1
|
Abdul-Rahman T, Roy P, Herrera-Calderón RE, Khidri FF, Omotesho QA, Rumide TS, Fatima M, Roy S, Wireko AA, Atallah O, Roy S, Amekpor F, Ghosh S, Agyigra IA, Horbas V, Teslyk T, Bumeister V, Papadakis M, Alexiou A. Extracellular vesicle-mediated drug delivery in breast cancer theranostics. Discov Oncol 2024; 15:181. [PMID: 38780753 PMCID: PMC11116322 DOI: 10.1007/s12672-024-01007-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Breast cancer (BC) continues to be a significant global challenge due to drug resistance and severe side effects. The increasing prevalence is alarming, requiring new therapeutic approaches to address these challenges. At this point, Extracellular vesicles (EVs), specifically small endosome-released nanometer-sized EVs (SEVs) or exosomes, have been explored by literature as potential theranostics. Therefore, this review aims to highlight the therapeutic potential of exosomes in BC, focusing on their advantages in drug delivery and their ability to mitigate metastasis. Following the review, we identified exosomes' potential in combination therapies, serving as miRNA carriers and contributing to improved anti-tumor effects. This is evident in clinical trials investigating exosomes in BC, which have shown their ability to boost chemotherapy efficacy by delivering drugs like paclitaxel (PTX) and doxorubicin (DOX). However, the translation of EVs into BC therapy is hindered by various challenges. These challenges include the heterogeneity of EVs, the selection of the appropriate parent cell, the loading procedures, and determining the optimal administration routes. Despite the promising therapeutic potential of EVs, these obstacles must be addressed to realize their benefits in BC treatment.
Collapse
Affiliation(s)
| | - Poulami Roy
- Department of Medicine, North Bengal Medical College and Hospital, Siliguri, India
| | - Ranferi Eduardo Herrera-Calderón
- Center for Research in Health Sciences (CICSA), Faculty of Medicine, Anahuac University North Campus, 52786, Huixquilucan, Mexico
| | | | | | | | | | - Sakshi Roy
- School of Medicine, Queens University Belfast, Northern Ireland, UK
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Subham Roy
- Hull York Medical School, University of York, York, UK
| | - Felix Amekpor
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Shankhaneel Ghosh
- Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan, Bhubaneswar, India
| | | | | | | | | | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Athanasios Alexiou
- University Centre for Research and Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India.
- Department of Research and Development, Funogen, 11741, Athens, Greece.
- Department of Research and Development, AFNP Med, 1030, Vienna, Austria.
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia.
| |
Collapse
|
2
|
Bidkar AP, Zerefa L, Yadav S, VanBrocklin HF, Flavell RR. Actinium-225 targeted alpha particle therapy for prostate cancer. Theranostics 2024; 14:2969-2992. [PMID: 38773983 PMCID: PMC11103494 DOI: 10.7150/thno.96403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
Targeted alpha particle therapy (TAT) has emerged as a promising strategy for the treatment of prostate cancer (PCa). Actinium-225 (225Ac), a potent alpha-emitting radionuclide, may be incorporated into targeting vectors, causing robust and in some cases sustained antitumor responses. The development of radiolabeling techniques involving EDTA, DOTA, DOTPA, and Macropa chelators has laid the groundwork for advancements in this field. At the forefront of clinical trials with 225Ac in PCa are PSMA-targeted TAT agents, notably [225Ac]Ac-PSMA-617, [225Ac]Ac-PSMA-I&T and [225Ac]Ac-J591. Ongoing investigations spotlight [225Ac]Ac-hu11B6, [225Ac]Ac-YS5, and [225Ac]Ac-SibuDAB, targeting hK2, CD46, and PSMA, respectively. Despite these efforts, hurdles in 225Ac production, daughter redistribution, and a lack of suitable imaging techniques hinder the development of TAT. To address these challenges and additional advantages, researchers are exploring alpha-emitting isotopes including 227Th, 223Ra, 211At, 213Bi, 212Pb or 149Tb, providing viable alternatives for TAT.
Collapse
Affiliation(s)
- Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
| | - Luann Zerefa
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
| | - Surekha Yadav
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA-94107, USA
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA-94107, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA-94107, USA
| |
Collapse
|
3
|
Liatsou I, Assefa B, Liyanage W, Surasinghe S, Nováková Z, Bařinka C, Gabrielson K, Raman V, Artemov D, Hapuarachchige S. Development and therapeutic evaluation of 5D3(CC-MLN8237) 3.2 antibody-theranostic conjugates for PSMA-positive prostate cancer therapy. Front Pharmacol 2024; 15:1385598. [PMID: 38751786 PMCID: PMC11094276 DOI: 10.3389/fphar.2024.1385598] [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: 02/13/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Prostate cancer (PC) is an aggressive cancer that can progress rapidly and eventually become castrate-resistant prostate cancer (CRPC). Stage IV metastatic castrate-resistant prostate cancer (mCRPC) is an incurable late-stage cancer type with a low 5-year overall survival rate. Targeted therapeutics such as antibody-drug conjugates (ADCs) based on high-affinity monoclonal antibodies and potent drugs conjugated via smart linkers are being developed for PC management. Conjugating further with in vitro or in vivo imaging agents, ADCs can be used as antibody-theranostic conjugates (ATCs) for diagnostic and image-guided drug delivery. In this study, we have developed a novel ATC for PSMA (+) PC therapy utilizing (a) anti-PSMA 5D3 mAb, (b) Aurora A kinase inhibitor, MLN8237, and (c) for the first time using tetrazine (Tz) and trans-cyclooctene (TCO) click chemistry-based conjugation linker (CC linker) in ADC development. The resulting 5D3(CC-MLN8237)3.2 was labeled with suitable fluorophores for in vitro and in vivo imaging. The products were characterized by SDS-PAGE, MALDI-TOF, and DLS and evaluated in vitro by optical imaging, flow cytometry, and WST-8 assay for cytotoxicity in PSMA (+/-) cells. Therapeutic efficacy was determined in human PC xenograft mouse models following a designed treatment schedule. After the treatment study animals were euthanized, and toxicological studies, complete blood count (CBC), blood clinical chemistry analysis, and H&E staining of vital organs were conducted to determine side effects and systemic toxicities. The IC50 values of 5D3(CC-MLN8237)3.2-AF488 in PSMA (+) PC3-PIP and PMSA (-) PC3-Flu cells are 8.17 nM and 161.9 nM, respectively. Pure MLN8237 shows 736.9 nM and 873.4 nM IC50 values for PC3-PIP and PC3-Flu cells, respectively. In vivo study in human xenograft mouse models confirmed high therapeutic efficacy of 5D3(CC-MLN8237)3.2-CF750 with significant control of PSMA (+) tumor growth with minimal systemic toxicity in the treated group compared to PSMA (-) treated and untreated groups. Approximately 70% of PSMA (+) PC3-PIP tumors did not exceed the threshold of the tumor size in the surrogate Kaplan-Meyer analysis. The novel ATC successfully controlled the growth of PSMA (+) tumors in preclinical settings with minimal systemic toxicities. The therapeutic efficacy and favorable safety profile of novel 5D3(CC-MLN8237)3.2 ATC demonstrates their potential use as a theranostic against aggressive PC.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Betelhem Assefa
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wathsala Liyanage
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sharmane Surasinghe
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Zora Nováková
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Cyril Bařinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Venu Raman
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dmitri Artemov
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sudath Hapuarachchige
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Zhong X, Yan J, Ding X, Su C, Xu Y, Yang M. Recent Advances in Bioorthogonal Click Chemistry for Enhanced PET and SPECT Radiochemistry. Bioconjug Chem 2023; 34:457-476. [PMID: 36811499 DOI: 10.1021/acs.bioconjchem.2c00583] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Due to their high reaction rate and reliable selectivity, bioorthogonal click reactions have been extensively investigated in numerous research fields, such as nanotechnology, drug delivery, molecular imaging, and targeted therapy. Previous reviews on bioorthogonal click chemistry for radiochemistry mainly focus on 18F-labeling protocols employed to produce radiotracers and radiopharmaceuticals. In fact, besides fluorine-18, other radionuclides such as gallium-68, iodine-125, and technetium-99m are also used in the field of bioorthogonal click chemistry. Herein, to provide a more comprehensive perspective, we provide a summary of recent advances in radiotracers prepared using bioorthogonal click reactions, including small molecules, peptides, proteins, antibodies, and nucleic acids as well as nanoparticles based on these radionuclides. The combination of pretargeting with imaging modalities or nanoparticles, as well as the clinical translations study, are also discussed to illustrate the effects and potential of bioorthogonal click chemistry for radiopharmaceuticals.
Collapse
Affiliation(s)
- Xinlin Zhong
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Junjie Yan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Xiang Ding
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Chen Su
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, P. R. China
| | - Yuping Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Min Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, P. R. China
| |
Collapse
|
5
|
Chen Z, Krishnamachary B, Mironchik Y, Ray Banerjee S, Pomper MG, Bhujwalla ZM. PSMA-specific degradable dextran for multiplexed immunotargeted siRNA therapeutics against prostate cancer. NANOSCALE 2022; 14:14014-14022. [PMID: 36093754 PMCID: PMC9844541 DOI: 10.1039/d2nr02200a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Small interfering RNA (siRNA) is ideal for gene silencing through a sequence-specific RNA interference process. The redundancy and complexity of molecular pathways in cancer create a need for multiplexed targeting that can be achieved with multiplexed siRNA delivery. Here, we delivered multiplexed siRNA with a PSMA-targeted biocompatible dextran nanocarrier to downregulate CD46 and PD-L1 in PSMA expressing prostate cancer cells. The selected gene targets, PD-L1 and CD46, play important roles in the escape of cancer cells from immune surveillance. PSMA, abundantly expressed by prostate cancer cells, allowed the prostate cancer-specific delivery of the nanocarrier. The nanocarrier was modified with acid cleavable acetal bonds for a rapid release of siRNA. Cell imaging and flow cytometry studies confirmed the PSMA-specific delivery of CD46 and PD-L1 siRNA to high PSMA expressing PC-3 PIP cells. Immunoblot, qRT-PCR and flow cytometry methods confirmed the downregulation of CD46 and PD-L1 following treatment with multiplexed siRNA.
Collapse
Affiliation(s)
- Zhihang Chen
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Balaji Krishnamachary
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Yelena Mironchik
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Sangeeta Ray Banerjee
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Zaver M Bhujwalla
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
6
|
Wang YC, Tian JY, Han YY, Liu YF, Chen SY, Guo FJ. Evaluation of the potential of ultrasound-mediated drug delivery for the treatment of ovarian cancer through preclinical studies. Front Oncol 2022; 12:978603. [PMID: 36132133 PMCID: PMC9483181 DOI: 10.3389/fonc.2022.978603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer (OC) has the greatest mortality rate among gynecological cancers, with a five-year survival rate of <50%. Contemporary adjuvant chemotherapy mostly fails in the case of OCs that are refractory, metastatic, recurrent, and drug-resistant. Emerging ultrasound (US)-mediated technologies show remarkable promise in overcoming these challenges. Absorption of US waves by the tissue results in the generation of heat due to its thermal effect causing increased diffusion of drugs from the carriers and triggering sonoporation by increasing the permeability of the cancer cells. Certain frequencies of US waves could also produce a cavitation effect on drug-filled microbubbles (MBs, phospholipid bilayers) thereby generating shear force and acoustic streaming that could assist drug release from the MBs, and promote the permeability of the cell membrane. A new class of nanoparticles that carry therapeutic agents and are guided by US contrast agents for precision delivery to the site of the ovarian tumor has been developed. Phase-shifting of nanoparticles by US sonication has also been engineered to enhance the drug delivery to the ovarian tumor site. These technologies have been used for targeting the ovarian cancer stem cells and protein moieties that are particularly elevated in OCs including luteinizing hormone-releasing hormone, folic acid receptor, and vascular endothelial growth factor. When compared to healthy ovarian tissue, the homeostatic parameters at the tissue microenvironment including pH, oxygen levels, and glucose metabolism differ significantly in ovarian tumors. US-based technologies have been developed to take advantage of these tumor-specific alterations for precision drug delivery. Preclinical efficacy of US-based targeting of currently used clinical chemotherapies presented in this review has the potential for rapid human translation, especially for formulations that use all substances that are deemed to be generally safe by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yi-Chao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing-Yan Tian
- Department of Urology, The Second Division of the First Hospital of Jilin University, Changchun, China
| | - Ying-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yun-Fei Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Si-Yao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Feng-Jun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Feng-Jun Guo,
| |
Collapse
|
7
|
Su Z, Xiao D, Xie F, Liu L, Wang Y, Fan S, Zhou X, Li S. Antibody-drug conjugates: Recent advances in linker chemistry. Acta Pharm Sin B 2021; 11:3889-3907. [PMID: 35024314 PMCID: PMC8727783 DOI: 10.1016/j.apsb.2021.03.042] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/17/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Antibody–drug conjugates (ADCs) are gradually revolutionizing clinical cancer therapy. The antibody–drug conjugate linker molecule determines both the efficacy and the adverse effects, and so has a major influence on the fate of ADCs. An ideal linker should be stable in the circulatory system and release the cytotoxic payload specifically in the tumor. However, existing linkers often release payloads nonspecifically and inevitably lead to off-target toxicity. This defect is becoming an increasingly important factor that restricts the development of ADCs. The pursuit of ADCs with optimal therapeutic windows has resulted in remarkable progress in the discovery and development of novel linkers. The present review summarizes the advance of the chemical trigger, linker‒antibody attachment and linker‒payload attachment over the last 5 years, and describes the ADMET properties of ADCs. This work also helps clarify future developmental directions for the linkers.
Collapse
Affiliation(s)
- Zheng Su
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Dian Xiao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fei Xie
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Lianqi Liu
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yanming Wang
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shiyong Fan
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Corresponding author. Tel: +86 10 66930603 (Shiyong Fan), +86 10 66930673 (Xinbo Zhou).
| | - Xinbo Zhou
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Corresponding author. Tel: +86 10 66930603 (Shiyong Fan), +86 10 66930673 (Xinbo Zhou).
| | - Song Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
8
|
Hapuarachchige S, Si G, Huang CT, Lesniak WG, Mease RC, Guo X, Gabrielson K, Artemov D. Dual-Modality PET-SPECT Image-Guided Pretargeting Delivery in HER2(+) Breast Cancer Models. Biomacromolecules 2021; 22:4606-4617. [PMID: 34704434 PMCID: PMC8578463 DOI: 10.1021/acs.biomac.1c00918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pretargeted drug delivery has been explored for decades as a promising approach in cancer therapy. An image-guided pretargeting strategy significantly enhances the intrinsic advantages of this approach since imaging the pretargeting step can be used for diagnostic purposes, while imaging of the drug delivery step can be utilized to evaluate drug distribution and assess therapeutic response. A trastuzumab (Tz)-based HER2 pretargeting component (Tz-TCO-[89Zr-DFO]) was developed by conjugating with trans-cyclooctene (TCO) bioorthogonal click chemistry functional groups and deferoxamine (DFO) to enable radiolabeling with a 89Zr PET tracer. The drug delivery component (HSA-DM1-Tt-[99mTc-HyNic]) was developed by conjugating human serum albumin (HSA) with mertansine (DM1), tetrazine (Tt) functional groups, and a HyNic chelator and radiolabeling with 99mTc. For ex vivo biodistribution studies, pretargeting and delivery components (without drug) were administered subsequently to mice bearing human HER2(+) breast cancer xenografts, and a high tumor uptake of Tz-TCO-[89Zr-DFO] (26.4% ID/g) and HSA-Tt-[99mTc-HyNic] (4.6% ID/g) was detected at 24 h postinjection. In vivo treatment studies were performed in the same HER2(+) breast cancer model using PET-SPECT image guidance. The increased tumor uptake of the pretargeting and drug delivery components was detected by PET-CT and SPECT-CT, respectively. The study showed a significant 92% reduction of the relative tumor volume in treated mice (RTV = 0.08 in 26 days), compared to the untreated control mice (RTV = 1.78 in 11 days) and to mice treated with only HSA-DM1-Tt-[99mTc-HyNic] (RTV = 1.88 in 16 days). Multimodality PET-SPECT image-guided and pretargeted drug delivery can be utilized to maximize efficacy, predict therapeutic response, and minimize systemic toxicity.
Collapse
Affiliation(s)
- Sudath Hapuarachchige
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, Maryland 21287, United States
| | - Ge Si
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Colin T Huang
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
| | - Wojciech G Lesniak
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
| | - Ronnie C Mease
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, Maryland 21287, United States
| | - Xin Guo
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Dmitri Artemov
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, Maryland 21287, United States
| |
Collapse
|
9
|
A Pretargeting Strategy Enabled by Bioorthogonal Reactions Towards Advanced Nuclear Medicines: Application and Perspective. Chem Res Chin Univ 2021. [DOI: 10.1007/s40242-021-1179-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
10
|
Zhou H, Fu J, Fu Q, Feng Y, Hong R, Li P, Wang Z, Huang X, Li F. Biotin-streptavidin-guided two-step pretargeting approach using PLGA for molecular ultrasound imaging and chemotherapy for ovarian cancer. PeerJ 2021; 9:e11486. [PMID: 34113492 PMCID: PMC8162236 DOI: 10.7717/peerj.11486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/27/2021] [Indexed: 01/18/2023] Open
Abstract
Background Ovarian cancer seriously threatens the lives and health of women, and early diagnosis and treatment are still challenging. Pre-targeting is a promising strategy to improve the treatment efficacy of ovarian cancer and the results of ultrasound imaging. Purpose To explore the effects of a pre-targeting strategy using streptavidin (SA) and paclitaxel (PTX)-loaded phase-shifting poly lactic-co-glycolic acid (PLGA) nanoparticles with perfluoro-n-pentane (PTX-PLGA-SA/PFPs) on the treatment and ultrasound imaging of ovarian cancer. Methods PTX-PLGA/PFPs were prepared with a single emulsion (O/W) solvent evaporation method and SA was attached using carbodiimide. The encapsulation efficiency of PTX and the release characteristics were assessed with high performance liquid chromatography. The phase-change characteristics of the PTX-PLGA-SA/PFPs were investigated. The anti-carcinoembryonic antigen (CEA) antibody (Ab) was covalently attached to PTX-PLGA/PFPs via carbodiimide to create PTX-PLGA-Ab/PFPs. The targeting efficiency of the nanoparticles and the viability of ovarian cancer SKOV3 cells were evaluated in each group using a microscope, flow cytometry, and cell counting kit 8 assays. Results THE PTX-PLGA-SA/PFPs were spheres with a size of 383.0 ± 75.59 nm. The encapsulation efficiency and loading capability of the nanoparticles for PTX were 71.56 ± 6.51% and 6.57 ± 0.61%, respectively. PTX was burst-released up to 70% in 2–3 d. When irradiated at 7.5 W for 3 min, the PTX-PLGA-SA/PFPs visibly enhanced the ultrasonography images (P < 0.05). At temperatures of 45°C and 60°C the nanoparticles phase-shifted into micro-bubbles and the sizes increased. The binding efficiencies of SA and Ab to the PTX-PLGA/PFPs were 97.16 ± 1.20% and 92.74 ± 5.75%, respectively. Pre-targeting resulted in a high binding efficacy and killing effect on SKOV3 cells (P < 0.05). Conclusions The two-step pre-targeting process can significantly enhance the targeting ability of PTX-loaded PLGA nanoparticles for ovarian cancer cells and substantially improve the therapeutic efficacy. This technique provides a new method for ultrasonic imaging and precise chemotherapy for ovarian cancer.
Collapse
Affiliation(s)
- Hang Zhou
- Ultrasound Medicine Department, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, Shapingba District, China
| | - Jing Fu
- Ultrasound Medicine Department, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, Shapingba District, China
| | - Qihuan Fu
- Ultrasound Medicine Department, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, Shapingba District, China
| | - Yujie Feng
- Ultrasound Medicine Department, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, Shapingba District, China
| | - Ruixia Hong
- Ultrasound Medicine Department, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, Shapingba District, China
| | - Pan Li
- Ultrasound Department, Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Yuzhong District, China
| | - Zhigang Wang
- Ultrasound Department, Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Yuzhong District, China
| | - Xiaoling Huang
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, Yuzhong District, China
| | - Fang Li
- Ultrasound Medicine Department, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, Shapingba District, China
| |
Collapse
|
11
|
Co CM, Izuagbe S, Zhou J, Zhou N, Sun X, Borrelli J, Tang L. Click chemistry-based pre-targeting cell delivery for cartilage regeneration. Regen Biomater 2021; 8:rbab018. [PMID: 34211730 DOI: 10.1093/rb/rbab018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/25/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
A fraction of the OA patient population is affected by post-traumatic osteoarthritis (PTOA) following acute joint injuries. Stopping or reversing the progression of PTOA following joint injury could improve long-term functional outcomes, reduced disability, and medical costs. To more effectively treat articular cartilage injury, we have developed a novel cell-based therapy that involves the pre-targeting of apoptotic chondrocytes and the delivery of healthy, metabolically active chondrocytes using click chemistry. Specifically, a pre-targeting agent was prepared via conjugating apoptotic binding peptide (ApoPep-1) and trans-cyclooctene (TCO) onto polyethylene glycol (PEG) polymer carrier. The pre-targeting agent would be introduced to injured areas of articular cartilage, leading to the accumulation of TCO groups on the injured areas from actively binding to apoptotic chondrocytes. Subsequently, methyltetrazine (Tz)-bearing chondrocytes would be immobilized on the surface of TCO-coated injured cartilage via Tz-TCO click chemistry reaction. Using an ex vivo human cartilage explant PTOA model, the effectiveness of this new approach was evaluated. Our studies show that this novel approach (Tz-TCO click chemistry) significantly enhanced the immobilization of healthy and metabolically active chondrocytes to the areas of apoptotic chondrocytes. Histological analyses demonstrated that this treatment regimen would significantly reduce the area of cartilage degeneration and enhance ECM regeneration. The results support that Tz-TCO click chemistry-mediated cell delivery approach has great potential in clinical applications for targeting and treatment of cartilage injury.
Collapse
Affiliation(s)
- Cynthia M Co
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Samira Izuagbe
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Jun Zhou
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Ning Zhou
- Department of Radiology, University of Texas Southwestern Medical, Dallas, TX 75390, USA
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical, Dallas, TX 75390, USA
| | - Joseph Borrelli
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Liping Tang
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| |
Collapse
|
12
|
Idiago-López J, Moreno-Antolín E, de la Fuente JM, Fratila RM. Nanoparticles and bioorthogonal chemistry joining forces for improved biomedical applications. NANOSCALE ADVANCES 2021; 3:1261-1292. [PMID: 36132873 PMCID: PMC9419263 DOI: 10.1039/d0na00873g] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/21/2021] [Indexed: 05/08/2023]
Abstract
Bioorthogonal chemistry comprises chemical reactions that can take place inside complex biological environments, providing outstanding tools for the investigation and elucidation of biological processes. Its use in combination with nanotechnology can lead to further developments in diverse areas of biomedicine, such as molecular bioimaging, targeted delivery, in situ drug activation, study of cell-nanomaterial interactions, biosensing, etc. Here, we summarise the recent efforts to bring together the unique properties of nanoparticles and the remarkable features of bioorthogonal reactions to create a toolbox of new or improved biomedical applications. We show how, by joining forces, bioorthogonal chemistry and nanotechnology can overcome some of the key current limitations in the field of nanomedicine, providing better, faster and more sensitive nanoparticle-based bioimaging and biosensing techniques, as well as therapeutic nanoplatforms with superior efficacy.
Collapse
Affiliation(s)
- Javier Idiago-López
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza Zaragoza 50009 Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Eduardo Moreno-Antolín
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza Zaragoza 50009 Spain
| | - Jesús M de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza Zaragoza 50009 Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza Zaragoza 50009 Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| |
Collapse
|
13
|
Cheal SM, McDevitt MR, Santich BH, Patel M, Yang G, Fung EK, Veach DR, Bell M, Ahad A, Vargas DB, Punzalan B, Pillarsetty NVK, Xu H, Guo HF, Monette S, Michel AO, Piersigilli A, Scheinberg DA, Ouerfelli O, Cheung NKV, Larson SM. Alpha radioimmunotherapy using 225Ac-proteus-DOTA for solid tumors - safety at curative doses. Theranostics 2020; 10:11359-11375. [PMID: 33052220 PMCID: PMC7546012 DOI: 10.7150/thno.48810] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
This is the initial report of an α-based pre-targeted radioimmunotherapy (PRIT) using 225Ac and its theranostic pair, 111In. We call our novel tumor-targeting DOTA-hapten PRIT system "proteus-DOTA" or "Pr." Herein we report the first results of radiochemistry development, radiopharmacology, and stoichiometry of tumor antigen binding, including the role of specific activity, anti-tumor efficacy, and normal tissue toxicity with the Pr-PRIT approach (as α-DOTA-PRIT). A series of α-DOTA-PRIT therapy studies were performed in three solid human cancer xenograft models of colorectal cancer (GPA33), breast cancer (HER2), and neuroblastoma (GD2), including evaluation of chronic toxicity at ~20 weeks of select survivors. Methods: Preliminary biodistribution experiments in SW1222 tumor-bearing mice revealed that 225Ac could not be efficiently pretargeted with current DOTA-Bn hapten utilized for 177Lu or 90Y, leading to poor tumor uptake in vivo. Therefore, we synthesized Pr consisting of an empty DOTA-chelate for 225Ac, tethered via a short polyethylene glycol linker to a lutetium-complexed DOTA for picomolar anti-DOTA chelate single-chain variable fragment (scFv) binding. Pr was radiolabeled with 225Ac and its imaging surrogate, 111In. In vitro studies verified anti-DOTA scFv recognition of [225Ac]Pr, and in vivo biodistribution and clearance studies were performed to evaluate hapten suitability and in vivo targeting efficiency. Results: Intravenously (i.v.) administered 225Ac- or 111In-radiolabeled Pr in mice showed rapid renal clearance and minimal normal tissue retention. In vivo pretargeting studies show high tumor accumulation of Pr (16.71 ± 5.11 %IA/g or 13.19 ± 3.88 %IA/g at 24 h p.i. for [225Ac]Pr and [111In]Pr, respectively) and relatively low uptake in normal tissues (all average ≤ 1.4 %IA/g at 24 h p.i.). Maximum tolerated dose (MTD) was not reached for either [225Ac]Pr alone or pretargeted [225Ac]Pr at administered activities up to 296 kBq/mouse. Single-cycle treatment consisting of α-DOTA-PRIT with either huA33-C825 bispecific anti-tumor/anti-DOTA-hapten antibody (BsAb), anti-HER2-C825 BsAb, or hu3F8-C825 BsAb for targeting GPA33, HER2, or GD2, respectively, was highly effective. In the GPA33 model, no complete responses (CRs) were observed but prolonged overall survival of treated animals was 42 d for α-DOTA-PRIT vs. 25 d for [225Ac]Pr only (P < 0.0001); for GD2, CRs (7/7, 100%) and histologic cures (4/7, 57%); and for HER2, CRs (7/19, 37%) and histologic cures (10/19, 56%) with no acute or chronic toxicity. Conclusions: [225Ac]Pr and its imaging biomarker [111In]Pr demonstrate optimal radiopharmacologic behavior for theranostic applications of α-DOTA-PRIT. For this initial evaluation of efficacy and toxicity, single-cycle treatment regimens were performed in all three systems. Histologic toxicity was not observed, so MTD was not observed. Prolonged overall survival, CRs, and histologic cures were observed in treated animals. In comparison to RIT with anti-tumor IgG antibodies, [225Ac]Pr has a much improved safety profile. Ultimately, these data will be used to guide clinical development of toxicity and efficacy studies of [225Ac]Pr, with the goal of delivering massive lethal doses of radiation to achieve a high probability of cure without toxicity.
Collapse
|
14
|
Engineered Fragments of the PSMA-Specific 5D3 Antibody and Their Functional Characterization. Int J Mol Sci 2020; 21:ijms21186672. [PMID: 32932591 PMCID: PMC7555429 DOI: 10.3390/ijms21186672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 11/29/2022] Open
Abstract
Prostate-Specific Membrane Antigen (PSMA) is an established biomarker for the imaging and experimental therapy of prostate cancer (PCa), as it is strongly upregulated in high-grade primary, androgen-independent, and metastatic lesions. Here, we report on the development and functional characterization of recombinant single-chain Fv (scFv) and Fab fragments derived from the 5D3 PSMA-specific monoclonal antibody (mAb). These fragments were engineered, heterologously expressed in insect S2 cells, and purified to homogeneity with yields up to 20 mg/L. In vitro assays including ELISA, immunofluorescence and flow cytometry, revealed that the fragments retain the nanomolar affinity and single target specificity of the parent 5D3 antibody. Importantly, using a murine xenograft model of PCa, we verified the suitability of fluorescently labeled fragments for in vivo imaging of PSMA-positive tumors and compared their pharmacokinetics and tissue distribution to the parent mAb. Collectively, our data provide an experimental basis for the further development of 5D3 recombinant fragments for future clinical use.
Collapse
|
15
|
Huang CT, Guo X, Bařinka C, Lupold SE, Pomper MG, Gabrielson K, Raman V, Artemov D, Hapuarachchige S. Development of 5D3-DM1: A Novel Anti-Prostate-Specific Membrane Antigen Antibody-Drug Conjugate for PSMA-Positive Prostate Cancer Therapy. Mol Pharm 2020; 17:3392-3402. [PMID: 32803984 DOI: 10.1021/acs.molpharmaceut.0c00457] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PC) is a potentially high-risk disease and the most common cancer in American men. It is a leading cause of cancer-related deaths in men in the US, second only to lung and bronchus cancer. Advanced and metastatic PC is initially treated with androgen deprivation therapy (ADT), but nearly all cases eventually progress to castrate-resistant prostate cancer (CRPC). CRPC is incurable in the metastatic stage but can be slowed by some conventional chemotherapeutics and second-generation ADT, such as enzalutamide and abiraterone. Therefore, novel therapeutic strategies are urgently needed. Prostate-specific membrane antigen (PSMA) is overexpressed in almost all aggressive PCs. PSMA is widely used as a target for PC imaging and drug delivery. Anti-PSMA monoclonal antibodies (mAbs) have been developed as bioligands for diagnostic imaging and targeted PC therapy. However, these mAbs are successfully used in PC imaging and only a few have gone beyond phase-I for targeted therapy. The 5D3 mAb is a novel, high-affinity, and fast-internalizing anti-PSMA antibody. Importantly, 5D3 mAb demonstrates a unique pattern of cellular localization to the centrosome after internalization in PSMA(+) PC3-PIP cells. These characteristics make 5D3 mAb an ideal bioligand to deliver tubulin inhibitors, such as mertansine, to the cell centrosome, leading to mitotic arrest and elimination of dividing PC cells. We have successfully developed a 5D3 mAb- and mertansine (DM1)-based antibody-drug conjugate (ADC) and evaluated it in vitro for binding affinity, internalization, and cytotoxicity. The in vivo therapeutic efficacy of 5D3-DM1 ADC was evaluated in PSMA(+) PC3-PIP and PSMA(-) PC3-Flu mouse models of human PC. This therapeutic study has revealed that this new anti-PSMA ADC can successfully control the growth of PSMA(+) tumors without inducing systemic toxicity.
Collapse
Affiliation(s)
- Colin T Huang
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
| | - Xin Guo
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Cyril Bařinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Shawn E Lupold
- The James Buchanan Brady Urologic Institute and Department of Urology, Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, Maryland 21287, United States
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States.,The James Buchanan Brady Urologic Institute and Department of Urology, Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, Maryland 21287, United States.,Department of Oncology, the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, Maryland 21287, United States
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Venu Raman
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States.,Department of Oncology, the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, Maryland 21287, United States
| | - Dmitri Artemov
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States.,Department of Oncology, the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, Maryland 21287, United States
| | - Sudath Hapuarachchige
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States
| |
Collapse
|
16
|
Hapuarachchige S, Artemov D. Theranostic Pretargeting Drug Delivery and Imaging Platforms in Cancer Precision Medicine. Front Oncol 2020; 10:1131. [PMID: 32793481 PMCID: PMC7387661 DOI: 10.3389/fonc.2020.01131] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/05/2020] [Indexed: 12/29/2022] Open
Abstract
Theranostics are nano-size or molecular-level agents serving for both diagnosis and therapy. Structurally, they are drug delivery systems integrated with molecular or targeted imaging agents. Theranostics are becoming popular because they are targeted therapeutics and can be used with no or minimal changes for diagnostic imaging to aid in precision medicine. Thus, there is a close relation between theranostics and image-guided therapy (IGT), and theranostics are actually a subclass of IGT in which both therapeutic and imaging functionalities are attributed to a single platform. An important theranostics strategy is biological pretargeting. In pretargeted IGT, first, the target is identified by a target-specific natural or synthetic bioligand followed by a nano-scale or molecular drug delivery component, which form therapeutic clusters by in situ conjugation reactions. If pretargeted drug delivery platforms are labeled with multimodal imaging probes, they can be used as theranostics for both diagnostic imaging and therapy. Optical and nuclear imaging techniques have mostly been used in proof-of-concept studies with pretargeted theranostics. The concept of pretargeting in theranostics is comparatively novel and generally requires a confirmed overexpression of surface receptors on targeted cells/tissue. In addition, the receptors should have natural or synthetic bioligands to be used as pretargeting components. Therefore, applications of pretargeting theranostics are still limited to several cancer types, which overexpress cell-surface markers on the target cancer cells. In this review, recent discoveries of pretargeting theranostics in breast, ovarian, prostate, and colorectal cancers are discussed to highlight main strengths and potential limitations the strategy.
Collapse
Affiliation(s)
- Sudath Hapuarachchige
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dmitri Artemov
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|