics.
Collapse
Affiliation(s)
- Quanxiao Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yu Kong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yuxuan Zhong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Ailing Huang
- College of Life SciencesHebei Agricultural UniversityBaodingChina
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yanling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| |
Collapse
|
15
|
Starnes HM, Jackson TW, Rock KD, Belcher SM. Quantitative cross-species comparison of serum albumin binding of per- and polyfluoroalkyl substances from five structural classes. Toxicol Sci 2024; 199:132-149. [PMID: 38518100 PMCID: PMC11057469 DOI: 10.1093/toxsci/kfae028] [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] [Indexed: 03/24/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a class of over 8000 chemicals, many of which are persistent, bioaccumulative, and toxic to humans, livestock, and wildlife. Serum protein binding affinity is instrumental in understanding PFAS toxicity, yet experimental binding data is limited to only a few PFAS congeners. Previously, we demonstrated the usefulness of a high-throughput, in vitro differential scanning fluorimetry assay for determination of relative binding affinities of human serum albumin for 24 PFAS congeners from 6 chemical classes. In the current study, we used this assay to comparatively examine differences in human, bovine, porcine, and rat serum albumin binding of 8 structurally informative PFAS congeners from 5 chemical classes. With the exception of the fluorotelomer alcohol 1H, 1H, 2H, 2H-perfluorooctanol (6:2 FTOH), each PFAS congener bound by human serum albumin was also bound by bovine, porcine, and rat serum albumin. The critical role of the charged functional headgroup in albumin binding was supported by the inability of albumin of each species tested to bind 6:2 FTOH. Significant interspecies differences in serum albumin binding affinities were identified for each of the bound PFAS congeners. Relative to human albumin, perfluoroalkyl carboxylic and sulfonic acids were bound with greater affinity by porcine and rat serum albumin, and the perfluoroalkyl ether acid congener bound with lower affinity to porcine and bovine serum albumin. These comparative affinity data for PFAS binding by serum albumin from human, experimental model, and livestock species reduce critical interspecies uncertainty and improve accuracy of predictive bioaccumulation and toxicity assessments for PFAS.
Collapse
Affiliation(s)
- Hannah M Starnes
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27607, USA
| | - Thomas W Jackson
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27607, USA
| | - Kylie D Rock
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27607, USA
| | - Scott M Belcher
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27607, USA
| |
Collapse
|
16
|
Du Y, Xu J, Han T, Jiang Z, Zhang Y, Li J, Chen X, Zhu S. Albumin-seeking dyes with adjustable assemblies in situ enable programmable imaging windows and targeting tumor imaging. Theranostics 2024; 14:2675-2686. [PMID: 38773981 PMCID: PMC11103493 DOI: 10.7150/thno.92991] [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: 12/07/2023] [Accepted: 04/04/2024] [Indexed: 05/24/2024] Open
Abstract
Cyanine dyes are widely used organic probes for in vivo imaging due to their tunable fluorescence. They can form complexes with endogenous albumin, resulting in enhanced brightness and photostability. However, this binding is uncontrollable and irreversible, leading to considerable nonspecific background signals and unregulated circulation time. Methods: Here, we connect varying numbers of 4-(4-iodophenyl) butanoic acid (IP) as albumin-binding moieties (ABM) to the cyanine dye, enabling dynamic and controllable binding with albumin. Meanwhile, we provide a blocking method to completely release the dye from covalent capture with albumin, resulting in specific targeting fluorescence. Furthermore, we evaluate the pharmacokinetics and tumor targeting of the developed dyes. Results: The engineered dyes can dynamically and selectively bind with multiple albumins to change the in situ size of assemblies and circulation time, providing programmable regulation over the imaging time window. The nucleophilic substitution of meso-Cl with water-soluble amino acids or targeting peptides for IP-engineered dye further addresses the nonspecific signals caused by albumin, allowing for adjustable angiography time and efficient tumor targeting. Conclusion: This study rationalizes the binding modes of dyes and proteins, applicable to a wide range of near-infrared (NIR) dyes for improving their in vivo molecular imaging.
Collapse
Affiliation(s)
- Yijing Du
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Jiajun Xu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Tianyang Han
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Zijian Jiang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
| | - Yuewei Zhang
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P.R. China
| | - Jia Li
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, 117597, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117609, Singapore
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| |
Collapse
|
17
|
Gao X, Kaluarachchi H, Zhang Y, Hwang S, Hannoush RN. A phage-displayed disulfide constrained peptide discovery platform yields novel human plasma protein binders. PLoS One 2024; 19:e0299804. [PMID: 38547072 PMCID: PMC10977726 DOI: 10.1371/journal.pone.0299804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/15/2024] [Indexed: 04/02/2024] Open
Abstract
Disulfide constrained peptides (DCPs) show great potential as templates for drug discovery. They are characterized by conserved cysteine residues that form intramolecular disulfide bonds. Taking advantage of phage display technology, we designed and generated twenty-six DCP phage libraries with enriched molecular diversity to enable the discovery of ligands against disease-causing proteins of interest. The libraries were designed based on five DCP scaffolds, namely Momordica charantia 1 (Mch1), gurmarin, Asteropsin-A, antimicrobial peptide-1 (AMP-1), and potato carboxypeptidase inhibitor (CPI). We also report optimized workflows for screening and producing synthetic and recombinant DCPs. Examples of novel DCP binders identified against various protein targets are presented, including human IgG Fc, serum albumin, vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor (PDGF). We identified DCPs against human IgG Fc and serum albumin with sub-micromolar affinity from primary panning campaigns, providing alternative tools for potential half-life extension of peptides and small protein therapeutics. Overall, the molecular diversity of the DCP scaffolds included in the designed libraries, coupled with their distinct biochemical and biophysical properties, enables efficient and robust identification of de novo binders to drug targets of therapeutic relevance.
Collapse
Affiliation(s)
- Xinxin Gao
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Harini Kaluarachchi
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| | - Yingnan Zhang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Biological Chemistry, Genentech, South San Francisco, California, United States of America
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| |
Collapse
|
18
|
De Felice S, Romanyuk Z, Chinellato M, Zoia G, Linciano S, Kumada Y, Pardon E, Steyaert J, Angelini A, Cendron L. Crystal structure of human serum albumin in complex with megabody reveals unique human and murine cross-reactive binding site. Protein Sci 2024; 33:e4887. [PMID: 38152025 PMCID: PMC10804666 DOI: 10.1002/pro.4887] [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: 07/31/2023] [Revised: 11/22/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
The pharmacokinetic properties of small biotherapeutics can be enhanced via conjugation to cross-reactive albumin-binding ligands in a process that improves their safety and accelerates testing through multiple pre-clinical animal models. In this context, the small and stable heavy-chain-only nanobody NbAlb1, capable of binding both human and murine albumin, has recently been successfully applied to improve the stability and prolong the in vivo plasma residence time of multiple small therapeutic candidates. Despite its clinical efficacy, the mechanism of cross-reactivity of NbAlb1 between human and murine serum albumins has not yet been investigated. To unveil the molecular basis of such an interaction, we solved the crystal structure of human serum albumin (hSA) in complex with NbAlb1. The structure was obtained by harnessing the unique features of a megabody chimeric protein, comprising NbAlb1 grafted onto a modified version of the circularly permutated and bacterial-derived protein HopQ. This structure showed that NbAlb1 contacts a yet unexplored binding site located in the peripheral region of domain II that is conserved in both human and mouse serum albumin proteins. Furthermore, we show that the binding of NbAlb1 to both serum albumin proteins is retained even at acidic pH levels, thus explaining its extended in vivo half-life. The elucidation of the molecular basis of NbAlb1 cross-reactivity to human and murine albumins might guide the design of novel nanobodies with broader reactivity toward a larger panel of serum albumins, thus facilitating the pre-clinical and clinical phases in humans.
Collapse
Affiliation(s)
| | - Zhanna Romanyuk
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
| | | | - Giulia Zoia
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
| | - Sara Linciano
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
| | - Yoichi Kumada
- Department of Functional Chemistry and EngineeringKyoto Institute of TechnologyKyotoJapan
| | - Els Pardon
- VIB‐VUB Center for Structural Biology, VIBBrusselsBelgium
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB)BrusselsBelgium
| | - Jan Steyaert
- VIB‐VUB Center for Structural Biology, VIBBrusselsBelgium
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB)BrusselsBelgium
| | - Alessandro Angelini
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
- European Centre for Living Technology (ECLT), Ca’ BottacinVeniceItaly
| | | |
Collapse
|
19
|
Chen LZ, Roos D, Philip E, Werth EG, Kostuk S, Yu H, Fuchs H. A Comprehensive Immunocapture-LC-MS/MS Bioanalytical Approach in Support of a Biotherapeutic Ocular PK Study. Pharmaceuticals (Basel) 2024; 17:193. [PMID: 38399408 PMCID: PMC10893151 DOI: 10.3390/ph17020193] [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: 11/07/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
BI-X, a therapeutic protein under development for the treatment of human ocular disease via intravitreal administration, binds to its therapeutic targets and endogenous albumin in the vitreous humor. A monkey ocular pharmacokinetic (PK) study following BI-X administration was conducted to measure drug and albumin levels in plasma, the vitreous humor, the aqueous humor, and retina tissue at various timepoints post-dose. A comprehensive bioanalytical approach was implemented in support of this study. Five immunocapture-LC-MS/MS assays were developed and qualified for quantitating BI-X in different matrices, while ELISA was used for albumin measurement. Immunocapture at the protein or peptide level was evaluated to achieve adequate assay sensitivity. Drug and albumin assays were applied for the analysis of the monkey study samples.
Collapse
Affiliation(s)
- Lin-Zhi Chen
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA (E.P.); (S.K.)
| | - David Roos
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA (E.P.); (S.K.)
| | - Elsy Philip
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA (E.P.); (S.K.)
| | - Emily G. Werth
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA (E.P.); (S.K.)
| | - Stephanie Kostuk
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA (E.P.); (S.K.)
| | - Hongbin Yu
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA (E.P.); (S.K.)
| | - Holger Fuchs
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany;
| |
Collapse
|
20
|
Bisht T, Adhikari A, Patil S, Dhoundiyal S. Bioconjugation Techniques for Enhancing Stability and Targeting Efficiency of Protein and Peptide Therapeutics. Curr Protein Pept Sci 2024; 25:226-243. [PMID: 37921168 DOI: 10.2174/0113892037268777231013154850] [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: 06/23/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 11/04/2023]
Abstract
Bioconjugation techniques have emerged as powerful tools for enhancing the stability and targeting efficiency of protein and peptide therapeutics. This review provides a comprehensive analysis of the various bioconjugation strategies employed in the field. The introduction highlights the significance of bioconjugation techniques in addressing stability and targeting challenges associated with protein and peptide-based drugs. Chemical and enzymatic bioconjugation methods are discussed, along with crosslinking strategies for covalent attachment and site-specific conjugation approaches. The role of bioconjugation in improving stability profiles is explored, showcasing case studies that demonstrate successful stability enhancement. Furthermore, bioconjugation techniques for ligand attachment and targeting are presented, accompanied by examples of targeted protein and peptide therapeutics. The review also covers bioconjugation approaches for prolonging circulation and controlled release, focusing on strategies to extend half-life, reduce clearance, and design-controlled release systems. Analytical characterization techniques for bioconjugates, including the evaluation of conjugation efficiency, stability, and assessment of biological activity and targeting efficiency, are thoroughly examined. In vivo considerations and clinical applications of bioconjugated protein and peptide therapeutics, including pharmacokinetic and pharmacodynamic considerations, as well as preclinical and clinical developments, are discussed. Finally, the review concludes with an overview of future perspectives, emphasizing the potential for novel conjugation methods and advanced targeting strategies to further enhance the stability and targeting efficiency of protein and peptide therapeutics.
Collapse
Affiliation(s)
- Tanuja Bisht
- Department of Pharmacy, Shree Dev Bhoomi Institute of Education, Science and Technology, Veer Madho Singh Bhandari Uttarakhand Technical University, Dehradun, Uttarakhand, India
| | - Anupriya Adhikari
- Department of Pharmacy, Shree Dev Bhoomi Institute of Education, Science and Technology, Veer Madho Singh Bhandari Uttarakhand Technical University, Dehradun, Uttarakhand, India
| | - Shivanand Patil
- Department of Pharmacy, Shree Dev Bhoomi Institute of Education, Science and Technology, Veer Madho Singh Bhandari Uttarakhand Technical University, Dehradun, Uttarakhand, India
| | - Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
21
|
Jindal S, Pedersen DV, Gera N, Chandler J, Patel R, Neill A, Cone J, Zhang Y, Yuan CX, Millman EE, Carlin D, Puffer B, Sheridan D, Andersen GR, Tamburini P. Characterization of the bispecific VHH antibody gefurulimab (ALXN1720) targeting complement component 5, and designed for low volume subcutaneous administration. Mol Immunol 2024; 165:29-41. [PMID: 38142486 DOI: 10.1016/j.molimm.2023.12.004] [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: 04/04/2023] [Revised: 09/29/2023] [Accepted: 12/09/2023] [Indexed: 12/26/2023]
Abstract
The bispecific antibody gefurulimab (also known as ALXN1720) was developed to provide patients with a subcutaneous treatment option for chronic disorders involving activation of the terminal complement pathway. Gefurulimab blocks the enzymatic cleavage of complement component 5 (C5) into the biologically active C5a and C5b fragments, which triggers activation of the terminal complement cascade. Heavy-chain variable region antigen-binding fragment (VHH) antibodies targeting C5 and human serum albumin (HSA) were isolated from llama immune-based libraries and humanized. Gefurulimab comprises an N-terminal albumin-binding VHH connected to a C-terminal C5-binding VHH via a flexible linker. The purified bispecific VHH antibody has the expected exact size by mass spectrometry and can be formulated at greater than 100 mg/mL. Gefurulimab binds tightly to human C5 and HSA with dissociation rate constants at pH 7.4 of 54 pM and 0.9 nM, respectively, and cross-reacts with C5 and serum albumin from cynomolgus monkeys. Gefurulimab can associate with C5 and albumin simultaneously, and potently inhibits the terminal complement activity from human serum initiated by any of the three complement pathways in Wieslab assays. Electron microscopy and X-ray crystallography revealed that the isolated C5-binding VHH recognizes the macroglobulin (MG) 4 and MG5 domains of the antigen and thereby is suggested to sterically prevent C5 binding to its activating convertase. Gefurulimab also inhibits complement activity supported by the rare C5 allelic variant featuring an R885H substitution in the MG7 domain. Taken together, these data suggest that gefurulimab may be a promising candidate for the potential treatment of complement-mediated disorders.
Collapse
Affiliation(s)
- Siddharth Jindal
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | | | - Nimish Gera
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Julian Chandler
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Rekha Patel
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Alyssa Neill
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Josh Cone
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Yuchun Zhang
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Chao-Xing Yuan
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Ellen E Millman
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Dan Carlin
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA.
| | - Bridget Puffer
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Douglas Sheridan
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics, Universitetsbyen 83, Aarhus University, Aarhus, Denmark
| | - Paul Tamburini
- Alexion, AstraZeneca Rare Disease, 100 College Street, New Haven, CT 06510, USA
| |
Collapse
|
22
|
Fakih HH, Tang Q, Summers A, Shin M, Buchwald JE, Gagnon R, Hariharan VN, Echeverria D, Cooper DA, Watts JK, Khvorova A, Sleiman HF. Dendritic amphiphilic siRNA: Selective albumin binding, in vivo efficacy, and low toxicity. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102080. [PMID: 38089931 PMCID: PMC10711485 DOI: 10.1016/j.omtn.2023.102080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/14/2023] [Indexed: 01/12/2024]
Abstract
Although an increasing number of small interfering RNA (siRNA) therapies are reaching the market, the challenge of efficient extra-hepatic delivery continues to limit their full therapeutic potential. Drug delivery vehicles and hydrophobic conjugates are being used to overcome the delivery bottleneck. Previously, we reported a novel dendritic conjugate that can be appended efficiently to oligonucleotides, allowing them to bind albumin with nanomolar affinity. Here, we explore the ability of this novel albumin-binding conjugate to improve the delivery of siRNA in vivo. We demonstrate that the conjugate binds albumin exclusively in circulation and extravasates to various organs, enabling effective gene silencing. Notably, we show that the conjugate achieves a balance between hydrophobicity and safety, as it significantly reduces the side effects associated with siRNA interactions with blood components, which are commonly observed in some hydrophobically conjugated siRNAs. In addition, it reduces siRNA monocyte uptake, which may lead to cytokine/inflammatory responses. This work showcases the potential of using this dendritic conjugate as a selective albumin binding handle for the effective and safe delivery of nucleic acid therapeutics. We envision that these properties may pave the way for new opportunities to overcome delivery hurdles of oligonucleotides in future applications.
Collapse
Affiliation(s)
- Hassan H. Fakih
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Qi Tang
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ashley Summers
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Minwook Shin
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- College of Pharmacy, Sookmyung Women’s University, Yongsan-gu, Seoul, Korea
| | - Julianna E. Buchwald
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Rosemary Gagnon
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Vignesh N. Hariharan
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dimas Echeverria
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - David A. Cooper
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jonathan K. Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hanadi F. Sleiman
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
| |
Collapse
|
23
|
Starnes HM, Jackson TW, Rock KD, Belcher SM. Quantitative Cross-Species Comparison of Serum Albumin Binding of Per- and Polyfluoroalkyl Substances from Five Structural Classes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566613. [PMID: 38014292 PMCID: PMC10680784 DOI: 10.1101/2023.11.10.566613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a class of over 8,000 chemicals that are persistent, bioaccumulative, and toxic to humans, livestock, and wildlife. Serum protein binding affinity is instrumental in understanding PFAS toxicity, yet experimental binding data is limited to only a few PFAS congeners. Previously, we demonstrated the usefulness of a high-throughput, in vitro differential scanning fluorimetry assay for determination of relative binding affinities of human serum albumin for 24 PFAS congeners from 6 chemical classes. In the current study, we used this differential scanning fluorimetry assay to comparatively examine differences in human, bovine, porcine, and rat serum albumin binding of 8 structurally informative PFAS congeners from 5 chemical classes. With the exception of the fluorotelomer alcohol 1H,1H,2H,2H-perfluorooctanol (6:2 FTOH), each PFAS congener bound by human serum albumin was also bound by bovine, porcine, and rat serum albumin. The critical role of the charged functional headgroup in albumin binding was supported by the inability of serum albumin of each species tested to bind 6:2 FTOH. Significant interspecies differences in serum albumin binding affinities were identified for each of the bound PFAS congeners. Relative to human albumin, perfluoroalkyl carboxylic and sulfonic acids were bound with greater affinity by porcine and rat serum albumin, and perfluoroalkyl ether congeners bound with lower affinity to porcine and bovine serum albumin. These comparative affinity data for PFAS binding by serum albumin from human, experimental model and livestock species reduce critical interspecies uncertainty and improve accuracy of predictive toxicity assessments for PFAS.
Collapse
Affiliation(s)
- Hannah M. Starnes
- Department of Biological Sciences, North Carolina State University, 127 David Clark Labs Campus Box 7617, Raleigh, NC 27607, USA
| | - Thomas W. Jackson
- Department of Biological Sciences, North Carolina State University, 127 David Clark Labs Campus Box 7617, Raleigh, NC 27607, USA
- Current address: Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Kylie D. Rock
- Department of Biological Sciences, North Carolina State University, 127 David Clark Labs Campus Box 7617, Raleigh, NC 27607, USA
- Current address: Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - Scott M. Belcher
- Department of Biological Sciences, North Carolina State University, 127 David Clark Labs Campus Box 7617, Raleigh, NC 27607, USA
| |
Collapse
|
24
|
Takeuchi T. Structural, nonclinical, and clinical features of ozoralizumab: A novel tumour necrosis factor inhibitor. Mod Rheumatol 2023; 33:1059-1067. [PMID: 37185766 DOI: 10.1093/mr/road038] [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: 01/05/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023]
Abstract
Tumour necrosis factor (TNF) inhibitors are currently the most widely used biological agents to treat rheumatoid arthritis. Ozoralizumab (OZR), a novel TNF inhibitor, is an antibody using variable heavy-chain domains of heavy-chain antibody (VHHs) and became the first VHH drug approved for the treatment of rheumatoid arthritis in September 2022. VHHs isolated from camelid heavy-chain antibodies can bind antigens with a single molecule. OZR is a trivalent VHH that consists of two anti-human TNFα VHHs and one anti-human serum albumin (anti-HSA) VHH. This review summarizes OZR's unique structural characteristics and nonclinical and clinical data. The clinical data outline the pharmacokinetics, efficacy, relationship between efficacy and pharmacokinetics, and safety of OZR, focusing on a Phase II/III confirmatory study (OHZORA trial).
Collapse
Affiliation(s)
- Tsutomu Takeuchi
- Saitama Medical University, Saitama, Japan
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
25
|
Kildemoes RJ, Backeljauw PF, Højby M, Blair JC, Miller BS, Mori J, Lyauk YK. Model-Based Analysis of IGF-I Response, Dosing, and Monitoring for Once-Weekly Somapacitan in Children With GH Deficiency. J Endocr Soc 2023; 7:bvad115. [PMID: 37818403 PMCID: PMC10561011 DOI: 10.1210/jendso/bvad115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Indexed: 10/12/2023] Open
Abstract
Context Growth hormone (GH) replacement therapy improves longitudinal growth and adult height in children with GH deficiency (GHD). GH stimulates insulin-like growth factor (IGF)-I release, the biomarker used for monitoring GH activity during treatment. Objective This study aims to provide model-based insights into the dose-IGF-I responses of once-weekly somapacitan, a novel long-acting GH, compared with daily GH in children with GHD. Methods Analyses included dosing information and 1473 pharmacokinetic samples from 210 somapacitan-treated pediatric patients with GHD across 3 trials, including phase 1 (NCT01973244), phase 2 (NCT02616562; REAL 3), and phase 3 (NCT03811535; REAL 4), as well as 1381 IGF-I samples from 186 patients with GHD treated with somapacitan in REAL 3 and REAL 4. Pharmacokinetic/pharmacodynamic modeling to characterize somapacitan dose-IGF-I response and predict the response to dosing day changes. Results Relationships were established between somapacitan dose, exposure, change from baseline IGF-I SD score (SDS), and height velocity (HV). A linear model permitted the development of a tool to calculate estimated average weekly IGF-I exposure from a single IGF-I sample obtained at any time within the somapacitan dosing interval at steady state. In practice, the use of this tool requires knowledge of somapacitan injection timing relative to IGF-I sample collection timing. IGF-I SDS simulations support flexible dosing day changes while maintaining at least 4 days between doses. Conclusion We characterized the dose-IGF-I response of somapacitan in children with GHD. To support physicians in IGF-I monitoring, we present a practical guide about expected weekly average IGF-I concentrations in these patients and provide insights on dosing day flexibility.
Collapse
Affiliation(s)
| | - Philippe F Backeljauw
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Michael Højby
- Clinical Drug Development, Novo Nordisk A/S, Søborg 2860, Denmark
| | - Joanne C Blair
- Department of Endocrinology, Alder Hey Children's NHS Foundation Trust, Liverpool L14 5AB, UK
| | - Bradley S Miller
- Division of Pediatric Endocrinology, University of Minnesota Medical School, MHealth Fairview Masonic Children’s Hospital, Minneapolis, MN 55454, USA
| | - Jun Mori
- Division of Pediatric Endocrinology and Metabolism, Children's Medical Center, Osaka City General Hospital, Osaka, 534-0021, Japan
| | - Yassine K Lyauk
- Clinical Drug Development, Novo Nordisk A/S, Søborg 2860, Denmark
| |
Collapse
|
26
|
Ullah A, Shin G, Lim SI. Human serum albumin binders: A piggyback ride for long-acting therapeutics. Drug Discov Today 2023; 28:103738. [PMID: 37591409 DOI: 10.1016/j.drudis.2023.103738] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/29/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023]
Abstract
Human serum albumin (HSA) is the most abundant protein in the blood and has desirable properties as a drug carrier. One of the most promising ways to exploit HSA as a carrier is to append an albumin-binding moiety (ABM) to a drug for in situ HSA binding upon administration. Nature- and library-derived ABMs vary in size, affinity, and epitope, differentially improving the pharmacokinetics of an appended drug. In this review, we evaluate the current state of knowledge regarding various aspects of ABMs and the unique advantages of ABM-mediated drug delivery. Furthermore, we discuss how ABMs can be specifically modulated to maximize potential benefits in clinical development.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, Khyber Pakhtunkhwa, Pakistan
| | - Goeun Shin
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Nbios Inc, 7, Jukheon-gil, Gangneung-si, Gangwon-do, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Marine BioResource Co., Ltd., 365, Sinseon-ro, Nam-gu, Busan 48548, Republic of Korea.
| |
Collapse
|
27
|
Wong JYK, Ekanayake AI, Kharchenko S, Kirberger SE, Qiu R, Kelich P, Sarkar S, Li J, Fernandez KX, Alvizo-Paez ER, Miao J, Kalhor-Monfared S, John JD, Kang H, Choi H, Nuss JM, Vederas JC, Lin YS, Macauley MS, Vukovic L, Pomerantz WCK, Derda R. Genetically encoded discovery of perfluoroaryl macrocycles that bind to albumin and exhibit extended circulation in vivo. Nat Commun 2023; 14:5654. [PMID: 37704629 PMCID: PMC10499988 DOI: 10.1038/s41467-023-41427-y] [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: 08/25/2022] [Accepted: 08/17/2023] [Indexed: 09/15/2023] Open
Abstract
Peptide-based therapeutics have gained attention as promising therapeutic modalities, however, their prevalent drawback is poor circulation half-life in vivo. In this paper, we report the selection of albumin-binding macrocyclic peptides from genetically encoded libraries of peptides modified by perfluoroaryl-cysteine SNAr chemistry, with decafluoro-diphenylsulfone (DFS). Testing of the binding of the selected peptides to albumin identified SICRFFC as the lead sequence. We replaced DFS with isosteric pentafluorophenyl sulfide (PFS) and the PFS-SICRFFCGG exhibited KD = 4-6 µM towards human serum albumin. When injected in mice, the concentration of the PFS-SICRFFCGG in plasma was indistinguishable from the reference peptide, SA-21. More importantly, a conjugate of PFS-SICRFFCGG and peptide apelin-17 analogue (N3-PEG6-NMe17A2) showed retention in circulation similar to SA-21; in contrast, apelin-17 analogue was cleared from the circulation after 2 min. The PFS-SICRFFC is the smallest known peptide macrocycle with a significant affinity for human albumin and substantial in vivo circulation half-life. It is a productive starting point for future development of compact macrocycles with extended half-life in vivo.
Collapse
Affiliation(s)
- Jeffrey Y K Wong
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Arunika I Ekanayake
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Serhii Kharchenko
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Steven E Kirberger
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ryan Qiu
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Payam Kelich
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Jiaqian Li
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Edgar R Alvizo-Paez
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Jiayuan Miao
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | | | - J Dwyer John
- Ferring Research Institute, San Diego, CA, 92121, USA
| | - Hongsuk Kang
- Quantum Intelligence Corp., 31F, One IFC, 10 Gukjegeumyung-ro, Yeongdeungpo-gu-Seoul, Republic of Korea
| | - Hwanho Choi
- Quantum Intelligence Corp., 31F, One IFC, 10 Gukjegeumyung-ro, Yeongdeungpo-gu-Seoul, Republic of Korea
| | - John M Nuss
- Ferring Research Institute, San Diego, CA, 92121, USA
| | - John C Vederas
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Lela Vukovic
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX, 79968, USA
| | | | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada.
| |
Collapse
|
28
|
Boinapally S, Alati S, Jiang Z, Yan Y, Lisok A, Singh R, Lofland G, Minn I, Hobbs RF, Pomper MG, Banerjee SR. Preclinical Evaluation of a New Series of Albumin-Binding 177Lu-Labeled PSMA-Based Low-Molecular-Weight Radiotherapeutics. Molecules 2023; 28:6158. [PMID: 37630410 PMCID: PMC10459686 DOI: 10.3390/molecules28166158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-based low-molecular-weight agents using beta(β)-particle-emitting radiopharmaceuticals is a new treatment paradigm for patients with metastatic castration-resistant prostate cancer. Although results have been encouraging, there is a need to improve the tumor residence time of current PSMA-based radiotherapeutics. Albumin-binding moieties have been used strategically to enhance the tumor uptake and retention of existing PSMA-based investigational agents. Previously, we developed a series of PSMA-based, β-particle-emitting, low-molecular-weight compounds. From this series, 177Lu-L1 was selected as the lead agent because of its reduced off-target radiotoxicity in preclinical studies. The ligand L1 contains a PSMA-targeting Lys-Glu urea moiety with an N-bromobenzyl substituent in the ε-amino group of Lys. Here, we structurally modified 177Lu-L1 to improve tumor targeting using two known albumin-binding moieties, 4-(p-iodophenyl) butyric acid moiety (IPBA) and ibuprofen (IBU), and evaluated the effects of linker length and composition. Six structurally related PSMA-targeting ligands (Alb-L1-Alb-L6) were synthesized based on the structure of 177Lu-L1. The ligands were assessed for in vitro binding affinity and were radiolabeled with 177Lu following standard protocols. All 177Lu-labeled analogs were studied in cell uptake and selected cell efficacy studies. In vivo pharmacokinetics were investigated by conducting tissue biodistribution studies for 177Lu-Alb-L2-177Lu-Alb-L6 (2 h, 24 h, 72 h, and 192 h) in male NSG mice bearing human PSMA+ PC3 PIP and PSMA- PC3 flu xenografts. Preliminary therapeutic ratios of the agents were estimated from the area under the curve (AUC0-192h) of the tumors, blood, and kidney uptake values. Compounds were obtained in >98% radiochemical yields and >99% purity. PSMA inhibition constants (Kis) of the ligands were in the ≤10 nM range. The long-linker-based agents, 177Lu-Alb-L4 and 177Lu-Alb-L5, displayed significantly higher tumor uptake and retention (p < 0.001) than the short-linker-bearing 177Lu-Alb-L2 and 177Lu-Alb-L3 and a long polyethylene glycol (PEG) linker-bearing agent, 177Lu-Alb-L6. The area under the curve (AUC0-192h) of the PSMA+ PC3 PIP tumor uptake of 177Lu-Alb-L4 and 177Lu-Alb-L5 were >4-fold higher than 177Lu-Alb-L2, 177Lu-Alb-L3, and 177Lu-Alb-L6, respectively. Also, the PSMA+ PIP tumor uptake (AUC0-192h) of 177Lu-Alb-L2 and 177Lu-Alb-L3 was ~1.5-fold higher than 177Lu-Alb-L6. However, the lowest blood AUC0-192h and kidney AUC0-192h were associated with 177Lu-Alb-L6 from the series. Consequently, 177Lu-Alb-L6 displayed the highest ratios of AUC(tumor)-to-AUC(blood) and AUC(tumor)-to-AUC(kidney) values from the series. Among the other agents, 177Lu-Alb-L4 demonstrated a nearly similar ratio of AUC(tumor)-to-AUC(blood) as 177Lu-Alb-L6. The tumor-to-blood ratio was the dose-limiting therapeutic ratio for all of the compounds. Conclusions: 177Lu-Alb-L4 and 177Lu-Alb-L6 showed high tumor uptake in PSMA+ tumors and tumor-to-blood ratios. The data suggest that linker length and composition can be modulated to generate an optimized therapeutic agent.
Collapse
Affiliation(s)
- Srikanth Boinapally
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Zirui Jiang
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Alla Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Gabriela Lofland
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Robert F. Hobbs
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
29
|
Cerofolini L, Vasa K, Bianconi E, Salobehaj M, Cappelli G, Bonciani A, Licciardi G, Pérez-Ràfols A, Padilla-Cortés L, Antonacci S, Rizzo D, Ravera E, Viglianisi C, Calderone V, Parigi G, Luchinat C, Macchiarulo A, Menichetti S, Fragai M. Combining Solid-State NMR with Structural and Biophysical Techniques to Design Challenging Protein-Drug Conjugates. Angew Chem Int Ed Engl 2023; 62:e202303202. [PMID: 37276329 DOI: 10.1002/anie.202303202] [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: 03/13/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
Several protein-drug conjugates are currently being used in cancer therapy. These conjugates rely on cytotoxic organic compounds that are covalently attached to the carrier proteins or that interact with them via non-covalent interactions. Human transthyretin (TTR), a physiological protein, has already been identified as a possible carrier protein for the delivery of cytotoxic drugs. Here we show the structure-guided development of a new stable cytotoxic molecule based on a known strong binder of TTR and a well-established anticancer drug. This example is used to demonstrate the importance of the integration of multiple biophysical and structural techniques, encompassing microscale thermophoresis, X-ray crystallography and NMR. In particular, we show that solid-state NMR has the ability to reveal effects caused by ligand binding which are more easily relatable to structural and dynamical alterations that impact the stability of macromolecular complexes.
Collapse
Affiliation(s)
- Linda Cerofolini
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Kristian Vasa
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Elisa Bianconi
- Department of Pharmaceutical Sciences, University of Perugia, Via Fabretti n.48, 06123, Perugia, Italy
| | - Maria Salobehaj
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Giulia Cappelli
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Alice Bonciani
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Giulia Licciardi
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Anna Pérez-Ràfols
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Giotto Biotech s.r.l, Sesto Fiorentino, Via della Madonna del Piano 6, 50019, Florence, Italy
| | - Luis Padilla-Cortés
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Sabrina Antonacci
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Domenico Rizzo
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Enrico Ravera
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Caterina Viglianisi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Vito Calderone
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Giacomo Parigi
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Claudio Luchinat
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
- Giotto Biotech s.r.l, Sesto Fiorentino, Via della Madonna del Piano 6, 50019, Florence, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Via Fabretti n.48, 06123, Perugia, Italy
| | - Stefano Menichetti
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Marco Fragai
- Magnetic Resonance Centre (CERM), University of Florence, Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
30
|
Huan T, Guan B, Li H, Tu X, Zhang C, Tang B. Principles and current clinical landscape of NK cell engaging bispecific antibody against cancer. Hum Vaccin Immunother 2023; 19:2256904. [PMID: 37772505 PMCID: PMC10543353 DOI: 10.1080/21645515.2023.2256904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/05/2023] [Indexed: 09/30/2023] Open
Abstract
Monoclonal antibody-based targeted therapies have greatly improved treatment options for patients by binding to the innate immune system. However, the long-term efficacy of such antibodies is limited by mechanisms of drug resistance. Over the last 50 years, with advances in protein engineering technology, more and more bispecific antibody (bsAb) platforms have been engineered to meet diverse clinical needs. Bispecific NK cell engagers (BiKEs) or tri-specific NK cell engagers (TriKEs) allow for direct targeting of immune cells to tumors, and therefore resistance and serious adverse effects are greatly reduced. Many preclinical and clinical trials are currently underway, depicting the promise of antibody-based natural killer cell engager therapeutics. In this review, we compile worldwide efforts to explore the involvement of NK cells in bispecific antibodies. With a particular emphasis on lessons learned, we focus on preclinical and clinical studies in malignancies and discuss the reasons for the limited success of NK-cell engagers against solid tumors, offering plausible new ideas for curing some advanced cancers shortly.
Collapse
Affiliation(s)
- Tian Huan
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bugao Guan
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Hongbo Li
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Xiu Tu
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Chi Zhang
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Bin Tang
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
- Department of Central Laboratory, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| |
Collapse
|
31
|
Song W, Wei W, Lan X, Cai W. Albumin binding improves nanobody pharmacokinetics for dual-modality PET/NIRF imaging of CEACAM5 in colorectal cancer models. Eur J Nucl Med Mol Imaging 2023; 50:2591-2594. [PMID: 37191678 PMCID: PMC10330897 DOI: 10.1007/s00259-023-06266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Affiliation(s)
- Wenyu Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
32
|
Pires IS, Berthiaume F, Palmer AF. Engineering Therapeutics to Detoxify Hemoglobin, Heme, and Iron. Annu Rev Biomed Eng 2023; 25:1-21. [PMID: 37289555 DOI: 10.1146/annurev-bioeng-081622-031203] [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] [Indexed: 06/10/2023]
Abstract
Hemolysis (i.e., red blood cell lysis) can increase circulatory levels of cell-free hemoglobin (Hb) and its degradation by-products, namely heme (h) and iron (Fe). Under homeostasis, minor increases in these three hemolytic by-products (Hb/h/Fe) are rapidly scavenged and cleared by natural plasma proteins. Under certain pathophysiological conditions, scavenging systems become overwhelmed, leading to the accumulation of Hb/h/Fe in the circulation. Unfortunately, these species cause various side effects such as vasoconstriction, hypertension, and oxidative organ damage. Therefore, various therapeutics strategies are in development, ranging from supplementation with depleted plasma scavenger proteins to engineered biomimetic protein constructs capable of scavenging multiple hemolytic species. In this review, we briefly describe hemolysis and the characteristics of the major plasma-derived protein scavengers of Hb/h/Fe. Finally, we present novel engineering approaches designed to address the toxicity of these hemolytic by-products.
Collapse
Affiliation(s)
- Ivan S Pires
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA;
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA;
| |
Collapse
|
33
|
Takeuchi T, Chino Y, Kawanishi M, Nakanishi M, Watase H, Mano Y, Sato Y, Uchida S, Tanaka Y. Efficacy and pharmacokinetics of ozoralizumab, an anti-TNFα NANOBODY ® compound, in patients with rheumatoid arthritis: 52-week results from the OHZORA and NATSUZORA trials. Arthritis Res Ther 2023; 25:60. [PMID: 37055803 PMCID: PMC10099673 DOI: 10.1186/s13075-023-03036-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/24/2023] [Indexed: 04/15/2023] Open
Abstract
INTRODUCTION Ozoralizumab (OZR), a tumor necrosis factor alpha (TNFα) inhibitor, is a NANOBODY® compound that binds to TNFα and human serum albumin. The main objective of this study was to analyze the pharmacokinetics (PK) of the drug and its correlation with clinical efficacy in patients with rheumatoid arthritis (RA). METHODS Efficacy data were analyzed from the OHZORA trial, in which OZR 30 or 80 mg was administered to Japanese patients with RA at 4-week intervals for 52 weeks in combination with methotrexate (MTX; n = 381), and the NATSUZORA trial, in which OZR 30 or 80 mg was administered without concomitant MTX (n = 140). Effects of patient baseline characteristics and anti-drug antibodies (ADAs) on the PK and efficacy of OZR were investigated, and a post hoc analysis of PK effects on drug efficacy was performed. RESULTS The maximum plasma concentration (Cmax) was reached in 6 days in both the 30 and 80 mg groups, with an elimination half-life of 18 days. The Cmax and area under the plasma concentration-time curve increased in a dose-dependent manner, and the trough concentration reached steady state by week 16. The exposure of OZR correlated negatively with patient body weight and was not affected by other patient baseline characteristics. Effects of ADAs on the exposure and efficacy of OZR were limited in both trials. However, antibodies that neutralize the binding to TNFα had some effect on the exposure and efficacy of OZR in the NATSUZORA trial. The receiver operating characteristic analysis of the effect of trough concentration on the American College of Rheumatology 20% and 50% improvement rates was retrospectively performed, and a cutoff trough concentration of approximately 1 μg/mL at week 16 was obtained in both trials. The efficacy indicators in the subgroup with trough concentration ≥ 1 μg/mL were higher than those in the < 1 μg/mL subgroup at week 16, while no clear cutoff was obtained at week 52 in both trials. CONCLUSIONS OZR showed a long half-life and favorable PK properties. A post hoc analysis suggested sustained efficacy independent of trough concentration by subcutaneous administration of OZR 30 mg at 4-week intervals for 52 weeks. TRIAL REGISTRATION JapicCTI, OHZORA trial: JapicCTI-184029, registration date July 9, 2018; NATSUZORA trial: JapicCTI-184031, registration date July 9, 2018.
Collapse
Affiliation(s)
- Tsutomu Takeuchi
- Keio University School of Medicine, Tokyo, Japan.
- Saitama Medical University, Saitama, Japan.
| | | | | | | | | | - Yoko Mano
- Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Yuri Sato
- Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | | | - Yoshiya Tanaka
- University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
34
|
Praveen P, Wang C, Handley TNG, Wu H, Samuel CS, Bathgate RAD, Hossain MA. A Lipidated Single-B-Chain Derivative of Relaxin Exhibits Improved In Vitro Serum Stability without Altering Activity. Int J Mol Sci 2023; 24:ijms24076616. [PMID: 37047588 PMCID: PMC10094921 DOI: 10.3390/ijms24076616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Human relaxin-2 (H2 relaxin) is therapeutically very important due to its strong anti-fibrotic, vasodilatory, and cardioprotective effects. Therefore, relaxin’s receptor, relaxin family peptide receptor 1 (RXFP1), is a potential target for the treatment of fibrosis and related disorders, including heart failure. H2 relaxin has a complex two-chain structure (A and B) and three disulfide bridges. Our laboratory has recently developed B7-33 peptide, a single-chain agonist based on the B-chain of H2 relaxin. However, the peptide B7-33 has a short circulation time in vitro in serum (t1/2 = ~6 min). In this study, we report structure-activity relationship studies on B7-33 utilizing different fatty-acid conjugations at different positions. We have shown that by fatty-acid conjugation with an appropriate spacer length, the in vitro half-life of B7-33 can be increased from 6 min to 60 min. In the future, the lead lipidated molecule will be studied in animal models to measure its PK/PD properties, which will lead to their pre-clinical applications.
Collapse
Affiliation(s)
- Praveen Praveen
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC 3168, Australia
| | - Thomas N. G. Handley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - Hongkang Wu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - Chrishan S. Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC 3168, Australia
| | - Ross A. D. Bathgate
- Department of Biochemistry and Pharmacology, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Mohammed Akhter Hossain
- Florey Department of Neuroscience and Mental, Florey Institute of Neuroscience and Mental Health, School of Chemistry, Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
35
|
Yong Joon Kim J, Sang Z, Xiang Y, Shen Z, Shi Y. Nanobodies: Robust miniprotein binders in biomedicine. Adv Drug Deliv Rev 2023; 195:114726. [PMID: 36754285 PMCID: PMC11725230 DOI: 10.1016/j.addr.2023.114726] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/30/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Variable domains of heavy chain-only antibodies (VHH), also known as nanobodies (Nbs), are monomeric antigen-binding domains derived from the camelid heavy chain-only antibodies. Nbs are characterized by small size, high target selectivity, and marked solubility and stability, which collectively facilitate high-quality drug development. In addition, Nbs are readily expressed from various expression systems, including E. coli and yeast cells. For these reasons, Nbs have emerged as preferred antibody fragments for protein engineering, disease diagnosis, and treatment. To date, two Nb-based therapies have been approved by the U.S. Food and Drug Administration (FDA). Numerous candidates spanning a wide spectrum of diseases such as cancer, immune disorders, infectious diseases, and neurodegenerative disorders are under preclinical and clinical investigation. Here, we discuss the structural features of Nbs that allow for specific, versatile, and strong target binding. We also summarize emerging technologies for identification, structural analysis, and humanization of Nbs. Our main focus is to review recent advances in using Nbs as a modular scaffold to facilitate the engineering of multivalent polymers for cutting-edge applications. Finally, we discuss remaining challenges for Nb development and envision new opportunities in Nb-based research.
Collapse
Affiliation(s)
- Jeffrey Yong Joon Kim
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine and Carnegie Mellon University, Pittsburgh, PA, USA
| | - Zhe Sang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA
| | - Yufei Xiang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA
| | - Zhuolun Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Shi
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA.
| |
Collapse
|
36
|
Szczukowski Ł, Maniewska J, Wiatrak B, Jawień P, Krzyżak E, Kotynia A, Marciniak A, Janeczek M, Redzicka A. Interactions of N-Mannich Bases of Pyrrolo[3,4- c]pyrrole with Artificial Models of Cell Membranes and Plasma Proteins, Evaluation of Anti-Inflammatory and Antioxidant Activity. MEMBRANES 2023; 13:349. [PMID: 36984737 PMCID: PMC10057445 DOI: 10.3390/membranes13030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
Despite the widespread and easy access to NSAIDs, effective and safe treatment of various inflammatory disorders is still a serious challenge because of the severe adverse effects distinctive to these drugs. The Mannich base derivatives of pyrrolo[3,4-c]pyrrole are potent, preferential COX-2 inhibitors with a COX-2/COX-1 inhibitory ratio better than meloxicam. Therefore, we chose the six most promising molecules and subjected them to further in-depth research. The current study presents the extensive biological, spectroscopic and in silico evaluation of the activity and physicochemical properties of pyrrolo[3,4-c]pyrrole derivatives. Aware of the advantages of dual COX-LOX inhibition, we investigated the 15-LOX inhibitory activity of these molecules. We also examined their antioxidant effect in several in vitro experiments in a protection and regeneration model. Furthermore, we defined how studied compounds interact with artificial models of cell membranes, which is extremely important for drugs administered orally with an intracellular target. The interactions and binding mode of the derivatives with the most abundant plasma proteins-human serum albumin and alpha-1-acid glycoprotein-are also described. Finally, we used computational techniques to evaluate their pharmacokinetic properties. According to the obtained results, we can state that pyrrolo[3,4-c]pyrrole derivatives are promising anti-inflammatory and antioxidant agents with potentially good membrane permeability.
Collapse
Affiliation(s)
- Łukasz Szczukowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Jadwiga Maniewska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland
| | - Paulina Jawień
- Department of Biostructure and Animal Physiology, Division of Animal Anatomy, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Kożuchowska 1, 51-631 Wroclaw, Poland
| | - Edward Krzyżak
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| | - Aleksandra Kotynia
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| | - Aleksandra Marciniak
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| | - Maciej Janeczek
- Department of Biostructure and Animal Physiology, Division of Animal Anatomy, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Kożuchowska 1, 51-631 Wroclaw, Poland
| | - Aleksandra Redzicka
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| |
Collapse
|
37
|
Interaction of Positively Charged Oligopeptides with Blood Plasma Proteins. Int J Mol Sci 2023; 24:ijms24032836. [PMID: 36769160 PMCID: PMC9918186 DOI: 10.3390/ijms24032836] [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: 11/21/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
In this project, we combine two areas of research, experimental characterization and molecular docking studies of the interaction of positively charged oligopeptides with crucial blood plasma proteins. The investigated peptides are rich in NH2 groups of amino acid side chains from Dap, Orn, Lys, and Arg residues, which are relevant in protein interaction. The peptides are 9- and 11-mer with the following sequences: (Lys-Dab-Dab-Gly-Orn-Pro-His-Lys-Arg-Lys-Dbt), (Lys-Dab-Ala-Gly-Orn-Pro-His-Lys-Arg), and (Lys-Dab-Dab-Gly-Orn-Pro-Phe(2-F)-Lys-Arg). The net charge of the compound strongly depends on the pH environment and it is an important aspect of protein binding. The studied oligopeptides exhibit therapeutic properties: anti-inflammatory activity and the capacity to diminish reactive oxygen species (ROS). Therefore, the mechanism of potential binding with blood plasma components is the next challenge. The binding interaction has been investigated under pseudo-physiological conditions with the main blood plasma proteins: albumin (BSA), α1-acid glycoprotein (AAG), and γ-globulin fraction (GGF). The biomolecular quenching constant (kq) and binding constant (Kb) were obtained by fluorescence spectroscopy at various temperatures. Simultaneously, the changes in the secondary structure of proteins were monitored by circular dichroism (CD) and infrared spectroscopy (IR) by quantity analysis. Moreover, molecular docking studies were conducted to estimate the binding affinity, the binding domain, and the chemical nature of these interactions. The results show that the investigated oligopeptides could be mainly transported by albumin, and the binding domain I is the most favored cavity. The BSA and GGF are able to form stable complexes with the studied compounds as opposed to AAG. The binding reactions are spontaneous processes. The highest binding constants were determined for Lys-Dab-Dab-Gly-Orn-Pro-His-Lys-Arg-Lys-Dbt peptide, in which the values of the binding constants Kb to BSA and GGF were 10.1 × 104 dm3mol-1 and 3.39 × 103 dm3mol-1, respectively. The positively charged surface of peptides participated in salt bridge interaction with proteins; however, hydrogen bonds were also formed. The secondary structure of BSA and GGF after contact with peptides was changed. A reduction in the α-helix structure was observed with an increase in the β-sheet and β-turn and random coil structures.
Collapse
|
38
|
Mamuti M, Chen W, Jiang X. Nanotechnology‐Assisted Immunoengineering for Cancer Vaccines. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Muhetaerjiang Mamuti
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| |
Collapse
|
39
|
Moro G, Liberi S, Vascon F, Linciano S, De Felice S, Fasolato S, Foresta C, De Toni L, Di Nisio A, Cendron L, Angelini A. Investigation of the Interaction between Human Serum Albumin and Branched Short-Chain Perfluoroalkyl Compounds. Chem Res Toxicol 2022; 35:2049-2058. [PMID: 36148994 PMCID: PMC9682524 DOI: 10.1021/acs.chemrestox.2c00211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The current trend dealing with the production of per- and polyfluoroalkyl substances (PFASs) involves the shifting toward branched short-chain fluorinated compounds known as new-generation PFASs. A key aspect to be clarified, to address the adverse health effects associated with the exposure to PFASs, is their binding mode to human serum albumin (hSA), the most abundant protein in plasma. In this study, we investigated the interaction between hSA and two representative branched short-chain PFASs, namely, HPFO-DA and C6O4. In-solution studies revealed that both compounds bind hSA with affinities and stoichiometries lower than that of the legacy long-chain perfluoroalkyl compound PFOA. Competition experiments using hSA-binding drugs with known site-selectivity revealed that both HPFO-DA and C6O4 bound to pockets located in subdomain IIIA. The crystal structure of hSA in complex with HPFO-DA unveiled the presence of two binding sites. The characterization and direct comparison of hSA interactions with new-generation PFASs may be key elements for the understanding of the toxicological impact of these compounds.
Collapse
Affiliation(s)
- Giulia Moro
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy.,Department of Medicine, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Stefano Liberi
- Department of Biology, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Filippo Vascon
- Department of Biology, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Sara Linciano
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Sofia De Felice
- Department of Biology, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Silvano Fasolato
- Department of Medicine, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Carlo Foresta
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Luca De Toni
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Andrea Di Nisio
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Laura Cendron
- Department of Biology, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy.,European Centre for Living Technology (ECLT), Ca' Bottacin, Dorsoduro 3911, Calle Crosera, 30123 Venice, Italy
| |
Collapse
|
40
|
Nakashima K, Iikuni S, Watanabe H, Ono M. Application of the Chelator-Based Clickable Radiotheranostic Platform to Moderate-Molecular-Weight Ligands. ACS Med Chem Lett 2022; 13:1642-1647. [PMID: 36262405 PMCID: PMC9575180 DOI: 10.1021/acsmedchemlett.2c00320] [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: 07/11/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022] Open
Abstract
We have reported that the chelator-based clickable radiotheranostic platform, ADIBO-DOTADG-ALB (ADA), has favorable properties as a radiotheranostic platform for low-molecular-weight ligands. In this study, we evaluated the applicability of ADA to moderate-molecular-weight ligands to expand the utility of the ADA platform. As a moderate-molecular-weight ligand, we selected exendin-4, a peptide-based agonist to glucagon-like peptide-1 receptor (GLP-1R). An exendin-4-incorporated ADA derivative, exendin-4-Cys40-triazole-DOTADG-ALB (EtDA), was radiolabeled with 111In by the conjugation of exendin-4-Cys40 azide to [111In]In-ADA. The click ligation of exendin-4-Cys40 azide to [111In]In-ADA was quantitatively completed in 10 min under ambient conditions. In the in vitro cell-binding assay and albumin-binding assay, [111In]In-EtDA showed strong binding to both a GLP-1R-expressing cell and albumin. In the biodistribution assay, [111In]In-EtDA showed markedly protracted tumor uptake, which was significantly decreased by the coinjection of exendin-4-Cys40. The single photon emission computed tomography (SPECT) image of [111In]In-EtDA visualized the tumor clearly. These results indicated the utility of [111In]In-EtDA as a radiotheranostic agent, suggesting the applicability of the ADA platform to moderate-molecular-weight ligands.
Collapse
Affiliation(s)
- Kazuma Nakashima
- Department of Patho-Functional Bioanalysis,
Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis,
Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis,
Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis,
Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
41
|
Xu L, Guan R, Yu B, Li Y, Liu H, Jiang Y. Fluorene methoxycarbonyl-PEG-deferoxamine conjugates "hitchhike" with albumin in situ for iron overload therapy. Int J Pharm 2022; 625:122136. [PMID: 36029994 DOI: 10.1016/j.ijpharm.2022.122136] [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: 06/29/2022] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 10/15/2022]
Abstract
Although deferoxamine (DFO) has been approved for the treatment the iron overloaded diseases, its clinical application is impeded by very short circulation time and its relating toxicity. In this work, the fluorene methoxycarbonyl (FMOC) for "albumin hitchhiking" was used to prolong the plasma circulation time of DFO and reduce toxicity. The designed FMOC-PEG-DFO conjugates were found to reversible bind to albumin and gradually release DFO in vivo. Herein, the FMOC-PEG1000-DFO conjugates could increase 30 times the blood circulation time of DFO with the improvement of the iron elimination efficacy. Meanwhile, the conjugates markedly reduced the cytotoxicity of DFO. Taken together, the result demonstrated the FMOC-PEG1000-DFO conjugates could be a potential therapeutic choice for iron-overload-related diseases.
Collapse
Affiliation(s)
- Linyi Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Rou Guan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Bohong Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Yicheng Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yiguo Jiang
- Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou 215153, China.
| |
Collapse
|
42
|
Shi X, Chen D, Liu G, Zhang H, Wang X, Wu Z, Wu Y, Yu F, Xu Q. Application of Elastin-Like Polypeptide in Tumor Therapy. Cancers (Basel) 2022; 14:cancers14153683. [PMID: 35954346 PMCID: PMC9367306 DOI: 10.3390/cancers14153683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Elastin-like Polypeptide (ELP) are widely applied in protein purification, drug delivery, tissue engineering, and even tumor therapy. Recent studies show that usage of ELP has made great progress in combination with peptide drugs or antibody drugs. The combination of ELP and photosensitizer in cancer therapy or imaging has made more progress and needs to be summarized. In this review, we summarize the specific application of ELP in cancer therapy, especially the latest developments in the combined use of ELP with photosensitizers. We seek to provide the most recent understanding of ELP and its new application in combination with Photosensitizer. Abstract Elastin-like polypeptides (ELPs) are stimulus-responsive artificially designed proteins synthesized from the core amino acid sequence of human tropoelastin. ELPs have good biocompatibility and biodegradability and do not systemically induce adverse immune responses, making them a suitable module for drug delivery. Design strategies can equip ELPs with the ability to respond to changes in temperature and pH or the capacity to self-assemble into nanoparticles. These unique tunable biophysicochemical properties make ELPs among the most widely studied biopolymers employed in protein purification, drug delivery, tissue engineering and even in tumor therapy. As a module for drug delivery and as a carrier to target tumor cells, the combination of ELPs with therapeutic drugs, antibodies and photo-oxidation molecules has been shown to result in improved pharmacokinetic properties (prolonged half-life, drug targeting, cell penetration and controlled release) while restricting the cytotoxicity of the drug to a confined infected site. In this review, we summarize the latest developments in the application methods of ELP employed in tumor therapy, with a focus on its conjugation with peptide drugs, antibodies and photosensitizers.
Collapse
Affiliation(s)
- Xianggang Shi
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; (X.S.); (D.C.); (Y.W.)
| | - Dongfeng Chen
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; (X.S.); (D.C.); (Y.W.)
| | - Guodong Liu
- Department of Gastroenterology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian 223800, China; (G.L.); (H.Z.); (X.W.)
| | - Hailing Zhang
- Department of Gastroenterology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian 223800, China; (G.L.); (H.Z.); (X.W.)
| | - Xiaoyan Wang
- Department of Gastroenterology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian 223800, China; (G.L.); (H.Z.); (X.W.)
| | - Zhi Wu
- Jiangsu Key Laboratory of High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China;
| | - Yan Wu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; (X.S.); (D.C.); (Y.W.)
| | - Feng Yu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; (X.S.); (D.C.); (Y.W.)
- Correspondence: (F.Y.); (Q.X.); Tel.: +86-139-5292-3250 (F.Y.); +86-159-5281-6017 (Q.X.)
| | - Qinggang Xu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; (X.S.); (D.C.); (Y.W.)
- Correspondence: (F.Y.); (Q.X.); Tel.: +86-139-5292-3250 (F.Y.); +86-159-5281-6017 (Q.X.)
| |
Collapse
|
43
|
Solomos K, Tarus V, Kotek V, Invernizzi G, Hoeg-Jensen T. Diboronate Fluorophore for the Measurement of l-Glucose and Other Carbohydrates and Its Interaction with Albumin. ACS OMEGA 2022; 7:24662-24668. [PMID: 35874210 PMCID: PMC9301969 DOI: 10.1021/acsomega.2c02519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
l-Glucose has recently been investigated as an artificial sweetener, but no facile method is established for the measurement of l-glucose. The commercial probe Eversense employs a fluorescent diboronate in a small device for the optical monitoring of d-glucose in people with diabetes. Being achiral, the Eversense probe should be able to detect l-glucose as well as native d-glucose, but the probe is designed for fixation under the skin, and our attempts to use the probe at laboratory conditions failed, as the probe was resetting when moved between compartments. We thus designed a water-soluble anthracene diboronate 8 similar to the fluorophore used in Eversense and found 8 to respond well to l-glucose and other carbohydrates and artificial sweeteners, thus enabling measurements of l-glucose with the limit of quantification of 12 μM. Notably, the fluorescent signal of diboronate 8 was largely quenched in buffers with the physiological concentration of albumin (0.5 mM), so the given analytical method would need more optimization to be useful for measuring l-glucose and other carbohydrates in blood samples. We suspect that other diboronate fluorophores from the literature may be similarly quenched if applied in the presence of albumin.
Collapse
Affiliation(s)
| | - Victoria Tarus
- Apigenex
s.r.o., Poděbradská 173/5, 190
00 Prague, Czech Republic
| | - Vladislav Kotek
- Apigenex
s.r.o., Poděbradská 173/5, 190
00 Prague, Czech Republic
| | - Gaetano Invernizzi
- Global
Research Technologies, Novo Nordisk A/S, Novo Nordisk Park H5.S.05, 2760 Maaloev, Denmark
| | - Thomas Hoeg-Jensen
- Global
Research Technologies, Novo Nordisk A/S, Novo Nordisk Park H5.S.05, 2760 Maaloev, Denmark
| |
Collapse
|
44
|
Palmitic Acid-Conjugated Radiopharmaceutical for Integrin αvβ3-Targeted Radionuclide Therapy. Pharmaceutics 2022; 14:pharmaceutics14071327. [PMID: 35890224 PMCID: PMC9321335 DOI: 10.3390/pharmaceutics14071327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is an emerging approach for patients with unresectable or metastatic tumors. Our previously optimized RGD peptide (3PRGD2) has excellent targeting specificity for a variety of integrin αvβ3/αvβ5-positive tumors and has been labeled with the therapeutic radionuclide [177Lu]LuCl3 for targeted radiotherapy of tumors. However, the rapid clearance of [177Lu]Lu-DOTA-3PRGD2 (177Lu-3PRGD2) in vivo requires two doses of 111 MBq/3 mCi to achieve effective tumor suppression, limiting its further clinical application. Albumin binders have been attached to drugs to facilitate binding to albumin in vivo to prolong the drug half-life in plasma and obtain long-term effects. In this study, we modified 3PRGD2 with albumin-binding palmitic acid (Palm-3PRGD2) and then radiolabeled Palm-3PRGD2 with 177Lu. [177Lu]Lu-DOTA-Palm-3PRGD2 (177Lu-Palm-3PRGD2) retained a specific binding affinity for integrin αvβ3/αvβ5, with an IC50 value of 5.13 ± 1.16 nM. Compared with 177Lu-3PRGD2, the 177Lu-Palm-3PRGD2 circulation time in blood was more than 6 times longer (slow half-life: 73.42 min versus 11.81 min), and the tumor uptake increased more than fivefold (21.34 ± 4.65 %IA/g and 4.11 ± 0.70 %IA/g at 12 h post-injection). Thus, the significant increase in tumor uptake and tumor retention resulted in enhanced efficacy of targeted radiotherapy, and tumor growth was completely inhibited by a single and relatively lowdose of 18.5 MBq/0.5 mCi. Thus, 177Lu-Palm-3PRGD2 shows great potential for clinical application.
Collapse
|
45
|
Meng L, Fang J, Zhao L, Wang T, Yuan P, Zhao Z, Zhuang R, Lin Q, Chen H, Chen X, Zhang X, Guo Z. Rational Design and Pharmacomodulation of Protein-Binding Theranostic Radioligands for Targeting the Fibroblast Activation Protein. J Med Chem 2022; 65:8245-8257. [PMID: 35658448 DOI: 10.1021/acs.jmedchem.1c02162] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The fibroblast activation protein (FAP), overexpressed on cancer-associated fibroblasts (CAFs), has become a valuable target for tumor diagnosis and therapy. However, most FAP-based radioligands show insufficient tumor uptake and retention. In this study, three novel albumin-binding FAP ligands (denoted as FSDD0I, FSDD1I, and FSDD3I) were labeled with 68Ga and 177Lu to overcome these limitations. Cell-based studies and molecular docking assays were performed to identify the specificity and protein-binding properties for FAP. Positron emission tomography (PET) scans in human hepatocellular carcinoma patient-derived xenografts (HCC-PDXs) animal models revealed longer blood retention of 68Ga-FSDD0I than 68Ga-FAPI-04, 68Ga-FSDD1I, and 68Ga-FSDD3I. Remarkably, 68Ga-FSDD3I had prominent tumor-to-nontarget (T/NT) ratios. The prominent tumor retention properties of 177Lu-FSDD0I in single photon emission computed tomography (SPECT) imaging and biodistribution studies were demonstrated. In summary, this study reports a proof-of-concept study of albumin-binding radioligands for FAP-targeted imaging and targeted radionuclide therapy (TRT).
Collapse
Affiliation(s)
- Lingxin Meng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jianyang Fang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Liang Zhao
- Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.,Department of Radiation Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Tingting Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Pu Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zuoquan Zhao
- Department of Nuclear Medicine, Cardiovascular Institute and FuWai Hospital, Chinese Academy of Medical Sciences, Beijing 100037, China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qin Lin
- Department of Radiation Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Haojun Chen
- Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
46
|
Linciano S, Moro G, Zorzi A, Angelini A. Molecular analysis and therapeutic applications of human serum albumin-fatty acid interactions. J Control Release 2022; 348:115-126. [PMID: 35643382 DOI: 10.1016/j.jconrel.2022.05.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/16/2022] [Accepted: 05/21/2022] [Indexed: 11/16/2022]
Abstract
Human serum albumin (hSA) is the major carrier protein for fatty acids (FAs) in plasma. Its ability to bind multiple FA moieties with moderate to high affinity has inspired the use of FA conjugation as a safe and natural platform to generate long-lasting therapeutics with enhanced pharmacokinetic properties and superior efficacy. In this frame, the choice of the FA is crucial and a comprehensive elucidation of the molecular interactions of FAs with hSA cannot be left out of consideration. To this intent, we report here a comparative analysis of the binding mode of different FA moieties with hSA. The choice among different albumin-binding FAs and how this influence the pharmacokinetics properties of a broad spectrum of therapeutic molecules will be discussed including a critical description of some clinically relevant FA conjugated therapeutics.
Collapse
Affiliation(s)
- Sara Linciano
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Giulia Moro
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; AXES Research Group, Department of Chemistry, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Alessandro Zorzi
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; European Centre for Living Technology (ECLT), Ca' Bottacin, Dorsoduro 3911, Calle Crosera, 30123 Venice, Italy.
| |
Collapse
|
47
|
Michot N, Guyochin A, Cinier M, Savignard C, Kitten O, Pascual MH, Pouzieux S, Ozoux ML, Verdier P, Vicat P, Dumas J. Albumin binding Nanofitins, a new scaffold to extend half-life of biologics - a case study with exenatide peptide. Peptides 2022; 152:170760. [PMID: 35150805 DOI: 10.1016/j.peptides.2022.170760] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 01/01/2023]
Abstract
A new strategy of peptide half-life extension has been evaluated. We investigated libraries of a small and very stable protein scaffold called Nanofitin, capable of high affinity for protein targets. We have identified Nanofitins targeting Human and mouse Serum Albumin, which could significantly improve the pharmacokinetics of an active associated peptide, mobilizing the patient's own albumin without external source. To demonstrate the impact of this approach on half-life extension, a genetic fusion of an Exenatide peptide with an Albumin Binding Nanofitin (ABNF) was performed. Specific activity of Exenatide-ABNF was measured and unaffected by the fusion. In vivo mice results provided convincing data (t½ of 8 min for Exenatide peptide compared to 20 h for Exenatide-ABNF) with sustained pharmacological activity over 3 days. This study constitutes a proof-of-concept of in vivo half-life extension of a biologic using an ABNF. Besides, the absence of cysteine in the Nanofitin scaffold, which is therefore devoid of structuring disulfide bonds, allows manufacturing in microbial cost effective systems.
Collapse
Affiliation(s)
- Nadine Michot
- Sanofi, Biologics Research, Vitry sur Seine, 94430, France
| | | | | | | | | | | | | | | | - Patrick Verdier
- Sanofi, Drug Safety & Animal Research, Alfortville 94430, France
| | - Pascale Vicat
- Sanofi, Drug Safety & Animal Research, Alfortville 94430, France
| | - Jacques Dumas
- Sanofi, Biologics Research, Vitry sur Seine, 94430, France
| |
Collapse
|
48
|
Hu H, Quintana J, Weissleder R, Parangi S, Miller M. Deciphering albumin-directed drug delivery by imaging. Adv Drug Deliv Rev 2022; 185:114237. [PMID: 35364124 PMCID: PMC9117484 DOI: 10.1016/j.addr.2022.114237] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/10/2022] [Accepted: 03/23/2022] [Indexed: 01/03/2023]
Abstract
Albumin is the most abundant plasma protein, exhibits extended circulating half-life, and its properties have long been exploited for diagnostics and therapies. Many drugs intrinsically bind albumin or have been designed to do so, yet questions remain about true rate limiting factors that govern albumin-based transport and their pharmacological impacts, particularly in advanced solid cancers. Imaging techniques have been central to quantifying - at a molecular and single-cell level - the impact of mechanisms such as phagocytic immune cell signaling, FcRn-mediated recycling, oncogene-driven macropinocytosis, and albumin-drug interactions on spatial albumin deposition and related pharmacology. Macroscopic imaging of albumin-binding probes quantifies vessel structure, permeability, and supports efficiently targeted molecular imaging. Albumin-based imaging in patients and animal disease models thus offers a strategy to understand mechanisms, guide drug development and personalize treatments.
Collapse
Affiliation(s)
- Huiyu Hu
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States; Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States; Department of General Surgery, Xiangya Hospital, Central South University, China
| | - Jeremy Quintana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States; Department of Systems Biology, Harvard Medical School, United States
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Miles Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States.
| |
Collapse
|
49
|
Jagusiak A, Chłopaś K, Zemanek G, Kościk I, Skorek P, Stopa B. Albumin Binds Doxorubicin via Self−Assembling Dyes as Specific Polymolecular Ligands. Int J Mol Sci 2022; 23:ijms23095033. [PMID: 35563426 PMCID: PMC9104453 DOI: 10.3390/ijms23095033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
Congo red (CR) type self–assembled ribbon–like structures (SRLS) were previously shown to interact with some proteins, including albumin. SRLS also complex with some drugs with a flat, ring–shaped structure with aromatic characteristics, intercalating them into their ribbon structure. The combination of interaction with proteins and drug binding by SRLS enables the use of such systems for immunotargeting. It is especially interesting in the case of chemotherapeutic agents. The present experiments aimed to show that the model carrier system composed of supramolecular albumin and Congo red efficiently binds doxorubicin (Dox) and that the drug can be released at reduced pH. The presented results come from the studies on such complexes differing in the molar ratio of CR to Dox. The following methods were used for the analysis: electrophoresis, dialysis, gel filtration, spectral analysis, and analysis of the size of the hydrodynamic radius using the dynamic light scattering method (DLS). The applied methods confirmed the formation of the CR–Dox complex, with large dimensions and changed properties compared with free CR. The presented results show that albumin binds both CR and its complex with Dox. Various CR–Dox molar ratios, 5:1, 2:1, and 1:1, were analyzed. The confirmation of the possibility of releasing the drug from the carriers thus formed was also obtained. The presented research is important due to the search for optimal solutions for the use of SRLS in drug immunotargeting, with particular emphasis on chemotherapeutic agents.
Collapse
Affiliation(s)
- Anna Jagusiak
- Chair of Medical Biochemistry, Faculty of Medicine, Medical College, Jagiellonian University, 31-034 Krakow, Poland; (G.Z.); (I.K.); (B.S.)
- Correspondence:
| | - Katarzyna Chłopaś
- Pulmonology and Allergology Clinical Department, University Hospital in Krakow, 30-688 Krakow, Poland;
| | - Grzegorz Zemanek
- Chair of Medical Biochemistry, Faculty of Medicine, Medical College, Jagiellonian University, 31-034 Krakow, Poland; (G.Z.); (I.K.); (B.S.)
| | - Izabela Kościk
- Chair of Medical Biochemistry, Faculty of Medicine, Medical College, Jagiellonian University, 31-034 Krakow, Poland; (G.Z.); (I.K.); (B.S.)
- Faculty of Biotechnology and Horticulture, University of Agriculture in Krakow, 31-425 Krakow, Poland
| | - Paweł Skorek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, 31-202 Krakow, Poland;
| | - Barbara Stopa
- Chair of Medical Biochemistry, Faculty of Medicine, Medical College, Jagiellonian University, 31-034 Krakow, Poland; (G.Z.); (I.K.); (B.S.)
| |
Collapse
|
50
|
Ng TSC, Hu H, Kronister S, Lee C, Li R, Gerosa L, Stopka SA, Burgenske DM, Khurana I, Regan MS, Vallabhaneni S, Putta N, Scott E, Matvey D, Giobbie-Hurder A, Kohler RH, Sarkaria JN, Parangi S, Sorger PK, Agar NYR, Jacene HA, Sullivan RJ, Buchbinder E, Mikula H, Weissleder R, Miller MA. Overcoming differential tumor penetration of BRAF inhibitors using computationally guided combination therapy. SCIENCE ADVANCES 2022; 8:eabl6339. [PMID: 35486732 PMCID: PMC9054019 DOI: 10.1126/sciadv.abl6339] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BRAF-targeted kinase inhibitors (KIs) are used to treat malignancies including BRAF-mutant non-small cell lung cancer, colorectal cancer, anaplastic thyroid cancer, and, most prominently, melanoma. However, KI selection criteria in patients remain unclear, as are pharmacokinetic/pharmacodynamic (PK/PD) mechanisms that may limit context-dependent efficacy and differentiate related drugs. To address this issue, we imaged mouse models of BRAF-mutant cancers, fluorescent KI tracers, and unlabeled drug to calibrate in silico spatial PK/PD models. Results indicated that drug lipophilicity, plasma clearance, faster target dissociation, and, in particular, high albumin binding could limit dabrafenib action in visceral metastases compared to other KIs. This correlated with retrospective clinical observations. Computational modeling identified a timed strategy for combining dabrafenib and encorafenib to better sustain BRAF inhibition, which showed enhanced efficacy in mice. This study thus offers principles of spatial drug action that may help guide drug development, KI selection, and combination.
Collapse
Affiliation(s)
- Thomas S. C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Huiyu Hu
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Stefan Kronister
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Chanseo Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Luca Gerosa
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Sylwia A. Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Ishaan Khurana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Michael S. Regan
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Sreeram Vallabhaneni
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Niharika Putta
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ella Scott
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Dylan Matvey
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter K. Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nathalie Y. R. Agar
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Heather A. Jacene
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryan J. Sullivan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Hannes Mikula
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Corresponding author.
| |
Collapse
|