1
|
Buckle T, Rietbergen DDD, de Wit-van der Veen L, Schottelius M. Lessons learned in application driven imaging agent design for image-guided surgery. Eur J Nucl Med Mol Imaging 2024; 51:3040-3054. [PMID: 38900308 PMCID: PMC11300579 DOI: 10.1007/s00259-024-06791-x] [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: 02/29/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
To meet the growing demand for intraoperative molecular imaging, the development of compatible imaging agents plays a crucial role. Given the unique requirements of surgical applications compared to diagnostics and therapy, maximizing translational potential necessitates distinctive imaging agent designs. For effective surgical guidance, exogenous signatures are essential and are achievable through a diverse range of imaging labels such as (radio)isotopes, fluorescent dyes, or combinations thereof. To achieve optimal in vivo utility a balanced molecular design of the tracer as a whole is required, which ensures a harmonious effect of the imaging label with the affinity and specificity (e.g., pharmacokinetics) of a pharmacophore/targeting moiety. This review outlines common design strategies and the effects of refinements in the molecular imaging agent design on the agent's pharmacological profile. This includes the optimization of affinity, pharmacokinetics (including serum binding and target mediated background), biological clearance route, the achievable signal intensity, and the effect of dosing hereon.
Collapse
Affiliation(s)
- Tessa Buckle
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
- Section Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Linda de Wit-van der Veen
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Rue du Bugnon 25A, Agora, Lausanne, CH-1011, Switzerland.
- Agora, pôle de recherche sur le cancer, Lausanne, Switzerland.
| |
Collapse
|
2
|
Quintana J, Kang M, Hu H, Ng TSC, Wojtkiewicz GR, Scott E, Parangi S, Schuemann J, Weissleder R, Miller MA. Extended Pharmacokinetics Improve Site-Specific Prodrug Activation Using Radiation. ACS CENTRAL SCIENCE 2024; 10:1371-1382. [PMID: 39071065 PMCID: PMC11273447 DOI: 10.1021/acscentsci.4c00354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024]
Abstract
Radiotherapy is commonly used to treat cancer, and localized energy deposited by radiotherapy has the potential to chemically uncage prodrugs; however, it has been challenging to demonstrate prodrug activation that is both sustained in vivo and truly localized to tumors without affecting off-target tissues. To address this, we developed a series of novel phenyl-azide-caged, radiation-activated chemotherapy drug-conjugates alongside a computational framework for understanding corresponding pharmacokinetic and pharmacodynamic (PK/PD) behaviors. We especially focused on an albumin-bound prodrug of monomethyl auristatin E (MMAE) and found it blocked tumor growth in mice, delivered a 130-fold greater amount of activated drug to irradiated tumor versus unirradiated tissue, was 7.5-fold more efficient than a non albumin-bound prodrug, and showed no appreciable toxicity compared to free or cathepsin-activatable drugs. These data guided computational modeling of drug action, which indicated that extended pharmacokinetics can improve localized and cumulative drug activation, especially for payloads with low vascular permeability and diffusivity and particularly in patients receiving daily treatments of conventional radiotherapy for weeks. This work thus offers a quantitative PK/PD framework and proof-of-principle experimental demonstration of how extending prodrug circulation can improve its localized activity in vivo.
Collapse
Affiliation(s)
- Jeremy
M. Quintana
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mikyung Kang
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Huiyu Hu
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Surgery, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Thomas S. C. Ng
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Gregory R. Wojtkiewicz
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
| | - Ella Scott
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
| | - Sareh Parangi
- Department
of Surgery, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Jan Schuemann
- Department
of Radiation Oncology, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Miles A. Miller
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
3
|
Khera E, Kim J, Stein A, Ratanapanichkich M, Thurber GM. Mechanistically Weighted Metric to Predict In Vivo Antibody-Receptor Occupancy: An Analytical Approach. J Pharmacol Exp Ther 2023; 387:78-91. [PMID: 37105581 PMCID: PMC11046736 DOI: 10.1124/jpet.122.001540] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/11/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
In situ clinical measurement of receptor occupancy (RO) is challenging, particularly for solid tumors, necessitating the use of mathematical models that predict tumor receptor occupancy to guide dose decisions. A potency metric, average free tissue target to initial target ratio (AFTIR), was previously described based on a mechanistic compartmental model and is informative for near-saturating dose regimens. However, the metric fails at clinically relevant subsaturating antibody doses, as compartmental models cannot capture the spatial heterogeneity of distribution faced by some antibodies in solid tumors. Here we employ a partial differential equation (PDE) Krogh cylinder model to simulate spatiotemporal receptor occupancy and derive an analytical solution, a mechanistically weighted global AFTIR, that can better predict receptor occupancy regardless of dosing regimen. In addition to the four key parameters previously identified, a fifth key parameter, the absolute receptor density (targets/cell), is incorporated into the mechanistic AFTIR metric. Receptor density can influence equilibrium intratumoral drug concentration relative to whether the dose is saturating or not, thereby influencing the tumor penetration depth of the antibody. We derive mechanistic RO predictions based on distinct patterns of antibody tumor penetration, presented as a global AFTIR metric guided by a Thiele Modulus and a local saturation potential (drug equivalent of binding potential for positron emissions tomography imaging) and validate the results using rigorous global and local sensitivity analysis. This generalized AFTIR serves as a more accurate analytical metric to aid clinical dose decisions and rational design of antibody-based therapeutics without the need for extensive PDE simulations. SIGNIFICANCE STATEMENT: Determining antibody-receptor occupancy (RO) is critical for dosing decisions in pharmaceutical development, but direct clinical measurement of RO is often challenging and invasive, particularly for solid tumors. Significant efforts have been made to develop mathematical models and simplified analytical metrics of RO, but these often require complex computer simulations. Here we present a mathematically rigorous but simplified analytical model to accurately predict RO across a range of affinities, doses, drug, and tumor properties.
Collapse
Affiliation(s)
- Eshita Khera
- Departments of Chemical Engineering (E.K., M.R., G.M.T.) and Biomedical Engineering (G.M.T.), University of Michigan, Ann Arbor, Michigan; and Novartis Institute for BioMedical Research, Cambridge, Massachusetts (J.K., A.S.)
| | - Jaeyeon Kim
- Departments of Chemical Engineering (E.K., M.R., G.M.T.) and Biomedical Engineering (G.M.T.), University of Michigan, Ann Arbor, Michigan; and Novartis Institute for BioMedical Research, Cambridge, Massachusetts (J.K., A.S.)
| | - Andrew Stein
- Departments of Chemical Engineering (E.K., M.R., G.M.T.) and Biomedical Engineering (G.M.T.), University of Michigan, Ann Arbor, Michigan; and Novartis Institute for BioMedical Research, Cambridge, Massachusetts (J.K., A.S.)
| | - Matt Ratanapanichkich
- Departments of Chemical Engineering (E.K., M.R., G.M.T.) and Biomedical Engineering (G.M.T.), University of Michigan, Ann Arbor, Michigan; and Novartis Institute for BioMedical Research, Cambridge, Massachusetts (J.K., A.S.)
| | - Greg M Thurber
- Departments of Chemical Engineering (E.K., M.R., G.M.T.) and Biomedical Engineering (G.M.T.), University of Michigan, Ann Arbor, Michigan; and Novartis Institute for BioMedical Research, Cambridge, Massachusetts (J.K., A.S.)
| |
Collapse
|
4
|
Xia W, Singh N, Goel S, Shi S. Molecular Imaging of Innate Immunity and Immunotherapy. Adv Drug Deliv Rev 2023; 198:114865. [PMID: 37182699 DOI: 10.1016/j.addr.2023.114865] [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: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The innate immune system plays a key role as the first line of defense in various human diseases including cancer, cardiovascular and inflammatory diseases. In contrast to tissue biopsies and blood biopsies, in vivo imaging of the innate immune system can provide whole body measurements of immune cell location and function and changes in response to disease progression and therapy. Rationally developed molecular imaging strategies can be used in evaluating the status and spatio-temporal distributions of the innate immune cells in near real-time, mapping the biodistribution of novel innate immunotherapies, monitoring their efficacy and potential toxicities, and eventually for stratifying patients that are likely to benefit from these immunotherapies. In this review, we will highlight the current state-of-the-art in noninvasive imaging techniques for preclinical imaging of the innate immune system particularly focusing on cell trafficking, biodistribution, as well as pharmacokinetics and dynamics of promising immunotherapies in cancer and other diseases; discuss the unmet needs and current challenges in integrating imaging modalities and immunology and suggest potential solutions to overcome these barriers.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
5
|
Zhang L, Wang Y, Homan KT, Gaudette SM, McCluskey AJ, Chan Y, Murphy J, Abdalla M, Nelson CM, Sun VZ, Erickson JE, Knight HL, Clabbers A, Sterman AJS, Mitra S. Imaging the Alternatively Spliced D Domain of Tenascin C in a Preclinical Model of Inflammatory Bowel Disease. Mol Imaging Biol 2023; 25:314-323. [PMID: 35906512 PMCID: PMC10006278 DOI: 10.1007/s11307-022-01758-6] [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: 04/20/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE To image colon-expressed alternatively spliced D domain of tenascin C in preclinical colitis models using near infrared (NIR)-labeled targeted molecular imaging agents. PROCEDURES A human IgG1 with nanomolar binding affinity specific to the alternatively spliced D domain of tenascin C was generated. Immunohistochemistry identified disease-specific expression of this extracellular matrix protein in the colon of mice given dextran sulfate sodium in the drinking water. The antibody reagent was labeled with the NIR fluorophore IRDye 800CW via amine chemistry and intravenously dosed to evaluate in vivo targeting specificity. Increasing doses of imaging agent were given to estimate the saturating dose. RESULTS The NIR-labeled proteins successfully targeted colonic lesions in a murine model of colitis. Co-administration of a molar excess competing unlabeled dose reduced normalized uptake in diseased colon by > 70%. Near infrared ex vivo images of colon resected from diseased animals showed saturation at doses exceeding 1 nmol and was confirmed with additional quantitative ex vivo biodistribution. Cellular-level specificity and protein stability were assessed via microscopy. CONCLUSIONS Our imaging data suggest the alternatively spliced D domain of tenascin C is a promising target for delivery-based applications in inflammatory bowel diseases.
Collapse
Affiliation(s)
- Liang Zhang
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA.
| | - Yuzhen Wang
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | | | - Stephanie M Gaudette
- Worcester Technical High School, 1 Officer Manny Familia Wy, Worcester, MA, 01605, USA
| | | | - Ying Chan
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | - Joanne Murphy
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | - Mary Abdalla
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | | | - Victor Z Sun
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | - Jamie E Erickson
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | - Heather L Knight
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | - Anca Clabbers
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| | | | - Soumya Mitra
- AbbVie Bioresearch Center, 100 Research Dr, Worcester, MA, 01605, USA
| |
Collapse
|
6
|
Luo Q, Shao N, Zhang AC, Chen CF, Wang D, Luo LP, Xiao ZY. Smart Biomimetic Nanozymes for Precise Molecular Imaging: Application and Challenges. Pharmaceuticals (Basel) 2023; 16:249. [PMID: 37259396 PMCID: PMC9965384 DOI: 10.3390/ph16020249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 04/06/2024] Open
Abstract
New nanotechnologies for imaging molecules are widely being applied to visualize the expression of specific molecules (e.g., ions, biomarkers) for disease diagnosis. Among various nanoplatforms, nanozymes, which exhibit enzyme-like catalytic activities in vivo, have gained tremendously increasing attention in molecular imaging due to their unique properties such as diverse enzyme-mimicking activities, excellent biocompatibility, ease of surface tenability, and low cost. In addition, by integrating different nanoparticles with superparamagnetic, photoacoustic, fluorescence, and photothermal properties, the nanoenzymes are able to increase the imaging sensitivity and accuracy for better understanding the complexity and the biological process of disease. Moreover, these functions encourage the utilization of nanozymes as therapeutic agents to assist in treatment. In this review, we focus on the applications of nanozymes in molecular imaging and discuss the use of peroxidase (POD), oxidase (OXD), catalase (CAT), and superoxide dismutase (SOD) with different imaging modalities. Further, the applications of nanozymes for cancer treatment, bacterial infection, and inflammation image-guided therapy are discussed. Overall, this review aims to provide a complete reference for research in the interdisciplinary fields of nanotechnology and molecular imaging to promote the advancement and clinical translation of novel biomimetic nanozymes.
Collapse
Affiliation(s)
| | | | | | | | | | - Liang-Ping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Ze-Yu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| |
Collapse
|
7
|
Dong S, Nessler I, Kopp A, Rubahamya B, Thurber GM. Predictive Simulations in Preclinical Oncology to Guide the Translation of Biologics. Front Pharmacol 2022; 13:836925. [PMID: 35308243 PMCID: PMC8927291 DOI: 10.3389/fphar.2022.836925] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Preclinical in vivo studies form the cornerstone of drug development and translation, bridging in vitro experiments with first-in-human trials. However, despite the utility of animal models, translation from the bench to bedside remains difficult, particularly for biologics and agents with unique mechanisms of action. The limitations of these animal models may advance agents that are ineffective in the clinic, or worse, screen out compounds that would be successful drugs. One reason for such failure is that animal models often allow clinically intolerable doses, which can undermine translation from otherwise promising efficacy studies. Other times, tolerability makes it challenging to identify the necessary dose range for clinical testing. With the ability to predict pharmacokinetic and pharmacodynamic responses, mechanistic simulations can help advance candidates from in vitro to in vivo and clinical studies. Here, we use basic insights into drug disposition to analyze the dosing of antibody drug conjugates (ADC) and checkpoint inhibitor dosing (PD-1 and PD-L1) in the clinic. The results demonstrate how simulations can identify the most promising clinical compounds rather than the most effective in vitro and preclinical in vivo agents. Likewise, the importance of quantifying absolute target expression and antibody internalization is critical to accurately scale dosing. These predictive models are capable of simulating clinical scenarios and providing results that can be validated and updated along the entire development pipeline starting in drug discovery. Combined with experimental approaches, simulations can guide the selection of compounds at early stages that are predicted to have the highest efficacy in the clinic.
Collapse
Affiliation(s)
- Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Ian Nessler
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Anna Kopp
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Greg M. Thurber,
| |
Collapse
|
8
|
|
9
|
CD24-targeted fluorescence imaging in patient-derived xenograft models of high-grade serous ovarian carcinoma. EBioMedicine 2020; 56:102782. [PMID: 32454401 PMCID: PMC7248428 DOI: 10.1016/j.ebiom.2020.102782] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/02/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The survival rate of patients with advanced high-grade serous ovarian carcinoma (HGSOC) remains disappointing. Clinically translatable orthotopic cell line xenograft models and patient-derived xenografts (PDXs) may aid the implementation of more personalised treatment approaches. Although orthotopic PDX reflecting heterogeneous molecular subtypes are considered the most relevant preclinical models, their use in therapeutic development is limited by lack of appropriate imaging modalities. METHODS We developed novel orthotopic xenograft and PDX models for HGSOC, and applied a near-infrared fluorescently labelled monoclonal antibody targeting the cell surface antigen CD24 for non-invasive molecular imaging of epithelial ovarian cancer. CD24-Alexa Fluor 680 fluorescence imaging was compared to bioluminescence imaging in three orthotopic cell line xenograft models of ovarian cancer (OV-90luc+, Skov-3luc+ and Caov-3luc+, n = 3 per model). The application of fluorescence imaging to assess treatment efficacy was performed in carboplatin-paclitaxel treated orthotopic OV-90 xenografts (n = 10), before the probe was evaluated to detect disease progression in heterogenous PDX models (n = 7). FINDINGS Application of the near-infrared probe, CD24-AF680, enabled both spatio-temporal visualisation of tumour development, and longitudinal therapy monitoring of orthotopic xenografts. Notably, CD24-AF680 facilitated imaging of multiple PDX models representing different histological subtypes of the disease. INTERPRETATION The combined implementation of CD24-AF680 and orthotopic PDX models creates a state-of-the-art preclinical platform which will impact the identification and validation of new targeted therapies, fluorescence image-guided surgery, and ultimately the outcome for HGSOC patients. FUNDING This study was supported by the H2020 program MSCA-ITN [675743], Helse Vest RHF, and Helse Bergen HF [911809, 911852, 912171, 240222, HV1269], as well as by The Norwegian Cancer Society [182735], and The Research Council of Norway through its Centers of excellence funding scheme [223250, 262652].
Collapse
|
10
|
Affibody Molecules as Targeting Vectors for PET Imaging. Cancers (Basel) 2020; 12:cancers12030651. [PMID: 32168760 PMCID: PMC7139392 DOI: 10.3390/cancers12030651] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
Affibody molecules are small (58 amino acids) engineered scaffold proteins that can be selected to bind to a large variety of proteins with a high affinity. Their small size and high affinity make them attractive as targeting vectors for molecular imaging. High-affinity affibody binders have been selected for several cancer-associated molecular targets. Preclinical studies have shown that radiolabeled affibody molecules can provide highly specific and sensitive imaging on the day of injection; however, for a few targets, imaging on the next day further increased the imaging sensitivity. A phase I/II clinical trial showed that 68Ga-labeled affibody molecules permit an accurate and specific measurement of HER2 expression in breast cancer metastases. This paper provides an overview of the factors influencing the biodistribution and targeting properties of affibody molecules and the chemistry of their labeling using positron emitters.
Collapse
|
11
|
Bartelink IH, Jones EF, Shahidi‐Latham SK, Lee PRE, Zheng Y, Vicini P, van ‘t Veer L, Wolf D, Iagaru A, Kroetz DL, Prideaux B, Cilliers C, Thurber GM, Wimana Z, Gebhart G. Tumor Drug Penetration Measurements Could Be the Neglected Piece of the Personalized Cancer Treatment Puzzle. Clin Pharmacol Ther 2019; 106:148-163. [PMID: 30107040 PMCID: PMC6617978 DOI: 10.1002/cpt.1211] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022]
Abstract
Precision medicine aims to use patient genomic, epigenomic, specific drug dose, and other data to define disease patterns that may potentially lead to an improved treatment outcome. Personalized dosing regimens based on tumor drug penetration can play a critical role in this approach. State-of-the-art techniques to measure tumor drug penetration focus on systemic exposure, tissue penetration, cellular or molecular engagement, and expression of pharmacological activity. Using in silico methods, this information can be integrated to bridge the gap between the therapeutic regimen and the pharmacological link with clinical outcome. These methodologies are described, and challenges ahead are discussed. Supported by many examples, this review shows how the combination of these techniques provides enhanced patient-specific information on drug accessibility at the tumor tissue level, target binding, and downstream pharmacology. Our vision of how to apply tumor drug penetration measurements offers a roadmap for the clinical implementation of precision dosing.
Collapse
Affiliation(s)
- Imke H. Bartelink
- Department of MedicineUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneSouth San FranciscoCaliforniaUSA
- Department of Clinical Pharmacology and PharmacyAmsterdam UMCVrije Universiteit AmsterdamThe Netherlands
| | - Ella F. Jones
- Department of Radiology and Biomedical ImagingUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Pei Rong Evelyn Lee
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Yanan Zheng
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneSouth San FranciscoCaliforniaUSA
| | - Paolo Vicini
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneCambridgeUK
| | - Laura van ‘t Veer
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Denise Wolf
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging at Stanford Health CareStanfordCaliforniaUSA
| | - Deanna L. Kroetz
- Department of Bioengineering and Therapeutic Sciences (BTS)School of PharmacyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Brendan Prideaux
- Rutgers New Jersey Medical SchoolPublic Health Research InstituteRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Cornelius Cilliers
- Departments of Chemical Engineering and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Greg M. Thurber
- Departments of Chemical Engineering and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Zena Wimana
- Institut Jules BordetUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| | - Geraldine Gebhart
- Institut Jules BordetUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| |
Collapse
|
12
|
Khera E, Zhang L, Roberts S, Nessler I, Sandoval D, Reiner T, Thurber GM. Blocking of Glucagonlike Peptide-1 Receptors in the Exocrine Pancreas Improves Specificity for β-Cells in a Mouse Model of Type 1 Diabetes. J Nucl Med 2019; 60:1635-1641. [PMID: 31076502 DOI: 10.2967/jnumed.118.224881] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 05/03/2019] [Indexed: 01/18/2023] Open
Abstract
The diabetes community has long desired an imaging agent to quantify the number of insulin-secreting β-cells, beyond just functional equivalents (insulin secretion), to help diagnose and monitor early stages of both type 1 and type 2 diabetes mellitus. Loss in the number of β-cells can be masked by a compensatory increase in function of the remaining cells. Since β-cells form only about 1% of the pancreas and decrease as the disease progresses, only a few imaging agents, such as exendin, have demonstrated clinical potential to detect a drop in the already scarce signal. However, clinical translation of imaging with exendin has been hampered by pancreatic uptake that is higher than expected in subjects with long-term diabetes who lack β-cells. Exendin binds glucagonlike peptide-1 receptor (GLP-1R), previously thought to be expressed only on β-cells, but recent studies report low levels of GLP-1R on exocrine cells, complicating β-cell mass quantification. Methods: Here, we used a GLP-1R knockout mouse model to demonstrate that exocrine binding of exendin is exclusively via GLP-1R (∼1,000/cell) and not any other receptor. We then used lipophilic Cy-7 exendin to selectively preblock exocrine GLP-1R in healthy and streptozotocin-induced diabetic mice. Results: Sufficient receptors remain on β-cells for subsequent labeling with a fluorescent- or 111In-exendin. Conclusion: Selective GLP-1R blocking, which improves contrast between healthy and diabetic pancreata and provides a potential avenue for achieving the long-standing goal of imaging β-cell mass in the clinic.
Collapse
Affiliation(s)
- Eshita Khera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Liang Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ian Nessler
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Darleen Sandoval
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York.,Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
13
|
Bhatnagar S, Khera E, Liao J, Eniola V, Hu Y, Smith DE, Thurber GM. Oral and Subcutaneous Administration of a Near-Infrared Fluorescent Molecular Imaging Agent Detects Inflammation in a Mouse Model of Rheumatoid Arthritis. Sci Rep 2019; 9:4661. [PMID: 30858419 PMCID: PMC6411963 DOI: 10.1038/s41598-019-38548-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/31/2018] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease that causes irreversible damage to the joints. However, effective drugs exist that can stop disease progression, leading to intense interest in early detection and treatment monitoring to improve patient outcomes. Imaging approaches have the potential for early detection, but current methods lack sensitivity and/or are time-consuming and expensive. We examined potential routes for self-administration of molecular imaging agents in the form of subcutaneous and oral delivery of an integrin binding near-infrared (NIR) fluorescent imaging agent in an animal model of RA with the long-term goal of increasing safety and patient compliance for screening. NIR imaging has relatively low cost, uses non-ionizing radiation, and provides minimally invasive spatial and molecular information. This proof-of-principle study shows significant uptake of an IRDye800CW agent in inflamed joints of a collagen antibody induced arthritis (CAIA) mouse model compared to healthy joints, irrespective of the method of administration. The imaging results were extrapolated to clinical depths in silico using a 3D COMSOL model of NIR fluorescence imaging in a human hand to examine imaging feasability. With target to background concentration ratios greater than 5.5, which are achieved in the mouse model, these probes have the potential to identify arthritic joints following oral delivery at clinically relevant depths.
Collapse
Affiliation(s)
- Sumit Bhatnagar
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Eshita Khera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Jianshan Liao
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Victoria Eniola
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Yongjun Hu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, United States
| | - David E Smith
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
14
|
Zhou EY, Knox HJ, Reinhardt CJ, Partipilo G, Nilges MJ, Chan J. Near-Infrared Photoactivatable Nitric Oxide Donors with Integrated Photoacoustic Monitoring. J Am Chem Soc 2018; 140:11686-11697. [PMID: 30198716 PMCID: PMC7331458 DOI: 10.1021/jacs.8b05514] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Photoacoustic (PA) tomography is a noninvasive technology that utilizes near-infrared (NIR) excitation and ultrasonic detection to image biological tissue at centimeter depths. While several activatable small-molecule PA sensors have been developed for various analytes, the use of PA molecules for deep-tissue analyte delivery and monitoring remains an underexplored area of research. Herein, we describe the synthesis, characterization, and in vivo validation of photoNOD-1 and photoNOD-2, the first organic, NIR-photocontrolled nitric oxide (NO) donors that incorporate a PA readout of analyte release. These molecules consist of an aza-BODIPY dye appended with an aryl N-nitrosamine NO-donating moiety. The photoNODs exhibit chemostability to various biological stimuli, including redox-active metals and CYP450 enzymes, and demonstrate negligible cytotoxicity in the absence of irradiation. Upon single-photon NIR irradiation, photoNOD-1 and photoNOD-2 release NO as well as rNOD-1 or rNOD-2, PA-active products that enable ratiometric monitoring of NO release. Our in vitro studies show that, upon irradiation, photoNOD-1 and photoNOD-2 exhibit 46.6-fold and 21.5-fold ratiometric turn-ons, respectively. Moreover, unlike existing NIR NO donors, the photoNODs do not require encapsulation or multiphoton activation for use in live animals. In this study, we use PA tomography to monitor the local, irradiation-dependent release of NO from photoNOD-1 and photoNOD-2 in mice after subcutaneous treatment. In addition, we use a murine model for breast cancer to show that photoNOD-1 can selectively affect tumor growth rates in the presence of NIR light stimulation following systemic administration.
Collapse
Affiliation(s)
- Effie Y. Zhou
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Hailey J. Knox
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Christopher J. Reinhardt
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Gina Partipilo
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Mark J. Nilges
- Illinois EPR Research Center, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
| | - Jefferson Chan
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| |
Collapse
|
15
|
Preclinical Evaluation of [ 68Ga]Ga-DFO-ZEGFR:2377: A Promising Affibody-Based Probe for Noninvasive PET Imaging of EGFR Expression in Tumors. Cells 2018; 7:cells7090141. [PMID: 30231504 PMCID: PMC6162391 DOI: 10.3390/cells7090141] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/13/2018] [Accepted: 09/15/2018] [Indexed: 01/10/2023] Open
Abstract
Radionuclide imaging of epidermal growth factor receptor (EGFR) expression in tumors may stratify patients for EGFR-targeting therapies and predict response or resistance to certain treatments. Affibody molecules, which are nonimmunoglobulin scaffold proteins, have a high potential as probes for molecular imaging. In this study, maleimido derivative of desferrioxamine B (DFO) chelator was site-specifically coupled to the C-terminal cysteine of the anti-EGFR affibody molecule ZEGFR:2377, and the DFO-ZEGFR:2377 conjugate was labeled with the generator-produced positron-emitting radionuclide 68Ga. Stability, specificity of binding to EGFR-expressing cells, and processing of [68Ga]Ga-DFO-ZEGFR:2377 by cancer cells after binding were evaluated in vitro. In vivo studies were performed in nude mice bearing human EGFR-expressing A431 epidermoid cancer xenografts. The biodistribution of [68Ga]Ga-DFO-ZEGFR:2377 was directly compared with the biodistribution of [89Zr]Zr-DFO-ZEGFR:2377. DFO-ZEGFR:2377 was efficiently (isolated yield of 73 ± 3%) and stably labeled with 68Ga. Binding of [68Ga]Ga-DFO-ZEGFR:2377 to EGFR-expressing cells in vitro was receptor-specific and proportional to the EGFR expression level. In vivo saturation experiment demonstrated EGFR-specific accumulation of [68Ga]Ga-DFO-ZEGFR:2377 in A431 xenografts. Compared to [89Zr]Zr-DFO-ZEGFR:2377, [68Ga]Ga-DFO-ZEGFR:2377 demonstrated significantly (p < 0.05) higher uptake in tumors and lower uptake in spleen and bones. This resulted in significantly higher tumor-to-organ ratios for [68Ga]Ga-DFO-ZEGFR:2377. In conclusion, [68Ga]Ga-DFO-ZEGFR:2377 is a promising probe for imaging of EGFR expression.
Collapse
|
16
|
MT1-MMP as a PET Imaging Biomarker for Pancreas Cancer Management. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:8382148. [PMID: 30224904 PMCID: PMC6129362 DOI: 10.1155/2018/8382148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/25/2018] [Indexed: 01/02/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to be one of the deadliest cancers for which optimal diagnostic tools are still greatly needed. Identification of PDAC-specific molecular markers would be extremely useful to improve disease diagnosis and follow-up. MT1-MMP has long been involved in pancreatic cancer, especially in tumour invasion and metastasis. In this study, we aim to ascertain the suitability of MT1-MMP as a biomarker for positron emission tomography (PET) imaging. Two probes were assessed and compared for this purpose, an MT1-MMP-specific binding peptide (MT1-AF7p) and a specific antibody (LEM2/15), labelled, respectively, with 68Ga and with 89Zr. PET imaging with both probes was conducted in patient-derived xenograft (PDX), subcutaneous and orthotopic, PDAC mouse models, and in a cancer cell line (CAPAN-2)-derived xenograft (CDX) model. Both radiolabelled tracers were successful in identifying, by means of PET imaging techniques, tumour tissues expressing MT1-MMP although they did so at different uptake levels. The 89Zr-DFO-LEM2/15 probe showed greater specific activity compared to the 68Ga-labelled peptide. The mean value of tumour uptake for the 89Zr-DFO-LEM2/15 probe (5.67 ± 1.11%ID/g, n=28) was 25-30 times higher than that of the 68Ga-DOTA-AF7p ones. Tumour/blood ratios (1.13 ± 0.51 and 1.44 ± 0.43 at 5 and 7 days of 89Zr-DFO-LEM2/15 after injection) were higher than those estimated for 68Ga-DOTA-AF7p probes (of approximately tumour/blood ratio = 0.5 at 90 min after injection). Our findings strongly point out that (i) the in vivo detection of MT1-MMP by PET imaging is a promising strategy for PDAC diagnosis and (ii) labelled LEM2/15 antibody is a better candidate than MT1-AF7p for PDAC detection.
Collapse
|
17
|
Shaghaghi Z, Abedi SM, Hosseinimehr SJ. Tricine co-ligand improved the efficacy of 99mTc-HYNIC-(Ser)3-J18 peptide for targeting and imaging of non-small-cell lung cancer. Biomed Pharmacother 2018; 104:325-331. [DOI: 10.1016/j.biopha.2018.05.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 11/26/2022] Open
|
18
|
Lindbo S, Garousi J, Mitran B, Vorobyeva A, Oroujeni M, Orlova A, Hober S, Tolmachev V. Optimized Molecular Design of ADAPT-Based HER2-Imaging Probes Labeled with 111In and 68Ga. Mol Pharm 2018; 15:2674-2683. [PMID: 29865791 DOI: 10.1021/acs.molpharmaceut.8b00204] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Radionuclide molecular imaging is a promising tool for visualization of cancer associated molecular abnormalities in vivo and stratification of patients for specific therapies. ADAPT is a new type of small engineered proteins based on the scaffold of an albumin binding domain of protein G. ADAPTs have been utilized to select and develop high affinity binders to different proteinaceous targets. ADAPT6 binds to human epidermal growth factor 2 (HER2) with low nanomolar affinity and can be used for its in vivo visualization. Molecular design of 111In-labeled anti-HER2 ADAPT has been optimized in several earlier studies. In this study, we made a direct comparison of two of the most promising variants, having either a DEAVDANS or a (HE)3DANS sequence at the N-terminus, conjugated with a maleimido derivative of DOTA to a GSSC amino acids sequence at the C-terminus. The variants (designated DOTA-C59-DEAVDANS-ADAPT6-GSSC and DOTA-C61-(HE)3DANS-ADAPT6-GSSC) were stably labeled with 111In for SPECT and 68Ga for PET. Biodistribution of labeled ADAPT variants was evaluated in nude mice bearing human tumor xenografts with different levels of HER2 expression. Both variants enabled clear discrimination between tumors with high and low levels of HER2 expression. 111In-labeled ADAPT6 derivatives provided higher tumor-to-organ ratios compared to 68Ga-labeled counterparts. The best performing variant was DOTA-C61-(HE)3DANS-ADAPT6-GSSC, which provided tumor-to-blood ratios of 208 ± 36 and 109 ± 17 at 3 h for 111In and 68Ga labels, respectively.
Collapse
Affiliation(s)
- Sarah Lindbo
- School of Engineering in Chemistry, Biotechnology and Health (CBH) , Division of Protein Science, KTH Royal Institute of Technology , SE-10691 Stockholm , Sweden
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology , Uppsala University , 751 85 Uppsala , Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry , Uppsala University , 751 23 Uppsala , Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology , Uppsala University , 751 85 Uppsala , Sweden
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology , Uppsala University , 751 85 Uppsala , Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry , Uppsala University , 751 23 Uppsala , Sweden
| | - Sophia Hober
- School of Engineering in Chemistry, Biotechnology and Health (CBH) , Division of Protein Science, KTH Royal Institute of Technology , SE-10691 Stockholm , Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology , Uppsala University , 751 85 Uppsala , Sweden
| |
Collapse
|
19
|
Buckle T, van Willigen DM, Spa SJ, Hensbergen AW, van der Wal S, de Korne CM, Welling MM, van der Poel HG, Hardwick JCH, van Leeuwen FWB. Tracers for Fluorescence-Guided Surgery: How Elongation of the Polymethine Chain in Cyanine Dyes Alters the Pharmacokinetics of a Dual-Modality c[RGDyK] Tracer. J Nucl Med 2018; 59:986-992. [PMID: 29449447 DOI: 10.2967/jnumed.117.205575] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/12/2018] [Indexed: 01/07/2023] Open
Abstract
The potential of receptor-mediated fluorescence-based image-guided surgery tracers is generally linked to the near-infrared emission profile and good-manufacturing-production availability of fluorescent dyes. Surprisingly, little is known about the critical interaction between the structural composition of the dyes and the pharmacokinetics of the tracers. In this study, a dual-modality tracer design was used to systematically and quantitatively evaluate the influence of elongation of the polymethine chain in a fluorescent cyanine dye on the imaging potential of a targeted tracer. Methods: As a model system, the integrin marker αvβ3 was targeted using arginylglycylaspartisc acid [RGD]-based vectors functionalized with a 111In-diethylenetriaminepentaacetic acid (DTPA) chelate and a fluorescent dye: (Cy3-(SO3)methyl-COOH [emission wavelength (λem), 580 nm], Cy5-(SO3)methyl-COOH [λem, 680 nm], or Cy7-(SO3)methyl-COOH [λem, 780 nm]). Tracers were analyzed for differences in photophysical properties, serum protein binding, chemical or optical stability, and signal penetration through tissue. Receptor affinities were evaluated using saturation and competition experiments. In vivo biodistribution (SPECT imaging and percentage injected dose per gram of tissue) was assessed in tumor-bearing mice and complemented with in vivo and ex vivo fluorescence images obtained using a clinical-grade multispectral fluorescence laparoscope. Results: Two carbon-atom-step variations in the polymethine chain of the fluorescent cyanine dyes were shown to significantly influence the chemical and photophysical characteristics (e.g., stability, brightness, and tissue penetration) of the hybrid RGD tracers. DTPA-Cy5-(SO3)methyl-COOH-c[RGDyK] structurally outperformed its Cy3 and Cy7 derivatives. Radioactivity-based evaluation of in vivo tracer pharmacokinetics yielded the lowest nonspecific uptake and highest tumor-to-background ratio for DTPA-Cy5-(SO3)methyl-COOH-c[RGDyK] (13.2 ± 1.7), with the Cy3 and Cy7 analogs trailing at respective tumor-to-background ratios of 5.7 ± 0.7 and 4.7 ± 0.7. Fluorescence-based assessment of tumor visibility revealed a similar trend. Conclusion: These findings underline that variations in the polymethine chain lengths of cyanine dyes have a profound influence on the photophysical properties, stability, and in vivo targeting capabilities of fluorescent imaging tracers. In a direct comparison, the intermediate-length dye (Cy5) yielded a superior c[RGDyK] tracer, compared with the shorter (Cy3) and longer (Cy7) analogs.
Collapse
Affiliation(s)
- Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Division of Molecular Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Danny M van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Silvia J Spa
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Albertus W Hensbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Steffen van der Wal
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clarize M de Korne
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mick M Welling
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - James C H Hardwick
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| |
Collapse
|
20
|
Sirois AR, Deny DA, Baierl SR, George KS, Moore SJ. Fn3 proteins engineered to recognize tumor biomarker mesothelin internalize upon binding. PLoS One 2018; 13:e0197029. [PMID: 29738555 PMCID: PMC5940182 DOI: 10.1371/journal.pone.0197029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/20/2018] [Indexed: 11/19/2022] Open
Abstract
Mesothelin is a cell surface protein that is overexpressed in numerous cancers, including breast, ovarian, lung, liver, and pancreatic tumors. Aberrant expression of mesothelin has been shown to promote tumor progression and metastasis through interaction with established tumor biomarker CA125. Therefore, molecules that specifically bind to mesothelin have potential therapeutic and diagnostic applications. However, no mesothelin-targeting molecules are currently approved for routine clinical use. While antibodies that target mesothelin are in development, some clinical applications may require a targeting molecule with an alternative protein fold. For example, non-antibody proteins are more suitable for molecular imaging and may facilitate diverse chemical conjugation strategies to create drug delivery complexes. In this work, we engineered variants of the fibronectin type III domain (Fn3) non-antibody protein scaffold to bind to mesothelin with high affinity, using directed evolution and yeast surface display. Lead engineered Fn3 variants were solubly produced and purified from bacterial culture at high yield. Upon specific binding to mesothelin on human cancer cell lines, the engineered Fn3 proteins internalized and co-localized to early endosomes. To our knowledge, this is the first report of non-antibody proteins engineered to bind mesothelin. The results validate that non-antibody proteins can be engineered to bind to tumor biomarker mesothelin, and encourage the continued development of engineered variants for applications such as targeted diagnostics and therapeutics.
Collapse
Affiliation(s)
- Allison R. Sirois
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts, United States of America
- Picker Engineering Program, Smith College, Northampton, Massachusetts, United States of America
| | - Daniela A. Deny
- Department of Biochemistry, Smith College, Northampton, Massachusetts, United States of America
| | - Samantha R. Baierl
- Picker Engineering Program, Smith College, Northampton, Massachusetts, United States of America
| | - Katia S. George
- Department of Biochemistry, Smith College, Northampton, Massachusetts, United States of America
| | - Sarah J. Moore
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts, United States of America
- Picker Engineering Program, Smith College, Northampton, Massachusetts, United States of America
- Department of Biological Sciences, Smith College, Northampton, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
21
|
Bhatnagar S, Verma KD, Hu Y, Khera E, Priluck A, Smith DE, Thurber GM. Oral Administration and Detection of a Near-Infrared Molecular Imaging Agent in an Orthotopic Mouse Model for Breast Cancer Screening. Mol Pharm 2018; 15:1746-1754. [PMID: 29696981 PMCID: PMC5941251 DOI: 10.1021/acs.molpharmaceut.7b00994] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
![]()
Molecular
imaging is advantageous for screening diseases such as
breast cancer by providing precise spatial information on disease-associated
biomarkers, something neither blood tests nor anatomical imaging can
achieve. However, the high cost and risks of ionizing radiation for
several molecular imaging modalities have prevented a feasible and
scalable approach for screening. Clinical studies have demonstrated
the ability to detect breast tumors using nonspecific probes such
as indocyanine green, but the lack of molecular information and required
intravenous contrast agent does not provide a significant benefit
over current noninvasive imaging techniques. Here we demonstrate that
negatively charged sulfate groups, commonly used to improve solubility
of near-infrared fluorophores, enable sufficient oral absorption and
targeting of fluorescent molecular imaging agents for completely noninvasive
detection of diseased tissue such as breast cancer. These functional
groups improve the pharmacokinetic properties of affinity ligands
to achieve targeting efficiencies compatible with clinical imaging
devices using safe, nonionizing radiation (near-infrared light). Together,
this enables development of a “disease screening pill”
capable of oral absorption and systemic availability, target binding,
background clearance, and imaging at clinically relevant depths for
breast cancer screening. This approach should be adaptable to other
molecular targets and diseases for use as a new class of screening
agents.
Collapse
|
22
|
Depalo N, Corricelli M, De Paola I, Valente G, Iacobazzi RM, Altamura E, Debellis D, Comegna D, Fanizza E, Denora N, Laquintana V, Mavelli F, Striccoli M, Saviano M, Agostiano A, Del Gatto A, Zaccaro L, Curri ML. NIR Emitting Nanoprobes Based on Cyclic RGD Motif Conjugated PbS Quantum Dots for Integrin-Targeted Optical Bioimaging. ACS APPLIED MATERIALS & INTERFACES 2017; 9:43113-43126. [PMID: 29148709 DOI: 10.1021/acsami.7b14155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Here, silica-coated PbS quantum dots (QDs) with photoluminescence emission properties in the near-infrared (NIR) region are proposed as potential effective single particle optical nanoprobes for future in vivo imaging of tumors. The dispersibility in aqueous medium of hydrophobic PbS QDs was accomplished by growing a silica shell on their surface by exploiting a base assisted water-in-oil microemulsion method. The silica-coated PbS QDs were then conjugated with a specifically designed cyclic arginine-glycine-aspartic acid (cRGD) peptide that is able to specifically recognize αvβ3 integrins, which are overexpressed in angiogenic tumor-induced vasculatures and on some solid tumors, to achieve tumor-specific targeting. The cRGD peptide PbS silica-coated QDs were systematically characterized, at each step of their preparation, by means of complementary optical and structural techniques, demonstrating appropriate colloidal stability and the maintenance of their optical futures in aqueous solutions. The cellular uptake of cRGD peptide functionalized luminescent nanostructures in human melanoma cells, where overexpression of αvβ3 was observed, was assessed by means of confocal microscopy analysis and cytometric study. The selectivity of the cRGD peptide PbS silica-coated QDs for the αvβ3 integrin was established, consequently highlighting the significant potential of the developed NIR emitting nanostructures as optically traceable nanoprobes for future αvβ3 integrin receptor in vivo targeting in the NIR region.
Collapse
Affiliation(s)
- N Depalo
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| | - M Corricelli
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| | - I De Paola
- Istituto di Biostrutture e Bioimmagini-CNR , Via Mezzocannone 16, 80134 Napoli, Italy
| | - G Valente
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| | - R M Iacobazzi
- Istituto Tumori IRCCS Giovanni Paolo II , Viale Orazio Flacco 65, 70124 Bari, Italy
| | | | | | - D Comegna
- Istituto di Biostrutture e Bioimmagini-CNR , Via Mezzocannone 16, 80134 Napoli, Italy
| | - E Fanizza
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| | - N Denora
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| | | | | | - M Striccoli
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| | - M Saviano
- Istituto di Cristallografia-CNR Bari , Via Amendola 122/O, 70126 Bari, Italy
| | - A Agostiano
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| | - A Del Gatto
- Istituto di Biostrutture e Bioimmagini-CNR , Via Mezzocannone 16, 80134 Napoli, Italy
| | - L Zaccaro
- Istituto di Biostrutture e Bioimmagini-CNR , Via Mezzocannone 16, 80134 Napoli, Italy
| | - M L Curri
- Istituto per i Processi Chimico-Fisici-CNR SS Bari , Via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
23
|
Cilliers C, Nessler I, Christodolu N, Thurber GM. Tracking Antibody Distribution with Near-Infrared Fluorescent Dyes: Impact of Dye Structure and Degree of Labeling on Plasma Clearance. Mol Pharm 2017; 14:1623-1633. [PMID: 28294622 PMCID: PMC5415873 DOI: 10.1021/acs.molpharmaceut.6b01091] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Monoclonal
antibodies labeled with near-infrared (NIR) fluorophores
have potential use in disease detection, intraoperative imaging, and
pharmacokinetic characterization of therapeutic antibodies in both
the preclinical and clinical setting. Recent work has shown conjugation
of NIR fluorophores to antibodies can potentially alter antibody disposition
at a sufficiently high degree of labeling (DoL); however, other reports
show minimal impact after labeling with NIR fluorophores. In this
work, we label two clinically approved antibodies, Herceptin (trastuzumab)
and Avastin (bevacizumab), with NIR dyes IRDye 800CW (800CW) or Alexa
Fluor 680 (AF680), at 1.2 and 0.3 dyes/antibody and examine the impact
of fluorophore conjugation on antibody plasma clearance and tissue
distribution. At 0.3 DoL, AF680 conjugates exhibited similar clearance
to unlabeled antibody over 17 days while 800CW conjugates diverged
after 4 days, suggesting AF680 is a more suitable choice for long-term
pharmacokinetic studies. At the 1.2 DoL, 800CW conjugates cleared
faster than unlabeled antibodies after several hours, in agreement
with other published reports. The tissue biodistribution for bevacizumab–800CW
and −AF680 conjugates agreed well with literature reported
biodistributions using radiolabels. However, the greater tissue autofluorescence
at 680 nm resulted in limited detection above background at low (∼2
mg/kg) doses and 0.3 DoL for AF680, indicating that 800CW is more
appropriate for short-term biodistribution measurements and intraoperative
imaging. Overall, our work shows a DoL of 0.3 or less for non-site-specifically
labeled antibodies (with a Poisson distribution) is ideal for limiting
the impact of NIR fluorophores on antibody pharmacokinetics.
Collapse
Affiliation(s)
- Cornelius Cilliers
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Ian Nessler
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Nikolas Christodolu
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Greg M Thurber
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
24
|
Comparative Evaluation of Anti-HER2 Affibody Molecules Labeled with 64Cu Using NOTA and NODAGA. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:8565802. [PMID: 29097939 PMCID: PMC5612711 DOI: 10.1155/2017/8565802] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/08/2017] [Indexed: 01/04/2023]
Abstract
Imaging using affibody molecules enables discrimination between breast cancer metastases with high and low expression of HER2, making appropriate therapy selection possible. This study aimed to evaluate if the longer half-life of 64Cu (T1/2 = 12.7 h) would make 64Cu a superior nuclide compared to 68Ga for PET imaging of HER2 expression using affibody molecules. The synthetic ZHER2:S1 affibody molecule was conjugated with the chelators NOTA or NODAGA and labeled with 64Cu. The tumor-targeting properties of 64Cu-NOTA-ZHER2:S1 and 64Cu-NODAGA-ZHER2:S1 were evaluated and compared with the targeting properties of 68Ga-NODAGA-ZHER2:S1 in mice. Both 64Cu-NOTA-ZHER2:S1 and 64Cu-NODAGA-ZHER2:S1 demonstrated specific targeting of HER2-expressing xenografts. At 2 h after injection of 64Cu-NOTA-ZHER2:S1, 64Cu-NODAGA-ZHER2:S1, and 68Ga-NODAGA-ZHER2:S1, tumor uptakes did not differ significantly. Renal uptake of 64Cu-labeled conjugates was dramatically reduced at 6 and 24 h after injection. Notably, radioactivity uptake concomitantly increased in blood, lung, liver, spleen, and intestines, which resulted in decreased tumor-to-organ ratios compared to 2 h postinjection. Organ uptake was lower for 64Cu-NODAGA-ZHER2:S1. The most probable explanation for this biodistribution pattern was the release and redistribution of renal radiometabolites. In conclusion, monoamide derivatives of NOTA and NODAGA may be suboptimal chelators for radiocopper labeling of anti-HER2 affibody molecules and, possibly, other scaffold proteins with high renal uptake.
Collapse
|
25
|
Oliveira C, Ferreira AS, Novoa-Carballal R, Nunes C, Pashkuleva I, Neves NM, Coimbra MA, Reis RL, Martins A, Silva TH. The Key Role of Sulfation and Branching on Fucoidan Antitumor Activity. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600340] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/16/2016] [Indexed: 01/11/2023]
Affiliation(s)
- Catarina Oliveira
- 3B's Research Group-Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Parque de Ciência e Tecnologia 4805-017 Barco Guimarães Portugal
- ICVS/3B's; PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Andreia S. Ferreira
- QOPNA, Department of Chemistry; University of Aveiro; Campus de Santiago 3810-193 Aveiro Portugal
| | - Ramon Novoa-Carballal
- 3B's Research Group-Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Parque de Ciência e Tecnologia 4805-017 Barco Guimarães Portugal
- ICVS/3B's; PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Cláudia Nunes
- QOPNA, Department of Chemistry; University of Aveiro; Campus de Santiago 3810-193 Aveiro Portugal
- CICECO, Department of Chemistry; University of Aveiro; Campus de Santiago 3810-193 Aveiro Portugal
| | - Iva Pashkuleva
- 3B's Research Group-Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Parque de Ciência e Tecnologia 4805-017 Barco Guimarães Portugal
- ICVS/3B's; PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Nuno M. Neves
- 3B's Research Group-Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Parque de Ciência e Tecnologia 4805-017 Barco Guimarães Portugal
- ICVS/3B's; PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Manuel A. Coimbra
- QOPNA, Department of Chemistry; University of Aveiro; Campus de Santiago 3810-193 Aveiro Portugal
| | - Rui L. Reis
- 3B's Research Group-Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Parque de Ciência e Tecnologia 4805-017 Barco Guimarães Portugal
- ICVS/3B's; PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Albino Martins
- 3B's Research Group-Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Parque de Ciência e Tecnologia 4805-017 Barco Guimarães Portugal
- ICVS/3B's; PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Tiago H. Silva
- 3B's Research Group-Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Parque de Ciência e Tecnologia 4805-017 Barco Guimarães Portugal
- ICVS/3B's; PT Government Associate Laboratory; Braga/Guimarães Portugal
| |
Collapse
|
26
|
Zhang L, Navaratna T, Thurber GM. A Helix-Stabilizing Linker Improves Subcutaneous Bioavailability of a Helical Peptide Independent of Linker Lipophilicity. Bioconjug Chem 2016; 27:1663-72. [PMID: 27327034 DOI: 10.1021/acs.bioconjchem.6b00209] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stabilized peptides address several limitations to peptide-based imaging agents and therapeutics such as poor stability and low affinity due to conformational flexibility. There is also active research in developing these compounds for intracellular drug targeting, and significant efforts have been invested to determine the effects of helix stabilization on intracellular delivery. However, much less is known about the impact on other pharmacokinetic parameters such as plasma clearance and bioavailability. We investigated the effect of different fluorescent helix-stabilizing linkers with varying lipophilicity on subcutaneous (sc) bioavailability using the glucagon-like peptide-1 (GLP-1) receptor ligand exendin as a model system. The stabilized peptides showed significantly higher protease resistance and increased bioavailability independent of linker hydrophilicity, and all subcutaneously delivered conjugates were able to successfully target the islets of Langerhans with high specificity. The lipophilic peptide variants had slower absorption and plasma clearance than their respective hydrophilic conjugates, and the absolute bioavailability was also lower likely due to the longer residence times in the skin. Their ease and efficiency make double-click helix stabilization chemistries a useful tool for increasing the bioavailability of peptide therapeutics, many of which suffer from rapid in vivo protease degradation. Helix stabilization using linkers of varying lipophilicity can further control sc absorption and clearance rates to customize plasma pharmacokinetics.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Tejas Navaratna
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Greg M Thurber
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|